251
|
Lindl KA, Marks DR, Kolson DL, Jordan-Sciutto KL. HIV-associated neurocognitive disorder: pathogenesis and therapeutic opportunities. J Neuroimmune Pharmacol 2010; 5:294-309. [PMID: 20396973 PMCID: PMC2914283 DOI: 10.1007/s11481-010-9205-z] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 03/05/2010] [Indexed: 01/08/2023]
Abstract
Human immunodeficiency virus type 1 (HIV) infection presently affects more that 40 million people worldwide, and is associated with central nervous system (CNS) disruption in at least 30% of infected individuals. The use of highly active antiretroviral therapy has lessened the incidence, but not the prevalence of mild impairment of higher cognitive and cortical functions (HIV-associated neurocognitive disorders) as well as substantially reduced a more severe form dementia (HIV-associated dementia). Furthermore, improving neurological outcomes will require novel, adjunctive therapies that are targeted towards mechanisms of HIV-induced neurodegeneration. Identifying such molecular and pharmacological targets requires an understanding of the events preceding irreversible neuronal damage in the CNS, such as actions of neurotoxins (HIV proteins and cellular factors), disruption of ion channel properties, synaptic damage, and loss of adult neurogenesis. By considering the specific mechanisms and consequences of HIV neuropathogenesis, unified approaches for neuroprotection will likely emerge using a tailored, combined, and non-invasive approach.
Collapse
Affiliation(s)
- Kathryn A. Lindl
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Room 312 Levy Building, Philadelphia, PA 19104-6030 USA
| | - David R. Marks
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Room 312 Levy Building, Philadelphia, PA 19104-6030 USA
| | - Dennis L. Kolson
- Department of Neurology School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Kelly L. Jordan-Sciutto
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Room 312 Levy Building, Philadelphia, PA 19104-6030 USA
| |
Collapse
|
252
|
Proline-rich synapse-associated protein-1 and 2 (ProSAP1/Shank2 and ProSAP2/Shank3)-scaffolding proteins are also present in postsynaptic specializations of the peripheral nervous system. Neuroscience 2010; 171:421-33. [PMID: 20800661 DOI: 10.1016/j.neuroscience.2010.08.041] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 08/16/2010] [Accepted: 08/18/2010] [Indexed: 12/17/2022]
Abstract
Proline-rich synapse-associated protein-1 and 2 (ProSAP1/Shank2 and ProSAP2/Shank3) were originally found as synapse-associated protein 90/postsynaptic density protein-95-associated protein (SAPAP)/guanylate-kinase-associated protein (GKAP) interaction partners and also isolated from synaptic junctional protein preparations of rat brain. They are essential components of the postsynaptic density (PSD) and are specifically targeted to excitatory asymmetric type 1 synapses. Functionally, the members of the ProSAP/Shank family are one of the postsynaptic key elements since they link and attach the postsynaptic signaling apparatus, for example N-methyl-d-aspartic acid (NMDA)-receptors via direct and indirect protein interactions to the actin-based cytoskeleton. The functional significance of ProSAP1/2 for synaptic transmission and the paucity of data with respect to the molecular composition of PSDs of the peripheral nervous system (PNS) stimulated us to investigate neuromuscular junctions (NMJs), synapses of the superior cervical ganglion (SCG), and synapses in myenteric ganglia as representative synaptic junctions of the PNS. Confocal imaging revealed ProSAP1/2-immunoreactivity (-iry) in NMJs of rat and mouse sternomastoid and tibialis anterior muscles. In contrast, ProSAP1/2-iry was only negligibly found in motor endplates of striated esophageal muscle probably caused by antigen masking or a different postsynaptic molecular anatomy at these synapses. ProSAP1/2-iry was furthermore detected in cell bodies and dendrites of superior cervical ganglion neurons and myenteric neurons in esophagus and stomach. Ultrastructural analysis of ProSAP1/2 expression in myenteric ganglia demonstrated that ProSAP1 and ProSAP2 antibodies specifically labelled PSDs of myenteric neurons. Thus, scaffolding proteins ProSAP1/2 were found within the postsynaptic specializations of synapses within the PNS, indicating a similar molecular assembly of central and peripheral postsynapses.
Collapse
|
253
|
Smith KR, Oliver PL, Lumb MJ, Arancibia-Carcamo IL, Revilla-Sanchez R, Brandon NJ, Moss SJ, Kittler JT. Identification and characterisation of a Maf1/Macoco protein complex that interacts with GABAA receptors in neurons. Mol Cell Neurosci 2010; 44:330-41. [PMID: 20417281 PMCID: PMC2931578 DOI: 10.1016/j.mcn.2010.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 03/09/2010] [Accepted: 04/09/2010] [Indexed: 01/16/2023] Open
Abstract
The majority of fast inhibitory synaptic transmission in the mammalian nervous system is mediated by GABA(A) receptors (GABA(A)Rs). Here we report a novel interaction between the protein Maf1 and GABA(A)R beta-subunit intracellular domains. We find Maf1 to be highly expressed in brain and enriched in the hippocampus and cortex. In heterologous cells and neurons we show Maf1 co-localises with GABA(A)Rs in intracellular compartments and at the cell surface. In neurons, Maf1 is found localised in the cytoplasm in dendrites, partially overlapping with GABA(A)Rs and inhibitory synapses and in addition is enriched in the neuronal nucleus. We also report that Maf1 interacts with a novel coiled-coil domain containing protein that we have called Macoco (for Maf1 interacting coiled-coil protein). Like Maf1, Macoco can also be found localised to inhibitory synapses and directly interacts with GABA(A)Rs. Expressing Macoco in neurons increases surface GABA(A)R levels. Our results suggest that Maf1 and Macoco are novel GABA(A)R interacting proteins important for regulating GABA(A)R surface expression and GABA(A)R signalling in the brain.
Collapse
Affiliation(s)
- Katharine R Smith
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | | | | | | | | | | | | |
Collapse
|
254
|
Tallafuss A, Constable JRL, Washbourne P. Organization of central synapses by adhesion molecules. Eur J Neurosci 2010; 32:198-206. [PMID: 20646051 DOI: 10.1111/j.1460-9568.2010.07340.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Synapses are the primary means for transmitting information from one neuron to the next. They are formed during the development of the nervous system, and the formation of appropriate synapses is crucial for the establishment of neuronal circuits that underlie behavior and cognition. Understanding how synapses form and are maintained will allow us to address developmental disorders such as autism, mental retardation and possibly also psychological disorders. A number of biochemical and proteomic studies have revealed a diverse and vast assortment of molecules that are present at the synapse. It is now important to untangle this large array of proteins and determine how it assembles into a functioning unit. Here we focus on recent reports describing how synaptic cell adhesion molecules interact with and organize the presynaptic and postsynaptic specializations of both excitatory and inhibitory central synapses.
Collapse
|
255
|
Pham E, Crews L, Ubhi K, Hansen L, Adame A, Cartier A, Salmon D, Galasko D, Michael S, Savas JN, Yates JR, Glabe C, Masliah E. Progressive accumulation of amyloid-beta oligomers in Alzheimer's disease and in amyloid precursor protein transgenic mice is accompanied by selective alterations in synaptic scaffold proteins. FEBS J 2010; 277:3051-67. [PMID: 20573181 PMCID: PMC2933033 DOI: 10.1111/j.1742-4658.2010.07719.x] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The cognitive impairment in patients with Alzheimer's disease is closely associated with synaptic loss in the neocortex and limbic system. Although the neurotoxic effects of aggregated amyloid-beta oligomers in Alzheimer's disease have been studied extensively in experimental models, less is known about the characteristics of these aggregates across the spectrum of Alzheimer's disease. In this study, postmortem frontal cortex samples from controls and patients with Alzheimer's disease were fractionated and analyzed for levels of oligomers and synaptic proteins. We found that the levels of oligomers correlated with the severity of cognitive impairment (blessed information-memory-concentration score and mini-mental state examination) and with the loss of synaptic markers. Reduced levels of the synaptic vesicle protein, vesicle-associated membrane protein-2, and the postsynaptic protein, postsynaptic density-95, correlated with the levels of oligomers in the various fractions analyzed. The strongest associations were found with amyloid-beta dimers and pentamers. Co-immunoprecipitation and double-labeling experiments supported the possibility that amyloid-beta and postsynaptic density-95 interact at synaptic sites. Similarly, in transgenic mice expressing high levels of neuronal amyloid precursor protein, amyloid-beta co-immunoprecipitated with postsynaptic density-95. This was accompanied by a decrease in the levels of the postsynaptic proteins Shank1 and Shank3 in patients with Alzheimer's disease and in the brains of amyloid precursor protein transgenic mice. In conclusion, this study suggests that the presence of a subpopulation of amyloid-beta oligomers in the brains of patients with Alzheimer's disease might be related to alterations in selected synaptic proteins and cognitive impairment.
Collapse
Affiliation(s)
- Emiley Pham
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Leslie Crews
- Department of Pathology, University of California, San Diego, La Jolla, California 92093
| | - Kiren Ubhi
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Lawrence Hansen
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
- Department of Pathology, University of California, San Diego, La Jolla, California 92093
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Anna Cartier
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - David Salmon
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Sarah Michael
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Jeffrey N. Savas
- Department of Chemical Physiology, 10550 North Torrey Pines Road, SR11, The Scripps Research Institute, La Jolla, CA 92037
| | - John R. Yates
- Department of Chemical Physiology, 10550 North Torrey Pines Road, SR11, The Scripps Research Institute, La Jolla, CA 92037
| | - Charles Glabe
- Department of Biochemistry, University of California, Irvine, Irvine CA 92697
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
- Department of Pathology, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
256
|
Tao-Cheng JH, Dosemeci A, Gallant PE, Smith C, Reese T. Activity induced changes in the distribution of Shanks at hippocampal synapses. Neuroscience 2010; 168:11-7. [PMID: 20347015 PMCID: PMC2871978 DOI: 10.1016/j.neuroscience.2010.03.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 03/18/2010] [Accepted: 03/19/2010] [Indexed: 01/15/2023]
Abstract
Dendritic spines contain a family of abundant scaffolding proteins known as Shanks, but little is known about how their distributions might change during synaptic activity. Here, pre-embedding immunogold electron microscopy is used to localize Shanks in synapses from cultured hippocampal neurons. We find that Shanks are preferentially located at postsynaptic densities (PSDs) as well as in a filamentous network near the PSD, extending up to 120 nm from the postsynaptic membrane. Application of sub-type specific antibodies shows that Shank2 is typically concentrated at and near PSDs while Shank1 is, in addition, distributed throughout the spine head. Depolarization with high K+ for 2 min causes transient, reversible translocation of Shanks towards the PSD that is dependent on extracellular Ca2+. The amount of activity-induced redistribution and subsequent recovery is pronounced for Shank1 but less so for Shank2. Thus, Shank1 appears to be a dynamic element within the spine, whose translocation could be involved in activity-induced, transient structural changes, while Shank2 appears to be a more stable element positioned at the interface of the PSD with the spine cytoplasm.
Collapse
Affiliation(s)
- J H Tao-Cheng
- Electron Microscopy (EM) Facility, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA.
| | | | | | | | | |
Collapse
|
257
|
Berkel S, Marshall CR, Weiss B, Howe J, Roeth R, Moog U, Endris V, Roberts W, Szatmari P, Pinto D, Bonin M, Riess A, Engels H, Sprengel R, Scherer SW, Rappold GA. Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental retardation. Nat Genet 2010; 42:489-91. [PMID: 20473310 DOI: 10.1038/ng.589] [Citation(s) in RCA: 404] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 04/15/2010] [Indexed: 12/15/2022]
Abstract
Using microarrays, we identified de novo copy number variations in the SHANK2 synaptic scaffolding gene in two unrelated individuals with autism-spectrum disorder (ASD) and mental retardation. DNA sequencing of SHANK2 in 396 individuals with ASD, 184 individuals with mental retardation and 659 unaffected individuals (controls) revealed additional variants that were specific to ASD and mental retardation cases, including a de novo nonsense mutation and seven rare inherited changes. Our findings further link common genes between ASD and intellectual disability.
Collapse
Affiliation(s)
- Simone Berkel
- Department of Molecular Human Genetics, Ruprecht-Karls-University, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
258
|
Miletic G, Dumitrascu CI, Honstad CE, Micic D, Miletic V. Loose ligation of the rat sciatic nerve elicits early accumulation of Shank1 protein in the post-synaptic density of spinal dorsal horn neurons. Pain 2010; 149:152-159. [PMID: 20171009 PMCID: PMC2836861 DOI: 10.1016/j.pain.2010.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 01/29/2010] [Accepted: 02/01/2010] [Indexed: 11/17/2022]
Abstract
Plasticity in the spinal dorsal horn may contribute to the development of pain following peripheral nerve injury. Shank proteins are a constituent family of the post-synaptic density (PSD), and they may play a role in synaptic plasticity through activity-dependent synaptic remodeling and growth. In this study we examined the early consequences of the loose ligation of the sciatic nerve on Shank1 protein and message levels in the PSD of spinal dorsal horn neurons. Four hours after sciatic ligation, the protein levels of Shank1 increased in the ipsilateral PSD of ligated animals. In contrast, no changes were detected in the contralateral PSD of these ligated animals, or either the ipsilateral or contralateral PSD of sham-operated animals. Shank1 was linked to the PSD marker protein PSD-95 and the NR2B subunit of NMDA receptors. The ligated animals also exhibited two early signs of pain behavior, a shift in weight distribution and thermal hyperalgesia. There was no overall change in Shank1 message in either ligated or sham-operated animals. The accumulation of Shank1 in the PSD was abolished by intrathecal pre-treatment with anisomycin or Shank1 siRNA, but not with non-target siRNA. The same pre-treatment prevented both the early signs of pain behavior. Intrathecal pre-treatment with either MK-801 or U0126 similarly prevented the Shank1 accumulation and alleviated both the behavioral signs of pain. The early accumulation of Shank1 in the PSD of dorsal horn neurons may be a necessary step in the injury-associated plasticity that in time leads to the development of persistent pain.
Collapse
Affiliation(s)
- Gordana Miletic
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Catalina I. Dumitrascu
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Christopher E. Honstad
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Daniela Micic
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Vjekoslav Miletic
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
259
|
DNA methylation in vulnerability to post-traumatic stress in rats: evidence for the role of the post-synaptic density protein Dlgap2. Int J Neuropsychopharmacol 2010; 13:347-59. [PMID: 19793403 DOI: 10.1017/s146114570999071x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is unique among psychiatric disorders since there is an explicit requirement for the presence of a well-defined precipitating environmental event. This suggests the participation of adaptable molecular processes such as epigenetic modifications, including acetylation and methylation of histones and DNA methylation. In the present study we investigated whether changes in DNA methylation are associated with the effects of traumatic stressor, using a validated PTSD rat model. Screening of genomic DNA methylation patterns revealed that maladaptation to traumatic stress is associated with numerous changes in the methylation pattern of rat hippocampus. Of the differentially methylated genes revealed by this global screening, Disks Large-Associated Protein (Dlgap2) was of special interest, demonstrating an increase in a specific methylation site which was associated with a reduction in its gene expression in PTSD-like compared to non-PTSD-like rats. The association between the methylation rate and Dlgap2 expression was further substantiated by re-dividing the rats according to their methylation state. A significantly higher expression was observed in the non-methylated compared to methylated rats. In addition, taking all rats as one group revealed a significant correlation between their behavioural stress responses and Dlgap2 transcript levels. These results suggest that alterations in global methylation pattern are involved in behavioural adaptation to environmental stress and pinpoint Dlgap2 as a possible target in PTSD.
Collapse
|
260
|
Li C, Naren AP. CFTR chloride channel in the apical compartments: spatiotemporal coupling to its interacting partners. Integr Biol (Camb) 2010; 2:161-77. [PMID: 20473396 PMCID: PMC2989726 DOI: 10.1039/b924455g] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-regulated chloride channel located primarily at the apical or luminal surfaces of epithelial cells in the airway, intestine, pancreas, kidney, sweat gland, as well as male reproductive tract, where it plays a crucial role in transepithelial fluid homeostasis. CFTR dysfunction can be detrimental and may result in life-threatening disorders. CFTR hypofunctioning because of genetic defects leads to cystic fibrosis, the most common lethal genetic disease in Caucasians, whereas CFTR hyperfunctioning resulting from various infections evokes secretory diarrhea, the leading cause of mortality in early childhood. Therefore, maintaining a dynamic balance between CFTR up-regulating processes and CFTR down-regulating processes is essential for maintaining fluid and body homeostasis. Accumulating evidence suggests that protein-protein interactions play a critical role in the fine-tuned regulation of CFTR function. A growing number of proteins have been reported to interact directly or indirectly with CFTR chloride channel, suggesting that CFTR might be coupled spatially and temporally to a wide variety of interacting partners including ion channels, receptors, transporters, scaffolding proteins, enzyme molecules, signaling molecules, and effectors. Most interactions occur primarily between the opposing terminal tails (amino or carboxyl) of CFTR protein and its binding partners, either directly or mediated through various PDZ scaffolding proteins. These dynamic interactions impact the channel function, as well as localization and processing of CFTR protein within cells. This article reviews the most recent progress and findings about the interactions between CFTR and its binding partners through PDZ scaffolding proteins, as well as the spatiotemporal regulation of CFTR-containing macromolecular signaling complexes in the apical compartments of polarized cells lining the secretory epithelia.
Collapse
Affiliation(s)
- Chunying Li
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, 540 E. Canfield Avenue, 5312 Scott Hall, Detroit, Michigan 48201, USA
| | - Anjaparavanda P. Naren
- Department of Physiology, University of Tennessee Health Science Center, 420 Nash, 894 Union Avenue, Memphis, Tennessee 38163, USA
| |
Collapse
|
261
|
Dhar S, del Gaudio D, German J, Peters S, Ou Z, Bader P, Berg J, Blazo M, Brown C, Graham B, Grebe T, Lalani S, Irons M, Sparagana S, Williams M, Phillips J, Beaudet A, Stankiewicz P, Patel A, Cheung S, Sahoo T. 22q13.3 deletion syndrome: clinical and molecular analysis using array CGH. Am J Med Genet A 2010; 152A:573-81. [PMID: 20186804 PMCID: PMC3119894 DOI: 10.1002/ajmg.a.33253] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The 22q13.3 deletion syndrome results from loss of terminal segments of varying sizes at 22qter. Few genotype-phenotype correlations have been found but all patients have mental retardation and severe delay, or absence of, expressive speech. We carried out clinical and molecular characterization of 13 patients. Developmental delay and speech abnormalities were common to all and comparable in frequency and severity to previously reported cases. Array-based comparative genomic hybridization showed the deletions to vary from 95 kb to 8.5 Mb. We also carried out high-resolution 244K array comparative genomic hybridization in 10 of 13 patients, that defined the proximal and distal breakpoints of each deletion and helped determine the size, extent, and gene content within the deletion. Two patients had a smaller 95 kb terminal deletion with breakpoints within the SHANK3 gene while three other patients had a similar 5.5 Mb deletion implying the recurrent nature of these deletions. The two largest deletions were found in patients with ring chromosome 22. No correlation could be made with deletion size and phenotype although complete/partial SHANK3 was deleted in all patients. There are very few reports on array comparative genomic hybridization analysis on patients with the 22q13.3 deletion syndrome, and we aim to accurately characterize these patients both clinically and at the molecular level, to pave the way for further genotype-phenotype correlations. (c) 2010 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- S.U. Dhar
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - D. del Gaudio
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - J.R. German
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - S.U. Peters
- Department of Pediatrics, Vanderbilt University, VKC for Research on Human Development
| | - Z. Ou
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - P.I. Bader
- Parkview Cytogenetic Laboratory, Fort Wayne, Indiana
| | - J.S. Berg
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - M. Blazo
- Division of Medical Genetics, Scott & White Clinic, Temple, Texas
| | - C.W. Brown
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - B.H. Graham
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - T.A. Grebe
- CHC Phoenix Genetics Program, St. Joseph’s Hospital & Medical Center, Phoenix, Arizona
| | - S. Lalani
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - M. Irons
- Division of Genetics, Children’s Hospital, Boston, Massachusetts
| | - S. Sparagana
- Department of Pediatric Neurology, Texas Scottish Rite Hospital for Children, Dallas, Texas
| | - M. Williams
- Division of Medical Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - J.A. Phillips
- Division of Medical Genetics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - A.L. Beaudet
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - P. Stankiewicz
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - A. Patel
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - S.W. Cheung
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
262
|
Structural Basis for Asymmetric Association of the βPIX Coiled Coil and Shank PDZ. J Mol Biol 2010; 397:457-66. [DOI: 10.1016/j.jmb.2010.01.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Revised: 01/19/2010] [Accepted: 01/21/2010] [Indexed: 11/21/2022]
|
263
|
Newly identified tumor-associated role of human Sharpin. Mol Cell Biochem 2010; 340:161-7. [DOI: 10.1007/s11010-010-0413-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 02/10/2010] [Indexed: 10/19/2022]
|
264
|
Lee JS, Lee YM, Kim JY, Park HW, Grinstein S, Orlowski J, Kim E, Kim KH, Lee MG. BetaPix up-regulates Na+/H+ exchanger 3 through a Shank2-mediated protein-protein interaction. J Biol Chem 2010; 285:8104-13. [PMID: 20080968 DOI: 10.1074/jbc.m109.055079] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Na(+)/H(+) exchanger 3 (NHE3) plays an important role in neutral Na(+) transport in mammalian epithelial cells. The Rho family of small GTPases and the PDZ (PSD-95/discs large/ZO-1) domain-based adaptor Shank2 are known to regulate the membrane expression and activity of NHE3. In this study we examined the role of betaPix, a guanine nucleotide exchange factor for the Rho GTPase and a strong binding partner to Shank2, in NHE3 regulation using integrated molecular and physiological approaches. Immunoprecipitation and pulldown assays revealed that NHE3, Shank2, and betaPix form a macromolecular complex when expressed heterologously in mammalian cells as well as endogenously in rat colon, kidney, and pancreas. In addition, these proteins co-segregated at the apical surface of rat colonic epithelial cells, as detected by immunofluorescence staining. When expressed in PS120/NHE3 cells, betaPix increased membrane expression and basal activity of NHE3. Interestingly, the effects of betaPix on NHE3 were abolished by cotransfection with dominant-negative Shank2 mutants and by treatment with Clostridium difficile toxin B, a Rho GTPase inhibitor, indicating that Shank2 and Rho GTPases are involved in betaPix-mediated NHE3 regulation. Knockdown of endogenous betaPix by RNA interference decreased Shank2-induced increase of NHE3 membrane expression in HEK 293T cells. These results indicate that betaPix up-regulates NHE3 membrane expression and activity by Shank2-mediated protein-protein interaction and by activating Rho GTPases in the apical regions of epithelial cells.
Collapse
Affiliation(s)
- Jung-Soo Lee
- Department of Pharmacology and Brain Korea 21 Project for Medical Science, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
265
|
Rubini C, Ruzza P, Spaller MR, Siligardi G, Hussain R, Udugamasooriya DG, Bellanda M, Mammi S, Borgogno A, Calderan A, Cesaro L, Brunati AM, Donella-Deana A. Recognition of lysine-rich peptide ligands by murine cortactin SH3 domain: CD, ITC, and NMR studies. Biopolymers 2009; 94:298-306. [DOI: 10.1002/bip.21350] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
266
|
Sykes NH, Toma C, Wilson N, Volpi EV, Sousa I, Pagnamenta AT, Tancredi R, Battaglia A, Maestrini E, Bailey AJ, Monaco AP, International Molecular Genetic Study of Autism Consortium (IMGSAC) 5. Copy number variation and association analysis of SHANK3 as a candidate gene for autism in the IMGSAC collection. Eur J Hum Genet 2009; 17:1347-53. [PMID: 19384346 PMCID: PMC2752466 DOI: 10.1038/ejhg.2009.47] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 01/28/2009] [Accepted: 02/17/2009] [Indexed: 11/13/2022] Open
Abstract
SHANK3 is located on chromosome 22q13.3 and encodes a scaffold protein that is found in excitatory synapses opposite the pre-synaptic active zone. SHANK3 is a binding partner of neuroligins, some of whose genes contain mutations in a small subset of individuals with autism. In individuals with autism spectrum disorders (ASDs), several studies have found SHANK3 to be disrupted by deletions ranging from hundreds of kilobases to megabases, suggesting that 1% of individuals with ASDs may have these chromosomal aberrations. To further analyse the involvement of SHANK3 in ASD, we screened the International Molecular Genetic Study of Autism Consortium (IMGSAC) multiplex family sample, 330 families, for SNP association and copy number variants (CNVs) in SHANK3. A collection of 76 IMGSAC Italian probands from singleton families was also examined by multiplex ligation-dependent probe amplification for CNVs. No CNVs or SNP associations were found within the sample set, although sequencing of the gene was not performed. Our data suggest that SHANK3 deletions may be limited to lower functioning individuals with autism.
Collapse
Affiliation(s)
- Nuala H Sykes
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Claudio Toma
- Department of Biology, University of Bologna, Bologna, Italy
| | - Natalie Wilson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Emanuela V Volpi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Inês Sousa
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Raffaella Tancredi
- Stella Maris Clinical Research Institute for Child and Adolescent Neuropsychiatry, Calambrone (Pisa), Italy
| | - Agatino Battaglia
- Stella Maris Clinical Research Institute for Child and Adolescent Neuropsychiatry, Calambrone (Pisa), Italy
| | - Elena Maestrini
- Department of Biology, University of Bologna, Bologna, Italy
| | - Anthony J Bailey
- University Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - Anthony P Monaco
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - International Molecular Genetic Study of Autism Consortium (IMGSAC)5
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Department of Biology, University of Bologna, Bologna, Italy
- Stella Maris Clinical Research Institute for Child and Adolescent Neuropsychiatry, Calambrone (Pisa), Italy
- University Department of Psychiatry, Warneford Hospital, Oxford, UK
| |
Collapse
|
267
|
Abstract
For more than three decades, the venom of the black widow spider and its principal active components, latrotoxins, have been used to induce release of neurotransmitters and hormones and to study the mechanisms of exocytosis. Given the complex nature of alpha--latrotoxin (alpha-LTX) actions, this research has been continuously overshadowed by many enigmas, misconceptions and perpetual changes of the underlying hypotheses. Some of the toxin's mechanisms of action are still not completely understood. Despite all these difficulties, the extensive work of several generations of neurobiologists has brought about a great deal of fascinating insights into pre-synaptic processes and has led to the discovery of several novel proteins and synaptic systems. For example, alpha-LTX studies have contributed to the widespread acceptance of the vesicular theory of transmitter release. Pre-synaptic receptors for alpha-LTX--neurexins, latrophilins and protein tyrosine phosphatase sigma--and their endogenous ligands have now become centrepieces of their own areas of research, with a potential of uncovering new mechanisms of synapse formation and regulation that may have medical implications. However, any future success of alpha-LTX research will require a better understanding of this unusual natural tool and a more precise dissection of its multiple mechanisms.
Collapse
Affiliation(s)
- John-Paul Silva
- Division of Cell and Molecular Biology, Imperial College London, Exhibition Road, London, UK
| | | | | |
Collapse
|
268
|
Abstract
Neurons are highly polarized cells that extend a single axon and several dendrites. Studies with cultured neurons indicate that the proximal portion of the axon, denoted as the axon initial segment (AIS), maintains neuronal polarity in vitro. The membrane-adaptor protein ankyrinG (ankG) is an essential component of the AIS. To determine the relevance of ankG for neuronal polarity in vivo, we studied mice with a cerebellum-specific ankG deficiency. Strikingly, ankG-depleted axons develop protrusions closely resembling dendritic spines. Such axonal spines are enriched with postsynaptic proteins, including ProSAP1/Shank2 and ionotropic and metabotropic glutamate receptors. In addition, immunofluorescence indicated that axonal spines are contacted by presynaptic glutamatergic boutons. For further analysis, double mutants were obtained by crossbreeding ankG(-/-) mice with L7/Purkinje cell-specific promoter 2 (PCP2) mice expressing enhanced green fluorescent protein (EGFP) in Purkinje cells (PCs). This approach allowed precise confocal microscopic mapping of EGFP-positive spiny axons and their subsequent identification at the electron microscopic level. Ultrastructurally, axonal spines contained a typical postsynaptic density and established asymmetric excitatory synapses with presynaptic boutons containing synaptic vesicles. In the shaft of spiny axons, typical ultrastructural features of the AIS, including the membrane-associated dense undercoating and cytoplasmic bundles of microtubules, were absent. Finally, using time-lapse imaging of organotypic cerebellar slice cultures, we demonstrate that nonspiny PC axons of EGFP-positive/ankG(-/-) mice acquire a spiny phenotype within a time range of only 3 days. Collectively, these findings demonstrate that axons of ankG-deficient mice acquire hallmark features of dendrites. AnkG thus is important for maintaining appropriate axo-dendritic polarity in vivo.
Collapse
|
269
|
Abstract
Understanding the evolutionary origins of behaviour is a central aim in the study of biology and may lead to insights into human disorders. Synaptic transmission is observed in a wide range of invertebrate and vertebrate organisms and underlies their behaviour. Proteomic studies of the molecular components of the highly complex mammalian postsynaptic machinery point to an ancestral molecular machinery in unicellular organisms--the protosynapse--that existed before the evolution of metazoans and neurons, and hence challenges existing views on the origins of the brain. The phylogeny of the molecular components of the synapse provides a new model for studying synapse diversity and complexity, and their implications for brain evolution.
Collapse
|
270
|
Schmeisser MJ, Grabrucker AM, Bockmann J, Boeckers TM. Synaptic cross-talk between N-methyl-D-aspartate receptors and LAPSER1-beta-catenin at excitatory synapses. J Biol Chem 2009; 284:29146-57. [PMID: 19703901 DOI: 10.1074/jbc.m109.020628] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Memory formation in the brain is thought to be depending upon long lasting plastic changes of synaptic contacts that require alterations on the transcriptional level. Here, we characterize LAPSER1, a putative cytokinetic tumor suppressor that binds directly to ProSAP2/Shank3 and the synaptic Rap-Gap protein SPAR1 as a novel postsynaptic density component. Postsynaptic LAPSER1 is in complex with all important members of the canonical Wnt pathway including beta-catenin. Upon N-methyl-D-aspartate receptor-dependent activation, LAPSER1 and beta-catenin comigrate from the postsynaptic density to the nucleus and induce the transcription and translation of known beta-catenin target genes, including Tcfe2a and c-Myc. The nuclear export and cytoplasmic redistribution of beta-catenin is tightly regulated by LAPSER1. We postulate a postsynaptic cross-talk between N-methyl-D-aspartate receptors and a LAPSER1-beta-catenin complex that results in a self-regulated, synaptic activity-dependent expression of beta-catenin target genes. This calls for a novel role of Tcfe2a and c-Myc in plastic changes of neural tissue.
Collapse
|
271
|
Qin J, Jia M, Wang L, Lu T, Ruan Y, Liu J, Guo Y, Zhang J, Yang X, Yue W, Zhang D. Association study of SHANK3 gene polymorphisms with autism in Chinese Han population. BMC MEDICAL GENETICS 2009; 10:61. [PMID: 19566951 PMCID: PMC2721832 DOI: 10.1186/1471-2350-10-61] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 06/30/2009] [Indexed: 12/27/2022]
Abstract
Background Autism, a heterogeneous disease, is described as a genetic psychiatry disorder. Recently, abnormalities at the synapse are supposed to be important for the etiology of autism.SHANK3 (SH3 and multiple ankyrin repeat domains protein) gene encodes a master synaptic scaffolding protein at postsynaptic density (PSD) of excitatory synapse. Rare mutations and copy number variation (CNV) evidence suggested SHANK3 as a strong candidate gene for the pathogenesis of autism. Methods We performed an association study between SHANK3 gene polymorphisms and autism in Chinese Han population. We analyzed the association between five single nucleotide polymorphisms (SNPs) of the SHANK3 gene and autism in 305 Chinese Han trios, using the family based association test (FBAT). Linkage disequilibrium (LD) analysis showed the presence of LD between pairwise markers across the locus. We also performed mutation screening for the rare de novo mutations reported previously. Results No significant evidence between any SNPs of SHANK3 and autism was observed. We did not detect any mutations described previously in our cohort. Conclusion We suggest that SHANK3 might not represent a major susceptibility gene for autism in Chinese Han population.
Collapse
Affiliation(s)
- Jian Qin
- Institute of Mental Health, Peking University, Beijing, PR China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
272
|
Balázs A, Csizmok V, Buday L, Rakács M, Kiss R, Bokor M, Udupa R, Tompa K, Tompa P. High levels of structural disorder in scaffold proteins as exemplified by a novel neuronal protein, CASK-interactive protein1. FEBS J 2009; 276:3744-56. [PMID: 19523119 DOI: 10.1111/j.1742-4658.2009.07090.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
CASK-interactive protein1 is a newly recognized post-synaptic density protein in mammalian neurons. Although its N-terminal region contains several well-known functional domains, its entire C-terminal proline-rich region of 800 amino acids lacks detectable sequence homology to any previously characterized protein. We used multiple techniques for the structural characterization of this region and its three fragments. By bioinformatics predictions, CD spectroscopy, wide-line and 1H-NMR spectroscopy, limited proteolysis and gel filtration chromatography, we provided evidence that the entire proline-rich region of CASK-interactive protein1 is intrinsically disordered. We also showed that the proline-rich region is biochemically functional, as it interacts with the adaptor protein Abl-interactor-2. To extend the finding of a high level of disorder in this scaffold protein, we collected 74 scaffold proteins (also including proteins denoted as anchor and docking), and predicted their disorder by three different algorithms. We found that a very high fraction (53.6; on average) of the residues fall into local disorder and their ordered domains are connected by linker regions which are mostly disordered (64.5 on average). Because of this high frequency of disorder, the usual design of scaffold proteins of short globular domains (86 amino acids on average) connected by longer linker regions (140 amino acids on average) and the noted binding functions of these regions in both CASK-interactive protein1 and the other proteins studied, we suggest that structurally disordered regions prevail and play key recognition roles in scaffold proteins.
Collapse
Affiliation(s)
- Annamária Balázs
- Department of Medical Chemistry, Semmelweis University Medical School, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
273
|
Homer and the ryanodine receptor. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2009; 39:91-102. [PMID: 19513708 DOI: 10.1007/s00249-009-0494-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 05/15/2009] [Accepted: 05/18/2009] [Indexed: 10/20/2022]
Abstract
Homer proteins have recently been identified as novel high-affinity ligands that modulate ryanodine receptor (RyR) Ca(2+) release channels in heart and skeletal muscle, through an EVH1 domain which binds to proline-rich regions in target proteins. Many Homer proteins can also self-associate through a coiled-coil domain that allows their multimerisation. In other tissues, especially neurons, Homer anchors proteins embedded in the surface membrane to the Ca(2+) release channel in the endoplasmic reticulum and can anchor membrane or cytosolic proteins to the cytoskeleton. Although this anchoring aspect of Homer function has not been extensively investigated in muscle, there are consensus sequences for Homer binding in the RyR and on many of the proteins that it interacts with in the massive RyR ion channel complex. In this review we explore the potential of Homer to contribute to a variety of cell processes in muscle and neurons that also involve RyR channels.
Collapse
|
274
|
Kim JH, Kim JH, Yang E, Park JH, Yu YS, Kim KW. Shank 2 expression coincides with neuronal differentiation in the developing retina. Exp Mol Med 2009; 41:236-42. [PMID: 19299912 DOI: 10.3858/emm.2009.41.4.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The retinal activity for vision requires a precise synaptic connectivity. Shank proteins at postsynaptic sites of excitatory synapses play roles in signal transmission into the postsynaptic neuron. However, the correlation of Shank 2 expression with neuronal differentiation in the developing retina remains to be elucidated regardless of previous evidences of Shank 2 expression in retina. Herein, we demonstrated that with progression of development, Shank 2 is initially detected in the inner plexiform layer at P2, and then intensively detected in inner plexiform layer, outer plexiform layer, and ganglion cell layer at P14, which was closely colocalized to the neurofilament expression. Shank 2 was, however, not colocalized with glial fibrillary acidic protein. Shank 2 expression was increased in the differentiated retinoblastoma cells, which was mediated by ERK 1/2 activation. Moreover, Shank 2 expression was colocalized with neurofilament at the dendritic region of cells. In conclusion, our data suggests that Shank 2 is expressed in the neurons of the developing retina and could play a critical role in the neuronal differentiation of the developing retina.
Collapse
Affiliation(s)
- Jeong Hun Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 110-744, Korea
| | | | | | | | | | | |
Collapse
|
275
|
Li HH, Chiang CS, Huang HY, Liaw GJ. mars and tousled-like kinase act in parallel to ensure chromosome fidelity in Drosophila. J Biomed Sci 2009; 16:51. [PMID: 19486529 PMCID: PMC2705347 DOI: 10.1186/1423-0127-16-51] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Accepted: 06/01/2009] [Indexed: 01/08/2023] Open
Abstract
Background High levels of Hepatoma Up-Regulated Protein (HURP) and Tousled-Like Kinase (TLK) transcripts are found in hepatocellular carcinoma. HURP overexpression induces anchorage-independent growth of 293-T cells and enhances a rough-eye phenotype resulting from tlk overexpression in Drosophila. In addition, both HURP and Mars, a Drosophila HURP sequence homologue, promote polymerization of mitotic spindles. Thus, the genetic interaction of mars with tlk might be required for accurate chromosome segregation. Methods To reveal whether chromosome fidelity was decreased, the frequency of gynandromorphy, an individual with both male and female characteristics, and of non-disjunction were measured in the progeny from parents with reduced mars and/or tlk activities and analyzed by Student's t-test. To show that the genetic interaction between mars and tlk is epistatic or parallel, a cytological analysis of embryos with either reduced or increased activities of mars and/or tlk was used to reveal defects in mitotic-spindle morphology and chromosome segregation. Results A significant but small fraction of the progeny from parents with reduced mars activity showed gynandromorphy and non-disjunction. Results of cytological analysis revealed that the decrease in chromosome fidelity was a result of delayed polymerization of the mitotic spindle, which led to asynchronous chromosome segregation in embryos that had reduced mars activity. By removing one copy of tousled-like kinase (tlk) from flies with reduced mars activity, chromosome fidelity was further reduced. This was indicated by an increased in the non-disjunction rate and more severe asynchrony. However, the morphology of the mitotic spindles in the embryos at metaphase where both gene activities were reduced was similar to that in mars embryos. Furthermore, tlk overexpression did not affect the morphology of the mitotic spindles and the cellular localization of Mars protein. Conclusion Chromosome fidelity in progeny from parents with reduced mars and/or tlk activity was impaired. The results from cytological studies revealed that mars and tlk function in parallel and that a balance between mars activity and tlk activity is required for cells to progress through mitosis correctly, thus ensuring chromosome fidelity.
Collapse
Affiliation(s)
- Hsing-Hsi Li
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, 112 Taiwan, ROC
| | | | | | | |
Collapse
|
276
|
Liebau S, Proepper C, Schmidt T, Schoen M, Bockmann J, Boeckers TM. ProSAPiP2, a novel postsynaptic density protein that interacts with ProSAP2/Shank3. Biochem Biophys Res Commun 2009; 385:460-5. [PMID: 19481056 DOI: 10.1016/j.bbrc.2009.05.098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 05/20/2009] [Indexed: 11/26/2022]
Abstract
The postsynaptic density (PSD) is a highly specialized structure that is located juxtaposed to the presynaptic active zone of excitatory synapses. It is composed of a variety of proteins that include receptors, signaling molecules, cytoskeletal components and scaffolding proteins. ProSAP/Shank proteins are large multidomain proteins that facilitate multiple functions within the PSD. They build large scaffolds that are the structural basis for the direct and/or indirect connection between receptor proteins and the actin based cytoskeleton. Here, we characterize a novel interaction partner of ProSAP2/Shank3, named ProSAP interacting protein 2 (ProSAPiP2) that does not show any close homology to other known proteins. It binds to the PDZ domain of ProSAP2/Shank3 and is highly expressed in the neuronal system. ProSAPiP2 is located in dendrites and spines, is enriched in the PSD and interacts with actin. Therefore ProSAPiP2 could be involved in the linkage between molecules of the PSD and the cytoskeleton.
Collapse
Affiliation(s)
- Stefan Liebau
- Institute of Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany
| | | | | | | | | | | |
Collapse
|
277
|
Sainlos M, Iskenderian WS, Imperiali B. A general screening strategy for peptide-based fluorogenic ligands: probes for dynamic studies of PDZ domain-mediated interactions. J Am Chem Soc 2009; 131:6680-2. [PMID: 19388649 PMCID: PMC2828873 DOI: 10.1021/ja900371q] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A systematic and general approach for identifying efficient probes for class I PDZ domains based on environment-sensitive chromophores is presented. A series of peptides derived from the C-terminal sequence of Stargazin was first used with PDZ domains of PSD-95 and Shank3 to identify the optimal position and linker length for the 4-DMAP chromophore. The results were applied to well-characterized ligand sequences for each set of domains to generate high affinity probes that retain their native sequence specificity and yield remarkable fluorescence increases upon binding. These probes constitute efficient tools to study the dynamics and regulatory mechanisms of PDZ domain-mediated interactions.
Collapse
Affiliation(s)
- Matthieu Sainlos
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139-4307, USA
| | - Wendy S. Iskenderian
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139-4307, USA
| | - Barbara Imperiali
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139-4307, USA
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139-4307, USA
| |
Collapse
|
278
|
Kim SM, Choi KY, Cho IH, Rhy JH, Kim SH, Park CS, Kim E, Song WK. Regulation of dendritic spine morphology by SPIN90, a novel Shank binding partner. J Neurochem 2009; 109:1106-17. [PMID: 19302483 DOI: 10.1111/j.1471-4159.2009.06039.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic spines are highly specialized actin-rich structures on which the majority of excitatory synapses are formed in the mammalian CNS. SPIN90 is an actin-binding protein known to be highly enriched in postsynaptic densities (PSDs), though little is known about its function there. Here, we show that SPIN90 is a novel binding partner for Shank proteins in the PSD. SPIN90 and Shank co-immunoprecipitate from brain lysates and co-localize in postsynaptic dendrites and act synergistically to mediate spine maturation and spine head enlargement. At the same time, SPIN90 causes accumulation of Shank and PSD-95 within dendritic spines. In addition, we found that the protein composition of PSDs in SPIN90 knockout mice is altered as is the actin cytoskeleton of cultured hippocampal SPIN90 knockout neurons. Taken together, these findings demonstrate that SPIN90 is a Shank1b binding partner and a key contributor to the regulation of dendritic spine morphogenesis and brain function.
Collapse
Affiliation(s)
- Seon-Myung Kim
- Cell Dynamics Research Center and Bioimaging Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | | | | | | | | | | | | | | |
Collapse
|
279
|
Steiner P, Higley MJ, Xu W, Czervionke BL, Malenka RC, Sabatini BL. Destabilization of the postsynaptic density by PSD-95 serine 73 phosphorylation inhibits spine growth and synaptic plasticity. Neuron 2008; 60:788-802. [PMID: 19081375 PMCID: PMC2671083 DOI: 10.1016/j.neuron.2008.10.014] [Citation(s) in RCA: 208] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 08/29/2008] [Accepted: 10/07/2008] [Indexed: 12/23/2022]
Abstract
Long-term potentiation (LTP) is accompanied by dendritic spine growth and changes in the composition of the postsynaptic density (PSD). We find that activity-dependent growth of apical spines of CA1 pyramidal neurons is accompanied by destabilization of the PSD that results in transient loss and rapid replacement of PSD-95 and SHANK2. Signaling through PSD-95 is required for activity-dependent spine growth and trafficking of SHANK2. N-terminal PDZ and C-terminal guanylate kinase domains of PSD-95 are required for both processes, indicating that PSD-95 coordinates multiple signals to regulate morphological plasticity. Activity-dependent trafficking of PSD-95 is triggered by phosphorylation at serine 73, a conserved calcium/calmodulin-dependent protein kinase II (CaMKII) consensus phosphorylation site, which negatively regulates spine growth and potentiation of synaptic currents. We propose that PSD-95 and CaMKII act at multiple steps during plasticity induction to initially trigger and later terminate spine growth by trafficking growth-promoting PSD proteins out of the active spine.
Collapse
Affiliation(s)
- Pascal Steiner
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michael J. Higley
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Weifeng Xu
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Brian L. Czervionke
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Robert C. Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Bernardo L. Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
280
|
Jeon D, Song I, Guido W, Kim K, Kim E, Oh U, Shin HS. Ablation of Ca2+ channel beta3 subunit leads to enhanced N-methyl-D-aspartate receptor-dependent long term potentiation and improved long term memory. J Biol Chem 2008; 283:12093-101. [PMID: 18339621 PMCID: PMC3774114 DOI: 10.1074/jbc.m800816200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 03/12/2008] [Indexed: 11/06/2022] Open
Abstract
The beta subunits of voltage-dependent Ca(2+) channels (VDCCs) have marked effects on the properties of the pore-forming alpha(1) subunits of VDCCs, including surface expression of channel complexes and modification of voltage-dependent kinetics. Among the four different beta subunits, the beta(3) subunit (Ca(v)beta3) is abundantly expressed in the hippocampus. However, the role of Ca(v)beta3 in hippocampal physiology and function in vivo has never been examined. Here, we investigated Ca(v)beta3-deficient mice for hippocampus-dependent learning and memory and synaptic plasticity at hippocampal CA3-CA1 synapses. Interestingly, the mutant mice exhibited enhanced performance in several hippocampus-dependent learning and memory tasks. However, electrophysiological studies revealed no alteration in the Ca(2+) current density, the frequency and amplitude of miniature excitatory postsynaptic currents, and the basal synaptic transmission in the mutant hippocampus. On the other hand, however, N-methyl-d-aspartate receptor (NMDAR)-mediated synaptic currents and NMDAR-dependent long term potentiation were significantly increased in the mutant. Protein blot analysis showed a slight increase in the level of NMDAR-2B in the mutant hippocampus. Our results suggest a possibility that, unrelated to VDCCs regulation, Ca(v)beta3 negatively regulates the NMDAR activity in the hippocampus and thus activity-dependent synaptic plasticity and cognitive behaviors in the mouse.
Collapse
Affiliation(s)
- Daejong Jeon
- From the Center for Neural Science, Korea Institute of Science and Technology, Seoul 136-791, Korea
- National Creative Research Initiative Center for Sensory Research, Seoul National University, College of Pharmacy, Seoul 151-742, Korea
| | - Inseon Song
- From the Center for Neural Science, Korea Institute of Science and Technology, Seoul 136-791, Korea
| | - William Guido
- the Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, and
| | - Karam Kim
- National Creative Research Initiative Center for Synaptogenesis, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Eunjoon Kim
- National Creative Research Initiative Center for Synaptogenesis, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Uhtaek Oh
- National Creative Research Initiative Center for Sensory Research, Seoul National University, College of Pharmacy, Seoul 151-742, Korea
| | - Hee-Sup Shin
- From the Center for Neural Science, Korea Institute of Science and Technology, Seoul 136-791, Korea
| |
Collapse
|
281
|
Scita G, Confalonieri S, Lappalainen P, Suetsugu S. IRSp53: crossing the road of membrane and actin dynamics in the formation of membrane protrusions. Trends Cell Biol 2008; 18:52-60. [DOI: 10.1016/j.tcb.2007.12.002] [Citation(s) in RCA: 194] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 12/04/2007] [Accepted: 12/04/2007] [Indexed: 11/16/2022]
|
282
|
Ueta Y, Yamamoto R, Sugiura S, Inokuchi K, Kato N. Homer 1a suppresses neocortex long-term depression in a cortical layer-specific manner. J Neurophysiol 2007; 99:950-7. [PMID: 18077661 DOI: 10.1152/jn.01101.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Homer1a/Vesl-1S is an activity-dependently induced member of the scaffold protein family Homer/Vesl, which is known to link group I metabotropic glutamate receptors (mGluRs) to endoplasmic calcium release channels and to regulate them. Here we studied roles of Homer 1a in inducing long-term depression (LTD) in rat visual cortex slices. Homer 1a protein was injected by diffusion from whole cell patch pipettes. In layer VI pyramidal cells, LTD was reduced in magnitude with Homer 1a. LTD in layer VI was suppressed by applying antagonists of mGluR5, a subtype of group I mGluRs expressed with higher density than mGluR1 in neocortex pyramidal cells, or inositol-1,4,5-triphosphate receptors (IP3Rs) but not that against N-methyl-d-aspartate receptors (NMDARs). In layer II/III or layer V, Homer 1a injection was unable to affect LTD, which is mostly dependent on NMDARs and not on group I mGluRs in these layers. To examine the effects of endogenous Homer 1a, electroconvulsive shock (ECS) was applied. Homer 1a thereby induced, as did Homer 1a injection, reduced LTD magnitude in layer VI pyramidal cells and failed to do so in layer II/III or layer V pyramidal cells. These results indicate that both exo- and endogenous Homer 1a suppressed LTD in a cortical layer-specific manner, and its layer-specificity may be explained by the high affinity of Homer 1a to group I mGluRs.
Collapse
Affiliation(s)
- Yoshifumi Ueta
- Department of Physiology, Kanazawa Medical University, 920-0293 Ishikawa, Japan
| | | | | | | | | |
Collapse
|
283
|
Kitamura C, Takahashi M, Kondoh Y, Tashiro H, Tashiro T. Identification of synaptic activity-dependent genes by exposure of cultured cortical neurons to tetrodotoxin followed by its withdrawal. J Neurosci Res 2007; 85:2385-99. [PMID: 17551986 DOI: 10.1002/jnr.21391] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Activity-dependent gene expression is one of the key mechanisms of synaptic plasticity that form the basis of higher order functions such as learning and memory. In the present study, we surveyed for activity-dependent genes by analyzing gene expression changes accompanying reversible inhibition of synaptic activity by tetrodotoxin (TTX) using two types of DNA microarrays; our focused oligo DNA microarray "Synaptoarray" and the commercially available high-density array. Cerebral cortical cells from E18 rat embryos were cultured for 14 days to ensure synaptogenesis, then treated with 1 muM TTX for 48 hr without detectable effect on cell viability. Synaptic density estimated by the amount of Synapsin I and Synaptotagmin I was decreased 21-24% by TTX treatment, but recovered to the control level 48 hr after TTX withdrawal. Comparison of gene expression profiles by competitive hybridization of fluorescently labeled cRNA from TTX-treated and control cells showed an overall downregulation of the genes on the Synaptoarray by TTX-treatment with different recovery rates after TTX withdrawal. With 16 representative genes, microarray data were validated by real-time PCR analysis. Genes most severely downregulated by TTX and upregulated above the control level at 5 hr after TTX withdrawal were munc13-1 (involved in docking and priming of synaptic vesicles) and Shank2 (involved in the postsynaptic scaffold). In addition, comprehensive screening at 5 hr after TTX withdrawal using high density arrays resulted in additional identification of Rgs2, a regulator of trimeric G-protein signaling, as an activity-dependent gene. These three genes are thus likely to be key factors in the regulation of synaptic plasticity. (c) 2007 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Chikara Kitamura
- Department of Chemistry and Biological Science, School of Science and Engineering, Aoyama-Gakuin University, Sagamihara, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
284
|
Abstract
Endocytosis, exocytosis, and lateral diffusion are key mechanisms for AMPA receptor trafficking. Endocytosis of AMPARs and other postsynaptic proteins has been proposed to occur at specific endocytic zones (EZs), but the mechanisms that regulate this process are not at all clear. In this issue of Neuron, Lu et al. show that correct synaptic EZ positioning requires links between the GTPase dynamin-3 and the Homer/Shank complex.
Collapse
Affiliation(s)
- Frédéric Jaskolski
- MRC Centre for Synaptic Plasticity, Department of Anatomy, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Stéphane Martin
- MRC Centre for Synaptic Plasticity, Department of Anatomy, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Jeremy M. Henley
- MRC Centre for Synaptic Plasticity, Department of Anatomy, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
285
|
Vessey JP, Karra D. More than just synaptic building blocks: scaffolding proteins of the post-synaptic density regulate dendritic patterning. J Neurochem 2007; 102:324-32. [PMID: 17596209 DOI: 10.1111/j.1471-4159.2007.04662.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The dendritic arbor is responsible for receiving and consolidating neuronal input. Outgrowth and morphogenesis of the arbor are complex stages of development that are poorly understood. However, recent findings have identified synaptic scaffolding proteins as novel regulators of these important events. Scaffolding proteins are enriched in the post-synaptic density where they bind and bring into close proximity neurotransmitter receptors, signaling molecules, and regulators of the actin cytoskeleton. This property is important for dendritic spine morphogenesis and maintenance in the mature neuron. Scaffolding proteins are now being described as key regulators of neurite outgrowth, dendritic development, and pattern formation in immature neurons. These proteins, which include post-synaptic-95, Shank and Densin-180, as well as many of their interacting partners, appear to regulate both the microtubule and actin cytoskeleton to influence dendrite morphology. Through a large array of protein-protein interaction domains, scaffolding proteins are able to form large macromolecular complexes that include cytoskeletal motor proteins as well as microtubule and actin regulatory molecules. Together, the new findings form a persuasive argument that scaffolding proteins deliver critical regulatory elements to sites of dendritic outgrowth and branching to modulate the formation and maintenance of the dendritic arbor.
Collapse
Affiliation(s)
- John P Vessey
- Department of Neural Cell Biology, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | | |
Collapse
|
286
|
Kneussel M, Loebrich S. Trafficking and synaptic anchoring of ionotropic inhibitory neurotransmitter receptors. Biol Cell 2007; 99:297-309. [PMID: 17504238 DOI: 10.1042/bc20060120] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neurotransmitter receptors are subject to microtubule-based transport between intracellular organelles and the neuronal plasma membrane. Receptors that arrive at plasma membrane compartments diffuse laterally within the plane of the cellular surface. To achieve immobilization at their sites of action, cytoplasmic receptor residues bind to submembrane proteins, which are coupled to the underlying cytoskeleton by multiprotein scaffolds. GABA(A)Rs (gamma-aminobutyric type A receptors) and GlyRs (glycine receptors) are the major inhibitory receptors in the central nervous system. At inhibitory postsynaptic sites, all GlyRs and the majority of GABA(A)Rs directly or indirectly couple to gephyrin, a multimeric PSD (postsynaptic density) component. In addition to cluster formations at axo-dendritic contacts, individual GABA(A)R subtypes also anchor and concentrate at extrasynaptic positions, either through association with gephyrin or direct interaction with the ERM (ezrin/radixin/moesin) family protein radixin. In addition to their role in diffusion trapping of surface receptors, scaffold components also undergo rapid exchange to/from and between postsynaptic specializations, leading to a dynamic equilibrium of receptor-scaffold complexes. Moreover, scaffold components serve as adaptor proteins that mediate specificity in intracellular transport complexes. In the present review, we discuss the dynamic delivery, stabilization and removal of inhibitory receptors at synaptic sites.
Collapse
Affiliation(s)
- Matthias Kneussel
- Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universität Hamburg, Falkenried 94, Germany.
| | | |
Collapse
|
287
|
Liebau S, Vaida B, Storch A, Boeckers TM. Maturation of synaptic contacts in differentiating neural stem cells. Stem Cells 2007; 25:1720-9. [PMID: 17379760 DOI: 10.1634/stemcells.2006-0823] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
NSCs are found in the developing brain, as well as in the adult brain. They are self-renewing cells that maintain the capacity to differentiate into all major brain-specific cell types, such as glial cells and neurons. However, it is still unclear whether these cells are capable of gaining full functionality, which is one of the major prerequisites for NSC-based cell replacement strategies of neurological diseases. The ability to establish and maintain polarized excitatory synaptic contacts would be one of the basic requirements for intercellular communication and functional integration into existing neuronal networks. In primary cultures of hippocampal neurons, it has already been shown that synaptogenesis is characterized by a well-ordered, time-dependent targeting and recruitment of pre- and postsynaptic proteins. In this study, we investigated the expression and localization of important pre- and postsynaptic proteins, including Bassoon and synaptophysin, as well as proteins of the ProSAP/Shank family, in differentiating rat fetal mesencephalic NSCs. Moreover, we analyzed the ultrastructural features of neuronal cell-cell contacts during synaptogenesis. We show that NSCs express and localize cytoskeletal and scaffolding molecules of the pre- and postsynaptic specializations in a well-defined temporal order, leading to mature synaptic contacts after 14 days of differentiation. The temporal and spatial pattern of synaptic maturation is comparable to synaptogenesis of hippocampal neurons grown in primary culture. Therefore, with respect to the general ability to create mature synaptic contacts, NSCs seem to be well equipped to potentially compensate for lost or injured brain tissue. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Stefan Liebau
- Institute of Anatomy and Cell Biology, Ulm University, Albert Einstein Allee 11, 89081 Ulm, Germany
| | | | | | | |
Collapse
|
288
|
Proepper C, Johannsen S, Liebau S, Dahl J, Vaida B, Bockmann J, Kreutz MR, Gundelfinger ED, Boeckers TM. Abelson interacting protein 1 (Abi-1) is essential for dendrite morphogenesis and synapse formation. EMBO J 2007; 26:1397-409. [PMID: 17304222 PMCID: PMC1817621 DOI: 10.1038/sj.emboj.7601569] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Accepted: 01/03/2007] [Indexed: 01/27/2023] Open
Abstract
Synaptogenesis and synaptic plasticity depend crucially on the dynamic and locally specific regulation of the actin cytoskeleton. We identified an important component for controlled actin assembly, abelson interacting protein-1 (Abi-1), as a binding partner for the postsynaptic density (PSD) protein ProSAP2/Shank3. During early neuronal development, Abi-1 is localized in neurites and growth cones; at later stages, the protein is enriched in dendritic spines and PSDs, as are components of a trimeric complex consisting of Abi-1, Eps8 and Sos-1. Abi-1 translocates upon NMDA application from PSDs to nuclei. Nuclear entry depends on abelson kinase activity. Abi-1 co-immunoprecipitates with the transcription factor complex of Myc/Max proteins and enhances E-box-regulated gene transcription. Downregulation of Abi-1 by small interfering RNA results in excessive dendrite branching, immature spine and synapse morphology and a reduction of synapses, whereas overexpression of Abi-1 has the opposite effect. Data show that Abi-1 can act as a specific synapto-nuclear messenger and is essentially involved in dendrite and synapse formation.
Collapse
Affiliation(s)
| | - Svenja Johannsen
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Stefan Liebau
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Janine Dahl
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Bianca Vaida
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Michael R Kreutz
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, IfN, Magdeburg, Germany
| | - Eckart D Gundelfinger
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, IfN, Magdeburg, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
- Institute for Anatomy and Cell Biology, Ulm University, Albert Einstein Allee 11, 89081 Ulm, Germany. Tel.: +49 731 5023220; Fax: +49 731 5023217; E-mail:
| |
Collapse
|
289
|
Tsuriel S, Geva R, Zamorano P, Dresbach T, Boeckers T, Gundelfinger ED, Garner CC, Ziv NE. Local sharing as a predominant determinant of synaptic matrix molecular dynamics. PLoS Biol 2007; 4:e271. [PMID: 16903782 PMCID: PMC1540708 DOI: 10.1371/journal.pbio.0040271] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Accepted: 06/14/2006] [Indexed: 01/03/2023] Open
Abstract
Recent studies suggest that central nervous system synapses can persist for weeks, months, perhaps lifetimes, yet little is known as to how synapses maintain their structural and functional characteristics for so long. As a step toward a better understanding of synaptic maintenance we examined the loss, redistribution, reincorporation, and replenishment dynamics of Synapsin I and ProSAP2/Shank3, prominent presynaptic and postsynaptic matrix molecules, respectively. Fluorescence recovery after photobleaching and photoactivation experiments revealed that both molecules are continuously lost from, redistributed among, and reincorporated into synaptic structures at time-scales of minutes to hours. Exchange rates were not affected by inhibiting protein synthesis or proteasome-mediated protein degradation, were accelerated by stimulation, and greatly exceeded rates of replenishment from somatic sources. These findings indicate that the dynamics of key synaptic matrix molecules may be dominated by local protein exchange and redistribution, whereas protein synthesis and degradation serve to maintain and regulate the sizes of local, shared pools of these proteins. To understand processes involved in synaptic maintenance, the authors examine the loss, redistribution, reincorporation and replenishment dynamics of two key synaptic proteins, Synapsin I and ProSAP2/Shank3.
Collapse
Affiliation(s)
- Shlomo Tsuriel
- The Rappaport Family Institute for Research in the Medical Sciences, Technion Faculty of Medicine, Haifa, Israel
- The Department of Physiology, Technion Faculty of Medicine, Haifa, Israel
| | - Ran Geva
- The Rappaport Family Institute for Research in the Medical Sciences, Technion Faculty of Medicine, Haifa, Israel
- The Department of Physiology, Technion Faculty of Medicine, Haifa, Israel
| | - Pedro Zamorano
- Department of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Thomas Dresbach
- Institute of Anatomy and Cell Biology II, University of Heidelberg, Heidelberg, Germany
| | - Tobias Boeckers
- Institute of Anatomy and Cell Biology, University of Ulm, Ulm, Germany
| | - Eckart D Gundelfinger
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Craig C Garner
- Department of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Noam E Ziv
- The Rappaport Family Institute for Research in the Medical Sciences, Technion Faculty of Medicine, Haifa, Israel
- The Department of Physiology, Technion Faculty of Medicine, Haifa, Israel
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
290
|
Lee JH, Richter W, Namkung W, Kim KH, Kim E, Conti M, Lee MG. Dynamic regulation of cystic fibrosis transmembrane conductance regulator by competitive interactions of molecular adaptors. J Biol Chem 2007; 282:10414-22. [PMID: 17244609 DOI: 10.1074/jbc.m610857200] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Disorganized ion transport caused by hypo- or hyperfunctioning of the cystic fibrosis transmembrane conductance regulator (CFTR) can be detrimental and may result in life-threatening diseases such as cystic fibrosis or secretory diarrhea. Thus, CFTR is controlled by elaborate positive and negative regulations for an efficient homeostasis. It has been shown that expression and activity of CFTR can be regulated either positively or negatively by PDZ (PSD-95/discs large/ZO-1) domain-based adaptors. Although a positive regulation by PDZ domain-based adaptors such as EBP50/NHERF1 is established, the mechanisms for negative regulation of the CFTR by Shank2, as well as the effects of multiple adaptor interactions, are not known. Here we demonstrate a physical and physiological competition between EBP50-CFTR and Shank2-CFTR associations and the dynamic regulation of CFTR activity by these positive and negative interactions using the surface plasmon resonance assays and consecutive patch clamp experiments. Furthermore whereas EBP50 recruits a cAMP-dependent protein kinase (PKA) complex to CFTR, Shank2 was found to be physically and functionally associated with the cyclic nucleotide phosphodiesterase PDE4D that precludes cAMP/PKA signals in epithelial cells and mouse brains. These findings strongly suggest that balanced interactions between the membrane transporter and multiple PDZ-based adaptors play a critical role in the homeostatic regulation of epithelial transport and possibly the membrane transport in other tissues.
Collapse
Affiliation(s)
- Ji Hyun Lee
- Department of Pharmacology, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | | | | | | | | | |
Collapse
|
291
|
Okamoto N, Kubota T, Nakamura Y, Murakami R, Nishikubo T, Tanaka I, Takahashi Y, Hayashi S, Imoto I, Inazawa J, Hosokai N, Kohsaka S, Uchino S. 22q13 microduplication in two patients with common clinical manifestations: A recognizable syndrome? Am J Med Genet A 2007; 143A:2804-9. [DOI: 10.1002/ajmg.a.31771] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
292
|
Tlili A, Masmoudi S, Dhouib H, Bouaziz S, Rebeh IB, Chouchen J, Turki K, Benzina Z, Charfedine I, Drira M, Ayadi H. Localization of a novel autosomal recessive non-syndromic hearing impairment locus DFNB63 to chromosome 11q13.3-q13.4. Ann Hum Genet 2006; 71:271-5. [PMID: 17166180 DOI: 10.1111/j.1469-1809.2006.00337.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hereditary hearing impairment is the most genetically heterogeneous trait known in humans. So far, 50 published autosomal recessive non-syndromic hearing impairment (ARNSHI) loci have been mapped, and 23 ARNSHI genes have been identified. Here, we report the mapping of a novel ARNSHI locus, DFNB63, to chromosome 11q13.3-q13.4 in a large consanguineous Tunisian family. A maximum LOD score of 5.33 was obtained with microsatellite markers D11S916 and D11S4207. Haplotype analysis defined a 5.55 Mb critical region between microsatellite markers D11S4136 and D11S4081. DFNB63 represents the sixth ARNSHI locus mapped to chromosome 11. We positionally excluded MYO7A from being the DFNB63-causative gene. In addition, the screening of two candidate genes, SHANK2 and KCNE3, failed to reveal any disease-causing mutations.
Collapse
Affiliation(s)
- A Tlili
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Tunisie
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
293
|
Lamprecht G, Seidler U. The emerging role of PDZ adapter proteins for regulation of intestinal ion transport. Am J Physiol Gastrointest Liver Physiol 2006; 291:G766-77. [PMID: 16798722 DOI: 10.1152/ajpgi.00135.2006] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In the gastrointestinal tract, CFTR, in conjunction with one or several members of the SLC26 anion exchanger family, mediates electrogenic Cl- and HCO3- secretion. Na+/H+ exchanger isoform NHE3, on the other hand, coupled to one or several of the SLC26 isoforms, mediates electroneutral NaCl absorption. The agonist-induced activation of anion secretion and inhibition of salt absorption causes secretory diarrhea. Current dogma sees the formation of a multiprotein complex of transport proteins, postsynaptic density-95/discs large/zonula occludens-1 (PDZ) adapter proteins, anchoring proteins, the cytoskeleton, and the involved protein kinases as one crucial step in the regulation of these transport processes. Data obtained in heterologous expression studies suggest an important role of these PDZ adapter proteins in trafficking, endocytic recycling, and membrane retention of the respective transmembrane proteins. This article reviews recent advances in our understanding of the role of the PDZ adapter proteins NHERF, E3KARP, PDZK1, IKEPP (NHERF-1 to NHERF-4), CAL, and Shank-2 that bind to CFTR, NHE3, and the intestinal SLC26 members in the regulation of intestinal fluid transport. Current concepts are mostly derived from heterologous expression studies and studies on their role in organ physiology are still in infancy. Recently, however, PDZ adapter protein-deficient mice and organ-specific cell lines have become available, and the first results suggest a more cell-type and possibly signal-specific role of these adapter proteins. This opens the potential for drug development targeted to PDZ domain interactions, which is, in theory, one of the most efficient antidiarrheal strategies.
Collapse
Affiliation(s)
- G Lamprecht
- First Medical Department, University of Tuebingen, Germany
| | | |
Collapse
|
294
|
Kiselyov K, Wang X, Shin DM, Zang W, Muallem S. Calcium signaling complexes in microdomains of polarized secretory cells. Cell Calcium 2006; 40:451-9. [PMID: 17034849 DOI: 10.1016/j.ceca.2006.08.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Accepted: 08/23/2006] [Indexed: 10/24/2022]
Abstract
The highly polarized nature of epithelial cells in exocrine glands necessitates targeting, assembly into complexes and confinement of the molecules comprising the Ca(2+) signaling apparatus, to cellular microdomains. Such high degree of polarized localization has been shown for all Ca(2+) signaling molecules tested, including G protein coupled receptors and their associated proteins, Ca(2+) pumps, Ca(2+) influx channels at the plasma membrane and Ca(2+) release channels in the endoplasmic reticulum. Although the physiological significance of polarized Ca(2+) signaling is clear, little is known about the mechanism of targeting, assembly and retention of Ca(2+) signaling complexes in cellular microdomains. The present review attempts to summarize the evidence in favor of polarized expression of Ca(2+) signaling proteins at the apical pole of secretory cells with emphasis on the role of scaffolding proteins in the assembly and function of the Ca(2+) signaling complexes. The consequence of polarized enrichment of Ca(2+) signaling complexes at the apical pole is generation of an apical to basal pole gradient of cell responsiveness that, at low physiological agonist concentrations, limits Ca(2+) spikes to the apical pole, and when a Ca(2+) wave occurs, it always propagates from the apical to the basal pole. Our understanding of Ca(2+) signaling in microdomains is likely to increase rapidly with the application of techniques to controllably and selectively disrupt components of the complexes and apply high resolution recording techniques, such as TIRF microscopy to this problem.
Collapse
Affiliation(s)
- Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA. kiselyov+@pitt.edu
| | | | | | | | | |
Collapse
|
295
|
Paquet M, Asay MJ, Fam SR, Inuzuka H, Castleberry AM, Oller H, Smith Y, Yun CC, Traynelis SF, Hall RA. The PDZ scaffold NHERF-2 interacts with mGluR5 and regulates receptor activity. J Biol Chem 2006; 281:29949-61. [PMID: 16891310 PMCID: PMC4670778 DOI: 10.1074/jbc.m602262200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The two members of the group I metabotropic glutamate receptor family, mGluR1 and mGluR5, both couple to G(q) to mediate rises in intracellular calcium. The alternatively spliced C termini (CT) of mGluRs1 and 5are known to be critical for regulating receptor activity and to terminate in motifs suggestive of potential interactions with PDZ domains. We therefore screened the CTs of both mGluR1a and mGluR5 against a PDZ domain proteomic array. Out of 96 PDZ domains examined, the domain that bound most strongly to mGluR5-CT was the second PDZ domain of the Na(+)/H(+) exchanger regulatory factor 2 (NHERF-2). This interaction was confirmed by reverse overlay, and a single point mutation to the mGluR5-CT was found to completely disrupt the interaction. Full-length mGluR5 robustly associated with full-length NHERF-2 in cells, as assessed by co-immunoprecipitation and confocal microscopy experiments. In contrast, mGluR1a was found to bind NHERF-2 in vitro with a weaker affinity than mGluR5, and furthermore mGluR1a did not detectably associate with NHERF-2 in a cellular context. Immunohistochemical experiments revealed that NHERF-2 and mGluR5 exhibit overlapping patterns of expression in mouse brain, being found most abundantly in astrocytic processes and postsynaptic neuronal elements. In functional experiments, the interaction of NHERF-2 with mGluR5 in cells was found to prolong mGluR5-mediated calcium mobilization and to also potentiate mGluR5-mediated cell death, whereas coexpression of mGluR1a with NHERF-2 had no evident effects on mGluR1a functional activity. These observations reveal that NHERF-2 can selectively modulate mGluR5 signaling, which may contribute to cell-specific regulation of mGluR5 activity.
Collapse
Affiliation(s)
- Maryse Paquet
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Matthew J. Asay
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Sami R. Fam
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Hiroyuki Inuzuka
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Amanda M. Castleberry
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Heide Oller
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Yoland Smith
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia 30322
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia 30322
| | - C. Chris Yun
- Division of Digestive Disease, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Stephen F. Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Randy A. Hall
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
296
|
Lea PM, Faden AI. Metabotropic glutamate receptor subtype 5 antagonists MPEP and MTEP. CNS DRUG REVIEWS 2006; 12:149-66. [PMID: 16958988 PMCID: PMC6494124 DOI: 10.1111/j.1527-3458.2006.00149.x] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Glutamate regulates the function of central nervous system (CNS), in part, through the cAMP and/or IP3/DAG second messenger-associated metabotropic glutamate receptors (mGluRs). The mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) has been extensively used to elucidate potential physiological and pathophysiological functions of mGluR5. Unfortunately, recent evidence indicates significant non-specific actions of MPEP, including inhibition of NMDA receptors. In contrast, in vivo and in vitro characterization of the newer mGluR5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) indicates that it is more highly selective for mGluR5 over mGluR1, has no effect on other mGluR subtypes, and has fewer off-target effects than MPEP. This article reviews literature on both of these mGluR5 antagonists, which suggests their possible utility in neurodegeneration, addiction, anxiety and pain management.
Collapse
Affiliation(s)
| | - Alan I. Faden
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
297
|
Abstract
Glutamatergic synapses in the central nervous system are characterized by an electron-dense web underneath the postsynaptic membrane; this web is called the postsynaptic density (PSD). PSDs are composed of a dense network of several hundred proteins, creating a macromolecular complex that serves a wide range of functions. Prominent PSD proteins such as members of the MaGuk or ProSAP/Shank family build up a dense scaffold that creates an interface between clustered membrane-bound receptors, cell adhesion molecules and the actin-based cytoskeleton. Moreover, kinases, phosphatases and several proteins of different signalling pathways are specifically localized within the spine/PSD compartment. Small GTPases and regulating proteins are also enriched in PSDs being the molecular basis for regulated structural changes of cytoskeletal components within the synapse in response to external or internal stimuli, e.g. synaptic activation. This synaptic rearrangement (structural plasticity) is a rapid process and is believed to underlie learning and memory formation. The characterization of synapse/PSD proteins is especially important in the light of recent data suggesting that several mental disorders have their molecular defect at the synapse/PSD level.
Collapse
Affiliation(s)
- T M Boeckers
- Department of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany.
| |
Collapse
|
298
|
Gundelfinger ED, Boeckers TM, Baron MK, Bowie JU. A role for zinc in postsynaptic density asSAMbly and plasticity? Trends Biochem Sci 2006; 31:366-73. [PMID: 16793273 DOI: 10.1016/j.tibs.2006.05.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Revised: 04/21/2006] [Accepted: 05/25/2006] [Indexed: 01/06/2023]
Abstract
Chemical synapses are asymmetric cell junctions that mediate communication between neurons. Multidomain scaffolding proteins of the Shank family act as major organizing elements of the "postsynaptic density"--that is, the cytoskeletal protein matrix associated with the postsynaptic membrane. A recent study has shown that the C-terminal sterile alpha-motif or "SAM domain" of Shank3 (also known as ProSAP2) can form two-dimensional sheets of helical fibers. Assembly and packaging of these fibers are markedly enhanced by the presence of Zn2+ ions. Zn2+ can be released together with glutamate from synaptic vesicles and can enter the postsynaptic cell through specific ionotropic receptors. Based on these observations, we propose a new model of synaptic plasticity in which Zn2+ influx directly and instantly modulates the structure and function of the postsynaptic density.
Collapse
Affiliation(s)
- Eckart D Gundelfinger
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Brenneckestrasse 6, 39118 Magdeburg, Germany.
| | | | | | | |
Collapse
|
299
|
Uchino S, Wada H, Honda S, Nakamura Y, Ondo Y, Uchiyama T, Tsutsumi M, Suzuki E, Hirasawa T, Kohsaka S. Direct interaction of post-synaptic density-95/Dlg/ZO-1 domain-containing synaptic molecule Shank3 with GluR1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor. J Neurochem 2006; 97:1203-14. [PMID: 16606358 DOI: 10.1111/j.1471-4159.2006.03831.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A class of scaffolding protein containing the post-synaptic density-95/Dlg/ZO-1 (PDZ) domain is thought to be involved in synaptic trafficking of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors during development. To clarify the molecular mechanism of AMPA receptor trafficking, we performed a yeast two-hybrid screening system using the cytoplasmic tail of the GluR1 subunit of AMPA receptor as a bait and identified a synaptic molecule, Shank3/ProSAP2, as a GluR1 subunit-interacting molecule. Shank3 is a PDZ domain-containing multidomain protein and is predominantly expressed in developing neurons. Using the glutathione S-transferase pull-down assay and immunoprecipitation technique we demonstrated that the GluR1 subunit directly binds to the PDZ domain of Shank3 via its carboxyl terminal PDZ-binding motif. We raised anti-Shank3 antibody to investigate the expression of Shank3 in cortical neurons. The pattern of Shank3 immunoreactivity was strikingly punctate, mainly observed in the spines, and closely matched the pattern of post-synaptic density-95 immunoreactivity, indicating that Shank3 is colocalized with post-synaptic density-95 in the same spines. When Shank3 and the GluR1 subunit were overexpressed in primary cortical neurons, they were also colocalized in the spines. Taken together with the biochemical interaction of Shank3 with the GluR1 subunit, these results suggest that Shank3 is an important molecule that interacts with GluR1 AMPA receptor at synaptic sites of developing neurons.
Collapse
Affiliation(s)
- Shigeo Uchino
- Department of Neurochemistry, National Institute of Neuroscience, Kodaira, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
300
|
Wendholt D, Spilker C, Schmitt A, Dolnik A, Smalla KH, Proepper C, Bockmann J, Sobue K, Gundelfinger ED, Kreutz MR, Boeckers TM. ProSAP-interacting protein 1 (ProSAPiP1), a novel protein of the postsynaptic density that links the spine-associated Rap-Gap (SPAR) to the scaffolding protein ProSAP2/Shank3. J Biol Chem 2006; 281:13805-13816. [PMID: 16522626 DOI: 10.1074/jbc.m601101200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ProSAPs/Shanks are a family of proteins that have a major scaffolding function for components of the postsynaptic density (PSD) of excitatory brain synapses. Members of the family harbor a variety of domains for protein-protein interactions, one of which is a unique PDZ domain that differs significantly from those of other proteins. We have identified a novel binding partner for this PDZ domain, termed ProSAPiP1, that is highly enriched in the PSD and shares significant sequence homology with the PSD protein PSD-Zip70. Both molecules code for a Fez1 domain that can be found in a total of four related proteins. ProSAPiP1 is widely expressed in rat brain and co-localizes with ProSAP2/Shank3 in excitatory spines and synapses. ProSAP2/Shank3 co-immunoprecipitates with ProSAPiP1 but not with PSD-Zip70. Both proteins, however, bind and recruit SPAR to synapses with a central coiled-coil region that harbors a leucine zipper motif. This region is also responsible for homo- and heteromultimerization of ProSAPiP1 and PSD-Zip70. Thus, ProSAPiP1 and PSD-Zip70 are founders of a novel family of scaffolding proteins, the "Fezzins," which adds further complexity to the organization of the PSD protein network.
Collapse
Affiliation(s)
- Doreen Wendholt
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Christina Spilker
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Angelika Schmitt
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Anna Dolnik
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Karl-Heinz Smalla
- AG Molecular Mechanisms of Plasticity, Department of Neurochemistry/Molecular Biology, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Christian Proepper
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Kenji Sobue
- Department of Neuroscience, Osaka University School of Medicine, Suita, Osaka 565, Japan
| | - Eckart D Gundelfinger
- AG Molecular Mechanisms of Plasticity, Department of Neurochemistry/Molecular Biology, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Michael R Kreutz
- AG Molecular Mechanisms of Plasticity, Department of Neurochemistry/Molecular Biology, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany,.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany.
| |
Collapse
|