Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Trends Biochem Sci ; 44(10): 827-836, 2019 10.
Article in English | MEDLINE | ID: mdl-31133362

ABSTRACT

The ability of mammalian cells to correctly identify and degrade misfolded secretory proteins, most of them bearing N-glycans, is crucial for their correct function and survival. An inefficient disposal mechanism results in the accumulation of misfolded proteins and consequent endoplasmic reticulum (ER) stress. N-glycan processing creates a code that reveals the folding status of each molecule, enabling continued folding attempts or targeting of the doomed glycoprotein for disposal. We review here the main steps involved in the accurate processing of unfolded glycoproteins. We highlight recent data suggesting that the processing is not stochastic, but that there is selective accelerated glycan trimming on misfolded glycoprotein molecules.


Subject(s)
Glycoproteins/metabolism , Endoplasmic Reticulum Stress , Glycoproteins/chemistry , Humans , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Folding
2.
J Biol Chem ; 297(5): 101299, 2021 11.
Article in English | MEDLINE | ID: mdl-34648767

ABSTRACT

The Sigma-1 receptor (S1R) is a transmembrane protein with important roles in cellular homeostasis in normal physiology and in disease. Especially in neurodegenerative diseases, S1R activation has been shown to provide neuroprotection by modulating calcium signaling, mitochondrial function and reducing endoplasmic reticulum (ER) stress. S1R missense mutations are one of the causes of the neurodegenerative Amyotrophic Lateral Sclerosis and distal hereditary motor neuronopathies. Although the S1R has been studied intensively, basic aspects remain controversial, such as S1R topology and whether it reaches the plasma membrane. To address these questions, we have undertaken several approaches. C-terminal tagging with a small biotin-acceptor peptide and BirA biotinylation in cells suggested a type II membrane orientation (cytosolic N-terminus). However, N-terminal tagging gave an equal probability for both possible orientations. This might explain conflicting reports in the literature, as tags may affect the protein topology. Therefore, we studied untagged S1R using a protease protection assay and a glycosylation mapping approach, introducing N-glycosylation sites. Both methods provided unambiguous results showing that the S1R is a type II membrane protein with a short cytosolic N-terminal tail. Assessments of glycan processing, surface fluorescence-activated cell sorting, and cell surface biotinylation indicated ER retention, with insignificant exit to the plasma membrane, in the absence or presence of S1R agonists or of ER stress. These findings may have important implications for S1R-based therapeutic approaches.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Receptors, sigma/metabolism , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , Receptors, sigma/genetics , Sigma-1 Receptor
3.
J Neurochem ; 158(2): 467-481, 2021 07.
Article in English | MEDLINE | ID: mdl-33871049

ABSTRACT

The endoplasmic reticulum (ER)-localized Sigma-1 receptor (S1R) is neuroprotective in models of neurodegenerative diseases, among them Huntington disease (HD). Recent clinical trials in HD patients and preclinical studies in cellular and mouse HD models suggest a therapeutic potential for the high-affinity S1R agonist pridopidine. However, the molecular mechanisms of the cytoprotective effect are unclear. We have previously reported strong induction of ER stress by toxic mutant huntingtin (mHtt) oligomers, which is reduced upon sequestration of these mHtt oligomers into large aggregates. Here, we show that pridopidine significantly ameliorates mHtt-induced ER stress in cellular HD models, starting at low nanomolar concentrations. Pridopidine reduced the levels of markers of the three branches of the unfolded protein response (UPR), showing the strongest effects on the PKR-like endoplasmic reticulum kinase (PERK) branch. The effect is S1R-dependent, as it is abolished in cells expressing mHtt in which the S1R was deleted using CRISPR/Cas9 technology. mHtt increased the level of the detergent-insoluble fraction of S1R, suggesting a compensatory cellular mechanism that responds to increased ER stress. Pridopidine further enhanced the levels of insoluble S1R, suggesting the stabilization of activated S1R oligomers. These S1R oligomeric species appeared in ER-localized patches, and not in the mitochondria-associated membranes nor the ER-derived quality control compartment. The colocalization of S1R with the chaperone BiP was significantly reduced by mHtt, and pridopidine restored this colocalization to normal, unstressed levels. Pridopidine increased toxic oligomeric mHtt recruitment into less toxic large sodium dodecyl sulfate-insoluble aggregates, suggesting that this in turn reduces ER stress and cytotoxicity.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Huntingtin Protein/genetics , Huntingtin Protein/toxicity , Piperidines/pharmacology , Receptors, sigma/drug effects , 3T3 Cells , Animals , CRISPR-Cas Systems , Endoplasmic Reticulum Chaperone BiP , Gene Knockout Techniques , HEK293 Cells , Heat-Shock Proteins , Humans , Mice , Mitochondrial Membranes/drug effects , Unfolded Protein Response/drug effects , Sigma-1 Receptor
4.
J Biol Chem ; 294(44): 15912-15913, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31676555

