1
|
Ritter MR, Oliveira MTD, Ardisson JS, Gonçalves RDCR, Kitagawa RR, Lima DDS, Seixas FAV, Nakamura CV, Mello JCPD. Trichilia catigua against Helicobacter pylori: An in vitro, molecular and in silico approach. Fitoterapia 2024; 177:106101. [PMID: 38945495 DOI: 10.1016/j.fitote.2024.106101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Helicobacter pylori is a bacterium that is present in the stomach of about 50% of the global population and is associated with several gastric disorders, including cancer. Natural products with antimicrobial activity have been tested against H. pylori, among them Trichilia catigua (catuaba), which is widely distributed in Brazil. This study aimed to evaluate extracts of T. catigua bark against H. pylori via determination of the minimum inhibitory and bactericidal concentrations (MIC and MBC); evaluation of virulence factors by real-time PCR, synergism with standard antimicrobials and morphology by scanning electron microscopy and simulations of the mechanism of action by molecular docking. The ethyl acetate fraction provided the best results, with an MIC50 of 250 μg/mL and a 42.34% reduction in urease activity, along with reduced expression of the CagA and VacA genes, which encode for the main virulence factors. This fraction presented synergistic activity with clarithromycin, reducing the MIC of the drug by four-fold. Docking simulations suggested that the extracts inhibit fatty acid synthesis by the FAS-II system, causing damage to the cell membrane. Therefore, T. catigua extracts have potential as an adjuvant to treatment and are promising for the development of new anti-H. pylori drugs.
Collapse
Affiliation(s)
- Mariane Roberta Ritter
- Laboratory of Pharmaceutical Biology, Palafito, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Brazil
| | - Marcelo Tempesta de Oliveira
- Laboratory of Pharmaceutical Biology, Palafito, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Brazil
| | - Juliana Santa Ardisson
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Espírito Santo, Vitória, Brazil
| | | | - Rodrigo Rezende Kitagawa
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Diego de Souza Lima
- Department of Technology, Universidade Estadual de Maringá, Umuarama, Brazil
| | | | - Celso Vataru Nakamura
- Technological Innovation Laboratory in the Development of Pharmaceuticals and Cosmetics, Universidade Estadual de Maringá, Brazil
| | - João Carlos Palazzo de Mello
- Laboratory of Pharmaceutical Biology, Palafito, Department of Pharmacy, Universidade Estadual de Maringá, Maringá, Brazil.
| |
Collapse
|
2
|
Norseeda K, Bin Aziz Pavel F, Rutherford JT, Meer HN, Dureja C, Hurdle JG, Hevener KE, Sun D. Synthesis and evaluation of phenylimidazole FabK inhibitors as new Anti-C. Difficile agents. Bioorg Med Chem 2023; 88-89:117330. [PMID: 37224699 PMCID: PMC10834300 DOI: 10.1016/j.bmc.2023.117330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/28/2023] [Accepted: 05/10/2023] [Indexed: 05/26/2023]
Abstract
Previously, 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(5-(pyridin-2-ylthio)thiazol-2-yl)urea bearing a p-bromine substitution was shown to possess selective inhibitory activity against the Clostridioides difficile enoyl-acyl carrier protein (ACP) reductase II enzyme, FabK. Inhibition of CdFabK by this compound translated to promising antibacterial activity in the low micromolar range. In these studies, we sought to expand our knowledge of the SAR of the phenylimidazole CdFabK inhibitor series while improving the potency of the compounds. Three main series of compounds were synthesized and evaluated based on: 1) pyridine head group modifications including the replacement with a benzothiazole moiety, 2) linker explorations, and 3) phenylimidazole tail group modifications. Overall, improvement in the CdFabK inhibition was achieved, while maintaining the whole cell antibacterial activity. Specifically, compounds 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(5-((3-(trifluoromethyl)pyridin-2-yl)thio)thiazol-2-yl)urea, 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(6-(trifluoromethyl)benzo[d]thiazol-2-yl)urea, and 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(6-chlorobenzo[d]thiazol-2-yl)urea showed CdFabK inhibition (IC50 = 0.10 to 0.24 μM), a 5 to 10-fold improvement in biochemical activity relative to 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(5-(pyridin-2-ylthio)thiazol-2-yl)urea, with anti-C. difficile activity ranging from 1.56 to 6.25 μg/mL. Detailed analysis of the expanded SAR, supported by computational analysis, is presented.
Collapse
Affiliation(s)
- Krissada Norseeda
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, United States
| | - Fahad Bin Aziz Pavel
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Jacob T Rutherford
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, United States
| | - Humna N Meer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Chetna Dureja
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, United States
| | - Julian G Hurdle
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, United States
| | - Kirk E Hevener
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| | - Dianqing Sun
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, United States.
| |
Collapse
|
3
|
Hasan AH, Shakya S, Hussain FHS, Murugesan S, Chander S, Pratama MRF, Jamil S, Das B, Biswas S, Jamalis J. Design, synthesis, anti-acetylcholinesterase evaluation and molecular modelling studies of novel coumarin-chalcone hybrids. J Biomol Struct Dyn 2023; 41:11450-11462. [PMID: 36591704 DOI: 10.1080/07391102.2022.2162583] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023]
Abstract
The major enzyme responsible for the hydrolytic breakdown of the neurotransmitter acetylcholine (ACh) is acetylcholinesterase (AChE). Acetylcholinesterase inhibitors (AChEIs) are the most prescribed class of medications for the treatment of Alzheimer's disease (AD) and dementia. The limitations of available therapy, like side effects, drug tolerance, and inefficacy in halting disease progression, drive the need for better, more efficacious, and safer drugs. In this study, a series of fourteen novel chalcone-coumarin derivatives (8a-n) were designed, synthesized and characterized by spectral techniques like FT-IR, NMR, and HR-MS. Subsequently, the synthesized compounds were tested for their ability to inhibit acetylcholinesterase (AChE) activity by Ellman's method. All tested compounds showed AChE inhibition with IC50 value ranging from 0.201 ± 0.008 to 1.047 ± 0.043 μM. Hybrid 8d having chloro substitution on ring-B of the chalcone scaffold showed relatively better potency, with IC50 value of 0.201 ± 0.008 μM compared to other members of the series. The reference drug, galantamine, exhibited an IC50 at 1.142 ± 0.027 μM. Computational studies revealed that designed compounds bind to the peripheral anionic site (PAS), the catalytic active site (CAS), and the mid-gorge site of AChE. Putative binding modes, ligand-enzyme interactions, and stability of the best active compound are studied using molecular docking, followed by molecular dynamics (MD) simulations. The cytotoxicity of the synthesised derivatives was determined using the MTT test at three concentrations (100 g/mL, 500 g/mL, and 1 mg/mL). None of the chemicals had a significant effect on the body at the highest dose of 1 mg/mL.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aso Hameed Hasan
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Department of Chemistry, College of Science, University of Garmian, Kalar, Kurdistan Region-Iraq, Iraq
| | - Sonam Shakya
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Faiq H S Hussain
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region-Iraq, Iraq
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Birla Institute of Technology & Science Pilani (BITS Pilani), Pilani, Rajasthan, India
| | - Subhash Chander
- Amity Institute of Phytochemistry and Phytomedicine, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Mohammad Rizki Fadhil Pratama
- Doctoral Program of Pharmaceutical Sciences, Universitas Airlangga, Surabaya, East Java, Indonesia
- Department of Pharmacy, Universitas Muhammadiyah Palangkaraya, Palangka Raya, Central Kalimantan, Indonesia
| | - Shajarahtunnur Jamil
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Basundhara Das
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Translational Cancer & Stem Cell Research Laboratory, Noida, Uttar Pradesh, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Translational Cancer & Stem Cell Research Laboratory, Noida, Uttar Pradesh, India
| | - Joazaizulfazli Jamalis
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| |
Collapse
|
4
|
Arif SM, Floto RA, Blundell TL. Using Structure-guided Fragment-Based Drug Discovery to Target Pseudomonas aeruginosa Infections in Cystic Fibrosis. Front Mol Biosci 2022; 9:857000. [PMID: 35433835 PMCID: PMC9006449 DOI: 10.3389/fmolb.2022.857000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) is progressive genetic disease that predisposes lungs and other organs to multiple long-lasting microbial infections. Pseudomonas aeruginosa is the most prevalent and deadly pathogen among these microbes. Lung function of CF patients worsens following chronic infections with P. aeruginosa and is associated with increased mortality and morbidity. Emergence of multidrug-resistant, extensively drug-resistant and pandrug-resistant strains of P. aeruginosa due to intrinsic and adaptive antibiotic resistance mechanisms has failed the current anti-pseudomonal antibiotics. Hence new antibacterials are urgently needed to treat P. aeruginosa infections. Structure-guided fragment-based drug discovery (FBDD) is a powerful approach in the field of drug development that has succeeded in delivering six FDA approved drugs over the past 20 years targeting a variety of biological molecules. However, FBDD has not been widely used in the development of anti-pseudomonal molecules. In this review, we first give a brief overview of our structure-guided FBDD pipeline and then give a detailed account of FBDD campaigns to combat P. aeruginosa infections by developing small molecules having either bactericidal or anti-virulence properties. We conclude with a brief overview of the FBDD efforts in our lab at the University of Cambridge towards targeting P. aeruginosa infections.
Collapse
Affiliation(s)
| | - R. Andres Floto
- Molecular Immunity Unit, Department of Medicine University of Cambridge, MRC-Laboratory of Molecular Biology, Cambridge, United Kingdom
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, United Kingdom
| | - Tom L. Blundell
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Tom L. Blundell,
| |
Collapse
|
5
|
Computer-Aided Design of Peptidomimetic Inhibitors of Falcipain-3: QSAR and Pharmacophore Models. Sci Pharm 2021. [DOI: 10.3390/scipharm89040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In this work, antiparasitic peptidomimetics inhibitors (PEP) of falcipain-3 (FP3) of Plasmodium falciparum (Pf) are proposed using structure-based and computer-aided molecular design. Beginning with the crystal structure of PfFP3-K11017 complex (PDB ID: 3BWK), three-dimensional (3D) models of FP3-PEPx complexes with known activities ( IC50exp) were prepared by in situ modification, based on molecular mechanics and implicit solvation to compute Gibbs free energies (GFE) of inhibitor-FP3 complex formation. This resulted in a quantitative structure–activity relationships (QSAR) model based on a linear correlation between computed GFE (ΔΔGcom) and the experimentally measured IC50exp. Apart from the structure-based relationship, a ligand-based quantitative pharmacophore model (PH4) of novel PEP analogues where substitutions were directed by comparative analysis of the active site interactions was derived using the proposed bound conformations of the PEPx. This provided structural information useful for the design of virtual combinatorial libraries (VL), which was virtually screened based on the 3D-QSAR PH4. The end results were predictive inhibitory activities falling within the low nanomolar concentration range.
Collapse
|
6
|
Ünsaldı E, Kurt-Kızıldoğan A, Özcan S, Becher D, Voigt B, Aktaş C, Özcengiz G. Proteomic analysis of a hom-disrupted, cephamycin C overproducing Streptomyces clavuligerus. Protein Pept Lett 2021; 28:205-220. [PMID: 32707026 DOI: 10.2174/0929866527666200723163655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Streptomyces clavuligerus is prolific producer of cephamycin C, a medically important antibiotic. In our former study, cephamycin C titer was 2-fold improved by disrupting homoserine dehydrogenase (hom) gene of aspartate pahway in Streptomyces clavuligerus NRRL 3585. OBJECTIVE In this article, we aimed to provide a comprehensive understanding at the proteome level on potential complex metabolic changes as a consequence of hom disruption in Streptomyces clavuligerus AK39. METHODS A comparative proteomics study was carried out between the wild type and its hom disrupted AK39 strain by 2 Dimensional Electrophoresis-Matrix Assisted Laser Desorption and Ionization Time-Of-Flight Mass Spectrometry (2DE MALDI-TOF/MS) and Nanoscale Liquid Chromatography- Tandem Mass Spectrometry (nanoLC-MS/MS) analyses. Clusters of Orthologous Groups (COG) database was used to determine the functional categories of the proteins. The theoretical pI and Mw values of the proteins were calculated using Expasy pI/Mw tool. RESULTS "Hypothetical/Unknown" and "Secondary Metabolism" were the most prominent categories of the differentially expressed proteins. Upto 8.7-fold increased level of the positive regulator CcaR was a key finding since CcaR was shown to bind to cefF promoter thereby direcly controlling its expression. Consistently, CeaS2, the first enzyme of CA biosynthetic pathway, was 3.3- fold elevated. There were also many underrepresented proteins associated with the biosynthesis of several Non-Ribosomal Peptide Synthases (NRPSs), clavams, hybrid NRPS/Polyketide synthases (PKSs) and tunicamycin. The most conspicuously underrepresented protein of amino acid metabolism was 4-Hydroxyphenylpyruvate dioxygenase (HppD) acting in tyrosine catabolism. The levels of a Two Component System (TCS) response regulator containing a CheY-like receiver domain and an HTH DNA-binding domain as well as DNA-binding protein HU were elevated while a TetR-family transcriptional regulator was underexpressed. CONCLUSION The results obtained herein will aid in finding out new targets for further improvement of cephamycin C production in Streptomyces clavuligerus.
Collapse
Affiliation(s)
- Eser Ünsaldı
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Turkey
| | | | - Servet Özcan
- Department of Biology, Erciyes University, Kayseri 38280, Turkey
| | - Dörte Becher
- Institute of Microbiology, Ernst- Moritz-Arndt-University of Greifswald, Greifswald D-17487, Germany
| | - Birgit Voigt
- Institute of Microbiology, Ernst- Moritz-Arndt-University of Greifswald, Greifswald D-17487, Germany
| | - Caner Aktaş
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Turkey
| | - Gülay Özcengiz
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Turkey
| |
Collapse
|
7
|
Shawon J, Khan AM, Shahriar I, Halim MA. Improving the binding affinity and interaction of 5-Pentyl-2-Phenoxyphenol against Mycobacterium Enoyl ACP reductase by computational approach. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
8
|
Aygün C, Mutlu Ö. Computational characterisation of Toxoplasma gondii FabG (3-oxoacyl-[acyl-carrier-protein] reductase): a combined virtual screening and all-atom molecular dynamics simulation study. J Biomol Struct Dyn 2020; 40:1952-1969. [PMID: 33063633 DOI: 10.1080/07391102.2020.1834456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii is an opportunistic obligate parasite, ubiquitous around the globe with seropositivity rates that range from 10% to 90% and infection by the parasite of pregnant women causes pre-natal death of the foetus in most cases and severe neurodegenerative syndromes in some. No vaccine is currently available, and since drug-resistance is common among T. gondii strains, discovering lead compounds for drug design using diverse tactics is necessary. In this study, the sole constituent isoform of an enzymatic 3-oxoacyl-[acyl-carrier-protein] reduction step in an apicoplast-located fatty acid biosynthesis pathway was chosen as a possible drug target. FASII is prokaryotic therefore, targeting it would pose fewer side-effects to human hosts. After a homology 3D modelling of TgFabG, a high-throughput virtual screening of 9867 compounds, the elimination of ligands was carried out by a flexible ligand molecular docking and 200 ns molecular dynamics simulations, with additional DCCM and PC plot analyses. Molecular Dynamics and related post-MD analyses of the top 3 TgFabG binders selected for optimal free binding energies, showed that L2 maintained strong H-bonds with TgFabG and facilitated structural reorientation expected of FabGs, namely an expansion of the Rossmann Fold and a flexible lid capping. The most flexible TgFabG sites were the α7 helix (the flexible lid region) and the β4-α4 and β5-α6 loops. For TgFabG-L2, the movements of these regions toward the active site enabled greater ligand stability. Thus, L2 ("Skimmine"; PubChem ID: 320361), was ultimately selected as the optimal candidate for the discovery of lead compounds for rational drug design.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Can Aygün
- Faculty of Arts and Sciences, Biology Department, Marmara University, Istanbul, Turkey
| | - Özal Mutlu
- Faculty of Arts and Sciences, Biology Department, Marmara University, Istanbul, Turkey
| |
Collapse
|
9
|
Trevisan DAC, da Silva PV, Farias ABP, Campanerut-Sá PAZ, Ribeiro TDVR, Faria DR, de Mendonça PSB, de Mello JCP, Seixas FAV, Mikcha JMG. Antibacterial activity of Barbatimão (Stryphnodendron adstringens) against Staphylococcus aureus: in vitro and in silico studies. Lett Appl Microbiol 2020; 71:259-271. [PMID: 32412089 DOI: 10.1111/lam.13317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/23/2022]
Abstract
We evaluated the activity of the aqueous fraction and the ethyl acetate fraction of Stryphnodendron adstringens against Staphylococcus aureus and proposed their mechanism of action. The antibacterial activity of S. adstringens fractions was evaluated against S. aureus and the cell targets were rated by docking. The fractions showed moderate antibacterial activity against S. aureus without toxicity on two mammalian cell lines. They also showed synergistic antibacterial activity with tannic acid (TA). In silico assays indicated FabG, FabZ and FabI as probable targets. The metabolic pathway for fatty acid biosynthesis in S. aureus was affected by components of S. adstringens. The synergistic effect when combining TA with S. adstringens fractions suggests a natural alternative to S. aureus control. SIGNIFICANCE AND IMPACT OF THE STUDY: This is the first study describing the possible targets of action of Stryphnodendron adstringens on Staphylococcus aureus. Molecular dynamics simulations showed that the components of S. adstringens affected the metabolic pathway for fatty acid biosynthesis (FAS II) in S. aureus, inhibiting the FabI, FabG and FabZ enzymes. As tannic acid (TA) is a known inhibitor of some targets identified, we showed synergistic antibacterial activity of S. adstringens in combination with TA. This combination did not show toxicity against HaCaT and Vero cells and based on all these results we suggest that S. adstringens can be a natural and sustainable alternative to S. aureus control.
Collapse
Affiliation(s)
- D A C Trevisan
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - P V da Silva
- Department of Technology, State University of Maringá, Maringá, Umuarama, PR, Brazil
| | - A B P Farias
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - P A Z Campanerut-Sá
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - T D V R Ribeiro
- Department of Pharmacy, State University of Maringá, Maringá, PR, Brazil
| | - D R Faria
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - P S B de Mendonça
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - J C P de Mello
- Department of Pharmacy, State University of Maringá, Maringá, PR, Brazil
| | - F A V Seixas
- Department of Technology, State University of Maringá, Maringá, Umuarama, PR, Brazil
| | - J M G Mikcha
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| |
Collapse
|
10
|
Padmini T, Bhikshapathi D, Suresh K, Kulkarni R, Kamal BR. Novel Aminopyrazole Tagged Hydrazones as Anti-Tubercular Agents: Synthesis and Molecular Docking Studies. Med Chem 2020; 17:344-351. [PMID: 32407282 DOI: 10.2174/1573406416666200514084747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pyrazole derivatives have been reported to possess numerous pharmacological activities viz., anti-inflammatory, antipsychotic, etc. Our group has disclosed that pyrazole benzamides display potent antibacterial and anti-tubercular activities. OBJECTIVE Synthesis of new pyrazole acetamides which possess hydrazone group to be evaluated for antitubercular activity. METHODS The key intermediate 5-aminopyrazole was synthesized with the known procedure, which is then converted into chloroacetamide. This compound than resulted in hydrazine derivative and finally converted into aromatic hydrazones. All the compounds were screened for antitubercular activity. RESULTS All the synthesized compounds have been characterized by their spectral data obtained and subjected to anti-tubercular activity. Among all the twenty tested compounds, three compounds, 5a5, 5b5 and 5b7 have demonstrated MIC value of 3.12 μg/mL against MTB H37Rv. Docking studies revealed important hydrogen bonding interactions with InhA. CONCLUSION Three compounds 5a5, 5b5 and 5b7 were found to be most potent among the series of compounds. Docking studies of compounds explained the presence of hydrogen bonding and π- π stacking interactions with InhA. Further synthesis of more such derivatives with optimized groups would produce compounds with more potent anti-tubercular activity.
Collapse
Affiliation(s)
| | - Darna Bhikshapathi
- Teegala Ram Reddy College of Pharmacy, Pragathi Colony, Meerpet, Hyderabad- 500097, Telangana, India
| | - Kandagatla Suresh
- Vijaya College of Pharmacy, Hayatnagar, Hyderabad-501511, Telangana, Hyderabad, India
| | - Ravindra Kulkarni
- Bharati Vidyapeeth's Poona College of Pharmacy, Erandwane, Pune-411038, Maharashtra, India
| | - Bigala R Kamal
- Research Supervisor, Mewar University, Chittorgarh, Rajasthan, India
| |
Collapse
|
11
|
Hasan AH, Amran SI, Saeed Hussain FH, Jaff BA, Jamalis J. Molecular Docking and Recent Advances in the Design and Development of Cholinesterase Inhibitor Scaffolds: Coumarin Hybrids. ChemistrySelect 2019. [DOI: 10.1002/slct.201903607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Aso Hameed Hasan
- Department of ChemistryFaculty of ScienceUniversiti Teknologi Malaysia 81310 Johor Bahru, Johor Malaysia
- Department of ChemistryCollege of ScienceUniversity of Garmian- Kalar, Kurdistan Region-Iraq Iraq
| | - Syazwani Itri Amran
- Department of BiosciencesFaculty of ScienceUniversiti Teknologi Malaysia 81310 Johor Bahru, Johor Malaysia
| | | | - Baram Ahmed Jaff
- Charmo Research CenterChemistry DepartmentCharmo University 46023 Chamchamal, Kurdistan Region-Iraq Iraq
| | - Joazaizulfazli Jamalis
- Department of ChemistryFaculty of ScienceUniversiti Teknologi Malaysia 81310 Johor Bahru, Johor Malaysia
| |
Collapse
|
12
|
Sun ZG, Xu YJ, Xu JF, Liu QX, Yang YS, Zhu HL. Introducing Broadened Antibacterial Activity to Rhodanine Derivatives Targeting Enoyl-Acyl Carrier Protein Reductase. Chem Pharm Bull (Tokyo) 2019; 67:125-129. [PMID: 30713272 DOI: 10.1248/cpb.c18-00663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Broadened antibacterial activity was introduced to rhodanine derivatives targeting Mycobacterial tuberculosis enoyl-acyl carrier protein reductase (Mtb InhA) by recruiting feature of xacins to bring DNA Gyrase B inhibitory capability. This is significant for preventing further bacterial injections in the tuberculosis treatment. The most potent compound Cy14 suggested comparable bioactivity (IC50 = 3.18 µM for Mtb InhA; IC50 = 10 nM for DNA Gyrase B) with positive controls. Structure-activity relationship discussion and molecular docking model revealed the significance of rhodanine moiety and derived methoxyl on meta-position, pointing out orientations for future modification.
Collapse
Affiliation(s)
- Zhi-Gang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University.,Central Laboratory, Linyi Central Hospital
| | - Yun-Jie Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| | - Jian-Fei Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| | - Qi-Xing Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| | - Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| |
Collapse
|
13
|
Wang T, Tang Y, Yang Y, An Q, Sang Z, Yang T, Liu P, Zhang T, Deng Y, Luo Y. Discovery of novel anti-tuberculosis agents with pyrrolo[1,2-a]quinoxaline-based scaffold. Bioorg Med Chem Lett 2018; 28:2084-2090. [PMID: 29748048 DOI: 10.1016/j.bmcl.2018.04.043] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/03/2018] [Accepted: 04/16/2018] [Indexed: 02/05/2023]
Abstract
A series of small molecules with novel pyrrolo[1,2-a]quinoxaline-based scaffold was designed via molecular hybridization of privileged agents active against Mycobacterium tuberculosis. Twenty-three compounds were synthesized and investigated for their antitubercular activities in vitro where ten compounds showed appreciable activities and moderate cytotoxicity. Compound 12g with MIC values of 5 μg/ml as a representative may possess better oral bioavailability and indicated high permeability by the parallel artificial membrane permeation assay of the blood-brain barrier (PAMPA-BBB). Further, the determination of enzyme inhibition and molecular docking study indicated that InhA may be the biological target of the active compounds. The results suggest the pyrrolo[1,2-a]quinoxaline hybrids as potential antitubercular leads for the development of new antitubercular agents.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunxiang Tang
- Institute of Physical Science and Information Technology, Anhui University, Hefei 230039, China
| | - Yang Yang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qi An
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zitai Sang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pingxian Liu
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510000, China
| | - Yong Deng
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Youfu Luo
- Department of Medicinal Chemistry, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Synthesis and biological evaluation of coumarin derivatives containing imidazole skeleton as potential antibacterial agents. Eur J Med Chem 2018; 143:958-969. [DOI: 10.1016/j.ejmech.2017.11.100] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/16/2017] [Accepted: 11/29/2017] [Indexed: 12/28/2022]
|
15
|
Lone MY, Manhas A, Athar M, Jha PC. Identification of InhA inhibitors: A combination of virtual screening, molecular dynamics simulations and quantum chemical studies. J Biomol Struct Dyn 2017; 36:2951-2965. [DOI: 10.1080/07391102.2017.1372313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Mohsin Y. Lone
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Anu Manhas
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Mohd. Athar
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Prakash C. Jha
- Centre for Applied Chemistry, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| |
Collapse
|
16
|
Lone MY, Athar M, Gupta VK, Jha PC. Identification of Mycobacterium tuberculosis enoyl-acyl carrier protein reductase inhibitors: A combined in-silico and in-vitro analysis. J Mol Graph Model 2017; 76:172-180. [DOI: 10.1016/j.jmgm.2017.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 01/20/2023]
|
17
|
Ahamad S, Rahman S, Khan FI, Dwivedi N, Ali S, Kim J, Imtaiyaz Hassan M. QSAR based therapeutic management of M. tuberculosis. Arch Pharm Res 2017; 40:676-694. [PMID: 28456911 DOI: 10.1007/s12272-017-0914-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 04/06/2017] [Indexed: 01/09/2023]
Abstract
Mycobacterium tuberculosis is responsible for severe mortality and morbidity worldwide but, under-developed and developing countries are more prone to infection. In search of effective and wide-spectrum anti-tubercular agents, interdisciplinary approaches are being explored. Of the several approaches used, computer based quantitative structure activity relationship (QSAR) have gained momentum. Structure-based drug design and discovery implies a combined knowledge of accurate prediction of ligand poses with the good prediction and interpretation of statistically validated models derived from the 3D-QSAR approach. The validated models are generally used to screen a small combinatorial library of potential synthetic candidates to identify hits which further subjected to docking to filter out compounds as novel potential emerging drug molecules to address multidrug-resistant tuberculosis. Several newer models are integrated to QSAR methods which include different types of chemical and biological data, and simultaneous prediction of pharmacological activities including toxicities and/or other safety profiles to get new compounds with desired activity. In the process, several newer molecules have been identified which are now being assessed for their clinical efficacy. Present review deals with the advances made in the field highlighting overall future prospects of the development of anti-tuberculosis drugs.
Collapse
Affiliation(s)
- Shahzaib Ahamad
- Department of Biotechnology, School of Engineering & Technology, IFTM University, Lodhipur-Rajput, Delhi Road, Moradabad, India
| | - Safikur Rahman
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, South Korea
| | - Faez Iqbal Khan
- School of Chemistry and Chemical Engineering, Henan University of Technology, Henan, 450001, China.,Department of Chemistry, Rhodes University, Grahamstown, 6140, South Africa
| | - Neeraja Dwivedi
- Department of Biotechnology, School of Engineering & Technology, IFTM University, Lodhipur-Rajput, Delhi Road, Moradabad, India
| | - Sher Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 10025, India
| | - Jihoe Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, South Korea.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 10025, India.
| |
Collapse
|
18
|
Slepikas L, Chiriano G, Perozzo R, Tardy S, Kranjc A, Patthey-Vuadens O, Ouertatani-Sakouhi H, Kicka S, Harrison CF, Scrignari T, Perron K, Hilbi H, Soldati T, Cosson P, Tarasevicius E, Scapozza L. In Silico Driven Design and Synthesis of Rhodanine Derivatives as Novel Antibacterials Targeting the Enoyl Reductase InhA. J Med Chem 2016; 59:10917-10928. [DOI: 10.1021/acs.jmedchem.5b01620] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Liudas Slepikas
- School
of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
- Faculty
of Pharmacy, Lithuanian University of Health Sciences, LT 44307 Kaunas, Lithuania
| | - Gianpaolo Chiriano
- School
of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| | - Remo Perozzo
- School
of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| | - Sébastien Tardy
- School
of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| | - Agata Kranjc
- School
of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| | - Ophélie Patthey-Vuadens
- School
of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| | - Hajer Ouertatani-Sakouhi
- Department
of Cell Physiology and Metabolism, CMU, Rue Michel-Servet 1 CH-1211 Geneva, Switzerland
| | - Sébastien Kicka
- Department
of Biochemistry, University of Geneva, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| | - Christopher F. Harrison
- Max
von Pettenkofer Institute, Department of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
| | - Tiziana Scrignari
- Microbiology
Unit, Department of Botany and Plant Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Karl Perron
- Microbiology
Unit, Department of Botany and Plant Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Hubert Hilbi
- Max
von Pettenkofer Institute, Department of Medicine, Ludwig-Maximilians University Munich, 80336 Munich, Germany
- Institute
of Medical Microbiology, Department of Medicine, University of Zürich, Gloriastrasse 30/32, CH-8006 Zürich, Switzerland
| | - Thierry Soldati
- Department
of Biochemistry, University of Geneva, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| | - Pierre Cosson
- Department
of Cell Physiology and Metabolism, CMU, Rue Michel-Servet 1 CH-1211 Geneva, Switzerland
| | - Eduardas Tarasevicius
- Faculty
of Pharmacy, Lithuanian University of Health Sciences, LT 44307 Kaunas, Lithuania
| | - Leonardo Scapozza
- School
of Pharmaceutical Sciences, Department of Pharmaceutical Biochemistry, University of Geneva and University of Lausanne, 30 Quai Ernest Ansermet, CH-1211 Geneva, Switzerland
| |
Collapse
|
19
|
Ünsaldı E, Kurt-Kızıldoğan A, Voigt B, Becher D, Özcengiz G. Proteome-wide alterations in an industrial clavulanic acid producing strain of Streptomyces clavuligerus. Synth Syst Biotechnol 2016; 2:39-48. [PMID: 29062960 PMCID: PMC5625738 DOI: 10.1016/j.synbio.2016.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/17/2016] [Accepted: 10/23/2016] [Indexed: 11/26/2022] Open
Abstract
The usefulness of genetic/metabolic engineering for further improvement of industrial strains is subject of discussion because of the general lack of knowledge on genetic alterations introduced by iterative cycles of random mutagenesis in such strains. An industrial clavulanic acid (CA)-overproducer Streptomyces clavuligerus DEPA was assessed to understand proteome-wide changes that have occurred in a local industrial CA overproducer developed through succesive mutagenesis programs. The proteins that could be identified corresponded to 33 distinct ORFs for underrepresented ones and 60 ORFs for overrepresented ones. Three CA biosynthetic enzymes were overrepresented in S. clavuligerus DEPA; carboxyethylarginine synthase (Ceas2), clavaldehyde dehydrogenase (Car) and carboxyethyl-arginine beta-lactam-synthase (Bls2) whereas the enzymes of two other secondary metabolites were underrepresented along with two important global regulators [two-component system (TCS) response regulator (SCLAV_2102) and TetR-family transcriptional regulator (SCLAV_3146)] that might be related with CA production and/or differentiation. γ-butyrolactone biosynthetic protein AvaA2 was 2.6 fold underrepresented in S. clavuligerus DEPA. The levels of two glycolytic enzymes, 2,3-bisphosphoglycerate-dependent phosphoglycerate mutase and phosophoglycerate kinase were found decreased while those of dihydrolipoyl dehydrogenase (E3) and isocitrate dehydrogenase, with two isoforms were found as significantly increased. A decrease of amino acid metabolism, methionine biosynthesis in particular, as well as S-adenosylmethionine synthetase appeared as one of the prominent mechanisms of success of S. clavuligerus DEPA strain as a prolific producer of CA. The levels of two enzymes of shikimate pathway that leads to the production of aromatic amino acids and aromatic secondary metabolites were also underrepresented. Some of the overrepresented stress proteins in S. clavuligerus DEPA included polynucleotide phosphorylase/polyadenylase (PNPase), ATP-dependent DNA helicase, two isoforms of an anti-sigma factor and thioredoxin reductase. Downregulation of important proteins of cell wall synthesis and division was recorded and a protein with β-lactamase domain (SCLAV_p1007) appeared in 12 isoforms, 5 of which were drastically overrepresented in DEPA strain. These results described herein provide useful information for rational engineering to improve CA production in Streptomyces clavuligerus.
Collapse
Affiliation(s)
- Eser Ünsaldı
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | - Aslıhan Kurt-Kızıldoğan
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey.,Department of Agricultural Biotechnology, Ondokuz Mayıs University, 55139, Samsun, Turkey
| | - Birgit Voigt
- Institute of Microbiology, Ernst-Moritz-Arndt-University of Greifswald, D-17487, Greifswald, Germany
| | - Dörte Becher
- Institute of Microbiology, Ernst-Moritz-Arndt-University of Greifswald, D-17487, Greifswald, Germany
| | - Gülay Özcengiz
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| |
Collapse
|
20
|
Jones JA, Virga KG, Gumina G, Hevener KE. Recent Advances in the Rational Design and Optimization of Antibacterial Agents. MEDCHEMCOMM 2016; 7:1694-1715. [PMID: 27642504 DOI: 10.1039/c6md00232c] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review discusses next-generation antibacterial agents developed using rational, or targeted, drug design strategies. The focus of this review is on small-molecule compounds that have been designed to bypass developing bacterial resistance, improve the antibacterial spectrum of activity, and/or to optimize other properties, including physicochemical and pharmacokinetic properties. Agents are discussed that affect known antibacterial targets, such as the bacterial ribosome, nucleic acid binding proteins, and proteins involved in cell-wall biosynthesis; as well as some affecting novel bacterial targets which do not have currently marketed agents. The discussion of the agents focuses on the rational design strategies employed and the synthetic medicinal chemistry and structure-based design techniques utilized by the scientists involved in the discoveries, including such methods as ligand- and structure-based strategies, structure-activity relationship (SAR) expansion strategies, and novel synthetic organic chemistry methods. As such, the discussion is limited to small-molecule therapeutics that have confirmed macromolecular targets and encompasses only a fraction of all antibacterial agents recently approved or in late-stage clinical trials. The antibacterial agents selected have been recently approved for use on the U.S. or European markets or have shown promising results in phase 2 or phase 3 U.S. CLINICAL TRIALS
Collapse
Affiliation(s)
- Jesse A Jones
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, 1311 E. Central Drive, Meridian, ID 83642-7991 (USA)
| | - Kristopher G Virga
- Department of Pharmaceutical Sciences, Presbyterian College School of Pharmacy, 307 North Broad Street, Clinton, SC 29325 (USA)
| | - Giuseppe Gumina
- Department of Pharmaceutical Sciences, Presbyterian College School of Pharmacy, 307 North Broad Street, Clinton, SC 29325 (USA)
| | - Kirk E Hevener
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, 1311 E. Central Drive, Meridian, ID 83642-7991 (USA)
| |
Collapse
|
21
|
Abstract
The pathways in Escherichia coli and (largely by analogy) S. enterica remain the paradigm of bacterial lipid synthetic pathways, although recently considerable diversity among bacteria in the specific areas of lipid synthesis has been demonstrated. The structural biology of the fatty acid synthetic proteins is essentially complete. However, the membrane-bound enzymes of phospholipid synthesis remain recalcitrant to structural analyses. Recent advances in genetic technology have allowed the essentialgenes of lipid synthesis to be tested with rigor, and as expected most genes are essential under standard growth conditions. Conditionally lethal mutants are available in numerous genes, which facilitates physiological analyses. The array of genetic constructs facilitates analysis of the functions of genes from other organisms. Advances in mass spectroscopy have allowed very accurate and detailed analyses of lipid compositions as well as detection of the interactions of lipid biosynthetic proteins with one another and with proteins outside the lipid pathway. The combination of these advances has resulted in use of E. coli and S. enterica for discovery of new antimicrobials targeted to lipid synthesis and in deciphering the molecular actions of known antimicrobials. Finally,roles for bacterial fatty acids other than as membrane lipid structural components have been uncovered. For example, fatty acid synthesis plays major roles in the synthesis of the essential enzyme cofactors, biotin and lipoic acid. Although other roles for bacterial fatty acids, such as synthesis of acyl-homoserine quorum-sensing molecules, are not native to E. coli introduction of the relevant gene(s) synthesis of these foreign molecules readily proceeds and the sophisticated tools available can used to decipher the mechanisms of synthesis of these molecules.
Collapse
|
22
|
Rajkhowa S, Jha AN, Deka RC. Anti-tubercular drug development: computational strategies to identify potential compounds. J Mol Graph Model 2015; 62:56-68. [PMID: 26386453 DOI: 10.1016/j.jmgm.2015.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/13/2015] [Accepted: 09/05/2015] [Indexed: 10/23/2022]
Abstract
InhA is an attractive target to combat tuberculosis (TB), which is targeted by many pro-drugs (isoniazid, etc.) and drugs such as triclosan. However, triclosan is less useful as an antitubercular drug due to its low bioavailability and therefore, in order to overcome this difficulty, many derivatives of triclosan were prepared. Here, we have combined various computational techniques to virtually screen out four potential triclosan derivatives. Molecular docking methods have been employed to screen out 32 out of 62 triclosan derivatives considering the mode of binding and the top re-rank scores. A comparative study on the chemical properties of triclosan and some of its derivatives has been performed using density functional theory (DFT) calculations. DFT based global reactivity descriptors (GRD), such as hardness, chemical potential, chemical softness, electrophilicity index, Fukui function, and local philicity calculated at the optimized geometries were used to investigate the usefulness of these descriptors for understanding the reactive nature and sites of the molecules. QSAR equations were built using these descriptors considering these 32 compounds. Four common compounds showing the best correlation and the best docking scores were considered for the ADMET property calculations and their dynamical movements have been studied using molecular dynamics simulations. Our results showed that these four compounds are chemically more active than triclosan and have the potential to inhibit the Mycobacterium tuberculosis enoyl acyl carrier protein reductase. This work shows that combination of different computational techniques may help to screen out potential drug candidates from a list of possible ones.
Collapse
Affiliation(s)
- Sanchaita Rajkhowa
- Department of Chemical Sciences, Tezpur University, Napaam, Tezpur 784028, Assam, India
| | - Anupam Nath Jha
- Department of Molecular Biology & Biotechnology, Tezpur University, Napaam, Tezpur 784028, Assam, India
| | - Ramesh Chandra Deka
- Department of Chemical Sciences, Tezpur University, Napaam, Tezpur 784028, Assam, India.
| |
Collapse
|
23
|
Owono Owono LC, Ntie-Kang F, Keita M, Megnassan E, Frecer V, Miertus S. Virtually Designed Triclosan-Based Inhibitors of Enoyl-Acyl Carrier Protein Reductase of Mycobacterium tuberculosis and of Plasmodium falciparum. Mol Inform 2015; 34:292-307. [PMID: 27490275 DOI: 10.1002/minf.201400141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/04/2014] [Indexed: 11/12/2022]
Abstract
We report here new chemical structures of predicted nanomolar triclosan-based inhibitors (TCLs) of Mycobacterium tuberculosis enoyl-acyl carrier protein reductase (InhA) virtually proposed by computer-assisted molecular design. 3D models of InhA-TCL complexes were prepared by in situ modifications of the reference crystal structure (PDB entry 1P45) for a training set of 15 TCLs with known InhA inhibitory activities. A QSAR model was built leading to linear correlation between the calculated free energies of complexation (ΔΔGcom ) and experimental values IC50 (exp) : pIC50 =-0.0657×ΔΔGcom +3.0502, R(2) =0.96. In addition, ligand-based quantitative pharmacophore model (PH4) was built from bound conformations of the training set compounds and confirmed the correlation between molecular models and observed activities: pIC50 (exp=) 0.8929×pIC50 (pre) -0.441, R(2) =0.95. Structural information from both models helped us to propose new TCL analogues. A virtual library of TCLs with known predicted activities against enoyl-acyl carrier protein reductase of Plasmodium falciparum (PfENR) was evaluated, revealing dual target TCLs. Moreover, analysis of binding site interactions suggested enriching substitutions, which led to more potent TCLs with predicted pIC50 (pre) as low as 7 nM. The computational approach, which used both free energy estimated from molecular modeling and 3D-QSAR pharmacophore model, was helpful in virtually proposing the dual-targeted drugs and provided valuable information for the design of novel potential antituberculotic agents.
Collapse
Affiliation(s)
- Luc C Owono Owono
- University of Yaoundé I, Advanced Teacher Training College, Laboratory for Simulation and Molecular Biophysics, P.O. Box 47 Yaoundé, Cameroon.,University of Douala, Centre for Atomic Molecular Physics and Quantum Optics (CEPAMOQ), P.O. Box 8580 Douala, Cameroon.,International Centre for Science and High Technology, UNIDO, AREA Science Park, Padriciano 99, Trieste, I-34012, Italy phone:+22502363008
| | - Fidele Ntie-Kang
- University of Douala, Centre for Atomic Molecular Physics and Quantum Optics (CEPAMOQ), P.O. Box 8580 Douala, Cameroon.,International Centre for Science and High Technology, UNIDO, AREA Science Park, Padriciano 99, Trieste, I-34012, Italy phone:+22502363008.,University of Buea, Chemical and Bioactivity Information Centre, Department of Chemistry, P.O. Box 63, Buea, Cameroon
| | - Melalie Keita
- International Centre for Science and High Technology, UNIDO, AREA Science Park, Padriciano 99, Trieste, I-34012, Italy phone:+22502363008.,University of Abobo Adjamé, UFR SFA, Laboratoire de Physique Fondamentale et Appliquée, 02 BP 801, Abidjan 02, Cote D'Ivoire
| | - Eugene Megnassan
- International Centre for Science and High Technology, UNIDO, AREA Science Park, Padriciano 99, Trieste, I-34012, Italy phone:+22502363008. .,University of Abobo Adjamé, UFR SFA, Laboratoire de Physique Fondamentale et Appliquée, 02 BP 801, Abidjan 02, Cote D'Ivoire.
| | - Vladimir Frecer
- International Centre for Science and High Technology, UNIDO, AREA Science Park, Padriciano 99, Trieste, I-34012, Italy phone:+22502363008.,Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University, SK-83232 Bratislava, Slovakia.,International Centre for Applied Research and Sustainable Technology, SK-84104 Bratislava, Slovakia
| | - Stanislav Miertus
- International Centre for Science and High Technology, UNIDO, AREA Science Park, Padriciano 99, Trieste, I-34012, Italy phone:+22502363008.,International Centre for Applied Research and Sustainable Technology, SK-84104 Bratislava, Slovakia.,Faculty of Natural Sciences, University of Ss. Cyril and Methodius, SK-91701 Trnava, Slovakia
| |
Collapse
|
24
|
Joshi SD, More UA, Koli D, Kulkarni MS, Nadagouda MN, Aminabhavi TM. Synthesis, evaluation and in silico molecular modeling of pyrroyl-1,3,4-thiadiazole inhibitors of InhA. Bioorg Chem 2015; 59:151-67. [DOI: 10.1016/j.bioorg.2015.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 10/23/2022]
|
25
|
Stec J, Vilchèze C, Lun S, Perryman AL, Wang X, Freundlich JS, Bishai W, Jacobs WR, Kozikowski AP. Biological evaluation of potent triclosan-derived inhibitors of the enoyl-acyl carrier protein reductase InhA in drug-sensitive and drug-resistant strains of Mycobacterium tuberculosis. ChemMedChem 2014; 9:2528-37. [PMID: 25165007 PMCID: PMC4213240 DOI: 10.1002/cmdc.201402255] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Indexed: 11/09/2022]
Abstract
New triclosan (TRC) analogues were evaluated for their activity against the enoyl-acyl carrier protein reductase InhA in Mycobacterium tuberculosis (Mtb). TRC is a well-known inhibitor of InhA, and specific modifications to its positions 5 and 4' afforded 27 derivatives; of these compounds, seven derivatives showed improved potency over that of TRC. These analogues were active against both drug-susceptible and drug-resistant Mtb strains. The most active compound in this series, 4-(n-butyl)-1,2,3-triazolyl TRC derivative 3, had an MIC value of 0.6 μg mL(-1) (1.5 μM) against wild-type Mtb. At a concentration equal to its MIC, this compound inhibited purified InhA by 98 %, and showed an IC50 value of 90 nM. Compound 3 and the 5-methylisoxazole-modified TRC 14 were able to inhibit the biosynthesis of mycolic acids. Furthermore, mc(2) 4914, an Mtb strain overexpressing inhA, was found to be less susceptible to compounds 3 and 14, supporting the notion that InhA is the likely molecular target of the TRC derivatives presented herein.
Collapse
Affiliation(s)
- Jozef Stec
- Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States, Fax: +1-312-996-7107
- Department of Pharmaceutical Sciences, College of Pharmacy, Chicago State University, 9501 S. King Drive, Chicago, Illinois 60628, United States
| | - Catherine Vilchèze
- Howard Hughes Medical Institute, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Shichun Lun
- Center for Tuberculosis Research, Department of Medicine, Division of Infectious Disease, Johns Hopkins School of Medicine, Baltimore, MD 21231-1044, United States
| | - Alexander L. Perryman
- Center for Emerging & Re-emerging Pathogens, Division of Infectious, Diseases, Department of Medicine, Rutgers University-New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, United States
| | - Xin Wang
- Center for Emerging & Re-emerging Pathogens, Division of Infectious, Diseases, Department of Medicine, Rutgers University-New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, United States
| | - Joel S. Freundlich
- Center for Emerging & Re-emerging Pathogens, Division of Infectious, Diseases, Department of Medicine, Rutgers University-New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, United States
- Department of Pharmacology and Physiology, Rutgers University-New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, United States
| | - William Bishai
- Center for Tuberculosis Research, Department of Medicine, Division of Infectious Disease, Johns Hopkins School of Medicine, Baltimore, MD 21231-1044, United States
| | - William R. Jacobs
- Howard Hughes Medical Institute, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Alan P. Kozikowski
- Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States, Fax: +1-312-996-7107
| |
Collapse
|
26
|
Punkvang A, Kamsri P, Saparpakorn P, Hannongbua S, Wolschann P, Irle S, Pungpo P. Key Structures and Interactions for Binding of Mycobacterium tuberculosis Protein Kinase B Inhibitors from Molecular Dynamics Simulation. Chem Biol Drug Des 2014; 86:91-101. [PMID: 25354564 DOI: 10.1111/cbdd.12465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 01/03/2023]
Abstract
Substituted aminopyrimidine inhibitors have recently been introduced as antituberculosis agents. These inhibitors show impressive activity against protein kinase B, a Ser/Thr protein kinase that is essential for cell growth of M. tuberculosis. However, up to now, X-ray structures of the protein kinase B enzyme complexes with the substituted aminopyrimidine inhibitors are currently unavailable. Consequently, structural details of their binding modes are questionable, prohibiting the structural-based design of more potent protein kinase B inhibitors in the future. Here, molecular dynamics simulations, in conjunction with molecular mechanics/Poisson-Boltzmann surface area binding free-energy analysis, were employed to gain insight into the complex structures of the protein kinase B inhibitors and their binding energetics. The complex structures obtained by the molecular dynamics simulations show binding free energies in good agreement with experiment. The detailed analysis of molecular dynamics results shows that Glu93, Val95, and Leu17 are key residues responsible to the binding of the protein kinase B inhibitors. The aminopyrazole group and the pyrimidine core are the crucial moieties of substituted aminopyrimidine inhibitors for interaction with the key residues. Our results provide a structural concept that can be used as a guide for the future design of protein kinase B inhibitors with highly increased antagonistic activity.
Collapse
Affiliation(s)
- Auradee Punkvang
- Faculty of Science, Nakhon Phanom University, Muang, Nakhon Phanom, 48000, Thailand
| | - Pharit Kamsri
- Department of Chemistry, Ubon Ratchathani University, 85 Sthollmark Road, Warinchamrap, Ubonratchathani, 34190, Thailand
| | | | - Supa Hannongbua
- Department of Chemistry, Kasetsart University, Chatuchak, Bangkok, 10900, Thailand
| | - Peter Wolschann
- Institute for Theoretical Chemistry, University of Vienna, Vienna, A-1090, Austria.,Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Stephan Irle
- Institute of Transformative Bio-Molecules (WPI-ITbM) and Department of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Pornpan Pungpo
- Department of Chemistry, Ubon Ratchathani University, 85 Sthollmark Road, Warinchamrap, Ubonratchathani, 34190, Thailand
| |
Collapse
|
27
|
Sippel KH, Vyas NK, Zhang W, Sankaran B, Quiocho FA. Crystal structure of the human fatty acid synthase enoyl-acyl carrier protein-reductase domain complexed with triclosan reveals allosteric protein-protein interface inhibition. J Biol Chem 2014; 289:33287-95. [PMID: 25301948 DOI: 10.1074/jbc.m114.608547] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human fatty acid synthase (FAS) is a large, multidomain protein that synthesizes long chain fatty acids. Because these fatty acids are primarily provided by diet, FAS is normally expressed at low levels; however, it is highly up-regulated in many cancers. Human enoyl-acyl carrier protein-reductase (hER) is one of the FAS catalytic domains, and its inhibition by drugs like triclosan (TCL) can increase cytotoxicity and decrease drug resistance in cancer cells. We have determined the structure of hER in the presence and absence of TCL. TCL was not bound in the active site, as predicted, but rather at the protein-protein interface (PPI). TCL binding induces a dimer orientation change that causes downstream structural rearrangement in critical active site residues. Kinetics studies indicate that TCL is capable of inhibiting the isolated hER domain with an IC50 of ∼ 55 μM. Given the hER-TCL structure and the inhibition observed in the hER domain, it seems likely that TCL is observed in the physiologically relevant binding site and that it acts as an allosteric PPI inhibitor. TCL may be a viable scaffold for the development of anti-cancer PPI FAS inhibitors.
Collapse
Affiliation(s)
- Katherine H Sippel
- From the Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030 and
| | - Nand K Vyas
- From the Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030 and
| | - Wei Zhang
- From the Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030 and
| | - Banumathi Sankaran
- the Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Florante A Quiocho
- From the Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030 and
| |
Collapse
|
28
|
Wang SF, Yin Y, Wu X, Qiao F, Sha S, Lv PC, Zhao J, Zhu HL. Synthesis, molecular docking and biological evaluation of coumarin derivatives containing piperazine skeleton as potential antibacterial agents. Bioorg Med Chem 2014; 22:5727-37. [PMID: 25306465 DOI: 10.1016/j.bmc.2014.09.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 09/06/2014] [Accepted: 09/23/2014] [Indexed: 11/24/2022]
Abstract
A series of 4-hydroxycoumarin derivatives were designed and synthesized in order to find some more potent antibacterial drugs. Their antibacterial activities against Escherichia coli, Pseudomonas aeruginosa, Bacillus subtilis and Staphylococcus aureus were tested. These compounds showed good antibacterial activities against Gram-positive strains. Compound 4 g represented the most potent antibacterial activity against Bacillus subtilis and S. aureus with MIC of 0.236, 0.355 μg/mL, respectively. What's more, it showed the most potent activity against SaFabI with IC50 of 0.57 μM. Molecular docking of 4 g into S. aureus Enoyl-ACP-reductase active site were performed to determine the probable binding mode, while the QSAR model was built to check the previous work as well as to introduce new directions.
Collapse
Affiliation(s)
- She-Feng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yong Yin
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Xun Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Fang Qiao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Shao Sha
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Peng-Cheng Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Jing Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China.
| |
Collapse
|
29
|
Elucidating the structural basis of diphenyl ether derivatives as highly potent enoyl-ACP reductase inhibitors through molecular dynamics simulations and 3D-QSAR study. J Mol Model 2014; 20:2319. [PMID: 24935113 DOI: 10.1007/s00894-014-2319-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/26/2014] [Indexed: 10/25/2022]
Abstract
Diphenyl ether derivatives are good candidates for anti-tuberculosis agents that display a promising potency for inhibition of InhA, an essential enoyl-acyl carrier protein (ACP) reductase involved in fatty acid biosynthesis pathways in Mycobacterium tuberculosis. In this work, key structural features for the inhibition were identified by 3D-QSAR CoMSIA models, constructed based on available experimental binding properties of diphenyl ether inhibitors, and a set of four representative compounds was subjected to MD simulations of inhibitor-InhA complexes for the calculation of binding free energies. The results show that bulky groups are required for the R1 substituent on the phenyl A ring of the inhibitors to favor a hydrophobic pocket formed by residues Phe149, Met155, Pro156, Ala157, Tyr158, Pro193, Met199, Val203, Leu207, Ile215, and Leu218. Small substituents with a hydrophilic property are required at the R3 and R4 positions of the inhibitor phenyl B rings to form hydrogen bonds with the backbones of Gly96 and Met98, respectively. For the R2 substituent, small substituents with simultaneous hydrophilic or hydrophobic properties are required to favor the interaction with the pyrophosphate moiety of NAD(+) and the methyl side chain of Ala198, respectively. The reported data provide structural guidance for the design of new and potent diphenyl ether-based inhibitors with high inhibitory activities against M. tuberculosis InhA.
Collapse
|
30
|
Kamsri P, Koohatammakun N, Srisupan A, Meewong P, Punkvang A, Saparpakorn P, Hannongbua S, Wolschann P, Prueksaaroon S, Leartsakulpanich U, Pungpo P. Rational design of InhA inhibitors in the class of diphenyl ether derivatives as potential anti-tubercular agents using molecular dynamics simulations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2014; 25:473-488. [PMID: 24785640 DOI: 10.1080/1062936x.2014.898690] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
A series of diphenyl ether derivatives were developed and showed promising potency for inhibiting InhA, an essential enoyl acyl carrier protein reductase involved in mycolic acid biosynthesis, leading to the lysis of Mycobacterium tuberculosis. To understand the structural basis of diphenyl ether derivatives for designing more potent inhibitors, molecular dynamics (MD) simulations were performed. Based on the obtained results, the dynamic behaviour in terms of flexibility, binding free energy, binding energy decomposition, conformation, and the inhibitor-enzyme interaction of diphenyl ether inhibitors were elucidated. Phe149, Tyr158, Met161, Met199, Val203 and NAD+ are the key residues for binding of diphenyl ether inhibitors in the InhA binding pocket. Our results could provide the structural concept to design new diphenyl ether inhibitors with better enzyme inhibitory activity against M. tuberculosis InhA. The present work facilitates the design of new and potentially more effective anti-tuberculosis agents.
Collapse
Affiliation(s)
- P Kamsri
- a Department of Chemistry, Faculty of Science , Ubon Ratchathani University , Ubonratchathani , Thailand
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Insights into the bonding pattern for characterizing the open and closed state of the substrate-binding loop in Mycobacterium tuberculosis InhA. Future Med Chem 2014; 6:605-16. [DOI: 10.4155/fmc.14.27] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Direct InhA inhibitors, which interact with the substrate-binding loop (SBL) and order it into a closed state, are thought to be potential anti-multidrug-resistant tuberculosis molecules. Thus, developing parameters to distinguish between the open and closed state of SBL can help in screening the potent inhibitors with loop ordering properties. Results: We report empirical parameters to differentiate the 'open' and 'closed' conformation of SBL by comprehensive analysis of InhA crystal structures. The 'open' state of SBL was observed with intra- and inter-loop H-bonding within the residues pair, G205–G208 and L207–I105, respectively, while the 'closed' conformation is found with H-bonding within the residues pair: L207–E210 and A206–I105. Moreover, potent inhibitors (IC50, 5.3–5160 nM) are observed to make hydrophobic interactions with residues of SBL, particularly with A198 in the structures with closed state of SBL. Conclusion: The observed set of H-bonding pattern and hydrophobic contact with residues of SBL can be utilized as a filter to evaluate novel inhibitors for their SBL ordering properties and potencies using the molecular dynamic simulation in the virtual screening of direct InhA inhibitors.
Collapse
|
32
|
Rayasam GV. MmpL3 a potential new target for development of novel anti-tuberculosis drugs. Expert Opin Ther Targets 2013; 18:247-56. [DOI: 10.1517/14728222.2014.859677] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
33
|
Cukier CD, Hope AG, Elamin AA, Moynie L, Schnell R, Schach S, Kneuper H, Singh M, Naismith JH, Lindqvist Y, Gray DW, Schneider G. Discovery of an allosteric inhibitor binding site in 3-Oxo-acyl-ACP reductase from Pseudomonas aeruginosa. ACS Chem Biol 2013; 8:2518-27. [PMID: 24015914 PMCID: PMC3833349 DOI: 10.1021/cb4005063] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
3-Oxo-acyl-acyl carrier protein (ACP) reductase (FabG) plays a key role in the bacterial fatty acid synthesis II system in pathogenic microorganisms, which has been recognized as a potential drug target. FabG catalyzes reduction of a 3-oxo-acyl-ACP intermediate during the elongation cycle of fatty acid biosynthesis. Here, we report gene deletion experiments that support the essentiality of this gene in P. aeruginosa and the identification of a number of small molecule FabG inhibitors with IC50 values in the nanomolar to low micromolar range and good physicochemical properties. Structural characterization of 16 FabG-inhibitor complexes by X-ray crystallography revealed that the compounds bind at a novel allosteric site located at the FabG subunit-subunit interface. Inhibitor binding relies primarily on hydrophobic interactions, but specific hydrogen bonds are also observed. Importantly, the binding cavity is formed upon complex formation and therefore would not be recognized by virtual screening approaches. The structure analysis further reveals that the inhibitors act by inducing conformational changes that propagate to the active site, resulting in a displacement of the catalytic triad and the inability to bind NADPH.
Collapse
Affiliation(s)
- Cyprian D. Cukier
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Ayssar A. Elamin
- LIONEX Diagnostics and Therapeutics GmbH, D-38126 Braunschweig, Germany
| | - Lucile Moynie
- Biomedical
Sciences Research Complex, University of St. Andrews, St. Andrews KY16 9ST, U.K
| | - Robert Schnell
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Susanne Schach
- LIONEX Diagnostics and Therapeutics GmbH, D-38126 Braunschweig, Germany
| | | | - Mahavir Singh
- LIONEX Diagnostics and Therapeutics GmbH, D-38126 Braunschweig, Germany
| | - James H. Naismith
- Biomedical
Sciences Research Complex, University of St. Andrews, St. Andrews KY16 9ST, U.K
| | - Ylva Lindqvist
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Gunter Schneider
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
34
|
Belluti F, Perozzo R, Lauciello L, Colizzi F, Kostrewa D, Bisi A, Gobbi S, Rampa A, Bolognesi ML, Recanatini M, Brun R, Scapozza L, Cavalli A. Design, synthesis, and biological and crystallographic evaluation of novel inhibitors of Plasmodium falciparum enoyl-ACP-reductase (PfFabI). J Med Chem 2013; 56:7516-26. [PMID: 24063369 DOI: 10.1021/jm400637m] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Malaria, a disease of worldwide significance, is responsible for over one million deaths annually. The liver-stage of Plasmodium's life cycle is the first, obligatory, but clinically silent step in malaria infection. The P. falciparum type II fatty acid biosynthesis pathway (PfFAS-II) has been found to be essential for complete liver-stage development and has been regarded as a potential antimalarial target for the development of drugs for malaria prophylaxis and liver-stage eradication. In this paper, new coumarin-based triclosan analogues are reported and their biological profile is explored in terms of inhibitory potency against enzymes of the PfFAS-II pathway. Among the tested compounds, 7 and 8 showed the highest inhibitory potency against Pf enoyl-ACP-reductase (PfFabI), followed by 15 and 3. Finally, we determined the crystal structures of compounds 7 and 11 in complex with PfFabI to identify their mode of binding and to confirm outcomes of docking simulations.
Collapse
Affiliation(s)
- Federica Belluti
- Department of Pharmacy and Biotechnology, University of Bologna , Via Belmeloro 6, I-40126 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Oblak EZ, Wright DL. Highly Substituted Oxabicyclic Derivatives from Furan: Synthesis of (±)-Platensimycin. Org Lett 2011; 13:2263-5. [DOI: 10.1021/ol2005775] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- E. Zachary Oblak
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Dennis L. Wright
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
36
|
Investigating the structural basis of arylamides to improve potency against M. tuberculosis strain through molecular dynamics simulations. Eur J Med Chem 2010; 45:5585-93. [DOI: 10.1016/j.ejmech.2010.09.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 08/13/2010] [Accepted: 09/04/2010] [Indexed: 11/18/2022]
|
37
|
Leung GYC, Li H, Toh QY, Ng AMY, Sum RJ, Bandow JE, Chen DYK. Total Synthesis and Biological Evaluation of the Fab-Inhibitory Antibiotic Platencin and Analogues Thereof. European J Org Chem 2010. [DOI: 10.1002/ejoc.201001281] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
Tipparaju SK, Muench SP, Mui EJ, Ruzheinikov SN, Lu JZ, Hutson SL, Kirisits MJ, Prigge ST, Roberts CW, Henriquez FL, Kozikowski AP, Rice DW, McLeod RL. Identification and development of novel inhibitors of Toxoplasma gondii enoyl reductase. J Med Chem 2010; 53:6287-300. [PMID: 20698542 DOI: 10.1021/jm9017724] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Toxoplasmosis causes significant morbidity and mortality, and yet available medicines are limited by toxicities and hypersensitivity. Because improved medicines are needed urgently, rational approaches were used to identify novel lead compounds effective against Toxoplasma gondii enoyl reductase (TgENR), a type II fatty acid synthase enzyme essential in parasites but not present in animals. Fifty-three compounds, including three classes that inhibit ENRs, were tested. Six compounds have antiparasite MIC(90)s < or = 6 microM without toxicity to host cells, three compounds have IC(90)s < 45 nM against recombinant TgENR, and two protect mice. To further understand the mode of inhibition, the cocrystal structure of one of the most promising candidate compounds in complex with TgENR has been determined to 2.7 A. The crystal structure reveals that the aliphatic side chain of compound 19 occupies, as predicted, space made available by replacement of a bulky hydrophobic residue in homologous bacterial ENRs by Ala in TgENR. This provides a paradigm, conceptual foundation, reagents, and lead compounds for future rational development and discovery of improved inhibitors of T. gondii.
Collapse
Affiliation(s)
- Suresh K Tipparaju
- Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Insight into crucial inhibitor–enzyme interaction of arylamides as novel direct inhibitors of the enoyl ACP reductase (InhA) from Mycobacterium tuberculosis: computer-aided molecular design. MONATSHEFTE FUR CHEMIE 2010. [DOI: 10.1007/s00706-010-0359-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
40
|
Kumar G, Banerjee T, Kapoor N, Surolia N, Surolia A. SAR and pharmacophore models for the rhodanine inhibitors of Plasmodium falciparum enoyl-acyl carrier protein reductase. IUBMB Life 2010; 62:204-13. [DOI: 10.1002/iub.306] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
41
|
Singh AP, Surolia N, Surolia A. Triclosan inhibit the growth of the late liver-stage ofPlasmodium. IUBMB Life 2009; 61:923-8. [DOI: 10.1002/iub.237] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
42
|
Cadieux PA, Chew BH, Nott L, Seney S, Elwood CN, Wignall GR, Goneau LW, Denstedt JD. Use of Triclosan-Eluting Ureteral Stents in Patients with Long-Term Stents. J Endourol 2009; 23:1187-94. [DOI: 10.1089/end.2008.0437] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Peter A. Cadieux
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Surgery/Division of Urology, University of Western Ontario, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Ben H. Chew
- The Stone Centre at Vancouver General Hospital, Department of Urological Sciences, University of British Columbia, Vancouver, Canada
| | - Linda Nott
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Surgery/Division of Urology, University of Western Ontario, London, Ontario, Canada
| | - Shannon Seney
- Lawson Health Research Institute, London, Ontario, Canada
| | | | - Geoffrey R. Wignall
- Department of Surgery/Division of Urology, University of Western Ontario, London, Ontario, Canada
| | - Lee W. Goneau
- Lawson Health Research Institute, London, Ontario, Canada
| | - John D. Denstedt
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Surgery/Division of Urology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
43
|
Nicolaou K, Chen J, Edmonds D, Estrada A. Fortschritte in der Chemie und Biologie natürlicher Antibiotika. Angew Chem Int Ed Engl 2009. [DOI: 10.1002/ange.200801695] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
44
|
Nicolaou KC, Chen JS, Edmonds DJ, Estrada AA. Recent advances in the chemistry and biology of naturally occurring antibiotics. Angew Chem Int Ed Engl 2009; 48:660-719. [PMID: 19130444 PMCID: PMC2730216 DOI: 10.1002/anie.200801695] [Citation(s) in RCA: 184] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Ever since the world-shaping discovery of penicillin, nature's molecular diversity has been extensively screened for new medications and lead compounds in drug discovery. The search for agents intended to combat infectious diseases has been of particular interest and has enjoyed a high degree of success. Indeed, the history of antibiotics is marked with impressive discoveries and drug-development stories, the overwhelming majority of which have their origin in natural products. Chemistry, and in particular chemical synthesis, has played a major role in bringing naturally occurring antibiotics and their derivatives to the clinic, and no doubt these disciplines will continue to be key enabling technologies. In this review article, we highlight a number of recent discoveries and advances in the chemistry, biology, and medicine of naturally occurring antibiotics, with particular emphasis on total synthesis, analogue design, and biological evaluation of molecules with novel mechanisms of action.
Collapse
Affiliation(s)
- K C Nicolaou
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
45
|
Banerjee T, Sharma SK, Surolia N, Surolia A. Epigallocatechin gallate is a slow-tight binding inhibitor of enoyl-ACP reductase from Plasmodium falciparum. Biochem Biophys Res Commun 2008; 377:1238-42. [DOI: 10.1016/j.bbrc.2008.10.135] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 10/28/2008] [Indexed: 01/19/2023]
|
46
|
Wignall GR, Goneau LW, Chew BH, Denstedt JD, Cadieux PA. The Effects of Triclosan on Uropathogen Susceptibility to Clinically Relevant Antibiotics. J Endourol 2008; 22:2349-56. [DOI: 10.1089/end.2008.9705] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Geoffrey R. Wignall
- Department of Surgery (Division of Urology), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Lee W. Goneau
- Lawson Health Research Institute, London, Ontario, Canada
| | - Ben H. Chew
- Department of Urology, University of British Columbia, Vancouver, British Columbia, Canada
| | - John D. Denstedt
- Department of Surgery (Division of Urology), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Peter A. Cadieux
- Department of Surgery (Division of Urology), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
47
|
Tipparaju SK, Mulhearn DC, Klein GM, Chen Y, Tapadar S, Bishop MH, Yang S, Chen J, Ghassemi M, Santarsiero BD, Cook JL, Johlfs M, Mesecar AD, Johnson ME, Kozikowski AP. Design and synthesis of aryl ether inhibitors of the Bacillus anthracis enoyl-ACP reductase. ChemMedChem 2008; 3:1250-68. [PMID: 18663709 PMCID: PMC2693028 DOI: 10.1002/cmdc.200800047] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Indexed: 11/11/2022]
Abstract
The problem of increasing bacterial resistance to the current generation of antibiotics is well documented. Known resistant pathogens such as methicillin-resistant Staphylococcus aureus are becoming more prevalent, while the potential exists for developing drug-resistant pathogens for use as bioweapons, such as Bacillus anthracis. The biphenyl ether antibacterial agent, triclosan, exhibits broad-spectrum activity by targeting the fatty acid biosynthetic pathway through inhibition of enoyl-acyl carrier protein reductase (ENR) and provides a potential scaffold for the development of new, broad-spectrum antibiotics. We used a structure-based approach to develop novel aryl ether analogues of triclosan that target ENR, the product of the fabI gene, from B. anthracis (BaENR). Structure-based design methods were used for the expansion of the compound series including X-ray crystal structure determination, molecular docking, and QSAR methods. Structural modifications were made to both phenyl rings of the 2-phenoxyphenyl core. A number of compounds exhibited improved potency against BaENR and increased efficacy against both the Sterne strain of B. anthracis and the methicillin-resistant strain of S. aureus. X-ray crystal structures of BaENR in complex with triclosan and two other compounds help explain the improved efficacy of the new compounds and suggest future rounds of optimization that might be used to improve their potency.
Collapse
Affiliation(s)
- Suresh K. Tipparaju
- Dr. S. K. Tipparaju, Dr. Y. Chen, Dr. S. Tapadar, Prof. Dr. A. P. Kozikowski, Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA Fax: (312) 413 0577
| | - Debbie C. Mulhearn
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - Gary M. Klein
- Dr. S. K. Tipparaju, Dr. Y. Chen, Dr. S. Tapadar, Prof. Dr. A. P. Kozikowski, Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA Fax: (312) 413 0577
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - Yufeng Chen
- Dr. S. K. Tipparaju, Dr. Y. Chen, Dr. S. Tapadar, Prof. Dr. A. P. Kozikowski, Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA Fax: (312) 413 0577
| | - Subhasish Tapadar
- Dr. S. K. Tipparaju, Dr. Y. Chen, Dr. S. Tapadar, Prof. Dr. A. P. Kozikowski, Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA Fax: (312) 413 0577
| | - Molly H. Bishop
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - Shuo Yang
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - Juan Chen
- Dr. J. Chen, Dr. M. Ghassemi, Dr. J. L. Cook, Department of Medicine, University of Illinois at Chicago, 808 S. Wood St., Chicago IL 60612, USA
| | - Mahmood Ghassemi
- Dr. J. Chen, Dr. M. Ghassemi, Dr. J. L. Cook, Department of Medicine, University of Illinois at Chicago, 808 S. Wood St., Chicago IL 60612, USA
| | - Bernard D. Santarsiero
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - James L. Cook
- Dr. J. Chen, Dr. M. Ghassemi, Dr. J. L. Cook, Department of Medicine, University of Illinois at Chicago, 808 S. Wood St., Chicago IL 60612, USA
| | - Mary Johlfs
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - Andrew D. Mesecar
- Dr. S. K. Tipparaju, Dr. Y. Chen, Dr. S. Tapadar, Prof. Dr. A. P. Kozikowski, Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA Fax: (312) 413 0577
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - Michael E. Johnson
- Dr. D. C. Mulhearn, G. M. Klein, M. H. Bishop, S. Yang, Dr. B. D. Santarsiero, Dr. M. Johlfs, Dr. A. D. Mesecar, Prof. Dr. M. E. Johnson, Center for Pharmaceutical Biotechnology, University of Illinois at Chicago, 900 S. Ashland Ave., Chicago, IL 60607–7173, USA, Fax: (312) 413 9303
| | - Alan P. Kozikowski
- Dr. S. K. Tipparaju, Dr. Y. Chen, Dr. S. Tapadar, Prof. Dr. A. P. Kozikowski, Drug Discovery Program, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA Fax: (312) 413 0577
| |
Collapse
|
48
|
Nicolaou KC, Toh QY, Chen DYK. An Expedient Asymmetric Synthesis of Platencin. J Am Chem Soc 2008; 130:11292-3. [DOI: 10.1021/ja804588r] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- K. C. Nicolaou
- Chemical Synthesis Laboratory@Biopolis, Institute of Chemical and Engineering Sciences (ICES), Agency for Science, Technology and Research (ASTAR), 11 Biopolis Way, The Helios Block, #03-08, Singapore 138667
| | - Qiao-Yan Toh
- Chemical Synthesis Laboratory@Biopolis, Institute of Chemical and Engineering Sciences (ICES), Agency for Science, Technology and Research (ASTAR), 11 Biopolis Way, The Helios Block, #03-08, Singapore 138667
| | - David Y.-K. Chen
- Chemical Synthesis Laboratory@Biopolis, Institute of Chemical and Engineering Sciences (ICES), Agency for Science, Technology and Research (ASTAR), 11 Biopolis Way, The Helios Block, #03-08, Singapore 138667
| |
Collapse
|
49
|
Kumar A, Siddiqi MI. CoMFA based de novo design of Pyrrolidine Carboxamides as Inhibitors of Enoyl Acyl Carrier Protein Reductase from Mycobacterium tuberculosis. J Mol Model 2008; 14:923-35. [DOI: 10.1007/s00894-008-0326-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Accepted: 05/29/2008] [Indexed: 11/29/2022]
|
50
|
Mishra S, Karmodiya K, Parasuraman P, Surolia A, Surolia N. Design, synthesis, and application of novel triclosan prodrugs as potential antimalarial and antibacterial agents. Bioorg Med Chem 2008; 16:5536-46. [DOI: 10.1016/j.bmc.2008.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Accepted: 04/04/2008] [Indexed: 10/22/2022]
|