5751
|
Ganguly K, Patel DK, Dutta SD, Lim KT. TEMPO-Cellulose Nanocrystal-Capped Gold Nanoparticles for Colorimetric Detection of Pathogenic DNA. ACS OMEGA 2021; 6:12424-12431. [PMID: 34056393 PMCID: PMC8154114 DOI: 10.1021/acsomega.1c00359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/11/2021] [Indexed: 05/06/2023]
Abstract
Nanocellulose-assisted gold nanoparticles are considered promising materials for developing eco-friendly diagnostic tools for biosensing applications. In this study, we synthesized 2,2,6,6-tetramethylpiperidin-1-piperidinyloxy (TEMPO)-oxidized cellulose nanocrystal (TEMPO-CNC)-capped gold nanoparticles (AuNPs) for the colorimetric detection of unamplified pathogenic DNA oligomers of methicillin-resistant Staphylococcus aureus. The fabricated TEMPO-CNC-AuNPs (TC-AuNPs) were characterized using UV-visible spectroscopy, transmission electron microscopy, atomic force microscopy, and dynamic light scattering. The average diameter of the synthesized AuNPs was approximately 30 nm. The aqueous solution of TC-AuNPs was stable and exhibited an absorption peak at 520 nm. The chemical interaction between TC-AuNPs and the surface charge of the target and non-target DNA determined the colorimetric differences under ionic conditions. A dramatic color change (red → blue) was observed in the TC-AuNP solution with the target DNA under ionic conditions due to the aggregation of AuNPs. However, no observable color change occurred in the TC-AuNP solution with the non-target DNA under similar conditions owing to the better shielding effects of the charged moieties. The colorimetric detection limit of the TC-AuNPs was demonstrated to be as low as 20 fM pathogenic DNA. Therefore, the use of TEMPO-oxidized CNC-capped AuNPs is efficient and straightforward as a biosensor for the colorimetric detection of pathogenic DNA.
Collapse
Affiliation(s)
- Keya Ganguly
- Department of Biosystems
Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Dinesh K. Patel
- Department of Biosystems
Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems
Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems
Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
5752
|
Mehndiratta S, Chen MC, Chao YH, Lee CH, Liou JP, Lai MJ, Lee HY. Effect of 3-subsitution of quinolinehydroxamic acids on selectivity of histone deacetylase isoforms. J Enzyme Inhib Med Chem 2021; 36:74-84. [PMID: 33161799 PMCID: PMC7655065 DOI: 10.1080/14756366.2020.1839446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A series of 3-subsituted quinolinehydroxamic acids has been synthesised and evaluated for their effect on human lung cancer cell line (A549), human colorectal cancer cell line (HCT116) and HDAC isoforms 1, 2, 6, and 8. The results indicated that substitution at C3 of quinoline is favoured for HDAC6 selectivity. Two compounds (25 and 26) were also found to be potent anti-proliferative compounds with IC50 values ranging from 1.29 to 2.13 µM against A549 and HCT116 cells. These compounds displayed remarkable selectivity for HDAC6 over other HDAC isoforms with nanomolar IC50 values. Western blot analysis revealed that compounds of this series activate apoptotic caspase pathway as indicated by cleavage of caspase 3, 8, and 9 and also increase phosphorylated H2AX thus inducing DNA double strand fragmentation in a concentration dependent manner. Flow cytometric analysis also displayed a dose dependent increase of cell population in sub G1 phase.
Collapse
Affiliation(s)
- Samir Mehndiratta
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Mei-Chuan Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei, Taiwan
| | - Yuh-Hsuan Chao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Hsin Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Mei-Jung Lai
- Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
5753
|
Lee W, Zhan X, Palma J, Vestfrid J, Gross Z, Churchill DG. Minding our P-block and Q-bands: paving inroads into main group corrole research to help instil broader potential. Chem Commun (Camb) 2021; 57:4605-4641. [PMID: 33881055 DOI: 10.1039/d1cc00105a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Main group chemistry is often considered less "dynamic" than transition metal (TM) chemistry because of predictable VSEPR-based central atom geometries, relatively slower redox switching and lack of electronic d-d transitions. However, we delineate what has been made possible with main group chemistry to give it its proper due and up-to-date treatment. The huge untapped potential regarding photophysical properties and functioning hereby spurred us to review a range of corrole reports addressing primarily photophysical trends, synthetic aspects, and important guidelines regarding substitution and inorganic principles. We also look at Ag and Au systems and also consider substitutions such as CF3, halogens, additives and also counterions. Throughout, as well as at the end of this review, we suggest various future directions; further future industrial catalytic and health science research is encouraged.
Collapse
Affiliation(s)
- Woohyun Lee
- Korea Advanced Institute of Science and Technology (KAIST), Department of Chemistry, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Xuan Zhan
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| | - Jaymee Palma
- Korea Advanced Institute of Science and Technology (KAIST), Department of Chemistry, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Jenya Vestfrid
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa 32000, Israel. and Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S3E5, Canada.
| | - Zeev Gross
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| | - David G Churchill
- Korea Advanced Institute of Science and Technology (KAIST), Department of Chemistry, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea. and Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS), Daejeon, Republic of Korea and KAIST Institute for Health Science and Technology (KIHST) (Therapeutic Bioengineering Section), Daejeon 34141, Republic of Korea
| |
Collapse
|
5754
|
Mack N, Mazzio E, Badisa R, Soliman KFA. Metabolic Response to the Mitochondrial Toxin 1-Methyl-4-phenylpyridinium (MPP+) in LDH-A/B Double-knockout LS174T Colon Cancer Cells. Cancer Genomics Proteomics 2021; 18:385-405. [PMID: 33994363 DOI: 10.21873/cgp.20267] [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: 02/15/2021] [Revised: 03/15/2021] [Accepted: 04/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Rapid glycolytic substrate-level phosphorylation (SLP) and accumulation of lactic acid are characteristics of diverse cancers. Recent advances in drug discovery have included the use of glycolytic inhibitors with mitochondrial targeting drugs to attempt to invoke an energy crisis in aggressive metabolically active chemo-resistant cancers. In this work, we examine the consequences of inhibiting mitochondrial oxidative phosphorylation (OXPHOS) with 1-methyl-4-phenylpyridinium (MPP+) in LS14T colon cancer cells containing a genetic double knock out (DKO) of lactic acid dehydrogenase (LDHA and LDHB). MATERIALS AND METHODS Several metabolic parameters were evaluated concomitant to whole transcriptomic (WT) mRNA, microRNA, and long intergenic non-coding RNAs using Affymetrix 2.1 human ST arrays. RESULTS MPP+ effectively blocked OXPHOS where a compensatory shift toward anaerobic SLP was only observed in the control vector (CV), and not observed in the LDH-A/B DKOs (lacking the ability to produce lactic acid). Despite this, there was an unexpected resilience to MPP+ in the latter in terms of energy, which displayed significantly higher resting baseline respiratory OXPHOS capacity relative to controls. At the transcriptome level, MPP+ invoked 1738 differential expressed genes (DEGs) out of 48,226; LDH-A/B DKO resulted in 855 DEGs while 349 DEGs were found to be overlapping in both groups versus respective controls, including loss of mitochondrial complex I (subunits 3 and 6), cell cycle transcripts and fluctuations in epigenetic chromatin remodeling systems. In terms of energy, the effects of MPP+ in the CV transcripts reflect the funneling of carbon intermediates toward glycolysis. The LDH-A/B DKO transcripts reflect a flow of carbons away from glycolysis toward the production of acetyl-CoA. CONCLUSION The findings from this study suggest a metabolic resilience to MPP+ in cancer cells devoid of LDH-A/B, explainable in-part by higher baseline OXPHOS respiratory ATP production, necessitating more toxin to suppress the electron transport chain.
Collapse
Affiliation(s)
- Nzinga Mack
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Elizabeth Mazzio
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Ramesh Badisa
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Karam F A Soliman
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A.
| |
Collapse
|
5755
|
Simurova NV, Maiboroda OI. Antiviral activity of 1,2,4-triazole derivatives (microreview). Chem Heterocycl Compd (N Y) 2021; 57:420-422. [PMID: 34007087 PMCID: PMC8118685 DOI: 10.1007/s10593-021-02919-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/05/2021] [Indexed: 11/24/2022]
Abstract
The microreview summarizes data published since 2015 on the antiviral properties and synthesis of compounds containing the 1,2,4-triazole ring.
Collapse
Affiliation(s)
- Nataliia V. Simurova
- National University of Food Technologies, 68 Volodymyrska St, Kyiv, 01601 Ukraine
| | - Olena I. Maiboroda
- National University of Food Technologies, 68 Volodymyrska St, Kyiv, 01601 Ukraine
| |
Collapse
|
5756
|
Kataev VE, Garifullin BF. Antiviral nucleoside analogs. Chem Heterocycl Compd (N Y) 2021; 57:326-341. [PMID: 34007086 PMCID: PMC8118684 DOI: 10.1007/s10593-021-02912-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
The minireview surveys the modification of native nucleosides as a result of which huge libraries of nucleoside analogs of various structures were synthesized. Particular attention is paid to the synthesis of the so-called prodrug forms of nucleoside analogs which ensure their penetration into the cell and metabolism to active 5'-triphosphate derivatives. All the best known antiviral cyclic nucleoside analogs approved for the treatment of HIV infections, hepatitis B, C, and influenza since the 1960s, as well as those in various stages of clinical trials in recent years, are listed. Nucleoside analogs that have shown the ability to inhibit the replication of SARS-CoV and MERS-CoV are discussed, including remdesivir, approved by the FDA for emergency use in the fight against COVID-19.
Collapse
Affiliation(s)
- Vladimir E. Kataev
- Arbuzov Institute of Organic and Physical Chemistry, Kazan Scientific Center, Russian Academy of Sciences, 8 Akademika Arbuzova St., Kazan, 420088 Tatarstan Russia
| | - Bulat F. Garifullin
- Arbuzov Institute of Organic and Physical Chemistry, Kazan Scientific Center, Russian Academy of Sciences, 8 Akademika Arbuzova St., Kazan, 420088 Tatarstan Russia
| |
Collapse
|
5757
|
Kumar B, Thakur A, Dwivedi AR, Kumar R, Kumar V. Multi-Target-Directed Ligands as an Effective Strategy for the Treatment of Alzheimer's Disease. Curr Med Chem 2021; 29:1757-1803. [PMID: 33982650 DOI: 10.2174/0929867328666210512005508] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder, and multiple pathological factors are believed to be involved in the genesis and progression of the disease. A number of hypotheses, including Acetylcholinesterase, Monoamine oxidase, β-Amyloid, Tau protein, etc., have been proposed for the initiation and progression of the disease. At present, acetylcholine esterase inhibitors and memantine (NMDAR antagonist) are the only approved therapies for the symptomatic management of AD. Most of these single-target drugs have miserably failed in the treatment or halting the progression of the disease. Multi-factorial diseases like AD require complex treatment strategies that involve simultaneous modulation of a network of interacting targets. Since the last few years, Multi-Target-Directed Ligands (MTDLs) strategy, drugs that can simultaneously hit multiple targets, is being explored as an effective therapeutic approach for the treatment of AD. In the current review article, the authors have briefly described various pathogenic pathways associated with AD. The importance of Multi-Target-Directed Ligands and their design strategies in recently reported articles have been discussed in detail. Potent leads are identified through various structure-activity relationship studies, and their drug-like characteristics are described. Recently developed promising compounds have been summarized in the article. Some of these MTDLs with balanced activity profiles against different targets have the potential to be developed as drug candidates for the treatment of AD.
Collapse
Affiliation(s)
- Bhupinder Kumar
- Central University of Punjab Department of Pharmaceutical Sciences and Natural Products, India
| | - Amandeep Thakur
- Central University of Punjab Department of Pharmaceutical Sciences and Natural Products, India
| | | | - Rakesh Kumar
- Central University of Punjab, Bathinda, Punjab-151001, India
| | - Vinod Kumar
- Department of Chemistry, Central University of Punjab, Bathinda, Punjab-151001, India
| |
Collapse
|
5758
|
Discovery of 3-(1H-indol-5-yl)-1,2,4-oxidizable derivatives as non-competitive α-glucosidase inhibitors. CHEMICAL PAPERS 2021. [DOI: 10.1007/s11696-021-01687-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
5759
|
Goyal R, Jerath G, Akhil R, Chandrasekharan A, Puppala ER, Ponneganti S, Sarma A, Naidu VGM, Santhoshkumar TR, Ramakrishnan V. Geometry encoded functional programming of tumor homing peptides for targeted drug delivery. J Control Release 2021; 333:16-27. [PMID: 33722612 DOI: 10.1016/j.jconrel.2021.03.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/24/2021] [Accepted: 03/07/2021] [Indexed: 01/10/2023]
Abstract
Poly-peptide molecules have shown promising applications in drug delivery and tumor targeting. A series of tumor homing peptides were designed by exhaustively sampling low energy geometrical basins of amino acids at specific sites of a peptide molecule to induce a conformational lock. This peptide library was pruned to a limited set of eight molecules, employing electrostatic interactions, docking, and molecular dynamics simulations. These designed and optimized peptides were synthesized and tested on various cell lines, including breast cancer (MDA-MB-231), cervical cancer (HeLa), osteosarcoma (U2-OS), and non-cancerous mammary epithelial cells (MCF-10A) using confocal microscopy and flow cytometry. Peptides show differential uptake in cancerous MDA-MB-231, HeLa, U2-OS, and non-cancerous MCF-10A cells. Confocal imaging verified their ability to penetrate even in 3D tumorospheres of MDA-MB-231 cells. Further, experiments of mitochondrial membrane potential depolarization and Caspase-3 activation confirmed that their cytotoxic effects are by apoptosis. Homing ability of the designed peptides in in vivo system and fluorescence imaging with clinical samples of human origin have further confirmed that the in vitro studies are qualitatively identical and quantitatively comparable in their ability to selectively recognize tumor cells. Overall, we present a roadmap for the functional programming of peptide-based homing and penetrating molecules that can perform selective tumor targeting.
Collapse
Affiliation(s)
- Ruchika Goyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Gaurav Jerath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - R Akhil
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Aneesh Chandrasekharan
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Eswara Rao Puppala
- National Institute of Pharmaceutical Education and Research Guwahati, Guwahati 781101, Assam, India
| | - Srikanth Ponneganti
- National Institute of Pharmaceutical Education and Research Guwahati, Guwahati 781101, Assam, India
| | - Anupam Sarma
- Dr. Bhubaneswar Borooah Cancer Institute, Tata Memorial Centre (Mumbai), Guwahati 781016, Assam, India
| | - V G M Naidu
- National Institute of Pharmaceutical Education and Research Guwahati, Guwahati 781101, Assam, India
| | - T R Santhoshkumar
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.
| | - Vibin Ramakrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
5760
|
Thakur A, Dhiman AK, Sumit, Kumar R, Sharma U. Rh(III)-Catalyzed Regioselective C8-Alkylation of Quinoline N-Oxides with Maleimides and Acrylates. J Org Chem 2021; 86:6612-6621. [PMID: 33881315 DOI: 10.1021/acs.joc.1c00393] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Herein, we disclose the Rh(III)-catalyzed selective C8-alkylation of quinoline N-oxides with maleimides and acrylates. The main features of the reaction include complete C8-selectivity and broad substrate scope with good to excellent yields. The reaction also proceeded well with unprotected maleimide. The applicability of the developed methodology is demonstrated with gram-scale synthesis and post-modification of the alkylated product. Preliminary mechanistic study revealed that the reaction proceeds through a five-membered rhodacycle and involves proto-demetalation step.
Collapse
Affiliation(s)
- Ankita Thakur
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource and Technology, Palampur 176061, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ankit Kumar Dhiman
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource and Technology, Palampur 176061, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumit
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource and Technology, Palampur 176061, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rakesh Kumar
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource and Technology, Palampur 176061, India
| | - Upendra Sharma
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource and Technology, Palampur 176061, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
5761
|
Ropponen HK, Bader CD, Diamanti E, Illarionov B, Rottmann M, Fischer M, Witschel M, Müller R, Hirsch AKH. Search for the Active Ingredients from a 2-Aminothiazole DMSO Stock Solution with Antimalarial Activity. ChemMedChem 2021; 16:2089-2093. [PMID: 33844432 PMCID: PMC8360061 DOI: 10.1002/cmdc.202100067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/17/2021] [Indexed: 12/31/2022]
Abstract
Chemical decomposition of DMSO stock solutions is a common incident that can mislead biological screening campaigns. Here, we share our case study of 2‐aminothiazole 1, originating from an antimalarial class that undergoes chemical decomposition in DMSO at room temperature. As previously measured biological activities observed against Plasmodium falciparum NF54 and for the target enzyme PfIspE were not reproducible for a fresh batch, we tackled the challenge to understand where the activity originated from. Solvent‐ and temperature‐dependent studies using HRMS and NMR spectroscopy to monitor the decomposition led to the isolation and in vitro evaluation of several fractions against PfIspE. After four days of decomposition, we successfully isolated the oxygenated and dimerised compounds using SFC purification and correlated the observed activities to them. Due to the unstable nature of the two isolates, it is likely that they undergo further decomposition contributing to the overall instability of the compound.
Collapse
Affiliation(s)
- Henni-Karoliina Ropponen
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Chantal D Bader
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Eleonora Diamanti
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany.,Helmholtz International Lab for Anti-Infectives, Campus E8.1, 66123, Saarbrücken, Germany
| | - Boris Illarionov
- Hamburg School of Food Science, University of Hamburg, Grindelallee 117, 20146, Hamburg, Germany
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002, Basel, Switzerland.,Universität Basel, Petersplatz 1, 4003, Basel, Switzerland
| | - Markus Fischer
- Hamburg School of Food Science, University of Hamburg, Grindelallee 117, 20146, Hamburg, Germany
| | | | - Rolf Müller
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany.,Helmholtz International Lab for Anti-Infectives, Campus E8.1, 66123, Saarbrücken, Germany
| | - Anna K H Hirsch
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany.,Helmholtz International Lab for Anti-Infectives, Campus E8.1, 66123, Saarbrücken, Germany
| |
Collapse
|
5762
|
Battistini L, Bugatti K, Sartori A, Curti C, Zanardi F. RGD Peptide‐Drug Conjugates as Effective Dual Targeting Platforms: Recent Advances. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100240] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Lucia Battistini
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27 A 43124 Parma Italy
| | - Kelly Bugatti
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27 A 43124 Parma Italy
| | - Andrea Sartori
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27 A 43124 Parma Italy
| | - Claudio Curti
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27 A 43124 Parma Italy
| | - Franca Zanardi
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27 A 43124 Parma Italy
| |
Collapse
|
5763
|
Kwon S, Chun HL, Ha HJ, Lee SY, Park HH. Heterogeneous multimeric structure of isocitrate lyase in complex with succinate and itaconate provides novel insights into its inhibitory mechanism. PLoS One 2021; 16:e0251067. [PMID: 33951112 PMCID: PMC8099091 DOI: 10.1371/journal.pone.0251067] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/20/2021] [Indexed: 11/18/2022] Open
Abstract
During the glyoxylate cycle, isocitrate lyases (ICLs) catalyze the lysis of isocitrate to glyoxylate and succinate. Itaconate has been reported to inhibit an ICL from Mycobacterium tuberculosis (tbICL). To elucidate the molecular mechanism of ICL inhibition, we determined the crystal structure of tbICL in complex with itaconate. Unexpectedly, succinate and itaconate were found to bind to the respective active sites in the dimeric form of tbICL. Our structure revealed the active site architecture as an open form, although the substrate and inhibitor were bound to the active sites. Our findings provide novel insights into the conformation of tbICL upon its binding to a substrate or inhibitor, along with molecular details of the inhibitory mechanism of itaconate.
Collapse
Affiliation(s)
- Sunghark Kwon
- Department of Biotechnology, Konkuk University, Chungju, Chungbuk, Republic of Korea
| | - Hye Lin Chun
- College of Pharmacy, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Hyun Ji Ha
- College of Pharmacy, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - So Yeon Lee
- College of Pharmacy, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| |
Collapse
|
5764
|
Kim H, Hwang YS, Kim M, Park SB. Recent advances in the development of covalent inhibitors. RSC Med Chem 2021; 12:1037-1045. [PMID: 34355176 DOI: 10.1039/d1md00068c] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/23/2021] [Indexed: 01/03/2023] Open
Abstract
The use of covalent inhibitors in the field of drug discovery has attracted considerable attention in the 2000s. As a result, more than 50 covalent drugs are currently on the market, and numerous covalent drug candidates are now under development. Therefore, interest in covalent drugs is expected to continue in the future. The purpose of this focused review is to provide an understanding of the development of covalent inhibitors by describing their inherent characteristics, possibilities, and limitations based on their mechanistic differences from noncovalent inhibitors. We also introduce the latest covalent warheads that can be applied to the development of potential covalent inhibitors.
Collapse
Affiliation(s)
- Hyunsoo Kim
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University Seoul 08826 Korea
| | - Yoon Soo Hwang
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University Seoul 08826 Korea
| | - Mingi Kim
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University Seoul 08826 Korea
| | - Seung Bum Park
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University Seoul 08826 Korea .,Department of Biophysics and Chemical Biology, Seoul National University Seoul 08826 Korea.,SPARK Biopharma, Inc. Seoul 08791 Korea
| |
Collapse
|
5765
|
Abstract
Porphyrin derivatives are ubiquitous in nature and have important biological roles, such as in light harvesting, oxygen transport, and catalysis. Owing to their intrinsic π-conjugated structure, porphyrin derivatives exhibit characteristic photophysical and electrochemical properties. In biological systems, porphyrin derivatives are associated with various protein molecules through noncovalent interactions. For example, hemoglobin, which is responsible for oxygen transport in most vertebrates, consists of four subunits of a globular protein with an iron porphyrin derivative prosthetic group. Furthermore, noncovalently arranged porphyrin derivatives are the fundamental chromophores in light-harvesting systems for photosynthesis in plants and algae. These biologically important roles originate from the functional versatility of porphyrin derivatives. Specifically, porphyrins are excellent host compounds, forming coordination complexes with various metal ions that adds functionality to the porphyrin unit, such as redox activity and additional ligand binding at the central metal ion. In addition, porphyrins are useful building blocks for functional supramolecular assemblies because of their flat and symmetrical molecular architectures, and their excellent photophysical properties are typically utilized for the fabrication of bioactive functional materials. In this Account, we summarize our endeavors over the past decade to develop functional materials based on porphyrin derivatives using bioinspired approaches. In the first section, we discuss several synthetic receptors that act as artificial allosteric host systems and can be used for the selective detection of various chemicals, such as cyanide, chloride, and amino acids. In the second section, we introduce multiporphyrin arrays as mimics of natural light-harvesting complexes. The active control of energy transfer processes by additional guest binding and the fabrication of organic photovoltaic devices using porphyrin derivatives are also introduced. In the third section, we introduce several types of porphyrin-based supramolecular assemblies. Through noncovalent interactions such as metal-ligand interaction, hydrogen bonding, and π-π interaction, porphyrin derivatives were constructed as supramolecular polymers with formation of fiber or toroidal assembly. In the last section, the application of porphyrin derivatives for biomedical nanodevice fabrication is introduced. Even though porphyrins were good candidates as photosensitizers for photodynamic therapy, they have limitations for biomedical application owing to aggregation in aqueous media. We suggested ionic dendrimer porphyrins and they showed excellent photodynamic therapy (PDT) efficacy.
Collapse
Affiliation(s)
- Jong Min Park
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kyeong-Im Hong
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hosoowi Lee
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Woo-Dong Jang
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
5766
|
Rudack T, Teuber C, Scherlo M, Güldenhaupt J, Schartner J, Lübben M, Klare J, Gerwert K, Kötting C. The Ras dimer structure. Chem Sci 2021; 12:8178-8189. [PMID: 34194708 PMCID: PMC8208300 DOI: 10.1039/d1sc00957e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/29/2021] [Indexed: 12/31/2022] Open
Abstract
Oncogenic mutated Ras is a key player in cancer, but despite intense and expensive approaches its catalytic center seems undruggable. The Ras dimer interface is a possible alternative drug target. Dimerization at the membrane affects cell growth signal transduction. In vivo studies indicate that preventing dimerization of oncogenic mutated Ras inhibits uncontrolled cell growth. Conventional computational drug-screening approaches require a precise atomic dimer model as input to successfully access drug candidates. However, the proposed dimer structural models are controversial. Here, we provide a clear-cut experimentally validated N-Ras dimer structural model. We incorporated unnatural amino acids into Ras to enable the binding of labels at multiple positions via click chemistry. This labeling allowed the determination of multiple distances of the membrane-bound Ras-dimer measured by fluorescence and electron paramagnetic resonance spectroscopy. In combination with protein-protein docking and biomolecular simulations, we identified key residues for dimerization. Site-directed mutations of these residues prevent dimer formation in our experiments, proving our dimer model to be correct. The presented dimer structure enables computational drug-screening studies exploiting the Ras dimer interface as an alternative drug target.
Collapse
Affiliation(s)
- Till Rudack
- Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr University Bochum 44801 Bochum Germany
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| | - Christian Teuber
- Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr University Bochum 44801 Bochum Germany
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| | - Marvin Scherlo
- Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr University Bochum 44801 Bochum Germany
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| | - Jörn Güldenhaupt
- Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr University Bochum 44801 Bochum Germany
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| | - Jonas Schartner
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| | - Mathias Lübben
- Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr University Bochum 44801 Bochum Germany
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| | - Johann Klare
- Department of Physics, Osnabrück University 49074 Osnabrück Germany
| | - Klaus Gerwert
- Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr University Bochum 44801 Bochum Germany
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| | - Carsten Kötting
- Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr University Bochum 44801 Bochum Germany
- Department of Biophysics, Ruhr University Bochum 44801 Bochum Germany
| |
Collapse
|
5767
|
van der Zouwen AJ, Witte MD. Modular Approaches to Synthesize Activity- and Affinity-Based Chemical Probes. Front Chem 2021; 9:644811. [PMID: 33937194 PMCID: PMC8082414 DOI: 10.3389/fchem.2021.644811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Combinatorial and modular methods to synthesize small molecule modulators of protein activity have proven to be powerful tools in the development of new drug-like molecules. Over the past decade, these methodologies have been adapted toward utilization in the development of activity- and affinity-based chemical probes, as well as in chemoproteomic profiling. In this review, we will discuss how methods like multicomponent reactions, DNA-encoded libraries, phage displays, and others provide new ways to rapidly screen novel chemical probes against proteins of interest.
Collapse
Affiliation(s)
- Antonie J van der Zouwen
- Chemical Biology II, Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| | - Martin D Witte
- Chemical Biology II, Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| |
Collapse
|
5768
|
Raskolupova VI, Popova TV, Zakharova OD, Nikotina AE, Abramova TV, Silnikov VN. Human Serum Albumin Labelling with a New BODIPY Dye Having a Large Stokes Shift. Molecules 2021; 26:2679. [PMID: 34063643 PMCID: PMC8124464 DOI: 10.3390/molecules26092679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 12/02/2022] Open
Abstract
BODIPY dyes are photostable neutral derivatives of 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene. These are widely used as chemosensors, laser materials, and molecular probes. At the same time, BODIPY dyes have small or moderate Stokes shifts like most other fluorophores. Large Stokes shifts are preferred for fluorophores because of higher sensitivity of such probes and sensors. The new boron containing BODIPY dye was designed and synthesized. We succeeded to perform an annulation of pyrrole ring with coumarin heterocyclic system and achieved a remarkable difference in absorption and emission maximum of obtained fluorophore up to 100 nm. This BODIPY dye was equipped with linker arm and was functionalized with a maleimide residue specifically reactive towards thiol groups of proteins. BODIPY residue equipped with a suitable targeting protein core can be used as a suitable imaging probe and agent for Boron Neutron Capture Therapy (BNCT). As the most abundant protein with a variety of physiological functions, human serum albumin (HSA) has been used extensively for the delivery and improvement of therapeutic molecules. Thiolactone chemistry provides a powerful tool to prepare albumin-based multimodal constructions. The released sulfhydryl groups of the homocysteine functional handle in thiolactone modified HSA were labeled with BODIPY dye to prepare a labeled albumin-BODIPY dye conjugate confirmed by MALDI-TOF-MS, UV-vis, and fluorescent emission spectra. Cytotoxicity of the resulting conjugate was investigated. This study is the basis for a novel BODIPY dye-albumin theranostic for BNCT. The results provide further impetus to develop derivatives of HSA for delivery of boron to cancer cells.
Collapse
Affiliation(s)
- Valeria I. Raskolupova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrent’ev Ave, 8, 630090 Novosibirsk, Russia; (V.I.R.); (T.V.P.); (O.D.Z.); (A.E.N.); (V.N.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova St., 2, 630090 Novosibirsk, Russia
| | - Tatyana V. Popova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrent’ev Ave, 8, 630090 Novosibirsk, Russia; (V.I.R.); (T.V.P.); (O.D.Z.); (A.E.N.); (V.N.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova St., 2, 630090 Novosibirsk, Russia
| | - Olga D. Zakharova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrent’ev Ave, 8, 630090 Novosibirsk, Russia; (V.I.R.); (T.V.P.); (O.D.Z.); (A.E.N.); (V.N.S.)
| | - Anastasia E. Nikotina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrent’ev Ave, 8, 630090 Novosibirsk, Russia; (V.I.R.); (T.V.P.); (O.D.Z.); (A.E.N.); (V.N.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova St., 2, 630090 Novosibirsk, Russia
| | - Tatyana V. Abramova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrent’ev Ave, 8, 630090 Novosibirsk, Russia; (V.I.R.); (T.V.P.); (O.D.Z.); (A.E.N.); (V.N.S.)
| | - Vladimir N. Silnikov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrent’ev Ave, 8, 630090 Novosibirsk, Russia; (V.I.R.); (T.V.P.); (O.D.Z.); (A.E.N.); (V.N.S.)
| |
Collapse
|
5769
|
Abreu Diaz AM, Drumeva GO, Laporte P, Alonso Martinez LM, Petrenyov DR, Carrier JF, DaSilva JN. Evaluation of the high affinity [ 18F]fluoropyridine-candesartan in rats for PET imaging of renal AT 1 receptors. Nucl Med Biol 2021; 96-97:41-49. [PMID: 33798796 DOI: 10.1016/j.nucmedbio.2021.03.003] [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: 01/20/2021] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Alterations in the expression of the Angiotensin II type 1 receptors (AT1R) have been demonstrated in the development of several heart and renal diseases. The aim of this study was to evaluate the novel compound [18F]fluoropyridine-candesartan as a PET imaging tracer of AT1R in rat kidneys. METHODS Competition binding assays were carried out with membranes from CHO-K1 cells expressing human AT1R. Binding to plasma proteins was assessed by ultrafiltration. Radiolabeled metabolites in rat plasma and kidneys of control and pretreated animals (candesartan 10 mg/kg or losartan 30 mg/kg) were analyzed by column-switch HPLC. Dynamic PET/CT images of [18F]fluoropyridine-candesartan in male Sprague-Dawley rats were acquired for 60 min at baseline, pre-treatment with the AT1R antagonist losartan (30 mg/kg) or the AT2R antagonist PD123,319 (5 mg/kg). RESULTS Fluoropyridine-candesartan bound with a high affinity for AT1R (Ki = 5.9 ± 1.1 nM), comparable to fluoropyridine-losartan but lower than the parent compound candesartan (Ki = 0.4 ± 0.1 nM). [18F]Fluoropyridine-candesartan bound strongly to plasma proteins (99.3%) and was mainly metabolized to radiolabeled hydrophilic compounds, displaying minimal interference on renal AT1R binding with 82% of unchanged tracer in the kidneys at 20 min post-injection. PET imaging displayed high renal and liver accumulations and slow clearances, with maximum tissue-to-blood ratios of 14 ± 3 and 54 ± 12 in kidney cortex and liver, respectively, at 10 min post-injection. Binding specificity for AT1R was demonstrated with marked reductions in kidney cortex (-84%) and liver (-93%) tissue-to-blood ratios at 20 min post-injection, when blocking with AT1R antagonist losartan (30 mg/kg). No change was observed in kidney cortex of rats pre-treated with AT2R antagonist PD 123,319 (5 mg/kg), confirming binding selectivity for AT1 over AT2 receptors. CONCLUSION High kidney-to-blood ratios and binding selectivity to renal AT1R combined with tracer in vivo stability displaying minimal interference from labeled metabolites support further PET imaging studies with [18F]fluoropyridine-candesartan.
Collapse
Affiliation(s)
- Aida M Abreu Diaz
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Département de pharmacologie et physiologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada; Institute de génie biomédical, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada; Departamento de Radioquímica, Instituto Superior de Tecnologías y Ciencias Aplicadas, Universidad de la Habana, La Habana, Cuba
| | - Gergana O Drumeva
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Département de pharmacologie et physiologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Philippe Laporte
- Institute de génie biomédical, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada; Département de physique, Faculté des arts et des sciences, Université de Montréal, Montréal, Québec, Canada
| | - Luis M Alonso Martinez
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Département de pharmacologie et physiologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada; Institute de génie biomédical, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Daniil R Petrenyov
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Jean-François Carrier
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Institute de génie biomédical, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada; Département de physique, Faculté des arts et des sciences, Université de Montréal, Montréal, Québec, Canada; Département de radiologie, radio-oncologie et médecine nucléaire, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean N DaSilva
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Département de pharmacologie et physiologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada; Institute de génie biomédical, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada; Département de radiologie, radio-oncologie et médecine nucléaire, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
5770
|
Mastering the Gram-negative bacterial barrier - Chemical approaches to increase bacterial bioavailability of antibiotics. Adv Drug Deliv Rev 2021; 172:339-360. [PMID: 33705882 DOI: 10.1016/j.addr.2021.02.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/08/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
To win the battle against resistant, pathogenic bacteria, novel classes of anti-infectives and targets are urgently needed. Bacterial uptake, distribution, metabolic and efflux pathways of antibiotics in Gram-negative bacteria determine what we here refer to as bacterial bioavailability. Understanding these mechanisms from a chemical perspective is essential for anti-infective activity and hence, drug discovery as well as drug delivery. A systematic and critical discussion of in bacterio, in vitro and in silico assays reveals that a sufficiently accurate holistic approach is still missing. We expect new findings based on Gram-negative bacterial bioavailability to guide future anti-infective research.
Collapse
|
5771
|
Shiryaev VA, Skomorohov MY, Leonova MV, Bormotov NI, Serova OA, Shishkina LN, Agafonov AP, Maksyutov RA, Klimochkin YN. Adamantane derivatives as potential inhibitors of p37 major envelope protein and poxvirus reproduction. Design, synthesis and antiviral activity. Eur J Med Chem 2021; 221:113485. [PMID: 33965861 PMCID: PMC9533879 DOI: 10.1016/j.ejmech.2021.113485] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 11/07/2022]
Abstract
Currently, smallpox, caused by the variola virus belonging to the poxvirus family, has been completely eradicated according to the WHO. However, other representatives of poxviruses, such as vaccinia virus, cowpox virus, ectromelia virus, monkeypox virus, mousepox virus and others, remain in the natural environment and can infect both animals and humans. The pathogens of animal diseases, belonging to the category with a high epidemic risk, have already caused several outbreaks among humans, and can, in an unfavorable combination of circumstances, cause not only an epidemic, but also a pandemic. Despite the fact that there are protocols for the treatment of poxvirus infections, the targeted design of new drugs will increase their availability and expand the arsenal of antiviral chemotherapeutic agents. One of the potential targets of poxviruses is the p37 protein, which is a tecovirimat target. This protein is relatively small, has no homologs among proteins of humans and other mammals and is necessary for the replication of viral particles, which makes it attractive target for virtual screening. Using the I-TASSER modelling and molecular dynamics refinement the p37 orthopox virus protein model was obtained and its was confirmed by ramachandran plot analysis and superimposition of the model with the template protein with similar function. A virtual library of adamantane containing compounds was generated and a number of potential inhibitors were chosen from virtual library using molecular docking. Several compounds bearing adamantane moiety were synthesized and their biological activity was tested in vitro on vaccinia, cowpox and mousepox viruses. The new compounds inhibiting vaccinia virus replication with IC50 concentrations between 0.133 and 0.515 μM were found as a result of the research. The applied approach can be useful in the search of new inhibitors of orthopox reproduction. The proposed approach may be suitable for the design of new poxvirus inhibitors containing cage structural moiety.
Collapse
Affiliation(s)
- Vadim A Shiryaev
- Department of Organic Chemistry, Samara State Technical University, 244 Molodogvardeyskaya St., Samara, Samara Region, 443100, Russia.
| | - Michael Yu Skomorohov
- Department of Organic Chemistry, Samara State Technical University, 244 Molodogvardeyskaya St., Samara, Samara Region, 443100, Russia
| | - Marina V Leonova
- Department of Organic Chemistry, Samara State Technical University, 244 Molodogvardeyskaya St., Samara, Samara Region, 443100, Russia
| | - Nikolai I Bormotov
- State Research Centre of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk Region, 630559, Russia
| | - Olga A Serova
- State Research Centre of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk Region, 630559, Russia
| | - Larisa N Shishkina
- State Research Centre of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk Region, 630559, Russia
| | - Alexander P Agafonov
- State Research Centre of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk Region, 630559, Russia
| | - Rinat A Maksyutov
- State Research Centre of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk Region, 630559, Russia
| | - Yuri N Klimochkin
- Department of Organic Chemistry, Samara State Technical University, 244 Molodogvardeyskaya St., Samara, Samara Region, 443100, Russia
| |
Collapse
|
5772
|
Faheem, Karan Kumar B, Venkata Gowri Chandra Sekhar K, Chander S, Kunjiappan S, Murugesan S. 1,2,3,4-Tetrahydroisoquinoline (THIQ) as privileged scaffold for anticancer de novo drug design. Expert Opin Drug Discov 2021; 16:1119-1147. [PMID: 33908322 DOI: 10.1080/17460441.2021.1916464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Cancer is a dreadful disorder that is emerging as one of the leading causes of mortality across the globe. The complex tumor environment, supplemented with drawbacks of the existing drugs, has made it a global health concern. The Tetrahydroisoquinoline (THIQ) ring holds an important position in medicinal chemistry due to its wide range of pharmacological properties. Several THIQ based natural products have been previously explored for their antitumor properties, making it a vital scaffold for anticancer drug design.Areas covered: This review article addresses the potential of THIQ as anticancer agents. Various medicinal chemistry strategies employed for the design and development of THIQ analogs as inhibitors or modulators of relevant anticancer targets have been discussed in detail. Moreover, the common strategies employed for the synthesis of the core scaffold are also highlighted.Expert opinion: Evidently, THIQs have tremendous potential in anticancer drug design. Some of these analogs exhibited potent activity against various cancer molecular targets. However, there are some drawbacks, such as selectivity that need addressing. The synthetic ease for constructing the core scaffold complimented with its reactivity makes it ideal for further structure-activity relationship studies. For these reasons, THIQ is a privileged scaffold for the design and development of novel anticancer agents.
Collapse
Affiliation(s)
- Faheem
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | - Banoth Karan Kumar
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | | | - Subhash Chander
- Amity Institute of Phytomedicine and Phytochemistry, Amity University Uttar Pradesh, Noida, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| |
Collapse
|
5773
|
Rani A, De Leon-Rodriguez LM, Kavianinia I, McGillivray DJ, Williams DE, Brimble MA. Synthesis and characterization of mono S-lipidated peptide hydrogels: a platform for the preparation of reactive oxygen species responsive materials. Org Biomol Chem 2021; 19:3665-3677. [PMID: 33908574 DOI: 10.1039/d1ob00355k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this work we report the synthesis of mono lipidated peptides containing a 3-mercaptopropionate linker in the N-terminus by means of a photoinitiated thiol-ene reaction (S-lipidation). We evaluate the self-assembling and hydrogelation properties of a library of mono S-lipidated peptides containing lipid chains of various lengths and demonstrate that hydrogelation was driven by a balance between the lipid chain's hydrophobicity and the peptide's facial hydrophobicity. We further postulate that a simple calculation using estimated values of log D could be used as a predictor of hydrogelation when designing similar systems. A mono S-lipidated peptide containing a short lipid chain that formed hydrogels was fully characterized and a mechanism for the peptide hydrogelation developed. Finally, we demonstrate that the presence of the thioether group in the mono S-lipidated peptide hydrogels, which is a feature lacking in conventional N-acyl lipidated systems, enables the controlled disassembly of the gel via oxidation to the sulfoxide by reactive oxygen species in accordance with a hydrophobicity-modulated strategy. Thus, we conclude that mono S-lipidated peptide hydrogels constitute a novel and simple tool for the development of tissue engineering and targeted drug delivery applications of diseases with overexpression of reactive oxygen species (e.g. degenerative and metabolic diseases, and cancers).
Collapse
Affiliation(s)
- Aakanksha Rani
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand. and School of Biological Sciences, The University of Auckland, 3A Symonds St., Auckland 1010, New Zealand and MacDiarmid Institute for Advanced Materials and Nanotechnology, New Zealand
| | - Luis M De Leon-Rodriguez
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand.
| | - Iman Kavianinia
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand. and School of Biological Sciences, The University of Auckland, 3A Symonds St., Auckland 1010, New Zealand and MacDiarmid Institute for Advanced Materials and Nanotechnology, New Zealand and Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3A Symonds St., Auckland 1010, New Zealand
| | - Duncan J McGillivray
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand. and School of Biological Sciences, The University of Auckland, 3A Symonds St., Auckland 1010, New Zealand and MacDiarmid Institute for Advanced Materials and Nanotechnology, New Zealand
| | - David E Williams
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand. and School of Biological Sciences, The University of Auckland, 3A Symonds St., Auckland 1010, New Zealand and MacDiarmid Institute for Advanced Materials and Nanotechnology, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand. and School of Biological Sciences, The University of Auckland, 3A Symonds St., Auckland 1010, New Zealand and MacDiarmid Institute for Advanced Materials and Nanotechnology, New Zealand and Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3A Symonds St., Auckland 1010, New Zealand
| |
Collapse
|
5774
|
Chen MH, Wang TJ, Chen LJ, Jiang MY, Wang YJ, Tseng GF, Chen JR. The effects of astaxanthin treatment on a rat model of Alzheimer's disease. Brain Res Bull 2021; 172:151-163. [PMID: 33932491 DOI: 10.1016/j.brainresbull.2021.04.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/03/2021] [Accepted: 04/25/2021] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder characterized by memory loss and dementia, could be a consequence of the abnormalities of cortical milieu, such as oxidative stress, inflammation, and/or accompanied with the aggregation of β-amyloid. The majority of AD patients are sporadic, late-onset AD, which predominantly occurs over 65 years of age. Our results revealed that the ferrous amyloid buthionine (FAB)-infused sporadic AD-like model showed deficits in spatial learning and memory and with apparent loss of choline acetyltransferase (ChAT) expression in medial septal (MS) nucleus. In hippocampal CA1 region, the loss of pyramidal neurons was accompanied with cholinergic fiber loss and neuroinflammatory responses including glial reaction and enhanced expression of inducible nitric oxide synthase (iNOS). Surviving hippocampal CA1 pyramidal neurons showed the reduction of dendritic spines as well. Astaxanthin (ATX), a potent antioxidant, reported to improve the outcome of oxidative-stress-related diseases. The ATX treatment in FAB-infused rats decreased neuroinflammation and restored the ChAT + fibers in hippocampal CA1 region and the ChAT expression in MS nucleus. It also partly recovered the spine loss on hippocampal CA1 pyramidal neurons and ameliorated the behavioral deficits in AD-like rats. From these data, we believed that the ATX can be a potential option for slowing the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Mu-Hsuan Chen
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Tsyr-Jiuan Wang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Li-Jin Chen
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Ming-Ying Jiang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Yueh-Jan Wang
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Guo-Fang Tseng
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan.
| | - Jeng-Rung Chen
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung-Hsing University, Taichung, Taiwan; Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan.
| |
Collapse
|
5775
|
Dextran based amphiphilic self-assembled biopolymeric macromolecule synthesized via RAFT polymerization as indomethacin carrier. Int J Biol Macromol 2021; 183:718-726. [PMID: 33930447 DOI: 10.1016/j.ijbiomac.2021.04.145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/28/2022]
Abstract
This work demonstrates a facile pathway to develop a biopolymer based amphiphilic macromolecule through reversible addition-fragmentation chain transfer (RAFT) polymerization, using dextran (a biopolymer) as starting material. Also, a new hydrophobic monomer [2-methyl-acrylic acid 1-benzyl-1H-[1,2,3] triazol-4-ylmethyl ester (MABTE)] has been synthesized using methacrylic acid via "click" approach. The resultant copolymer displays controlled radical polymerization characteristics: narrow polydispersity (Ð) and controlled molecular weight as obtained through advanced polymer chromatography (APC) analysis. In aqueous solution, the copolymer can proficiently be self-assembled to provide micellar structure, which has been evidenced from field emission scanning electron microscopy (FESEM) and transmission electron microscopy (TEM) analyses. The in-vitro cytotoxicity study illustrates the nontoxic nature of the copolymer up to 100 μg/mL polymer concentration. The copolymer has been found to be worthy as an efficient carrier for the sustained release of hydrophobic drug: Indomethacin (IND).
Collapse
|
5776
|
Amendola G, Ettari R, Previti S, Di Chio C, Messere A, Di Maro S, Hammerschmidt SJ, Zimmer C, Zimmermann RA, Schirmeister T, Zappalà M, Cosconati S. Lead Discovery of SARS-CoV-2 Main Protease Inhibitors through Covalent Docking-Based Virtual Screening. J Chem Inf Model 2021; 61:2062-2073. [PMID: 33784094 PMCID: PMC8029447 DOI: 10.1021/acs.jcim.1c00184] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Indexed: 12/13/2022]
Abstract
During almost all 2020, coronavirus disease 2019 (COVID-19) pandemic has constituted the major risk for the worldwide health and economy, propelling unprecedented efforts to discover drugs for its prevention and cure. At the end of the year, these efforts have culminated with the approval of vaccines by the American Food and Drug Administration (FDA) and the European Medicines Agency (EMA) giving new hope for the future. On the other hand, clinical data underscore the urgent need for effective drugs to treat COVID-19 patients. In this work, we embarked on a virtual screening campaign against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Mpro chymotrypsin-like cysteine protease employing our in-house database of peptide and non-peptide ligands characterized by different types of warheads acting as Michael acceptors. To this end, we employed the AutoDock4 docking software customized to predict the formation of a covalent adduct with the target protein. In vitro verification of the inhibition properties of the most promising candidates allowed us to identify two new lead inhibitors that will deserve further optimization. From the computational point of view, this work demonstrates the predictive power of AutoDock4 and suggests its application for the in silico screening of large chemical libraries of potential covalent binders against the SARS-CoV-2 Mpro enzyme.
Collapse
Affiliation(s)
- Giorgio Amendola
- DiSTABiF, University of Campania Luigi
Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical,
and Environmental Sciences, University of Messina, Viale
Annunziata, Messina 98168, Italy
| | - Santo Previti
- Department of Chemical, Biological, Pharmaceutical,
and Environmental Sciences, University of Messina, Viale
Annunziata, Messina 98168, Italy
| | - Carla Di Chio
- Department of Chemical, Biological, Pharmaceutical,
and Environmental Sciences, University of Messina, Viale
Annunziata, Messina 98168, Italy
| | - Anna Messere
- DiSTABiF, University of Campania Luigi
Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Salvatore Di Maro
- DiSTABiF, University of Campania Luigi
Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Stefan J. Hammerschmidt
- Institute of Pharmaceutical and Biomedical Sciences,
University of Mainz, Staudingerweg 5, Mainz 55128,
Germany
| | - Collin Zimmer
- Institute of Pharmaceutical and Biomedical Sciences,
University of Mainz, Staudingerweg 5, Mainz 55128,
Germany
| | - Robert A. Zimmermann
- Institute of Pharmaceutical and Biomedical Sciences,
University of Mainz, Staudingerweg 5, Mainz 55128,
Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences,
University of Mainz, Staudingerweg 5, Mainz 55128,
Germany
| | - Maria Zappalà
- Department of Chemical, Biological, Pharmaceutical,
and Environmental Sciences, University of Messina, Viale
Annunziata, Messina 98168, Italy
| | - Sandro Cosconati
- DiSTABiF, University of Campania Luigi
Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| |
Collapse
|
5777
|
Design, synthesis, and biological evaluation of novel dual inhibitors targeting lysine specific demethylase 1 (LSD1) and histone deacetylases (HDAC) for treatment of gastric cancer. Eur J Med Chem 2021; 220:113453. [PMID: 33957387 DOI: 10.1016/j.ejmech.2021.113453] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022]
Abstract
LSD1 and HDAC are physical and functional related to each other in various human cancers and simultaneous pharmacological inhibition of LSD1 and HDAC exerts synergistic anti-cancer effects. In this work, a series of novel LSD1/HDAC bifunctional inhibitors with a styrylpyridine skeleton were designed and synthesized based on our previously reported LSD1 inhibitors. The representative compounds 5d and 5m showed potent activity against LSD1 and HDAC at both molecular and cellular level and displayed high selectivity against MAO-A/B. Moreover, compounds 5d and 5m demonstrated potent antiproliferative activities against MGC-803 and HCT-116 cancer cell lines. Notably, compound 5m showed superior in vitro anticancer potency against a panel of gastric cancer cell lines than ORY-1001 and SP-2509 with IC50 values ranging from 0.23 to 1.56 μM. Compounds 5d and 5m significantly modulated the expression of Bcl-2, Bax, Vimentin, ZO-1 and E-cadherin, induced apoptosis, reduced colony formation and suppressed migration in MGC-803 cancer cells. In addition, preliminary absorption, distribution, metabolism, excretion (ADME) studies revealed that compounds 5d and 5m showed acceptable metabolic stability in human liver microsomes with minimal inhibition of cytochrome P450s (CYPs). Those results indicated that compound 5m could be a promising lead compound for further development as a therapeutic agent in gastric cancers via LSD1 and HDAC dual inhibition.
Collapse
|
5778
|
Regulation of Cardiac PKA Signaling by cAMP and Oxidants. Antioxidants (Basel) 2021; 10:antiox10050663. [PMID: 33923287 PMCID: PMC8146537 DOI: 10.3390/antiox10050663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/31/2022] Open
Abstract
Pathologies, such as cancer, inflammatory and cardiac diseases are commonly associated with long-term increased production and release of reactive oxygen species referred to as oxidative stress. Thereby, protein oxidation conveys protein dysfunction and contributes to disease progression. Importantly, trials to scavenge oxidants by systemic antioxidant therapy failed. This observation supports the notion that oxidants are indispensable physiological signaling molecules that induce oxidative post-translational modifications in target proteins. In cardiac myocytes, the main driver of cardiac contractility is the activation of the β-adrenoceptor-signaling cascade leading to increased cellular cAMP production and activation of its main effector, the cAMP-dependent protein kinase (PKA). PKA-mediated phosphorylation of substrate proteins that are involved in excitation-contraction coupling are responsible for the observed positive inotropic and lusitropic effects. PKA-actions are counteracted by cellular protein phosphatases (PP) that dephosphorylate substrate proteins and thus allow the termination of PKA-signaling. Both, kinase and phosphatase are redox-sensitive and susceptible to oxidation on critical cysteine residues. Thereby, oxidation of the regulatory PKA and PP subunits is considered to regulate subcellular kinase and phosphatase localization, while intradisulfide formation of the catalytic subunits negatively impacts on catalytic activity with direct consequences on substrate (de)phosphorylation and cardiac contractile function. This review article attempts to incorporate the current perception of the functionally relevant regulation of cardiac contractility by classical cAMP-dependent signaling with the contribution of oxidant modification.
Collapse
|
5779
|
Electrophilic Natural Products as Drug Discovery Tools. Trends Pharmacol Sci 2021; 42:434-447. [PMID: 33902949 DOI: 10.1016/j.tips.2021.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/22/2022]
Abstract
Electrophilic natural products (ENPs) are a rich source of bioactive molecules with tremendous therapeutic potential. While their synthetic complexity may hinder their direct use as therapeutics, they represent tools for elucidation of suitable molecular targets and serve as inspiration for the design of simplified synthetic counterparts. Here, we review the recent use of various activity-based protein profiling methods to uncover molecular targets of ENPs. Beyond target identification, these examples also showcase further development of synthetic ligands from natural product starting points. Two examples demonstrate how ENPs can progress the emerging fields of targeted protein degradation and molecular glues. Though challenges still remain in the synthesis of ENP-based probes, and in their synthetic simplification, their potential for discovery of novel mechanisms of action makes it well worth the effort.
Collapse
|
5780
|
Kneller D, Phillips G, Weiss KL, Zhang Q, Coates L, Kovalevsky A. Direct Observation of Protonation State Modulation in SARS-CoV-2 Main Protease upon Inhibitor Binding with Neutron Crystallography. J Med Chem 2021; 64:4991-5000. [PMID: 33755450 PMCID: PMC8009097 DOI: 10.1021/acs.jmedchem.1c00058] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Indexed: 02/08/2023]
Abstract
The main protease (3CL Mpro) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, is an essential enzyme for viral replication with no human counterpart, making it an attractive drug target. To date, no small-molecule clinical drugs are available that specifically inhibit SARS-CoV-2 Mpro. To aid rational drug design, we determined a neutron structure of Mpro in complex with the α-ketoamide inhibitor telaprevir at near-physiological (22 °C) temperature. We directly observed protonation states in the inhibitor complex and compared them with those in the ligand-free Mpro, revealing modulation of the active-site protonation states upon telaprevir binding. We suggest that binding of other α-ketoamide covalent inhibitors can lead to the same protonation state changes in the Mpro active site. Thus, by studying the protonation state changes induced by inhibitors, we provide crucial insights to help guide rational drug design, allowing precise tailoring of inhibitors to manipulate the electrostatic environment of SARS-CoV-2 Mpro.
Collapse
Affiliation(s)
- Daniel
W. Kneller
- Neutron
Scattering Division, Oak Ridge National
Laboratory, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831, United States
- National
Virtual Biotechnology Laboratory, US Department of Energy, Washington, D.C. 20585, United States
| | - Gwyndalyn Phillips
- Neutron
Scattering Division, Oak Ridge National
Laboratory, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831, United States
- National
Virtual Biotechnology Laboratory, US Department of Energy, Washington, D.C. 20585, United States
| | - Kevin L. Weiss
- Neutron
Scattering Division, Oak Ridge National
Laboratory, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831, United States
- National
Virtual Biotechnology Laboratory, US Department of Energy, Washington, D.C. 20585, United States
| | - Qiu Zhang
- Neutron
Scattering Division, Oak Ridge National
Laboratory, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831, United States
- National
Virtual Biotechnology Laboratory, US Department of Energy, Washington, D.C. 20585, United States
| | - Leighton Coates
- National
Virtual Biotechnology Laboratory, US Department of Energy, Washington, D.C. 20585, United States
- Second
Target Station, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831, United States
| | - Andrey Kovalevsky
- Neutron
Scattering Division, Oak Ridge National
Laboratory, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831, United States
- National
Virtual Biotechnology Laboratory, US Department of Energy, Washington, D.C. 20585, United States
| |
Collapse
|
5781
|
Serafini M, Cargnin S, Massarotti A, Tron GC, Pirali T, Genazzani AA. What's in a Name? Drug Nomenclature and Medicinal Chemistry Trends using INN Publications. J Med Chem 2021; 64:4410-4429. [PMID: 33847110 PMCID: PMC8154580 DOI: 10.1021/acs.jmedchem.1c00181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 12/13/2022]
Abstract
The World Health Organization assigns international nonproprietary names (INN), also known as common names, to compounds upon request from drug developers. Structures of INNs are publicly available and represent a source, albeit underused, to understand trends in drug research and development. Here, we explain how a common drug name is composed and analyze chemical entities from 2000 to 2021. In the analysis, we describe some changes that intertwine chemical structure, newer therapeutic targets (e.g., kinases), including a significant increase in the use of fluorine and of heterocycles, and some other evolutionary modifications, such as the progressive increase in molecular weight. Alongside these, small signs of change can be spotted, such as the rise in spirocyclic scaffolds and small rings and the emergence of unconventional structural moieties that might forecast the future to come.
Collapse
Affiliation(s)
| | | | | | - Gian Cesare Tron
- Department of Pharmaceutical
Sciences, Università del Piemonte
Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Tracey Pirali
- Department of Pharmaceutical
Sciences, Università del Piemonte
Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical
Sciences, Università del Piemonte
Orientale, Largo Donegani 2, 28100 Novara, Italy
| |
Collapse
|
5782
|
Meng R, Zhang X, Wang H, Zhang D, Zhao X. Different Inductive Effects of Praziquantel Racemate and its Enantiomers on the Enzyme CYP3A4 Mediated by Pregnane X Receptor and its Variants. Curr Drug Metab 2021; 22:232-239. [PMID: 33397228 DOI: 10.2174/1389200221999210104204057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Praziquantel (PZQ), which possesses an asymmetric center, is classified as a pyrazinoisoquinoline and has been the mainstay in the treatment of schistosomiasis since 1980. PZQ undergoes a pronounced first-pass metabolism in the liver through the CYP450 system which could be mediated by nuclear receptors. OBJECTIVE The purpose of this study was to investigate the possible different induction effects of CYP3A4 by PZQ racemate and enantiomers via the pregnane X receptor (PXR) and the effect of PXR polymorphism on the induction potency of PZQs. METHODS The dual-luciferase reporter gene systems constructed in HepG2 cells were used to measure the abilities of PZQs to induce CYP3A4 expression mediated by PXR. The mRNA and protein levels of CYP3A4 were evaluated by polymerase chain reaction (PCR) and western blotting, respectively. RESULTS In HepG2 cells transfected with PXRwt, PXR158, PXR163, PXR370 or PXR403 expression plasmids, PZQ racemate and its enantiomers up-regulated the luciferase activity in a concentration-dependent manner, while reaching saturation after transfected with PXR379 expression plasmids. The mRNA and protein expression of CYP3A4 was effectively activated in PXR-transfected HepG2 cells. The induction ability of CYP3A4 mediated by PXR activation by PZQ racemate and its enantiomers were statistically different between the same PXR group and different PXR groups. CONCLUSION The enantioselective induction effects of PZQs on CYP3A4 were related to the enantioselective activations of PXR by PZQs and were influenced by the PXR gene polymorphism. These findings provide a basis for further understanding the enantiomeric metabolism and the variable efficacy of PZQs.
Collapse
Affiliation(s)
- Ran Meng
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueli Zhang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haina Wang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Danlu Zhang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Zhao
- School of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
5783
|
Down-Regulation of the Proteoglycan Decorin Fills in the Tumor-Promoting Phenotype of Ionizing Radiation-Induced Senescent Human Breast Stromal Fibroblasts. Cancers (Basel) 2021; 13:cancers13081987. [PMID: 33924197 PMCID: PMC8074608 DOI: 10.3390/cancers13081987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Ionizing radiation (a typical remedy for breast cancer) results in the premature senescence of the adjacent to the neoplastic cells stromal fibroblasts. Here, we showed that these senescent fibroblasts are characterized by the down-regulation of the small leucine-rich proteoglycan decorin, a poor prognostic factor for the progression of the disease. Decorin down-regulation is mediated by secreted growth factors in an autocrine and paracrine (due to the interaction with breast cancer cells) manner, with bFGF and VEGF being the key players of this regulation in young and senescent breast stromal fibroblasts. Autophagy activation increases decorin mRNA levels, indicating that impaired autophagy is implicated in the reduction in decorin in this cell model. Decorin down-regulation acts additively to the already tumor-promoting phenotype of ionizing radiation-induced prematurely senescent human stromal fibroblasts, confirming that stromal senescence is a side-effect of radiotherapy that should be taken into account in the design of anticancer treatments. Abstract Down-regulation of the small leucine-rich proteoglycan decorin in the stroma is considered a poor prognostic factor for breast cancer progression. Ionizing radiation, an established treatment for breast cancer, provokes the premature senescence of the adjacent to the tumor stromal fibroblasts. Here, we showed that senescent human breast stromal fibroblasts are characterized by the down-regulation of decorin at the mRNA and protein level, as well as by its decreased deposition in the pericellular extracellular matrix in vitro. Senescence-associated decorin down-regulation is a long-lasting process rather than an immediate response to γ-irradiation. Growth factors were demonstrated to participate in an autocrine manner in decorin down-regulation, with bFGF and VEGF being the critical mediators of the phenomenon. Autophagy inhibition by chloroquine reduced decorin mRNA levels, while autophagy activation using the mTOR inhibitor rapamycin enhanced decorin transcription. Interestingly, the secretome from a series of both untreated and irradiated human breast cancer cell lines with different molecular profiles inhibited decorin expression in young and senescent stromal fibroblasts, which was annulled by SU5402, a bFGF and VEGF inhibitor. The novel phenotypic trait of senescent human breast stromal fibroblasts revealed here is added to their already described cancer-promoting role via the formation of a tumor-permissive environment.
Collapse
|
5784
|
Teresa Borrello M, Benelkebir H, Lee A, Hin Tam C, Shafat M, Rushworth SA, Bowles KM, Douglas L, Duriez PJ, Bailey S, Crabb SJ, Packham G, Ganesan A. Synthesis of Carboxamide-Containing Tranylcypromine Analogues as LSD1 (KDM1A) Inhibitors Targeting Acute Myeloid Leukemia. ChemMedChem 2021; 16:1316-1324. [PMID: 33533576 DOI: 10.1002/cmdc.202000754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/18/2020] [Indexed: 01/14/2023]
Abstract
Lysine-specific demethylase 1 (LSD1/KDM1A) oxidatively removes methyl groups from histone proteins, and its aberrant activity has been correlated with cancers including acute myeloid leukemia (AML). We report a novel series of tranylcypromine analogues with a carboxamide at the 4-position of the aryl ring. These compounds, such as 5 a and 5 b with benzyl and phenethylamide substituents, respectively, had potent sub-micromolar IC50 values for the inhibition of LSD1 as well as cell proliferation in a panel of AML cell lines. The dose-dependent increase in cellular expression levels of H3K4me2, CD86, CD11b and CD14 supported a mechanism involving LSD1 inhibition. The tert-butyl and ethyl carbamate derivatives of these tranylcypromines, although inactive in LSD1 inhibition, were of similar potency in cell-based assays with a more rapid onset of action. This suggests that carbamates can act as metabolically labile tranylcypromine prodrugs with superior pharmacokinetics.
Collapse
Affiliation(s)
| | - Hanae Benelkebir
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Adam Lee
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Chak Hin Tam
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Manar Shafat
- Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, UK
| | | | - Kristian M Bowles
- Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Leon Douglas
- Protein Core Facility and Cancer Sciences, Cancer Research UK Centre and Experimental Cancer Medicines Centre University of Southampton Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Patrick J Duriez
- Protein Core Facility and Cancer Sciences, Cancer Research UK Centre and Experimental Cancer Medicines Centre University of Southampton Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Sarah Bailey
- Protein Core Facility and Cancer Sciences, Cancer Research UK Centre and Experimental Cancer Medicines Centre University of Southampton Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Simon J Crabb
- Protein Core Facility and Cancer Sciences, Cancer Research UK Centre and Experimental Cancer Medicines Centre University of Southampton Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Graham Packham
- Protein Core Facility and Cancer Sciences, Cancer Research UK Centre and Experimental Cancer Medicines Centre University of Southampton Southampton General Hospital, Southampton, SO16 6YD, UK
| | - A Ganesan
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ, UK
| |
Collapse
|
5785
|
Bhagat DS, Chawla PA, Gurnule WB, Shejul SK, Bumbrah GS. An Insight into Synthesis and Anticancer Potential of Thiazole and 4-thiazolidinone Containing Motifs. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825999210101234704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Over the years, the branch of oncology has reached a mature stage, and substantial
development and advancement have been achieved in this dimension of medical science. The
synthesis and isolation of numerous novel anticancer agents of natural and synthetic origins
have been reported. Thiazole and 4-thiazolidinone containing heterocyclic compounds, having
a broad spectrum of pharmaceutical activities, represent a significant class of medicinal
chemistry. Thiazole and 4-thiazolidinone are five-membered unique heterocyclic motifs containing
S and N atoms as an essential core scaffold and have commendable medicinal significance.
Thiazoles and 4-thiazolidinones containing heterocyclic compounds are used as building
blocks for the next generation of pharmaceuticals. Thiazole precursors have been frequently
used due to their capabilities to bind to numerous cancer-specific protein targets.
Suitably, thiazole motifs have a biological suit via inhibition of different signaling pathways involved in cancer
causes. The scientific community has always tried to synthesize novel thiazole-based heterocycles by carrying out
different replacements of functional groups or skeleton around thiazole moiety. Herein, we report the current trend of
research and development in anticancer activities of thiazoles and 4-thiazolidinones containing scaffolds. In the current
study, we have also highlighted some other significant biological properties of thiazole, novel protocols of synthesis
for the synthesis of the new candidates, along with a significant broad spectrum of the anticancer activities of
thiazole containing scaffolds. This study facilitates the development of novel thiazole and 4-thiazolidinone containing
candidates with potent, efficient anticancer activity and less cytotoxic property.
Collapse
Affiliation(s)
- Devidas S. Bhagat
- Department of Forensic Chemistry and Toxicology, Government Institute of Forensic Science, Aurangabad 431 004, (MS), India
| | - Pooja A. Chawla
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Wasudeo B. Gurnule
- Department of Chemistry, Kamla Nehru Mahavidyalaya, Nagpur-440024, (MS), India
| | - Sampada K. Shejul
- Department of Life Science, Vivekanand Arts, Sardar Dalipsingh Commerce and Science College, Aurangabad 431 001, (MS), India
| | - Gurvinder S. Bumbrah
- Department of Chemistry, Biochemistry and Forensic Science, Amity School of Applied Sciences, Amity University, 122413, Haryana, India
| |
Collapse
|
5786
|
Verma SK, Verma R, Kumar KSS, Banjare L, Shaik AB, Bhandare RR, Rakesh KP, Rangappa KS. A key review on oxadiazole analogs as potential methicillin-resistant Staphylococcus aureus (MRSA) activity: Structure-activity relationship studies. Eur J Med Chem 2021; 219:113442. [PMID: 33878562 DOI: 10.1016/j.ejmech.2021.113442] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/22/2021] [Accepted: 04/02/2021] [Indexed: 01/03/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is becoming dangerous to human beings due to easy transmission mode and leading to the difficult-to-treat situation. The rapid resistance development of MRSA to many approved antibiotics is of major concern. There is a lot of scope to develop novel, efficient, specific, and nontoxic drug candidates to fight against MRSA isolates. The interesting molecular structure and adaptable feature of oxadiazole moiety which are bioisosteres of esters and amides, and these functional groups show improved resistance to esterases mediated hydrolytic cleavage, attracting researchers to develop required novel antibiotics based on oxadiazole core. This review summarizes the developments of oxadiazole-containing derivatives as potent antibacterial agents against multidrug-resistant MRSA strains and discussing the structure-activity relationship (SAR) in various directions. The current survey is the highlight of the present scenario of oxadiazole hybrids on MRSA studies, covering articles published from 2011 to 2020. This collective information may become a good platform to plan and develop new oxadiazole-based small molecule growth inhibitors of MRSA with minimal side effects.
Collapse
Affiliation(s)
- Santosh Kumar Verma
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, 719000, Shaanxi, PR China; Shaanxi Key Laboratory of Low Metamorphic Coal Clean Utilization, Yulin University, Yulin, 719000, Shaanxi, PR China
| | - Rameshwari Verma
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, 719000, Shaanxi, PR China; Shaanxi Key Laboratory of Low Metamorphic Coal Clean Utilization, Yulin University, Yulin, 719000, Shaanxi, PR China.
| | | | - Laxmi Banjare
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur, Koni, 495009, Chhattisgarh, India
| | - Afzal B Shaik
- Department of Pharmaceutical Chemistry, Vignan Pharmacy College, Jawaharlal Nehru Technological University, Vadlamudi, 522213, Andhra Pradesh, India
| | - Richie R Bhandare
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates; Centre of Medical and Bio-allied Health Sciences Research, Ajman Uniersity, Ajman, United Arab Emirates
| | - Kadalipura P Rakesh
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, 430073, PR China
| | | |
Collapse
|
5787
|
Du YE, Bae ES, Lim Y, Cho JC, Nam SJ, Shin J, Lee SK, Nam SI, Oh DC. Svalbamides A and B, Pyrrolidinone-Bearing Lipodipeptides from Arctic Paenibacillus sp. Mar Drugs 2021; 19:229. [PMID: 33920625 PMCID: PMC8073366 DOI: 10.3390/md19040229] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 11/23/2022] Open
Abstract
Two new secondary metabolites, svalbamides A (1) and B (2), were isolated from a culture extract of Paenibacillus sp. SVB7 that was isolated from surface sediment from a core (HH17-1085) taken in the Svalbard archipelago in the Arctic Ocean. The combinational analysis of HR-MS and NMR spectroscopic data revealed the structures of 1 and 2 as being lipopeptides bearing 3-amino-2-pyrrolidinone, d-valine, and 3-hydroxy-8-methyldecanoic acid. The absolute configurations of the amino acid residues in svalbamides A and B were determined using the advanced Marfey's method, in which the hydrolysates of 1 and 2 were derivatized with l- and d- forms of 1-fluoro-2,4-dinitrophenyl-5-alanine amide (FDAA). The absolute configurations of 1 and 2 were completely assigned by deducing the stereochemistry of 3-hydroxy-8-methyldecanoic acid based on DP4 calculations. Svalbamides A and B induced quinone reductase activity in Hepa1c1c7 murine hepatoma cells, indicating that they represent chemotypes with a potential for functioning as chemopreventive agents.
Collapse
Affiliation(s)
- Young Eun Du
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (Y.E.D.); (E.S.B.); (J.S.); (S.K.L.)
| | - Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (Y.E.D.); (E.S.B.); (J.S.); (S.K.L.)
| | - Yeonjung Lim
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (Y.L.); (J.-C.C.)
| | - Jang-Cheon Cho
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (Y.L.); (J.-C.C.)
| | - Sang-Jip Nam
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea;
| | - Jongheon Shin
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (Y.E.D.); (E.S.B.); (J.S.); (S.K.L.)
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (Y.E.D.); (E.S.B.); (J.S.); (S.K.L.)
| | - Seung-Il Nam
- Korea Polar Research Institute, Incheon 21990, Korea;
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (Y.E.D.); (E.S.B.); (J.S.); (S.K.L.)
| |
Collapse
|
5788
|
Johe P, Jung S, Endres E, Kersten C, Zimmer C, Ye W, Sönnichsen C, Hellmich UA, Sotriffer C, Schirmeister T, Neuweiler H. Warhead Reactivity Limits the Speed of Inhibition of the Cysteine Protease Rhodesain. ACS Chem Biol 2021; 16:661-670. [PMID: 33719398 DOI: 10.1021/acschembio.0c00911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Viral and parasitic pathogens rely critically on cysteine proteases for host invasion, replication, and infectivity. Their inhibition by synthetic inhibitors, such as vinyl sulfone compounds, has emerged as a promising treatment strategy. However, the individual reaction steps of protease inhibition are not fully understood. Using the trypanosomal cysteine protease rhodesain as a medically relevant target, we design photoinduced electron transfer (PET) fluorescence probes to detect kinetics of binding of reversible and irreversible vinyl sulfones directly in solution. Intriguingly, the irreversible inhibitor, apart from its unlimited residence time in the enzyme, reacts 5 times faster than the reversible one. Results show that the reactivity of the warhead, and not binding of the peptidic recognition unit, limits the rate constant of protease inhibition. The use of a reversible inhibitor decreases the risk of off-target side effects not only by allowing its release from an off-target but also by reducing the rate constant of binding.
Collapse
Affiliation(s)
- Patrick Johe
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128 Mainz, Germany
| | - Sascha Jung
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128 Mainz, Germany
- TU Dortmund University, Chemical Biology, Otto-Hahn-Str. 6, D-44227 Dortmund, Germany
| | - Erik Endres
- Institute for Pharmacy and Food Chemistry, Julius Maximilians University Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Christian Kersten
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128 Mainz, Germany
| | - Collin Zimmer
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128 Mainz, Germany
| | - Weixiang Ye
- Department of Chemistry, Nanobiotechnology, Johannes Gutenberg University Mainz, Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Carsten Sönnichsen
- Department of Chemistry, Nanobiotechnology, Johannes Gutenberg University Mainz, Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Ute A. Hellmich
- Department of Chemistry, Biochemistry, Johannes Gutenberg University Mainz, Johann-Joachim-Becherweg 30, D-55128 Mainz, Germany
- Centre for Biomolecular Magnetic Resonance, Goethe-University Frankfurt, Max von Laue Str. 9, D-60438 Frankfurt, Germany
| | - Christoph Sotriffer
- Institute for Pharmacy and Food Chemistry, Julius Maximilians University Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, D-55128 Mainz, Germany
| | - Hannes Neuweiler
- Institute for Biotechnology & Biophysics, Julius Maximilians University Würzburg, Am Hubland, D-97074 Würzburg, Germany
| |
Collapse
|
5789
|
Jamshaid H, Din FU, Khan GM. Nanotechnology based solutions for anti-leishmanial impediments: a detailed insight. J Nanobiotechnology 2021; 19:106. [PMID: 33858436 PMCID: PMC8051083 DOI: 10.1186/s12951-021-00853-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
As a neglected tropical disease, Leishmaniasis is significantly instigating morbidity and mortality across the globe. Its clinical spectrum varies from ulcerative cutaneous lesions to systemic immersion causing hyperthermic hepato-splenomegaly. Curbing leishmanial parasite is toughly attributable to the myriad obstacles in existing chemotherapy and immunization. Since the 1990s, extensive research has been conducted for ameliorating disease prognosis, by resolving certain obstacles of conventional therapeutics viz. poor efficacy, systemic toxicity, inadequate drug accumulation inside the macrophage, scarce antigenic presentation to body's immune cells, protracted length and cost of the treatment. Mentioned hurdles can be restricted by designing nano-drug delivery system (nano-DDS) of extant anti-leishmanials, phyto-nano-DDS, surface modified-mannosylated and thiolated nano-DDS. Likewise, antigen delivery with co-transportation of suitable adjuvants would be achievable through nano-vaccines. In the past decade, researchers have engineered nano-DDS to improve the safety profile of existing drugs by restricting their release parameters. Polymerically-derived nano-DDS were found as a suitable option for oral delivery as well as SLNs due to pharmacokinetic re-modeling of drugs. Mannosylated nano-DDS have upgraded macrophage internalizing of nanosystem and the entrapped drug, provided with minimal toxicity. Cutaneous Leishmaniasis (CL) was tackling by the utilization of nano-DDS designed for topical delivery including niosomes, liposomes, and transfersomes. Transfersomes, however, appears to be superior for this purpose. The nanotechnology-based solution to prevent parasitic resistance is the use of Thiolated drug-loaded and multiple drugs loaded nano-DDS. These surfaces amended nano-DDS possess augmented IC50 values in comparison to conventional drugs and un-modified nano-DDS. Phyto-nano-DDS, another obscure horizon, have also been evaluated for their anti-leishmanial response, however, more intense assessment is a prerequisite. Impoverished Cytotoxic T-cells response followed by Leishmanial antigen proteins delivery have also been vanquished using nano-adjuvants. The eminence of nano-DDS for curtailment of anti-leishmanial chemotherapy and immunization associated challenges are extensively summed up in this review. This expedited approach is ameliorating the Leishmaniasis management successfully. Alongside, total to partial eradication of this disease can be sought along with associated co-morbidities.
Collapse
Affiliation(s)
- Humzah Jamshaid
- Nanomedicine Research Group, Department of Pharmacy, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Fakhar Ud Din
- Nanomedicine Research Group, Department of Pharmacy, Quaid-I-Azam University, Islamabad, 45320, Pakistan.
| | - Gul Majid Khan
- Nanomedicine Research Group, Department of Pharmacy, Quaid-I-Azam University, Islamabad, 45320, Pakistan.
- Islamia College University, Peshawar, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
5790
|
Bokosi FRB, Beteck RM, Mbaba M, Mtshare TE, Laming D, Hoppe HC, Khanye SD. Design, synthesis and biological evaluation of mono- and bisquinoline methanamine derivatives as potential antiplasmodial agents. Bioorg Med Chem Lett 2021; 38:127855. [PMID: 33609655 DOI: 10.1016/j.bmcl.2021.127855] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 11/29/2022]
Abstract
Several classes of antimalarial drugs are currently available, although issues of toxicity and the emergence of drug resistant malaria parasites have reduced their overall therapeutic efficiency. Quinoline based antiplasmodial drugs have unequivocally been long-established and continue to inspire the design of new antimalarial agents. Herein, a series of mono- and bisquinoline methanamine derivatives were synthesised through sequential steps; Vilsmeier-Haack, reductive amination, and nucleophilic substitution, and obtained in low to excellent yields. The resulting compounds were investigated for in vitro antiplasmodial activity against the 3D7 chloroquine-sensitive strain of Plasmodium falciparum, and compounds 40 and 59 emerged as the most promising with IC50 values of 0.23 and 0.93 µM, respectively. The most promising compounds were also evaluated in silico by molecular docking protocols for binding affinity to the {001} fast-growing face of a hemozoin crystal model.
Collapse
Affiliation(s)
- Fostino R B Bokosi
- Department of Chemistry, Faculty of Science, Rhodes University, Makhanda 6140, South Africa.
| | - Richard M Beteck
- Department of Chemistry, Faculty of Science, Rhodes University, Makhanda 6140, South Africa; Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Mziyanda Mbaba
- Department of Chemistry, Faculty of Science, Rhodes University, Makhanda 6140, South Africa; Department of Chemistry, Faculty of Science, University of Cape Town, Rondebosch 7701, South Africa
| | - Thanduxolo E Mtshare
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa
| | - Dustin Laming
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Makhanda 6140, South Africa
| | - Heinrich C Hoppe
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Makhanda 6140, South Africa; Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Makhanda 6140, South Africa
| | - Setshaba D Khanye
- Department of Chemistry, Faculty of Science, Rhodes University, Makhanda 6140, South Africa; Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Rhodes University, Makhanda 6140, South Africa; Centre for Chemico- and Biomedicinal Research, Rhodes University, Makhanda 6140, South Africa.
| |
Collapse
|
5791
|
Guan I, Williams K, Pan J, Liu X. New Cysteine Covalent Modification Strategies Enable Advancement of Proteome‐wide Selectivity of Kinase Modulators. ASIAN J ORG CHEM 2021. [DOI: 10.1002/ajoc.202100036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ivy Guan
- School of Chemistry The Heart Research Institute The University of Sydney Sydney New South Wales 2006 Australia
| | - Kayla Williams
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Jolyn Pan
- Faculty of Science & Engineering The University of Waikato 124 Hillcrest Road, Hillcrest Hamilton 3216 New Zealand
| | - Xuyu Liu
- School of Chemistry The Heart Research Institute The University of Sydney Sydney New South Wales 2006 Australia
| |
Collapse
|
5792
|
Mottamal M, Kang B, Peng X, Wang G. From Pure Antagonists to Pure Degraders of the Estrogen Receptor: Evolving Strategies for the Same Target. ACS OMEGA 2021; 6:9334-9343. [PMID: 33869913 PMCID: PMC8047716 DOI: 10.1021/acsomega.0c06362] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/19/2021] [Indexed: 05/08/2023]
Abstract
Pure antiestrogens, or selective estrogen receptor degraders (SERDs), have proven to be effective in treating breast cancer that has progressed on tamoxifen and/or aromatase inhibitors. However, the only FDA-approved pure antiestrogen, fulvestrant, is limited in efficacy by its low bioavailability. The search for orally bioavailable SERDs has continued for nearly as long as the clinical history of the injection-only fulvestrant. Oral SERDs that have been developed and tested in patients ranged from nonsteroidal ER binders containing an acrylic acid or amino side chain to bifunctional proteolysis-targeting chimera (PROTAC) pure ER degraders. Structural evolution in the development of oral SERD molecules has been closely associated with quantifiable ER-degrading potency, as seen in the structural comparison analysis of acrylic acid and basic amino side-chain-bearing SERDs. Failure to improve on fulvestrant in the clinical trials by numerous acidic SERDs and early basic SERDs is blamed on tolerability and/or insufficient efficacy, which will likely be overcome by the new-generation basic SERD molecules and PROTAC ER degraders with improved oral bioavailability, low toxicity, and superior efficacy of receptor degradation.
Collapse
|
5793
|
Disch JS, Duffy JM, Lee ECY, Gikunju D, Chan B, Levin B, Monteiro MI, Talcott SA, Lau AC, Zhou F, Kozhushnyan A, Westlund NE, Mullins PB, Yu Y, von Rechenberg M, Zhang J, Arnautova YA, Liu Y, Zhang Y, McRiner AJ, Keefe AD, Kohlmann A, Clark MA, Cuozzo JW, Huguet C, Arora S. Bispecific Estrogen Receptor α Degraders Incorporating Novel Binders Identified Using DNA-Encoded Chemical Library Screening. J Med Chem 2021; 64:5049-5066. [PMID: 33844532 DOI: 10.1021/acs.jmedchem.1c00127] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bispecific degraders (PROTACs) of ERα are expected to be advantageous over current inhibitors of ERα signaling (aromatase inhibitors/SERMs/SERDs) used to treat ER+ breast cancer. Information from DNA-encoded chemical library (DECL) screening provides a method to identify novel PROTAC binding features as the linker positioning, and binding elements are determined directly from the screen. After screening ∼120 billion DNA-encoded molecules with ERα WT and 3 gain-of-function (GOF) mutants, with and without estradiol to identify features that enrich ERα competitively, the off-DNA synthesized small molecule exemplar 7 exhibited nanomolar ERα binding, antagonism, and degradation. Click chemistry synthesis on an alkyne E3 ligase engagers panel and an azide variant of 7 rapidly generated bispecific nanomolar degraders of ERα, with PROTACs 18 and 21 inhibiting ER+ MCF7 tumor growth in a mouse xenograft model of breast cancer. This study validates this approach toward identifying novel bispecific degrader leads from DECL screening with minimal optimization.
Collapse
Affiliation(s)
- Jeremy S Disch
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Jennifer M Duffy
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Esther C Y Lee
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Diana Gikunju
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Betty Chan
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Benjamin Levin
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Michael I Monteiro
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Sarah A Talcott
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Anthony C Lau
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Fei Zhou
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Anton Kozhushnyan
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Neil E Westlund
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Patrick B Mullins
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Yan Yu
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | | | - Junyi Zhang
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Yelena A Arnautova
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Yanbin Liu
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Ying Zhang
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Andrew J McRiner
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Anthony D Keefe
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Anna Kohlmann
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Matthew A Clark
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - John W Cuozzo
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Christelle Huguet
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Shilpi Arora
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| |
Collapse
|
5794
|
Dodero A, Schlatter G, Hébraud A, Vicini S, Castellano M. Polymer-free cyclodextrin and natural polymer-cyclodextrin electrospun nanofibers: A comprehensive review on current applications and future perspectives. Carbohydr Polym 2021; 264:118042. [PMID: 33910745 DOI: 10.1016/j.carbpol.2021.118042] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023]
Abstract
The present review discusses the use of cyclodextrins and their derivatives to prepare electrospun nanofibers with specific features. Cyclodextrins, owing to their unique capability to form inclusion complexes with hydrophobic and volatile molecules, can indeed facilitate the encapsulation of bioactive compounds in electrospun nanofibers allowing fast-dissolving products for food, biomedical, and pharmaceutical purposes, filtering materials for wastewater and air purification, as well as a variety of other technological applications. Additionally, cyclodextrins can improve the processability of naturally occurring biopolymers helping the fabrication of "green" materials with a strong industrial relevance. Hence, this review provides a comprehensive state-of-the-art of different cyclodextrins-based nanofibers including those made of pure cyclodextrins, of polycyclodextrins, and those made of natural biopolymer functionalized with cyclodextrins. To this end, the advantages and disadvantages of such approaches and their possible applications are investigated along with the current limitations in the exploitation of electrospinning at the industrial level.
Collapse
Affiliation(s)
- Andrea Dodero
- Department of Chemistry and Industrial Chemistry, University of Genoa, Via Dodecaneso 31, Genoa, 16146, Italy
| | - Guy Schlatter
- Institute of Chemistry and Processes for Energy, Environment and Health (ICPEES), CNRS UMR 7515, ECPM - University of Strasbourg, 25 Rue Becquerel, Strasbourg, 67087, France.
| | - Anne Hébraud
- Institute of Chemistry and Processes for Energy, Environment and Health (ICPEES), CNRS UMR 7515, ECPM - University of Strasbourg, 25 Rue Becquerel, Strasbourg, 67087, France
| | - Silvia Vicini
- Department of Chemistry and Industrial Chemistry, University of Genoa, Via Dodecaneso 31, Genoa, 16146, Italy
| | - Maila Castellano
- Department of Chemistry and Industrial Chemistry, University of Genoa, Via Dodecaneso 31, Genoa, 16146, Italy.
| |
Collapse
|
5795
|
Li JQ, Gao H, Zhai L, Sun LY, Chen C, Chigan JZ, Ding HH, Yang KW. Dipyridyl-substituted thiosemicarbazone as a potent broad-spectrum inhibitor of metallo-β-lactamases. Bioorg Med Chem 2021; 38:116128. [PMID: 33862468 DOI: 10.1016/j.bmc.2021.116128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 11/17/2022]
Abstract
To combat the superbug infection caused by metallo-β-lactamases (MβLs), a dipyridyl-substituted thiosemicarbazone (DpC), was identified to be the broad-spectrum inhibitor of MβLs (NDM-1, VIM-2, IMP-1, ImiS, L1), with an IC50 value in the range of 0.021-1.08 µM. It reversibly and competitively inhibited NDM-1 with a Ki value of 10.2 nM. DpC showed broad-spectrum antibacterial effect on clinical isolate K. pneumonia, CRE, VRE, CRPA and MRSA, with MIC value ranged from 16 to 32 µg/mL, and exhibited synergistic antibacterial effect with meropenem on MβLs-producing bacteria, resulting in a 2-16-, 2-8-, and 8-fold reduction in MIC of meropenem against EC-MβLs, EC01-EC24, K. pneumonia, respectively. Moreover, mice experiments showed that DpC also had synergistic antibacterial action with meropenem. In this work, DpC was identified to be a potent scaffold for the development of broad-spectrum inhibitors of MβLs.
Collapse
Affiliation(s)
- Jia-Qi Li
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, PR China
| | - Han Gao
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, PR China
| | - Le Zhai
- Shaanxi Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji 72101, Shaanxi Province, PR China
| | - Le-Yun Sun
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, PR China
| | - Cheng Chen
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, PR China
| | - Jia-Zhu Chigan
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, PR China
| | - Huan-Huan Ding
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, PR China
| | - Ke-Wu Yang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, PR China.
| |
Collapse
|
5796
|
Van Bocxlaer K, McArthur KN, Harris A, Alavijeh M, Braillard S, Mowbray CE, Croft SL. Film-Forming Systems for the Delivery of DNDI-0690 to Treat Cutaneous Leishmaniasis. Pharmaceutics 2021; 13:516. [PMID: 33918099 PMCID: PMC8069359 DOI: 10.3390/pharmaceutics13040516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 11/18/2022] Open
Abstract
In cutaneous leishmaniasis (CL), parasites reside in the dermis, creating an opportunity for local drug administration potentially reducing adverse effects and improving treatment adherence compared to current therapies. Polymeric film-forming systems (FFSs) are directly applied to the skin and form a thin film as the solvent evaporates. In contrast to conventional topical dosage forms, FFSs strongly adhere to the skin, favouring sustained drug delivery to the affected site, reducing the need for frequent applications, and enhancing patient compliance. This study reports the first investigation of the use of film-forming systems for the delivery of DNDI-0690, a nitroimidazole compound with potent activity against CL-causing Leishmania species. A total of seven polymers with or without plasticiser were evaluated for drying time, stickiness, film-flexibility, and cosmetic attributes; three FFSs yielded a positive evaluation for all test parameters. The impact of each of these FFSs on the permeation of the model skin permeant hydrocortisone (hydrocortisone, 1% (w/v) across the Strat-M membrane was evaluated, and the formulations resulting in the highest and lowest permeation flux (Klucel LF with triethyl citrate and Eudragit RS with dibutyl sebacate, respectively) were selected as the FFS vehicle for DNDI-0690. The release and skin distribution of the drug upon application to Leishmania-infected and uninfected BALB/c mouse skin were examined using Franz diffusion cells followed by an evaluation of the efficacy of both DNDI-0690 FFSs (1% (w/v)) in an experimental CL model. Whereas the Eudragit film resulted in a higher permeation of DNDI-0690, the Klucel film was able to deposit four times more drug into the skin, where the parasite resides. Of the FFSs formulations, only the Eudragit system resulted in a reduced parasite load, but not reduced lesion size, when compared to the vehicle only control. Whereas drug delivery into the skin was successfully modulated using different FFS systems, the FFS systems selected were not effective for the topical application of DNDI-0690. The convenience and aesthetic of FFS systems alongside their ability to modulate drug delivery to and into the skin merit further investigation using other promising antileishmanial drugs.
Collapse
Affiliation(s)
- Katrien Van Bocxlaer
- Department of Biology, York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Kerri-Nicola McArthur
- Pharmidex Pharmaceutical Services Ltd., London EC2V 8AU, UK; (K.-N.M.); (A.H.); (M.A.)
| | - Andy Harris
- Pharmidex Pharmaceutical Services Ltd., London EC2V 8AU, UK; (K.-N.M.); (A.H.); (M.A.)
| | - Mo Alavijeh
- Pharmidex Pharmaceutical Services Ltd., London EC2V 8AU, UK; (K.-N.M.); (A.H.); (M.A.)
| | - Stéphanie Braillard
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.B.); (C.E.M.)
| | - Charles E. Mowbray
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.B.); (C.E.M.)
| | - Simon L. Croft
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK;
| |
Collapse
|
5797
|
Tordonato C, Marzi MJ, Giangreco G, Freddi S, Bonetti P, Tosoni D, Di Fiore PP, Nicassio F. miR-146 connects stem cell identity with metabolism and pharmacological resistance in breast cancer. J Cell Biol 2021; 220:211945. [PMID: 33819341 PMCID: PMC8025236 DOI: 10.1083/jcb.202009053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/26/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
Although ectopic overexpression of miRNAs can influence mammary normal and cancer stem cells (SCs/CSCs), their physiological relevance remains uncertain. Here, we show that miR-146 is relevant for SC/CSC activity. MiR-146a/b expression is high in SCs/CSCs from human/mouse primary mammary tissues, correlates with the basal-like breast cancer subtype, which typically has a high CSC content, and specifically distinguishes cells with SC/CSC identity. Loss of miR-146 reduces SC/CSC self-renewal in vitro and compromises patient-derived xenograft tumor growth in vivo, decreasing the number of tumor-initiating cells, thus supporting its pro-oncogenic function. Transcriptional analysis in mammary SC-like cells revealed that miR-146 has pleiotropic effects, reducing adaptive response mechanisms and activating the exit from quiescent state, through a complex network of finely regulated miRNA targets related to quiescence, transcription, and one-carbon pool metabolism. Consistent with these findings, SCs/CSCs display innate resistance to anti-folate chemotherapies either in vitro or in vivo that can be reversed by miR-146 depletion, unmasking a “hidden vulnerability” exploitable for the development of anti-CSC therapies.
Collapse
Affiliation(s)
- Chiara Tordonato
- European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università Degli Studi di Milano, Milano, Italy
| | - Matteo Jacopo Marzi
- Center for Genomic Science of Istituto Italiano di Tecnologia at European School of Molecular Medicine, Istituto Italiano di Tecnologia, Milan, Italy
| | - Giovanni Giangreco
- European Institute of Oncology IRCCS, Milan, Italy.,Tumour Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | | | - Paola Bonetti
- Center for Genomic Science of Istituto Italiano di Tecnologia at European School of Molecular Medicine, Istituto Italiano di Tecnologia, Milan, Italy
| | | | - Pier Paolo Di Fiore
- European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università Degli Studi di Milano, Milano, Italy
| | - Francesco Nicassio
- Center for Genomic Science of Istituto Italiano di Tecnologia at European School of Molecular Medicine, Istituto Italiano di Tecnologia, Milan, Italy
| |
Collapse
|
5798
|
Winkler DA. Use of Artificial Intelligence and Machine Learning for Discovery of Drugs for Neglected Tropical Diseases. Front Chem 2021; 9:614073. [PMID: 33791277 PMCID: PMC8005575 DOI: 10.3389/fchem.2021.614073] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/18/2021] [Indexed: 12/11/2022] Open
Abstract
Neglected tropical diseases continue to create high levels of morbidity and mortality in a sizeable fraction of the world’s population, despite ongoing research into new treatments. Some of the most important technological developments that have accelerated drug discovery for diseases of affluent countries have not flowed down to neglected tropical disease drug discovery. Pharmaceutical development business models, cost of developing new drug treatments and subsequent costs to patients, and accessibility of technologies to scientists in most of the affected countries are some of the reasons for this low uptake and slow development relative to that for common diseases in developed countries. Computational methods are starting to make significant inroads into discovery of drugs for neglected tropical diseases due to the increasing availability of large databases that can be used to train ML models, increasing accuracy of these methods, lower entry barrier for researchers, and widespread availability of public domain machine learning codes. Here, the application of artificial intelligence, largely the subset called machine learning, to modelling and prediction of biological activities and discovery of new drugs for neglected tropical diseases is summarized. The pathways for the development of machine learning methods in the short to medium term and the use of other artificial intelligence methods for drug discovery is discussed. The current roadblocks to, and likely impacts of, synergistic new technological developments on the use of ML methods for neglected tropical disease drug discovery in the future are also discussed.
Collapse
Affiliation(s)
- David A Winkler
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Latrobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia.,School of Pharmacy, University of Nottingham, Nottingham, United Kingdom.,CSIRO Data61, Pullenvale, QLD, Australia
| |
Collapse
|
5799
|
He Y, Li SG, Mbaezue II, Reddy AC, Tsantrizos YS. Copper-boryl mediated transfer hydrogenation of N-sulfonyl imines using methanol as the hydrogen donor. Tetrahedron 2021. [DOI: 10.1016/j.tet.2021.132063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
5800
|
Lu W, Kostic M, Zhang T, Che J, Patricelli MP, Jones LH, Chouchani ET, Gray NS. Fragment-based covalent ligand discovery. RSC Chem Biol 2021; 2:354-367. [PMID: 34458789 PMCID: PMC8341086 DOI: 10.1039/d0cb00222d] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022] Open
Abstract
Targeted covalent inhibitors have regained widespread attention in drug discovery and have emerged as powerful tools for basic biomedical research. Fueled by considerable improvements in mass spectrometry sensitivity and sample processing, chemoproteomic strategies have revealed thousands of proteins that can be covalently modified by reactive small molecules. Fragment-based drug discovery, which has traditionally been used in a target-centric fashion, is now being deployed on a proteome-wide scale thereby expanding its utility to both the discovery of novel covalent ligands and their cognate protein targets. This powerful approach is allowing 'high-throughput' serendipitous discovery of cryptic pockets leading to the identification of pharmacological modulators of proteins previously viewed as "undruggable". The reactive fragment toolkit has been enabled by recent advances in the development of new chemistries that target residues other than cysteine including lysine and tyrosine. Here, we review the emerging area of covalent fragment-based ligand discovery, which integrates the benefits of covalent targeting and fragment-based medicinal chemistry. We discuss how the two strategies synergize to facilitate the efficient discovery of new pharmacological modulators of established and new therapeutic target proteins.
Collapse
Affiliation(s)
- Wenchao Lu
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA 02215 USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA 02215 USA
| | - Milka Kostic
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA 02215 USA
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA 02215 USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA 02215 USA
| | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA 02215 USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA 02215 USA
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA 02215 USA
| | | | - Lyn H Jones
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA 02215 USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA 02215 USA
- Department of Cell Biology, Harvard Medical School Boston MA 02215 USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA 02215 USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA 02215 USA
| |
Collapse
|