首页 > 最新文献

In silico pharmacology最新文献

英文 中文
Anthelmintic activity of Calotropis gigantea: in silico investigation on novel target site nematode kinases. 石菖蒲的驱虫活性:线虫激酶新靶点的硅学研究。
Pub Date : 2025-04-04 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00331-7
Velan Gopalakrishnan, Thiruvenkadam Mahendiran

Over one billion peoples are currently infected with a parasitic nematode, particularly in tropical regions. Helminthiasis is a medical condition characterized by an infection of parasitic worms in the human body. Calotropis gigantea Linn. Belongs to the Asclepiadaceae family, commonly known as "Gaint milkweed" or "Crown flower" and recognized as a traditional medicinal plant. In this study, anthelminthic potential of Calotropis gigantea phytoconstituents were determined. Nematode kinases EGFR, MEK1, and PLK1 have diverged from their vertebrate counterparts in their drug-binding pockets. The development of novel anthelmintics targets Kinases that hold potential as targets. This pipeline revealed three potential anthelmintic targets that include epidermal growth factor tyrosine kinase transmembrane receptor [EGFR], the Mitogen-activate protein kinase [MEK1], and polo-like kinase [PLK1]. The software used in the in silico study investigation is the pyRx virtual screening tool, Biovia Discovery Studio, mol-inspiration, and RCSB [Protein Data Bank (PDB)]. The various phytoconstituents of Calotropis gigantea were docked Stigmasterol, beta-sitosterol, Desmosterol, alpha-amyrin, asclepin, and others showed high binding energies in EGFR, MEK1, and PLK1 receptors. This investigational study research highlights the potential of phytoconstituents from Calotropis gigantea as anthelmintic activity.

Graphic abstract:

{"title":"Anthelmintic activity of <i>Calotropis gigantea</i>: in silico investigation on novel target site nematode kinases.","authors":"Velan Gopalakrishnan, Thiruvenkadam Mahendiran","doi":"10.1007/s40203-025-00331-7","DOIUrl":"10.1007/s40203-025-00331-7","url":null,"abstract":"<p><p>Over one billion peoples are currently infected with a parasitic nematode, particularly in tropical regions. Helminthiasis is a medical condition characterized by an infection of parasitic worms in the human body. <i>Calotropis gigantea</i> Linn. Belongs to the Asclepiadaceae family, commonly known as \"Gaint milkweed\" or \"Crown flower\" and recognized as a traditional medicinal plant. In this study, anthelminthic potential of <i>Calotropis gigantea</i> phytoconstituents were determined. Nematode kinases EGFR, MEK1, and PLK1 have diverged from their vertebrate counterparts in their drug-binding pockets. The development of novel anthelmintics targets Kinases that hold potential as targets. This pipeline revealed three potential anthelmintic targets that include epidermal growth factor tyrosine kinase transmembrane receptor [EGFR], the Mitogen-activate protein kinase [MEK1], and polo-like kinase [PLK1]. The software used in the in silico study investigation is the pyRx virtual screening tool, Biovia Discovery Studio, mol-inspiration, and RCSB [Protein Data Bank (PDB)]. The various phytoconstituents of <i>Calotropis gigantea</i> were docked Stigmasterol, beta-sitosterol, Desmosterol, alpha-amyrin, asclepin, and others showed high binding energies in EGFR, MEK1, and PLK1 receptors. This investigational study research highlights the potential of phytoconstituents from <i>Calotropis gigantea</i> as anthelmintic activity.</p><p><strong>Graphic abstract: </strong></p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"54"},"PeriodicalIF":0.0,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968633/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143797512","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Alpha-bisabolol inhibits yeast to hyphal form transition and biofilm development in Candida albicans: in vitro and in silico studies.
Pub Date : 2025-04-01 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00335-3
Sayali Chougule, Shivani Patil, Tanjila Gavandi, Sargun Basrani, Ashwini K Jadhav, S Mohan Karuppayil

In recent years, there has been growing concern about infections caused by Candida albicans, which pose a significant threat to human health. This intensifies the concern that can be largely attributed to the increasing number of people with compromised immune systems and the emergence of drug-resistant strains. Natural molecules are considered to be good alternatives to synthetic antifungal agents. The present study explored the effectiveness of alpha-bisabolol as an antifungal agent and its mechanism of action against C. albicans ATCC90028. α-bisabolol effectively inhibited various pathogenic traits of C. albicans like, adhesion, yeast to hyphal switching, and development of biofilm at 1 mg/ml, 0.25 mg/ml, and 0.125 mg/ml concentration, respectively. In addition, α-bisabolol demonstrated inhibition of cell cycle propagation at the G1 phase. Ergosterol production in the C. albicans was suppressed by α-bisabolol treatment in a dose-dependent manner. The molecular docking study revealed α-bisabolol has a good binding energy of - 7.11 kcal/mol with 14-α-demethylase enzyme, which is crucial for ergosterol synthesis. Therefore, the cell membrane integrity may be affected by treatment with α-bisabolol. qRT-PCR studies proved that α-bisabolol treatment affects gene expression in C. albicans. In silico binding affinity was also analyzed for RAS1, TUP1 and CST20 in the signal transduction pathway and exhibited binding affinities for at - 7.7 kcal/mol, - 8.21 kcal/mol, and for - 5.79 kcal/mol respectively. In conclusion, α-bisabolol caused reduced biofilm, ergosterol synthesis along with altered gene expressions in C. albicans with no hemolysis. This study proposed α-bisabolol as an alternative antifungal agent.

{"title":"Alpha-bisabolol inhibits yeast to hyphal form transition and biofilm development in <i>Candida albicans</i>: in vitro and in silico studies.","authors":"Sayali Chougule, Shivani Patil, Tanjila Gavandi, Sargun Basrani, Ashwini K Jadhav, S Mohan Karuppayil","doi":"10.1007/s40203-025-00335-3","DOIUrl":"10.1007/s40203-025-00335-3","url":null,"abstract":"<p><p>In recent years, there has been growing concern about infections caused by <i>Candida albicans</i>, which pose a significant threat to human health. This intensifies the concern that can be largely attributed to the increasing number of people with compromised immune systems and the emergence of drug-resistant strains. Natural molecules are considered to be good alternatives to synthetic antifungal agents. The present study explored the effectiveness of alpha-bisabolol as an antifungal agent and its mechanism of action against <i>C. albicans</i> ATCC90028. α-bisabolol effectively inhibited various pathogenic traits of <i>C. albicans</i> like, adhesion, yeast to hyphal switching, and development of biofilm at 1 mg/ml, 0.25 mg/ml, and 0.125 mg/ml concentration, respectively. In addition, α-bisabolol demonstrated inhibition of cell cycle propagation at the G1 phase. Ergosterol production in the <i>C. albicans</i> was suppressed by α-bisabolol treatment in a dose-dependent manner. The molecular docking study revealed α-bisabolol has a good binding energy of - 7.11 kcal/mol with 14-α-demethylase enzyme, which is crucial for ergosterol synthesis. Therefore, the cell membrane integrity may be affected by treatment with α-bisabolol. qRT-PCR studies proved that α-bisabolol treatment affects gene expression in <i>C. albicans</i>. In silico binding affinity was also analyzed for <i>RAS1</i>, <i>TUP1</i> and <i>CST20</i> in the signal transduction pathway and exhibited binding affinities for at - 7.7 kcal/mol, - 8.21 kcal/mol, and for - 5.79 kcal/mol respectively. In conclusion, α-bisabolol caused reduced biofilm, ergosterol synthesis along with altered gene expressions in <i>C. albicans</i> with no hemolysis. This study proposed α-bisabolol as an alternative antifungal agent.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"53"},"PeriodicalIF":0.0,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11961840/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143782365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular dynamics and experimental evaluation of piperine as a potential mTOR inhibitor in colon cancer cells.
Pub Date : 2025-03-28 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00339-z
Ifat Jan, Tabasum Ali, Rafat Ali, Nida Jamil Khan, Khurshid Iqbal Andrabi, Ghulam Nabi Bader

Piperine, a natural alkaloid found in black pepper (Piper nigrum), has the chemical formula C₁₇H₁₉NO₃ and a molecular weight of 285.34 g/mol. This research investigated its effect on the mTOR protein, which plays a crucial role in cancer development, using molecular docking, dynamic simulations, MTT, and scratch wound assays on the HCT-116 colon cancer cell line. Molecular docking revealed that piperine exhibited a binding affinity of - 8.3 kcal/mol to the mTOR protein, which is significantly comparable to rapamycin's binding affinity of - 8.8 kcal/mol, a well-known mTOR inhibitor. This comparison highlights that piperine demonstrates a substantial ability to interact with the mTOR binding site, making it a potential candidate for further evaluation. Molecular dynamics simulation studies over 100 ns confirmed that piperine remains stable and firmly bound to the mTOR active site, binding in an ATP-competitive mode. MTT assay results revealed that piperine significantly reduced cancer cell viability, with IC50 values of 84.5 ± 0.5 µM at 24 h, 46.3 ± 0.26 µM at 48 h, and 19.73 ± 0.25 µM at 72 h, while the scratch wound assay confirmed its inhibition of cancer cell migration, suggesting potential to suppress metastasis. These findings indicate that piperine is a promising mTOR inhibitor with potential applications in cancer therapy, though further research is needed.

{"title":"Molecular dynamics and experimental evaluation of piperine as a potential mTOR inhibitor in colon cancer cells.","authors":"Ifat Jan, Tabasum Ali, Rafat Ali, Nida Jamil Khan, Khurshid Iqbal Andrabi, Ghulam Nabi Bader","doi":"10.1007/s40203-025-00339-z","DOIUrl":"10.1007/s40203-025-00339-z","url":null,"abstract":"<p><p>Piperine, a natural alkaloid found in black pepper (<i>Piper nigrum</i>), has the chemical formula C₁₇H₁₉NO₃ and a molecular weight of 285.34 g/mol. This research investigated its effect on the mTOR protein, which plays a crucial role in cancer development, using molecular docking, dynamic simulations, MTT, and scratch wound assays on the HCT-116 colon cancer cell line. Molecular docking revealed that piperine exhibited a binding affinity of - 8.3 kcal/mol to the mTOR protein, which is significantly comparable to rapamycin's binding affinity of - 8.8 kcal/mol, a well-known mTOR inhibitor. This comparison highlights that piperine demonstrates a substantial ability to interact with the mTOR binding site, making it a potential candidate for further evaluation. Molecular dynamics simulation studies over 100 ns confirmed that piperine remains stable and firmly bound to the mTOR active site, binding in an ATP-competitive mode. MTT assay results revealed that piperine significantly reduced cancer cell viability, with IC50 values of 84.5 ± 0.5 µM at 24 h, 46.3 ± 0.26 µM at 48 h, and 19.73 ± 0.25 µM at 72 h, while the scratch wound assay confirmed its inhibition of cancer cell migration, suggesting potential to suppress metastasis. These findings indicate that piperine is a promising mTOR inhibitor with potential applications in cancer therapy, though further research is needed.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"52"},"PeriodicalIF":0.0,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11953494/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143756789","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of phyto-compounds from Mangifera indica as inhibitors of 17β-hydroxysteroid dehydrogenase: a computational approach against prostate cancer.
Pub Date : 2025-03-28 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00332-6
Adedotun Olayemi Oluwatuyi, Olusola Olalekan Elekofehinti, Hannah Oluwaseun Popoola, Moses Orimoloye Akinjiyan, Idayat Oyinkansola Kehinde, Ifeoluwa Racheal Adetoyi, Olufemi Adebisi Akinola, Folasade O Ayodeji, Olabimpe Omolola Apeji, Adeola Victor Kolawole, Akinola Oluwadamilola Dorcas, Alonge Sunday Ayodeji

Prostate cancer (PrCa) is a serious health concern for the affected people and, there is an increasing demand for a viable therapy that can address the limitations of current treatments with minimal or no adverse effects. This study aims to evaluate phytocompounds extracts of Mangifera indica as a potential therapy development for prostate cancer. Herein, molecular docking, QSAR, molecular mechanics/generalized born surface area (MM/GBSA) estimation, ADME screening, and molecular dynamics (MD) simulation were performed using the Schrodinger suite to identify 17β-hydroxysteroid dehydrogenase antagonist from Mangifera indica. The results showed that fisetin (-11.669), riboflavin (-10.918), quercetin (-10.843), gallic acid 6-phenylhexyl ester (-10.817), cianidanol (-10.608), (-)-epicatechin (-10.603), ellagic acid (-10.522), Butin (-10.124) in kcal/mol were predicted to possess greater inhibitory activities against the protein target based on their high binding energies and remarkable stability compared to the standard drug, docetaxel (-7.374 kcal/mol). Fisetin (-718.37), and riboflavin (-722.37) also have better induce fit score than docetaxel (-714.02) in kcal/mol with better pharmacokinetics profile compared to the standard drug.MD simulation over 100 ns predicts that Fisetin forms stable interactions with vital residues at the catalytic site of the protein. The observations from this study predict fisetin as a putative antagonist of 17β-hydroxysteroid dehydrogenase and should be experimentally verified as a lead compound for prostate cancer therapy.

{"title":"Identification of phyto-compounds from <i>Mangifera indica</i> as inhibitors of 17β-hydroxysteroid dehydrogenase: a computational approach against prostate cancer.","authors":"Adedotun Olayemi Oluwatuyi, Olusola Olalekan Elekofehinti, Hannah Oluwaseun Popoola, Moses Orimoloye Akinjiyan, Idayat Oyinkansola Kehinde, Ifeoluwa Racheal Adetoyi, Olufemi Adebisi Akinola, Folasade O Ayodeji, Olabimpe Omolola Apeji, Adeola Victor Kolawole, Akinola Oluwadamilola Dorcas, Alonge Sunday Ayodeji","doi":"10.1007/s40203-025-00332-6","DOIUrl":"10.1007/s40203-025-00332-6","url":null,"abstract":"<p><p>Prostate cancer (PrCa) is a serious health concern for the affected people and, there is an increasing demand for a viable therapy that can address the limitations of current treatments with minimal or no adverse effects. This study aims to evaluate phytocompounds extracts of <i>Mangifera indica</i> as a potential therapy development for prostate cancer. Herein, molecular docking, QSAR, molecular mechanics/generalized born surface area (MM/GBSA) estimation, ADME screening, and molecular dynamics (MD) simulation were performed using the Schrodinger suite to identify 17β-hydroxysteroid dehydrogenase antagonist from <i>Mangifera indica</i>. The results showed that fisetin (-11.669), riboflavin (-10.918), quercetin (-10.843), gallic acid 6-phenylhexyl ester (-10.817), cianidanol (-10.608), (-)-epicatechin (-10.603), ellagic acid (-10.522), Butin (-10.124) in kcal/mol were predicted to possess greater inhibitory activities against the protein target based on their high binding energies and remarkable stability compared to the standard drug, docetaxel (-7.374 kcal/mol). Fisetin (-718.37), and riboflavin (-722.37) also have better induce fit score than docetaxel (-714.02) in kcal/mol with better pharmacokinetics profile compared to the standard drug.MD simulation over 100 ns predicts that Fisetin forms stable interactions with vital residues at the catalytic site of the protein. The observations from this study predict fisetin as a putative antagonist of 17β-hydroxysteroid dehydrogenase and should be experimentally verified as a lead compound for prostate cancer therapy.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"50"},"PeriodicalIF":0.0,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11953514/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143756785","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Computational investigation of antiviral peptide interactions with Mpox DNA polymerase.
Pub Date : 2025-03-28 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00342-4
Harshit Tiwari, Ashal Ilyas, Pankaj Kumar Rai, Shashank Upadhyay, Subhomoi Borkotoky

The Mpox DNA polymerase (DNA pol) plays a crucial role in the viral replication process, making it an ideal target for antiviral therapies. It facilitates the synthetic process of viral DNA, which is an integral stage in the life of a virus. The inhibition of the operation of Mpox DNA pol would interfere with the multiplication of the virus and help manage the disease. Peptides have emerged as a possible therapeutic alternative against viruses due to their distinct characteristics. Peptides have broad-spectrum antiviral activity, being effective against a variety of viruses. Using computational techniques, we attempted to explore the molecular details of the interaction between antiviral peptides and Mpox DNA pol. Two databases of antiviral peptides were screened in this study. This study used molecular docking, followed by molecular dynamics (MD) simulation and post-simulation binding energy predictions. From the 19 selected peptides with activity against DNA polymerases, two peptides-DRAVPe01393 and DRAVPe01399-were identified as particularly promising candidates. These peptides exhibited stable interactions with Mpox DNA pol and demonstrated good cell penetration potential as evident from the MD simulation studies. Notably, the peptides DRAVPe01399 and DRAVPe01393 have a better binding affinity of - 60.86 kcal/mol and - 47.92 kcal/mol respectively than the control ligand Cidofovir diphosphate (- 10.79 kcal/mol). These findings could lead to the development of innovative antiviral treatments to prevent monkeypox, helping global efforts to battle this emerging infectious disease.

{"title":"Computational investigation of antiviral peptide interactions with Mpox DNA polymerase.","authors":"Harshit Tiwari, Ashal Ilyas, Pankaj Kumar Rai, Shashank Upadhyay, Subhomoi Borkotoky","doi":"10.1007/s40203-025-00342-4","DOIUrl":"10.1007/s40203-025-00342-4","url":null,"abstract":"<p><p>The Mpox DNA polymerase (DNA pol) plays a crucial role in the viral replication process, making it an ideal target for antiviral therapies. It facilitates the synthetic process of viral DNA, which is an integral stage in the life of a virus. The inhibition of the operation of Mpox DNA pol would interfere with the multiplication of the virus and help manage the disease. Peptides have emerged as a possible therapeutic alternative against viruses due to their distinct characteristics. Peptides have broad-spectrum antiviral activity, being effective against a variety of viruses. Using computational techniques, we attempted to explore the molecular details of the interaction between antiviral peptides and Mpox DNA pol. Two databases of antiviral peptides were screened in this study. This study used molecular docking, followed by molecular dynamics (MD) simulation and post-simulation binding energy predictions. From the 19 selected peptides with activity against DNA polymerases, two peptides-DRAVPe01393 and DRAVPe01399-were identified as particularly promising candidates. These peptides exhibited stable interactions with Mpox DNA pol and demonstrated good cell penetration potential as evident from the MD simulation studies. Notably, the peptides DRAVPe01399 and DRAVPe01393 have a better binding affinity of - 60.86 kcal/mol and - 47.92 kcal/mol respectively than the control ligand Cidofovir diphosphate (- 10.79 kcal/mol). These findings could lead to the development of innovative antiviral treatments to prevent monkeypox, helping global efforts to battle this emerging infectious disease.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"49"},"PeriodicalIF":0.0,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11953516/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143756579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Harnessing computational methods for uncovering structural insights into Leishmania donovani 3-MST: implications for drug design and target specificity.
Pub Date : 2025-03-28 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00340-6
Ravi Ranjan, Rashmi Kumari, Ashish Kumar, Saravanan Vijayakumar

3-Mercaptopyruvate sulfurtransferase (3-MST) is an enzyme that plays integral roles in various biological processes. In the realm of Leishmania, the role of 3-MST is less explored. It is a critical player in maintaining oxidative homeostasis in Leishmania during stress for survival. This highlights the potential of Ld3-MST as an appealing drug target. However, recognising structural disparities becomes essential when a protein is present in the host and parasite. This study delves into the structural distinctions between Ld3-MST and Hs3-MST, providing valuable insights with direct implications for drug design. A standout feature of Ld3-MST is the elongated 70 amino acid C-terminal mainly contributing to a lid-like domain above the active site cavity, setting it apart from Hs3-MST. The RMSD analysis shows fluctuation due to the extended tail, while Rg and SASA confirm the open and solvent-accessible nature of Ld3-MST, especially in its active site, suggesting its ability to accommodate larger molecules. PC and FEL analysis reveals unique internal molecular dynamics of Ld3-MST, particularly in its active site. Docking studies demonstrate that Ld3-MST's active site can effectively accommodate molecules, highlighting its potential as a drug target. This comprehensive investigation lays the foundation for developing precise Ld3-MST inhibitors with promising therapeutic applications.

Graphical abstract:

Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00340-6.

{"title":"Harnessing computational methods for uncovering structural insights into <i>Leishmania donovani</i> 3-MST: implications for drug design and target specificity.","authors":"Ravi Ranjan, Rashmi Kumari, Ashish Kumar, Saravanan Vijayakumar","doi":"10.1007/s40203-025-00340-6","DOIUrl":"10.1007/s40203-025-00340-6","url":null,"abstract":"<p><p>3-Mercaptopyruvate sulfurtransferase (3-MST) is an enzyme that plays integral roles in various biological processes. In the realm of <i>Leishmania</i>, the role of 3-MST is less explored. It is a critical player in maintaining oxidative homeostasis in <i>Leishmania</i> during stress for survival. This highlights the potential of <i>Ld</i>3-MST as an appealing drug target. However, recognising structural disparities becomes essential when a protein is present in the host and parasite. This study delves into the structural distinctions between <i>Ld</i>3-MST and <i>Hs</i>3-MST, providing valuable insights with direct implications for drug design. A standout feature of <i>Ld</i>3-MST is the elongated 70 amino acid C-terminal mainly contributing to a lid-like domain above the active site cavity, setting it apart from <i>Hs</i>3-MST. The RMSD analysis shows fluctuation due to the extended tail, while Rg and SASA confirm the open and solvent-accessible nature of Ld3-MST, especially in its active site, suggesting its ability to accommodate larger molecules. PC and FEL analysis reveals unique internal molecular dynamics of Ld3-MST, particularly in its active site. Docking studies demonstrate that <i>Ld</i>3-MST's active site can effectively accommodate molecules, highlighting its potential as a drug target. This comprehensive investigation lays the foundation for developing precise <i>Ld</i>3-MST inhibitors with promising therapeutic applications.</p><p><strong>Graphical abstract: </strong></p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00340-6.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"51"},"PeriodicalIF":0.0,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11953493/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143756706","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unraveling the mechanisms of antitumor action of Sophora flavescens flavonoids via network pharmacology and molecular simulation.
Pub Date : 2025-03-20 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00338-0
Zhongyuan Guo, Gaoyue Dong, Xiaoqian Liu, Liangmian Chen, Hong Yang, Zhimin Wang
<p><p>Cancer, particularly lung, liver, and other malignancies, remains a major global health challenge due to their high incidence, complex etiology, and resistance to conventional therapies. Flavonoids derived from <i>Sophora flavescens</i> (Kushen) have gained attention for their potential in cancer prevention and treatment. This study uses network pharmacology, based on Traditional Chinese Medicine's holistic approach, and molecular simulation techniques to explore the anticancer mechanisms of <i>Sophora flavescens</i> flavonoids, aiming to provide a theoretical basis for developing plant-based anticancer agents. Active compounds and their targets were identified through literature screening and target identification methods. A cancer-related protein-protein interaction (PPI) network was constructed to identify key therapeutic targets, helping to understand how these flavonoids exert multitarget anticancer effects. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses elucidated mechanisms related to cancer cell proliferation inhibition, apoptosis induction, and signaling pathway regulation. Network pharmacology analysis identified TP53, ESR1, SRC, AKT1, and MAPK1 as key anticancer targets, involved in essential biological processes like phosphorylation and protein kinase activity. KEGG analysis showed that these flavonoids modulate critical pathways, particularly PI3K-Akt and RAS/RAF/MEK/ERK. Molecular docking revealed that rutin and luteolin-7-O-gentiobioside strongly bind to MAPK1, with interaction energies of 77.1466 kcal/mol and 79.2011 kcal/mol, respectively, indicating promising anticancer effects. Additionally, compounds with different substitution positions, such as those with glycosylation at the 7-OH position or isoprenyl groups at the C-8 positions, exhibited significantly higher interaction energies. Non-covalent interaction analysis further clarified how these flavonoids enhance anticancer effects through stable binding, with hydrogen bonds and hydrophobic contacts stabilizing interactions with MAPK1. Molecular dynamics simulations also confirmed the stability of these interactions, and binding free energy calculations revealed that luteolin-7-O-gentiobioside and rutin exhibited the lowest binding free energies (- 153.7841 kcal/mol and - 132.7434 kcal/mol, respectively), significantly outperforming the original ligand (- 57.7209 kcal/mol), further supporting the therapeutic potential of these compounds. To complement these findings, in silico ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) predictions were employed to evaluate the pharmacokinetic and safety profiles of rutin and luteolin-7-O-gentiobioside. The systematic analysis revealed critical issues in solubility, blood-brain barrier penetration, and hepatotoxicity, providing strategic guidance for structural optimization and formulation design. This study provides valuable insights into the multitarget anticancer me
{"title":"Unraveling the mechanisms of antitumor action of <i>Sophora flavescens</i> flavonoids via network pharmacology and molecular simulation.","authors":"Zhongyuan Guo, Gaoyue Dong, Xiaoqian Liu, Liangmian Chen, Hong Yang, Zhimin Wang","doi":"10.1007/s40203-025-00338-0","DOIUrl":"10.1007/s40203-025-00338-0","url":null,"abstract":"&lt;p&gt;&lt;p&gt;Cancer, particularly lung, liver, and other malignancies, remains a major global health challenge due to their high incidence, complex etiology, and resistance to conventional therapies. Flavonoids derived from &lt;i&gt;Sophora flavescens&lt;/i&gt; (Kushen) have gained attention for their potential in cancer prevention and treatment. This study uses network pharmacology, based on Traditional Chinese Medicine's holistic approach, and molecular simulation techniques to explore the anticancer mechanisms of &lt;i&gt;Sophora flavescens&lt;/i&gt; flavonoids, aiming to provide a theoretical basis for developing plant-based anticancer agents. Active compounds and their targets were identified through literature screening and target identification methods. A cancer-related protein-protein interaction (PPI) network was constructed to identify key therapeutic targets, helping to understand how these flavonoids exert multitarget anticancer effects. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses elucidated mechanisms related to cancer cell proliferation inhibition, apoptosis induction, and signaling pathway regulation. Network pharmacology analysis identified TP53, ESR1, SRC, AKT1, and MAPK1 as key anticancer targets, involved in essential biological processes like phosphorylation and protein kinase activity. KEGG analysis showed that these flavonoids modulate critical pathways, particularly PI3K-Akt and RAS/RAF/MEK/ERK. Molecular docking revealed that rutin and luteolin-7-O-gentiobioside strongly bind to MAPK1, with interaction energies of 77.1466 kcal/mol and 79.2011 kcal/mol, respectively, indicating promising anticancer effects. Additionally, compounds with different substitution positions, such as those with glycosylation at the 7-OH position or isoprenyl groups at the C-8 positions, exhibited significantly higher interaction energies. Non-covalent interaction analysis further clarified how these flavonoids enhance anticancer effects through stable binding, with hydrogen bonds and hydrophobic contacts stabilizing interactions with MAPK1. Molecular dynamics simulations also confirmed the stability of these interactions, and binding free energy calculations revealed that luteolin-7-O-gentiobioside and rutin exhibited the lowest binding free energies (- 153.7841 kcal/mol and - 132.7434 kcal/mol, respectively), significantly outperforming the original ligand (- 57.7209 kcal/mol), further supporting the therapeutic potential of these compounds. To complement these findings, in silico ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) predictions were employed to evaluate the pharmacokinetic and safety profiles of rutin and luteolin-7-O-gentiobioside. The systematic analysis revealed critical issues in solubility, blood-brain barrier penetration, and hepatotoxicity, providing strategic guidance for structural optimization and formulation design. This study provides valuable insights into the multitarget anticancer me","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"48"},"PeriodicalIF":0.0,"publicationDate":"2025-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11926300/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143694962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting cardiotoxicity: the potential of Annona squamosa L. in doxorubicin therapy.
Pub Date : 2025-03-17 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00333-5
Kshitij A Lele, Priyanka P Patil, Sneha V Kakade, Naveen R Maledavar, Shriram D Ranade, Shankar G Alegaon, Prakash R Biradar, Nayeem A Khatib

