1
|
Zhou C, Qin W, Tu C, Chen Y, Fu S, Liu B. Total Synthesis of Euphane Triterpenoids Using Metal-Catalyzed Hydrogen Atom Transfer. Angew Chem Int Ed Engl 2025; 64:e202503943. [PMID: 40110973 DOI: 10.1002/anie.202503943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/22/2025]
Abstract
Euphane triterpenoids are widely distributed in nature and show various intriguing bioactivities, but relatively few synthetic routes to them have been described. Here, we report asymmetric convergent total syntheses of euphanes involving two triterpenoids and two nortriterpenoids: euphol, 25,26,27-trisnor-3β-hydroxy-euphan-24-al, euphorbiumrin D, and 3-oxo-tirucall-7-ene-3,20-dione. The syntheses employ an enantioselective Antilla allylboration and intramolecular radical cyclization to construct ring A, a palladium-catalyzed Liebeskind stannane-thioester coupling to connect ring A with the bicyclic CD system, and a novel radical cascade with metal-catalyzed hydrogen atom transfer (MHAT) to complete the polycyclic architecture. The late-stage syntheses of both triterpenoids feature a diimide reduction and a MHAT/1,5-hydrogen transfer cascade to diastereoselectively forge the C20 and C17 stereogenic centers.
Collapse
Affiliation(s)
- Chengying Zhou
- Key Laboratory of Green Chemistry and Technology of the Ministry of Education, College of Chemistry Sichuan University, 29 Wangjiang Rd., Chengdu, Sichuan, 610064, China
| | - Weitian Qin
- Key Laboratory of Green Chemistry and Technology of the Ministry of Education, College of Chemistry Sichuan University, 29 Wangjiang Rd., Chengdu, Sichuan, 610064, China
| | - Canhui Tu
- Key Laboratory of Green Chemistry and Technology of the Ministry of Education, College of Chemistry Sichuan University, 29 Wangjiang Rd., Chengdu, Sichuan, 610064, China
| | - Yunxuan Chen
- Key Laboratory of Green Chemistry and Technology of the Ministry of Education, College of Chemistry Sichuan University, 29 Wangjiang Rd., Chengdu, Sichuan, 610064, China
| | - Shaomin Fu
- Key Laboratory of Green Chemistry and Technology of the Ministry of Education, College of Chemistry Sichuan University, 29 Wangjiang Rd., Chengdu, Sichuan, 610064, China
| | - Bo Liu
- Key Laboratory of Green Chemistry and Technology of the Ministry of Education, College of Chemistry Sichuan University, 29 Wangjiang Rd., Chengdu, Sichuan, 610064, China
| |
Collapse
|
2
|
Uti DE, Atangwho IJ, Alum EU, Ntaobeten E, Obeten UN, Bawa I, Agada SA, Ukam CIO, Egbung GE. Antioxidants in cancer therapy mitigating lipid peroxidation without compromising treatment through nanotechnology. DISCOVER NANO 2025; 20:70. [PMID: 40272665 PMCID: PMC12021792 DOI: 10.1186/s11671-025-04248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/27/2025]
Abstract
BACKGROUND Cancer treatments often exploit oxidative stress to selectively kill tumour cells by disrupting their lipid peroxidation membranes and inhibiting antioxidant enzymes. However, lipid peroxidation plays a dual role in cancer progression, acting as both a tumour promoter and a suppressor. Balancing oxidative stress through antioxidant therapy remains a challenge, as excessive antioxidant activity may compromise the efficacy of chemotherapy and radiotherapy. AIM This review explores the role of antioxidants in mitigating lipid peroxidation in cancer therapy while maintaining treatment efficacy. It highlights recent advancements in nanotechnology-based targeted antioxidant delivery to optimize therapeutic outcomes. METHODS A comprehensive literature review was conducted using reputable databases, including PubMed, Scopus, Web of Science, and ScienceDirect. The search focused on publications from the past five years (2020-2025), supplemented by relevant studies from earlier years. Keywords such as "antioxidants," "lipid peroxidation," "nanotechnology in cancer therapy," and "oxidative stress" were utilized. Relevant articles were critically analysed, and graphical illustrations were created. RESULTS Emerging evidence suggests that nanoparticles, including liposomes, polymeric nanoparticles, metal-organic frameworks, and others, can effectively encapsulate and control the release of antioxidants in tumour cells while minimizing systemic toxicity. Stimuli-responsive carriers with tumour-specific targeting mechanisms further enhance antioxidant delivery. Studies indicate that these strategies help preserve normal cells, mitigate oxidative stress-related damage, and improve treatment efficacy. However, challenges such as bioavailability, stability, and potential interactions with standard therapies remain. CONCLUSION Integrating nanotechnology with antioxidant-based interventions presents a promising approach for optimizing cancer therapy. Future research should focus on refining lipid peroxidation modulation strategies, assessing oxidative stress profiles during treatment, and employing biomarkers to determine optimal antioxidant dosing. A balanced approach to antioxidant use may enhance therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Daniel Ejim Uti
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria.
| | - Item Justin Atangwho
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Esther Ugo Alum
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda
| | - Emmanuella Ntaobeten
- Department of Cancer and Haematology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Uket Nta Obeten
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | - Inalegwu Bawa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | - Samuel A Agada
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | | | - Godwin Eneji Egbung
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
3
|
Gadiya Y, Genilloud O, Bilitewski U, Brönstrup M, von Berlin L, Attwood M, Gribbon P, Zaliani A. Predicting Antimicrobial Class Specificity of Small Molecules Using Machine Learning. J Chem Inf Model 2025; 65:2416-2431. [PMID: 39987507 PMCID: PMC11898080 DOI: 10.1021/acs.jcim.4c02347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/25/2025]
Abstract
While the useful armory of antibiotic drugs is continually depleted due to the emergence of drug-resistant pathogens, the development of novel therapeutics has also slowed down. In the era of advanced computational methods, approaches like machine learning (ML) could be one potential solution to help reduce the high costs and complexity of antibiotic drug discovery and attract collaboration across organizations. In our work, we developed a large antimicrobial knowledge graph (AntiMicrobial-KG) as a repository for collecting and visualizing public in vitro antibacterial assay. Utilizing this data, we build ML models to efficiently scan compound libraries to identify compounds with the potential to exhibit antimicrobial activity. Our strategy involved training seven classic ML models across six compound fingerprint representations, of which the Random Forest trained on the MHFP6 fingerprint outperformed, demonstrating an accuracy of 75.9% and Cohen's Kappa score of 0.68. Finally, we illustrated the model's applicability for predicting the antimicrobial properties of two small molecule screening libraries. First, the EU-OpenScreen library was tested against a panel of Gram-positive, Gram-negative, and Fungal pathogens. Here, we unveiled that the model was able to correctly predict more than 30% of active compounds for Gram-positive, Gram-negative, and Fungal pathogens. Second, with the Enamine library, a commercially available HTS compound collection with claimed antibacterial properties, we predicted its antimicrobial activity and pathogen class specificity. These results may provide a means for accelerating research in AMR drug discovery efforts by carefully filtering out compounds from commercial libraries with lower chances of being active.
Collapse
Affiliation(s)
- Yojana Gadiya
- Fraunhofer
Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, Hamburg 22525, Germany
- Bonn-Aachen
International Center for Information Technology (B-IT), University of Bonn, Bonn 53113, Germany
| | - Olga Genilloud
- Fundación
MEDINA, Centro de Excelencia En Investigación de Medicamentos
Innovadores En Andalucía, Avenida Del Conocimiento 34, Armilla 18016, Spain
| | - Ursula Bilitewski
- Helmholtz
Centre for Infection Research, Braunschweig 38124, Germany
| | - Mark Brönstrup
- Helmholtz
Centre for Infection Research, Braunschweig 38124, Germany
- German
Center for Infection Research, Hannover-Braunschweig Site, Hannover 38124, Germany
| | - Leonie von Berlin
- Fraunhofer
Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, Hamburg 22525, Germany
| | - Marie Attwood
- PK/PD Laboratory, North Bristol, NHS Trust, Southmead Hospital, Bristol BS10 5NB, U.K.
| | - Philip Gribbon
- Fraunhofer
Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, Hamburg 22525, Germany
| | - Andrea Zaliani
- Fraunhofer
Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, Hamburg 22525, Germany
| |
Collapse
|
4
|
Khadem S, Marles RJ. Biological activity of natural 2-quinolinones. Nat Prod Res 2025; 39:1359-1373. [PMID: 38824680 DOI: 10.1080/14786419.2024.2359545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/18/2024] [Accepted: 05/18/2024] [Indexed: 06/04/2024]
Abstract
While natural products have undeniably played a crucial role in drug discovery, challenges such as limited availability and complex synthesis methods have hindered the identification of lead compounds. At the core of numerous natural and synthetic compounds, each displaying distinct biological behaviours, lies the foundational structure of 2-quinolinone. Compounds with this structural motif exhibit a broad range of effects in different tissues. Furthermore, specific members showcase therapeutic potential for various disorders. Despite the significance of these compounds, the current review literature has not provided a comprehensive overview, underscoring the essential contribution of this article in exploring their biological functions. This study examines the biological activity of selected 2-quinolinone alkaloids across diverse organisms, unveiling their potential as a source of innovative bioactive natural products.
Collapse
Affiliation(s)
- Shahriar Khadem
- Safe Environments Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, Canada
| | - Robin J Marles
- Retired Senior Scientific Advisor, Health Canada, Ottawa, Canada
| |
Collapse
|
5
|
Sharma S, Beg MA, Latief I, Aboti J, Jamal S, Juneja P, Tanwar S, Sharma K, Rehman SU, Selvapandiyan A, Shafi S. Design, synthesis and biological activity of peptidyl β-nitrostyrenes as cysteine protease inhibitors against Leishmania donovani. RSC Adv 2025; 15:5703-5719. [PMID: 39981004 PMCID: PMC11837772 DOI: 10.1039/d4ra06510g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/15/2024] [Indexed: 02/22/2025] Open
Abstract
Cysteine proteases are essential for the survival of Leishmania parasites that cause several clinical forms of leishmaniases. Inhibiting cysteine protease can be a promising strategy against parasitic diseases because of their essential functions in the life cycles of these pathogens. The aim of the present study was to synthesize and evaluate peptidyl -nitrostyrenes as antipromastigote inhibitors against Leishmania donovani promastigotes. A library of 12 peptidyl β-nitrostyrenes was synthesized and evaluated for anti-promastigote activity. Most of the compounds exhibited comparable activity to the standard, with IC50 values ranging from 1.468 to 16.81 μM. Notably, compounds 14a, 14e, 14f, and 14g showed significant activity against both L. donovani promastigotes and intracellular amastigotes. Compounds 14e and 14f displayed superior anti-promastigote activity with IC50 values of 1.468 μM and 1.551 μM, respectively, compared to the standard (IC50 = 3.073 μM). Moreover, compounds 14e and 14f demonstrated better inhibitory potential against intracellular amastigotes, with IC50 values of 1.28 μM and 0.64 μM, respectively, outperforming AmphoB (IC50 = 3.07 μM). Additionally, compounds 14a and 14g showed negligible cytotoxicity to mammalian macrophages even at a concentration of 28 μM. Given their high activity, favorable safety profiles, and cost-effective synthesis, this class of compounds holds promise for the development of anti-leishmanial drugs.
Collapse
Affiliation(s)
- Sweta Sharma
- Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| | - Mirza A Beg
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard New Delhi 110062 India
| | - Insha Latief
- Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| | - Jyoti Aboti
- Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| | - Samra Jamal
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| | - Pallavi Juneja
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| | - Supriya Tanwar
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy Moga Punjab India
| | - Sayeed Ur Rehman
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| | - Angamuthu Selvapandiyan
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard New Delhi 110062 India
| | - Syed Shafi
- Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard New Delhi India
| |
Collapse
|
6
|
Vrablova L, Gonec T, Kauerova T, Oravec M, Jendrzejewska I, Kollar P, Cizek A, Jampilek J. Antimicrobial and ADME properties of methoxylated, methylated and nitrated 2-hydroxynaphthalene-1 carboxanilides. ADMET AND DMPK 2025; 13:2642. [PMID: 40161889 PMCID: PMC11954145 DOI: 10.5599/admet.2642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/04/2025] [Indexed: 04/02/2025] Open
Abstract
Background and purpose Many new compounds are being prepared to overcome the problem of increasing microbial resistance and the increasing number of infections. Experimental approach This study includes a series of twenty-seven mono-, di- and trisubstituted 2-hydroxynaphthalene-1-carboxanilides designed as multitarget agents. The compounds are substituted with methoxy, methyl, and nitro groups, as well as additionally with chlorine, bromine, and trifluoromethyl at various positions. All the compounds were evaluated for antibacterial activities against Gram-positive and Gram-negative bacteria and mycobacteria. Cytotoxicity on human cells was also tested. Key results Three compounds showed activity comparable to clinically used drugs. N-(3,5-Dimethylphenyl)-2-hydroxynaphthalene-1-carboxamide (13) showed only antistaphylococcal activity (minimum inhibitory concentration (MIC) = 54.9 μM); 2-hydroxy-N-[2-methyl-5-(trifluoromethyl)phenyl]naphthalene-1-carboxamide (22) and 2-hydroxy-N-[4-nitro-3-(trifluoromethyl)phenyl]naphthalene-1-carboxamide (27) were active across the entire spectrum of tested bacteria/mycobacteria, both against the sensitive set and against resistant isolates (MICs range 0.3 to 92.6 μM). Compound 22 was even active against E. coli (MIC = 23.2 μM). The active agents showed no in vitro cytotoxicity up to a concentration of 30 μM. Conclusion Compounds with trifluoromethyl in the meta-anilide position, experimental lipophilicity expressed as log k (logarithm of the capacity factor) in the range of 0.31 to 0.34 and calculated electron σ parameter for the anilide substituent higher than 0.59 were effective. The investigated compounds meet the definition of Michael acceptors. Based on ADME screening, the investigated compounds 13, 22 and 27 should have suitable physicochemical parameters for good bioavailability in the organism. Therefore, these are promising agents for further study.
Collapse
Affiliation(s)
- Lucia Vrablova
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 00 Brno, Czech Republic
| | - Tereza Kauerova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 00 Brno, Czech Republic
| | - Michal Oravec
- Global Change Research Institute CAS, Belidla 986/4a, 603 00 Brno, Czech Republic
| | | | - Peter Kollar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 00 Brno, Czech Republic
| | - Alois Cizek
- Department of Infectious Diseases and Microbiology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Palackeho tr. 1946/1, 612 42 Brno, Czech Republic
| | - Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 779 00 Olomouc, Czech Republic
| |
Collapse
|
7
|
Nunes JA, Santos-Júnior PFDS, Gomes MC, Ferreira LAS, Padilha EKA, Teixeira TR, Stanger EJ, Kaur Y, Silva EBD, Costa CACB, Freitas JDD, Araújo-Júnior JXD, Mendonça-Junior FJB, Giardini MA, Siqueira-Neto JL, Caffrey CR, Zhan P, Cardoso SH, Silva-Júnior EFD. Nanomolar activity of coumarin-3-thiosemicarbazones targeting Trypanosoma cruzi cruzain and the T. brucei cathepsin L-like protease. Eur J Med Chem 2025; 283:117109. [PMID: 39653622 DOI: 10.1016/j.ejmech.2024.117109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025]
Abstract
Trypanosoma cruzi (T. cruzi) and Trypanosoma brucei (T. brucei) urgently demand innovative drug development due to their impact on public health worldwide. Their cysteine proteases, Cruzain (CRZ) and the T. brucei Cathepsin L-like protease (TbrCATL) are established drug targets for these parasites. In this study, our coumarin-3-thiosemicarbazones demonstrated nanomolar IC50 values (22-698 nM) toward these proteases. Against T. cruzi amastigotes and T. brucei trypomastigotes, LASF-01 displayed a promising result. Herein, MCG-02, the most potent TbrCATL inhibitor, underwent comprehensive analyses, including cytotoxicity assessments and in vitro tests. Molecular dynamics (MD) simulations and a multiscale Quantum Mechanics/Quantum Mechanics (QM/QM) approach were used to generate insights into their binding modes. These suggested that MCG-02 could be a reversible, competitive covalent inhibitor. Further, confirmatory assays were experimentally performed changing different parameters to prove its efficacy. Additionally, the predicted pharmacokinetic profile showed that there is no violation of the Lipinski rule of five. Notably, coumarin-3-thiosemicarbazone hybrids emerged as promising candidates for designing highly active inhibitors against CRZ and TbrCATL. Overall, the integration of in silico and experimental approaches enhanced our understanding regarding the binding modes of MCG-02, which were experimentally corroborated, providing valuable insights for future drug development.
Collapse
Affiliation(s)
- Jéssica Alves Nunes
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, Alagoas, Maceió, 57072-970, Brazil
| | - Paulo Fernando da Silva Santos-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, Alagoas, Maceió, 57072-970, Brazil
| | - Midiane Correa Gomes
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, Alagoas, Maceió, 57072-970, Brazil
| | - Luiz Alberto Santos Ferreira
- Laboratory of Organic and Medicinal Synthesis, Federal University of Alagoas, Campus Arapiraca, Manoel Severino Barbosa Avenue, Arapiraca, 57309-005, Brazil
| | - Emanuelly Karla Araújo Padilha
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, Alagoas, Maceió, 57072-970, Brazil
| | - Thaiz Rodrigues Teixeira
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Emily J Stanger
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Yashpreet Kaur
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Elany Barbosa da Silva
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Johnnatan Duarte de Freitas
- Department of Chemistry, Federal Institute of Alagoas, Maceió Campus, Mizael Domingues Street, 57020-600, Maceió, Alagoas, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, Alagoas, Maceió, 57072-970, Brazil
| | | | - Miriam A Giardini
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jair L Siqueira-Neto
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Sílvia Helena Cardoso
- Laboratory of Organic and Medicinal Synthesis, Federal University of Alagoas, Campus Arapiraca, Manoel Severino Barbosa Avenue, Arapiraca, 57309-005, Brazil.
| | - Edeildo Ferreira da Silva-Júnior
- Biological and Molecular Chemistry Research Group, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões Campus, Alagoas, Maceió, 57072-970, Brazil.
| |
Collapse
|
8
|
Shaff AB, Hazra A, Gardner BW, Lalic G. Selective Synthesis of Z-Michael Acceptors via Hydroalkylation of Conjugated Alkynes. J Am Chem Soc 2025; 147:27-32. [PMID: 39721993 DOI: 10.1021/jacs.4c09755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Hydroalkylation of terminal alkynes is a powerful approach to the synthesis of disubstituted alkenes. However, its application is largely unexplored in the synthesis of α,β-unsaturated carbonyls, which are common among synthetic intermediates and biologically active molecules. The thermodynamically less stable Z-isomers of activated alkenes have been particularly challenging to access because of their propensity for isomerization and the paucity of reliable Z-selective hydroalkylation methods. We developed a highly Z-selective silver-catalyzed hydroalkylation of terminal conjugated alkynes using alkyl boranes as coupling partners. The reaction allows access to (Z)-α,β-unsaturated esters, secondary and tertiary alkyl amides, aryl amides, and alkyl and aryl ketones and tolerates a wide range of functional groups. The reaction can be performed successfully in the presence of alkyl and aryl halides, esters, protected alcohols, and amines. The hydroalkylation involves the formation of an alkynylboronate complex followed by a 1,2-metalate shift. This sequence of steps mechanistically constrains the stereochemical outcome, which, together with mild reaction conditions, ensures high Z-selectivity.
Collapse
Affiliation(s)
- Austin B Shaff
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Avijit Hazra
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Bradley W Gardner
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Gojko Lalic
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
9
|
Danilov RD, Smirnova IE, Galimova ZI, Sokolova EV, Lukyanov AV, Kalitin KY, Mukha OY, Babkov DA, Kazakova OB, Spasov AA. A Novel Dipterocarpol Derivative That Targets Alpha-Glucosidase and NLRP3 Inflammasome Activity for Treatment of Diabetes Mellitus. Chem Biodivers 2025; 22:e202401626. [PMID: 39269647 DOI: 10.1002/cbdv.202401626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 09/15/2024]
Abstract
Type 2 diabetes mellitus is a chronic metabolic disorder characterized by persistent hyperglycemia, chronic inflammation, impaired insulin secretion, and/or peripheral insulin resistance. Current α-glucosidase inhibitors approved for clinical use exhibit limited efficacy compared to other glucose-lowering agents. In this study, a series of mono- and bis-benzylidene derivatives were synthesized via aldol condensation of 3-oxo-dammarane triterpenoids with terephthalic aldehyde. The target mono- and bis-benzylidene derivatives, based on the dammarane triterpenoids hollongdione 1, (20S)-23,24-epoxy-25,26,27-trinordammar-3,24-dione 2, and 24(R,S)-20(S)-epoxy-25-hydroxy-dammar-3-one 3, were successfully synthesized. Several of these inhibitors demonstrated significantly greater efficacy than the reference drug acarbose. Notably, compound 4 inhibited S. cerevisiae α-glucosidase with an IC50 of 2.67 μM. Furthermore, the target compounds effectively inhibited NLRP3 inflammasome activation, reducing IL-1β production in LPS+ATP-stimulated murine peritoneal macrophages without detectable cytotoxicity. Compound 8, which exhibited dual activity, was further characterized as an inhibitor of NLRP3 activation in peripheral blood mononuclear cells, leading to the prevention of pyroptosis and IL-1β release. Additionally, compound 8 was shown to promote neuronal survival in LPS+ATP-treated rat hippocampal slices, highlighting its potential as a promising antidiabetic agent that targets both postprandial hyperglycemia and metaflammation.
Collapse
Affiliation(s)
- Roman D Danilov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39, Novorossiyskaya, Volgograd, 400087, Russian Federation
| | - Irina E Smirnova
- Ufa Institute of Chemistry, UFRC RAS, 71, pr. Oktyabrya, Ufa, 450054, Russian Federation
| | - Zarema I Galimova
- Ufa Institute of Chemistry, UFRC RAS, 71, pr. Oktyabrya, Ufa, 450054, Russian Federation
| | - Elena V Sokolova
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39, Novorossiyskaya, Volgograd, 400087, Russian Federation
| | - Andrey V Lukyanov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39, Novorossiyskaya, Volgograd, 400087, Russian Federation
| | - Konstantin Y Kalitin
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39, Novorossiyskaya, Volgograd, 400087, Russian Federation
| | - Olga Y Mukha
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39, Novorossiyskaya, Volgograd, 400087, Russian Federation
| | - Denis A Babkov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39, Novorossiyskaya, Volgograd, 400087, Russian Federation
| | - Oxana B Kazakova
- Ufa Institute of Chemistry, UFRC RAS, 71, pr. Oktyabrya, Ufa, 450054, Russian Federation
| | - Alexander A Spasov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39, Novorossiyskaya, Volgograd, 400087, Russian Federation
| |
Collapse
|
10
|
Kos J, Strharsky T, Tosso R, Gutierrez L, Kos D, Jurica J, Zendulka O, Pes O, Gregorova J, Degotte G, Gonec T, Oravec M, Vojackova V, Krystof V, Cizek A, Francotte P, Frederich M, Jampilek J, Enriz D. Trifluoromethylcinnamanilides - Effective dual inhibitors of Mycobacterium smegmatis and Plasmodium falciparum. Bioorg Chem 2025; 154:107957. [PMID: 39615279 DOI: 10.1016/j.bioorg.2024.107957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 01/15/2025]
Abstract
A series of eighteen new 2-trifluoromethylcinnamanilides (1a-r) were synthesized by microwave synthesis and investigated for their antimycobacterial and antimalarial activities, along with the complementary (2E)-3-[3-(trifluoromethyl)phenyl]-N-arylprop-2-enanilides (2a-r) and (2E)-3-[4-(trifluoromethyl)phenyl]-N-arylprop-2-enanilides (3a-r) prepared earlier. All the compounds were evaluated in vitro against Mycobacterium smegmatis ATCC 700084 and a chloroquine-sensitive strain of Plasmodium falciparum 3D7/MRA-102. The most active compounds against M. smegmatis (MIC values in the range of 1.17-11.1 µM, more effective than rifampicin) were anilides substituted by 3,5-CF3 (1q, 2q, 3q), 4-OCF3 (1k), and 4-CF3 (1j, 2j). The most effective agents against P. falciparum (IC50 values in the range of 0.32-4.5 µM, comparable to chloroquine) were anilides substituted by 3,5-CF3 (1q, 2q, 3q), 2-Br-4-OCF3 (1r), 4-CF3 (1j, 3j), 4-F (2d), 4-Cl (2g), 2-Cl (1e, 2e). A preliminary in vitro cytotoxicity screening was assessed using human leukemic cell lines and human dermal fibroblasts, revealing the toxic effect of 3,5-CF3 substituted anilides. On the other hand, the other investigated agents showed insignificant cytotoxic effects. Stability assays using rat liver microsomes demonstrated that compounds 1r (R = 2-Br-4-OCF3) and 1q (R = 3,5-CF3) are neither metabolized nor affect cytochrome P450 metabolizing capacity in vitro. Furthermore, complex in silico studies were performed - a combined approach (docking/MD simulations/QTAIM calculations) helped to define the molecular interactions that were applied during the binding of active agents and the subsequent inhibition of their molecular targets - InhA (activity against M. smegmatis) and arginase (activity against P. falciparum). In conclusion, promising active agents with dual antimycobacterial and antimalarial effects were identified.
Collapse
Affiliation(s)
- Jiri Kos
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Tomas Strharsky
- Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho 1946/1, 612 00 Brno, Czech Republic.
| | - Rodrigo Tosso
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, IMIBIO-CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina.
| | - Lucas Gutierrez
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, IMIBIO-CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina.
| | - Dominika Kos
- Department of Molecular Pharmacy, Faculty of Pharmacy, Masaryk University, Palackeho 1946/1, 612 00 Brno, Czech Republic.
| | - Jan Jurica
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Ondrej Zendulka
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Ondrej Pes
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Jana Gregorova
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Gilles Degotte
- Renslo's Lab, Department of Pharmaceutical Chemistry, University California San Francisco, 600 16(th) Street, 94143 San Francisco, CA, USA.
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho 1946/1, 612 00 Brno, Czech Republic.
| | - Michal Oravec
- Global Change Research Institute CAS, Belidla 986/4a, 603 00 Brno, Czech Republic.
| | - Veronika Vojackova
- Department of Experimental Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 779 00 Olomouc, Czech Republic.
| | - Vladimir Krystof
- Department of Experimental Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 779 00 Olomouc, Czech Republic.
| | - Alois Cizek
- Department of Infectious Diseases and Microbiology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Palackeho tr. 1946/1, 612 42 Brno, Czech Republic.
| | - Pierre Francotte
- Laboratory of Medicinal Chemistry, CIRM - Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, 4000 Liege, Belgium.
| | - Michel Frederich
- Laboratory of Pharmacognosy, CIRM - Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, 4000 Liege, Belgium.
| | - Josef Jampilek
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 779 00 Olomouc, Czech Republic; Institute of Chemistry, University of Silesia, Szkolna 9, 40-007 Katowice, Poland.
| | - Daniel Enriz
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, IMIBIO-CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina.
| |
Collapse
|
11
|
Chakkittukandiyil A, Sajini DV, Rymbai E, Sugumar D, Mathew J, Arumugam S, Ramachandran V, Selvaraj D. Synthesis and evaluation of novel ethyl ferulate derivatives as potent Keap1 inhibitors to activate the Nrf2/ARE pathway in Parkinson's disease. Toxicol Appl Pharmacol 2025; 494:117172. [PMID: 39603427 DOI: 10.1016/j.taap.2024.117172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
The Kelch-like ECH-associated protein 1/Nuclear factor erythroid 2 related factor 2/Antioxidant Response Elements (Keap1/Nrf2/ARE) pathway is essential for neuronal resilience against the complex pathogenesis of Parkinson's disease (PD). Activating this pathway by covalently modifying Keap1 cysteine residues is a promising strategy for regulating neuroprotective gene expression. Our study aimed to identify phytochemicals that could irreversibly inhibit Keap1. A preliminary docking analysis revealed that ethyl ferulate could covalently bind with Cys151 of Keap1 by Michael's addition reaction. Further, we designed several ethyl ferulate derivatives with improved lipophilicity and assessed their binding affinity with Keap1. The molecules with good binding scores were synthesized and structures were confirmed through 1H NMR, 13C NMR, FT-IR, and mass spectroscopy. Neuroprotection screening was conducted in all-trans retinoic acid differentiated SH-SY5Y cells using rotenone as a disease-inducing agent. Pre-treatment with compounds C2 and C4 significantly mitigated rotenone toxicity. Additionally, C2 and C4 decreased rotenone-induced ROS production and mitochondrial membrane potential loss. C2 and C4 also induced Nrf2 nuclear translocation in SH-SY5Y cells and increased mRNA expression of heme oxygenase-1, an Nrf2-regulated antioxidant response element. In vivo, pretreatment with C2 (50, 100 mg/kg, p.o.) and C4 (50, 100 mg/kg, p.o.) protected against neurodegenerative phenotypes associated with rotenone (1.5 mg/kg, s.c.) induction in Wistar rats. Results indicate, C2 and C4 dose-dependently improved muscle rigidity, catalepsy, and cognitive deficits in rotenone-induced Wistar rats, and mitigated dopaminergic neurodegeneration in the substantia nigra. These findings highlight the potential of ethyl ferulate derivatives in modulating oxidative stress and neurodegeneration in PD via activation of Nrf2.
Collapse
Affiliation(s)
- Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Deepak Vasudevan Sajini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Deepa Sugumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Jinu Mathew
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Suresh Arumugam
- Department of Pharmacology, The Kaavery Pharmacy College, Mecheri, Salem, Tamil Nadu, India
| | - Vadivelan Ramachandran
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India.
| |
Collapse
|
12
|
Wu J, Han M, Tan X, Zeng L, Yang Z, Zhong H, Jiang X, Yao S, Liu W, Li W, Liu X, Wu W. Green synthesis of neuroprotective spirocyclic chalcone derivatives and their role in protecting against traumatic optic nerve injury. Eur J Med Chem 2024; 280:116933. [PMID: 39368262 DOI: 10.1016/j.ejmech.2024.116933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/30/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
For clinically prevalent traumatic optic neuropathy (TON) and other retinal and optic nerve injuries lacking effective therapeutic agents, there is an urgent clinical demand for developing highly efficient and safe neuroprotective agents. Here, we have integrated naturally sourced chalcone with isatin through a catalyst-free green synthesis method, reporting a series of spirocyclic chalcone derivatives with significantly lower cytotoxicity than chalcone itself. Following in vitro cell protection assays in models of hydrogen peroxide and glutamic acid-induced damage, multiple active compounds capable of combating both forms of damage were identified. Among these, candidate compound X38 demonstrated promising neuroprotective prospects: in vitro, it attenuated glutamate-induced cell apoptosis, while in vivo, it effectively ameliorated retinal thinning and loss of optic nerve electrophysiological function induced by optic nerve injury. Preliminary mechanistic studies suggest that X38 exerts its neuroprotective effects by mitigating intracellular ROS accumulation, inhibiting JNK phosphorylation, and alleviating oxidative stress. Additionally, acute toxicity studies (intraperitoneal injection, 500 mg/kg) underscored the favorable in vivo safety profile of X38. Taken together, this study has designed a class of safe, neuroprotective spirocyclic chalcone derivatives that can be synthesized using green methods, offering an attractive candidate for treating retinal and optic nerve injuries.
Collapse
Affiliation(s)
- Jianzhang Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
| | - Meiting Han
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiangpeng Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Ling Zeng
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhenzhen Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Hongliang Zhong
- The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiaohui Jiang
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shuang Yao
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Weibin Liu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wulan Li
- The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xin Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| | - Wencan Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| |
Collapse
|
13
|
Tong L, Zha ML, Hu J, Li HY, Kuai L, Li B, Dang Y, Zhao Q, Liao R, Lin GQ, He QL. Adenanthin exhibits anti-inflammatory effects by covalently targeting the p65 subunit in the NF-κB signaling pathway. Eur J Med Chem 2024; 280:116946. [PMID: 39383653 DOI: 10.1016/j.ejmech.2024.116946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Adenanthin is a structurally unique ent-kaurane diterpenoid isolated from Rabdosia adenantha, a traditional Chinese medicinal plant with potent anti-cancer and anti-inflammatory activities. However, its anti-inflammatory molecular mechanism remains largely elusive to date. Here, we developed an affinity-based label-free protein profiling (ALFPP) to identify potential covalent targets of electrophilic natural products with ketone or aldehyde groups. Using ALFPP, we identified 27 potential covalent targets of adenanthin, among which p65 (RelA) has been associated with its anti-inflammatory activities. Through a series of experiments, including LC-MS/MS, molecular docking, electrophoretic mobility shift assays (EMSA), and genome editing, we demonstrated that adenanthin could covalently modify the Cys38 residue of p65 to affect the binding of DNA to p65, thereby inhibiting the NF-κB signaling pathway. ALFPP will facilitate the target identification of electrophilic carbonylated natural products, especially those containing α, β-unsaturated keto groups. Furthermore, the elucidation of the molecular mechanism of adenanthin will contribute to new drug development of adenanthin to treat inflammations and cancers, enhancing the possibility for its clinical application.
Collapse
Affiliation(s)
- Lu Tong
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Meng-Li Zha
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Junchi Hu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Hai-Yang Li
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Bin Li
- Shanghai Skin Disease Hospital, Tongji University, Shanghai, 200443, China
| | - Yongjun Dang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Qunfei Zhao
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Rijing Liao
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.
| | - Guo-Qiang Lin
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Qing-Li He
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
14
|
Wang X, Li X, Zhang X, Wang X, Yang J, Liu G. Design, synthesis and biological evaluation of novel curcumin-fluorouracil hybrids as potential anti-cancer agents. Biochem Pharmacol 2024; 230:116559. [PMID: 39326677 DOI: 10.1016/j.bcp.2024.116559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
The latest global cancer data statistics report shows that cancer poses a serious threat to human life and health; The number of new cancer and death cases worldwide is severe. Molecular hybridization is considered an effective strategy for developing new anti-cancer drugs. Curcumin (Cur) is a natural active compound containing Michael receptors that target thioredoxin reductase (TrxR). Fluorouracil (5-FU) is the first anti-metabolic drug synthesized based on certain assumptions for tumor treatment, acting on thymidylate synthase (TS). This study synthesized a series of novel hybrid derivatives of Cur and 5-FU, and evaluated their anti-tumor cell proliferation effects. Several compounds with good cytotoxic activity against tumor cells were discovered; and they exhibited high selectivity towards A549 cells, compared to normal THLE cells. Among them, the hybrid derivative F-4 has the best anti-proliferative activity in tumor cells. F-4 can target TrxR, increase reactive oxygen species levels in tumor cells, and lead to tumor cell apoptosis, which may be related to the Michael receptor structure in the chemical structure of F-4; F-4 can also target TS, leading to cell cycle arrest in G0/G1 phase, which may be related to the 5-FU structure in the chemical structure of F-4. Moreover, F-4 can effectively exert anti-tumor activity in mice, significantly reduce tumor volume and weight, and has low toxic side effects. These results indicate that Cur-5-FU hybrid derivative F-4 is a novel lead compound with in vivo anti-tumor activity and minimal side effects, which deserves further investigation.
Collapse
Affiliation(s)
- Xiaotong Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Xin Li
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Xu Zhang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Xuekun Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China.
| | - Jie Yang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China; Liaocheng Key Laboratory of Quality Control and Pharmacodynamic Evaluation of Ganoderma lucidum, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252059, China.
| | - Guoyun Liu
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China; Liaocheng Key Laboratory of Quality Control and Pharmacodynamic Evaluation of Ganoderma lucidum, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252059, China.
| |
Collapse
|
15
|
Acharya A, Nagpure M, Roy N, Gupta V, Patranabis S, Guchhait SK. How to nurture natural products to create new therapeutics: Strategic innovations and molecule-to-medicinal insights into therapeutic advancements. Drug Discov Today 2024; 29:104221. [PMID: 39481593 DOI: 10.1016/j.drudis.2024.104221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Natural products (NPs) are privileged structures interacting with biomacromolecular targets and exhibiting biological effects important for human health. In this review, we have presented NP-inspired strategic innovations that are promising for addressing preclinical and clinical challenges. An analysis of 'molecule-to-medicinal' properties for improvement of P3 and absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiles has been illustrated. The strategies include chemical evolution through knowledge of structure-medicinal properties, truncation of NPs to avoid molecular obesity, pseudo-NPs, selection of common structural features of NPs, medicinophore installation, scaffold hopping, and induced proximity. Molecule-to-medicinal property analysis can guide the development of 'nature-to-new' chemical therapeutics. Coupled with scientific advances and innovations in instrumentation, these strategies hold great potential for enhancing drug design and discovery.
Collapse
Affiliation(s)
- Ayan Acharya
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Mithilesh Nagpure
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Nibedita Roy
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Vaibhav Gupta
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Soumyadeep Patranabis
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Sankar K Guchhait
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India.
| |
Collapse
|
16
|
Al-Awadhi F, Kokkaliari S, Ratnayake R, Paul VJ, Luesch H. Isolation and Characterization of the Cyanobacterial Macrolide Glycoside Moorenaside, an Anti-Inflammatory Analogue of Aurisides Targeting the Keap1/Nrf2 Pathway. JOURNAL OF NATURAL PRODUCTS 2024; 87:2355-2365. [PMID: 39315953 PMCID: PMC11519913 DOI: 10.1021/acs.jnatprod.4c00420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/07/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024]
Abstract
A new 14-membered ring brominated macrolide glycoside, named moorenaside (1), was discovered from a marine cyanobacterial sample collected from Shands Key in Florida. The structure of 1 was established by analysis of spectroscopic data including its relative configuration. The absolute configuration was inferred from optical rotation data and comparison with related compounds. The structure of 1 features an α,β-unsaturated carbonyl system, which is also found in aurisides. The presence of this motif in 1 prompted us to evaluate its effect on Keap1/Nrf2 signaling, a cytoprotective pathway culminating in the activation of antioxidant genes activated upstream by the cysteine alkylation of Keap1. Moorenaside exhibited moderate ARE luciferase activity at 32 μM. Due to the established crosstalk between Nrf2 and NF-κB pathways, we investigated the anti-inflammatory effects of 1 in LPS-induced mouse macrophages (RAW264.7 cells), a commonly used model for inflammation. Moorenaside significantly upregulated Nqo1 (Nrf2 target gene) and downregulated iNos (NF-κB target gene) at 32 μM by 5.0- and 2.5-fold, respectively, resulting in a significant reduction of nitric oxide (NO) levels. Furthermore, we performed RNA-sequencing and demonstrated the transcriptional activity of 1 on a global level and identified canonical pathways and upstream regulators involved in inflammation, immune response, and certain oxidative-stress-underlying diseases such as multiple sclerosis and chronic kidney disease.
Collapse
Affiliation(s)
- Fatma
H. Al-Awadhi
- Department
of Medicinal Chemistry and Center for Natural Products, Drug Discovery
and Development (CNPD3), University of Florida, Gainesville, Florida 32610, United States
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| | - Sofia Kokkaliari
- Department
of Medicinal Chemistry and Center for Natural Products, Drug Discovery
and Development (CNPD3), University of Florida, Gainesville, Florida 32610, United States
| | - Ranjala Ratnayake
- Department
of Medicinal Chemistry and Center for Natural Products, Drug Discovery
and Development (CNPD3), University of Florida, Gainesville, Florida 32610, United States
| | - Valerie J. Paul
- Smithsonian
Marine Station, Fort Pierce, Florida 34949, United States
| | - Hendrik Luesch
- Department
of Medicinal Chemistry and Center for Natural Products, Drug Discovery
and Development (CNPD3), University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
17
|
Chen K, Zhang L, Ding Y, Sun Z, Meng J, Luo R, Zhou X, Liu L, Yang S. Activity-based protein profiling in drug/pesticide discovery: Recent advances in target identification of antibacterial compounds. Bioorg Chem 2024; 151:107655. [PMID: 39032407 DOI: 10.1016/j.bioorg.2024.107655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/18/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Given the escalating incidence of bacterial diseases and the challenge posed by pathogenic bacterial resistance, it is imperative to identify appropriate methodologies for conducting proteomic investigations on bacteria, and thereby promoting the target-based drug/pesticide discovery. Interestingly, a novel technology termed "activity-based protein profiling" (ABPP) has been developed to identify the target proteins of active molecules. However, few studies have summarized advancements in ABPP for identifying the target proteins in antibacterial-active compounds. In order to accelerate the discovery and development of new drug/agrochemical discovery, we provide a concise overview of ABPP and its recent applications in antibacterial agent discovery. Diversiform cases were cited to demonstrate the potential of ABPP for target identification though highlighting the design strategies and summarizing the reported target protein of antibacterial compounds. Overall, this review is an excellent reference for probe design towards antibacterial compounds, and offers a new perspective of ABPP in bactericide development.
Collapse
Affiliation(s)
- Kunlun Chen
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Ling Zhang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Yue Ding
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Zhaoju Sun
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Jiao Meng
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Rongshuang Luo
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Xiang Zhou
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| | - Liwei Liu
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Song Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| |
Collapse
|
18
|
Chihab A, El Brahmi N, Hamdoun G, El Abbouchi A, Ghammaz H, Touil N, Bousmina M, El Fahime E, El Kazzouli S. Development of a new experimental NMR strategy for covalent cysteine protease inhibitors screening: toward enhanced drug discovery. RSC Adv 2024; 14:26829-26836. [PMID: 39184001 PMCID: PMC11342919 DOI: 10.1039/d4ra04938a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
In the development of antiviral drugs, proteases and polymerases are among the most important targets. Cysteine proteases, also known as thiol proteases, catalyze the degradation of proteins by cleaving peptide bonds using the nucleophilic thiol group of cysteine. As part of our research, we are examining how cysteine, an essential amino acid found in the active site of the main protease (Mpro) enzyme in SARS-CoV-2, interacts with electrophilic groups present in ethacrynic acid (EA) and compounds 4, 6, and 8 to form sulfur-carbon bonds. Nuclear magnetic resonance (NMR) spectroscopy was used to monitor the reaction rate between cysteine and Michael acceptors. We found that the inhibitory activity of these compounds towards Mpro is correlated to their chemical reactivity toward cysteine. This approach may serve as a valuable tool in drug development for detecting potential covalent inhibitors of Mpro and other cysteine proteases.
Collapse
Affiliation(s)
| | | | | | | | - Hamza Ghammaz
- Centre National de la Recherche Scientifique et Technique (CNRST) Angle avenues des FAR et Allal El Fassi, Hay Ryad 10102 Rabat Morocco
| | - Nadia Touil
- Cell Culture Unit, Center of Virology, Infectious, and Tropical Diseases Mohammed V Military Hospital Rabat Morocco
| | | | - Elmostafa El Fahime
- Centre National de la Recherche Scientifique et Technique (CNRST) Angle avenues des FAR et Allal El Fassi, Hay Ryad 10102 Rabat Morocco
| | | |
Collapse
|
19
|
Sun Y, Li Q, Huang Y, Yang Z, Li G, Sun X, Gu X, Qiao Y, Wu Q, Xie T, Sui X. Natural products for enhancing the sensitivity or decreasing the adverse effects of anticancer drugs through regulating the redox balance. Chin Med 2024; 19:110. [PMID: 39164783 PMCID: PMC11334420 DOI: 10.1186/s13020-024-00982-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024] Open
Abstract
Redox imbalance is reported to play a pivotal role in tumorigenesis, cancer development, and drug resistance. Severe oxidative damage is a general consequence of cancer cell responses to treatment and may cause cancer cell death or severe adverse effects. To maintain their longevity, cancer cells can rescue redox balance and enter a state of resistance to anticancer drugs. Therefore, targeting redox signalling pathways has emerged as an attractive and prospective strategy for enhancing the efficacy of anticancer drugs and decreasing their adverse effects. Over the past few decades, natural products (NPs) have become an invaluable source for developing new anticancer drugs due to their high efficacy and low toxicity. Increasing evidence has demonstrated that many NPs exhibit remarkable antitumour effects, whether used alone or as adjuvants, and are emerging as effective approaches to enhance sensitivity and decrease the adverse effects of conventional cancer therapies by regulating redox balance. Among them are several novel anticancer drugs based on NPs that have entered clinical trials. In this review, we summarize the synergistic anticancer effects and related redox mechanisms of the combination of NPs with conventional anticancer drugs. We believe that NPs targeting redox regulation will represent promising novel candidates and provide prospects for cancer treatment in the future.
Collapse
Affiliation(s)
- Yitian Sun
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Qinyi Li
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yufei Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Zijing Yang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Guohua Li
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiaoyu Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiaoqing Gu
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yunhao Qiao
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
| | - Tian Xie
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Xinbing Sui
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
20
|
Osorio Reineke N, Elsen FAV, Grab HA, Mostert D, Sieber SA, Bach T. Synthesis and biological evaluation of vioprolide B and its dehydrobutyrine-glycine analogue. Chem Commun (Camb) 2024; 60:8272-8275. [PMID: 39015034 DOI: 10.1039/d4cc02946a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Herein, we describe the total synthesis of the depsipeptide vioprolide B and of an analogue, in which the (E)-dehydrobutyrine amino acid was replaced by glycine. The compounds were studied in biological assays which revealed cytotoxicity solely for vioprolide B presumably by covalent binding to cysteine residues of elongation factor eEF1A1 and of chromatin assembly factor CHAF1A.
Collapse
Affiliation(s)
- Noé Osorio Reineke
- Technische Universität München, School of Natural Sciences, Department of Chemistry and Catalysis Research Center, Lichtenbergstraße 4, 85747 Garching, Germany.
| | - Franziska A V Elsen
- Technische Universität München, School of Natural Sciences, Department of Bioscience and Center for Functional Protein Assemblies, Ernst-Otto-Fischer-Straße 8, 85747 Garching, Germany
| | - Hanusch A Grab
- Technische Universität München, School of Natural Sciences, Department of Chemistry and Catalysis Research Center, Lichtenbergstraße 4, 85747 Garching, Germany.
| | - Dietrich Mostert
- Technische Universität München, School of Natural Sciences, Department of Bioscience and Center for Functional Protein Assemblies, Ernst-Otto-Fischer-Straße 8, 85747 Garching, Germany
| | - Stephan A Sieber
- Technische Universität München, School of Natural Sciences, Department of Bioscience and Center for Functional Protein Assemblies, Ernst-Otto-Fischer-Straße 8, 85747 Garching, Germany
| | - Thorsten Bach
- Technische Universität München, School of Natural Sciences, Department of Chemistry and Catalysis Research Center, Lichtenbergstraße 4, 85747 Garching, Germany.
| |
Collapse
|
21
|
Tuzi A, Carbone M, Ciavatta ML, Evidente A. Structure Revision of the Fungal Phytotoxin Cavoxin and of Its Corresponding Chroman-4-one Cavoxone by X-ray Crystallography. JOURNAL OF NATURAL PRODUCTS 2024; 87:1888-1892. [PMID: 38967603 DOI: 10.1021/acs.jnatprod.4c00531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Cavoxin (1) was isolated as the main phytotoxin produced by Phoma cava Schulzer, a toxigenic fungus isolated from Castanea spp. Its structure was determined by 1D NMR and MS in 1985 along with that of the corresponding chroman-4-one cavoxone (2), an artifact formed by acid treatment of 1. Since that time cavoxin was shown to be phytotoxic, antifungal, antifeedant, herbicidal, and antirust with potential application in agriculture and medicine. During a study aimed at improving cavoxin's production by P. cava, single crystals for X-ray diffractometric analysis were obtained. The X-ray crystallography characterization confirmed only in part the structure proposed for cavoxin (1), revealing a different substitution pattern on the aromatic ring, as depicted in the revised structure 3.
Collapse
Affiliation(s)
- Angela Tuzi
- Dipartimento di Scienze Chimiche, Università di Napoli "Federico II", Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Napoli, Italy
| | - Marianna Carbone
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Maria Letizia Ciavatta
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Antonio Evidente
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| |
Collapse
|
22
|
Zeng X, Tian Y, Kong H, Li Z, Gu Z, Li C, Hong Y, Cheng L, Ban X. Catalytic Mode and Product Specificity of an α-Agarase Reveal Its Direct Catalysis for the Production of Agarooligosaccharides. Foods 2024; 13:2351. [PMID: 39123543 PMCID: PMC11311870 DOI: 10.3390/foods13152351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Many α-agarases have been characterized and are utilized for producing agarooligosaccharides through the degradation of agar and agarose, which are considered valuable for applications in the food and medicine industries. However, the catalytic mechanism and product transformation process of α-agarase remain unclear, limiting further enzyme engineering for industrial applications. In this study, an α-agarase from Catenovulum maritimus STB14 (Cm-AGA) was employed to degrade agarose oligosaccharides (AGOs) with varying degrees of polymerization (DPs) to investigate the catalytic mechanism of α-agarases. The results demonstrated that Cm-AGA could degrade agarose into agarotetraose and agarohexaose. The reducing ends of agarotetraose and agarohexaose spontaneously release unstable 3,6-anhydro-α-l-galactose molecules, which were further degraded into agarotriose and agaropentose. Cm-AGA cannot act on α-1,3-glucoside bonds in agarotriose, agarotetraose, neoagarobiose, and neoagarotetraose but can act on AGOs with a DP greater than four. The product analysis was further verified by β-galactosidase hydrolysis, which specifically cleaves the non-reducing glycosidic bond of agarooligosaccharides. Multiple sequence alignment results showed that two conserved residues, Asp994 and Glu1129, were proposed as catalytic residues and were further identified by site-directed mutagenesis. Molecular docking of Cm-AGA with agaroheptose revealed the potential substrate binding mode of the α-agarase. These findings enhance the understanding of Cm-AGA's catalytic mode and could guide enzyme engineering for modulating the production of agarooligosaccharides.
Collapse
Affiliation(s)
- Xiaofeng Zeng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
| | - Yixiong Tian
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
| | - Haocun Kong
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Zhaofeng Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Caiming Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
| | - Li Cheng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
| | - Xiaofeng Ban
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (X.Z.); (Y.T.); (H.K.); (Z.L.); (Z.G.); (C.L.); (Y.H.); (L.C.)
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
23
|
Jiang Z, Chen JA, Mohamed OG, Huynh J, Chen A, Tripathi A, La Clair JJ, Burkart MD. Cryptic Cerulenin Rearrangement in Ketosynthase Covalent Inhibition. J Am Chem Soc 2024; 146:20370-20378. [PMID: 38981108 DOI: 10.1021/jacs.4c05938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
The antibiotic cerulenin is a fungal natural product identified as a covalent inhibitor of ketosynthases within fatty acid and polyketide biosynthesis. Due to its selective and potent inhibitory activity, cerulenin has found significant utility in multidisciplinary biochemical, biomedical, and clinical studies. Although its covalent inhibition profile has been confirmed, cerulenin's mechanism has not been fully determined at a molecular level, frustrating the drug development of related analogues. Herein, we describe the use of stable isotopic tracking with NMR and MS methods to unravel the covalent mechanism of cerulenin against type II fatty acid ketosynthases. We detail the discovery of a unique C2-C3 retro-aldol bond cleavage and a structural rearrangement upon covalent inhibition of cerulenin at the active cysteine residue in E. coli type II fatty acid ketosynthases FabB and FabF.
Collapse
Affiliation(s)
- Ziran Jiang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Jeffrey A Chen
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Osama G Mohamed
- Natural product Discovery Core - Life Sciences Institute, University of Michigan, Mary Sue Coleman Hall, 210 Washtenaw Avenue, Ann Arbor, Michigan 48109, United States
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr el Aini Street, Cairo 11562, Egypt
| | - Jennifer Huynh
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Aochiu Chen
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Ashootosh Tripathi
- Natural product Discovery Core - Life Sciences Institute, University of Michigan, Mary Sue Coleman Hall, 210 Washtenaw Avenue, Ann Arbor, Michigan 48109, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 1600 Huron Parkway, NCRC B520, Ann Arbor, Michigan 48109, United States
| | - James J La Clair
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Michael D Burkart
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| |
Collapse
|
24
|
Santos-Júnior PFDS, Batista VDM, Nascimento IJDS, Nunes IC, Silva LR, Costa CACB, Freitas JDD, Quintans-Júnior LJ, Araújo-Júnior JXD, Freitas MEGD, Zhan P, Green KD, Garneau-Tsodikova S, Mendonça-Júnior FJB, Rodrigues-Junior VS, Silva-Júnior EFD. A consensus reverse docking approach for identification of a competitive inhibitor of acetyltransferase enhanced intracellular survival protein from Mycobacterium tuberculosis. Bioorg Med Chem 2024; 108:117774. [PMID: 38833750 DOI: 10.1016/j.bmc.2024.117774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/08/2024] [Accepted: 05/24/2024] [Indexed: 06/06/2024]
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb), which remains a significant global health challenge. The emergence of multidrug-resistant (MDR) Mtb strains imposes the development of new therapeutic strategies. This study focuses on the identification and evaluation of potential inhibitors against Mtb H37Ra through a comprehensive screening of an in-house chemolibrary. Subsequently, a promising pyrimidine derivative (LQM495) was identified as promising and then further investigated by experimental and in silico approaches. In this context, computational techniques were used to elucidate the potential molecular target underlying the inhibitory action of LQM495. Then, a consensus reverse docking (CRD) protocol was used to investigate the interactions between this compound and several Mtb targets. Out of 98 Mtb targets investigated, the enhanced intracellular survival (Eis) protein emerged as a target for LQM495. To gain insights into the stability of the LQM495-Eis complex, molecular dynamics (MD) simulations were conducted over a 400 ns trajectory. Further insights into its binding modes within the Eis binding site were obtained through a Quantum mechanics (QM) approach, using density functional theory (DFT), with B3LYP/D3 basis set. These calculations shed light on the electronic properties and reactivity of LQM495. Subsequently, inhibition assays and kinetic studies of the Eis activity were used to investigate the activity of LQM495. Then, an IC50 value of 11.0 ± 1.4 µM was found for LQM495 upon Eis protein. Additionally, its Vmax, Km, and Ki parameters indicated that it is a competitive inhibitor. Lastly, this study presents LQM495 as a promising inhibitor of Mtb Eis protein, which could be further explored for developing novel anti-TB drugs in the future.
Collapse
Affiliation(s)
| | - Vitoria de Melo Batista
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | - Igor José Dos Santos Nascimento
- Post-Graduation Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina Grande, Brazil
| | - Isabelle Cavalcante Nunes
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | - Leandro Rocha Silva
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | | | - Johnnatan Duarte de Freitas
- Department of Chemistry, Federal Institute of Alagoas, Maceió campus, Mizael Domingues Street, 57020-600 Maceió, Alagoas, Brazil
| | - Lucindo José Quintans-Júnior
- Pharmaceutical Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, Sergipe 49100-001, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | | | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Keith D Green
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, 40536-0596, United States
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, 40536-0596, United States
| | | | - Valnês S Rodrigues-Junior
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil; Post-Graduation Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, João Pessoa, Brazil
| | - Edeildo Ferreira da Silva-Júnior
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil.
| |
Collapse
|
25
|
Andrés CMC, Pérez de la Lastra JM, Bustamante Munguira E, Andrés Juan C, Pérez-Lebeña E. Michael Acceptors as Anti-Cancer Compounds: Coincidence or Causality? Int J Mol Sci 2024; 25:6099. [PMID: 38892287 PMCID: PMC11172677 DOI: 10.3390/ijms25116099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Michael acceptors represent a class of compounds with potential anti-cancer properties. They act by binding to nucleophilic sites in biological molecules, thereby disrupting cancer cell function and inducing cell death. This mode of action, as well as their ability to be modified and targeted, makes them a promising avenue for advancing cancer therapy. We are investigating the molecular mechanisms underlying Michael acceptors and their interactions with cancer cells, in particular their ability to interfere with cellular processes and induce apoptosis. The anti-cancer properties of Michael acceptors are not accidental but are due to their chemical structure and reactivity. The electrophilic nature of these compounds allows them to selectively target nucleophilic residues on disease-associated proteins, resulting in significant therapeutic benefits and minimal toxicity in various diseases. This opens up new perspectives for the development of more effective and precise cancer drugs. Nevertheless, further studies are essential to fully understand the impact of our discoveries and translate them into clinical practice.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | | | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
| | | |
Collapse
|
26
|
Shkil DO, Muhamedzhanova AA, Petrov PI, Skorb EV, Aliev TA, Steshin IS, Tumanov AV, Kislinskiy AS, Fedorov MV. Expanding Predictive Capacities in Toxicology: Insights from Hackathon-Enhanced Data and Model Aggregation. Molecules 2024; 29:1826. [PMID: 38675645 PMCID: PMC11055041 DOI: 10.3390/molecules29081826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
In the realm of predictive toxicology for small molecules, the applicability domain of QSAR models is often limited by the coverage of the chemical space in the training set. Consequently, classical models fail to provide reliable predictions for wide classes of molecules. However, the emergence of innovative data collection methods such as intensive hackathons have promise to quickly expand the available chemical space for model construction. Combined with algorithmic refinement methods, these tools can address the challenges of toxicity prediction, enhancing both the robustness and applicability of the corresponding models. This study aimed to investigate the roles of gradient boosting and strategic data aggregation in enhancing the predictivity ability of models for the toxicity of small organic molecules. We focused on evaluating the impact of incorporating fragment features and expanding the chemical space, facilitated by a comprehensive dataset procured in an open hackathon. We used gradient boosting techniques, accounting for critical features such as the structural fragments or functional groups often associated with manifestations of toxicity.
Collapse
Affiliation(s)
- Dmitrii O. Shkil
- Syntelly LLC, Moscow 121205, Russia; (A.A.M.); (I.S.S.); (A.V.T.); (A.S.K.)
- Moscow Institute of Physics and Technology, Moscow 141700, Russia
| | | | | | - Ekaterina V. Skorb
- Infochemistry Scientific Center, ITMO University, Saint-Petersburg 191002, Russia; (E.V.S.); (T.A.A.)
| | - Timur A. Aliev
- Infochemistry Scientific Center, ITMO University, Saint-Petersburg 191002, Russia; (E.V.S.); (T.A.A.)
| | - Ilya S. Steshin
- Syntelly LLC, Moscow 121205, Russia; (A.A.M.); (I.S.S.); (A.V.T.); (A.S.K.)
| | | | | | - Maxim V. Fedorov
- Kharkevich Institute for Information Transmission Problems of Russian Academy of Sciences, Moscow 127994, Russia
| |
Collapse
|
27
|
Ha KS. Transglutaminase 2 in diabetes mellitus: Unraveling its multifaceted role and therapeutic implications for vascular complications. Theranostics 2024; 14:2329-2344. [PMID: 38646650 PMCID: PMC11024853 DOI: 10.7150/thno.95742] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/17/2024] [Indexed: 04/23/2024] Open
Abstract
Diabetes, a severe metabolic disease characterized by chronic hypoglycemia, poses debilitating and life-threatening risks of microvascular and macrovascular complications, including blindness, kidney failure, heart attacks, and limb amputation. Addressing these complications is paramount, urging the development of interventions targeting diabetes-associated vascular dysfunctions. To effectively combat diabetes, a comprehensive understanding of the pathological mechanisms underlying complications and identification of precise therapeutic targets are imperative. Transglutaminase 2 (TGase2) is a multifunctional enzyme implicated in the pathogenesis of diverse diseases such as neurodegenerative disorders, fibrosis, and inflammatory conditions. TGase2 has recently emerged as a key player in both the pathogenesis and therapeutic intervention of diabetic complications. This review highlights TGase2 as a therapeutic target for diabetic complications and explores TGase2 inhibition as a promising therapeutic approach in their treatment.
Collapse
Affiliation(s)
- Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
| |
Collapse
|
28
|
Saquib Q, Bakheit AH, Ahmed S, Ansari SM, Al-Salem AM, Al-Khedhairy AA. Identification of Phytochemicals from Arabian Peninsula Medicinal Plants as Strong Binders to SARS-CoV-2 Proteases (3CL Pro and PL Pro) by Molecular Docking and Dynamic Simulation Studies. Molecules 2024; 29:998. [PMID: 38474509 DOI: 10.3390/molecules29050998] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/04/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
We provide promising computational (in silico) data on phytochemicals (compounds 1-10) from Arabian Peninsula medicinal plants as strong binders, targeting 3-chymotrypsin-like protease (3CLPro) and papain-like proteases (PLPro) of SARS-CoV-2. Compounds 1-10 followed the Lipinski rules of five (RO5) and ADMET analysis, exhibiting drug-like characters. Non-covalent (reversible) docking of compounds 1-10 demonstrated their binding with the catalytic dyad (CYS145 and HIS41) of 3CLPro and catalytic triad (CYS111, HIS272, and ASP286) of PLPro. Moreover, the implementation of the covalent (irreversible) docking protocol revealed that only compounds 7, 8, and 9 possess covalent warheads, which allowed the formation of the covalent bond with the catalytic dyad (CYS145) in 3CLPro and the catalytic triad (CYS111) in PLPro. Root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF), and radius of gyration (Rg) analysis from molecular dynamic (MD) simulations revealed that complexation between ligands (compounds 7, 8, and 9) and 3CLPro and PLPro was stable, and there was less deviation of ligands. Overall, the in silico data on the inherent properties of the above phytochemicals unravel the fact that they can act as reversible inhibitors for 3CLPro and PLPro. Moreover, compounds 7, 8, and 9 also showed their novel properties to inhibit dual targets by irreversible inhibition, indicating their effectiveness for possibly developing future drugs against SARS-CoV-2. Nonetheless, to confirm the theoretical findings here, the effectiveness of the above compounds as inhibitors of 3CLPro and PLPro warrants future investigations using suitable in vitro and in vivo tests.
Collapse
Affiliation(s)
- Quaiser Saquib
- Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ahmed H Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sarfaraz Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sabiha M Ansari
- Botany & Microbiology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdullah M Al-Salem
- Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdulaziz A Al-Khedhairy
- Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
29
|
Mu K, Kitts DD. Intestinal polyphenol antioxidant activity involves redox signaling mechanisms facilitated by aquaporin activity. Redox Biol 2023; 68:102948. [PMID: 37922763 PMCID: PMC10643476 DOI: 10.1016/j.redox.2023.102948] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Ascertaining whether dietary polyphenols evoke an antioxidant or prooxidant activity, which translates to a functional role required to maintain intestinal cell homeostasis continues to be an active and controversial area of research for food chemists and biochemists alike. We have proposed that the paradoxical function of polyphenols to autoxidize to generate H2O2 is a required first step in the capacity of some plant phenolics to function as intracellular antioxidants. This is based on the fact that cell redox homeostasis is achieved by a balance between H2O2 formation and subsequent outcomes of antioxidant systems function. Maintaining optimal extracellular and intracellular H2O2 concentrations is required for cell survival, since low levels are important to upregulate endogenous antioxidant capacity; whereas, concentrations that go beyond homeostatic control typically result in an inflammatory response, growth arrest, or eventual cell death. Aquaporins (AQPs) are a family of water channel membrane proteins that facilitate cellular transportation of water and other small molecule-derived solutes, such as H2O2, in all organisms. In the intestine, AQPs act as gatekeepers to regulate intracellular uptake of H2O2, generated from extracellular polyphenol autoxidation, thus enabling an intracellular cell signaling responses to mitigate onset of oxidative stress and intestinal inflammation. In this review, we highlight the potential role of AQPs to control important underlying mechanisms that define downstream regulation of intestinal redox homeostasis, specifically. It has been established that polyphenols that undergo oxidation to the quinone form, resulting in subsequent adduction to a thiol group on Keap1-Nrf2 complex, trigger Nrf2 activation and a cascade of indirect intracellular antioxidant effects. Here, we propose a similar mechanism that involves H2O2 generated from specific dietary polyphenols with a predisposition to undergo autoxidation. The ultimate bioactivity is regulated and expressed by AQP membrane function and thus, by extension, represents expression of an intracellular antioxidant chemoprotection mechanism.
Collapse
Affiliation(s)
- Kaiwen Mu
- Food Science, Food Nutrition and Health Program. Faculty of Land and Food System, The University of British Columbia, 2205 East Mall, Vancouver, B.C, V6T 1Z4, Canada
| | - David D Kitts
- Food Science, Food Nutrition and Health Program. Faculty of Land and Food System, The University of British Columbia, 2205 East Mall, Vancouver, B.C, V6T 1Z4, Canada.
| |
Collapse
|
30
|
Al-Awadhi FH, Simon EF, Liu N, Ratnayake R, Paul VJ, Luesch H. Discovery and Anti-Inflammatory Activity of a Cyanobacterial Fatty Acid Targeting the Keap1/Nrf2 Pathway. Mar Drugs 2023; 21:553. [PMID: 37999377 PMCID: PMC10672429 DOI: 10.3390/md21110553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
The monounsaturated fatty acid 7(E)-9-keto-hexadec-7-enoic acid (1) and three structurally related analogues with different oxidation states and degrees of unsaturation (2-4) were discovered from a marine benthic cyanobacterial mat collected from Delta Shoal, Florida Keys. Their structures were elucidated using NMR spectroscopy and mass spectrometry. The structure of 1 contained an α,β-unsaturated carbonyl system, a key motif required for the activation of the Keap1/Nrf2-ARE pathway that is involved in the activation of antioxidant and phase II detoxification enzymes. Compounds 1-4 were screened in ARE-luciferase reporter gene assay using stably transfected HEK293 cells, and only 1 significantly induced Nrf2 activity at 32 and 10 µM, whereas 2-4 were inactive. As there is crosstalk between inflammation and oxidative stress, subsequent biological studies were focused on 1 to investigate its anti-inflammatory potential. Compound 1 induced Nqo1, a well-known target gene of Nrf2, and suppressed iNos transcript levels, which translated into reduced levels of nitric oxide in LPS-activated mouse macrophage RAW264.7 cells, a more relevant model for inflammation. RNA sequencing was performed to capture the effects of 1 on a global level and identified additional canonical pathways and upstream regulators involved in inflammation and immune response, particularly those related to multiple sclerosis. A targeted survey of marine cyanobacterial samples from other geographic locations, including Guam, suggested the widespread occurrence of 1. Furthermore, the previous isolation of 1 from marine diatoms and green algae implied a potentially important ecological role across marine algal eukaryotes and prokaryotes. The previous isolation from sea lettuce raises the possibility of dietary intervention to attenuate inflammation and related disease progression.
Collapse
Affiliation(s)
- Fatma H. Al-Awadhi
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, USA or (F.H.A.-A.); (E.F.S.); (N.L.); (R.R.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| | - Emily F. Simon
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, USA or (F.H.A.-A.); (E.F.S.); (N.L.); (R.R.)
| | - Na Liu
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, USA or (F.H.A.-A.); (E.F.S.); (N.L.); (R.R.)
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Ranjala Ratnayake
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, USA or (F.H.A.-A.); (E.F.S.); (N.L.); (R.R.)
| | | | - Hendrik Luesch
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, USA or (F.H.A.-A.); (E.F.S.); (N.L.); (R.R.)
| |
Collapse
|
31
|
Phetruen T, van Dam B, Chanarat S. Andrographolide Induces ROS-Mediated Cytotoxicity, Lipid Peroxidation, and Compromised Cell Integrity in Saccharomyces cerevisiae. Antioxidants (Basel) 2023; 12:1765. [PMID: 37760068 PMCID: PMC10525756 DOI: 10.3390/antiox12091765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Andrographolide, a bioactive compound found in Andrographis paniculata, has gained significant attention for its potential therapeutic properties. Despite its promising benefits, the understanding of its side effects and underlying mechanisms remains limited. Here, we investigated the impact of andrographolide in Saccharomyces cerevisiae and observed that andrographolide induced cytotoxicity, particularly when oxidative phosphorylation was active. Furthermore, andrographolide affected various cellular processes, including vacuole fragmentation, endoplasmic reticulum stress, lipid droplet accumulation, reactive oxygen species levels, and compromised cell integrity. Moreover, we unexpectedly observed that andrographolide induced the precipitation of biomolecules secreted from yeast cells, adding an additional source of stress. Overall, this study provides insights into the cellular effects and potential mechanisms of andrographolide in yeast, shedding light on its side effects and underlying cytotoxicity pathways.
Collapse
Affiliation(s)
| | | | - Sittinan Chanarat
- Laboratory of Molecular Cell Biology, Department of Biochemistry, Center for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
32
|
Martinez-Banaclocha MA. Targeting the Cysteine Redox Proteome in Parkinson's Disease: The Role of Glutathione Precursors and Beyond. Antioxidants (Basel) 2023; 12:1373. [PMID: 37507913 PMCID: PMC10376658 DOI: 10.3390/antiox12071373] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Encouraging recent data on the molecular pathways underlying aging have identified variants and expansions of genes associated with DNA replication and repair, telomere and stem cell maintenance, regulation of the redox microenvironment, and intercellular communication. In addition, cell rejuvenation requires silencing some transcription factors and the activation of pluripotency, indicating that hidden molecular networks must integrate and synchronize all these cellular mechanisms. Therefore, in addition to gene sequence expansions and variations associated with senescence, the optimization of transcriptional regulation and protein crosstalk is essential. The protein cysteinome is crucial in cellular regulation and plays unexpected roles in the aging of complex organisms, which show cumulative somatic mutations, telomere attrition, epigenetic modifications, and oxidative dysregulation, culminating in cellular senescence. The cysteine thiol groups are highly redox-active, allowing high functional versatility as structural disulfides, redox-active disulfides, active-site nucleophiles, proton donors, and metal ligands to participate in multiple regulatory sites in proteins. Also, antioxidant systems control diverse cellular functions, including the transcription machinery, which partially depends on the catalytically active cysteines that can reduce disulfide bonds in numerous target proteins, driving their biological integration. Since we have previously proposed a fundamental role of cysteine-mediated redox deregulation in neurodegeneration, we suggest that cellular rejuvenation of the cysteine redox proteome using GSH precursors, like N-acetyl-cysteine, is an underestimated multitarget therapeutic approach that would be particularly beneficial in Parkinson's disease.
Collapse
|
33
|
Ahmad Mir S, Shahid Maqbool M, Bharitkar YP, Malik FA, Khalid Yousuf S. Suzuki coupling-based synthesis and in vitro cytotoxic evaluation of C-2 aryl derivatives of withaferin A. Steroids 2023; 195:109246. [PMID: 37141979 DOI: 10.1016/j.steroids.2023.109246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/06/2023]
Affiliation(s)
- Shabir Ahmad Mir
- Natural Product and Medicinal Chemistry Division, Indian Institute of Integrative Medicine (CSIR-IIIM), Srinagar190005, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mir Shahid Maqbool
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR-IIIM), Srinagar 190005, India
| | - Yogesh P Bharitkar
- Natural Product and Medicinal Chemistry Division, Indian Institute of Integrative Medicine (CSIR-IIIM), Srinagar190005, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Fayaz A Malik
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR-IIIM), Srinagar 190005, India
| | - Syed Khalid Yousuf
- Natural Product and Medicinal Chemistry Division, Indian Institute of Integrative Medicine (CSIR-IIIM), Srinagar190005, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Quality Management & Instrumentation Division, Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu180001, India.
| |
Collapse
|
34
|
Yang G, Mi X, Wang Y, Li S, Yu L, Huang X, Tan S, Yu H. Fusion of Michael-acceptors enhances the anti-inflammatory activity of ginsenosides as potential modulators of the NLRP3 signaling pathway. Bioorg Chem 2023; 134:106467. [PMID: 36933337 DOI: 10.1016/j.bioorg.2023.106467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023]
Abstract
Ginsenosides are a promising group of secondary metabolites for developing anti-inflammatory agents. In this study, Michael acceptor was fused into the aglycone A-ring of protopanoxadiol (PPD)-type ginsenosides (MAAG), the main pharmacophore of ginseng, and its liver metabolites to produce novel derivatives and assess their anti-inflammatory activity in vitro. The structure-activity relationship of MAAG derivatives was assessed based on their NO-inhibition activities. Of these, a 4-nitrobenzylidene derivative of PPD (2a) was the most effective and dose-dependently inhibited the release of proinflammatory cytokines. Further studies indicated that 2a-induced downregulation on lipopolysaccharide (LPS)-induced iNOS protein expression and cytokine release may be related to its inhibitory effect on MAPK and NF-κB signaling pathways. Importantly, 2a almost completely inhibited LPS-induced production of mitochondrial reactive oxygen species (mtROS) and LPS-induced NLRP3 upregulation. This inhibition was higher than that by hydrocortisone sodium succinate, a glucocorticoid drug. Overall, the fusion of Michael acceptors into the aglycone of ginsenosides greatly enhanced the anti-inflammatory activities of the derivatives, and 2a alleviated inflammation considerably. These findings could be attributed to the inhibition of LPS-induced mtROS to block abnormal activation of the NLRP3 pathway.
Collapse
Affiliation(s)
- Gangqiang Yang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China.
| | - Xiaoliang Mi
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Yunxiao Wang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Shuang Li
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Liping Yu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Xinru Huang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Shuai Tan
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China
| | - Hui Yu
- College of Food Engineering, Ludong University, Yantai 264025, China.
| |
Collapse
|
35
|
McCord JM, Gao B, Hybertson BM. The Complex Genetic and Epigenetic Regulation of the Nrf2 Pathways: A Review. Antioxidants (Basel) 2023; 12:antiox12020366. [PMID: 36829925 PMCID: PMC9952775 DOI: 10.3390/antiox12020366] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Nrf2 is a major transcription factor that significantly regulates-directly or indirectly-more than 2000 genes. While many of these genes are involved in maintaining redox balance, others are involved in maintaining balance among metabolic pathways that are seemingly unrelated to oxidative stress. In the past 25 years, the number of factors involved in the activation, nuclear translocation, and deactivation of Nrf2 has continued to expand. The purpose of this review is to provide an overview of the remarkable complexity of the tortuous sequence of stop-and-go signals that not only regulate expression or repression, but may also modify transcriptional intensity as well as the specificity of promoter recognition, allowing fluidity of its gene expression profile depending on the various structural modifications the transcription factor encounters on its journey to the DNA. At present, more than 45 control points have been identified, many of which represent sites of action of the so-called Nrf2 activators. The complexity of the pathway and the synergistic interplay among combinations of control points help to explain the potential advantages seen with phytochemical compositions that simultaneously target multiple control points, compared to the traditional pharmaceutical paradigm of "one-drug, one-target".
Collapse
Affiliation(s)
- Joe M. McCord
- Pathways Bioscience, Aurora, CO 80045, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Correspondence:
| | - Bifeng Gao
- Pathways Bioscience, Aurora, CO 80045, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brooks M. Hybertson
- Pathways Bioscience, Aurora, CO 80045, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
36
|
Trifluoromethylcinnamanilide Michael Acceptors for Treatment of Resistant Bacterial Infections. Int J Mol Sci 2022; 23:ijms232315090. [PMID: 36499415 PMCID: PMC9737391 DOI: 10.3390/ijms232315090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
A series of thirty-two anilides of 3-(trifluoromethyl)cinnamic acid (series 1) and 4-(trifluoromethyl)cinnamic acid (series 2) was prepared by microwave-assisted synthesis. All the compounds were tested against reference strains Staphylococcus aureus ATCC 29213 and Enterococcus faecalis ATCC 29212 and resistant clinical isolates of methicillin-resistant S. aureus (MRSA) and vancomycin-resistant E. faecalis (VRE). All the compounds were evaluated in vitro against Mycobacterium smegmatis ATCC 700084 and M. marinum CAMP 5644. (2E)-3-[3-(Trifluoromethyl)phenyl]-N-[4-(trifluoromethyl)phenyl]prop-2-enamide (1j), (2E)-N-(3,5-dichlorophenyl)-3-[3-(trifluoromethyl)phenyl]prop-2-enamide (1o) and (2E)-N-[3-(trifluoromethyl)phenyl]-3-[4-(trifluoromethyl)-phenyl]prop-2-enamide (2i), (2E)-N-[3,5-bis(trifluoromethyl)phenyl]-3-[4-(trifluoromethyl)phenyl]-prop-2-enamide (2p) showed antistaphylococcal (MICs/MBCs 0.15-5.57 µM) as well as anti-enterococcal (MICs/MBCs 2.34-44.5 µM) activity. The growth of M. marinum was strongly inhibited by compounds 1j and 2p in a MIC range from 0.29 to 2.34 µM, while all the agents of series 1 showed activity against M. smegnatis (MICs ranged from 9.36 to 51.7 µM). The performed docking study demonstrated the ability of the compounds to bind to the active site of the mycobacterial enzyme InhA. The compounds had a significant effect on the inhibition of bacterial respiration, as demonstrated by the MTT assay. The compounds showed not only bacteriostatic activity but also bactericidal activity. Preliminary in vitro cytotoxicity screening was assessed using the human monocytic leukemia cell line THP-1 and, except for compound 2p, all effective agents did show insignificant cytotoxic effect. Compound 2p is an interesting anti-invasive agent with dual (cytotoxic and antibacterial) activity, while compounds 1j and 1o are the most interesting purely antibacterial compounds within the prepared molecules.
Collapse
|