Pub Date : 2025-12-31DOI: 10.1016/j.bmcl.2025.130531
Jufeng Sun , Yixiao Wang , Rufei Wang , Yuan Yao , Jiarui Wang , Guojun Pan , Renshuai Zhang , Xiaoguang Zhao
Dioscin, a natural steroidal saponin derived from Dioscorea species, has been reported to exhibit lipid-lowering activity despite its extremely low oral bioavailability. This study aimed to elucidate whether dioscin exerts its lipid-lowering effects through intestinal mechanisms. Using both high-fat diet (HFD)-induced and acute lipid-challenge rat models, dioscin significantly reduced plasma triglyceride (TG) and total cholesterol (TC) levels, indicating an inhibitory effect on intestinal lipid absorption. In vitro assays demonstrated that dioscin inhibited pancreatic triglyceride lipase (PTL) activity with moderate potency and suppressed Niemann-Pick C1-Like 1 (NPC1L1)-mediated cholesterol uptake in Caco-2 cells. Surface plasmon resonance (SPR) analysis and molecular docking suggested direct interactions between dioscin and both PTL and NPC1L1, suggesting that dioscin interferes with intestinal lipid absorption through mechanisms involving TG hydrolysis and cholesterol transport. These findings provide mechanistic insight into the intestinal lipid-lowering activity of dioscin and highlight its potential as a natural lead compound for the development of safe and intestine- restricted lipid-lowering agents.
{"title":"Dioscin attenuates hyperlipidemia by dual modulation of intestinal triglyceride and cholesterol absorption","authors":"Jufeng Sun , Yixiao Wang , Rufei Wang , Yuan Yao , Jiarui Wang , Guojun Pan , Renshuai Zhang , Xiaoguang Zhao","doi":"10.1016/j.bmcl.2025.130531","DOIUrl":"10.1016/j.bmcl.2025.130531","url":null,"abstract":"<div><div>Dioscin, a natural steroidal saponin derived from <em>Dioscorea</em> species, has been reported to exhibit lipid-lowering activity despite its extremely low oral bioavailability. This study aimed to elucidate whether dioscin exerts its lipid-lowering effects through intestinal mechanisms. Using both high-fat diet (HFD)-induced and acute lipid-challenge rat models, dioscin significantly reduced plasma triglyceride (TG) and total cholesterol (TC) levels, indicating an inhibitory effect on intestinal lipid absorption. In vitro assays demonstrated that dioscin inhibited pancreatic triglyceride lipase (PTL) activity with moderate potency and suppressed Niemann-Pick C1-Like 1 (NPC1L1)-mediated cholesterol uptake in Caco-2 cells. Surface plasmon resonance (SPR) analysis and molecular docking suggested direct interactions between dioscin and both PTL and NPC1L1, suggesting that dioscin interferes with intestinal lipid absorption through mechanisms involving TG hydrolysis and cholesterol transport. These findings provide mechanistic insight into the intestinal lipid-lowering activity of dioscin and highlight its potential as a natural lead compound for the development of safe and intestine- restricted lipid-lowering agents.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130531"},"PeriodicalIF":2.2,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145891827","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}
Pub Date : 2025-12-31DOI: 10.1016/j.bmcl.2025.130530
Nooshin Mesbahi , Brenna C. McAllister , Hosog Yoon , Aaron T. Hendricksen , Melody D. Fulton , Leslie A. Caromile , Clifford E. Berkman
Targeted payload release in cancer cells can be modulated by tuning both the linker, spacer, and the payload chemistries. In previous studies, a PSMA-targeted probe incorporating a 7-amino-4-methylcoumarin (AMC) payload and a PEG linker resulted in predominant payload release in the lysosome (pH ∼5.0). Here, we introduce a second-generation PSMA-targeted turn-on probe with a shorter, hydrophobic linker and a 7-hydroxy-4-methylcoumarin (HMC) payload. Based on pH-dependent kinetic studies, the HMC payload exhibits faster cleavage at a slightly higher pH (pH 5.5), suggesting an earlier release—potentially more in early endosomes than lysosomes. Our results demonstrate that subtle changes in linker and payload structures can alter intracellular release kinetics, offering improved control over the cellular release site, which is critical for optimizing targeted therapeutic and imaging strategies in prostate cancer cells.
{"title":"Second generation PSMA-targeted turn-on probe for imaging cargo release in prostate cancer cells","authors":"Nooshin Mesbahi , Brenna C. McAllister , Hosog Yoon , Aaron T. Hendricksen , Melody D. Fulton , Leslie A. Caromile , Clifford E. Berkman","doi":"10.1016/j.bmcl.2025.130530","DOIUrl":"10.1016/j.bmcl.2025.130530","url":null,"abstract":"<div><div>Targeted payload release in cancer cells can be modulated by tuning both the linker, spacer, and the payload chemistries. In previous studies, a PSMA-targeted probe incorporating a 7-amino-4-methylcoumarin (AMC) payload and a PEG linker resulted in predominant payload release in the lysosome (pH ∼5.0). Here, we introduce a second-generation PSMA-targeted turn-on probe with a shorter, hydrophobic linker and a 7-hydroxy-4-methylcoumarin (HMC) payload. Based on pH-dependent kinetic studies, the HMC payload exhibits faster cleavage at a slightly higher pH (pH 5.5), suggesting an earlier release—potentially more in early endosomes than lysosomes. Our results demonstrate that subtle changes in linker and payload structures can alter intracellular release kinetics, offering improved control over the cellular release site, which is critical for optimizing targeted therapeutic and imaging strategies in prostate cancer cells.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130530"},"PeriodicalIF":2.2,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145891838","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}
Pub Date : 2025-12-30DOI: 10.1016/j.bmcl.2025.130529
Vineetha K. Unnikrishnan , Niranjana sri Sundaramoorthy , Ramaravinth Manivannan , Markabandhu Shanthi , Divya Prakash Gnanadhas , Soundarya Alexander , Venkatasubramanian Ulaganathan , Ramesh Subburethinam , Saisubramanian Nagarajan
Candida albicans is an opportunistic fungus that causes infections in people with weakened immune system. Candida has the propensity to gain drug resistance by mutation, and its ability to extrude antifungals through drug transporters. Efflux pump inhibitors increase the intracellular drug concentration and restore drug sensitivity in resistant strains. We synthesized and screened 10 azole heterocyclic derivatives (pyrazole-5-amine and 2H-indazole) against Mdr1p/Cdr1p overexpressing and knock-out strains of Candida albicans. Among the derivatives screened, sulfenylated pyrazole-5-amine (designated as 4a) displayed strong efflux inhibitory potential and caused a 64-fold reduction in the fluconazole MIC. Compound 4a inhibited the Mdr1p pump, as MIC reversal was not observed in the Mdr1p knock-out strain. 4a exhibited synergy with fluconazole only against Mdr1p harboring strains, as discerned by the Checkerboard assay. Combining 4a with fluconazole restricted microbial growth and reduced cell counts by 4 log fold relative to treatment with fluconazole in a time-kill assay. Infection of RAW macrophages followed by treatment with a combination of sulfenylated aminopyrazole & fluconazole resulted in a significant 2 log reduction in intracellular cell counts relative to treatment with fluconazole. 4a also inhibited biofilm formation and yeast to hyphal shift in Candida, thereby reducing virulence. Combining EPIs with antifungal drugs has the potential to improve the treatment of C. albicans infections and reduce the risk of drug resistance. However, further research is needed to determine the safety and efficacy of these compounds in humans.
{"title":"Evaluation of the efflux inhibitory potential of aminopyrazole derivative to restore azole sensitivity in Candida albicans","authors":"Vineetha K. Unnikrishnan , Niranjana sri Sundaramoorthy , Ramaravinth Manivannan , Markabandhu Shanthi , Divya Prakash Gnanadhas , Soundarya Alexander , Venkatasubramanian Ulaganathan , Ramesh Subburethinam , Saisubramanian Nagarajan","doi":"10.1016/j.bmcl.2025.130529","DOIUrl":"10.1016/j.bmcl.2025.130529","url":null,"abstract":"<div><div><em>Candida albicans</em> is an opportunistic fungus that causes infections in people with weakened immune system. Candida has the propensity to gain drug resistance by mutation, and its ability to extrude antifungals through drug transporters. Efflux pump inhibitors increase the intracellular drug concentration and restore drug sensitivity in resistant strains. We synthesized and screened 10 azole heterocyclic derivatives (pyrazole-5-amine and 2H-indazole) against Mdr1p/Cdr1p overexpressing and knock-out strains of <em>Candida albicans.</em> Among the derivatives screened, sulfenylated pyrazole-5-amine (designated as <strong>4a)</strong> displayed strong efflux inhibitory potential and caused a 64-fold reduction in the fluconazole MIC. Compound <strong>4a</strong> inhibited the Mdr1p pump, as MIC reversal was not observed in the Mdr1p knock-out strain. <strong>4a</strong> exhibited synergy with fluconazole only against Mdr1p harboring strains, as discerned by the Checkerboard assay. Combining <strong>4a</strong> with fluconazole restricted microbial growth and reduced cell counts by 4 log fold relative to treatment with fluconazole in a time-kill assay. Infection of RAW macrophages followed by treatment with a combination of sulfenylated aminopyrazole & fluconazole resulted in a significant 2 log reduction in intracellular cell counts relative to treatment with fluconazole. <strong>4a</strong> also inhibited biofilm formation and yeast to hyphal shift in <em>Candida</em>, thereby reducing virulence. Combining EPIs with antifungal drugs has the potential to improve the treatment of <em>C. albicans</em> infections and reduce the risk of drug resistance. However, further research is needed to determine the safety and efficacy of these compounds in humans.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130529"},"PeriodicalIF":2.2,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145888389","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}
Pub Date : 2025-12-30DOI: 10.1016/j.bmcl.2025.130526
Yuichiro Kawamoto , Shohei Takase , Kumar Ashutosh , Hiroki Maruo , Yuki Maemoto , Kam Y.J. Zhang , Hisanaka Ito , Akihiro Ito
CBX2 protein plays an important role in methyl-lysine recognition involved in epigenetic regulation. In contrast to CBX7 modulators, there are few reported CBX2 inhibitors, most of which are peptidergic molecules. This fact encouraged us to search non-peptidic small molecule CBX2 inhibitors with sufficient cell permeability by in silico screening followed by synthetic evolution. In virtual screening, we identified a 4-aminobenzamide scaffold with high in silico score and Nano BRET activities. Based on several in silico hit compounds, we synthesized a series of 4-aminobenzamide derivatives and tested their Nano BRET activities. Among them, compound 37 showed moderate CBX2 inhibitory activity with an IC50 value of 9.6 μM, a potential lead compound. This is a first small molecule CBX2 inhibitor identified as a result of SAR studies and lead optimization by medicinal chemistry.
{"title":"Discovery of novel 4-aminobenzamide derivatives as small molecule CBX2 inhibitors","authors":"Yuichiro Kawamoto , Shohei Takase , Kumar Ashutosh , Hiroki Maruo , Yuki Maemoto , Kam Y.J. Zhang , Hisanaka Ito , Akihiro Ito","doi":"10.1016/j.bmcl.2025.130526","DOIUrl":"10.1016/j.bmcl.2025.130526","url":null,"abstract":"<div><div>CBX2 protein plays an important role in methyl-lysine recognition involved in epigenetic regulation. In contrast to CBX7 modulators, there are few reported CBX2 inhibitors, most of which are peptidergic molecules. This fact encouraged us to search non-peptidic small molecule CBX2 inhibitors with sufficient cell permeability by <em>in silico</em> screening followed by synthetic evolution. In virtual screening, we identified a 4-aminobenzamide scaffold with high <em>in silico</em> score and Nano BRET activities. Based on several <em>in silico</em> hit compounds, we synthesized a series of 4-aminobenzamide derivatives and tested their Nano BRET activities. Among them, compound <strong>37</strong> showed moderate CBX2 inhibitory activity with an IC<sub>50</sub> value of 9.6 μM, a potential lead compound. This is a first small molecule CBX2 inhibitor identified as a result of SAR studies and lead optimization by medicinal chemistry.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130526"},"PeriodicalIF":2.2,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145888371","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}
Pub Date : 2025-12-27DOI: 10.1016/j.bmcl.2025.130527
Natsumi Yamada , Sora Suzuki , Nanae Fukahori , Yoshiyuki Yamaoka , Yuta Komiyama , Ninako Kimura , Koji Umezawa , Isao Kii
A folding intermediate of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) in the non-native state autophosphorylates intramolecularly, whereas the folded conformation of DYRK1A in the native state no longer catalyzes this reaction. We identified FINDY, a small molecule that selectively inhibits the DYRK1A folding intermediate but not its fully folded conformation. These findings indicated the presence of a unique binding site that is exposed only in the folding intermediate. In the previous study, we developed a facile method for screening of inhibitors that target the folding intermediate in the non-native state using recombinant folded proteins in the native state. We found that FINDY and its derivatives target both DYRK1A and DYRK1B in their non-native states. In this study, we examined the potency of these inhibitors on DYRK2, another member of the DYRK family. FINDY inhibited DYRK2 in the non-native state slightly greater than that in the native state. Although RD0448, a FINDY derivative, selectively targets DYRK1A/1B in their non-native states, RD0448 exhibited no selectivity between the native and non-native states of DYRK2, indicating that the RD0448-binding site is hidden in the native state of DYRK1A/1B but is exposed in the native state of DYRK2. Furthermore, RD0448 bound tightly to DYRK1A/1B, whereas RD0448 showed weak affinity for DYRK2 and rapidly dissociated from the binding complex. These results suggest a correlation between the tight binding of the inhibitors and their target selectivity toward the non-native state in DYRK family kinases.
{"title":"Correlation between tight binding of inhibitors and their target selectivity toward the non-native state in the DYRK family of kinases","authors":"Natsumi Yamada , Sora Suzuki , Nanae Fukahori , Yoshiyuki Yamaoka , Yuta Komiyama , Ninako Kimura , Koji Umezawa , Isao Kii","doi":"10.1016/j.bmcl.2025.130527","DOIUrl":"10.1016/j.bmcl.2025.130527","url":null,"abstract":"<div><div>A folding intermediate of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) in the non-native state autophosphorylates intramolecularly, whereas the folded conformation of DYRK1A in the native state no longer catalyzes this reaction. We identified FINDY, a small molecule that selectively inhibits the DYRK1A folding intermediate but not its fully folded conformation. These findings indicated the presence of a unique binding site that is exposed only in the folding intermediate. In the previous study, we developed a facile method for screening of inhibitors that target the folding intermediate in the non-native state using recombinant folded proteins in the native state. We found that FINDY and its derivatives target both DYRK1A and DYRK1B in their non-native states. In this study, we examined the potency of these inhibitors on DYRK2, another member of the DYRK family. FINDY inhibited DYRK2 in the non-native state slightly greater than that in the native state. Although RD0448, a FINDY derivative, selectively targets DYRK1A/1B in their non-native states, RD0448 exhibited no selectivity between the native and non-native states of DYRK2, indicating that the RD0448-binding site is hidden in the native state of DYRK1A/1B but is exposed in the native state of DYRK2. Furthermore, RD0448 bound tightly to DYRK1A/1B, whereas RD0448 showed weak affinity for DYRK2 and rapidly dissociated from the binding complex. These results suggest a correlation between the tight binding of the inhibitors and their target selectivity toward the non-native state in DYRK family kinases.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130527"},"PeriodicalIF":2.2,"publicationDate":"2025-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145852768","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}
Pub Date : 2025-12-27DOI: 10.1016/j.bmcl.2025.130528
Meiling Zhang , Shijia Xiao , Kexin Yan , Jiyue Sun , Chen Xi , Yuhong Qin , Cheng Dong , Dongxing Chen
The proteolysis-targeting chimera (PROTAC) technology utilizes heterobifunctional molecules to induce targeted protein degradation through the ubiquitin-proteasome system. Structurally, PROTAC molecules consist of a target protein ligand and an E3 ligase ligand covalently linked by a suitable linker arm, forming the target protein-PROTAC-E3 ligase stable ternary complex and bringing the target protein in proximity to the E3 ligase for ubiquitination and subsequent proteasomal degradation. However, only a few E3 ligases have been used to generate effective PROTACs with limited small molecule E3 ligase ligands. Therefore, there is an urgent need to discover novel E3 ligase ligands to expand the toolbox for PROTACs. Unlike traditional E3 ligases such as CRBN and VHL, GID4 E3 ligase recognizes substrates bearing N-terminal proline or other small residues through the Pro/N-degron pathway, and has already been successfully leveraged in PROTAC technology. Here, we reported the development of a fluorescent probe YG11, with a Kd value of 8.1 ± 0.7 nM for GID4. With this probe, we established a robust fluorescence polarization (FP)-based competition assay for evaluation of GID4 ligands. The assay exhibited a high signal-to-noise ratio of over 20, 2.5 % DMSO tolerance, and a Z'-factor of 0.84, confirming its suitability and robustness for high-throughput screening. Thus, by enabling rapid identification of GID4 ligands, this FP competition assay promises to substantially advance PROTAC development initiatives.
{"title":"Development of the fluorescence polarization-based competition assay for the E3 ligase GID4","authors":"Meiling Zhang , Shijia Xiao , Kexin Yan , Jiyue Sun , Chen Xi , Yuhong Qin , Cheng Dong , Dongxing Chen","doi":"10.1016/j.bmcl.2025.130528","DOIUrl":"10.1016/j.bmcl.2025.130528","url":null,"abstract":"<div><div>The proteolysis-targeting chimera (PROTAC) technology utilizes heterobifunctional molecules to induce targeted protein degradation through the ubiquitin-proteasome system. Structurally, PROTAC molecules consist of a target protein ligand and an E3 ligase ligand covalently linked by a suitable linker arm, forming the target protein-PROTAC-E3 ligase stable ternary complex and bringing the target protein in proximity to the E3 ligase for ubiquitination and subsequent proteasomal degradation. However, only a few E3 ligases have been used to generate effective PROTACs with limited small molecule E3 ligase ligands. Therefore, there is an urgent need to discover novel E3 ligase ligands to expand the toolbox for PROTACs. Unlike traditional E3 ligases such as CRBN and VHL, GID4 E3 ligase recognizes substrates bearing N-terminal proline or other small residues through the Pro/N-degron pathway, and has already been successfully leveraged in PROTAC technology. Here, we reported the development of a fluorescent probe <strong>YG11</strong>, with a K<sub>d</sub> value of 8.1 ± 0.7 nM for GID4. With this probe, we established a robust fluorescence polarization (FP)-based competition assay for evaluation of GID4 ligands. The assay exhibited a high signal-to-noise ratio of over 20, 2.5 % DMSO tolerance, and a Z'-factor of 0.84, confirming its suitability and robustness for high-throughput screening. Thus, by enabling rapid identification of GID4 ligands, this FP competition assay promises to substantially advance PROTAC development initiatives.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130528"},"PeriodicalIF":2.2,"publicationDate":"2025-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145852801","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}
Pub Date : 2025-12-25DOI: 10.1016/j.bmcl.2025.130525
Hao Sun , Xu Yan , Chunhuan Mo , Jia Chen , Yan Liu , Yaohui Liu , Xia Han , Jianqin Gao , Lehua Yin , Jinghua Xu , Yuan Chen , Xinhua He
Monophosphorylation is a key rate-limiting step for the efficacy of broad-spectrum nucleoside antiviral drugs, and designing phosphate prodrugs is an effective method to enhance antiviral efficacy. N4-hydroxycytidine (NHC) has a broad-spectrum antiviral effect. Based on the mechanism of action and metabolic inactivation characteristics of NHC, we designed a series of dual ester prodrugs targeting the N4-hydroxyl group and the 5′-phosphate of the ribose ring. Twenty-nine compounds were designed and synthesized. The cytotoxicity assay results showed low cytotoxicity (no significant toxicity at 160 μM). Using the Vesicular Stomatitis Virus expressing Green Fluorescent Protein (VSV-GFP) virus screen, it was found that all of them have antiviral activity. Compounds 2328 and 2322 showed better antiviral activity than molnupiravir. The EC50s of 2328 against VSV, HMPV-A2, and RSV-A2 viruses were 2.19 μM, 35.6 μM, and 53.5 μM, respectively, and the EC50s of 2322 against VSV, HMPV-A2, and RSV-A2 were 6.23 μM, 52.8 μM, and 28.2 μM, respectively. Structure-activity relationships indicate that the antiviral activity of compounds at the cellular level is closely linked to the structure of the prodrug's phosphate ester, whereas the ester group on the N4-hydroxyl is more tolerant of diverse functional groups.
{"title":"Design, synthesis, and antiviral activity evaluation of the bis-ester prodrug of N4-hydroxycytidine phosphate","authors":"Hao Sun , Xu Yan , Chunhuan Mo , Jia Chen , Yan Liu , Yaohui Liu , Xia Han , Jianqin Gao , Lehua Yin , Jinghua Xu , Yuan Chen , Xinhua He","doi":"10.1016/j.bmcl.2025.130525","DOIUrl":"10.1016/j.bmcl.2025.130525","url":null,"abstract":"<div><div>Monophosphorylation is a key rate-limiting step for the efficacy of broad-spectrum nucleoside antiviral drugs, and designing phosphate prodrugs is an effective method to enhance antiviral efficacy. <em>N4</em>-hydroxycytidine (NHC) has a broad-spectrum antiviral effect. Based on the mechanism of action and metabolic inactivation characteristics of NHC, we designed a series of dual ester prodrugs targeting the N4-hydroxyl group and the 5′-phosphate of the ribose ring. Twenty-nine compounds were designed and synthesized. The cytotoxicity assay results showed low cytotoxicity (no significant toxicity at 160 μM). Using the Vesicular Stomatitis Virus expressing Green Fluorescent Protein (VSV-GFP) virus screen, it was found that all of them have antiviral activity. Compounds 2328 and 2322 showed better antiviral activity than molnupiravir. The EC<sub>50</sub>s of 2328 against VSV, HMPV-A2, and RSV-A2 viruses were 2.19 μM, 35.6 μM, and 53.5 μM, respectively, and the EC<sub>50</sub>s of 2322 against VSV, HMPV-A2, and RSV-A2 were 6.23 μM, 52.8 μM, and 28.2 μM, respectively. Structure-activity relationships indicate that the antiviral activity of compounds at the cellular level is closely linked to the structure of the prodrug's phosphate ester, whereas the ester group on the <em>N4</em>-hydroxyl is more tolerant of diverse functional groups.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130525"},"PeriodicalIF":2.2,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145846166","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}
Pub Date : 2025-12-24DOI: 10.1016/j.bmcl.2025.130523
Renan Augusto Gomes , Leonardo Luiz Gomes Ferreira , Witor Ribeiro Ferraz , Rodrigo Alves Heleno , Fernando Moura Gatti , Mariana Laureano de Souza , Cleydson Breno Rodrigues dos Santos , Adriano Defini Andricopulo , Gustavo Henrique Goulart Trossini
Parasitic diseases like leishmaniasis and Chagas pose significant global health challenges due to limited treatment options and drug resistance. In this study, a series of novel cinnamoyl aryl hydrazone derivatives was synthesized and tested against Leishmania donovani and Trypanosoma cruzi. Four compounds showed promising antileishmanial activity (from 1.27 to 19.53 μM), with two analogues displaying superior potency compared to some current first-line treatments, such as sodium stibogluconate and paromomycin. Computational analyses revealed that activity is driven by specific electronic properties rather than steric factors, while a methyl group consistently reduced potency. The compounds demonstrated favorable predicted ADME properties and were not flagged as aggregators. This research identifies cinnamoyl aryl hydrazones as a promising scaffold for leishmanicidal drug discovery, providing a rational basis for future optimization efforts.
{"title":"Cinnamoyl aryl hydrazones as potent leishmanicidal agents: design, synthesis, and structure–activity relationships","authors":"Renan Augusto Gomes , Leonardo Luiz Gomes Ferreira , Witor Ribeiro Ferraz , Rodrigo Alves Heleno , Fernando Moura Gatti , Mariana Laureano de Souza , Cleydson Breno Rodrigues dos Santos , Adriano Defini Andricopulo , Gustavo Henrique Goulart Trossini","doi":"10.1016/j.bmcl.2025.130523","DOIUrl":"10.1016/j.bmcl.2025.130523","url":null,"abstract":"<div><div>Parasitic diseases like leishmaniasis and Chagas pose significant global health challenges due to limited treatment options and drug resistance. In this study, a series of novel cinnamoyl aryl hydrazone derivatives was synthesized and tested against <em>Leishmania donovani</em> and <em>Trypanosoma cruzi</em>. Four compounds showed promising antileishmanial activity (from 1.27 to 19.53 μM), with two analogues displaying superior potency compared to some current first-line treatments, such as sodium stibogluconate and paromomycin. Computational analyses revealed that activity is driven by specific electronic properties rather than steric factors, while a methyl group consistently reduced potency. The compounds demonstrated favorable predicted ADME properties and were not flagged as aggregators. This research identifies cinnamoyl aryl hydrazones as a promising scaffold for leishmanicidal drug discovery, providing a rational basis for future optimization efforts.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130523"},"PeriodicalIF":2.2,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145843331","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}
Abiraterone, a CYP17A1 inhibitor, is approved by the FDA for castration-resistance prostate cancer. To mimic the structure of abiraterone, we designed and synthesized nine tetrahydroindeno[4,5-c]chromen-4(3H)-one derivatives with pyridine congeners at C-1 position. Notably, 13c and 14c exhibited the GI50 of 10 nM against PC-3 cells compared to 21.4 μM for abiraterone. The docking studies further revealed that 14c shares a similar binding mode with abiraterone at the CYP17A1 active site.
{"title":"Design and synthesis of Tetrahydroindeno[4,5-c]chromen-4(3H)-one derivatives as antiproliferative agents for prostate Cancer cells","authors":"Hui-Yu Chan , Pin-Shuo Huang , Pei-Fang Chiu , Tzung-Sheng Lin , Li-Jou Huang , Pi-Hui Liang , Jih-Hwa Guh , Mei-Hsiang Lin","doi":"10.1016/j.bmcl.2025.130515","DOIUrl":"10.1016/j.bmcl.2025.130515","url":null,"abstract":"<div><div>Abiraterone, a CYP17A1 inhibitor, is approved by the FDA for castration-resistance prostate cancer. To mimic the structure of abiraterone, we designed and synthesized nine tetrahydroindeno[4,5-<em>c</em>]chromen-4(3<em>H</em>)-one derivatives with pyridine congeners at C-1 position. Notably, <strong>13c</strong> and <strong>14c</strong> exhibited the GI<sub>50</sub> of 10 nM against PC-3 cells compared to 21.4 μM for abiraterone. The docking studies further revealed that <strong>14c</strong> shares a similar binding mode with abiraterone at the CYP17A1 active site.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130515"},"PeriodicalIF":2.2,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145843385","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}
Pub Date : 2025-12-23DOI: 10.1016/j.bmcl.2025.130513
Yanlin He , Yuwan Zhang , Wei Wang , Chaoyong Cui , Qianyun Hao , Hongguan Xie , Rui Yang
The endophytic fungus Alternaria sp. S4, derived from Ruta graveolens, shows significant antimicrobial potential, but its active components and mode of action require clarification. Through bioassay-guided fractionation, alternariol (1) and alternariol-5-O-methyl ether (2) were isolated from the ethyl acetate fraction of Alternaria sp. S4 and characterized spectroscopically. Compound 1 exhibited potent bactericidal activity against Staphylococcus aureus (MIC = 4 μg/mL, MBC = 16 μg/mL), whereas compound 2 was inactive. Time-growth assays confirmed concentration-dependent bacterial eradication, with 2× MIC of compound 1 achieving complete growth inhibition. Mechanistic investigations revealed that compound 1 targets the bacterial membrane, inducing depolarization, compromising permeability (evidenced by ion leakage and release of intracellular constituents), and causing ultrastructural damage visualized via SEM. Furthermore, compound 1 prevented biofilm formation via bacterial eradication and disrupted preformed biofilms. Checkerboard assays indicated additive effects when combined with conventional antibiotics. Notably, compound 1 displayed no hemolytic activity even at 1024 μg/mL, highlighting its membrane selectivity. This work identifies alternariol as a promising membrane-targeting bactericidal agent from an endophytic source, capable of preventing biofilm establishment, with potential for combinatorial therapy against S. aureus.
从芦笋中提取的内生真菌Alternaria sp. S4显示出显著的抗菌潜力,但其有效成分和作用方式尚不清楚。采用生物测定引导分离的方法,从Alternaria sp. S4的乙酸乙酯部位分离得到交替蒿醇(1)和交替蒿醇-5- o -甲基醚(2),并对其进行了光谱表征。化合物1对金黄色葡萄球菌具有较强的杀菌活性(MIC = 4 μg/mL, MBC = 16 μg/mL),而化合物2对金黄色葡萄球菌无活性。时间生长试验证实了浓度依赖的细菌根除,化合物1的2倍MIC达到完全生长抑制。机制研究表明,化合物1靶向细菌膜,诱导去极化,损害渗透性(通过离子泄漏和细胞内成分释放证明),并通过扫描电镜观察造成超微结构损伤。此外,化合物1通过细菌根除阻止生物膜的形成,并破坏预先形成的生物膜。棋盘试验表明,当与常规抗生素联合使用时,会产生叠加效应。值得注意的是,化合物1即使在1024 μg/mL浓度下也没有溶血活性,表明其具有膜选择性。这项工作确定了交替蒿醇是一种有前途的内生膜靶向杀菌剂,能够阻止生物膜的形成,具有联合治疗金黄色葡萄球菌的潜力。
{"title":"Alternariol from the endophytic fungus Alternaria sp. S4 acts as a membrane-targeting bactericidal agent against Staphylococcus aureus","authors":"Yanlin He , Yuwan Zhang , Wei Wang , Chaoyong Cui , Qianyun Hao , Hongguan Xie , Rui Yang","doi":"10.1016/j.bmcl.2025.130513","DOIUrl":"10.1016/j.bmcl.2025.130513","url":null,"abstract":"<div><div>The endophytic fungus <em>Alternaria</em> sp. S4, derived from <em>Ruta graveolens</em>, shows significant antimicrobial potential, but its active components and mode of action require clarification. Through bioassay-guided fractionation, alternariol (<strong>1</strong>) and alternariol-5-<em>O</em>-methyl ether (<strong>2</strong>) were isolated from the ethyl acetate fraction of <em>Alternaria</em> sp. S4 and characterized spectroscopically. Compound <strong>1</strong> exhibited potent bactericidal activity against <em>Staphylococcus aureus</em> (MIC = 4 μg/mL, MBC = 16 μg/mL), whereas compound <strong>2</strong> was inactive. Time-growth assays confirmed concentration-dependent bacterial eradication, with 2× MIC of compound <strong>1</strong> achieving complete growth inhibition. Mechanistic investigations revealed that compound <strong>1</strong> targets the bacterial membrane, inducing depolarization, compromising permeability (evidenced by ion leakage and release of intracellular constituents), and causing ultrastructural damage visualized via SEM. Furthermore, compound <strong>1</strong> prevented biofilm formation via bacterial eradication and disrupted preformed biofilms. Checkerboard assays indicated additive effects when combined with conventional antibiotics. Notably, compound <strong>1</strong> displayed no hemolytic activity even at 1024 μg/mL, highlighting its membrane selectivity. This work identifies alternariol as a promising membrane-targeting bactericidal agent from an endophytic source, capable of preventing biofilm establishment, with potential for combinatorial therapy against <em>S. aureus</em>.</div></div>","PeriodicalId":256,"journal":{"name":"Bioorganic & Medicinal Chemistry Letters","volume":"133 ","pages":"Article 130513"},"PeriodicalIF":2.2,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145831710","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}