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










Publication year range
1.
J Neurogenet ; 34(1): 92-105, 2020 03.
Article in English | MEDLINE | ID: mdl-31965876

ABSTRACT

Amyloid precursor protein (APP), the precursor of amyloid beta peptide, plays a central role in Alzheimer's disease (AD), a pathology characterized by memory decline and synaptic loss upon aging. Understanding the physiological role of APP is fundamental in deciphering the progression of AD, and several studies suggest a synaptic function via protein-protein interactions. Nevertheless, it remains unclear whether and how these interactions contribute to memory. In Drosophila, we previously showed that APP-like (APPL), the fly APP homolog, is required for aversive associative memory in the olfactory memory center, the mushroom body (MB). In the present study, we show that APPL is required for appetitive long-term memory (LTM), another form of associative memory, in a specific neuronal subpopulation of the MB, the α'/ß' Kenyon cells. Using a biochemical approach, we identify the synaptic MAGUK (membrane-associated guanylate kinase) proteins X11, CASK, Dlgh2 and Dlgh4 as interactants of the APP intracellular domain (AICD). Next, we show that the Drosophila homologs CASK and Dlg are also required for appetitive LTM in the α'/ß' neurons. Finally, using a double RNAi approach, we demonstrate that genetic interactions between APPL and CASK, as well as between APPL and Dlg, are critical for appetitive LTM. In summary, our results suggest that APPL contributes to associative long-term memory through its interactions with the main synaptic scaffolding proteins CASK and Dlg. This function should be conserved across species.


Subject(s)
Appetitive Behavior/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Memory, Long-Term/physiology , Mushroom Bodies/physiology , Nerve Tissue Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Animals, Genetically Modified , Drosophila melanogaster/physiology
2.
Cell Rep ; 20(9): 2087-2099, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854360

ABSTRACT

Clathrin/adaptor protein-1-coated carriers connect the secretory and the endocytic pathways. Carrier biogenesis relies on distinct protein networks changing membrane shape at the trans-Golgi network, each regulating coat assembly, F-actin-based mechanical forces, or the biophysical properties of lipid bilayers. How these different hubs are spatiotemporally coordinated remains largely unknown. Using in vitro reconstitution systems, quantitative proteomics, and lipidomics, as well as in vivo cell-based assays, we characterize the protein networks controlling membrane lipid composition, membrane shape, and carrier scission. These include PIP5K1A and phospholipase C-beta 3 controlling the conversion of PI[4]P into diacylglycerol. PIP5K1A binding to RAC1 provides a link to F-actin-based mechanical forces needed to tubulate membranes. Tubular membranes then recruit the BAR-domain-containing arfaptin-1/2 guiding carrier scission. These findings provide a framework for synchronizing the chemical/biophysical properties of lipid bilayers, F-actin-based mechanical forces, and the activity of proteins sensing membrane shape during clathrin/adaptor protein-1-coated carrier biogenesis.


Subject(s)
Actins/metabolism , Adaptor Protein Complex 1/metabolism , Clathrin-Coated Vesicles/metabolism , Lipid Metabolism , Animals , Biomechanical Phenomena , Carrier Proteins/metabolism , Clathrin/metabolism , Diglycerides/biosynthesis , HeLa Cells , Humans , Mice , Phosphatidylinositol Phosphates/metabolism , Phospholipase C beta/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polymerization , rac1 GTP-Binding Protein/metabolism
3.
PLoS One ; 11(10): e0164829, 2016.
Article in English | MEDLINE | ID: mdl-27760174

ABSTRACT

Bone resorption in vertebrates relies on the ability of osteoclasts to assemble F-actin-rich podosomes that condense into podosomal belts, forming sealing zones. Sealing zones segregate bone-facing ruffled membranes from other membrane domains, and disassemble when osteoclasts migrate to new areas. How podosome/sealing zone dynamics is regulated remains unknown. We illustrate the essential role of the membrane scaffolding F-BAR-Proline-Serine-Threonine Phosphatase Interacting Proteins (PSTPIP) 1 and 2 in this process. Whereas PSTPIP2 regulates podosome assembly, PSTPIP1 regulates their disassembly. PSTPIP1 recruits, through its F-BAR domain, the protein tyrosine phosphatase non-receptor type 6 (PTPN6) that de-phosphophorylates the phosphatidylinositol 5-phosphatases SHIP1/2 bound to the SH3 domain of PSTPIP1. Depletion of any component of this complex prevents sealing zone disassembly and increases osteoclast activity. Thus, our results illustrate the importance of BAR domain proteins in podosome structure and dynamics, and identify a new PSTPIP1/PTPN6/SHIP1/2-dependent negative feedback mechanism that counterbalances Src and PI(3,4,5)P3 signalling to control osteoclast cell polarity and activity during bone resorption.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Cytoskeletal Proteins/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Podosomes/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Mice , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Protein Domains , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proteomics , RAW 264.7 Cells , RNA Interference
4.
Nat Commun ; 7: 13233, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27775035

ABSTRACT

The glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) facilitates fast axonal transport in neurons. However, given that GAPDH does not produce ATP, it is unclear whether glycolysis per se is sufficient to propel vesicles. Although many proteins regulating transport have been identified, the molecular composition of transported vesicles in neurons has yet to be fully elucidated. Here we selectively enrich motile vesicles and perform quantitative proteomic analysis. In addition to the expected molecular motors and vesicular proteins, we find an enrichment of all the glycolytic enzymes. Using biochemical approaches and super-resolution microscopy, we observe that most glycolytic enzymes are selectively associated with vesicles and facilitate transport of vesicles in neurons. Finally, we provide evidence that mouse brain vesicles produce ATP from ADP and glucose, and display movement in a reconstituted in vitro transport assay of native vesicles. We conclude that transport of vesicles along microtubules can be autonomous.


Subject(s)
Brain/metabolism , Energy Metabolism , Glycolysis , Neurons/metabolism , Transport Vesicles/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Mice , Mice, Transgenic , Microtubules/metabolism , Neurons/cytology , Proteome/metabolism , Proteomics/methods , Rats
5.
PLoS One ; 11(8): e0159824, 2016.
Article in English | MEDLINE | ID: mdl-27490675

ABSTRACT

Multipotent mesenchymal stromal cells (MSCs) are promising tools for regenerative medicine. They can be isolated from different sources based on their plastic-adherence property. The identification of reliable cell surface markers thus becomes the Holy Grail for their prospective isolation. Here, we determine the cell surface proteomes of human dental pulp-derived MSCs isolated from single donors after culture expansion in low (2%) or high (10%) serum-containing media. Cell surface proteins were tagged on intact cells using cell impermeable, cleavable sulfo-NHS-SS-biotin, which allows their enrichment by streptavidin pull-down. For the proteomic analyses, we first compared label-free methods to analyze cell surface proteomes i.e. composition, enrichment and proteomic differences, and we developed a new mathematical model to determine cell surface protein enrichment using a combinatorial gene ontology query. Using this workflow, we identified 101 cluster of differentiation (CD) markers and 286 non-CD cell surface proteins. Based on this proteome profiling, we identified 14 cell surface proteins, which varied consistently in abundance when cells were cultured under low or high serum conditions. Collectively, our analytical methods provide a basis for identifying the cell surface proteome of dental pulp stem cells isolated from single donors and its evolution during culture or differentiation. Our data provide a comprehensive cell surface proteome for the precise identification of dental pulp-derived MSC populations and their isolation for potential therapeutic intervention.


Subject(s)
Dental Pulp/metabolism , Mass Spectrometry , Proteome/analysis , Adolescent , Antigens, CD/analysis , Antigens, CD/genetics , Biotin/chemistry , Cells, Cultured , Dental Pulp/cytology , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Membrane Proteins/analysis , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Proteome/genetics , Proteomics , Real-Time Polymerase Chain Reaction , Young Adult
6.
PLoS One ; 10(6): e0130485, 2015.
Article in English | MEDLINE | ID: mdl-26125944

ABSTRACT

While the Amyloid Precursor Protein (APP) plays a central role in Alzheimer's disease, its cellular function still remains largely unclear. It was our goal to establish APP function which will provide insights into APP's implication in Alzheimer's disease. Using our recently developed proteo-liposome assay we established the interactome of APP's intracellular domain (known as AICD), thereby identifying novel APP interactors that provide mechanistic insights into APP function. By combining biochemical, cell biological and genetic approaches we validated the functional significance of one of these novel interactors. Here we show that APP binds the PIKfyve complex, an essential kinase for the synthesis of the endosomal phosphoinositide phosphatidylinositol-3,5-bisphosphate. This signalling lipid plays a crucial role in endosomal homeostasis and receptor sorting. Loss of PIKfyve function by mutation causes profound neurodegeneration in mammals. Using C. elegans genetics we demonstrate that APP functionally cooperates with PIKfyve in vivo. This regulation is required for maintaining endosomal and neuronal function. Our findings establish an unexpected role for APP in the regulation of endosomal phosphoinositide metabolism with dramatic consequences for endosomal biology and important implications for our understanding of Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Endosomes/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Neurons/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositols/metabolism , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vacuoles/metabolism
7.
J Biol Chem ; 289(26): 18347-59, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24821726

ABSTRACT

The initial step of bone digestion is the adhesion of osteoclasts onto bone surfaces and the assembly of podosomal belts that segregate the bone-facing ruffled membrane from other membrane domains. During bone digestion, membrane components of the ruffled border also need to be recycled after macropinocytosis of digested bone materials. How osteoclast polarity and membrane recycling are coordinated remains unknown. Here, we show that the Cdc42-guanine nucleotide exchange factor FGD6 coordinates these events through its Src-dependent interaction with different actin-based protein networks. At the plasma membrane, FGD6 couples cell adhesion and actin dynamics by regulating podosome formation through the assembly of complexes comprising the Cdc42-interactor IQGAP1, the Rho GTPase-activating protein ARHGAP10, and the integrin interactors Talin-1/2 or Filamin A. On endosomes and transcytotic vesicles, FGD6 regulates retromer-dependent membrane recycling through its interaction with the actin nucleation-promoting factor WASH. These results provide a mechanism by which a single Cdc42-exchange factor controlling different actin-based processes coordinates cell adhesion, cell polarity, and membrane recycling during bone degradation.


Subject(s)
Endosomes/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Membranes/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Animals , Bone and Bones/metabolism , Cell Adhesion , Cell Line , Cell Polarity , Guanine Nucleotide Exchange Factors/genetics , Mice , Protein Binding , cdc42 GTP-Binding Protein/metabolism
8.
Methods Enzymol ; 534: 223-43, 2014.
Article in English | MEDLINE | ID: mdl-24359957

ABSTRACT

Transport carriers regulate the bidirectional flow of membrane between the compartments of the secretory and endocytic pathways. Their biogenesis relies on the recruitment of a number of cytosolic proteins and protein complexes on specific membrane microdomains with defined protein and lipid compositions. The timely assembly of these cellular machines onto membranes involves multiple protein-protein and protein-lipid interactions and is necessary to select membrane proteins and lipids into nascent carriers, to bend the flat membrane of the donor compartment, to change the shape of this nascent carrier into a tubular-vesicular structure, and to operate its scission from the donor compartment. A challenge in this field of membrane cell biology has been to identify these machineries and to understand their precise function, in particular by studying their spatial and temporal dynamics during carrier biogenesis. During the past years, liposome-based synthetic biology fully recapitulating the fidelity of carrier biogenesis as seen in vivo has proved to be instrumental to identify these key cytosolic components using mass spectrometry and their dynamics using fluorescence microscopy. We describe here the methods to isolate on synthetic membranes the protein networks needed for carrier biogenesis, to identify them using label-free quantitative proteomics, and to visualize their dynamics on giant unilamellar vesicles.


Subject(s)
Cell Membrane/metabolism , Cytosol/metabolism , Golgi Apparatus/metabolism , Liposomes/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , Brain Chemistry , Cell Membrane/chemistry , Clathrin/genetics , Clathrin/metabolism , Cytosol/chemistry , Electrophoresis, Polyacrylamide Gel , Gene Expression , Golgi Apparatus/chemistry , Liposomes/chemistry , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/metabolism , Mass Spectrometry , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Peptides/chemistry , Phospholipids/chemistry , Phospholipids/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Staining and Labeling , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
9.
Nat Methods ; 10(8): 788-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23793238

ABSTRACT

A major obstacle in defining the exact role of extracellular matrix (ECM) in stem cell niches is the lack of suitable in vitro methods that recapitulate complex ECM microenvironments. Here we describe a methodology that permits reliable anchorage of native cell-secreted ECM to culture carriers. We validated our approach by fabricating two types of human bone marrow-specific ECM substrates that were robust enough to support human mesenchymal stem cells (MSCs) and hematopoietic stem and progenitor cells in vitro. We characterized the molecular composition, structural features and nanomechanical properties of the MSC-derived ECM preparations and demonstrated their ability to support expansion and differentiation of bone marrow stem cells. Our methodology enables the deciphering and modulation of native-like multicomponent ECMs of tissue-resident stem cells and will therefore prepare the ground for a more rational design of engineered stem cell niches.


Subject(s)
Bone Marrow Cells/physiology , Extracellular Matrix/physiology , Hematopoietic Stem Cells/physiology , Mesenchymal Stem Cells/physiology , Stem Cell Niche/physiology , Animals , Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Specific Pathogen-Free Organisms
10.
Curr Biol ; 21(13): 1111-7, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21700461

ABSTRACT

The evolutionarily conserved apical determinant Crumbs (Crb) is essential for maintaining apicobasal polarity and integrity of many epithelial tissues [1]. Crb levels are crucial for cell polarity and homeostasis, yet strikingly little is known about its trafficking or the mechanism of its apical localization. Using a newly established, liposome-based system described here, we determined Crb to be an interaction partner and cargo of the retromer complex. Retromer is essential for the retrograde transport of numerous transmembrane proteins from endosomes to the trans-Golgi network (TGN) and is conserved between plants, fungi, and animals [2]. We show that loss of retromer function results in a substantial reduction of Crb in Drosophila larvae, wing discs, and the follicle epithelium. Moreover, loss of retromer phenocopies loss of crb by preventing apical localization of key polarity molecules, such as atypical protein kinase C (aPKC) and Par6 in the follicular epithelium, an effect that can be rescued by overexpression of Crb. Additionally, loss of retromer results in multilayering of the follicular epithelium, indicating that epithelial integrity is severely compromised. Our data reveal a mechanism for Crb trafficking by retromer that is vital for maintaining Crb levels and localization. We also show a novel function for retromer in maintaining epithelial cell polarity.


Subject(s)
Cell Polarity , Drosophila Proteins/metabolism , Drosophila/metabolism , Epithelial Cells/cytology , Membrane Proteins/metabolism , Animals , Drosophila/cytology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Epithelial Cells/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation , Protein Structure, Tertiary , Protein Transport , RNA Interference , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/metabolism , trans-Golgi Network/metabolism
11.
PLoS One ; 6(5): e20399, 2011.
Article in English | MEDLINE | ID: mdl-21637820

ABSTRACT

BACKGROUND: Multipotent human mesenchymal stromal cells (hMSCs) are considered as promising biological tools for regenerative medicine. Their antibody-based isolation relies on the identification of reliable cell surface markers. METHODOLOGY/PRINCIPAL FINDINGS: To obtain a comprehensive view of the cell surface proteome of bone marrow-derived hMSCs, we have developed an analytical pipeline relying on cell surface biotinylation of intact cells using cell impermeable, cleavable sulfo-NHS-SS-biotin to enrich the plasma membrane proteins and mass spectrometry for identification with extremely high confidence. Among the 888 proteins identified, we found ≈200 bona fide plasma membrane proteins including 33 cell adhesion molecules and 26 signaling receptors. In total 41 CD markers including 5 novel ones (CD97, CD112, CD239, CD276, and CD316) were identified. The CD markers are distributed homogenously within plastic-adherent hMSC populations and their expression is modulated during the process of adipogenesis or osteogenesis. Moreover, our in silico analysis revealed a significant difference between the cell surface proteome of hMSCs and that of human embryonic stem cells reported previously. CONCLUSIONS/SIGNIFICANCE: Collectively, our analytical methods not only provide a basis for further studies of mechanisms maintaining the multipotency of hMSCs within their niches and triggering their differentiation after signaling, but also a toolbox for a refined antibody-based identification of hMSC populations from different tissues and their isolation for therapeutic intervention.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Proteome/metabolism , Adipogenesis , Adult , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Lineage , Embryonic Stem Cells/metabolism , Flow Cytometry , Humans , Membrane Proteins/isolation & purification , Osteogenesis , Proteome/isolation & purification , Stromal Cells/cytology , Stromal Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL