1
|
Dhameliya TM, Vekariya DD, Bhatt PR, Kachroo T, Virani KD, Patel KR, Bhatt S, Dholakia SP. Synthetic account on indoles and their analogues as potential anti-plasmodial agents. Mol Divers 2025; 29:871-897. [PMID: 38709459 DOI: 10.1007/s11030-024-10842-8] [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: 10/23/2023] [Accepted: 03/07/2024] [Indexed: 05/07/2024]
Abstract
Malaria caused by P. falciparum, has been recognized as one of the major infectious diseases causing the death of several patients as per the reports from the World Health Organization. In search of effective therapeutic agents against malaria, several research groups have started working on the design and development of novel heterocycles as anti-malarial agents. Heterocycles have been recognized as the pharmacophoric features for the different types of medicinally important activities. Among all these heterocycles, nitrogen containing aza-heterocycles should not be underestimated owing to their wide therapeutic window. Amongst the aza-heterocycles, indoles and fused indoles such as marinoquinolines, isocryptolepines and their regioisomers, manzamines, neocryptolenines, and indolones have been recognized as anti-malarial agents active against P. falciparum. The present work unleashes the synthetic attempts of anti-malarial indoles and fused indoles through cyclocondensation, Fischer-indole synthesis, etc. along with the brief discussions on structure-activity relationships, in vitro or in vivo studies for the broader interest of these medicinal chemists, working on their design and development as potential anti-malarial agents.
Collapse
Affiliation(s)
- Tejas M Dhameliya
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India.
- Present Address: Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| | - Drashtiben D Vekariya
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| | - Pooja R Bhatt
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| | - Tarun Kachroo
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| | - Kumkum D Virani
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| | - Khushi R Patel
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| | - Shelly Bhatt
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| | - Sandip P Dholakia
- Department of Pharmaceutical Chemistry and Quality Assurance, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| |
Collapse
|
2
|
Patel H, Nagani A, Patel M, Patel M, Yadav MR. Design, synthesis and biological evaluation of some imidazo[1,2- a]pyridine derivatives as anti-tubercular agents: an in silico - in vitro approach. J Biomol Struct Dyn 2024:1-18. [PMID: 39663643 DOI: 10.1080/07391102.2024.2436554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/14/2024] [Indexed: 12/13/2024]
Abstract
In this study, we designed, synthesized and evaluated some novel imidazo[1,2-a]pyridine derivatives as potential anti-TB agents. Preliminary in vitro screening for anti-TB activity of the synthesized compounds was performed against H37Rv strain using the microplate Alamar Blue assay (MABA). Network pharmacology was used to identify the possible targets and pathways of these compounds against Mtb infection. Molecular docking and molecular dynamics simulations were also performed to investigate the binding modes and stability of these compounds with the selected targets. The results showed that some of the synthesized compounds (6b, 6c, 6e, 6f, 6h, 6i, 6j, 6n and 6o) exhibited potent anti-TB activity, with minimum inhibitory concentrations (MICs) ranging from 1.6 to 6.25 μg/mL. The network pharmacology analysis revealed that among the 455 putative targets of imidazo[1,2-a]pyridine derivatives, 24 targets are the potential targets for treatment of Mtb infection. Among these 24 targets, 10 hub-targets were identified (TLR4, ICAM1, TLR9, STAT3, TNFRSF1A, ERBB2, CXCR3, ACE, IKBKG and NOS2) which were significantly involved in GO processes such as positive regulation of DNA-binding transcription factor activity, peptidyl-tyrosine phosphorylation, positive regulation of inflammatory response, mononuclear cell proliferation, regulation of hemopoiesis and cytokine production involved in inflammatory response and KEGG pathways such as pathways in Tuberculosis, NF-kappa B signalling, HIF-1 signalling PD-L1 expression, and PD-1 checkpoint pathway in cancer. Molecular docking and dynamics simulations confirmed the stable interactions of imidazo[1,2-a]pyridine derivatives with core target active sites, highlighting their potential as novel anti-TB drug candidates.
Collapse
Affiliation(s)
- Harnisha Patel
- Parul Institute of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Afzal Nagani
- Parul Institute of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Mirav Patel
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Research & Development Cell, Parul University, Vadodara, Gujarat, India
| | - Mitesh Patel
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Research & Development Cell, Parul University, Vadodara, Gujarat, India
| | - Mange Ram Yadav
- Research & Development Cell, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
3
|
El-Shoukrofy MS, Atta A, Fahmy S, Sriram D, Shehat MG, Labouta IM, Mahran MA. Challenging the Biginelli scaffold to surpass the first line antitubercular drugs: Mycobacterium tuberculosis thymidine monophosphate kinase (TMPK mt) inhibition activity and molecular modelling studies. J Enzyme Inhib Med Chem 2024; 39:2386668. [PMID: 39258667 PMCID: PMC11391879 DOI: 10.1080/14756366.2024.2386668] [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: 05/06/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 09/12/2024] Open
Abstract
New Biginelli adducts were rationalised, via the introduction of selected anti-tubercular (TB) pharmacophores into the dihydropyrimidine (DHPM) ring of deoxythymidine monophosphate (dTMP), the natural substrate of Mycobacterium tuberculosis thymidine monophosphate kinase (TMPKmt). Repurposing was one of the design rationale strategies for some selected mimics of the designed compounds. The anti-TB activity was screened against the Mtb H37Rv strain where 11a was superior to ethambutol (EMB), and was 9-fold more potent than pyrazinamide (PZA). Additionally, compounds 11b, 4a, 4b, 13a, 13b and 14a elicited higher anti-TB activity than PZA, showing better safety profiles than EMB against RAW 264.7 cells' growth. The in vitro TMPKmt inhibition assay released compounds 11a, 11b and 13b as the most potent inhibitors. Docking studies presumed the binding modes and molecular dynamics (MD) simulation revealed the dynamic stability of 11a-TMPKmt complex over 100 ns. In silico prediction of the chemo-informatics properties of the most active compounds was conducted.
Collapse
Affiliation(s)
- Mai S. El-Shoukrofy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amal Atta
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Salwa Fahmy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Dharmarajan Sriram
- Medicinal Chemistry Research Laboratory, Pharmacy Group, Birla Institute of Technology and Science, Pilani, India
| | - Michael G. Shehat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ibrahim M. Labouta
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mona A. Mahran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
Samoon R, Sau S, Roy A, Parida KK, Sharma K, Yakkala PA, Dewangan RP, Abdin MZ, Kalia NP, Shafi S. Development and Evaluation of Bis-benzothiazoles as a New Class of Benzothiazoles Targeting DprE1 as Antitubercular Agents. ACS Infect Dis 2024; 10:3320-3331. [PMID: 39150887 DOI: 10.1021/acsinfecdis.4c00415] [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] [Indexed: 08/18/2024]
Abstract
Benzothiazole-bearing compounds have emerged as potential noncovalent DprE1 (decaprenylphosphoryl-β-d-ribose-2'-epimerase) inhibitors active against Mycobacterium tuberculosis. Based on structure-based virtual screening (PDB ID: 4KW5), a focused library of thirty-one skeletally diverse benzothiazole amides was prepared, and the compounds were assessed for their antitubercular activity against M.tb H37Ra. Most potent compounds 3b and 3n were further evaluated against the M.tb H37Rv strain by the microdilution assay method. Among the compounds evaluated, bis-benzothiazole amide 3n emerged as a hit molecule and demonstrated promising antitubercular activity with minimum inhibitory concentration (MIC) values of 0.45 μg/mL and 8.0 μg/mL against H37Ra and H37Rv, respectively. Based on the preliminary hit molecule (3n), a focused library of 12 more bis-benzothiazole amide derivatives was further prepared by varying the substituents on either side to obtain new leads and generate a structure-activity relationship (SAR). Among these compounds, 6a, 6c, and 6d demonstrated remarkable antitubercular activity with MIC values of 0.5 μg/mL against H37Ra and 1.0, 2.0, and 8.0 μg/mL against H37Rv, respectively. The most active compound, 6a, also displayed significant efficacy against four drug-resistant tuberculosis strains. Compound 6a was assessed for in vitro cytotoxicity against the HepG2 cell line, and it displayed insignificant cytotoxicity. Furthermore, time-kill kinetic studies demonstrated time- and dose-dependent bactericidal activity of this compound. The GFP release assay revealed that compound 6a targets the inhibition of a cell wall component. SNPs in dprE-1 gene assessment revealed that compound 6a binds to tyrosine at position 314 of DprE1 and replaces it with histidine, causing resistance similar to that of standard TCA1. In silico docking studies further suggest that the strong noncovalent interactions of these compounds may lead to the development of potent noncovalent DprE1 inhibitors.
Collapse
Affiliation(s)
- Rabiya Samoon
- Department of Chemistry, School of Chemical and Life science, Jamia Hamdard, New Delhi 110062, India
| | - Shashikanta Sau
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Arnab Roy
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Kishan Kumar Parida
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab 142001, India
| | - Prasanna Anjaneyulu Yakkala
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Rikeshwer Prasad Dewangan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Malik Zainul Abdin
- Centre for Transgenic Plants, Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard India, New Delhi 110062, India
| | - Nitin Pal Kalia
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Syed Shafi
- Department of Chemistry, School of Chemical and Life science, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
5
|
Khalilzadeh M, Saberi S, Noori G, Vaziri M, Sepehri S, Bakherad H, Esmaeili-Fallah M, Mirzayi S, Farhadi G. Synthesis, biological assessment, and computational investigations of nifedipine and monastrol analogues as anti-leishmanial major and anti-microbial agents. Mol Divers 2023; 27:2555-2575. [PMID: 36417095 DOI: 10.1007/s11030-022-10569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/11/2022] [Indexed: 11/24/2022]
Abstract
Leishmaniasis includes a range of parasitic diseases caused by numerous types of the protozoan kinetoplastid parasite. Fungal and bacterial pathogens have led to infectious illnesses causing some main public health problem in current years. A series of dihydropyridine and tetrahydropyrimidine derivatives having fluoro, bromo, and nitro substituents at para-phenyl ring on C4 of dihydropyridine and tetrahydropyrimidine rings were synthesized. Then, anti-leishmanial and antimicrobial potencies of compounds were assessed. All compounds were synthesized via Hantzsch and Biginelli reactions. All derivatives were evaluated for their anti-leishmanial and antimicrobial activities. Moreover, docking and molecular dynamics simulation calculations of the compounds in PRT1 binding site were performed to report the results of anti-leishmanial and antimicrobial activities. Compounds 4a and 4b showed the highest anti-amastigote and anti-promastigote activities. Compound 4a revealed the highest antimicrobial activity against E. coli, P. aeruginosa, and C. albicans strains. In addition, compound 4c showed the highest activity against S. aureus. The fluoro, bromo, and nitro substituents in para-position of phenyl group at C4 of dihydropyridine and tetrahydropyrimidine moieties as well as the bulk and length of the chain linking to the ester moieties are essential for anti-leishmanial and anti-microbial activities of these derivatives. Low cytotoxicity was shown by most of derivatives against macrophages. The molecular docking studies were in agreement with in vitro assay. Moreover, hydrogen binds, RMSF, RMSD, and Rg, strongly showed the steady binding of 4a and 4b compounds in PRT1 active site.
Collapse
Affiliation(s)
- Mahdieh Khalilzadeh
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sedigheh Saberi
- Department of Mycology and Parasitology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghazal Noori
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mostafa Vaziri
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saghi Sepehri
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hamid Bakherad
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Esmaeili-Fallah
- Department of Mycology and Parasitology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sahar Mirzayi
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghazaleh Farhadi
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
6
|
Shekhar, Alcaraz M, Seboletswe P, Manhas N, Kremer L, Singh P, Kumar V. Tailoring selective triclosan azo-adducts: Design, synthesis, and anti-mycobacterial evaluation. Heliyon 2023; 9:e22182. [PMID: 38034623 PMCID: PMC10685269 DOI: 10.1016/j.heliyon.2023.e22182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
A series of triclosan azo-adducts were synthesized to investigate their structure-activity relationship against Mycobacterium tuberculosis and non-tuberculous mycobacteria. The series' most potent compound was four and sixteen times more active than triclosan and rifabutin against drug-resistant Mycobacterium abscessus, respectively, while being less cytotoxic to human macrophages than triclosan on day one. Additionally, one of the azo-adducts was twice as efficient against M. tuberculosis as triclosan and twice as effective against Mycobacterium marinum as isoniazid. Furthermore, the synthesized azo-adducts were equally effective against M. abscessus strains overexpressing InhA, suggesting that these compounds work through a distinct mechanism.
Collapse
Affiliation(s)
- Shekhar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
| | - Pule Seboletswe
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Neha Manhas
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| | - Parvesh Singh
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
7
|
Dhameliya TM, Vekariya DD, Patel HY, Patel JT. Comprehensive coverage on anti-mycobacterial endeavour reported during 2022. Eur J Med Chem 2023; 255:115409. [PMID: 37120997 DOI: 10.1016/j.ejmech.2023.115409] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 05/02/2023]
Abstract
TB being one of the deadliest diseases and second most common infectious cause of deaths, poses the severe threat to global health. The extended duration of therapy owing to resistance and its upsurge in immune-compromised patients have been the driving force for the development of novel of anti-TB scaffolds. Recently, we have compiled the account of anti-mycobacterial scaffolds published during 2015-2020 and updated them in 2021. The present work involves the insights on the anti-mycobacterial scaffolds reported in 2022 with their mechanism of action, structure activity relationships, along with the key perceptions for the design of newer anti-TB agents for the broader interests of medicinal chemists.
Collapse
Affiliation(s)
- Tejas M Dhameliya
- L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India.
| | | | - Heta Y Patel
- L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| | - Janvi T Patel
- L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India
| |
Collapse
|
8
|
Rathod S, Chavan P, Mahuli D, Rochlani S, Shinde S, Pawar S, Choudhari P, Dhavale R, Mudalkar P, Tamboli F. Exploring biogenic chalcones as DprE1 inhibitors for antitubercular activity via in silico approach. J Mol Model 2023; 29:113. [PMID: 36971900 DOI: 10.1007/s00894-023-05521-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 03/17/2023] [Indexed: 03/28/2023]
Abstract
Cases of drug-resistant tuberculosis (TB) have increased worldwide in the last few years, and it is a major threat to global TB control strategies and the human population. Mycobacterium tuberculosis is a common causative agent responsible for increasing cases of TB and as reported by WHO, approximately, 1.5 million death occurred from TB in 2020. Identification of new therapies against drug-resistant TB is an urgent need to be considered primarily. The current investigation aims to find the potential biogenic chalcone against the potential targets of drug-resistant TB via in silico approach. The ligand library of biogenic chalcones was screened against DprE1. Results of molecular docking and in silico ADMET prediction revealed that ZINC000005158606 has lead-like properties against the targeted protein. Pharmacophore modeling was done to identify the pharmacophoric features and their geometric distance present in ZINC000005158606. The binding stability study performed using molecular dynamics (MD) simulation of the DprE1-ZINC000005158606 complex revealed the conformational stability of the complex system over 100 ns with minimum deviation. Further, the in silico anti-TB sensitivity of ZINC000005158606 was found to be higher as compared to the standards against Mycobacterium tuberculosis. The overall in silico investigation indicated the potential of identified hit to act as a lead molecule against Mycobacterium tuberculosis.
Collapse
|
9
|
Computational design of MmpL3 inhibitors for tuberculosis therapy. Mol Divers 2023; 27:357-369. [PMID: 35477825 DOI: 10.1007/s11030-022-10436-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/07/2022] [Indexed: 02/08/2023]
Abstract
Tuberculosis is a chronic communicable disease caused by Mycobacterium tuberculosis (Mtb) and spreads from lungs to lymphatic system. The cell wall of mycobacterium plays a prominent role in maintaining the virulence and pathogenicity and also acts as prime target for drug discovery. Hence, this study has put into emphasis with target MmpLs (Mycobacterial membrane proteins Large) which are significant for the growth and survival of Mycobacterium tuberculosis. MmpLs belongs to the resistance, nodulation and division (RND) protein superfamily. MmpL3 is the only MmpL deemed essential for the replication and viability of mycobacterial cells. For the study, we have selected SQ109 derivatives as Mmpl3 inhibitor, which holds non-covalent property. Structure-based pharmacophore model of MmpL3 target protein with SQ109 as co-crystallized ligand (PDB: 6AJG) was generated to screen the ligand database. Compounds with decent fitness score and pharmacophoric features were compared with standard drug and taken for molecular docking studies. Further prime molecular mechanics-Poisson-Boltzmann surface area (MM-GBSA) and induced fit calculations identified potential molecules for further drug-likeness screening. Overall computational calculations identified ZINC000000016638 and ZINC000000003594 as potential in silico MmpL3 inhibitors. Molecular dynamics simulations integrated with MM-PBSA free energy calculations identified that MmpL3-ZINC000000016638 complex was more stable. Study can be further extended for synthesis and biological evaluation, derivatization of active compound to identify potential and safe lead compounds for effective tuberculosis therapy.
Collapse
|
10
|
Chudasama SJ, Shah BJ, Patel KM, Dhameliya TM. The spotlight review on ionic liquids catalyzed synthesis of aza- and oxa-heterocycles reported in 2021. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119664] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
11
|
Sureja DK, Shah AP, Gajjar ND, Jadeja SB, Bodiwala KB, Dhameliya TM. In-silico Computational Investigations of AntiViral Lignan Derivatives as Potent Inhibitors of SARS CoV-2. ChemistrySelect 2022; 7:e202202069. [PMID: 35942360 PMCID: PMC9349937 DOI: 10.1002/slct.202202069] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/05/2022] [Indexed: 11/11/2022]
Abstract
Due to alarming outbreak of pandemic COVID-19 in recent times, there is a strong need to discover and identify new antiviral agents acting against SARS CoV-2. Among natural products, lignan derivatives have been found effective against several viral strains including SARS CoV-2. Total of twenty-seven reported antiviral lignan derivatives of plant origin have been selected for computational studies to identify the potent inhibitors of SARS CoV-2. Molecular docking study has been carried out in order to predict and describe molecular interaction between active site of enzyme and lignan derivatives. Out of identified hits, clemastatin B and erythro-strebluslignanol G demonstrated stronger binding and high affinity with all selected proteins. Molecular dynamics simulation studies of clemastin B and savinin against promising targets of SARS CoV-2 have revealed their inhibitory potential against SARS CoV-2. In fine, in-silico computational studies have provided initial breakthrough in design and discovery of potential SARS CoV-2 inhibitors.
Collapse
Affiliation(s)
- Dipen K. Sureja
- Department of Pharmaceutical Chemistry and Quality AssuranceL. M. College of Pharmacy, NavrangpuraAhmedabad380009, GujaratIndia
| | - Ashish P. Shah
- Department of Pharmacy, Sumandeep VidyapeethVadodara391760, GujaratIndia
| | - Normi D. Gajjar
- Department of Pharmaceutical Chemistry and Quality AssuranceL. M. College of Pharmacy, NavrangpuraAhmedabad380009, GujaratIndia
| | - Shwetaba B. Jadeja
- Department of Pharmaceutical Chemistry and Quality AssuranceL. M. College of Pharmacy, NavrangpuraAhmedabad380009, GujaratIndia
| | - Kunjan B. Bodiwala
- Department of Pharmaceutical Chemistry and Quality AssuranceL. M. College of Pharmacy, NavrangpuraAhmedabad380009, GujaratIndia
| | - Tejas M. Dhameliya
- Department of Pharmaceutical Chemistry and Quality AssuranceL. M. College of Pharmacy, NavrangpuraAhmedabad380009, GujaratIndia
| |
Collapse
|
12
|
Si A, Landgraf AD, Geden S, Sucheck SJ, Rohde KH. Synthesis and Evaluation of Marine Natural Product-Inspired Meroterpenoids with Selective Activity toward Dormant Mycobacterium tuberculosis. ACS OMEGA 2022; 7:23487-23496. [PMID: 35847331 PMCID: PMC9281309 DOI: 10.1021/acsomega.2c01887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Tuberculosis is a disease caused primarily by the organism Mycobacterium tuberculosis (Mtb), which claims about 1.5 million lives every year. A challenge that impedes the elimination of this pathogen is the ability of Mtb to remain dormant after primary infection, thus creating a reservoir for the disease in the population that reactivates under more ideal conditions. A better understanding of the physiology of dormant Mtb and therapeutics able to kill these phenotypically tolerant bacilli will be critical for completely eradicating Mtb. Our groups are focusing on characterizing the activity of derivatives of the marine natural product (+)-puupehenone (1). Recently, the Rohde group reported that puupehedione (2) and 15-α-methoxypuupehenol (3) exhibit enhanced activity in an in vitro multi-stress dormancy model of Mtb. To optimize the antimycobacterial activity of these terpenoids, novel 15-α-methoxy- and 15-α-acetoxy-puupehenol esters were prepared from (+)-puupehenone (1) accessed through a (+)-sclareolide-derived β-hydroxyl aldehyde. For added diversity, various congeners related to (1) were also prepared from a common borono-sclareolide donor, which resulted in the synthesis of epi-puupehenol and the natural products (+)-chromazonarol and (+)-yahazunol. In total, we generated a library of 24 compounds, of which 14 were found to be active against Mtb, and the most active compounds retained the enhanced activity against dormant Mtb seen in the parent compound. Several of the 15-α-methoxy- and 15-α-acetoxy-puupehenol esters possessed potent activity against actively dividing and dormant Mtb. Intriguingly, the closely related triisobutyl derivative 16 showed similar activity to 1 in actively dividing Mtb but lost about 178-fold activity against dormant Mtb. However, the monopivaloyl compound 13 showed a modest 3- to 4-fold loss in activity in both actively dividing and dormant Mtb relative to the activity of 1 revealing the importance of the free OH at C19 supporting the potential role of quinone methide formation as critical for activity in dormant Mtb. Elucidating important structure-activity relationships and the mechanism of action of this natural product-inspired chemical series may yield insights into vulnerable drug targets in dormant bacilli and new therapeutics to more effectively target dormant Mtb.
Collapse
Affiliation(s)
- Anshupriya Si
- Department
of Chemistry and Biochemistry, University
of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United
States
| | - Alexander D. Landgraf
- Department
of Chemistry and Biochemistry, University
of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United
States
| | - Sandra Geden
- Division
of Immunity and Pathogenesis, Burnett School of Biomedical Sciences,
College of Medicine, University of Central
Florida, Orlando, Florida 32827, United
States
| | - Steven J. Sucheck
- Department
of Chemistry and Biochemistry, University
of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United
States
| | - Kyle H. Rohde
- Division
of Immunity and Pathogenesis, Burnett School of Biomedical Sciences,
College of Medicine, University of Central
Florida, Orlando, Florida 32827, United
States
| |
Collapse
|
13
|
Bhakhar KA, Vaghela PV, Varakala SD, Chudasma SJ, Gajjar ND, Nagar PR, Sriram D, Dhameliya TM. Indole‐2‐carboxamides as New Anti‐Mycobacterial Agents: Design, Synthesis, Biological Evaluation and Molecular Modeling against mmpL3. ChemistrySelect 2022. [DOI: 10.1002/slct.202201813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Kaushikkumar A. Bhakhar
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura Ahmedabad 380009 Gujarat India
| | - Punit V. Vaghela
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura Ahmedabad 380009 Gujarat India
| | - Saiprasad D. Varakala
- Department of Pharmacy Birla Institute of Technology & Science - Pilani Hyderabad Campus, Jawahar Nagar Hyderabad 500 078 India
| | - Shrdhhaba J. Chudasma
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura Ahmedabad 380009 Gujarat India
| | - Normi D. Gajjar
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura Ahmedabad 380009 Gujarat India
| | - Prinsa R. Nagar
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura Ahmedabad 380009 Gujarat India
| | - Dharmarajan Sriram
- Department of Pharmacy Birla Institute of Technology & Science - Pilani Hyderabad Campus, Jawahar Nagar Hyderabad 500 078 India
| | - Tejas M. Dhameliya
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura Ahmedabad 380009 Gujarat India
| |
Collapse
|
14
|
|
15
|
Kumar A, Dhameliya TM, Sharma K, Patel KA, Hirani RV. Environmentally Benign Approaches towards the Synthesis of Quinolines. ChemistrySelect 2022. [DOI: 10.1002/slct.202201059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Asim Kumar
- Amity Institute of Pharmacy Amity University Haryana, Panchgaon, Manesar 122 413 Haryana India
| | - Tejas M. Dhameliya
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura, Ahmedabad 380 009 Gujarat India
| | - Kirti Sharma
- Amity Institute of Pharmacy Amity University Haryana, Panchgaon, Manesar 122 413 Haryana India
| | - Krupa A. Patel
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura, Ahmedabad 380 009 Gujarat India
| | - Rajvi V. Hirani
- Department of Pharmaceutical Chemistry and Quality Assurance L. M. College of Pharmacy, Navrangpura, Ahmedabad 380 009 Gujarat India
| |
Collapse
|
16
|
Dhameliya TM, Devani AA, Patel KA, Shah KC. Comprehensive Coverage on Anti‐mycobacterial Endeavour Reported in 2021. ChemistrySelect 2022. [DOI: 10.1002/slct.202200921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Aanal A. Devani
- L. M. College of Pharmacy, Navrangpura Ahmedabad 380 009 Gujarat India
| | - Krupa A. Patel
- L. M. College of Pharmacy, Navrangpura Ahmedabad 380 009 Gujarat India
| | - Kashvi C. Shah
- L. M. College of Pharmacy, Navrangpura Ahmedabad 380 009 Gujarat India
| |
Collapse
|
17
|
A review on synthetic account of 1,2,4-oxadiazoles as anti-infective agents. Mol Divers 2022; 26:2967-2980. [PMID: 34984590 PMCID: PMC8727175 DOI: 10.1007/s11030-021-10375-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/24/2021] [Indexed: 11/03/2022]
Abstract
Most of the currently marketed drugs consist of heterocyclic scaffolds containing nitrogen and or oxygen as heteroatoms in their structures. Several research groups have synthesized diversely substituted 1,2,4-oxadiazoles as anti-infective agents having anti-bacterial, anti-viral, anti-leishmanial, etc. activities. For the first time, the present review article will provide the coverage of synthetic account of 1,2,4-oxadiazoles as anti-infective agents along with their potential for SAR, activity potential, promising target for mode of action. The efforts have been made to provide the chemical intuitions to the reader to design new chemical entity with potential of anti-infective activity. This review will mark the impact as the valuable, comprehensive and pioneered work along with the library of synthetic strategies for the organic and medicinal chemists for further refinement of 1,2,4-oxadiazole as anti-infective agents.
Collapse
|
18
|
Bhakhar KA, Sureja DK, Dhameliya TM. Synthetic account of indoles in search of potential anti-mycobacterial agents: A review and future insights. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131522] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|