Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
J Microbiol Biotechnol ; 34(5): 1126-1134, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38563095

ABSTRACT

The production of disulfide bond-containing recombinant proteins in Escherichia coli has traditionally been done by either refolding from inclusion bodies or by targeting the protein to the periplasm. However, both approaches have limitations. Two broad strategies were developed to allow the production of proteins with disulfide bonds in the cytoplasm of E. coli: i) engineered strains with deletions in the disulfide reduction pathways, e.g. SHuffle, and ii) the co-expression of oxidative folding catalysts, e.g. CyDisCo. However, to our knowledge, the relative effectiveness of these strategies has not been properly evaluated. Here, we systematically compare the purified yields of 14 different proteins of interest (POI) that contain disulfide bonds in their native state when expressed in both systems. We also compared the effects of different background strains, commonly used promoters, and two media types: defined and rich autoinduction. In rich autoinduction media, POI which can be produced in a soluble (non-native) state without a system for disulfide bond formation were produced in higher purified yields from SHuffle, whereas all other proteins were produced in higher purified yields using CyDisCo. In chemically defined media, purified yields were at least 10x higher in all cases using CyDisCo. In addition, the quality of the three POI tested was superior when produced using CyDisCo.


Subject(s)
Cytoplasm , Disulfides , Escherichia coli Proteins , Escherichia coli , Protein Folding , Recombinant Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Disulfides/metabolism , Disulfides/chemistry , Cytoplasm/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Oxidation-Reduction , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Periplasm/metabolism , Periplasm/genetics , Culture Media/chemistry
2.
J Biol Chem ; 300(3): 105746, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38354787

ABSTRACT

In the methylotrophic yeast Komagataella phaffii, we identified an endoplasmic reticulum-resident protein disulfide isomerase (PDI) family member, Erp41, with a peculiar combination of active site motifs. Like fungal ERp38, it has two thioredoxin-like domains which contain active site motifs (a and a'), followed by an alpha-helical ERp29c C-terminal domain (c domain). However, while the a domain has a typical PDI-like active site motif (CGHC), the a' domain instead has CGYC, a glutaredoxin-like motif which confers to the protein an exceptional affinity for GSH/GSSG. This combination of active site motifs has so far been unreported in PDI-family members. Homology searches revealed ERp41 is present in the genome of some plants, fungal parasites, and a few nonconventional yeasts, among which are Komagataella spp. and Yarrowia lipolytica. These yeasts are both used for the production of secreted recombinant proteins. Here, we analyzed the activity of K. phaffii Erp41. We report that it is nonessential in K. phaffii, and that it can catalyze disulfide bond formation in partnership with the sulfhydryl oxidase Ero1 in vitro with higher turnover rates than the canonical PDI from K. phaffii, Pdi1, but slower activation times. We show how Erp41 has unusually fast glutathione-coupled oxidation activity and relate it to its unusual combination of active sites in its thioredoxin-like domains. We further describe how this determines its unusually efficient catalysis of dithiol oxidation in peptide and protein substrates.


Subject(s)
Protein Disulfide-Isomerases , Protein Folding , Saccharomycetales , Disulfides/chemistry , Glutathione/metabolism , Oxidation-Reduction , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Protein Structure, Tertiary , Saccharomycetales/enzymology , Thioredoxins/metabolism
3.
Protein Expr Purif ; 215: 106404, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37979630

ABSTRACT

Fragment of antigen-binding region (Fab) of antibodies are important biomolecules, with a broad spectrum of functionality in the biomedical field. While full length antibodies are usually produced in mammalian cells, the smaller size, lack of N-glycosylation and less complex structure of Fabs make production in microbial cell factories feasible. Since Fabs contain disulfide bonds, such production is often done in the periplasm, but there the formation of the inter-molecular disulfide bond between light and heavy chains can be problematic. Here we studied the use of the CyDisCo system (cytoplasmic disulfide bond formation in E. coli) to express two Fabs (Herceptin and Maa48) in the cytoplasm of E. coli in fed-batch fermentation using a generic chemically defined media. We were able to solubly express both Fabs with purified yields of 565 mg/L (Maa48) and 660 mg/L (Herceptin) from low density fermentation. Both proteins exhibited CD spectra consistent with natively folded protein and both were biologically active. To our knowledge this is the first demonstration of high-level production of biological active Fabs in the cytoplasm of E. coli in industrially relevant fermentation conditions.


Subject(s)
Escherichia coli , Immunoglobulin Fab Fragments , Animals , Cytoplasm/metabolism , Disulfides/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Fermentation , Trastuzumab , Immunoglobulin Fab Fragments/biosynthesis
4.
Sci Rep ; 13(1): 14188, 2023 08 30.
Article in English | MEDLINE | ID: mdl-37648872

ABSTRACT

With increased accessibility and tissue penetration, smaller antibody formats such as antibody fragments (Fab) and single chain variable fragments (scFv) show potential as effective and low-cost choices to full-length antibodies. These formats derived from the modular architecture of antibodies could prove to be game changers for certain therapeutic and diagnostic applications. Microbial hosts have shown tremendous promise as production hosts for antibody fragment formats. However, low target protein yields coupled with the complexity of protein folding result in production limitations. Here, we report an alternative antibody fragment format 'FabH3' designed to overcome some key bottlenecks associated with the folding and production of Fabs. The FabH3 molecule is based on the Fab format with the constant domains replaced by engineered immunoglobulin G1 (IgG1) CH3 domains capable of heterodimerization based on the electrostatic steering approach. We show that this alternative antibody fragment format can be efficiently produced in the cytoplasm of E. coli using the catalyzed disulfide-bond formation system (CyDisCo) in a natively folded state with higher soluble yields than its Fab counterpart and a comparable binding affinity against the target antigen.


Subject(s)
Escherichia coli , Single-Chain Antibodies , Escherichia coli/genetics , Cytoplasm , Diet, Protein-Restricted , Immunoglobulin G , Single-Chain Antibodies/genetics
5.
FEBS Lett ; 597(13): 1792-1801, 2023 07.
Article in English | MEDLINE | ID: mdl-37247262

ABSTRACT

Human aldehyde oxidase (hAOX1) is a molybdoenzyme that oxidizes aldehydes and N-heterocyclic compounds, thereby generating hydrogen peroxide (H2 O2 ) and superoxide during turnover. hAOX1 has been shown previously to be inactivated under turnover conditions by H2 O2 . Here, we investigated the effect of exogenously added H2 O2 on the activity of hAOX1. We show that exogenously added H2 O2 did not affect the enzyme activity under aerobic conditions, but completely inactivated the enzyme under anaerobic conditions. We propose that this effect is based on the reducing power of H2 O2 and the susceptibility of the reduced molybdenum cofactor (Moco) to lose the sulfido ligand. When oxygen is present, the enzyme is rapidly reoxidized. We believe that our study is significant in understanding the detailed effect of reactive oxygen species on the inactivation of hAOX1 and other molybdoenzymes.


Subject(s)
Aldehyde Oxidase , Coenzymes , Humans , Reactive Oxygen Species , Superoxides , Oxygen , Hydrogen Peroxide/pharmacology
6.
Microbiologyopen ; 12(2): e1350, 2023 04.
Article in English | MEDLINE | ID: mdl-37186227

ABSTRACT

High-value heterologous proteins produced in Escherichia coli that contain disulfide bonds are almost invariably targeted to the periplasm via the Sec pathway as it, among other advantages, enables disulfide bond formation and simplifies downstream processing. However, the Sec system cannot transport complex or rapidly folding proteins, as it only transports proteins in an unfolded state. The Tat system also transports proteins to the periplasm, and it has significant potential as an alternative means of recombinant protein production because it transports fully folded proteins. Most of the studies related to Tat secretion have used the well-studied TorA signal peptide that is Tat-specific, but this signal peptide also tends to induce degradation of the protein of interest, resulting in lower yields. This makes it difficult to use Tat in the industry. In this study, we show that a model disulfide bond-containing protein, YebF, can be exported to the periplasm and media at a very high level by the Tat pathway in a manner almost completely dependent on cytoplasmic disulfide formation, by other two putative Tat SPs: those of MdoD and AmiC. In contrast, the TorA SP exports YebF at a low level.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Periplasm , Recombinant Proteins , Twin-Arginine-Translocation System , Protein Transport , Periplasm/metabolism , Disulfides/chemistry , Escherichia coli/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Protein Folding , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Protein Sorting Signals , Twin-Arginine-Translocation System/metabolism , Culture Media , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
7.
Drug Metab Dispos ; 51(6): 764-770, 2023 06.
Article in English | MEDLINE | ID: mdl-37012073

ABSTRACT

Human aldehyde oxidase (hAOX1) is a molybdoflavoenzyme that belongs to the xanthine oxidase (XO) family. hAOX1 is involved in phase I drug metabolism, but its physiologic role is not fully understood to date, and preclinical studies consistently underestimated hAOX1 clearance. In the present work, we report an unexpected effect of the common sulfhydryl-containing reducing agents, e.g., dithiothreitol (DTT), on the activity of hAOX1 and mouse aldehyde oxidases. We demonstrate that this effect is due to the reactivity of the sulfido ligand bound at the molybdenum cofactor with the sulfhydryl groups. The sulfido ligand coordinated to the Mo atom in the XO family of enzymes plays a crucial role in the catalytic cycle and its removal results in the total inactivation of these enzymes. Because liver cytosols, S9 fractions, and hepatocytes are commonly used to screen the drug candidates for hAOX1, our study suggests that DTT treatment of these samples should be avoided, otherwise false negative results by an inactivated hAOX1 might be obtained. SIGNIFICANCE STATEMENT: This work characterizes the inactivation of human aldehyde oxidase (hAOX1) by sulfhydryl-containing agents and identifies the site of inactivation. The role of dithiothreitol in the inhibition of hAOX1 should be considered for the preparation of hAOX1-containing fractions for pharmacological studies on drug metabolism and drug clearance.


Subject(s)
Aldehyde Oxidase , Reducing Agents , Humans , Animals , Mice , Aldehyde Oxidase/metabolism , Ligands , Dithiothreitol/pharmacology , Coenzymes , Xanthine Oxidase
8.
Sci Rep ; 13(1): 4408, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36927743

ABSTRACT

Global health challenges such as the coronavirus pandemic warrant the urgent need for a system that allows efficient production of diagnostic and therapeutic interventions. Antibody treatments against SARS-CoV-2 were developed with an unprecedented pace and this enormous progress was achieved mainly through recombinant protein production technologies combined with expeditious screening approaches. A heterologous protein production system that allows efficient soluble production of therapeutic antibody candidates against rapidly evolving variants of deadly pathogens is an important step in preparedness towards future pandemic challenges. Here, we report cost and time-effective soluble production of SARS-CoV-2 receptor binding domain (RBD) variants as well as an array of neutralizing antibody fragments (Fabs) based on Casirivimab and Imdevimab using the CyDisCo system in the cytoplasm of E. coli. We also report variants of the two Fabs with higher binding affinity against SARS-CoV-2 RBD and suggest this cytoplasmic production of disulfide containing antigens and antibodies can be broadly applied towards addressing future global public health threats.


Subject(s)
Antibodies, Neutralizing , COVID-19 , Humans , SARS-CoV-2 , Escherichia coli/metabolism , Antibodies, Viral , Cytoplasm/metabolism
9.
Int J Mol Sci ; 23(23)2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36499069

ABSTRACT

Cost-effective production of therapeutic proteins in microbial hosts is an indispensable tool towards accessible healthcare. Many of these heterologously expressed proteins, including all antibody formats, require disulfide bond formation to attain their native and functional state. A system for catalyzed disulfide bond formation (CyDisCo) has been developed allowing efficient production of recombinant proteins in the cytoplasm of one of the most used microbial expression systems, Escherichia coli. Here, we report high-yield production (up to 230 mg/L from 3 mL cultures) of in-demand therapeutics such as IgG1-based Fc fusion proteins in the E. coli cytoplasm. However, the production of this drug class using the CyDisCo system faces bottlenecks related to redox heterogeneity during oxidative folding. Our investigations identified and addressed one of the major causes of redox heterogeneity during CyDisCo-based production of Fc fusion proteins, i.e., disulfide bond formation in the IgG1 CH3 domain. Here, we communicate that mutating the cysteines in the CH3 domain of target Fc fusion proteins allows their production in a homogeneous redox state in the cytoplasm of E. coli without compromising on yields and thermal stability.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Escherichia coli/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Disulfides/chemistry , Recombinant Proteins/metabolism , Cytoplasm/metabolism , Immunoglobulin G/metabolism , Recombinant Fusion Proteins/chemistry
10.
Antioxidants (Basel) ; 11(10)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36290643

ABSTRACT

Proteins in the thioredoxin superfamily share a similar fold, contain a -CXXC- active site, and catalyze oxidoreductase reactions by dithiol-disulfide exchange mechanisms. Protein disulfide isomerase (PDI) has two -CGHC- active sites. For in vitro studies, oxidation/reduction of PDI during the catalytic cycle is accomplished with glutathione. Glutathione may act as electron donor/acceptor for PDI also in vivo, but at least for oxidation reactions, GSSG probably is not the major electron acceptor and PDI may not have evolved to react with glutathione with high affinity, but merely having adequate affinity for both glutathione and folding proteins/peptides. Glutaredoxins, on the other hand, have a high affinity for glutathione. They commonly have -CXFC- or -CXYC- active site, where the tyrosine residue forms part of the GSH binding groove. Mutating the active site of PDI to a more glutaredoxin-like motif increased its reactivity with glutathione. All such variants showed an increased rate in GSH-dependent reduction or GSSG-dependent oxidation of the active site, as well as a decreased rate of the native disulfide bond formation, with the magnitude of the effect increasing with glutathione concentration. This suggests that these variants lead to competition in binding between glutathione and folding protein substrates.

11.
J Biol Chem ; 298(12): 102614, 2022 12.
Article in English | MEDLINE | ID: mdl-36265586

ABSTRACT

Collagen prolyl 4-hydroxylases (C-P4H) are α2ß2 tetramers, which catalyze the prolyl 4-hydroxylation of procollagen, allowing for the formation of the stable triple-helical collagen structure in the endoplasmic reticulum. The C-P4H α-subunit provides the N-terminal dimerization domain, the middle peptide-substrate-binding (PSB) domain, and the C-terminal catalytic (CAT) domain, whereas the ß-subunit is identical to the enzyme protein disulfide isomerase (PDI). The structure of the N-terminal part of the α-subunit (N-terminal region and PSB domain) is known, but the structures of the PSB-CAT linker region and the CAT domain as well as its mode of assembly with the ß/PDI subunit, are unknown. Here, we report the crystal structure of the CAT domain of human C-P4H-II complexed with the intact ß/PDI subunit, at 3.8 Å resolution. The CAT domain interacts with the a, b', and a' domains of the ß/PDI subunit, such that the CAT active site is facing bulk solvent. The structure also shows that the C-P4H-II CAT domain has a unique N-terminal extension, consisting of α-helices and a ß-strand, which is the edge strand of its major antiparallel ß-sheet. This extra region of the CAT domain interacts tightly with the ß/PDI subunit, showing that the CAT-PDI interface includes an intersubunit disulfide bridge with the a' domain and tight hydrophobic interactions with the b' domain. Using this new information, the structure of the mature C-P4H-II α2ß2 tetramer is predicted. The model suggests that the CAT active-site properties are modulated by α-helices of the N-terminal dimerization domains of both subunits of the α2-dimer.


Subject(s)
Prolyl Hydroxylases , Protein Disulfide-Isomerases , Humans , Catalytic Domain , Collagen/metabolism , Peptides/metabolism , Procollagen-Proline Dioxygenase/metabolism , Prolyl Hydroxylases/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Conformation
12.
EMBO J ; 41(2): e105531, 2022 12 17.
Article in English | MEDLINE | ID: mdl-34904718

ABSTRACT

Recessive gene mutations underlie many developmental disorders and often lead to disabling neurological problems. Here, we report identification of a homozygous c.170G>A (p.Cys57Tyr or C57Y) mutation in the gene coding for protein disulfide isomerase A3 (PDIA3, also known as ERp57), an enzyme that catalyzes formation of disulfide bonds in the endoplasmic reticulum, to be associated with syndromic intellectual disability. Experiments in zebrafish embryos show that PDIA3C57Y expression is pathogenic and causes developmental defects such as axonal disorganization as well as skeletal abnormalities. Expression of PDIA3C57Y in the mouse hippocampus results in impaired synaptic plasticity and memory consolidation. Proteomic and functional analyses reveal that PDIA3C57Y expression leads to dysregulation of cell adhesion and actin cytoskeleton dynamics, associated with altered integrin biogenesis and reduced neuritogenesis. Biochemical studies show that PDIA3C57Y has decreased catalytic activity and forms disulfide-crosslinked aggregates that abnormally interact with chaperones in the endoplasmic reticulum. Thus, rare disease gene variant can provide insight into how perturbations of neuronal proteostasis can affect the function of the nervous system.


Subject(s)
Developmental Disabilities/genetics , Endoplasmic Reticulum/metabolism , Protein Disulfide-Isomerases/genetics , Proteostasis , Adolescent , Adult , Animals , Axons/metabolism , Axons/pathology , Cell Adhesion , Cells, Cultured , Child , Cytoskeleton/metabolism , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Integrins/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Neuronal Outgrowth , Neuronal Plasticity , Pedigree , Protein Disulfide-Isomerases/metabolism , Zebrafish
13.
Essays Biochem ; 65(2): 247-260, 2021 07 26.
Article in English | MEDLINE | ID: mdl-33955451

ABSTRACT

Recombinant proteins have been extensively employed as therapeutics for the treatment of various critical and life-threatening diseases and as industrial enzymes in high-value industrial processes. Advances in genetic engineering and synthetic biology have broadened the horizon of heterologous protein production using multiple expression platforms. Selection of a suitable expression system depends on a variety of factors ranging from the physicochemical properties of the target protein to economic considerations. For more than 40 years, Escherichia coli has been an established organism of choice for protein production. This review aims to provide a stepwise approach for any researcher embarking on the journey of recombinant protein production in E. coli. We present an overview of the challenges associated with heterologous protein expression, fundamental considerations connected to the protein of interest (POI) and designing expression constructs, as well as insights into recently developed technologies that have contributed to this ever-growing field.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Genetic Engineering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synthetic Biology
14.
J Mol Biol ; 433(5): 166826, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33453188

ABSTRACT

The folding of disulfide bond containing proteins in the endoplasmic reticulum (ER) is a complex process that requires protein folding factors, some of which are protein-specific. The ER resident saposin-like protein pERp1 (MZB1, CNPY5) is crucial for the correct folding of IgA, IgM and integrins. pERp1 also plays a role in ER calcium homeostasis and plasma cell mobility. As an important factor for proper IgM maturation and hence immune function, pERp1 is upregulated in many auto-immune diseases. This makes it a potential therapeutic target. pERp1 belongs to the CNPY family of ER resident saposin-like proteins. To date, five of these proteins have been identified. All are implicated in protein folding and all contain a saposin-like domain. All previously structurally characterized saposins are involved in lipid binding. However, there are no reports of CNPY family members interacting with lipids, suggesting a novel function for the saposin fold. However, the molecular mechanisms of their function remain elusive. To date, no structure of any CNPY protein has been reported. Here, we present the high-resolution (1.4 Å) crystal structure of human pERp1 and confirm that it has a saposin-fold with unique structural elements not present in other saposin-fold structures. The implications for the role of CNPY proteins in protein folding in the ER are discussed.


Subject(s)
Immunoglobulin A/chemistry , Immunoglobulin M/chemistry , Molecular Chaperones/chemistry , Saposins/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Sequence , Binding Sites , Calcium/metabolism , Cloning, Molecular , Crystallography, X-Ray , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Immunity, Humoral , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Models, Molecular , Molecular Chaperones/genetics , Molecular Chaperones/immunology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Saposins/genetics , Saposins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
15.
Protein Expr Purif ; 180: 105809, 2021 04.
Article in English | MEDLINE | ID: mdl-33338588

ABSTRACT

The major cat allergen Fel d 1 is one of the most common and potent causes of animal related allergy. Medical treatment of cat allergy has relied on immunotherapy carried out with cat dander extract. This approach has been problematic, mainly due to inconsistent levels of the major allergen in the produced extracts. Recombinant DNA technology has been proposed as an alternative method to produce more consistent pharmaceuticals for immunotherapy and diagnostics of allergy. Current approaches to produce recombinant Fel d 1 (recFel d 1) in the cytoplasm of Escherichia coli have however resulted in protein folding deficiencies and insoluble inclusion body formation, requiring elaborate in vitro processing to acquire folded material. In this study, we introduce an efficient method for cytoplasmic production of recFel d 1 that utilizes eukaryotic folding factors to aid recFel d 1 to fold and be produced in the soluble fraction of E. coli. The solubly expressed recFel d 1 is shown by biophysical in vitro experiments to contain structural disulfides, is extremely stable, and has a sensitivity for methionine sulfoxidation. The latter is discussed in the context of functional relevance.


Subject(s)
Allergens , Glycoproteins , Protein Folding , Allergens/biosynthesis , Allergens/chemistry , Allergens/genetics , Allergens/isolation & purification , Animals , Cats , Glycoproteins/biosynthesis , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
16.
Free Radic Biol Med ; 160: 103-110, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32768572

ABSTRACT

Abnormal protein accumulations are typical pathological features for neurodegenerative diseases. Protein disulfide isomerase (PDI) is a critical enzyme in oxidative protein folding. S-nitrosylated PDI has been detected in the postmortem brain in neurodegenerative disease patients, but the effect of S-nitrosylation on PDI function and developing neurodegeneration was not clarified in detail. In this study, we identified that in vitro and in vivo S-nitrosylation of C343 in the b' domain of PDI occurs. Reduced recombinant human PDI (hPDI) reacted quickly with S-nitrosocompounds, with an observed increase in the expected S-nitrosylated species and the appearance of the disulfide state of the active sites. Both Mononitrosylated and dinitrosylated were observed from the S-nitrosylation of hPDI. Dinitrosylated species were S-nitrosylated both cysteines at active site. But, at least in part, mononitrosylated species were S-nitrosylated on cysteine 343 in the substrate binding b' domain. Although active site S-nitrosylation is reversible by reduced glutathione, S-nitrosylation of C343 is comparative stable. S-nitrosylation of PDI in SH-SY5Y cells was observed after the S-nitrosocysteine (SNOC) treatment and S-nitrosylated PDI was still detected 24 h after removing SNOC. While wild-type PDI was S-nitrosylated, the level of S-nitrosylation of the C343S mutant in over-expressed cells was substantially lower and only wild-type PDI of S-nitrosylation remained 24 h after removing SNOC in over-expressed cells. Both of in vitro and in vivo results suggested that S-nitrosylation of C343 in PDI may be the causative effect on physiological changes in neurodegerenative disease patients, and may be useful for the drug development for neurodegenerative diseases.


Subject(s)
Cysteine , Neurodegenerative Diseases , Protein Disulfide-Isomerases , Brain/metabolism , Humans , Protein Binding , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Folding
17.
J Bone Miner Res ; 35(12): 2381-2392, 2020 12.
Article in English | MEDLINE | ID: mdl-32722848

ABSTRACT

We studied a family with severe primary osteoporosis carrying a heterozygous p.Arg8Phefs*14 deletion in COL1A2, leading to haploinsufficiency. Three affected individuals carried the mutation and presented nearly identical spinal fractures but lacked other typical features of either osteogenesis imperfecta or Ehlers-Danlos syndrome. Although mutations leading to haploinsufficiency in COL1A2 are rare, mutations in COL1A1 that lead to less protein typically result in a milder phenotype. We hypothesized that other genetic factors may contribute to the severe phenotype in this family. We performed whole-exome sequencing in five family members and identified in all three affected individuals a rare nonsense variant (c.1282C > T/p.Arg428*, rs150257846) in ZNF528. We studied the effect of the variant using qPCR and Western blot and its subcellular localization with immunofluorescence. Our results indicate production of a truncated ZNF528 protein that locates in the cell nucleus as per the wild-type protein. ChIP and RNA sequencing analyses on ZNF528 and ZNF528-c.1282C > T indicated that ZNF528 binding sites are linked to pathways and genes regulating bone morphology. Compared with the wild type, ZNF528-c.1282C > T showed a global shift in genomic binding profile and pathway enrichment, possibly contributing to the pathophysiology of primary osteoporosis. We identified five putative target genes for ZNF528 and showed that the expression of these genes is altered in patient cells. In conclusion, the variant leads to expression of truncated ZNF528 and a global change of its genomic occupancy, which in turn may lead to altered expression of target genes. ZNF528 is a novel candidate gene for bone disorders and may function as a transcriptional regulator in pathways affecting bone morphology and contribute to the phenotype of primary osteoporosis in this family together with the COL1A2 deletion. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Osteogenesis Imperfecta , Osteoporosis , Transcription Factors/genetics , Collagen Type I/genetics , Exome/genetics , Humans , Mutation , Osteogenesis Imperfecta/genetics , Osteoporosis/genetics , Phenotype , Sequence Deletion , Exome Sequencing
18.
J Biol Chem ; 295(26): 8647-8655, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32102847

ABSTRACT

Protein maturation in the endoplasmic reticulum (ER) depends on a fine balance between oxidative protein folding and quality control mechanisms, which together ensure high-capacity export of properly folded proteins from the ER. Oxidative protein folding needs to be regulated to avoid hyperoxidation. The folding capacity of the ER is regulated by the unfolded protein response (UPR) and ER-associated degradation (ERAD). The UPR is triggered by unfolded protein stress and leads to up-regulation of cellular components such as chaperones and folding catalysts. These components relieve stress by increasing folding capacity and up-regulating ERAD components that remove non-native proteins. Although oxidative protein folding and the UPR/ERAD pathways each are well-understood, very little is known about any direct cross-talk between them. In this study, we carried out comprehensive in vitro activity and binding assays, indicating that the oxidative protein folding relay formed by ER oxidoreductin 1 (Ero1), and protein disulfide-isomerase can be inactivated by a feedback inhibition mechanism involving unfolded proteins and folding intermediates when their levels exceed the folding capacity of the system. This mechanism allows client proteins to remain mainly in the reduced state and thereby minimizes potential futile oxidation-reduction cycles and may also enhance ERAD, which requires reduced protein substrates. Relief from excess levels of non-native proteins by increasing the levels of folding factors removed the feedback inhibition. These results reveal regulatory cross-talk between the oxidative protein folding and UPR and ERAD pathways.


Subject(s)
Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Folding , Endoplasmic Reticulum-Associated Degradation , Humans , Membrane Glycoproteins/chemistry , Oxidation-Reduction , Oxidoreductases/chemistry , Oxygen Consumption , Protein Disulfide-Isomerases/chemistry , Unfolded Protein Response
19.
Int J Mol Sci ; 21(3)2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31973001

ABSTRACT

Escherichia coli is the most widely used protein production host in academia and a major host for industrial protein production. However, recombinant production of eukaryotic proteins in prokaryotes has challenges. One of these is post-translational modifications, including native disulfide bond formation. Proteins containing disulfide bonds have traditionally been made by targeting to the periplasm or by in vitro refolding of proteins made as inclusion bodies. More recently, systems for the production of disulfide-containing proteins in the cytoplasm have been introduced. However, it is unclear if these systems have the capacity for the production of disulfide-rich eukaryotic proteins. To address this question, we tested the capacity of one such system to produce domain constructs, containing up to 44 disulfide bonds, of the mammalian extracellular matrix proteins mucin 2, alpha tectorin, and perlecan. All were successfully produced with purified yields up to 6.5 mg/L. The proteins were further analyzed using a variety of biophysical techniques including circular dichroism spectrometry, thermal stability assay, and mass spectrometry. These analyses indicated that the purified proteins are most likely correctly folded to their native state. This greatly extends the use of E. coli for the production of eukaryotic proteins for structural and functional studies.


Subject(s)
Cytoplasm/metabolism , Escherichia coli Proteins/biosynthesis , Escherichia coli/metabolism , Extracellular Matrix Proteins/biosynthesis , Animals , Disulfides/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Heparan Sulfate Proteoglycans , Inclusion Bodies/metabolism , Mucin-2/metabolism , Periplasm/metabolism , Protein Processing, Post-Translational , Protein Stability
20.
Protein Expr Purif ; 165: 105498, 2020 01.
Article in English | MEDLINE | ID: mdl-31521797

ABSTRACT

Candida antarctica lipase B (CalB) is a very efficient catalyst and is used in a wide range of industries from food flavour to pharmaceutical, and biodiesel manufacturing. It has a high degree of enantioselective and regioselective substrate specificity and is stable over a wide range of biophysical conditions including pH, temperature and solvent conditions. High-level expression of biologically active wild-type CalB has been problematic, partly due to folding events. Consequently, focus has been on modified CalB, which has allowed orders of magnitude increase in yields of protein. However, these modifications alter the quaternary structure of the protein. Here we produce soluble wild-type CalB in high yields in the cytoplasm of E.coli using a catalyzed system for cytoplasmic disulfide bond formation both in shake flasks and in fermentation in chemically defined media. The CalB produced had the expected stereospecific activity and had a higher activity than CalB from a commercial source.


Subject(s)
Cytoplasm/metabolism , Escherichia coli/metabolism , Fungal Proteins/genetics , Fungal Proteins/isolation & purification , Lipase/genetics , Lipase/isolation & purification , Base Sequence , Catalysis , Cloning, Molecular , Disulfides/metabolism , Escherichia coli/ultrastructure , Fermentation , Fungal Proteins/metabolism , Gene Expression Regulation, Bacterial , Lipase/metabolism , Oxidation-Reduction , Protein Conformation , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...