Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.329
Filter
1.
J Med Chem ; 65(3): 2329-2341, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35072486

ABSTRACT

In recent years, the function of pharmacological chaperones as a "thermodynamic stabilizer" has been attracting attention in combination therapy. The coadministration of a pharmacological chaperone and recombinant human acid α-glucosidase (rhGAA) leads to improved stability and maturation by binding to the folded state of the rhGAA and thereby promotes enzyme delivery. This study provides the first example of a strategy to design a high-affinity ligand toward lysosomal acid α-glucosidase (GAA) focusing on alkyl branches on 1-deoxynojirimycin (DNJ); 5-C-heptyl-DNJ produced a nanomolar affinity for GAA with a Ki value of 0.0047 µM, which is 13-fold more potent than DNJ. The protein thermal shift assay revealed that 10 µM 5-C-heptyl-DNJ increased the midpoint of the protein denaturation temperature (Tm) to 73.6 °C from 58.6 °C in the absence of the ligand, significantly improving the thermal stability of rhGAA. Furthermore, 5-C-heptyl-DNJ dose dependency increased intracellular GAA activities in Pompe patient's fibroblasts with the M519V mutation. The introduction of C5 alkyl branches on DNJ provides a new molecular strategy for pharmacological chaperone therapy for Pompe disease, which may lead to the development of higher-affinity and practically useful chaperones.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , Enzyme Inhibitors/pharmacology , alpha-Glucosidases/metabolism , Alkylation , Enzyme Inhibitors/chemical synthesis , Fibroblasts/metabolism , Glycogen Storage Disease Type II , Humans , Molecular Dynamics Simulation , Molecular Structure , Mutation , Protein Conformation/drug effects , Protein Stability/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , alpha-Glucosidases/drug effects , alpha-Glucosidases/genetics
2.
STAR Protoc ; 2(4): 100896, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34723213

ABSTRACT

Identification of selective deubiquitinase (DUB) inhibitors is critical for probe development to further understand and explore DUB biological function. Here, we detail the optimization and deployment of an in vitro fluorogenic ubiquitin-rhodamine assay to conduct high-throughput screening of a small molecule library against a panel of DUBs. In screening the compound library against multiple DUBs in parallel, we describe an approach for identifying selective DUB inhibitors and provide a roadmap for enabling selective DUB inhibitor discovery. For complete details on the use and execution of this protocol, please refer to Varca et al. (2021).


Subject(s)
Deubiquitinating Enzymes , Enzyme Inhibitors , High-Throughput Screening Assays/methods , Rhodamines/metabolism , Ubiquitin/metabolism , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/metabolism , Enzyme Assays , Escherichia coli/genetics , Fluorescent Dyes/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Small Molecule Libraries
3.
Int J Mol Sci ; 22(13)2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34281290

ABSTRACT

Plasmodium falciparum's resistance to available antimalarial drugs highlights the need for the development of novel drugs. Pyrimidine de novo biosynthesis is a validated drug target for the prevention and treatment of malaria infection. P. falciparum dihydroorotate dehydrogenase (PfDHODH) catalyzes the oxidation of dihydroorotate to orotate and utilize ubiquinone as an electron acceptor in the fourth step of pyrimidine de novo biosynthesis. PfDHODH is targeted by the inhibitor DSM265, which binds to a hydrophobic pocket located at the N-terminus where ubiquinone binds, which is known to be structurally divergent from the mammalian orthologue. In this study, we screened 40,400 compounds from the Kyoto University chemical library against recombinant PfDHODH. These studies led to the identification of 3,4-dihydro-2H,6H-pyrimido[1,2-c][1,3]benzothiazin-6-imine and its derivatives as a new class of PfDHODH inhibitor. Moreover, the hit compounds identified in this study are selective for PfDHODH without inhibition of the human enzymes. Finally, this new scaffold of PfDHODH inhibitors showed growth inhibition activity against P. falciparum 3D7 with low toxicity to three human cell lines, providing a new starting point for antimalarial drug development.


Subject(s)
Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Imines/pharmacology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Antimalarials/chemistry , Antimalarials/toxicity , Cell Line , Dihydroorotate Dehydrogenase , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Humans , Imines/chemistry , Imines/toxicity , Plasmodium falciparum/growth & development , Pyrimidines/chemistry , Pyrimidines/toxicity , Recombinant Proteins/drug effects , Structure-Activity Relationship , Triazoles/pharmacology
4.
Biosci Rep ; 41(5)2021 05 28.
Article in English | MEDLINE | ID: mdl-33950219

ABSTRACT

Selective modulation of retinaldehyde dehydrogenases (RALDHs)-the main aldehyde dehydrogenase (ALDH) enzymes converting retinal into retinoic acid (RA), is very important not only in the RA signaling pathway but also for the potential regulatory effects on RALDH isozyme-specific processes and RALDH-related cancers. However, very few selective modulators for RALDHs have been identified, partly due to variable overexpression protocols of RALDHs and insensitive activity assay that needs to be addressed. In the present study, deletion of the N-terminal disordered regions is found to enable simple preparation of all RALDHs and their closest paralog ALDH2 using a single protocol. Fluorescence-based activity assay was employed for enzymatic activity investigation and screening for RALDH-specific modulators from extracts of various spices and herbs that are well-known for containing many phyto-derived anti-cancer constituents. Under the established conditions, spice and herb extracts exhibited differential regulatory effects on RALDHs/ALDH2 with several extracts showing potential selective inhibition of the activity of RALDHs. In addition, the presence of magnesium ions was shown to significantly increase the activity for the natural substrate retinal of RALDH3 but not the others, while His-tag cleavage considerably increased the activity of ALDH2 for the non-specific substrate retinal. Altogether we propose a readily reproducible workflow to find selective modulators for RALDHs and suggest potential sources of selective modulators from spices and herbs.


Subject(s)
Enzyme Assays/methods , Plant Extracts/pharmacology , Retinal Dehydrogenase/metabolism , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli , Humans , Plant Extracts/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinal Dehydrogenase/chemistry , Retinal Dehydrogenase/drug effects , Retinal Dehydrogenase/genetics , Sequence Homology
5.
Protein Expr Purif ; 185: 105906, 2021 09.
Article in English | MEDLINE | ID: mdl-33991675

ABSTRACT

Nanobodies are single-domain antibody constructs derived from the variable regions of heavy chain only (VHH) camelid IgGs. Their small size and single gene format make them amenable to various molecular biology applications that require a protein affinity-based approach. These features, in addition to their high solubility, allows their periplasmic expression, extraction and purification in E. coli systems with relative ease, using standardized protocols. However, some Nanobodies are recalcitrant to periplasmic expression, extraction and purification within E. coli systems. To improve their expression would require either a change in the expression host, vector or an increased scale of expression, all of which entail an increase in the complexity of their expression, and production cost. However, as shown here, specific changes in the existing standard E. coli culture protocol, aimed at reducing breakdown of selective antibiotic pressure, increasing the initial culture inoculum and improving transport to the periplasmic space, rescued the expression of several such refractory Nanobodies. The periplasmic extraction protocol was also changed to ensure efficient osmolysis, prevent both protein degradation and prevent downstream chelation of Ni2+ ions during IMAC purification. Adoption of this protocol will lead to an improvement of the expression of Nanobodies in general, and specifically, those that are recalcitrant.


Subject(s)
Escherichia coli/metabolism , Periplasm/metabolism , Recombinant Proteins/isolation & purification , Single-Domain Antibodies/biosynthesis , Amino Acid Sequence , Cloning, Molecular , Culture Media/chemistry , Culture Media/pharmacology , Escherichia coli/genetics , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Osmotic Pressure , Periplasm/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Single-Domain Antibodies/genetics , Single-Domain Antibodies/isolation & purification
6.
Biochem Biophys Res Commun ; 545: 125-131, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33548625

ABSTRACT

Bromodomain and PHD finger containing transcription factor (BPTF) is a multidomain protein that regulates the transcription of chromatin and is related to many cancers. Herein, we report the screening-based discovery of Cpd1, a compound with micromolar affinity to the BPTF bromodomain. Through structure-guided optimization, we synthesized a variety of new inhibitors. Among these compounds, Cpd8 and Cpd10 were highly potent and selective inhibitors, with KD values of 428 nM and 655 nM in ITC assays, respectively. The high activity was explained by the cocrystal structure of Cpd8 in complex with the BPTF bromodomain protein. Cpd8 and Cpd10 were able to stabilize the BPTF bromodomain protein in cells in a cellular thermal shift assay (CETSA). Cpd8 downregulated c-MYC expression in A549 cells. All experiments prove that these two compounds are potential BPTF inhibitors.


Subject(s)
Nerve Tissue Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , A549 Cells , Antigens, Nuclear/chemistry , Antigens, Nuclear/genetics , Calorimetry , Crystallography, X-Ray , Drug Design , Drug Discovery , Drug Evaluation, Preclinical , Fluorometry , Gene Expression Regulation/drug effects , Genes, myc , HEK293 Cells , Humans , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Domains , Protein Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/genetics
7.
Biochem J ; 478(3): 647-668, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33459338

ABSTRACT

Pseudomonas aeruginosa has recently been highlighted by the World Health Organisation (WHO) as a major threat with high priority for the development of new therapies. In severe P. aeruginosa infections, the phospholipase activity of the type 3 secretion system toxin, ExoU, induces lysis of target host cells and results in the poorest clinical outcomes. We have developed an integrated pipeline to evaluate small molecule inhibitors of ExoU in vitro and in cultured cell models, including a disease-relevant corneal epithelial (HCE-T) scratch and infection model using florescence microscopy and cell viability assays. Compounds Pseudolipasin A, compound A and compound B were effective in vitro inhibitors of ExoU and mitigated P. aeruginosa ExoU-dependent cytotoxicity after infection of HCE-T cells at concentrations as low as 0.5 µM. Addition of the antimicrobial moxifloxacin controlled bacterial load, allowing these assays to be extended from 6 h to 24 h. P. aeruginosa remained cytotoxic to HCE-T cells with moxifloxacin, present at the minimal inhibitory concentration for 24 h, but, when used in combination with either Pseudolipasin A, compound A or compound B, a greater amount of viable cells and scratch healing were observed. Thus, our pipeline provides evidence that ExoU inhibitors could be used in combination with certain antimicrobials as a novel means to treat infections due to ExoU producing P. aeruginosa, as well as the means to identify more potent ExoU inhibitors for future therapeutics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Evaluation, Preclinical/methods , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Cells, Cultured , Drug Synergism , Epithelial Cells , Epithelium, Corneal/cytology , HeLa Cells , High-Throughput Screening Assays , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Moxifloxacin/pharmacology , Protein Conformation , Recombinant Proteins/drug effects , Transfection
8.
Nucleic Acids Res ; 49(2): 700-712, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33410883

ABSTRACT

Aptamers are single-stranded oligonucleotides that bind to a specific target with high affinity, and are widely applied in biomedical diagnostics and drug development. However, the use of aptamers has largely been limited to simple binders or inhibitors that interfere with the function of a target protein. Here, we show that an aptamer can also act as a positive allosteric modulator that enhances the activation of a receptor by stabilizing the binding of a ligand to that receptor. We developed an aptamer, named IR-A43, which binds to the insulin receptor, and confirmed that IR-A43 and insulin bind to the insulin receptor with mutual positive cooperativity. IR-A43 alone is inactive, but, in the presence of insulin, it potentiates autophosphorylation and downstream signaling of the insulin receptor. By using the species-specific activity of IR-A43 at the human insulin receptor, we demonstrate that residue Q272 in the cysteine-rich domain is directly involved in the insulin-enhancing activity of IR-A43. Therefore, we propose that the region containing residue Q272 is a hotspot that can be used to enhance insulin receptor activation. Moreover, our study implies that aptamers are promising reagents for the development of allosteric modulators that discriminate a specific conformation of a target receptor.


Subject(s)
Antigens, CD/drug effects , Aptamers, Nucleotide/pharmacology , Receptor, Insulin/drug effects , Allosteric Regulation , Animals , Antigens, CD/chemistry , Antigens, CD/metabolism , Cells, Cultured , Cricetinae , Glutamine/chemistry , Humans , Insulin/metabolism , Mice , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Protein Processing, Post-Translational , Rats , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , SELEX Aptamer Technique , Stimulation, Chemical
9.
Biochimie ; 180: 43-53, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33122104

ABSTRACT

Entamoeba histolytica is an intestinal protozoan parasite of humans and is endemic in developing countries. E. histolytica has two low molecular weight protein tyrosine phosphatase (LMW-PTP) genes, EhLMW-PTP1 and EhLMW-PTP2, which are expressed in cultured trophozoites, clinical isolates, and cysts. The amino acid sequences of proteins EhLMW-PTP1 and EhLMW-PTP2 showed only one amino acid difference between them at position A85V, respectively. Both genes are expressed in cultured trophozoites, mainly EhLMW-PTP2, and in trophozoites recovered from amoebic liver abscess, the expression of EhLMW-PTP1 is downregulated. We cloned the two genes and purified the corresponding recombinant (rEhLMW-PTPs) proteins. Antibodies anti-rEhLMW-PTP2 showed that during red blood cells uptake by E. histolytica, the EhLMW-PTPs were found in the phagocytic cups based on analysis of fluorescence signals. On the other hand, rEhLMW-PTPs showed an optimum phosphatase activity at pH 6.0 with p-nitrophenyl phosphate as the substrate. They dephosphorylate phosphotyrosine and 3-O-methylfluorescein phosphate, but not phosphoserine or phosphothreonine, and the enzymatic activity is inhibited by orthovanadate. rEhLMW-PTP1 and rEhLMW-PTP2 exhibited optimum temperatures of activities at 60 °C and 58 °C, respectively, with high thermal stability at 50 °C. Also, the rEhLMW-PTPs showed high specific activities and specific km value with pNPP or OMFP as the substrates at the physiological temperature (37 °C).


Subject(s)
Entamoeba histolytica/enzymology , Liver Abscess, Amebic/enzymology , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Animals , Chelating Agents/pharmacology , Cricetinae , Entamoeba histolytica/genetics , Enzyme Inhibitors/pharmacology , Enzyme Stability , Erythrocytes/parasitology , Female , Humans , Hydrogen-Ion Concentration , Liver Abscess, Amebic/genetics , Mice, Inbred BALB C , Molecular Weight , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Temperature , Trophozoites/cytology , Trophozoites/enzymology , Trophozoites/genetics
10.
Molecules ; 25(24)2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33316879

ABSTRACT

Previously, a hypothetical protein (HP) termed Bleg1_2437 (currently named Bleg1_2478) from Bacillus lehensis G1 was discovered to be an evolutionary divergent B3 subclass metallo-ß-lactamase (MBL). Due to the scarcity of clinical inhibitors for B3 MBLs and the divergent nature of Bleg1_2478, this study aimed to design and characterise peptides as inhibitors against Bleg1_2478. Through in silico docking, RSWPWH and SSWWDR peptides with comparable binding energy to ampicillin were obtained. In vitro assay results showed RSWPWH and SSWWDR inhibited the activity of Bleg1_2478 by 50% at concentrations as low as 0.90 µM and 0.50 µM, respectively. At 10 µM of RSWPWH and 20 µM of SSWWDR, the activity of Bleg1_2478 was almost completely inhibited. Isothermal titration calorimetry (ITC) analyses showed slightly improved binding properties of the peptides compared to ampicillin. Docked peptide-protein complexes revealed that RSWPWH bound near the vicinity of the Bleg1_2478 active site while SSWWDR bound at the center of the active site itself. We postulate that the peptides caused the inhibition of Bleg1_2478 by reducing or blocking the accessibility of its active site from ampicillin, thus hampering its catalytic function.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/chemical synthesis , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/chemical synthesis , beta-Lactamases/drug effects , Amino Acid Sequence , Ampicillin/chemistry , Ampicillin/pharmacology , Bacillus/enzymology , Bacillus/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Chemical Phenomena , Drug Design , Evolution, Molecular , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Oligopeptides/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Thermodynamics , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/chemistry , beta-Lactamases/genetics
11.
Biochem Biophys Res Commun ; 533(4): 952-957, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33008592

ABSTRACT

Quercetin is a natural flavonoid which has been reported to be analgesic in different animal models of pain. However, the mechanism underlying the pain-relieving effects is still unclear. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play critical roles in controlling pacemaker activity in cardiac and nervous systems, making the channel a new target for therapeutic exploration. In this study, we explored a series of flavonoids for their modulation on HCN channels. Among all tested flavonoids, quercetin was the most potent inhibitor for HCN channels with an IC50 value of 27.32 ± 1.19 µM for HCN2. Furthermore, quercetin prominently left shifted the voltage-dependent activation curves of HCN channels and decelerated deactivation process. The results presented herein firstly characterize quercetin as a novel and potent inhibitor for HCN channels, which represents a novel structure for future drug design of HCN channel inhibitors.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Quercetin/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Drug Evaluation, Preclinical , Electrophysiological Phenomena , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonols/chemistry , Flavonols/pharmacology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels/metabolism , Quercetin/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship
12.
PLoS One ; 15(9): e0238999, 2020.
Article in English | MEDLINE | ID: mdl-32915889

ABSTRACT

Aberrant expression of the transcription factor ERG is a key driving event in approximately one-half of all of prostate cancers. Lacking an enzymatic pocket and mainly disordered, the structure of ERG is difficult to exploit for therapeutic design. We recently identified EWS as a specific interacting partner of ERG that is required for oncogenic function. In this study, we aimed to target this specific protein-protein interaction with small molecules. A high-throughput screening (HTS) strategy was implemented to identify potential protein-protein interaction inhibitors. Secondary assays verified the function of several hit compounds, and one lead compound inhibited ERG-mediated phenotypes in prostate cells. This is the first study aimed at targeting the ERG-EWS protein-protein interaction for the development of a small molecule-based prostate cancer therapy.


Subject(s)
High-Throughput Screening Assays/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Feasibility Studies , Humans , Male , Prostatic Neoplasms/genetics , Protein Interaction Domains and Motifs/drug effects , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Small Molecule Libraries , Transcriptional Regulator ERG/antagonists & inhibitors , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
13.
J Cell Mol Med ; 24(18): 10693-10704, 2020 09.
Article in English | MEDLINE | ID: mdl-32725958

ABSTRACT

Endometriosis is a benign gynaecological disease appearing with pelvic pain, rising dysmenorrhoea and infertility seriously impacting on 10% of reproductive-age females. This research attempts to demonstrate the function and molecular mechanism of RhoA/ROCK pathway on epithelial-mesenchymal transition (EMT) and proliferation in endometriosis. The expression of Rho family was abnormally changed in endometriotic lesions; in particular, RhoA and ROCK1/2 were significantly elevated. Overexpression of RhoA in human eutopic endometrial epithelial cells (eutopic EECs) enhanced the cell mobility, epithelial-mesenchymal transition (EMT) and proliferation, and RhoA knockdown exhibited the opposite function. Oestrogen up-regulated the RhoA activity and expression of RhoA and ROCK1/2. RhoA overexpression reinforced the effect of oestrogen on promoting EMT and proliferation, and RhoA knockdown impaired the effect of oestrogen. oestrogen receptor α (ERα) was involved with the regulation of oestrogen on EMT and proliferation and up-regulated RhoA activity and expression of RhoA and ROCK1/2. The function of ERα was modulated by the change in RhoA expression. Furthermore, phosphorylated ERK that was enhanced by oestrogen and ERα promoted the protein expression of RhoA/ROCK pathway. Endometriosis mouse model revealed that oestrogen enhanced the size and weight of endometriotic lesions. The expression of RhoA and phosphorylated ERK in mouse endometriotic lesions was significantly elevated by oestrogen. We conclude that abnormal activated RhoA/ROCK pathway in endometriosis is responsible for the function of oestrogen/ERα/ERK signalling, which promoted EMT and proliferation and resulted in the development of endometriosis.


Subject(s)
Endometriosis/pathology , Endometrium/pathology , Epithelial-Mesenchymal Transition/physiology , Estrogens/physiology , Signal Transduction/physiology , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/physiology , Adult , Animals , Cells, Cultured , Disease Models, Animal , Endometriosis/surgery , Endometrium/drug effects , Endometrium/transplantation , Epithelial-Mesenchymal Transition/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Female , Gene Expression Regulation , Humans , Mice , Mice, Inbred BALB C , Ovarian Cysts/etiology , Ovarian Cysts/surgery , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Signal Transduction/drug effects , rho-Associated Kinases/biosynthesis , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/biosynthesis , rhoA GTP-Binding Protein/genetics
14.
Sci Rep ; 10(1): 11058, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32632152

ABSTRACT

An actinomycete strain CSR-4 was isolated from the rhizosphere soil of Zingiber montanum. Taxonomic characterization revealed strain CSR-4 was a member of the genus Microbispora. Whole-genome sequence analysis exhibited the highest average nucleotide identity (ANI) value (95.34%) and digital DNA-DNA hybridization (DDH) value (74.7%) between strain CSR-4 and the closest relative M. hainanensis DSM 45428T, which was in line with the assignment to same species. In addition, a new diterpene compound, 2α-hydroxy-8(14), 15-pimaradien-17, 18-dioic acid, and nine known compounds were isolated from the ethyl acetate crude extract of fermentation broth. Interestingly, a new diterpene displayed the suppressive effect on the recombinant human acetylcholinesterase (rhAChE) enzymes (IC50 96.87 ± 2.31 µg/ml). In silico studies based on molecular docking and molecular dynamics (MD) simulations were performed to predict a binding mode of the new compound into the binding pocket of the rhAChE enzyme and revealed that some amino acids in the peripheral anions site (PAS), anionic subsite, oxyanion site and catalytic active site (CAS) of the rhAChE have interacted with the compound. Therefore, our new compound could be proposed as a potential active human AChE inhibitor. Moreover, the new compound can protect significantly the neuron cells (% neuron viability = 88.56 ± 5.19%) from oxidative stress induced by serum deprivation method at 1 ng/ml without both neurotoxicities on murine P19-derived neuron cells and cytotoxicity against Vero cells.


Subject(s)
Actinobacteria/chemistry , Cholinesterase Inhibitors/pharmacology , Diterpenes/pharmacology , Acetylcholinesterase/chemistry , Acetylcholinesterase/drug effects , Actinobacteria/classification , Actinobacteria/genetics , Animals , Antioxidants/chemistry , Antioxidants/isolation & purification , Antioxidants/pharmacology , Catalytic Domain , Chlorocebus aethiops , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/isolation & purification , Computer Simulation , Diterpenes/chemistry , Diterpenes/isolation & purification , Humans , In Vitro Techniques , Mice , Molecular Dynamics Simulation , Molecular Structure , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacology , Phylogeny , RNA, Ribosomal, 16S/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Vero Cells
15.
Methods Mol Biol ; 2141: 755-775, 2020.
Article in English | MEDLINE | ID: mdl-32696388

ABSTRACT

Tau is an intrinsically disordered protein implicated in the pathogenesis of Alzheimer's disease and other neurodegenerative disorders. Here we describe the application of single-molecule Förster resonance energy transfer (smFRET) for the characterization of the interactions between tau and polyphosphate, an intracellular polymer that accelerates tau aggregation. We describe the design of tau constructs, purification and fluorescent labeling of tau, and details of acquisition and analysis of smFRET data. The protocols provided here outline an approach that may be applied to the study of other intrinsically disordered proteins and their binding partners.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Intrinsically Disordered Proteins/chemistry , Polyphosphates/pharmacology , Protein Aggregates , Single Molecule Imaging/methods , tau Proteins/drug effects , Alzheimer Disease/metabolism , Calibration , Cell Line , Cloning, Molecular/methods , Cysteine/chemistry , Fluorescence Resonance Energy Transfer/instrumentation , Fluorescent Dyes , Humans , Mutagenesis, Site-Directed , Organic Chemicals , Protein Domains , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Single Molecule Imaging/instrumentation , Spectrometry, Fluorescence/methods , tau Proteins/genetics
16.
Bioorg Chem ; 101: 103999, 2020 08.
Article in English | MEDLINE | ID: mdl-32563966

ABSTRACT

In this study, we have discovered small druglike molecules as selective inhibitors of human tissue-nonspecific alkaline phosphatase (h-TNAP), an enzyme critical for the regulation of extracellular matrix calcification. The upregulation of h-TNAP is associated with various pathologies particularly the vascular calcification (VC). Selective inhibition of h-TNAP over h-NPP1 may serve as a useful therapeutic strategy against vascular calcification. A series of novel triazolyl pyrazole derivatives (10a-y) in which thiol bearing triazole moiety as the zinc binding functional group was introduced to a pyrazole based pharmacophore was synthesized and evaluated as potent and selective inhibitors of h-TNAP over h-NPP1. The biological screening against h-TNAP, h-IAP, h-NPP1 and h-NPP3 showed that many of the synthesized compounds are selective inhibitors of TNAP. Particularly, the compounds 10a-h, 10j, 10m-q, 10u, 10w and 10x displayed high potency and complete selectivity towards h-TNAP over h-NPP1. Compound 10q emerged as a highly potent inhibitor (IC50 = 0.16 µM or 160 nM) against h-TNAP with 127-fold increased inhibition compared to levamisole. On the other hand, compound 10e was found to be most selective inhibitor against the tested APs and NPPs (IC50 = 1.59 ± 0.36 µM). Binding sites architecture analysis, molecular-docking and molecular dynamics simulations (MDS), revealed the basis for h-TNAP and h-IAP ligand selectivity as well as selectivity towards h-TNAP over h-NPP1. These newly discovered inhibitors are believed to represent valuable lead structures to further streamline the generation of candidate compounds to target VC.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Vascular Calcification/prevention & control , Computational Chemistry , Enzyme Inhibitors/chemistry , Humans , Ligands , Molecular Dynamics Simulation , Recombinant Proteins/drug effects , Structure-Activity Relationship , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology
17.
Mol Cancer Ther ; 19(8): 1589-1597, 2020 08.
Article in English | MEDLINE | ID: mdl-32430490

ABSTRACT

The camptothecin derivatives topoisomerase I (TOP1) inhibitors, irinotecan and topotecan, are FDA approved for the treatment of colorectal, ovarian, lung and breast cancers. Because of the chemical instability of camptothecins, short plasma half-life, drug efflux by the multidrug-resistance ABC transporters, and the severe diarrhea produced by irinotecan, indenoisoquinoline TOP1 inhibitors (LMP400, LMP776, and LMP744), which overcome these limitations, have been developed and are in clinical development. Further modifications of the indenoisoquinolines led to the fluoroindenoisoquinolines, one of which, LMP517, is the focus of this study. LMP517 showed better antitumor activity than its parent compound LMP744 against H82 (small cell lung cancer) xenografts. Genetic analyses in DT40 cells showed a dual TOP1 and TOP2 signature with selectivity of LMP517 for DNA repair-deficient tyrosyl DNA phosphodiesterase 2 (TDP2)- and Ku70-knockout cells. RADAR assays revealed that LMP517, and to a lesser extent LMP744, induce TOP2 cleavage complexes (TOP2cc) in addition to TOP1ccs. Histone γH2AX detection showed that, unlike classical TOP1 inhibitors, LMP517 targets cells independently of their position in the cell cycle. Our study establishes LMP517 as a dual TOP1 and TOP2 inhibitor with therapeutic potential.


Subject(s)
Indans/therapeutic use , Isoquinolines/therapeutic use , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Topoisomerase I Inhibitors/therapeutic use , Topoisomerase II Inhibitors/therapeutic use , Animals , Camptothecin/pharmacology , Carcinoma, Small Cell/drug therapy , Cell Line, Tumor , Chickens , DNA Topoisomerases, Type I , DNA Topoisomerases, Type II , Etoposide/pharmacology , Female , Histones/analysis , Humans , Indans/pharmacology , Isoquinolines/pharmacology , Lung Neoplasms/drug therapy , Lymphoma/pathology , Lymphoma/veterinary , Mice , Mice, Nude , Poultry Diseases/pathology , Random Allocation , Recombinant Proteins/drug effects , Substrate Specificity , Xenograft Model Antitumor Assays
18.
J Enzyme Inhib Med Chem ; 35(1): 1233-1239, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32441172

ABSTRACT

Vascular Endothelial Growth Factor (VEGF) promotes angiogenesis in tumours of various cancers. Monoclonal antibodies and nanobodies are one of the potent agents in the treatment of cancer. Due to their high costs, researchers are considering to design and produce peptides as a substitute approach in recent years. The aim of the current study was designing a mimotope against VEGF and evaluate its effects on cell proliferation and tube formation in the HUVEC cell line. For this, a peptide was designed against VEGF and chemically produced. The effects of synthetic peptide and nanobody on the inhibition of proliferation of HUVEC cells were examined using MTT and tube formation assays. The data indicate that the peptide was able to significantly inhibit both HUVEC cell proliferation and tube formation through inhibition of VEGF, highlighting the potential of peptides as a 'novel' class of candidate drugs to inhibit angiogenesis.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Single-Domain Antibodies/chemistry , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Cell Proliferation/drug effects , Computational Biology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Single-Domain Antibodies/pharmacology , Vascular Endothelial Growth Factor A/chemistry
19.
Microb Pathog ; 146: 104239, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32376360

ABSTRACT

Staphylococcus aureus expresses many Microbial Surface Recognizing Adhesive Matrix Molecules (MSCRAMM's) to recognize host extracellular matrix (ECM) molecules to initiate colonization. The MSCRAMM, fibronectin binding protein A (FnBPA), is an important adhesin for S. aureus infection. FnBPA also binds with fibrinogen (Fg) by using a unique ligand binding mechanism called dock, lock and latch. Nanoparticles, especially nanosilver particles have been widely used in a variety of biomedical applications which includes disease diagnosis and treatment, drug delivery and implanted medical device coating. In a biological system, when protein molecules encounter nanoparticle, they can be absorbed onto its surface which results in the formation of protein corona. In the present study, we have analysed the fibrinogen binding ability of rFnBPA(189-512) in the presence of silver nanoparticles by employing techniques like gel shift assay, Western blot, size exclusion chromatography, enzyme-linked immunosorbent assay, bio-layer interferometry and circular dichroism spectroscopy. The results indicate that rFnBPA(189-512) is unable to bind to Fg in the presence of a nanoparticle. This could be due to the inaccessibility of the Fg binding site and conformational change in rFnBPA(189-512). With nanoparticles, rFnBPA(189-512) undergoes significant structural changes as the ß-sheet content has drastically reduced to 10% from the initial 60% at higher concentration of the nanoparticle. Pathogenic bacteria interact with its surrounding environment through their surface molecules which includes MSCRAMMs. Therefore MSCRAMMs play an important role when bacteria encounter nanoparticles. The results of the present study suggest that the orientation of the protein during the absorption on the surface of a nanoparticle as well as the concentration of the nanoparticle, will dictate the function of the absorbed protein and in this case the Fg binding property of rFnBPA(189-512).


Subject(s)
Adhesins, Bacterial , Bacterial Adhesion/drug effects , Metal Nanoparticles , Staphylococcus aureus/metabolism , Adhesins, Bacterial/biosynthesis , Adhesins, Bacterial/drug effects , Adhesins, Bacterial/isolation & purification , Adhesins, Bacterial/metabolism , Bacterial Proteins/drug effects , Bacterial Proteins/metabolism , Fibrinogen/drug effects , Fibrinogen/metabolism , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Protein Binding , Recombinant Proteins/drug effects , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Staphylococcal Infections/drug therapy
20.
Neuromolecular Med ; 22(3): 359-369, 2020 09.
Article in English | MEDLINE | ID: mdl-32086739

ABSTRACT

Glioblastoma is the most common primary tumor of the central nervous system that develops chemotherapy resistance. Previous studies showed that Allicin could inhibit multiple cancer cells including glioblastoma, but the function of Allicin in glioblastoma is still unclear. Our work aimed to investigate the underlying molecular mechanism. The results showed that miR-486-3p levels were greatly increased in glioblastoma during Allicin treatment. Overexpression of miR-486-3p increased chemosensitivity to temozolomide (TMZ) in vitro and in vivo. O6-methylguanine-DNA methyltransferase (MGMT) was identified as a direct target of miR-486-3p, and miR-486-3p overexpression prevented the protein translation of MGMT. Moreover, overexpression of MGMT restored miR-486-3p-induced chemosensitivity to TMZ. Taken together, our studies revealed that Allicin could upregulate miR-486-3p and enhance TMZ sensitivity in glioblastoma. The results suggested that in the future, Allicin can be used as an adjuvant therapy with TMZ to improve the prognosis of patients, and miR-486-3p may be a potential target for glioblastoma treatment to improve the curative effects.


Subject(s)
Brain Neoplasms/drug therapy , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Disulfides/pharmacology , Glioblastoma/drug therapy , MicroRNAs/physiology , Neoplasm Proteins/antagonists & inhibitors , RNA, Neoplasm/physiology , Sulfinic Acids/pharmacology , Temozolomide/therapeutic use , Tumor Suppressor Proteins/antagonists & inhibitors , Adult , Animals , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Disulfides/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Glioblastoma/pathology , Humans , Mice , Mice, Nude , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recombinant Proteins/drug effects , Specific Pathogen-Free Organisms , Sulfinic Acids/therapeutic use , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...