Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 353
Filter
1.
Int J Biol Macromol ; 186: 414-423, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34246679

ABSTRACT

Polyhydroxyalkanoates (PHAs) are biopolyesters synthesized by microorganisms as intracellular energy reservoirs under stressful environmental conditions. PHA synthase (PhaC) is the key enzyme responsible for PHA biosynthesis, but the importance of its N- and C-terminal ends still remains elusive. Six plasmid constructs expressing truncation variants of Aquitalea sp. USM4 PhaC (PhaC1As) were generated and heterologously expressed in Cupriavidus necator PHB-4. Removal of the first six residues at the N-terminus enabled the modulation of PHA composition without altering the PHA content in cells. Meanwhile, deletion of 13 amino acids from the C-terminus greatly affected the catalytic activity of PhaC1As, retaining only 1.1-7.4% of the total activity. Truncation(s) at the N- and/or C-terminus of PhaC1As gradually diminished the incorporation of comonomer units, and revealed that the N-terminal region is essential for PhaC1As dimerization whereas the C-terminal region is required for stabilization. Notably, transmission electron microscopy analysis showed that PhaC modification affected the morphology of intracellular PHA granules, which until now is only known to be regulated by phasins. This study provided substantial evidence and highlighted the significance of both the N- and C-termini of PhaC1As in regulating intracellular granule morphology, activity, substrate specificity, dimerization and stability of the synthase.


Subject(s)
Acyltransferases/metabolism , Betaproteobacteria/enzymology , Inclusion Bodies/enzymology , Polyhydroxyalkanoates/metabolism , Acyltransferases/chemistry , Acyltransferases/genetics , Betaproteobacteria/genetics , Betaproteobacteria/ultrastructure , Binding Sites , Catalytic Domain , Enzyme Stability , Inclusion Bodies/genetics , Inclusion Bodies/ultrastructure , Protein Domains , Protein Multimerization , Structure-Activity Relationship , Substrate Specificity
2.
Bioprocess Biosyst Eng ; 44(10): 2129-2139, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34036441

ABSTRACT

Tobacco etch virus protease (TEVp) is an enzymatic reagent to remove fusion tag, but additional purification steps are required for removing the TEVp after cleavage reaction is finished. Use of carrier-free and dependent TEVp immobilizates can eliminate protease contamination. In this work, we identified that, among the four constructed missense variants, the insoluble variant with the highest activity was correspondent with the soluble one tested formerly. The activities of the insoluble 15 codon variants were assayed and the variant with highest activity was selected. The K45F and/or E106G mutations have been reported on slightly improving protein stability of the wild-type TEVp, but only E106G mutation enhanced soluble production and activity of the selected TEVp variant, and it increased soluble amounts of two codon variants with the impaired folding. The decreased activity and use efficiency of the optimized TEVp variant in inclusion bodies was balanced by the determined high level production, lower leaking amounts of the protein, the enhanced resistance to the limited proteolysis mediated by protease K and trypsin, and the increased inhibition of auto-cleavage, as comparison to those of the immobilized soluble one. Thus, the TEVp construct is a potential alternate for simplifying protein purification protocols after tag-removal.


Subject(s)
Endopeptidases/metabolism , Inclusion Bodies/enzymology , Mutation , Affinity Labels , Amino Acid Sequence , Chromatography, Affinity , Endopeptidases/genetics , Endopeptidases/isolation & purification , Enzymes, Immobilized/genetics , Enzymes, Immobilized/isolation & purification , Enzymes, Immobilized/metabolism
3.
Infect Immun ; 88(12)2020 11 16.
Article in English | MEDLINE | ID: mdl-32900818

