Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cancer Res ; 83(23): 3861-3867, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37668528

ABSTRACT

International cancer registries make real-world genomic and clinical data available, but their joint analysis remains a challenge. AACR Project GENIE, an international cancer registry collecting data from 19 cancer centers, makes data from >130,000 patients publicly available through the cBioPortal for Cancer Genomics (https://genie.cbioportal.org). For 25,000 patients, additional real-world longitudinal clinical data, including treatment and outcome data, are being collected by the AACR Project GENIE Biopharma Collaborative using the PRISSMM data curation model. Several thousand of these cases are now also available in cBioPortal. We have significantly enhanced the functionalities of cBioPortal to support the visualization and analysis of this rich clinico-genomic linked dataset, as well as datasets generated by other centers and consortia. Examples of these enhancements include (i) visualization of the longitudinal clinical and genomic data at the patient level, including timelines for diagnoses, treatments, and outcomes; (ii) the ability to select samples based on treatment status, facilitating a comparison of molecular and clinical attributes between samples before and after a specific treatment; and (iii) survival analysis estimates based on individual treatment regimens received. Together, these features provide cBioPortal users with a toolkit to interactively investigate complex clinico-genomic data to generate hypotheses and make discoveries about the impact of specific genomic variants on prognosis and therapeutic sensitivities in cancer. SIGNIFICANCE: Enhanced cBioPortal features allow clinicians and researchers to effectively investigate longitudinal clinico-genomic data from patients with cancer, which will improve exploration of data from the AACR Project GENIE Biopharma Collaborative and similar datasets.


Subject(s)
Genomics , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine
2.
Acta Neuropathol Commun ; 10(1): 100, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35799292

ABSTRACT

Frontotemporal dementia is characterized by progressive atrophy of frontal and/or temporal cortices at an early age of onset. The disorder shows considerable clinical, pathological, and genetic heterogeneity. Here we investigated the proteomic signatures of frontal and temporal cortex from brains with frontotemporal dementia due to GRN and MAPT mutations to identify the key cell types and molecular pathways in their pathophysiology. We compared patients with mutations in the GRN gene (n = 9) or with mutations in the MAPT gene (n = 13) with non-demented controls (n = 11). Using quantitative proteomic analysis on laser-dissected tissues we identified brain region-specific protein signatures for both genetic subtypes. Using published single cell RNA expression data resources we deduced the involvement of major brain cell types in driving these different protein signatures. Subsequent gene ontology analysis identified distinct genetic subtype- and cell type-specific biological processes. For the GRN subtype, we observed a distinct role for immune processes related to endothelial cells and for mitochondrial dysregulation in neurons. For the MAPT subtype, we observed distinct involvement of dysregulated RNA processing, oligodendrocyte dysfunction, and axonal impairments. Comparison with an in-house protein signature of Alzheimer's disease brains indicated that the observed alterations in RNA processing and oligodendrocyte function are distinct for the frontotemporal dementia MAPT subtype. Taken together, our results indicate the involvement of different brain cell types and biological mechanisms in genetic subtypes of frontotemporal dementia. Furthermore, we demonstrate that comparison of proteomic profiles of different disease entities can separate general neurodegenerative processes from disease-specific pathways, which may aid the development of disease subtype-specific treatment strategies.


Subject(s)
Frontotemporal Dementia , Pick Disease of the Brain , Endothelial Cells/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mutation/genetics , Progranulins/genetics , Proteomics , tau Proteins/genetics , tau Proteins/metabolism
3.
Neuron ; 103(2): 217-234.e4, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31171447

ABSTRACT

Synapses are fundamental information-processing units of the brain, and synaptic dysregulation is central to many brain disorders ("synaptopathies"). However, systematic annotation of synaptic genes and ontology of synaptic processes are currently lacking. We established SynGO, an interactive knowledge base that accumulates available research about synapse biology using Gene Ontology (GO) annotations to novel ontology terms: 87 synaptic locations and 179 synaptic processes. SynGO annotations are exclusively based on published, expert-curated evidence. Using 2,922 annotations for 1,112 genes, we show that synaptic genes are exceptionally well conserved and less tolerant to mutations than other genes. Many SynGO terms are significantly overrepresented among gene variations associated with intelligence, educational attainment, ADHD, autism, and bipolar disorder and among de novo variants associated with neurodevelopmental disorders, including schizophrenia. SynGO is a public, universal reference for synapse research and an online analysis platform for interpretation of large-scale -omics data (https://syngoportal.org and http://geneontology.org).


Subject(s)
Brain/cytology , Gene Ontology , Proteomics , Software , Synapses/physiology , Animals , Brain/physiology , Databases, Genetic , Humans , Knowledge Bases , Synaptic Potentials/physiology , Synaptosomes
4.
Acta Neuropathol Commun ; 6(1): 46, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29860944

ABSTRACT

Alzheimer's disease (AD) is characterized by amyloid beta (Aß) deposits as plaques in the parenchyma and in the walls of cortical and leptomeningeal blood vessels of the brain called cerebral amyloid angiopathy (CAA). It is suggested that CAA type-1, which refers to amyloid deposition in both capillaries and larger vessels, adds to the symptomatic manifestation of AD and correlates with disease severity. Currently, CAA cannot be diagnosed pre-mortem and disease mechanisms involved in CAA are elusive. To obtain insight in the disease mechanism of CAA and to identify marker proteins specifically associated with CAA we performed a laser dissection microscopy assisted mass spectrometry analysis of post-mortem human brain tissue of (I) AD cases with only amyloid deposits in the brain parenchyma and no vascular related amyloid, (II) AD cases with severe CAA type-1 and no or low numbers of parenchymal amyloid deposits and (III) cognitively healthy controls without amyloid deposits. By contrasting the quantitative proteomics data between the three groups, 29 potential CAA-selective proteins were identified. A selection of these proteins was analysed by immunoblotting and immunohistochemistry to confirm regulation and to determine protein localization and their relation to brain pathology. In addition, specificity of these markers in relation to other small vessel diseases including prion CAA, CADASIL, CARASAL and hypertension related small vessel disease was assessed using immunohistochemistry.Increased levels of clusterin (CLU), apolipoprotein E (APOE) and serum amyloid P-component (APCS) were observed in AD cases with CAA. In addition, we identified norrin (NDP) and collagen alpha-2(VI) (COL6A2) as highly selective markers that are clearly present in CAA yet virtually absent in relation to parenchymal amyloid plaque pathology. NDP showed the highest specificity to CAA when compared to other small vessel diseases. The specific changes in the proteome of CAA provide new insight in the pathogenesis and yields valuable selective biomarkers for the diagnosis of CAA.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/etiology , Cerebral Amyloid Angiopathy/metabolism , Proteomics , Aged , Aged, 80 and over , Apolipoproteins E/blood , Brain/metabolism , Cerebral Amyloid Angiopathy/blood , Clusterin/blood , Female , Humans , Laser Capture Microdissection , Male , Mass Spectrometry , Serum Amyloid P-Component/metabolism , Statistics, Nonparametric
5.
PLoS One ; 13(5): e0196979, 2018.
Article in English | MEDLINE | ID: mdl-29787571

ABSTRACT

To identify coding and non-coding suppressor genes of anchorage-independent proliferation by efficient loss-of-function screening, we have developed a method for enzymatic production of low complexity shRNA libraries from subtracted transcriptomes. We produced and screened two LEGO (Low-complexity by Enrichment for Genes shut Off) shRNA libraries that were enriched for shRNA vectors targeting coding and non-coding polyadenylated transcripts that were reduced in transformed Mouse Embryonic Fibroblasts (MEFs). The LEGO shRNA libraries included ~25 shRNA vectors per transcript which limited off-target artifacts. Our method identified 79 coding and non-coding suppressor transcripts. We found that taurine-responsive GABAA receptor subunits, including GABRA5 and GABRB3, were induced during the arrest of non-transformed anchor-deprived MEFs and prevented anchorless proliferation. We show that taurine activates chloride currents through GABAA receptors on MEFs, causing seclusion of cell volume in large membrane protrusions. Volume seclusion from cells by taurine correlated with reduced proliferation and, conversely, suppression of this pathway allowed anchorage-independent proliferation. In human cholangiocarcinomas, we found that several proteins involved in taurine signaling via GABAA receptors were repressed. Low GABRA5 expression typified hyperproliferative tumors, and loss of taurine signaling correlated with reduced patient survival, suggesting this tumor suppressive mechanism operates in vivo.


Subject(s)
Cholangiocarcinoma/pathology , Embryo, Mammalian/pathology , Receptors, GABA-A/metabolism , Taurine/pharmacology , Transcriptome/drug effects , Animals , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cell Proliferation/drug effects , Cells, Cultured , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Humans , Mice , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Receptors, GABA-A/genetics , Signal Transduction/drug effects , Subtractive Hybridization Techniques , Survival Rate
6.
Sci Transl Med ; 9(421)2017 Dec 20.
Article in English | MEDLINE | ID: mdl-29263233

ABSTRACT

Patients with depression often suffer from cognitive impairments that contribute to disease burden. We used social defeat-induced persistent stress (SDPS) to induce a depressive-like state in rats and then studied long-lasting memory deficits in the absence of acute stressors in these animals. The SDPS rat model showed reduced short-term object location memory and maintenance of long-term potentiation (LTP) in CA1 pyramidal neurons of the dorsal hippocampus. SDPS animals displayed increased expression of synaptic chondroitin sulfate proteoglycans in the dorsal hippocampus. These effects were abrogated by a 3-week treatment with the antidepressant imipramine starting 8 weeks after the last defeat encounter. Next, we observed an increase in the number of perineuronal nets (PNNs) surrounding parvalbumin-expressing interneurons and a decrease in the frequency of inhibitory postsynaptic currents (IPSCs) in the hippocampal CA1 region in SDPS animals. In vivo breakdown of the hippocampus CA1 extracellular matrix by the enzyme chondroitinase ABC administered intracranially restored the number of PNNs, LTP maintenance, hippocampal inhibitory tone, and memory performance on the object place recognition test. Our data reveal a causal link between increased hippocampal extracellular matrix and the cognitive deficits associated with a chronic depressive-like state in rats exposed to SDPS.


Subject(s)
Cognitive Dysfunction/pathology , Depression/pathology , Extracellular Matrix/metabolism , Hippocampus/pathology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Chondroitin ABC Lyase/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Chronic Disease , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Depression/complications , Depression/drug therapy , Depression/physiopathology , Hippocampus/drug effects , Hippocampus/physiopathology , Imipramine/pharmacology , Imipramine/therapeutic use , Interneurons/drug effects , Interneurons/pathology , Male , Memory/drug effects , Neural Inhibition/drug effects , Rats, Wistar , Social Behavior , Stress, Psychological/drug therapy , Stress, Psychological/etiology , Stress, Psychological/physiopathology , Synapses/drug effects , Synapses/metabolism
7.
Front Aging Neurosci ; 9: 138, 2017.
Article in English | MEDLINE | ID: mdl-28567012

ABSTRACT

Aging is the most important risk factor for neurodegenerative diseases associated with pathological protein aggregation such as Alzheimer's disease. Although aging is an important player, it remains unknown which molecular changes are relevant for disease initiation. Recently, it has become apparent that widespread protein aggregation is a common feature of aging. Indeed, several studies demonstrate that 100s of proteins become highly insoluble with age, in the absence of obvious disease processes. Yet it remains unclear how these misfolded proteins aggregating with age affect neurodegenerative diseases. Importantly, several of these aggregation-prone proteins are found as minor components in disease-associated hallmark aggregates such as amyloid-ß plaques or neurofibrillary tangles. This co-localization raises the possibility that age-dependent protein aggregation directly contributes to pathological aggregation. Here, we show for the first time that highly insoluble proteins from aged Caenorhabditis elegans or aged mouse brains, but not from young individuals, can initiate amyloid-ß aggregation in vitro. We tested the seeding potential at four different ages across the adult lifespan of C. elegans. Significantly, protein aggregates formed during the early stages of aging did not act as seeds for amyloid-ß aggregation. Instead, we found that changes in protein aggregation occurring during middle-age initiated amyloid-ß aggregation. Mass spectrometry analysis revealed several late-aggregating proteins that were previously identified as minor components of amyloid-ß plaques and neurofibrillary tangles such as 14-3-3, Ubiquitin-like modifier-activating enzyme 1 and Lamin A/C, highlighting these as strong candidates for cross-seeding. Overall, we demonstrate that widespread protein misfolding and aggregation with age could be critical for the initiation of pathogenesis, and thus should be targeted by therapeutic strategies to alleviate neurodegenerative diseases.

8.
Sci Rep ; 6: 35456, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27748445

ABSTRACT

Development of the brain involves the formation and maturation of numerous synapses. This process requires prominent changes of the synaptic proteome and potentially involves thousands of different proteins at every synapse. To date the proteome analysis of synapse development has been studied sparsely. Here, we analyzed the cortical synaptic membrane proteome of juvenile postnatal days 9 (P9), P15, P21, P27, adolescent (P35) and different adult ages P70, P140 and P280 of C57Bl6/J mice. Using a quantitative proteomics workflow we quantified 1560 proteins of which 696 showed statistically significant differences over time. Synaptic proteins generally showed increased levels during maturation, whereas proteins involved in protein synthesis generally decreased in abundance. In several cases, proteins from a single functional molecular entity, e.g., subunits of the NMDA receptor, showed differences in their temporal regulation, which may reflect specific synaptic development features of connectivity, strength and plasticity. SNARE proteins, Snap 29/47 and Stx 7/8/12, showed higher expression in immature animals. Finally, we evaluated the function of Cxadr that showed high expression levels at P9 and a fast decline in expression during neuronal development. Knock down of the expression of Cxadr in cultured primary mouse neurons revealed a significant decrease in synapse density.


Subject(s)
Brain/metabolism , Cerebral Cortex/metabolism , Proteome , Proteomics , Synapses/metabolism , Animals , Brain/growth & development , Cell Adhesion Molecules/metabolism , Cerebral Cortex/growth & development , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Protein Interaction Mapping , Protein Interaction Maps , Proteomics/methods
9.
Anal Chem ; 88(9): 4825-32, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27046509

ABSTRACT

This study describes a new platform for the fast and efficient functional screening for bioactive compounds in complex natural mixtures using a cell-based assay. The platform combines reversed-phase liquid chromatography (LC) with online flow cytometry (FC) and mass spectrometry (MS). As a model (an example or proof-of-concept study) we have used a functional calcium-flux assay in human neuroblastoma SH-SY5Y cells stably overexpressing the α-7 nicotinic acetylcholine receptor (α7-nAChR), a potential therapeutic target for central nervous system (CNS) related diseases. We have designed the coupled LC-FC system employing the neuroblastoma cells followed by analytical and pharmacological evaluation of the hyphenated setup in agonist and mixed antagonist-agonist assay modes. Using standard receptor ligands we have validated pharmacological responses and standardized good assay quality parameters. The applicability of the screening system was evaluated by analysis of various types of natural samples, such as a tobacco plant extract (in agonist assay mode) and snake venoms (in mixed antagonist-agonist assay mode). The bioactivity responses were correlated directly to the respective accurate masses of the compounds. Using simultaneous functional agonist and antagonist responses nicotine and known neurotoxins were detected from tobacco extract and snake venoms, respectively. Thus, the developed analytical screening technique represents a new tool for rapid measurement of functional cell-based responses and parallel separation and identification of compounds in complex mixtures targeting the α7-nAChR. It is anticipated that other fast-response cell-based assays (e.g., other ion flux assays) can be incorporated in this analytical setup.


Subject(s)
Biological Assay/methods , Chromatography, Liquid , Flow Cytometry , Mass Spectrometry , Online Systems , Humans , Tumor Cells, Cultured
10.
Alzheimers Dement ; 12(6): 654-68, 2016 06.
Article in English | MEDLINE | ID: mdl-26772638

ABSTRACT

INTRODUCTION: We performed a comprehensive quantitative proteomics study on human hippocampus tissue involving all Braak stages to assess changes in protein abundance over the various stages of Alzheimer's disease (AD). METHODS: Hippocampal subareas CA1 and subiculum of 40 cases were isolated using laser capture microdissection and analyzed using mass spectrometry. Immunoblotting and immunohistochemistry were used for validation. RESULTS: Over the Braak stages, an altered expression was found for 372 proteins including changes in levels of extracellular matrix components, and in calcium-dependent signaling proteins. Early changes were observed in levels of proteins related to cytoskeletal dynamics and synaptic components including an increase in RIMS1 and GRIK4. Several synaptic proteins, such as BSN, LIN7A, DLG2, -3, and -4, exhibit an early-up, late-down expression pattern. DISCUSSION: This study provides new insight into AD-dependent changes in protein levels in the hippocampus during AD pathology, identifying potential novel therapeutic targets and biomarkers.


Subject(s)
Alzheimer Disease/pathology , CA1 Region, Hippocampal/metabolism , Proteome/metabolism , Aged , Aged, 80 and over , Extracellular Matrix Proteins/metabolism , Female , Humans , Laser Capture Microdissection , Male , Mass Spectrometry , Middle Aged , Nerve Tissue Proteins/metabolism , Proteomics , Psychiatric Status Rating Scales , Signal Transduction
11.
J Cell Biol ; 211(3): 569-86, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26527743

ABSTRACT

Synaptic plasticity requires remodeling of the actin cytoskeleton. Although two actin isoforms, ß- and γ-actin, are expressed in dendritic spines, the specific contribution of γ-actin in the expression of synaptic plasticity is unknown. We show that synaptic γ-actin levels are regulated by the E3 ubiquitin ligase TRIM3. TRIM3 protein and Actg1 transcript are colocalized in messenger ribonucleoprotein granules responsible for the dendritic targeting of messenger RNAs. TRIM3 polyubiquitylates γ-actin, most likely cotranslationally at synaptic sites. Trim3(-/-) mice consequently have increased levels of γ-actin at hippocampal synapses, resulting in higher spine densities, increased long-term potentiation, and enhanced short-term contextual fear memory consolidation. Interestingly, hippocampal deletion of Actg1 caused an increase in long-term fear memory. Collectively, our findings suggest that temporal control of γ-actin levels by TRIM3 is required to regulate the timing of hippocampal plasticity. We propose a model in which TRIM3 regulates synaptic γ-actin turnover and actin filament stability and thus forms a transient inhibitory constraint on the expression of hippocampal synaptic plasticity.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Hippocampus/metabolism , Neuronal Plasticity/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Actin Cytoskeleton/metabolism , Animals , Dendritic Spines/metabolism , Long-Term Potentiation/physiology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synapses/metabolism
12.
J Biol Chem ; 290(25): 15635-15645, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25911096

ABSTRACT

The inbred strains C57BL/6J and DBA/2J (DBA) display striking differences in a number of behavioral tasks depending on hippocampal function, such as contextual memory. Historically, this has been explained through differences in postsynaptic protein expression underlying synaptic transmission and plasticity. We measured the synaptic hippocampal protein content (iTRAQ (Isobaric Tags for Relative and Absolute Quantitation) and mass spectrometry), CA1 synapse ultrastructural morphology, and synaptic functioning in adult C57BL/6J and DBA mice. DBA mice showed a prominent decrease in the Ras-GAP calcium-sensing protein RASAL1. Furthermore, expression of several presynaptic markers involved in exocytosis, such as syntaxin (Stx1b), Ras-related proteins (Rab3a/c), and rabphilin (Rph3a), was reduced. Ultrastructural analysis of CA1 hippocampal synapses showed a significantly lower number of synaptic vesicles and presynaptic cluster size in DBA mice, without changes in postsynaptic density or active zone. In line with this compromised presynaptic morphological and molecular phenotype in DBA mice, we found significantly lower paired-pulse facilitation and enhanced short term depression of glutamatergic synapses, indicating a difference in transmitter release and/or refilling mechanisms. Taken together, our data suggest that in addition to strain-specific postsynaptic differences, the change in dynamic properties of presynaptic transmitter release may underlie compromised synaptic processing related to cognitive functioning in DBA mice.


Subject(s)
Cognition/physiology , Hippocampus , Memory/physiology , Nerve Tissue Proteins/metabolism , Post-Synaptic Density , Proteome/metabolism , Animals , Hippocampus/physiology , Hippocampus/ultrastructure , Mice , Mice, Inbred DBA , Proteome/physiology , Proteome/ultrastructure , Proteomics , Species Specificity
13.
Hippocampus ; 25(11): 1250-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25708624

ABSTRACT

A change in efficacy of hippocampal synapses is critical for memory formation. So far, the molecular analysis of synapses during learning has focused on small groups of proteins, whereas the dynamic global changes at these synapses have remained unknown. Here, we analyzed the temporal changes of the mouse hippocampal synaptic membrane proteome 1 and 4 h after contextual fear learning, comparing two groups; (1) a fear memory forming "delayed-shock" group and (2) a fear memory-deficient "immediate-shock" group. No changes in protein expression were observed 1 h after conditioning between the two experimental groups. However, 423 proteins were significantly regulated 4 h later of which 164 proteins showed a temporal regulation after a delayed shock and 273 proteins after the stress of an immediate shock. From the proteins that were differentially regulated between the delayed- and the immediate-shock groups at 4 h, 48 proteins, most prominently representing endocytosis, (amphiphysin, dynamin, and synaptojanin1), glutamate signaling (glutamate [NMDA] receptor subunit epsilon-1, disks large homolog 3), and neurotransmitter metabolism (excitatory amino acid transporter 1, excitatory amino acid transporter 2, sodium- and chloride-dependent GABA transporter 3) were regulated in both protocols, but in opposite directions, pointing toward an interaction of learning and stress. Taken together, this data set yields novel insight into diverse and dynamic changes that take place at hippocampal synapses over the time course of contextual fear-memory learning.


Subject(s)
Behavior, Animal/physiology , Conditioning, Psychological/physiology , Fear/physiology , Hippocampus/metabolism , Proteome/metabolism , Stress, Psychological/metabolism , Synaptic Membranes/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Time Factors
14.
J Proteome Res ; 13(12): 5918-27, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25308431

ABSTRACT

Astrocytes are being increasingly recognized as crucial contributors to neuronal function at synapses, axons, and somas. Reliable methods that can provide insight into astrocyte proteins at the neuron-astrocyte functional interface are highly desirable. Here, we conducted a mass spectrometry analysis of Percoll gradient-isolated gliosomes, a viable preparation of glial subcellular particles often used to study mechanisms of astrocytic transmitter uptake and release and their regulation. Gliosomes were compared with synaptosomes, a preparation containing the neurotransmitter release machinery, and, accordingly, synaptosomes were enriched for proteins involved in synaptic vesicle-mediated transport. Interestingly, gliosome preparations were found to be enriched for different classes of known astrocyte proteins, such as VAMP3 (involved in astrocyte exocytosis), Ezrin (perisynaptic astrocyte cytoskeletal protein), and Basigin (astrocyte membrane glycoprotein), as well as for G-protein-mediated signaling proteins. Mass spectrometry data are available via ProteomeXchange with the identifier PXD001375. Together, these data provide the first detailed description of the gliosome proteome and show that gliosomes can be a useful preparation to study glial membrane proteins and associated processes.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Membrane Proteins/metabolism , Proteome/metabolism , Animals , Brain/cytology , Gene Ontology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proteome/genetics , Proteomics , Synaptosomes/metabolism , Tandem Mass Spectrometry
15.
Mol Cell Proteomics ; 13(11): 2975-85, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25044018

ABSTRACT

Age-related cognitive decline is a serious health concern in our aging society. Decreased cognitive function observed during healthy brain aging is most likely caused by changes in brain connectivity and synaptic dysfunction in particular brain regions. Here we show that aged C57BL/6J wild-type mice have hippocampus-dependent spatial memory impairments. To identify the molecular mechanisms that are relevant to these memory deficits, we investigated the temporal profile of mouse hippocampal synaptic proteome changes at 20, 40, 50, 60, 70, 80, 90, and 100 weeks of age. Extracellular matrix proteins were the only group of proteins that showed robust and progressive up-regulation over time. This was confirmed by immunoblotting and histochemical analysis, which indicated that the increased levels of hippocampal extracellular matrix might limit synaptic plasticity as a potential cause of age-related cognitive decline. In addition, we observed that stochasticity in synaptic protein expression increased with age, in particular for proteins that were previously linked with various neurodegenerative diseases, whereas low variance in expression was observed for proteins that play a basal role in neuronal function and synaptic neurotransmission. Together, our findings show that both specific changes and increased variance in synaptic protein expression are associated with aging and may underlie reduced synaptic plasticity and impaired cognitive performance in old age.


Subject(s)
Cognitive Dysfunction/physiopathology , Extracellular Matrix Proteins/metabolism , Hippocampus/physiology , Maze Learning/physiology , Memory Disorders/physiopathology , Aging/physiology , Animals , Cognition/physiology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/biosynthesis , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neuronal Plasticity/physiology , Proteome/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stochastic Processes , Tandem Mass Spectrometry
16.
Acta Neuropathol Commun ; 2: 76, 2014 Jun 29.
Article in English | MEDLINE | ID: mdl-24974208

ABSTRACT

Alzheimer's disease is caused by increased production or reduced clearance of amyloid-ß, which results in the formation amyloid-ß plaques and triggers a cascade of downstream events leading to progressive neurodegeneration. The earliest clinical symptoms of Alzheimer's disease, i.e., memory loss, are however poorly understood from a molecular and cellular perspective. Here we used APPswe/PS1dE9 (APP/PS1) transgenic mice to study the early pre-pathological effects of increased amyloid-ß levels on hippocampal synaptic plasticity and memory. Using an unbiased proteomics approach we show that the early increase in amyloid-ß levels in APP/PS1 mice at three months of age coincides with a robust and significant upregulation of several protein components of the extracellular matrix in hippocampal synaptosome preparations. This increase in extracellular matrix levels occurred well before the onset of plaque formation and was paralleled by impairments in hippocampal long-term potentiation and contextual memory. Direct injection into the hippocampus of the extracellular matrix inactivating enzyme chondroitinase ABC restored both long-term potentiation and contextual memory performance. These findings indicate an important role for the extracellular matrix in causing early memory loss in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Hippocampus/metabolism , Memory Disorders/metabolism , Alzheimer Disease/pathology , Animals , Chondroitin ABC Lyase/pharmacology , Conditioning, Psychological , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Humans , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid , Proteomics
17.
PLoS One ; 9(2): e87360, 2014.
Article in English | MEDLINE | ID: mdl-24498314

ABSTRACT

Shisa9 (initially named CKAMP44) has been identified as auxiliary subunit of the AMPA-type glutamate receptors and was shown to modulate its physiological properties. Shisa9 is a type-I transmembrane protein and contains a C-terminal PDZ domain that potentially interacts with cytosolic proteins. In this study, we performed a yeast two-hybrid screening that yielded eight PDZ domain-containing interactors of Shisa9, which were independently validated. The identified interactors are known scaffolding proteins residing in the neuronal postsynaptic density. To test whether C-terminal scaffolding interactions of Shisa9 affect synaptic AMPA receptor function in the hippocampus, we disrupted these interactions using a Shisa9 C-terminal mimetic peptide. In the absence of scaffolding interactions of Shisa9, glutamatergic AMPA receptor-mediated synaptic currents in the lateral perforant path of the mouse hippocampus had a faster decay time, and paired-pulse facilitation was reduced. Furthermore, disruption of the PDZ interactions between Shisa9 and its binding partners affected hippocampal network activity. Taken together, our data identifies novel interaction partners of Shisa9, and shows that the C-terminal interactions of Shisa9 through its PDZ domain interaction motif are important for AMPA receptor synaptic and network functions.


Subject(s)
Amino Acid Motifs , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Receptors, AMPA/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , HEK293 Cells , Hippocampus/metabolism , Hippocampus/physiology , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Net/physiology , Nerve Tissue Proteins/genetics , Neuronal Plasticity/physiology , Peptides/genetics , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, AMPA/genetics , Synapses/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Two-Hybrid System Techniques
18.
Nucleic Acids Res ; 41(Database issue): D834-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23143269

ABSTRACT

We created SynSysNet, available online at http://bioinformatics.charite.de/synsysnet, to provide a platform that creates a comprehensive 4D network of synaptic interactions. Neuronal synapses are fundamental structures linking nerve cells in the brain and they are responsible for neuronal communication and information processing. These processes are dynamically regulated by a network of proteins. New developments in interaction proteomics and yeast two-hybrid methods allow unbiased detection of interactors. The consolidation of data from different resources and methods is important to understand the relation to human behaviour and disease and to identify new therapeutic approaches. To this end, we established SynSysNet from a set of ∼1000 synapse specific proteins, their structures and small-molecule interactions. For two-thirds of these, 3D structures are provided (from Protein Data Bank and homology modelling). Drug-target interactions for 750 approved drugs and 50 000 compounds, as well as 5000 experimentally validated protein-protein interactions, are included. The resulting interaction network and user-selected parts can be viewed interactively and exported in XGMML. Approximately 200 involved pathways can be explored regarding drug-target interactions. Homology-modelled structures are downloadable in Protein Data Bank format, and drugs are available as MOL-files. Protein-protein interactions and drug-target interactions can be viewed as networks; corresponding PubMed IDs or sources are given.


Subject(s)
Databases, Protein , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Protein Interaction Mapping , Synapses/drug effects , Synapses/metabolism , Humans , Internet , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Conformation , User-Computer Interface
19.
J Proteome Res ; 11(3): 1855-67, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22289077

ABSTRACT

Cognitive decline is one of the earliest hallmarks of both normal and pathological brain aging. Here we used Ercc1 mutant mice, which are impaired in multiple DNA repair systems and consequently show accelerated aging and progressive memory deficits, to identify changes in the levels of hippocampal synaptic proteins that potentially underlie these age-dependent deficits. Aged Ercc1 mutant mice show normal gross hippocampal dendritic morphology and synapse numbers, and Ercc1 mutant hippocampal neurons displayed normal outgrowth and synapse formation in vitro. However, using isobaric tag for relative and absolute quantification (iTRAQ) of hippocampal synaptic proteins at two different ages, postnatal days 28 and 112, we observed a progressive decrease in synaptic ionotropic glutamate receptor levels and increased levels of G-proteins and of cell adhesion proteins. These together may cause long-term changes in synapse function. In addition, we observed a downregulation of mitochondrial proteins and concomitant upregulation of Na,K-ATPase subunits, which might compensate for reduced mitochondrial activity. Thus, our findings show that under conditions of apparent intact neuronal connectivity, levels of specific synaptic proteins are already affected during the early stages of DNA damage-induced aging, which might contribute to age-dependent cognitive decline.


Subject(s)
Aging/metabolism , DNA Repair , DNA-Binding Proteins/genetics , Endonucleases/genetics , Nerve Tissue Proteins/metabolism , Proteome/metabolism , Synapses/metabolism , Aging/genetics , Animals , Cells, Cultured , Cognition Disorders/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/physiology , Proteome/genetics , Synapses/pathology
20.
Mol Cell Proteomics ; 10(5): M110.005413, 2011 May.
Article in English | MEDLINE | ID: mdl-21398567

ABSTRACT

During brain development, the neocortex shows periods of enhanced plasticity, which enables the acquisition of knowledge and skills that we use and build on in adult life. Key to persistent modifications of neuronal connectivity and plasticity of the neocortex are molecular changes occurring at the synapse. Here we used isobaric tag for relative and absolute quantification to measure levels of 467 synaptic proteins in a well-established model of plasticity in the mouse visual cortex and the regulation of its critical period. We found that inducing visual cortex plasticity by monocular deprivation during the critical period increased levels of kinases and proteins regulating the actin-cytoskeleton and endocytosis. Upon closure of the critical period with age, proteins associated with transmitter vesicle release and the tubulin- and septin-cytoskeletons increased, whereas actin-regulators decreased in line with augmented synapse stability and efficacy. Maintaining the visual cortex in a plastic state by dark rearing mice into adulthood only partially prevented these changes and increased levels of G-proteins and protein kinase A subunits. This suggests that in contrast to the general belief, dark rearing does not simply delay cortical development but may activate signaling pathways that specifically maintain or increase the plasticity potential of the visual cortex. Altogether, this study identified many novel candidate plasticity proteins and signaling pathways that mediate synaptic plasticity during critical developmental periods or restrict it in adulthood.


Subject(s)
Neuronal Plasticity , Proteome/metabolism , Synapses/metabolism , Visual Cortex/growth & development , Visual Cortex/physiology , Age Factors , Animals , Clathrin/genetics , Clathrin/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Darkness , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Proteome/genetics , Sensory Deprivation , Synapses/genetics , Vision, Monocular/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...