151
|
Abstract
Small GTPases use GDP/GTP alternation to actuate a variety of functional switches that are pivotal for cell dynamics. The GTPase switch is turned on by GEFs, which stimulate dissociation of the tightly bound GDP, and turned off by GAPs, which accelerate the intrinsically sluggish hydrolysis of GTP. For Ras, Rho, and Rab GTPases, this switch incorporates a membrane/cytosol alternation regulated by GDIs and GDI-like proteins. The structures and core mechanisms of representative members of small GTPase regulators from most families have now been elucidated, illuminating their general traits combined with scores of unique features. Recent studies reveal that small GTPase regulators have themselves unexpectedly sophisticated regulatory mechanisms, by which they process cellular signals and build up specific cell responses. These mechanisms include multilayered autoinhibition with stepwise release, feedback loops mediated by the activated GTPase, feed-forward signaling flow between regulators and effectors, and a phosphorylation code for RhoGDIs. The flipside of these highly integrated functions is that they make small GTPase regulators susceptible to biochemical abnormalities that are directly correlated with diseases, notably a striking number of missense mutations in congenital diseases, and susceptible to bacterial mimics of GEFs, GAPs, and GDIs that take command of small GTPases in infections. This review presents an overview of the current knowledge of these many facets of small GTPase regulation.
Collapse
Affiliation(s)
- Jacqueline Cherfils
- Laboratoire d’Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique, Centre deRecherche de Gif, Gif-sur-Yvette, France
| | | |
Collapse
|
152
|
Lu H, Jiao Q, Wang Y, Yang Z, Feng M, Wang L, Chen X, Jin W, Liu Y. The mental retardation-associated protein srGAP3 regulates survival, proliferation, and differentiation of rat embryonic neural stem/progenitor cells. Stem Cells Dev 2013; 22:1709-16. [PMID: 23311320 DOI: 10.1089/scd.2012.0455] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The mental retardation-associated protein, srGAP3 is highly expressed in neurogenic sites. It is thought to regulate the key aspects of neuronal development and functions. Little is known about the interaction between srGAP3 and immature neural stem cells/neural progenitor cells (NSCs/NPCs). In the current study, the expression of srGAP3 in NSCs/NPCs was detected. Then, survival, proliferation, differentiation, and morphological alteration of NSCs/NPCs were assessed after a lentivirus-mediated knockdown of srGAP3. The results showed that srGAP3 is highly expressed in NSCs/NPCs both in vitro and in vivo. After knockdown of srGAP3 (LV3-srGAP3 infection), viability and proliferation of NSCs/NPCs dramatically decreased, approximately 85% displayed a similar morphology with type I cells that have no or only few indistinguishable processes. After 7 days culture in a differentiation medium, 62.5%±8.3% of cells in the srGAP3 knockdown group were nestin-positive and 24.8%±5.8% of them were β-tubulin III-positive, which are significantly higher (30.2%±9.9% and 14.6%±2.7%) than in the control group (LV3-NC infection). In addition, cells in the knockdown group had significantly fewer, but longer processes. Our results demonstrate that srGAP3 knockdown negatively regulates NSCs/NPCs survival, proliferation, differentiation, and morphological alteration, particularly, process formation. Taken together, our results provide strong evidence that srGAP3 is involved in the regulation of biological behavior and the morphological features in rat NSCs/NPCs in vitro.
Collapse
Affiliation(s)
- Haixia Lu
- Institute of Neurobiology, Xi'an Jiaotong University College of Medicine, Xi'an, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Ding Y, Zhang R, Zhang K, Lv X, Chen Y, Li A, Wang L, Zhang X, Xia Q. Nischarin is differentially expressed in rat brain and regulates neuronal migration. PLoS One 2013; 8:e54563. [PMID: 23342172 PMCID: PMC3546990 DOI: 10.1371/journal.pone.0054563] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 12/13/2012] [Indexed: 11/19/2022] Open
Abstract
Nischarin is a protein known to inhibit breast cancer cell motility by regulating the signaling of the Rho GTPase family. However, little is known about its location and function in the nervous system. The aim of the present study was to investigate the regional and cellular expression and functions of Nischarin in the adult rodent brain. As assessed by real-time PCR, Western blot analysis and immunostaining, we found that Nischarin was widely distributed throughout the brain, with a higher expression in the cerebral cortex and hippocampus. Double-labeling showed that Nischarin was expressed in neurons and was mainly located in the perinuclear region and F-actin-rich protrusions. The expression pattern of Nischarin in the brain was thought to be closely associated with its function. This was verified by our findings from cell migration assays that Nischarin regulated neuronal migration. These results provide a preliminary survey of the distribution of Nischarin in different regions and cell types in the rat brain. This might help to elucidate its physiological roles, and to evaluate its potential clinical implications.
Collapse
Affiliation(s)
- Yuemin Ding
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Ruyi Zhang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Kena Zhang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyou Lv
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanan Chen
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Aiqing Li
- Gastroenterology Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linlin Wang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiong Zhang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Xia
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
- * E-mail:
| |
Collapse
|
154
|
Bacon C, Endris V, Rappold GA. The cellular function of srGAP3 and its role in neuronal morphogenesis. Mech Dev 2012; 130:391-5. [PMID: 23127797 DOI: 10.1016/j.mod.2012.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 10/15/2012] [Accepted: 10/17/2012] [Indexed: 10/27/2022]
Abstract
The Slit-Robo GTPase activating protein 3 (srGAP3) dynamically regulates cytoskeletal reorganisation through inhibition of the Rho GTPase Rac1 and interaction with actin remodelling proteins. SrGAP3-mediated reorganisation of the actin cytoskeleton is crucial for the normal development of dendritic spines and loss of srGAP3 leads to abnormal synaptic activity and impaired cognitive behaviours in mice, which is reminiscent of an association between disrupted srGAP3 and intellectual disability in humans. Additionally, srGAP3 has been implicated to act downstream of Slit-Robo signalling in commissural axons of the spinal cord. Thus, srGAP3-mediated cytoskeletal reorganisation has an important influence on a variety of neurodevelopmental processes, which may be required for normal cognitive function.
Collapse
Affiliation(s)
- Claire Bacon
- Department of Human Molecular Genetics, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
155
|
Soria Fregozo C, Pérez Vega M. Actin-binding proteins and signalling pathways associated with the formation and maintenance of dendritic spines. NEUROLOGÍA (ENGLISH EDITION) 2012. [DOI: 10.1016/j.nrleng.2011.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
156
|
Siudeja K, Grzeschik NA, Rana A, de Jong J, Sibon OCM. Cofilin/Twinstar phosphorylation levels increase in response to impaired coenzyme a metabolism. PLoS One 2012; 7:e43145. [PMID: 22912811 PMCID: PMC3422318 DOI: 10.1371/journal.pone.0043145] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 07/19/2012] [Indexed: 11/19/2022] Open
Abstract
Coenzyme A (CoA) is a pantothenic acid-derived metabolite essential for many fundamental cellular processes including energy, lipid and amino acid metabolism. Pantothenate kinase (PANK), which catalyses the first step in the conversion of pantothenic acid to CoA, has been associated with a rare neurodegenerative disorder PKAN. However, the consequences of impaired PANK activity are poorly understood. Here we use Drosophila and human neuronal cell cultures to show how PANK deficiency leads to abnormalities in F-actin organization. Cells with reduced PANK activity are characterized by abnormally high levels of phosphorylated cofilin, a conserved actin filament severing protein. The increased levels of phospho-cofilin coincide with morphological changes of PANK-deficient Drosophila S2 cells and human neuronal SHSY-5Y cells. The latter exhibit also markedly reduced ability to form neurites in culture--a process that is strongly dependent on actin remodeling. Our results reveal a novel and conserved link between a metabolic biosynthesis pathway, and regulation of cellular actin dynamics.
Collapse
Affiliation(s)
| | | | | | | | - Ody C. M. Sibon
- Department of Cell Biology, Radiation and Stress Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
157
|
Asensio-Juan E, Gallego C, Martínez-Balbás MA. The histone demethylase PHF8 is essential for cytoskeleton dynamics. Nucleic Acids Res 2012; 40:9429-40. [PMID: 22850744 PMCID: PMC3479184 DOI: 10.1093/nar/gks716] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
PHF8 is a histone demethylase associated with X-linked mental retardation. It has been described as a transcriptional co-activator involved in cell cycle progression, but its physiological role is still poorly understood. Here we show that PHF8 controls the expression of genes involved in cell adhesion and cytoskeleton organization such as RhoA, Rac1 and GSK3β. A lack of PHF8 not only results in a cell cycle delay but also in a disorganized actin cytoskeleton and impaired cell adhesion. Our data demonstrate that PHF8 directly regulates the expression of these genes by demethylating H4K20me1 at promoters. Moreover, c-Myc transcription factor cooperates with PHF8 to regulate the analysed promoters. Further analysis in neurons shows that depletion of PHF8 results in down-regulation of cytoskeleton genes and leads to a deficient neurite outgrowth. Overall, our results suggest that the mental retardation phenotype associated with loss of function of PHF8 could be due to abnormal neuronal connections as a result of alterations in cytoskeleton function.
Collapse
Affiliation(s)
- Elena Asensio-Juan
- Department of Molecular Genomics, Instituto de Biología Molecular de Barcelona, Spanish Research Council (CSIC), Barcelona Science Park, Barcelona 08028, Spain
| | | | | |
Collapse
|
158
|
Rosário M, Schuster S, Jüttner R, Parthasarathy S, Tarabykin V, Birchmeier W. Neocortical dendritic complexity is controlled during development by NOMA-GAP-dependent inhibition of Cdc42 and activation of cofilin. Genes Dev 2012; 26:1743-57. [PMID: 22810622 DOI: 10.1101/gad.191593.112] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neocortical neurons have highly branched dendritic trees that are essential for their function. Indeed, defects in dendritic arborization are associated with human neurodevelopmental disorders. The molecular mechanisms regulating dendritic arbor complexity, however, are still poorly understood. Here, we uncover the molecular basis for the regulation of dendritic branching during cortical development. We show that during development, dendritic branching requires post-mitotic suppression of the RhoGTPase Cdc42. By generating genetically modified mice, we demonstrate that this is catalyzed in vivo by the novel Cdc42-GAP NOMA-GAP. Loss of NOMA-GAP leads to decreased neocortical volume, associated specifically with profound oversimplification of cortical dendritic arborization and hyperactivation of Cdc42. Remarkably, dendritic complexity and cortical thickness can be partially restored by genetic reduction of post-mitotic Cdc42 levels. Furthermore, we identify the actin regulator cofilin as a key regulator of dendritic complexity in vivo. Cofilin activation during late cortical development depends on NOMA-GAP expression and subsequent inhibition of Cdc42. Strikingly, in utero expression of active cofilin is sufficient to restore postnatal dendritic complexity in NOMA-GAP-deficient animals. Our findings define a novel cell-intrinsic mechanism to regulate dendritic branching and thus neuronal complexity in the cerebral cortex.
Collapse
Affiliation(s)
- Marta Rosário
- Neurocure Excellence Cluster, Institute of Cell and Neurobiology, Charité Universitätsmedizin Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
159
|
Pozueta J, Lefort R, Shelanski ML. Synaptic changes in Alzheimer's disease and its models. Neuroscience 2012; 251:51-65. [PMID: 22687952 DOI: 10.1016/j.neuroscience.2012.05.050] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 05/14/2012] [Accepted: 05/17/2012] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder characterized by a progressive loss of cognition and the presence of two hallmark lesions, senile plaques (SP) and neurofibrillary tangles (NFT), which result from the accumulation and deposition of the β-amyloid peptide (Aβ) and the aggregation of hyperphosphorylated tau protein, respectively. Initially, it was thought that Aβ fibrils, which make up SP, were the root cause of the massive neurodegeneration usual found in AD brains. Over time, the longstanding emphasis on fibrillar Aβ deposits and neuronal death slowly gave way to a new paradigm involving soluble oligomeric forms of Aβ, which play a prominent role in triggering the cognitive deficits by specifically targeting synapses and disrupting synaptic signaling pathways. While this paradigm is widely accepted today in the AD field, the molecular details have not been fully elucidated. In this review, we address some of the important evidence, which has led to the Aβ oligomer-centric hypothesis as well as some of the key findings concerning the effects of Aβ oligomers on synapses at a morphological and functional level. Understanding how Aβ oligomers target synapses provides an important framework for ongoing AD research, which can lead to the development of successful therapeutic strategies designed to alter or perhaps reverse the course of the disease.
Collapse
Affiliation(s)
- J Pozueta
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain and Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, United States
| | | | | |
Collapse
|
160
|
Borrelli S, Musilli M, Martino A, Diana G. Long-lasting efficacy of the cognitive enhancer cytotoxic necrotizing factor 1. Neuropharmacology 2012; 64:74-80. [PMID: 22659407 DOI: 10.1016/j.neuropharm.2012.05.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/08/2012] [Accepted: 05/22/2012] [Indexed: 11/29/2022]
Abstract
Rho GTPases are key regulators of the activity-dependent changes of neural circuits. Besides being involved in nervous system development and repair, this neural structural plasticity is believed to constitute the cellular basis of learning and memory. Here we report that concurrent modulation of cerebral Rho GTPases, including Rac, Rho and Cdc42 subfamilies, by Cytotoxic Necrotizing Factor 1 (CNF1, 10 fmol/kg intracerebroventricularly) improves object recognition in both C57BL/6J and CD1 mice. The improvement is long lasting, as it is still observed 90 days post treatment. At this time, the treatment is associated with enhancement of neurotransmission and long-term potentiation. The effects depend on changes in Rho GTPase status, since the recombinant molecule CNF1 C866S, in which the enzymatic activity was abolished through substitution of serine to cysteine at position 866, is ineffective. The study confirms the role of Rho GTPases in learning and suggests that a single administration of CNF1 is effective for a long time after administration. In general, the long-lasting cognition enhancing effect of CNF1 might be beneficial for the treatment of CNS disorders. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Sonia Borrelli
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | | | | | | |
Collapse
|
161
|
Liu M, Bi F, Zhou X, Zheng Y. Rho GTPase regulation by miRNAs and covalent modifications. Trends Cell Biol 2012; 22:365-73. [PMID: 22572609 DOI: 10.1016/j.tcb.2012.04.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/05/2012] [Accepted: 04/10/2012] [Indexed: 12/15/2022]
Abstract
To date, most studies of Rho GTPase regulation have focused on the classic GTPase cycle - GTP binding and hydrolysis - controlled by guanine nucleotide exchange factors (GEFs), GTPase-activating proteins (GAPs) and GDP-dissociation inhibitors (GDIs). Recent investigations have unveiled important additional regulatory mechanisms: microRNA (miRNA) regulating post-transcriptional processing of Rho GTPase-encoding mRNAs; palmitoylation and nuclear targeting affecting intracellular distribution; post-translational phosphorylation, transglutamination and AMPylation impacting Rho GTPase signaling; and ubiquitination controlling Rho GTPase protein stability and turnover. These modes of regulation add to the complexity of the Rho GTPase signaling network and allow precise spatiotemporal control of individual Rho GTPases. This review discusses these 'unconventional' modes of regulation and their contribution to cellular function, focusing on post-transcriptional and post-translational events beyond the classic GTPase cycle regulatory model.
Collapse
Affiliation(s)
- Ming Liu
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
162
|
Sensitive time-windows for susceptibility in neurodevelopmental disorders. Trends Neurosci 2012; 35:335-44. [PMID: 22542246 DOI: 10.1016/j.tins.2012.03.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 02/22/2012] [Accepted: 03/09/2012] [Indexed: 11/20/2022]
Abstract
Many neurodevelopmental disorders (NDDs) are characterized by age-dependent symptom onset and regression, particularly during early postnatal periods of life. The neurobiological mechanisms preceding and underlying these developmental cognitive and behavioral impairments are, however, not clearly understood. Recent evidence using animal models for monogenic NDDs demonstrates the existence of time-regulated windows of neuronal and synaptic impairments. We propose that these developmentally-dependent impairments can be unified into a key concept: namely, time-restricted windows for impaired synaptic phenotypes exist in NDDs, akin to critical periods during normal sensory development in the brain. Existence of sensitive time-windows has significant implications for our understanding of early brain development underlying NDDs and may indicate vulnerable periods when the brain is more susceptible to current therapeutic treatments.
Collapse
|
163
|
5-HT7R/G12 signaling regulates neuronal morphology and function in an age-dependent manner. J Neurosci 2012; 32:2915-30. [PMID: 22378867 DOI: 10.1523/jneurosci.2765-11.2012] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The common neurotransmitter serotonin controls different aspects of early neuronal differentiation, although the underlying mechanisms are poorly understood. Here we report that activation of the serotonin 5-HT(7) receptor promotes synaptogenesis and enhances synaptic activity in hippocampal neurons at early postnatal stages. An analysis of Gα(12)-deficient mice reveals a critical role of G(12)-protein for 5-HT(7) receptor-mediated effects in neurons. In organotypic preparations from the hippocampus of juvenile mice, stimulation of 5-HT(7)R/G(12) signaling potentiates formation of dendritic spines, increases neuronal excitability, and modulates synaptic plasticity. In contrast, in older neuronal preparations, morphogenetic and synaptogenic effects of 5-HT(7)/G(12) signaling are abolished. Moreover, inhibition of 5-HT(7) receptor had no effect on synaptic plasticity in hippocampus of adult animals. Expression analysis reveals that the production of 5-HT(7) and Gα(12)-proteins in the hippocampus undergoes strong regulation with a pronounced transient increase during early postnatal stages. Thus, regulated expression of 5-HT(7) receptor and Gα(12)-protein may represent a molecular mechanism by which serotonin specifically modulates formation of initial neuronal networks during early postnatal development.
Collapse
|
164
|
Bae J, Sung BH, Cho IH, Kim SM, Song WK. NESH regulates dendritic spine morphology and synapse formation. PLoS One 2012; 7:e34677. [PMID: 22485184 PMCID: PMC3317636 DOI: 10.1371/journal.pone.0034677] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/06/2012] [Indexed: 11/18/2022] Open
Abstract
Background Dendritic spines are small membranous protrusions on the neuronal dendrites that receive synaptic input from axon terminals. Despite their importance for integrating the enormous information flow in the brain, the molecular mechanisms regulating spine morphogenesis are not well understood. NESH/Abi-3 is a member of the Abl interactor (Abi) protein family, and its overexpression is known to reduce cell motility and tumor metastasis. NESH is prominently expressed in the brain, but its function there remains unknown. Methodology/Principal Findings NESH was strongly expressed in the hippocampus and moderately expressed in the cerebral cortex, cerebellum and striatum, where it co-localized with the postsynaptic proteins PSD95, SPIN90 and F-actin in dendritic spines. Overexpression of NESH reduced numbers of mushroom-type spines and synapse density but increased thin, filopodia-like spines and had no effect on spine density. siRNA knockdown of NESH also reduced mushroom spine numbers and inhibited synapse formation but it increased spine density. The N-terminal region of NESH co-sedimented with filamentous actin (F-actin), which is an essential component of dendritic spines, suggesting this interaction is important for the maturation of dendritic spines. Conclusions/Significance NESH is a novel F-actin binding protein that likely plays important roles in the regulation of dendritic spine morphogenesis and synapse formation.