ABSTRACT

The obligate intracellular pathogen Chlamydia trachomatis is the leading cause of noncongenital blindness and causative agent of the most common sexually transmitted infection of bacterial origin. With a reduced genome, C. trachomatis is dependent on its host for survival, in part due to a need for the host cell to compensate for incomplete bacterial metabolic pathways. However, relatively little is known regarding how C. trachomatis is able to hijack host cell metabolism. In this study, we show that two host glycolytic enzymes, aldolase A and pyruvate kinase, as well as lactate dehydrogenase, are enriched at the C. trachomatis inclusion membrane during infection. Inclusion localization was not species specific, since a similar phenotype was observed with C. muridarum Time course experiments showed that the number of positive inclusions increased throughout the developmental cycle. In addition, these host enzymes colocalized to the same inclusion, and their localization did not appear to be dependent on sustained bacterial protein synthesis or on intact host actin, vesicular trafficking, or microtubules. Depletion of the host glycolytic enzyme aldolase A resulted in decreased inclusion size and infectious progeny production, indicating a role for host glycolysis in bacterial growth. Finally, quantitative PCR analysis showed that expression of C. trachomatis glycolytic enzymes inversely correlated with host enzyme localization at the inclusion. We discuss potential mechanisms leading to inclusion localization of host glycolytic enzymes and how it could benefit the bacteria. Altogether, our findings provide further insight into the intricate relationship between host and bacterial metabolism during Chlamydia infection.


Subject(s)
Chlamydia Infections/metabolism , Chlamydia trachomatis/metabolism , Fructose-Bisphosphate Aldolase/metabolism , Glycolysis , Host Microbial Interactions , Inclusion Bodies/metabolism , L-Lactate Dehydrogenase/metabolism , Pyruvate Kinase/metabolism , Actins/metabolism , Bacterial Outer Membrane/enzymology , Bacterial Outer Membrane/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chlamydia Infections/enzymology , Chlamydia Infections/genetics , Chlamydia muridarum/metabolism , Chlamydia trachomatis/enzymology , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/pathogenicity , Fructose-Bisphosphate Aldolase/genetics , HeLa Cells , Humans , Inclusion Bodies/enzymology , Inclusion Bodies/microbiology , L-Lactate Dehydrogenase/genetics , Microtubules/metabolism , Protein Biosynthesis/drug effects , Pyruvate Kinase/genetics
4.
Biotechnol Bioeng ; 117(5): 1446-1457, 2020 05.
Article in English | MEDLINE | ID: mdl-32043560

ABSTRACT

Enzyme clustering into compact agglomerates could accelerate the processing of intermediates to enhance metabolic pathway flux. However, enzyme clustering is still a challenging task due to the lack of universal assembly strategy applicable to all enzymes. Therefore, we proposed an alternative enzyme assembly strategy based on functional inclusion bodies. First, functional inclusion bodies in cells were formed by the fusion expression of stomatin/prohibitin/flotillin/HflK/C (SPFH) domain and enhanced green fluorescent protein, as observed visually and by transmission electron microscopy. The formation of SPFH-induced functional inclusion bodies enhanced intermolecular polymerization as revealed by further analysis combined with Förster resonance energy transfer and bimolecular fluorescent complimentary. Finally, the functional inclusion bodies significantly improved the enzymatic catalysis in living cells, as proven by the examples with whole-cell biocatalysis of phenyllactic acid by Escherichia coli, and the production of N-acetylglucosamine by Bacillus subtilis. Our findings suggest that SPFH-induced functional inclusion bodies can enhance the cascade reaction of enzymes, to serve as a potential universal strategy for the construction of efficient microbial cell factories.


Subject(s)
Enzymes , Inclusion Bodies , Metabolic Engineering/methods , Recombinant Fusion Proteins , Acetylglucosamine/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Biocatalysis , Enzymes/chemistry , Enzymes/genetics , Enzymes/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inclusion Bodies/enzymology , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Lactates/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
Appl Microbiol Biotechnol ; 104(2): 643-652, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31788710

ABSTRACT

Immobilization of the enzyme benefits the catalytic industry a lot. The gram-positive enhancer matrix (GEM) particles could purify and immobilize the recombinant α-amylase in one step without changing the enzymatic character. The enzyme immobilized by GEM particles exhibited good reusability and storage stability. The denaturants dissolved some of the GEM particles and a part of the GEM particles could bear the denaturants. The GEM particles had strong binding ability to the recombination protein with the AcmA tag even when the denaturants existed. The inclusion body was dissolved by urea and then bound by the GEM particles. The GEM particles binding the recombination protein were separated by centrifugation and resuspended in the renaturation solution. GEM particles were recycled by repeating the boiling procedure used in preparing them. The recombination α-amylase without any tag was obtained by digestion and separated via centrifugation. Altogether, our findings suggest that GEM particles have the potential to function as both immobilization and purification materials to bind the soluble recombinant protein with the AcmA tag and the inclusion body dissolved in the denaturants.


Subject(s)
Enzymes, Immobilized/isolation & purification , Inclusion Bodies/enzymology , Recombinant Proteins/isolation & purification , alpha-Amylases/isolation & purification , Enzyme Stability , Escherichia coli/enzymology , Protein Binding
7.
Protein J ; 38(1): 30-36, 2019 02.
Article in English | MEDLINE | ID: mdl-30666487

ABSTRACT

HIV-1 is an infectious virus that causes acquired immunodeficiency syndrome (AIDS) and it is one of the major causes of deaths worldwide. The production of HIV-1 protease (PR) on a large scale has been a problem for scientists due to its cytotoxicity, low yield, insolubility, and low activity. HIV-1 C-SA protease has been cloned, expressed, and purified previously, however, with low recovery (0.25 mg/L). Herein we report an optimal expression and solubilisation procedure to recover active HIV-1 C-SA protease enzyme from inclusion bodies. The HIV protease was expressed in seven different vectors (pET11b, pET15b, pET28a pET32a, pET39b, pET41b and pGEX 6P-1). The highest expression was achieved when the vector pET32a (Trx tag) was employed. A total of 19.5 mg of fusion protein was refolded of which 5.5 mg of active protease was obtained after cleavage. The free protease had a high specific activity of 2.81 µmoles/min/mg. Interestingly the Trx-fusion protein also showed activity closer (1.24 µmoles/min/mg) to that of the free protease suggesting that the pET32a vector (Trx tag) expressed in BL21(DE3) pLysS provides a more efficient way to obtain HIV-1 protease.


Subject(s)
HIV Protease/chemistry , HIV Protease/isolation & purification , HIV-1/enzymology , Inclusion Bodies/enzymology , Escherichia coli/chemistry , Escherichia coli/genetics , HIV Protease/genetics , HIV-1/genetics , Inclusion Bodies/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
8.
Colloids Surf B Biointerfaces ; 173: 769-775, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30384274

ABSTRACT

Bacterial inclusion bodies (IBs) were historically considered one of the major obstacles in protein production through recombinant DNA techniques and conceived as amorphous deposits formed by passive and rather unspecific structures of unfolded proteins aggregates. Subsequent studies demonstrated that IBs contained an important quantity of active protein. In this work, we proved that recombinant ß-galactosidase inclusion bodies (IBß-Gal) are functional aggregates. Moreover, they exhibit particular features distinct to the soluble version of the enzyme. The particulate enzyme was highly active against lactose in physiological and in acid pH and also retained its activity upon a pre-incubation at high temperature. IBß-Gal washing or dilution induced the spontaneous release of active enzymes from the supramolecular aggregates. Along this process, we observed a continuous change in the values of several kinetic parameters, including specific activity and Michaelis-Menten constant, measured in the IBß-Gal suspensions. Simultaneously, IBß-Gal turned into a more heterogeneous population where smaller particles appeared. The released protein exhibited secondary structure features more similar to those of the soluble species than to the aggregated enzyme. Concluding, IBß-Gal represents a reservoir and packed source of highly active and stable enzyme.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Inclusion Bodies/enzymology , Lactose/chemistry , beta-Galactosidase/chemistry , Cloning, Molecular , Enzyme Stability , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hot Temperature , Hydrogen-Ion Concentration , Inclusion Bodies/chemistry , Kinetics , Lactose/metabolism , Protein Aggregates , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility , Structure-Activity Relationship , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
9.
Biotechnol Lett ; 41(1): 159-169, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30446859

ABSTRACT

OBJECTIVE: To obtain active lipases for biodiesel production by refolding Proteus sp. lipase inclusion bodies expressed in E. coli. RESULTS: A lipase gene lipPN1 was cloned from Proteus sp. NH 2-2 and expressed in E. coli BL21(DE3). Non-reducing SDS-PAGE revealed that recombinant LipPN1(rLipPN1) were prone to form inclusion bodies as disulfide-linked dimers in E. coli. Site-directed mutagenesis confirmed that Cys85 in LipPN1 was involved in the dimer formation. After optimizing the inclusion body refolding conditions, the maximum lipase activity reached 1662 U/L. The refolded rLipPN1 exhibited highest activity toward p-nitrophenyl butyrate at pH 9.0 and 40 °C. It could be activated by Ca2+ with moderate tolerance to organic solvents. It could also convert soybean oil into biodiesel at a conversion ratio of 91.5%. CONCLUSION: Preventing the formation of disulfide bond could enhance the refolding efficiency of rLipPN1 inclusion bodies.


Subject(s)
Biofuels , Escherichia coli , Protein Refolding , Proteus , Amino Acid Substitution , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Inclusion Bodies/enzymology , Inclusion Bodies/genetics , Lipase/biosynthesis , Lipase/chemistry , Lipase/genetics , Mutagenesis, Site-Directed , Proteus/enzymology , Proteus/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
10.
mSphere ; 3(6)2018 12 19.
Article in English | MEDLINE | ID: mdl-30567898

ABSTRACT

Here we report on a new nanoscale secondary ion mass spectrometry (nanoSIMS) approach based on enzyme-mediated oxygen isotope exchange, which combines the visualization of general metabolic activity in the cytoplasm with insights into the activity of enzymes related to polyphosphate (polyP) inclusions. The polyP-accumulating strain of the large sulfur bacterium Beggiatoa was used as a model organism. Beggiatoa cultures were grown under oxic and anoxic conditions when exposed to either low- or high-sulfide conditions, which are known to influence polyP metabolism in this strain. Subsequent incubation with 18O-labeled water led to high 18O enrichments above the natural background in the cytoplasm and polyP granules derived from enzymatically mediated oxygen isotope exchange. The relative importance of polyP under the different sulfide regimes became evident by an apparent continued metabolic activity at polyP inclusions under stressfully high sulfide concentrations, in contrast to a decreased general metabolic activity in the cytoplasm. This finding confirms the role of polyP as a critical component in bacterial stress response and maintenance of a survival metabolism.IMPORTANCE Microbial organisms exert a large influence on the environment as they directly affect the turnover of essential elements. This is particularly true for polyphosphate-accumulating large sulfur bacteria, which can either accumulate phosphate as polyphosphate or degrade it and release phosphate into the environment, depending on environmental conditions. This study presents a new approach to simultaneously visualize general metabolic activity and enzymatic activity at polyphosphate granules by incubation with 18O-labeled water as the only stable isotope tracer. For this purpose, the well-studied Beggiatoa sp. strain 35Flor was used as a model organism and was exposed to different stress regimes. General metabolic activity was strongly impaired during high-stress regimes. In contrast, intense intracellular polyP cycling was not restricted to favorable or stressful conditions, highlighting the importance of polyP for general cell physiology, especially during hostile conditions. The nanoSIMS approach adds a new tool to study microorganisms involved in phosphorus cycling in the environment together with the identification of general metabolic activity.


Subject(s)
Beggiatoa/enzymology , Cytoplasm/enzymology , Enzymes/analysis , Inclusion Bodies/enzymology , Isotope Labeling , Oxygen Isotopes/metabolism , Spectrometry, Mass, Secondary Ion/methods , Polyphosphates/analysis
11.
Biologicals ; 56: 45-53, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30327235

ABSTRACT

The present work elucidates the production of recombinant human asparaginase (rhASP) under optimized fermentation and downstream processes in Escherichia coli. The maximum biomass yield of 6.7 g/L was achieved with fed-batch fermentation. The highest rhASP inclusion bodies recovery yield (91%) was achieved with the optimized lysis conditions. The 8.0 M urea at pH 8.5 has shown efficient solubilization (94%) of rhASP inclusion bodies. The refolding efficiency of rhASP increased at pH 8.5 (84%) and temperature 25°C (86%). The diluted rhASP solution was concentrated and partially purified (92%) using cross flow filtration. A single step ion exchange chromatography is successfully achieved the maximum purity of ≥ 97%. The molecular mass of purified rhASP is confirmed as 34.1 kDa by mass spectrometry. The secondary structure of rhASP is characterized by FT-IR spectroscopy based on the structural elements. Finally, cell proliferative assay of purified rhASP is signifies the similar biological activity over the standard.


Subject(s)
Asparaginase/biosynthesis , Autoantigens/biosynthesis , Recombinant Proteins/biosynthesis , Asparaginase/chemistry , Asparaginase/isolation & purification , Asparaginase/pharmacology , Autoantigens/chemistry , Autoantigens/isolation & purification , Autoantigens/pharmacology , Batch Cell Culture Techniques , Cell Proliferation/drug effects , Chromatography, Ion Exchange , Escherichia coli , Fermentation , Humans , Inclusion Bodies/enzymology , Protein Refolding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
12.
J Biochem ; 164(6): 407-414, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30101295

ABSTRACT

An active recombinant human pancreatic lipase (recHPL) was successfully prepared for the first time from the Escherichia coli expression system using short Strep-tag II (ST II). The recHPL-ST II was solubilized using 8 M urea from E.coli lysate and purified on a Strep-Tactin-Sepharose column. After refolding by stepwise dialyses in the presence of glycerol and Ca2+ for 2 days followed by gel filtration, 1.8-6 mg of active recHPL-ST II was obtained from 1 L of culture. The recHPL was non-glycosylated, but showed almost equal specific activity, pH-dependency and time-dependent stability compared to those of native porcine pancreatic lipase (PPL) at 37°C. However, the recHPL lost its lipolytic activity above 50°C, showing a lower heat-stability than that of native PPL, which retained half its activity at this temperature.


Subject(s)
Lipase/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Circular Dichroism , Dietary Supplements/adverse effects , Enzyme Inhibitors/pharmacology , Enzyme Replacement Therapy/adverse effects , Enzyme Stability , Escherichia coli/growth & development , Escherichia coli/metabolism , Glycosylation , Hot Temperature/adverse effects , Humans , Inclusion Bodies/enzymology , Inclusion Bodies/metabolism , Kinetics , Lipase/adverse effects , Lipase/antagonists & inhibitors , Lipase/chemistry , Lipase/genetics , Lipase/isolation & purification , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/isolation & purification , Oligopeptides/metabolism , Orlistat/pharmacology , Protein Conformation , Protein Processing, Post-Translational , Protein Refolding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Solubility , Sus scrofa
13.
Sci Rep ; 8(1): 5856, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29643457

ABSTRACT

Sustainable and eco-efficient alternatives for the production of platform chemicals, fuels and chemical building blocks require the development of stable, reusable and recyclable biocatalysts. Here we present a novel concept for the biocatalytic production of 1,5-diaminopentane (DAP, trivial name: cadaverine) using catalytically active inclusion bodies (CatIBs) of the constitutive L-lysine decarboxylase from E. coli (EcLDCc-CatIBs) to process L-lysine-containing culture supernatants from Corynebacterium glutamicum. EcLDCc-CatIBs can easily be produced in E. coli followed by a simple purification protocol yielding up to 43% dry CatIBs per dry cell weight. The stability and recyclability of EcLDCc-CatIBs was demonstrated in (repetitive) batch experiments starting from L-lysine concentrations of 0.1 M and 1 M. EcLDC-CatIBs exhibited great stability under reaction conditions with an estimated half-life of about 54 h. High conversions to DAP of 87-100% were obtained in 30-60 ml batch reactions using approx. 180-300 mg EcLDCc-CatIBs, respectively. This resulted in DAP titres of up to 88.4 g l-1 and space-time yields of up to 660 gDAP l-1 d-1 per gram dry EcLDCc-CatIBs. The new process for DAP production can therefore compete with the currently best fermentative process as described in the literature.


Subject(s)
Cadaverine/biosynthesis , Carboxy-Lyases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Inclusion Bodies/enzymology , Batch Cell Culture Techniques/methods , Biocatalysis , Bioreactors/microbiology , Carboxy-Lyases/genetics , Carboxy-Lyases/isolation & purification , Corynebacterium glutamicum/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Lysine/metabolism , Metabolic Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
14.
Int J Biol Macromol ; 111: 186-192, 2018 May.
Article in English | MEDLINE | ID: mdl-29305883

ABSTRACT

Protein aggregation is a major hindrance in many in vivo and in vitro studies of proteins. It results in the formation of inclusion bodies and non-functional aggregates. Chemical chaperones also known as osmolytes which are accumulated during the stress conditions in the cells can influence the protein stability through various mechanisms. They act as osmoprotectants and contribute to the protein folding by enabling the protein to bury the backbone into the core of protein fold. In the current study, we observed the effect of chemical chaperones from four different classes on the stability and functionality of aggregation prone protein zebrafish dihydrofolate reductase (zDHFR). We also used UV-visible and circular dichroism (CD) spectroscopy to explore the protecting action of chemical chaperones on the structure and activity of zDHFR in vitro and in vivo conditions.


Subject(s)
Molecular Chaperones/chemistry , Protein Folding , Tetrahydrofolate Dehydrogenase/chemistry , Animals , Inclusion Bodies/enzymology , Kinetics , Protein Stability , Zebrafish
15.
Bioengineered ; 9(1): 6-11, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28463573

ABSTRACT

The need for cost-effectively produced and improved biocatalysts for industrial, pharmaceutical and environmental processes is steadily increasing. While enzyme properties themselves can be improved via protein engineering, immobilization by attachment to carrier materials remains a critical step for stabilization and process implementation. A new emerging immobilization approach, the in situ immobilization, enables simultaneous production of highly active enzymes and carrier materials using bioengineering/synthetic biology of microbial cells. In situ enzyme immobilization holds the promise of cost-effective production of highly functional immobilized biocatalysts for uses such as in bioremediation, drug synthesis, bioenergy and food processing.


Subject(s)
Enzymes, Immobilized/chemistry , Inclusion Bodies/enzymology , Magnetosomes/enzymology , Polyhydroxyalkanoates/chemistry , Protein Engineering/methods , Adsorption , Biocatalysis , Biodegradation, Environmental , Cross-Linking Reagents/chemistry , Enzymes, Immobilized/genetics , Enzymes, Immobilized/metabolism , Food Handling/methods , Gene Expression , Inclusion Bodies/genetics , Magnetosomes/genetics , Proteolipids/chemical synthesis , Proteolipids/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Biotechnol Lett ; 39(11): 1733-1740, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28836020

ABSTRACT

OBJECTIVES: To optimize the production of active inclusion bodies (IBs) containing human D-amino acid oxidase (hDAAO) in Escherichia coli. RESULTS: The optimized initial codon region combined with the coexpressed rare tRNAs, fusion of each of the N-terminal partners including cellulose-binding module, thioredoxin, glutathione S-transferase and expressivity tag, deletion of the incorporated linker, and improvement of tRNA abundance affected the production and activity for oxidizing D-alanine of the hDAAO in IBs. Compared with the optimized fusion constructs and expression host, IBs yields and activity were increased to 2.6- and 2.8-fold respectively by changing the N-terminal codon bias of the hDAAO. The insoluble hDAAO codon variant displayed the same substrate specificity as the soluble one for oxidizing D-alanine, D-serine and D-aspartic acid. The freshly prepared hDAAO codon variant was used for analyzing the L-serine racemization activity of the bacterially expressed maize serine racemase. CONCLUSIONS: Optimization of the N-terminal codon bias combined with the coexpression of rare tRNAs is a novel and efficient approach to produce active IBs of the hDAAO.


Subject(s)
D-Amino-Acid Oxidase/genetics , Inclusion Bodies/enzymology , RNA, Transfer/genetics , Codon , D-Amino-Acid Oxidase/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Humans , Recombinant Fusion Proteins/metabolism
17.
Biochem Biophys Res Commun ; 490(4): 1250-1253, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28684315

ABSTRACT

Inclusion bodies are often formed when the foreign protein is over expressed in Escherichia coli. Since proteins in inclusion bodies are inactive, denaturing and refolding of inclusion body proteins are necessary to obtain the active form. Instead of the conventional denaturants, urea and guanidine hydrochloride, a strong anionic detergent SDS was used to solubilize C-terminal His-tag form of ulvan lyase in the inclusion bodies. Solution containing SDS-solubilized enzyme were kept on ice to precipitate SDS, followed by SDS-KCl insoluble crystal formation to remove SDS completely. After removing the precipitate by centrifugation, the supernatant was applied to Ni-NTA column to purify His-tagged ulvan lyase. The purified protein showed a dimeric form and ulvan lyase activity, demonstrating that SDS-denatured protein was renatured and recovered enzyme activity. This simple method could be useful for refolding other inclusion body proteins.


Subject(s)
Detergents/pharmacology , Inclusion Bodies/enzymology , Polysaccharide-Lyases/metabolism , Sodium Dodecyl Sulfate/pharmacology , Escherichia coli/drug effects , Polysaccharide-Lyases/genetics , Protein Denaturation/drug effects , Time Factors , Ulva/enzymology
18.
Biophys J ; 112(12): 2540-2551, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28636911

ABSTRACT

The acylphosphatase from Sulfolobus solfataricus (Sso AcP) is a globular protein able to aggregate in vitro from a native-like conformational ensemble without the need for a transition across the major unfolding energy barrier. This process leads to the formation of assemblies in which the protein retains its native-like structure, which subsequently convert into amyloid-like aggregates. Here, we investigate the mechanism by which Sso AcP aggregates in vivo to form bacterial inclusion bodies after expression in E. coli. Shortly after the initiation of expression, Sso AcP is incorporated into inclusion bodies as a native-like protein, still exhibiting small but significant enzymatic activity. Additional experiments revealed that this overall process of aggregation is enhanced by the presence of the unfolded N-terminal region of the sequence and by destabilization of the globular segment of the protein. At later times, the Sso AcP molecules in the inclusion bodies lose their native-like properties and convert into ß-sheet-rich amyloid-like structures, as indicated by their ability to bind thioflavin T and Congo red. These results show that the aggregation behavior of this protein is similar in vivo to that observed in vitro, and that, at least for a predominant part of the protein population, the transition from a native to an amyloid-like structure occurs within the aggregate state.


Subject(s)
Acid Anhydride Hydrolases/chemistry , Archaeal Proteins/chemistry , Inclusion Bodies/enzymology , Protein Aggregates , Sulfolobus solfataricus/enzymology , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , Amyloid/chemistry , Amyloid/metabolism , Archaeal Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Stability , Escherichia coli , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Aggregation, Pathological , Protein Folding , Protein Structure, Secondary , Spectroscopy, Fourier Transform Infrared , Acylphosphatase
19.
J Biotechnol ; 258: 136-147, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28465211

ABSTRACT

Bacterial inclusion bodies (IBs) consist of unfolded protein aggregates and represent inactive waste products often accumulating during heterologous overexpression of recombinant genes in Escherichia coli. This general misconception has been challenged in recent years by the discovery that IBs, apart from misfolded polypeptides, can also contain substantial amounts of active and thus correctly or native-like folded protein. The corresponding catalytically-active inclusion bodies (CatIBs) can be regarded as a biologically-active sub-micrometer sized biomaterial or naturally-produced carrier-free protein immobilizate. Fusion of polypeptide (protein) tags can induce CatIB formation paving the way towards the wider application of CatIBs in synthetic chemistry, biocatalysis and biomedicine. In the present review we summarize the history of CatIBs, present the molecular-biological tools that are available to induce CatIB formation, and highlight potential lines of application. In the second part findings regarding the formation, architecture, and structure of (Cat)IBs are summarized. Finally, an overview is presented about the available bioinformatic tools that potentially allow for the prediction of aggregation and thus (Cat)IB formation. This review aims at demonstrating the potential of CatIBs for biotechnology and hopefully contributes to a wider acceptance of this promising, yet not widely utilized, protein preparation.


Subject(s)
Enzymes, Immobilized/metabolism , Inclusion Bodies/enzymology , Inclusion Bodies/metabolism , Recombinant Proteins/metabolism , Biotechnology , Enzymes, Immobilized/chemistry , Escherichia coli/metabolism , Recombinant Proteins/chemistry
20.
Microb Cell Fact ; 16(1): 16, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28137256

ABSTRACT

BACKGROUND: Through functional screening of a fosmid library, generated from a phytopathogen-suppressive soil metagenome, the novel antifungal chitinase-named Chi18H8 and belonging to family 18 glycosyl hydrolases-was previously discovered. The initial extremely low yield of Chi18H8 recombinant production and purification from Escherichia coli cells (21 µg/g cell) limited its characterization, thus preventing further investigation on its biotechnological potential. RESULTS: We report on how we succeeded in producing hundreds of milligrams of pure and biologically active Chi18H8 by developing and scaling up to a high-yielding, 30 L bioreactor process, based on a novel method of mild solubilization of E. coli inclusion bodies in lactic acid aqueous solution, coupled with a single step purification by hydrophobic interaction chromatography. Chi18H8 was characterized as a Ca2+-dependent mesophilic chitobiosidase, active on chitin substrates at acidic pHs and possessing interesting features, such as solvent tolerance, long-term stability in acidic environment and antifungal activity against the phytopathogens Fusarium graminearum and Rhizoctonia solani. Additionally, Chi18H8 was found to operate according to a non-processive endomode of action on a water-soluble chitin-like substrate. CONCLUSIONS: Expression screening of a metagenomic library may allow access to the functional diversity of uncultivable microbiota and to the discovery of novel enzymes useful for biotechnological applications. A persisting bottleneck, however, is the lack of methods for large scale production of metagenome-sourced enzymes from genes of unknown origin in the commonly used microbial hosts. To our knowledge, this is the first report on a novel metagenome-sourced enzyme produced in hundreds-of-milligram amount by recovering the protein in the biologically active form from recombinant E. coli inclusion bodies.


Subject(s)
Antifungal Agents/pharmacology , Chitinases/metabolism , Chitinases/pharmacology , Escherichia coli/genetics , Hexosaminidases/metabolism , Hexosaminidases/pharmacology , Soil Microbiology , Antifungal Agents/isolation & purification , Antifungal Agents/metabolism , Bioreactors , Chitin/metabolism , Chitinases/genetics , Chitinases/isolation & purification , Cloning, Molecular , Escherichia coli/metabolism , Fusarium/drug effects , Gene Library , Hexosaminidases/genetics , Hexosaminidases/isolation & purification , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Inclusion Bodies/enzymology , Lactic Acid/metabolism , Metagenome , Metagenomics/methods , Phylogeny , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Rhizoctonia/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...