Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 10(1): 603, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30723194

ABSTRACT

Zinc ions (Zn2+) are imported into the early secretory pathway by Golgi-resident transporters, but their handling and functions are not fully understood. Here, we show that Zn2+ binds with high affinity to the pH-sensitive chaperone ERp44, modulating its localization and ability to retrieve clients like Ero1α and ERAP1 to the endoplasmic reticulum (ER). Silencing the Zn2+ transporters that uptake Zn2+ into the Golgi led to ERp44 dysfunction and increased secretion of Ero1α and ERAP1. High-resolution crystal structures of Zn2+-bound ERp44 reveal that Zn2+ binds to a conserved histidine-cluster. The consequent large displacements of the regulatory C-terminal tail expose the substrate-binding surface and RDEL motif, ensuring client capture and retrieval. ERp44 also forms Zn2+-bridged homodimers, which dissociate upon client binding. Histidine mutations in the Zn2+-binding sites compromise ERp44 activity and localization. Our findings reveal a role of Zn2+ as a key regulator of protein quality control at the ER-Golgi interface.


Subject(s)
Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Secretory Pathway , Zinc/metabolism , Aminopeptidases/metabolism , Binding Sites/genetics , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Crystallography, X-Ray , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Hep G2 Cells , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Minor Histocompatibility Antigens/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Oxidoreductases/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Quality Control , RNA Interference , Zinc/chemistry
2.
J Cell Sci ; 127(Pt 19): 4260-9, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25097228

ABSTRACT

ERp44 is a pH-regulated chaperone of the secretory pathway. In the acidic milieu of the Golgi, its C-terminal tail changes conformation, simultaneously exposing the substrate-binding site for cargo capture and the RDEL motif for ER retrieval through interactions with cognate receptors. Protonation of cysteine 29 in the active site allows tail movements in vitro and in vivo. Here, we show that conserved histidine residues in the C-terminal tail also regulate ERp44 in vivo. Mutants lacking these histidine residues retain substrates more efficiently. Surprisingly, they are also O-glycosylated and partially secreted. Co-expression of client proteins prevents secretion of the histidine mutants, forcing tail opening and RDEL accessibility. Client-induced RDEL exposure allows retrieval of proteins from distinct stations along the secretory pathway, as indicated by the changes in O-glycosylation patterns upon overexpression of different partners. The ensuing gradients might help to optimize folding and assembly of different cargoes. Endogenous ERp44 is O-glycosylated and secreted by human primary endometrial cells, suggesting possible pathophysiological roles of these processes.


Subject(s)
Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Humans , Molecular Chaperones/genetics , Quality Control , Secretory Pathway
3.
J Biol Chem ; 289(39): 27004-27018, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25122773

ABSTRACT

Bisphenol A (BPA) is an endocrine disruptor that may have adverse effects on human health. We recently isolated protein-disulfide isomerase (PDI) as a BPA-binding protein from rat brain homogenates and found that BPA markedly inhibited PDI activity. To elucidate mechanisms of this inhibition, detailed structural, biophysical, and functional analyses of PDI were performed in the presence of BPA. BPA binding to PDI induced significant rearrangement of the N-terminal thioredoxin domain of PDI, resulting in more compact overall structure. This conformational change led to closure of the substrate-binding pocket in b' domain, preventing PDI from binding to unfolded proteins. The b' domain also plays an essential role in the interplay between PDI and ER oxidoreduclin 1α (Ero1α), a flavoenzyme responsible for reoxidation of PDI. We show that BPA inhibited Ero1α-catalyzed PDI oxidation presumably by inhibiting the interaction between the b' domain of PDI and Ero1α; the phenol groups of BPA probably compete with a highly conserved tryptophan residue, located in the protruding ß-hairpin of Ero1α, for binding to PDI. Consistently, BPA slowed down the reoxidation of PDI and caused the reduction of PDI in HeLa cells, indicating that BPA has a great impact on the redox homeostasis of PDI within cells. However, BPA had no effect on the interaction between PDI and peroxiredoxin-4 (Prx4), another PDI family oxidase, suggesting that the interaction between Prx4 and PDI is different from that of Ero1α and PDI. These results indicate that BPA, a widely distributed and potentially harmful chemical, inhibits Ero1-PDI-mediated disulfide bond formation.


Subject(s)
Benzhydryl Compounds/pharmacology , Estrogens, Non-Steroidal/pharmacology , Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Phenols/pharmacology , Protein Disulfide-Isomerases/metabolism , Animals , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Oxidation-Reduction/drug effects , Oxidoreductases/genetics , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Protein Binding/drug effects , Protein Disulfide-Isomerases/genetics , Protein Structure, Tertiary , Rats
4.
Structure ; 22(3): 431-43, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24462249

ABSTRACT

The mammalian endoplasmic reticulum (ER) contains a diverse oxidative protein folding network in which ERp46, a member of the protein disulfide isomerase (PDI) family, serves as an efficient disulfide bond introducer together with Peroxiredoxin-4 (Prx4). We revealed a radically different molecular architecture of ERp46, in which the N-terminal two thioredoxin (Trx) domains with positively charged patches near their peptide-binding site and the C-terminal Trx are linked by unusually long loops and arranged extendedly, forming an opened V-shape. Whereas PDI catalyzes native disulfide bond formation by the cooperative action of two mutually facing redox-active sites on folding intermediates bound to the central cleft, ERp46 Trx domains are separated, act independently, and engage in rapid but promiscuous disulfide bond formation during early oxidative protein folding. Thus, multiple PDI family members likely contribute to different stages of oxidative folding and work cooperatively to ensure the efficient production of multi-disulfide proteins in the ER.


Subject(s)
Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Aprotinin/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Disulfides/chemistry , Models, Molecular , Mutation , Oxygen/metabolism , Protein Disulfide-Isomerases/genetics , Protein Folding , Protein Structure, Tertiary , Scattering, Small Angle , Thioredoxins/chemistry , X-Ray Diffraction
5.
Sci Rep ; 3: 2456, 2013.
Article in English | MEDLINE | ID: mdl-23949117

ABSTRACT

The mammalian endoplasmic reticulum (ER) harbors disulfide bond-generating enzymes, including Ero1α and peroxiredoxin 4 (Prx4), and nearly 20 members of the protein disulfide isomerase family (PDIs), which together constitute a suitable environment for oxidative protein folding. Here, we clarified the Prx4 preferential recognition of two PDI family proteins, P5 and ERp46, and the mode of interaction between Prx4 and P5 thioredoxin domain. Detailed analyses of oxidative folding catalyzed by the reconstituted Prx4-PDIs pathways demonstrated that, while P5 and ERp46 are dedicated to rapid, but promiscuous, disulfide introduction, PDI is an efficient proofreader of non-native disulfides. Remarkably, the Prx4-dependent formation of native disulfide bonds was accelerated when PDI was combined with ERp46 or P5, suggesting that PDIs work synergistically to increase the rate and fidelity of oxidative protein folding. Thus, the mammalian ER seems to contain highly systematized oxidative networks for the efficient production of large quantities of secretory proteins.


Subject(s)
Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Peroxiredoxins/chemistry , Peroxiredoxins/metabolism , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Binding Sites , HEK293 Cells , Humans , Oxidation-Reduction , Peroxiredoxins/ultrastructure , Protein Binding , Protein Disulfide-Isomerases/ultrastructure , Protein Folding
6.
Mol Cell ; 50(6): 783-92, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23685074

ABSTRACT

To warrant the quality of the secretory proteome, stringent control systems operate at the endoplasmic reticulum (ER)-Golgi interface, preventing the release of nonnative products. Incompletely assembled oligomeric proteins that are deemed correctly folded must rely on additional quality control mechanisms dedicated to proper assembly. Here we unveil how ERp44 cycles between cisGolgi and ER in a pH-regulated manner, patrolling assembly of disulfide-linked oligomers such as IgM and adiponectin. At neutral, ER-equivalent pH, the ERp44 carboxy-terminal tail occludes the substrate-binding site. At the lower pH of the cisGolgi, conformational rearrangements of this peptide, likely involving protonation of ERp44's active cysteine, simultaneously unmask the substrate binding site and -RDEL motif, allowing capture of orphan secretory protein subunits and ER retrieval via KDEL receptors. The ERp44 assembly control cycle couples secretion fidelity and efficiency downstream of the calnexin/calreticulin and BiP-dependent quality control cycles.


Subject(s)
Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Protein Multimerization , Amino Acid Motifs , Amino Acid Substitution , Catalytic Domain , Cell Cycle , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Membrane Glycoproteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Mutagenesis, Site-Directed , Oxidoreductases/metabolism , Protein Transport , Secretory Pathway
7.
J Biol Chem ; 286(18): 16261-71, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21398518

ABSTRACT

In the endoplasmic reticulum (ER) of human cells, ERO1α and protein-disulfide isomerase (PDI) constitute one of the major electron flow pathways that catalyze oxidative folding of secretory proteins. Specific and limited PDI oxidation by ERO1α is essential to avoid ER hyperoxidation. To investigate how ERO1α oxidizes PDI selectively among more than 20 ER-resident PDI family member proteins, we performed docking simulations and systematic biochemical analyses. Our findings reveal that a protruding ß-hairpin of ERO1α specifically interacts with the hydrophobic pocket present in the redox-inactive PDI b'-domain through the stacks between their aromatic residues, leading to preferred oxidation of the C-terminal PDI a'-domain. ERO1α associated preferentially with reduced PDI, explaining the stepwise disulfide shuttle mechanism, first from ERO1α to PDI and then from oxidized PDI to an unfolded polypeptide bound to its hydrophobic pocket. The interaction of ERO1α with ERp44, another PDI family member protein, was also analyzed. Notably, ERO1α-dependent PDI oxidation was inhibited by a hyperactive ERp44 mutant that lacks the C-terminal tail concealing the substrate-binding hydrophobic regions. The potential ability of ERp44 to inhibit ERO1α activity may suggest its physiological role in ER redox and protein homeostasis.


Subject(s)
Disulfides/chemistry , Membrane Glycoproteins/chemistry , Oxidoreductases/chemistry , Protein Disulfide-Isomerases/chemistry , Protein Folding , Binding Sites , Catalysis , Disulfides/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Structure, Secondary
8.
EMBO J ; 29(19): 3330-43, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20834232

ABSTRACT

In the endoplasmic reticulum (ER) of eukaryotic cells, Ero1 flavoenzymes promote oxidative protein folding through protein disulphide isomerase (PDI), generating reactive oxygen species (hydrogen peroxide) as byproducts. Therefore, Ero1 activity must be strictly regulated to avoid futile oxidation cycles in the ER. Although regulatory mechanisms restraining Ero1α activity ensure that not all PDIs are oxidized, its specificity towards PDI could allow other resident oxidoreductases to remain reduced and competent to carry out isomerization and reduction of protein substrates. In this study, crystal structures of human Ero1α were solved in its hyperactive and inactive forms. Our findings reveal that human Ero1α modulates its oxidative activity by properly positioning regulatory cysteines within an intrinsically flexible loop, and by fine-tuning the electron shuttle ability of the loop through disulphide rearrangements. Specific PDI targeting is guaranteed by electrostatic and hydrophobic interactions of Ero1α with the PDI b'-domain through its substrate-binding pocket. These results reveal the molecular basis of the regulation and specificity of protein disulphide formation in human cells.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Models, Molecular , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Disulfide-Isomerases/metabolism , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Oxidation-Reduction , Protein Binding , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...