Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38836811

ABSTRACT

During ER-associated decay, unfolded membrane-resident proteins are targeted for removal and degradation by ubiquitin ligases whose identities and precise operations remain unclear. In this issue, Guerriero and Brodsky discuss new results from Kamada et al. (https://doi.org/10.1083/jcb.202308003) showing the clearance of misfolded CFTR by the E3 ligase HERC3.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Membrane Proteins , Ubiquitination , Humans , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Membrane Proteins/chemistry , Protein Folding , Ubiquitin-Protein Ligases/metabolism
2.
Mol Biol Cell ; 35(4): ar59, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38446639

ABSTRACT

GRP170 (Hyou1) is required for mouse embryonic development, and its ablation in kidney nephrons leads to renal failure. Unlike most chaperones, GRP170 is the lone member of its chaperone family in the ER lumen. However, the cellular requirement for GRP170, which both binds nonnative proteins and acts as nucleotide exchange factor for BiP, is poorly understood. Here, we report on the isolation of mouse embryonic fibroblasts obtained from mice in which LoxP sites were engineered in the Hyou1 loci (Hyou1LoxP/LoxP). A doxycycline-regulated Cre recombinase was stably introduced into these cells. Induction of Cre resulted in depletion of Grp170 protein which culminated in cell death. As Grp170 levels fell we observed a portion of BiP fractionating with insoluble material, increased binding of BiP to a client with a concomitant reduction in its turnover, and reduced solubility of an aggregation-prone BiP substrate. Consistent with disrupted BiP functions, we observed reactivation of BiP and induction of the unfolded protein response (UPR) in futile attempts to provide compensatory increases in ER chaperones and folding enzymes. Together, these results provide insights into the cellular consequences of controlled Grp170 loss and provide hypotheses as to why mutations in the Hyou1 locus are linked to human disease.


Subject(s)
Embryonic Development , Endoplasmic Reticulum Chaperone BiP , HSP70 Heat-Shock Proteins , Animals , Humans , Mice , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Molecular Chaperones/metabolism
3.
bioRxiv ; 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37905119

ABSTRACT

GRP170, a product of the Hyou1 gene, is required for mouse embryonic development, and its ablation in kidney nephrons leads to renal failure. Unlike most chaperones, GRP170 is the lone member of its chaperone family in the ER lumen. However, the cellular requirement for GRP170, which both binds non-native proteins and acts as nucleotide exchange factor for BiP, is poorly understood. Here, we report on the isolation of embryonic fibroblasts from mice in which LoxP sites were engineered in the Hyou1 loci ( Hyou1 LoxP/LoxP ). A doxycycline-regulated Cre recombinase was also stably introduced into these cells. Induction of Cre resulted in excision of Hyou1 and depletion of Grp170 protein, culminating in apoptotic cell death. As Grp170 levels fell we observed increased steady-state binding of BiP to a client, slowed degradation of a misfolded BiP substrate, and BiP accumulation in NP40-insoluble fractions. Consistent with disrupted BiP functions, we observed reactivation of BiP storage pools and induction of the unfolded protein response (UPR) in futile attempts to provide compensatory increases in ER chaperones and folding enzymes. Together, these results provide insights into the cellular consequences of controlled Grp170 loss and insights into mutations in the Hyou1 locus and human disease.

4.
Biochem J ; 480(18): 1459-1473, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37702403

ABSTRACT

Transmembrane proteins have unique requirements to fold and integrate into the endoplasmic reticulum (ER) membrane. Most notably, transmembrane proteins must fold in three separate environments: extracellular domains fold in the oxidizing environment of the ER lumen, transmembrane domains (TMDs) fold within the lipid bilayer, and cytosolic domains fold in the reducing environment of the cytosol. Moreover, each region is acted upon by a unique set of chaperones and monitored by components of the ER associated quality control machinery that identify misfolded domains in each compartment. One factor is the ER lumenal Hsp70-like chaperone, Lhs1. Our previous work established that Lhs1 is required for the degradation of the unassembled α-subunit of the epithelial sodium channel (αENaC), but not the homologous ß- and γENaC subunits. However, assembly of the ENaC heterotrimer blocked the Lhs1-dependent ER associated degradation (ERAD) of the α-subunit, yet the characteristics that dictate the specificity of Lhs1-dependent ERAD substrates remained unclear. We now report that Lhs1-dependent substrates share a unique set of features. First, all Lhs1 substrates appear to be unglycosylated, and second they contain two TMDs. Each substrate also contains orphaned or unassembled TMDs. Additionally, interfering with inter-subunit assembly of the ENaC trimer results in Lhs1-dependent degradation of the entire complex. Finally, our work suggests that Lhs1 is required for a subset of ERAD substrates that also require the Hrd1 ubiquitin ligase. Together, these data provide hints as to the identities of as-yet unconfirmed substrates of Lhs1 and potentially of the Lhs1 homolog in mammals, GRP170.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum , Animals , Cytosol , Lipid Bilayers , Membrane Proteins/genetics , Mammals
5.
Cell Chem Biol ; 29(8): 1303-1316.e3, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35830852

ABSTRACT

The potential of small molecules to localize within subcellular compartments is rarely explored. To probe this question, we measured the localization of Hsp70 inhibitors using fluorescence microscopy. We found that even closely related analogs had dramatically different distributions, with some residing predominantly in the mitochondria and others in the ER. CRISPRi screens supported this idea, showing that different compounds had distinct chemogenetic interactions with Hsp70s of the ER (HSPA5/BiP) and mitochondria (HSPA9/mortalin) and their co-chaperones. Moreover, localization seemed to determine function, even for molecules with conserved binding sites. Compounds with distinct partitioning have distinct anti-proliferative activity in breast cancer cells compared with anti-viral activity in cellular models of Dengue virus replication, likely because different sets of Hsp70s are required in these processes. These findings highlight the contributions of subcellular partitioning and chemogenetic interactions to small molecule activity, features that are rarely explored during medicinal chemistry campaigns.


Subject(s)
HSP70 Heat-Shock Proteins , Molecular Chaperones , Binding Sites , Endoplasmic Reticulum Chaperone BiP , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Domains
6.
Cell Rep ; 36(12): 109717, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551305

ABSTRACT

To maintain secretory pathway fidelity, misfolded proteins are commonly retained in the endoplasmic reticulum (ER) and selected for ER-associated degradation (ERAD). Soluble misfolded proteins use ER chaperones for retention, but the machinery that restricts aberrant membrane proteins to the ER is unclear. In fact, some misfolded membrane proteins escape the ER and traffic to the lysosome/vacuole. To this end, we describe a model substrate, SZ∗, that contains an ER export signal but is also targeted for ERAD. We observe decreased ER retention when chaperone-dependent SZ∗ ubiquitination is compromised. In addition, appending a linear tetra-ubiquitin motif onto SZ∗ overrides ER export. By screening known ubiquitin-binding proteins, we then positively correlate SZ∗ retention with Ubx2 binding. Deletion of Ubx2 also inhibits the retention of another misfolded membrane protein. Our results indicate that polyubiquitination is sufficient to retain misfolded membrane proteins in the ER prior to ERAD.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , HSP40 Heat-Shock Proteins/metabolism , Leupeptins/pharmacology , Membrane Proteins/chemistry , Protein Binding , Protein Folding , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity , Ubiquitin/metabolism , Ubiquitination
7.
FEBS Lett ; 595(18): 2383-2394, 2021 09.
Article in English | MEDLINE | ID: mdl-34358326

ABSTRACT

Maintenance of the proteome (proteostasis) is essential for cellular homeostasis and prevents cytotoxic stress responses that arise from protein misfolding. However, little is known about how different types of misfolded proteins impact homeostasis, especially when protein degradation pathways are compromised. We examined the effects of misfolded protein expression on yeast growth by characterizing a suite of substrates possessing the same aggregation-prone domain but engaging different quality control pathways. We discovered that treatment with a proteasome inhibitor was more toxic in yeast expressing misfolded membrane proteins, and this growth defect was mirrored in yeast lacking a proteasome-specific transcription factor, Rpn4p. These results highlight weaknesses in the proteostasis network's ability to handle the stress arising from an accumulation of misfolded membrane proteins.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Protein Folding , Saccharomyces cerevisiae Proteins/classification , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Cell Growth Processes/drug effects , Cytoplasm/metabolism , DNA-Binding Proteins/deficiency , Endoplasmic Reticulum-Associated Degradation , Heat-Shock Proteins/metabolism , Nucleotides/metabolism , Proteasome Inhibitors/pharmacology , Protein Binding , Protein Domains , Proteolysis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Transcription Factors/deficiency
8.
Biophys J ; 117(4): 668-678, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31399214

ABSTRACT

Membrane proteins must adopt their proper topologies within biological membranes, but achieving the correct topology is compromised by the presence of marginally hydrophobic transmembrane helices (TMHs). In this study, we report on a new model membrane protein in yeast that harbors two TMHs fused to an unstable nucleotide-binding domain. Because the second helix (TMH2) in this reporter has an unfavorable predicted free energy of insertion, we employed established methods to generate variants that alter TMH2 insertion free energy. We first found that altering TMH2 did not significantly affect the extent of protein degradation by the cellular quality control machinery. Next, we correlated predicted insertion free energies from a knowledge-based energy scale with the measured apparent free energies of TMH2 insertion. Although the predicted and apparent insertion energies showed a similar trend, the predicted free-energy changes spanned an unanticipated narrow range. By instead using a physics-based model, we obtained a broader range of free energies that agreed considerably better with the magnitude of the experimentally derived values. Nevertheless, some variants still inserted better in yeast than predicted from energy-based scales. Therefore, molecular dynamics simulations were performed and indicated that the corresponding mutations induced conformational changes within TMH2, which altered the number of stabilizing hydrogen bonds. Together, our results offer insight into the ability of the cellular quality control machinery to recognize conformationally distinct misfolded topomers, provide a model to assess TMH insertion in vivo, and indicate that TMH insertion energy scales may be limited depending on the specific protein and the mutation present.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Cell Membrane/chemistry , Molecular Dynamics Simulation , Saccharomyces cerevisiae Proteins/chemistry , ATP-Binding Cassette Transporters/metabolism , Cell Membrane/metabolism , Protein Domains , Protein Folding , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism
9.
Protein Sci ; 28(7): 1290-1306, 2019 07.
Article in English | MEDLINE | ID: mdl-31050848

ABSTRACT

Misfolded proteins in the endoplasmic reticulum (ER) are selected for ER-associated degradation (ERAD). More than 60 disease-associated proteins are substrates for the ERAD pathway due to the presence of missense or nonsense mutations. In yeast, the Hsp104 molecular chaperone disaggregates detergent-insoluble ERAD substrates, but the spectrum of disease-associated ERAD substrates that may be aggregation prone is unknown. To determine if Hsp104 recognizes aggregation-prone ERAD substrates associated with human diseases, we developed yeast expression systems for a hydrophobic lipid-binding protein, apolipoprotein B (ApoB), along with a chimeric protein harboring a nucleotide-binding domain from the cystic fibrosis transmembrane conductance regulator (CFTR) into which disease-causing mutations were introduced. We discovered that Hsp104 facilitates the degradation of ER-associated ApoB as well as a truncated CFTR chimera in which a premature stop codon corresponds to a disease-causing mutation. Chimeras containing a wild-type version of the CFTR domain or a different mutation were stable and thus Hsp104 independent. We also discovered that the detergent solubility of the unstable chimera was lower than the stable chimeras, and Hsp104 helped retrotranslocate the unstable chimera from the ER, consistent with disaggregase activity. To determine why the truncated chimera was unstable, we next performed molecular dynamics simulations and noted significant unraveling of the CFTR nucleotide-binding domain. Because human cells lack Hsp104, these data indicate that an alternate disaggregase or mechanism facilitates the removal of aggregation-prone, disease-causing ERAD substrates in their native environments.


Subject(s)
Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum-Associated Degradation , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Dynamics Simulation , Mutation , Protein Aggregates , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
10.
Article in English | MEDLINE | ID: mdl-30670468

ABSTRACT

Misfolded proteins compromise cellular homeostasis. This is especially problematic in the endoplasmic reticulum (ER), which is a high-capacity protein-folding compartment and whose function requires stringent protein quality-control systems. Multiprotein complexes in the ER are able to identify, remove, ubiquitinate, and deliver misfolded proteins to the 26S proteasome for degradation in the cytosol, and these events are collectively termed ER-associated degradation, or ERAD. Several steps in the ERAD pathway are facilitated by molecular chaperone networks, and the importance of ERAD is highlighted by the fact that this pathway is linked to numerous protein conformational diseases. In this review, we discuss the factors that constitute the ERAD machinery and detail how each step in the pathway occurs. We then highlight the underlying pathophysiology of protein conformational diseases associated with ERAD.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Conformation , Proteostasis Deficiencies/pathology , Cytoplasm/metabolism , Genome, Human , Homeostasis , Humans , Mutation , Peptides/metabolism , Polysaccharides/chemistry , Protein Folding , Proteostasis , Quality Control
11.
J Cell Sci ; 131(17)2018 09 05.
Article in English | MEDLINE | ID: mdl-30131440

