1
|
Saeed M, Alamri MA, Rashid MAR, Javed MR, Azeem F, Bashir Z, Alanzi AR, Muhseen ZT, Almusallam SY, Hussain K. Identification of novel inhibitors against VP40 protein of Marburg virus by integrating molecular modeling and dynamics approaches. J Biomol Struct Dyn 2025; 43:3942-3955. [PMID: 38178383 DOI: 10.1080/07391102.2023.2300134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Marburg virus (MV) is a highly etiological agent of haemorrhagic fever in humans and has spread across the world. Its outbreaks caused a 23-90% human death rate. However, there are currently no authorized preventive or curative measures yet. VP40 is the MV matrix protein, which builds protein shell underneath the viral envelope and confers hallmark filamentous. VP40 alone is able to induce assembly and budding of filamentous virus-like particles (VLPs), which resemble authentic virions. As a result, this research is credited with clarifying the function of VP40 and leading to the discovery of new therapeutic targets effective in combating MV disease (MVD). Virtual screening, molecular docking and molecular dynamics (MD) simulation were used to find the putative active chemicals based on a 3D pharmacophore model of the protein's active site cavity. Initially, andrographidine-C, a potent inhibitor was selected for the development of the pharmacophore model. Later, a library of 30,000 compounds along with the andrographidine-C was docked against VP40 protein. Three best hits including avanafil, diuvaretin and macrourone were subjected to further MD simulation analysis, as these compounds had better binding affinities as compared to andrographidine-C. Furthermore, throughout the 100 ns simulations, the back bone of VP40 protein in presence of avanafil, diuvaretin and macrourone remained stable which was further validated by MM-PBSA analysis. Additionally, all of these compounds depict maximum drug-like properties. The predicted drugs based on the ligand, avanafil, diuvaretin and macrourone could be exploited and developed as an alternative or complementary therapy for the treatment of MVD.
Collapse
Affiliation(s)
- Muhammad Saeed
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Mubarak A Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Muhammad Rizwan Javed
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farrukh Azeem
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zarmina Bashir
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Abdullah R Alanzi
- Department of Pharmacogonsy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | - Shahad Youseff Almusallam
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Khadim Hussain
- Plant Protection Department, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Joshi CP, Baldi A, Kumar N, Pradhan J. Harnessing network pharmacology in drug discovery: an integrated approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4689-4703. [PMID: 39621088 DOI: 10.1007/s00210-024-03625-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 04/11/2025]
Abstract
Traditional drug discovery approach is based on one drug-one target, that is associated with very lengthy timelines, high costs and very low success rates. Network pharmacology (NP) is a novel method of drug designing, that is based on a multiple-target approach. NP integrates systems such as biology, pharmacology and computational techniques to address the limitations of traditional methods of drug discovery. With help of mapping biological networks, it provides deep insights into biological molecules' interactions and enhances our understanding to the mechanism of drugs, polypharmacology and disease etiology. This review explores the theoretical framework of network pharmacology, discussing the principles and methodologies that enable the construction of drug-target and disease-gene networks. It highlights how data mining, bioinformatics tools and computational models are utilised to predict drug behaviour, repurpose existing drugs and identify novel therapeutic targets. Applications of network pharmacology in the treatment of complex diseases-such as cancer, neurodegenerative disorders, cardiovascular diseases and infectious diseases-are extensively covered, demonstrating its potential to identify multi-target drugs for multifaceted disease mechanisms. Despite the promising results, NP faces challenges due to incomplete and quality of biological data, computational complexities and biological system redundancy. It also faces regulatory challenges in drug approval, demanding revision in regulatory guidelines towards multi-target therapies. Advancements in AI and machine learning, dynamic network modelling and global collaboration can further enhance the efficacy of network pharmacology. This integrative approach has the potential to revolutionise drug discovery, offering new solutions for personalised medicine, drug repurposing and tackling the complexities of modern diseases.
Collapse
Affiliation(s)
- Chandra Prakash Joshi
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Ashish Baldi
- Pharma Innovation Lab, Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India.
| | - Neeraj Kumar
- B N College of Pharmacy, B. N. University, Udaipur, Rajasthan, India
| | - Joohee Pradhan
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan, India.
| |
Collapse
|
3
|
Priyamvada P, Ashok G, Joshi T, Anbarasu S, Anbarasu A, Ramaiah S. Unravelling the molecular mechanistic pathway underlying the anticancer effects of kaempferol in colorectal cancer: a reverse pharmacology network approach. Mol Divers 2025; 29:1049-1067. [PMID: 38795259 DOI: 10.1007/s11030-024-10890-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/30/2024] [Indexed: 05/27/2024]
Abstract
Colorectal cancer (CRC) is the third most diagnosed and highly fatal malignancy, presenting serious health concerns worldwide. The search for an effective cure for CRC is challenging and poses a serious concern. Kaempferol is a potent anti-cancerous bioactive compound often suggested for treating various cancers, including CRC. However, its underlying molecular mechanism against CRC remains unclear. The present study delves into kaempferol's molecular pathways and underlying molecular mechanisms against CRC targets. The target protein-coding genes for kaempferol were retrieved, and the CRC-associated genes were curated. Twelve common targets with a disease specificity index of > 0.6 were validated for their protein expression at different stages of CRC. Over-expressed USP1, SETD7, POLH, TDP1 and RACGAP1 were selected for further studies. The binding affinities of kaempferol to the corresponding proteins were evaluated using molecular docking and Molecular Dynamics (MD) simulations. SETD7 exhibited the highest binding affinity with the lowest binding energy (- 8.06 kcal/mol). Additionally, the MD simulation, and MM-PBSA conferred SETD7-kaempferol complex had the least root-mean-square deviation with lower interaction energy and higher conformational stability. The protein-protein interaction of SETD7 constructed revealed direct interactors, namely, DNMT1, FOXO1, FOXO3, FOXO4, H3-3B, H3-4, H3C12, H3C13, SETD7, SIRT1 and TP53, have a potential role in cancer progression through FOXO signalling. In summary, our study revealed kaempferol's multi-target and synergistic effect on multiple CRC targets and its underlying mechanisms. Finally, the study recommends in-vitro and in-vivo trials for validation of anti-cancerous drugs for CRC.
Collapse
Affiliation(s)
- P Priyamvada
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Gayathri Ashok
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Tushar Joshi
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Suvitha Anbarasu
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India.
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India.
| |
Collapse
|
4
|
Anshori I, Marcius D, Syaifie PH, Siregar KAAK, Syakuran LA, Jauhar MM, Arda AG, Shalannanda W, Mardliyati E. Therapeutic Potential of Propolis Extract in Managing Hyperinflammation and Long COVID-19: A Bioinformatics Study. Chem Biodivers 2025; 22:e202401947. [PMID: 39576127 DOI: 10.1002/cbdv.202401947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024]
Abstract
Hyperinflammation is a significant factor in long COVID, impacting over 65 million post-COVID-19 individuals globally. Herbal remedies, including propolis, show promise in reducing severity and pro-inflammatory cytokines. However, the natural pharmacological role of propolis in COVID-19 management remains underexplored. Employing network pharmacology and in silico techniques, we assessed propolis extract's potential in countering SARS-CoV-2-induced inflammation. We identified 80 flavonoids via LC-MS/MS QTOF and employed 11 anti-inflammatory drugs as references for inflammation target fishing. Utilizing in silico techniques encompassing target fishing, molecular docking, and dynamics, we examined propolis' effects. We identified 1105 gene targets connected to inflammation through multiple validated target predictors. By integrating SARS-CoV-2 DEGs from GSE147507 with these targets, we identify 25 inflammation-COVID-19-associated propolis targets, including STAT1, NOS2, CFB, EIF2K2, NPY5R, and BTK. Enrichment analyses highlighted primary pharmacological pathways related to Epstein-Barr virus infection and COVID-19. Molecular docking validated isokaempferide, iristectorigenin B, 3'-methoxypuerarin, cosmosiin, and baicalein-7-O-β-d-glucopyranoside, which exhibited strong binding affinity and stability with relevant genes. Moreover, our findings indicate that propolis ligands could potentially suppress reactivation of Epstein-Barr Virus infections in post-COVID-19 cases. However, this study has a limitation in that the concentration of each propolis compound has not been quantified. Therefore, further exploration of propolis compounds quantification and experimental validation are needed to support these findings.
Collapse
Affiliation(s)
- Isa Anshori
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
- Research Center for Nanosciences and Nanotechnology (RCNN), Bandung Institute of Technology, Bandung, Indonesia
| | - Donny Marcius
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| | - Putri Hawa Syaifie
- Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
| | - Khalish Arsy Al Khairy Siregar
- Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
- Faculty of Pharmacy, Universitas Muhammadiyah Kalimantan Timur, Samarinda, East Borneo, Indonesia
| | | | | | | | - Wervyan Shalannanda
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| | - Etik Mardliyati
- Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
- Research Center for Vaccine and Drugs, National Research and Innovation Agency (BRIN), Cibinong, Indonesia
| |
Collapse
|
5
|
Chen Q, Chen G, Wang Q. Application of Network Pharmacology in the Treatment of Neurodegenerative Diseases with Traditional Chinese Medicine. PLANTA MEDICA 2025; 91:226-237. [PMID: 39778593 PMCID: PMC12021452 DOI: 10.1055/a-2512-8928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025]
Abstract
In recent years, the incidence of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, has exhibited a steadily rising trend, which has posed a major challenge to the global public health. Traditional Chinese medicine, with its multicomponent and multitarget characteristics, offers a promising approach to treating neurodegenerative diseases. However, comprehensively elucidating the complex mechanisms underlying traditional Chinese medicine formulations remains challenging. As an emerging systems biology method, network pharmacology has provided a vital tool for revealing the multitarget mechanisms of traditional Chinese medicine through high-throughput technologies, molecular docking, and network analysis. This paper reviews the advancements in the application of network pharmacology in treating neurodegenerative diseases using traditional Chinese medicine, analyzes the current status of relevant databases and technological methods, discusses the limitations, and proposes future directions to promote the modernization of traditional Chinese medicine and the development of precision medicine.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Pharmacy, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guanghui Chen
- Department of Pharmacy, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Qianyan Wang
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
6
|
Siregar KAAK, Syaifie PH, Jauhar MM, Arda AG, Rochman NT, Kustiawan PM, Mardliyati E. Revealing curcumin therapeutic targets on SRC, PPARG, MAPK8 and HSP90 as liver cirrhosis therapy based on comprehensive bioinformatic study. J Biomol Struct Dyn 2025; 43:3172-3189. [PMID: 38217310 DOI: 10.1080/07391102.2023.2301534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/09/2023] [Indexed: 01/15/2024]
Abstract
Cirrhosis naturally progresses through three stages: compensated, decompensated, and late decompensated, which carry an elevated risk of death. Although curcumin's anti-cirrhosis effects have been studied, underlying mechanism in preventing cirrhosis progression and the correlation between curcumin's action with upregulated genes remains insufficiently explored. In this study, we employed network pharmacology approach to construct a drug-target-disease network through bioinformatics and validate the findings with molecular docking and dynamic simulation. The curcumin-targeted liver cirrhosis network encompassed 54 nodes with 282 edges in protein-protein interactions (PPI) network. By utilizing network centrality analysis, we identified eight crucial genes. KEGG enrichment pathway revealed that these crucial genes are involved in pathway of cancer, endocrine resistance, estrogen signaling, chemical carcinogenesis-receptor activation, lipid metabolism, and atherosclerosis. Notably, these eight genes predominantly participate in cancer-related pathways. Further investigation revealed upregulation of four genes and downregulation of four others in hepatocellular carcinoma patients. These upregulated genes-MAPK8, SRC, PPARG, and HSP90AA1-strongly correlated with reduced survival probability in liver hepatocellular carcinoma patients with survival times approximately under 4000 days (∼11 years). Molecular docking and molecular dynamic results exhibited curcumin's superior binding affinities and stability compared to native ligands of MAPK8, SRC, PPARG, and HSP90AA1 within 50 ns simulations. Moreover, MM-GBSA analysis showed stronger binding energy of curcumin to MAPK8, SRC, and HSP90AA1 than native ligand. In conclusion, this study provides valuable insights into curcumin's potential mechanisms in preventing liver cirrhosis progression, specifically in HCC. These findings offer a theoretical basis for further pharmacological research into anti-HCC effect of curcumin.
Collapse
Affiliation(s)
- Khalish Arsy Al Khairy Siregar
- Faculty of Pharmacy, Universitas Muhammadiyah Kalimantan Timur, Samarinda, Indonesia
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
| | - Putri Hawa Syaifie
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
| | | | - Adzani Gaisani Arda
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
| | - Nurul Taufiqu Rochman
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
- Research Center for Advanced Material, National Research and Innovation Agency (BRIN), South Tangerang, Indonesia
| | | | - Etik Mardliyati
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, Indonesia
- Research Center for Vaccine and Drug, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| |
Collapse
|
7
|
Zubair A, Zaib S, Zhu M, Mohany M. Elemental Profiling of Common Anti-diabetic Medicinal Plants of Swat and Peshawar Districts of Khyber Pakhtunkhwa (KPK) Province of Pakistan: An Investigation Using PIXE and ICP-OES. Biol Trace Elem Res 2025; 203:2072-2083. [PMID: 39037503 DOI: 10.1007/s12011-024-04305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/07/2024] [Indexed: 07/23/2024]
Abstract
The use of medicinal plants is integral to addressing liver, heart, lung, and other metabolic issues. These plants are rich in vitamins, minerals, flavonoids, and alkaloids, which collectively help in lowering intestinal glucose absorption and increasing insulin secretion by pancreatic tissues. Elemental analysis, encompassing major, minor, and trace elements, was performed on various parts (leaves, roots, and seeds) of 16 anti-diabetic medicinal plants collected from different regions of Swat and Peshawar in Khyber Pakhtunkhwa (KPK), Pakistan. This analysis utilized proton-induced X-ray emission (PIXE) and inductively coupled plasma optical emission spectroscopy (ICP-OES) techniques. Our PIXE and ICP-OES analysis revealed the presence of major (Ca, K, S, P), minor (Si, Cl), and trace (Al, Mn, Fe, Ni, Cu, Zn, Se, Cr, and Sc) elements in various parts (leaves, roots, and seeds) of the 16 anti-diabetic medicinal plants studied. Specifically, elements such as Ca, K, Cr, Cu, Mn, Zn, and Se were detected, all of which are known to contribute in maintaining normal glucose metabolism. Notably, Zn and Se are crucial trace elements for the synthesis, secretion, and action of insulin. Significant Zn concentrations were observed in ten anti-diabetic medicinal plants: Albizia lebbeck (AL), Atropa acuminata (AA), Avena fatua (AF), Citrus medica (CM), Commiphora wightii (CW), Cymbopogon citratus (CC), Daucus carota (DC), Ziziphus mauritiana (FM), Hyoscyamus niger (HN), and Martynia annua (MA), and significant Se concentrations were observed in twelve medicinal plants, i.e., Albizia lebbeck (AL), Allium sativum (AS), Atropa acuminata (AA), Avena fatua (AF), Cannabis sativa (CS), Capparis spinosa (CaS), Commiphora wightii (CW), Cymbopogon citratus (CC), Datura alba (DA), Daucus carota (DC), Ziziphus mauritiana (FM), and Hyoscyamus niger (HN). Our study's elemental analysis using PIXE and ICP-OES on various parts of 16 medicinal plants identified a significant number of useful elements. Elements such as Ca, K, S, P, Al, Si, Cl, Mn, Fe, Ni, Cu, Zn, Se, and Cr were identified and quantified. These findings support the potential use of these plants in managing diabetes and highlight the importance of elemental profiling in understanding their therapeutic properties.
Collapse
Affiliation(s)
- Akmal Zubair
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, 45320, Pakistan.
| | - Sania Zaib
- Department of Biological Sciences, Faculty of Sciences, International Islamic University, Islamabad, 44000, Pakistan
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Mingkun Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Zheng G, Cao X, Jing Y, Wang L, Yan R, Ji Y, Zhang Y, Li H, Wang Y, Shi Y, Yu Y, Xiong Q. An integrative approach for mechanistic insights into the atherosclerotic plaque-stabilizing properties of Danggui Buxue decoction. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119450. [PMID: 39933661 DOI: 10.1016/j.jep.2025.119450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danggui Buxue Decoction (DBD), a traditional Chinese medicinal formula, has historically been used for cardiovascular health, including managing atherosclerotic plaques (ASP). However, its precise mechanisms remain elusive. AIM OF THE STUDY The purpose of this study was to use a novel integrative bioinformatics analysis and experimental validation approach to provide a molecular basis for ASP's stabilization by DBD. MATERIALS AND METHODS A mice model of ApoE-deficient atherosclerosis fed with a high-fat diet was employed to evaluate the efficacy of DBD in stabilizing ASP. The potential mechanism underlying the stabilization effect of DBD on ASP was systematically investigated using an integrated approach combining network pharmacology, molecular docking, and molecular dynamics simulation. Additionally, an ox-LDL-induced macrophage foam cell model and multivariate statistical analysis were utilized to validate the pharmacodynamic material basis and target of DBD in stabilizing ASP. RESULTS Firstly, it was found that DBD can significantly alleviate ASP, which was manifested as a significant reduction in the atherosclerosis index, ratio of area for plaque to lumen, and vulnerability index. Afterwards, network pharmacology investigation identified quercetin and kaempferol as the primary active compounds in DBD anti-ASP. Key core targets mainly involved TP53, AKT1, IL-6 and TNF. The main action pathways included lipid and atherosclerosis, PI3K-Akt signaling, and other pathways. Subsequently, molecular docking and molecular dynamics simulation results confirmed the strong stability of the main active compounds with the key target. Finally, the cell validation experiment in vitro revealed that both quercetin and kaempferol could significantly inhibit RAW264.7 macrophage foaming formation induced by ox-LDL and improve its lipid metabolism disorder. Meanwhile, they could also significantly reverse ox-LDL induced abnormal expression of core protein predicted by network pharmacology in RAW264.7 foam cells. Further correlation analysis revealed that the improvement effect of quercetin and kaempferol on macrophage foaming had a close correlation with the inhibition of core protein expression. CONCLUSION DBD mainly utilized active ingredients such as quercetin and kaempferol, through regulating multiple targets like TP53, AKT1, IL-6 and TNF, to stabilize ASP.
Collapse
Affiliation(s)
- Guangzhen Zheng
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Xiangyang Cao
- Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223003, Jiangsu, PR China.
| | - Yi Jing
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Ling Wang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Ruixue Yan
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Yan Ji
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Yuhan Zhang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Heng Li
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Yunpeng Wang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Yingying Shi
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China
| | - Yadong Yu
- Department of Neurology, Lianshui County People's Hospital, Huai'an, 223400, Jiangsu, PR China.
| | - Qingping Xiong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, PR China.
| |
Collapse
|
9
|
Akter S, Ahmad SU, Bhuiyan MA, Dewan I, Reza R, Morshed N, Samdani MN, Reza MS, Kumer A, Naina Mohamed I. Network Pharmacology, Molecular Docking and Experimental Validation on Potential Application of Diabetic Wound Healing of Cinnamomum zeylanicum Through Matrix Metalloproteinases-8 And 9 (MMP-8 And MMP-9). Drug Des Devel Ther 2025; 19:1753-1782. [PMID: 40093644 PMCID: PMC11910940 DOI: 10.2147/dddt.s489113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/08/2025] [Indexed: 03/19/2025] Open
Abstract
Background Diabetic wounds are a significant clinical challenge due to impaired healing processes often exacerbated by elevated matrix metalloproteinases (MMPs). Cinnamomum zeylanicum, known for its anti-inflammatory and antioxidant properties, has shown potential in promoting wound healing. This study investigates the molecular docking and experimental validation of Cinnamomum zeylanicum's effects on diabetic wound healing, focusing on its interaction with matrix metalloproteinases-8 (MMP-8) and 9 (MMP-9). Methods Molecular docking studies were performed to predict the binding affinity of Cinnamomum zeylanicum compounds to MMP-8 and MMP-9. Diabetic wound healing was evaluated using in vivo models where wounds were induced and treated with Cinnamomum zeylanicum extract. Various parameters were measured, including wound contraction, hydroxyproline content, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and malondialdehyde (MDA) levels. Biochemical analyses included glucose levels, fasting blood glucose (FBG), oral glucose tolerance test (OGTT), and histomorphological examination of skin tissues. Results Molecular docking results indicated a high binding affinity of Cinnamomum zeylanicum's bioactive compounds with MMP-8 and MMP-9, suggesting potential inhibition. Experimental validation showed significant improvement in wound contraction and increased hydroxyproline content, indicating enhanced collagen synthesis. Antioxidant enzyme activities (SOD, GPx, CAT) were significantly elevated, while MDA levels were reduced, reflecting decreased oxidative stress. Biochemical analysis demonstrated improved glucose homeostasis with reduced FBG and enhanced OGTT responses. Histomorphological studies revealed improved tissue architecture and re-epithelialization in treated wounds. Conclusion Cinnamomum zeylanicum exhibits promising potential in diabetic wound healing by modulating MMP-8 and MMP-9 activities, enhancing antioxidant defenses, and improving glucose regulation. These findings support its therapeutic application for diabetic wounds, providing a foundation for further clinical investigations.
Collapse
Affiliation(s)
- Sharmin Akter
- Department of Pharmacy, School of Medicine, University of Asia Pacific, Dhaka, 1215, Bangladesh
| | - Shihab Uddin Ahmad
- Department of Pharmacy, School of Medicine, University of Asia Pacific, Dhaka, 1215, Bangladesh
| | - Mohiuddin Ahmed Bhuiyan
- Department of Pharmacy, School of Medicine, University of Asia Pacific, Dhaka, 1215, Bangladesh
| | - Irin Dewan
- Department of Pharmacy, School of Medicine, University of Asia Pacific, Dhaka, 1215, Bangladesh
| | - Rumman Reza
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Niaz Morshed
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Md Nazmus Samdani
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Md Selim Reza
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Ajoy Kumer
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
- Department of Chemistry, College of Arts and Sciences, International University of Business Agriculture and Technology, Dhaka, 1230, Bangladesh
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, 56000, Malaysia
| |
Collapse
|
10
|
Mohanty D, Padhee S, Priyadarshini A, Kerry RG, Dash B, Sahoo A, Jena S, Panda PC, Khan HA, Nayak S, Ray A. Integrative approach to decipher pharmacological mechanism of Cinnamomum zeylanicum essential oil in prostate cancer. Med Oncol 2025; 42:100. [PMID: 40072751 DOI: 10.1007/s12032-025-02665-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Prostate cancer has garnered much importance in recent years due to its rising incidence and mortality among men worldwide. The ineffectiveness of existing therapies and adverse events associated with conventional treatment have led patients to turn towards traditional medicine for the management of prostate cancer. Cinnamomum zeylanicum bark essential oil (CZEO) possesses promising anticancer properties, yet the exact mechanism of action of CZEO for the management of prostate cancer remains unclear. Therefore, the current study tried to elucidate the bioactive components and key potential targets through which CZEO may exert its anticancer effect for treating prostate cancer. Fifty-nine constituents were identified by GC-MS, of which 52 were drug-like constituents. A total of 2847 targets related to CZEO and 2283 targets related to prostate cancer were obtained from public databases and the GEO dataset. Twenty-three overlapping targets exist between CZEO and disease targets. Compound-disease-target network analysis revealed camphor, eugenol, methyl eugenol, trans farnesyl acetate and nerol as the core bioactive ingredients of CZEO. The topological screening of the PPI network revealed BCL2, TNF, NFKBIA, CREBBP and IL7R as potential hub targets. These hub targets were validated based on mRNA expression level, pathological stages, overall survival, immune infiltrate and genetic alteration analysis in prostate adenocarcinoma and normal patients. KEGG enrichment analysis proposed that CZEO exhibits its anticancer effect mainly by modulating the PI3-AKT and MAPK signalling pathway. Moreover, molecular docking and dynamics simulation studies revealed a good binding affinity of these core compounds with TNF, NFKBIA and BCL2. CZEO exhibited a remarkable anti-proliferative effect against PC-3 cells with an IC50 value of 13.56 µg/mL. CZEO promoted apoptosis and cell cycle arrest in the G2/M phase in PC-3 cells. CZEO-induced apoptosis was due to loss of mitochondrial membrane potential, increase in reactive oxygen species levels and activation of caspases (caspase 3, caspase 8 and caspase 9). RT-qPCR analysis revealed that CZEO modulated the mRNA expression level of hub genes (BCL2, TNF, NFKBIA, CREBBP, and IL7R, caspase 3, caspase 8 and caspase 9). The present study provides a mechanistic approach of Cinnamomum zeylanicum essential oil against prostate cancer.
Collapse
Affiliation(s)
- Debajani Mohanty
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Sucheesmita Padhee
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Arpita Priyadarshini
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Rout George Kerry
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Biswabhusan Dash
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Ambika Sahoo
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Sudipta Jena
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Pratap Chandra Panda
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Haseeb Ahmad Khan
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Sanghamitra Nayak
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Asit Ray
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India.
| |
Collapse
|
11
|
Jadhav PA, Thomas AB, Pathan MK, Chaudhari SY, Wavhale RD, Chitlange SS. Unlocking the therapeutic potential of unexplored phytocompounds as hepatoprotective agents through integration of network pharmacology and in-silico analysis. Sci Rep 2025; 15:8425. [PMID: 40069278 PMCID: PMC11897136 DOI: 10.1038/s41598-025-92868-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Liver diseases account for over two million deaths annually, amounting to 4% of mortality worldwide, underscoring the need for development of novel preventive and therapeutic strategies. The growing interest in natural hepatoprotective agents highlights the potential of traditional medicine for modern drug discovery, though unlocking their molecular complexity requires advanced tools. This study integrates cutting-edge computational techniques with traditional herbal knowledge to identify potential hepatoprotective compounds. Protein targets implicated in liver disorders were identified through network pharmacology and by leveraging the rich molecular diversity inherent in herbal compounds, phytocompounds were selected. The Gene Ontology, Kyoto Encyclopedia of Genes and Genome data were compiled and enrichment analysis was performed using the DAVID database. Molecular docking of selected phytocompounds with top five protein targets helped identify 14 compounds which were employed for building the pharmacophore model. In virtual screening, among 1089 compounds screened, 10 compounds were identified as potential hits based on their predicted scores and alignment with pharmacophore features. The interactions of resulting hits were then analyzed through redocking studies and validated through molecular dynamics simulation and ADMET studies. Notably, (2S,5E)-2-(3,4-Dihydroxybenzyl)-6-(3,4-dihydroxyphenyl)-4-oxo-5-hexenoic acid and 5'-hydroxymorin emerged as lead compounds for further investigation. Both compounds exhibited significant binding affinities with specific amino acids in selected targets, suggesting their potential to modulate key pathways involved in hepatic disorders. Our findings demonstrate the utility of this integrated approach which transits beyond traditional trial-and-error methods. This approach will accelerate the discovery of novel hepatoprotective compounds, providing deeper insights into their mechanistic pathways and action.
