Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Mol Psychiatry ; 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36450866

ABSTRACT

Postsynaptic scaffold proteins such as Shank, PSD-95, Homer and SAPAP/GKAP family members establish the postsynaptic density of glutamatergic synapses through a dense network of molecular interactions. Mutations in SHANK genes are associated with neurodevelopmental disorders including autism and intellectual disability. However, no SHANK missense mutations have been described which interfere with the key functions of Shank proteins believed to be central for synapse formation, such as GKAP binding via the PDZ domain, or Zn2+-dependent multimerization of the SAM domain. We identify two individuals with a neurodevelopmental disorder carrying de novo missense mutations in SHANK2. The p.G643R variant distorts the binding pocket for GKAP in the Shank2 PDZ domain and prevents interaction with Thr(-2) in the canonical PDZ ligand motif of GKAP. The p.L1800W variant severely delays the kinetics of Zn2+-dependent polymerization of the Shank2-SAM domain. Structural analysis shows that Trp1800 dislodges one histidine crucial for Zn2+ binding. The resulting conformational changes block the stacking of helical polymers of SAM domains into sheets through side-by-side contacts, which is a hallmark of Shank proteins, thereby disrupting the highly cooperative assembly process induced by Zn2+. Both variants reduce the postsynaptic targeting of Shank2 in primary cultured neurons and alter glutamatergic synaptic transmission. Super-resolution microscopy shows that both mutants interfere with the formation of postsynaptic nanoclusters. Our data indicate that both the PDZ- and the SAM-mediated interactions of Shank2 contribute to the compaction of postsynaptic protein complexes into nanoclusters, and that deficiencies in this process interfere with normal brain development in humans.

2.
Hum Genet ; 141(2): 257-272, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34907471

ABSTRACT

Bain type of X-linked syndromic intellectual developmental disorder, caused by pathogenic missense variants in HRNRPH2, was initially described in six female individuals affected by moderate-to-severe neurodevelopmental delay. Although it was initially postulated that the condition would not be compatible with life in males, several affected male individuals harboring pathogenic variants in HNRNPH2 have since been documented. However, functional in-vitro analyses of identified variants have not been performed and, therefore, possible genotype-phenotype correlations remain elusive. Here, we present eight male individuals, including a pair of monozygotic twins, harboring pathogenic or likely pathogenic HNRNPH2 variants. Notably, we present the first individuals harboring nonsense or frameshift variants who, similarly to an individual harboring a de novo p.(Arg29Cys) variant within the first quasi-RNA-recognition motif (qRRM), displayed mild developmental delay, and developed mostly autistic features and/or psychiatric co-morbidities. Additionally, we present two individuals harboring a recurrent de novo p.(Arg114Trp), within the second qRRM, who had a severe neurodevelopmental delay with seizures. Functional characterization of the three most common HNRNPH2 missense variants revealed dysfunctional nucleocytoplasmic shuttling of proteins harboring the p.(Arg206Gln) and p.(Pro209Leu) variants, located within the nuclear localization signal, whereas proteins with p.(Arg114Trp) showed reduced interaction with members of the large assembly of splicing regulators (LASR). Moreover, RNA-sequencing of primary fibroblasts of the individual harboring the p.(Arg114Trp) revealed substantial alterations in the regulation of alternative splicing along with global transcriptome changes. Thus, we further expand the clinical and variant spectrum in HNRNPH2-associated disease in males and provide novel molecular insights suggesting the disorder to be a spliceopathy on the molecular level.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Mutation , Neurodevelopmental Disorders/genetics , Adolescent , Alternative Splicing/genetics , Amino Acid Substitution , Brain/diagnostic imaging , Child , Child, Preschool , Chromosomes, Human, X/genetics , Codon, Nonsense , Diseases in Twins/diagnostic imaging , Diseases in Twins/genetics , Female , Frameshift Mutation , Genetic Association Studies , Genetic Variation , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Male , Mutation, Missense , Neurodevelopmental Disorders/diagnostic imaging , Phenotype , RNA-Seq , Twins, Monozygotic , Young Adult
3.
J Neurochem ; 157(4): 1331-1350, 2021 05.
Article in English | MEDLINE | ID: mdl-33090494

ABSTRACT

Mutations in the X-linked gene coding for the calcium-/calmodulin-dependent serine protein kinase (CASK) are associated with severe neurological disorders ranging from intellectual disability (in males) to mental retardation and microcephaly with pontine and cerebellar hypoplasia. CASK is involved in transcription control, in the regulation of trafficking of the post-synaptic NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, and acts as a presynaptic scaffolding protein. For CASK missense mutations, it is mostly unclear which of CASK's molecular interactions and cellular functions are altered and contribute to patient phenotypes. We identified five CASK missense mutations in male patients affected by neurodevelopmental disorders. These and five previously reported mutations were systematically analysed with respect to interaction with CASK interaction partners by co-expression and co-immunoprecipitation. We show that one mutation in the L27 domain interferes with binding to synapse-associated protein of 97 kDa. Two mutations in the guanylate kinase (GK) domain affect binding of CASK to the nuclear factors CASK-interacting nucleosome assembly protein (CINAP) and T-box, brain, 1 (Tbr1). A total of five mutations in GK as well as PSD-95/discs large/ZO-1 (PDZ) domains affect binding of CASK to the pre-synaptic cell adhesion molecule Neurexin. Upon expression in neurons, we observe that binding to Neurexin is not required for pre-synaptic localization of CASK. We show by bimolecular fluorescence complementation assay that Neurexin induces oligomerization of CASK, and that mutations in GK and PDZ domains interfere with the Neurexin-induced oligomerization of CASK. Our data are supported by molecular modelling, where we observe that the cooperative activity of PDZ, SH3 and GK domains is required for Neurexin binding and oligomerization of CASK.


Subject(s)
Guanylate Kinases/genetics , Neural Cell Adhesion Molecules/metabolism , Neurodevelopmental Disorders/metabolism , Animals , Humans , Male , Models, Molecular , Mutation, Missense , Protein Binding , Rats
4.
Pharmacol Rev ; 70(4): 763-835, 2018 10.
Article in English | MEDLINE | ID: mdl-30232095

ABSTRACT

Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.


Subject(s)
Receptors, Somatostatin/metabolism , Animals , Gene Expression Regulation , Humans , Ligands , Protein Conformation , Protein Transport , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Receptors, Somatostatin/physiology , Signal Transduction , Terminology as Topic
5.
J Neurochem ; 155(3): 250-263, 2020 11.
Article in English | MEDLINE | ID: mdl-32202324

ABSTRACT

Mutations in SHANK3, coding for a large scaffold protein of excitatory synapses in the CNS, are associated with neurodevelopmental disorders including autism spectrum disorders and intellectual disability (ID). Several cases have been identified in which the mutation leads to truncation of the protein, eliminating C-terminal sequences required for post-synaptic targeting of the protein. We identify here a patient with a truncating mutation in SHANK3, affected by severe global developmental delay and intellectual disability. By analyzing the subcellular distribution of this truncated form of Shank3, we identified a nuclear localization signal (NLS) in the N-terminal part of the protein which is responsible for targeting Shank3 fragments to the nucleus. To determine the relevance of Shank3 for nuclear signaling, we analyze how it affects signaling by ß-catenin, a component of the Wnt pathway. We show that full length as well as truncated variants of Shank3 interact with ß-catenin via the PDZ domain of Shank3, and the armadillo repeats of ß-catenin. As a result of this interaction, truncated forms of Shank3 and ß-catenin strictly co-localize in small intra-nuclear bodies both in 293T cells and in rat hippocampal neurons. On a functional level, the sequestration of both proteins in these nuclear bodies is associated with a strongly repressed transcriptional activation by ß-catenin owing to interaction with the truncated Shank3 fragment found in patients. Our data suggest that truncating mutations in SHANK3 may not only lead to a reduction in Shank3 protein available at postsynaptic sites but also negatively affect the Wnt signaling pathway.


Subject(s)
Cell Nucleus/metabolism , Developmental Disabilities/metabolism , Mutation/physiology , Nerve Tissue Proteins/metabolism , beta Catenin/metabolism , Animals , Cell Nucleus/genetics , Cells, Cultured , Developmental Disabilities/genetics , Female , HEK293 Cells , Humans , Male , Nerve Tissue Proteins/genetics , Pregnancy , Rats , Rats, Wistar , Signal Transduction/physiology
6.
Am J Hum Genet ; 101(5): 716-724, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100085

ABSTRACT

DHX30 is a member of the family of DExH-box helicases, which use ATP hydrolysis to unwind RNA secondary structures. Here we identified six different de novo missense mutations in DHX30 in twelve unrelated individuals affected by global developmental delay (GDD), intellectual disability (ID), severe speech impairment and gait abnormalities. While four mutations are recurrent, two are unique with one affecting the codon of one recurrent mutation. All amino acid changes are located within highly conserved helicase motifs and were found to either impair ATPase activity or RNA recognition in different in vitro assays. Moreover, protein variants exhibit an increased propensity to trigger stress granule (SG) formation resulting in global translation inhibition. Thus, our findings highlight the prominent role of translation control in development and function of the central nervous system and also provide molecular insight into how DHX30 dysfunction might cause a neurodevelopmental disorder.


Subject(s)
Developmental Disabilities/genetics , Mutation, Missense/genetics , RNA Helicases/genetics , Adenosine Triphosphatases/genetics , Adolescent , Amino Acids/genetics , Cell Line , Cell Line, Tumor , Central Nervous System/pathology , Child , Child, Preschool , Female , HEK293 Cells , Humans , Intellectual Disability/genetics , Male , RNA/genetics
7.
J Biol Chem ; 290(10): 6120-9, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25614626

ABSTRACT

Many G-protein-coupled receptors carry C-terminal ligand motifs for PSD-95/discs large/ZO-1 (PDZ) domains; via interaction with PDZ domain-containing scaffold proteins, this allows for integration of receptors into signaling complexes. However, the presence of PDZ domain proteins attached to intracellular membranes suggests that PDZ-type interactions may also contribute to subcellular sorting of receptors. The protein interacting specifically with Tc10 (PIST; also known as GOPC) is a trans-Golgi-associated protein that interacts through its single PDZ domain with a variety of cell surface receptors. Here we show that PIST controls trafficking of the interacting ß1-adrenergic receptor both in the anterograde, biosynthetic pathway and during postendocytic recycling. Overexpression and knockdown experiments show that PIST leads to retention of the receptor in the trans-Golgi network (TGN), to the effect that overexpressed PIST reduces activation of the MAPK pathway by ß1-adrenergic receptor (ß1AR) agonists. Receptors can be released from retention in the TGN by coexpression of the plasma membrane-associated scaffold PSD-95, which allows for transport of receptors to the plasma membrane. Stimulation of ß1 receptors and activation of the cAMP pathway lead to relocation of PIST from the TGN to an endosome-like compartment. Here PIST colocalizes with SNX1 and the internalized ß1AR and protects endocytosed receptors from lysosomal degradation. In agreement, ß1AR levels are decreased in hippocampi of PIST-deficient mice. Our data suggest that PIST contributes to the fine-tuning of ß1AR sorting both during biosynthetic and postendocytic trafficking.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , PDZ Domains/genetics , Receptors, Adrenergic, beta-1/metabolism , trans-Golgi Network/genetics , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Endocytosis/genetics , Endosomes/genetics , Gene Expression Regulation , Golgi Matrix Proteins , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Transport Proteins , Mice , Receptors, Adrenergic, beta-1/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , trans-Golgi Network/metabolism
8.
J Neurochem ; 136(4): 752-763, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26560964

ABSTRACT

Learning and memory is dependent on postsynaptic architecture and signaling processes in forebrain regions. The insulin receptor substrate protein of 53 kDa (IRSp53, also known as Baiap2) is a signaling and adapter protein in forebrain excitatory synapses. Mice deficient in IRSp53 display enhanced levels of postsynaptic N-methyl-D-aspartate receptors (NMDARs) and long-term potentiation (LTP) associated with severe learning deficits. In humans, reduced IRSp53/Baiap2 expression is associated with a variety of neurological disorders including autism, schizophrenia, and Alzheimer's disease. Here, we analyzed mice lacking one copy of the gene coding for IRSp53 using behavioral tests including contextual fear conditioning and the puzzle box. We show that a 50% reduction in IRSp53 levels strongly affects the performance in fear-evoking learning paradigms. This correlates with increased targeting of NMDARs to the postsynaptic density (PSD) in hippocampi of both heterozygous and knock out (ko) mice at the expense of extrasynaptic NMDARs. As hippocampal NMDAR-dependent LTP is enhanced in IRSp53-deficient mice, we investigated signaling cascades important for the formation of fear-evoked memories. Here, we observed a dramatic increase in cAMP response element-binding protein-dependent signaling in heterozygous and IRSp53-deficient mice, necessary for the transcriptional dependent phase of LTP. In contrast, activation of the MAPK and Akt kinase pathways required for translation-dependent phase of LTP are reduced. Our data suggest that loss or even the reduction in IRSp53 increases NMDAR-dependent cAMP responsive element-binding protein activation in the hippocampus, and interferes with the ability of mice to learn upon anxiety-related stimuli. We show here that a moderate reduction in the postsynaptic protein IRSp53 in mice leads to an increase in postsynaptic NMDA receptors. Both in heterozygous and IRSp53 deficient mice, this is associated with altered postsynaptic signal transduction, and poor performance of mice in fear-associated learning paradigms, indicating that precise control of postsynaptic NMDA receptor density is essential for memory formation.

10.
J Biol Chem ; 288(37): 26697-708, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23897824

ABSTRACT

Shank/ProSAP proteins are major scaffold proteins of the postsynaptic density; mutations in the human SHANK3 gene are associated with intellectual disability or autism spectrum disorders. We have analyzed the functional relevance of several SHANK3 missense mutations affecting the N-terminal portion of the protein by expression of wild-type and mutant Shank3 in cultured neurons and by binding assays in heterologous cells. Postsynaptic targeting of recombinant Shank3 was unaltered. In electrophysiological experiments, both wild-type and L68P mutant forms of Shank3 were equally effective in restoring synaptic function after knockdown of endogenous Shank3. We observed that several mutations affected binding to interaction partners of the Shank3 ankyrin repeat region. One of these mutations, L68P, improved binding to both ligands. Leu-68 is located N-terminal to the ankyrin repeats, in a highly conserved region that we identify here as a novel domain termed the Shank/ProSAP N-terminal (SPN) domain. We show that the SPN domain interacts with the ankyrin repeats in an intramolecular manner, thereby restricting access of either Sharpin or α-fodrin. The L68P mutation disrupts this blockade, thus exposing the Shank3 ankyrin repeat region to its ligands. Our data identify a new type of regulation of Shank proteins and suggest that mutations in the SHANK3 gene do not necessarily induce a loss of function, but may represent a gain of function with respect to specific interaction partners.


Subject(s)
Ankyrin Repeat/genetics , Autistic Disorder/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Animals , Autistic Disorder/metabolism , Carrier Proteins/metabolism , Electrophysiology , HEK293 Cells , Hippocampus/cytology , Humans , Leucine/chemistry , Ligands , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Mutation, Missense , Neurons/metabolism , Patch-Clamp Techniques , Protein Binding , Protein Structure, Tertiary , Rats , Synaptic Transmission , Two-Hybrid System Techniques , Ubiquitins/metabolism
11.
Hum Mol Genet ; 21(2): 268-86, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21989057

ABSTRACT

Mutations in the ARHGEF6 gene, encoding the guanine nucleotide exchange factor αPIX/Cool-2 for the Rho GTPases Rac1 and Cdc42, cause X-linked intellectual disability (ID) in humans. We show here that αPix/Arhgef6 is primarily expressed in neuropil regions of the hippocampus. To study the role of αPix/Arhgef6 in neuronal development and plasticity and gain insight into the pathogenic mechanisms underlying ID, we generated αPix/Arhgef6-deficient mice. Gross brain structure in these mice appeared to be normal; however, analysis of Golgi-Cox-stained pyramidal neurons revealed an increase in both dendritic length and spine density in the hippocampus, accompanied by an overall loss in spine synapses. Early-phase long-term potentiation was reduced and long-term depression was increased in the CA1 hippocampal area of αPix/Arhgef6-deficient animals. Knockout animals exhibited impaired spatial and complex learning and less behavioral control in mildly stressful situations, suggesting that this model mimics the human ID phenotype. The structural and electrophysiological alterations in the hippocampus were accompanied by a significant reduction in active Rac1 and Cdc42, but not RhoA. In conclusion, we suggest that imbalance in activity of different Rho GTPases may underlie altered neuronal connectivity and impaired synaptic function and cognition in αPix/Arhgef6 knockout mice.


Subject(s)
Cognition Disorders/genetics , Disease Models, Animal , Genetic Diseases, X-Linked/genetics , Guanine Nucleotide Exchange Factors/genetics , Intellectual Disability/genetics , Neuronal Plasticity/genetics , rho GTP-Binding Proteins/metabolism , Animals , Maze Learning , Mice , Mice, Knockout , Rho Guanine Nucleotide Exchange Factors
12.
Am J Hum Genet ; 88(3): 382-90, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21397065

ABSTRACT

Posterior microphthalmos (MCOP) is a rare isolated developmental anomaly of the eye characterized by extreme hyperopia due to short axial length. The population of the Faroe Islands shows a high prevalence of an autosomal-recessive form (arMCOP) of the disease. Based on published linkage data, we refined the position of the disease locus (MCOP6) in an interval of 250 kb in chromosome 2q37.1 in two large Faroese families. We detected three different mutations in PRSS56. Patients of the Faroese families were either homozygous for c.926G>C (p.Trp309Ser) or compound heterozygous for c.926G>C and c.526C>G (p.Arg176Gly), whereas a homozygous 1 bp duplication (c.1066dupC) was identified in five patients with arMCOP from a consanguineous Tunisian family. In one patient with MCOP from the Faroe Islands and in another one from Turkey, no PRSS56 mutation was detected, suggesting nonallelic heterogeneity of the trait. Using RT-PCR, PRSS56 transcripts were detected in samples derived from the human adult retina, cornea, sclera, and optic nerve. The expression of the mouse ortholog could be first detected in the eye at E17 and was maintained into adulthood. The predicted PRSS56 protein is a 603 amino acid long secreted trypsin-like serine peptidase. The c.1066dupC is likely to result in a functional null allele, whereas the two point mutations predict the replacement of evolutionary conserved and functionally important residues. Molecular modeling of the p.Trp309Ser mutant suggests that both the affinity and reactivity of the enzyme toward in vivo protein substrates are likely to be substantially reduced.


Subject(s)
Genes, Recessive/genetics , Microphthalmos/genetics , Mutation/genetics , Serine Endopeptidases/genetics , Serine Proteases/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Mutational Analysis , Eye/enzymology , Eye/pathology , Family , Gene Expression Regulation, Enzymologic , Genetic Loci/genetics , Humans , Meiosis/genetics , Mice , Microphthalmos/enzymology , Models, Molecular , Molecular Sequence Data , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Serine Proteases/chemistry , Serine Proteases/metabolism
13.
Mol Neurobiol ; 61(2): 693-706, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37656313

ABSTRACT

Members of the Shank family of postsynaptic scaffold proteins (Shank1-3) link neurotransmitter receptors to the actin cytoskeleton in dendritic spines through establishing numerous interactions within the postsynaptic density (PSD) of excitatory synapses. Large Shank isoforms carry at their N-termini a highly conserved domain termed the Shank/ProSAP N-terminal (SPN) domain, followed by a set of Ankyrin repeats. Both domains are involved in an intramolecular interaction which is believed to regulate accessibility for additional interaction partners, such as Ras family G-proteins, αCaMKII, and cytoskeletal proteins. Here, we analyze the functional relevance of the SPN-Ank module; we show that binding of active Ras or Rap1a to the SPN domain can differentially regulate the localization of Shank3 in dendrites. In Shank1 and Shank3, the linker between the SPN and Ank domains binds to inactive αCaMKII. Due to this interaction, both Shank1 and Shank3 exert a negative effect on αCaMKII activity at postsynaptic sites in mice in vivo. The relevance of the SPN-Ank intramolecular interaction was further analyzed in primary cultured neurons; here, we observed that in the context of full-length Shank3, a closed conformation of the SPN-Ank tandem is necessary for proper clustering of Shank3 on the head of dendritic spines. Shank3 variants carrying Ank repeats which are not associated with the SPN domain lead to the atypical formation of postsynaptic clusters on dendritic shafts, at the expense of clusters in spine-like protrusions. Our data show that the SPN-Ank tandem motif contributes to the regulation of postsynaptic signaling and is also necessary for proper targeting of Shank3 to postsynaptic sites. Our data also suggest how missense variants found in autistic patients which alter SPN and Ank domains affect the synaptic function of Shank3.


Subject(s)
Nerve Tissue Proteins , Signal Transduction , Mice , Humans , Animals , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , Cytoskeletal Proteins/metabolism , Microfilament Proteins/metabolism
14.
J Neurochem ; 124(5): 670-84, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23121659

ABSTRACT

Dendritic targeting of mRNAs encoding the microtubule-associated protein 2 (MAP2) in neurons involves a cis-acting dendritic targeting element. Two rat brain proteins, MAP2-RNA trans-acting protein (MARTA)1 and MARTA2, bind to the cis-element with both high affinity and specificity. In this study, affinity-purified MARTA2 was identified as orthologue of human far-upstream element binding protein 3. In neurons, it resides in somatodendritic granules and dendritic spines and associates with MAP2 mRNAs. Expression of a dominant-negative variant of MARTA2 disrupts dendritic targeting of endogenous MAP2 mRNAs, while not noticeably altering the level and subcellular distribution of polyadenylated mRNAs as a whole. Finally, MAP2 transcripts associate with the microtubule-based motor KIF5 and inhibition of KIF5, but not cytoplasmic dynein function disrupts extrasomatic trafficking of MAP2 mRNA granules. Thus, in neurons MARTA2 appears to represent a key trans-acting factor involved in KIF5-mediated dendritic targeting of MAP2 mRNAs.


Subject(s)
Dendrites/metabolism , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Blotting, Western , Dendrites/ultrastructure , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Mass Spectrometry , Microscopy, Immunoelectron , Neurons/ultrastructure , Protein Transport/physiology , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS Biol ; 7(6): e1000138, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19564905

ABSTRACT

The regulation of filopodia plays a crucial role during neuronal development and synaptogenesis. Axonal filopodia, which are known to originate presynaptic specializations, are regulated in response to neurotrophic factors. The structural components of filopodia are actin filaments, whose dynamics and organization are controlled by ensembles of actin-binding proteins. How neurotrophic factors regulate these latter proteins remains, however, poorly defined. Here, using a combination of mouse genetic, biochemical, and cell biological assays, we show that genetic removal of Eps8, an actin-binding and regulatory protein enriched in the growth cones and developing processes of neurons, significantly augments the number and density of vasodilator-stimulated phosphoprotein (VASP)-dependent axonal filopodia. The reintroduction of Eps8 wild type (WT), but not an Eps8 capping-defective mutant, into primary hippocampal neurons restored axonal filopodia to WT levels. We further show that the actin barbed-end capping activity of Eps8 is inhibited by brain-derived neurotrophic factor (BDNF) treatment through MAPK-dependent phosphorylation of Eps8 residues S624 and T628. Additionally, an Eps8 mutant, impaired in the MAPK target sites (S624A/T628A), displays increased association to actin-rich structures, is resistant to BDNF-mediated release from microfilaments, and inhibits BDNF-induced filopodia. The opposite is observed for a phosphomimetic Eps8 (S624E/T628E) mutant. Thus, collectively, our data identify Eps8 as a critical capping protein in the regulation of axonal filopodia and delineate a molecular pathway by which BDNF, through MAPK-dependent phosphorylation of Eps8, stimulates axonal filopodia formation, a process with crucial impacts on neuronal development and synapse formation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cytoskeletal Proteins/metabolism , Neurons/drug effects , Pseudopodia/physiology , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Axons/metabolism , Axons/physiology , Cell Line , Cells, Cultured , Cytoskeletal Proteins/genetics , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism , RNA Interference , Rats , Transfection
17.
Adv Exp Med Biol ; 970: 285-305, 2012.
Article in English | MEDLINE | ID: mdl-22351061

ABSTRACT

Selective targeting of specific mRNAs into neuronal dendrites and their locally regulated translation at particular cell contact sites contribute to input-specific synaptic plasticity. Thus, individual synapses become decision-making units, which control gene expression in a spatially restricted and nucleus-independent manner. Dendritic targeting of mRNAs is achieved by active, microtubule-dependent transport. For this purpose, mRNAs are packaged into large ribonucleoprotein (RNP) particles containing an array of trans-acting RNA-binding proteins. These are attached to molecular motors, which move their RNP cargo into dendrites. A variety of proteins may be synthesized in dendrites, including signalling and scaffold proteins of the synapse and neurotransmitter receptors. In some cases, such as the alpha subunit of the calcium/calmodulin-dependent protein kinase II (αCaMKII) and the activity-regulated gene of 3.1 kb (Arg3.1, also referred to as activity-regulated cDNA, Arc), their local synthesis at synapses can modulate long-term changes in synaptic efficiency. Local dendritic translation is regulated by several signalling cascades including Akt/mTOR and Erk/MAP kinase pathways, which are triggered by synaptic activity. More recent findings show that miRNAs also play an important role in protein synthesis at synapses. Disruption of local translation control at synapses, as observed in the fragile X syndrome (FXS) and its mouse models and possibly also in autism spectrum disorders, interferes with cognitive abilities in mice and men.


Subject(s)
Dendrites/genetics , Gene Expression Regulation/physiology , Protein Biosynthesis/physiology , RNA, Messenger/genetics , Signal Transduction/genetics , Synapses/physiology , Animals , Biological Transport, Active/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dendrites/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Humans , Long-Term Potentiation/physiology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
18.
Life Sci Alliance ; 5(10)2022 10.
Article in English | MEDLINE | ID: mdl-36137748

ABSTRACT

CASK is a unique membrane-associated guanylate kinase (MAGUK) because of its Ca2+/calmodulin-dependent kinase (CaMK) domain. We describe four male patients with a severe neurodevelopmental disorder with microcephaly carrying missense variants affecting the CaMK domain. One boy who carried the p.E115K variant and died at an early age showed pontocerebellar hypoplasia (PCH) in addition to microcephaly, thus exhibiting the classical MICPCH phenotype observed in individuals with CASK loss-of-function variants. All four variants selectively weaken the interaction of CASK with Liprin-α2, a component of the presynaptic active zone. Liprin-α proteins form spherical phase-separated condensates, which we observe here in Liprin-α2 overexpressing HEK293T cells. Large Liprin-α2 clusters were also observed in transfected primary-cultured neurons. Cluster formation of Liprin-α2 is reversed in the presence of CASK; this is associated with altered phosphorylation of Liprin-α2. The p.E115K variant fails to interfere with condensate formation. As the individual carrying this variant had the severe MICPCH disorder, we suggest that regulation of Liprin-α2-mediated phase condensate formation is a new functional feature of CASK which must be maintained to prevent PCH.


Subject(s)
Microcephaly , Calmodulin/genetics , Calmodulin/metabolism , Cerebellar Diseases , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , HEK293 Cells , Humans , Male , Mental Retardation, X-Linked , Microcephaly/genetics , Mutation
19.
Sci Rep ; 12(1): 902, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35042901

ABSTRACT

Shank proteins are major scaffolds of the postsynaptic density of excitatory synapses. Mutations in SHANK genes are associated with autism and intellectual disability. The effects of missense mutations on Shank3 function, and therefore the pathomechanisms are unclear. Several missense mutations in SHANK3 affect the N-terminal region, consisting of the Shank/ProSAP N-terminal (SPN) domain and a set of Ankyrin (Ank) repeats. Here we identify a novel SHANK3 missense mutation (p.L270M) in the Ankyrin repeats in patients with an ADHD-like phenotype. We functionally analysed this and a series of other mutations, using biochemical and biophysical techniques. We observe two major effects: (1) a loss of binding to δ-catenin (e.g. in the p.L270M variant), and (2) interference with the intramolecular interaction between N-terminal SPN domain and the Ank repeats. This also interferes with binding to the α-subunit of the calcium-/calmodulin dependent kinase II (αCaMKII), and appears to be associated with a more severe neurodevelopmental pathology.


Subject(s)
Synapses
20.
Traffic ; 10(7): 844-57, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19416473

ABSTRACT

Dendritic mRNA transport coupled with local regulation of translation enables neurons to selectively alter the protein composition of individual postsynaptic sites. We have analyzed dendritic localization of shank1 mRNAs; shank proteins (shank1-3) are scaffolding molecules of the postsynaptic density (PSD) of excitatory synapses, which are crucial for PSD assembly and the formation of dendritic spines. Live cell imaging demonstrates saltatory movements of shank1 mRNA containing granules along microtubules in both anterograde and retrograde directions. A population of brain messenger ribonucleoprotein particles (mRNPs) containing shank1 mRNAs associates with the cargo-binding domain of the motor protein KIF5C. Through expression of dominant negative proteins, we show that dendritic targeting of shank1 mRNA granules involves KIF5C and the KIF5-associated RNA-binding protein staufen1. While transport of shank1 mRNAs follows principles previously outlined for other dendritic transcripts, shank1 mRNAs are distinguished by their translational regulation. Translation is strongly inhibited by a GC-rich 5(')untranslated region; in addition, internal ribosomal entry sites previously detected in other dendritic transcripts are absent in the shank1 mRNA. A concept emerges from our data in which dendritic transport of different mRNAs occurs collectively via a staufen1- and KIF5-dependent pathway, whereas their local translation is controlled individually by unique cis-acting elements.


Subject(s)
5' Untranslated Regions , Dendrites/metabolism , Kinesins/metabolism , Membrane Proteins , Protein Biosynthesis , RNA, Messenger/metabolism , Biological Transport/physiology , Cells, Cultured , Gene Expression Regulation , Humans , Kinesins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL