2,584 research outputs found

    Molecular mechanism of orlistat hydrolysis by the thioesterase of human fatty acid synthase for targeted drug discovery

    Get PDF
    Indiana University-Purdue University Indianapolis (IUPUI)Fatty acid synthase (FASN) is over-expressed in many cancers, and novel inhibitors that target FASN may find use in the treatment of cancers. It has been shown that orlistat, an FDA approved drug for weight loss, inhibits the thioesterase (TE) of FASN, but can be hydrolyzed by TE. To understand the mechanisms of TE action and for designing better FASN inhibitors, I examined the mechanism of orlistat hydrolysis by TE using molecular dynamics simulations. I found that the hexyl tail of orlistat undergoes a conformational transition, destabilizing a hydrogen bond that forms between orlistat and the active site histidine. A water molecule can then hydrogen bond with histidine and become activated to hydrolyze orlistat. These findings suggest that rational design of inhibitors that block hexyl tail transition may lead to a more potent TE inhibitor. To search for novel inhibitors of TE, I performed virtual DOCK screening of FDA approved drugs followed by a fluorogenic assay using recombinant TE protein and found that proton pump inhibitors (PPIs) can competitively inhibit TE. PPIs, which are used for the treatment of gastroesophageal reflux and peptic ulcers, work to decrease gastric acid production by binding irreversibly with gastric hydrogen potassium ATPase in the stomach. Recently, PPIs have been reported to reduce drug resistance in cancer cells when used in combination with chemotherapeutics, although the mechanism of resistance reduction is unknown. Further investigation showed that PPIs are able to decrease FASN activity and cancer cell proliferation in a dose-dependent manner. These findings provide new evidence that FDA approved PPIs may synergistically suppress cancer cells by inhibiting TE of FASN and suggests that the use of PPIs in combinational therapies for the treatment of many types of cancer, including pancreatic cancer, warrants further investigation

    Molecular Docking Investigation of new inhibitors of falciparum vivax

    Get PDF
    Peer reviewedPublisher PD

    Curcumin and 10-undecenoic acid as natural quorum sensing inhibitors of LuxS/AI-2 of Bacillus subtilis and LasI/LasR of Pseudomonas aeruginosa

    Get PDF
    The quorum sensing (QS) system is related to cell-to-cell communication as a function of population density, which regulates several physiological functions including biofilm formation and virulence gene expression. QS inhibitors have emerged as a promising strategy to tackle virulence and biofilm development. Among a wide variety of phytochemicals, many of them have been described as QS inhibitors. Driven by their promising clues, this study aimed to identify active phytochemicals against LuxS/autoinducer-2 (AI-2) (as the universal QS system) from Bacillus subtilis and LasI/LasR (as a specific QS system) of Pseudomonas aeruginosa, through in silico analysis followed by in vitro validation. The optimized virtual screening protocols were applied to screen a phytochemical database containing 3479 drug-like compounds. The most promising phytochemicals were curcumin, pioglitazone hydrochloride, and 10-undecenoic acid. In vitro analysis corroborated the QS inhibitory activity of curcumin and 10-undecenoic acid, however, pioglitazone hydrochloride showed no relevant effect. Inhibitory effects on LuxS/AI-2 QS system triggered reduction of 33-77% by curcumin (at 1.25-5 mu g/mL) and 36-64% by 10-undecenoic acid (at 12.5-50 mu g/mL). Inhibition of LasI/LasR QS system was 21% by curcumin (at 200 mu g/mL) and 10-54% by 10-undecenoic acid (at 15.625-250 mu g/mL). In conclusion, in silico analysis allowed the identification of curcumin and, for the first time, 10-undecenoic acid (showing low cost, high availability, and low toxicity) as alternatives to counteract bacterial pathogenicity and virulence, avoiding the imposition of selective pressure usually related to classic industrial disinfection and antibiotics therapy

    Blocking The Entry Of HIV Into Host Cells Through Co-receptor Inhibition

    Get PDF
    The goal of our project is to synthesize a dual CXCR4/CCR5 co-receptor inhibitor to block the entry of HIV into host cells. In the early phase of the HIV-1 replication cycle, HIV-1 binds to host cells through the CD4 protein present on the host cell surface. To infect the cell, HIV-1 requires further interactions that promote fusion of the viral and cellular membranes. This can occur through binding to the chemokine co-receptors such as CXCR4 and CCR5. We are choosing dual inhibition since under selective pressure of a CCR5 antagonist, CXCR4-using strains have been shown to predominate. Through dual inhibition, we want to minimize possible resistance development that could occur when inhibiting only one of the two co-receptors. Using a computational screen, a compound predicted to bind to both CCR5 and CXCR4 was identified (Hit 1). We established a reaction scheme to synthesize an analog of this compound (target compound 4) through solid phase peptide synthesis. We confirmed the successful synthesis of this compound through LCMS and HPLC. We also made progress towards synthesizing two analogs of target compound (4) by varying the last Fmoc-phenylalanine. The first analog contained Fmoc-4-fluoro-phenylalanine while the second analog contained Fmoc-4-methoxy-phenylalanine. Lastly, we ran a docking screen to determine FDA approved compounds that could be repurposed as potential CCR5 inhibitors. In this, 40 compounds were identified to bind CCR5 with a higher affinity than FDA-approved CCR5 inhibitor, Maraviroc

    Chemoinformatic Identification of Novel Inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase

    Get PDF
    L-Aspartate α-decarboxylase (ADC) belongs to a class of pyruvoyl dependent enzymes and catalyzes the conversion of aspartate to β-alanine in the pantothenate pathway, which is critical for the growth of several micro-organisms, including Mycobacterium tuberculosis (Mtb). Its presence only in micro-organisms, fungi and plants and its absence in animals, particularly human, make it a promising drug target. We have followed a chemoinformatics-based approach to identify potential drug-like inhibitors against Mycobacterium tuberculosis L-aspartate α-decarboxylase (MtbADC). The structure-based high throughput virtual screening (HTVS) mode of the Glide program was used to screen 333,761 molecules of the Maybridge, National Cancer Institute (NCI) and Food and Drug Administration (FDA) approved drugs databases. Ligands were rejected if they cross-reacted with S-adenosylmethionine (SAM) decarboxylase, a human pyruvoyl dependent enzyme. The lead molecules were further analyzed for physicochemical and pharmacokinetic parameters, based on Lipinski's rule of five, and ADMET (absorption, distribution, metabolism, excretion and toxicity) properties. This analysis resulted in eight small potential drug-like inhibitors that are in agreement with the binding poses of the crystallographic ADC:fumarate and ADC:isoasparagine complex structures and whose backbone scaffolds seem to be suitable for further experimental studies in therapeutic development against tuberculosis

    Computational Molecular Docking Studies of Small Molecule Inhibitors With the SARS-CoV-2 Spike Protein Variants: In-Silico Drug Discovery Using Virtual Screening and Drug Repurposing Approaches

    Get PDF
    The pandemic caused by the emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 has caused a global public health crisis of nearly unprecedented scale. In the years following the outbreak, the scientific community has mobilized to develop several vaccines and treatments. Drug repurposing as a strategy for drug development has produced many of the current therapeutic options. The greatest challenge to designing a therapeutic inhibitor of SARS-CoV-2 is the shifting mutational landscape of the virus as it evolves. In this study, we focus on the spike protein as a target for potential inhibitors. We explore two methods of inhibiting spike function, allosteric inhibition and direct inhibition. In the study of allosteric inhibition, we screened two compound libraries against two allosteric sites. In the study of direct inhibition, several top-performing direct inhibitors of the wildtype spike were evaluated against five variants, B.1.1.7, B.1.351, P.1, B.1.617.2, and B.1.1.529. In summary, we identify four potential allosteric inhibitors that warrant further in-vitro study. We also find that the direct potential inhibitors of the wildtype spike had the most similar performance against the B.1.617.2 and B.1.1.7 variants

    Repositioning proton pump inhibitors as anticancer drugs by targeting the thioesterase domain of human fatty acid synthase

    Get PDF
    Fatty acid synthase (FASN), the enzyme responsible for de novo synthesis of free fatty acids, is up-regulated in many cancers. FASN is essential for cancer cell survival and contributes to drug resistance and poor prognosis. However, it is not expressed in most nonlipogenic normal tissues. Thus, FASN is a desirable target for drug discovery. Although different FASN inhibitors have been identified, none has successfully moved into clinical use. In this study, using in silico screening of an FDA-approved drug database, we identified proton pump inhibitors (PPIs) as effective inhibitors of the thioesterase activity of human FASN. Further investigation showed that PPIs inhibited proliferation and induced apoptosis of cancer cells. Supplementation of palmitate, the end product of FASN catalysis, rescued cancer cells from PPI-induced cell death. These findings provide new evidence for the mechanism by which this FDA-approved class of compounds may be acting on cancer cells

    Synergistic Effect of Chlorogenic Acid and Caffeic Acid with Fosfomycin on Growth Inhibition of a Resistant Listeria monocytogenes Strain

    Get PDF
    Listeria monocytogenes, a human foodborne pathogen that causes listeriosis with high-rate mortality, has been reported to be resistant to commonly used antibiotics. New antibiotics or cocktails of existing antibiotics with synergistic compounds are in high demand for treating this multi-drug-resistant pathogen. Fosfomycin is one of the novel and promising therapeutic antibiotics for the treatment of listeriosis. However, some L. monocytogenes strains with the FosX gene were recently reported to survive from the fosfomycin treatment. This work aims to identify FosX inhibitors that can revive fosfomycin in treating resistant L. monocytogenes. Since structures and activities of the FosX protein in L. monocytogenes have been well studied, we used an integrated computational and experimental approach to identify FosX inhibitors that show synergistic effect with fosfomycin in treating resistant L. monocytogenes. Specifically, automated ligand docking was implemented to perform virtual screening of the Indofine natural-product database and FDA-approved drugs to identify potential inhibitors. An in vitro bacterial growth inhibition test was then utilized to verify the effectiveness of identified compounds combined with fosfomycin in inhibiting the resistant L. monocytogenes strains. Two phenolic acids, i.e., caffeic acid and chlorogenic acid, were predicted as high-affinity FosX inhibitors from the ligand-docking platform. Experiments with these compounds indicated that the cocktail of either caffeic acid (1.5 mg/mL) or chlorogenic acid (3 mg/mL) with fosfomycin (50 mg/L) was able to significantly inhibit the growth of the pathogen. The finding of this work implies that the combination of fosfomycin with either caffeic acid or chlorogenic acid is of potential to be used in the clinical treatment of Listeria infections

    ANTI-TUBERCULAR ACTIVITY OF EDTA AND HOUSEHOLD CHEMICALS AGAINST MYCOBACTERIUM SMEGMATIS, A SURROGATE FOR MULTI-DRUG RESISTANT TUBERCULOSIS

    Get PDF
    Drug discovery efforts are increasingly being directed at re-purposing old drugs for use in treatment of multidrug resistant tuberculosis (MDRTB) which continues to pose serious health challenges worldwide. Recent studies suggest that Mycobacterium smegmatis, with innate resistance to isoniazid, could be used as a surrogate for MDRTB screening in laboratories without adequate containment facilities. The current study utilized resazurin microtiter assay plate (REMA) to screen common household chemicals and non-steroidal anti-inflammatory drugs (NSAIDs) for anti-tubercular activity against Mycobacterium smegmatis, as a non-pathogenic surrogate of MDRTB. A ligand-based approach was adopted in selecting household chemicals; using molecular docking tools to probe for binding affinity to Enoyl-[acyl-carrier-protein] reductase (InhA), the main target of isoniazid. Among household chemicals and analgesics studied, EDTA and lauric acid showed the highest activity with minimum inhibitory concentration (MIC) of 31 μg/mL and 7.8 μg/mL respectively, while Ibuprofen and aspirin exhibited activity with MIC of 125 μg/mL. Taken together, this study demonstrates the potential use of EDTA, lauric acid or NSAIDs in treatment of drug-resistant tuberculosis, a major contribution of the current stud

    Development of novel anticancer agents targeting G protein coupled receptor: GPR120

    Get PDF
    The G-protein coupled receptor, GPR120, has ubiquitous expression and multifaceted roles in modulating metabolic and anti-inflammatory processes. GPR120 - also known as Free Fatty Acid Receptor 4 (FFAR4) is classified as a free fatty acid receptor of the Class A GPCR family. GPR120 has recently been implicated as a novel target for cancer management. GPR120 gene knockdown in breast cancer studies revealed a role of GPR120-induced chemoresistance in epirubicin and cisplatin-induced DNA damage in tumour cells. Higher expression and activation levels of GPR120 is also reported to promote tumour angiogenesis and cell migration in colorectal cancer. A number of agonists targeting GPR120 have been reported, such as TUG891 and Compound39, but to date development of small-molecule inhibitors of GPR120 is limited. This research applied a rational drug discovery approach to discover and design novel anticancer agents targeting the GPR120 receptor. A homology model of GPR120 (short isoform) was generated to identify potential anticancer compounds using a combined in silico docking-based virtual screening (DBVS), molecular dynamics (MD) assisted pharmacophore screenings, structure–activity relationships (SAR) and in vitro screening approach. A pharmacophore hypothesis was derived from analysis of 300 ns all-atomic MD simulations on apo, TUG891-bound and Compound39-bound GPR120 (short isoform) receptor models and was used to screen for ligands interacting with Trp277 and Asn313 of GPR120. Comparative analysis of 100 ns all-atomic MD simulations of 9 selected compounds predicted the effects of ligand binding on the stability of the “ionic lock” – a characteristic of Class A GPCRs activation and inactivation. The “ionic lock” between TM3(Arg136) and TM6(Asp) is known to prevent G-protein recruitment while GPCR agonist binding is coupled to outward movement of TM6 breaking the “ionic lock” which facilitates G-protein recruitment. The MD-assisted pharmacophore hypothesis predicted Cpd 9, (2-hydroxy-N-{4-[(6-hydroxy-2-methylpyrimidin-4-yl) amino] phenyl} benzamide) to act as a GPR120S antagonist which can be evaluated and characterised in future studies. Additionally, DBVS of a small molecule database (~350,000 synthetic chemical compounds) against the developed GPR120 (short isoform) model led to selection of the 13 hit molecules which were then tested in vitro to evaluate their cytotoxic, colony forming and cell migration activities against SW480 – human CRC cell line expressing GPR120. Two of the DBVS hit molecules showed significant (\u3e 90%) inhibitory effects on cell growth with micromolar affinities (at 100 μM) - AK-968/12713190 (dihydrospiro(benzo[h]quinazoline-5,1′-cyclopentane)-4(3H)-one) and AG-690/40104520 (fluoren-9-one). SAR analysis of these two test compounds led to the identification of more active compounds in cell-based cytotoxicity assays – AL-281/36997031 (IC50 = 5.89–6.715 μM), AL-281/36997034 (IC50 = 6.789 to 7.502 μM) and AP-845/40876799 (IC50 = 14.16-18.02 μM). In addition, AL-281/36997031 and AP-845/40876799 were found to be significantly target-specific during comparative cytotoxicity profiling in GPR120-silenced and GPR120-expressing SW480 cells. In wound healing assays, AL-281/36997031 was found to be the most active at 3 μM (IC25) and prevented cell migration. As well as in the assessment of the proliferation ability of a single cell to survive and form colonies through clonogenic assays, AL-281/36997031 was found to be the most potent of all three test compounds with the survival rate of ~ 30% at 3 μM. The inter-disciplinary approach applied in this work identified potential chemical scaffolds –spiral benzo-quinazoline and fluorenone, targeting GPR120 which can be further explored for designing anti-cancer drug development studies
    corecore