Doxorubicin, a potent anthracycline used in chemotherapy, is limited by dose-dependent cardiotoxicity, leading to irreversible heart damage and heart failure. Common symptoms include fatigue, dyspnea, lower limb edema, hypotension, tachycardia, and transient arrhythmias. Annona squamosa L. (AS), traditionally used in medicine, was investigated for its cardioprotective action against doxorubicin-induced cardiotoxicity through computational studies. Phytocompounds were identified using literature reviews, Dr. Duke's, IMPPAT, and PubChem databases. Targets associated with Doxorubicin induced cardiotoxicity were accessed from GeneCards, and protein-protein interactions were analyzed using the STRING database. Cytoscape was used for network visualization, revealing 18 bioactives targeting 67 proteins across 14 pathways. PIK3R1 emerged as a key target with the highest interaction count among 767 targets. Molecular docking showed that the PIK3R1-Rutin complex had the lowest binding energy (- 11.873 kcal/mol), and a 100 ns molecular dynamics (MD) simulation confirmed its stability. LC-MS analysis of the crude extract indicated the presence of bioactives. In vitro antioxidant activity of AS, assessed using the DPPH assay, showed significant radical scavenging activity, correlating with the high total phenol (TPC) and total flavonoid content (TFC) detected. This integrated approach highlights AS's potential in mitigating doxorubicin-induced cardiotoxicity.

Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00333-5.

{"title":"Targeting cardiotoxicity: the potential of <i>Annona squamosa</i> L. in doxorubicin therapy.","authors":"Kshitij A Lele, Priyanka P Patil, Sneha V Kakade, Naveen R Maledavar, Shriram D Ranade, Shankar G Alegaon, Prakash R Biradar, Nayeem A Khatib","doi":"10.1007/s40203-025-00333-5","DOIUrl":"10.1007/s40203-025-00333-5","url":null,"abstract":"<p><p>Doxorubicin, a potent anthracycline used in chemotherapy, is limited by dose-dependent cardiotoxicity, leading to irreversible heart damage and heart failure. Common symptoms include fatigue, dyspnea, lower limb edema, hypotension, tachycardia, and transient arrhythmias. <i>Annona squamosa</i> L. (AS), traditionally used in medicine, was investigated for its cardioprotective action against doxorubicin-induced cardiotoxicity through computational studies. Phytocompounds were identified using literature reviews, Dr. Duke's, IMPPAT, and PubChem databases. Targets associated with Doxorubicin induced cardiotoxicity were accessed from GeneCards, and protein-protein interactions were analyzed using the STRING database. Cytoscape was used for network visualization, revealing 18 bioactives targeting 67 proteins across 14 pathways. PIK3R1 emerged as a key target with the highest interaction count among 767 targets. Molecular docking showed that the PIK3R1-Rutin complex had the lowest binding energy (- 11.873 kcal/mol), and a 100 ns molecular dynamics (MD) simulation confirmed its stability. LC-MS analysis of the crude extract indicated the presence of bioactives. In vitro antioxidant activity of AS, assessed using the DPPH assay, showed significant radical scavenging activity, correlating with the high total phenol (TPC) and total flavonoid content (TFC) detected. This integrated approach highlights AS's potential in mitigating doxorubicin-induced cardiotoxicity.</p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00333-5.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"47"},"PeriodicalIF":0.0,"publicationDate":"2025-03-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11914696/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143665780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Natural compounds as therapeutic candidates for spinocerebellar ataxia type 1: a computational approach.
Pub Date : 2025-03-15 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00308-6
Surbhi Singh, Suchitra Singh, Deepika Joshi, C Mohanty, Royana Singh

Spinocerebellar Ataxia Type 1 (SCA1) is a progressive neurodegenerative disorder caused by the expansion and aggregation of polyglutamine (polyQ) in the Ataxin-1 (ATXN1) protein, leading to severe neuronal dysfunction. Currently, only symptomatic treatments are available, highlighting the requirement for disease-modifying therapies. This study employed a detailed in silico approach to identify potential neuroprotective natural compounds targeting the Ataxin-1 protein implicated in SCA1. The three-dimensional structure of Ataxin-1 was retrieved, validated, and optimized to achieve a stable structural model. Validation using a Ramachandran plot indicated that 77% of the residues were in favored regions, confirming the reliability of the protein structure. Active site residues were identified using CASTp, and receptor grids were generated for molecular docking studies. A library of 50 natural compounds was screened, among which 21 satisfied Lipinski's rule of five. Molecular docking using PyRx and AutoDock 4.2 identified Withanolide A as the top candidate, exhibiting the highest binding affinity (- 10.14 kcal/mol) and forming four hydrogen bonds with key active site residues. The top six ligands were further assessed for ADMET properties, with Withanolide A showing optimal drug-likeness, high gastrointestinal and blood-brain absorption, and non-toxic profiles. Molecular dynamics simulations over 200 ns demonstrated the stability of the Ataxin-1-Withanolide A complex, supported by RMSD, RMSF, RoG, and SASA analyses. PCA revealed reduced conformational flexibility, indicating enhanced structural stability of the ligand-bound complex. Additionally, MM-PBSA analysis confirmed that Van der Waals interactions were the primary stabilizing forces, complemented by electrostatic contributions. This integrated computational approach highlights the therapeutic potential of Withanolide A as a neuroprotective agent for SCA1, providing a base for future experimental validation and drug development.

Graphical abstract:

{"title":"Natural compounds as therapeutic candidates for spinocerebellar ataxia type 1: a computational approach.","authors":"Surbhi Singh, Suchitra Singh, Deepika Joshi, C Mohanty, Royana Singh","doi":"10.1007/s40203-025-00308-6","DOIUrl":"10.1007/s40203-025-00308-6","url":null,"abstract":"<p><p>Spinocerebellar Ataxia Type 1 (SCA1) is a progressive neurodegenerative disorder caused by the expansion and aggregation of polyglutamine (polyQ) in the Ataxin-1 (ATXN1) protein, leading to severe neuronal dysfunction. Currently, only symptomatic treatments are available, highlighting the requirement for disease-modifying therapies. This study employed a detailed in silico approach to identify potential neuroprotective natural compounds targeting the Ataxin-1 protein implicated in SCA1. The three-dimensional structure of Ataxin-1 was retrieved, validated, and optimized to achieve a stable structural model. Validation using a Ramachandran plot indicated that 77% of the residues were in favored regions, confirming the reliability of the protein structure. Active site residues were identified using CASTp, and receptor grids were generated for molecular docking studies. A library of 50 natural compounds was screened, among which 21 satisfied Lipinski's rule of five. Molecular docking using PyRx and AutoDock 4.2 identified Withanolide A as the top candidate, exhibiting the highest binding affinity (- 10.14 kcal/mol) and forming four hydrogen bonds with key active site residues. The top six ligands were further assessed for ADMET properties, with Withanolide A showing optimal drug-likeness, high gastrointestinal and blood-brain absorption, and non-toxic profiles. Molecular dynamics simulations over 200 ns demonstrated the stability of the Ataxin-1-Withanolide A complex, supported by RMSD, RMSF, RoG, and SASA analyses. PCA revealed reduced conformational flexibility, indicating enhanced structural stability of the ligand-bound complex. Additionally, MM-PBSA analysis confirmed that Van der Waals interactions were the primary stabilizing forces, complemented by electrostatic contributions. This integrated computational approach highlights the therapeutic potential of Withanolide A as a neuroprotective agent for SCA1, providing a base for future experimental validation and drug development.</p><p><strong>Graphical abstract: </strong></p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"46"},"PeriodicalIF":0.0,"publicationDate":"2025-03-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11910456/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143652794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Deciphering the the molecular mechanism of aloe-emodin in managing type II diabetes mellitus using network pharmacology, molecular docking, and molecular dynamics simulation approaches.
Pub Date : 2025-03-15 eCollection Date: 2025-01-01 DOI: 10.1007/s40203-025-00337-1
Samuel Baker Obakiro, Kenedy Kiyimba, Yahaya Gavamukulya, Richard Maseruka, Catherine Nabitandikwa, Ronald Kibuuka, Jalia Lulenzi, Tonny Wotoyitide Lukwago, Mercy Chebijira, Moses Opio, Edeya Sharon Tracy, Dan Kibuule, Richard Owor Oriko, Paul Waako, Angela Makaye, Daniel M Shadrack, Moses Andima

Aloe-emodin (AE) has drawn interest due to its potential activity against type II diabetes mellitus (T2DM). However, the mechanisms underlying its antidiabetic activity are not well explored. Using network pharmacology, molecular docking and molecular dynamics simulation studies, we investigated its molecular mechanisms in the management of T2DM. Potential target genes of AE were predicted using the Swiss Target Prediction (http://www.swisstargetprediction.ch/) database. The GeneCards, OMIM and DisGeNET databases were used to compile a comprehensive list of genes associated with T2DM. A compound-disease-target network was constructed, and protein-protein interaction networks were analysed to identify hub genes. Finally, molecular docking and interaction analysis between AE and the identified proteins were performed using AutoDock tools. Investigation of AE targets and genes associated with T2DM identified 32 overlapping genes. Gene ontology studies revealed that AE may exert its anti-diabetic effects by modulating glucose metabolism and enhancing cellular response to glucose. Furthermore, KEGG pathway analysis suggested that AE influences these processes by targeting pathways related to apoptosis, insulin resistance, and T2DM signaling. The core target proteins identified were TNF, ALB, TP53, PPARG, BCL2, CASP3, and EGFR. AE interaction with each of these proteins exhibited a binding energy of > - 5 kcal/mol, with TNF showing the lowest binding energy (- 7.75 kcal/mol). Molecular dynamics simulation further validated the molecular docking results with TNF and EGFR exhibiting a strong affinity for AE and forming stable interactions. AE exerts its antidiabetic activity through multiple mechanisms, with the most significant being the amelioration of pancreatic β-cell apoptosis by binding to and inhibiting the actions of TNFα. Further cellular and molecular studies are needed to validate these findings.

Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00337-1.

芦荟大黄素(AE)因其对 II 型糖尿病(T2DM)的潜在活性而备受关注。然而,其抗糖尿病活性的机制尚未得到很好的探索。我们利用网络药理学、分子对接和分子动力学模拟研究,探讨了 AE 在治疗 T2DM 中的分子机制。我们利用瑞士目标预测数据库(http://www.swisstargetprediction.ch/)预测了 AE 的潜在目标基因。我们利用 GeneCards、OMIM 和 DisGeNET 数据库编制了与 T2DM 相关的全面基因列表。构建了化合物-疾病-靶点网络,并分析了蛋白质-蛋白质相互作用网络,以确定枢纽基因。最后,使用 AutoDock 工具进行了 AE 与所识别蛋白质之间的分子对接和相互作用分析。对AE靶点和T2DM相关基因的研究发现了32个重叠基因。基因本体研究显示,AE可能通过调节葡萄糖代谢和增强细胞对葡萄糖的反应来发挥其抗糖尿病作用。此外,KEGG 通路分析表明,AE 通过靶向与细胞凋亡、胰岛素抵抗和 T2DM 信号转导相关的通路来影响这些过程。确定的核心靶蛋白包括 TNF、ALB、TP53、PPARG、BCL2、CASP3 和表皮生长因子受体。AE 与这些蛋白的结合能均大于 - 5 kcal/mol,其中 TNF 的结合能最低(- 7.75 kcal/mol)。分子动力学模拟进一步验证了分子对接结果,TNF 和表皮生长因子受体与 AE 的亲和力很强,并能形成稳定的相互作用。AE 通过多种机制发挥其抗糖尿病活性,其中最重要的机制是通过与 TNFα 结合并抑制 TNFα 的作用来改善胰腺 β 细胞的凋亡。需要进一步的细胞和分子研究来验证这些发现:在线版本包含补充材料,可查阅 10.1007/s40203-025-00337-1。
{"title":"Deciphering the the molecular mechanism of aloe-emodin in managing type II diabetes mellitus using network pharmacology, molecular docking, and molecular dynamics simulation approaches.","authors":"Samuel Baker Obakiro, Kenedy Kiyimba, Yahaya Gavamukulya, Richard Maseruka, Catherine Nabitandikwa, Ronald Kibuuka, Jalia Lulenzi, Tonny Wotoyitide Lukwago, Mercy Chebijira, Moses Opio, Edeya Sharon Tracy, Dan Kibuule, Richard Owor Oriko, Paul Waako, Angela Makaye, Daniel M Shadrack, Moses Andima","doi":"10.1007/s40203-025-00337-1","DOIUrl":"10.1007/s40203-025-00337-1","url":null,"abstract":"<p><p>Aloe-emodin (AE) has drawn interest due to its potential activity against type II diabetes mellitus (T2DM). However, the mechanisms underlying its antidiabetic activity are not well explored. Using network pharmacology, molecular docking and molecular dynamics simulation studies, we investigated its molecular mechanisms in the management of T2DM. Potential target genes of AE were predicted using the Swiss Target Prediction (http://www.swisstargetprediction.ch/) database. The GeneCards, OMIM and DisGeNET databases were used to compile a comprehensive list of genes associated with T2DM. A compound-disease-target network was constructed, and protein-protein interaction networks were analysed to identify hub genes. Finally, molecular docking and interaction analysis between AE and the identified proteins were performed using AutoDock tools. Investigation of AE targets and genes associated with T2DM identified 32 overlapping genes. Gene ontology studies revealed that AE may exert its anti-diabetic effects by modulating glucose metabolism and enhancing cellular response to glucose. Furthermore, KEGG pathway analysis suggested that AE influences these processes by targeting pathways related to apoptosis, insulin resistance, and T2DM signaling. The core target proteins identified were TNF, ALB, TP53, PPARG, BCL2, CASP3, and EGFR. AE interaction with each of these proteins exhibited a binding energy of > - 5 kcal/mol, with TNF showing the lowest binding energy (- 7.75 kcal/mol). Molecular dynamics simulation further validated the molecular docking results with TNF and EGFR exhibiting a strong affinity for AE and forming stable interactions. AE exerts its antidiabetic activity through multiple mechanisms, with the most significant being the amelioration of pancreatic β-cell apoptosis by binding to and inhibiting the actions of TNFα. Further cellular and molecular studies are needed to validate these findings.</p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00337-1.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 1","pages":"45"},"PeriodicalIF":0.0,"publicationDate":"2025-03-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11910477/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143652864","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
In silico pharmacology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1