ABSTRACT

The generation of free N-glycans, or unconjugated oligosaccharides derived from N-linked glycoproteins, is well understood, but whether a similar fate awaits O-linked glycoprotein carbohydrates was unknown. Hirayama et al. now reveal, by using only mannose as an energy source, the generation of free O-glycans in Saccharomyces cerevisiae, in the lumen of a secretory compartment, possibly the vacuole. These findings uncover the presence of a possible regulated degradation pathway for O-mannosylated glycoproteins.


Subject(s)
Glycoproteins , Saccharomyces cerevisiae , Glycosylation , Mannose , Oligosaccharides , Polysaccharides
5.
Acta Neuropathol ; 140(5): 737-764, 2020 11.
Article in English | MEDLINE | ID: mdl-32642868

ABSTRACT

Impaired neuronal proteostasis is a salient feature of many neurodegenerative diseases, highlighting alterations in the function of the endoplasmic reticulum (ER). We previously reported that targeting the transcription factor XBP1, a key mediator of the ER stress response, delays disease progression and reduces protein aggregation in various models of neurodegeneration. To identify disease modifier genes that may explain the neuroprotective effects of XBP1 deficiency, we performed gene expression profiling of brain cortex and striatum of these animals and uncovered insulin-like growth factor 2 (Igf2) as the major upregulated gene. Here, we studied the impact of IGF2 signaling on protein aggregation in models of Huntington's disease (HD) as proof of concept. Cell culture studies revealed that IGF2 treatment decreases the load of intracellular aggregates of mutant huntingtin and a polyglutamine peptide. These results were validated using induced pluripotent stem cells (iPSC)-derived medium spiny neurons from HD patients and spinocerebellar ataxia cases. The reduction in the levels of mutant huntingtin was associated with a decrease in the half-life of the intracellular protein. The decrease in the levels of abnormal protein aggregation triggered by IGF2 was independent of the activity of autophagy and the proteasome pathways, the two main routes for mutant huntingtin clearance. Conversely, IGF2 signaling enhanced the secretion of soluble mutant huntingtin species through exosomes and microvesicles involving changes in actin dynamics. Administration of IGF2 into the brain of HD mice using gene therapy led to a significant decrease in the levels of mutant huntingtin in three different animal models. Moreover, analysis of human postmortem brain tissue and blood samples from HD patients showed a reduction in IGF2 level. This study identifies IGF2 as a relevant factor deregulated in HD, operating as a disease modifier that buffers the accumulation of abnormal protein species.


Subject(s)
Huntington Disease/metabolism , Huntington Disease/pathology , Insulin-Like Growth Factor II/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Insulin-Like Growth Factor II/pharmacology , Mice , Mice, Transgenic , Protein Aggregates/drug effects
6.
Semin Cell Dev Biol ; 41: 99-109, 2015 May.
Article in English | MEDLINE | ID: mdl-25460542

ABSTRACT

The internal environment of the eukaryotic cell is divided by membranes into various organelles, containing diverse functional subcompartments, which allow complex cellular life. The quality control of newly made secretory proteins relies on the ability of the endoplasmic reticulum (ER) to segregate and compartmentalize molecules at different folding states. These folding states are communicated by N-glycans present on most secretory proteins. In ER-associated degradation (ERAD), protein molecules that have been identified as terminally misfolded are sent for degradation at the cytosolic proteasomes after being dislocated from the ER to the cytosol. This review will focus on how misfolded glycoprotein molecules are segregated from their properly folded counterparts and targeted to ERAD. The pathway involves compartmentalization, which is intimately linked to differential N-glycan processing. Recent data suggests that these processes are very dynamic, and include transient assembly of ERAD machinery complexes.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Glycoproteins/metabolism , Polysaccharides/metabolism , Cell Compartmentation , Glycoproteins/chemistry , Humans , Models, Biological , Protein Folding , Protein Processing, Post-Translational , Protein Transport
7.
J Cell Sci ; 126(Pt 17): 3893-903, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23843619

ABSTRACT

The palmitoylation of calnexin serves to enrich calnexin on the mitochondria-associated membrane (MAM). Given a lack of information on the significance of this finding, we have investigated how this endoplasmic reticulum (ER)-internal sorting signal affects the functions of calnexin. Our results demonstrate that palmitoylated calnexin interacts with sarcoendoplasmic reticulum (SR) Ca(2+) transport ATPase (SERCA) 2b and that this interaction determines ER Ca(2+) content and the regulation of ER-mitochondria Ca(2+) crosstalk. In contrast, non-palmitoylated calnexin interacts with the oxidoreductase ERp57 and performs its well-known function in quality control. Interestingly, our results also show that calnexin palmitoylation is an ER-stress-dependent mechanism. Following a short-term ER stress, calnexin quickly becomes less palmitoylated, which shifts its function from the regulation of Ca(2+) signaling towards chaperoning and quality control of known substrates. These changes also correlate with a preferential distribution of calnexin to the MAM under resting conditions, or the rough ER and ER quality control compartment (ERQC) following ER stress. Our results have therefore identified the switch that assigns calnexin either to Ca(2+) signaling or to protein chaperoning.


Subject(s)
Calnexin/metabolism , Endoplasmic Reticulum Stress/physiology , Lipoylation/physiology , Mitochondrial Membranes/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , 3T3 Cells , Animals , Calcium/metabolism , Calcium Signaling , Cell Line , Endoplasmic Reticulum/metabolism , Fibroblasts , HEK293 Cells , HeLa Cells , Humans , Mice , Mitochondria/metabolism , Protein Disulfide-Isomerases/metabolism
8.
J Biol Chem ; 288(4): 2167-78, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23233672

ABSTRACT

Studies of misfolded protein targeting to endoplasmic reticulum-associated degradation (ERAD) have largely focused on glycoproteins, which include the bulk of the secretory proteins. Mechanisms of targeting of nonglycosylated proteins are less clear. Here, we studied three nonglycosylated proteins and analyzed their use of known glycoprotein quality control and ERAD components. Similar to an established glycosylated ERAD substrate, the uncleaved precursor of asialoglycoprotein receptor H2a, its nonglycosylated mutant, makes use of calnexin, EDEM1, and HRD1, but only glycosylated H2a is a substrate for the cytosolic SCF(Fbs2) E3 ubiquitin ligase with lectin activity. Two nonglycosylated BiP substrates, NS-1κ light chain and truncated Igγ heavy chain, interact with the ERAD complex lectins OS-9 and XTP3-B and require EDEM1 for degradation. EDEM1 associates through a region outside of its mannosidase-like domain with the nonglycosylated proteins. Similar to glycosylated substrates, proteasomal inhibition induced accumulation of the nonglycosylated proteins and ERAD machinery in the endoplasmic reticulum-derived quality control compartment. Our results suggest a shared ERAD pathway for glycosylated and nonglycosylated proteins composed of luminal lectin machinery components also capable of protein-protein interactions.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Animals , Chaperonins/chemistry , Cytosol/metabolism , Glycosylation , HEK293 Cells , Humans , Immunoglobulin kappa-Chains/chemistry , Lectins/chemistry , Mannosidases/chemistry , Mice , NIH 3T3 Cells , Polysaccharides/chemistry , Protein Denaturation , Protein Folding
9.
Neurotherapeutics ; 21(2): e00335, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38368172

ABSTRACT

There is currently no disease-modifying therapy for Huntington's disease (HD). We recently described a small molecule, MK-28, which restored homeostasis in HD models by specifically activating PKR-like ER kinase (PERK). This activation boosts the unfolded protein response (UPR), thereby reducing endoplasmic reticulum (ER) stress, a central cytotoxic mechanism in HD and other neurodegenerative diseases. Here, we have tested the long-term effects of MK-28 in HD model mice. R6/2 CAG (160) mice were treated by lifetime intraperitoneal injections 3 times a week. CatWalk measurements of motor function showed strong improvement compared to untreated mice after only two weeks of MK-28 treatment and continued with time, most significantly at 1 â€‹mg/kg MK-28, approaching WT values. Seven weeks treatment significantly improved paw grip strength. Body weight recovered and glucose levels, which are elevated in HD mice, were significantly reduced. Treatment with another PERK activator, CCT020312 at 1 â€‹mg/kg, also caused amelioration, consistent with PERK activation. Lifespan, measured in more resilient R6/2 CAG (120) mice with daily IP injection, was much extended by 16 days (20%) with 0.3 â€‹mg/kg MK-28, and by 38 days (46%) with 1 â€‹mg/kg MK-28. No toxicity, measured by weight, blood glucose levels and blood liver function markers, was detectable in WT mice treated for 6 weeks with 6 â€‹mg/kg MK-28. Boosting of PERK activity by long-term treatment with MK-28 could be a safe and promising therapeutic approach for HD.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Mice , Animals , Huntington Disease/metabolism , Mice, Transgenic , Disease Models, Animal , Endoplasmic Reticulum Stress
10.
iScience ; 26(3): 106232, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36876137

ABSTRACT

Misfolded proteins and components of the endoplasmic reticulum (ER) quality control and ER associated degradation (ERAD) machineries concentrate in mammalian cells in the pericentriolar ER-derived quality control compartment (ERQC), suggesting it as a staging ground for ERAD. By tracking the chaperone calreticulin and an ERAD substrate, we have now determined that the trafficking to the ERQC is reversible and recycling back to the ER is slower than the movement in the ER periphery. The dynamics suggest vesicular trafficking rather than diffusion. Indeed, using dominant negative mutants of ARF1 and Sar1 or the drugs Brefeldin A and H89, we observed that COPI inhibition causes accumulation in the ERQC and increases ERAD, whereas COPII inhibition has the opposite effect. Our results suggest that targeting of misfolded proteins to ERAD involves COPII-dependent transport to the ERQC and that they can be retrieved to the peripheral ER in a COPI-dependent manner.

11.
J Biol Chem ; 286(2): 1292-300, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21062743

ABSTRACT

Although the trimming of α1,2-mannose residues from precursor N-linked oligosaccharides is an essential step in the delivery of misfolded glycoproteins to endoplasmic reticulum (ER)-associated degradation (ERAD), the exact role of this trimming is unclear. EDEM1 was initially suggested to bind N-glycans after mannose trimming, a role presently ascribed to the lectins OS9 and XTP3-B, because of their in vitro affinities for trimmed oligosaccharides. We have shown before that ER mannosidase I (ERManI) is required for the trimming and concentrates together with the ERAD substrate and ERAD machinery in the pericentriolar ER-derived quality control compartment (ERQC). Inhibition of mannose trimming prevents substrate accumulation in the ERQC. Here, we show that the mannosidase inhibitor kifunensine or ERManI knockdown do not affect binding of an ERAD substrate glycoprotein to EDEM1. In contrast, substrate association with XTP3-B and with the E3 ubiquitin ligases HRD1 and SCF(Fbs2) was inhibited. Consistently, whereas the ERAD substrate partially colocalized upon proteasomal inhibition with EDEM1, HRD1, and Fbs2 at the ERQC, colocalization was repressed by mannosidase inhibition in the case of the E3 ligases but not for EDEM1. Interestingly, association and colocalization of the substrate with Derlin-1 was independent of mannose trimming. The HRD1 adaptor protein SEL1L had been suggested to play a role in N-glycan-dependent substrate delivery to OS9 and XTP3-B. However, substrate association with XTP3-B was still dependent on mannose trimming upon SEL1L knockdown. Our results suggest that mannose trimming enables delivery of a substrate glycoprotein from EDEM1 to late ERAD steps through association with XTP3-B.


Subject(s)
Endoplasmic Reticulum/metabolism , Glycoproteins/metabolism , Lectins/metabolism , Mannose/metabolism , Membrane Proteins/metabolism , Animals , Glycoproteins/chemistry , Glycosylation , HEK293 Cells , Humans , Lectins/chemistry , Mannosidases/metabolism , Membrane Proteins/chemistry , Mice , NIH 3T3 Cells , Neoplasm Proteins/metabolism , Protein Folding , Ubiquitin-Protein Ligases/metabolism
12.
Biomolecules ; 11(3)2021 02 26.
Article in English | MEDLINE | ID: mdl-33652720

ABSTRACT

With the extension of life span in recent decades, there is an increasing burden of late-onset neurodegenerative diseases, for which effective treatments are lacking. Neurodegenerative diseases include the widespread Alzheimer's disease (AD) and Parkinson's disease (PD), the less frequent Huntington's disease (HD) and Amyotrophic Lateral Sclerosis (ALS) and also rare early-onset diseases linked to mutations that cause protein aggregation or loss of function in genes that maintain protein homeostasis. The difficulties in applying gene therapy approaches to tackle these diseases is drawing increasing attention to strategies that aim to inhibit cellular toxicity and restore homeostasis by intervening in cellular pathways. These include the unfolded protein response (UPR), activated in response to endoplasmic reticulum (ER) stress, a cellular affliction that is shared by these diseases. Special focus is turned to the PKR-like ER kinase (PERK) pathway of the UPR as a target for intervention. However, the complexity of the pathway and its ability to promote cell survival or death, depending on ER stress resolution, has led to some confusion in conflicting studies. Both inhibition and activation of the PERK pathway have been reported to be beneficial in disease models, although there are also some reports where they are counterproductive. Although with the current knowledge a definitive answer cannot be given on whether it is better to activate or to inhibit the pathway, the most encouraging strategies appear to rely on boosting some steps without compromising downstream recovery.


Subject(s)
Neurodegenerative Diseases/metabolism , eIF-2 Kinase/metabolism , Animals , Endoplasmic Reticulum/metabolism , Humans , Signal Transduction/physiology , Unfolded Protein Response/physiology
13.
Trends Biochem Sci ; 30(6): 297-303, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950873

ABSTRACT

Of the many post-translational modifications of proteins, ubiquitination and N-glycosylation stand out because they are polymeric additions. In contrast to single-unit modifications, the fate of the modified protein is determined by the dynamic equilibrium of polymerization versus depolymerization, rather than by the initial addition itself. Notably, it is the trimming of sugar chains and elongation of polyubiquitin that target the protein to degradation. Recent research suggests that, for each process, special receptors recognize chains that reach an appropriate length and commit the conjugated substrate for proteasomal disposal. We propose that the 'magic numbers' are loss of at least three mannose residues from the initial chain, or extension to at least four ubiquitins. Although these processes are compartmentalized to either side of the endoplasmic reticulum (ER) membrane, some proteins are sequentially subjected to both because they transverse this membrane for ER-associated degradation.


Subject(s)
Carbohydrate Metabolism , Endoplasmic Reticulum/metabolism , Proteins/metabolism , Ubiquitin/metabolism , Calnexin/metabolism , Carbohydrate Sequence , Glycoproteins/metabolism , Glycosylation , Mannose/metabolism , Membrane Proteins/metabolism , Molecular Sequence Data
14.
Cells ; 9(9)2020 09 22.
Article in English | MEDLINE | ID: mdl-32971745

ABSTRACT

N-linked glycosylation and sugar chain processing, as well as disulfide bond formation, are among the most common post-translational protein modifications taking place in the endoplasmic reticulum (ER). They are essential modifications that are required for membrane and secretory proteins to achieve their correct folding and native structure. Several oxidoreductases responsible for disulfide bond formation, isomerization, and reduction have been shown to form stable, functional complexes with enzymes and chaperones that are involved in the initial addition of an N-glycan and in folding and quality control of the glycoproteins. Some of these oxidoreductases are selenoproteins. Recent studies also implicate glycan machinery-oxidoreductase complexes in the recognition and processing of misfolded glycoproteins and their reduction and targeting to ER-associated degradation. This review focuses on the intriguing cooperation between the glycoprotein-specific cell machineries and ER oxidoreductases, and highlights open questions regarding the functions of many members of this large family.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/genetics , Molecular Chaperones/metabolism , Oxidoreductases/metabolism , Polysaccharides/metabolism , Protein Processing, Post-Translational , Selenoproteins/metabolism , Calnexin/genetics , Calnexin/metabolism , Disulfides/metabolism , Endoplasmic Reticulum/enzymology , Eukaryotic Cells/cytology , Eukaryotic Cells/enzymology , Glycosylation , Humans , Mannosidases/genetics , Mannosidases/metabolism , Molecular Chaperones/genetics , Oxidation-Reduction , Oxidoreductases/genetics , Protein Folding , Selenoproteins/genetics
15.
Sci Rep ; 10(1): 6875, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327686

ABSTRACT

One of the pathways of the unfolded protein response, initiated by PKR-like endoplasmic reticulum kinase (PERK), is key to neuronal homeostasis in neurodegenerative diseases. PERK pathway activation is usually accomplished by inhibiting eIF2α-P dephosphorylation, after its phosphorylation by PERK. Less tried is an approach involving direct PERK activation without compromising long-term recovery of eIF2α function by dephosphorylation. Here we show major improvement in cellular (STHdhQ111/111) and mouse (R6/2) Huntington's disease (HD) models using a potent small molecule PERK activator that we developed, MK-28. MK-28 showed PERK selectivity in vitro on a 391-kinase panel and rescued cells (but not PERK-/- cells) from ER stress-induced apoptosis. Cells were also rescued by the commercial PERK activator CCT020312 but MK-28 was significantly more potent. Computational docking suggested MK-28 interaction with the PERK activation loop. MK-28 exhibited remarkable pharmacokinetic properties and high BBB penetration in mice. Transient subcutaneous delivery of MK-28 significantly improved motor and executive functions and delayed death onset in R6/2 mice, showing no toxicity. Therefore, PERK activation can treat a most aggressive HD model, suggesting a possible approach for HD therapy and worth exploring for other neurodegenerative disorders.


Subject(s)
Enzyme Activators/pharmacology , Huntington Disease/enzymology , eIF-2 Kinase/metabolism , Animals , Apoptosis/drug effects , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Enzyme Activators/chemistry , Eukaryotic Initiation Factor-2/metabolism , Huntingtin Protein/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Mice , Models, Biological , Neostriatum/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects , Survival Analysis
16.
Front Mol Biosci ; 6: 20, 2019.
Article in English | MEDLINE | ID: mdl-31001537

ABSTRACT

Increasing evidence in recent years indicates that protein misfolding and aggregation, leading to ER stress, are central factors of pathogenicity in neurodegenerative diseases. This is particularly true in Huntington's disease (HD), where in contrast with other disorders, the cause is monogenic. Mutant huntingtin interferes with many cellular processes, but the fact that modulation of ER stress and of the unfolded response pathways reduces the toxicity, places these mechanisms at the core and gives hope for potential therapeutic approaches. There is currently no effective treatment for HD and it has a fatal outcome a few years after the start of symptoms of cognitive and motor impairment. Here we will discuss recent findings that shed light on the mechanisms of protein misfolding and aggregation that give origin to ER stress in neurodegenerative diseases, focusing on Huntington's disease, on the cellular response and on how to use this knowledge for possible therapeutic strategies.

17.
Biochem J ; 404(3): 509-16, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17338678

ABSTRACT

The UPR (unfolded protein response) activates transcription of genes involved in proteasomal degradation. However, we found that in its early stages the UPR leads to a transient inhibition of proteasomal disposal of cytosolic substrates (p53 and p27kip1) and of those targeted to ER (endoplasmic reticulum)-associated degradation (uncleaved precursor of asialoglycoprotein receptor H2a). Degradation resumed soon after the protein synthesis arrest that occurs in early UPR subsided. Consistent with this, protein synthesis inhibitors blocked ubiquitin/proteasomal degradation. Ubiquitination was inhibited during the translation block, suggesting short-lived E3 ubiquitin ligases as candidate depleted proteins. This was indeed the case for p53 whose E3 ligase, Mdm2 (murine double minute 2), when overexpressed, restored the degradation, whereas a mutant Mdm2 in its acidic domain restored the ubiquitination but did not completely restore the degradation. Inhibition of proteasomal degradation early in UPR may prevent depletion of essential short-lived factors during the translation arrest. Stabilization of p27 through this mechanism may explain the cell cycle arrest in G1 when translation is blocked by inhibitors or by the UPR.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis , Protein Conformation , Animals , Asialoglycoprotein Receptor/genetics , Asialoglycoprotein Receptor/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cycloheximide/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mice , NIH 3T3 Cells , Protein Folding , Protein Synthesis Inhibitors/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism
18.
Commun Biol ; 1: 172, 2018.
Article in English | MEDLINE | ID: mdl-30374462

ABSTRACT

Extensive mannose trimming of nascent glycoprotein N-glycans signals their targeting to endoplasmic reticulum-associated degradation (ERAD). ER mannosidase I (ERManI) and the EDEM protein family participate in this process. However, whether the EDEMs are truly mannosidases can be addressed only by measuring mannosidase activity in vitro. Here, we reveal EDEM1 and EDEM2 mannosidase activities in vitro. Whereas ERManI significantly trims free N-glycans, activity of the EDEMs is modest on free oligosaccharides and on glycoproteins. However, mannosidase activity of ERManI and the EDEMs is significantly higher on a denatured glycoprotein. The EDEMs associate with oxidoreductases, protein disulfide isomerase, and especially TXNDC11, enhancing mannosidase activity on glycoproteins but not on free N-glycans. The finding that substrate unfolded status increases mannosidase activity solves an important conundrum, as current models suggest general slow mannose trimming. As we show, misfolded or unfolded glycoproteins are subject to differentially faster trimming (and targeting to ERAD) than well-folded ones.

19.
Cell Stress Chaperones ; 12(4): 373-83, 2007.
Article in English | MEDLINE | ID: mdl-18229456

ABSTRACT

Inhibition of protein folding in the endoplasmic reticulum (ER) causes ER stress, which triggers the unfolded protein response (UPR). To decrease the biosynthetic burden on the ER, the UPR inhibits in its initial stages protein synthesis. At later stages it upregulates components of ER-associated degradation (ERAD) and of the ubiquitin/proteasome system, which targets ER as well as cytosolic proteins for disposal. Here we report that, at later stages, the UPR also activates an alternative nonproteasomal pathway of degradation, which is resistant to proteasome inhibitors and is specific for ER substrates (assessed with uncleaved precursor of asialoglycoprotein receptor H2a and unassembled CD3delta) and not for cytosolic ones (p53). To mimic the initial inhibition of translation during UPR, we incubated cells with cycloheximide. After this treatment, degradation of ERAD substrates was no longer effected by proteasomal inhibition, similarly to the observed outcome of UPR. The degradation also became insensitive to abrogation of ubiquitination in a cell line carrying a thermosensitive E1 ubiquitin activating enzyme mutant. Of all protease inhibitors tested, only the metal chelator o-phenanthroline could block this nonproteasomal degradation. Preincubation of o-phenanthroline with Mn2+ or Co2+, but not with other cations, reversed the inhibition. Our results suggest that, upon inhibition of translation, an alternative nonproteasomal pathway is activated for degradation of proteins from the ER. This involves a Mn2+/Co2+-dependent metalloprotease or other metalloprotein. The alternative pathway selectively targets ERAD substrates to reduce the ER burden, but does not affect p53, the levels of which remain dependent on proteasomal control.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Folding , Protein Processing, Post-Translational , Proteins/metabolism , Animals , CHO Cells , Cobalt , Cricetinae , Cricetulus , Cycloheximide/pharmacology , Cytosol/drug effects , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Manganese , Metalloproteins/metabolism , Mice , Models, Biological , Mutation/genetics , NIH 3T3 Cells , Phenanthrolines/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , Temperature , Tumor Suppressor Protein p53/metabolism , Ubiquitination/drug effects
20.
Mol Biol Cell ; 15(5): 2133-42, 2004 May.
Article in English | MEDLINE | ID: mdl-14978212

ABSTRACT

The thiol oxidoreductase endoplasmic reticulum (ER)p57 interacts with newly synthesized glycoproteins through ternary complexes with the chaperones/lectins calnexin or calreticulin. On proteasomal inhibition calnexin and calreticulin concentrate in the pericentriolar endoplasmic reticulum-derived quality control compartment that we recently described. Surprisingly, ERp57 remained in an endoplasmic reticulum pattern. Using asialoglycoprotein receptor H2a and H2b as models, we determined in pulse-chase experiments that both glycoproteins initially bind to calnexin and ERp57. However, H2b, which will exit to the Golgi, dissociated from calnexin and remained bound for a longer period to ERp57, whereas the opposite was true for the endoplasmic reticulum-associated degradation substrate H2a that will go to the endoplasmic reticulum-derived quality control compartment. At 15 degrees C, ERp57 colocalized with H2b adjacent to an endoplasmic reticulum-Golgi intermediate compartment marker. Posttranslational inhibition of glucose excision prolonged association of H2a precursor to calnexin but not to ERp57. Preincubation with a low concentration (15 microg/ml) of the glucosidase inhibitor castanospermine prevented the association of H2a to ERp57 but not to calnexin. This low concentration of castanospermine accelerated the degradation of H2a, suggesting that ERp57 protects the glycoprotein from degradation and not calnexin. Our results suggest an early chaperone-mediated sorting event with calnexin being involved in the quality control retention of molecules bound for endoplasmic reticulum-associated degradation and ERp57 giving initial protection from degradation and later assisting the maturation of molecules that will exit to the Golgi.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Calnexin/physiology , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/physiology , Isomerases/physiology , Animals , Calnexin/metabolism , Glucose/analysis , Glucose/metabolism , Golgi Apparatus/metabolism , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Indolizines/pharmacology , Isomerases/antagonists & inhibitors , Isomerases/metabolism , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Mice , NIH 3T3 Cells , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Disulfide-Isomerases , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL