Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Front Mol Neurosci ; 10: 87, 2017.
Article in English | MEDLINE | ID: mdl-28424586

ABSTRACT

Understanding the molecular mechanisms underlying amyloid precursor protein family (APP/APP-like proteins, APLP) function in the nervous system can be achieved by studying the APP/APLP interactome. In this review article, we focused on intracellular APP interacting proteins that bind the YENPTY internalization motif located in the last 15 amino acids of the C-terminal region. These proteins, which include X11/Munc-18-interacting proteins (Mints) and FE65/FE65Ls, represent APP cytosolic binding partners exhibiting different neuronal functions. A comparison of FE65 and APP family member mutant mice revealed a shared function for APP/FE65 protein family members in neurogenesis and neuronal positioning. Accumulating evidence also supports a role for membrane-associated APP/APLP proteins in synapse formation and function. Therefore, it is tempting to speculate that APP/APLP C-terminal interacting proteins transmit APP/APLP-dependent signals at the synapse. Herein, we compare our current knowledge of the synaptic phenotypes of APP/APLP mutant mice with those of mice lacking different APP/APLP interaction partners and discuss the possible downstream effects of APP-dependent FE65/FE65L or X11/Mint signaling on synaptic vesicle release, synaptic morphology and function. Given that the role of X11/Mint proteins at the synapse is well-established, we propose a model highlighting the role of FE65 protein family members for transduction of APP/APLP physiological function at the synapse.

2.
Sci Rep ; 6: 25652, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27734846

ABSTRACT

The FE65 adaptor proteins (FE65, FE65L1 and FE65L2) bind proteins that function in diverse cellular pathways and are essential for specific biological processes. Mice lacking both FE65 and FE65L1 exhibit ectopic neuronal positioning in the cortex and muscle weakness. p97FE65-KO mice, expressing a shorter FE65 isoform able to bind amyloid precursor protein family members (APP, APLP1, APLP2), develop defective long-term potentiation (LTP) and aged mice display spatial learning and memory deficits that are absent from young mice. Here, we examined the central and peripheral nervous systems of FE65-KO, FE65L1-KO and FE65/FE65L1-DKO mice. We find spatial learning and memory deficits in FE65-KO and FE65L1-KO mice. Severe motor impairments, anxiety, hippocampal LTP deficits and neuromuscular junction (NMJ) abnormalities, characterized by decreased size and reduced apposition of pre- and postsynaptic sites, are observed in FE65/FE65L1-DKO mice. As their NMJ deficits resemble those of mutant APP/APLP2-DKO mice lacking the FE65/FE65L1 binding site, the NMJs of APLP2/FE65-DKO and APLP2/FE65L1-DKO mice were analyzed. NMJ deficits are aggravated in these mice when compared to single FE65- and FE65L1-KO mice. Together, our data demonstrate a role for FE65 proteins at central and peripheral synapses possibly occurring downstream of cell surface-associated APP/APLPs.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Carrier Proteins/metabolism , Epistasis, Genetic , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Nuclear Proteins/metabolism , Synapses/metabolism , Adaptor Proteins, Signal Transducing , Animals , Anxiety , Dendritic Spines/metabolism , Genotype , Hippocampus/metabolism , Hippocampus/physiopathology , Learning , Long-Term Potentiation , Male , Maze Learning , Memory Disorders/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Motor Activity , Neuromuscular Junction/physiopathology , Pyramidal Cells/metabolism
3.
FASEB J ; 29(6): 2628-39, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25757569

ABSTRACT

FE65 and FE65L1 are cytoplasmic adaptor proteins that bind a variety of proteins, including the amyloid precursor protein, and that mediate the assembly of multimolecular complexes. We previously reported that FE65/FE65L1 double knockout (DKO) mice display disorganized laminin in meningeal fibroblasts and a cobblestone lissencephaly-like phenotype in the developing cortex. Here, we examined whether loss of FE65 and FE65L1 causes ocular and muscular deficits, 2 phenotypes that frequently accompany cobblestone lissencephaly. Eyes of FE65/FE65L1 DKO mice develop normally, but lens degeneration becomes apparent in young adult mice. Abnormal lens epithelial cell migration, widespread small vacuole formation, and increased laminin expression underneath lens capsules suggest impaired interaction between epithelial cells and capsular extracellular matrix in DKO lenses. Cortical cataracts develop in FE65L1 knockout (KO) mice aged 16 months or more but are absent in wild-type or FE65 KO mice. FE65 family KO mice show attenuated grip strength, and the nuclei of DKO muscle cells frequently locate in the middle of muscle fibers. These findings reveal that FE65 and FE65L1 are essential for the maintenance of lens transparency, and their loss produce phenotypes in brain, eye, and muscle that are comparable to the clinical features of congenital muscular dystrophies in humans.


Subject(s)
Carrier Proteins/genetics , Cataract/genetics , Muscle Weakness/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis , Blotting, Western , Carrier Proteins/metabolism , Cataract/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunohistochemistry , Laminin/metabolism , Lens Capsule, Crystalline/metabolism , Lens Capsule, Crystalline/pathology , Lens Diseases/genetics , Lens Diseases/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle Weakness/metabolism , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Nerve Tissue Proteins/deficiency , Nuclear Proteins/deficiency
4.
PLoS One ; 9(2): e88365, 2014.
Article in English | MEDLINE | ID: mdl-24516642

ABSTRACT

Antigen presentation by MHC class I molecules requires degradation of epitope source proteins in the cytosol. Although the preeminent role of the proteasome is clearly established, evidence suggesting a significant role for proteasome-independent generation of class I ligands has been reported repeatedly. However, an enzyme responsible for such a role has not been identified. Recently insulin-degrading enzyme (IDE) was shown to produce an antigenic peptide derived from the tumor antigen MAGE-A3 in an entirely proteasome-independent manner, raising the question of the global impact of IDE in MHC class I antigen processing. Here we report that IDE knockdown in human cell lines, or knockout in two different mouse strains, has no effect on cell surface expression of various MHC class I molecules, including allomorphs such as HLA-A3 and HLA-B27 suggested to be loaded in an at least a partly proteasome-independent manner. Moreover, reduced or absent IDE expression does not affect presentation of five epitopes including epitopes derived from beta amyloid and proinsulin, two preferred IDE substrates. Thus, IDE does not play a major role in MHC class I antigen processing, confirming the dominant and almost exclusive role of the proteasome in cytosolic production of MHC class I ligands.


Subject(s)
Antigen Presentation/immunology , Genes, MHC Class I/immunology , Insulysin/metabolism , Animals , Cell Line, Tumor , Cytosol/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Insulysin/genetics , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism
5.
Mol Cell Proteomics ; 13(2): 475-88, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24284412

ABSTRACT

FE65 is a cytosolic adapter protein and an important binding partner of amyloid precursor protein. Dependent on Thr668 phosphorylation in amyloid precursor protein, which influences amyloidogenic amyloid precursor protein processing, FE65 undergoes nuclear translocation, thereby transmitting a signal from the cell membrane to the nucleus. As this translocation may be relevant in Alzheimer disease, and as FE65 consists of three protein-protein interaction domains able to bind and affect a variety of other proteins and downstream signaling pathways, the identification of the FE65 interactome is of central interest in Alzheimer disease research. In this study, we identified 121 proteins as new potential FE65 interacting proteins in a pulldown/mass spectrometry approach using human post-mortem brain samples as protein pools for recombinantly expressed FE65. Co-immunoprecipitation assays further validated the interaction of FE65 with the candidates SV2A and SERCA2. In parallel, we investigated the whole cell proteome of primary hippocampal neurons from FE65/FE65L1 double knockout mice. Notably, the validated FE65 binding proteins were also found to be differentially abundant in neurons derived from the FE65 knockout mice relative to wild-type control neurons. SERCA2 is an important player in cellular calcium homeostasis, which was found to be up-regulated in double knockout neurons. Indeed, knock-down of FE65 in HEK293T cells also evoked an elevated sensitivity to thapsigargin, a stressor specifically targeting the activity of SERCA2. Thus, our results suggest that FE65 is involved in the regulation of intracellular calcium homeostasis. Whereas transfection of FE65 alone caused a typical dot-like phenotype in the nucleus, co-transfection of SV2A significantly reduced the percentage of FE65 dot-positive cells, pointing to a possible role for SV2A in the modulation of FE65 intracellular targeting. Given that SV2A has a signaling function at the presynapse, its effect on FE65 intracellular localization suggests that the SV2A/FE65 interaction might play a role in synaptic signal transduction.


Subject(s)
Brain/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Interaction Maps , Animals , Brain/pathology , Cells, Cultured , Embryo, Mammalian , HEK293 Cells , Humans , Immunoprecipitation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/isolation & purification , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/genetics , Protein Binding , Protein Interaction Maps/genetics , Synapses/genetics , Synapses/metabolism
6.
Mol Neurodegener ; 7: 9, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22429478

ABSTRACT

BACKGROUND: Several studies found that FE65, a cytoplasmic adaptor protein, interacts with APP and LRP1, altering the trafficking and processing of APP. We have previously shown that FE65 interacts with the ApoE receptor, ApoER2, altering its trafficking and processing. Interestingly, it has been shown that FE65 can act as a linker between APP and LRP1 or ApoER2. In the present study, we tested whether FE65 can interact with another ApoE receptor, VLDLR, thereby altering its trafficking and processing, and whether FE65 can serve as a linker between APP and VLDLR. RESULTS: We found that FE65 interacted with VLDLR using GST pull-down and co-immunoprecipitation assays in COS7 cells and in brain lysates. This interaction occurs via the PTB1 domain of FE65. Co-transfection with FE65 and full length VLDLR increased secreted VLDLR (sVLDLR); however, the levels of VLDLR C-terminal fragment (CTF) were undetectable as a result of proteasomal degradation. Additionally, FE65 increased cell surface levels of VLDLR. Moreover, we identified a novel complex between VLDLR and APP, which altered trafficking and processing of both proteins. Furthermore, immunoprecipitation results demonstrated that the presence of FE65 increased the interaction between APP and VLDLR in vitro and in vivo. CONCLUSIONS: These data suggest that FE65 can regulate VLDLR trafficking and processing. Additionally, the interaction between VLDLR and APP altered both protein's trafficking and processing. Finally, our data suggest that FE65 serves as a link between VLDLR and APP. This novel interaction adds to a growing body of literature indicating trimeric complexes with various ApoE Receptors and APP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, LDL/metabolism , Animals , COS Cells , Chlorocebus aethiops , Immunoprecipitation , Mice , Mice, Knockout , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Transfection
7.
J Neurochem ; 119(2): 377-88, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21824144

ABSTRACT

Amyloid precursor protein (APP) family members and their proteolytic products are implicated in normal nervous system function and Alzheimer's disease pathogenesis. APP processing and Aß secretion are regulated by neuronal activity. Various data suggest that NMDA receptor (NMDAR) activity plays a role in both non-amyloidogenic and amyloidogenic APP processing depending on whether synaptic or extrasynaptic NMDARs are activated, respectively. The APP-interacting FE65 proteins modulate APP trafficking and processing in cell lines, but little is known about their contribution to APP trafficking and processing in neurons, either in vivo or in vitro. In this study, we examined the contribution of the FE65 protein family to APP trafficking and processing in WT and FE65/FE65L1 double knockout neurons under basal conditions and following NMDAR activation. We report that FE65 proteins facilitate neuronal Aß secretion without affecting APP fast axonal transport to pre-synaptic terminals. In addition, FE65 proteins facilitate an NMDAR-dependent non-amyloidogenic APP processing pathway. Generation of high-molecular weight (HMW) species bearing an APP C-terminal epitope was also observed following NMDAR activation. These HMW species require proteasomal and calpain activities for their accumulation. Recovery of APP polypeptide fragments from electroeluted HMW species having molecular weights consistent with calpain I cleavage of APP suggests that HMW species are complexes formed from APP metabolic products. Our results indicate that the FE65 proteins contribute to physiological APP processing and accumulation of APP metabolic products resulting from NMDAR activation.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Amyloid beta-Peptides/metabolism , Animals , Axonal Transport/physiology , Blotting, Western , Calpain/pharmacology , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Glycosylation , Mice , Mice, Inbred ICR , Mice, Knockout , Molecular Weight , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Peptide Fragments/metabolism , Phosphorylation , Polysaccharides/chemistry , Proteasome Endopeptidase Complex/drug effects , Protein Processing, Post-Translational , Receptors, N-Methyl-D-Aspartate/drug effects , Reverse Transcriptase Polymerase Chain Reaction
8.
J Neurosci ; 31(2): 480-91, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21228158

ABSTRACT

Gonadotropin-releasing hormone-1 (GnRH-1) neurons migrate from the nasal placode to the forebrain where they control gonadal function via the hypothalamic-pituitary-gonadal axis. The birth of GnRH-1-expressing neurons is one of the first neurogenic events in the developing nasal placode. By gene expression screening on single GnRH-1 neurons, amyloid precursor binding protein-1 (FE65) was identified in migratory GnRH-1 neurons. FE65 has been shown to modulate ß1-integrin dynamics, actin cytoskeleton, cell motility, and FE65/amyloid precursor protein signaling has been described in neuro/glial cell fate determination as well as in modulating neurogenesis. Analysis of two mouse lines, one deficient for the 97 kDa FE65 isoform and a second deficient for the 97 and 60 kDa forms of FE65, showed overlapping phenotypes. In both lines, no migratory defects of the GnRH-1 neurons were observed, but a 25% increase in GnRH-1 neuronal number during embryonic development was found. Bromodeoxyuridine birth tracing and spatiotemporal tracking of GnRH-1 cell precursors demonstrated that the lack of the N-terminal portion of FE65, which includes part of the functional nuclear translocation/gene transcription domain of FE65 (WW domain), extends the timing of GnRH-1 neurogenesis in the developing nasal placode without affecting proliferation of GnRH-1 neuronal progenitors or cell death. The observed changes in the dynamics of GnRH-1 neurogenesis highlight a unique role for the 97 kDa isoform of FE65 and suggest that GnRH-1 cells, which have a short neurogenic window, originate from multipotent progenitors able to generate distinct cell types as GnRH-1 neurogenesis declines in response to environmental changes.


Subject(s)
Brain/metabolism , Gonadotropin-Releasing Hormone/metabolism , Nerve Tissue Proteins/physiology , Neurogenesis , Nuclear Proteins/physiology , Animals , Brain/cytology , Brain/embryology , Cell Count , Cell Death , Cell Movement , Cell Proliferation , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurons/cytology , Neurons/physiology , Nuclear Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Structure, Tertiary , Vomeronasal Organ/cytology , Vomeronasal Organ/embryology , Vomeronasal Organ/metabolism
9.
J Neurochem ; 114(1): 160-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20374429

ABSTRACT

Alternative splicing of tau exon 10 influences microtubule assembly and stability during development and in pathological processes of the central nervous system. However, the cellular events that underlie this pre-mRNA splicing remain to be delineated. In this study, we examined the possibility that ischemic injury, known to change the cellular distribution and expression of several RNA splicing factors, alters the splicing of tau exon 10. Transient occlusion of the middle cerebral artery reduced tau exon 10 inclusion in the ischemic cortical area within 12 h, resulting in the induction of three-repeat (3R) tau in cortical neurons. Ubiquitinated protein aggregates and reduced proteasome activity were also observed. Administration of proteasome inhibitors such as MG132, proteasome inhibitor I and lactacystin reduced tau exon 10 splicing in cortical cell cultures. Decreased levels of Tra2beta, an RNA splicing factor responsible for tau exon 10 inclusion, were detected both in cortical cell cultures exposed to MG132 and in cerebral cortex after ischemic injury. Taken together, these findings suggest that transient focal cerebral ischemia reduces tau exon 10 splicing through a mechanism involving proteasome-ubiquitin dysfunction and down-regulation of Tra2beta.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Ischemic Attack, Transient/metabolism , Proteasome Endopeptidase Complex/physiology , tau Proteins/metabolism , Alternative Splicing , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Exons , Male , Neurons/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Proteasome Inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Serine-Arginine Splicing Factors , Ubiquitination , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL