Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(2): e105531, 2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34904718

RESUMEN

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.


Asunto(s)
Discapacidades del Desarrollo/genética , Retículo Endoplásmico/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteostasis , Adolescente , Adulto , Animales , Axones/metabolismo , Axones/patología , Adhesión Celular , Células Cultivadas , Niño , Citoesqueleto/metabolismo , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Proyección Neuronal , Plasticidad Neuronal , Linaje , Proteína Disulfuro Isomerasas/metabolismo , Pez Cebra
2.
J Biol Chem ; 300(3): 105746, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38354787

RESUMEN

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.


Asunto(s)
Proteína Disulfuro Isomerasas , Pliegue de Proteína , Saccharomycetales , Disulfuros/química , Glutatión/metabolismo , Oxidación-Reducción , Proteína Disulfuro Isomerasas/química , Proteína Disulfuro Isomerasas/metabolismo , Estructura Terciaria de Proteína , Saccharomycetales/enzimología , Tiorredoxinas/metabolismo
3.
J Biol Chem ; 300(3): 105726, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38325741

RESUMEN

Hyperlipidemia predisposes individuals to cardiometabolic diseases, the most common cause of global mortality. Microsomal triglyceride transfer protein (MTP) transfers multiple lipids and is essential for the assembly of apolipoprotein B-containing lipoproteins. MTP inhibition lowers plasma lipids but causes lipid retention in the liver and intestine. Previous studies suggested two lipid transfer domains in MTP and that specific inhibition of triglyceride (TG) and not phospholipid (PL) transfer can lower plasma lipids without significant tissue lipid accumulation. However, how MTP transfers different lipids and the domains involved in these activities are unknown. Here, we tested a hypothesis that two different ß-sandwich domains in MTP transfer TG and PL. Mutagenesis of charged amino acids in ß2-sandwich had no effect on PL transfer activity indicating that they are not critical. In contrast, amino acids with bulky hydrophobic side chains in ß1-sandwich were critical for both TG and PL transfer activities. Substitutions of these residues with smaller hydrophobic side chains or positive charges reduced, whereas negatively charged side chains severely attenuated MTP lipid transfer activities. These studies point to a common lipid transfer domain for TG and PL in MTP that is enriched with bulky hydrophobic amino acids. Furthermore, we observed a strong correlation in different MTP mutants with respect to loss of both the lipid transfer activities, again implicating a common binding site for TG and PL in MTP. We propose that targeting of areas other than the identified common lipid transfer domain might reduce plasma lipids without causing cellular lipid retention.


Asunto(s)
Proteínas Portadoras , Interacciones Hidrofóbicas e Hidrofílicas , Fosfolípidos , Triglicéridos , Humanos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Apolipoproteínas B/química , Apolipoproteínas B/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Fosfolípidos/sangre , Fosfolípidos/metabolismo , Triglicéridos/sangre , Triglicéridos/metabolismo , Dominios Proteicos , Mutación , Relación Estructura-Actividad , Sitios de Unión
4.
Protein Expr Purif ; 215: 106404, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37979630

RESUMEN

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.


Asunto(s)
Escherichia coli , Fragmentos Fab de Inmunoglobulinas , Animales , Citoplasma/metabolismo , Disulfuros/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fermentación , Trastuzumab , Fragmentos Fab de Inmunoglobulinas/biosíntesis
5.
J Biol Chem ; 298(12): 102614, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36265586

RESUMEN

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.


Asunto(s)
Prolil Hidroxilasas , Proteína Disulfuro Isomerasas , Humanos , Dominio Catalítico , Colágeno/metabolismo , Péptidos/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Prolil Hidroxilasas/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Conformación Proteica
6.
Drug Metab Dispos ; 51(6): 764-770, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37012073

RESUMEN

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.


Asunto(s)
Aldehído Oxidasa , Sustancias Reductoras , Humanos , Animales , Ratones , Aldehído Oxidasa/metabolismo , Ligandos , Ditiotreitol/farmacología , Coenzimas , Xantina Oxidasa
7.
PLoS Genet ; 16(8): e1008941, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32760060

RESUMEN

Apolipoprotein B-containing lipoproteins (B-lps) are essential for the transport of hydrophobic dietary and endogenous lipids through the circulation in vertebrates. Zebrafish embryos produce large numbers of B-lps in the yolk syncytial layer (YSL) to move lipids from yolk to growing tissues. Disruptions in B-lp production perturb yolk morphology, readily allowing for visual identification of mutants with altered B-lp metabolism. Here we report the discovery of a missense mutation in microsomal triglyceride transfer protein (Mtp), a protein that is essential for B-lp production. This mutation of a conserved glycine residue to valine (zebrafish G863V, human G865V) reduces B-lp production and results in yolk opacity due to aberrant accumulation of cytoplasmic lipid droplets in the YSL. However, this phenotype is milder than that of the previously reported L475P stalactite (stl) mutation. MTP transfers lipids, including triglycerides and phospholipids, to apolipoprotein B in the ER for B-lp assembly. In vitro lipid transfer assays reveal that while both MTP mutations eliminate triglyceride transfer activity, the G863V mutant protein unexpectedly retains ~80% of phospholipid transfer activity. This residual phospholipid transfer activity of the G863V mttp mutant protein is sufficient to support the secretion of small B-lps, which prevents intestinal fat malabsorption and growth defects observed in the mttpstl/stl mutant zebrafish. Modeling based on the recent crystal structure of the heterodimeric human MTP complex suggests the G865V mutation may block triglyceride entry into the lipid-binding cavity. Together, these data argue that selective inhibition of MTP triglyceride transfer activity may be a feasible therapeutic approach to treat dyslipidemia and provide structural insight for drug design. These data also highlight the power of yolk transport studies to identify proteins critical for B-lp biology.


Asunto(s)
Proteínas Portadoras/genética , Lípidos/genética , Lipoproteínas/genética , Triglicéridos/genética , Animales , Hígado Graso/genética , Hígado Graso/patología , Tracto Gastrointestinal/metabolismo , Humanos , Inmunoprecipitación , Gotas Lipídicas/metabolismo , Lipoproteínas/metabolismo , Mutación Missense/genética , Mutación Puntual/genética , Transporte de Proteínas/genética , Triglicéridos/metabolismo , Pez Cebra/genética
8.
Proc Natl Acad Sci U S A ; 116(35): 17251-17260, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31395737

RESUMEN

Microsomal triglyceride transfer protein (MTP) plays an essential role in lipid metabolism, especially in the biogenesis of very low-density lipoproteins and chylomicrons via the transfer of neutral lipids and the assembly of apoB-containing lipoproteins. Our understanding of the molecular mechanisms of MTP has been hindered by a lack of structural information of this heterodimeric complex comprising an MTPα subunit and a protein disulfide isomerase (PDI) ß-subunit. The structure of MTP presented here gives important insights into the potential mechanisms of action of this essential lipid transfer molecule, structure-based rationale for previously reported disease-causing mutations, and a means for rational drug design against cardiovascular disease and obesity. In contrast to the previously reported structure of lipovitellin, which has a funnel-like lipid-binding cavity, the lipid-binding site is encompassed in a ß-sandwich formed by 2 ß-sheets from the C-terminal domain of MTPα. The lipid-binding cavity of MTPα is large enough to accommodate a single lipid. PDI independently has a major role in oxidative protein folding in the endoplasmic reticulum. Comparison of the mechanism of MTPα binding by PDI with previously published structures gives insights into large protein substrate binding by PDI and suggests that the previous structures of human PDI represent the "substrate-bound" and "free" states rather than differences arising from redox state.


Asunto(s)
Proteínas Portadoras/química , Sitios de Unión , Cristalografía por Rayos X , Humanos , Conformación Proteica en Lámina beta
9.
Int J Mol Sci ; 23(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36499069

RESUMEN

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.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/química , Disulfuros/química , Proteínas Recombinantes/metabolismo , Citoplasma/metabolismo , Inmunoglobulina G/metabolismo , Proteínas Recombinantes de Fusión/química
10.
J Biol Chem ; 295(26): 8647-8655, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32102847

RESUMEN

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.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Oxidorreductasas/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Pliegue de Proteína , Degradación Asociada con el Retículo Endoplásmico , Humanos , Glicoproteínas de Membrana/química , Oxidación-Reducción , Oxidorreductasas/química , Consumo de Oxígeno , Proteína Disulfuro Isomerasas/química , Respuesta de Proteína Desplegada
11.
Protein Expr Purif ; 180: 105809, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33338588

RESUMEN

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.


Asunto(s)
Alérgenos , Glicoproteínas , Pliegue de Proteína , Alérgenos/biosíntesis , Alérgenos/química , Alérgenos/genética , Alérgenos/aislamiento & purificación , Animales , Gatos , Glicoproteínas/biosíntesis , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
12.
Hum Mol Genet ; 27(24): 4288-4302, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30239752

RESUMEN

The development of tissue fibrosis is complex and at the present time, not fully understood. Fibrosis, neurodegeneration and cerebral angiomatosis (FINCA disease) have been described in patients with mutations in NHL repeat-containing protein 2 (NHLRC2). However, the molecular functions of NHLRC2 are uncharacterized. Herein, we identified putative interacting partners for NHLRC2 using proximity-labeling mass spectrometry. We also investigated the function of NHLRC2 using immortalized cells cultured from skin biopsies of FINCA patients and normal fibroblasts with NHLRC2 knock-down and NHLRC2 overexpressing gene modifications. Transmission electron microscopy analysis of immortalized cell cultures from three FINCA patients demonstrated multilamellar bodies and distinctly organized vimentin filaments. Additionally, two of three cultures derived from patient skin biopsies contained cells that exhibited features characteristic of myofibroblasts. Altogether, the data presented in this study show for the first time that NHLRC2 is involved in cellular organization through regulation of the cytoskeleton and vesicle transport. We conclude that compound heterozygous p.Asp148Tyr and p.Arg201GlyfsTer6 mutations in NHLRC2 lead to severe tissue fibrosis in humans by enhancing the differentiation of fibroblasts to myofibroblasts.


Asunto(s)
Angiomatosis/patología , Encefalopatías/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miofibroblastos/patología , Degeneración Nerviosa/genética , Actinas/genética , Angiomatosis/genética , Encefalopatías/genética , Diferenciación Celular/genética , Células Cultivadas , Fibrosis , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación/genética , Miofibroblastos/metabolismo , Piel/metabolismo , Piel/patología
13.
EMBO J ; 35(8): 845-65, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26869642

RESUMEN

Disturbance of endoplasmic reticulum (ER) proteostasis is a common feature of amyotrophic lateral sclerosis (ALS). Protein disulfide isomerases (PDIs) areERfoldases identified as possibleALSbiomarkers, as well as neuroprotective factors. However, no functional studies have addressed their impact on the disease process. Here, we functionally characterized fourALS-linked mutations recently identified in two majorPDIgenes,PDIA1 andPDIA3/ERp57. Phenotypic screening in zebrafish revealed that the expression of thesePDIvariants induce motor defects associated with a disruption of motoneuron connectivity. Similarly, the expression of mutantPDIs impaired dendritic outgrowth in motoneuron cell culture models. Cellular and biochemical studies identified distinct molecular defects underlying the pathogenicity of thesePDImutants. Finally, targetingERp57 in the nervous system led to severe motor dysfunction in mice associated with a loss of neuromuscular synapses. This study identifiesERproteostasis imbalance as a risk factor forALS, driving initial stages of the disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Neuronas Motoras/patología , Procolágeno-Prolina Dioxigenasa/genética , Proteína Disulfuro Isomerasas/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Animales Modificados Genéticamente , Electromiografía , Embrión no Mamífero , Estrés del Retículo Endoplásmico/genética , Humanos , Ratones Noqueados , Mutación , Neuritas/patología , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
14.
Protein Expr Purif ; 165: 105498, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31521797

RESUMEN

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.


Asunto(s)
Citoplasma/metabolismo , Escherichia coli/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/aislamiento & purificación , Lipasa/genética , Lipasa/aislamiento & purificación , Secuencia de Bases , Catálisis , Clonación Molecular , Disulfuros/metabolismo , Escherichia coli/ultraestructura , Fermentación , Proteínas Fúngicas/metabolismo , Regulación Bacteriana de la Expresión Génica , Lipasa/metabolismo , Oxidación-Reducción , Conformación Proteica , Transducción de Señal , Transfección
15.
Int J Mol Sci ; 21(3)2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-31973001

RESUMEN

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.


Asunto(s)
Citoplasma/metabolismo , Proteínas de Escherichia coli/biosíntesis , Escherichia coli/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Animales , Disulfuros/química , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Proteoglicanos de Heparán Sulfato , Cuerpos de Inclusión/metabolismo , Mucina 2/metabolismo , Periplasma/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica
16.
Biochem Soc Trans ; 47(5): 1223-1231, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31671179

RESUMEN

Disulfide bond formation is an essential post-translational modification required for many proteins to attain their native, functional structure. The formation of disulfide bonds, otherwise known as oxidative protein folding, occurs in the endoplasmic reticulum and mitochondrial inter-membrane space in eukaryotes and the periplasm of prokaryotes. While there are differences in the molecular mechanisms of oxidative folding in different compartments, it can essentially be broken down into two steps, disulfide formation and disulfide isomerization. For both steps, catalysts exist in all compartments where native disulfide bond formation occurs. Due to the importance of disulfide bonds for a plethora of proteins, considerable effort has been made to generate cell factories which can make them more efficiently and cheaper. Recently synthetic biology has been used to transfer catalysts of native disulfide bond formation into the cytoplasm of prokaryotes such as Escherichia coli. While these engineered systems cannot yet rival natural systems in the range and complexity of disulfide-bonded proteins that can be made, a growing range of proteins have been made successfully and yields of homogenously folded eukaryotic proteins exceeding g/l yields have been obtained. This review will briefly give an overview of such systems, the uses reported to date and areas of future potential development, including combining with engineered systems for cytoplasmic glycosylation.


Asunto(s)
Citoplasma/metabolismo , Disulfuros/metabolismo , Catálisis , Escherichia coli/metabolismo , Glicosilación , Procesamiento Proteico-Postraduccional
17.
Acta Neuropathol ; 135(5): 727-742, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29423877

RESUMEN

A novel multi-organ disease that is fatal in early childhood was identified in three patients from two non-consanguineous families. These children were born asymptomatic but at the age of 2 months they manifested progressive multi-organ symptoms resembling no previously known disease. The main clinical features included progressive cerebropulmonary symptoms, malabsorption, progressive growth failure, recurrent infections, chronic haemolytic anaemia and transient liver dysfunction. In the affected children, neuropathology revealed increased angiomatosis-like leptomeningeal, cortical and superficial white matter vascularisation and congestion, vacuolar degeneration and myelin loss in white matter, as well as neuronal degeneration. Interstitial fibrosis and previously undescribed granuloma-like lesions were observed in the lungs. Hepatomegaly, steatosis and collagen accumulation were detected in the liver. A whole-exome sequencing of the two unrelated families with the affected children revealed the transmission of two heterozygous variants in the NHL repeat-containing protein 2 (NHLRC2); an amino acid substitution p.Asp148Tyr and a frameshift 2-bp deletion p.Arg201GlyfsTer6. NHLRC2 is highly conserved and expressed in multiple organs and its function is unknown. It contains a thioredoxin-like domain; however, an insulin turbidity assay on human recombinant NHLRC2 showed no thioredoxin activity. In patient-derived fibroblasts, NHLRC2 levels were low, and only p.Asp148Tyr was expressed. Therefore, the allele with the frameshift deletion is likely non-functional. Development of the Nhlrc2 null mouse strain stalled before the morula stage. Morpholino knockdown of nhlrc2 in zebrafish embryos affected the integrity of cells in the midbrain region. This is the first description of a fatal, early-onset disease; we have named it FINCA disease based on the combination of pathological features that include fibrosis, neurodegeneration, and cerebral angiomatosis.


Asunto(s)
Angiomatosis/genética , Encefalopatías/genética , Variación Genética , Péptidos y Proteínas de Señalización Intracelular/genética , Enfermedades Neurodegenerativas/genética , Fibrosis Pulmonar/genética , Angiomatosis/patología , Angiomatosis/fisiopatología , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Encéfalo/patología , Encefalopatías/patología , Encefalopatías/fisiopatología , Células Cultivadas , Familia , Resultado Fatal , Humanos , Lactante , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hepatopatías/genética , Hepatopatías/patología , Hepatopatías/fisiopatología , Masculino , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Estudios Prospectivos , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/fisiopatología , Síndrome , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
18.
Microb Cell Fact ; 16(1): 108, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28619018

RESUMEN

BACKGROUND: The production of recombinant proteins containing disulfide bonds in Escherichia coli is challenging. In most cases the protein of interest needs to be either targeted to the oxidizing periplasm or expressed in the cytoplasm in the form of inclusion bodies, then solubilized and re-folded in vitro. Both of these approaches have limitations. Previously we showed that soluble expression of disulfide bonded proteins in the cytoplasm of E. coli is possible at shake flask scale with a system, known as CyDisCo, which is based on co-expression of a protein of interest along with a sulfhydryl oxidase and a disulfide bond isomerase. With CyDisCo it is possible to produce disulfide bonded proteins in the presence of intact reducing pathways in the cytoplasm. RESULTS: Here we scaled up production of four disulfide bonded proteins to stirred tank bioreactors and achieved high cell densities and protein yields in glucose fed-batch fermentations, using an E. coli strain (BW25113) with the cytoplasmic reducing pathways intact. Even without process optimization production of purified human single chain IgA1 antibody fragment reached 139 mg/L and hen avidin 71 mg/L, while purified yields of human growth hormone 1 and interleukin 6 were around 1 g/L. Preliminary results show that human growth hormone 1 was also efficiently produced in fermentations of W3110 strain and when glucose was replaced with glycerol as the carbon source. CONCLUSIONS: Our results show for the first time that efficient production of high yields of soluble disulfide bonded proteins in the cytoplasm of E. coli with the reducing pathways intact is feasible to scale-up to bioreactor cultivations on chemically defined minimal media.


Asunto(s)
Citoplasma/química , Disulfuros/química , Escherichia coli/genética , Animales , Avidina/análisis , Avidina/biosíntesis , Avidina/genética , Reactores Biológicos , Pollos , Medios de Cultivo/química , Citoplasma/metabolismo , Escherichia coli/química , Escherichia coli/citología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Femenino , Fermentación , Glucosa/metabolismo , Glicerol/metabolismo , Hormona de Crecimiento Humana/biosíntesis , Hormona de Crecimiento Humana/genética , Humanos , Fragmentos de Inmunoglobulinas/biosíntesis , Fragmentos de Inmunoglobulinas/genética , Cuerpos de Inclusión/química , Cuerpos de Inclusión/metabolismo , Interleucina-6/biosíntesis , Interleucina-6/genética , Oxidación-Reducción , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química
19.
Biochim Biophys Acta ; 1853(3): 756-63, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25554517

RESUMEN

Numerous therapeutic proteins are expressed in Escherichia coli and targeted to the periplasm in order to facilitate purification and enable disulfide bond formation. Export is normally achieved by the Sec pathway, which transports proteins through the plasma membrane in a reduced, unfolded state. The Tat pathway is a promising alternative means of export, because it preferentially exports correctly folded proteins; however, the reducing cytoplasm of standard strains has been predicted to preclude export by Tat of proteins that contain disulfide bonds in the native state because, in the reduced state, they are sensed as misfolded and rejected. Here, we have tested a series of disulfide-bond containing biopharmaceuticals for export by the Tat pathway in CyDisCo strains that do enable disulfide bond formation in the cytoplasm. We show that interferon α2b, human growth hormone (hGH) and two antibody fragments are exported with high efficiency; surprisingly, however, they are efficiently exported even in the absence of cytoplasmic disulfide formation. The exported proteins acquire disulfide bonds in the periplasm, indicating that the normal disulfide oxidation machinery is able to act on the proteins. Tat-dependent export of hGH proceeds even when the disulfide bonds are removed by substitution of the Cys residues involved, suggesting that these substrates adopt tertiary structures that are accepted as fully-folded by the Tat machinery.


Asunto(s)
Disulfuros/metabolismo , Proteínas de Escherichia coli/fisiología , Hormona de Crecimiento Humana/metabolismo , Fragmentos de Inmunoglobulinas/metabolismo , Interferón-alfa/metabolismo , Proteínas de Transporte de Membrana/fisiología , Periplasma/metabolismo , Secuencia de Aminoácidos , Anticuerpos/química , Anticuerpos/metabolismo , Disulfuros/química , Escherichia coli/metabolismo , Humanos , Interferón alfa-2 , Redes y Vías Metabólicas , Datos de Secuencia Molecular , Organismos Modificados Genéticamente , Oxidación-Reducción , Transporte de Proteínas , Proteínas Recombinantes/metabolismo
20.
Microb Cell Fact ; 15: 22, 2016 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-26809624

RESUMEN

BACKGROUND: Disulfide bonds are the most common structural, post-translational modification found in proteins. Antibodies contain up to 25 disulfide bonds depending on type, with scFv fragments containing two disulfides and Fab fragments containing five or six disulfide bonds. The production of antibody fragments that contain native disulfide bonds can be challenging, especially on a large scale. The protein needs to be targeted to prokaryotic periplasm or the eukaryotic endoplasmic reticulum. These compartments are specialised for disulfide bond formation, but both compartments have limitations. RESULTS: Here we show that the introduction into the cytoplasm of a catalyst of disulfide bond formation and a catalyst of disulfide bond isomerization allows the efficient formation of natively folded scFv and Fab antibody fragments in the cytoplasm of Escherichia coli with intact reducing pathways. Eleven scFv and eleven Fab fragments were screened and ten of each were obtained in yields of >5 mg/L from deep-well plates. Production of eight of the scFv and all ten of the Fab showed a strong dependence on the addition of the folding factors. Yields of purified scFv of up to 240 mg/L and yields of purified Fab fragments of up to 42 mg/L were obtained. Purified fragments showed circular dichroism spectra consistent with being natively folded and were biologically active. CONCLUSIONS: Our results show that the efficient production of soluble, biologically active scFv and Fab antibody fragments in the cytoplasm of E. coli is not only possible, but facile. The required components can be easily transferred between different E. coli strains.


Asunto(s)
Citoplasma/metabolismo , Escherichia coli/metabolismo , Fragmentos de Inmunoglobulinas/metabolismo , Ingeniería de Proteínas/métodos , Animales , Anticuerpos/aislamiento & purificación , Vectores Genéticos/metabolismo , Humanos , Fragmentos de Inmunoglobulinas/aislamiento & purificación , Ratones , Peso Molecular , Plásmidos/metabolismo , Anticuerpos de Cadena Única/metabolismo , Solubilidad , Espectrometría de Masa por Ionización de Electrospray
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA