Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Cell Mol Life Sci ; 81(1): 334, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39115595

ABSTRACT

Mutations in the IER3IP1 (Immediate Early Response-3 Interacting Protein 1) gene can give rise to MEDS1 (Microcephaly with Simplified Gyral Pattern, Epilepsy, and Permanent Neonatal Diabetes Syndrome-1), a severe condition leading to early childhood mortality. The small endoplasmic reticulum (ER)-membrane protein IER3IP1 plays a non-essential role in ER-Golgi transport. Here, we employed secretome and cell-surface proteomics to demonstrate that the absence of IER3IP1 results in the mistrafficking of proteins crucial for neuronal development and survival, including FGFR3, UNC5B and SEMA4D. This phenomenon correlates with the distension of ER membranes and increased lysosomal activity. Notably, the trafficking of cargo receptor ERGIC53 and KDEL-receptor 2 are compromised, with the latter leading to the anomalous secretion of ER-localized chaperones. Our investigation extended to in-utero knock-down of Ier3ip1 in mouse embryo brains, revealing a morphological phenotype in newborn neurons. In summary, our findings provide insights into how the loss or mutation of a 10 kDa small ER-membrane protein can cause a fatal syndrome.


Subject(s)
Endoplasmic Reticulum , Golgi Apparatus , Microcephaly , Endoplasmic Reticulum/metabolism , Animals , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Mice , Golgi Apparatus/metabolism , Humans , Mutation , Protein Transport , Membrane Proteins/metabolism , Membrane Proteins/genetics , Neurons/metabolism , Neurons/pathology
2.
Bioessays ; 44(10): e2200064, 2022 10.
Article in English | MEDLINE | ID: mdl-35986435

ABSTRACT

An entirely different mechanism and localization were recently proposed for the COPII coat complex, challenging its well-accepted function to select and concentrate cargo into small COPII-coated spherical transport vesicles. Instead, the COPII complex is suggested to form a dynamic yet stationary collar that forms a boundary between the ER and the ER export membrane domain. This membrane domain, the ER exit site (ERES), is the site of COPII-mediated sorting and concentration of transport competent proteins. Subsequently, the ERES is implicated to mature and bud to form a sizeable pleiomorphic transport carrier that translocate on microtubules to fuse with the Golgi apparatus. Despite this drastic mechanistic dogma shift, most of the underlying protein-protein and protein-membrane interactions remain unchanged. Here, we attempt to provide a detailed description of the newly proposed model of how ER to Golgi transport works by describing the role of several essential proteins of the transport machinery.


Subject(s)
COP-Coated Vesicles , Golgi Apparatus , COP-Coated Vesicles/metabolism , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Protein Transport
3.
Cell Mol Life Sci ; 79(3): 168, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35235058

ABSTRACT

ß-Site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1) is the major described ß-secretase to generate Aß peptides in Alzheimer's disease (AD). However, all therapeutic attempts to block BACE1 activity and to improve AD symptoms have so far failed. A potential candidate for alternative Aß peptides generation is the metalloproteinase meprin ß, which cleaves APP predominantly at alanine in p2 and in this study we can detect an increased meprin ß expression in AD brain. Here, we report the generation of the transgenic APP/lon mouse model of AD lacking the functional Mep1b gene (APP/lon × Mep1b-/-). We examined levels of canonical and truncated Aß species using urea-SDS-PAGE, ELISA and immunohistochemistry in brains of APP/lon mouse × Mep1b-/-. Additionally, we investigated the cognitive abilities of these mice during the Morris water maze task. Aß1-40 and 1-42 levels are reduced in APP/lon mice when meprin ß is absent. Immunohistochemical staining of mouse brain sections revealed that N-terminally truncated Aß2-x peptide deposition is decreased in APP/lon × Mep1b-/- mice. Importantly, loss of meprin ß improved cognitive abilities and rescued learning behavior impairments in APP/lon mice. These observations indicate an important role of meprin ß within the amyloidogenic pathway and Aß production in vivo.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Learning , Memory Disorders/pathology , Metalloendopeptidases/deficiency , Aged , Amyloid Precursor Protein Secretases/metabolism , Animals , Astrocytes/metabolism , Brain/pathology , Crosses, Genetic , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Metalloendopeptidases/metabolism , Mice, Knockout , Peptides/metabolism , Protein Processing, Post-Translational
4.
J Cell Sci ; 133(17)2020 09 01.
Article in English | MEDLINE | ID: mdl-32873699

ABSTRACT

The quote "bring it back, bring it back, don't take it away from me" from Queen's Love of my life describes the function of the sorting receptor RER1, a 23 kDa protein with four transmembrane domains (TMDs) that localizes to the intermediate compartment and the cis-Golgi. From there it returns escaped proteins that are not supposed to leave the endoplasmic reticulum (ER) back to it. Unique about RER1 is its ability to recognize its ligands through binding motifs in TMDs. Among its substrates are ER-resident proteins, as well as unassembled subunits of multimeric complexes that are retrieved back into the ER, this way guarding the full assembly of their respective complexes. The basic mechanisms for RER1-dependent retrieval have been already elucidated some years ago in yeast. More recently, several important cargoes of RER1 have been described in mammalian cells, and the in vivo role of RER1 is being unveiled by using mouse models. In this Review, we give an overview of the cell biology of RER1 in different models, discuss its controversial role in the brain and provide an outlook on future directions for RER1 research.


Subject(s)
Golgi Apparatus , Membrane Glycoproteins , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Membrane Glycoproteins/metabolism , Mice , Protein Transport , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
5.
Nucleic Acids Res ; 48(19): 10924-10939, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33010171

ABSTRACT

NBS1 is a critical component of the MRN (MRE11/RAD50/NBS1) complex, which regulates ATM- and ATR-mediated DNA damage response (DDR) pathways. Mutations in NBS1 cause the human genomic instability syndrome Nijmegen Breakage Syndrome (NBS), of which neuronal deficits, including microcephaly and intellectual disability, are classical hallmarks. Given its function in the DDR to ensure proper proliferation and prevent death of replicating cells, NBS1 is essential for life. Here we show that, unexpectedly, Nbs1 deletion is dispensable for postmitotic neurons, but compromises their arborization and migration due to dysregulated Notch signaling. We find that Nbs1 interacts with NICD-RBPJ, the effector of Notch signaling, and inhibits Notch activity. Genetic ablation or pharmaceutical inhibition of Notch signaling rescues the maturation and migration defects of Nbs1-deficient neurons in vitro and in vivo. Upregulation of Notch by Nbs1 deletion is independent of the key DDR downstream effector p53 and inactivation of each MRN component produces a different pattern of Notch activity and distinct neuronal defects. These data indicate that neuronal defects and aberrant Notch activity in Nbs1-deficient cells are unlikely to be a direct consequence of loss of MRN-mediated DDR function. This study discloses a novel function of NBS1 in crosstalk with the Notch pathway in neuron development.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Neurogenesis , Neurons/metabolism , Receptors, Notch/metabolism , Acid Anhydride Hydrolases/metabolism , Animals , Cells, Cultured , DNA Damage , DNA Repair , Embryo, Mammalian , Fibroblasts , MRE11 Homologue Protein/metabolism , Mice , Neurons/cytology
6.
Neurobiol Dis ; 155: 105400, 2021 07.
Article in English | MEDLINE | ID: mdl-34019998

ABSTRACT

Mutations in the ER-network forming GTPase atlastin3 (ATL3) can cause axon degeneration of sensory neurons by not fully understood mechanisms. We here show that the hereditary sensory and autonomous neuropathy (HSAN)-causing ATL3 Y192C or P338R are excluded from distal axons by a barrier at the axon initial segment (AIS). This barrier is selective for mutated ATL3, but not wildtype ATL3 or unrelated ER-membrane proteins. Actin-depolymerization partially restores the transport of ATL3 Y192C into distal axons. The results point to the existence of a selective diffusion barrier in the ER membrane at the AIS, analogous to the AIS-based barriers for plasma membrane and cytosolic proteins. Functionally, the absence of ATL3 at the distal axon reduces axonal autophagy and the ER network deformation in the soma causes a reduction in axonal lysosomes. Both could contribute to axonal degeneration and eventually to HSAN.


Subject(s)
Autophagy/physiology , Axons/physiology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mutation/physiology , Animals , Axons/pathology , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL
7.
Org Biomol Chem ; 19(3): 574-578, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33406188

ABSTRACT

An efficient synthesis for silicon-rhodamines was developed, enabling the preparation and evaluation of silicon-rhodamine isothiocyanate (SITC) as a novel tool for facile fluorescent labeling. Ease of use in conjugation to amino groups, high stability and excellent photophysical properties are demonstrated. SITC-actin was found to be neutral to F-actin polymerization induction and well suited for high resolution fluorescence microscopy.

8.
J Biol Chem ; 294(31): 11741-11750, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31186352

ABSTRACT

The Notch receptor is a key mediator of developmental programs and cell-fate decisions. Imbalanced Notch signaling leads to developmental disorders and cancer. To fully characterize the Notch signaling pathway and exploit it in novel therapeutic interventions, a comprehensive view on the regulation and requirements of Notch signaling is needed. Notch is regulated at different levels, ranging from ligand binding, stability to endocytosis. Using an array of different techniques, including reporter gene assays, immunocytochemistry, and ChIP-qPCR we show here, to the best of our knowledge for the first time, regulation of Notch signaling at the level of the nuclear pore. We found that the nuclear pore protein Nup214 (nucleoporin 214) and its interaction partner Nup88 negatively regulate Notch signaling in vitro and in vivo in zebrafish. In mammalian cells, loss of Nup88/214 inhibited nuclear export of recombination signal-binding protein for immunoglobulin κJ region (RBP-J), the DNA-binding component of the Notch pathway. This inhibition increased binding of RBP-J to its cognate promoter regions, resulting in increased downstream Notch signaling. Interestingly, we also found that NUP214 fusion proteins, causative for certain cases of T-cell acute lymphatic leukemia, potentially contribute to tumorigenesis via a Notch-dependent mechanism. In summary, the nuclear pore components Nup88/214 suppress Notch signaling in vitro, and in zebrafish, nuclear RBP-J levels are rate-limiting factors for Notch signaling in mammalian cells, and regulation of nucleocytoplasmic transport of RBP-J may contribute to fine-tuning Notch activity in cells.


Subject(s)
Nuclear Pore Complex Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Morpholinos/genetics , Morpholinos/metabolism , Nuclear Pore Complex Proteins/antagonists & inhibitors , Nuclear Pore Complex Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Notch/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Cell Mol Life Sci ; 76(7): 1433-1445, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30666337

ABSTRACT

Atlastins (ATLs) are membrane-bound GTPases involved in shaping of the endoplasmic reticulum (ER). Mutations in ATL1 and ATL3 cause spastic paraplegia and hereditary sensory neuropathy. We here show that the sensory neuropathy causing ATL3 Y192C mutation reduces the complexity of the tubular ER-network. ATL3 Y192C delays ER-export by reducing the number of ER exit sites, reduces autophagy, fragments the Golgi and causes malformation of the nucleus. In cultured primary neurons, ATL3 Y192C does not localize to the growing axon, resulting in axon growth deficits. Patient-derived fibroblasts possess a tubular ER with reduced complexity and have a reduced number of autophagosomes. The data suggest that the disease-causing ATL3 Y192C mutation affects multiple ER-related pathways, possibly as a consequence of the distorted ER morphology.


Subject(s)
Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/metabolism , Animals , Autophagosomes , Autophagy , Axons/metabolism , Cells, Cultured , Endoplasmic Reticulum/pathology , GTP Phosphohydrolases/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutagenesis, Site-Directed , Neurons/cytology , Neurons/metabolism
10.
J Cell Sci ; 129(20): 3868-3877, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27587840

ABSTRACT

Export out of the endoplasmic reticulum (ER) involves the Sar1 and COPII machinery acting at ER exit sites (ERES). Whether and how cargo proteins are recruited upstream of Sar1 and COPII is unclear. Two models are conceivable, a recruitment model where cargo is actively transported through a transport factor and handed over to the Sar1 and COPII machinery in ERES, and a capture model, where cargo freely diffuses into ERES where it is captured by the Sar1 and COPII machinery. Using the novel secretion inhibitor FLI-06, we show that recruitment of the cargo VSVG to ERES is an active process upstream of Sar1 and COPII. Applying FLI-06 before concentration of VSVG in ERES completely abolishes its recruitment. In contrast, applying FLI-06 after VSVG concentration in ERES does not lead to dispersal of the concentrated VSVG, arguing that it inhibits recruitment to ERES as opposed to capture in ERES. FLI-06 also inhibits export out of the trans-Golgi network (TGN), suggesting that similar mechanisms might orchestrate cargo selection and concentration at the ER and TGN. FLI-06 does not inhibit autophagosome biogenesis and the ER-peroxisomal transport route, suggesting that these rely on different mechanisms.


Subject(s)
Endoplasmic Reticulum/metabolism , Quinolines/pharmacology , trans-Golgi Network/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Endocytosis/drug effects , Exocytosis/drug effects , HeLa Cells , Humans , Peroxisomes/drug effects , Peroxisomes/metabolism , Protein Folding/drug effects , Protein Transport/drug effects , trans-Golgi Network/drug effects
11.
J Biol Chem ; 291(44): 23068-23083, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27613868

ABSTRACT

Genetic rearrangements are a hallmark of several forms of leukemia and can lead to oncogenic fusion proteins. One example of an affected chromosomal region is the gene coding for Nup214, a nucleoporin that localizes to the cytoplasmic side of the nuclear pore complex (NPC). We investigated two such fusion proteins, SET-Nup214 and SQSTM1 (sequestosome)-Nup214, both containing C-terminal portions of Nup214. SET-Nup214 nuclear bodies containing the nuclear export receptor CRM1 were observed in the leukemia cell lines LOUCY and MEGAL. Overexpression of SET-Nup214 in HeLa cells leads to the formation of similar nuclear bodies that recruit CRM1, export cargo proteins, and certain nucleoporins and concomitantly affect nuclear protein and poly(A)+ RNA export. SQSTM1-Nup214, although mostly cytoplasmic, also forms nuclear bodies and inhibits nuclear protein but not poly(A)+ RNA export. The interaction of the fusion proteins with CRM1 is RanGTP-dependent, as shown in co-immunoprecipitation experiments and binding assays. Further analysis revealed that the Nup214 parts mediate the inhibition of nuclear export, whereas the SET or SQSTM1 part determines the localization of the fusion protein and therefore the extent of the effect. SET-Nup214 nuclear bodies are highly mobile structures, which are in equilibrium with the nucleoplasm in interphase and disassemble during mitosis or upon treatment of cells with the CRM1-inhibitor leptomycin B. Strikingly, we found that nucleoporins can be released from nuclear bodies and reintegrated into existing NPC. Our results point to nuclear bodies as a means of preventing the formation of potentially insoluble and harmful protein aggregates that also may serve as storage compartments for nuclear transport factors.


Subject(s)
Histone Chaperones/metabolism , Intranuclear Inclusion Bodies/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/metabolism , Poly A/metabolism , RNA, Messenger/metabolism , Sequestosome-1 Protein/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , DNA-Binding Proteins , Histone Chaperones/genetics , Humans , Intranuclear Inclusion Bodies/genetics , Karyopherins/genetics , Karyopherins/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Proteins/genetics , Poly A/genetics , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sequestosome-1 Protein/genetics , Transcription Factors/genetics , Exportin 1 Protein
12.
Biochem Biophys Res Commun ; 478(4): 1751-7, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27608597

ABSTRACT

The γ-secretase complex comprises presenilin (PS), nicastrin (NCT), anterior pharynx-defective 1 (Aph1), and presenilin enhancer 2 (Pen2). PS has two homologues, PS1 and PS2. Aph1 has two isoforms, Aph1a and Aph1b, with the former existing as two splice variants Aph1aL and Aph1aS. Each complex consists of one subunit each, resulting in six different γ-secretases. To better understand the functional differences among the γ-secretases, we reconstituted them using a yeast system and compared Notch1-cleavage and amyloid precursor protein (APP)-cleavage activities. Intriguingly, PS2/Aph1b had a clear substrate specificity: APP-Gal4, but not Notch-Gal4, was cleaved. In HEK cell lines expressing defined γ-secretase subunits, we showed that PS1/Aph1b, PS2/Aph1aL, PS2/Aph1aS and PS2/Aph1b γ-secretase produced amyloid ß peptide (Aß) with a higher Aß42+Aß43-to-Aß40 (Aß42(43)/Aß40) ratio than the other γ-secretases. In addition, PS2/Aph1aS γ-secretase produced less Notch intracellular domain (NICD) than did the other 5 γ-secretases. Considering that the Aß42(43)/Aß40 ratio is relevant in the pathogenesis of Alzheimer's disease (AD), and that inhibition of Notch cleavage causes severe side effect, these results suggest that the PS2/Aph1aS γ-secretase complex is a potential therapeutic target in AD.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism , Presenilin-1/metabolism , Presenilin-2/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Blotting, Western , Endopeptidases , HEK293 Cells , Humans , Membrane Proteins/genetics , Peptide Fragments/metabolism , Peptide Hydrolases/genetics , Presenilin-1/genetics , Presenilin-2/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Substrate Specificity
13.
Biol Res ; 49(1): 34, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27464526

