Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.385
Filter
1.
Int J Biol Macromol ; 270(Pt 1): 131917, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679252

ABSTRACT

Enzymatic degradation of alginate for the preparation of alginate oligosaccharides (AOS) is currently receiving significant attention in the field. AOS has been shown to promote crop growth and improve plant resistance to abiotic stresses. In this study, two PL6 family alginate lyases, AlyRmA and AlyRmB, were expressed and characterized. These enzymes demonstrate exceptional activity and stable thermophilicity compared to other known alginate lyases. AlyRmA (8855.34 U/mg) and AlyRmB (7879.44 U/mg) exhibited excellent degradation activity towards sodium alginate even at high temperatures (70 °C). The AlyRmA and AlyRmB were characterized and utilized to efficiently produce AOS. The study investigated the promotional effect of AOS on the growth of Brassica napus L. seedlings in a saline-alkaline environment. The results of this study demonstrate the high activity and thermal stability of AlyRmA and AlyRmB, highlighting their potential in the preparation of AOS. Moreover, the application of AOS prepared by AlyRmB could enhance the resistance of Brassica napus L. to saline-alkali environments, thereby broadening the potential applications of AOS.


Subject(s)
Alginates , Brassica napus , Oligosaccharides , Polysaccharide-Lyases , Brassica napus/enzymology , Alginates/chemistry , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Polysaccharide-Lyases/metabolism , Polysaccharide-Lyases/chemistry , Alkalies/chemistry , Enzyme Stability/drug effects , Temperature , Hydrogen-Ion Concentration , Salinity , Seedlings/drug effects , Seedlings/growth & development , Seedlings/metabolism
2.
Sci Rep ; 11(1): 23584, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34880340

ABSTRACT

Transketolase (TK) has been previously engineered, using semi-rational directed evolution and substrate walking, to accept increasingly aliphatic, cyclic, and then aromatic substrates. This has ultimately led to the poor water solubility of new substrates, as a potential bottleneck to further exploitation of this enzyme in biocatalysis. Here we used a range of biophysical studies to characterise the response of both E. coli apo- and holo-TK activity and structure to a range of polar organic co-solvents: acetonitrile (AcCN), n-butanol (nBuOH), ethyl acetate (EtOAc), isopropanol (iPrOH), and tetrahydrofuran (THF). The mechanism of enzyme deactivation was found to be predominantly via solvent-induced local unfolding. Holo-TK is thermodynamically more stable than apo-TK and yet for four of the five co-solvents it retained less activity than apo-TK after exposure to organic solvents, indicating that solvent tolerance was not simply correlated to global conformational stability. The co-solvent concentrations required for complete enzyme inactivation was inversely proportional to co-solvent log(P), while the unfolding rate was directly proportional, indicating that the solvents interact with and partially unfold the enzyme through hydrophobic contacts. Small amounts of aggregate formed in some cases, but this was not sufficient to explain the enzyme inactivation. TK was found to be tolerant to 15% (v/v) iPrOH, 10% (v/v) AcCN, or 6% (v/v) nBuOH over 3 h. This work indicates that future attempts to engineer the enzyme to better tolerate co-solvents should focus on increasing the stability of the protein to local unfolding, particularly in and around the cofactor-binding loops.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Escherichia coli/metabolism , Solvents/chemistry , Transketolase/chemistry , Water/chemistry , Biocatalysis , Enzyme Stability/drug effects
3.
Cell Death Dis ; 12(9): 828, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34480022

ABSTRACT

Lung epithelial cell death is a prominent feature of acute lung injury and acute respiratory distress syndrome (ALI/ARDS), which results from severe pulmonary infection leading to respiratory failure. Multiple mechanisms are believed to contribute to the death of epithelia; however, limited data propose a role for epigenetic modifiers. In this study, we report that a chromatin modulator protein arginine N-methyltransferase 4/coactivator-associated arginine methyltransferase 1 (PRMT4/CARM1) is elevated in human lung tissues with pneumonia and in experimental lung injury models. Here PRMT4 is normally targeted for its degradation by an E3 ubiquitin ligase, SCFFBXO9, that interacts with PRMT4 via a phosphodegron to ubiquitinate the chromatin modulator at K228 leading to its proteasomal degradation. Bacterial-derived endotoxin reduced levels of SCFFBXO9 thus increasing PRMT4 cellular concentrations linked to epithelial cell death. Elevated PRMT4 protein caused substantial epithelial cell death via caspase 3-mediated cell death signaling, and depletion of PRMT4 abolished LPS-mediated epithelial cell death both in cellular and murine injury models. These findings implicate a unique molecular interaction between SCFFBXO9 and PRMT4 and its regulation by endotoxin that impacts the life span of lung epithelia, which may play a key role in the pathobiology of tissue injury observed during critical respiratory illness.


Subject(s)
Endotoxins/toxicity , Epithelial Cells/enzymology , Epithelial Cells/pathology , Lung/pathology , Protein-Arginine N-Methyltransferases/metabolism , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Caspase 3/metabolism , Cell Death/drug effects , Cell Line , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Epithelial Cells/drug effects , F-Box Proteins/metabolism , Humans , Lysine/metabolism , Mice , Models, Biological , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Ubiquitin/metabolism , Ubiquitination/drug effects
4.
Int J Biol Macromol ; 188: 1012-1024, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34375665

ABSTRACT

The oxygenases have attracted considerable attention in enzyme-mediated bioremediation of xenobiotic compounds due to their high specificity, cost-effectiveness, and targeted field applications. Here, we performed a functional metagenomics approach to cope with culturability limitations to isolate a novel extradiol dioxygenase. Fosmid clone harboring dioxygenase gene was sequenced and analyzed by bioinformatics tools. One ring-cleaving dioxygenase RW4-MPC (metapyrocatechase) was purified and characterized to examine its degradation efficiency. The RW4-MPC was significantly active in the temperature and pH range of 5 to 40 °C, and 7-10, respectively, with an optimum temperature of 25 °C and pH 8. To gain insight into observed differential activity, Small-Angle X-ray Scattering (SAXS) data of the protein samples were analyzed, which brought forth that the RW4-MPC molecules form tight globular tetramers in solution. This native association was stable till 35 °C, and protein started to associate at higher temperatures, explaining heat-induced loss of function. Similarly, RW4-MPC aggregated or lost globular profile below pH 7 or at pH 10, respectively. The kinetic parameters showed the six folds high catalytic efficiency of RW4-MPC towards 2,3-dihydroxy biphenyl than catechol and its derivatives. RW4-MPC molecules showed remarkable retention of functionality in hypersaline conditions with more than 70% activity in a buffer having 3 M NaCl concentration. In concordance, SAXS data analysis showed retention of functional shape profile in hypersaline conditions. The halotolerant and oxygen insensitive nature of this enzyme makes it a potential candidate for bioremediation.


Subject(s)
Catechol 2,3-Dioxygenase/chemistry , Catechol 2,3-Dioxygenase/metabolism , Metagenomics , Scattering, Small Angle , X-Ray Diffraction , Amino Acid Sequence , Catechol 2,3-Dioxygenase/isolation & purification , Circular Dichroism , Clone Cells , Enzyme Stability/drug effects , Hydrogen-Ion Concentration , Ions , Kinetics , Metals/pharmacology , Molecular Weight , Phylogeny , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sodium Chloride/pharmacology , Substrate Specificity/drug effects , Temperature
5.
Int J Biol Macromol ; 183: 1939-1947, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34097957

ABSTRACT

Protein aggregation, such as amyloid fibril formation, is molecular hallmark of many neurodegenerative disorders including Alzheimer's, Parkinson's, and Prion disease. Indole alkaloids are well-known as the compounds having the ability to inhibit protein fibrillation. In this study, we experimentally and computationally have investigated the anti-amyloid property of a derivative of a synthesized tetracyclic indole alkaloid (TCIA), possessing capable functional groups. The fibrillation reaction of Hen White Egg Lysozyme (HEWL) was performed in absence and presence of the indole alkaloid. For quantitative analysis, we used Thioflovin T binding assay which showed ~50% reduction in fibril formation in the presence of 20 µM TCIA. Using TEM imaging, we observed a significant morphological change in our model protein in the presence of TCIA. In addition, we exploited FT-IR assay by which Amide I peak's shifting toward lower wavenumber was clearly observed. Using Molecular Docking, the interaction of the inhibitor (TCIA) with the protein's amyloidogenic region was modeled. Also, different biophysical parameters were calculated by Molecular Dynamics (MD) simulation. Various biochemical assays, conformational change, and hydrophobicity exposure of the protein during amyloid formation indicated that the compound assists HEWL to keep its native structure via destabilizing ß-sheet structure.


Subject(s)
Benzothiazoles/chemistry , Indole Alkaloids/pharmacology , Muramidase/chemistry , Animals , Chickens , Enzyme Stability/drug effects , Hydrophobic and Hydrophilic Interactions , Indole Alkaloids/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Muramidase/drug effects , Protein Aggregates/drug effects , Protein Structure, Secondary/drug effects , Spectroscopy, Fourier Transform Infrared
6.
SLAS Discov ; 26(8): 1029-1039, 2021 09.
Article in English | MEDLINE | ID: mdl-34167376

ABSTRACT

Triose phosphate isomerase deficiency (TPI Df) is an untreatable, childhood-onset glycolytic enzymopathy. Patients typically present with frequent infections, anemia, and muscle weakness that quickly progresses with severe neuromusclar dysfunction requiring aided mobility and often respiratory support. Life expectancy after diagnosis is typically ~5 years. There are several described pathogenic mutations that encode functional proteins; however, these proteins, which include the protein resulting from the "common" TPIE105D mutation, are unstable due to active degradation by protein quality control (PQC) pathways. Previous work has shown that elevating mutant TPI levels by genetic or pharmacological intervention can ameliorate symptoms of TPI Df in fruit flies. To identify compounds that increase levels of mutant TPI, we have developed a human embryonic kidney (HEK) stable knock-in model expressing the common TPI Df protein fused with green fluorescent protein (HEK TPIE105D-GFP). To directly address the need for lead TPI Df therapeutics, these cells were developed into an optical drug discovery platform that was implemented for high-throughput screening (HTS) and validated in 3-day variability tests, meeting HTS standards. We initially used this assay to screen the 446-member National Institutes of Health (NIH) Clinical Collection and validated two of the hits in dose-response, by limited structure-activity relationship studies with a small number of analogs, and in an orthogonal, non-optical assay in patient fibroblasts. The data form the basis for a large-scale phenotypic screening effort to discover compounds that stabilize TPI as treatments for this devastating childhood disease.


Subject(s)
Drug Discovery/methods , Enzyme Stability/drug effects , High-Throughput Screening Assays/methods , Small Molecule Libraries , Triose-Phosphate Isomerase/chemistry , Drug Evaluation, Preclinical/methods , Genes, Reporter , HEK293 Cells , Humans , Mutation , Triose-Phosphate Isomerase/antagonists & inhibitors , Triose-Phosphate Isomerase/deficiency , Triose-Phosphate Isomerase/genetics
7.
Molecules ; 26(10)2021 May 14.
Article in English | MEDLINE | ID: mdl-34069161

ABSTRACT

The dimeric dihydropyrimidine dehydrogenase (DPD), metalloenzyme, an adjunct anti-cancer drug target, contains highly specialized 4 × Fe2+4S2-4 clusters per chain. These clusters facilitate the catalysis of the rate-limiting step in the pyrimidine degradation pathway through a harmonized electron transfer cascade that triggers a redox catabolic reaction. In the process, the bulk of the administered 5-fluorouracil (5-FU) cancer drug is inactivated, while a small proportion is activated to nucleic acid antimetabolites. The occurrence of missense mutations in DPD protein within the general population, including those of African descent, has adverse toxicity effects due to altered 5-FU metabolism. Thus, deciphering mutation effects on protein structure and function is vital, especially for precision medicine purposes. We previously proposed combining molecular dynamics (MD) and dynamic residue network (DRN) analysis to decipher the molecular mechanisms of missense mutations in other proteins. However, the presence of Fe2+4S2-4 clusters in DPD poses a challenge for such in silico studies. The existing AMBER force field parameters cannot accurately describe the Fe2+ center coordination exhibited by this enzyme. Therefore, this study aimed to derive AMBER force field parameters for DPD enzyme Fe2+ centers, using the original Seminario method and the collation features Visual Force Field Derivation Toolkit as a supportive approach. All-atom MD simulations were performed to validate the results. Both approaches generated similar force field parameters, which accurately described the human DPD protein Fe2+4S2-4 cluster architecture. This information is crucial and opens new avenues for in silico cancer pharmacogenomics and drug discovery related research on 5-FU drug efficacy and toxicity issues.


Subject(s)
Antineoplastic Agents/pharmacology , Computer Simulation , Dihydrouracil Dehydrogenase (NADP)/metabolism , Fluorouracil/pharmacology , Iron-Sulfur Proteins/metabolism , Neoplasms/drug therapy , Pharmacogenetics , Animals , Antineoplastic Agents/therapeutic use , Enzyme Stability/drug effects , Fluorouracil/therapeutic use , Humans , Molecular Conformation , Molecular Dynamics Simulation , Principal Component Analysis , Protons , Quantum Theory , Structural Homology, Protein , Swine , Thermodynamics
8.
Sci Rep ; 11(1): 12768, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140593

ABSTRACT

As an approach to the exploration of cold-active enzymes, in this study, we isolated a cold-active protease produced by psychrotrophic bacteria from glacial soils of Thajwas Glacier, Himalayas. The isolated strain BO1, identified as Bacillus pumilus, grew well within a temperature range of 4-30 °C. After its qualitative and quantitative screening, the cold-active protease (Apr-BO1) was purified. The Apr-BO1 had a molecular mass of 38 kDa and showed maximum (37.02 U/mg) specific activity at 20 °C, with casein as substrate. It was stable and active between the temperature range of 5-35 °C and pH 6.0-12.0, with an optimum temperature of 20 °C at pH 9.0. The Apr-BO1 had low Km value of 1.0 mg/ml and Vmax 10.0 µmol/ml/min. Moreover, it displayed better tolerance to organic solvents, surfactants, metal ions and reducing agents than most alkaline proteases. The results exhibited that it effectively removed the stains even in a cold wash and could be considered a decent detergent additive. Furthermore, through protein modelling, the structure of this protease was generated from template, subtilisin E of Bacillus subtilis (PDB ID: 3WHI), and different methods checked its quality. For the first time, this study reported the protein sequence for psychrotrophic Apr-BO1 and brought forth its novelty among other cold-active proteases.


Subject(s)
Bacteria/enzymology , Cold Temperature , Ice Cover/microbiology , Peptide Hydrolases/isolation & purification , Soil Microbiology , Amino Acid Sequence , Bacteria/isolation & purification , Caseins/metabolism , Catalytic Domain , Enzyme Inhibitors/pharmacology , Enzyme Stability/drug effects , Hydrogen-Ion Concentration , Hydrolysis , India , Ions , Kinetics , Metals/pharmacology , Models, Molecular , Molecular Weight , Oxidants/pharmacology , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Phylogeny , Protein Domains , Solvents/pharmacology , Substrate Specificity/drug effects , Surface-Active Agents/pharmacology
9.
J Biol Chem ; 297(1): 100841, 2021 07.
Article in English | MEDLINE | ID: mdl-34058201

ABSTRACT

SGNH-type acetyl xylan esterases (AcXEs) play important roles in marine and terrestrial xylan degradation, which are necessary for removing acetyl side groups from xylan. However, only a few cold-adapted AcXEs have been reported, and the underlying mechanisms for their cold adaptation are still unknown because of the lack of structural information. Here, a cold-adapted AcXE, AlAXEase, from the Arctic marine bacterium Arcticibacterium luteifluviistationis SM1504T was characterized. AlAXEase could deacetylate xylooligosaccharides and xylan, which, together with its homologs, indicates a novel SGNH-type carbohydrate esterase family. AlAXEase showed the highest activity at 30 °C and retained over 70% activity at 0 °C but had unusual thermostability with a Tm value of 56 °C. To explain the cold adaption mechanism of AlAXEase, we next solved its crystal structure. AlAXEase has similar noncovalent stabilizing interactions to its mesophilic counterpart at the monomer level and forms stable tetramers in solutions, which may explain its high thermostability. However, a long loop containing the catalytic residues Asp200 and His203 in AlAXEase was found to be flexible because of the reduced stabilizing hydrophobic interactions and increased destabilizing asparagine and lysine residues, leading to a highly flexible active site. Structural and enzyme kinetic analyses combined with molecular dynamics simulations at different temperatures revealed that the flexible catalytic loop contributes to the cold adaptation of AlAXEase by modulating the distance between the catalytic His203 in this loop and the nucleophilic Ser32. This study reveals a new cold adaption strategy adopted by the thermostable AlAXEase, shedding light on the cold adaption mechanisms of AcXEs.


Subject(s)
Acetylesterase/chemistry , Acetylesterase/metabolism , Adaptation, Physiological , Cold Temperature , Acetylesterase/antagonists & inhibitors , Acetylesterase/genetics , Amino Acid Sequence , Bacteria/enzymology , Catalytic Domain , Enzyme Inhibitors/pharmacology , Enzyme Stability/drug effects , Kinetics , Metals/pharmacology , Models, Molecular , Molecular Dynamics Simulation , Mutation/genetics , Phylogeny , Protein Multimerization , Substrate Specificity/drug effects , Temperature
10.
Int J Mol Sci ; 22(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806499

ABSTRACT

Diabetic nephropathy (DN) ranks among the most detrimental long-term effects of diabetes, affecting more than 30% of all patients. Within the diseased kidney, intraglomerular mesangial cells play a key role in facilitating the pro-fibrotic turnover of extracellular matrix components and a progredient glomerular hyperproliferation. These pathological effects are in part caused by an impaired functionality of soluble guanylate cyclase (sGC) and a consequentially reduced synthesis of anti-fibrotic messenger 3',5'-cyclic guanosine monophosphate (cGMP). Bay 58-2667 (cinaciguat) is able to re-activate defective sGC; however, the drug suffers from poor bioavailability and its systemic administration is linked to adverse events such as severe hypotension, which can hamper the therapeutic effect. In this study, cinaciguat was therefore efficiently encapsulated into virus-mimetic nanoparticles (NPs) that are able to specifically target renal mesangial cells and therefore increase the intracellular drug accumulation. NP-assisted drug delivery thereby increased in vitro potency of cinaciguat-induced sGC stabilization and activation, as well as the related downstream signaling 4- to 5-fold. Additionally, administration of drug-loaded NPs provided a considerable suppression of the non-canonical transforming growth factor ß (TGF-ß) signaling pathway and the resulting pro-fibrotic remodeling by 50-100%, making the system a promising tool for a more refined therapy of DN and other related kidney pathologies.


Subject(s)
Benzoates/administration & dosage , Drug Delivery Systems , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , Benzoates/pharmacokinetics , Biomimetic Materials , Cells, Cultured , Cyclic GMP/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Fibrosis , Humans , Mesangial Cells/pathology , Models, Biological , Nanoparticles/administration & dosage , Rats , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
11.
Biochem Biophys Res Commun ; 551: 38-45, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33714758

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) mutant R132H, promoting the oncometabolite D-2-hydroxyglutarate (D2HG), is a driver mutation and an emerging therapeutic target in glioma. This study identified a novel mutant IDH1 inhibitor, WM17, by virtual screening and enzymatic confirmation. It could bind to and increase mutant IDH1 protein's thermostability in both endogenous heterozygous cells and exogenous overexpressed cells. Consequently, WM17 reversed the accumulation of D2HG and histone hypermethylation in IDH1 mutated cells. Finally, we concluded that WM17 significantly inhibited cell migration in IDH1 mutated glioma cells, although it has no apparent effect on cell proliferation. Further studies are guaranteed toward the development of WM17 as a therapeutic agent for IDH1 mutated glioma.


Subject(s)
Glioma/drug therapy , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Mutant Proteins/antagonists & inhibitors , Mutation , Benzeneacetamides/pharmacology , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Stability/drug effects , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Histones/metabolism , Humans , Imidazoles/pharmacology , Methylation/drug effects , Models, Molecular , Molecular Targeted Therapy , Mutant Proteins/genetics , Protein Binding
12.
Molecules ; 26(4)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673063

ABSTRACT

This paper aims to investigate the effects of some salts (NaCl, (NH4)2SO4 and Na2SO4) at pH 5.0, 7.0 and 9.0 on the stability of 13 different immobilized enzymes: five lipases, three proteases, two glycosidases, and one laccase, penicillin G acylase and catalase. The enzymes were immobilized to prevent their aggregation. Lipases were immobilized via interfacial activation on octyl agarose or on glutaraldehyde-amino agarose beads, proteases on glyoxyl agarose or glutaraldehyde-amino agarose beads. The use of high concentrations of salts usually has some effects on enzyme stability, but the intensity and nature of these effects depends on the inactivation pH, nature and concentration of the salt, enzyme and immobilization protocol. The same salt can be a stabilizing or a destabilizing agent for a specific enzyme depending on its concentration, inactivation pH and immobilization protocol. Using lipases, (NH4)2SO4 generally permits the highest stabilities (although this is not a universal rule), but using the other enzymes this salt is in many instances a destabilizing agent. At pH 9.0, it is more likely to find a salt destabilizing effect than at pH 7.0. Results confirm the difficulty of foreseeing the effect of high concentrations of salts in a specific immobilized enzyme.


Subject(s)
Enzyme Stability/drug effects , Enzymes, Immobilized/chemistry , Salts/chemistry , Catalase/chemistry , Enzymes, Immobilized/antagonists & inhibitors , Glycoside Hydrolases/chemistry , Hydrogen-Ion Concentration , Kinetics , Laccase/chemistry , Lipase/chemistry , Organic Chemicals/chemistry , Penicillin Amidase/chemistry , Peptide Hydrolases/chemistry , Salts/pharmacology , Solutions/chemistry , Solutions/pharmacology , Temperature
13.
Blood Cancer J ; 11(3): 61, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737511

ABSTRACT

T-cell and B-cell acute lymphoblastic leukemias (T-ALL, B-ALL) are aggressive hematological malignancies characterized by an accumulation of immature T- or B-cells. Although patient outcomes have improved, novel targeted therapies are needed to reduce the intensity of chemotherapy and improve the prognosis of high-risk patients. Using cell lines, primary cells and patient-derived xenograft (PDX) models, we demonstrate that ALL cells viability is sensitive to NVP-BEP800, an ATP-competitive inhibitor of Heat shock protein 90 (HSP90). Furthermore, we reveal that lymphocyte-specific SRC family kinases (SFK) are important clients of the HSP90 chaperone in ALL. When PDX mice are treated with NVP-BEP800, we found that there is a decrease in ALL progression. Together, these results demonstrate that the chaperoning of SFK by HSP90 is involved in the growth of ALL. These novel findings provide an alternative approach to target SRC kinases and could be used for the development of new treatment strategies for ALL.


Subject(s)
Antineoplastic Agents/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyrimidines/therapeutic use , src-Family Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Stability/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrimidines/pharmacology , Tumor Cells, Cultured , src-Family Kinases/antagonists & inhibitors
14.
PLoS One ; 16(3): e0248878, 2021.
Article in English | MEDLINE | ID: mdl-33740023

ABSTRACT

Garlic is a well-known example of natural self-defence system consisting of an inactive substrate (alliin) and enzyme (alliinase) which, when combined, produce highly antimicrobial allicin. Increase of alliinase stability and its activity are of paramount importance in various applications relying on its use for in-situ synthesis of allicin or its analogues, e.g., pulmonary drug delivery, treatment of superficial injuries, or urease inhibitors in fertilizers. Here, we discuss the effect of temperature, pH, buffers, salts, and additives, i.e. antioxidants, chelating agents, reducing agents and cosolvents, on the stability and the activity of alliinase extracted from garlic. The effects of the storage temperature and relative humidity on the stability of lyophilized alliinase was demonstrated. A combination of the short half-life, high reactivity and non-specificity to particular proteins are reasons most bacteria cannot deal with allicin's mode of action and develop effective defence mechanism, which could be the key to sustainable drug design addressing serious problems with escalating emergence of multidrug-resistant (MDR) bacterial strains.


Subject(s)
Carbon-Sulfur Lyases/metabolism , Chemical Phenomena , Disulfides/metabolism , Garlic/enzymology , Sulfinic Acids/metabolism , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/ultrastructure , Biocatalysis/drug effects , Buffers , Disulfides/chemistry , Enzyme Stability/drug effects , Freeze Drying , Hydrogen-Ion Concentration , Kinetics , Microbial Sensitivity Tests , Microbial Viability/drug effects , Stereoisomerism , Sulfinic Acids/chemistry , Temperature , Time Factors
15.
Sci Rep ; 11(1): 4573, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33633359

ABSTRACT

Thermostability and substrate specificity of proteases are major factors in their industrial applications. rEla is a novel recombinant cysteine protease obtained from a thermophilic bacterium, Cohnella sp.A01 (PTCC No: 1921). Herein, we were interested in recombinant production and characterization of the enzyme and finding the novel features in comparison with other well-studied cysteine proteases. The bioinformatics analysis showed that rEla is allosteric cysteine protease from DJ-1/ThiJ/PfpI superfamily. The enzyme was heterologously expressed and characterized and the recombinant enzyme molecular mass was 19.38 kD which seems to be smaller than most of the cysteine proteases. rEla exhibited acceptable activity in broad pH and temperature ranges. The optimum activity was observed at 50℃ and pH 8 and the enzyme showed remarkable stability by keeping 50% of residual activity after 100 days storage at room temperature. The enzyme Km and Vmax values were 21.93 mM, 8 U/ml, respectively. To the best of our knowledge, in comparison with the other characterized cysteine proteases, rEla is the only reported cysteine protease with collagen specificity. The enzymes activity increases up to 1.4 times in the presence of calcium ion (2 mM) suggesting it as the enzyme's co-factor. When exposed to surfactants including Tween20, Tween80, Triton X-100 and SDS (1% and 4% v/v) the enzyme activity surprisingly increased up to 5 times.


Subject(s)
Bacillales/enzymology , Cysteine Proteases/metabolism , Amino Acid Sequence , Bacillales/drug effects , Bacillales/genetics , Binding Sites , Cysteine Proteases/chemistry , Cysteine Proteases/genetics , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Hydrogen-Ion Concentration , Molecular Docking Simulation , Molecular Dynamics Simulation , Phylogeny , Protein Binding , Protein Conformation , Sequence Analysis, DNA , Structure-Activity Relationship , Temperature
16.
Biomolecules ; 11(1)2021 01 18.
Article in English | MEDLINE | ID: mdl-33477596

ABSTRACT

A novel temperature stable alkaline protease yielding bacteria was isolated from the soils of Dachigam National Park, which is known to be inhabited by a wide variety of endemic plant and animal species of Western Himalaya. This high-potential protease producing isolate was characterized and identified as Bacillus amyloliquefaciens strain HM48 by morphological, Gram's staining and biochemical techniques followed by molecular characterization using 16S rRNA approach. The extracellular protease of B. amyloliquefaciens HM48 was purified by precipitating with ammonium sulfate (80%), followed by dialysis and Gel filtration chromatography increasing its purity by 5.8-fold. The SDS-PAGE analysis of the purified enzyme confirmed a molecular weight of about ≈25 kDa. The enzyme displayed exceptional activity in a broad temperature range (10-90 °C) at pH 8.0, retaining its maximum at 70 °C, being the highest reported for this proteolytic Bacillus sp., with KM and Vmax of 11.71 mg/mL and 357.14 µmol/mL/min, respectively. The enzyme exhibited remarkable activity and stability against various metal ions, surfactants, oxidizing agent (H2O2), organic solvents and displayed outstanding compatibility with widely used detergents. This protease showed effective wash performance by exemplifying complete blood and egg-yolk stains removal at 70 °C and efficiently disintegrated chicken feathers making it of vital importance for laundry purpose and waste management. For functional analysis, protease gene amplification of strain HM48 yielded a nucleotide sequence of about 700 bp, which, when checked against the available sequences in NCBI, displayed similarity with subtilisin-like serine protease of B. amyloliquefaciens. The structure of this protease and its highest-priority substrate ß-casein was generated through protein modeling. These protein models were validated through futuristic algorithms following which protein-protein (protease from HM48 and ß-casein) docking was performed. The interaction profile of these proteins in the docked state with each other was also generated, shedding light on their finer details. Such attributes make this thermally stable protease novel and suitable for high-temperature industrial and environmental applications.


Subject(s)
Bacillus amyloliquefaciens/enzymology , Hot Temperature , Peptide Hydrolases/metabolism , Soil Microbiology , Animals , Bacillus amyloliquefaciens/cytology , Bacillus amyloliquefaciens/genetics , Bacillus amyloliquefaciens/isolation & purification , Caseins/metabolism , Chickens , Edetic Acid/pharmacology , Enzyme Stability/drug effects , Feathers , Geography , Hydrogen-Ion Concentration , India , Ions , Kinetics , Metals/pharmacology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Weight , Oxidants/pharmacology , Peptide Hydrolases/genetics , Proteolysis/drug effects , RNA, Ribosomal, 16S/genetics , Reproducibility of Results , Solvents , Substrate Specificity/drug effects , Surface-Active Agents/pharmacology
17.
Biomolecules ; 11(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33466452

ABSTRACT

The gene coding for a novel cold-active esterase PMGL3 was previously obtained from a Siberian permafrost metagenomic DNA library and expressed in Escherichia coli. We elucidated the 3D structure of the enzyme which belongs to the hormone-sensitive lipase (HSL) family. Similar to other bacterial HSLs, PMGL3 shares a canonical α/ß hydrolase fold and is presumably a dimer in solution but, in addition to the dimer, it forms a tetrameric structure in a crystal and upon prolonged incubation at 4 °C. Detailed analysis demonstrated that the crystal tetramer of PMGL3 has a unique architecture compared to other known tetramers of the bacterial HSLs. To study the role of the specific residues comprising the tetramerization interface of PMGL3, several mutant variants were constructed. Size exclusion chromatography (SEC) analysis of D7N, E47Q, and K67A mutants demonstrated that they still contained a portion of tetrameric form after heat treatment, although its amount was significantly lower in D7N and K67A compared to the wild type. Moreover, the D7N and K67A mutants demonstrated a 40 and 60% increase in the half-life at 40 °C in comparison with the wild type protein. Km values of these mutants were similar to that of the wt PMGL3. However, the catalytic constants of the E47Q and K67A mutants were reduced by ~40%.


Subject(s)
Cold Temperature , Esterases/chemistry , Protein Multimerization , Amino Acid Sequence , Catalytic Domain , Detergents/pharmacology , Enzyme Stability/drug effects , Esterases/metabolism , Ions , Metals/pharmacology , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Sodium Chloride/pharmacology , Solvents , Structural Homology, Protein
18.
Int J Biol Macromol ; 166: 557-566, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33186653

ABSTRACT

In this study, serine alkaline protease from halotolerant alkaliphilic Salipaludibacillus agaradhaerens strain AK-R was purified and immobilized onto double mesoporous core-shell silica (DMCSS) nanospheres. Covalent immobilization of AK-R protease onto activated DMCSS-NH2 nanospheres was more efficient than physical adsorption and was applied in further studies. DMCSS-NH2 nanospheres showed high loading capacity of 103.8 µg protein/mg nanospheres. Relative to free AK-R protease, the immobilized enzyme exhibited shifts in the optimal temperature and pH from 60 to 65 °C and pH 10.0 to 10.5, respectively. While the soluble enzyme retained 47.2% and 9.1% of its activity after treatment for 1 h at 50 and 60 °C, the immobilized protease maintained 87.7% and 48.3%, respectively. After treatment for 2 h at pH 5 and 13, the immobilized protease maintained 73.6% and 53.4% of its activity, whereas the soluble enzyme retained 32.9% and 1.4%, respectively. Furthermore, the immobilized AK-R protease showed significant improvement of enzyme stability in high concentration of NaCl, organic solvents, surfactants, and commercial detergents. In addition, the immobilized protease exhibited a very good operational stability, retaining 79.8% of its activity after ten cycles. The results clearly suggest that the developed immobilized protease system is a promising nanobiocatalyst for various protease applications.


Subject(s)
Bacillaceae/enzymology , Bacterial Proteins/metabolism , Endopeptidases/metabolism , Enzymes, Immobilized/metabolism , Nanospheres/chemistry , Biocatalysis/drug effects , Detergents/pharmacology , Enzyme Stability/drug effects , Hydrogen-Ion Concentration , Nanospheres/ultrastructure , Oxidants/pharmacology , Porosity , Salinity , Silicon Dioxide/chemistry , Solvents/chemistry , Surface-Active Agents/pharmacology , Temperature
19.
Nature ; 588(7839): 712-716, 2020 12.
Article in English | MEDLINE | ID: mdl-33328633

ABSTRACT

Altered expression of mitochondrial DNA (mtDNA) occurs in ageing and a range of human pathologies (for example, inborn errors of metabolism, neurodegeneration and cancer). Here we describe first-in-class specific inhibitors of mitochondrial transcription (IMTs) that target the human mitochondrial RNA polymerase (POLRMT), which is essential for biogenesis of the oxidative phosphorylation (OXPHOS) system1-6. The IMTs efficiently impair mtDNA transcription in a reconstituted recombinant system and cause a dose-dependent inhibition of mtDNA expression and OXPHOS in cell lines. To verify the cellular target, we performed exome sequencing of mutagenized cells and identified a cluster of amino acid substitutions in POLRMT that cause resistance to IMTs. We obtained a cryo-electron microscopy (cryo-EM) structure of POLRMT bound to an IMT, which further defined the allosteric binding site near the active centre cleft of POLRMT. The growth of cancer cells and the persistence of therapy-resistant cancer stem cells has previously been reported to depend on OXPHOS7-17, and we therefore investigated whether IMTs have anti-tumour effects. Four weeks of oral treatment with an IMT is well-tolerated in mice and does not cause OXPHOS dysfunction or toxicity in normal tissues, despite inducing a strong anti-tumour response in xenografts of human cancer cells. In summary, IMTs provide a potent and specific chemical biology tool to study the role of mtDNA expression in physiology and disease.


Subject(s)
Mitochondria/drug effects , Mitochondria/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Transcription, Genetic/drug effects , Animals , Cell Proliferation/drug effects , Cryoelectron Microscopy , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , DNA-Directed RNA Polymerases/metabolism , Down-Regulation/drug effects , Enzyme Stability/drug effects , Female , Gene Expression Regulation/drug effects , Genes, Mitochondrial/drug effects , Humans , Male , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Substrate Specificity/drug effects , Xenograft Model Antitumor Assays
20.
Mol Oncol ; 14(12): 3030-3047, 2020 12.
Article in English | MEDLINE | ID: mdl-33025742

ABSTRACT

Metabolic rewiring is a mechanism of adaptation to unfavorable environmental conditions and tumor progression. TRAP1 is an HSP90 molecular chaperone upregulated in human colorectal carcinomas (CRCs) and responsible for downregulation of oxidative phosphorylation (OXPHOS) and adaptation to metabolic stress. The mechanism by which TRAP1 regulates glycolytic metabolism and the relevance of this regulation in resistance to EGFR inhibitors were investigated in patient-derived CRC spheres, human CRC cells, samples, and patients. A linear correlation was observed between TRAP1 levels and 18 F-fluoro-2-deoxy-glucose (18 F-FDG) uptake upon PET scan or GLUT1 expression in human CRCs. Consistently, TRAP1 enhances GLUT1 expression, glucose uptake, and lactate production and downregulates OXPHOS in CRC patient-derived spheroids and cell lines. Mechanistically, TRAP1 maximizes lactate production to balance low OXPHOS through the regulation of the glycolytic enzyme phosphofructokinase-1 (PFK1); this depends on the interaction between TRAP1 and PFK1, which favors PFK1 glycolytic activity and prevents its ubiquitination/degradation. By contrast, TRAP1/PFK1 interaction is lost in conditions of enhanced OXPHOS, which results in loss of TRAP1 regulation of PFK1 activity and lactate production. Notably, TRAP1 regulation of glycolysis is involved in resistance of RAS-wild-type CRCs to EGFR monoclonals. Indeed, either TRAP1 upregulation or high glycolytic metabolism impairs cetuximab activity in vitro, whereas TRAP1 targeting and/or inhibition of glycolytic pathway enhances cell response to cetuximab. Finally, a linear correlation between 18 F-FDG PET uptake and poor response to cetuximab in first-line therapy in human metastatic CRCs was observed. These results suggest that TRAP1 is a key determinant of CRC metabolic rewiring and favors resistance to EGFR inhibitors through regulation of glycolytic metabolism.


Subject(s)
Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , HSP90 Heat-Shock Proteins/metabolism , Phosphofructokinase-1/metabolism , Protein Kinase Inhibitors/pharmacology , Warburg Effect, Oncologic , Cell Line, Tumor , Cell Respiration/drug effects , Cetuximab/pharmacology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Stability/drug effects , ErbB Receptors/metabolism , Fluorodeoxyglucose F18/metabolism , Glucose Transporter Type 1/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Phenotype , Protein Binding/drug effects , Proto-Oncogene Proteins B-raf/metabolism , Warburg Effect, Oncologic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...