Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Brain ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38743588

ABSTRACT

Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited peripheral neuropathy caused by a 1.5 megabase tandem duplication of chromosome 17 harboring the PMP22 gene. This dose-dependent overexpression of PMP22 results in disrupted Schwann cell myelination of peripheral nerves. To get better insights into the underlying pathogenic mechanisms in CMT1A, we investigated the role of PMP22 duplication on cellular homeostasis in CMT1A mouse models and in patient-derived induced pluripotent stem cells differentiated into Schwann cell precursors (iPSC-SCPs). We performed lipidomic profiling and bulk RNA sequencing on sciatic nerves of two developing CMT1A mouse models and on CMT1A patient derived iPSC-SCPs. For the sciatic nerves of the CMT1A mice, cholesterol and lipid metabolism was dose-dependently downregulated throughout development. For the CMT1A iPSC-SCPs, transcriptional analysis unveiled a strong suppression of genes related to autophagy and lipid metabolism. Gene ontology enrichment analysis identified disturbances in pathways related to plasma membrane components and cell receptor signaling. Lipidomic analysis confirmed the severe dysregulation in plasma membrane lipids, particularly sphingolipids, in CMT1A iPSC-SCPs. Furthermore, we identified reduced lipid raft dynamics, disturbed plasma membrane fluidity, and impaired cholesterol incorporation and storage, all of which could result from altered lipid storage homeostasis in the patient-derived CMT1A iPSC-SCPs. Importantly, this phenotype could be rescued by stimulating autophagy and lipolysis. We conclude that PMP22 duplication disturbs intracellular lipid storage and leads to a more disordered plasma membrane due to an alteration in the lipid composition, which ultimately may lead to impaired axo-glial interactions. Moreover, targeting lipid handling and metabolism could hold promise for the treatment of CMT1A patients.

2.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34292306

ABSTRACT

γ-Secretase affects many physiological processes through targeting >100 substrates; malfunctioning links γ-secretase to cancer and Alzheimer's disease. The spatiotemporal regulation of its stoichiometric assembly remains unresolved. Fractionation, biochemical assays, and imaging support prior formation of stable dimers in the ER, which, after ER exit, assemble into full complexes. In vitro ER budding shows that none of the subunits is required for the exit of others. However, knockout of any subunit leads to the accumulation of incomplete subcomplexes in COPII vesicles. Mutating a DPE motif in presenilin 1 (PSEN1) abrogates ER exit of PSEN1 and PEN-2 but not nicastrin. We explain this by the preferential sorting of PSEN1 and nicastrin through Sec24A and Sec24C/D, respectively, arguing against full assembly before ER exit. Thus, dimeric subcomplexes aided by Sec24 paralog selectivity support a stepwise assembly of γ-secretase, controlling final levels in post-Golgi compartments.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , COP-Coated Vesicles/metabolism , Endopeptidases/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Presenilin-1/metabolism , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/genetics , Animals , Biological Transport , COP-Coated Vesicles/chemistry , COP-Coated Vesicles/genetics , Cell Line , Cell Line, Tumor , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Endopeptidases/chemistry , Endopeptidases/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Golgi Apparatus/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Models, Molecular , Neurons/cytology , Presenilin-1/chemistry , Presenilin-1/genetics , Primary Cell Culture , Protein Binding , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Rats , Rats, Wistar , Signal Transduction , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
3.
Cell ; 166(1): 193-208, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27293189

ABSTRACT

γ-Secretases are a family of intramembrane-cleaving proteases involved in various signaling pathways and diseases, including Alzheimer's disease (AD). Cells co-express differing γ-secretase complexes, including two homologous presenilins (PSENs). We examined the significance of this heterogeneity and identified a unique motif in PSEN2 that directs this γ-secretase to late endosomes/lysosomes via a phosphorylation-dependent interaction with the AP-1 adaptor complex. Accordingly, PSEN2 selectively cleaves late endosomal/lysosomal localized substrates and generates the prominent pool of intracellular Aß that contains longer Aß; familial AD (FAD)-associated mutations in PSEN2 increased the levels of longer Aß further. Moreover, a subset of FAD mutants in PSEN1, normally more broadly distributed in the cell, phenocopies PSEN2 and shifts its localization to late endosomes/lysosomes. Thus, localization of γ-secretases determines substrate specificity, while FAD-causing mutations strongly enhance accumulation of aggregation-prone Aß42 in intracellular acidic compartments. The findings reveal potentially important roles for specific intracellular, localized reactions contributing to AD pathogenesis.


Subject(s)
Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/analysis , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Presenilin-2/analysis , Adaptor Protein Complex 1/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Motifs , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Endosomes/chemistry , Humans , Lysosomes/chemistry , Mice , Presenilin-1/analysis , Presenilin-1/chemistry , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/chemistry , Presenilin-2/genetics , Presenilin-2/metabolism , Rats , Substrate Specificity
4.
J Cell Biol ; 198(1): 23-35, 2012 Jul 09.
Article in English | MEDLINE | ID: mdl-22753898

ABSTRACT

Presenilin (PSEN) deficiency is accompanied by accumulation of endosomes and autophagosomes, likely caused by impaired endo-lysosomal fusion. Recently, Lee et al. (2010. Cell. doi: http://dx.doi.org/10.1016/j.cell.2010.05.008) attributed this phenomenon to PSEN1 enabling the transport of mature V0a1 subunits of the vacuolar ATPase (V-ATPase) to lysosomes. In their view, PSEN1 mediates the N-glycosylation of V0a1 in the endoplasmic reticulum (ER); consequently, PSEN deficiency prevents V0a1 glycosylation, compromising the delivery of unglycosylated V0a1 to lysosomes, ultimately impairing V-ATPase function and lysosomal acidification. We show here that N-glycosylation is not a prerequisite for proper targeting and function of this V-ATPase subunit both in vitro and in vivo in Drosophila melanogaster. We conclude that endo-lysosomal dysfunction in PSEN(-/-) cells is not a consequence of failed N-glycosylation of V0a1, or compromised lysosomal acidification. Instead, lysosomal calcium storage/release is significantly altered in PSEN(-/-) cells and neurons, thus providing an alternative hypothesis that accounts for the impaired lysosomal fusion capacity and accumulation of endomembranes that accompanies PSEN deficiency.


Subject(s)
Calcium/metabolism , Drosophila Proteins/metabolism , Homeostasis/physiology , Lysosomes/metabolism , Presenilin-1/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Biological Transport , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Fibroblasts/metabolism , Glycosylation , Hippocampus/metabolism , Humans , Mice , Mice, Knockout , Neurons/metabolism , Vacuolar Proton-Translocating ATPases/genetics
5.
EMBO J ; 31(15): 3252-69, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22781129

ABSTRACT

Dendritic filopodia are dynamic structures thought to be the precursors of spines during synapse development. Morphological maturation to spines is associated with the stabilization and strengthening of synapses, and can be altered in various neurological disorders. Telencephalin (TLN/intercellular adhesion molecule-5 (ICAM5)) localizes to dendritic filopodia, where it facilitates their formation/maintenance, thereby slowing spine morphogenesis. As spines are largely devoid of TLN, its exclusion from the filopodia surface appears to be required in this maturation process. Using HeLa cells and primary hippocampal neurons, we demonstrate that surface removal of TLN involves internalization events mediated by the small GTPase ADP-ribosylation factor 6 (ARF6), and its activator EFA6A. This endocytosis of TLN affects filopodia-to-spine transition, and requires Rac1-mediated dephosphorylation/release of actin-binding ERM proteins from TLN. At the somato-dendritic surface, TLN and EFA6A are confined to distinct, flotillin-positive membrane subdomains. The co-distribution of TLN with this lipid raft marker also persists during its endosomal targeting to CD63-positive late endosomes. This suggests a specific microenvironment facilitating ARF6-mediated mobilization of TLN that contributes to promotion of dendritic spine development.


Subject(s)
ADP-Ribosylation Factors/physiology , Cell Adhesion Molecules/metabolism , Dendrites/physiology , Dendritic Spines/metabolism , Endosomes/metabolism , Nerve Tissue Proteins/metabolism , Pseudopodia/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Amino Acid Sequence , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Cellular Microenvironment/genetics , Cellular Microenvironment/physiology , Dendrites/genetics , Dendrites/metabolism , Dendritic Spines/genetics , Dendritic Spines/physiology , HeLa Cells , Humans , Models, Biological , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Primary Cell Culture , Protein Transport/genetics , Pseudopodia/genetics , Pseudopodia/physiology , Sequence Homology, Amino Acid
6.
Hum Mol Genet ; 17(8): 1175-83, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18182442

ABSTRACT

Mutations in ZFHX1B cause Mowat-Wilson syndrome (MWS) but the precise mechanisms underlying the aberrant functions of mutant ZFHX1B proteins (also named Smad-interacting protein-1, SIP1) in patients are unknown. Using mass spectrometry analysis, we identified subunits of the NuRD corepressor complex in affinity-purified Zfhx1b complexes. We find that Zfhx1b associates with NuRD through its N-terminal domain, which contains a previously postulated NuRD interacting motif. Interestingly, this motif is substituted by an unrelated sequence in a recently described MWS patient. We show here that such aberrant ZFHX1B protein is unable to recruit NuRD subunits and displays reduced transcriptional repression activity on the XBMP4 gene promoter, a target of Zfhx1b. We further demonstrate that the NuRD component Mi-2beta is involved in repression of the Zfhx1b target gene E-cadherin as well as in Zfhx1b-induced neural induction in animal caps from Xenopus embryos. Thus, NuRD and Zfhx1b functionally interact, and defective NuRD recruitment by mutant human ZFHX1B can be a MWS-causing mechanism. This is the first study providing mechanistic insight into the aberrant function of a single domain of the multi-domain protein ZFHX1B/SIP1 in human disease.


Subject(s)
Abnormalities, Multiple/metabolism , Histone Deacetylases/metabolism , Intellectual Disability/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Adenosine Triphosphatases/metabolism , Animals , Autoantigens/metabolism , Cadherins/metabolism , Cell Line , DNA Helicases/metabolism , Embryo, Nonmammalian/metabolism , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Protein Structure, Tertiary , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/isolation & purification , Syndrome , Xenopus
7.
Dev Biol ; 306(1): 34-49, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17442301

ABSTRACT

The DNA-binding transcription factor Smad-interacting protein-1 (Sip1) (also named Zfhx1b/ZEB2) plays essential roles in vertebrate embryogenesis. In Xenopus, XSip1 is essential at the gastrula stage for neural tissue formation, but the precise molecular mechanisms that underlie this process have not been fully identified yet. Here we show that XSip1 functions as a transcriptional repressor during neural induction. We observed that constitutive activation of BMP signaling prevents neural induction by XSip1 but not the inhibition of several epidermal genes. We provide evidence that XSip1 binds directly to the BMP4 proximal promoter and modulates its activity. Finally, by deletion and mutational analysis, we show that XSip1 possesses multiple repression domains and that CtBPs contribute to its repression activity. Consistent with this, interference with XCtBP function reduced XSip1 neuralizing activity. These results suggest that Sip1 acts in neural tissue formation through direct repression of BMP4 but that BMP-independent mechanisms are involved as well. Our data also provide the first demonstration of the importance of CtBP binding in Sip1 transcriptional activity in vivo.


Subject(s)
Alcohol Oxidoreductases/metabolism , Bone Morphogenetic Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Nervous System/embryology , Repressor Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Alcohol Oxidoreductases/genetics , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Ectoderm/metabolism , Epidermis/metabolism , Homeodomain Proteins/genetics , Nervous System/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Repressor Proteins/genetics , Xenopus/anatomy & histology , Xenopus/genetics , Xenopus Proteins/genetics
8.
Dev Dyn ; 235(6): 1491-500, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16518800

ABSTRACT

The zinc finger/homeo-domain transcription factor (zfh x 1) family in vertebrates consists of two members, deltaEF1 and SIP1. They have been proposed to display antagonistic activities in the interpretation of Smad-dependent TGFbeta signaling during mesoderm formation. We cloned Xenopus deltaEF1 cDNA, analyzed the expression profile of the gene, and compared the inducing and interacting properties of the protein to that of XSIP1. Whereas XSIP1 RNA is selectively expressed in the early developing nervous system, we show that XdeltaEF1 gene transcription is only activated during neurulation and that its expression is restricted to the paraxial mesoderm. From early tail bud stage, XdeltaEF1 and XSIP1 are coexpressed in migratory cranial neural crest, in the retina, and in the neural tube. Overproduction of XdeltaEF1 in RNA-injected embryos, like that of XSIP1, reduced the expression of BMP-dependent genes but only XSIP1 has the ability to induce neural markers. We find that XdeltaEF1 and XSIP1 can both form complexes, although with different efficiency, with Smad3, with the coactivators p300 and pCAF, and with the corepressor CtBP1. Together, these results indicate that deltaEF1 and SIP1 do not function as antagonists during Xenopus early embryogenesis but do display different repression efficiencies and interaction properties.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Xenopus Proteins/genetics , Amino Acid Sequence , Animals , Embryonic Development/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/physiology , Mesoderm/metabolism , Molecular Sequence Data , Repressor Proteins/biosynthesis , Repressor Proteins/physiology , Transcription Factors/biosynthesis , Xenopus , Xenopus Proteins/biosynthesis , Xenopus Proteins/physiology , Zinc Fingers/genetics
9.
J Biol Chem ; 278(28): 26135-45, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12714599

ABSTRACT

deltaEF1 and SIP1 (or Zfhx1a and Zfhx1b, respectively) are the only known members of the vertebrate Zfh1 family of homeodomain/zinc finger-containing proteins. Similar to other transcription factors, both Smad-interacting protein-1 (SIP1) and deltaEF1 are capable of repressing E-cadherin transcription through binding to the E2 boxes located in its promoter. In the case of deltaEF1, this repression has been proposed to occur via interaction with the corepressor C-terminal binding protein (CtBP). In this study, we show by coimmunoprecipitation that SIP1 and CtBP interact in vivo and that an isolated CtBP-binding SIP1 fragment depends on CtBP for transcriptional repression. However, and most importantly, full-length SIP1 and deltaEF1 proteins do not depend on their interaction with CtBP to repress transcription from the E-cadherin promoter. Furthermore, in E-cadherin-positive kidney epithelial cells, the conditional synthesis of mutant SIP1 that cannot bind to CtBP abrogates endogenous E-cadherin expression in a similar way as wild-type SIP1. Our results indicate that full-length SIP1 can repress E-cadherin in a CtBP-independent manner.


Subject(s)
Cadherins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Homeodomain Proteins/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Alcohol Oxidoreductases , Amino Acid Motifs , Animals , Binding Sites , Blotting, Western , Cell Line , Cells, Cultured , Dogs , Down-Regulation , Genes, Reporter , Humans , Luciferases/metabolism , Mice , Microscopy, Fluorescence , Models, Genetic , Mutation , Peptides/chemistry , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Transfection , Tumor Cells, Cultured , Two-Hybrid System Techniques , Up-Regulation , Zinc Finger E-box Binding Homeobox 2
SELECTION OF CITATIONS
SEARCH DETAIL
...