Pub Date : 2024-12-01Epub Date: 2024-03-15DOI: 10.1080/14756366.2024.2318830
Jung Hun Lee, Sang-Gyu Kim, Kyung-Min Jang, Kyoungmin Shin, Hyeonku Jin, Dae-Wi Kim, Byeong Chul Jeong, Sang Hee Lee
The urgent demand for effective countermeasures against metallo-β-lactamases (MBLs) necessitates development of novel metallo-β-lactamase inhibitors (MBLIs). This study is dedicated to identifying critical chemical moieties within previously developed MBLIs, and critical MBLs should serve as the target in MBLI evaluations. Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA), a systematic literature analysis was conducted, and the NCBI RefSeq genome database was exploited to access the abundance profile and taxonomic distribution of MBLs and their variant types. Through the implementation of two distinct systematic approaches, we elucidated critical chemical moieties of MBLIs, providing pivotal information for rational drug design. We also prioritised MBLs and their variant types, highlighting the imperative need for comprehensive testing to ensure the potency and efficacy of the newly developed MBLIs. This approach contributes valuable information to advance the field of antimicrobial drug discovery.
{"title":"Elucidation of critical chemical moieties of metallo-β-lactamase inhibitors and prioritisation of target metallo-β-lactamases.","authors":"Jung Hun Lee, Sang-Gyu Kim, Kyung-Min Jang, Kyoungmin Shin, Hyeonku Jin, Dae-Wi Kim, Byeong Chul Jeong, Sang Hee Lee","doi":"10.1080/14756366.2024.2318830","DOIUrl":"10.1080/14756366.2024.2318830","url":null,"abstract":"<p><p>The urgent demand for effective countermeasures against metallo-β-lactamases (MBLs) necessitates development of novel metallo-β-lactamase inhibitors (MBLIs). This study is dedicated to identifying critical chemical moieties within previously developed MBLIs, and critical MBLs should serve as the target in MBLI evaluations. Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA), a systematic literature analysis was conducted, and the NCBI RefSeq genome database was exploited to access the abundance profile and taxonomic distribution of MBLs and their variant types. Through the implementation of two distinct systematic approaches, we elucidated critical chemical moieties of MBLIs, providing pivotal information for rational drug design. We also prioritised MBLs and their variant types, highlighting the imperative need for comprehensive testing to ensure the potency and efficacy of the newly developed MBLIs. This approach contributes valuable information to advance the field of antimicrobial drug discovery.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10946278/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140131585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-06-14DOI: 10.1080/14756366.2024.2360063
Xianting Wang, Yidan Deng, Jianmei Wang, Lin Qin, Yimei Du, Qianru Zhang, Di Wu, Xingdong Wu, Jian Xie, Yuqi He, Daopeng Tan
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease mainly caused by insulin resistance, which can lead to a series of complications such as cardiovascular disease, retinopathy, and its typical clinical symptom is hyperglycaemia. Glucosidase inhibitors, including Acarbose, Miglitol, are commonly used in the clinical treatment of hypoglycaemia. In addition, Protein tyrosine phosphatase 1B (PTP1B) is also an important promising target for the treatment of T2DM. Gynostemma pentaphyllum is a well-known oriental traditional medicinal herbal plant, and has many beneficial effects on glucose and lipid metabolism. In the present study, three new and nine known dammarane triterpenoids isolated from G. pentaphyllum, and their structures were elucidated by spectroscopic methods including HR-ESI-MS,1H and 13C NMR and X-ray crystallography. All these compounds were evaluated for inhibitory activity against α-glucosidase, α-amylase and PTP1B. The results suggested that compounds 7∼10 were potential antidiabetic agents with significantly inhibition activity against PTP1B in a dose-dependent manner.
