5601
|
Synthesis, Crystal Structure and Bioactivity of Phenazine-1-carboxylic Acylhydrazone Derivatives. Molecules 2021; 26:molecules26175320. [PMID: 34500750 PMCID: PMC8434039 DOI: 10.3390/molecules26175320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
A phenazine-1-carboxylic acid intermediate was synthesized from the reaction of aniline and 2-bromo-3-nitro-benzoic acid. It was then esterified and reacted with hydrazine hydrate to afford phenazine-1-carboxylic hydrazine. Finally, 10 new hydrazone compounds 3a–3j were obtained by the condensation reaction of phenazine-1-carboxylic acid hydrazide and the respective aldehyde-containing compound. The structures were characterized by 1H and 13C NMR spectroscopy, MS and single crystal X-ray diffraction. The antitumor activity of the target compounds in vitro (HeLa and A549) was determined by thiazolyl blue tetrazolium bromide. The results showed that compound (E)-N′-(2-hydroxy-4-(2-(piperidine-1-yl) ethoxy) benzyl) phenazine-1-carbonyl hydrazide 3d exhibited good cytotoxic activity.
Collapse
|
5602
|
Gomes ATPC, Neves MGPMS, Fernandes R, Ribeiro CF, Cavaleiro JAS, Moura NMM. Unraveling the Photodynamic Activity of Cationic Benzoporphyrin-Based Photosensitizers against Bladder Cancer Cells. Molecules 2021; 26:5312. [PMID: 34500746 PMCID: PMC8434352 DOI: 10.3390/molecules26175312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 02/01/2023] Open
Abstract
In this study, we report the preparation of new mono-charged benzoporphyrin complexes by reaction of the appropriate neutral benzoporphyrin with (2,2'-bipyridine)dichloroplatinum(II) and of the analogs' derivatives synthesized through alkylation of the neutral scaffold with iodomethane. All derivatives were incorporated into polyvinylpyrrolidone (PVP) micelles. The ability of the resultant formulations to generate reactive oxygen species was evaluated, mainly the singlet oxygen formation. Then, the capability of the PVP formulations to act as photosensitizers against bladder cancer cells was assessed. Some of the studied formulations were the most active photosensitizers causing a decrease in HT-1376 cells' viability. This creates an avenue to further studies related to bladder cancer cells.
Collapse
Affiliation(s)
- Ana T. P. C. Gomes
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (R.F.); (C.F.R.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Interdisciplinary Research in Health (CIIS), Faculty of Dental Medicine, Universidade Católica Portuguesa, 3504-505 Viseu, Portugal
| | | | - Rosa Fernandes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (R.F.); (C.F.R.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carlos F. Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (R.F.); (C.F.R.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - José A. S. Cavaleiro
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Nuno M. M. Moura
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
5603
|
Dyavar SR, Singh R, Emani R, Pawar GP, Chaudhari VD, Podany AT, Avedissian SN, Fletcher CV, Salunke DB. Role of toll-like receptor 7/8 pathways in regulation of interferon response and inflammatory mediators during SARS-CoV2 infection and potential therapeutic options. Biomed Pharmacother 2021; 141:111794. [PMID: 34153851 PMCID: PMC8189763 DOI: 10.1016/j.biopha.2021.111794] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/17/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) is the causative agent of Corona Virus Disease 2019 (COVID-19). Lower production of type I and III interferons and higher levels of inflammatory mediators upon SARS-CoV2 infection contribute to COVID-19 pathogenesis. Optimal interferon production and controlled inflammation are essential to limit COVID-19 pathogenesis. However, the aggravated inflammatory response observed in COVID-19 patients causes severe damage to the host and frequently advances to acute respiratory distress syndrome (ARDS). Toll-like receptor 7 and 8 (TLR7/8) signaling pathways play a central role in regulating induction of interferons (IFNs) and inflammatory mediators in dendritic cells. Controlled inflammation is possible through regulation of TLR mediated response without influencing interferon production to reduce COVID-19 pathogenesis. This review focuses on inflammatory mediators that contribute to pathogenic effects and the role of TLR pathways in the induction of interferon and inflammatory mediators and their contribution to COVID-19 pathogenesis. We conclude that potential TLR7/8 agonists inducing antiviral interferon response and controlling inflammation are important therapeutic options to effectively eliminate SARS-CoV2 induced pathogenesis. Ongoing and future studies may provide additional evidence on their safety and efficacy to treat COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Shetty Ravi Dyavar
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA.
| | - Rahul Singh
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Rohini Emani
- Buck Institute for Research on Ageing, Novato, CA, USA
| | - Ganesh P Pawar
- Division of Medicinal Chemistry, CSIR-Institute of Microbiology Technology Chandigarh, Sector-39A, Chandigarh,160036, India
| | - Vinod D Chaudhari
- Division of Medicinal Chemistry, CSIR-Institute of Microbiology Technology Chandigarh, Sector-39A, Chandigarh,160036, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anthony T Podany
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Sean N Avedissian
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Courtney V Fletcher
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India; National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
5604
|
Zhou Y, Shen W, Peng J, Deng Y, Li X. Identification of isoform/domain-selective fragments from the selection of DNA-encoded dynamic library. Bioorg Med Chem 2021; 45:116328. [PMID: 34364223 DOI: 10.1016/j.bmc.2021.116328] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 12/18/2022]
Abstract
DNA-encoded chemical library (DEL) has emerged to be a powerful ligand screening technology in drug discovery. Recently, we reported a DNA-encoded dynamic library (DEDL) approach that combines the principle of traditional dynamic combinatorial library (DCL) with DEL. DEDL has shown excellent potential in fragment-based ligand discovery with a variety of protein targets. Here, we further tested the utility of DEDL in identifying low molecular weight fragments that are selective for different isoforms or domains of the same protein family. A 10,000-member DEDL was selected against sirtuin-1, 2, and 5 (SIRT1, 2, 5) and the BD1 and BD2 domains of bromodomain 4 (BRD4), respectively. Albeit with modest potency, a series of isoform/domain-selective fragments were identified and the corresponding inhibitors were derived by fragment linking.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Wenyin Shen
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Jianzhao Peng
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Yuqing Deng
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Xiaoyu Li
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region; Laboratory for Synthetic Chemistry and Chemical Biology, Health@InnoHK, Innovation and Technology Commission, Hong Kong Special Administrative Region
| |
Collapse
|
5605
|
Kumar M, Reddy NC, Rai V. Chemical technologies for precise protein bioconjugation interfacing biology and medicine. Chem Commun (Camb) 2021; 57:7083-7095. [PMID: 34180471 DOI: 10.1039/d1cc02268g] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteins provide an excellent means to monitor and regulate biological processes. Hence, a precise chemical toolbox for their modification becomes indispensable. In this perspective, this feature article outlines our efforts to establish the core principles of chemoselectivity, site-selectivity, site-specificity, site-modularity, residue-modularity, and protein-specificity. With the knowledge to systematically regulate these parameters, the field has access to technological platforms that can address multiple challenges at the interface of chemistry, biology, and medicine.
Collapse
Affiliation(s)
- Mohan Kumar
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, MP 462 066, India.
| | - Neelesh C Reddy
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, MP 462 066, India.
| | - Vishal Rai
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, MP 462 066, India.
| |
Collapse
|
5606
|
Neveselý T, Wienhold M, Molloy JJ, Gilmour R. Advances in the E → Z Isomerization of Alkenes Using Small Molecule Photocatalysts. Chem Rev 2021; 122:2650-2694. [PMID: 34449198 DOI: 10.1021/acs.chemrev.1c00324] [Citation(s) in RCA: 222] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Geometrical E → Z alkene isomerization is intimately entwined in the historical fabric of organic photochemistry and is enjoying a renaissance (Roth et al. Angew. Chem., Int. Ed. Engl. 1989 28, 1193-1207). This is a consequence of the fundamental stereochemical importance of Z-alkenes, juxtaposed with frustrations in thermal reactivity that are rooted in microscopic reversibility. Accessing excited state reactivity paradigms allow this latter obstacle to be circumnavigated by exploiting subtle differences in the photophysical behavior of the substrate and product chromophores: this provides a molecular basis for directionality. While direct irradiation is operationally simple, photosensitization via selective energy transfer enables augmentation of the alkene repertoire to include substrates that are not directly excited by photons. Through sustained innovation, an impressive portfolio of tailored small molecule catalysts with a range of triplet energies are now widely available to facilitate contra-thermodynamic and thermo-neutral isomerization reactions to generate Z-alkene fragments. This review is intended to serve as a practical guide covering the geometric isomerization of alkenes enabled by energy transfer catalysis from 2000 to 2020, and as a logical sequel to the excellent treatment by Dugave and Demange (Chem. Rev. 2003 103, 2475-2532). The mechanistic foundations underpinning isomerization selectivity are discussed together with induction models and rationales to explain the counterintuitive directionality of these processes in which very small energy differences distinguish substrate from product. Implications for subsequent stereospecific transformations, application in total synthesis, regioselective polyene isomerization, and spatiotemporal control of pre-existing alkene configuration in a broader sense are discussed.
Collapse
Affiliation(s)
- Tomáš Neveselý
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Max Wienhold
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - John J Molloy
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Ryan Gilmour
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
5607
|
Frühauf A, Meyer-Almes FJ. Non-Hydroxamate Zinc-Binding Groups as Warheads for Histone Deacetylases. Molecules 2021; 26:5151. [PMID: 34500583 PMCID: PMC8434074 DOI: 10.3390/molecules26175151] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Histone deacetylases (HDACs) remove acetyl groups from acetylated lysine residues and have a large variety of substrates and interaction partners. Therefore, it is not surprising that HDACs are involved in many diseases. Most inhibitors of zinc-dependent HDACs (HDACis) including approved drugs contain a hydroxamate as a zinc-binding group (ZBG), which is by far the biggest contributor to affinity, while chemical variation of the residual molecule is exploited to create more or less selectivity against HDAC isozymes or other metalloproteins. Hydroxamates have a propensity for nonspecificity and have recently come under considerable suspicion because of potential mutagenicity. Therefore, there are significant concerns when applying hydroxamate-containing compounds as therapeutics in chronic diseases beyond oncology due to unwanted toxic side effects. In the last years, several alternative ZBGs have been developed, which can replace the critical hydroxamate group in HDACis, while preserving high potency. Moreover, these compounds can be developed into highly selective inhibitors. This review aims at providing an overview of the progress in the field of non-hydroxamic HDACis in the time period from 2015 to present. Formally, ZBGs are clustered according to their binding mode and structural similarity to provide qualitative assessments and predictions based on available structural information.
Collapse
Affiliation(s)
| | - Franz-Josef Meyer-Almes
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Haardtring 100, 64295 Darmstadt, Germany;
| |
Collapse
|
5608
|
Štambuk N, Konjevoda P, Pavan J. Antisense Peptide Technology for Diagnostic Tests and Bioengineering Research. Int J Mol Sci 2021; 22:9106. [PMID: 34502016 PMCID: PMC8431130 DOI: 10.3390/ijms22179106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 01/01/2023] Open
Abstract
Antisense peptide technology (APT) is based on a useful heuristic algorithm for rational peptide design. It was deduced from empirical observations that peptides consisting of complementary (sense and antisense) amino acids interact with higher probability and affinity than the randomly selected ones. This phenomenon is closely related to the structure of the standard genetic code table, and at the same time, is unrelated to the direction of its codon sequence translation. The concept of complementary peptide interaction is discussed, and its possible applications to diagnostic tests and bioengineering research are summarized. Problems and difficulties that may arise using APT are discussed, and possible solutions are proposed. The methodology was tested on the example of SARS-CoV-2. It is shown that the CABS-dock server accurately predicts the binding of antisense peptides to the SARS-CoV-2 receptor binding domain without requiring predefinition of the binding site. It is concluded that the benefits of APT outweigh the costs of random peptide screening and could lead to considerable savings in time and resources, especially if combined with other computational and immunochemical methods.
Collapse
Affiliation(s)
- Nikola Štambuk
- Center for Nuclear Magnetic Resonance, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| | - Paško Konjevoda
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| | - Josip Pavan
- Department of Ophthalmology, University Hospital Dubrava, Avenija Gojka Šuška 6, HR-10000 Zagreb, Croatia
| |
Collapse
|
5609
|
Yan F, Gao F. An overview of potential inhibitors targeting non-structural proteins 3 (PL pro and Mac1) and 5 (3CL pro/M pro) of SARS-CoV-2. Comput Struct Biotechnol J 2021; 19:4868-4883. [PMID: 34457214 PMCID: PMC8382591 DOI: 10.1016/j.csbj.2021.08.036] [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: 04/25/2021] [Revised: 08/02/2021] [Accepted: 08/21/2021] [Indexed: 12/11/2022] Open
Abstract
There is an urgent need to develop effective treatments for coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The rapid spread of SARS-CoV-2 has resulted in a global pandemic that has not only affected the daily lives of individuals but also had a significant impact on the global economy and public health. Although extensive research has been conducted to identify inhibitors targeting SARS-CoV-2, there are still no effective treatment strategies to combat COVID-19. SARS-CoV-2 comprises two important proteolytic enzymes, namely, the papain-like proteinase, located within non-structural protein 3 (nsp3), and nsp5, both of which cleave large replicase polypeptides into multiple fragments that are required for viral replication. Moreover, a domain within nsp3, known as the macrodomain (Mac1), also plays an important role in viral replication. Inhibition of their functions should be able to significantly interfere with the replication cycle of the virus, and therefore these key proteins may serve as potential therapeutic targets. The functions of the above viral targets and their corresponding inhibitors have been summarized in the current review. This review provides comprehensive updates of nsp3 and nsp5 inhibitor development and would help advance the discovery of novel anti-viral therapeutics against SARS-CoV-2.
Collapse
Affiliation(s)
- Fangfang Yan
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Feng Gao
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| |
Collapse
|
5610
|
Nikitidis G, Carlsson ACC, Karlsson S, Campbell AD, Cook C, Dai K, Emtenäs H, Jonson AC, Leek H, Malmgren M, Moravčík Š, Pithani S, Tatton MR, Zhao H, Öhlén K. Synthetic and Chromatographic Challenges and Strategies for Multigram Manufacture of KRASG12C Inhibitors. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.1c00179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Grigorios Nikitidis
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Anna-Carin C. Carlsson
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Staffan Karlsson
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Andrew D. Campbell
- Chemical Development, Pharmaceutical Technology & Development, Operations, AstraZeneca Macclesfield, Macclesfield SK10 2NA, United Kingdom
| | - Calum Cook
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Macclesfield, Macclesfield SK10 2NA, United Kingdom
| | - Kuangchu Dai
- Changzhou SynTheAll Pharmaceutical Co., Ltd, No 589, North Yulong Road, Chunjiang town, Xinbei District, Changzhou 213127, Jiangsu, China
| | - Hans Emtenäs
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Anna C. Jonson
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Hanna Leek
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Marcus Malmgren
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Štefan Moravčík
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Subhash Pithani
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| | - Matthew R. Tatton
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Macclesfield, Macclesfield SK10 2NA, United Kingdom
| | - Hucheng Zhao
- Changzhou SynTheAll Pharmaceutical Co., Ltd, No 589, North Yulong Road, Chunjiang town, Xinbei District, Changzhou 213127, Jiangsu, China
| | - Kristina Öhlén
- Early Chemical Development, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca Gothenburg, SE-431 83 Mölndal, Sweden
| |
Collapse
|
5611
|
Franz M, Mörchen B, Degenhart C, Gülden D, Shkura O, Wolters D, Koch U, Klebl B, Stoll R, Helfrich I, Scherkenbeck J. Sequence-Selective Covalent CaaX-Box Receptors Prevent Farnesylation of Oncogenic Ras Proteins and Impact MAPK/PI3 K Signaling. ChemMedChem 2021; 16:2504-2514. [PMID: 33899342 PMCID: PMC8453727 DOI: 10.1002/cmdc.202100167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Indexed: 01/21/2023]
Abstract
Oncogenic Ras proteins are implicated in the most common life-threatening cancers. Despite intense research over the past two decades, the progress towards small-molecule inhibitors has been limited. One reason for this failure is that Ras proteins interact with their effectors only via protein-protein interactions, which are notoriously difficult to address with small organic molecules. Herein we describe an alternative strategy, which prevents farnesylation and subsequent membrane insertion, a prerequisite for the activation of Ras proteins. Our approach is based on sequence-selective supramolecular receptors which bind to the C-terminal farnesyl transferase recognition unit of Ras and Rheb proteins and covalently modify the essential cysteine in the so-called CaaX-box.
Collapse
Affiliation(s)
- Matthias Franz
- Faculty of Mathematics and Natural SciencesUniversity of Wuppertal42119WuppertalGermany
| | - Britta Mörchen
- Vascular Oncology & MetastasisUniversity Hospital Essen45147EssenGermany
| | | | - Daniel Gülden
- Faculty of Mathematics and Natural SciencesUniversity of Wuppertal42119WuppertalGermany
| | - Oleksandr Shkura
- Faculty of Chemistry and BiochemistryRuhr-University Bochum44780BochumGermany
| | - Dirk Wolters
- Faculty of Chemistry and BiochemistryRuhr-University Bochum44780BochumGermany
| | - Uwe Koch
- Lead Discovery Center GmbH44227DortmundGermany
| | - Bert Klebl
- Lead Discovery Center GmbH44227DortmundGermany
| | - Raphael Stoll
- Faculty of Chemistry and BiochemistryRuhr-University Bochum44780BochumGermany
| | - Iris Helfrich
- Vascular Oncology & MetastasisUniversity Hospital Essen45147EssenGermany
| | - Jürgen Scherkenbeck
- Faculty of Mathematics and Natural SciencesUniversity of Wuppertal42119WuppertalGermany
| |
Collapse
|
5612
|
Le Bescont J, Mouawad L, Boddaert T, Bombard S, Piguel S. Photoactivatable Small‐Molecule Inhibitors for Light‐Controlled TAM Kinase Activity. CHEMPHOTOCHEM 2021. [DOI: 10.1002/cptc.202100131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Julie Le Bescont
- Institut Curie Université PSL CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
- Université Paris-Saclay CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
| | - Liliane Mouawad
- Institut Curie Université PSL CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
- Université Paris-Saclay CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
| | - Thomas Boddaert
- Université Paris-Saclay CNRS UMR 8182 ICMMO CP3A Organic Synthesis Group 91405 Orsay France
| | - Sophie Bombard
- Institut Curie Université PSL CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
- Université Paris-Saclay CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
| | - Sandrine Piguel
- Institut Curie Université PSL CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
- Université Paris-Saclay CNRS UMR 9187 INSERM U1196 Chimie et Modélisation pour la Biologie du Cancer 91405 Orsay France
| |
Collapse
|
5613
|
Wang Z, Yang L, Zhao XE. Co-crystallization and structure determination: An effective direction for anti-SARS-CoV-2 drug discovery. Comput Struct Biotechnol J 2021; 19:4684-4701. [PMID: 34426762 PMCID: PMC8373586 DOI: 10.1016/j.csbj.2021.08.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/29/2021] [Accepted: 08/17/2021] [Indexed: 01/18/2023] Open
Abstract
Safer and more-effective drugs are urgently needed to counter infections with the highly pathogenic SARS-CoV-2, cause of the COVID-19 pandemic. Identification of efficient inhibitors to treat and prevent SARS-CoV-2 infection is a predominant focus. Encouragingly, using X-ray crystal structures of therapeutically relevant drug targets (PLpro, Mpro, RdRp, and S glycoprotein) offers a valuable direction for anti-SARS-CoV-2 drug discovery and lead optimization through direct visualization of interactions. Computational analyses based primarily on MMPBSA calculations have also been proposed for assessing the binding stability of biomolecular structures involving the ligand and receptor. In this study, we focused on state-of-the-art X-ray co-crystal structures of the abovementioned targets complexed with newly identified small-molecule inhibitors (natural products, FDA-approved drugs, candidate drugs, and their analogues) with the assistance of computational analyses to support the precision design and screening of anti-SARS-CoV-2 drugs.
Collapse
Key Words
- 3CLpro, 3C-Like protease
- ACE2, angiotensin-converting enzyme 2
- COVID-19, coronavirus disease 2019
- Candidate drugs
- Co-crystal structures
- DyKAT, dynamic kinetic asymmetric transformation
- EBOV, Ebola virus
- EC50, half maximal effective concentration
- EMD, Electron Microscopy Data
- FDA, U.S. Food and Drug Administration
- FDA-approved drugs
- HCoV-229E, human coronavirus 229E
- HPLC, high-performance liquid chromatography
- IC50, half maximal inhibitory concentration
- MD, molecular dynamics
- MERS-CoV, Middle East respiratory syndrome coronavirus
- MMPBSA, molecular mechanics Poisson-Boltzmann surface area
- MTase, methyltransferase
- Mpro, main protease
- Natural products
- Nsp, nonstructural protein
- PDB, Protein Data Bank
- PLpro, papain-like protease
- RTP, ribonucleoside triphosphate
- RdRp, RNA-dependent RNA polymerase
- SAM, S-adenosylmethionine
- SARS-CoV, severe acute respiratory syndrome coronavirus
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SI, selectivity index
- Ugi-4CR, Ugi four-component reaction
- cryo-EM, cryo-electron microscopy
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, PR China
| | - Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, PR China
| | - Xian-En Zhao
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China
| |
Collapse
|
5614
|
Hamilton G, Plangger A. Cytotoxic activity of KRAS inhibitors in combination with chemotherapeutics. Expert Opin Drug Metab Toxicol 2021; 17:1065-1074. [PMID: 34347509 DOI: 10.1080/17425255.2021.1965123] [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: 02/07/2023]
Abstract
INTRODUCTION KRAS is the most frequently mutated oncogenic driver in pancreatic, lung, and colon cancer. Recently, KRAS inhibitors in clinical use show promising activity but most responses are partial and drug resistance develops. The use of therapeutics in combination with KRAS inhibitors are expected to improve outcomes. AREAS COVERED This review describes the KRAS G12C mutation-specific inhibitors and the SOS1-targeting inhibitors that reduce the GTP-loading of wildtype and mutated KRAS. Both types of compounds reduce tumor cell proliferation in vitro and in vivo. The combinations of the various KRAS inhibitors with downstream signaling effectors, modulators of KRAS-associated metabolic alterations and chemotherapeutics are summarized. EXPERT OPINION The clinical potency of mutated KRAS-specific inhibitors needs to be improved by suitable drug combinations. Inhibition of downstream signaling cascades increases toxicity and other combinations exploited comprise G12C-directed inhibitors with SOS1 inhibitors, glucose/glutamine metabolic modulators, classical chemotherapeutics, and others. The most suitable inhibitor combinations corroborated in preclinical development await clinical verification.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department Of Vascular Surgery, Medical University Of Vienna, Vienna, Austria
| | - Adelina Plangger
- Department Of Vascular Surgery, Medical University Of Vienna, Vienna, Austria
| |
Collapse
|
5615
|
Kappler-Gratias S, Bucher L, Top S, Quentin-Froignant C, Desbois N, Bertagnoli S, Louison M, Monge E, Bousquet-Melou A, Lacroix M, Gros CP, Gallardo F. Antipoxvirus Activity Evaluation of Optimized Corroles Based on Development of Autofluorescent ANCHOR Myxoma Virus. ACS Infect Dis 2021; 7:2370-2382. [PMID: 34048219 DOI: 10.1021/acsinfecdis.1c00068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A series of 43 antiviral corrole-based molecules have been tested on myxoma virus (Lausanne-like T1MYXV strain). An autofluorescent MYXV, with an ANCHOR cassette, has been used for the studies. A2B-fluorocorroles display various toxicities, from 40 being very toxic (CC50 = 1.7 μM) to nontoxic 38 (CC50 > 50 μM), whereas A3-fluorocorroles, with one to three fluorine atoms, are not toxic (with the exception of corroles 9, 10, and 22). In vitro, these compounds show a good selectivity index when used alone. Corrole 35 seems to be the most promising compound, which displays a high selectivity index with the lowest IC50. Interestingly, this "Hit" corrole is easy to synthesize in a two-step reaction. Upscaling production up to 25 g has been carried out for in vivo tests. In vivo studies on New Zealand white rabbits infected with myxoma virus show that symptoms are delayed and animal weight is increased upon treatment, while no acute toxicity of the corrole molecule was detected.
Collapse
Affiliation(s)
| | - Léo Bucher
- Institut de Chimie Moléculaire de l’Université de Bourgogne (ICMUB, UMR CNRS 6302), Université Bourgogne Franche-Comté, 9 avenue Alain Savary, BP 47870, 21078 Dijon Cedex, France
| | - Sokunthea Top
- NeoVirTech, SAS, 1 place Pierre Potier, Oncopole, 31106 Toulouse, France
- IHAP, Université de Toulouse, INRAE, ENVT, 31076 Toulouse Cedex 3, France
| | - Charlotte Quentin-Froignant
- NeoVirTech, SAS, 1 place Pierre Potier, Oncopole, 31106 Toulouse, France
- IHAP, Université de Toulouse, INRAE, ENVT, 31076 Toulouse Cedex 3, France
| | - Nicolas Desbois
- Institut de Chimie Moléculaire de l’Université de Bourgogne (ICMUB, UMR CNRS 6302), Université Bourgogne Franche-Comté, 9 avenue Alain Savary, BP 47870, 21078 Dijon Cedex, France
| | | | - Matthieu Louison
- IHAP, Université de Toulouse, INRAE, ENVT, 31076 Toulouse Cedex 3, France
| | - Emma Monge
- IHAP, Université de Toulouse, INRAE, ENVT, 31076 Toulouse Cedex 3, France
| | | | - Marlène Lacroix
- INTHERES, Université de Toulouse, INRAE, ENVT, 31076 Toulouse Cedex 3, France
| | - Claude P. Gros
- Institut de Chimie Moléculaire de l’Université de Bourgogne (ICMUB, UMR CNRS 6302), Université Bourgogne Franche-Comté, 9 avenue Alain Savary, BP 47870, 21078 Dijon Cedex, France
| | - Franck Gallardo
- NeoVirTech, SAS, 1 place Pierre Potier, Oncopole, 31106 Toulouse, France
| |
Collapse
|
5616
|
Turner LD, Nielsen AL, Lin L, Campedelli AJ, Silvaggi NR, Chen JS, Wakefield AE, Allen KN, Janda KD. Use of Crystallography and Molecular Modeling for the Inhibition of the Botulinum Neurotoxin A Protease. ACS Med Chem Lett 2021; 12:1318-1324. [PMID: 34413962 DOI: 10.1021/acsmedchemlett.1c00325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/20/2021] [Indexed: 01/14/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are extremely toxic and have been deemed a Tier 1 potential bioterrorism agent. The most potent and persistent of the BoNTs is the "A" serotype, with strategies to counter its etiology focused on designing small-molecule inhibitors of its light chain (LC), a zinc-dependent metalloprotease. The successful structure-based drug design of inhibitors has been confounded as the LC is highly flexible with significant morphological changes occurring upon inhibitor binding. To achieve greater success, previous and new cocrystal structures were evaluated from the standpoint of inhibitor enantioselectivity and their effect on active-site morphology. Based upon these structural insights, we designed inhibitors that were predicted to take advantage of π-π stacking interactions present in a cryptic hydrophobic subpocket. Structure-activity relationships were defined, and X-ray crystal structures and docking models were examined to rationalize the observed potency differences between inhibitors.
Collapse
Affiliation(s)
- Lewis D. Turner
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Alexander L. Nielsen
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Lucy Lin
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Antonio J. Campedelli
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Nicholas R. Silvaggi
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Jason S. Chen
- Automated Synthesis Facility, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Amanda E. Wakefield
- Department of Biomedical Engineering and Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Karen N. Allen
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Kim D. Janda
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
5617
|
Lasso JD, Castillo-Pazos DJ, Li CJ. Green chemistry meets medicinal chemistry: a perspective on modern metal-free late-stage functionalization reactions. Chem Soc Rev 2021; 50:10955-10982. [PMID: 34382989 DOI: 10.1039/d1cs00380a] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The progress of drug discovery and development is paced by milestones reached in organic synthesis. In the last decade, the advent of late-stage functionalization (LSF) reactions has represented a valuable breakthrough. Recent literature has defined these reactions as the chemoselective modification of complex molecules by means of C-H functionalization or the manipulation of endogenous functional groups. Traditionally, these diversifications have been accomplished by organometallic means. However, the presence of metals carries disadvantages related to their cost, environmental hazard and health risks. Fundamentally, green chemistry directives can help minimize such hazards through the development of metal-free LSF methodologies. In this review, we expand the current discussion on metal-free LSF reactions by providing an overview of C(sp2)-H, and C(sp3)-H functionalizations, as well as the utilization of heteroatom-containing functional groups as chemical handles. Selected topics such as metal-free cross-dehydrogenative coupling (CDC) reactions, organocatalysis, electrochemistry and photochemistry are also discussed. By writing the first review on metal-free LSF methodologies, we aim to highlight current advances in the field with examples that reveal specific challenges and solutions, as well as future research opportunities.
Collapse
Affiliation(s)
- Juan D Lasso
- Department of Chemistry, FRQNT Centre for Green Chemistry and Catalysis, McGill University, 801 Sherbrooke St. W., Montreal, Quebec H3A 0B8, Canada.
| | - Durbis J Castillo-Pazos
- Department of Chemistry, FRQNT Centre for Green Chemistry and Catalysis, McGill University, 801 Sherbrooke St. W., Montreal, Quebec H3A 0B8, Canada.
| | - Chao-Jun Li
- Department of Chemistry, FRQNT Centre for Green Chemistry and Catalysis, McGill University, 801 Sherbrooke St. W., Montreal, Quebec H3A 0B8, Canada.
| |
Collapse
|
5618
|
Li L, Chenna BC, Yang KS, Cole TR, Goodall ZT, Giardini M, Moghadamchargari Z, Hernandez EA, Gomez J, Calvet CM, Bernatchez JA, Mellott DM, Zhu J, Rademacher A, Thomas D, Blankenship LR, Drelich A, Laganowsky A, Tseng CTK, Liu WR, Wand AJ, Cruz-Reyes J, Siqueira-Neto JL, Meek TD. Self-Masked Aldehyde Inhibitors: A Novel Strategy for Inhibiting Cysteine Proteases. J Med Chem 2021; 64:11267-11287. [PMID: 34288674 PMCID: PMC10504874 DOI: 10.1021/acs.jmedchem.1c00628] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cysteine proteases comprise an important class of drug targets, especially for infectious diseases such as Chagas disease (cruzain) and COVID-19 (3CL protease, cathepsin L). Peptide aldehydes have proven to be potent inhibitors for all of these proteases. However, the intrinsic, high electrophilicity of the aldehyde group is associated with safety concerns and metabolic instability, limiting the use of aldehyde inhibitors as drugs. We have developed a novel class of self-masked aldehyde inhibitors (SMAIs) for cruzain, the major cysteine protease of the causative agent of Chagas disease-Trypanosoma cruzi. These SMAIs exerted potent, reversible inhibition of cruzain (Ki* = 18-350 nM) while apparently protecting the free aldehyde in cell-based assays. We synthesized prodrugs of the SMAIs that could potentially improve their pharmacokinetic properties. We also elucidated the kinetic and chemical mechanism of SMAIs and applied this strategy to the design of anti-SARS-CoV-2 inhibitors.
Collapse
Affiliation(s)
- Linfeng Li
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Bala C Chenna
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Kai S Yang
- Department of Chemistry, Texas A&M University, 580 Ross Street, College Station, Texas 77843, United States
| | - Taylor R Cole
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Zachary T Goodall
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Miriam Giardini
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Dr, La Jolla, California 92093, United States
| | - Zahra Moghadamchargari
- Department of Chemistry, Texas A&M University, 580 Ross Street, College Station, Texas 77843, United States
| | - Elizabeth A Hernandez
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Jana Gomez
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Claudia M Calvet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Dr, La Jolla, California 92093, United States
| | - Jean A Bernatchez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Dr, La Jolla, California 92093, United States
| | - Drake M Mellott
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Jiyun Zhu
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Andrew Rademacher
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Diane Thomas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Dr, La Jolla, California 92093, United States
| | - Lauren R Blankenship
- Department of Chemistry, Texas A&M University, 580 Ross Street, College Station, Texas 77843, United States
| | - Aleksandra Drelich
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, 580 Ross Street, College Station, Texas 77843, United States
| | - Chien-Te K Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Wenshe R Liu
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
- Department of Chemistry, Texas A&M University, 580 Ross Street, College Station, Texas 77843, United States
| | - A Joshua Wand
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Jorge Cruz-Reyes
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| | - Jair L Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Dr, La Jolla, California 92093, United States
| | - Thomas D Meek
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd, College Station, Texas 77843, United States
| |
Collapse
|
5619
|
Hassanisaadi M, Bonjar GHS, Rahdar A, Pandey S, Hosseinipour A, Abdolshahi R. Environmentally Safe Biosynthesis of Gold Nanoparticles Using Plant Water Extracts. NANOMATERIALS 2021; 11:nano11082033. [PMID: 34443864 PMCID: PMC8400837 DOI: 10.3390/nano11082033] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/18/2022]
Abstract
Due to their simplicity of synthesis, stability, and functionalization, low toxicity, and ease of detection, gold nanoparticles (AuNPs) are a natural choice for biomedical applications. AuNPs’ unique optoelectronic features have subsequently been investigated and used in high-tech applications such as organic photovoltaics, sensory probes, therapeutic agents, the administration of drugs in biological and medical applications, electronic devices, catalysis, etc. Researchers have demonstrated the biosynthesis of AuNPs using plants. The present study evaluates 109 plant species used in the traditional medicine of Middle East countries as new sources of AuNPs in a wide variety of laboratory environments. In this study, dried samples of bark, bulb, flower, fruit, gum, leaf, petiole, rhizome, root, seed, stamen, and above-ground parts were evaluated in water extracts. About 117 plant parts were screened from 109 species in 54 plant families, with 102 extracts demonstrating a bioreduction of Au3+ to Au0, revealing 37 new plant species in this regard. The color change of biosynthesized AuNPs to gray, violet, or red was confirmed by UV-Visible spectroscopy, TEM, FSEM, DLS, and EDAX of six plants. In this study, AuNPs of various sizes were measured from 27 to 107 nm. This study also includes an evaluation of the potency of traditional East Asian medicinal plants used in this biosynthesis of AuNPs. An environmentally safe procedure such as this could act as a foundation for cosmetic industries whose quality assessment systems give a high priority to non-chemically synthesized products. It is crucial that future optimizations are adequately documented to scale up the described process.
Collapse
Affiliation(s)
- Mohadeseh Hassanisaadi
- Department of Plant Protection, Shahid Bahonar University of Kerman, Kerman 7618411764, Iran; (M.H.); (A.H.)
| | - Gholam Hosein Shahidi Bonjar
- Department of Plant Protection, Shahid Bahonar University of Kerman, Kerman 7618411764, Iran; (M.H.); (A.H.)
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 7616913555, Iran
- Correspondence: (G.H.S.B.); or (S.P.)
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 98615-538, Iran;
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
- Correspondence: (G.H.S.B.); or (S.P.)
| | - Akbar Hosseinipour
- Department of Plant Protection, Shahid Bahonar University of Kerman, Kerman 7618411764, Iran; (M.H.); (A.H.)
| | - Roohollah Abdolshahi
- Department of Agronomy and Plant Breeding, Shahid Bahonar University of Kerman, Kerman 7618411764, Iran;
| |
Collapse
|
5620
|
Terekhova N, Khailova LS, Rokitskaya TI, Nazarov PA, Islamov DR, Usachev KS, Tatarinov DA, Mironov VF, Kotova EA, Antonenko YN. Trialkyl(vinyl)phosphonium Chlorophenol Derivatives as Potent Mitochondrial Uncouplers and Antibacterial Agents. ACS OMEGA 2021; 6:20676-20685. [PMID: 34396013 PMCID: PMC8359139 DOI: 10.1021/acsomega.1c02909] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/19/2021] [Indexed: 05/08/2023]
Abstract
Trialkyl phosphonium derivatives of vinyl-substituted p-chlorophenol were synthesized here by a recently developed method of preparing quaternary phosphonium salts from phosphine oxides using Grignard reagents. All the derivatives with a number (n) of carbon atoms in phosphonium alkyl substituents varying from 4 to 7 showed pronounced uncoupling activity in isolated rat liver mitochondria at micromolar concentrations, with a tripentyl derivative being the most effective both in accelerating respiration and causing membrane potential collapse, as well as in provoking mitochondrial swelling in a potassium-acetate medium. Remarkably, the trialkyl phosphonium derivatives with n from 4 to 7 also proved to be rather potent antibacterial agents. Methylation of the chlorophenol hydroxyl group suppressed the effects of P555 and P444 on the respiration and membrane potential of mitochondria but not those of P666, thereby suggesting a mechanistic difference in the mitochondrial uncoupling by these derivatives, which was predominantly protonophoric (carrier-like) in the case of P555 and P444 but detergent-like with P666. The latter was confirmed by the carboxyfluorescein leakage assay on model liposomal membranes.
Collapse
Affiliation(s)
- Natalia
V. Terekhova
- Arbuzov
Institute of Organic and Physical Chemistry, FRC Kazan Scientific
Center of Russian Academy of Sciences, Arbuzov Str. 8, Kazan 420088, Russian Federation
| | - Lyudmila S. Khailova
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, Leninskie Gory 1, Moscow 119991, Russian Federation
| | - Tatyana I. Rokitskaya
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, Leninskie Gory 1, Moscow 119991, Russian Federation
| | - Pavel A. Nazarov
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, Leninskie Gory 1, Moscow 119991, Russian Federation
| | - Daut R. Islamov
- Arbuzov
Institute of Organic and Physical Chemistry, FRC Kazan Scientific
Center of Russian Academy of Sciences, Arbuzov Str. 8, Kazan 420088, Russian Federation
| | - Konstantin S. Usachev
- Institute
of Fundamental Medicine and Biology, Kazan
Federal University, Kremlevskaya 18, Kazan 420008, Russian Federation
| | - Dmitry A. Tatarinov
- Arbuzov
Institute of Organic and Physical Chemistry, FRC Kazan Scientific
Center of Russian Academy of Sciences, Arbuzov Str. 8, Kazan 420088, Russian Federation
| | - Vladimir F. Mironov
- Arbuzov
Institute of Organic and Physical Chemistry, FRC Kazan Scientific
Center of Russian Academy of Sciences, Arbuzov Str. 8, Kazan 420088, Russian Federation
| | - Elena A. Kotova
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, Leninskie Gory 1, Moscow 119991, Russian Federation
| | - Yuri N. Antonenko
- Belozersky
Institute of Physico-Chemical Biology, Lomonosov
Moscow State University, Leninskie Gory 1, Moscow 119991, Russian Federation
| |
Collapse
|
5621
|
Patel SC, Smith MW, Mercer JAM, Suzuki K, Burns NZ. Enantioselective Cyclobutenylation of Olefins Using N-Sulfonyl-1,2,3-Triazoles as Vicinal Dicarbene Equivalents. Org Lett 2021; 23:6530-6535. [PMID: 34374544 DOI: 10.1021/acs.orglett.1c02331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cyclobutenes are highly useful synthetic intermediates as well as important motifs in bioactive small molecules. Herein, we report a regio-, chemo-, and enantioselective synthesis of cyclobutenes from olefins using N-sulfonyl-1,2,3-triazoles as vicinal dicarbene equivalents or alkyne [2 + 2] cycloaddition surrogates. Terminal and cis-olefins can be transformed into enantioenriched cyclopropanes via rhodium catalysis. Then, in one pot, treatment of these intermediates with tosyl hydrazide and base effects diazo formation followed by rhodium-catalyzed ring expansion to yield enantioenriched cyclobutenes. These cyclobutenes can be transformed into highly substituted, enantioenriched cyclobutanes, including structures relevant to natural product scaffolds.
Collapse
Affiliation(s)
- Sajan C Patel
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Myles W Smith
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Jaron A M Mercer
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Kensuke Suzuki
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Noah Z Burns
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
5622
|
|
5623
|
Lundquist KP, Panchal V, Gotfredsen CH, Brenk R, Clausen MH. Fragment-Based Drug Discovery for RNA Targets. ChemMedChem 2021; 16:2588-2603. [PMID: 34101375 DOI: 10.1002/cmdc.202100324] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Indexed: 12/26/2022]
Abstract
Rapid development within the fields of both fragment-based drug discovery (FBDD) and medicinal targeting of RNA provides possibilities for combining technologies and methods in novel ways. This review provides an overview of fragment-based screening (FBS) against RNA targets, including a discussion of the most recently used screening and hit validation methods such as NMR spectroscopy, X-ray crystallography, and virtual screening methods. A discussion of fragment library design based on research from small-molecule RNA binders provides an overview on both the currently limited guidelines within RNA-targeting fragment library design, and future possibilities. Finally, future perspectives are provided on screening and hit validation methods not yet used in combination with both fragment screening and RNA targets.
Collapse
Affiliation(s)
- Kasper P Lundquist
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800, Kgs. Lyngby, Denmark
| | - Vipul Panchal
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5020, Bergen, Norway
| | - Charlotte H Gotfredsen
- NMR Center ⋅ DTU, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800, Kgs. Lyngby, Denmark
| | - Ruth Brenk
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5020, Bergen, Norway
| | - Mads H Clausen
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800, Kgs. Lyngby, Denmark
| |
Collapse
|
5624
|
Sutanto F, Shaabani S, Oerlemans R, Eris D, Patil P, Hadian M, Wang M, Sharpe ME, Groves MR, Dömling A. Combining High-Throughput Synthesis and High-Throughput Protein Crystallography for Accelerated Hit Identification. Angew Chem Int Ed Engl 2021; 60:18231-18239. [PMID: 34097796 PMCID: PMC8456925 DOI: 10.1002/anie.202105584] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/31/2021] [Indexed: 12/24/2022]
Abstract
Protein crystallography (PX) is widely used to drive advanced stages of drug optimization or to discover medicinal chemistry starting points by fragment soaking. However, recent progress in PX could allow for a more integrated role into early drug discovery. Here, we demonstrate for the first time the interplay of high throughput synthesis and high throughput PX. We describe a practical multicomponent reaction approach to acrylamides and -esters from diverse building blocks suitable for mmol scale synthesis on 96-well format and on a high-throughput nanoscale format in a highly automated fashion. High-throughput PX of our libraries efficiently yielded potent covalent inhibitors of the main protease of the COVID-19 causing agent, SARS-CoV-2. Our results demonstrate, that the marriage of in situ HT synthesis of (covalent) libraires and HT PX has the potential to accelerate hit finding and to provide meaningful strategies for medicinal chemistry projects.
Collapse
Affiliation(s)
- Fandi Sutanto
- University of GroningenDepartment of Drug DesignA. Deusinglaan 19713AVGroningenThe Netherlands
| | - Shabnam Shaabani
- University of GroningenDepartment of Drug DesignA. Deusinglaan 19713AVGroningenThe Netherlands
| | - Rick Oerlemans
- University of GroningenDepartment of Drug DesignA. Deusinglaan 19713AVGroningenThe Netherlands
| | - Deniz Eris
- Photon Science DivisionPaul Scherrer InstituteSwitzerland
| | - Pravin Patil
- University of GroningenDepartment of Drug DesignA. Deusinglaan 19713AVGroningenThe Netherlands
| | - Mojgan Hadian
- University of GroningenDepartment of Drug DesignA. Deusinglaan 19713AVGroningenThe Netherlands
| | - Meitian Wang
- Photon Science DivisionPaul Scherrer InstituteSwitzerland
| | | | - Matthew R. Groves
- University of GroningenDepartment of Drug DesignA. Deusinglaan 19713AVGroningenThe Netherlands
| | - Alexander Dömling
- University of GroningenDepartment of Drug DesignA. Deusinglaan 19713AVGroningenThe Netherlands
| |
Collapse
|
5625
|
Sutanto F, Shaabani S, Oerlemans R, Eris D, Patil P, Hadian M, Wang M, Sharpe ME, Groves MR, Dömling A. Combining High‐Throughput Synthesis and High‐Throughput Protein Crystallography for Accelerated Hit Identification. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202105584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Fandi Sutanto
- University of Groningen Department of Drug Design A. Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Shabnam Shaabani
- University of Groningen Department of Drug Design A. Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Rick Oerlemans
- University of Groningen Department of Drug Design A. Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Deniz Eris
- Photon Science Division Paul Scherrer Institute Switzerland
| | - Pravin Patil
- University of Groningen Department of Drug Design A. Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Mojgan Hadian
- University of Groningen Department of Drug Design A. Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Meitian Wang
- Photon Science Division Paul Scherrer Institute Switzerland
| | | | - Matthew R. Groves
- University of Groningen Department of Drug Design A. Deusinglaan 1 9713 AV Groningen The Netherlands
| | - Alexander Dömling
- University of Groningen Department of Drug Design A. Deusinglaan 1 9713 AV Groningen The Netherlands
| |
Collapse
|
5626
|
Charzewski Ł, Krzyśko KA, Lesyng B. Exploring Covalent Docking Mechanisms of Boron-Based Inhibitors to Class A, C and D β-Lactamases Using Time-dependent Hybrid QM/MM Simulations. Front Mol Biosci 2021; 8:633181. [PMID: 34434961 PMCID: PMC8380965 DOI: 10.3389/fmolb.2021.633181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Recently, molecular covalent docking has been extensively developed to design new classes of inhibitors that form chemical bonds with their biological targets. This strategy for the design of such inhibitors, in particular boron-based inhibitors, holds great promise for the vast family of β-lactamases produced, inter alia, by Gram-negative antibiotic-resistant bacteria. However, the description of covalent docking processes requires a quantum-mechanical approach, and so far, only a few studies of this type have been presented. This study accurately describes the covalent docking process between two model inhibitors - representing two large families of inhibitors based on boronic-acid and bicyclic boronate scaffolds, and three β-lactamases which belong to the A, C, and D classes. Molecular fragments containing boron can be converted from a neutral, trigonal, planar state with sp2 hybridization to the anionic, tetrahedral sp3 state in a process sometimes referred to as morphing. This study applies multi-scale modeling methods, in particular, the hybrid QM/MM approach which has predictive power reaching well beyond conventional molecular modeling. Time-dependent QM/MM simulations indicated several structural changes and geometric preferences, ultimately leading to covalent docking processes. With current computing technologies, this approach is not computationally expensive, can be used in standard molecular modeling and molecular design works, and can effectively support experimental research which should allow for a detailed understanding of complex processes important to molecular medicine. In particular, it can support the rational design of covalent boron-based inhibitors for β-lactamases as well as for many other enzyme systems of clinical relevance, including SARS-CoV-2 proteins.
Collapse
Affiliation(s)
| | | | - Bogdan Lesyng
- Department of Biophysics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| |
Collapse
|
5627
|
Lan L, Sun Y, Jin X, Xie L, Liu L, Cheng L. A Light‐Controllable Chemical Modulation of m
6
A RNA Methylation. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ling Lan
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Laboratory of Molecular Recognition and Function CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Ying‐Jie Sun
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Laboratory of Molecular Recognition and Function CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Xiao‐Yang Jin
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Laboratory of Molecular Recognition and Function CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Li‐Jun Xie
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Laboratory of Molecular Recognition and Function CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Li Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Laboratory of Molecular Recognition and Function CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Liang Cheng
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Laboratory of Molecular Recognition and Function CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
- Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
5628
|
Antiviral, Cytotoxic, and Antioxidant Activities of Three Edible Agaricomycetes Mushrooms: Pleurotus columbinus, Pleurotus sajor-caju, and Agaricus bisporus. J Fungi (Basel) 2021; 7:jof7080645. [PMID: 34436184 PMCID: PMC8399653 DOI: 10.3390/jof7080645] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 01/02/2023] Open
Abstract
In this study, we investigated aqueous extracts of three edible mushrooms: Agaricus bisporus (white button mushroom), Pleurotus columbinus (oyster mushroom), and Pleurotus sajor-caju (grey oyster mushroom). The extracts were biochemically characterized for total carbohydrate, phenolic, flavonoid, vitamin, and protein contents besides amino acid analysis. Triple TOF proteome analysis showed 30.1% similarity between proteomes of the two Pleurotus spp. All three extracts showed promising antiviral activities. While Pleurotus columbinus extract showed potent activity against adenovirus (Ad7, selectivity index (SI) = 4.2), Agaricus bisporus showed strong activity against herpes simplex II (HSV-2; SI = 3.7). The extracts showed low cytotoxicity against normal human peripheral blood mononuclear cells (PBMCs) and moderate cytotoxicity against prostate (PC3, DU-145); colorectal (Colo-205); cecum carcinoma (LS-513); liver carcinoma (HepG2); cervical cancer (HeLa); breast adenocarcinoma (MDA-MB-231 and MCF-7) as well as leukemia (CCRF-CEM); acute monocytic leukemia (THP1); acute promyelocytic leukemia (NB4); and lymphoma (U937) cell lines. Antioxidant activity was evaluated using 2,2-diphenyl-1-picryl-hydrazyl-hydrate (DPPH) radical scavenging, 2,2′-Azinobis-(3-Ethylbenzthiazolin-6-Sulfonic Acid) ABTS radical cation scavenging, and oxygen radical absorbance capacity (ORAC) assays. The three extracts showed potential antioxidant activities with the maximum activity recorded for Pleurotus columbinus (IC50 µg/mL) = 35.13 ± 3.27 for DPPH, 13.97 ± 4.91 for ABTS, and 29.42 ± 3.21 for ORAC assays.
Collapse
|
5629
|
Suzuki S, Yonesaka K, Teramura T, Takehara T, Kato R, Sakai H, Haratani K, Tanizaki J, Kawakami H, Hayashi H, Sakai K, Nishio K, Nakagawa K. KRAS inhibitor-resistance in MET-amplified KRAS G12C non-small cell lung cancer induced by RAS- and non-RAS-mediated cell signaling mechanisms. Clin Cancer Res 2021; 27:5697-5707. [PMID: 34365406 DOI: 10.1158/1078-0432.ccr-21-0856] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/21/2021] [Accepted: 07/30/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Treatment with KRAS G12C inhibitors such as sotorasib can produce substantial regression of tumors in some patients with non-small cell lung cancer (NSCLC). These patients require alternative treatment after acquiring resistance to the inhibitor. The mechanisms underlying this acquired resistance are unclear. The purpose of this study was to identify the mechanisms underlying acquired sotorasib resistance, and to explore potential treatments for rescuing patients with sotorasib-resistant KRAS G12C NSCLC cells. EXPERIMENTAL DESIGN Clones of sotorasib-sensitive KRAS G12C NSCLC H23 cells exposed to different concentrations of sotorasib were examined using whole-genomic transcriptome analysis, multiple receptor kinase phosphorylation analysis, and gene copy number evaluation. The underlying mechanisms of resistance were investigated using immunological examination, and a treatment aimed at overcoming resistance was tested in vitro and in vivo Results: Unbiased screening detected subclonal evolution of MET amplification in KRAS G12C NSCLC cells that had developed resistance to sotorasib in vitro MET knockdown using siRNA restored susceptibility to sotorasib in these resistant cells. MET activation by its amplification reinforced RAS cycling from its inactive form to its active form. In addition to RAS-mediated MEK-ERK induction, MET induced AKT activation independently of RAS. Crizotinib, a MET inhibitor, restored sensitivity to sotorasib by eliminating RAS-MEK-ERK as well as AKT signaling. MET/KRAS G12C dual inhibition led to tumor shrinkage in sotorasib-resistant xenograft mice. CONCLUSIONS MET amplification leads to the development of resistance to KRAS G12C inhibitors in NSCLC. Dual blockade of MET and KRAS G12C could be a treatment option for MET amplified, KRAS G12C-mutated NSCLC.
Collapse
Affiliation(s)
- Shinichiro Suzuki
- Department of Medical Oncology, Kindai University Faculty of Medicine
| | - Kimio Yonesaka
- Department of Medical Oncology, Kindai University Faculty of Medicine
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University
| | - Toshiyuki Takehara
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine
| | | | - Hitomi Sakai
- Department of Medical Oncology, Kindai University Faculty of Medicine
| | - Koji Haratani
- Department of Medical Oncology, Kindai University Faculty of Medicine
| | - Junko Tanizaki
- Department of Medical Oncology, Kindai University Faculty of Medicine
| | | | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine
| | - Kazuko Sakai
- Department of Genome Biology, Kinki University Faculty of Medicine
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine
| | | |
Collapse
|
5630
|
Altarejos J, Sucunza D, Vaquero JJ, Carreras J. Enantioselective Copper-Catalyzed Synthesis of Trifluoromethyl-Cyclopropylboronates. Org Lett 2021; 23:6174-6178. [PMID: 34320310 PMCID: PMC8895459 DOI: 10.1021/acs.orglett.1c02420] [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] [Indexed: 11/28/2022]
Abstract
![]()
A copper-catalyzed
enantioselective cyclopropanation involving
trifluorodiazoethane in the presence of alkenyl boronates has been
developed. This transformation enables the preparation of 2-substituted-3-(trifluoromethyl)cyclopropylboronates
with high levels of stereocontrol. The products are valuable synthetic
intermediates by transformation of the boronate group. This methodology
can be applied to the synthesis of novel trifluoromethylated analogues
of trans-2-arylcyclopropylamines, which are prevalent
motifs in biologically active compounds.
Collapse
Affiliation(s)
- Julia Altarejos
- Universidad de Alcalá, Departamento de Química Orgánica y Química Inorgánica,, Alcalá de Henares 28805, Spain.,Instituto de Investigación Química Andrés Manuel del Río (IQAR), Universidad de Alcalá, Alcalá de Henares 28805, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - David Sucunza
- Universidad de Alcalá, Departamento de Química Orgánica y Química Inorgánica,, Alcalá de Henares 28805, Spain.,Instituto de Investigación Química Andrés Manuel del Río (IQAR), Universidad de Alcalá, Alcalá de Henares 28805, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Juan J Vaquero
- Universidad de Alcalá, Departamento de Química Orgánica y Química Inorgánica,, Alcalá de Henares 28805, Spain.,Instituto de Investigación Química Andrés Manuel del Río (IQAR), Universidad de Alcalá, Alcalá de Henares 28805, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Javier Carreras
- Universidad de Alcalá, Departamento de Química Orgánica y Química Inorgánica,, Alcalá de Henares 28805, Spain.,Instituto de Investigación Química Andrés Manuel del Río (IQAR), Universidad de Alcalá, Alcalá de Henares 28805, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| |
Collapse
|
5631
|
Affiliation(s)
- Patricia Cuervo
- Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brazil.
| | - Gabriel Padrón
- Center for Genetic Engineering & Biotechnology, La Habana, Cuba.
| |
Collapse
|
5632
|
Retamal MA, Fernandez-Olivares A, Stehberg J. Over-activated hemichannels: A possible therapeutic target for human diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166232. [PMID: 34363932 DOI: 10.1016/j.bbadis.2021.166232] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022]
Abstract
In our body, all the cells are constantly sharing chemical and electrical information with other cells. This intercellular communication allows them to respond in a concerted way to changes in the extracellular milieu. Connexins are transmembrane proteins that have the particularity of forming two types of channels; hemichannels and gap junction channels. Under normal conditions, hemichannels allow the controlled release of signaling molecules to the extracellular milieu. However, under certain pathological conditions, over-activated hemichannels can induce and/or exacerbate symptoms. In the last decade, great efforts have been put into developing new tools that can modulate these over-activated hemichannels. Small molecules, antibodies and mimetic peptides have shown a potential for the treatment of human diseases. In this review, we summarize recent findings in the field of hemichannel modulation via specific tools, and how these tools could improve patient outcome in certain pathological conditions.
Collapse
Affiliation(s)
- Mauricio A Retamal
- Universidad del Desarrollo, Programa de Comunicación Celular en Cáncer, Santiago, Chile; Universidad del Desarrollo, Centro de Fisiología Celular e Integrativa, Santiago, Chile.
| | | | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
5633
|
Santiago Á, Guzmán-Ocampo DC, Aguayo-Ortiz R, Dominguez L. Characterizing the Chemical Space of γ-Secretase Inhibitors and Modulators. ACS Chem Neurosci 2021; 12:2765-2775. [PMID: 34291906 DOI: 10.1021/acschemneuro.1c00313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
γ-Secretase (GS) is one of the most attractive molecular targets for the treatment of Alzheimer's disease (AD). Its key role in the final step of amyloid-β peptides generation and its relationship in the cascade of events for disease development have caught the attention of many pharmaceutical groups. Over the past years, different inhibitors and modulators have been evaluated as promising therapeutics against AD. However, despite the great chemical diversity of the reported compounds, a global classification and visual representation of the chemical space for GS inhibitors and modulators remain unavailable. In the present work, we carried out a two-dimensional (2D) chemical space analysis from different classes and subclasses of GS inhibitors and modulators based on their structural similarity. Along with the novel structural information available for GS complexes, our analysis opens the possibility to identify compounds with high molecular similarity, critical to finding new chemical structures through the optimization of existing compounds and relating them with a potential binding site.
Collapse
Affiliation(s)
- Ángel Santiago
- Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Dulce C. Guzmán-Ocampo
- Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Rodrigo Aguayo-Ortiz
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Laura Dominguez
- Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
5634
|
Yamada T, Komatsu T. Methemoglobin-Albumin Cluster Incorporating Protoporphyrin IX: Dual Functional Protein Drug for Photodynamic Therapy. Chembiochem 2021; 22:2526-2529. [PMID: 34156148 DOI: 10.1002/cbic.202100213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/18/2021] [Indexed: 11/10/2022]
Abstract
We describe the synthesis, photophysical properties, and photodynamic activity of a methemoglobin (metHb) wrapped covalently by human serum albumins (HSAs) incorporating protoporphyrin IX (PPIX): a metHb-HSA3 -PPIX2 cluster. The metHb core catalyzes H2 O2 disproportionation to generate O2 in tumor tissue. The HSA3 -PPIX2 shell acts as a photosensitizer for 1 O2 formation. The metHb-HSA3 -PPIX2 cluster acts as a dual functional protein drug for photodynamic therapy.
Collapse
Affiliation(s)
- Taiga Yamada
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga Bunkyo-ku, Tokyo, 112-8551, Japan
| | - Teruyuki Komatsu
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga Bunkyo-ku, Tokyo, 112-8551, Japan
| |
Collapse
|
5635
|
Sun J, Wang J, Wang X, Hu X, Cao H, Bai J, Li D, Hua H. Design and synthesis of β-carboline derivatives with nitrogen mustard moieties against breast cancer. Bioorg Med Chem 2021; 45:116341. [PMID: 34365102 DOI: 10.1016/j.bmc.2021.116341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022]
Abstract
To discover the promising antitumor agents, a series of β-carboline derivatives with nitrogen mustard moieties were designed and synthesized. Most target derivatives showed antiproliferative activity against MCF-7 and MDA-MB-231 cells. Among them, (1-methyl-9H-pyrido[3,4-b]indol-3-yl)methyl (S)-3-(4-(bis(2-chloroethyl)amino)phenyl)-2-formamidopropanoate possessed the most potent antiproliferative activity with IC50 values of 1.79 μM and 4.96 μM, respectively, which were significantly higher than that of the parent compounds, and the efficacy was comparable to that of the positive control doxorubicin. More importantly, it showed weak cytotoxicity against human normal breast cell line MCF-10A (IC50 > 20 μM), exhibiting certain selectivity. Subsequently, further mechanism exploration indicated that it induced G2/M phase cell cycle arrest and apoptosis in MDA-MB-231 cells. The DCFH-DA fluorescent probe assay and comet assay showed that this compound could cause intracellular ROS accumulation and DNA damage. In addition, it exerted potent inhibitory effect on the migration, invasion and adhesion of MDA-MB-231 cells in vitro. In short, (1-methyl-9H-pyrido[3,4-b]indol-3-yl)methyl (S)-3-(4-(bis(2-chloroethyl)amino)phenyl)-2-formamidopropanoate was considered as a promising compound for anti-breast cancer.
Collapse
Affiliation(s)
- Jianan Sun
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Jiesen Wang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Xinyan Wang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Xu Hu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Hao Cao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Jiao Bai
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| |
Collapse
|
5636
|
Xu P, Chang JC, Zhou X, Wang W, Bamkole M, Wong E, Bettayeb K, Jiang LL, Huang T, Luo W, Xu H, Nairn AC, Flajolet M, Ip NY, Li YM, Greengard P. GSAP regulates lipid homeostasis and mitochondrial function associated with Alzheimer's disease. J Exp Med 2021; 218:e20202446. [PMID: 34156424 PMCID: PMC8222926 DOI: 10.1084/jem.20202446] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/22/2021] [Accepted: 05/26/2021] [Indexed: 11/04/2022] Open
Abstract
Biochemical, pathogenic, and human genetic data confirm that GSAP (γ-secretase activating protein), a selective γ-secretase modulatory protein, plays important roles in Alzheimer's disease (AD) and Down's syndrome. However, the molecular mechanism(s) underlying GSAP-dependent pathogenesis remains largely elusive. Here, through unbiased proteomics and single-nuclei RNAseq, we identified that GSAP regulates multiple biological pathways, including protein phosphorylation, trafficking, lipid metabolism, and mitochondrial function. We demonstrated that GSAP physically interacts with the Fe65-APP complex to regulate APP trafficking/partitioning. GSAP is enriched in the mitochondria-associated membrane (MAM) and regulates lipid homeostasis through the amyloidogenic processing of APP. GSAP deletion generates a lipid environment unfavorable for AD pathogenesis, leading to improved mitochondrial function and the rescue of cognitive deficits in an AD mouse model. Finally, we identified a novel GSAP single-nucleotide polymorphism that regulates its brain transcript level and is associated with an increased AD risk. Together, our findings indicate that GSAP impairs mitochondrial function through its MAM localization and that lowering GSAP expression reduces pathological effects associated with AD.
Collapse
Affiliation(s)
- Peng Xu
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Jerry C. Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science and Technology Parks, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease, and Drug Development, Shenzhen–Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Wei Wang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Michael Bamkole
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karima Bettayeb
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Lu-Lin Jiang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Timothy Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Wenjie Luo
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Angus C. Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, CT
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Nancy Y. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science and Technology Parks, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease, and Drug Development, Shenzhen–Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Program of Pharmacology and Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| |
Collapse
|
5637
|
Yue J, Li L, Jiang C, Mei Q, Dong WF, Yan R. Riboflavin-based carbon dots with high singlet oxygen generation for photodynamic therapy. J Mater Chem B 2021; 9:7972-7978. [PMID: 34338706 DOI: 10.1039/d1tb01291f] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Photodynamic therapy, as an effective treatment for superficial tumors, has attracted more and more attention. The development of safe, biocompatible and in vivo photosensitive materials is helpful to promote photodynamic therapy. Here we report green fluorescent carbon quantum dots prepared from a natural vitamin, riboflavin (VB2), as a photosensitizer. The VB2-based carbon dots have excellent water solubility and biocompatibility, and their singlet oxygen generation ability is much stronger than that of riboflavin itself. Through endocytosis, the carbon dots can easily enter the cells and show bright green fluorescence. In vivo experiments show that after photodynamic therapy the carbon dots can significantly inhibit the growth of tumors, and will not have toxic and side effects on other organs.
Collapse
Affiliation(s)
- Juan Yue
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China and The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou 215153, P. R. China. and CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, P. R. China.
| | - Li Li
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, P. R. China.
| | - Chenyu Jiang
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, P. R. China. and Jinan Guokeyigong Science and Technology Development Co., Ltd, Jinan 250103, P. R. China.
| | - Qian Mei
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, P. R. China.
| | - Wen-Fei Dong
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, P. R. China. and Jinan Guokeyigong Science and Technology Development Co., Ltd, Jinan 250103, P. R. China.
| | - Ruhong Yan
- The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou 215153, P. R. China. and CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), 88 Keling Road, Suzhou 215163, P. R. China.
| |
Collapse
|
5638
|
Litowczenko J, Woźniak-Budych MJ, Staszak K, Wieszczycka K, Jurga S, Tylkowski B. Milestones and current achievements in development of multifunctional bioscaffolds for medical application. Bioact Mater 2021; 6:2412-2438. [PMID: 33553825 PMCID: PMC7847813 DOI: 10.1016/j.bioactmat.2021.01.007] [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: 11/13/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering (TE) is a rapidly growing interdisciplinary field, which aims to restore or improve lost tissue function. Despite that TE was introduced more than 20 years ago, innovative and more sophisticated trends and technologies point to new challenges and development. Current challenges involve the demand for multifunctional bioscaffolds which can stimulate tissue regrowth by biochemical curves, biomimetic patterns, active agents and proper cell types. For those purposes especially promising are carefully chosen primary cells or stem cells due to its high proliferative and differentiation potential. This review summarized a variety of recently reported advanced bioscaffolds which present new functions by combining polymers, nanomaterials, bioactive agents and cells depending on its desired application. In particular necessity of study biomaterial-cell interactions with in vitro cell culture models, and studies using animals with in vivo systems were discuss to permit the analysis of full material biocompatibility. Although these bioscaffolds have shown a significant therapeutic effect in nervous, cardiovascular and muscle, tissue engineering, there are still many remaining unsolved challenges for scaffolds improvement.
Collapse
Affiliation(s)
- Jagoda Litowczenko
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Marta J. Woźniak-Budych
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Katarzyna Staszak
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Stefan Jurga
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Bartosz Tylkowski
- Eurecat, Centre Tecnològic de Catalunya, Chemical Technologies Unit, Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| |
Collapse
|
5639
|
Elhusseiny SM, El-Mahdy TS, Awad MF, Elleboudy NS, Farag MMS, Yassein MA, Aboshanab KM. Proteome Analysis and In Vitro Antiviral, Anticancer and Antioxidant Capacities of the Aqueous Extracts of Lentinula edodes and Pleurotus ostreatus Edible Mushrooms. Molecules 2021; 26:4623. [PMID: 34361776 PMCID: PMC8348442 DOI: 10.3390/molecules26154623] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/16/2022] Open
Abstract
In this study, we examined aqueous extracts of the edible mushrooms Pleurotus ostreatus (oyster mushroom) and Lentinula edodes (shiitake mushroom). Proteome analysis was conducted using LC-Triple TOF-MS and showed the expression of 753 proteins by Pleurotus ostreatus, and 432 proteins by Lentinula edodes. Bioactive peptides: Rab GDP dissociation inhibitor, superoxide dismutase, thioredoxin reductase, serine proteinase and lectin, were identified in both mushrooms. The extracts also included promising bioactive compounds including phenolics, flavonoids, vitamins and amino acids. The extracts showed promising antiviral activities, with a selectivity index (SI) of 4.5 for Pleurotus ostreatus against adenovirus (Ad7), and a slight activity for Lentinula edodes against herpes simplex-II (HSV-2). The extracts were not cytotoxic to normal human peripheral blood mononuclear cells (PBMCs). On the contrary, they showed moderate cytotoxicity against various cancer cell lines. Additionally, antioxidant activity was assessed using DPPH radical scavenging, ABTS radical cation scavenging and ORAC assays. The two extracts showed potential antioxidant activities, with the maximum activity seen for Pleurotus ostreatus (IC50 µg/mL) = 39.46 ± 1.27 for DPPH; 11.22 ± 1.81 for ABTS; and 21.40 ± 2.20 for ORAC assays. This study encourages the use of these mushrooms in medicine in the light of their low cytotoxicity on normal PBMCs vis à vis their antiviral, antitumor and antioxidant capabilities.
Collapse
Affiliation(s)
- Shaza M. Elhusseiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University (ACU), 4th Industrial Area, 6th of October City, Cairo 2566, Egypt; (S.M.E.); (T.S.E.-M.)
| | - Taghrid S. El-Mahdy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University (ACU), 4th Industrial Area, 6th of October City, Cairo 2566, Egypt; (S.M.E.); (T.S.E.-M.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Mohamed F. Awad
- Department of Biology, College of Science, Taif University, Taif 11099, Saudi Arabia;
| | - Nooran S. Elleboudy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Cairo 11566, Egypt; (N.S.E.); (M.A.Y.)
| | - Mohamed M. S. Farag
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo 11884, Egypt;
| | - Mahmoud A. Yassein
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Cairo 11566, Egypt; (N.S.E.); (M.A.Y.)
| | - Khaled M. Aboshanab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Cairo 11566, Egypt; (N.S.E.); (M.A.Y.)
| |
Collapse
|
5640
|
Xiong Y, Zhu GH, Zhang YN, Hu Q, Wang HN, Yu HN, Qin XY, Guan XQ, Xiang YW, Tang H, Ge GB. Flavonoids in Ampelopsis grossedentata as covalent inhibitors of SARS-CoV-2 3CL pro: Inhibition potentials, covalent binding sites and inhibitory mechanisms. Int J Biol Macromol 2021; 187:976-987. [PMID: 34333006 PMCID: PMC8322037 DOI: 10.1016/j.ijbiomac.2021.07.167] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 10/25/2022]
Abstract
Coronavirus 3C-like protease (3CLpro) is a crucial target for treating coronavirus diseases including COVID-19. Our preliminary screening showed that Ampelopsis grossedentata extract (AGE) displayed potent SARS-CoV-2-3CLpro inhibitory activity, but the key constituents with SARS-CoV-2-3CLpro inhibitory effect and their mechanisms were unrevealed. Herein, a practical strategy via integrating bioactivity-guided fractionation and purification, mass spectrometry-based peptide profiling and time-dependent biochemical assay, was applied to identify the crucial constituents in AGE and to uncover their inhibitory mechanisms. The results demonstrated that the flavonoid-rich fractions (10-17.5 min) displayed strong SARS-CoV-2-3CLpro inhibitory activities, while the constituents in these fractions were isolated and their SARS-CoV-2-3CLpro inhibitory activities were investigated. Among all isolated flavonoids, dihydromyricetin, isodihydromyricetin and myricetin strongly inhibited SARS-CoV-2 3CLpro in a time-dependent manner. Further investigations demonstrated that myricetin could covalently bind on SARS-CoV-2 3CLpro at Cys300 and Cys44, while dihydromyricetin and isodihydromyricetin covalently bound at Cys300. Covalent docking coupling with molecular dynamics simulations showed the detailed interactions between the orthoquinone form of myricetin and two covalent binding sites (surrounding Cys300 and Cys44) of SARS-CoV-2 3CLpro. Collectively, the flavonoids in AGE strongly and time-dependently inhibit SARS-CoV-2 3CLpro, while the newly identified SARS-CoV-2 3CLpro inhibitors in AGE offer promising lead compounds for developing novel antiviral agents.
Collapse
Affiliation(s)
- Yuan Xiong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, China
| | - Guang-Hao Zhu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ya-Ni Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Hu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao-Nan Wang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao-Nan Yu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Ya Qin
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, China
| | - Xiao-Qing Guan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan-Wei Xiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Tang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, China.
| | - Guang-Bo Ge
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
5641
|
Yadav V, Balaraman E, Mhaske SB. Phosphine‐Free Manganese(II)‐Catalyst Enables Acceptorless Dehydrogenative Coupling of Alcohols with Indoles. Adv Synth Catal 2021. [DOI: 10.1002/adsc.202100621] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Vinita Yadav
- Division of Organic Chemistry CSIR-National Chemical Laboratory (CSIR-NCL) Pune 411008 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Ekambaram Balaraman
- Department of Chemistry Indian Institute of Science Education and Research (IISER) Tirupati Tirupati 517507 India
| | - Santosh B. Mhaske
- Division of Organic Chemistry CSIR-National Chemical Laboratory (CSIR-NCL) Pune 411008 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| |
Collapse
|
5642
|
Trebino MA, Shingare RD, MacMillan JB, Yildiz FH. Strategies and Approaches for Discovery of Small Molecule Disruptors of Biofilm Physiology. Molecules 2021; 26:molecules26154582. [PMID: 34361735 PMCID: PMC8348372 DOI: 10.3390/molecules26154582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/02/2022] Open
Abstract
Biofilms, the predominant growth mode of microorganisms, pose a significant risk to human health. The protective biofilm matrix, typically composed of exopolysaccharides, proteins, nucleic acids, and lipids, combined with biofilm-grown bacteria’s heterogenous physiology, leads to enhanced fitness and tolerance to traditional methods for treatment. There is a need to identify biofilm inhibitors using diverse approaches and targeting different stages of biofilm formation. This review discusses discovery strategies that successfully identified a wide range of inhibitors and the processes used to characterize their inhibition mechanism and further improvement. Additionally, we examine the structure–activity relationship (SAR) for some of these inhibitors to optimize inhibitor activity.
Collapse
Affiliation(s)
- Michael A. Trebino
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA 95064, USA;
| | - Rahul D. Shingare
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA;
| | - John B. MacMillan
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA;
- Correspondence: (J.B.M.); (F.H.Y.)
| | - Fitnat H. Yildiz
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA 95064, USA;
- Correspondence: (J.B.M.); (F.H.Y.)
| |
Collapse
|
5643
|
Pseudomonas aeruginosa Consumption of Airway Metabolites Promotes Lung Infection. Pathogens 2021; 10:pathogens10080957. [PMID: 34451421 PMCID: PMC8401524 DOI: 10.3390/pathogens10080957] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/03/2022] Open
Abstract
Prevailing dogma indicates that the lung of cystic fibrosis (CF) individuals is infected by multiple pathogens due to the abundant accumulation of mucus, which traps most of inhaled organisms. However, this hypothesis does not explain how specific opportunists, like Pseudomonas aeruginosa, are selected in the CF lung to cause chronic disease. This strongly suggests that other factors than mucus are accrued in the human airway and might predispose to bacterial disease, especially by P. aeruginosa. In this review we discuss the role of macrophage metabolites, like succinate and itaconate, in P. aeruginosa pneumonia. We analyze how dysfunction of the CF transmembrane conductance regulator (CFTR) favors release of these metabolites into the infected airway, and how P. aeruginosa exploits these elements to induce transcriptomic and metabolic changes that increase its capacity to cause intractable disease. We describe the host and pathogen pathways associated with succinate and itaconate catabolism, mechanisms of bacterial adaptation to these determinants, and suggest how both experimental settings and future therapies should consider macrophage metabolites abundance to better study P. aeruginosa pathogenesis.
Collapse
|
5644
|
Spiridon IA, Ciobanu DGA, Giușcă SE, Ferariu D, Pleşca IC, Căruntu ID. GIST and Ghrelin: To Be or Not to Be? Diagnostics (Basel) 2021; 11:1361. [PMID: 34441296 PMCID: PMC8393501 DOI: 10.3390/diagnostics11081361] [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: 06/08/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ghrelin is the orexigenic hormone secreted mainly by the stomach. Its involvement in neoplastic development has been studied in gastrointestinal adenocarcinomas. Our paper aims to evaluate the influence of the ghrelin axis in gastrointestinal stromal tumors (GISTs). MATERIALS AND METHODS The study design included two groups of patients, 46 with gastric GISTs and 30 with obesity. Archived tissue samples were evaluated for the presence of gastritis and H. pylori. Immunohistochemical expression of ghrelin and its receptor (GHS-R) was assessed. RESULTS All GISTs showed absent immunohistochemical expression for ghrelin, while GHS-R displayed a particular pattern, with notable differences in intensity (p = 0.0256) and percentage of stained cells (p < 0.00001) in the periphery vs. core of tumors. Positive ghrelin expression was lower in the gastric mucosa of the first group compared to the second group (p < 0.001). CONCLUSION The ghrelin axis can influence GISTs carcinogenesis through activation of GHS-R. A previously described direct autocrine/paracrine mechanism is not supported by our findings.
Collapse
Affiliation(s)
- Irene Alexandra Spiridon
- Department of Morpho-Functional Sciences I—Morphopathology, “Grigore T. Popa” University of Medicine and Pharmacy, Strada Universității 16, 700115 Iași, Romania;
| | - Delia Gabriela Apostol Ciobanu
- Department of Morpho-Functional Sciences I—Morphopathology, “Grigore T. Popa” University of Medicine and Pharmacy, Strada Universității 16, 700115 Iași, Romania;
| | - Simona Eliza Giușcă
- Department of Morpho-Functional Sciences I—Morphopathology, “Grigore T. Popa” University of Medicine and Pharmacy, Strada Universității 16, 700115 Iași, Romania;
| | - Dan Ferariu
- Department of Pathology, Regional Institute of Oncology, Str. General Henri Mathias Berthelot 2-4, 700483 Iași, Romania;
| | - Iulia Cătălina Pleşca
- Science Research Department, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University, Strada Lascăr Catargi 54, 700107 Iași, Romania;
| | - Irina Draga Căruntu
- Department of Morpho-Functional Sciences I—Histology, “Grigore T. Popa” University of Medicine and Pharmacy, Strada Universității 16, 700115 Iași, Romania;
| |
Collapse
|
5645
|
Davis TR, Pierce MR, Novak SX, Hougland JL. Ghrelin octanoylation by ghrelin O-acyltransferase: protein acylation impacting metabolic and neuroendocrine signalling. Open Biol 2021; 11:210080. [PMID: 34315274 PMCID: PMC8316800 DOI: 10.1098/rsob.210080] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The acylated peptide hormone ghrelin impacts a wide range of physiological processes but is most well known for controlling hunger and metabolic regulation. Ghrelin requires a unique posttranslational modification, serine octanoylation, to bind and activate signalling through its cognate GHS-R1a receptor. Ghrelin acylation is catalysed by ghrelin O-acyltransferase (GOAT), a member of the membrane-bound O-acyltransferase (MBOAT) enzyme family. The ghrelin/GOAT/GHS-R1a system is defined by multiple unique aspects within both protein biochemistry and endocrinology. Ghrelin serves as the only substrate for GOAT within the human proteome and, among the multiple hormones involved in energy homeostasis and metabolism such as insulin and leptin, acts as the only known hormone in circulation that directly stimulates appetite and hunger signalling. Advances in GOAT enzymology, structural modelling and inhibitor development have revolutionized our understanding of this enzyme and offered new tools for investigating ghrelin signalling at the molecular and organismal levels. In this review, we briefly summarize the current state of knowledge regarding ghrelin signalling and ghrelin/GOAT enzymology, discuss the GOAT structural model in the context of recently reported MBOAT enzyme superfamily member structures, and highlight the growing complement of GOAT inhibitors that offer options for both ghrelin signalling studies and therapeutic applications.
Collapse
Affiliation(s)
- Tasha R Davis
- Department of Chemistry, Syracuse University, Syracuse, NY 13244 USA
| | - Mariah R Pierce
- Department of Chemistry, Syracuse University, Syracuse, NY 13244 USA
| | - Sadie X Novak
- Department of Chemistry, Syracuse University, Syracuse, NY 13244 USA
| | - James L Hougland
- Department of Chemistry, Syracuse University, Syracuse, NY 13244 USA.,BioInspired Syracuse, Syracuse University, Syracuse, NY 13244 USA
| |
Collapse
|
5646
|
Selective MOR activity of DAPEA and Endomorphin-2 analogues containing a (R)-γ-Freidinger lactam in position two. Bioorg Chem 2021; 115:105219. [PMID: 34343741 DOI: 10.1016/j.bioorg.2021.105219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/17/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022]
Abstract
The use of α-amino-γ lactam of Freidinger (Agl) may serve as an impressive method to increase the biological stability of peptides and an appropriate tool to elucidate their structure-activity relationships. The endomorphin-2 (EM-2) and [D-Ala2, des-Leu5] enkephalin amide (DAPEA) are two linear opioid tetrapeptides agonists of MOR and MOR/DOR respectively. Herein, we investigated the influence of the incorporation of (R/S)-Agl in position 2 and 3 on the biological profile of the aforementioned products in vitro and in vivo. Receptor radiolabeled displacement and functional assays were used to measure in vitro the binding affinity and receptors activation of the novel analogues. The mouse tail flick and formalin tests allowed to observe their antinociceptive effect in vivo. Data revealed that peptide A2D was able to selectively bind and activate MOR with a potent antinociceptive effect after intracerebroventricular (i.c.v.) administration, performing better than the parent compounds EM-2 and DAPEA. Molecular docking calculations helped us to understand the key role exerted by the Freidinger Agl moiety in A2D for the interaction with the MOR binding pocket.
Collapse
|
5647
|
Yang W, Yoon Y, Lee Y, Oh H, Choi J, Shin S, Lee S, Lee H, Lee Y, Seo J. Photosensitizer-peptoid conjugates for photoinactivation of Gram-negative bacteria: structure-activity relationship and mechanistic studies. Org Biomol Chem 2021; 19:6546-6557. [PMID: 34259297 DOI: 10.1039/d1ob00926e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Multitarget engagement is considered an effective strategy to overcome the threat of bacterial infection, and antimicrobials with multiple mechanisms of action have been successful as natural chemical weaponry. Here, we synthesized a library of photosensitizer-peptoid conjugates (PsPCs) as novel antimicrobial photodynamic therapy (aPDT) agents. The peptoids, linkers, and photosensitizers were varied, and their structure-antimicrobial activity relationships against Escherichia coli were evaluated; PsPC 9 was indicated to be the most promising photoresponsive antimicrobial agent among the synthesized PsPCs. Spectroscopic analyses indicated that 9 generated singlet oxygen upon absorption of visible light (420 nm) while maintaining the weakly helical conformation of the peptoid. Mechanistic studies suggested that damage to the bacterial membrane and cleavage of DNA upon light irradiation were the main causes of bactericidal activity, which was supported by flow cytometry and DNA gel electrophoresis experiments. We demonstrated that the optimal combination of membrane-active peptoids and photosensitizers can generate an efficient aPDT agent that targets multiple sites of bacterial components and kills bacteria by membrane disruption and reactive oxygen species generation.
Collapse
Affiliation(s)
- Woojin Yang
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Younggun Yoon
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Yunjee Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Hyeongyeol Oh
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Jieun Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Sujin Shin
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), 49 Dosicheomdansaneopro, Nam-gu, Gwangju 61751, South Korea
| | - Hohjai Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Yunho Lee
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| | - Jiwon Seo
- Department of Chemistry, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, South Korea.
| |
Collapse
|
5648
|
Transcriptomics-Based Phenotypic Screening Supports Drug Discovery in Human Glioblastoma Cells. Cancers (Basel) 2021; 13:cancers13153780. [PMID: 34359681 PMCID: PMC8345128 DOI: 10.3390/cancers13153780] [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: 06/09/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) remains a particularly challenging cancer, with an aggressive phenotype and few promising treatment options. Future therapy will rely heavily on diagnosing and targeting aggressive GBM cellular phenotypes, both before and after drug treatment, as part of personalized therapy programs. Here, we use a genome-wide drug-induced gene expression (DIGEX) approach to define the cellular drug response phenotypes associated with two clinical drug candidates, the phosphodiesterase 10A inhibitor Mardepodect and the multi-kinase inhibitor Regorafenib. We identify genes encoding specific drug targets, some of which we validate as effective antiproliferative agents and combination therapies in human GBM cell models, including HMGCoA reductase (HMGCR), salt-inducible kinase 1 (SIK1), bradykinin receptor subtype B2 (BDKRB2), and Janus kinase isoform 2 (JAK2). Individual, personalized treatments will be essential if we are to address and overcome the pharmacological plasticity that GBM exhibits, and DIGEX will play a central role in validating future drugs, diagnostics, and possibly vaccine candidates for this challenging cancer. Abstract We have used three established human glioblastoma (GBM) cell lines—U87MG, A172, and T98G—as cellular systems to examine the plasticity of the drug-induced GBM cell phenotype, focusing on two clinical drugs, the phosphodiesterase PDE10A inhibitor Mardepodect and the multi-kinase inhibitor Regorafenib, using genome-wide drug-induced gene expression (DIGEX) to examine the drug response. Both drugs upregulate genes encoding specific growth factors, transcription factors, cellular signaling molecules, and cell surface proteins, while downregulating a broad range of targetable cell cycle and apoptosis-associated genes. A few upregulated genes encode therapeutic targets already addressed by FDA approved drugs, but the majority encode targets for which there are no approved drugs. Amongst the latter, we identify many novel druggable targets that could qualify for chemistry-led drug discovery campaigns. We also observe several highly upregulated transmembrane proteins suitable for combined drug, immunotherapy, and RNA vaccine approaches. DIGEX is a powerful way of visualizing the complex drug response networks emerging during GBM drug treatment, defining a phenotypic landscape which offers many new diagnostic and therapeutic opportunities. Nevertheless, the extreme heterogeneity we observe within drug-treated cells using this technique suggests that effective pan-GBM drug treatment will remain a significant challenge for many years to come.
Collapse
|
5649
|
In vitro, in vivo, and ADME evaluation of SF 5-containing N,N'-diarylureas as antischistosomal agents. Antimicrob Agents Chemother 2021; 65:e0061521. [PMID: 34310210 DOI: 10.1128/aac.00615-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In recent years, N,N'-diarylureas have emerged as a promising chemotype for the treatment of schistosomiasis, a disease that poses a considerable health burden to millions of people worldwide. Here, we report a novel series of N,N'-diarylureas featuring the scarcely explored pentafluorosulfanyl group. Low IC50 values for Schistosoma mansoni newly transformed schistosomula (0.6 - 7.7 μM) and adult worms (0.1 - 1.6 μM) were observed. Four selected compounds, highly active in presence of albumin (>70% at 10 μM), endowed with decent cytotoxicity profile (SI against L6 cells >8.5) and good microsomal hepatic stability (>62.5% of drug remaining after 60 min), were tested in S. mansoni infected mice. Despite the promising in vitro worm killing potency, none of them showed significant activity in vivo. Pharmacokinetic data showed a slow absorption, with maximal drug concentrations reached after 24 h of exposure. Finally, no direct correlation between drug exposure and in vivo activity was found. Thus, further investigations are needed to better understand the underlying mechanisms of SF5-containing N,N'-diarylureas.
Collapse
|
5650
|
Suto N, Kamoshita S, Hosoya S, Sakurai K. Exploration of the Reactivity of Multivalent Electrophiles for Affinity Labeling: Sulfonyl Fluoride as a Highly Efficient and Selective Label. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202104347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Nanako Suto
- Department of Bioengineering and Life Science Tokyo University of Agriculture and Technology 4-24-16, Naka-cho, Koganei-shi Tokyo 184-8588 Japan
| | - Shione Kamoshita
- Department of Bioengineering and Life Science Tokyo University of Agriculture and Technology 4-24-16, Naka-cho, Koganei-shi Tokyo 184-8588 Japan
| | - Shoichi Hosoya
- Institute of Research Tokyo Medical and Dental University 1-5-45, Yushima, Bunkyo-ku Tokyo 113-8510 Japan
| | - Kaori Sakurai
- Department of Bioengineering and Life Science Tokyo University of Agriculture and Technology 4-24-16, Naka-cho, Koganei-shi Tokyo 184-8588 Japan
| |
Collapse
|