Bharat Sahu, Sanheeta Chakrabarty, Vaishali Saini, Meenakshi Kandpal, Bharat Goel, Sanju Kumari, Ijaz Ahmed, Hem Chandra Jha, Shreyans K. Jain
{"title":"Exploring the cytotoxic potential of biflavones of Araucaria cunninghamii: Precise identification combined by LC-HRMS-metabolomics and database mining, targeted isolation, network pharmacology, in vitro cytotoxicity, and docking studies","authors":"Bharat Sahu, Sanheeta Chakrabarty, Vaishali Saini, Meenakshi Kandpal, Bharat Goel, Sanju Kumari, Ijaz Ahmed, Hem Chandra Jha, Shreyans K. Jain","doi":"10.1111/cbdd.14564","DOIUrl":"10.1111/cbdd.14564","url":null,"abstract":"<p>The leaves of <i>Araucaria cunninghamii</i> are known to be nonedible and toxic. Previous studies have identified biflavones in various <i>Araucaria</i> species. This study aimed to investigate the in vitro cytotoxicity of the isolated compounds from <i>Araucaria cunninghamii</i> after metabolomics and network pharmacological analysis. Methanol extract of <i>Araucaria cunninghamii</i> leaves was subjected to bioassay-guided fractionation. The active fraction was analyzed using LC-HRMS, through strategic database mining, by comparing the data to the Dictionary of Natural Products to identify 12 biflavones, along with abietic acid, beta-sitosterol, and phthalate. Eight compounds were screened for network pharmacology study, where in silico ADME analysis, prediction of gene targets, compound-gene-pathway network and hierarchical network analysis, protein–protein interaction, KEGG pathway, and Gene Ontology analyses were done, that showed PI3KR1, EGFR, GSK3B, and ABCB1 as the common targets for all the compounds that may act in the gastric cancer pathway. Simultaneously, four biflavones were isolated via chromatography and identified through NMR as dimeric apigenin with varying methoxy substitutions. Cytotoxicity study against the AGS cell line for gastric cancer showed that AC1 biflavone (IC<sub>50</sub> 90.58 μM) exhibits the highest cytotoxicity and monomeric apigenin (IC<sub>50</sub> 174.5 μM) the lowest. Besides, the biflavones were docked to the previously identified targets to analyze their binding affinities, and all the ligands were found to bind with energy ≤−7 Kcal/mol.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141285664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
JingXu Chen, JiaLi Rao, Hao Lu, Min Lu, ChengCheng Wang, Yan Cao
{"title":"Network pharmacology and experimental verification to explore the effect of Hedyotis diffusa on Alzheimer's disease","authors":"JingXu Chen, JiaLi Rao, Hao Lu, Min Lu, ChengCheng Wang, Yan Cao","doi":"10.1111/cbdd.14558","DOIUrl":"10.1111/cbdd.14558","url":null,"abstract":"<p>This study aimed to explore the active components and the effect of <i>Hedyotis diffusa</i> (HD) against Alzheimer's disease (AD) via network pharmacology, molecular docking, and experimental evaluations. We conducted a comprehensive screening process using the TCMSP, Swiss Target Prediction, and PharmMapper databases to identify the active components and their related targets in HD. In addition, we collected potential therapeutic targets of AD from the Gene Cards, Drugbank, and OMIM databases. Afterward, we utilized Cytoscape to establish both protein–protein interaction (PPI) networks and compound-target (C-T) networks. To gain further insights into the functional aspect, we performed GO and KEGG pathway analyses using the David database. Next, we employed Autodock vina to estimate the binding force between the components and the hub genes. To validate our network pharmacology findings, we conducted relevant experiments on <i>Caenorhabditis elegans</i>, further confirming the reliability of our results. Then a total of six active compounds and 149 therapeutic targets were detected. Through the analysis of the association between active compounds, therapeutic targets, and signaling pathways, it was observed that the therapeutic effect of HD primarily encompassed the inhibition of Aβ, suppression of AChE activity, and mitigating oxidative stress. Additionally, our investigation revealed that the key active compounds in HD primarily consisted of iridoids, which exhibited resistance against AD by acting on the Alzheimer's disease pathway and the AGE-RAGE signaling pathway in diabetic complications.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ji Jiang, Yan Yang, Fuhuan Wang, Wei Mao, Zhongjun Wang, Zegang Liu
{"title":"Quercetin inhibits breast cancer cell proliferation and survival by targeting Akt/mTOR/PTEN signaling pathway","authors":"Ji Jiang, Yan Yang, Fuhuan Wang, Wei Mao, Zhongjun Wang, Zegang Liu","doi":"10.1111/cbdd.14557","DOIUrl":"10.1111/cbdd.14557","url":null,"abstract":"<p>Recently, natural compounds such as quercetin have gained an increasing amount of attention in treating breast cancer. However, the exact mechanisms responsible for the antiproliferative functions of quercetin are not completely understood. Therefore, we aimed to examine quercetin impacts on breast cancer cell proliferation and survival and the involvement of PI3K/Akt/mTOR pathway. Breast cancer MDA-MB-231 and MCF-7 cells were exposed to quercetin, and cell proliferation was assessed by MTT assay. ELISA was applied to evaluate cell apoptosis. The expression levels of apoptotic mediators such as caspase-3, Bcl-2, Bax and PI3K, Akt, mTOR, and PTEN were assessed via qRT-PCR and western blot. We found that quercetin suppressed dose dependently cell growth capacity in MDA-MB-231 and MCF-7 cells. In addition, quercetin treatment increase apoptosis in both cells lines via modulating the pro- and antiapoptotic markers. Quercetin upregulated PTEN and downregulated PI3K, Akt, and mTOR, hence suppressing this signaling pathway in cells. In conclusion, we showed antiproliferative and pro-apoptotic function of quercetin in breast cancer cell lines, which is mediated by targeting and suppressing PI3K/Akt/mTOR signal transduction.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Upasana Sharma, Rajnish Kumar, Avijit Mazumder, Salahuddin, Neelima Kukreti, Rashmi Mishra, M. V. N. L. Chaitanya
{"title":"Substrate-based synthetic strategies and biological activities of 1,3,4-oxadiazole: A review","authors":"Upasana Sharma, Rajnish Kumar, Avijit Mazumder, Salahuddin, Neelima Kukreti, Rashmi Mishra, M. V. N. L. Chaitanya","doi":"10.1111/cbdd.14552","DOIUrl":"10.1111/cbdd.14552","url":null,"abstract":"<p>The five-membered 1,3,4-oxadiazole heterocyclic ring has received considerable attention because of its unique bio-isosteric properties and an unusually wide spectrum of biological activities. After a century since 1,3,4-oxadiazole was discovered, its uncommon potential attracted medicinal chemist's attention, leading to the discovery of a few presently accessible drugs containing 1,3,4-oxadiazole units, and a large number of patents have been granted on research related to 1,3,4-oxadiazole. It is worth noting that interest in 1,3,4-oxadiazoles' biological applications has doubled in the last few years. Herein, this review presents a comprehensive overview of the recent achievements in the synthesis of 1,3,4-oxadiazole-based compounds and highlights the major advances in their biological applications in the last 10 years, as well as brief remarks on prospects for further development. We hope that researchers across the scientific streams will benefit from the presented review articles for designing their work related to 1,3,4-oxadiazoles.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201594","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Peimenine unleashes therapeutic promise in urothelial bladder cancer: inhibition of proliferation, induction of cell death and modulation of key pathways","authors":"Zhao Yang, Rui Guo, Ying Bi, Wenkai Xu, Mingxuan Hao, Youfeng Liang, Yongchao Li, Haifeng Wang, Jun Zhang, Jianxin Xie, Chuanxing Wan, Jirui Sun","doi":"10.1111/cbdd.14528","DOIUrl":"10.1111/cbdd.14528","url":null,"abstract":"<p>Peimenine (PEI) is a steroid alkaloid substance isolated from <i>Fritillaria thunbergii</i> bulbs. It has various pharmacological activities, such as relief from coughs and asthma, expectorant properties, antibacterial effects, sedative qualities, and anti-inflammatory properties. Notably, PEI can effectively inhibit the proliferation and tumor formation of liver cancer and osteosarcoma cells by inducing autophagic cell death. However, the precise effect and mechanisms of PEI on urothelial bladder cancer (UBC) cells remain uncertain. Thus, this study aims to investigate the impact of PEI on UBC cells both in vivo and in vitro. The IC<sub>50</sub> values of BIU-87 and EJ-1 cells after 48 h were 710.3 and 651.1 μg/mL, respectively. Additionally, PEI blocked the cell cycle in BIU-87 and EJ-1 cells during the G1 phase. Furthermore, it hindered the migration of BIU-87 and EJ-1 cells substantially. PEI significantly inhibited the tumor development of EJ-1 cells within the xenograft tumor model in vivo. Mechanically, PEI augmented the protein and mRNA expression of <i>BIM</i>, <i>BAK1</i>, and <i>Cytochrome C</i> (<i>CYCS)</i> in UBC cells. Taken together, PEI suppressed the proliferation of UBC cells both in vitro and in vivo by inducing cell death and cell cycle arrest, suggesting that PEI could be applied in the treatment of UBC.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141173788","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Design, synthesis, and antibacterial activity of pleuromutilin derivatives","authors":"Hui-xian Liu, Wen-yu Yao, Ge Cui, Jing Zhou, Hao Yan, Hui Guo, Yu-wei Wang, Yue Zhang","doi":"10.1111/cbdd.14554","DOIUrl":"10.1111/cbdd.14554","url":null,"abstract":"<p>This paper reports the design, synthesis, and antibacterial activity study of pleuromutilin derivatives with 2-methyl-4-nitroaniline and 2-methoxy-4-nitroaniline side chains at the C22 position. The structures of the new compounds were characterized by <sup>1</sup>H-NMR, <sup>13</sup>C-NMR and HRMS. The inhibitory activity of the compounds against MSSA, <i>pyogeniccoccus</i>, <i>streptococcus</i>, and MRSA strains was determined using the micro broth dilution method. The results showed that the compounds exhibited certain activity against Gram-positive bacteria, among which compounds A8a, A8b, A8c, A8d, and A7 demonstrated superior antibacterial activity against MSSA, MRSA, and <i>pyogeniccoccus</i> compared to tiamulin, although the derivatives showed lower antibacterial activity against <i>streptococcus</i> compared to the control drug. Based on the favorable in vitro activity of A8c, the time-kill kinetics against MRSA were evaluated, revealing that compound A8c could inhibit bacterial proliferation in a concentration-dependent manner.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141162867","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dmitry I. Osolodkin, Liubov I. Kozlovskaya, Ildar R. Iusupov, Alexander V. Kurkin, Elena Y. Shustova, Alexey A. Orlov, Evgeny V. Khvatov, Elena S. Mutnykh, Svetlana S. Kurashova, Anna N. Vetrova, Darya O. Yatsenko, Alexander S. Goryashchenko, Vladimir N. Ivanov, Evgeny R. Lukyanenko, Evgenia V. Karpova, Daria A. Stepanova, Viktor P. Volok, Svetlana E. Sotskova, Tamara K. Dzagurova, Galina G. Karganova, Alexander N. Lukashev, Aydar A. Ishmukhametov
{"title":"Phenotypic assessment of antiviral activity for spiro-annulated oxepanes and azepenes","authors":"Dmitry I. Osolodkin, Liubov I. Kozlovskaya, Ildar R. Iusupov, Alexander V. Kurkin, Elena Y. Shustova, Alexey A. Orlov, Evgeny V. Khvatov, Elena S. Mutnykh, Svetlana S. Kurashova, Anna N. Vetrova, Darya O. Yatsenko, Alexander S. Goryashchenko, Vladimir N. Ivanov, Evgeny R. Lukyanenko, Evgenia V. Karpova, Daria A. Stepanova, Viktor P. Volok, Svetlana E. Sotskova, Tamara K. Dzagurova, Galina G. Karganova, Alexander N. Lukashev, Aydar A. Ishmukhametov","doi":"10.1111/cbdd.14553","DOIUrl":"10.1111/cbdd.14553","url":null,"abstract":"<p>Evolutionary potential of viruses can result in outbreaks of well-known viruses and emergence of novel ones. Pharmacological methods of intervening the reproduction of various less popular, but not less important viruses are not available, as well as the spectrum of antiviral activity for most known compounds. In the framework of chemical biology paradigm, characterization of antiviral activity spectrum of new compounds allows to extend the antiviral chemical space and provides new important structure–activity relationships for data-driven drug discovery. Here we present a primary assessment of antiviral activity of spiro-annulated derivatives of seven-membered heterocycles, oxepane and azepane, in phenotypic assays against viruses with different genomes, virion structures, and genome realization schemes: orthoflavivirus (tick-borne encephalitis virus, TBEV), enteroviruses (poliovirus, enterovirus A71, echovirus 30), adenovirus (human adenovirus C5), hantavirus (Puumala virus). Hit compounds inhibited reproduction of adenovirus C5, the only DNA virus in the studied set, in the yield reduction assay, and did not inhibit reproduction of RNA viruses.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141093454","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Juliana da Trindade Granato, Emerson Teixeira da Silva, Ari Sérgio de Oliveira Lemos, Patrícia de Almeida Machado, Victor do Valle Midlej, Luciana Maria Ribeiro Antinarelli, Adolfo Firmino da Silva Neto, Marcus Vinícius Nora Souza, Elaine Soares Coimbra
{"title":"4-Quinolinylhydrazone analogues kill Leishmania (Leishmania) amazonensis by inducing apoptosis and mitochondria-dependent pathway cell death","authors":"Juliana da Trindade Granato, Emerson Teixeira da Silva, Ari Sérgio de Oliveira Lemos, Patrícia de Almeida Machado, Victor do Valle Midlej, Luciana Maria Ribeiro Antinarelli, Adolfo Firmino da Silva Neto, Marcus Vinícius Nora Souza, Elaine Soares Coimbra","doi":"10.1111/cbdd.14535","DOIUrl":"10.1111/cbdd.14535","url":null,"abstract":"<p>Despite efforts, available alternatives for the treatment of leishmaniasis are still scarce. In this work we tested a class of 15 quinolinylhydrazone analogues and presented data that support the use of the most active compound in cutaneous leishmaniasis caused by <i>Leishmania amazonensis</i>. In general, the compounds showed activity at low concentrations for both parasitic forms (5.33–37.04 μM to promastigotes, and 14.31–61.98 μM to amastigotes). In addition, the best compound (<b>MHZ15</b>) is highly selective for the parasite. Biochemical studies indicate that the treatment of promastigotes with <b>MHZ15</b> leads the loss of mitochondrial potential and increase in ROS levels as the primary effects, which triggers accumulation of lipid droplets, loss of plasma membrane integrity and apoptosis hallmarks, including DNA fragmentation and phosphatidylserine exposure. These effects were similar in the intracellular form of the parasite. However, in this parasitic form there is no change in plasma membrane integrity in the observed treatment time, which can be attributed to metabolic differences and the resilience of the amastigote. Also, ultrastructural changes such as vacuolization suggesting autophagy were observed. The in vivo effectiveness of <b>MHZ15</b> in the experimental model of cutaneous leishmaniasis was carried out in mice of the BALB/c strain infected with <i>L</i>. <i>amazonensis</i>. The treatment by intralesional route showed that <b>MHZ15</b> acted with great efficiency with significantly reduction in the parasite load in the injured paws and draining lymph nodes, without clinical signs of distress or compromise of animal welfare. In vivo toxicity was also evaluated and null alterations in the levels of hepatic enzymes aspartate aminotransferase, and alanine aminotransferase was observed. The data presented herein demonstrates that <b>MHZ15</b> exhibits a range of favorable characteristics conducive to the development of an antileishmanial agent.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141077130","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Design, synthesis and neuroprotective biological evaluation of novel HDAC6 inhibitors incorporating benzothiadiazinyl systems as cap groups","authors":"Bo Han, Xiu Gu, Mengfei Wang, Huihao Wang, Niubing Sun, Xuezhi Yang, Qingwei Zhang","doi":"10.1111/cbdd.14556","DOIUrl":"10.1111/cbdd.14556","url":null,"abstract":"<p>Histone deacetylase 6 (HDAC6), as the key regulatory enzyme, plays an important role in the development of the nervous system. More and more studies indicate that HDAC6 has become a promising therapeutic target for CNS diseases. Herein we designed and synthesized a series of novel HDAC6 inhibitors with benzothiadiazinyl systems as cap groups and evaluated their activity in vitro and in vivo. Among them, compound <b>3</b> exhibited superior selective inhibitory activity against HDAC6 (IC<sub>50</sub> = 5.1 nM, about 30-fold selectivity over HDAC1). The results of docking showed that compound <b>3</b> can interact well with the key amino acid residues of HDAC6. Compound <b>3</b> showed lower cytotoxicity (20 μM to SH-SY5Y cells, inhibition rate = 25.75%) and better neuroprotective activity against L-glutamate-induced SH-SY5Y cell injury model in vitro. Meanwhile, compound <b>3</b> exhibited weak cardiotoxicity (10 μM hERG inhibition rate = 17.35%) and possess good druggability properties. Especially, compound <b>3</b> could significantly reduce cerebral infarction from 49.87% to 32.18%, and similar with butylphthalide in MCAO model, indicating potential clinical application prospects for alleviating ischemic stroke-induced brain infarction.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141077131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gayathri Seenivasan, Sarwat Asma Ziya Ahmad, Nikhil Kumar Tuti, Unnikrishnan P. Shaji, Susmita Das, Faiz Ahmed Khan, Roy Anindya
{"title":"Evaluation of a panel of furochromenones as the activator and inhibitor of tyrosinase","authors":"Gayathri Seenivasan, Sarwat Asma Ziya Ahmad, Nikhil Kumar Tuti, Unnikrishnan P. Shaji, Susmita Das, Faiz Ahmed Khan, Roy Anindya","doi":"10.1111/cbdd.14539","DOIUrl":"10.1111/cbdd.14539","url":null,"abstract":"<p>Tyrosinase is a copper-containing enzyme involved in the biosynthesis of melanin pigment. While the excess production of melanin causes hyperpigmentation of human skin, hypopigmentation results in medical conditions like vitiligo. Tyrosinase inhibitors could be used as efficient skin whitening agents and tyrosinase agonists could be used for enhanced melanin synthesis and skin protection from UV exposure. Among a wide range of tyrosinase-regulating compounds, natural and synthetic derivatives of furochromenones, such as 8-methoxypsoralen (8-MOP), are known to both activate and inhibit tyrosinase. We recently reported a synthetic approach to generate a variety of dihydrofuro[3,2-c]chromenones and furo[3,2-c]chromenones in a metal-free condition. In the present study, we investigated these compounds for their potential as antagonists or agonists of tyrosinase. Using fungal tyrosinase-based in vitro biochemical assay, we obtained one compound (<b>3k</b>) which could inhibit tyrosinase activity, and the other compound (<b>4f</b>) that stimulated tyrosinase activity. The kinetic studies revealed that compound <b>3k</b> caused ‘mixed’ type tyrosinase inhibition and <b>4f</b> stimulated the catalytic efficiency. Studying the mechanisms of these compounds may provide a basis for the development of new effective tyrosinase inhibitors or activators.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140961105","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}