Background: Lymphatic filariasis, or elephantiasis, is a neglected tropical disease caused by filarial nematodes such as Brugia malayi. Current antifilarial drugs-diethylcarbamazine (DEC), albendazole, and ivermectin form the basis of mass drug administration (MDA) programs for lymphatic filariasis. While effective against microfilarial stages, these agents show little or no macrofilaricidal activity, necessitating repeated treatment rounds to interrupt transmission. Growing evidence of reduced efficacy and emerging resistance further threatens the sustainability of these regimens. Thioredoxin reductase (TrxR) and β-tubulin are critical for parasite survival: TrxR maintains redox balance and protects against oxidative stress, while β-tubulin supports cytoskeletal integrity, intracellular transport, and cell division. Their combined roles in stress adaptation and structural stability make them compelling dual targets. This study employed structure-based virtual screening, molecular docking, and molecular dynamics simulations (MDS) to identify novel thiol-based inhibitors against both proteins.
Results: A total of 467 compounds were virtually screened, leading to the identification of seven lead candidates with superior docking scores (- 8.5 to - 4.0 kcal/mol) compared to the standard drug albendazole (- 5.3 to - 4.5 kcal/mol). Notably, compound 15 demonstrated the strongest binding affinity coupled with an optimal toxicity profile. Pharmacokinetic analysis using ADME assays confirmed drug-likeness and oral bioavailability of the top ligands, with minimal Lipinski's rule violations. Molecular dynamics simulations exceeding 100 ns revealed sustained stability of the protein-ligand complex, which was further supported by RMSD and RMSF analyses, demonstrating conformational stability. Principal interactions comprised hydrogen bonding, hydrophobic contacts, and π-stacking with conserved residues within the active sites of the target proteins.
Conclusion: The integrated in silico approach combining docking, pharmacokinetic profiling, and MDS successfully identified potent thiol-based ligands with high affinity for β-tubulin and TrxR in B. malayi. Among these, HI/CYR/TH-15 emerged as the most promising lead. These findings provide a foundation for the development of next-generation anti-filarial therapies targeting multiple life stages, warranting further in vitro and in vivo validation to confirm therapeutic potential.
Graphical abstract:
Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00474-7.
{"title":"Structure-based virtual screening of β-tubulin and thioredoxin reductase in <i>Brugia malayi</i>: a step toward novel anti-filarial agents.","authors":"Vrushali Patil, Ashish Asrondkar, Harshada Sawant, Chetan M Jain, Dattatray Nandaram Thorat, Bhagwat Ramdas Patil","doi":"10.1007/s40203-025-00474-7","DOIUrl":"https://doi.org/10.1007/s40203-025-00474-7","url":null,"abstract":"<p><strong>Background: </strong>Lymphatic filariasis, or elephantiasis, is a neglected tropical disease caused by filarial nematodes such as <i>Brugia malayi</i>. Current antifilarial drugs-diethylcarbamazine (DEC), albendazole, and ivermectin form the basis of mass drug administration (MDA) programs for lymphatic filariasis. While effective against microfilarial stages, these agents show little or no macrofilaricidal activity, necessitating repeated treatment rounds to interrupt transmission. Growing evidence of reduced efficacy and emerging resistance further threatens the sustainability of these regimens. Thioredoxin reductase (TrxR) and β-tubulin are critical for parasite survival: TrxR maintains redox balance and protects against oxidative stress, while β-tubulin supports cytoskeletal integrity, intracellular transport, and cell division. Their combined roles in stress adaptation and structural stability make them compelling dual targets. This study employed structure-based virtual screening, molecular docking, and molecular dynamics simulations (MDS) to identify novel thiol-based inhibitors against both proteins.</p><p><strong>Results: </strong>A total of 467 compounds were virtually screened, leading to the identification of seven lead candidates with superior docking scores (- 8.5 to - 4.0 kcal/mol) compared to the standard drug albendazole (- 5.3 to - 4.5 kcal/mol). Notably, compound 15 demonstrated the strongest binding affinity coupled with an optimal toxicity profile. Pharmacokinetic analysis using ADME assays confirmed drug-likeness and oral bioavailability of the top ligands, with minimal Lipinski's rule violations. Molecular dynamics simulations exceeding 100 ns revealed sustained stability of the protein-ligand complex, which was further supported by RMSD and RMSF analyses, demonstrating conformational stability. Principal interactions comprised hydrogen bonding, hydrophobic contacts, and π-stacking with conserved residues within the active sites of the target proteins.</p><p><strong>Conclusion: </strong>The integrated in silico approach combining docking, pharmacokinetic profiling, and MDS successfully identified potent thiol-based ligands with high affinity for β-tubulin and TrxR in <i>B. malayi</i>. Among these, HI/CYR/TH-15 emerged as the most promising lead. These findings provide a foundation for the development of next-generation anti-filarial therapies targeting multiple life stages, warranting further in vitro and in vivo validation to confirm therapeutic potential.</p><p><strong>Graphical abstract: </strong></p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00474-7.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"178"},"PeriodicalIF":0.0,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12618773/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544021","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}
Pub Date : 2025-11-14eCollection Date: 2025-01-01DOI: 10.1007/s40203-025-00469-4
Sarumathi Ravishankar, Sankaranarayanan Chandrasekaran, Mohammed Jalaludeen Abdulkadhar
Diabetic peripheral neuropathy is a devasting microvascular complication that is associated with chronic pain. Currently transient receptor potential vanilloid 1 (TRPV1), serotonin transporter (SERT) and voltage gated calcium channel (VGCC) are considered as potential targets for neuropathic pain management. The present study investigated the interactions of terpinolene with pain targets by molecular docking and simulation studies. The structure and physicochemical properties of terpinolene were obtained from the PubChem, SwissADME and pkCSM databases respectively. The crystallographic structures of the molecular targets retrieved from PDB database (5IS0, 6AWO and 7MIY) were used for docking analysis using Schrodinger software. Docking studies revealed that terpinolene docks well with pain targets related to neuropathy. Molecular dynamics (MD) simulations of protein-ligand complexes were carried out using CABS-flex V2.0 and the iMOD server. These tools were used to assess the root-mean-square fluctuations (RMSFs) and the stability of the protein structures, respectively. The findings indicated that the docked models exhibited greater flexibility and stability with terpinolene. Thus, terpinolene can be considered as a potential drug in the management of pain associated with diabetic peripheral neuropathy.
{"title":"Studies on the role of Terpinolene a monoterpene on neuropathic pain: an in Silico and molecular dynamic approach.","authors":"Sarumathi Ravishankar, Sankaranarayanan Chandrasekaran, Mohammed Jalaludeen Abdulkadhar","doi":"10.1007/s40203-025-00469-4","DOIUrl":"https://doi.org/10.1007/s40203-025-00469-4","url":null,"abstract":"<p><p>Diabetic peripheral neuropathy is a devasting microvascular complication that is associated with chronic pain. Currently transient receptor potential vanilloid 1 (TRPV1), serotonin transporter (SERT) and voltage gated calcium channel (VGCC) are considered as potential targets for neuropathic pain management. The present study investigated the interactions of terpinolene with pain targets by molecular docking and simulation studies. The structure and physicochemical properties of terpinolene were obtained from the PubChem, SwissADME and pkCSM databases respectively. The crystallographic structures of the molecular targets retrieved from PDB database (5IS0, 6AWO and 7MIY) were used for docking analysis using Schrodinger software. Docking studies revealed that terpinolene docks well with pain targets related to neuropathy. Molecular dynamics (MD) simulations of protein-ligand complexes were carried out using CABS-flex V2.0 and the iMOD server. These tools were used to assess the root-mean-square fluctuations (RMSFs) and the stability of the protein structures, respectively. The findings indicated that the docked models exhibited greater flexibility and stability with terpinolene. Thus, terpinolene can be considered as a potential drug in the management of pain associated with diabetic peripheral neuropathy.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"183"},"PeriodicalIF":0.0,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12618763/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544062","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}
Ulcerative colitis (UC) is a colon inflammatory illness that causes significant harm to the colon tissues. Nelumbo nucifera G. has been used for hundreds of years in old-style medication for treating gastrointestinal issues like inflammation and ulcers. To explore the therapeutic effects of Nelumbo nucifera ethanolic extract (NNEE) on 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced UC in a rat model and to reveal the probable mechanisms intricate in the anti-colitic activities of NNEE. Network pharmacology analysis of Nelumbo nucifera G. was performed to determine the targets and pathways. The UC was prompted by intrarectal treatment of TNBS and rats were given with the treatment of NNEE via oral gavage. Body weight, colon weight, length, disease activity index, spleen weight, thymus weight, Macroscopic, adhesion and histopathological scores, myeloperoxidase, nitric oxide, haematological, and antioxidant parameters were assessed in colon tissues. Inflammatory cytokines were also assessed in serum samples. A comprehensive set of 11 active components was determined to be essential to NNEE effectiveness against UC treatment based on network pharmacology. The study also identified 152 putative core therapeutic targets in addition to the top 5 targets, which are ESR1, NFKB1, SLC6A4, CREBBP, and HMGCR. After NNEE treatment, the body weights and thymus weight of rats significantly increased, as colon weight, spleen weight, disease activity index score and histological score. The haematological parameters were significantly restored such as an increase in haemoglobin, red blood cells, platelet count and a reduction in white blood cell count. However, nitric oxide and myeloperoxidase were found to be decreased with the restoration of antioxidant markers and a reduction in inflammatory cytokines levels when matched to TNBS-treated rats. NNEE decreases colonic injury in the TNBS colitis model and alleviates inflammatory and oxidative events. It does this by boosting mucus production, decreasing leukocyte migration, lowering serum levels of inflammatory cytokines, and restoring antioxidant parameters, all of which suggest that NNEE could be used as a medicinal plant to treat UC.
Graphical abstract:
Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00458-7.
{"title":"Network pharmacology and molecular docking of <i>Nelumbo nucifera</i> Gaertn. to reveal the pharmacological mechanisms in treating TNBS-induced ulcerative colitis in rats.","authors":"Deepa Mandlik, Rutuparna Joshi, Chaitanya Kalsekar, Abhishek Kamble, Manoj Magre, Amol Muthal, Satish Mandlik, Vaibhav Shinde","doi":"10.1007/s40203-025-00458-7","DOIUrl":"https://doi.org/10.1007/s40203-025-00458-7","url":null,"abstract":"<p><p>Ulcerative colitis (UC) is a colon inflammatory illness that causes significant harm to the colon tissues. <i>Nelumbo nucifera</i> G. has been used for hundreds of years in old-style medication for treating gastrointestinal issues like inflammation and ulcers. To explore the therapeutic effects of <i>Nelumbo nucifera</i> ethanolic extract (NNEE) on 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced UC in a rat model and to reveal the probable mechanisms intricate in the anti-colitic activities of NNEE. Network pharmacology analysis of <i>Nelumbo nucifera</i> G. was performed to determine the targets and pathways. The UC was prompted by intrarectal treatment of TNBS and rats were given with the treatment of NNEE via oral gavage. Body weight, colon weight, length, disease activity index, spleen weight, thymus weight, Macroscopic, adhesion and histopathological scores, myeloperoxidase, nitric oxide, haematological, and antioxidant parameters were assessed in colon tissues. Inflammatory cytokines were also assessed in serum samples. A comprehensive set of 11 active components was determined to be essential to NNEE effectiveness against UC treatment based on network pharmacology. The study also identified 152 putative core therapeutic targets in addition to the top 5 targets, which are ESR1, NFKB1, SLC6A4, CREBBP, and HMGCR. After NNEE treatment, the body weights and thymus weight of rats significantly increased, as colon weight, spleen weight, disease activity index score and histological score. The haematological parameters were significantly restored such as an increase in haemoglobin, red blood cells, platelet count and a reduction in white blood cell count. However, nitric oxide and myeloperoxidase were found to be decreased with the restoration of antioxidant markers and a reduction in inflammatory cytokines levels when matched to TNBS-treated rats. NNEE decreases colonic injury in the TNBS colitis model and alleviates inflammatory and oxidative events. It does this by boosting mucus production, decreasing leukocyte migration, lowering serum levels of inflammatory cytokines, and restoring antioxidant parameters, all of which suggest that NNEE could be used as a medicinal plant to treat UC.</p><p><strong>Graphical abstract: </strong></p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00458-7.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"180"},"PeriodicalIF":0.0,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12618775/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544710","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}
Pub Date : 2025-11-14eCollection Date: 2025-01-01DOI: 10.1007/s40203-025-00471-w
Sushma Jahagirdar, Smitha S Bhat, R Sindhu, Sarana Rose Sommano, Rashmi Doddabassappa, Pradeep Matam, Nishanth Bhat, Shashanka K Prasad
Background: The growing incidence of cancer worldwide necessitates the development of novel, effective, and affordable antineoplastic drugs. Clinacanthus nutans, an ethnomedicinal plant known for its diverse pharmacological properties, has shown promising anticancer potential. This study investigates the anticancer, anti-inflammatory, and antioxidant activities of a fraction isolated from Clinacanthus nutans leaves.
Results: Quantification of the total phenolic content of CNM.CF5.5 revealed a substantial presence of phenolic compounds. Antioxidant potential, assessed through DPPH, FRAP, and ABTS assays, demonstrated a clear dose-dependent increase in activity. The BSA denaturation assay further indicated marked anti-inflammatory effects, comparable to those of the diclofenac sodium standard. Cytotoxic evaluation by MTT assay confirmed potent growth inhibition of MCF-7 and A549 cells, with IC₅₀ values of 61.67 µg/mL and 81.62 µg/mL, respectively. HR-LCMS profiling of CNM.CF5.5 identified a diverse array of phytochemicals. Subsequent molecular docking studies, performed using the Maestro interface of the Schrödinger Suite, revealed favorable binding interactions between ligands from the fraction and cancer-associated proteins. Notably, vitexin and isovitexin exhibited strong interactions with CDK1, CDK2, and human NAD[P]H-quinone oxidoreductase, supporting their potential role as key anticancer constituents of the fraction.
Conclusion: These findings support the potential of CNM.CF 5.5 bioactives as suitable agents in cancer therapy.
背景:世界范围内癌症发病率的增长要求开发新型、有效和负担得起的抗肿瘤药物。山茱萸是一种以其多种药理特性而闻名的民族药用植物,具有良好的抗癌潜力。本研究研究了从芒萁叶中分离得到的抗肿瘤、抗炎和抗氧化活性。结果:对CNM.CF5.5的总酚含量进行定量分析,发现含有大量酚类化合物。通过DPPH、FRAP和ABTS测试评估的抗氧化潜力显示出明显的剂量依赖性活性增加。BSA变性试验进一步显示了显著的抗炎作用,与双氯芬酸钠标准相当。MTT法的细胞毒性评估证实了MCF-7和A549细胞的有效生长抑制,IC₅0值分别为61.67µg/mL和81.62µg/mL。CNM.CF5.5的HR-LCMS分析鉴定出多种植物化学物质。随后使用Schrödinger Suite的Maestro界面进行的分子对接研究显示,该组分的配体与癌症相关蛋白之间存在良好的结合相互作用。值得注意的是,牡荆素和异牡荆素表现出与CDK1、CDK2和人类NAD[P] h -醌氧化还原酶的强相互作用,支持它们作为关键抗癌成分的潜在作用。结论:这些发现支持CNM的潜力。CF 5.5生物活性物在癌症治疗中的应用
{"title":"Isolation and characterization of a potent anticancer fraction from <i>Clinacanthus nutans</i> targeting MCF-7 and A549 cells: an integrated in vitro and in silico study.","authors":"Sushma Jahagirdar, Smitha S Bhat, R Sindhu, Sarana Rose Sommano, Rashmi Doddabassappa, Pradeep Matam, Nishanth Bhat, Shashanka K Prasad","doi":"10.1007/s40203-025-00471-w","DOIUrl":"https://doi.org/10.1007/s40203-025-00471-w","url":null,"abstract":"<p><strong>Background: </strong>The growing incidence of cancer worldwide necessitates the development of novel, effective, and affordable antineoplastic drugs. <i>Clinacanthus nutans</i>, an ethnomedicinal plant known for its diverse pharmacological properties, has shown promising anticancer potential. This study investigates the anticancer, anti-inflammatory, and antioxidant activities of a fraction isolated from <i>Clinacanthus nutans</i> leaves.</p><p><strong>Results: </strong>Quantification of the total phenolic content of CNM.CF5.5 revealed a substantial presence of phenolic compounds. Antioxidant potential, assessed through DPPH, FRAP, and ABTS assays, demonstrated a clear dose-dependent increase in activity. The BSA denaturation assay further indicated marked anti-inflammatory effects, comparable to those of the diclofenac sodium standard. Cytotoxic evaluation by MTT assay confirmed potent growth inhibition of MCF-7 and A549 cells, with IC₅₀ values of 61.67 µg/mL and 81.62 µg/mL, respectively. HR-LCMS profiling of CNM.CF5.5 identified a diverse array of phytochemicals. Subsequent molecular docking studies, performed using the Maestro interface of the Schrödinger Suite, revealed favorable binding interactions between ligands from the fraction and cancer-associated proteins. Notably, vitexin and isovitexin exhibited strong interactions with CDK1, CDK2, and human NAD[P]H-quinone oxidoreductase, supporting their potential role as key anticancer constituents of the fraction.</p><p><strong>Conclusion: </strong>These findings support the potential of CNM.CF 5.5 bioactives as suitable agents in cancer therapy.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"182"},"PeriodicalIF":0.0,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12618782/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544725","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}
Marine algae are known for producing a wide variety of bioactive secondary metabolites. The present study investigated both in vitro and in silico approaches to evaluate the antioxidant, antibacterial, and anticancer properties of brown seaweed Sargassum cristaefolium, aim of assessing its biological activities. Preliminary phytochemical analysis revealed the presence of eight phytochemical constituents exhibiting notable biological properties. The seaweed extract was characterized using FT-IR and GC-MS, which identified various functional groups, including alkanes and anhydrides as well as bioactive compounds. Among these, phytol (3.66%) was the major bioactive compound in the ethanolic extract. The extract also demonstrated strong antioxidant activity, with an IC₅₀ value of less than 127.96 µg/mL. Furthermore, ethanolic extract exhibited significant antibacterial activity against Mycobacterium smegmatis (15.3 ± 0.50 mm) and anticancer efficiency against the A549 lung cancer cell line with an IC50 value of 222 µg/mL. Computational analysis identified key compounds from S. cristaefolium, which showed strong binding affinities towards specific proteins such as DNA gyrase, human peroxiredoxin 6, and epidermal growth factor receptor (EGFR), supporting their potential as antioxidant, anticancer, and antibacterial agents for novel drug development.
Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00465-8.
{"title":"Exploring the therapeutic potential of brown seaweed <i>Sargassum cristaefolium</i>: in vitro and in silico insights into antioxidant, antibacterial, and anticancer properties.","authors":"Nandhini Selvaraj, Aravinth Annamalai, Mishel Francis, Saroja Ramasubbu Sivakumar","doi":"10.1007/s40203-025-00465-8","DOIUrl":"https://doi.org/10.1007/s40203-025-00465-8","url":null,"abstract":"<p><p>Marine algae are known for producing a wide variety of bioactive secondary metabolites. The present study investigated both in vitro and in silico approaches to evaluate the antioxidant, antibacterial, and anticancer properties of brown seaweed <i>Sargassum cristaefolium,</i> aim of assessing its biological activities. Preliminary phytochemical analysis revealed the presence of eight phytochemical constituents exhibiting notable biological properties. The seaweed extract was characterized using FT-IR and GC-MS, which identified various functional groups, including alkanes and anhydrides as well as bioactive compounds. Among these, phytol (3.66%) was the major bioactive compound in the ethanolic extract. The extract also demonstrated strong antioxidant activity, with an IC₅₀ value of less than 127.96 µg/mL. Furthermore, ethanolic extract exhibited significant antibacterial activity against <i>Mycobacterium smegmatis</i> (15.3 ± 0.50 mm) and anticancer efficiency against the A549 lung cancer cell line with an IC<sub>50</sub> value of 222 µg/mL. Computational analysis identified key compounds from <i>S. cristaefolium</i>, which showed strong binding affinities towards specific proteins such as DNA gyrase, human peroxiredoxin 6, and epidermal growth factor receptor (EGFR), supporting their potential as antioxidant, anticancer, and antibacterial agents for novel drug development.</p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00465-8.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"181"},"PeriodicalIF":0.0,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12618761/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544688","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}
Pub Date : 2025-11-14eCollection Date: 2025-01-01DOI: 10.1007/s40203-025-00472-9
O O Fawibe, B R Lawal, A A Mustafa, A S Oyelakin, O F Akinyemi, A O Bankole, S A Poku, F Boudou, D Das
Tuberculosis (TB) is a significant global public health challenge. Targeting enoyl-acyl carrier protein (ACP) reductase (InhA), an enzyme involved in mycolic acid biosynthesis, is a promising path to discovering an effective treatment for tuberculosis. This study assessed the inhibitory potential of bioactive compounds from four medicinal plants (Garcinia kola, Moringa oleifera, Newbouldia laevis, and Ocimum gratissimum) and control drugs (Isoniazid and Ethionamide) against InhA. Molecular docking and computational tools were used to evaluate the binding affinities and interactions with InhA's active site. Drug-likeness, binding affinities, bioactivity, and absorption, distribution, metabolism, excretion, and toxicity (ADMET) attributes were determined using online tools. Ten out of twenty-three bioactive compounds studied were screened out due to violating Lipinski's, Ghose's, Veber's, Egan's, or Muegge's rules. The remaining thirteen compounds showed stronger binding affinities with InhA than the control drugs. The binding energy of the bioactive compounds ranged from - 8.0 to - 9.5 kcal/mol, while those of Isoniazid and Ethionamide were 6.1 kcal/mol and - 6.0 kcal/mol, respectively. The compounds also exhibited hydrogen bonds, hydrophobic, and π stacking interactions with the protein residues. Molecular dynamic simulations confirmed that 5,7-dihydroxydehydroiso-α-lapachone had a compact and more stable complex with InhA than the hit ligands with high binding energies. The ADMET property of each hit ligand predicted its ability to effectively reach and remain at the target protein to exert its therapeutic influence. The study shows that the screened bioactive compounds, especially 5,7-dihydroxydehydroiso-α-lapachone, exhibit drug-like properties capable of inhibiting InhA, hence, could serve as a novel anti-tuberculosis drug.
Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00472-9.
{"title":"Molecular docking and pharmacokinetic studies of bioactive compounds from medicinal plants as promising inhibitory agents against <i>Mycobacterium tuberculosis</i> enoyl-acyl carrier protein (ACP)-reductase (InhA).","authors":"O O Fawibe, B R Lawal, A A Mustafa, A S Oyelakin, O F Akinyemi, A O Bankole, S A Poku, F Boudou, D Das","doi":"10.1007/s40203-025-00472-9","DOIUrl":"https://doi.org/10.1007/s40203-025-00472-9","url":null,"abstract":"<p><p>Tuberculosis (TB) is a significant global public health challenge. Targeting enoyl-acyl carrier protein (ACP) reductase (InhA), an enzyme involved in mycolic acid biosynthesis, is a promising path to discovering an effective treatment for tuberculosis. This study assessed the inhibitory potential of bioactive compounds from four medicinal plants (<i>Garcinia kola</i>, <i>Moringa oleifera</i>, <i>Newbouldia laevis</i>, and <i>Ocimum gratissimum</i>) and control drugs (Isoniazid and Ethionamide) against InhA. Molecular docking and computational tools were used to evaluate the binding affinities and interactions with InhA's active site. Drug-likeness, binding affinities, bioactivity, and absorption, distribution, metabolism, excretion, and toxicity (ADMET) attributes were determined using online tools. Ten out of twenty-three bioactive compounds studied were screened out due to violating Lipinski's, Ghose's, Veber's, Egan's, or Muegge's rules. The remaining thirteen compounds showed stronger binding affinities with InhA than the control drugs. The binding energy of the bioactive compounds ranged from - 8.0 to - 9.5 kcal/mol, while those of Isoniazid and Ethionamide were 6.1 kcal/mol and - 6.0 kcal/mol, respectively. The compounds also exhibited hydrogen bonds, hydrophobic, and π stacking interactions with the protein residues. Molecular dynamic simulations confirmed that 5,7-dihydroxydehydroiso-α-lapachone had a compact and more stable complex with InhA than the hit ligands with high binding energies. The ADMET property of each hit ligand predicted its ability to effectively reach and remain at the target protein to exert its therapeutic influence. The study shows that the screened bioactive compounds, especially 5,7-dihydroxydehydroiso-α-lapachone, exhibit drug-like properties capable of inhibiting InhA, hence, could serve as a novel anti-tuberculosis drug.</p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00472-9.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"179"},"PeriodicalIF":0.0,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12618755/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544686","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}
Pub Date : 2025-11-06eCollection Date: 2025-01-01DOI: 10.1007/s40203-025-00481-8
Lutfat A Usman, Emmanuel O Ajani, Afolabi C Akinmoladun, Rasheed B Ibrahim, Hassan T Abdulameed
Background: Neurodegenerative diseases, marked by progressive brain cell damage, pose a major health challenge due to complex causes like oxidative stress and inflammation. Tamarindus indica, a plant rich in bioactive flavonoids and terpenoids, is underexplored for its neuroprotective potential against chemical-induced neuronal injury.
Methods: We used computational tools (network pharmacology, molecular docking, and molecular dynamics simulations) to investigate how Tamarindus indica fruit pulp ethanol extract's bioactive compounds might protect against neurodegeneration. Additionally, we tested the extract in a rat model of acrylonitrile-induced neuronal damage. Seventy male albino rats were divided into seven groups, receiving either acrylonitrile alone, Tamarindus indica extract (200 or 400 mg/kg) before or after acrylonitrile, or a standard drug (donepezil). Over the course of 28 days, we measured oxidative stress, inflammation, and brain enzyme activity.
Results: Network pharmacology, molecular docking, and molecular dynamics simulations mapped and validated interactions between T. indica bioactives and neurodegenerative pathway targets, revealing strong binding affinities and stability of T. indica bioactives, particularly apigenin, to key neuroprotective targets. In vivo, the extract significantly enhanced antioxidant enzyme activity, reduced lipid peroxidation, downregulated pro-inflammatory cytokines, and inhibited acetylcholinesterase, a key enzyme in neurodegeneration.
Conclusions: These findings position T. indica extract as a promising multi-targeted therapy for neurodegenerative disorders, leveraging antioxidant and anti-inflammatory mechanisms. Further preclinical and clinical studies are needed to translate these insights into novel treatments, bridging traditional phytotherapy with modern pharmacology.
{"title":"<i>Tamarindus indica</i> fruit pulp in neurodegeneration: a multimodal pharmacological study.","authors":"Lutfat A Usman, Emmanuel O Ajani, Afolabi C Akinmoladun, Rasheed B Ibrahim, Hassan T Abdulameed","doi":"10.1007/s40203-025-00481-8","DOIUrl":"10.1007/s40203-025-00481-8","url":null,"abstract":"<p><strong>Background: </strong>Neurodegenerative diseases, marked by progressive brain cell damage, pose a major health challenge due to complex causes like oxidative stress and inflammation. <i>Tamarindus indica</i>, a plant rich in bioactive flavonoids and terpenoids, is underexplored for its neuroprotective potential against chemical-induced neuronal injury.</p><p><strong>Methods: </strong>We used computational tools (network pharmacology, molecular docking, and molecular dynamics simulations) to investigate how <i>Tamarindus indica</i> fruit pulp ethanol extract's bioactive compounds might protect against neurodegeneration. Additionally, we tested the extract in a rat model of acrylonitrile-induced neuronal damage. Seventy male albino rats were divided into seven groups, receiving either acrylonitrile alone, <i>Tamarindus indica</i> extract (200 or 400 mg/kg) before or after acrylonitrile, or a standard drug (donepezil). Over the course of 28 days, we measured oxidative stress, inflammation, and brain enzyme activity.</p><p><strong>Results: </strong>Network pharmacology, molecular docking, and molecular dynamics simulations mapped and validated interactions between <i>T. indica</i> bioactives and neurodegenerative pathway targets, revealing strong binding affinities and stability of <i>T. indica</i> bioactives, particularly apigenin, to key neuroprotective targets. In vivo, the extract significantly enhanced antioxidant enzyme activity, reduced lipid peroxidation, downregulated pro-inflammatory cytokines, and inhibited acetylcholinesterase, a key enzyme in neurodegeneration.</p><p><strong>Conclusions: </strong>These findings position <i>T. indica</i> extract as a promising multi-targeted therapy for neurodegenerative disorders, leveraging antioxidant and anti-inflammatory mechanisms. Further preclinical and clinical studies are needed to translate these insights into novel treatments, bridging traditional phytotherapy with modern pharmacology.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"171"},"PeriodicalIF":0.0,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12592624/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145484320","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}
West Nile Virus (WNV), a globally distributed mosquito-borne flavivirus, poses a significant public health threat due to its potential to cause severe neurological complications and the absence of licensed vaccines or specific antiviral treatments. In this study, we applied an integrated artificial intelligence, structural, and immunoinformatics-driven approach to design a multi-epitope subunit vaccine (MESV) targeting the non-structural proteins NS1 and NS4B, which play key roles in viral replication and immune evasion. B-cell, cytotoxic T lymphocyte (CTL), and helper T lymphocyte (HTL) epitopes were computationally predicted and screened for high antigenicity, non-allergenicity, and non-toxicity. Selected epitopes were assembled into a 307-amino-acid construct incorporating the 50S ribosomal protein L7/L12 and RS09 as adjuvants, along with a PADRE sequence to enhance T-helper responses. Structural modeling, physicochemical analysis, and mRNA structure prediction confirmed stability, solubility, and immunogenic potential. Molecular docking and 100-ns molecular dynamics simulations demonstrated strong, sustained interactions with TLR3 and TLR4, with principal component analysis supporting conformational stability. Immune simulations predicted robust humoral and cellular responses, including elevated IgM, IgG1, IgG2, cytokine production, and memory formation. Population coverage analysis revealed broad HLA representation, particularly in Europe (99.55%). Codon optimization and in silico cloning confirmed suitability for E. coli expression. Collectively, these AI-assisted computational insights highlight the MESV as a promising WNV vaccine candidate and provide a rational framework for future experimental evaluation.
Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00459-6.
{"title":"Artificial intelligence, structural, and immunoinformatics-driven multi-epitope vaccine design targeting non-structural proteins of West Nile Virus.","authors":"Rahul Mallick, Guneswar Sethi, Satyanarayan Sethi, Jeong Ho Hwang, Ramadas Krishna","doi":"10.1007/s40203-025-00459-6","DOIUrl":"10.1007/s40203-025-00459-6","url":null,"abstract":"<p><p>West Nile Virus (WNV), a globally distributed mosquito-borne flavivirus, poses a significant public health threat due to its potential to cause severe neurological complications and the absence of licensed vaccines or specific antiviral treatments. In this study, we applied an integrated artificial intelligence, structural, and immunoinformatics-driven approach to design a multi-epitope subunit vaccine (MESV) targeting the non-structural proteins NS1 and NS4B, which play key roles in viral replication and immune evasion. B-cell, cytotoxic T lymphocyte (CTL), and helper T lymphocyte (HTL) epitopes were computationally predicted and screened for high antigenicity, non-allergenicity, and non-toxicity. Selected epitopes were assembled into a 307-amino-acid construct incorporating the 50S ribosomal protein L7/L12 and RS09 as adjuvants, along with a PADRE sequence to enhance T-helper responses. Structural modeling, physicochemical analysis, and mRNA structure prediction confirmed stability, solubility, and immunogenic potential. Molecular docking and 100-ns molecular dynamics simulations demonstrated strong, sustained interactions with TLR3 and TLR4, with principal component analysis supporting conformational stability. Immune simulations predicted robust humoral and cellular responses, including elevated IgM, IgG1, IgG2, cytokine production, and memory formation. Population coverage analysis revealed broad HLA representation, particularly in Europe (99.55%). Codon optimization and in silico cloning confirmed suitability for <i>E. coli</i> expression. Collectively, these AI-assisted computational insights highlight the MESV as a promising WNV vaccine candidate and provide a rational framework for future experimental evaluation.</p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00459-6.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"177"},"PeriodicalIF":0.0,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12592618/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145484345","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}
Areca catechu nut exhibits potential antidiabetic properties due to its bioactive compounds, including alkaloids, flavonoids, and phenolic compounds, which enhance insulin sensitivity and glucose uptake. However, the specific compounds responsible and their underlying mechanisms remain undefined. This study employed an integrated computational and experimental approach to elucidate the antidiabetic action of Areca catechu. Phytochemicals were retrieved from Dr. Duke's and IMPPAT databases, and molecular targets were predicted using SwissTargetPrediction, SEA Search Server, and Binding Database. Disease-associated targets were obtained from DisGeNet, GeneCards, and MalaCards databases. Network pharmacology analysis identified carbonic anhydrase II (CA-II) as a key therapeutic target. Molecular docking using BIOVIA Discovery Studio revealed -CDOCKER Interaction Energy of chrysophanic acid (64.20 kcal/mol), isorhamnetin (62.52 kcal/mol), quercetin (54.54 kcal/mol), and ferulic acid (52.13 kcal/mol) had stronger binding affinities for CA-II than the reference inhibitor, acetazolamide (36.80 kcal/mol). These compounds were quantified using LC-MS/MS in nut extract, and their CA-II inhibitory activity was assessed using an in vitro assay. Chrysophanic acid exhibited the highest inhibitory activity (IC₅₀ = 0.3125 ± 0.03 µM), demonstrating a potency 17.9-fold greater than the reference drug, acetazolamide. Additionally, 100ns molecular dynamics simulations confirmed the stability of chrysophanic acid-CA-II interactions. Taken together, these results suggest that CA-II inhibition is a potential mechanism for the antidiabetic effects of Areca catechu. They also identify key lead compounds for future development as antidiabetic agents.
Graphical abstract:
Supplementary information: The online version contains supplementary material available at 10.1007/s40203-025-00464-9.
{"title":"Network pharmacology, molecular docking and in vitro experimental validation to unveil antidiabetic mechanism of <i>Areca Catechu</i>.","authors":"Sanket Jadhav, RajaSekhar Reddy Alavala, Ashish Kanhed, Prashant Kurkate, Amisha Vora","doi":"10.1007/s40203-025-00464-9","DOIUrl":"10.1007/s40203-025-00464-9","url":null,"abstract":"<p><p><i>Areca catechu</i> nut exhibits potential antidiabetic properties due to its bioactive compounds, including alkaloids, flavonoids, and phenolic compounds, which enhance insulin sensitivity and glucose uptake. However, the specific compounds responsible and their underlying mechanisms remain undefined. This study employed an integrated computational and experimental approach to elucidate the antidiabetic action of <i>Areca catechu</i>. Phytochemicals were retrieved from Dr. Duke's and IMPPAT databases, and molecular targets were predicted using SwissTargetPrediction, SEA Search Server, and Binding Database. Disease-associated targets were obtained from DisGeNet, GeneCards, and MalaCards databases. Network pharmacology analysis identified carbonic anhydrase II (CA-II) as a key therapeutic target. Molecular docking using BIOVIA Discovery Studio revealed -CDOCKER Interaction Energy of chrysophanic acid (64.20 kcal/mol), isorhamnetin (62.52 kcal/mol), quercetin (54.54 kcal/mol), and ferulic acid (52.13 kcal/mol) had stronger binding affinities for CA-II than the reference inhibitor, acetazolamide (36.80 kcal/mol). These compounds were quantified using LC-MS/MS in nut extract, and their CA-II inhibitory activity was assessed using an in vitro assay. Chrysophanic acid exhibited the highest inhibitory activity (IC₅₀ = 0.3125 ± 0.03 µM), demonstrating a potency 17.9-fold greater than the reference drug, acetazolamide. Additionally, 100ns molecular dynamics simulations confirmed the stability of chrysophanic acid-CA-II interactions. Taken together, these results suggest that CA-II inhibition is a potential mechanism for the antidiabetic effects of <i>Areca catechu</i>. They also identify key lead compounds for future development as antidiabetic agents.</p><p><strong>Graphical abstract: </strong></p><p><strong>Supplementary information: </strong>The online version contains supplementary material available at 10.1007/s40203-025-00464-9.</p>","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"173"},"PeriodicalIF":0.0,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12592626/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145484380","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}
<p><p>Colorectal cancer (CRC) remains a leading cause of cancer-related morbidity and mortality globally. Even with significant advances in the synthesis of chemically based anti-cancer drugs, the unfavorable prognosis of the disease continues to pose a significant challenge. The most frequently encountered limitation of standard cancer treatments is drug resistance. Furthermore, newer target-specific therapies such as immunotherapy and stem cell therapy are costly enough and beyond the reach of most patients. Natural products, being affordable and less toxic, offer multi-targeted strategies to overcome drug resistance and improve therapeutic outcomes. Therefore, this study aims to investigate the therapeutic potential of glycoside compounds derived from <i>Moringa oleifera</i>: Niazirinin (NZR), Niazimicin A (NZA), 4-(Rhamnosyloxy) phenylacetonitrile (RPA), and Moringyne (MRG) in the context of CRC. An integrative network pharmacology strategy was used with the help of multiple databases and web tools. Glycoside compounds derived from <i>Moringa oleifera</i> were selected from the IMPPAT 2.0 database. Compound-target prediction was performed with Super Pred and STRING, while colorectal cancer (CRC)-associated genes were retrieved from the Gene Cards database. Overlapping genes between the compounds and CRC were determined with Venny 2.1. Protein-protein interaction (PPI) enrichment and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were carried out with the STRING database (v12.0). Network building and identification of hub genes were carried out in Cytoscape (v3.10.3) and Cyto Hubba plugin respectively. Validation of the hub genes included mRNA expression profiling, overall survival, and tumor stage-specific analysis using GEPIA 2 webserver. Lastly, molecular docking of <i>Moringa oleifera</i> glycosides with the key hub CRC proteins was performed using the CB-Dock2 web server. Eleven hub genes (NFKB1, PIK3R1, PIK3CD, PIK3CB, CHUK, GRB2, NOS2, SLC2A1, ABL1, PDGFRA, and STAT1) were identified. Five major pathways, PI3K-Akt, cAMP, Ras, HIF-1 signaling, and MicroRNAs in cancer were highly enriched. Of the eleven hub genes, six (PIK3R1, NOS2, SLC2A1, ABL1, PDGFRA, and STAT1) were significantly dysregulated in colon (COAD) and rectal (READ) cancer tissues. In particular, NOS2, SLC2A1, and STAT1 were significantly upregulated, and PIK3R1, ABL1, and PDGFRA were significantly down regulated, indicating possible oncogenic and tumor-suppressive functions, respectively. Stage-specific analysis identified that expression of SLC2A1 differed significantly among pathological stages (F = 4.31, <i>p</i> = 0.00531), which warrants its consideration as a stage-specific prognostic biomarker. Molecular docking revealed that NOS2 and SLC2A1 exhibited high-affinity interactions with all <i>Moringa oleifera</i> glycosides, suggesting their potent inhibitory potential against metabolic and inflammatory targets in CRC. NOS2 and
结直肠癌(CRC)仍然是全球癌症相关发病率和死亡率的主要原因。即使在化学抗癌药物合成方面取得了重大进展,但该疾病的不良预后仍然构成重大挑战。标准癌症治疗最常见的限制是耐药性。此外,新的靶向特异性疗法,如免疫疗法和干细胞疗法,价格昂贵,大多数患者无法承受。天然产品价格合理且毒性较低,可提供多目标策略来克服耐药性并改善治疗结果。因此,本研究旨在探讨辣木苷类化合物Niazimicin (NZR)、Niazimicin A (NZA)、4-(鼠李糖氧基)苯乙腈(RPA)和Moringyne (MRG)在结直肠癌中的治疗潜力。在多个数据库和网络工具的帮助下,采用了综合网络药理学策略。从IMPPAT 2.0数据库中选择辣木苷类化合物。使用Super Pred和STRING进行化合物靶标预测,而从Gene Cards数据库检索结直肠癌(CRC)相关基因。用Venny 2.1检测化合物与结直肠癌之间的重叠基因。利用STRING数据库(v12.0)进行蛋白质-蛋白质相互作用(PPI)富集和基因本体(GO)和京都基因与基因组百科全书(KEGG)途径富集。在Cytoscape (v3.10.3)和Cyto Hubba插件中分别进行网络构建和枢纽基因的鉴定。中心基因的验证包括mRNA表达谱、总生存率和使用GEPIA 2网络服务器的肿瘤分期特异性分析。最后,利用CB-Dock2 web服务器进行辣木苷与关键枢纽CRC蛋白的分子对接。鉴定出11个枢纽基因(NFKB1、PIK3R1、PIK3CD、PIK3CB、CHUK、GRB2、NOS2、SLC2A1、ABL1、PDGFRA和STAT1)。肿瘤中PI3K-Akt、cAMP、Ras、HIF-1信号通路和microrna等5条主要通路均高度富集。在11个枢纽基因中,6个(PIK3R1、NOS2、SLC2A1、ABL1、PDGFRA和STAT1)在结肠(COAD)和直肠(READ)癌组织中显著失调。其中NOS2、SLC2A1和STAT1显著上调,PIK3R1、ABL1和PDGFRA显著下调,分别提示可能具有致癌和抑瘤功能。分期特异性分析发现,SLC2A1的表达在病理分期之间存在显著差异(F = 4.31, p = 0.00531),这证明了其作为分期特异性预后生物标志物的可行性。分子对接发现,NOS2和SLC2A1与辣木苷均表现出高亲和力相互作用,提示其对结直肠癌代谢和炎症靶点具有强大的抑制潜力。NOS2和SLC2A1是CRC中上调的关键枢纽基因,与所有选择的辣木苷具有很强的结合亲和性,显示出其治疗潜力。值得注意的是,SLC2A1也出现了分期特异性表达,突出了其作为分期特异性预后生物标志物的潜力。此外,PDGFRA,一个下调的枢纽基因,表现出强烈的相互作用,并可能作为这些糖苷调节的肿瘤抑制靶点。图片摘要:补充资料:在线版本包含补充资料,网址为10.1007/s40203-025-00461-y。
{"title":"Integrated network pharmacology and molecular docking reveal therapeutic potential of <i>Moringa oleifera</i> glycosides, targeting key regulatory genes in colorectal cancer.","authors":"Anupam Sharma, Abhinav Sharma, Devinder Kumar Maheshwari, Sunil Kumar, Anil Kumar Sharma","doi":"10.1007/s40203-025-00461-y","DOIUrl":"10.1007/s40203-025-00461-y","url":null,"abstract":"<p><p>Colorectal cancer (CRC) remains a leading cause of cancer-related morbidity and mortality globally. Even with significant advances in the synthesis of chemically based anti-cancer drugs, the unfavorable prognosis of the disease continues to pose a significant challenge. The most frequently encountered limitation of standard cancer treatments is drug resistance. Furthermore, newer target-specific therapies such as immunotherapy and stem cell therapy are costly enough and beyond the reach of most patients. Natural products, being affordable and less toxic, offer multi-targeted strategies to overcome drug resistance and improve therapeutic outcomes. Therefore, this study aims to investigate the therapeutic potential of glycoside compounds derived from <i>Moringa oleifera</i>: Niazirinin (NZR), Niazimicin A (NZA), 4-(Rhamnosyloxy) phenylacetonitrile (RPA), and Moringyne (MRG) in the context of CRC. An integrative network pharmacology strategy was used with the help of multiple databases and web tools. Glycoside compounds derived from <i>Moringa oleifera</i> were selected from the IMPPAT 2.0 database. Compound-target prediction was performed with Super Pred and STRING, while colorectal cancer (CRC)-associated genes were retrieved from the Gene Cards database. Overlapping genes between the compounds and CRC were determined with Venny 2.1. Protein-protein interaction (PPI) enrichment and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were carried out with the STRING database (v12.0). Network building and identification of hub genes were carried out in Cytoscape (v3.10.3) and Cyto Hubba plugin respectively. Validation of the hub genes included mRNA expression profiling, overall survival, and tumor stage-specific analysis using GEPIA 2 webserver. Lastly, molecular docking of <i>Moringa oleifera</i> glycosides with the key hub CRC proteins was performed using the CB-Dock2 web server. Eleven hub genes (NFKB1, PIK3R1, PIK3CD, PIK3CB, CHUK, GRB2, NOS2, SLC2A1, ABL1, PDGFRA, and STAT1) were identified. Five major pathways, PI3K-Akt, cAMP, Ras, HIF-1 signaling, and MicroRNAs in cancer were highly enriched. Of the eleven hub genes, six (PIK3R1, NOS2, SLC2A1, ABL1, PDGFRA, and STAT1) were significantly dysregulated in colon (COAD) and rectal (READ) cancer tissues. In particular, NOS2, SLC2A1, and STAT1 were significantly upregulated, and PIK3R1, ABL1, and PDGFRA were significantly down regulated, indicating possible oncogenic and tumor-suppressive functions, respectively. Stage-specific analysis identified that expression of SLC2A1 differed significantly among pathological stages (F = 4.31, <i>p</i> = 0.00531), which warrants its consideration as a stage-specific prognostic biomarker. Molecular docking revealed that NOS2 and SLC2A1 exhibited high-affinity interactions with all <i>Moringa oleifera</i> glycosides, suggesting their potent inhibitory potential against metabolic and inflammatory targets in CRC. NOS2 and ","PeriodicalId":94038,"journal":{"name":"In silico pharmacology","volume":"13 3","pages":"166"},"PeriodicalIF":0.0,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12592598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145484328","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}