Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Proteome Res ; 11(8): 4075-90, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22731840

ABSTRACT

Knowledge of the protein networks interacting with the amyloid precursor protein (APP) in vivo can shed light on the physiological function of APP. To date, most proteins interacting with the APP intracellular domain (AICD) have been identified by Yeast Two Hybrid screens which only detect direct interaction partners. We used a proteomics-based approach by biochemically isolating tagged APP from the brains of transgenic mice and subjecting the affinity-purified complex to mass spectrometric (MS) analysis. Using two different quantitative MS approaches, we compared the protein composition of affinity-purified samples isolated from wild-type mice versus transgenic mice expressing tagged APP. This enabled us to assess truly enriched proteins in the transgenic sample and yielded an overlapping set of proteins containing the major proteins involved in synaptic vesicle endo- and exocytosis. Confocal microscopy analyses of cotransfected primary neurons showed colocalization of APP with synaptic vesicle proteins in vesicular structures throughout the neurites. We analyzed the interaction of APP with these proteins using pulldown experiments from transgenic mice or cotransfected cells followed by Western blotting. Synaptotagmin-1 (Stg1), a resident synaptic vesicle protein, was found to directly bind to APP. We fused Citrine and Cerulean to APP and the candidate proteins and measured fluorescence resonance energy transfer (FRET) in differentiated SH-SY5Y cells. Differentially tagged APPs showed clear sensitized FRET emission, in line with the described dimerization of APP. Among the candidate APP-interacting proteins, again only Stg1 was in close proximity to APP. Our results strongly argue for a function of APP in synaptic vesicle turnover in vivo. Thus, in addition to the APP cleavage product Aß, which influences synaptic transmission at the postsynapse, APP interacts with the calcium sensor of synaptic vesicles and might thus play a role in the regulation of synaptic vesicle exocytosis.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Protein Interaction Maps , Proteome/metabolism , Synaptic Vesicles/metabolism , Synaptotagmin I/metabolism , Amyloid beta-Protein Precursor/isolation & purification , Animals , Chromatography, Affinity , Exocytosis , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Protein Interaction Mapping , Protein Transport , Proteome/isolation & purification
2.
Neurobiol Aging ; 31(1): 58-73, 2010 Jan.
Article in English | MEDLINE | ID: mdl-18403052

ABSTRACT

The beta-amyloid precursor protein (APP) plays a major role in Alzheimer's disease. The APP intracellular domain (AICD), together with Fe65 and Tip60, localizes to spherical nuclear AFT complexes, which may represent sites of transcription. Despite a lack of co-localization with several described nuclear compartments, we have identified a close apposition between AFT complexes and splicing speckles, Cajal bodies and PML bodies. Live imaging revealed that AFT complexes were highly mobile within nuclei and following pharmacological inhibition of transcription fused into larger assemblies. We have previously shown that AICD regulates the expression of its own precursor APP. In support of our earlier findings, transfection of APP promoter plasmids as substrates resulted in cytosolic AFT complex formation at labeled APP promoter plasmids. In addition, identification of chromosomal APP or KAI1 gene loci by fluorescence in situ hybridization showed their close association with nuclear AFT complexes. The transcriptional activator Notch intracellular domain (NICD) localized to the same nuclear spots as occupied by AFT complexes suggesting that these nuclear compartments correspond to transcription factories. Fe65 and Tip60 also co-localized with APP in the neurites of primary neurons. Pre-assembled AFT complexes may serve to assist fast nuclear signaling upon endoproteolytic APP cleavage.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Cell Nucleus/metabolism , Neurons/metabolism , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Brain/physiopathology , Cell Line , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Cells, Cultured , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , In Situ Hybridization, Fluorescence , Lysine Acetyltransferase 5 , Macromolecular Substances/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Plasmids , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary/physiology , Receptors, Notch/chemistry , Signal Transduction/physiology , Trans-Activators , Transcriptional Activation/physiology
3.
J Cell Sci ; 122(Pt 20): 3703-14, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19773363

ABSTRACT

Proteolytic processing of the amyloid precursor protein (APP) occurs via two alternative pathways, localized to different subcellular compartments, which result in functionally distinct outcomes. Cleavage by a beta-gamma sequence generates the Abeta peptide that plays a central role in Alzheimer's disease. In the case of alpha-gamma cleavage, a secreted neurotrophic molecule is generated and the Abeta peptide cleaved and destroyed. In both cases, a cytosolic APP intracellular domain (AICD) is generated. We have previously shown that coexpression of APP with the APP-binding protein Fe65 and the histone acetyltransferase Tip60 results in the formation of nuclear complexes (termed AFT complexes), which localize to transcription sites. We now show that blocking endocytosis or the pharmacological or genetic inhibition of the endosomal beta-cleavage pathway reduces translocation of AICD to these nuclear AFT complexes. AICD signaling further depends on active transport along microtubules and can be modulated by interference with both anterograde and retrograde transport systems. Nuclear signaling by endogenous AICD in primary neurons could similarly be blocked by inhibiting beta-cleavage but not by alpha-cleavage inhibition. This suggests that amyloidogenic cleavage, despite representing the minor cleavage pathway of APP, is predominantly responsible for AICD-mediated nuclear signaling.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Cell Nucleus/metabolism , Intracellular Space/metabolism , Protein Processing, Post-Translational , Signal Transduction , Active Transport, Cell Nucleus , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cell Line , Cell Membrane/metabolism , Endocytosis , Endosomes/metabolism , Gene Knockout Techniques , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Protein Transport
4.
Rapid Commun Mass Spectrom ; 19(5): 589-96, 2005.
Article in English | MEDLINE | ID: mdl-15685682

ABSTRACT

In liquid chromatography/tandem mass spectrometry (LC/MS/MS) analyses of complex peptide mixtures, dynamic exclusion functions are used to minimize repeat selections of identical precursors for collision induced dissociation (CID). We describe a new algorithm for the dynamic exclusion of m/z values during LC/MS/MS. Full-scan based peak exclusion (Fulspec) uses a simplified model of chromatographic peak formation to detect and exclude contaminants present throughout the run or that lead to broad peaks. Therefore, instead of excluding peptides from fragment analysis according to a rigidly predefined time window, the chromatographic properties of the detected analytes are used. The algorithm was tested on two datasets derived from previously published experiments. Fulspec achieves a distribution of CID spectra with minimal tailing on the retention time axis, without resorting to rigid exclusion of m/z values. The procedure further excludes intensities with a bias towards low-quality CID spectra. This combination frees up valuable analytical capacity. The underlying intensity vs. quality analyses challenge the assumption that abundant precursors automatically give the best identifications. Further validation of the algorithm will require its incorporation by equipment manufacturers into the instrument control programs.


Subject(s)
Algorithms , Chromatography, Liquid/methods , Complex Mixtures/analysis , Complex Mixtures/chemistry , Mass Spectrometry/methods , Pattern Recognition, Automated/methods , Peptides/analysis , Peptides/chemistry , Anatomy, Comparative , Sample Size
5.
J Cell Sci ; 117(Pt 19): 4435-48, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15331662

ABSTRACT

The physiological functions of the beta-amyloid precursor protein (APP) may include nuclear signaling. To characterize the role of the APP adaptor proteins Fe65, Jip1b, X11alpha (MINT1) and the chromatin-associated protein Tip60, we analyzed their interactions by confocal microscopy and co-immunoprecipitations. AICD corresponding to S3-cleaved APP bound to Fe65 that transported it to nuclei and docked it to Tip60. These proteins formed AICD-Fe65-Tip60 (AFT) complexes that were concentrated in spherical nuclear spots. gamma-Secretase inhibitors prevented AFT-complex formation with AICD derived from full-length APP. The APP adaptor protein Jip1b also transported AICD to nuclei and docked it to Tip60, but AICD-Jip1b-Tip60 (AJT) complexes had different, speckle-like morphology. By contrast, X11alpha trapped AICD in the cytosol. Induced AICD expression identified the APP-effector genes APP, BACE, Tip60, GSK3beta and KAI1, but not the Notch-effector gene Hes1 as transcriptional targets. These data establish a role for APP in nuclear signaling, and they suggest that therapeutic strategies designed to modulate the cleavage of APP affect AICD-dependent signaling.


Subject(s)
Active Transport, Cell Nucleus/physiology , Amyloid beta-Protein Precursor/metabolism , Cell Nucleus/metabolism , Multiprotein Complexes/metabolism , Transcription, Genetic/physiology , Acetyltransferases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , Basic Helix-Loop-Helix Transcription Factors , Cells, Cultured , Fluorescence Resonance Energy Transfer , Histone Acetyltransferases , Homeodomain Proteins/metabolism , Humans , Kangai-1 Protein , Lysine Acetyltransferase 5 , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , Transcription Factor HES-1
6.
Protein Expr Purif ; 28(2): 362-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12699702

ABSTRACT

Rubredoxin (Rub) from Thermotoga maritima, a 6.1-kDa red protein containing an Fe(III)-cysteine(4) center, was evaluated for its usefulness as a colored fusion tag for expression of recombinant proteins in E. coli. Here, we describe the Rub features relevant to accelerating screening for optimal high yield soluble expression conditions and automating the ensuing purification process. Spectroscopic properties and the yield of Rub fused to a typical target protein were compared to analogous GFP and Flavodoxin constructs, showing Rub absorption to be sufficient for structural genomics purposes while being produced at much higher soluble levels than GFP constructs. Based entirely on Rub absorption at 380 nm, both generic and affinity purification of crude cell lysate were performed: thus guided anion exchange purification of a Rub fusion construct as well as automated Ni-NTA purification resulted in pure protein. Rub is stable over a wide range of pH, temperature, and buffer environments, enabling robust purification protocols. Across a variety of fusion constructs, including N- and C-terminal Rub, quantitation via the Rub signal was shown to reliably correlate with analytical HPLC data obtained at 220 nm. We propose the "RubyTag" as an alternative to conventional protein fusion tags, as it combines a specific absorption signal with convenient biochemical and biological properties. Further, it allows direct on-line readout on conventional chromatography systems, holding promise for automated multi-step chromatography.


Subject(s)
Nitrilotriacetic Acid/analogs & derivatives , Recombinant Fusion Proteins/metabolism , Rubredoxins/metabolism , CD11 Antigens/genetics , CD11 Antigens/isolation & purification , CD11 Antigens/metabolism , Chromatography, High Pressure Liquid/methods , Chromatography, Ion Exchange/methods , Cloning, Molecular , Escherichia coli/genetics , Flavodoxin/genetics , Flavodoxin/isolation & purification , Flavodoxin/metabolism , Gene Expression , Green Fluorescent Proteins , Humans , Hydrogen-Ion Concentration , Luminescent Proteins/genetics , Luminescent Proteins/isolation & purification , Luminescent Proteins/metabolism , Nitrilotriacetic Acid/chemistry , Organometallic Compounds/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Rubredoxins/genetics , Rubredoxins/isolation & purification , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...