Collapse
Affiliation(s)
- Jeomil Bae
- Cell Dynamics and Bioimaging Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Bong Hwan Sung
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - In Ha Cho
- Cell Dynamics and Bioimaging Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Seon-Myung Kim
- Department of Cell Biology and Neurology, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Woo Keun Song
- Cell Dynamics and Bioimaging Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
- * E-mail:
| |
Collapse
|
165
|
Iyer SC, Wang D, Iyer EPR, Trunnell SA, Meduri R, Shinwari R, Sulkowski MJ, Cox DN. The RhoGEF trio functions in sculpting class specific dendrite morphogenesis in Drosophila sensory neurons. PLoS One 2012; 7:e33634. [PMID: 22442703 PMCID: PMC3307743 DOI: 10.1371/journal.pone.0033634] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 02/16/2012] [Indexed: 12/23/2022] Open
Abstract
Background As the primary sites of synaptic or sensory input in the nervous system, dendrites play an essential role in processing neuronal and sensory information. Moreover, the specification of class specific dendrite arborization is critically important in establishing neural connectivity and the formation of functional networks. Cytoskeletal modulation provides a key mechanism for establishing, as well as reorganizing, dendritic morphology among distinct neuronal subtypes. While previous studies have established differential roles for the small GTPases Rac and Rho in mediating dendrite morphogenesis, little is known regarding the direct regulators of these genes in mediating distinct dendritic architectures. Methodology/Principal Findings Here we demonstrate that the RhoGEF Trio is required for the specification of class specific dendritic morphology in dendritic arborization (da) sensory neurons of the Drosophila peripheral nervous system (PNS). Trio is expressed in all da neuron subclasses and loss-of-function analyses indicate that Trio functions cell-autonomously in promoting dendritic branching, field coverage, and refining dendritic outgrowth in various da neuron subtypes. Moreover, overexpression studies demonstrate that Trio acts to promote higher order dendritic branching, including the formation of dendritic filopodia, through Trio GEF1-dependent interactions with Rac1, whereas Trio GEF-2-dependent interactions with Rho1 serve to restrict dendritic extension and higher order branching in da neurons. Finally, we show that de novo dendritic branching, induced by the homeodomain transcription factor Cut, requires Trio activity suggesting these molecules may act in a pathway to mediate dendrite morphogenesis. Conclusions/Significance Collectively, our analyses implicate Trio as an important regulator of class specific da neuron dendrite morphogenesis via interactions with Rac1 and Rho1 and indicate that Trio is required as downstream effector in Cut-mediated regulation of dendrite branching and filopodia formation.
Collapse
Affiliation(s)
- Srividya Chandramouli Iyer
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Dennis Wang
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Eswar Prasad R. Iyer
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Sarah A. Trunnell
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Ramakrishna Meduri
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Riaz Shinwari
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Mikolaj J. Sulkowski
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Daniel N. Cox
- School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
166
|
Li L, Zhang W, Cheng S, Cao D, Parent M. Isoprenoids and related pharmacological interventions: potential application in Alzheimer's disease. Mol Neurobiol 2012; 46:64-77. [PMID: 22418893 DOI: 10.1007/s12035-012-8253-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/28/2012] [Indexed: 12/18/2022]
Abstract
Two major isoprenoids, farnesyl pyrophosphate and geranylgeranyl pyrophosphate, serve as lipid donors for the posttranslational modification (known as prenylation) of proteins that possess a characteristic C-terminal motif. The prenylation reaction is catalyzed by prenyltransferases. The lipid prenyl group facilitates to anchor the proteins in cell membranes and mediates protein-protein interactions. A variety of important intracellular proteins undergo prenylation, including almost all members of small GTPase superfamilies as well as heterotrimeric G protein subunits and nuclear lamins. These prenylated proteins are involved in regulating a wide range of cellular processes and functions, such as cell growth, differentiation, cytoskeletal organization, and vesicle trafficking. Prenylated proteins are also implicated in the pathogenesis of different types of diseases. Consequently, isoprenoids and/or prenyltransferases have emerged as attractive therapeutic targets for combating various disorders. This review attempts to summarize the pharmacological agents currently available or under development that control isoprenoid availability and/or the process of prenylation, mainly focusing on statins, bisphosphonates, and prenyltransferase inhibitors. Whereas statins and bisphosphonates deplete the production of isoprenoids by inhibiting the activity of upstream enzymes, prenyltransferase inhibitors directly block the prenylation of proteins. As the importance of isoprenoids and prenylated proteins in health and disease continues to emerge, the therapeutic potential of these pharmacological agents has expanded across multiple disciplines. This review mainly discusses their potential application in Alzheimer's disease.
Collapse
Affiliation(s)
- Ling Li
- Department of Experimental and Clinical Pharmacology, University of Minnesota, 2001 6th St SE, MTRF 4-208, Minneapolis, MN 55455, USA.
| | | | | | | | | |
Collapse
|
167
|
Iwai T, Saitoh A, Yamada M, Takahashi K, Hashimoto E, Ukai W, Saito T, Yamada M. Rhotekin modulates differentiation of cultured neural stem cells to neurons. J Neurosci Res 2012; 90:1359-66. [DOI: 10.1002/jnr.23029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 12/19/2011] [Accepted: 12/19/2011] [Indexed: 11/10/2022]
|
168
|
Christoffel DJ, Golden SA, Russo SJ. Structural and synaptic plasticity in stress-related disorders. Rev Neurosci 2012; 22:535-49. [PMID: 21967517 DOI: 10.1515/rns.2011.044] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract Stress can have a lasting impact on the structure and function of brain circuitry that results in long-lasting changes in the behavior of an organism. Synaptic plasticity is the mechanism by which information is stored and maintained within individual synapses, neurons, and neuronal circuits to guide the behavior of an organism. Although these mechanisms allow the organism to adapt to its constantly evolving environment, not all of these adaptations are beneficial. Under prolonged bouts of physical or psychological stress, these mechanisms become dysregulated, and the connectivity between brain regions becomes unbalanced, resulting in pathological behaviors. In this review, we highlight the effects of stress on the structure and function of neurons within the mesocorticolimbic brain systems known to regulate mood and motivation. We then discuss the implications of these spine adaptations on neuronal activity and pathological behaviors implicated in mood disorders. Finally, we end by discussing recent brain imaging studies in human depression within the context of these basic findings to provide insight into the underlying mechanisms leading to neural dysfunction in depression.
Collapse
Affiliation(s)
- Daniel J Christoffel
- Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029-6574, USA
| | | | | |
Collapse
|
169
|
Seabold GK, Wang PY, Petralia RS, Chang K, Zhou A, McDermott MI, Wang YX, Milgram SL, Wenthold RJ. Dileucine and PDZ-binding motifs mediate synaptic adhesion-like molecule 1 (SALM1) trafficking in hippocampal neurons. J Biol Chem 2012; 287:4470-84. [PMID: 22174418 PMCID: PMC3281672 DOI: 10.1074/jbc.m111.279661] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/22/2011] [Indexed: 12/18/2022] Open
Abstract
Synaptic adhesion-like molecules (SALMs) are a family of cell adhesion molecules involved in neurite outgrowth and synapse formation. Of the five family members, only SALM1, -2, and -3 contain a cytoplasmic C-terminal PDZ-binding motif. We have found that SALM1 is unique among the SALMs because deletion of its PDZ-binding motif (SALM1ΔPDZ) blocks its surface expression in heterologous cells. When expressed in hippocampal neurons, SALM1ΔPDZ had decreased surface expression in dendrites and the cell soma but not in axons, suggesting that the PDZ-binding domain may influence cellular trafficking of SALMs to specific neuronal locations. Endoglycosidase H digestion assays indicated that SALM1ΔPDZ is retained in the endoplasmic reticulum (ER) in heterologous cells. However, when the entire C-terminal tail of SALM1 was deleted, SALM1 was detected on the cell surface. Using serial deletions, we identified a region of SALM1 that contains a putative dileucine ER retention motif, which is not present in the other SALMs. Mutation of this DXXXLL motif allowed SALM1 to leave the ER and enhanced its surface expression in heterologous cells and neurons. An increase in the number of protrusions at the dendrites and cell body was observed when this SALM1 mutant was expressed in hippocampal neurons. With electron microscopy, these protrusions appeared to be irregular, enlarged spines and filopodia. Thus, enrichment of SALM1 on the cell surface affects dendritic arborization, and intracellular motifs regulate its dendritic versus axonal localization.
Collapse
Affiliation(s)
- Gail K Seabold
- Laboratory of Neurochemistry, NIDCD/National Institutes of Health, 50 South Dr., Bldg. 50, Rm. 4144, Bethesda, MD20892-8027, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Dobrin SE, Fahrbach SE. Rho GTPase activity in the honey bee mushroom bodies is correlated with age and foraging experience. JOURNAL OF INSECT PHYSIOLOGY 2012; 58:228-234. [PMID: 22108023 PMCID: PMC3256268 DOI: 10.1016/j.jinsphys.2011.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 11/08/2011] [Accepted: 11/09/2011] [Indexed: 05/31/2023]
Abstract
Foraging experience is correlated with structural plasticity of the mushroom bodies of the honey bee brain. While several neurotransmitter and intracellular signaling pathways have been previously implicated as mediators of these structural changes, none interact directly with the cytoskeleton, the ultimate effector of changes in neuronal morphology. The Rho family of GTPases are small, monomeric G proteins that, when activated, initiate a signaling cascade that reorganizes the neuronal cytoskeleton. In this study, we measured activity of two members of the Rho family of GTPases, Rac and RhoA, in the mushroom bodies of bees with different durations of foraging experience. A transient increase in Rac activity coupled with a transient decrease in RhoA activity was found in honey bees with 4 days foraging experience compared with same-aged new foragers. These observations are in accord with previous reports based on studies of other species of a growth supporting role for Rac and a growth opposing role for RhoA. This is the first report of Rho GTPase activation in the honey bee brain.
Collapse
Affiliation(s)
- Scott E Dobrin
- Neuroscience Program, Wake Forest University, Graduate School of Arts and Sciences, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
171
|
Perez SE, Getova DP, He B, Counts SE, Geula C, Desire L, Coutadeur S, Peillon H, Ginsberg SD, Mufson EJ. Rac1b increases with progressive tau pathology within cholinergic nucleus basalis neurons in Alzheimer's disease. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:526-40. [PMID: 22142809 PMCID: PMC3349868 DOI: 10.1016/j.ajpath.2011.10.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 10/05/2011] [Accepted: 10/11/2011] [Indexed: 01/19/2023]
Abstract
Cholinergic basal forebrain (CBF) nucleus basalis (NB) neurons display neurofibrillary tangles (NFTs) during Alzheimer's disease (AD) progression, yet the mechanisms underlying this selective vulnerability are currently unclear. Rac1, a member of the Rho family of GTPases, may interact with the proapoptotic pan-neurotrophin receptor p75(NTR) to induce neuronal cytoskeletal abnormalities in AD NB neurons. Herein, we examined the expression of Rac1b, a constitutively active splice variant of Rac1, in NB cholinergic neurons during AD progression. CBF tissues harvested from people who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment, or AD were immunolabeled for both p75(NTR) and Rac1b. Rac1b appeared as cytoplasmic diffuse granules, loosely aggregated filaments, or compact spheres in p75(NTR)-positive NB neurons. Although Rac1b colocalized with tau cytoskeletal markers, the percentage of p75(NTR)-immunoreactive neurons expressing Rac1b was significantly increased only in AD compared with both mild cognitive impairment and NCI. Furthermore, single-cell gene expression profiling with custom-designed microarrays showed down-regulation of caveolin 2, GNB4, and lipase A in AD Rac1b-positive/p75(NTR)-labeled NB neurons compared with Rac1b-negative/p75(NTR)-positive perikarya in NCI. These proteins are involved in Rac1 pathway/cell cycle progression and lipid metabolism. These data suggest that Rac1b expression acts as a modulator or transducer of various signaling pathways that lead to NFT formation and membrane dysfunction in a subgroup of CBF NB neurons in AD.
Collapse
Affiliation(s)
- Sylvia E. Perez
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Damianka P. Getova
- Department of Pharmacology and Clinical Pharmacology, Medical University, Plovdiv, Bulgaria
| | - Bin He
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Scott E. Counts
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Changiz Geula
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Chicago Illinois
| | | | | | | | - Stephen D. Ginsberg
- Center for Dementia Research and Departments of Psychiatry and Physiology and Neuroscience, Nathan Kline Institute/New York University Langone Medical Center, New York, New York
| | - Elliott J. Mufson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
172
|
Long H, Zhu X, Yang P, Gao Q, Chen Y, Ma L. Myo9b and RICS modulate dendritic morphology of cortical neurons. ACTA ACUST UNITED AC 2012; 23:71-9. [PMID: 22250289 DOI: 10.1093/cercor/bhr378] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Regulated growth and branching of dendritic processes is critical for the establishment of neuronal circuitry and normal brain functions. Rho family GTPases, including RhoA, Rac1, and Cdc42, play a prominent role in dendritic development. RhoA inhibits dendritic branching and growth, whereas Rac1/Cdc42 does the opposite. It has been suggested that the activity of RhoA must be kept low to allow dendritic growth. However, how neurons restrict the activation of RhoA for proper dendritic development is not clear. In the present study, we undertook a comprehensive loss-of-function analysis of putative RhoA GTPase-activating proteins (RhoA GAPs) in the cortical neurons. The expression of 16 RhoA GAPs was detected in the developing rat brain, and RNA interference experiments suggest that 2 of them, Myo9b and RICS, are critical regulators of dendritic morphogenesis. Knockdown of either Myo9b or RICS in cultured cortical neurons or developing cortex resulted in decreased dendrite length and number. Inhibition of RhoA/ROCK signaling restores the defects of dendritic morphology induced by knockdown of Myo9b or RICS. These data demonstrate that Myo9b and RICS repress RhoA/Rock signaling and modulate dendritic morphogenesis in cortical neurons, providing evidence for critical physiological function of RhoA GAPs in regulation of dendritic development.
Collapse
Affiliation(s)
- Hui Long
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, Shanghai Medical College and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
173
|
GluN3 subunit-containing NMDA receptors: not just one-trick ponies. Trends Neurosci 2012; 35:240-9. [PMID: 22240240 DOI: 10.1016/j.tins.2011.11.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 10/16/2011] [Accepted: 11/27/2011] [Indexed: 02/06/2023]
Abstract
The two GluN3 subunits were the last NMDA receptor subunits to be cloned some 15 years ago. Strikingly, despite the steadily growing interest in their function, their physiological role remains elusive. The original billing as dominant-negative modulators of classical NMDA receptors composed of GluN1 and GluN2 subunits has given way to proposals of much more complex functions, including roles in synaptogenesis and synaptic plasticity. In addition, GluN3 subunits in the absence of GluN2 surprisingly assemble with GluN1 into excitatory glycine receptors. This review provides an overview of the unique spatial and temporal expression patterns of the GluN3 subunits, discusses proposed functions and physiological roles for receptors comprising these subunits, and briefly summarizes their putative involvement in several neural diseases.
Collapse
|
174
|
Abstract
Polarized cellular responses, for example, cell migration, require the co-ordinated assembly of signalling complexes at a particular subcellular location, such as the leading edge of cells. Small GTPases of the Ras superfamily play central roles in many (polarized) responses to growth factors, chemokines or integrin ligands. These small GTPases are functionally distinct, yet remarkably homologous in their primary sequence and especially in their effector domains. Therefore it has long been unclear how GTPase signalling specificity is regulated. Small GTPases carry a lipid anchor, in the context of a hypervariable region, which mediates membrane association. However, whereas the lipid has long been proposed to be the critical regulator of subcellular GTPase targeting, there is now increasing evidence that specific protein-protein interactions are important as well. This review discusses recent findings on GTPase targeting and proposes a revised model for GTPase signalling. In this model, the hypervariable domain acts in conjunction with the lipid tail to target the GTPase to specific membrane-associated protein complexes. Here, local GTPase activation occurs, leading to subsequent exposure of the effector domain, binding to effector proteins and the initiation of downstream signalling.
Collapse
Affiliation(s)
- Jean Paul ten Klooster
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
175
|
Levenga J, Willemsen R. Perturbation of dendritic protrusions in intellectual disability. PROGRESS IN BRAIN RESEARCH 2012; 197:153-68. [PMID: 22541292 DOI: 10.1016/b978-0-444-54299-1.00008-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intellectual disability (ID) affects 1-3% of the general population and is defined by an intelligence quotient score under 70 and the presence of two or more adaptive behaviors. Learning and memory involves the change in the transmission efficacy at the synapse (synaptic plasticity). Synaptic plasticity is the ability of the connection, or synapse, between two functional neurons to change in strength. Many molecular mechanisms are involved in the change in synaptic strength, which can result in changes in spine morphology. Spines are specialized dendritic protrusions and their change in morphology is implicated in learning and memory. In several cases of ID, the link between spine abnormalities (abnormal in number, size, and shape) and ID is well described, including nonsyndromic ID and Down, Fragile X, and Rett syndromes. This chapter discusses the underlying molecular mechanisms of this altered spine phenotype.
Collapse
Affiliation(s)
- Josien Levenga
- Department of Physiology and Neuroscience, New York University, School of Medicine, New York, NY, USA
| | | |
Collapse
|
176
|
Synaptic dysfunction and intellectual disability. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:433-49. [PMID: 22351067 DOI: 10.1007/978-3-7091-0932-8_19] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intellectual disability (ID) is a common and highly heterogeneous paediatric disorder with a very severe social impact. Intellectual disability can be caused by environmental and/or genetic factors. Although in the last two decades a number of genes have been discovered whose mutations cause mental retardation, we are still far from identifying the impact of these mutations on brain functions. Many of the genes mutated in ID code for several proteins with a variety of functions: chromatin remodelling, pre-/post-synaptic activity, and intracellular trafficking. The prevailing hypothesis suggests that the ID phenotype could emerge from abnormal cellular processing leading to pre- and/or post-synaptic dysfunction. In this chapter, we focus on the role of small GTPases and adhesion molecules, and we discuss the mechanisms through which they lead to synaptic network dysfunction.
Collapse
|
177
|
Ligeti E, Welti S, Scheffzek K. Inhibition and Termination of Physiological Responses by GTPase Activating Proteins. Physiol Rev 2012; 92:237-72. [DOI: 10.1152/physrev.00045.2010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Physiological processes are strictly organized in space and time. However, in cell physiology research, more attention is given to the question of space rather than to time. To function as a signal, environmental changes must be restricted in time; they need not only be initiated but also terminated. In this review, we concentrate on the role of one specific protein family involved in biological signal termination. GTPase activating proteins (GAPs) accelerate the endogenously low GTP hydrolysis rate of monomeric guanine nucleotide-binding proteins (GNBPs), limiting thereby their prevalence in the active, GTP-bound form. We discuss cases where defective or excessive GAP activity of specific proteins causes significant alteration in the function of the nervous, endocrine, and hemopoietic systems, or contributes to development of infections and tumors. Biochemical and genetic data as well as observations from human pathology support the notion that GAPs represent vital elements in the spatiotemporal fine tuning of physiological processes.
Collapse
Affiliation(s)
- Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary; Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; and Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Stefan Welti
- Department of Physiology, Semmelweis University, Budapest, Hungary; Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; and Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Klaus Scheffzek
- Department of Physiology, Semmelweis University, Budapest, Hungary; Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; and Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
178
|
Kalirin signaling: implications for synaptic pathology. Mol Neurobiol 2011; 45:109-18. [PMID: 22194219 DOI: 10.1007/s12035-011-8223-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 11/29/2011] [Indexed: 01/31/2023]
Abstract
Spine morphogenesis and plasticity are intimately linked to cognition, and there is strong evidence that aberrant regulation of spine plasticity is associated with physiological, behavioral, and pathological conditions. The neuronal guanine nucleotide exchange factor (GEF) kalirin is emerging as a key regulator of structural and functional plasticity at dendritic spines. Here, we review recent studies that have genetically and functionally linked kalirin signaling to a number of human disorders. Kalirin signaling may thus represent a disease mechanism and provide a novel therapeutic target.
Collapse
|
179
|
Soria Fregozo C, Pérez Vega MI. Actin-binding proteins and signalling pathways associated with the formation and maintenance of dendritic spines. Neurologia 2011; 27:421-31. [PMID: 22178050 DOI: 10.1016/j.nrl.2011.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 10/09/2011] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Dendritic spines are the main sites of excitatory synaptic contacts. Moreover, they present plastic responses to different stimuli present in synaptic activity or damage, ranging from an increase or decrease in their total number, to redistribution of progenitor dendritic spines, to variations in their size or shape. However, the spines can remain stable for a long time. BACKGROUND The use of experimental models has shown that different molecules of the F-actin binding and signalling pathways are closely related to the development, maintenance and plasticity of excitatory synapses, which could affect the number, size and shape of the dendritic spines; these mechanisms affect and depend on the reorganisation of the actin cytoskeleton. DEVELOPMENT It is proposed that the filopodia are precursors of dendritic spines. Drebrin is an F-actin binding protein, and it is responsible for concentrating F-actin and PSD-95 in filopodia that will guide the formation of the new spines. CONCLUSION The specific mechanisms of actin regulation are an integral part in the formation, maturing process and plasticity of dendritic spines in association with the various actin cytoskeleton-binding proteins The signalling pathways mediated by small GTPases and the equilibrium between G-actin and F-actin are also involved.
Collapse
Affiliation(s)
- C Soria Fregozo
- Laboratorio de Psicobiología y Biología Molecular, Departamento de Ciencias de la Tierra y de la Vida, Centro Universitario de los Lagos, Universidad de Guadalajara, Guadalajara, Mexico.
| | | |
Collapse
|
180
|
Sheng M, Kim E. The postsynaptic organization of synapses. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a005678. [PMID: 22046028 DOI: 10.1101/cshperspect.a005678] [Citation(s) in RCA: 390] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The postsynaptic side of the synapse is specialized to receive the neurotransmitter signal released from the presynaptic terminal and transduce it into electrical and biochemical changes in the postsynaptic cell. The cardinal functional components of the postsynaptic specialization of excitatory and inhibitory synapses are the ionotropic receptors (ligand-gated channels) for glutamate and γ-aminobutyric acid (GABA), respectively. These receptor channels are concentrated at the postsynaptic membrane and embedded in a dense and rich protein network comprised of anchoring and scaffolding molecules, signaling enzymes, cytoskeletal components, as well as other membrane proteins. Excitatory and inhibitory postsynaptic specializations are quite different in molecular organization. The postsynaptic density of excitatory synapses is especially complex and dynamic in composition and regulation; it contains hundreds of different proteins, many of which are required for cognitive function and implicated in psychiatric illness.
Collapse
Affiliation(s)
- Morgan Sheng
- The Department of Neuroscience, Genentech Incorporated, San Francisco, California 94080, USA
| | | |
Collapse
|
181
|
Powell AD, Gill KK, Saintot PP, Jiruska P, Chelly J, Billuart P, Jefferys JGR. Rapid reversal of impaired inhibitory and excitatory transmission but not spine dysgenesis in a mouse model of mental retardation. J Physiol 2011; 590:763-76. [PMID: 22124149 DOI: 10.1113/jphysiol.2011.219907] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Intellectual disability affects 2-3% of the population: those due to mutations of the X-chromosome are a major cause of moderate to severe cases (1.8/1000 males). Established theories ascribe the cellular aetiology of intellectual disability to malformations of dendritic spines. Recent work has identified changes in synaptic physiology in some experimental models. Here, we investigated the pathophysiology of a mouse model of intellectual disability using electrophysiological recordings combined with confocal imaging of dentate gyrus granule neurons. Lack of oligophrenin-1 resulted in reductions in dendritic tree complexity and mature dendritic spine density and in evoked and spontaneous EPSCs and IPSCs. In the case of inhibitory transmission, the physiological change was associated with a reduction in the readily releasable pool and vesicle recycling which impaired the efficiency of inhibitory synaptic transmission. Acute inhibition of the downstream signalling pathway of oligophrenin-1 fully reversed the functional changes in synaptic transmission but not the dendritic abnormalities. The impaired inhibitory (as well as excitatory) synaptic transmission at frequencies associated with cognitive function suggests a cellular mechanism for the intellectual disability, because cortical oscillations associated with cognition normally depend on inhibitory neurons firing on every cycle.
Collapse
Affiliation(s)
- Andrew D Powell
- School of Clinical and Experimental Medicine (Neuronal Networks Group), College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | | | | | | | | | |
Collapse
|
182
|
Chen Y, Wang F, Long H, Chen Y, Wu Z, Ma L. GRK5 promotes F-actin bundling and targets bundles to membrane structures to control neuronal morphogenesis. ACTA ACUST UNITED AC 2011; 194:905-20. [PMID: 21930777 PMCID: PMC3207290 DOI: 10.1083/jcb.201104114] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neuronal morphogenesis requires extensive membrane remodeling and cytoskeleton dynamics. In this paper, we show that GRK5, a G protein-coupled receptor kinase, is critically involved in neurite outgrowth, dendrite branching, and spine morphogenesis through promotion of filopodial protrusion. Interestingly, GRK5 is not acting as a kinase but rather provides a key link between the plasma membrane and the actin cytoskeleton. GRK5 promoted filamentous actin (F-actin) bundling at the membranes of dynamic neuronal structures by interacting with both F-actin and phosphatidylinositol-4,5-bisphosphate. Moreover, separate domains of GRK5 mediated the coupling of actin cytoskeleton dynamics and membrane remodeling and were required for its effects on neuronal morphogenesis. Accordingly, GRK5 knockout mice exhibited immature spine morphology and deficient learning and memory. Our findings identify GRK5 as a critical mediator of dendritic development and suggest that coordinated actin cytoskeleton and membrane remodeling mediated by bifunctional actin-bundling and membrane-targeting molecules, such as GRK5, is crucial for proper neuronal morphogenesis and the establishment of functional neuronal circuitry.
Collapse
Affiliation(s)
- Yuejun Chen
- The State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
183
|
Maintenance of dendritic spine morphology by partitioning-defective 1b through regulation of microtubule growth. J Neurosci 2011; 31:12094-103. [PMID: 21865452 DOI: 10.1523/jneurosci.0751-11.2011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Dendritic spines are postsynaptic structures that receive excitatory synaptic input from presynaptic terminals. Actin and its regulatory proteins play a central role in morphogenesis of dendritic spines. In addition, recent studies have revealed that microtubules are indispensable for the maintenance of mature dendritic spine morphology by stochastically invading dendritic spines and regulating dendritic localization of p140Cap, which is required for actin reorganization. However, the regulatory mechanisms of microtubule dynamics remain poorly understood. Partitioning-defective 1b (PAR1b), a cell polarity-regulating serine/threonine protein kinase, is thought to regulate microtubule dynamics by inhibiting microtubule binding of microtubule-associated proteins. Results from the present study demonstrated that PAR1b participates in the maintenance of mature dendritic spine morphology in mouse hippocampal neurons. Immunofluorescent analysis revealed PAR1b localization in the dendrites, which was concentrated in dendritic spines of mature neurons. PAR1b knock-down cells exhibited decreased mushroom-like dendritic spines, as well as increased filopodia-like dendritic protrusions, with no effect on the number of protrusions. Live imaging of microtubule plus-end tracking proteins directly revealed decreases in distance and duration of microtubule growth following PAR1b knockdown in a neuroblastoma cell line and in dendrites of hippocampal neurons. In addition, reduced accumulation of GFP-p140Cap in dendritic protrusions was confirmed in PAR1b knock-down neurons. In conclusion, the present results suggested a novel function for PAR1b in the maintenance of mature dendritic spine morphology by regulating microtubule growth and the accumulation of p140Cap in dendritic spines.
Collapse
|
184
|
Sultana R, Robinson RAS, Di Domenico F, Mohmmad Abdul H, St. Clair DK, Markesbery WR, Cai J, Pierce WM, Butterfield DA. Proteomic identification of specifically carbonylated brain proteins in APP(NLh)/APP(NLh) × PS-1(P264L)/PS-1(P264L) human double mutant knock-in mice model of Alzheimer disease as a function of age. J Proteomics 2011; 74:2430-40. [PMID: 21726674 PMCID: PMC3199338 DOI: 10.1016/j.jprot.2011.06.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/10/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
Abstract
Alzheimer disease (AD) is the most common type of dementia and is characterized pathologically by the presence of neurofibrillary tangles (NFTs), senile plaques (SPs), and loss of synapses. The main component of SP is amyloid-beta peptide (Aβ), a 39 to 43 amino acid peptide, generated by the proteolytic cleavage of amyloid precursor protein (APP) by the action of beta- and gamma-secretases. The presenilins (PS) are components of the γ-secretase, which contains the protease active center. Mutations in PS enhance the production of the Aβ42 peptide. To date, more than 160 mutations in PS1 have been identified. Many PS mutations increase the production of the β-secretase-mediated C-terminal (CT) 99 amino acid-long fragment (CT99), which is subsequently cleaved by γ-secretase to yield Aβ peptides. Aβ has been proposed to induce oxidative stress and neurotoxicity. Previous studies from our laboratory and others showed an age-dependent increase in oxidative stress markers, loss of lipid asymmetry, and Aβ production and amyloid deposition in the brain of APP/PS1 mice. In the present study, we used APP (NLh)/APP(NLh) × PS-1(P246L)/PS-1(P246L) human double mutant knock-in APP/PS-1 mice to identify specific targets of brain protein carbonylation in an age-dependent manner. We found a number of proteins that are oxidatively modified in APP/PS1 mice compared to age-matched controls. The relevance of the identified proteins to the progression and pathogenesis of AD is discussed.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Renã A. S. Robinson
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Hafiz Mohmmad Abdul
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Daret K. St. Clair
- Graduate Center of Toxicology, University of Kentucky, Lexington, KY 40536, USA
| | | | - Jian Cai
- Department of Pharmacology, University of Louisville, Louisville, Kentucky 40292
| | - William M. Pierce
- Department of Pharmacology, University of Louisville, Louisville, Kentucky 40292
| | - D. Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| |
Collapse
|
185
|
Ramakers GJA, Wolfer D, Rosenberger G, Kuchenbecker K, Kreienkamp HJ, Prange-Kiel J, Rune G, Richter K, Langnaese K, Masneuf S, Bösl MR, Fischer KD, Krugers HJ, Lipp HP, van Galen E, Kutsche K. Dysregulation of Rho GTPases in the αPix/Arhgef6 mouse model of X-linked intellectual disability is paralleled by impaired structural and synaptic plasticity and cognitive deficits. Hum Mol Genet 2011; 21:268-86. [PMID: 21989057 DOI: 10.1093/hmg/ddr457] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mutations in the ARHGEF6 gene, encoding the guanine nucleotide exchange factor αPIX/Cool-2 for the Rho GTPases Rac1 and Cdc42, cause X-linked intellectual disability (ID) in humans. We show here that αPix/Arhgef6 is primarily expressed in neuropil regions of the hippocampus. To study the role of αPix/Arhgef6 in neuronal development and plasticity and gain insight into the pathogenic mechanisms underlying ID, we generated αPix/Arhgef6-deficient mice. Gross brain structure in these mice appeared to be normal; however, analysis of Golgi-Cox-stained pyramidal neurons revealed an increase in both dendritic length and spine density in the hippocampus, accompanied by an overall loss in spine synapses. Early-phase long-term potentiation was reduced and long-term depression was increased in the CA1 hippocampal area of αPix/Arhgef6-deficient animals. Knockout animals exhibited impaired spatial and complex learning and less behavioral control in mildly stressful situations, suggesting that this model mimics the human ID phenotype. The structural and electrophysiological alterations in the hippocampus were accompanied by a significant reduction in active Rac1 and Cdc42, but not RhoA. In conclusion, we suggest that imbalance in activity of different Rho GTPases may underlie altered neuronal connectivity and impaired synaptic function and cognition in αPix/Arhgef6 knockout mice.
Collapse
Affiliation(s)
- Ger J A Ramakers
- Department of Neurons and Networks, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Abstract
Mutations in more than 450 different genes have been associated with intellectual disability (ID) and related cognitive disorders (CDs), such as autism. It is to be expected that this number will increase three to fourfold in the next years due to the rapid implementation of innovative high-throughput sequencing technology in genetics labs. Numerous functional relationships have been identified between the products of individual ID genes, and common molecular and cellular pathways onto which these networks converge are beginning to emerge. Prominent examples are genes involved in synaptic plasticity, Ras and Rho GTPase signaling, and epigenetic genes that encode modifiers of the chromatin structure. It thus seems that there might be common pathological patterns in ID, despite its bewildering genetic heterogeneity. These common pathways provide attractive opportunities for knowledge-based therapeutic interventions.
Collapse
Affiliation(s)
- Hans van Bokhoven
- Molecular Neurogenetics Unit, Department of Human Genetics, Nijmegen Center for Molecular Life Sciences, Radboud University, Nijmegen Medical Center, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
187
|
Myosin IIb activity and phosphorylation status determines dendritic spine and post-synaptic density morphology. PLoS One 2011; 6:e24149. [PMID: 21887379 PMCID: PMC3162601 DOI: 10.1371/journal.pone.0024149] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 08/01/2011] [Indexed: 12/12/2022] Open
Abstract
Dendritic spines in hippocampal neurons mature from a filopodia-like precursor into a mushroom-shape with an enlarged post-synaptic density (PSD) and serve as the primary post-synaptic location of the excitatory neurotransmission that underlies learning and memory. Using myosin II regulatory mutants, inhibitors, and knockdowns, we show that non-muscle myosin IIB (MIIB) activity determines where spines form and whether they persist as filopodia-like spine precursors or mature into a mushroom-shape. MIIB also determines PSD size, morphology, and placement in the spine. Local inactivation of MIIB leads to the formation of filopodia-like spine protrusions from the dendritic shaft. However, di-phosphorylation of the regulatory light chain on residues Thr18 and Ser19 by Rho kinase is required for spine maturation. Inhibition of MIIB activity or a mono-phosphomimetic mutant of RLC similarly prevented maturation even in the presence of NMDA receptor activation. Expression of an actin cross-linking, non-contractile mutant, MIIB R709C, showed that maturation into a mushroom-shape requires contractile activity. Loss of MIIB also leads to an elongated PSD morphology that is no longer restricted to the spine tip; whereas increased MIIB activity, specifically through RLC-T18, S19 di-phosphorylation, increases PSD area. These observations support a model whereby myosin II inactivation forms filopodia-like protrusions that only mature once NMDA receptor activation increases RLC di-phosphorylation to stimulate MIIB contractility, resulting in mushroom-shaped spines with an enlarged PSD.
Collapse
|
188
|
Ghiretti AE, Paradis S. The GTPase Rem2 regulates synapse development and dendritic morphology. Dev Neurobiol 2011; 71:374-89. [PMID: 21485012 DOI: 10.1002/dneu.20868] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rem2 is a member of the Rad/Rem/Rem2/Gem/Kir subfamily of small Ras-like GTPases that was identified as an important mediator of synapse development. We performed a comprehensive, loss- of-function analysis of Rem2 function in cultured hippocampal neurons using RNAi to substantially decrease Rem2 protein levels. We found that knockdown of Rem2 decreases the density and maturity of dendritic spines, the primary site of excitatory synapses onto pyramidal neurons in the hippocampus. Knockdown of Rem2 also alters the gross morphology of dendritic arborizations, increasing the number of dendritic branches without altering total neurite length. Thus, Rem2 functions to inhibit dendritic branching and promote the development of dendritic spines and excitatory synapses. Interestingly, binding to the calcium-binding protein calmodulin is required for the Rem2 regulation of dendritic branching. However, this interaction is completely dispensable for synapse development. Overall, our results suggest that Rem2 regulates dendritic branching and synapse development via distinct and overlapping signal transduction pathways.
Collapse
Affiliation(s)
- Amy E Ghiretti
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, USA
| | | |
Collapse
|
189
|
Chapleau CA, Larimore JL, Theibert A, Pozzo-Miller L. Modulation of dendritic spine development and plasticity by BDNF and vesicular trafficking: fundamental roles in neurodevelopmental disorders associated with mental retardation and autism. J Neurodev Disord 2011; 1:185-96. [PMID: 19966931 PMCID: PMC2788955 DOI: 10.1007/s11689-009-9027-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The process of axonal and dendritic development establishes the synaptic circuitry of the central nervous system (CNS) and is the result of interactions between intrinsic molecular factors and the external environment. One growth factor that has a compelling function in neuronal development is the neurotrophin brain-derived neurotrophic factor (BDNF). BDNF participates in axonal and dendritic differentiation during embryonic stages of neuronal development, as well as in the formation and maturation of dendritic spines during postnatal development. Recent studies have also implicated vesicular trafficking of BDNF via secretory vesicles, and both secretory and endosomal trafficking of vesicles containing synaptic proteins, such as neurotransmitter and neurotrophin receptors, in the regulation of axonal and dendritic differentiation, and in dendritic spine morphogenesis. Several genes that are either mutated or deregulated in neurodevelopmental disorders associated with mental retardation have now been identified, and several mouse models of these disorders have been generated and characterized. Interestingly, abnormalities in dendritic and synaptic structure are consistently observed in human neurodevelopmental disorders associated with mental retardation, and in mouse models of these disorders as well. Abnormalities in dendritic and synaptic differentiation are thought to underlie altered synaptic function and network connectivity, thus contributing to the clinical outcome. Here, we review the roles of BDNF and vesicular trafficking in axonal and dendritic differentiation in the context of dendritic and axonal morphological impairments commonly observed in neurodevelopmental disorders associated with mental retardation.
Collapse
Affiliation(s)
- Christopher A Chapleau
- Department of Neurobiology, Civitan International Research Center, Evelyn McKnight Brain Institute, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
190
|
Portera-Cailliau C. Which Comes First in Fragile X Syndrome, Dendritic Spine Dysgenesis or Defects in Circuit Plasticity? Neuroscientist 2011; 18:28-44. [DOI: 10.1177/1073858410395322] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The salient neuropathological defect in fragile X syndrome is the overabundance of immature dendritic spines in cortical pyramidal neurons. This review examines this anatomical synaptic defect in the context of other alterations in synaptic and circuit plasticity in fragile X mice. In theory, abnormal spines could lead to dysfunctional circuits and vice versa, so it is still not clear which problem comes first. Because of the tight structure-function relationships at the synapse, and given the significant overlap between signaling pathways that regulate spine shape/dynamics and long-term synaptic plasticity (both of which involve proteins regulated by fragile X mental retardation protein [FMRP]), it is argued that the two defects cannot be separated. It will be critical to determine whether neurons that lack FMRP and demonstrate alterations in long-term potentiation/depression also fail to undergo the expected enlargement/shrinkage of dendritic spines associated with those forms of synaptic plasticity or to establish clear links from FMRP signaling to either spine instability or defective synaptic plasticity, especially during critical periods of brain development. The resulting data will be vital in guiding translational research that can identify novel molecular targets for therapy in this devastating disorder.
Collapse
Affiliation(s)
- Carlos Portera-Cailliau
- Departments of Neurology and Neurobiology, David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| |
Collapse
|
191
|
Synaptic Wnt signaling-a contributor to major psychiatric disorders? J Neurodev Disord 2011; 3:162-74. [PMID: 21533542 PMCID: PMC3180925 DOI: 10.1007/s11689-011-9083-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 04/05/2011] [Indexed: 02/08/2023] Open
Abstract
Wnt signaling is a key pathway that helps organize development of the nervous system. It influences cell proliferation, cell fate, and cell migration in the developing nervous system, as well as axon guidance, dendrite development, and synapse formation. Given this wide range of roles, dysregulation of Wnt signaling could have any number of deleterious effects on neural development and thereby contribute in many different ways to the pathogenesis of neurodevelopmental disorders. Some major psychiatric disorders, including schizophrenia, bipolar disorder, and autism spectrum disorders, are coming to be understood as subtle dysregulations of nervous system development, particularly of synapse formation and maintenance. This review will therefore touch on the importance of Wnt signaling to neurodevelopment generally, while focusing on accumulating evidence for a synaptic role of Wnt signaling. These observations will be discussed in the context of current understanding of the neurodevelopmental bases of major psychiatric diseases, spotlighting schizophrenia, bipolar disorder, and autism spectrum disorder. In short, this review will focus on the potential role of synapse formation and maintenance in major psychiatric disorders and summarize evidence that defective Wnt signaling could contribute to their pathogenesis via effects on these late neural differentiation processes.
Collapse
|
192
|
Tolias KF, Duman JG, Um K. Control of synapse development and plasticity by Rho GTPase regulatory proteins. Prog Neurobiol 2011; 94:133-48. [PMID: 21530608 DOI: 10.1016/j.pneurobio.2011.04.011] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 04/13/2011] [Accepted: 04/15/2011] [Indexed: 12/21/2022]
Abstract
Synapses are specialized cell-cell contacts that mediate communication between neurons. Most excitatory synapses in the brain are housed on dendritic spines, small actin-rich protrusions extending from dendrites. During development and in response to environmental stimuli, spines undergo marked changes in shape and number thought to underlie processes like learning and memory. Improper spine development, in contrast, likely impedes information processing in the brain, since spine abnormalities are associated with numerous brain disorders. Elucidating the mechanisms that regulate the formation and plasticity of spines and their resident synapses is therefore crucial to our understanding of cognition and disease. Rho-family GTPases, key regulators of the actin cytoskeleton, play essential roles in orchestrating the development and remodeling of spines and synapses. Precise spatio-temporal regulation of Rho GTPase activity is critical for their function, since aberrant Rho GTPase signaling can cause spine and synapse defects as well as cognitive impairments. Rho GTPases are activated by guanine nucleotide exchange factors (GEFs) and inhibited by GTPase-activating proteins (GAPs). We propose that Rho-family GEFs and GAPs provide the spatiotemporal regulation and signaling specificity necessary for proper Rho GTPase function based on the following features they possess: (i) existence of multiple GEFs and GAPs per Rho GTPase, (ii) developmentally regulated expression, (iii) discrete localization, (iv) ability to bind to and organize specific signaling networks, and (v) tightly regulated activity, perhaps involving GEF/GAP interactions. Recent studies describe several Rho-family GEFs and GAPs that uniquely contribute to spinogenesis and synaptogenesis. Here, we highlight several of these proteins and discuss how they occupy distinct biochemical niches critical for synaptic development.
Collapse
Affiliation(s)
- Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | |
Collapse
|
193
|
Ceman S, Saugstad J. MicroRNAs: Meta-controllers of gene expression in synaptic activity emerge as genetic and diagnostic markers of human disease. Pharmacol Ther 2011; 130:26-37. [PMID: 21256154 PMCID: PMC3043141 DOI: 10.1016/j.pharmthera.2011.01.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 01/05/2011] [Indexed: 12/18/2022]
Abstract
MicroRNAs are members of the non-protein-coding family of RNAs. They serve as regulators of gene expression by modulating the translation and/or stability of messenger RNA targets. The discovery of microRNAs has revolutionized the field of cell biology, and has permanently altered the prevailing view of a linear relationship between gene and protein expression. The increased complexity of gene regulation is both exciting and daunting, as emerging evidence supports a pervasive role for microRNAs in virtually every cellular process. This review briefly describes microRNA processing and formation of RNA-induced silencing complexes, with a focus on the role of RNA binding proteins in this process. We also discuss mechanisms for microRNA-mediated regulation of translation, particularly in dendritic spine formation and function, and the role of microRNAs in synaptic plasticity. We then discuss the evidence for altered microRNA function in cognitive brain disorders, and the effect of gene mutations revealed by single nucleotide polymorphism analysis on altered microRNA function and human disease. Further, we present evidence that altered microRNA expression in circulating fluids such as plasma/serum can correlate with, and serve as, novel diagnostic biomarkers of human disease.
Collapse
Affiliation(s)
- Stephanie Ceman
- University of Illinois, Department of Cell & Developmental Biology, Urbana IL 61801, United States
| | - Julie Saugstad
- Legacy Research Institute, RS Dow Neurobiology Labs, Portland, OR 97232, United States
| |
Collapse
|
194
|
NMDA receptor-dependent regulation of dendritic spine morphology by SAP102 splice variants. J Neurosci 2011; 31:89-96. [PMID: 21209193 DOI: 10.1523/jneurosci.1034-10.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Membrane-associated guanylate kinases (MAGUKs) are major components of the postsynaptic density and play important roles in synaptic organization and plasticity. Most excitatory synapses are located on dendritic spines, which are dynamic structures that undergo morphological changes during synapse formation and plasticity. Synapse-associated protein 102 (SAP102) is a MAGUK that is highly expressed early in development and mediates receptor trafficking during synaptogenesis. Mutations in human SAP102 cause mental retardation, which is often accompanied with abnormalities in dendritic spines. However, little is known about the role of SAP102 in regulating synapse formation or spine morphology. We now find that SAP102 contains a novel NMDA receptor binding site in the N-terminal domain, which is specific for the NR2B subunit. The interaction between SAP102 and NR2B is PDZ (postsynaptic density-95/Discs large/zona occludens-1) domain independent and is regulated by alternative splicing of SAP102. We show that SAP102 that possesses an N-terminal insert is developmentally regulated at both mRNA and protein levels. In addition, expression of SAP102 increases synapse formation. Furthermore, the alternative splicing of SAP102 regulates dendritic spine morphology. SAP102 containing the N-terminal insert promotes lengthening of dendritic spines and preferentially promotes the formation of synapses at long spines, whereas a short hairpin RNA knockdown of the same SAP102 splice variant causes spine shrinkage. Finally, blocking NMDA receptor activity prevents the spine lengthening induced by the N-terminal splice variant of SAP102. Thus, our data provide the first evidence that SAP102 links NMDA receptor activation to alterations in spine morphology.
Collapse
|
195
|
Lemkuil BP, Head BP, Pearn ML, Patel HH, Drummond JC, Patel PM. Isoflurane neurotoxicity is mediated by p75NTR-RhoA activation and actin depolymerization. Anesthesiology 2011; 114:49-57. [PMID: 21169791 PMCID: PMC3037980 DOI: 10.1097/aln.0b013e318201dcb3] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The mechanisms by which isoflurane injured the developing brain are not clear. Recent work has demonstrated that it is mediated in part by activation of p75 neurotrophin receptor. This receptor activates RhoA, a small guanosine triphosphatase that can depolymerize actin. It is therefore conceivable that inhibition of RhoA or prevention of cytoskeletal depolymerization might attenuate isoflurane neurotoxicity. This study was conducted to test these hypotheses using primary cultured neurons and hippocampal slice cultures from neonatal mouse pups. METHODS Primary neuron cultures (days in vitro, 4-7) and hippocampal slice cultures from postnatal day 4-7 mice were exposed to 1.4% isoflurane (4 h). Neurons were pretreated with TAT-Pep5, an intracellular inhibitor of p75 neurotrophin receptor, the cytoskeletal stabilizer jasplakinolide, or their corresponding vehicles. Hippocampal slice cultures were pretreated with TAT-Pep5 before isoflurane exposure. RhoA activation was evaluated by immunoblot. Cytoskeletal depolymerization and apoptosis were evaluated with immunofluorescence microscopy using drebrin and cleaved caspase-3 staining, respectively. RESULTS RhoA activation was increased after 30 and 120 min of isoflurane exposure in neurons; TAT-Pep5 (10 μm) decreased isoflurane-mediated RhoA activation at both time intervals. Isoflurane decreased drebrin immunofluorescence and enhanced cleaved caspase-3 in neurons, effects that were attenuated by pretreatment with either jasplakinolide (1 μm) or TAT-Pep5. TAT-Pep5 attenuated the isoflurane-mediated decrease in phalloidin immunofluorescence. TAT-Pep5 significantly attenuated isoflurane-mediated loss of drebrin immunofluorescence in hippocampal slices. CONCLUSIONS Isoflurane results in RhoA activation, cytoskeletal depolymerization, and apoptosis. Inhibition of RhoA activation or prevention of downstream actin depolymerization significantly attenuated isoflurane-mediated neurotoxicity in developing neurons.
Collapse
Affiliation(s)
- Brian P Lemkuil
- Department of Anesthesiology and Veterans Affairs San Diego Healthcare System, University of California, San Diego, La Jolla, California 92161-5085, USA.
| | | | | | | | | | | |
Collapse
|
196
|
Moreno-López B, Sunico CR, González-Forero D. NO orchestrates the loss of synaptic boutons from adult "sick" motoneurons: modeling a molecular mechanism. Mol Neurobiol 2010; 43:41-66. [PMID: 21190141 DOI: 10.1007/s12035-010-8159-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 12/02/2010] [Indexed: 12/14/2022]
Abstract
Synapse elimination is the main factor responsible for the cognitive decline accompanying many of the neuropathological conditions affecting humans. Synaptic stripping of motoneurons is also a common hallmark of several motor pathologies. Therefore, knowledge of the molecular basis underlying this plastic process is of central interest for the development of new therapeutic tools. Recent advances from our group highlight the role of nitric oxide (NO) as a key molecule triggering synapse loss in two models of motor pathologies. De novo expression of the neuronal isoform of NO synthase (nNOS) in motoneurons commonly occurs in response to the physical injury of a motor nerve and in the course of amyotrophic lateral sclerosis. In both conditions, this event precedes synaptic withdrawal from motoneurons. Strikingly, nNOS-synthesized NO is "necessary" and "sufficient" to induce synaptic detachment from motoneurons. The mechanism involves a paracrine/retrograde action of NO on pre-synaptic structures, initiating a downstream signaling cascade that includes sequential activation of (1) soluble guanylyl cyclase, (2) cyclic guanosine monophosphate-dependent protein kinase, and (3) RhoA/Rho kinase (ROCK) signaling. Finally, ROCK activation promotes phosphorylation of regulatory myosin light chain, which leads to myosin activation and actomyosin contraction. This latter event presumably contributes to the contractile force to produce ending axon retraction. Several findings support that this mechanism may operate in the most prevalent neurodegenerative diseases.
Collapse
Affiliation(s)
- Bernardo Moreno-López
- Grupo de NeuroDegeneración y NeuroReparación (GRUNEDERE), Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Plaza Falla, 9, 11003 Cádiz, Spain.
| | | | | |
Collapse
|
197
|
Regulation of the postsynaptic cytoskeleton: roles in development, plasticity, and disorders. J Neurosci 2010; 30:14937-42. [PMID: 21068295 DOI: 10.1523/jneurosci.4276-10.2010] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The small size of dendritic spines belies the elaborate role they play in excitatory synaptic transmission and ultimately complex behaviors. The cytoskeletal architecture of the spine is predominately composed of actin filaments. These filaments, which at first glance might appear simple, are also surprisingly complex. They dynamically assemble into different structures and serve as a platform for orchestrating the elaborate responses of the spine during experience-dependent plasticity. This mini-symposium review will feature ongoing research into how spines are regulated by actin-signaling pathways during development and plasticity. It will also highlight evolving studies into how disruptions to these pathways might be functionally coupled to congenital disorders such as mental retardation.
Collapse
|
198
|
von Bohlen Und Halbach O. Dendritic spine abnormalities in mental retardation. Cell Tissue Res 2010; 342:317-23. [PMID: 21080001 DOI: 10.1007/s00441-010-1070-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 10/04/2010] [Indexed: 12/13/2022]
Abstract
Abnormalities in dendritic spine morphologies are often associated with mental retardation. Since dendritic spines are thought to represent a morphological correlate of neuronal plasticity, altered spine morphologies may underlie or contribute to cognitive deficits seen in mental retardation. Signaling cascades that are important for cytoskeletal regulation may have an impact upon spine morphologies. The Rho GTPase signaling pathway has been shown to be involved in the regulation of the cytoskeleton and to play fundamental roles in the structural plasticity of dendritic spines. Moreover, alterations in the Rho GTPase signaling pathway have been shown to contribute to mental retardation. Recently, different mental retardation-associated genes have been identified that encode modulators of the Rho GTPases. Disturbances in these genes can lead to mental retardation and-on the morphological level-to alterations in dendritic spines. Thus, getting more insight into the Rho GTPase signaling pathways, and the molecules involved, would not only help in understanding the basic mechanisms by which the morphologies of dendritic spines are modulated but may also allow the development of therapeutic strategies to counteract some aspects of mental retardation.
Collapse
Affiliation(s)
- Oliver von Bohlen Und Halbach
- Institute for Anatomy and Cell Biology, Ernst Moritz Arndt University of Greifswald, Friedrich Loeffler Strasse 23c, 17487 Greifswald, Germany.
| |
Collapse
|
199
|
Regulation of synaptic Rac1 activity, long-term potentiation maintenance, and learning and memory by BCR and ABR Rac GTPase-activating proteins. J Neurosci 2010; 30:14134-44. [PMID: 20962234 DOI: 10.1523/jneurosci.1711-10.2010] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rho family small GTPases are important regulators of neuronal development. Defective Rho regulation causes nervous system dysfunctions including mental retardation and Alzheimer's disease. Rac1, a member of the Rho family, regulates dendritic spines and excitatory synapses, but relatively little is known about how synaptic Rac1 is negatively regulated. Breakpoint cluster region (BCR) is a Rac GTPase-activating protein known to form a fusion protein with the c-Abl tyrosine kinase in Philadelphia chromosome-positive chronic myelogenous leukemia. Despite the fact that BCR mRNAs are abundantly expressed in the brain, the neural functions of BCR protein have remained obscure. We report here that BCR and its close relative active BCR-related (ABR) localize at excitatory synapses and directly interact with PSD-95, an abundant postsynaptic scaffolding protein. Mice deficient for BCR or ABR show enhanced basal Rac1 activity but only a small increase in spine density. Importantly, mice lacking BCR or ABR exhibit a marked decrease in the maintenance, but not induction, of long-term potentiation, and show impaired spatial and object recognition memory. These results suggest that BCR and ABR have novel roles in the regulation of synaptic Rac1 signaling, synaptic plasticity, and learning and memory, and that excessive Rac1 activity negatively affects synaptic and cognitive functions.
Collapse
|
200
|
Ye X, Carew TJ. Small G protein signaling in neuronal plasticity and memory formation: the specific role of ras family proteins. Neuron 2010; 68:340-61. [PMID: 21040840 PMCID: PMC3008420 DOI: 10.1016/j.neuron.2010.09.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2010] [Indexed: 01/04/2023]
Abstract
Small G proteins are an extensive family of proteins that bind and hydrolyze GTP. They are ubiquitous inside cells, regulating a wide range of cellular processes. Recently, many studies have examined the role of small G proteins, particularly the Ras family of G proteins, in memory formation. Once thought to be primarily involved in the transduction of a variety of extracellular signals during development, it is now clear that Ras family proteins also play critical roles in molecular processing underlying neuronal and behavioral plasticity. We here review a number of recent studies that explore how the signaling of Ras family proteins contributes to memory formation. Understanding these signaling processes is of fundamental importance both from a basic scientific perspective, with the goal of providing mechanistic insights into a critical aspect of cognitive behavior, and from a clinical perspective, with the goal of providing effective therapies for a range of disorders involving cognitive impairments.
Collapse
Affiliation(s)
- Xiaojing Ye
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697, USA
| | | |
Collapse
|