{"title":"New natural protein tyrosine phosphatase 1B inhibitors from <i>Gynostemma pentaphyllum</i>.","authors":"Xianting Wang, Yidan Deng, Jianmei Wang, Lin Qin, Yimei Du, Qianru Zhang, Di Wu, Xingdong Wu, Jian Xie, Yuqi He, Daopeng Tan","doi":"10.1080/14756366.2024.2360063","DOIUrl":"10.1080/14756366.2024.2360063","url":null,"abstract":"<p><p>Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease mainly caused by insulin resistance, which can lead to a series of complications such as cardiovascular disease, retinopathy, and its typical clinical symptom is hyperglycaemia. Glucosidase inhibitors, including Acarbose, Miglitol, are commonly used in the clinical treatment of hypoglycaemia. In addition, Protein tyrosine phosphatase 1B (PTP1B) is also an important promising target for the treatment of T2DM. <i>Gynostemma pentaphyllum</i> is a well-known oriental traditional medicinal herbal plant, and has many beneficial effects on glucose and lipid metabolism. In the present study, three new and nine known dammarane triterpenoids isolated from <i>G. pentaphyllum</i>, and their structures were elucidated by spectroscopic methods including HR-ESI-MS,<sup>1</sup>H and <sup>13</sup>C NMR and X-ray crystallography. All these compounds were evaluated for inhibitory activity against α-glucosidase, α-amylase and PTP1B. The results suggested that compounds <b>7</b>∼<b>10</b> were potential antidiabetic agents with significantly inhibition activity against PTP1B in a dose-dependent manner.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11182071/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141317384","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-06-21DOI: 10.1080/14756366.2024.2358934
Mater H Mahnashi, Mohammed Nahari, Hassan Almasoudi, Abdulaziz Alhasaniah, Sara Elgazwi, Mahrous A Abou-Salim
Novel series of nitric oxide-releasing thiazolidine-2,4-diones (NO-TZD-3a-d,5,6) and 3,4,5-trimethoxychalcone-based multifunctional 1,4-dihydropyrimidines (CDHPM-10a-g) have been designed and synthesised as potent broad-spectrum anticancer agents with potential VEGFR-2 inhibition. The designed analogs were evaluated for their anticancer activities towards a full panel of NCI-60 tumour cell lines and CDHPM-10a-g emerged mean %inhibitions ranging from 76.40 to 147.69%. Among them, CDHPM-10e and CDHPM-10f demonstrated the highest MGI% of 147.69 and 140.24%, respectively. Compounds CDHPM-10a,b,d-f showed higher mean %inhibitory activity than the reference drug sorafenib (MGI% = 105.46%). Superiorly, the hybrid CDHPM-10e displayed the highest potencies towards all the herein tested subpanels of nine types of cancer with MGI50 of 1.83 µM. Also, it revealed potent cytostatic single-digit micromolar activity towards the herein examined cancer cell lines. The designed compounds CDHPM-10a-g were exposed as potent non-selective broad-spectrum anticancer agents over all NCI subpanels with an SI range of 0.66-1.97. In addition, the target analog CDHPM-10e revealed potency towards VEGFR-2 kinase comparable to that of sorafenib with a sub-micromolar IC50 value of 0.11 µM. Also, CDHPM-10e could effectively induce Sub-G1-phase arrest and prompt apoptosis via caspase and p53-dependent mechanisms. Furthermore, CDHPM-10e revealed significant anti-metastatic activity as detected by wound healing assay. The modelling study implies that CDHPM-10e overlaid well with sorafenib and formed a strong H-bond in the DFG binding domain. The ADMET studies hinted out that CDHPM-10e met Pfizer's drug-likeness criteria. The presented novel potent anticancer agent merits further devotion as a new lead product in developing more chalcone-based VEGFR-2 inhibitors.
{"title":"Novel NO-TZDs and trimethoxychalcone-based DHPMs: design, synthesis, and biological evaluation as potential VEGFR-2 inhibitors.","authors":"Mater H Mahnashi, Mohammed Nahari, Hassan Almasoudi, Abdulaziz Alhasaniah, Sara Elgazwi, Mahrous A Abou-Salim","doi":"10.1080/14756366.2024.2358934","DOIUrl":"10.1080/14756366.2024.2358934","url":null,"abstract":"<p><p>Novel series of nitric oxide-releasing thiazolidine-2,4-diones (<b>NO-TZD-3a-d,5,6</b>) and 3,4,5-trimethoxychalcone-based multifunctional 1,4-dihydropyrimidines (<b>CDHPM-10a-g</b>) have been designed and synthesised as potent broad-spectrum anticancer agents with potential VEGFR-2 inhibition. The designed analogs were evaluated for their anticancer activities towards a full panel of NCI-60 tumour cell lines and <b>CDHPM-10a-g</b> emerged mean %inhibitions ranging from 76.40 to 147.69%. Among them, <b>CDHPM-10e</b> and <b>CDHPM-10f</b> demonstrated the highest MGI% of 147.69 and 140.24%, respectively. Compounds <b>CDHPM-10a,b,d-f</b> showed higher mean %inhibitory activity than the reference drug sorafenib (MGI% = 105.46%). Superiorly, the hybrid <b>CDHPM-10e</b> displayed the highest potencies towards all the herein tested subpanels of nine types of cancer with MGI<sub>50</sub> of 1.83 µM. Also, it revealed potent cytostatic single-digit micromolar activity towards the herein examined cancer cell lines. The designed compounds <b>CDHPM-10a-g</b> were exposed as potent non-selective broad-spectrum anticancer agents over all NCI subpanels with an SI range of 0.66-1.97. In addition, the target analog <b>CDHPM-10e</b> revealed potency towards VEGFR-2 kinase comparable to that of sorafenib with a sub-micromolar IC<sub>50</sub> value of 0.11 µM. Also, <b>CDHPM-10e</b> could effectively induce Sub-G1-phase arrest and prompt apoptosis <i>via</i> caspase and p53-dependent mechanisms. Furthermore, <b>CDHPM-10e</b> revealed significant anti-metastatic activity as detected by wound healing assay. The modelling study implies that <b>CDHPM-10e</b> overlaid well with sorafenib and formed a strong H-bond in the DFG binding domain. The ADMET studies hinted out that <b>CDHPM-10e</b> met Pfizer's drug-likeness criteria. The presented novel potent anticancer agent merits further devotion as a new lead product in developing more chalcone-based VEGFR-2 inhibitors.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC467104/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141432072","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-01-23DOI: 10.1080/14756366.2024.2305852
Zhuoying Chen, Quan Wang, Yao Yao Yan, Dalong Jin, Yumeng Wang, Xing Xing Zhang, Xin Hua Liu
It has been reported that CDK8 plays a key role in acute myeloid leukaemia. Here, a total of 40 compounds were rational designed and synthesised based on the previous SAR. Among them, compound 12 (3-(3-(furan-3-yl)-1H-pyrrolo[2,3-b]pyridin-5-yl)benzamide) showed the most potent inhibiting activity against CDK8 with an IC50 value of 39.2 ± 6.3 nM and anti AML cell proliferation activity (molm-13 GC50 = 0.02 ± 0.01 μM, MV4-11 GC50 = 0.03 ± 0.01 μM). Mechanistic studies revealed that this compound 12 could inhibit the phosphorylation of STAT-1 and STAT-5. Importantly, compound 12 showed relative good bioavailability (F = 38.80%) and low toxicity in vivo. This study has great significance for the discovery of more efficient CDK8 inhibitors and the development of drugs for treating AML in the future.
{"title":"Discovery of novel and potent CDK8 inhibitors for the treatment of acute myeloid leukaemia.","authors":"Zhuoying Chen, Quan Wang, Yao Yao Yan, Dalong Jin, Yumeng Wang, Xing Xing Zhang, Xin Hua Liu","doi":"10.1080/14756366.2024.2305852","DOIUrl":"10.1080/14756366.2024.2305852","url":null,"abstract":"<p><p>It has been reported that CDK8 plays a key role in acute myeloid leukaemia. Here, a total of 40 compounds were rational designed and synthesised based on the previous SAR. Among them, compound <b>12</b> (<i>3-(3-(furan-3-yl)-1H-pyrrolo[2,3-b]pyridin-5-yl)benzamide</i>) showed the most potent inhibiting activity against CDK8 with an IC<sub>50</sub> value of 39.2 ± 6.3 nM and anti AML cell proliferation activity (molm-13 GC<sub>50</sub> = 0.02 ± 0.01 <i>μ</i>M, MV4-11 GC<sub>50</sub> = 0.03 ± 0.01 <i>μ</i>M). Mechanistic studies revealed that this compound <b>12</b> could inhibit the phosphorylation of STAT-1 and STAT-5. Importantly, compound <b>12</b> showed relative good bioavailability (<i>F</i> = 38.80%) and low toxicity <i>in vivo</i>. This study has great significance for the discovery of more efficient CDK8 inhibitors and the development of drugs for treating AML in the future.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10810651/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139521031","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2023-12-07DOI: 10.1080/14756366.2023.2288810
Keting Li, Wenshu Hu, Yingjie Wang, Wenxing Chen, Hongmei Wen, Jian Liu, Wei Li, Bo Wang
Disruption of p53-MDM2/MDMX interaction by smaller inhibitors is a promising therapeutic intervention gaining tremendous interest. However, no MDM2/MDMX inhibitors have been marketed so far. Drug repurposing is a validated, practical approach to drug discovery. In this regard, we employed structure-based virtual screening in a reservoir of marketed drugs and identified nintedanib as a new MDM2/MDMX dual inhibitor. The computational structure analysis and biochemical experiments uncover that nintedanib binds MDM2/MDMX similarly to RO2443, a dual MDM2/MDMX inhibitor. Furthermore, the mechanistic study reveals that nintedanib disrupts the physical interaction of p53-MDM2/MDMX, enabling the transcriptional activation of p53 and the subsequent cell cycle arrest and growth inhibition in p53+/+ cancer cells. Lastly, structural minimisation of nintedanib yields H3 with the equivalent potency. In summary, this work provides a solid foundation for reshaping nintedanib as a valuable lead compound for the further design of MDM2/MDMX dual inhibitors.
{"title":"Searching for novel MDM2/MDMX dual inhibitors through a drug repurposing approach.","authors":"Keting Li, Wenshu Hu, Yingjie Wang, Wenxing Chen, Hongmei Wen, Jian Liu, Wei Li, Bo Wang","doi":"10.1080/14756366.2023.2288810","DOIUrl":"10.1080/14756366.2023.2288810","url":null,"abstract":"<p><p>Disruption of p53-MDM2/MDMX interaction by smaller inhibitors is a promising therapeutic intervention gaining tremendous interest. However, no MDM2/MDMX inhibitors have been marketed so far. Drug repurposing is a validated, practical approach to drug discovery. In this regard, we employed structure-based virtual screening in a reservoir of marketed drugs and identified nintedanib as a new MDM2/MDMX dual inhibitor. The computational structure analysis and biochemical experiments uncover that nintedanib binds MDM2/MDMX similarly to RO2443, a dual MDM2/MDMX inhibitor. Furthermore, the mechanistic study reveals that nintedanib disrupts the physical interaction of p53-MDM2/MDMX, enabling the transcriptional activation of p53 and the subsequent cell cycle arrest and growth inhibition in p53<sup>+/+</sup> cancer cells. Lastly, structural minimisation of nintedanib yields H3 with the equivalent potency. In summary, this work provides a solid foundation for reshaping nintedanib as a valuable lead compound for the further design of MDM2/MDMX dual inhibitors.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138498513","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-01-17DOI: 10.1080/14756366.2023.2296355
Yong-Xin Luo, Xu Gong, Zhi-Cheng Su, Jin-Feng Mo, Dong-Li Li, Ri-Hui Wu, Jing-Wei Jin, Ming Lang, Jin-Ping Wang, Xue-Tao Xu, Li-She Gan
Orthosiphon aristatus is a well-known folkloric medicine and herb for Guangdong soup for the treatment of rheumatism in China. Eight isopimarane-type and migrated pimarane-type diterpenoids (1-8), including a new one with a rarely occurring α,β-unsaturated diketone C-ring, were isolated from O. aristatus. Their structures were determined by spectroscopic methods and quantum chemical calculations. Furthermore, the most abundant compound, orthosiphol K, was structurally modified by modern synthetic techniques to give seven new derivatives (9-15). The anti-rheumatoid arthritis activity of these diterpenoids were evaluated on a TNF-α induced MH7A human rheumatoid fibroblast-like synoviocyte model. Compound 10 showed the most potent activity among these compounds. Based on their inhibitory effects on the release levels of IL-1β, the preliminary structure-activity relationships were concluded. Furthermore, western blot analysis revealed that 10 could increase the expression of IκBα and decrease the expression of NF-κB p65, and the expression levels of COX-2 and NLRP3 proteins were consequently down-regulated.
{"title":"Isolation, structure modification, and anti-rheumatoid arthritis activity of isopimarane-type diterpenoids from <i>Orthosiphon aristatus</i>.","authors":"Yong-Xin Luo, Xu Gong, Zhi-Cheng Su, Jin-Feng Mo, Dong-Li Li, Ri-Hui Wu, Jing-Wei Jin, Ming Lang, Jin-Ping Wang, Xue-Tao Xu, Li-She Gan","doi":"10.1080/14756366.2023.2296355","DOIUrl":"10.1080/14756366.2023.2296355","url":null,"abstract":"<p><p><i>Orthosiphon aristatus</i> is a well-known folkloric medicine and herb for Guangdong soup for the treatment of rheumatism in China. Eight isopimarane-type and migrated pimarane-type diterpenoids (<b>1</b>-<b>8</b>), including a new one with a rarely occurring α,β-unsaturated diketone C-ring, were isolated from <i>O. aristatus</i>. Their structures were determined by spectroscopic methods and quantum chemical calculations. Furthermore, the most abundant compound, orthosiphol K, was structurally modified by modern synthetic techniques to give seven new derivatives (<b>9-15</b>). The anti-rheumatoid arthritis activity of these diterpenoids were evaluated on a TNF-α induced MH7A human rheumatoid fibroblast-like synoviocyte model. Compound <b>10</b> showed the most potent activity among these compounds. Based on their inhibitory effects on the release levels of IL-1β, the preliminary structure-activity relationships were concluded. Furthermore, western blot analysis revealed that <b>10</b> could increase the expression of IκBα and decrease the expression of NF-κB p65, and the expression levels of COX-2 and NLRP3 proteins were consequently down-regulated.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10798283/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139485391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-01-30DOI: 10.1080/14756366.2024.2309171
Mostafa M M El-Miligy, Ahmed K Al-Kubeisi, Rasha A Nassra, Saad R El-Zemity, Aly A Hazzaa
New thymol-3,4-disubstitutedthiazole hybrids were synthesised as dual COX-2/5-LOX inhibitors. Compounds 6b, 6d, 6e, and 6f displayed in vitro inhibitory activity against COX-2 (IC50= 0.037, 0.042, 0.046, and 0.039 µM) nearly equal to celecoxib (IC50= 0.045 µM). 6b, 6d, and 6f showed SI (379, 341, and 374, respectively) higher than that of celecoxib (327). 6a-l elicited in vitro 5-LOX inhibitory activity higher than quercetin. 6a-f, 6i-l, 7a, and 7c possessed in vivo inhibition of formalin induced paw edoema higher than celecoxib. 6a, 6b, 6f, 6h-l, and 7b showed gastrointestinal safety profile as celecoxib and diclofenac sodium in the population of fasted rats. Induced fit docking and molecular dynamics simulation predicted good fitting of 6b and 6f without changing the packing and globularity of the apo protein. In conclusion, 6b and 6f achieved the target goal as multitarget inhibitors of inflammation.
{"title":"Discovery of new thymol-3,4-disubstituted thiazole hybrids as dual COX-2/5-LOX inhibitors with <i>in vivo</i> proof.","authors":"Mostafa M M El-Miligy, Ahmed K Al-Kubeisi, Rasha A Nassra, Saad R El-Zemity, Aly A Hazzaa","doi":"10.1080/14756366.2024.2309171","DOIUrl":"10.1080/14756366.2024.2309171","url":null,"abstract":"<p><p>New thymol-3,4-disubstitutedthiazole hybrids were synthesised as dual COX-2/5-LOX inhibitors. Compounds <b>6b</b>, <b>6d</b>, <b>6e</b>, and <b>6f</b> displayed <i>in vitro</i> inhibitory activity against COX-2 (IC<sub>50</sub>= 0.037, 0.042, 0.046, and 0.039 µM<b>)</b> nearly equal to celecoxib (IC<sub>50</sub>= 0.045 µM<b>)</b>. <b>6b</b>, <b>6d</b>, and <b>6f</b> showed SI (379, 341, and 374, respectively) higher than that of celecoxib (327). <b>6a</b>-<b>l</b> elicited <i>in vitro</i> 5-LOX inhibitory activity higher than quercetin. <b>6a</b>-<b>f</b>, <b>6i</b>-<b>l</b>, <b>7a</b>, and <b>7c</b> possessed <i>in vivo</i> inhibition of formalin induced paw edoema higher than celecoxib. <b>6a</b>, <b>6b</b>, <b>6f</b>, <b>6h</b>-<b>l</b>, and <b>7b</b> showed gastrointestinal safety profile as celecoxib and diclofenac sodium in the population of fasted rats. Induced fit docking and molecular dynamics simulation predicted good fitting of <b>6b</b> and <b>6f</b> without changing the packing and globularity of the apo protein. In conclusion, <b>6b</b> and <b>6f</b> achieved the target goal as multitarget inhibitors of inflammation.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10833116/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139642292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2023-12-07DOI: 10.1080/14756366.2023.2287420
Shiu-Wen Huang, Ming Jen Hsu, Hsiu-Chen Chen, Rita Meleddu, Simona Distinto, Elias Maccioni, Filippo Cottiglia
The phytochemical investigation of the methanol extract of the seeds of Magydaris pastinacea afforded two undescribed benzofuran glycosides, furomagydarins A-B (1, 2), together with three known coumarins. The structures of the new isolates were elucidated after extensive 1D and 2D NMR experiments as well as HR MS. Compound 1 was able to inhibit the COX-2 expression in RAW264.7 macrophages exposed to lipopolysaccharide, a pro-inflammatory stimulus. RT-qPCR and luciferase reporter assays suggested that compound 1 reduces COX-2 expression at the transcriptional level. Further studies highlighted the capability of compound 1 to suppress the LPS-induced p38MAPK, JNK, and C/EBPβ phosphorylation, leading to COX-2 down-regulation in RAW264.7 macrophages.
{"title":"Suppression of lipopolysaccharide-induced COX-2 expression via p38MAPK, JNK, and C/EBPβ phosphorylation inhibition by furomagydarin A, a benzofuran glycoside from <i>Magydaris pastinacea</i>.","authors":"Shiu-Wen Huang, Ming Jen Hsu, Hsiu-Chen Chen, Rita Meleddu, Simona Distinto, Elias Maccioni, Filippo Cottiglia","doi":"10.1080/14756366.2023.2287420","DOIUrl":"10.1080/14756366.2023.2287420","url":null,"abstract":"<p><p>The phytochemical investigation of the methanol extract of the seeds of <i>Magydaris pastinacea</i> afforded two undescribed benzofuran glycosides, furomagydarins A-B (<b>1</b>, <b>2</b>), together with three known coumarins. The structures of the new isolates were elucidated after extensive 1D and 2D NMR experiments as well as HR MS. Compound <b>1</b> was able to inhibit the COX-2 expression in RAW264.7 macrophages exposed to lipopolysaccharide, a pro-inflammatory stimulus. RT-qPCR and luciferase reporter assays suggested that compound <b>1</b> reduces COX-2 expression at the transcriptional level. Further studies highlighted the capability of compound <b>1</b> to suppress the LPS-induced p38MAPK, JNK, and C/EBPβ phosphorylation, leading to COX-2 down-regulation in RAW264.7 macrophages.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138498514","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-05-03DOI: 10.1080/14756366.2024.2343352
Anna Szymanowska, Dominika Radomska, Robert Czarnomysy, Mariusz Mojzych, Katarzyna Kotwica-Mojzych, Krzysztof Bielawski, Anna Bielawska
In the last decade, an increasing interest in compounds containing pyrazolo[4,3-e][1,2,4]triazine moiety is observed. Therefore, the aim of the research was to synthesise a novel sulphonyl pyrazolo[4,3-e][1,2,4]triazines (2a, 2b) and pyrazolo[4,3-e]tetrazolo[1,5-b][1,2,4]triazine sulphonamide derivatives (3a, 3b) to assess their anticancer activity. The MTT assay showed that 2a, 2b, 3a, 3b have stronger cytotoxic activity than cisplatin in both breast cancer cells (MCF-7 and MDA-MB-231) and exhibited weaker effect on normal breast cells (MCF-10A). The obtained results showed that the most active compound 3b increased apoptosis via caspase 9, caspase 8, and caspase 3/7. It is worth to note that compound 3b suppressed NF-κB expression and promoted p53, Bax, and ROS which play important role in activation of apoptosis. Moreover, our results confirmed that compound 3b triggers autophagy through increased formation of autophagosomes, expression of beclin-1 and mTOR inhibition. Thus, our study defines a possible mechanism underlying 3b-induced anti-cancer activity against breast cancer cell lines.
{"title":"The activity of pyrazolo[4,3-<i>e</i>][1,2,4]triazine and pyrazolo[4,3-<i>e</i>]tetrazolo[1,5-<i>b</i>][1,2,4]triazine sulphonamide derivatives in monolayer and spheroid breast cancer cell cultures.","authors":"Anna Szymanowska, Dominika Radomska, Robert Czarnomysy, Mariusz Mojzych, Katarzyna Kotwica-Mojzych, Krzysztof Bielawski, Anna Bielawska","doi":"10.1080/14756366.2024.2343352","DOIUrl":"10.1080/14756366.2024.2343352","url":null,"abstract":"<p><p>In the last decade, an increasing interest in compounds containing pyrazolo[4,3-<i>e</i>][1,2,4]triazine moiety is observed. Therefore, the aim of the research was to synthesise a novel sulphonyl pyrazolo[4,3-<i>e</i>][1,2,4]triazines (<b>2a</b>, <b>2b</b>) and pyrazolo[4,3-<i>e</i>]tetrazolo[1,5-<i>b</i>][1,2,4]triazine sulphonamide derivatives (<b>3a</b>, <b>3b</b>) to assess their anticancer activity. The MTT assay showed that <b>2a</b>, <b>2b</b>, <b>3a</b>, <b>3b</b> have stronger cytotoxic activity than cisplatin in both breast cancer cells (MCF-7 and MDA-MB-231) and exhibited weaker effect on normal breast cells (MCF-10A). The obtained results showed that the most active compound <b>3b</b> increased apoptosis via caspase 9, caspase 8, and caspase 3/7. It is worth to note that compound <b>3b</b> suppressed NF-κB expression and promoted p53, Bax, and ROS which play important role in activation of apoptosis. Moreover, our results confirmed that compound <b>3b</b> triggers autophagy through increased formation of autophagosomes, expression of beclin-1 and mTOR inhibition. Thus, our study defines a possible mechanism underlying <b>3b</b>-induced anti-cancer activity against breast cancer cell lines.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11073428/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140862942","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Acyl-CoA: cholesterol acyltransferase (ACAT), a pivotal enzyme in the absorption and metabolism of cholesterol, is primarily responsible for intracellular esterification. ACAT inhibition is expected to diminish plasma lipid levels by impeding intestinal cholesterol absorption, thereby preventing the progression of atherosclerotic lesions. A previous study shows that selective inhibition of ACAT2 significantly mitigated hypercholesterolaemia and atherosclerosis in mouse models. Therefore, the need for ACAT2 selective inhibitors becomes particularly urgent. In this study, we established a multilayer virtual screening workflow and subjected biologically evaluated representative compounds to enzyme inhibitory assays. The experimental results indicated that the two compounds, STL565001 (inhibition rate at 25 μM: 75.7 ± 27.8%, selectivity = 6) and STL528213 (inhibition rate at 25 μM: 87.8 ± 12.4%, selectivity = 13), demonstrated robust activity against ACAT2, displaying greater selectivity for ACAT2 than for ACAT1. The molecular mechanisms governing the inhibitory activities of the selected compounds were systematically elucidated using computational approaches. In addition, hotspot residues in ACAT2 that are crucial for ligand binding were successfully identified. In summary, we devised a multilayer screening scheme to expeditiously and efficiently identify compounds with enzyme inhibitory activity, offering novel scaffolds for subsequent drug design centred on ACAT2 targets.
{"title":"Discovery of selective ACAT2 antagonist via a combination strategy based on deep docking, pharmacophore modelling, and molecular dynamics simulation.","authors":"Yanfeng Liu, Feng Ding, Liangying Deng, Shuran Zhang, Lixing Wu, Huangjin Tong","doi":"10.1080/14756366.2024.2403736","DOIUrl":"https://doi.org/10.1080/14756366.2024.2403736","url":null,"abstract":"<p><p>Acyl-CoA: cholesterol acyltransferase (ACAT), a pivotal enzyme in the absorption and metabolism of cholesterol, is primarily responsible for intracellular esterification. ACAT inhibition is expected to diminish plasma lipid levels by impeding intestinal cholesterol absorption, thereby preventing the progression of atherosclerotic lesions. A previous study shows that selective inhibition of ACAT2 significantly mitigated hypercholesterolaemia and atherosclerosis in mouse models. Therefore, the need for ACAT2 selective inhibitors becomes particularly urgent. In this study, we established a multilayer virtual screening workflow and subjected biologically evaluated representative compounds to enzyme inhibitory assays. The experimental results indicated that the two compounds, STL565001 (inhibition rate at 25 μM: 75.7 ± 27.8%, selectivity = 6) and STL528213 (inhibition rate at 25 μM: 87.8 ± 12.4%, selectivity = 13), demonstrated robust activity against ACAT2, displaying greater selectivity for ACAT2 than for ACAT1. The molecular mechanisms governing the inhibitory activities of the selected compounds were systematically elucidated using computational approaches. In addition, hotspot residues in ACAT2 that are crucial for ligand binding were successfully identified. In summary, we devised a multilayer screening scheme to expeditiously and efficiently identify compounds with enzyme inhibitory activity, offering novel scaffolds for subsequent drug design centred on ACAT2 targets.</p>","PeriodicalId":15769,"journal":{"name":"Journal of Enzyme Inhibition and Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11423527/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142347992","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}