ABSTRACT

BACKGROUND: Cellular senescence is induced either internally, for example by replication exhaustion and cell division, or externally, for example by irradiation. In both cases, cellular damages accumulate which, if not successfully repaired, can result in senescence induction. Recently, we determined the transcriptional changes combined with the transition into replicative senescence in primary human fibroblast strains. Here, by γ-irradiation we induced premature cellular senescence in the fibroblast cell strains (HFF and MRC-5) and determined the corresponding transcriptional changes by high-throughput RNA sequencing. RESULTS: Comparing the transcriptomes, we found a high degree of similarity in differential gene expression in replicative as well as in irradiation induced senescence for both cell strains suggesting, in each cell strain, a common cellular response to error accumulation. On the functional pathway level, "Cell cycle" was the only pathway commonly down-regulated in replicative and irradiation-induced senescence in both fibroblast strains, confirming the tight link between DNA repair and cell cycle regulation. However, "DNA repair" and "replication" pathways were down-regulated more strongly in fibroblasts undergoing replicative exhaustion. We also retrieved genes and pathways in each of the cell strains specific for irradiation induced senescence. CONCLUSION: We found the pathways associated with "DNA repair" and "replication" less stringently regulated in irradiation induced compared to replicative senescence. The strong regulation of these pathways in replicative senescence highlights the importance of replication errors for its induction.


Subject(s)
Cellular Senescence/physiology , Fibroblasts/radiation effects , Aborted Fetus , Analysis of Variance , Cells, Cultured , Cellular Senescence/genetics , Cellular Senescence/radiation effects , DNA Damage , DNA Repair/radiation effects , DNA Replication/radiation effects , Down-Regulation/radiation effects , Fibroblasts/physiology , Gamma Rays , Gene Expression Profiling , Humans , Immunoblotting , Lung , Male , Sequence Analysis, RNA , Time Factors , Up-Regulation/radiation effects , beta-Galactosidase/metabolism
14.
J Cell Biochem ; 116(7): 1222-31, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25546737

ABSTRACT

The internalization of near-infrared fluorescently labeled cargos into living cells and tissues allows a highly sensitive detection without interference from skin, porphins or other fluorescent cell and tissue compounds. In this study, the uptake of labeled bovine serum albumin and an antibody, into fibrosarcoma (HT-1080) cells was triggered by the formation of non-covalent complexes with different cell-penetrating peptides; uptake efficiency and intracellular localization were determined. To improve selectivity of internalization into tumor cells, a fluorescent activatable cell-penetrating peptide (ACPP) was synthesized and functionally characterized. This 25-mer peptide was designed to be activatable by Matrix-Metallo-Proteases (MMPs). Its uptake selectivity was estimated using cells with different MMP activities.


Subject(s)
Carbocyanines/chemistry , Cell-Penetrating Peptides/pharmacology , Indoles/chemistry , Serum Albumin, Bovine/chemistry , Trastuzumab/chemistry , Cell Line, Tumor , Cell-Penetrating Peptides/chemical synthesis , Drug Delivery Systems/methods , Fluorescent Dyes/chemistry , Humans , Matrix Metalloproteinases/metabolism , Trastuzumab/metabolism
15.
Nat Chem Biol ; 9(11): 731-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24077179

ABSTRACT

Notch signaling has a pivotal role in numerous cell-fate decisions, and its aberrant activity leads to developmental disorders and cancer. To identify molecules that influence Notch signaling, we screened nearly 17,000 compounds using automated microscopy to monitor the trafficking and processing of a ligand-independent Notch-enhanced GFP (eGFP) reporter. Characterization of hits in vitro by biochemical and cellular assays and in vivo using zebrafish led to five validated compounds, four of which induced accumulation of the reporter at the plasma membrane by inhibiting γ-secretase. One compound, the dihydropyridine FLI-06, disrupted the Golgi apparatus in a manner distinct from that of brefeldin A and golgicide A. FLI-06 inhibited general secretion at a step before exit from the endoplasmic reticulum (ER), which was accompanied by a tubule-to-sheet morphological transition of the ER, rendering FLI-06 the first small molecule acting at such an early stage in secretory traffic. These data highlight the power of phenotypic screening to enable investigations of central cellular signaling pathways.


Subject(s)
Dihydropyridines/pharmacology , Endoplasmic Reticulum/drug effects , Receptors, Notch/antagonists & inhibitors , Secretory Pathway/drug effects , Signal Transduction/drug effects , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Dihydropyridines/chemistry , Endoplasmic Reticulum/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , HeLa Cells , Humans , Molecular Structure , Receptors, Notch/metabolism , Structure-Activity Relationship , Zebrafish/metabolism
16.
Brain ; 137(Pt 3): 683-92, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24459106

ABSTRACT

Many neurodegenerative disorders present with sensory loss. In the group of hereditary sensory and autonomic neuropathies loss of nociception is one of the disease hallmarks. To determine underlying factors of sensory neurodegeneration we performed whole-exome sequencing in affected individuals with the disorder. In a family with sensory neuropathy with loss of pain perception and destruction of the pedal skeleton we report a missense mutation in a highly conserved amino acid residue of atlastin GTPase 3 (ATL3), an endoplasmic reticulum-shaping GTPase. The same mutation (p.Tyr192Cys) was identified in a second family with similar clinical outcome by screening a large cohort of 115 patients with hereditary sensory and autonomic neuropathies. Both families show an autosomal dominant pattern of inheritance and the mutation segregates with complete penetrance. ATL3 is a paralogue of ATL1, a membrane curvature-generating molecule that is involved in spastic paraplegia and hereditary sensory neuropathy. ATL3 proteins are enriched in three-way junctions, branch points of the endoplasmic reticulum that connect membranous tubules to a continuous network. Mutant ATL3 p.Tyr192Cys fails to localize to branch points, but instead disrupts the structure of the tubular endoplasmic reticulum, suggesting that the mutation exerts a dominant-negative effect. Identification of ATL3 as novel disease-associated gene exemplifies that long-term sensory neuronal maintenance critically depends on the structural organisation of the endoplasmic reticulum. It emphasizes that alterations in membrane shaping-proteins are one of the major emerging pathways in axonal degeneration and suggests that this group of molecules should be considered in neuroprotective strategies.


Subject(s)
Bone Diseases/genetics , Endoplasmic Reticulum/genetics , GTP Phosphohydrolases/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Adult , Age of Onset , Bone Diseases/etiology , Bone Diseases/physiopathology , Cohort Studies , Cough/genetics , Cough/pathology , Cough/physiopathology , Endoplasmic Reticulum/pathology , Exome/genetics , Female , Fractures, Bone/genetics , Fractures, Bone/pathology , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/pathology , Gastroesophageal Reflux/physiopathology , Genes, Dominant/genetics , Haplotypes/genetics , Hereditary Sensory and Autonomic Neuropathies/complications , Hereditary Sensory and Autonomic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Intracellular Space/genetics , Male , Mutation , Mutation, Missense/genetics , Pedigree , Phenotype , Young Adult
17.
Proc Natl Acad Sci U S A ; 108(2): 621-5, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187406

ABSTRACT

The nicotinic acetylcholine receptor of skeletal muscle is composed of five subunits that are assembled in a stepwise manner. Quality control mechanisms ensure that only fully assembled receptors reach the cell surface. Here, we show that Rer1, a putative Golgi-ER retrieval receptor, is involved in the biogenesis of acetylcholine receptors. Rer1 is expressed in the early secretory pathway in the myoblast line C2C12 and in mouse skeletal muscle, and up-regulated during myogenesis. Upon down-regulation of Rer1 in C2C12 cells, unassembled acetylcholine receptor α-subunits escape from the ER and are transported to the plasma membrane and lysosomes, where they are degraded. As a result, the amount of fully assembled receptor at the cell surface is reduced. In vivo Rer1 knockdown and genetic inactivation of one Rer1 allele lead to significantly smaller neuromuscular junctions in mice. Our data show that Rer1 is a functionally important unique factor that controls surface expression of muscle acetylcholine receptors by localizing unassembled α-subunits to the early secretory pathway.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Membrane Glycoproteins/physiology , Muscles/metabolism , Receptors, Cholinergic/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Adaptor Proteins, Vesicular Transport , Alleles , Animals , Down-Regulation , Lysosomes/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Protein Transport , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
18.
J Neurosci ; 32(2): 738-45, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22238109

ABSTRACT

Disrupted-in-Schizophrenia 1 (DISC1) is a prominent susceptibility gene for major psychiatric disorders. Previous work indicated that DISC1 plays an important role during neuronal proliferation and differentiation in the cerebral cortex and that it affects the positioning of radial migrating pyramidal neurons. Here we show that in mice, DISC1 is necessary for the migration of the cortical interneurons generated in the medial ganglionic eminence (MGE). RT-PCR, in situ hybridizations, and immunocytochemical data revealed expression of DISC1 transcripts and protein in MGE-derived cells. To study the possible functional role of DISC1 during tangential migration, we performed in utero and ex utero electroporation to suppress DISC1 in the MGE in vivo and in vitro. Results indicate that after DISC1 knockdown, the proportion of tangentially migrating MGE neurons that reached their cortical target was strongly reduced. In addition, there were profound alterations in the morphology of DISC1-deficient neurons, which exhibited longer and less branched leading processes than control cells. These findings provide a possible link between clinical studies reporting alterations of cortical interneurons in schizophrenic patients and the current notion of schizophrenia as a neurodevelopmental disorder.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/embryology , Interneurons/physiology , Nerve Tissue Proteins/physiology , Telencephalon/embryology , Animals , Cerebral Cortex/abnormalities , Cerebral Cortex/physiology , Female , Ganglia/cytology , Ganglia/physiology , Interneurons/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Pregnancy , Primary Cell Culture , Telencephalon/abnormalities , Telencephalon/physiology
19.
Proc Natl Acad Sci U S A ; 107(5): 1942-7, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20133839

ABSTRACT

The formation of extracellular amyloid plaques is a common patho-biochemical event underlying several debilitating human conditions, including Alzheimer's disease (AD). Considerable evidence implies that AD damage arises primarily from small oligomeric amyloid forms of Abeta peptide, but the precise mechanism of pathogenicity remains to be established. Using a cell culture system that reproducibly leads to the formation of Alzheimer's Abeta amyloid plaques, we show here that the formation of a single amyloid plaque represents a template-dependent process that critically involves the presence of endocytosis- or phagocytosis-competent cells. Internalized Abeta peptide becomes sorted to multivesicular bodies where fibrils grow out, thus penetrating the vesicular membrane. Upon plaque formation, cells undergo cell death and intracellular amyloid structures become released into the extracellular space. These data imply a mechanism where the pathogenic activity of Abeta is attributed, at least in part, to intracellular aggregates.


Subject(s)
Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Freeze Fracturing , Humans , Intracellular Fluid/metabolism , Mice , Microscopy, Electron, Scanning , Microscopy, Video , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Plaque, Amyloid/chemistry , Plaque, Amyloid/ultrastructure
20.
Biomedicines ; 11(1)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36672654

ABSTRACT

Proteins of the secretory pathway undergo glycosylation in the endoplasmic reticulum (ER) and the Golgi apparatus. Altered protein glycosylation can manifest in serious, sometimes fatal malfunctions. We recently showed that mutations in GDP-mannose pyrophosphorylase A (GMPPA) can cause a syndrome characterized by alacrima, achalasia, mental retardation, and myopathic alterations (AAMR syndrome). GMPPA acts as a feedback inhibitor of GDP-mannose pyrophosphorylase B (GMPPB), which provides GDP-mannose as a substrate for protein glycosylation. Loss of GMPPA thus enhances the incorporation of mannose into glycochains of various proteins, including α-dystroglycan (α-DG), a protein that links the extracellular matrix with the cytoskeleton. Here, we further characterized the consequences of loss of GMPPA for the secretory pathway. This includes a fragmentation of the Golgi apparatus, which comes along with a regulation of the abundance of several ER- and Golgi-resident proteins. We further show that the activity of the Golgi-associated endoprotease furin is reduced. Moreover, the fraction of α-DG, which is retained in the ER, is increased. Notably, WT cells cultured at a high mannose concentration display similar changes with increased retention of α-DG, altered structure of the Golgi apparatus, and a decrease in furin activity. In summary, our data underline the importance of a balanced mannose homeostasis for the secretory pathway.

SELECTION OF CITATIONS
SEARCH DETAIL