ABSTRACT

Cancer cells thrive when challenged with proteotoxic stress by inducing components of the protein folding, proteasome, autophagy and unfolded protein response (UPR) pathways. Consequently, specific molecular chaperones have been validated as targets for anti-cancer therapies. For example, inhibition of Hsp70 family proteins (hereafter Hsp70) in rhabdomyosarcoma triggers UPR induction and apoptosis. To define how these cancer cells respond to compromised proteostasis, we compared rhabdomyosarcoma cells that were sensitive (RMS13) or resistant (RMS13-R) to the Hsp70 inhibitor MAL3-101. We discovered that endoplasmic reticulum-associated degradation (ERAD) and autophagy were activated in RMS13-R cells, suggesting that resistant cells overcome Hsp70 ablation by increasing misfolded protein degradation. Indeed, RMS13-R cells degraded ERAD substrates more rapidly than RMS cells and induced the autophagy pathway. Surprisingly, inhibition of the proteasome or ERAD had no effect on RMS13-R cell survival, but silencing of select autophagy components or treatment with autophagy inhibitors restored MAL3-101 sensitivity and led to apoptosis. These data indicate a route through which cancer cells overcome a chaperone-based therapy, define how cells can adapt to Hsp70 inhibition, and demonstrate the value of combined chaperone and autophagy-based therapies.This article has an associated First Person interview with the first author of the paper.


Subject(s)
HSP70 Heat-Shock Proteins/antagonists & inhibitors , Proteostasis , Rhabdomyosarcoma/physiopathology , Apoptosis , Autophagy , Cell Line, Tumor , Endoplasmic Reticulum-Associated Degradation , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Proteasome Endopeptidase Complex/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Small Molecule Libraries/pharmacology , Unfolded Protein Response
12.
Mol Cell ; 70(2): 242-253.e6, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677492

ABSTRACT

Misfolded proteins in the endoplasmic reticulum (ER) are destroyed by ER-associated degradation (ERAD). Although the retrotranslocation of misfolded proteins from the ER has been reconstituted, how a polypeptide is initially selected for ERAD remains poorly defined. To address this question while controlling for the diverse nature of ERAD substrates, we constructed a series of truncations in a single ER-tethered domain. We observed that the truncated proteins exhibited variable degradation rates and discovered a positive correlation between ERAD substrate instability and detergent insolubility, which demonstrates that aggregation-prone species can be selected for ERAD. Further, Hsp104 facilitated degradation of an insoluble species, consistent with the chaperone's disaggregase activity. We also show that retrotranslocation of the ubiquitinated substrate from the ER was inhibited in the absence of Hsp104. Therefore, chaperone-mediated selection frees the ER membrane of potentially toxic, aggregation-prone species.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/enzymology , Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Heat-Shock Proteins/genetics , Protein Aggregates , Protein Aggregation, Pathological , Protein Folding , Protein Transport , Proteolysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Solubility , Substrate Specificity , Ubiquitination
13.
Mol Biol Cell ; 28(15): 2076-2090, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28539401

ABSTRACT

Integral membrane proteins fold inefficiently and are susceptible to turnover via the endoplasmic reticulum-associated degradation (ERAD) pathway. During ERAD, misfolded proteins are recognized by molecular chaperones, polyubiquitinated, and retrotranslocated to the cytoplasm for proteasomal degradation. Although many aspects of this pathway are defined, how transmembrane helices (TMHs) are removed from the membrane and into the cytoplasm before degradation is poorly understood. In this study, we asked whether the hydrophobic character of a TMH acts as an energetic barrier to retrotranslocation. To this end, we designed a dual-pass model ERAD substrate, Chimera A*, which contains the cytoplasmic misfolded domain from a characterized ERAD substrate, Sterile 6* (Ste6p*). We found that the degradation requirements for Chimera A* and Ste6p* are similar, but Chimera A* was retrotranslocated more efficiently than Ste6p* in an in vitro assay in which retrotranslocation can be quantified. We then constructed a series of Chimera A* variants containing synthetic TMHs with a range of ΔG values for membrane insertion. TMH hydrophobicity correlated inversely with retrotranslocation efficiency, and in all cases, retrotranslocation remained Cdc48p dependent. These findings provide insight into the energetic restrictions on the retrotranslocation reaction, as well as a new computational approach to predict retrotranslocation efficiency.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Membrane Proteins/metabolism , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Endoplasmic Reticulum/metabolism , Hydrophobic and Hydrophilic Interactions , Membranes/metabolism , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Folding , Protein Translocation Systems/metabolism , Protein Transport , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
14.
Proc Natl Acad Sci U S A ; 113(32): 9015-20, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27450084

ABSTRACT

Cytosolic and organelle-based heat-shock protein (HSP) chaperones ensure proper folding and function of nascent and injured polypeptides to support cell growth. Under conditions of cellular stress, including oncogenic transformation, proteostasis components maintain homeostasis and prevent apoptosis. Although this cancer-relevant function has provided a rationale for therapeutically targeting proteostasis regulators (e.g., HSP90), cancer-subtype dependencies upon particular proteostasis components are relatively undefined. Here, we show that human rhabdomyosarcoma (RMS) cells, but not several other cancer cell types, depend upon heat-shock protein 70 kDA (HSP70) for survival. HSP70-targeted therapy (but not chemotherapeutic agents) promoted apoptosis in RMS cells by triggering an unfolded protein response (UPR) that induced PRKR-like endoplasmic reticulum kinase (PERK)-eukaryotic translation initiation factor α (eIF2α)-CEBP homologous protein (CHOP) signaling and CHOP-mediated cell death. Intriguingly, inhibition of only cytosolic HSP70 induced the UPR, suggesting that the essential activity of HSP70 in RMS cells lies at the endoplasmic reticulum-cytosol interface. We also found that increased CHOP mRNA in clinical specimens was a biomarker for poor outcomes in chemotherapy-treated RMS patients. The data suggest that, like human epidermal growth factor receptor 2 (HER2) amplification in breast cancer, increased CHOP in RMS is a biomarker of decreased response to chemotherapy but enhanced response to targeted therapy. Our findings identify the cytosolic HSP70-UPR axis as an unexpected regulator of RMS pathogenesis, revealing HSP70-targeted therapy as a promising strategy to engage CHOP-mediated apoptosis and improve RMS treatment. Our study highlights the utility of dissecting cancer subtype-specific dependencies on proteostasis networks to uncover unanticipated cancer vulnerabilities.


Subject(s)
HSP70 Heat-Shock Proteins/physiology , Rhabdomyosarcoma/etiology , Apoptosis , Cell Line, Tumor , Cell Survival , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Humans , PAX3 Transcription Factor/physiology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Transcription Factor CHOP/physiology , Unfolded Protein Response
15.
Structure ; 23(8): 1526-1537, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26118532

ABSTRACT

The electrostatic properties of membrane proteins often reveal many of their key biophysical characteristics, such as ion channel selectivity and the stability of charged membrane-spanning segments. The Poisson-Boltzmann (PB) equation is the gold standard for calculating protein electrostatics, and the software APBSmem enables the solution of the PB equation in the presence of a membrane. Here, we describe significant advances to APBSmem, including full automation of system setup, per-residue energy decomposition, incorporation of PDB2PQR, calculation of membrane-induced pKa shifts, calculation of non-polar energies, and command-line scripting for large-scale calculations. We highlight these new features with calculations carried out on a number of membrane proteins, including the recently solved structure of the ion channel TRPV1 and a large survey of 1,614 membrane proteins of known structure. This survey provides a comprehensive list of residues with large electrostatic penalties for being embedded in the membrane, potentially revealing interesting functional information.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Molecular Dynamics Simulation , Saccharomyces cerevisiae Proteins/chemistry , Sodium Channels/chemistry , Software , TRPV Cation Channels/chemistry , Voltage-Dependent Anion Channel 1/chemistry , Amino Acid Motifs , Data Mining , Humans , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Static Electricity , Structural Homology, Protein , Thermodynamics
16.
Mol Biol Cell ; 25(2): 276-89, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227888

ABSTRACT

Protein quality control (PQC) is required to ensure cellular health. PQC is recognized for targeting the destruction of defective polypeptides, whereas regulated protein degradation mechanisms modulate the concentration of specific proteins in concert with physiological demands. For example, ion channel levels are physiologically regulated within tight limits, but a system-wide approach to define which degradative systems are involved is lacking. We focus on the Kir2.1 potassium channel because altered Kir2.1 levels lead to human disease and Kir2.1 restores growth on low-potassium medium in yeast mutated for endogenous potassium channels. Using this system, first we find that Kir2.1 is targeted for endoplasmic reticulum-associated degradation (ERAD). Next a synthetic gene array identifies nonessential genes that negatively regulate Kir2.1. The most prominent gene family that emerges from this effort encodes members of endosomal sorting complex required for transport (ESCRT). ERAD and ESCRT also mediate Kir2.1 degradation in human cells, with ESCRT playing a more prominent role. Thus multiple proteolytic pathways control Kir2.1 levels at the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum-Associated Degradation/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Potassium Channels, Inwardly Rectifying/genetics , Cell Membrane/genetics , Endosomes/genetics , Endosomes/metabolism , Gene Expression Regulation , Humans , Potassium Channels, Inwardly Rectifying/metabolism , Protein Transport , Surface Properties
17.
PLoS One ; 8(6): e65779, 2013.
Article in English | MEDLINE | ID: mdl-23776545

ABSTRACT

Many inflammatory diseases may be linked to pathologically elevated signaling via the receptor for lipopolysaccharide (LPS), toll-like receptor 4 (TLR4). There has thus been great interest in the discovery of TLR4 inhibitors as potential anti-inflammatory agents. Recently, the structure of TLR4 bound to the inhibitor E5564 was solved, raising the possibility that novel TLR4 inhibitors that target the E5564-binding domain could be designed. We utilized a similarity search algorithm in conjunction with a limited screening approach of small molecule libraries to identify compounds that bind to the E5564 site and inhibit TLR4. Our lead compound, C34, is a 2-acetamidopyranoside (MW 389) with the formula C17H27NO9, which inhibited TLR4 in enterocytes and macrophages in vitro, and reduced systemic inflammation in mouse models of endotoxemia and necrotizing enterocolitis. Molecular docking of C34 to the hydrophobic internal pocket of the TLR4 co-receptor MD-2 demonstrated a tight fit, embedding the pyran ring deep inside the pocket. Strikingly, C34 inhibited LPS signaling ex-vivo in human ileum that was resected from infants with necrotizing enterocolitis. These findings identify C34 and the ß-anomeric cyclohexyl analog C35 as novel leads for small molecule TLR4 inhibitors that have potential therapeutic benefit for TLR4-mediated inflammatory diseases.


Subject(s)
Acetylglucosamine/analogs & derivatives , Drug Discovery/methods , Inflammation/drug therapy , Small Molecule Libraries , Toll-Like Receptor 4/antagonists & inhibitors , Acetylglucosamine/chemical synthesis , Acetylglucosamine/chemistry , Acetylglucosamine/pharmacology , Analysis of Variance , Animals , Binding Sites/genetics , DNA Primers/genetics , Enterocolitis, Necrotizing/drug therapy , Enterocytes/metabolism , Humans , Lipid A/analogs & derivatives , Lipid A/metabolism , Macrophages/metabolism , Mice , Protein Binding , Real-Time Polymerase Chain Reaction , Tritium
18.
J Biol Chem ; 288(25): 18506-20, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23653356

ABSTRACT

Accumulation of misfolded proteins in cellular compartments can result in stress-induced cell death. In the endoplasmic reticulum (ER), ER-associated degradation clears aberrant proteins from the secretory pathway. In the cytoplasm and nucleus, this job is left to the cytoplasmic quality control (CytoQC) machinery. Both processes utilize chaperones and the ubiquitin-proteasome system to aid in protein elimination. Previous studies in yeast have drawn comparisons between these processes using data from structurally and topologically different substrates. We sought to draw a direct comparison between ERAD and CytoQC by studying the elimination of a single misfolded domain that, depending on its residence, is disposed by either of these pathways. The truncated, second nucleotide binding domain (NBD2*) from a yeast ERAD substrate, Ste6p*, resides at the cytoplasmic face of the ER. We show that a soluble form of NBD2* is cytoplasmic and unlike wild-type NBD2 is targeted for proteasome-mediated degradation. In contrast to Ste6p*, which employs the ER-localized Doa10p ubiquitin ligase, NBD2* is ubiquitinated by a nuclear E3 ligase San1p, a factor that is also required for its degradation. Although the yeast cytoplasmic Hsp70 chaperone, Ssa1p, has been thought to facilitate the nuclear import or to maintain the solubility of most CytoQC substrates, we discovered that Ssa1p facilitates the interaction between San1p and NBD2*, demonstrating that chaperones can aid in substrate recognition and San1p-dependent protein degradation. These results emphasize the diverse action of molecular chaperones during CytoQC.


Subject(s)
Cytoplasm/metabolism , HSP70 Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligases/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Active Transport, Cell Nucleus , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Binding Sites/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation , HSP70 Heat-Shock Proteins/genetics , Immunoblotting , Microscopy, Fluorescence , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Valosin Containing Protein
19.
ACS Chem Biol ; 8(7): 1460-8, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23594135

ABSTRACT

Misfolding and aggregation of α-synuclein (α-syn) is associated with the development of a number of neurodegenerative diseases including Parkinson's disease (PD). Analyses of post mortem tissues revealed the presence of molecular chaperones within α-syn aggregates, suggesting that chaperones play a role in α-syn misfolding and aggregation. In fact, inhibition of chaperone activity aggravates α-syn toxicity, and the overexpression of chaperones, particularly 70-kDa heat shock protein (Hsp70), protects against α-syn-induced toxicity. In this study, we investigated the effect of carbenoxolone (CBX), a glycyrrhizic acid derivative previously reported to upregulate Hsp70, in human neuroglioma cells overexpressing α-syn. We report that CBX treatment lowers α-syn aggregation and prevents α-syn-induced cytotoxicity. We demonstrate further that Hsp70 induction by CBX arises from activation of heat shock factor 1 (HSF1). The Hsp70 inhibitor MAL3-101 and the Hsp70 enhancer 115-7c led to an increase or decrease in α-syn aggregation, respectively, in agreement with these findings. In summary, this study provides a proof-of-principle demonstration that chemical modulation of the Hsp70 machine is a promising strategy to prevent α-syn aggregation.


Subject(s)
Carbenoxolone/pharmacology , Gene Expression Regulation/drug effects , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , alpha-Synuclein/metabolism , Anti-Ulcer Agents/pharmacology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Humans , Molecular Structure , Protein Binding/drug effects , Protein Folding , Tumor Cells, Cultured
20.
PLoS One ; 8(1): e53790, 2013.
Article in English | MEDLINE | ID: mdl-23342003

ABSTRACT

Localized synthesis of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] at clathrin coated pits (CCPs) is crucial for the recruitment of adaptors and other components of the internalization machinery, as well as for regulating actin dynamics during endocytosis. PtdIns(4,5)P(2) is synthesized from phosphatidylinositol 4-phosphate by any of three phosphatidylinositol 5-kinase type I (PIP5KI) isoforms (α, ß or γ). PIP5KIß localizes almost exclusively to the apical surface in polarized mouse cortical collecting duct cells, whereas the other isoforms have a less polarized membrane distribution. We therefore investigated the role of PIP5KI isoforms in endocytosis at the apical and basolateral domains. Endocytosis at the apical surface is known to occur more slowly than at the basolateral surface. Apical endocytosis was selectively stimulated by overexpression of PIP5KIß whereas the other isoforms had no effect on either apical or basolateral internalization. We found no difference in the affinity for PtdIns(4,5)P(2)-containing liposomes of the PtdIns(4,5)P(2) binding domains of epsin and Dab2, consistent with a generic effect of elevated PtdIns(4,5)P(2) on apical endocytosis. Additionally, using apical total internal reflection fluorescence imaging and electron microscopy we found that cells overexpressing PIP5KIß have fewer apical CCPs but more internalized coated structures than control cells, consistent with enhanced maturation of apical CCPs. Together, our results suggest that synthesis of PtdIns(4,5)P(2) mediated by PIP5KIß is rate limiting for apical but not basolateral endocytosis in polarized kidney cells. PtdIns(4,5)P(2) may be required to overcome specific structural constraints that limit the efficiency of apical endocytosis.


Subject(s)
Cell Polarity , Endocytosis , Epithelial Cells/cytology , Kidney/cytology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Clathrin-Coated Vesicles/metabolism , Dogs , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Madin Darby Canine Kidney Cells , Mice , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...