Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neuron ; 111(17): 2660-2674.e9, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37385246

ABSTRACT

Many RNA-binding proteins (RBPs), particularly those associated with RNA granules, promote pathological protein aggregation in neurodegenerative diseases. Here, we demonstrate that G3BP2, a core component of stress granules, directly interacts with Tau and inhibits Tau aggregation. In the human brain, the interaction of G3BP2 and Tau is dramatically increased in multiple tauopathies, and it is independent of neurofibrillary tangle (NFT) formation in Alzheimer's disease (AD). Surprisingly, Tau pathology is significantly elevated upon loss of G3BP2 in human neurons and brain organoids. Moreover, we found that G3BP2 masks the microtubule-binding region (MTBR) of Tau, thereby inhibiting Tau aggregation. Our study defines a novel role for RBPs as a line of defense against Tau aggregation in tauopathies.


Subject(s)
Alzheimer Disease , Tauopathies , Humans , tau Proteins/metabolism , Tauopathies/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Neurons/metabolism , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism
2.
Cells ; 11(22)2022 11 17.
Article in English | MEDLINE | ID: mdl-36429079

ABSTRACT

The AMPA glutamate receptor (AMPAR) is the major type of synaptic excitatory ionotropic receptor in the brain. AMPARs have four different subunits, GluA1-4 (each encoded by different genes, Gria1, Gria2, Gria3 and Gria4), that can form distinct tetrameric assemblies. The most abundant AMPAR subtypes in the hippocampus are GluA1/2 and GluA2/3 heterotetramers. Each subtype contributes differentially to mechanisms of synaptic plasticity, which may be in part caused by how these receptors are regulated by specific associated proteins. A broad range of AMPAR interacting proteins have been identified, including the well-studied transmembrane AMPA receptor regulatory proteins TARP-γ2 (also known as Stargazin) and TARP-γ8, Cornichon homolog 2 (CNIH-2) and many others. Several interactors were shown to affect biogenesis, AMPAR trafficking, and channel properties, alone or in distinct assemblies, and several revealed preferred binding to specific AMPAR subunits. To date, a systematic specific interactome analysis of the major GluA1/2 and GluA2/3 AMPAR subtypes separately is lacking. To reveal interactors belonging to specific AMPAR subcomplexes, we performed both expression and interaction proteomics on hippocampi of wildtype and Gria1- or Gria3 knock-out mice. Whereas GluA1/2 receptors co-purified TARP-γ8, synapse differentiation-induced protein 4 (SynDIG4, also known as Prrt1) and CNIH-2 with highest abundances, GluA2/3 receptors revealed strongest co-purification of CNIH-2, TARP-γ2, and Noelin1 (or Olfactomedin-1). Further analysis revealed that TARP-γ8-SynDIG4 interact directly and co-assemble into an AMPAR subcomplex especially at synaptic sites. Together, these data provide a framework for further functional analysis into AMPAR subtype specific pathways in health and disease.


Subject(s)
Proteomics , Receptors, AMPA , Animals , Mice , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synapses/metabolism , Neuronal Plasticity/physiology , Hippocampus/metabolism , Mice, Knockout
3.
Mol Ther Nucleic Acids ; 29: 625-642, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36090761

ABSTRACT

Tau is a microtubule-associated protein (MAPT, tau) implicated in the pathogenesis of tauopathies, a spectrum of neurodegenerative disorders characterized by accumulation of hyperphosphorylated, aggregated tau. Because tau pathology can be distinct across diseases, a pragmatic therapeutic approach may be to intervene at the level of the tau transcript, as it makes no assumptions to mechanisms of tau toxicity. Here we performed a large library screen of locked-nucleic-acid (LNA)-modified antisense oligonucleotides (ASOs), where careful tiling of the MAPT locus resulted in the identification of hot spots for activity in the 3' UTR. Further modifications to the LNA design resulted in the generation of ASO-001933, which selectively and potently reduces tau in primary cultures from hTau mice, monkey, and human neurons. ASO-001933 was well tolerated and produced a robust, long-lasting reduction in tau protein in both mouse and cynomolgus monkey brain. In monkey, tau protein reduction was maintained in brain for 20 weeks post injection and corresponded with tau protein reduction in the cerebrospinal fluid (CSF). Our results demonstrate that LNA-ASOs exhibit excellent drug-like properties and sustained efficacy likely translating to infrequent, intrathecal dosing in patients. These data further support the development of LNA-ASOs against tau for the treatment of tauopathies.

4.
Mol Psychiatry ; 27(5): 2590-2601, 2022 05.
Article in English | MEDLINE | ID: mdl-35264729

ABSTRACT

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by the loss of neuronal E3 ligase UBE3A. Restoring UBE3A levels is a potential disease-modifying therapy for AS and has recently entered clinical trials. There is paucity of data regarding the molecular changes downstream of UBE3A hampering elucidation of disease therapeutics and biomarkers. Notably, UBE3A plays an important role in the nucleus but its targets have yet to be elucidated. Using proteomics, we assessed changes during postnatal cortical development in an AS mouse model. Pathway analysis revealed dysregulation of proteasomal and tRNA synthetase pathways at all postnatal brain developmental stages, while synaptic proteins were altered in adults. We confirmed pathway alterations in an adult AS rat model across multiple brain regions and highlighted region-specific differences. UBE3A reinstatement in AS model mice resulted in near complete and partial rescue of the proteome alterations in adolescence and adults, respectively, supporting the notion that restoration of UBE3A expression provides a promising therapeutic option. We show that the nuclear enriched transketolase (TKT), one of the most abundantly altered proteins, is a novel direct UBE3A substrate and is elevated in the neuronal nucleus of rat brains and human iPSC-derived neurons. Taken together, our study provides a comprehensive map of UBE3A-driven proteome remodeling in AS across development and species, and corroborates an early UBE3A reinstatement as a viable therapeutic option. To support future disease and biomarker research, we present an accessible large-scale multi-species proteomic resource for the AS community ( https://www.angelman-proteome-project.org/ ).


Subject(s)
Angelman Syndrome , Proteomics , Angelman Syndrome/drug therapy , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Animals , Disease Models, Animal , Mice , Proteome , Rats , Signal Transduction , Ubiquitin-Protein Ligases/genetics
5.
Cell Rep Med ; 2(8): 100360, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34467244

ABSTRACT

Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of maternal UBE3A, a ubiquitin protein ligase E3A. Here, we study neurons derived from patients with AS and neurotypical individuals, and reciprocally modulate UBE3A using antisense oligonucleotides. Unbiased proteomics reveal proteins that are regulated by UBE3A in a disease-specific manner, including PEG10, a retrotransposon-derived GAG protein. PEG10 protein increase, but not RNA, is dependent on UBE3A and proteasome function. PEG10 binds to both RNA and ataxia-associated proteins (ATXN2 and ATXN10), localizes to stress granules, and is secreted in extracellular vesicles, modulating vesicle content. Rescue of AS patient-derived neurons by UBE3A reinstatement or PEG10 reduction reveals similarity in transcriptome changes. Overexpression of PEG10 during mouse brain development alters neuronal migration, suggesting that it can affect brain development. These findings imply that PEG10 is a secreted human UBE3A target involved in AS pathophysiology.


Subject(s)
Angelman Syndrome/metabolism , Angelman Syndrome/physiopathology , Apoptosis Regulatory Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Products, gag/chemistry , RNA-Binding Proteins/metabolism , Retroviridae/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Movement , Child, Preschool , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Female , Humans , Induced Pluripotent Stem Cells/pathology , Male , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Domains , Retroelements/genetics , Stress Granules/metabolism , Stress Granules/ultrastructure , Transcriptome/genetics
6.
Proc Natl Acad Sci U S A ; 117(33): 19854-19865, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32759214

ABSTRACT

The blood-retina barrier and blood-brain barrier (BRB/BBB) are selective and semipermeable and are critical for supporting and protecting central nervous system (CNS)-resident cells. Endothelial cells (ECs) within the BRB/BBB are tightly coupled, express high levels of Claudin-5 (CLDN5), a junctional protein that stabilizes ECs, and are important for proper neuronal function. To identify novel CLDN5 regulators (and ultimately EC stabilizers), we generated a CLDN5-P2A-GFP stable cell line from human pluripotent stem cells (hPSCs), directed their differentiation to ECs (CLDN5-GFP hPSC-ECs), and performed flow cytometry-based chemogenomic library screening to measure GFP expression as a surrogate reporter of barrier integrity. Using this approach, we identified 62 unique compounds that activated CLDN5-GFP. Among them were TGF-ß pathway inhibitors, including RepSox. When applied to hPSC-ECs, primary brain ECs, and retinal ECs, RepSox strongly elevated barrier resistance (transendothelial electrical resistance), reduced paracellular permeability (fluorescein isothiocyanate-dextran), and prevented vascular endothelial growth factor A (VEGFA)-induced barrier breakdown in vitro. RepSox also altered vascular patterning in the mouse retina during development when delivered exogenously. To determine the mechanism of action of RepSox, we performed kinome-, transcriptome-, and proteome-profiling and discovered that RepSox inhibited TGF-ß, VEGFA, and inflammatory gene networks. In addition, RepSox not only activated vascular-stabilizing and barrier-establishing Notch and Wnt pathways, but also induced expression of important tight junctions and transporters. Taken together, our data suggest that inhibiting multiple pathways by selected individual small molecules, such as RepSox, may be an effective strategy for the development of better BRB/BBB models and novel EC barrier-inducing therapeutics.


Subject(s)
Endothelial Cells/drug effects , Pluripotent Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/metabolism , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Claudin-5/genetics , Claudin-5/metabolism , Drug Evaluation, Preclinical , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Editing , Genome , Humans , Mice , Mice, Knockout , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
Front Mol Neurosci ; 11: 364, 2018.
Article in English | MEDLINE | ID: mdl-30333727

ABSTRACT

Key to the human brain's unique capacities are a myriad of neural cell types, specialized molecular expression signatures, and complex patterns of neuronal connectivity. Neurons in the human brain communicate via well over a quadrillion synapses. Their specific contribution might be key to the dynamic activity patterns that underlie primate-specific cognitive function. Recently, functional differences were described in transmission capabilities of human and rat synapses. To test whether unique expression signatures of synaptic proteins are at the basis of this, we performed a quantitative analysis of the hippocampal synaptic proteome of four mammalian species, two primates, human and marmoset, and two rodents, rat and mouse. Abundance differences down to 1.15-fold at an FDR-corrected p-value of 0.005 were reliably detected using SWATH mass spectrometry. The high measurement accuracy of SWATH allowed the detection of a large group of differentially expressed proteins between individual species and rodent vs. primate. Differentially expressed proteins between rodent and primate were found highly enriched for plasticity-related proteins.

8.
Cell Rep ; 24(5): 1218-1230, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067977

ABSTRACT

Lateral diffusion on the neuronal plasma membrane of the AMPA-type glutamate receptor (AMPAR) serves an important role in synaptic plasticity. We investigated the role of the secreted glycoprotein Noelin1 (Olfactomedin-1 or Pancortin) in AMPAR lateral mobility and its dependence on the extracellular matrix (ECM). We found that Noelin1 interacts with the AMPAR with high affinity, however, without affecting rise- and decay time and desensitization properties. Noelin1 co-localizes with synaptic and extra-synaptic AMPARs and is expressed at synapses in an activity-dependent manner. Single-particle tracking shows that Noelin1 reduces lateral mobility of both synaptic and extra-synaptic GluA1-containing receptors and affects short-term plasticity. While the ECM does not constrain the synaptic pool of AMPARs and acts only extrasynaptically, Noelin1 contributes to synaptic potentiation by limiting AMPAR mobility at synaptic sites. This is the first evidence for the role of a secreted AMPAR-interacting protein on mobility of GluA1-containing receptors and synaptic plasticity.


Subject(s)
Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Neuronal Plasticity , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Cells, Cultured , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Protein Binding , Protein Transport
9.
Sci Rep ; 7(1): 12107, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28935861

ABSTRACT

Protein correlation profiling might assist in defining co-assembled proteins and subcellular distribution. Here, we quantified the proteomes of five biochemically isolated mouse brain cellular sub-fractions, with emphasis on synaptic compartments, from three brain regions, hippocampus, cortex and cerebellum. We demonstrated the expected co-fractionation of canonical synaptic proteins belonging to the same functional groups. The enrichment profiles also suggested the presence of many novel pre- and post-synaptic proteins. Using super-resolution microscopy on primary neuronal culture we confirmed the postsynaptic localization of PLEKHA5 and ADGRA1. We further detected profound brain region specific differences in the extent of enrichment for some functionally associated proteins. This is exemplified by different AMPA receptor subunits and substantial differences in sub-fraction distribution of their potential interactors, which implicated the differences of AMPA receptor complex compositions. This resource aids the identification of proteins partners and subcellular distribution of synaptic proteins.


Subject(s)
Brain Chemistry , Neurons/chemistry , Proteome/analysis , Proteomics/methods , Receptors, G-Protein-Coupled/metabolism , Synapses/chemistry , Animals , Cells, Cultured , Cerebellum/chemistry , Cerebral Cortex/chemistry , Hippocampus/chemistry , Intracellular Signaling Peptides and Proteins/analysis , Mice , Mice, Inbred C57BL , Neurons/cytology , Receptors, G-Protein-Coupled/analysis , Subcellular Fractions/chemistry , Tandem Mass Spectrometry/methods
10.
Proteomics ; 16(20): 2698-2705, 2016 10.
Article in English | MEDLINE | ID: mdl-27392515

ABSTRACT

The group 1 metabotropic glutamate receptors 1 and 5 (mGluR1/5) have been implicated in mechanisms of synaptic plasticity and may serve as potential therapeutic targets in autism spectrum disorders. The interactome of group 1 mGluRs has remained largely unresolved. Using a knockout-controlled interaction proteomics strategy we examined the mGluR5 protein complex in two brain regions, hippocampus and cortex, and identified mGluR1 as its major interactor in addition to the well described Homer proteins. We confirmed the presence of mGluR1/5 complex by (i) reverse immunoprecipitation using an mGluR1 antibody to pulldown mGluR5 from hippocampal tissue, (ii) coexpression in HEK293 cells followed by coimmunoprecipitation to reveal the direct interaction of mGluR1 and 5, and (iii) superresolution microscopy imaging of hippocampal primary neurons to show colocalization of the mGluR1/5 in the synapse.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , HEK293 Cells , Humans , Immunoprecipitation , Mice , Receptor, Metabotropic Glutamate 5/analysis , Receptors, Metabotropic Glutamate/analysis
11.
J Proteome Res ; 13(12): 5695-706, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25337787

ABSTRACT

Fast excitatory synaptic transmission in the brain is mediated by glutamate acting on postsynaptic AMPA receptors. Recent studies have revealed a substantial number of AMPA receptor auxiliary proteins, which potentially contribute to the regulation of AMPA receptor trafficking, subcellular receptor localization, and receptor gating properties. Here we examined the AMPA receptor interactomes from cortex, hippocampus, and cerebellum by comprehensive interaction proteomics. The study reveals that AMPA receptor auxiliary proteins are engaged in distinct brain region-specific AMPA receptors subcomplexes, which might underlie brain region-specific differential regulation of AMPA receptor properties. Depending on the brain region, an interacting protein can be involved in an AMPA and a non-AMPA receptor complex.


Subject(s)
Brain/metabolism , Multiprotein Complexes/metabolism , Proteomics/methods , Receptors, AMPA/metabolism , Animals , Blotting, Western , Carrier Proteins/metabolism , Cerebellum/metabolism , Cerebral Cortex/metabolism , Chromatography, Liquid , Hippocampus/metabolism , Immunoprecipitation , Mice, Inbred C57BL , Protein Binding , Protein Subunits/metabolism , Proteome/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...