151
|
Dosemeci A, Weinberg RJ, Reese TS, Tao-Cheng JH. The Postsynaptic Density: There Is More than Meets the Eye. Front Synaptic Neurosci 2016; 8:23. [PMID: 27594834 PMCID: PMC4990544 DOI: 10.3389/fnsyn.2016.00023] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/25/2016] [Indexed: 11/28/2022] Open
Abstract
The postsynaptic density (PSD), apparent in electron micrographs as a dense lamina just beneath the postsynaptic membrane, includes a deeper layer, the “pallium”, containing a scaffold of Shank and Homer proteins. Though poorly defined in traditionally prepared thin-section electron micrographs, the pallium becomes denser and more conspicuous during intense synaptic activity, due to the reversible addition of CaMKII and other proteins. In this Perspective article, we review the significance of CaMKII-mediated recruitment of proteins to the pallium with respect to both the trafficking of receptors and the remodeling of spine shape that follow synaptic stimulation. We suggest that the level and duration of CaMKII translocation and activation in the pallium will shape activity-induced changes in the spine.
Collapse
Affiliation(s)
- Ayse Dosemeci
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH) Bethesda, MD, USA
| | - Richard J Weinberg
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill Chapel Hill, NC, USA
| | - Thomas S Reese
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH) Bethesda, MD, USA
| | - Jung-Hwa Tao-Cheng
- Electron Microscopy Facility, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH) Bethesda, MD, USA
| |
Collapse
|
152
|
Jones OD. Do group I metabotropic glutamate receptors mediate LTD? Neurobiol Learn Mem 2016; 138:85-97. [PMID: 27545442 DOI: 10.1016/j.nlm.2016.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 08/01/2016] [Accepted: 08/17/2016] [Indexed: 01/31/2023]
Abstract
Synapses undergo significant structural and functional reorganization in response to varying patterns of stimulation. These forms of plasticity are considered fundamental to cognition and neuronal homeostasis. An increasing number of reports highlight the importance of activity-dependent synaptic strengthening (long term potentiation: LTP) for learning. However, the functional significance of activity-dependent weakening of synapses (long term depression: LTD) remains relatively poorly understood. One form of synaptic weakening, induced by group I metabotropic glutamate receptors (mGluRs), has received significant attention from a mechanistic point of view and because of its augmentation in a murine model of Fragile X Syndrome. Yet, studies of this form of plasticity often yield confusing, contradictory results. These conflicting findings are likely attributable to the bulk stimulation and recording techniques often used to study synaptic plasticity (typically involving evoked extracellular recordings, which represent the summed activity of many synapses). Such studies inherently blur the identity of the synapses undergoing change, thus giving the illusion that synapses per se are being modified when in fact this may only be true of a specific subset of synapses. Indeed, studies employing minimal synaptic activation paint a fundamentally different picture of what is commonly called "mGluR-LTD". Here, I review the evidence in favour of group I mGluRs as mediators of various forms of synaptic downregulation and attempt to explain discrepancies in the literature. I argue that, while multiple forms of synaptic weakening may be triggered by these receptors, the canonical form of group I mGluR-mediated depression, mGluR-LTD, is in fact not a depression of basal synaptic responses. Rather, it is a reversal of established LTP and thus a form of depotentiation. Far from being arbitrary, this distinction has significant implications for the role of group I mGluRs in cognition, both in the healthy brain and in pathological conditions. Further, the differential actions of group I mGluRs at naïve and potentiated synapses suggest these receptors signal in a state-dependent manner to regulate various stages of the learning process.
Collapse
Affiliation(s)
- Owen D Jones
- Department of Psychology, Brain Health Research Centre & Brain Research New Zealand, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
153
|
Stanika RI, Flucher BE, Obermair GJ. Regulation of Postsynaptic Stability by the L-type Calcium Channel CaV1.3 and its Interaction with PDZ Proteins. Curr Mol Pharmacol 2016; 8:95-101. [PMID: 25966696 PMCID: PMC5384370 DOI: 10.2174/1874467208666150507103716] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/29/2015] [Accepted: 04/20/2015] [Indexed: 01/24/2023]
Abstract
Alterations in dendritic spine morphology and postsynaptic structure are a hallmark of neurological disorders. Particularly spine pruning of striatal medium spiny neurons and aberrant rewiring of corticostriatal synapses have been associated with the pathology of Parkinson’s disease and L-DOPA induced dyskinesia, respectively. Owing to its low activation threshold the neuronal L-type calcium channel CaV1.3 is particularly critical in the control of neuronal excitability and thus in the calcium-dependent regulation of neuronal functions. CaV1.3 channels are located in dendritic spines and contain a C-terminal class 1 PDZ domain-binding sequence. Until today the postsynaptic PDZ domain proteins shank, densin-180, and erbin have been shown to interact with CaV1.3 channels and to modulate their current properties. Interestingly experimental evidence suggests an involvement of all three PDZ proteins as well as CaV1.3 itself in regulating dendritic and postsynaptic morphology. Here we briefly review the importance of CaV1.3 and its proposed interactions with PDZ proteins for the stability of dendritic spines. With a special focus on the pathology associated with Parkinson’s disease, we discuss the hypothesis that CaV1.3 L-type calcium channels may be critical modulators of dendritic spine stability.
Collapse
Affiliation(s)
| | | | - Gerald J Obermair
- Division of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Fritz-Pregl-Str. 3, 6020 Innsbruck, Austria.
| |
Collapse
|
154
|
Abstract
Autism spectrum disorders (ASD) are highly heterogeneous pediatric developmental disorders with estimated heritability more than 70%. Although the genetic factors in ASD are mainly unknown, a large number of gene mutations have been found, especially in genes involved in neurogenesis. The Neurexin-Neuroligin-Shank (NRXN-NLGN-SHANK) pathway plays a key role in the formation, maturation and maintenance of synapses, consistent with the hypothesis of neurodevelopmental abnormality in ASD. Presynaptic NRXNs interact with postsynaptic NLGNs in excitatory glutamatergic synapses. SHANK proteins function as core components of the postsynaptic density (PSD) by interacting with multiple proteins. Recently, deletions and point mutations of the SHANK1 gene have been detected in ASD individuals, indicating the involvement of SHANK1 in ASD. This review focuses on the function of SHANK1 protein, Shank1 mouse models, and the molecular genetics of the SHANK1 gene in human ASD.
Collapse
Affiliation(s)
- XiaoHong Gong
- MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| | - HongYan Wang
- MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
155
|
Borralleras C, Mato S, Amédée T, Matute C, Mulle C, Pérez-Jurado LA, Campuzano V. Synaptic plasticity and spatial working memory are impaired in the CD mouse model of Williams-Beuren syndrome. Mol Brain 2016; 9:76. [PMID: 27485321 PMCID: PMC4971717 DOI: 10.1186/s13041-016-0258-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/29/2016] [Indexed: 01/22/2023] Open
Abstract
Mice heterozygous for a complete deletion (CD) equivalent to the most common deletion found in individuals with Williams-Beuren syndrome (WBS) recapitulate relevant features of the neurocognitive phenotype, such as hypersociability, along with some neuroanatomical alterations in specific brain areas. However, the pathophysiological mechanisms underlying these phenotypes still remain largely unknown. We have studied the synaptic function and cognition in CD mice using hippocampal slices and a behavioral test sensitive to hippocampal function. We have found that long-term potentiation (LTP) elicited by theta burst stimulation (TBS) was significantly impaired in hippocampal field CA1 of CD animals. This deficit might be associated with the observed alterations in spatial working memory. However, we did not detect changes in presynaptic function, LTP induction mechanisms or AMPA and NMDA receptor function. Reduced levels of Brain-derived neurotrophic factor (BDNF) were present in the CA1-CA3 hippocampal region of CD mice, which could account for LTP deficits in these mice. Taken together, these results suggest a defect of CA1 synapses in CD mice to sustain synaptic strength after stimulation. These data represent the first description of synaptic functional deficits in CD mice and further highlights the utility of the CD model to study the mechanisms underlying the WBS neurocognitive profile.
Collapse
Affiliation(s)
- Cristina Borralleras
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain.,Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Susana Mato
- Department of Neuroscience, Neurotek-University of the Basque Country, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Zamudio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Thierry Amédée
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 - University of Bordeaux, F-33000, Bordeaux, France
| | - Carlos Matute
- Department of Neuroscience, Neurotek-University of the Basque Country, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Zamudio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 - University of Bordeaux, F-33000, Bordeaux, France
| | - Luis A Pérez-Jurado
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain.,Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Victoria Campuzano
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain. .,Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain.
| |
Collapse
|
156
|
SALM4 suppresses excitatory synapse development by cis-inhibiting trans-synaptic SALM3-LAR adhesion. Nat Commun 2016; 7:12328. [PMID: 27480238 PMCID: PMC4974644 DOI: 10.1038/ncomms12328] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 06/23/2016] [Indexed: 12/01/2022] Open
Abstract
Synaptic adhesion molecules regulate various aspects of synapse development, function and plasticity. These functions mainly involve trans-synaptic interactions and positive regulations, whereas cis-interactions and negative regulation are less understood. Here we report that SALM4, a member of the SALM/Lrfn family of synaptic adhesion molecules, suppresses excitatory synapse development through cis inhibition of SALM3, another SALM family protein with synaptogenic activity. Salm4-mutant (Salm4−/−) mice show increased excitatory synapse numbers in the hippocampus. SALM4 cis-interacts with SALM3, inhibits trans-synaptic SALM3 interaction with presynaptic LAR family receptor tyrosine phosphatases and suppresses SALM3-dependent presynaptic differentiation. Importantly, deletion of Salm3 in Salm4−/− mice (Salm3−/−; Salm4−/−) normalizes the increased excitatory synapse number. These results suggest that SALM4 negatively regulates excitatory synapses via cis inhibition of the trans-synaptic SALM3–LAR adhesion. Synaptic adhesion molecules regulate synapse development and function by both cis and trans-interactions. Here, Lie et al. show that postsynaptic SALM4 regulates excitatory synapse numbers by cis inhibition of the SALM3-LAR transynaptic interaction.
Collapse
|
157
|
Sarowar T, Chhabra R, Vilella A, Boeckers TM, Zoli M, Grabrucker AM. Activity and circadian rhythm influence synaptic Shank3 protein levels in mice. J Neurochem 2016; 138:887-95. [DOI: 10.1111/jnc.13709] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/01/2016] [Accepted: 06/05/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Tasnuva Sarowar
- WG Molecular Analysis of Synaptopathies, Department of Neurology; Neurocenter of Ulm University; Ulm Germany
| | - Resham Chhabra
- WG Molecular Analysis of Synaptopathies, Department of Neurology; Neurocenter of Ulm University; Ulm Germany
| | - Antonietta Vilella
- Department of Biomedical; Metabolic and Neural Sciences; University of Modena and Reggio Emilia; Modena Italy
| | | | - Michele Zoli
- Department of Biomedical; Metabolic and Neural Sciences; University of Modena and Reggio Emilia; Modena Italy
| | - Andreas M. Grabrucker
- WG Molecular Analysis of Synaptopathies, Department of Neurology; Neurocenter of Ulm University; Ulm Germany
- Institute for Anatomy and Cell Biology; Ulm University; Ulm Germany
| |
Collapse
|
158
|
Bal-Price A, Lein PJ, Keil KP, Sethi S, Shafer T, Barenys M, Fritsche E, Sachana M, Meek MEB. Developing and applying the adverse outcome pathway concept for understanding and predicting neurotoxicity. Neurotoxicology 2016; 59:240-255. [PMID: 27212452 DOI: 10.1016/j.neuro.2016.05.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/13/2016] [Accepted: 05/13/2016] [Indexed: 12/12/2022]
Abstract
The Adverse Outcome Pathway (AOP) concept has recently been proposed to support a paradigm shift in regulatory toxicology testing and risk assessment. This concept is similar to the Mode of Action (MOA), in that it describes a sequence of measurable key events triggered by a molecular initiating event in which a stressor interacts with a biological target. The resulting cascade of key events includes molecular, cellular, structural and functional changes in biological systems, resulting in a measurable adverse outcome. Thereby, an AOP ideally provides information relevant to chemical structure-activity relationships as a basis for predicting effects of structurally similar compounds. AOPs could potentially also form the basis for qualitative and quantitative predictive modeling of the human adverse outcome resulting from molecular initiating or other key events for which higher-throughput testing methods are available or can be developed. A variety of cellular and molecular processes are known to be critical for normal function of the central (CNS) and peripheral nervous systems (PNS). Because of the biological and functional complexity of the CNS and PNS, it has been challenging to establish causative links and quantitative relationships between key events that comprise the pathways leading from chemical exposure to an adverse outcome in the nervous system. Following introduction of the principles of MOA and AOPs, examples of potential or putative adverse outcome pathways specific for developmental or adult neurotoxicity are summarized and aspects of their assessment considered. Their possible application in developing mechanistically informed Integrated Approaches to Testing and Assessment (IATA) is also discussed.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy.
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Kimberly P Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Timothy Shafer
- Integrated Systems Toxicology Division, Office of Research and Development, U.S. Environmental Protection Agency, RTP, USA
| | - Marta Barenys
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Ellen Fritsche
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Magdalini Sachana
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy
| | - M E Bette Meek
- McLaughlin Centre for Risk Science, University of Ottawa, Ottawa, Canada
| |
Collapse
|
159
|
A binding site outside the canonical PDZ domain determines the specific interaction between Shank and SAPAP and their function. Proc Natl Acad Sci U S A 2016; 113:E3081-90. [PMID: 27185935 DOI: 10.1073/pnas.1523265113] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Shank and SAPAP (synapse-associated protein 90/postsynaptic density-95-associated protein) are two highly abundant scaffold proteins that directly interact with each other to regulate excitatory synapse development and plasticity. Mutations of SAPAP, but not other reported Shank PDZ domain binders, share a significant overlap on behavioral abnormalities with the mutations of Shank both in patients and in animal models. The molecular mechanism governing the exquisite specificity of the Shank/SAPAP interaction is not clear, however. Here we report that a sequence preceding the canonical PDZ domain of Shank, together with the elongated PDZ BC loop, form another binding site for a sequence upstream of the SAPAP PDZ-binding motif, leading to a several hundred-fold increase in the affinity of the Shank/SAPAP interaction. We provide evidence that the specific interaction afforded by this newly identified site is required for Shank synaptic targeting and the Shank-induced synaptic activity increase. Our study provides a molecular explanation of how Shank and SAPAP dosage changes due to their gene copy number variations can contribute to different psychiatric disorders.
Collapse
|
160
|
Ko HG, Oh SB, Zhuo M, Kaang BK. Reduced acute nociception and chronic pain in Shank2-/- mice. Mol Pain 2016; 12:12/0/1744806916647056. [PMID: 27145803 PMCID: PMC4956181 DOI: 10.1177/1744806916647056] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/04/2016] [Indexed: 01/29/2023] Open
Abstract
Autism spectrum disorder is a debilitating mental illness and social issue. Autism spectrum disorder patients suffer from social isolation, cognitive deficits, compulsive behavior, and sensory deficits, including hyposensitivity to pain. However, recent studies argued that autism spectrum disorder patients show physiological pain response and, in some cases, even extremely intense pain response to harmless stimulation. Recently, Shank gene family was reported as one of the genetic risk factors of autism spectrum disorder. Thus, in this study, we used Shank2−/− (Shank2 knock-out, KO) mice to investigate the controversial pain sensitivity issue and found that Shank2 KO mice showed reduced tactile perception and analgesia to chronic pain.
Collapse
Affiliation(s)
- Hyoung-Gon Ko
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Korea
| | - Seog-Bae Oh
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Pain Cognitive Function Research Center, Dental Research Institute Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul, Korea
| | - Min Zhuo
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Korea Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
161
|
Homberg JR, Kyzar EJ, Scattoni ML, Norton WH, Pittman J, Gaikwad S, Nguyen M, Poudel MK, Ullmann JFP, Diamond DM, Kaluyeva AA, Parker MO, Brown RE, Song C, Gainetdinov RR, Gottesman II, Kalueff AV. Genetic and environmental modulation of neurodevelopmental disorders: Translational insights from labs to beds. Brain Res Bull 2016; 125:79-91. [PMID: 27113433 DOI: 10.1016/j.brainresbull.2016.04.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/25/2016] [Accepted: 04/20/2016] [Indexed: 01/12/2023]
Abstract
Neurodevelopmental disorders (NDDs) are a heterogeneous group of prevalent neuropsychiatric illnesses with various degrees of social, cognitive, motor, language and affective deficits. NDDs are caused by aberrant brain development due to genetic and environmental perturbations. Common NDDs include autism spectrum disorder (ASD), intellectual disability, communication/speech disorders, motor/tic disorders and attention deficit hyperactivity disorder. Genetic and epigenetic/environmental factors play a key role in these NDDs with significant societal impact. Given the lack of their efficient therapies, it is important to gain further translational insights into the pathobiology of NDDs. To address these challenges, the International Stress and Behavior Society (ISBS) has established the Strategic Task Force on NDDs. Summarizing the Panel's findings, here we discuss the neurobiological mechanisms of selected common NDDs and a wider NDD+ spectrum of associated neuropsychiatric disorders with developmental trajectories. We also outline the utility of existing preclinical (animal) models for building translational and cross-diagnostic bridges to improve our understanding of various NDDs.
Collapse
Affiliation(s)
- Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Evan J Kyzar
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; The International Stress and Behavior Society (ISBS) and ZENEREI Research Center, Slidell, LA, USA
| | - Maria Luisa Scattoni
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanita, Rome, Italy
| | | | - Julian Pittman
- Department of Biological and Environmental Sciences, Troy University, Troy, AL, USA
| | - Siddharth Gaikwad
- The International Stress and Behavior Society (ISBS) and ZENEREI Research Center, Slidell, LA, USA
| | - Michael Nguyen
- The International Stress and Behavior Society (ISBS) and ZENEREI Research Center, Slidell, LA, USA; New York University School of Medicine, NY, NY, USA
| | - Manoj K Poudel
- The International Stress and Behavior Society (ISBS) and ZENEREI Research Center, Slidell, LA, USA
| | - Jeremy F P Ullmann
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia; Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - David M Diamond
- Department of Psychology, University of South Florida, Tampa, FL, USA; J.A. Haley Veterans Hospital, Research and Development Service, Tampa, FL, USA
| | - Aleksandra A Kaluyeva
- The International Stress and Behavior Society (ISBS) and ZENEREI Research Center, Slidell, LA, USA
| | - Matthew O Parker
- School of Health Sciences and Social Work, University of Portsmouth, Portsmouth, UK
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Cai Song
- Research Institute of Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong, China; Graduate Institute of Neural and Cognitive Sciences, China Medical University Hospital, Taichung, Taiwan
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Skolkovo Institute of Science and Technology, Skolkovo, Moscow Region, Russia
| | | | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| |
Collapse
|
162
|
Sarowar T, Grabrucker S, Föhr K, Mangus K, Eckert M, Bockmann J, Boeckers TM, Grabrucker AM. Enlarged dendritic spines and pronounced neophobia in mice lacking the PSD protein RICH2. Mol Brain 2016; 9:28. [PMID: 26969129 PMCID: PMC4788860 DOI: 10.1186/s13041-016-0206-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/26/2016] [Indexed: 02/16/2023] Open
Abstract
Background The majority of neurons within the central nervous system receive their excitatory inputs via small, actin-rich protrusions called dendritic spines. Spines can undergo rapid morphological alterations according to synaptic activity. This mechanism is implicated in learning and memory formation as it is ultimately altering the number and distribution of receptors and proteins at the post-synaptic membrane, thereby regulating synaptic input. The Rho-family GTPases play an important role in regulating this spine plasticity by the interaction with cytoskeletal components and several signaling pathways within the spine compartment. Rho-GAP interacting CIP4 homologue2/RICH2 is a Rho-GAP protein regulating small GTPases and was identified as an interaction partner of the scaffolding protein SHANK3 at post-synaptic densities. Results Here, we characterize the loss of RICH2 in a novel mouse model. Our results show that RICH2 KO animals display a selective and highly significant fear of novel objects and increased stereotypic behavior as well as impairment of motor learning. We found an increase in multiple spine synapses in the hippocampus and cerebellum along with alterations in receptor composition and actin polymerization. Furthermore, we observed that the loss of RICH2 leads to a disinhibition of synaptic RAC1 in vivo. Conclusions The results are in line with the reported role of RAC1 activity being essential for activity-dependent spine enlargement. Since SHANK3 mutations are known to be causative for neuropsychiatric diseases of the Autism Spectrum (ASD), a disintegrated SHANK3/RICH2 complex at synaptic sites might at least in part be responsible for abnormal spine formation and plasticity in ASDs. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0206-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tasnuva Sarowar
- WG Molecular Analysis of Synaptopathies, Neurology Department, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Stefanie Grabrucker
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Karl Föhr
- Department of Anesthesiology, University of Ulm, 89081, Ulm, Germany
| | - Katharina Mangus
- WG Molecular Analysis of Synaptopathies, Neurology Department, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Matti Eckert
- WG Molecular Analysis of Synaptopathies, Neurology Department, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany.
| | - Andreas M Grabrucker
- WG Molecular Analysis of Synaptopathies, Neurology Department, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany. .,Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, D-89081, Ulm, Germany.
| |
Collapse
|
163
|
Liu J, Liu M, Zheng B, Yao Z, Xia J. Affinity Enhancement by Ligand Clustering Effect Inspired by Peptide Dendrimers-Shank PDZ Proteins Interactions. PLoS One 2016; 11:e0149580. [PMID: 26918521 PMCID: PMC4769301 DOI: 10.1371/journal.pone.0149580] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/01/2016] [Indexed: 01/28/2023] Open
Abstract
High-affinity binders are desirable tools to probe the function that specific protein−protein interactions play in cell. In the process of seeking a general strategy to design high-affinity binders, we found a clue from the βPIX (p21-activated kinase interacting exchange factor)−Shank PDZ interaction in synaptic assembly: three PDZ-binding sites are clustered by a parallel coiled-coil trimer but bind to Shank PDZ protein with 1:1 stoichiometry (1 trimer/1 PDZ). Inspired by this architecture, we proposed that peptide dendrimer, mimicking the ligand clustering in βPIX, will also show enhanced binding affinity, yet with 1:1 stoichiometry. This postulation has been proven here, as we synthesized a set of monomeric, dimeric and trimeric peptides and measured their binding affinity and stoichiometry with Shank PDZ domains by isothermal titration calorimetry, native mass spectrometry and surface plasmon resonance. This affinity enhancement, best explained by proximity effect, will be useful to guide the design of high-affinity blockers for protein−protein interactions.
Collapse
Affiliation(s)
- Jiahui Liu
- Department of Chemistry, Centre of Novel Biomaterials, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Miao Liu
- Department of Chemistry, Centre of Novel Biomaterials, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Bo Zheng
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Zhongping Yao
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Jiang Xia
- Department of Chemistry, Centre of Novel Biomaterials, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- * E-mail:
| |
Collapse
|
164
|
Lee K, Goodman L, Fourie C, Schenk S, Leitch B, Montgomery JM. AMPA Receptors as Therapeutic Targets for Neurological Disorders. ION CHANNELS AS THERAPEUTIC TARGETS, PART A 2016; 103:203-61. [DOI: 10.1016/bs.apcsb.2015.10.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
165
|
MacGillavry HD, Kerr JM, Kassner J, Frost NA, Blanpied TA. Shank-cortactin interactions control actin dynamics to maintain flexibility of neuronal spines and synapses. Eur J Neurosci 2015; 43:179-93. [PMID: 26547831 DOI: 10.1111/ejn.13129] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/31/2022]
Abstract
The family of Shank scaffolding molecules (comprising Shank1, 2 and 3) are core components of the postsynaptic density (PSD) in neuronal synapses. Shanks link surface receptors to other scaffolding molecules within the PSD, as well as to the actin cytoskeleton. However, determining the function of Shank proteins in neurons has been complicated because the different Shank isoforms share a very high degree of sequence and domain homology. Therefore, to control Shank content while minimizing potential compensatory effects, a miRNA-based knockdown strategy was developed to reduce the expression of all synaptically targeted Shank isoforms simultaneously in rat hippocampal neurons. Using this approach, a strong (>75%) reduction in total Shank protein levels was achieved at individual dendritic spines, prompting an approximately 40% decrease in mushroom spine density. Furthermore, Shank knockdown reduced spine actin levels and increased sensitivity to the actin depolymerizing agent Latrunculin A. A SHANK2 mutant lacking the proline-rich cortactin-binding motif (SHANK2-ΔPRO) was unable to rescue these defects. Furthermore, Shank knockdown reduced cortactin levels in spines and increased the mobility of spine cortactin as measured by single-molecule tracking photoactivated localization microscopy, suggesting that Shank proteins recruit and stabilize cortactin at the synapse. Furthermore, it was found that Shank knockdown significantly reduced spontaneous remodelling of synapse morphology that could not be rescued by the SHANK2-ΔPRO mutant. It was concluded that Shank proteins are key intermediates between the synapse and the spine interior that, via cortactin, permit the actin cytoskeleton to dynamically regulate synapse morphology and function.
Collapse
Affiliation(s)
- Harold D MacGillavry
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584, CH Utrecht, the Netherlands
| | - Justin M Kerr
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Josh Kassner
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Nicholas A Frost
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
166
|
Waller JA, Chen F, Sánchez C. Vortioxetine promotes maturation of dendritic spines in vitro: A comparative study in hippocampal cultures. Neuropharmacology 2015; 103:143-54. [PMID: 26702943 DOI: 10.1016/j.neuropharm.2015.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/03/2015] [Accepted: 12/13/2015] [Indexed: 10/22/2022]
Abstract
Cognitive dysfunction is prevalent in patients with major depressive disorder (MDD), and cognitive impairments can persist after relief of depressive symptoms. The multimodal-acting antidepressant vortioxetine is an antagonist at 5-HT3, 5-HT7, and 5-HT1D receptors, a partial agonist at 5-HT1B receptors, an agonist at 5-HT1A receptors, and an inhibitor of the serotonin (5-HT) transporter (SERT) and has pro-cognitive properties. In preclinical studies, vortioxetine enhances long-term potentiation (LTP), a cellular correlate of neuroplasticity, and enhances memory in various cognitive tasks. However, the molecular mechanisms by which vortioxetine augments LTP and memory remain unknown. Dendritic spines are specialized, actin-rich microdomains on dendritic shafts and are major sites of most excitatory synapses. Since dendritic spine remodeling is implicated in synaptic plasticity and spine size dictates the strength of synaptic transmission, we assessed if vortioxetine, relative to other antidepressants including ketamine, duloxetine, and fluoxetine, plays a role in the maintenance of dendritic spine architecture in vitro. We show that vortioxetine, ketamine, and duloxetine induce spine enlargement. However, only vortioxetine treatment increased the number of spines in contact with presynaptic terminals. In contrast, fluoxetine had no effect on spine remodeling. These findings imply that the various 5-HT receptor mechanisms of vortioxetine may play a role in its effect on spine dynamics and in increasing the proportion of potentially functional synaptic contacts.
Collapse
Affiliation(s)
- Jessica A Waller
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Paramus, NJ 07652, USA
| | - Fenghua Chen
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, DK-8000 Aarhus C, Denmark; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark
| | - Connie Sánchez
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Paramus, NJ 07652, USA.
| |
Collapse
|
167
|
Queenan BN, Lee KJ, Tan H, Huganir RL, Vicini S, Pak DTS. Mapping homeostatic synaptic plasticity using cable properties of dendrites. Neuroscience 2015; 315:206-16. [PMID: 26701298 DOI: 10.1016/j.neuroscience.2015.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 11/26/2022]
Abstract
When chronically silenced, cortical and hippocampal neurons homeostatically upregulate excitatory synaptic function. However, the subcellular position of such changes on the dendritic tree is not clear. We exploited the cable-filtering properties of dendrites to derive a parameter, the dendritic filtering index (DFI), to map the spatial distribution of synaptic currents. Our analysis indicates that young rat cortical neurons globally scale AMPA receptor-mediated currents, while mature hippocampal neurons do not, revealing distinct homeostatic strategies between brain regions and developmental stages. The DFI presents a useful tool for mapping the dendritic origin of synaptic currents and the location of synaptic plasticity changes.
Collapse
Affiliation(s)
- B N Queenan
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, United States; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - K J Lee
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, United States; Research Division, Korea Brain Research Institute, Daegu, Republic of Korea
| | - H Tan
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - R L Huganir
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - S Vicini
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, United States; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - D T S Pak
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, United States; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States.
| |
Collapse
|
168
|
Zhou Y, Kaiser T, Monteiro P, Zhang X, Van der Goes MS, Wang D, Barak B, Zeng M, Li C, Lu C, Wells M, Amaya A, Nguyen S, Lewis M, Sanjana N, Zhou Y, Zhang M, Zhang F, Fu Z, Feng G. Mice with Shank3 Mutations Associated with ASD and Schizophrenia Display Both Shared and Distinct Defects. Neuron 2015; 89:147-62. [PMID: 26687841 DOI: 10.1016/j.neuron.2015.11.023] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 08/27/2015] [Accepted: 11/02/2015] [Indexed: 11/30/2022]
Abstract
Genetic studies have revealed significant overlaps of risk genes among psychiatric disorders. However, it is not clear how different mutations of the same gene contribute to different disorders. We characterized two lines of mutant mice with Shank3 mutations linked to ASD and schizophrenia. We found both shared and distinct synaptic and behavioral phenotypes. Mice with the ASD-linked InsG3680 mutation manifest striatal synaptic transmission defects before weaning age and impaired juvenile social interaction, coinciding with the early onset of ASD symptoms. On the other hand, adult mice carrying the schizophrenia-linked R1117X mutation show profound synaptic defects in prefrontal cortex and social dominance behavior. Furthermore, we found differential Shank3 mRNA stability and SHANK1/2 upregulation in these two lines. These data demonstrate that different alleles of the same gene may have distinct phenotypes at molecular, synaptic, and circuit levels in mice, which may inform exploration of these relationships in human patients.
Collapse
Affiliation(s)
- Yang Zhou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Tobias Kaiser
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Patrícia Monteiro
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; PhD Program in Experimental Biology and Biomedicine (PDBEB), Center for Neuroscience and Cell Biology, University of Coimbra, 3000-214 Coimbra, Portugal
| | - Xiangyu Zhang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Marie S Van der Goes
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dongqing Wang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Boaz Barak
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Menglong Zeng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Life Science, Center of Systems Biology and Human Health, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong
| | - Chenchen Li
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Congyi Lu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael Wells
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Aldo Amaya
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shannon Nguyen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael Lewis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Neville Sanjana
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yongdi Zhou
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Mingjie Zhang
- Division of Life Science, Center of Systems Biology and Human Health, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong
| | - Feng Zhang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zhanyan Fu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
169
|
Harony-Nicolas H, De Rubeis S, Kolevzon A, Buxbaum JD. Phelan McDermid Syndrome: From Genetic Discoveries to Animal Models and Treatment. J Child Neurol 2015; 30:1861-70. [PMID: 26350728 PMCID: PMC5321557 DOI: 10.1177/0883073815600872] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/15/2015] [Indexed: 01/16/2023]
Abstract
Phelan-McDermid syndrome or 22q13.3 deletion syndrome is a rare neurodevelopmental disorder characterized by generalized developmental delay, intellectual disability, absent or delayed speech, seizures, autism spectrum disorder, neonatal hypotonia, physical dysmorphic features, and recurrent medical comorbidities. Individuals with Phelan-McDermid syndrome have terminal deletions of the chromosomal region 22q13.3 encompassing SHANK3, a gene encoding a structural component of excitatory synapses indispensable for proper synaptogenesis and neuronal physiology, or point mutations within the gene. Here, we review the clinical aspects of the syndrome and the genetic findings shedding light onto the underlying etiology. We also provide an overview on the evidence from genetic studies and mouse models that supports SHANK3 haploinsufficiency as a major contributor of the neurobehavioral manifestations of Phelan-McDermid syndrome. Finally, we discuss how all these discoveries are uncovering the pathophysiology of Phelan-McDermid syndrome and are being translated into clinical trials for novel therapeutics ameliorating the core symptoms of the disorder.
Collapse
Affiliation(s)
- Hala Harony-Nicolas
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
170
|
PSD-95 family MAGUKs are essential for anchoring AMPA and NMDA receptor complexes at the postsynaptic density. Proc Natl Acad Sci U S A 2015; 112:E6983-92. [PMID: 26604311 DOI: 10.1073/pnas.1517045112] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The postsynaptic density (PSD)-95 family of membrane-associated guanylate kinases (MAGUKs) are major scaffolding proteins at the PSD in glutamatergic excitatory synapses, where they maintain and modulate synaptic strength. How MAGUKs underlie synaptic strength at the molecular level is still not well understood. Here, we explore the structural and functional roles of MAGUKs at hippocampal excitatory synapses by simultaneous knocking down PSD-95, PSD-93, and synapse-associated protein (SAP)102 and combining electrophysiology and transmission electron microscopic (TEM) tomography imaging to analyze the resulting changes. Acute MAGUK knockdown greatly reduces synaptic transmission mediated by α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptors (AMPARs) and N-methyl-d-aspartate receptors (NMDARs). This knockdown leads to a significant rise in the number of silent synapses, diminishes the size of PSDs without changes in pre- or postsynaptic membrane, and depletes the number of membrane-associated PSD-95-like vertical filaments and transmembrane structures, identified as AMPARs and NMDARs by EM tomography. The differential distribution of these receptor-like structures and dependence of their abundance on PSD size matches that of AMPARs and NMDARs in the hippocampal synapses. The loss of these structures following MAGUK knockdown tracks the reduction in postsynaptic AMPAR and NMDAR transmission, confirming the structural identities of these two types of receptors. These results demonstrate that MAGUKs are required for anchoring both types of glutamate receptors at the PSD and are consistent with a structural model where MAGUKs, corresponding to membrane-associated vertical filaments, are the essential structural proteins that anchor and organize both types of glutamate receptors and govern the overall molecular organization of the PSD.
Collapse
|
171
|
Burkhardt P. The origin and evolution of synaptic proteins - choanoflagellates lead the way. ACTA ACUST UNITED AC 2015; 218:506-14. [PMID: 25696814 DOI: 10.1242/jeb.110247] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The origin of neurons was a key event in evolution, allowing metazoans to evolve rapid behavioral responses to environmental cues. Reconstructing the origin of synaptic proteins promises to reveal their ancestral functions and might shed light on the evolution of the first neuron-like cells in metazoans. By analyzing the genomes of diverse metazoans and their closest relatives, the evolutionary history of diverse presynaptic and postsynaptic proteins has been reconstructed. These analyses revealed that choanoflagellates, the closest relatives of metazoans, possess diverse synaptic protein homologs. Recent studies have now begun to investigate their ancestral functions. A primordial neurosecretory apparatus in choanoflagellates was identified and it was found that the mechanism, by which presynaptic proteins required for secretion of neurotransmitters interact, is conserved in choanoflagellates and metazoans. Moreover, studies on the postsynaptic protein homolog Homer revealed unexpected localization patterns in choanoflagellates and new binding partners, both which are conserved in metazoans. These findings demonstrate that the study of choanoflagellates can uncover ancient and previously undescribed functions of synaptic proteins.
Collapse
Affiliation(s)
- Pawel Burkhardt
- Marine Biological Association, The Laboratory, Citadel Hill, Plymouth PL1 2PB, UK
| |
Collapse
|
172
|
Jaramillo TC, Speed HE, Xuan Z, Reimers JM, Liu S, Powell CM. Altered Striatal Synaptic Function and Abnormal Behaviour in Shank3 Exon4-9 Deletion Mouse Model of Autism. Autism Res 2015; 9:350-75. [PMID: 26559786 DOI: 10.1002/aur.1529] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/07/2015] [Accepted: 07/21/2015] [Indexed: 12/17/2022]
Abstract
Shank3 is a multi-domain, synaptic scaffolding protein that organizes proteins in the postsynaptic density of excitatory synapses. Clinical studies suggest that ∼ 0.5% of autism spectrum disorder (ASD) cases may involve SHANK3 mutation/deletion. Patients with SHANK3 mutations exhibit deficits in cognition along with delayed/impaired speech/language and repetitive and obsessive/compulsive-like (OCD-like) behaviors. To examine how mutation/deletion of SHANK3 might alter brain function leading to ASD, we have independently created mice with deletion of Shank3 exons 4-9, a region implicated in ASD patients. We find that homozygous deletion of exons 4-9 (Shank3(e4-9) KO) results in loss of the two highest molecular weight isoforms of Shank3 and a significant reduction in other isoforms. Behaviorally, both Shank3(e4-9) heterozygous (HET) and Shank3(e4-9) KO mice display increased repetitive grooming, deficits in novel and spatial object recognition learning and memory, and abnormal ultrasonic vocalizations. Shank3(e4-9) KO mice also display abnormal social interaction when paired with one another. Analysis of synaptosome fractions from striata of Shank3(e4-9) KO mice reveals decreased Homer1b/c, GluA2, and GluA3 expression. Both Shank3(e4-9) HET and KO demonstrated a significant reduction in NMDA/AMPA ratio at excitatory synapses onto striatal medium spiny neurons. Furthermore, Shank3(e4-9) KO mice displayed reduced hippocampal LTP despite normal baseline synaptic transmission. Collectively these behavioral, biochemical and physiological changes suggest Shank3 isoforms have region-specific roles in regulation of AMPAR subunit localization and NMDAR function in the Shank3(e4-9) mutant mouse model of autism.
Collapse
Affiliation(s)
- Thomas C Jaramillo
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Haley E Speed
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhong Xuan
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jeremy M Reimers
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shunan Liu
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Craig M Powell
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Psychiatry and Neuroscience Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
173
|
Maiti P, Manna J, Ilavazhagan G, Rossignol J, Dunbar GL. Molecular regulation of dendritic spine dynamics and their potential impact on synaptic plasticity and neurological diseases. Neurosci Biobehav Rev 2015; 59:208-37. [PMID: 26562682 DOI: 10.1016/j.neubiorev.2015.09.020] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/20/2015] [Accepted: 09/07/2015] [Indexed: 12/12/2022]
Abstract
The structure and dynamics of dendritic spines reflect the strength of synapses, which are severely affected in different brain diseases. Therefore, understanding the ultra-structure, molecular signaling mechanism(s) regulating dendritic spine dynamics is crucial. Although, since last century, dynamics of spine have been explored by several investigators in different neurological diseases, but despite countless efforts, a comprehensive understanding of the fundamental etiology and molecular signaling pathways involved in spine pathology is lacking. The purpose of this review is to provide a contextual framework of our current understanding of the molecular mechanisms of dendritic spine signaling, as well as their potential impact on different neurodegenerative and psychiatric diseases, as a format for highlighting some commonalities in function, as well as providing a format for new insights and perspectives into this critical area of research. Additionally, the potential strategies to restore spine structure-function in different diseases are also pointed out. Overall, these informations should help researchers to design new drugs to restore the structure-function of dendritic spine, a "hot site" of synaptic plasticity.
Collapse
Affiliation(s)
- Panchanan Maiti
- Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, USA; Department of Psychology and Neurosciences Program, Central Michigan University, Mt. Pleasant, MI, USA.
| | - Jayeeta Manna
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - G Ilavazhagan
- Hindustan University, Rajiv Gandhi Salai (OMR), Padur, Kelambakam, Chennai, TN, India.
| | - Julien Rossignol
- Department of Psychology and Neurosciences Program, Central Michigan University, Mt. Pleasant, MI, USA; College of Medicine, Central Michigan University, Mt. Pleasant, MI, USA.
| | - Gary L Dunbar
- Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, USA; Department of Psychology and Neurosciences Program, Central Michigan University, Mt. Pleasant, MI, USA.
| |
Collapse
|
174
|
Hatanaka Y, Watase K, Wada K, Nagai Y. Abnormalities in synaptic dynamics during development in a mouse model of spinocerebellar ataxia type 1. Sci Rep 2015; 5:16102. [PMID: 26531852 PMCID: PMC4632040 DOI: 10.1038/srep16102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/07/2015] [Indexed: 11/18/2022] Open
Abstract
Late-onset neurodegenerative diseases are characterized by neurological symptoms and progressive neuronal death. Accumulating evidence suggests that neuronal dysfunction, rather than neuronal death, causes the symptoms of neurodegenerative diseases. However, the mechanisms underlying the dysfunction that occurs prior to cell death remain unclear. To investigate the synaptic basis of this dysfunction, we employed in vivo two-photon imaging to analyse excitatory postsynaptic dendritic protrusions. We used Sca1154Q/2Q mice, an established knock-in mouse model of the polyglutamine disease spinocerebellar ataxia type 1 (SCA1), which replicates human SCA1 features including ataxia, cognitive impairment, and neuronal death. We found that Sca1154Q/2Q mice exhibited greater synaptic instability than controls, without synaptic loss, in the cerebral cortex, where obvious neuronal death is not observed, even before the onset of distinct symptoms. Interestingly, this abnormal synaptic instability was evident in Sca1154Q/2Q mice from the synaptic developmental stage, and persisted into adulthood. Expression of synaptic scaffolding proteins was also lower in Sca1154Q/2Q mice than controls before synaptic maturation. As symptoms progressed, synaptic loss became evident. These results indicate that aberrant synaptic instability, accompanied by decreased expression of scaffolding proteins during synaptic development, is a very early pathology that precedes distinct neurological symptoms and neuronal cell death in SCA1.
Collapse
Affiliation(s)
- Yusuke Hatanaka
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan.,CREST, JST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Kei Watase
- Center for Brain Integration Research, Tokyo Medical &Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8510, Japan
| | - Keiji Wada
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan.,CREST, JST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Yoshitaka Nagai
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan.,CREST, JST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| |
Collapse
|
175
|
Zhang Y, Gaetano CM, Williams KR, Bassell GJ, Mihailescu MR. FMRP interacts with G-quadruplex structures in the 3'-UTR of its dendritic target Shank1 mRNA. RNA Biol 2015; 11:1364-74. [PMID: 25692235 DOI: 10.1080/15476286.2014.996464] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Fragile X syndrome (FXS), the most common cause of inherited intellectual disability, is caused by the loss of expression of the fragile X mental retardation protein (FMRP). FMRP, which regulates the transport and translation of specific mRNAs, uses its RGG box domain to bind mRNA targets that form G-quadruplex structures. One of the FMRP in vivo targets, Shank1 mRNA, encodes the master scaffold proteins of the postsynaptic density (PSD) which regulate the size and shape of dendritic spines because of their capacity to interact with many different PSD components. Due to their effect on spine morphology, altered translational regulation of Shank1 transcripts may contribute to the FXS pathology. We hypothesized that the FMRP interactions with Shank1 mRNA are mediated by the recognition of the G quadruplex structure, which has not been previously demonstrated. In this study we used biophysical techniques to analyze the Shank1 mRNA 3'-UTR and its interactions with FMRP and its phosphorylated mimic FMRP S500D. We found that the Shank1 mRNA 3 ' -UTR adopts two very stable intramolecular G-quadruplexes which are bound specifically and with high affinity by FMRP both in vitro and in vivo. These results suggest a role of G-quadruplex RNA motif as a structural element in the common mechanism of FMRP regulation of its dendritic mRNA targets.
Collapse
Affiliation(s)
- Yang Zhang
- a Graduate School of Pharmaceutical Sciences; Mylan School of Pharmacy ; Duquesne University ; Pittsburgh , PA USA
| | | | | | | | | |
Collapse
|
176
|
Rozeboom AM, Queenan BN, Partridge JG, Farnham C, Wu JY, Vicini S, Pak DTS. Evidence for glycinergic GluN1/GluN3 NMDA receptors in hippocampal metaplasticity. Neurobiol Learn Mem 2015; 125:265-73. [PMID: 26477834 DOI: 10.1016/j.nlm.2015.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/25/2015] [Accepted: 10/07/2015] [Indexed: 10/22/2022]
Abstract
Hebbian, or associative, forms of synaptic plasticity are considered the molecular basis of learning and memory. However, associative synaptic modifications, including long-term potentiation (LTP) and depression (LTD), can form positive feedback loops which must be constrained for neural networks to remain stable. One proposed constraint mechanism is metaplasticity, a process whereby synaptic changes shift the threshold for subsequent plasticity. Metaplasticity has been functionally observed but the molecular basis is not well understood. Here, we report that stimulation which induces LTP recruits GluN2B-lacking GluN1/GluN3 NMDA receptors (NMDARs) to excitatory synapses of hippocampal pyramidal neurons. These unconventional receptors may compete against conventional GluN1/GluN2 NMDARs to favor synaptic depotentiation in response to subsequent "LTP-inducing" stimulation. These results implicate glycinergic GluN1/GluN3 NMDAR as molecular brakes on excessive synaptic strengthening, suggesting a role for these receptors in the brain that has previously been elusive.
Collapse
Affiliation(s)
- Aaron M Rozeboom
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | - Bridget N Queenan
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057-1464, USA; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057-1464, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - John G Partridge
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | - Christina Farnham
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | - Jian-Young Wu
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057-1464, USA; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | - Stefano Vicini
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057-1464, USA; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | - Daniel T S Pak
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057-1464, USA; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057-1464, USA.
| |
Collapse
|
177
|
Smothers CT, Szumlinski KK, Worley PF, Woodward JJ. Altered NMDA receptor function in primary cultures of hippocampal neurons from mice lacking the Homer2 gene. Synapse 2015; 70:33-9. [PMID: 26426435 DOI: 10.1002/syn.21869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/24/2015] [Accepted: 08/28/2015] [Indexed: 12/17/2022]
Abstract
N-Methyl-D-Aspartate (NMDA) receptors are inhibited during acute exposure to ethanol and are involved in changes in neuronal plasticity following repeated ethanol exposure. The postsynaptic scaffolding protein Homer2 can regulate the cell surface expression of NMDA receptors in vivo, and mice with a null mutation of the Homer2 gene exhibit an alcohol-avoiding and -intolerant phenotype that is accompanied by a lack of ethanol-induced glutamate sensitization. Thus, Homer2 deletion may perturb the function or acute ethanol sensitivity of the NMDA receptor. In this study, the function and ethanol sensitivity of glutamate receptors in cultured hippocampal neurons from wild-type (WT) and Homer2 knock-out (KO) mice were examined at 7 and 14 days in vitro (DIV) using standard whole-cell voltage-clamp electrophysiology. As compared with wild-type controls, NMDA receptor current density was reduced in cultured hippocampal neurons from Homer2 KO mice at 14 DIV, but not at 7 DIV. There were no genotype-dependent changes in whole-cell capacitance or in currents evoked by kainic acid. The GluN2B-selective antagonist ifenprodil inhibited NMDA-evoked currents to a similar extent in both wild-type and Homer2 KO neurons and inhibition was greater at 7 versus 14 DIV. NMDA receptor currents from both WT and KO mice were inhibited by ethanol (10-100 mM) and the degree of inhibition did not differ as a function of genotype. In conclusion, NMDA receptor function, but not ethanol sensitivity, is reduced in hippocampal neurons lacking the Homer2 gene.
Collapse
Affiliation(s)
- C Thetford Smothers
- Department of Neuroscience and Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, MSC 861, Charleston, South Carolina, 29425
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute, University of California, Santa Barbara, California, 93106-9660
| | - Paul F Worley
- Departments of Neuroscience & Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - John J Woodward
- Departments of Neuroscience & Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| |
Collapse
|
178
|
de Bartolomeis A, Errico F, Aceto G, Tomasetti C, Usiello A, Iasevoli F. D-aspartate dysregulation in Ddo(-/-) mice modulates phencyclidine-induced gene expression changes of postsynaptic density molecules in cortex and striatum. Prog Neuropsychopharmacol Biol Psychiatry 2015; 62:35-43. [PMID: 25979765 DOI: 10.1016/j.pnpbp.2015.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
Abstract
N-methyl-D-aspartate receptor (NMDAR) hypofunction has been considered a key alteration in schizophrenia pathophysiology. Thus, several strategies aimed at enhancing glutamatergic transmission, included the introduction in therapy of D-amino acids, such as D-serine and D-cycloserine augmentation, have been proposed to counteract difficult-to-treat symptoms or treatment-resistant forms of schizophrenia. Another D-amino acid, D-aspartate, has recently gained increasing interest for its role in NMDAR activation and has been found reduced in post-mortem cortex of schizophrenia patients. NMDAR is the core of the postsynaptic density (PSD), a postsynaptic site involved in glutamate signaling and responsive to antipsychotic treatment. In this study, we investigated striatal and cortical gene expression of key PSD transcripts (i.e. Homer1a, Homer1b/c, and PSD-95) in mice with persistently elevated brain D-aspartate-levels, i.e. the D-aspartate-oxidase knockout mice (Ddo(-/-)). These animal models were analyzed both in naive condition and after phencyclidine (PCP) treatment. Naive Ddo(-/-) mice showed decreased Homer1a expression in the prefrontal cortex, increased Homer1b/c expression in the striatum, and decreased PSD-95 expression in the striatum and in the cortex. Acute PCP treatment restored, and even potentiated, Homer1a expression in the prefrontal cortex of mutant mice, while it had limited effects on the other genes. These results suggest that persistently elevated D-aspartate, by enhancing NMDA transmission, may cause complex adaptive mechanisms affecting Homer1a, which in turn may explain the recently demonstrated protective effects of this D-amino acid against PCP-induced behavioral alterations, such as ataxic behavior.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy.
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples (SUN), Caserta, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy
| |
Collapse
|
179
|
Du Y, Ge MM, Xue W, Yang QQ, Wang S, Xu Y, Wang HL. Chronic Lead Exposure and Mixed Factors of Gender×Age×Brain Regions Interactions on Dendrite Growth, Spine Maturity and NDR Kinase. PLoS One 2015; 10:e0138112. [PMID: 26368815 PMCID: PMC4569283 DOI: 10.1371/journal.pone.0138112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 08/26/2015] [Indexed: 02/01/2023] Open
Abstract
NDR1/2 kinase is essential in dendrite morphology and spine formation, which is regulated by cellular Ca2+. Lead (Pb) is a potent blocker of L-type calcium channel and our recent work showed Pb exposure impairs dendritic spine outgrowth in hippocampal neurons in rats. But the sensitivity of Pb-induced spine maturity with mixed factors (gender×age×brain regions) remains unknown. This study aimed to systematically investigate the effect of Pb exposure on spine maturity in rat brain with three factors (gender×age×brain regions), as well as the NDR1/2 kinase expression. Sprague–Dawley rats were exposed to Pb from parturition to postnatal day 30, 60, 90, respectively. Golgi-Cox staining was used to examine spine maturity. Western blot assay was applied to measure protein expression and real-time fluorescence quantitative PCR assay was used to examine mRNA levels. The results showed chronic Pb exposure significantly decreased dendritic length and impaired spine maturity in both rat hippocampus and medial prefrontal cortex. The impairment of dendritic length induced by Pb exposure tended to adolescence > adulthood, hippocampus > medial prefrontal cortex and female > male. Pb exposure induced significant damage in spine maturity during adolescence and early adult while little damage during adult in male rat brain and female medial prefrontal cortex. Besides, there was sustained impairment from adolescence to adulthood in female hippocampus. Interestingly, impairment of spine maturity followed by Pb exposure was correlated with NDR1/2 kinase. The reduction of NDR1/2 kinase protein expression after Pb exposure was similar to the result of spine maturity. In addition, NDR2 and their substrate Rabin3 mRNA levels were significantly decreased by Pb exposure in developmental rat brain. Taken together, Pb exposure impaired dendrite growth and maturity which was subject to gender×age×brain regions effects and related to NDR1/2 signal expression.
Collapse
Affiliation(s)
- Yang Du
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Meng-Meng Ge
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Weizhen Xue
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Qian-Qian Yang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Shuang Wang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Yi Xu
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
| | - Hui-Li Wang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei, Anhui 230009, P. R. China
- * E-mail:
| |
Collapse
|
180
|
PKA Inhibitor H89 (N-[2-p-bromocinnamylamino-ethyl]-5-isoquinolinesulfonamide) Attenuates Synaptic Dysfunction and Neuronal Cell Death following Ischemic Injury. Neural Plast 2015; 2015:374520. [PMID: 26448879 PMCID: PMC4584069 DOI: 10.1155/2015/374520] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/27/2015] [Accepted: 03/17/2015] [Indexed: 11/26/2022] Open
Abstract
The cyclic AMP-dependent protein kinase (PKA), which activates prosurvival signaling proteins, has been implicated in the expression of long-term potentiation and hippocampal long-term memory. It has come to light that H89 commonly known as the PKA inhibitor have diverse roles in the nervous system that are unrelated to its role as a PKA inhibitor. We have investigated the role of H89 in ischemic and reperfusion injury. First, we examined the expression of postsynaptic density protein 95 (PSD95), microtubule-associated protein 2 (MAP2), and synaptophysin in mouse brain after middle cerebral artery occlusion injury. Next, we examined the role of H89 pretreatment on the expression of brain-derived neurotrophic factor (BDNF), PSD95, MAP2, and the apoptosis regulators Bcl2 and cleaved caspase-3 in cultured neuroblastoma cells exposed to hypoxia and reperfusion injury. In addition, we investigated the alteration of AKT activation in H89 pretreated neuroblastoma cells under hypoxia and reperfusion injury. The data suggest that H89 may contribute to brain recovery after ischemic stroke by regulating neuronal death and proteins related to synaptic plasticity.
Collapse
|
181
|
Sala C, Vicidomini C, Bigi I, Mossa A, Verpelli C. Shank synaptic scaffold proteins: keys to understanding the pathogenesis of autism and other synaptic disorders. J Neurochem 2015; 135:849-58. [PMID: 26338675 DOI: 10.1111/jnc.13232] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 02/01/2023]
Abstract
Shank/ProSAP proteins are essential to synaptic formation, development, and function. Mutations in the family of SHANK genes are strongly associated with autism spectrum disorders (ASD) and other neurodevelopmental and neuropsychiatric disorders, such as intellectual disability (ID), and schizophrenia. Thus, the term 'Shankopathies' identifies a number of neuronal diseases caused by alteration of Shank protein expression leading to abnormal synaptic development. With this review we want to summarize the major genetic, molecular, behavior and electrophysiological studies that provide new clues into the function of Shanks and pave the way for the discovery of new therapeutic drugs targeted to treat patients with SHANK mutations and also patients affected by other neurodevelopmental and neuropsychiatric disorders. Shank/ProSAP proteins are essential to synaptic formation, development, and function. Mutations in the family of SHANK genes are strongly associated with autism spectrum disorders (ASD) and other neurodevelopmental and neuropsychiatric disorders, such as intellectual disability (ID), and schizophrenia (SCZ). With this review we want to summarize the major genetic, molecular, behavior and electrophysiological studies that provide new clues into the function of Shanks and pave the way for the discovery of new therapeutic drugs targeted to treat patients with SHANK mutations.
Collapse
Affiliation(s)
- Carlo Sala
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Cinzia Vicidomini
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Ilaria Bigi
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Adele Mossa
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
182
|
Kang J, Park H, Kim E. IRSp53/BAIAP2 in dendritic spine development, NMDA receptor regulation, and psychiatric disorders. Neuropharmacology 2015; 100:27-39. [PMID: 26275848 DOI: 10.1016/j.neuropharm.2015.06.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/26/2015] [Accepted: 06/28/2015] [Indexed: 01/08/2023]
Abstract
IRSp53 (also known as BAIAP2) is a multi-domain scaffolding and adaptor protein that has been implicated in the regulation of membrane and actin dynamics at subcellular structures, including filopodia and lamellipodia. Accumulating evidence indicates that IRSp53 is an abundant component of the postsynaptic density at excitatory synapses and an important regulator of actin-rich dendritic spines. In addition, IRSp53 has been implicated in diverse psychiatric disorders, including autism spectrum disorders, schizophrenia, and attention deficit/hyperactivity disorder. Mice lacking IRSp53 display enhanced NMDA (N-methyl-d-aspartate) receptor function accompanied by social and cognitive deficits, which are reversed by pharmacological suppression of NMDA receptor function. These results suggest the hypothesis that defective actin/membrane modulation in IRSp53-deficient dendritic spines may lead to social and cognitive deficits through NMDA receptor dysfunction. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
Affiliation(s)
- Jaeseung Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea
| | - Haram Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 305-701, South Korea.
| |
Collapse
|
183
|
Abstract
Phelan-McDermid syndrome (PMS), also called 22q13.3 deletion syndrome, is a neurodevelopmental disorder characterized by global developmental delay, intellectual disability, severe speech delays, poor motor tone and function, and autism spectrum disorder (ASD). Although the overall prevalence of PMS is unknown, there have been at least 1200 cases reported worldwide, according to the Phelan-McDermid Syndrome Foundation. PMS is now considered to be a relatively common cause of ASD and intellectual disability, accounting for between 0.5% and 2.0% of cases. The cause of PMS has been isolated to loss of function of one copy of SHANK3, which codes for a master scaffolding protein found in the postsynaptic density of excitatory synapses. Reduced expression of SH3 and multiple ankyrin repeat domains 3 (SHANK3) leads to reduced numbers of dendrites, and impaired synaptic transmission and plasticity. Recent mouse and human neuronal models of PMS have led to important opportunities to develop novel therapeutics, and at least 2 clinical trials are underway, one in the USA, and one in the Netherlands. The SHANK3 pathway may also be relevant to other forms of ASD, and many of the single-gene causes of ASD identified to date appear to converge on several common molecular pathways that underlie synaptic neurotransmission. As a result, treatments developed for PMS may also affect other forms of ASD.
Collapse
Affiliation(s)
- Jesse L. Costales
- />Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alexander Kolevzon
- />Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
- />Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA
- />Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY USA
- />Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- />Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
184
|
Ultrastructural analyses in the hippocampus CA1 field in Shank3-deficient mice. Mol Autism 2015; 6:41. [PMID: 26137200 PMCID: PMC4486760 DOI: 10.1186/s13229-015-0036-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 06/22/2015] [Indexed: 11/10/2022] Open
Abstract
Background The genetics of autism spectrum disorder (hereafter referred to as “autism”) are rapidly unfolding, with a significant increase in the identification of genes implicated in the disorder. Many of these genes are part of a complex landscape of genetic variants that are thought to act together to cause the behavioral phenotype associated with autism. One of the few single-locus causes of autism involves a mutation in the SH3 and multiple ankyrin repeat domains 3 (SHANK3) gene. Previous electrophysiological studies in mice with Shank3 mutations demonstrated impairment in synaptic long-term potentiation, suggesting a potential disruption at the synapse. Methods To understand how variants in SHANK3 would lead to such impairments and manifest in the brain of patients with autism, we assessed the presence of synaptic pathology in Shank3-deficient mice at 5 weeks and 3 months of age, focusing on the stratum radiatum of the CA1 field. This study analyzed both Shank3 heterozygous and homozygous mice using an electron microscopy approach to determine whether there is a morphological correlate to the synaptic functional impairment. Results As both synaptic strength and plasticity are affected in Shank3-deficient mice, we hypothesized that there would be a reduction in synapse density, postsynaptic density length, and perforated synapse density. No differences were found in most parameters assessed. However, Shank3 heterozygotes had significantly higher numbers of perforated synapses at 5 weeks compared to 3 months of age and significantly higher numbers of perforated synapses compared to 5-week-old wildtype and Shank3 homozygous mice. Conclusions Although this finding represents preliminary evidence for ultrastructural alterations, it suggests that while major structural changes seem to be compensated for in Shank3-deficient mice, more subtle morphological alterations, affecting synaptic structure, may take place in an age-dependent manner.
Collapse
|
185
|
Duffney LJ, Zhong P, Wei J, Matas E, Cheng J, Qin L, Ma K, Dietz DM, Kajiwara Y, Buxbaum JD, Yan Z. Autism-like Deficits in Shank3-Deficient Mice Are Rescued by Targeting Actin Regulators. Cell Rep 2015; 11:1400-1413. [PMID: 26027926 DOI: 10.1016/j.celrep.2015.04.064] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 03/05/2015] [Accepted: 04/30/2015] [Indexed: 02/07/2023] Open
Abstract
Haploinsufficiency of the Shank3 gene, which encodes a scaffolding protein at glutamatergic synapses, is a highly prevalent and penetrant risk factor for autism. Using combined behavioral, electrophysiological, biochemical, imaging, and molecular approaches, we find that Shank3-deficient mice exhibit autism-like social deficits and repetitive behaviors, as well as the significantly diminished NMDA receptor (NMDAR) synaptic function and synaptic distribution in prefrontal cortex. Concomitantly, Shank3-deficient mice have a marked loss of cortical actin filaments, which is associated with the reduced Rac1/PAK activity and increased activity of cofilin, the major actin depolymerizing factor. The social deficits and NMDAR hypofunction are rescued by inhibiting cofilin or activating Rac1 in Shank3-deficient mice and are induced by inhibiting PAK or Rac1 in wild-type mice. These results indicate that the aberrant regulation of synaptic actin filaments and loss of synaptic NMDARs contribute to the manifestation of autism-like phenotypes. Thus, targeting actin regulators provides a strategy for autism treatment.
Collapse
Affiliation(s)
- Lara J Duffney
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| | - Ping Zhong
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| | - Jing Wei
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| | - Emmanuel Matas
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| | - Jia Cheng
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| | - Luye Qin
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| | - Kaijie Ma
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| | - David M Dietz
- Department of Pharmacology & Toxicology, State University of New York at Buffalo, Buffalo, NY, 14214
| | - Yuji Kajiwara
- Seaver Autism Center for Research and Treatment, Department of Psychiatry, Friedman Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Department of Psychiatry, Friedman Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Zhen Yan
- Department of Physiology & Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214
| |
Collapse
|
186
|
Wu J, Pipathsouk A, Keizer-Gunnink A, Fusetti F, Alkema W, Liu S, Altschuler S, Wu L, Kortholt A, Weiner OD. Homer3 regulates the establishment of neutrophil polarity. Mol Biol Cell 2015; 26:1629-39. [PMID: 25739453 PMCID: PMC4436775 DOI: 10.1091/mbc.e14-07-1197] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 01/10/2023] Open
Abstract
Most chemoattractants rely on activation of the heterotrimeric G-protein Gαi to regulate directional cell migration, but few links from Gαi to chemotactic effectors are known. Through affinity chromatography using primary neutrophil lysate, we identify Homer3 as a novel Gαi2-binding protein. RNA interference-mediated knockdown of Homer3 in neutrophil-like HL-60 cells impairs chemotaxis and the establishment of polarity of phosphatidylinositol 3,4,5-triphosphate (PIP3) and the actin cytoskeleton, as well as the persistence of the WAVE2 complex. Most previously characterized proteins that are required for cell polarity are needed for actin assembly or activation of core chemotactic effectors such as the Rac GTPase. In contrast, Homer3-knockdown cells show normal magnitude and kinetics of chemoattractant-induced activation of phosphoinositide 3-kinase and Rac effectors. Chemoattractant-stimulated Homer3-knockdown cells also exhibit a normal initial magnitude of actin polymerization but fail to polarize actin assembly and intracellular PIP3 and are defective in the initiation of cell polarity and motility. Our data suggest that Homer3 acts as a scaffold that spatially organizes actin assembly to support neutrophil polarity and motility downstream of GPCR activation.
Collapse
Affiliation(s)
- Julie Wu
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158
| | - Anne Pipathsouk
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158
| | - A Keizer-Gunnink
- Department of Cell Biochemistry, Groningen Biological Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - F Fusetti
- Department of Biochemistry and Netherlands Proteomics Centre, Groningen Biological Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - W Alkema
- NIZO Food Research, 6718 ZB Ede, Netherlands Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, 6525 GA Nijmegen, Netherlands
| | - Shanshan Liu
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Steven Altschuler
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lani Wu
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Arjan Kortholt
- Department of Cell Biochemistry, Groningen Biological Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Orion D Weiner
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
187
|
miR-26a and miR-384-5p are required for LTP maintenance and spine enlargement. Nat Commun 2015; 6:6789. [PMID: 25858512 PMCID: PMC4403380 DOI: 10.1038/ncomms7789] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/27/2015] [Indexed: 02/06/2023] Open
Abstract
Long-term potentiation (LTP) is a form of synaptic plasticity that results in enhanced synaptic strength. It is associated with the formation and enlargement of dendritic spines—tiny protrusions accommodating excitatory synapses. Both LTP and spine remodelling are crucial for brain development, cognition and the pathophysiology of neurological disorders. The role of microRNAs (miRNAs) in the maintenance of LTP, however, is not well understood. Using next-generation sequencing to profile miRNA transcriptomes, we demonstrate that miR-26a and miR-384-5p specifically affect the maintenance, but not induction, of LTP and different stages of spine enlargement by regulating the expression of RSK3. Using bioinformatics, we also examine the global effects of miRNA transcriptome changes during LTP on gene expression and cellular activities. This study reveals a novel miRNA-mediated mechanism for gene-specific regulation of translation in LTP, identifies two miRNAs required for long-lasting synaptic and spine plasticity and presents a catalogue of candidate ‘LTP miRNAs'. Long-term potentiation (LTP) is a form of synaptic plasticity that results in enhanced synaptic strength. Here, the authors demonstrate that miR-26a and miR-384-5p affect the maintenance, but not induction, of LTP as well as spine enlargement by regulating the expression of RSK3.
Collapse
|
188
|
Abstract
Accumulating data, including those from large genetic association studies, indicate that alterations in glutamatergic synapse structure and function represent a common underlying pathology in many symptomatically distinct cognitive disorders. In this review, we discuss evidence from human genetic studies and data from animal models supporting a role for aberrant glutamatergic synapse function in the etiology of intellectual disability (ID), autism spectrum disorder (ASD), and schizophrenia (SCZ), neurodevelopmental disorders that comprise a significant proportion of human cognitive disease and exact a substantial financial and social burden. The varied manifestations of impaired perceptual processing, executive function, social interaction, communication, and/or intellectual ability in ID, ASD, and SCZ appear to emerge from altered neural microstructure, function, and/or wiring rather than gross changes in neuron number or morphology. Here, we review evidence that these disorders may share a common underlying neuropathy: altered excitatory synapse function. We focus on the most promising candidate genes affecting glutamatergic synapse function, highlighting the likely disease-relevant functional consequences of each. We first present a brief overview of glutamatergic synapses and then explore the genetic and phenotypic evidence for altered glutamate signaling in ID, ASD, and SCZ.
Collapse
Affiliation(s)
- Lenora Volk
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | | | | | | |
Collapse
|
189
|
Mao W, Watanabe T, Cho S, Frost JL, Truong T, Zhao X, Futai K. Shank1 regulates excitatory synaptic transmission in mouse hippocampal parvalbumin-expressing inhibitory interneurons. Eur J Neurosci 2015; 41:1025-35. [PMID: 25816842 DOI: 10.1111/ejn.12877] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/30/2015] [Accepted: 02/18/2015] [Indexed: 01/25/2023]
Abstract
The Shank genes (SHANK1, 2, 3) encode scaffold proteins highly enriched in postsynaptic densities where they regulate synaptic structure in spiny neurons. Mutations in human Shank genes are linked to autism spectrum disorder and schizophrenia. Shank1 mutant mice exhibit intriguing cognitive phenotypes reminiscent of individuals with autism spectrum disorder. However, the molecular mechanisms leading to the human pathophysiological phenotypes and mouse behaviors have not been elucidated. In this study it is shown that Shank1 protein is highly localized in parvalbumin-expressing (PV+) fast-spiking inhibitory interneurons in the hippocampus. Importantly, a lack of Shank1 in hippocampal CA1 PV+ neurons reduced excitatory synaptic inputs and inhibitory synaptic outputs to pyramidal neurons. Furthermore, it is demonstrated that hippocampal CA1 pyramidal neurons in Shank1 mutant mice exhibit a shift in the excitatory and inhibitory balance (E-I balance), a pathophysiological hallmark of autism spectrum disorder. The mutant mice also exhibit lower expression of gephyrin (a scaffold component of inhibitory synapses), supporting the dysregulation of E-I balance in the hippocampus. These results suggest that Shank1 scaffold in PV+ interneurons regulates excitatory synaptic strength and participates in the maintenance of E-I balance in excitatory neurons.
Collapse
Affiliation(s)
- Wenjie Mao
- Brudnick Neuropsychiatric Research Institute and Program in Neuroscience, University of Massachusetts Medical School, 303 Belmont Street, Worcester, MA, 01604, USA
| | | | | | | | | | | | | |
Collapse
|
190
|
Tao-Cheng JH, Yang Y, Reese TS, Dosemeci A. Differential distribution of Shank and GKAP at the postsynaptic density. PLoS One 2015; 10:e0118750. [PMID: 25775468 PMCID: PMC4361712 DOI: 10.1371/journal.pone.0118750] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/06/2015] [Indexed: 11/18/2022] Open
Abstract
Shank and GKAP are scaffold proteins and binding partners at the postsynaptic density (PSD). The distribution and dynamics of Shank and GKAP were studied in dissociated hippocampal cultures by pre-embedding immunogold electron microscopy. Antibodies against epitopes containing their respective mutual binding sites were used to verify the expected juxtapositioning of Shank and GKAP. If all Shank and GKAP molecules at the PSD were bound to each other, the distribution of label for the two proteins should coincide. However, labels for the mutual binding sites showed significant differences in distribution, with a narrow distribution for GKAP located close to the postsynaptic membrane, and a wider distribution for Shank extending deeper into the cytoplasm. Upon depolarization with high K+, neither the intensity nor distribution of label for GKAP changed, but labeling intensity for Shank at the PSD increased to ~150% of controls while the median distance of label from postsynaptic membrane increased by 7.5 nm. These results indicate a preferential recruitment of Shank to more distal parts of the PSD complex. Conversely, upon incubation in Ca2+-free medium containing EGTA, the labeling intensity of Shank at the PSD decreased to ~70% of controls and the median distance of label from postsynaptic membrane decreased by 9 nm, indicating a preferential loss of Shank molecules in more distal parts of the PSD complex. These observations identify two pools of Shank at the PSD complex, one relatively stable pool, closer to the postsynaptic membrane that can bind to GKAP, and another more dynamic pool at a location too far away to bind to GKAP.
Collapse
Affiliation(s)
- Jung-Hwa Tao-Cheng
- EM Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Yijung Yang
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas S. Reese
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ayse Dosemeci
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
191
|
Yang Y, Wei M, Xiong Y, Du X, Zhu S, Yang L, Zhang C, Liu JJ. Endophilin A1 regulates dendritic spine morphogenesis and stability through interaction with p140Cap. Cell Res 2015; 25:496-516. [PMID: 25771685 DOI: 10.1038/cr.2015.31] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 11/24/2014] [Accepted: 12/12/2014] [Indexed: 02/07/2023] Open
Abstract
Dendritic spines are actin-rich membrane protrusions that are the major sites of excitatory synaptic input in the mammalian brain, and their morphological plasticity provides structural basis for learning and memory. Here we report that endophilin A1, with a well-established role in clathrin-mediated synaptic vesicle endocytosis at the presynaptic terminal, also localizes to dendritic spines and is required for spine morphogenesis, synapse formation and synaptic function. We identify p140Cap, a regulator of cytoskeleton reorganization, as a downstream effector of endophilin A1 and demonstrate that disruption of their interaction impairs spine formation and maturation. Moreover, we demonstrate that knockdown of endophilin A1 or p140Cap impairs spine stabilization and synaptic function. We further show that endophilin A1 regulates the distribution of p140Cap and its downstream effector, the F-actin-binding protein cortactin as well as F-actin enrichment in dendritic spines. Together, these results reveal a novel function of postsynaptic endophilin A1 in spine morphogenesis, stabilization and synaptic function through the regulation of p140Cap.
Collapse
Affiliation(s)
- Yanrui Yang
- 1] State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Beijing 100101, China [2] CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Mengping Wei
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Beijing 100871, China [2] PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Ying Xiong
- School of Life Sciences, University of Science and Technology of China, Hefei Anhui 230026, China
| | - Xiangyang Du
- 1] State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Beijing 100101, China [2] CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Beijing 100101, China [3] University of Chinese Academy of Sciences, Beijing 100039, China
| | - Shaoxia Zhu
- 1] State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Beijing 100101, China [2] CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Beijing 100101, China
| | - Chen Zhang
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Beijing 100871, China [2] PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jia-Jia Liu
- 1] State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Beijing 100101, China [2] CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
192
|
McGuier NS, Padula AE, Mulholland PJ, Chandler LJ. Homer2 deletion alters dendritic spine morphology but not alcohol-associated adaptations in GluN2B-containing N-methyl-D-aspartate receptors in the nucleus accumbens. Front Pharmacol 2015; 6:28. [PMID: 25755642 PMCID: PMC4337331 DOI: 10.3389/fphar.2015.00028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/03/2015] [Indexed: 12/01/2022] Open
Abstract
Repeated exposure to ethanol followed by withdrawal leads to alterations in glutamatergic signaling and impaired synaptic plasticity in the nucleus accumbens (NAc) in both clinical and preclinical models of ethanol exposure. Homer2 is a member of a family of postsynaptic density (PSD) scaffolding proteins that functions in part to cluster N-methyl-D-aspartate (NMDA) signaling complexes in the PSD, and has been shown to be critically important for plasticity in multiple models of drug and alcohol abuse. Here we used Homer2 knockout (KO) mice and a chronic intermittent intraperitoneal (IP) ethanol injection model to investigate a potential role for the protein in ethanol-induced adaptations in dendritic spine morphology and PSD protein expression. While deletion of Homer2 was associated with increased density of long spines on medium spiny neurons of the NAc core of saline treated mice, ethanol exposure had no effect on dendritic spine morphology in either wild-type (WT) or Homer2 KO mice. Western blot analysis of tissue samples from the NAc enriched for PSD proteins revealed a main effect of ethanol treatment on the expression of GluN2B, but there was no effect of genotype or treatment on the expression other glutamate receptor subunits or PSD95. These data indicate that the global deletion of Homer2 leads to aberrant regulation of dendritic spine morphology in the NAc core that is associated with an increased density of long, thin spines. Unexpectedly, intermittent IP ethanol did not affect spine morphology in either WT or KO mice. Together these data implicate Homer2 in the formation of long, thin spines and further supports its role in neuronal structure.
Collapse
Affiliation(s)
- Natalie S McGuier
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina , Charleston, SC, USA
| | - Audrey E Padula
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina , Charleston, SC, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina , Charleston, SC, USA
| | - L Judson Chandler
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina , Charleston, SC, USA
| |
Collapse
|
193
|
Fossati G, Morini R, Corradini I, Antonucci F, Trepte P, Edry E, Sharma V, Papale A, Pozzi D, Defilippi P, Meier JC, Brambilla R, Turco E, Rosenblum K, Wanker EE, Ziv NE, Menna E, Matteoli M. Reduced SNAP-25 increases PSD-95 mobility and impairs spine morphogenesis. Cell Death Differ 2015; 22:1425-36. [PMID: 25678324 DOI: 10.1038/cdd.2014.227] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 10/22/2014] [Accepted: 11/26/2014] [Indexed: 12/24/2022] Open
Abstract
Impairment of synaptic function can lead to neuropsychiatric disorders collectively referred to as synaptopathies. The SNARE protein SNAP-25 is implicated in several brain pathologies and, indeed, brain areas of psychiatric patients often display reduced SNAP-25 expression. It has been recently found that acute downregulation of SNAP-25 in brain slices impairs long-term potentiation; however, the processes through which this occurs are still poorly defined. We show that in vivo acute downregulation of SNAP-25 in CA1 hippocampal region affects spine number. Consistently, hippocampal neurons from SNAP-25 heterozygous mice show reduced densities of dendritic spines and defective PSD-95 dynamics. Finally, we show that, in brain, SNAP-25 is part of a molecular complex including PSD-95 and p140Cap, with p140Cap being capable to bind to both SNAP-25 and PSD-95. These data demonstrate an unexpected role of SNAP-25 in controlling PSD-95 clustering and open the possibility that genetic reductions of the protein levels - as occurring in schizophrenia - may contribute to the pathology through an effect on postsynaptic function and plasticity.
Collapse
Affiliation(s)
- G Fossati
- 1] Department of Biotechnology and Translational Medicine, University of Milan, Milano 20129, Italy [2] Humanitas Clinical and Research Center, Laboratory of Pharmacology and Brain Pathology, Via Manzoni 56, Rozzano, 20089 Milano, Italy
| | - R Morini
- 1] Department of Biotechnology and Translational Medicine, University of Milan, Milano 20129, Italy [2] Humanitas Clinical and Research Center, Laboratory of Pharmacology and Brain Pathology, Via Manzoni 56, Rozzano, 20089 Milano, Italy
| | - I Corradini
- 1] Department of Biotechnology and Translational Medicine, University of Milan, Milano 20129, Italy [2] Istituto di Neuroscienze del CNR, Milano 20129, Italy
| | - F Antonucci
- 1] Department of Biotechnology and Translational Medicine, University of Milan, Milano 20129, Italy [2] Istituto di Neuroscienze del CNR, Milano 20129, Italy
| | - P Trepte
- Neuroproteomics, Max Delbrueck Center for Molecular Medicine (MDC), Berlin 13125, Germany
| | - E Edry
- Sagol Department of Neurobiology, Center for Gene Manipulation in the Adult Brain (CGMB), Haifa University, Haifa, Israel
| | - V Sharma
- Sagol Department of Neurobiology, Center for Gene Manipulation in the Adult Brain (CGMB), Haifa University, Haifa, Israel
| | - A Papale
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute and University, Milano 20132, Italy
| | - D Pozzi
- Humanitas Clinical and Research Center, Laboratory of Pharmacology and Brain Pathology, Via Manzoni 56, Rozzano, 20089 Milano, Italy
| | - P Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10124, Italy
| | - J C Meier
- 1] RNA Editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany [2] TU Braunschweig, Zoological Institute, Division of Cell Biology and Cell Physiology, Braunschweig, Germany
| | - R Brambilla
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute and University, Milano 20132, Italy
| | - E Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10124, Italy
| | - K Rosenblum
- Sagol Department of Neurobiology, Center for Gene Manipulation in the Adult Brain (CGMB), Haifa University, Haifa, Israel
| | - E E Wanker
- Neuroproteomics, Max Delbrueck Center for Molecular Medicine (MDC), Berlin 13125, Germany
| | - N E Ziv
- Network Biology Labs and Faculty of Medicine, Technion, 33000 Haifa, Israel
| | - E Menna
- 1] Humanitas Clinical and Research Center, Laboratory of Pharmacology and Brain Pathology, Via Manzoni 56, Rozzano, 20089 Milano, Italy [2] Istituto di Neuroscienze del CNR, Milano 20129, Italy
| | - M Matteoli
- 1] Department of Biotechnology and Translational Medicine, University of Milan, Milano 20129, Italy [2] Humanitas Clinical and Research Center, Laboratory of Pharmacology and Brain Pathology, Via Manzoni 56, Rozzano, 20089 Milano, Italy
| |
Collapse
|
194
|
Braude JP, Vijayakumar S, Baumgarner K, Laurine R, Jones TA, Jones SM, Pyott SJ. Deletion of Shank1 has minimal effects on the molecular composition and function of glutamatergic afferent postsynapses in the mouse inner ear. Hear Res 2015; 321:52-64. [PMID: 25637745 DOI: 10.1016/j.heares.2015.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/24/2014] [Accepted: 01/13/2015] [Indexed: 01/06/2023]
Abstract
Shank proteins (1-3) are considered the master organizers of glutamatergic postsynaptic densities in the central nervous system, and the genetic deletion of either Shank1, 2, or 3 results in altered composition, form, and strength of glutamatergic postsynapses. To investigate the contribution of Shank proteins to glutamatergic afferent synapses of the inner ear and especially cochlea, we used immunofluorescence and quantitative real time PCR to determine the expression of Shank1, 2, and 3 in the cochlea. Because we found evidence for expression of Shank1 but not 2 and 3, we investigated the morphology, composition, and function of afferent postsynaptic densities from defined tonotopic regions in the cochlea of Shank1(-/-) mice. Using immunofluorescence, we identified subtle changes in the morphology and composition (but not number and localization) of cochlear afferent postsynaptic densities at the lower frequency region (8 kHz) in Shank1(-/-) mice compared to Shank1(+/+) littermates. However, we detected no differences in auditory brainstem responses at matching or higher frequencies. We also identified Shank1 in the vestibular afferent postsynaptic densities, but detected no differences in vestibular sensory evoked potentials in Shank1(-/-) mice compared to Shank1(+/+) littermates. This work suggests that Shank proteins play a different role in the development and maintenance of glutamatergic afferent synapses in the inner ear compared to the central nervous system.
Collapse
Affiliation(s)
- Jeremy P Braude
- University of North Carolina Wilmington, Department of Biology and Marine Biology, 601 South College Road, Wilmington, NC 28403, USA
| | - Sarath Vijayakumar
- University of Nebraska Lincoln, Department of Special Education and Communication Disorders, 304B Barkley Memorial Center, Lincoln, NE 68583-0738, USA
| | - Katherine Baumgarner
- University of North Carolina Wilmington, Department of Biology and Marine Biology, 601 South College Road, Wilmington, NC 28403, USA
| | - Rebecca Laurine
- University of North Carolina Wilmington, Department of Biology and Marine Biology, 601 South College Road, Wilmington, NC 28403, USA
| | - Timothy A Jones
- University of Nebraska Lincoln, Department of Special Education and Communication Disorders, 304B Barkley Memorial Center, Lincoln, NE 68583-0738, USA
| | - Sherri M Jones
- University of Nebraska Lincoln, Department of Special Education and Communication Disorders, 304B Barkley Memorial Center, Lincoln, NE 68583-0738, USA
| | - Sonja J Pyott
- University of North Carolina Wilmington, Department of Biology and Marine Biology, 601 South College Road, Wilmington, NC 28403, USA.
| |
Collapse
|
195
|
Vogl AM, Brockmann MM, Giusti SA, Maccarrone G, Vercelli CA, Bauder CA, Richter JS, Roselli F, Hafner AS, Dedic N, Wotjak CT, Vogt-Weisenhorn DM, Choquet D, Turck CW, Stein V, Deussing JM, Refojo D. Neddylation inhibition impairs spine development, destabilizes synapses and deteriorates cognition. Nat Neurosci 2015; 18:239-51. [PMID: 25581363 DOI: 10.1038/nn.3912] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 12/04/2014] [Indexed: 02/08/2023]
Abstract
Neddylation is a ubiquitylation-like pathway that controls cell cycle and proliferation by covalently conjugating Nedd8 to specific targets. However, its role in neurons, nonreplicating postmitotic cells, remains unexplored. Here we report that Nedd8 conjugation increased during postnatal brain development and is active in mature synapses, where many proteins are neddylated. We show that neddylation controls spine development during neuronal maturation and spine stability in mature neurons. We found that neddylated PSD-95 was present in spines and that neddylation on Lys202 of PSD-95 is required for the proactive role of the scaffolding protein in spine maturation and synaptic transmission. Finally, we developed Nae1(CamKIIα-CreERT2) mice, in which neddylation is conditionally ablated in adult excitatory forebrain neurons. These mice showed synaptic loss, impaired neurotransmission and severe cognitive deficits. In summary, our results establish neddylation as an active post-translational modification in the synapse regulating the maturation, stability and function of dendritic spines.
Collapse
Affiliation(s)
- Annette M Vogl
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Marisa M Brockmann
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Sebastian A Giusti
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Giuseppina Maccarrone
- Department for Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Claudia A Vercelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Corinna A Bauder
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Julia S Richter
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Francesco Roselli
- 1] Department of Neurobiology of Stress and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany. [2] Department of Neuroscience, Section of Neurology, University of Bari, Bari, Italy
| | - Anne-Sophie Hafner
- 1] University of Bordeaux, Interdisciplinary Institute for Neuroscience, Bordeaux, France. [2] CNRS UMR 5297, Bordeaux, France
| | - Nina Dedic
- 1] Department of Neurobiology of Stress and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany. [2] Institute of Developmental Genetics, Helmholtz-Zentrum München, Neuherberg, Germany
| | - Carsten T Wotjak
- Department of Neurobiology of Stress and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Daniel Choquet
- 1] University of Bordeaux, Interdisciplinary Institute for Neuroscience, Bordeaux, France. [2] CNRS UMR 5297, Bordeaux, France
| | - Christoph W Turck
- Department for Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Valentin Stein
- Institute of Physiology, University of Bonn, Bonn, Germany
| | - Jan M Deussing
- Department of Neurobiology of Stress and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Damian Refojo
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
196
|
Washbourne P. Synapse assembly and neurodevelopmental disorders. Neuropsychopharmacology 2015; 40:4-15. [PMID: 24990427 PMCID: PMC4262893 DOI: 10.1038/npp.2014.163] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/23/2014] [Accepted: 06/26/2014] [Indexed: 12/31/2022]
Abstract
In this review we examine the current understanding of how genetic deficits associated with neurodevelopmental disorders may impact synapse assembly. We then go on to discuss how the critical periods for these genetic deficits will shape the nature of future clinical interventions.
Collapse
Affiliation(s)
- Philip Washbourne
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA,Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA, Tel: +1 541 346 4138, Fax: +1 541 346 4548, E-mail:
| |
Collapse
|
197
|
Liu ZH, Yang Y, Ge MM, Xu L, Tang Y, Hu F, Xu Y, Wang HL. Bisphenol-A exposure alters memory consolidation and hippocampal CA1 spine formation through Wnt signaling in vivo and in vitro. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00093e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Based on Wnt signaling pathway, this study aims to further mechanistically understand memory alteration after BPA exposure.
Collapse
Affiliation(s)
- Zhi-Hua Liu
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| | - Ye Yang
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| | - Meng-Meng Ge
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| | - Li Xu
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| | - Yuqing Tang
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| | - Fan Hu
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| | - Yi Xu
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| | - Hui-Li Wang
- School of Biotechnology and Food Engineering
- Hefei University of Technology
- Hefei
- PR China
| |
Collapse
|
198
|
Ashhad S, Johnston D, Narayanan R. Activation of InsP₃ receptors is sufficient for inducing graded intrinsic plasticity in rat hippocampal pyramidal neurons. J Neurophysiol 2014; 113:2002-13. [PMID: 25552640 DOI: 10.1152/jn.00833.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/29/2014] [Indexed: 11/22/2022] Open
Abstract
The synaptic plasticity literature has focused on establishing necessity and sufficiency as two essential and distinct features in causally relating a signaling molecule to plasticity induction, an approach that has been surprisingly lacking in the intrinsic plasticity literature. In this study, we complemented the recently established necessity of inositol trisphosphate (InsP3) receptors (InsP3R) in a form of intrinsic plasticity by asking if InsP3R activation was sufficient to induce intrinsic plasticity in hippocampal neurons. Specifically, incorporation of d-myo-InsP3 in the recording pipette reduced input resistance, maximal impedance amplitude, and temporal summation but increased resonance frequency, resonance strength, sag ratio, and impedance phase lead. Strikingly, the magnitude of plasticity in all these measurements was dependent on InsP3 concentration, emphasizing the graded dependence of such plasticity on InsP3R activation. Mechanistically, we found that this InsP3-induced plasticity depended on hyperpolarization-activated cyclic nucleotide-gated channels. Moreover, this calcium-dependent form of plasticity was critically reliant on the release of calcium through InsP3Rs, the influx of calcium through N-methyl-d-aspartate receptors and voltage-gated calcium channels, and on the protein kinase A pathway. Our results delineate a causal role for InsP3Rs in graded adaptation of neuronal response dynamics, revealing novel regulatory roles for the endoplasmic reticulum in neural coding and homeostasis.
Collapse
Affiliation(s)
- Sufyan Ashhad
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India; and
| | - Daniel Johnston
- Center for Learning and Memory, The University of Texas at Austin, Austin, Texas
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India; and
| |
Collapse
|
199
|
Saraceno C, Marcello E, Di Marino D, Borroni B, Claeysen S, Perroy J, Padovani A, Tramontano A, Gardoni F, Di Luca M. SAP97-mediated ADAM10 trafficking from Golgi outposts depends on PKC phosphorylation. Cell Death Dis 2014; 5:e1547. [PMID: 25429624 PMCID: PMC4260750 DOI: 10.1038/cddis.2014.492] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 10/02/2014] [Indexed: 11/09/2022]
Abstract
A disintegrin and metalloproteinase 10 (ADAM10) is the major α-secretase that catalyzes the amyloid precursor protein (APP) ectodomain shedding in the brain and prevents amyloid formation. Its activity depends on correct intracellular trafficking and on synaptic membrane insertion. Here, we describe that in hippocampal neurons the synapse-associated protein-97 (SAP97), an excitatory synapse scaffolding element, governs ADAM10 trafficking from dendritic Golgi outposts to synaptic membranes. This process is mediated by a previously uncharacterized protein kinase C phosphosite in SAP97 SRC homology 3 domain that modulates SAP97 association with ADAM10. Such mechanism is essential for ADAM10 trafficking from the Golgi outposts to the synapse, but does not affect ADAM10 transport from the endoplasmic reticulum. Notably, this process is altered in Alzheimer's disease brains. These results help in understanding the mechanism responsible for the modulation of ADAM10 intracellular path, and can constitute an innovative therapeutic strategy to finely tune ADAM10 shedding activity towards APP.
Collapse
Affiliation(s)
- C Saraceno
- Department of Pharmacological and Biomolecular Sciences, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - E Marcello
- Department of Pharmacological and Biomolecular Sciences, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - D Di Marino
- Department of Physics, Sapienza University of Rome, P.le A. Moro, 5-00187 Rome, Italy
| | - B Borroni
- Department of Neurological Sciences, University of Brescia, 25125 Brescia, Italy
| | - S Claeysen
- 1] CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier, France [2] Inserm, U661, Montpellier, France [3] Universités de Montpellier 1 and 2, UMR-5203, Montpellier, France
| | - J Perroy
- 1] CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier, France [2] Inserm, U661, Montpellier, France [3] Universités de Montpellier 1 and 2, UMR-5203, Montpellier, France
| | - A Padovani
- Department of Neurological Sciences, University of Brescia, 25125 Brescia, Italy
| | - A Tramontano
- 1] Department of Physics, Sapienza University of Rome, P.le A. Moro, 5-00187 Rome, Italy [2] Institute Pasteur Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le A. Moro, 5-00187 Rome, Italy
| | - F Gardoni
- Department of Pharmacological and Biomolecular Sciences, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - M Di Luca
- Department of Pharmacological and Biomolecular Sciences, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
200
|
Gomez AM, Froemke RC, Burden SJ. Synaptic plasticity and cognitive function are disrupted in the absence of Lrp4. eLife 2014; 3:e04287. [PMID: 25407677 PMCID: PMC4270049 DOI: 10.7554/elife.04287] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 11/19/2014] [Indexed: 12/13/2022] Open
Abstract
Lrp4, the muscle receptor for neuronal Agrin, is expressed in the hippocampus and areas involved in cognition. The function of Lrp4 in the brain, however, is unknown, as Lrp4−/− mice fail to form neuromuscular synapses and die at birth. Lrp4−/− mice, rescued for Lrp4 expression selectively in muscle, survive into adulthood and showed profound deficits in cognitive tasks that assess learning and memory. To learn whether synapses form and function aberrantly, we used electrophysiological and anatomical methods to study hippocampal CA3–CA1 synapses. In the absence of Lrp4, the organization of the hippocampus appeared normal, but the frequency of spontaneous release events and spine density on primary apical dendrites were reduced. CA3 input was unable to adequately depolarize CA1 neurons to induce long-term potentiation. Our studies demonstrate a role for Lrp4 in hippocampal function and suggest that patients with mutations in Lrp4 or auto-antibodies to Lrp4 should be evaluated for neurological deficits. DOI:http://dx.doi.org/10.7554/eLife.04287.001 LRP4 is a muscle protein that is found in the hippocampus, a region of the brain that controls cognitive processes such as learning and memory. However, we know very little about what exactly LRP4 does in the hippocampus, and how it affects learning and memory. A standard way to figure out what a protein does is to study mice that have been genetically modified so that they cannot produce that protein. However, deleting the gene for LRP4 leads to muscle problems that kill these mutant mice at birth. To get around this problem, Gomez et al. have developed a method to restore the production of LRP4 in the muscles of mutant mice but not in their brains. These mutant mice were then subjected to a battery of tests to measure their ability to learn and recall new memories. These tests showed that LRP4 must be present in the brain, otherwise learning and memory are impaired. Gomez et al. also explored a process known as long-term potentiation. This process, which involves strengthening the functional connections between neurons, is believed to be essential for learning and other cognitive process. Gomez et al. demonstrated that long-term potentiation was disrupted by the lack of LRP4. Further experiments are needed to work out how LRP4 controls the learning process in the hippocampus and to explore the connection between LRP4 and various neuromuscular and neurological diseases. DOI:http://dx.doi.org/10.7554/eLife.04287.002
Collapse
Affiliation(s)
- Andrea M Gomez
- Graduate Program in Developmental Genetics, Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, NYU Medical Center, New York, United States
| | - Robert C Froemke
- Graduate Program in Developmental Genetics, Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, NYU Medical Center, New York, United States
| | - Steven J Burden
- Graduate Program in Developmental Genetics, Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, NYU Medical Center, New York, United States
| |
Collapse
|