Collapse
Affiliation(s)
- Pranali A Jadhav
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India
| | - Asha B Thomas
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India.
| | | | - Somdatta Y Chaudhari
- Department of Pharmaceutical Chemistry, PES's Modern College of Pharmacy, Nigdi, Pune, Maharashtra, India
| | - Ravindra D Wavhale
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India
| | - Sohan S Chitlange
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India
| |
Collapse
|
12
|
Xu QH, Wang YL, Wang C, Jiang SS, Zhang BR, Tian J. Exploring the active ingredients and potential mechanisms of Pingchan granules in Parkinson's disease treatment through network pharmacology and transcriptomics. Sci Rep 2025; 15:7847. [PMID: 40050654 PMCID: PMC11885611 DOI: 10.1038/s41598-025-91344-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Parkinson's disease (PD), the second most prevalent neurodegenerative disorder, poses significant challenges to single-target therapeutic strategies due to its complex etiology. This has driven interest in multi-target approaches, particularly those leveraging natural compounds. Pingchan granules (PCG), a traditional Chinese medicine composed of plant- and animal-derived compounds, have shown efficacy in alleviating PD symptoms. Here, we identify 96 PCG-associated anti-PD targets, enriched in neuronal synaptic signaling and G protein-coupled receptor pathways. Through protein-protein interaction network analysis of anti-PD targets and random forest modeling of substantia nigra transcriptomic data from PD patients, SLC6A3 and SRC emerged as central hub targets, with Mendelian randomization further validating SRC as a potential therapeutic target. Molecular docking and single-cell sequencing reveal that dauricine, PCG's principal active compound, binds strongly to SLC6A3 and SRC, modulating glucose metabolism pathways in dopaminergic neurons. These findings illuminate the molecular basis of PCG's therapeutic effects, offer a foundation for future drug development, and underscore the potential of dauricine as a targeted treatment for PD.
Collapse
Affiliation(s)
- Qiu-Han Xu
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Yi-Ling Wang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Cheng Wang
- Department of Neurosurgey, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Si-Si Jiang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Bao-Rong Zhang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
| | - Jun Tian
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
13
|
Chintaluri PG, Ganapathy Vilasam Sreekala A, Gupta KK, Sivadasan A, Nathan VK. Network pharmacological evaluation of Cressa cretica L.- an integrated approach of modern and ancient pharmacology. J Biomol Struct Dyn 2025:1-16. [PMID: 40035562 DOI: 10.1080/07391102.2025.2472403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/24/2024] [Indexed: 03/05/2025]
Abstract
Cressa cretica L. is immensely valuable in pharmacology. Computational approach through network pharmacology has been attempted to understand lead molecules of Cressa and their interactions with multiple targets. The phytochemical components of methanolic extracts of Cressa leaves were identified using GC-MS analysis, revealing 16 compounds. Using the identified lead molecules, target proteins were predicted using SWISS-target prediction and were analyzed using Cytoscape. This led to the identification of 56 candidate protein targets, which were used to construct a network using CytoHubba, Centiscape, MCODE, and KEGG pathways. The STRING network was created using Cytoscape for analyzing protein-protein interactions, and the top 5 genes were chosen from a total of 12 algorithms in CytoHubba. The antioxidant effects of C. cretica were investigated using 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging activity, which showed an increase in the trend activity of the plant extract with an inhibition percentage of 51.53 ± 0.003%. This was further validated by ferric reducing antioxidant power (FRAP) assay that resulted in an antioxidant activity of 6.64 µg/mL at a high concentration of 500 µg/mL. Molecular docking and simulation were performed to study the interaction of human cyclooxygenase-2 (PDB ID: 5KIR) with Cressa metabolites. 5KIR exhibited a higher interaction with methyl stearate, forming two H-bond interactions with Arg 120 and Tyr 355. Molecular dynamics simulation analysis confirmed the stability of the protein-ligand complex. The network pharmacology analysis of putative proteins obtained from C. cretica revealed that the peroxisome proliferator-activated receptor gamma (PPARG) gene is found in numerous cancer pathways and can be inhibited.
Collapse
Affiliation(s)
- Pratham Gour Chintaluri
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thirumalasamudram, Thanjavur, India
| | | | - Krishna Kant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thirumalasamudram, Thanjavur, India
- National Centre for Cell Science, NCCS Complex, University of Pune Campus, Pune, India
| | - Aparna Sivadasan
- Department of Surgery, Ayurvedic practitioner & Intern, Rashtriya Ayurveda Vidyapeeth, New Delhi, India
| | - Vinod Kumar Nathan
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thirumalasamudram, Thanjavur, India
| |
Collapse
|
14
|
Naseem A, Majeed Khan H, Umar A, Elshikh MS, Aljowaie RM, Gancarz M. Mechanistic insights of methylcinnamate in improving oxidative stress and inflammation in acetaminophen-induced hepatotoxic mice by upregulating Nrf2 pathway. J Pharm Pharmacol 2025; 77:418-429. [PMID: 39851237 DOI: 10.1093/jpp/rgaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025]
Abstract
BACKGROUND Methylcinnamate (MC), a safe flavoring agent naturally found in Occimum basilicum L. is reported to have an anti-inflammatory responses in various disease models. Acetaminophen (APAP) toxicity is a significant contributor to acute liver injury, which leads to oxidative stress and inflammation. The transcriptional factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulated the cellular defense mechanisms aid to antioxidant response facilitation and reduction in inflammation against various disorders. METHODOLOGY This study evaluated the protective effects of MC in APAP-induced hepatotoxicity in mice and its anti-oxidant, anti-inflammatory, and Nrf2 mechanisms were studied. In-vitro 2,2-diphenyl-1-picrylhydrazyl assay showed the antioxidant capacity of MC. Mice were pretreated with MC (25, 50, 75, and 100 mg/kg) orally for 7 days. After a fasting period of 16 h, hepatotoxicity was induced by injecting APAP 300 mg/kg intraperitoneal on day 7. Liver profile, oxidative test, and histopathological changes were studied. Gene expression of interlukin-1β (IL-1β), interlukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), cytochrome P450 2E1 (CYP2E1), Nrf2, and NAD(P)H dehydrogenase (quinone) 1 (NQO-1) were estimated by real time quantitative polymerase chain reaction (RT-qPCR). IL-1β, IL-6, and TNF-α concentrations were also analyzed by enzyme-linked immunosorbent assay (ELISA). RESULTS The MC treatment showed a notable reduction in alanine transaminase, aspartate aminotransferase and alkaline phosphatase activities, and total bilirubin level of serum. Moreover, MC significantly attenuated oxidative stress by rising the antioxidant enzymes catalase, glutathione, and superoxide dismutase and reducing the malondialdehyde and nitric oxide levels in the liver. Furthermore, MC successfully mitigated the levels of IL-1β, IL-6, and TNF-α, which were estimated through RT-qPCR and ELISA. The RT-qPCR revealed a CYP2E1 enzyme inhibition and significant upregulation of hepatic Nrf2 and NQO-1 levels after MC therapy. Histopathological analysis showed improvement in liver injury within the MC treatment groups. CONCLUSION It was concluded from this study that pretreatment of MC had successfully protected the liver through anti-inflammatory, anti-oxidant activity upon subsequent activation of Nrf2.
Collapse
Affiliation(s)
- Afshan Naseem
- Department of Pharmacology, Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore 54000, Pakistan
| | - Humaira Majeed Khan
- Department of Pharmacology, Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore 54000, Pakistan
| | - Aisha Umar
- Institute of Botany, University of the Punjab, Quaid-e-Azam campus, Lahore, 54590, Pakistan
| | - Mohamed S Elshikh
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| | - Reem M Aljowaie
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| | - Marek Gancarz
- Faculty of Production and Power Engineering, University of Agriculture in Krakow, Balicka 116B, 30-149 Krakow, Poland
| |
Collapse
|
15
|
Wang Y, Cai S, Wen W, Tan Y, Wang W, Xu J, Xiong P. A Network Pharmacology Study and In Vitro Evaluation of the Bioactive Compounds of Kadsura coccinea Leaf Extract for the Treatment of Type 2 Diabetes Mellitus. Molecules 2025; 30:1157. [PMID: 40076380 PMCID: PMC11901907 DOI: 10.3390/molecules30051157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Kadsura coccinea is a traditional Chinese medicine whose roots have long been used to treat various ailments, but little is known about the efficacy of its leaves. In this study, the antidiabetic activity of K. coccinea leaf extract (KCLE) was determined, the main components of KCLE were identified using UPLC-TOF-MS, and network pharmacology and molecular docking were integrated to elucidate the antidiabetic mechanism of KCLE. The results showed that KCLE effectively increased the glucose consumption of IR-HepG2 cells through pyruvate kinase (PK) and hexokinase (HK), promoted glycogen synthesis, and inhibited α-glucosidase and α-amylase activities. KCLE also improves diabetes by regulating AKT1, TNF, EGFR, and GSK3β. These targets (especially AKT1 and TNF) have a high binding affinity with the main active ingredients of KCLE (rutin, luteolin, demethylwedelolactone, maritimetin, and polydatin). Pathway enrichment analysis showed that the antidiabetic effect of KCLE was closely related to the PI3K-Akt signaling pathway, MAPK signaling pathway, AGE-RAGE signaling pathway, and FoxO signaling pathway. These findings provide a theoretical basis for promoting the pharmacodynamic development of K. coccinea and its application in treating diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ping Xiong
- Department of Pharmaceutical Engineering, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
16
|
Fan Y, Wang Q, Zhang Y, Wang Y, Li W, Jiang S, Duan JN. Mechanism of Guishao Yigong decoction in treating colorectal cancer based on network pharmacology and experimental validation. J Pharm Pharmacol 2025; 77:430-445. [PMID: 39352002 DOI: 10.1093/jpp/rgae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/05/2024] [Indexed: 03/06/2025]
Abstract
OBJECTIVES To explore the effective components of Guishao Yigong decoction (GYD) in the treatment of colorectal cancer and reveal its potential mechanism of action. METHODS Through network pharmacology, the main target and signaling pathway of GYD therapy for colorectal cancer (CRC) were found. Subsequently, the effect of GYD was verified by in vitro cell viability measurements, colony formation, and scratch healing tests. The effects of GYD on metabolic pathways in vivo were found through plasma metabolomics. Finally, flow cytometry and qPCR experiments were used to verify the cycle-blocking effect of GYD on CRC cells. KEY FINDINGS Based on the network pharmacological analysis and molecular docking technology, it was found that GYD could restrain the growth of CRC cells by affecting lipid metabolic pathways and mitogen-activated protein kinase (MAPK) signaling pathways. A series of cell experiments showed that GYD could inhibit the proliferation, migration and clonogenic ability of CRC cells. Furthermore, the plasma metabolomics results showed that GYD could affect the production of unsaturated fatty acids in mice. Flow cytometry and qPCR experiments further proved that GYD blocked the CRC cells in the G1 phase and modulated the expression of cell cycle-related targets, such as AKT, TP53, CDKN1A, and CDK2. CONCLUSIONS All the results indicated that GYD could regulate the related metabolism of unsaturated fatty acids. Thus, the cell cycle was blocked and the expressions of the key proteins such as AKT and TP53 were regulated, which achieved the purpose of intervention in colorectal cancer.
Collapse
Affiliation(s)
- Yuwen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Quyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Wenwen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Ji-Nao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
17
|
Shan G, Jiang J, Ji L, Li S, Wang Z, Yang S, Shen Q. Evaluation of active substances in gamboge and their mechanisms for the treatment of colorectal cancer by UPLC-MS/MS integrated with network pharmacology. Anal Biochem 2025; 698:115747. [PMID: 39647807 DOI: 10.1016/j.ab.2024.115747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 12/10/2024]
Abstract
Gamboge exhibits anti-colorectal cancer (CRC) activity, however, its active compounds and the underlying mechanisms remain unclear. Herein, a liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method for determining gambogellic acid, β-morellic acid, isogambogenic acid, gambogenic acid, R-gambogic acid, S-gambogic acid, and hydroxygambogic acid in gamboge was established. The key parameters including ion transitions, voltages, LOD, and LOQ were determined, with LOD ranging from 0.8 to 2.0 ng mL-1 and LOQ from 2.7 to 6.7 ng mL-1. The recovery rates were found to be between 95.6 % and 103.5 %. Furthermore, the active compounds were successfully determined, and molecular mechanisms of gamboge in treating CRC were explored. Network pharmacology revealed a "compound-target-pathway" network where the seven compounds could target key proteins, modulate PI3K-Akt and JAK-STAT pathways, and inhibit CRC development. Molecular docking validated SRC, SATA3, PIK3CA, among others, as potential targets for the active compounds in CRC intervention. In conclusion, this method significantly reduces analysis time and improves efficiency relative to existing approaches, making it highly suitable for the effective determination of multiple compounds in the quality control of gamboge materials.
Collapse
Affiliation(s)
- Guodong Shan
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiajun Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Liting Ji
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Shiyan Li
- Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Zejun Wang
- Laboratory of Medicine-Food Homology Innovation and Achievement Transformation, Linping Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, 311110, China.
| | - Shaohui Yang
- Department of Colorectal Surgery, Lihuili Hospital, Ningbo Medical Center, Ningbo, 315040, China.
| | - Qing Shen
- Panvascular Diseases Research Center, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China; Laboratory of Food Nutrition and Clinical Research, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310012, China.
| |
Collapse
|
18
|
Rasul HO, Ghafour DD, Aziz BK, Hassan BA, Rashid TA, Kivrak A. Decoding Drug Discovery: Exploring A-to-Z In Silico Methods for Beginners. Appl Biochem Biotechnol 2025; 197:1453-1503. [PMID: 39630336 DOI: 10.1007/s12010-024-05110-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 03/29/2025]
Abstract
The drug development process is a critical challenge in the pharmaceutical industry due to its time-consuming nature and the need to discover new drug potentials to address various ailments. The initial step in drug development, drug target identification, often consumes considerable time. While valid, traditional methods such as in vivo and in vitro approaches are limited in their ability to analyze vast amounts of data efficiently, leading to wasteful outcomes. To expedite and streamline drug development, an increasing reliance on computer-aided drug design (CADD) approaches has merged. These sophisticated in silico methods offer a promising avenue for efficiently identifying viable drug candidates, thus providing pharmaceutical firms with significant opportunities to uncover new prospective drug targets. The main goal of this work is to review in silico methods used in the drug development process with a focus on identifying therapeutic targets linked to specific diseases at the genetic or protein level. This article thoroughly discusses A-to-Z in silico techniques, which are essential for identifying the targets of bioactive compounds and their potential therapeutic effects. This review intends to improve drug discovery processes by illuminating the state of these cutting-edge approaches, thereby maximizing the effectiveness and duration of clinical trials for novel drug target investigation.
Collapse
Affiliation(s)
- Hezha O Rasul
- Department of Pharmaceutical Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq.
| | - Dlzar D Ghafour
- Department of Medical Laboratory Science, College of Science, Komar University of Science and Technology, 46001, Sulaimani, Iraq
- Department of Chemistry, College of Science, University of Sulaimani, 46001, Sulaimani, Iraq
| | - Bakhtyar K Aziz
- Department of Nanoscience and Applied Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Bryar A Hassan
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
- Department of Computer Science, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Tarik A Rashid
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
| | - Arif Kivrak
- Department of Chemistry, Faculty of Sciences and Arts, Eskisehir Osmangazi University, Eskişehir, 26040, Turkey
| |
Collapse
|
19
|
Mangral ZA, Bhat BA, Sheikh S, Islam SU, Tariq L, Dar R, Varadharajan V, Hassan Dar TU. Exploring the therapeutic potential of Rhododendron anthopogon D.Don essential oil constituents against lung cancer: A network pharmacology-based analysis with molecular docking and experimental studies. Comput Biol Med 2025; 187:109827. [PMID: 39933268 DOI: 10.1016/j.compbiomed.2025.109827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/11/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
Rhododendron anthopogon D.Don is an evergreen shrub used by Himalayan healers to treat many ailments most notably lung problems. However, the mechanism by which R. anthopogon essential oil fights lung cancer has not been well studied. Here, in the present study, we used network pharmacology in combination with chemical profiling, molecular docking, and in-vitro experimental studies to uncover the mechanism of R. anthopogon essential oil constituents against lung cancer. By employing network pharmacology-based analysis, a total of 266 potential target genes obtained for 12 active components of R. anthopogon interacted with 260 common targets and 17,731 disease targets associated with lung cancer were retrieved. Using protein-protein interaction network (PPI), search tool for the retrieval of interacting genes/proteins (STRING) and database for annotation, visualization, and integrated discovery (DAVID) databases, we predicted that the main signaling pathways involved in the association of lung cancer with R. anthopogon essential oil constituents are the cancer signaling pathway and vascular endothelial growth factor and its receptor (VEGFR) cancer signalling pathway. Using TIMER 2.0 analysis and University of Alabama Cancer Database (UALCAN) findings, the expression pattern of EGFR was investigated across all TCGA (The cancer genome atlas) datasets. The study revealed that EGFR expression was elevated in various cancers especially in lung adenocarcinoma. Molecular docking analysis revealed that linalool, α-bisabolol, and guaiol possessed strong binding affinity with TNF-α, MAPK3, and EGFR protein drug targets. Our results predicted that TNF-α, MAPK3, and EGFR may be potential molecular targets of R. anthopogon essential oil constituents for the treatment of lung cancer. Furthermore, our study verified that R. anthopogon essential oil constituents inhibit proliferation, and induces apoptosis in lung cancer cell lines. Therefore, the present study highlights anti-lung cancer activity of the constituents of R. anthopogon essential oil and its potential involvement in comprehending therapeutic mechanism that may be applied in the lung cancer therapy.
Collapse
Affiliation(s)
- Zahid Ahmed Mangral
- Department of Biotechnology, School of Biosciences and Biotechnology, BGSB University, Rajouri, Jammu and Kashmir, India
| | - Basharat Ahmad Bhat
- Department of Bio-Resources, Govt. Degree College for Women, Pulwama, J & K, India
| | - Shagufta Sheikh
- Department of Biochemistry, University of Kashmir Srinagar, Jammu and Kashmir, India
| | - Shahid Ul Islam
- Department of Biotechnology, School of Biosciences and Biotechnology, BGSB University, Rajouri, Jammu and Kashmir, India
| | - Lubna Tariq
- Department of Biotechnology, School of Biosciences and Biotechnology, BGSB University, Rajouri, Jammu and Kashmir, India
| | - Rubiya Dar
- Centre of Research for Development, University of Kashmir, Jammu and Kashmir, India
| | | | - Tanvir Ul Hassan Dar
- Department of Biotechnology, School of Biosciences and Biotechnology, BGSB University, Rajouri, Jammu and Kashmir, India.
| |
Collapse
|
20
|
Lyu C, Kang SY, Shao H, Kim D, Jung HW. Ameliorative effects of Asiasarum root and rhizome extract on high fat diet‑induced obesity in mice through regulation of the SIRT1/PGC1α/AMPK pathways in muscle and liver tissues. Mol Med Rep 2025; 31:76. [PMID: 39886968 PMCID: PMC11795245 DOI: 10.3892/mmr.2025.13440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/27/2024] [Indexed: 02/01/2025] Open
Abstract
Asiasarum root and rhizome (Asarum) is commonly used as a diaphoretic. Due to its warm and pungent characteristics in traditional Chinese and Korean medicine, it is considered as having the potential to prevent disease. The present study investigated the effects of Asarum extract on the symptoms of obesity in mice, and the regulation of energy metabolism in the liver and skeletal muscle tissues. In addition, to identify the potential molecular targets and signaling pathways involved in the mechanism of action of Asarum extract in obesity, network pharmacological and molecular docking analysis was performed. In vitro studies demonstrated that Asarum extract significantly increased the expression of regulators of energy metabolism [sirtuin 1 (SIRT1), peroxisome proliferator‑activated receptor γ coactivator 1‑α (PGC1α), nuclear respiratory factor 1, AMP‑activated protein kinase (AMPK) and glucose transporter type 4 (GLUT4)] and myogenic regulatory factors (MyoD, myogenin and myosin heavy chain) in C2C12 myotubes. Furthermore, the in vivo studies demonstrated that Asarum extract could reduce increases in body weight, and the levels of blood glucose, insulin, total cholesterol, triglycerides and low‑density lipoprotein cholesterol in the sera of obese mice. Asarum extract also improved pathological changes in the liver and pancreatic tissues of obese mice, and significantly increased the ratio of brown fat mass to body weight. In addition, Asarum extract reversed the expression of energy metabolism regulators and myogenic regulatory factors in the gastrocnemius tissues of obese mice. Asarum extract also activated the expression of SIRT1, PGC1α and AMPK in the liver tissues of obese mice. These findings indicated that Asarum extract may exert anti‑obesity effects, such as body weight loss, decreases in lipid metabolite levels, and inhibition of pancreatic and liver damage. Using network pharmacological analysis, the mechanisms underlying the effects of Asarum extract on the regulation of energy metabolism were explored, particularly in skeletal muscle and liver tissues.
Collapse
Affiliation(s)
- Chenzi Lyu
- Department of Herbology, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongsangbuk 38066, Republic of Korea
| | - Seok Yong Kang
- Korean Medicine R&D Center, Dongguk University, Gyeongju, Gyeongsangbuk 38066, Republic of Korea
| | - Haifeng Shao
- Department of Herbology, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongsangbuk 38066, Republic of Korea
| | - Dongeun Kim
- Department of Herbology, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongsangbuk 38066, Republic of Korea
| | - Hyo Won Jung
- Department of Herbology, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongsangbuk 38066, Republic of Korea
- Korean Medicine R&D Center, Dongguk University, Gyeongju, Gyeongsangbuk 38066, Republic of Korea
| |
Collapse
|
21
|
Martins-Silva C, Anderson CL, Boyce AKJ, Andrade TES, Tizziani T, Lopes KHS, Micke GA, Cregan SP, Dos Santos ARS, Thompson RJ. The Ethanolic Extract of Polygala paniculata L. Blocks Panx1 Channels and Reduces Ischemic Brain Infarct in a Dose- and Sex-Dependent Way. Mol Neurobiol 2025; 62:3258-3275. [PMID: 39271622 DOI: 10.1007/s12035-024-04453-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Polygala paniculata L. is a native plant from tropical America. The therapeutic potential of the hydroalcoholic extract of P. paniculata (HEPp) has been scientifically explored due to folk medicine reports on its action against several afflictions. HEPp contains several bioactive molecules with neuroprotective activities, making it a promising candidate for stroke treatment. This study used electrophysiological, biochemical, and in vivo experiments to evaluate the molecular mechanisms underlying HEPp as a neuroprotective therapy for stroke targeting Pannexin-1 (Panx1). Panx1 is a non-selective channel that opens during ischemia and contributes to neuronal death. HEPp was not toxic to cortical neurons and pre-treatment with the extract reduced neuronal death promoted by oxygen and glucose deprivation in a dose-dependent manner. Additionally, HEPp blocked Panx1 currents in a dose-dependent manner and the effect, which was shown to be partially due to rutin. Animals submitted to photothrombosis and post-treated with HEPp had reduced infarct volume, and the effective dose was lower in males (1 mg/kg) than in females (10 mg/kg). On the other hand, in Panx1 KD mice (50% Panx1 levels), the acute treatment reduced the infarct volume only in males. Upon chronic treatment with HEPp, a reduction in Panx1 protein levels was observed. The current study provides reliable evidence of the neuroprotective properties of HEPp in both in vitro and in vivo models of stroke. The underlying mechanism involves, at least in part, the inhibition of Panx1 channel function and possibly downregulation of protein levels, suppressing the secondary events that lead to apoptosis and inflammation.
Collapse
Affiliation(s)
- Cristina Martins-Silva
- Department of Physiological Sciences, Health Sciences Center, Laboratory of Neurochemistry and Behaviour (LabNeC), Graduate Program in Biochemistry, Federal University of Espirito Santo, Vitoria, ES, 29043910, Brazil.
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada.
| | - Connor L Anderson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Andrew K J Boyce
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Tassiane E S Andrade
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Neuroscience Program, Department of Physiology and Pharmacology, University of Western Ontario, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, ON, N6A 5K8, Canada
| | - Tiago Tizziani
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Kheytiany H S Lopes
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Gustavo A Micke
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Sean P Cregan
- Neuroscience Program, Department of Physiology and Pharmacology, University of Western Ontario, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, ON, N6A 5K8, Canada
| | - Adair Roberto Soares Dos Santos
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Roger J Thompson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
22
|
Silambarasan R, Kasthuri Nair A, Maniyan G, Vijaya R, Nair RV, Hareendran Nair J, Nishanth Kumar S, Sasidharan S. Exploring the molecular mechanism of Dioscorea alata L. for the treatment of menstrual disorders using network pharmacology and molecular docking. Heliyon 2025; 11:e42582. [PMID: 40028534 PMCID: PMC11870275 DOI: 10.1016/j.heliyon.2025.e42582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
Menstrual disorders (MDs), including premenstrual syndrome, amenorrhea, and dysmenorrhea, affect women globally. Dioscorea alata L., a traditional yam species, has been used medicinally, but its potential in treating MDs remains understudied. This study employs a network pharmacology approach to examine the effects of D. alata's secondary metabolites on MDs via multi-target mechanisms. Compounds were identified from literature and PubChem, while disease-related targets were gathered from GeneCards, DisGeNET, and CTD databases. Swiss target prediction was used to link compounds to targets. A protein-protein interaction (PPI) network was constructed using STRING, and Gene Ontology (GO) and KEGG enrichment analyses were conducted to predict functional pathways. Eighteen bioactive compounds and 120 therapeutic targets specific to MDs were identified. KEGG analysis revealed 20 significant pathways related to menstrual disturbances. Among the 120 targets, TNF α, PPARG, ESR1, and AKT1 were highlighted as key therapeutic targets. Molecular docking showed strong interactions between Daidzein and ESR1, Diosgenin and TNF α, Alatanin and AKT1, and PPARG. The findings suggest that D. alata's bioactive compounds, such as Diosgenin, Daidzein, Genistin, Cycloartane, and Alatanin, could modulate pathways involved in ovarian follicle formation, hormone regulation, estrogen receptor signaling, and the stress-activated MAP kinase pathway. This study provides new insights into the multi-target potential of D. alata for treating menstrual disorders, supporting further investigation and therapeutic development.
Collapse
Affiliation(s)
- Rajendran Silambarasan
- Department of R&D, Pankajakasthuri Herbal Research Foundation, Pankajakasthuri Ayurveda Medical College Campus, Trivandrum, India
| | - A. Kasthuri Nair
- Department of Kayachikitsa, Pankajakasthuri Ayurveda Medical College & PG Centre, Killy, Kattakada, Thiruvananthapuram, Kerala, India
| | - Gomathi Maniyan
- Native Women Food Products Foundation, Research and Development Department, SMIDS Campus, Nagercoil, Tamil Nadu, India
| | - R. Vijaya
- Department of Dravyagunavijnanam, Pankajakasthuri Ayurveda Medical College & P.G. Centre, Killy, Kattakada, Thiruvananthapuram, Kerala, India
| | - Reshma V.R. Nair
- Department of R&D, Pankajakasthuri Herbal Research Foundation, Pankajakasthuri Ayurveda Medical College Campus, Trivandrum, India
| | - J. Hareendran Nair
- Department of R&D, Pankajakasthuri Herbal Research Foundation, Pankajakasthuri Ayurveda Medical College Campus, Trivandrum, India
| | - S. Nishanth Kumar
- Department of R&D, Pankajakasthuri Herbal Research Foundation, Pankajakasthuri Ayurveda Medical College Campus, Trivandrum, India
| | - Shan Sasidharan
- HCEMM-SU Cardiovascular Comorbidities Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary
| |
Collapse
|
23
|
Martins Fernandes Pereira K, de Carvalho AC, Ventura Fernandes BH, Dos Santos Grecco S, Rodrigues E, da Silva Fernandes MJ, de Carvalho LRS, Nakamura MU, Guo S, Hernández RB. Systems toxicology studies reveal important insights about chronic exposure of zebrafish to Kalanchoe pinnata (Lam.) Pers leaf - KPL: Implications for medicinal use. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119044. [PMID: 39532221 DOI: 10.1016/j.jep.2024.119044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The prevalence of depression and anxiety is high during pregnancy. Several traditional medicines use the plant Kalanchoe pinnata (Lam.) Pers. (KP) to treat emotional disorders, inflammation, and to prevent preterm delivery, but the effects on the exposed offspring and the mechanism behind these events remain unknown. AIM OF THE STUDY In this work, integrated systems toxicology (INSYSTA) was used to investigate traditional toxicological outcomes and behavioral performance in zebrafish larvae after chronic exposure (from 2 to 96 hpf) to K. pinnata leaf extracts (KPL). MATERIALS AND METHODS We investigated light/dark preference, thigmotaxis and locomotor activity parameters, followed by gene expression and systems biology approaches to discover the mechanisms behind toxicological endpoint and phenomics. RESULTS The embryos exposed to 700 mg/L KPL showed retarded development including hatching delay. Larvae exposed to 500 mg/L KPL resulted in decreased dark avoidance and increased locomotor activity, while 700 mg/L showed opposite effects. The INSYSTA revealed sixteen genes down-regulated after KPL chronic treatment; they are involved in folding, sorting, and degradation of proteins as well as DNA replication and repair mechanisms. This may result in deregulation of the organismal functions, including those of immune and endocrine systems. These physiological changes appear to make embryos more sensitive to infections and disorders that resemble 47 human diseases. CONCLUSION These findings suggest that the medicinal use of plant extracts requires strict toxicological, pharmacological, and medical supervision. At the same time, it suggests a polypharmacological pathway for KPL extract that goes beyond preventing premature delivery and controlling anxiety.
Collapse
Affiliation(s)
- Kássia Martins Fernandes Pereira
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Universidade Federal de São Paulo, 04021-001, São Paulo, SP, Brazil.
| | | | - Bianca H Ventura Fernandes
- Technical Directorate of Support for Teaching, Research and Innovation at the Faculty of Medicine of the University of São Paulo, São Paulo, SP, Brazil.
| | - Simone Dos Santos Grecco
- Department of Chemistry, Universidade Federal de São Paulo, 09972-270, Diadema, SP, Brazil; Triplet Biotechnology Solutions, São Paulo, Brazil.
| | - Eliana Rodrigues
- Center for Ethnobotanical and Ethnopharmacological Studies, Department of Environmental Sciences, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Maria José da Silva Fernandes
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Universidade Federal de São Paulo, 04021-001, São Paulo, SP, Brazil.
| | - Luciani Renata Silveira de Carvalho
- Technical Directorate of Support for Teaching, Research and Innovation at the Faculty of Medicine of the University of São Paulo, São Paulo, SP, Brazil; Discipline of Endocrinology, Laboratory of Hormones and Molecular Genetics-LIM42, Hospital das Clínicas of the University of São Paulo, São Paulo, SP, Brazil.
| | - Mary Uchiyama Nakamura
- Department of Obstetrics, Universidade Federal de São Paulo, São Paulo, SP, 04021-001, Brazil.
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, Programs in Biological Sciences and Human Genetics, University of California, San Francisco, CA, 94158-2811, USA.
| | - Raúl Bonne Hernández
- Laboratory of Bioinorganic and Environmental Toxicology - LABITA, Department of Exact and Earth Sciences, Universidade Federal de São Paulo, 09972-270, Diadema, SP, Brazil.
| |
Collapse
|
24
|
Nascimento Júnior JAC, Oliveira AMS, Porras KDL, Menezes PDP, Araujo AADS, Nunes PS, Aragón DM, Serafini MR. Exploring trends in natural product-based treatments to skin burn: A comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156481. [PMID: 39951972 DOI: 10.1016/j.phymed.2025.156481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/13/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Burns are traumatic injuries caused by thermal, chemical, or other external factor, significantly impacting organic tissue. They are among the most common and severe types of trauma worldwide, often resulting in considerable morbidity and mortality. Natural products, owing to their pharmacological properties, present promising avenues for burn management and treatment. PURPOSE This study aims to provide a comprehensive review of patented pharmaceutical formulations containing natural products for burn treatment and to define trends in the market. METHODS Patent documents were identified through searches in the World Intellectual Property Organization (WIPO) and European Patent Office (EPO) databases using "burn*" as a keyword in the title and/or abstract and International Patent Classification (IPC) code A61K36/00. The review also examines clinical trials and SWOT analyses to evaluate strengths, weaknesses, opportunities, and threats in this field. RESULTS A total of 82 patents were selected, highlighting the use of natural products, such as Aloe vera, Coptis chinensis, borneol, menthol, and propolis, predominantly derived from Traditional Chinese Medicine. These findings are supplemented with clinical trial data and market insights. The results underscore both the therapeutic efficacy and challenges, such as standardization and regulatory hurdles, of using natural products. CONCLUSION This patent review highlights the potential of natural-origin formulations in addressing the limitations of conventional burn treatments. Continued research is essential to overcome existing barriers, ensuring broader accessibility and enhanced therapeutic outcomes.
Collapse
Affiliation(s)
| | - Ana Maria Santos Oliveira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil
| | | | - Paula Dos Passos Menezes
- Postgraduate Program in Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil; SejaPhD, Brazil
| | - Adriano Antunes de Souza Araujo
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil; Postgraduate Program in Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
| | - Paula Santos Nunes
- Postgraduate Program in Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil
| | - Diana Marcela Aragón
- Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional da Colombia, Bogotá D.C., Colombia
| | - Mairim Russo Serafini
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil; Postgraduate Program in Health Sciences, Federal University of Sergipe, São Cristóvão, Brazil.
| |
Collapse
|
25
|
Feifei W, Wenrou S, Jinyue S, Qiaochu D, Jingjing L, Jin L, Junxiang L, Xuhui L, Xiao L, Congfen H. Anti-ageing mechanism of topical bioactive ingredient composition on skin based on network pharmacology. Int J Cosmet Sci 2025; 47:134-154. [PMID: 39246148 DOI: 10.1111/ics.13005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/16/2024] [Accepted: 06/28/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE To elucidate the anti-ageing mechanism of the combination of eight ingredients on the skin from a multidimensional view of the skin. METHODS The target pathway mechanisms of composition to delay skin ageing were investigated by a network pharmacology approach and experimentally validated at three levels: epidermal, dermal, and tissue. RESULTS We identified 24 statistically significant skin ageing-related pathways, encompassing crucial processes such as epidermal barrier repair, dermal collagen and elastin production, inhibition of reactive oxygen species (ROS), as well as modulation of acetylcholine and acetylcholine receptor binding. Furthermore, our in vitro experimental findings exhibited the following outcomes: the composition promotes fibroblast proliferation and the expression of barrier-related genes in the epidermis; it also stimulated the expression of collagen I, collagen III, and elastic fibre while inhibiting ROS and β-Gal levels in HDF cells within the dermis. Additionally, Spilanthol in the Acmella oleracea extract contained in the composition demonstrated neuro-relaxing activity in Zebrafish embryo, suggesting its potential as an anti-wrinkle ingredient at the hypodermis level. CONCLUSIONS In vitro experiments validated the anti-ageing mechanism of composition at multiple skin levels. This framework can be extended to unravel the functional mechanisms of other clinically validated compositions, including traditional folk recipes utilized in cosmeceuticals.
Collapse
Affiliation(s)
- Wang Feifei
- Yunnan Botanee Bio-Technology Group Co., Ltd., Yunnan, China
- Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan, China
| | - Su Wenrou
- Yunnan Botanee Bio-Technology Group Co., Ltd., Yunnan, China
- Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan, China
| | - Sun Jinyue
- AGECODE R&D Center, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, China
- Beijing Key Lab of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China
| | - Du Qiaochu
- Yunnan Botanee Bio-Technology Group Co., Ltd., Yunnan, China
- Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan, China
| | - Li Jingjing
- Yunnan Botanee Bio-Technology Group Co., Ltd., Yunnan, China
- Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan, China
| | - Liu Jin
- Yunnan Botanee Bio-Technology Group Co., Ltd., Yunnan, China
- Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan, China
| | - Li Junxiang
- AGECODE R&D Center, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, China
- Harvest Biotech (Zhejiang) Co., Ltd., Zhejiang, China
| | - Li Xuhui
- AGECODE R&D Center, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, China
| | - Lin Xiao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - He Congfen
- Beijing Key Lab of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
26
|
Xiong Y, Huang X, Li Y, Nie Y, Yu H, Shi Y, Xue J, Ji Z, Rong K, Zhang X. Integrating larval zebrafish model and network pharmacology for screening and identification of edible herbs with therapeutic potential for MAFLD: A promising drug Smilax glabra Roxb. Food Chem 2025; 464:141470. [PMID: 39406145 DOI: 10.1016/j.foodchem.2024.141470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 11/21/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is becoming a prevalent chronic liver disease. Many medicinal and edible herbs exhibit remarkable biological activities in ameliorating MAFLD but lack a comprehensive assessment of their therapeutic efficacy. This study determined total phenolic and flavonoid contents and in vitro antioxidant properties of 34 edible herbs. Smilax glabra Roxb. (SGR), Coreopsis tinctoria Nutt., and Smilax china L. were obtained with the best bioactivity and antioxidant capacity. The high-cholesterol diet-induced larval zebrafish model was established to compare the anti-MAFLD activity of the three herb extracts mentioned above. In vivo experiments revealed that SGR intervention significantly decreased lipid accumulation, alleviated oxidative stress, and modulated intestinal microbiota composition in zebrafish. Furthermore, three potential active components in SGR and their possible mechanisms were explored through network pharmacology and molecular docking. Our study suggested that SGR is a potential candidate for developing new drugs or dietary supplements for MAFLD therapy.
Collapse
Affiliation(s)
- Yinjuan Xiong
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Xixuan Huang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yuxin Li
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yukang Nie
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Haodong Yu
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yaqi Shi
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Jiajie Xue
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Zhehui Ji
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Keming Rong
- Research Institute of Huanong-Tianchen, Wuhan 430070, China
| | - Xuezhen Zhang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China; Research Institute of Huanong-Tianchen, Wuhan 430070, China.
| |
Collapse
|
27
|
Jiang T, Li C, Li Y, Hu W, Guo J, Du X, Meng Q, Zhu X, Song W, Guo J, Su X. Multi-omics and bioinformatics for the investigation of therapeutic mechanism of roucongrong pill against postmenopausal osteoporosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118873. [PMID: 39362330 DOI: 10.1016/j.jep.2024.118873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Roucongrong Pill (RCRP), originating from the historical General Medical Collection of Royal Benevolence, is frequently used to treat postmenopausal osteoporosis (PMOP). Despite its prevalent application, the specific anti-osteoporotic mechanisms of RCRP remain to be elucidated. AIM OF THE STUDY This study aims to elucidate the therapeutic mechanism of RCRP in the context of ovariectomy (OVX)-induced PMOP in rats. By employing an integrative approach, the research combines medicinal chemistry, gut microbiota (GM) profiling, metabolomics, MetOrigin traceability, network pharmacology, molecular docking, and molecular dynamics simulations to deliver a comprehensive analysis. MATERIALS AND METHODS Sprague-Dawley (SD) rats underwent bilateral OVX to establish a PMOP model. The therapeutic efficacy of RCRP was evaluated through bone metrics (BMD, bone strength, BV/TV, Tb.Sp), hematoxylin and eosin (H&E) histological assessment, and bone metabolism markers (OPG, BALP, TRACP-5b, β-CTX, RANKL). Fecal metabolomics and 16S rDNA sequencing were employed to assess the influence of RCRP on GM and metabolite profiles. Furthermore, MetOrigin facilitated the traceability analysis of relevant metabolites. Molecular docking identified potential RCRP compounds with anti-PMOP activity, while their stability and protein interactions were assessed through molecular dynamics simulations. Network pharmacology further confirms the targets of action. RESULTS RCRP alleviated PMOP in rats, enhancing bone strength, cortical and trabecular BMD, BV/TV, and serum OPG levels, while reducing Tb.Sp, serum BALP, TRACP-5b, β-CTX, and RANKL concentrations. A total of twenty-six distinct metabolites were identified, of which ten-tribufos, sulfoacetic acid, betamethasone dipropionate, 9-oxooctadeca-10,12,15-trienoic acid, menatetrenone, piperlongumine, maltopentaose, enol-phenylpyruvate, catechol, pentaacetate, and (+)-2-methylpropanoic acid-exhibited correlations with six GM species: Turicibacter, Roseburia, Colidextribacter, Helicobacter, Odoribacter, and Lachnoclostridium, as determined by Spearman's correlation analysis. Notably, MetOrigin revealed the microbial metabolism of taurine and hypotaurine, along with host-specific steroid hormone synthesis. Computational docking studies demonstrated robust interactions between five RCRP-derived steroids (hydroxyecdysone, corticosterone, trilostane, 5α-androstan-3,6,17-trione, and cortisol) and key enzymes (estradiol 17α-dehydrogenase and UDP-glucuronosyltransferase), suggesting a potential enhancement of therapeutic efficacy against PMOP. Furthermore, molecular dynamics simulations indicated stable interactions between hydroxyecdysone and two proteins, with binding free energies of -67.427 kJ/mol and -156.948 kJ/mol, respectively. Through network pharmacology and molecular docking approaches, potential targets of these metabolites were identified, including estrogen receptors ESR1 and ESR2, dual specificity phosphatase 6 (DUSP6), sex hormone-binding globulin (SHBG), prostaglandin E receptor 4 (PTGER4), cannabinoid receptor 2 (CNR2), cathepsin K (CTSK), and androgen receptor (AR). CONCLUSIONS RCRP effectively mitigates OVX-induced bone loss in PMOP rats by modulating GM and associated metabolites, along with their potential targets and key metabolic pathways, including taurine and hypotaurine metabolism, as well as steroid hormone biosynthesis. These findings offer new insights into the therapeutic mechanisms by which RCRP may alleviate PMOP.
Collapse
Affiliation(s)
- Tao Jiang
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Chenhao Li
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Yufen Li
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Wanli Hu
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Jiurui Guo
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xingchen Du
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Qianting Meng
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xiaojuan Zhu
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Wu Song
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Junpeng Guo
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xin Su
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
28
|
Zhou C, Xiang T, Yu Y, Ma H, Liu C, Yang F, Yang L. Dose-Weighted Network Pharmacology: Evaluating Traditional Chinese Medicine Formulations for Lumbar Disc Herniation. J Inflamm Res 2025; 18:1281-1300. [PMID: 39897525 PMCID: PMC11784360 DOI: 10.2147/jir.s496124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/18/2025] [Indexed: 02/04/2025] Open
Abstract
Background and Purpose Lumbar disc herniation (LDH) significantly impacts individuals, particularly those aged 40-45. Traditional Chinese Medicine (TCM) formulations such as Taohong Siwu Decoction (TSD), Yaotong Jizheng Decoction (YJD), and Panlong Qi Tablet (PQT) are widely used for treatment. This study introduces dose-weighted network pharmacology, a novel approach that incorporates drug dosage as a quantitative factor into network analysis to evaluate better and compare the therapeutic potential of TCM formulations. Methods This study combines drug dosage with the PPI network to propose a theoretical algorithm for comparing the therapeutic efficacy of different traditional Chinese medicine formulations. The VIKOR method was used to assess the importance of therapeutic targets, with weights assigned based on both drug and disease perspectives. TSD, YJD, and PQT were evaluated in animal experiments, and the algorithm's feasibility was validated through GO and KEGG pathway analysis, Thermal Hyperalgesia Test, H&E staining, Western blotting (WB), RT-PCR, and ELISA assays. Results The computational model indicated that YJD and PQT had higher predicted efficacy compared to TSD. These predictions were confirmed in animal studies, where YJD demonstrated the greatest reduction in thermal hyperalgesia and the most significant decrease in inflammatory markers, surpassing both TSD and PQT. GO and KEGG pathway analyses highlighted key pathways related to oxidative stress and inflammation, providing mechanistic insights into the effectiveness of the treatments. Conclusion Incorporating dosage as a reference factor into network pharmacology research proved feasible and effective, emphasizing the importance of precise dosage control in TCM formulations for treating LDH. The new algorithm provided reliable predictions, demonstrating its potential to enhance the design and evaluation of TCM formulations. Future improvements, such as establishing a target acceptance rate database, could further refine the algorithm, expanding its application in personalized medicine and targeted therapy.
Collapse
Affiliation(s)
- Changwen Zhou
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Shaanxi, People’s Republic of China
| | - Ting Xiang
- Rehabilitation Department, Xiang Xi Autonomous Prefecture National Hospital, Hunan, People’s Republic of China
| | - Yu Yu
- Department of Chinese Medicine, Minda Hospital of Hubei Minzu University, Hubei, People’s Republic of China
| | - Hongzhong Ma
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Shaanxi, People’s Republic of China
| | - Ce Liu
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Shaanxi, People’s Republic of China
| | - Feng Yang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Shaanxi, People’s Republic of China
- Department of Orthopedic Hospital, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, People’s Republic of China
| | - Lixue Yang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Shaanxi, People’s Republic of China
- Department of Orthopedic Hospital, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, People’s Republic of China
| |
Collapse
|
29
|
Lee MJ, Jeong D, Lee JH, Kang J, Choi J, Seo J, Kim HI, Seo J, Ko K, Nam DH, Lee HL, Kang KS. The Effects of a Novel Astragalus-Based Extract (Keyfobell Powder (KFB)) on Longitudinal Bone Growth via IGF-1 Upregulation: A Potential Growth Hormone Alternative. Nutrients 2025; 17:416. [PMID: 39940274 PMCID: PMC11820268 DOI: 10.3390/nu17030416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/12/2025] [Accepted: 01/18/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study evaluated the effects of a novel Astragalus extract (Keyfobell powder [KFB]) composed of Astragalus membranaceus, red ginseng (Panax ginseng C. A. Meyer), and Cervi Parvum Cornu as a potential growth hormone (GH) alternative. The primary focus was placed on its impact on longitudinal bone growth through the upregulation of circulatory insulin-like growth factor (IGF)-1. METHODS We performed in vitro and in vivo experiments using a hypothalamic cell line and Sprague-Dawley (SD) rats. Quantitative RT-PCR was performed to determine growth hormone-releasing hormone (GHRH) and ghrelin mRNA expressions in GT1-7 cells. The treatment groups were administered KFB at various dosages, and the positive controls received recombinant human GH. Body weight, bone length, and density were assessed, along with serum levels of insulin-like growth factor binding protein (IGFBP)-3 and IGF-1. RESULTS KFB and somatropin exhibited no cytotoxic effect in GT1-7 cells and increased GHRH and ghrelin mRNA levels in a dose-dependent manner. KFB administration resulted in a significant dose-dependent increase in body weight and bone growth (femur and tibia). Changes in IGF-1 and IGFBP-3 levels were comparable to those observed in the GH-treated group. Based on network pharmacological analysis, multiple compounds in KFB ((20S)-20-hydroxypregn-4-en-3-one, 2-isopropyl-3-methoxypyrazine, caproic acid, daidzein, furfuryl alcohol, lauric acid, octanal, and salicylic acid) may synergistically regulate the PI3K-Akt, Ras, and Rap1 signaling pathways linked to growth control and cartilage formation, leading to a possible increase in height. CONCLUSIONS Our results suggest that KFB can function as a GH-mimetic agent that promotes bone growth through IGF-1 upregulation.
Collapse
Affiliation(s)
- Myong Jin Lee
- Department of Preventive Medicine, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (M.J.L.); (J.H.L.)
| | - Daesik Jeong
- College of Convergence Engineering, Sangmyung University, Seoul 03016, Republic of Korea;
| | - Ji Hwan Lee
- Department of Preventive Medicine, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (M.J.L.); (J.H.L.)
| | - Jaeha Kang
- Department of Computer Science, Sangmyung University, Seoul 03016, Republic of Korea;
| | - Jihye Choi
- Chong Kun Dang (CKD) Pharm Research Institute, Yongin 16995, Republic of Korea; (J.C.); (J.S.); (H.I.K.); (J.S.); (K.K.); (D.H.N.)
| | - Jaeok Seo
- Chong Kun Dang (CKD) Pharm Research Institute, Yongin 16995, Republic of Korea; (J.C.); (J.S.); (H.I.K.); (J.S.); (K.K.); (D.H.N.)
| | - Hong Il Kim
- Chong Kun Dang (CKD) Pharm Research Institute, Yongin 16995, Republic of Korea; (J.C.); (J.S.); (H.I.K.); (J.S.); (K.K.); (D.H.N.)
| | - Jisoo Seo
- Chong Kun Dang (CKD) Pharm Research Institute, Yongin 16995, Republic of Korea; (J.C.); (J.S.); (H.I.K.); (J.S.); (K.K.); (D.H.N.)
| | - Kiseong Ko
- Chong Kun Dang (CKD) Pharm Research Institute, Yongin 16995, Republic of Korea; (J.C.); (J.S.); (H.I.K.); (J.S.); (K.K.); (D.H.N.)
| | - Dong Hyuk Nam
- Chong Kun Dang (CKD) Pharm Research Institute, Yongin 16995, Republic of Korea; (J.C.); (J.S.); (H.I.K.); (J.S.); (K.K.); (D.H.N.)
| | - Hye Lim Lee
- Department of Pediatrics, College of Korean Medicine, Daejeon University, Daejeon 34520, Republic of Korea
| | - Ki Sung Kang
- Department of Preventive Medicine, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (M.J.L.); (J.H.L.)
| |
Collapse
|
30
|
Zhang R, Wang P, Jin Y, Xie Q, Xiao P. Imperatorin's Effect on Myocardial Infarction Based on Network Pharmacology and Molecular Docking. Cardiovasc Ther 2025; 2025:7551459. [PMID: 39834616 PMCID: PMC11745561 DOI: 10.1155/cdr/7551459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025] Open
Abstract
Purpose: Myocardial infarction (MI), a severe cardiovascular disease, is the result of insufficient blood supply to the myocardium. Despite the improvements of conventional therapies, new approaches are needed to improve the outcome post-MI. Imperatorin is a natural compound with multiple pharmacological properties and potential cardioprotective effects. Therefore, this work investigated imperatorin's therapeutic effects and its mechanism of action in an MI mouse model. Methods: Network pharmacology, molecular docking, and experimental validation were performed for exploring the pharmacokinetic properties, therapeutic effects, and molecular targets of imperatorin in MI. Permanent ligation of the left anterior descending artery was performed in male C57BL/6 mice to induce MI before treatment with imperatorin once per day for 1 week. Echocardiography, heart histology, RNA sequencing, and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) as well as western blotting were carried out for evaluating cardiac function and structure, as well as gene and protein expression. Results: Imperatorin significantly improved cardiac function, preserved cardiac structure, attenuated cardiac remodeling and fibrosis, and reduced cardiomyocyte apoptosis in MI mice. Eight differentially expressed genes overlapping with key target genes were found, two upregulated and six downregulated. A key target in signaling pathways associated with imperatorin effect in MI was angiotensin-converting enzyme (ACE). Imperatorin inhibited ACE-angiotensin II (Ang II)-angiotensin II Type 1 receptor (AT1R) axis in MI mice. Conclusion: Imperatorin attenuated MI by inhibiting the ACE-Ang II-AT1R axis. Thus, imperatorin might be considered a potential therapeutic agent to cure MI.
Collapse
MESH Headings
- Animals
- Myocardial Infarction/drug therapy
- Myocardial Infarction/pathology
- Myocardial Infarction/metabolism
- Myocardial Infarction/physiopathology
- Myocardial Infarction/genetics
- Male
- Furocoumarins/pharmacology
- Furocoumarins/chemistry
- Mice, Inbred C57BL
- Disease Models, Animal
- Network Pharmacology
- Molecular Docking Simulation
- Apoptosis/drug effects
- Signal Transduction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/metabolism
- Ventricular Remodeling/drug effects
- Fibrosis
- Ventricular Function, Left/drug effects
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/drug effects
- Angiotensin II
- Gene Expression Regulation
- Gene Regulatory Networks
Collapse
Affiliation(s)
- Ruizhe Zhang
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yao Jin
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qingya Xie
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Pingxi Xiao
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Liu CY, Li Z, Cheng FE, Nan Y, Li WQ. Radix Codonopsis: a review of anticancer pharmacological activities. Front Pharmacol 2025; 15:1498707. [PMID: 39840099 PMCID: PMC11747557 DOI: 10.3389/fphar.2024.1498707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Radix Codonopsis (Dangshen), derived from the dried root of plants in the Campanulaceae family, is a widely used Chinese herbal medicine. It is renowned for its pharmacological effects, including tonifying the middle qi, invigorating the spleen, benefiting the lungs, enhancing immunity, and nourishing the blood. Codonopsis extract is frequently incorporated into health products such as tablets and capsules, making it accessible for daily health maintenance. Additionally, it is commonly used in dietary applications like soups, teas, and porridges to nourish qi, enrich blood, and promote overall vitality. In recent years, increasing attention has been given to the anti-cancer potential of Radix Codonopsis. Studies have identified key active components such as luteolin, stigmasterol, polyacetylenes, lobetyolin, and glycitein, which exhibit anti-tumor properties through mechanisms like inhibiting cancer cell growth and proliferation, suppressing epithelial-mesenchymal transition (EMT), and inducing apoptosis. This review highlights the research progress on Radix Codonopsis, including its active constituents, anti-cancer mechanisms, and its role in the convergence of medicine and food in modern life. By doing so, it aims to provide valuable insights and references for future scientific studies and clinical applications of Radix Codonopsis.
Collapse
Affiliation(s)
- Cai-Yue Liu
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
| | - Zheng Li
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
| | - Fan-E. Cheng
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
| | - Yi Nan
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
- Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Wei-Qiang Li
- Ningxia Medical University, Ningxia of Traditional Chinese Medicine, Yinchuan, China
- Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
32
|
Kim DU, Kweon B, Oh J, Lim Y, Noh G, Yu J, Kang HR, Kwon T, Lee KY, Bae GS. A Network Pharmacology Study and Experimental Validation to Identify the Potential Mechanism of Heparan Sulfate on Alzheimer's Disease-Related Neuroinflammation. Biomedicines 2025; 13:103. [PMID: 39857687 PMCID: PMC11761859 DOI: 10.3390/biomedicines13010103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Heparan sulfate (HS) is a polysaccharide that is found on the surface of cells and has various biological functions in the body. METHODS The purpose of this study was to predict the pharmacological effects and molecular mechanisms of HS on Alzheimer's disease (AD) and neuroinflammation (NI) through a network pharmacology analysis and to experimentally verify them. RESULTS We performed functional enrichment analysis of common genes between HS target genes and AD-NI gene sets and obtained items such as the "Cytokine-Mediated Signaling Pathway", "Positive Regulation Of MAPK Cascade", and "MAPK signaling pathway". To confirm the predicted results, the anti-inflammatory effect of HS was investigated using lipopolysaccharide (LPS)-stimulated BV2 microglia cells. HS inhibited the production of nittic oxide, interleukin (IL)-6, and tumor necrosis factor-α in LPS-stimulated BV2 cells, but not IL-1β. In addition, HS inactivated P38 in the MAPK signaling pathway. CONCLUSIONS These findings suggest the potential for HS to become a new treatment for AD and NI.
Collapse
Affiliation(s)
- Dong-Uk Kim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Jinyoung Oh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Yebin Lim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
| | - Gyeongran Noh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
| | - Jihyun Yu
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
| | - Hyang-Rin Kang
- Woori B&B Life Science Laboratory, Jeonju 54853, Republic of Korea; (H.-R.K.)
| | - Tackmin Kwon
- Woori B&B Life Science Laboratory, Jeonju 54853, Republic of Korea; (H.-R.K.)
| | - Kwang youll Lee
- Woori B&B Life Science Laboratory, Jeonju 54853, Republic of Korea; (H.-R.K.)
| | - Gi-Sang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (B.K.)
- Hanbang Cardio-Renal Syndrome Research Center, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
33
|
Sreeharsha N, Basavarajappa GM, Aloufi B, Shiroorkar PN, Anwer MK, Rehman A. An Integrative Network Pharmacology and Bioinformatics Approach for Deciphering the Multi-target Effect of Nyctanthes arbortristis L. against COVID-19. Curr Pharm Des 2025; 31:855-872. [PMID: 38877861 DOI: 10.2174/0113816128298950240428013723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/06/2024] [Indexed: 04/24/2025]
Abstract
INTRODUCTION The COVID-19 pandemic represents a significant challenge across scientific, medical, and societal dimensions. The unpredictability of the disease progression, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), underscores the urgent need for identifying compounds that target multiple aspects of the virus to ensure swift and effective treatment. Nyctanthes arbortristis L., a delicate, perennial, deciduous shrub found across various Asian regions, has been recognized for its wide range of pharmacological benefits, including hepatoprotective, antimalarial, antibacterial, anti-inflammatory, antioxidant, and antiviral properties. METHODS Various in vitro studies revealed the therapeutic significance of Nyctanthes arbortristis against COVID-19. However, the exact molecular mechanism remains unclarified. In the present study, a network pharmacology approach was employed to uncover the active ingredients, their potential targets, and signaling pathways in Nyctanthes arbortristis for the treatment of COVID-19. In the framework of this study, we explored the active ingredient-target-pathway network and figured out that naringetol, ursolic acid, betasitosterol, and daucosterol decisively contributed to the development of COVID-19 by affecting IL6, MAPK3, and MDM2 genes. RESULTS The results of molecular docking analysis indicated that Nyctanthes arbortristis exerted effective binding capacity in COVID-19. Further, we disclosed the targets, biological functions, and signaling pathways of Nyctanthes arbortristis in COVID-19. The analysis indicated that Nyctanthes arbortristis could help treat COVID-19 through the enhancement of immunologic functions, inhibition of inflammatory reactions and regulation of the cellular microenvironment. In short, the current study used a series of network pharmacologybased and computational analyses to understand and characterize the binding capacity, biological functions, pharmacological targets and therapeutic mechanisms of Nyctanthes arbortristis in COVID-19. CONCLUSION However, the findings were not validated in actual COVID-19 patients, so further investigation is needed to confirm the potential use of Nyctanthes arbortristis for treating COVID-19.
Collapse
Affiliation(s)
- Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bangalore 560035, India
| | | | - Bandar Aloufi
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | | | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Alkharj 11942, Saudi Arabia
| | - Abdur Rehman
- Department of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
34
|
Ayari A, Dakhli N, Jedidi S, Sammari H, Arrari F, Sebai H. Laxative and purgative actions of phytoactive compounds from beetroot juice against loperamide-induced constipation, oxidative stress, and inflammation in rats. Neurogastroenterol Motil 2025; 37:e14935. [PMID: 39370602 DOI: 10.1111/nmo.14935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Chronic constipation is a gastrointestinal functional disorder which affects patient quality of life. Therefore, many studies were oriented to search herbal laxative agents. In this study, we investigated the phytochemical composition of beetroot juice (BJ) and its laxative potential in an experimental model of constipation and colonic dysmotility induced by loperamide (LOP) in Wistar rats. METHODS Animals were concurrently pretreated with LOP (3 mg/kg, b.w., i.p.) and BJ (5 and 10 mL/kg, b.w., p.o.), or yohimbine (2 mg/kg, b.w., i.p.), during 1 week. The laxative activity was determined based on the weight, frequency, and water content of the feces matter. The gastric-emptying test and intestinal transit were determined. Colon histology was examined, and oxidative status was evaluated using biochemical-colorimetric methods. KEY RESULTS The in vivo study revealed that LOP induced a significant inhibition of gastrointestinal motility, negative consequences on defecation parameters, oxidative stress, and colonic mucosa lesions. Conversely, administration of BJ reestablished these parameters and restored colonic oxidative balance. Importantly, BJ treatment protected against LOP-induced inflammatory markers (pro-inflammatory cytokines and WBC) and the increase in intracellular mediators such as hydrogen peroxide, free iron, and calcium levels. CONCLUSIONS & INFERENCES This study demonstrated that the bioactive compounds in BJ provided an anti-constipation effect by modulating intestinal motility and regulating oxidative stress and inflammation induced by LOP intoxication.
Collapse
Affiliation(s)
- Ala Ayari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Nouha Dakhli
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
- Sylvo-Pastoral Institute of Tabarka, Laboratory of Sylvo-Pastoral Resources, University of Jendouba, Tabarka, Tunisia
| | - Houcem Sammari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Fatma Arrari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| |
Collapse
|
35
|
Luo P, Feng X, Liu S, Jiang Y. Traditional Uses, Phytochemistry, Pharmacology and Toxicology of Ruta graveolens L.: A Critical Review and Future Perspectives. Drug Des Devel Ther 2024; 18:6459-6485. [PMID: 39758226 PMCID: PMC11697671 DOI: 10.2147/dddt.s494417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
Medicinal plants are fundamental sources of natural products with high chemical diversity and specificity as novel lead compounds with diverse pharmacological activities. Ruta graveolens L. is an important traditional Chinese medicine used to treat fever caused by cold, wind-fire toothache, headache, bruises and sprains, irregular menstruation, and infantile eczema. Although various traditional uses and chemical constituent activity evaluations have been reported, no systematic review and future perspective of R. graveolens has been published. A total of 113 literature about R. graveolens were collected from online scientific databases, including SciFinder, PubMed, CNKI, Web of Science, and Google Scholar. Additional information was obtained from other sources of literature, such as the Chinese Pharmacopoeia, Flora of China, classical Chinese herbal books and local prints and scripts. Herein, we comprehensively review the traditional uses, phytochemistry, pharmacology, and toxicology of R. graveolens, and provide critical comments and meaningful perspectives for the future development of this medicinal plant.
Collapse
Affiliation(s)
- Ping Luo
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Xu Feng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- College of Pharmacy, Changsha Medical University, Changsha, Hunan, 410219, People’s Republic of China
| |
Collapse
|
36
|
Zoghebi K, Sabei FY, Safhi AY. Exploring the anti-cancer properties of Carissa carandas as a multi-targeted approach against breast cancer. J Biomol Struct Dyn 2024:1-25. [PMID: 39660546 DOI: 10.1080/07391102.2024.2437548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/22/2024] [Indexed: 12/12/2024]
Abstract
The escalating incidence of breast cancer globally presents a formidable challenge within oncology. Our research pursued an examination of the anti-cancer potential of Carissa carandas, a shrub traditionally used for medicinal purposes and known for its composition of vital nutrients and phytochemicals. We employed a network pharmacology strategy combined with molecular docking and molecular dynamics simulations to elucidate the intricate relationships between the phytochemical constituents of C. carandas, critical breast cancer proteins, and associated signaling pathways. The study highlighted a complex network of protein interactions, identifying AKT1, HIF1A, PTGS2, and GSK3B as key nodes within this network. These proteins are engaged by numerous investigated compounds from C. carandas and are fundamental in modulating crucial signaling pathways such as those involving Estrogen, HIF-1, Prolactin, VEGF, and Th17 cell differentiation-each of which plays a recognized role in breast cancer progression, affecting tumor growth, proliferation, and metastatic potential. Our analysis suggests that the phytochemicals in C. carandas may exert anti-cancer activity by synergistically modulating these pathways, highlighting the benefit of multi-targeted therapeutic approaches over single-targeted ones. In summary, through the application of advanced network pharmacology, molecular docking, MD simulations, and MM/PBSA analysis, our study offers a detailed exploration of the potential mechanisms by which C. carandas may exert anti-cancer effects. This sets a foundation for further in-depth experimental and clinical trials to validate these mechanisms and support the advancement of novel plant-derived therapeutic options towards breast cancer, with the possibility of significantly advancing the therapeutic options for this prevalent disease.
Collapse
Affiliation(s)
- Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Fahad Y Sabei
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
37
|
Kulshrestha S, Goel A, Banerjee S, Sharma R, Khan MR, Chen KT. Metabolomics and network pharmacology-guided analysis of TNF-α expression by Argemone mexicana (Linn) targeting NF-kB the signalling pathway in cancer cell lines. Front Oncol 2024; 14:1502819. [PMID: 39687882 PMCID: PMC11648424 DOI: 10.3389/fonc.2024.1502819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/04/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Cancer has emerged as one of the leading causes of fatality all over the world. Phytoconstituents are being studied for their synergistic effects, which include disease prevention by altering molecular pathways and immunomodulation without side effects. The present experiment aims to explore the cancer preventive activities of Argemone mexicana Linn leaves extract in skin cancer cell lines (A431) and colon cancer cell lines (COLO 320DM)). In addition, TNF-α expression patterns and NF-kB signaling pathways have been examined. Methods LC/MS study of Argemone mexicana Linn extracts in various solvents revealed anti-cancerous phytoconstituents. Network pharmacology analysis used Binding DB, STRING, DAVID, and KEGG for data mining to evaluate predicted compounds using functional annotation analysis. Cytoscape 3.2.1 created "neighbourhood approach" and networks. The MNTD of these extracts was tested on L929 fibroblasts. Skin cancer (A431) and colon cancer (COLO 320DM) cell lines were tested for IC50 inhibition. Evaluation of TNF-α and NF-kB expression in cell culture supernatants and homogenates reveals anti-cancerous effects. Results LC-MS analysis of extracts predicted the presence of anticancer alkaloids Berberine, Atropine, Argemexicin, and Argemonin. In Network pharmacology analysis, enrichment was linked to the PI3-AKT pathway for both cancer types. MNTD was calculated at 1000μg/ml in L929. The ethanolic extract at 1000μg/ml significantly inhibited skin cancer cell proliferation by 67% and colon cancer cells by 75%. Ethanolic extract significantly reduced TNF-α expression in both cell lines (p<0.001), with the highest inhibition at 1000μg/ml. In TNF-α stimulated cell lines, 1000μg/ml ethanolic extract significantly reduced the regulation of the NF-kB pathway, which plays a role in cancer progression (p<0.001). Conclusion Argemone mexicana Linn. known as 'swarnkshiri' in Ayurveda has been reported to be used by the traditional healers for the treatment of psoriasis and its anti-inflammatory and anti-cancerous properties, according to the Indian Medicinal Plant dictionary. In the experiment, the abatement in the expression of inflammatory cytokine TNF-α and inhibition of NF-kB transcription factor activation could be linked with the downregulation of cancer cell proliferation. The study revealed the anticancer activity of Argemone mexicana Linn in the cancer cell lines and paved a pathway for molecular approaches that could be explored more in In vivo studies.
Collapse
Affiliation(s)
| | - Anjana Goel
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Subhadip Banerjee
- School of Natural Products Studies, Jadavpur University, Kolkata, West Bengal, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi, India
| | - Mohammad Rashid Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Kow-Tong Chen
- Department of Occupational Medicine, Tainan Municipal Hospital (managed by ShowChwan Medical Care Corporation), Tainan, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
38
|
Chen Q, Mo J, Li Y, Gao L, Wu K. Anti-Inflammatory Action and Molecular Mechanism of Fucoidan Against Cystitis Glandularis. Food Sci Nutr 2024; 12:10255-10261. [PMID: 39723046 PMCID: PMC11666824 DOI: 10.1002/fsn3.4560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/09/2024] [Accepted: 10/10/2024] [Indexed: 12/28/2024] Open
Abstract
Cystitis glandularis (CG), known as a pre-gradual lesion in the bladder, is the pathological changes in the vesical mucosa characterized by inflammatory invasion and chronic obstruction. Clinically, effective treatment against CG is prescribed only when using drug therapy. Fucoidan, the naturally extractive polysaccharide, is well-reported bioactive compound with anti-inflammatory and immunoregulatory properties. In this research, an emerging computational approach was applied to explicate anti-CG actions and pharmacological targets exhibited by fucoidan in detail. Current network pharmacology data showed that 16 intersection genes of fucoidan and CG were identified, whereas all 6 core targets, including interleukin-6 (IL-6), tumor necrosis factor (TNF), interleukin-1B (IL-1B), matrix metalloproteinase-9 (MMP-9), interleukin-10 (IL-10), matrix metalloproteinase-2 (MMP-2), biological processes, and signaling pathways of fucoidan against CG were characterized, respectively. As revealed in the underlying mechanism, the anti-CG actions achieved by fucoidan were chiefly implicated in the reduction of inflammatory reactions and enhancement of immunoregulation. Taken together, these network bioinformatics findings may be used to reveal anti-CG effects and the pharmacological mechanism of fucoidan before further experimental validation. Furthermore, those core genes identified may be therapeutic targets for research and development of fucoidan-anti-CG.
Collapse
Affiliation(s)
- Qingting Chen
- The Third Affiliated Hospital of Guangxi Medical University, The Second People's Hospital of Nanning CityNanningChina
| | - Jie Mo
- The Third Affiliated Hospital of Guangxi Medical University, The Second People's Hospital of Nanning CityNanningChina
| | - Yu Li
- Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Li Gao
- Department of Urology SurgeryThe Second Affiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Ka Wu
- The Third Affiliated Hospital of Guangxi Medical University, The Second People's Hospital of Nanning CityNanningChina
| |
Collapse
|
39
|
Sailah I, Tallei TE, Safitri L, Tamala Y, Halimatushadyah E, Ekatanti D, Maulydia NB, Celik I. A network pharmacology approach to elucidate the anti-inflammatory and antioxidant effects of bitter leaf ( Vernonia amygdalina Del.). NARRA J 2024; 4:e1016. [PMID: 39816097 PMCID: PMC11731942 DOI: 10.52225/narra.v4i3.1016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 01/18/2025]
Abstract
The therapeutic potential of bitter leaf (Vernonia amygdalina Del.) has been established both empirically and in various scientific investigations. However, the molecular pathways related to its possible anti-inflammatory and antioxidant properties remain unclear. Therefore, the aim of this study was to elucidate the molecular interactions between bitter leaf's bioactive compounds and cellular targets involved in these activities. The compounds in bitter leaf were identified using gas chromatography-mass spectrometry (GC-MS) analysis, and subsequently, a network pharmacology approach was employed together with molecular docking and dynamics simulations. Acetonitrile (4.5%) and dimethylamine (4.972%) were the most prevalent compounds among the 38 identified by the GC-MS analysis of bitter leaf extract. The proto-oncogene tyrosine-protein kinase (SRC) demonstrated significant connectivity within the antioxidant network, highlighting its pivotal role in facilitating inter-protein communication. It also exhibited strategic positioning in anti-inflammatory mechanisms based on closeness centrality (0.385). The enrichment analysis suggested multifaceted mechanisms of bitter leaf compounds, including transcriptional regulation and diverse cellular targeting, indicating broad antioxidant and anti-inflammatory effects. Eicosapentaenoyl ethanolamide (EPEA) displayed strong interactions with multiple proteins, including SRC (-7.17 kcal/mol) and CYP3A4 (-6.88 kcal/mol). Moreover, EPEA demonstrated to form a stable interaction with SRC during a 100 ns simulation. In conclusion, the computational simulations revealed that the hypothetical antioxidant and anti-inflammatory actions of bitter leaf compounds were achieved by specifically targeting SRC. However, confirmation using either in vitro or in vivo techniques is necessary.
Collapse
Affiliation(s)
- Illah Sailah
- Agroindustrial Engineering Study Program, Department of Agroindustrial Technology, Faculty of Agricultural Engineering and Technology, Institut Pertanian Bogor, Bogor, Indonesia
| | - Trina E. Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sam Ratulangi, Manado, Indonesia
| | - Linda Safitri
- Agroindustrial Engineering Study Program, Department of Agroindustrial Technology, Faculty of Agricultural Engineering and Technology, Institut Pertanian Bogor, Bogor, Indonesia
| | - Yulianida Tamala
- Agroindustrial Engineering Study Program, Department of Agroindustrial Technology, Faculty of Agricultural Engineering and Technology, Institut Pertanian Bogor, Bogor, Indonesia
| | - Ernie Halimatushadyah
- Pharmacy Study Program, Faculty of Health Sciences and Technology, Universitas Binawan, Jakarta, Indonesia
| | - Dewi Ekatanti
- Pharmacy Study Program, Faculty of Mathematics and Natural Sciences, Universitas Sam Ratulangi, Manado, Indonesia
| | - Nur B. Maulydia
- Graduate School of Mathematics and Applied Sciences, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| |
Collapse
|
40
|
Ferjani W, Kouki A, Dang PMC, Fetoui H, Chtourou Y, Ghanem-Boughanmi N, Ben-Attia M, El-Benna J, Souli A. Opuntia ficus-indica cladodes extract inhibits human neutrophil pro-inflammatory functions and protects rats from acetic acid-induced ulcerative colitis. Inflammopharmacology 2024; 32:3825-3844. [PMID: 39369123 DOI: 10.1007/s10787-024-01577-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024]
Abstract
The increased production of reactive oxygen species (ROS) by human neutrophils can lead to oxidative imbalances and several diseases, such as inflammatory bowel disease (IBD). Opuntia ficus-indica (O. ficus-indica) is rich in bioactive substances with anti-inflammatory properties. This study aimed to identify the bioactive compounds present in aqueous cladodes extract (ACE) of O. ficus-indica using high-performance liquid chromatography (HPLC) and to test its effects on human neutrophil inflammatory functions and on ulcerative colitis (UC) induced by acetic acid (Aa) in rats. ROS production and degranulation by neutrophils were assessed by luminol-amplified chemiluminescence, enzymatic techniques, and western blotting. In vivo, the experiment involved seven groups of rats: a negative control group (NaCl), the acetic acid group (Aa), and groups treated with oral sulfasalazine (150 mg/kg) or various doses of ACE for 7 days. Colonic lesions were induced by an intra-rectal Aa injection, and inflammation was assessed. HPLC analysis identified gallic acid, catechin, caffeic acid, and ferulic acid as major compounds in ACE. In vitro, ACE inhibited neutrophil ROS production, including superoxide anion produced by NADPH oxidase, and significantly reduced myeloperoxidase activity and neutrophil degranulation. In vivo, ACE protected rats from Aa-induced histopathological damage of the colonic mucosa, significantly increased catalase, superoxide dismutase and reduced glutathione levels, and significantly suppressed the increases of plasma cytokines (TNF-α and IL-1β) observed in the Aa group. In conclusion, O. ficus-indica ACE has significant anti-inflammatory properties by restoring oxidative balance, indicating that it could be a potential source of therapeutic agents for inflammatory diseases, particularly UC.
Collapse
Affiliation(s)
- Wafa Ferjani
- INSERM-U1149, CNRS-ERL8252, Inflammation Research Center, Inflamex Excellence Laboratory, X. Bichat Faculty of Medicine, University of Paris-Cité, 75018, Paris, France
- Environment Biomonitoring Laboratory (LR01/ES14), Sciences Faculty of Bizerte, University of Carthage, Zarzouna, 7021, Bizerte, Tunisia
| | - Ahmed Kouki
- INSERM-U1149, CNRS-ERL8252, Inflammation Research Center, Inflamex Excellence Laboratory, X. Bichat Faculty of Medicine, University of Paris-Cité, 75018, Paris, France
- Environment Biomonitoring Laboratory (LR01/ES14), Sciences Faculty of Bizerte, University of Carthage, Zarzouna, 7021, Bizerte, Tunisia
| | - Pham My-Chan Dang
- INSERM-U1149, CNRS-ERL8252, Inflammation Research Center, Inflamex Excellence Laboratory, X. Bichat Faculty of Medicine, University of Paris-Cité, 75018, Paris, France
| | - Hamadi Fetoui
- Laboratory of Toxicology-Microbiology and Environmental Health (UR17/ES06), Sciences Faculty of Sfax, Soukra Street Km 3.5, 3000, BP1171, Sfax, Tunisia
| | - Yassine Chtourou
- Laboratory of Toxicology-Microbiology and Environmental Health (UR17/ES06), Sciences Faculty of Sfax, Soukra Street Km 3.5, 3000, BP1171, Sfax, Tunisia
| | - Néziha Ghanem-Boughanmi
- Environmental Stress Risks Unit (UR17/ES20), Sciences Faculty of Bizerta, University of Carthage, Zarzouna, 7021, Bizerte, Tunisia
| | - Mossadok Ben-Attia
- Environment Biomonitoring Laboratory (LR01/ES14), Sciences Faculty of Bizerte, University of Carthage, Zarzouna, 7021, Bizerte, Tunisia
| | - Jamel El-Benna
- INSERM-U1149, CNRS-ERL8252, Inflammation Research Center, Inflamex Excellence Laboratory, X. Bichat Faculty of Medicine, University of Paris-Cité, 75018, Paris, France
| | - Abdelaziz Souli
- Environment Biomonitoring Laboratory (LR01/ES14), Sciences Faculty of Bizerte, University of Carthage, Zarzouna, 7021, Bizerte, Tunisia.
| |
Collapse
|
41
|
Ding X, Liu Z, Li H, Yue P, Jia Y, Li E, Lv N, Chen T, Fang R, Zhou H, Song X. Binding with HSP90β, cimifugin ameliorates fibrotic cataracts in vitro and in vivo by inhibiting TGFβ signaling pathways. Exp Eye Res 2024; 249:110127. [PMID: 39424221 DOI: 10.1016/j.exer.2024.110127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Fibrotic cataracts, the most frequent complications after phacoemulsification, cannot be cured by drugs in clinic. The primary mechanism underlying the disease is the epithelial-mesenchymal transition (EMT). Cimifugin is a natural monomer component of traditional Chinese medicines. Previous researches have demonstrated the effect of cimifugin inhibiting EMT in the lung. The purpose of this work is to evaluate the impact of cimifugin on EMT in the lens and elucidate its precise mechanism. The pathogenesis of fibrotic cataracts was simulated using TGFβ2-induced cell model of EMT and the injury-induced anterior subcapsular cataract animal model. Through H&E staining and immunofluorescence of mice eyeballs, we discovered that cimifugin can inhibit the expansion of fibrotic lesions in vivo. Furthermore, at mRNA and protein levels, we confirmed that cimifugin can allay EMT of lens epithelial cells (LECs) in vitro and in vivo. Additionally, the inhibition of cimifugin on the activation of TGFβ-related signaling pathways was certified by immunoblot. HSP90β, the target of cimifugin, was predicted by network pharmacology and verified by drug affinity responsive target stability, the cellular thermal shift assay, and microscale thermophoresis. Moreover, co-immunoprecipitation revealed the interaction between HSP90β and TGFβ receptor (TGFβR) II. Together, our findings showed that by weakening the binding of HSP90β and TGFβRII, cimifugin suppressed the TGFβ signaling pathways to alleviate fibrotic cataracts. Cimifugin is a promising medication for the treatment of fibrotic cataracts.
Collapse
Affiliation(s)
- Xuefei Ding
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Zhaochuan Liu
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Hailong Li
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nan kai University, Tianjin, 300071, China
| | - Peilin Yue
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Yuxuan Jia
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Enjie Li
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Ningxin Lv
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Ting Chen
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Rui Fang
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China
| | - Honggang Zhou
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nan kai University, Tianjin, 300071, China.
| | - Xudong Song
- Beijing Tongren Hospital, Beijing, 100730, China; Capital Medical University, Beijing, 100730, China; Beijing Tongren Eye Center, Beijing, 100730, China; Beijing Ophthalmology&Visual Sciences Key Lab, Beijing, 100730, China.
| |
Collapse
|
42
|
Wang X, Wu L, Luo D, He L, Wang H, Peng B. Mechanism of action of Salvia miltiorrhiza on avascular necrosis of the femoral head determined by integrated network pharmacology and molecular dynamics simulation. Sci Rep 2024; 14:28479. [PMID: 39558045 PMCID: PMC11574184 DOI: 10.1038/s41598-024-79532-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
Avascular necrosis of the femoral head (ANFH) is a progressive, multifactorial, and challenging clinical condition that often leads to hip dysfunction and deterioration. The pathogenesis of ANFH is complex, and there is no foolproof treatment strategy. Although some pharmacologic and surgical treatments have been shown to improve ANFH, the associated side effects and poor prognosis are of concern. Therefore, there is an urgent need to explore therapeutic interventions with superior efficacy and safety to improve the quality of life of patients with ANFH. Salvia miltiorrhiza (SM), a traditional Chinese medicine with a long history, is widely used for the treatment of cardiovascular and musculoskeletal diseases due to its multiple pharmacological activities. However, the molecular mechanism of SM for the treatment of ANFH is still unclear. Therefore, this study aimed to explore the potential targets and mechanisms of SM for the treatment of ANFH using network pharmacology and molecular modeling techniques. By searching multiple databases, we screened 52 compounds and 42 common targets involved in ANFH therapy and identified dan-shexinkum d, cryptotanshinone, tanshinone iia, and dihydrotanshinlactone as key compounds. Based on the protein-protein interaction (PPI) network, TP53, AKT1, EGFR, STAT3, BCL2, IL6, and TNF were identified as core targets. Subsequent enrichment analysis revealed that these targets were mainly enriched in the AGE-RAGE, IL-17, and TNF pathways, which were mainly associated with inflammatory responses, apoptosis, and oxidative stress. In addition, molecular docking and 100 nanoseconds molecular dynamics (MD) simulations showed that the bioactive compounds of SM had excellent affinity and binding strength to the core targets. Among them, dan-shexinkum d possessed the lowest binding free energy (-215.874 kcal/mol and - 140.277 kcal/mol, respectively) for AKT1 and EGFR. These results demonstrated the multi-component, multi-target, and multi-pathway intervention mechanism of SM in the treatment of ANFH, which provided theoretical basis and clues for further experimental validation and development of anti-ANFH drugs.
Collapse
Affiliation(s)
- Xiangjin Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Lijiao Wu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Dan Luo
- Basic Medical College of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Langyu He
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Hao Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Bo Peng
- Department of Respiratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China.
| |
Collapse
|
43
|
Jang B, Kim Y, Song J, Kim YW, Lee WY. Identifying Herbal Candidates and Active Ingredients Against Postmenopausal Osteoporosis Using Biased Random Walk on a Multiscale Network. Int J Mol Sci 2024; 25:12322. [PMID: 39596387 PMCID: PMC11594441 DOI: 10.3390/ijms252212322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Postmenopausal osteoporosis is a major global health concern, particularly affecting aging women, and necessitates innovative treatment options. Herbal medicine, with its multi-compound, multi-target characteristics, offers a promising approach for complex diseases. In this study, we applied multiscale network and random walk-based analyses to identify candidate herbs and their active ingredients for postmenopausal osteoporosis, focusing on their underlying mechanisms. A dataset of medicinal herbs, their active ingredients, and protein targets was compiled, and diffusion profiles were calculated to assess the propagation effects. Through correlation analysis, we prioritized herbs based on their relevance to osteoporosis, identifying the top candidates like Benincasae Semen, Glehniae Radix, Corydalis Tuber, and Houttuyniae Herba. Gene Set Enrichment Analysis (GSEA) revealed that the 49 core protein targets of these herbs were significantly associated with pathways related to inflammation, osteoclast differentiation, and estrogen metabolism. Notably, compounds such as falcarindiol from Glehniae Radix and tetrahydrocoptisine from Corydalis Tuber-previously unstudied for osteoporosis-were predicted to interact with inflammation-related proteins, including IL6, IL1B, and TNF, affecting key biological processes like apoptosis and cell proliferation. This study advances the understanding of herbal therapies for osteoporosis and offers a framework for discovering novel therapeutic agents.
Collapse
Affiliation(s)
- Boyun Jang
- IntegroMediLab Co., Ltd., Seoul 04626, Republic of Korea
| | - Youngsoo Kim
- IntegroMediLab Co., Ltd., Seoul 04626, Republic of Korea
| | - Jungbin Song
- Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Young-Woo Kim
- School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Won-Yung Lee
- School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
44
|
Yang HJ, Zhang T, Kim MJ, Hur HJ, Wu X, Jang DJ, Park S. Efficacy and Mechanism of Schisandra chinensis Fructus Water Extract in Alzheimer's Disease: Insights from Network Pharmacology and Validation in an Amyloid-β Infused Animal Model. Nutrients 2024; 16:3751. [PMID: 39519586 PMCID: PMC11547720 DOI: 10.3390/nu16213751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Schisandra chinensis Fructus (SCF) is a traditional medicinal herb containing lignans that improves glucose metabolism by mitigating insulin resistance. We aimed to investigate the therapeutic potential and action mechanism of SCF for Alzheimer's disease (AD) using a network pharmacology analysis, followed by experimental validation in an AD rat model. METHODS The biological activities of SCF's bioactive compounds were assessed through a network pharmacology analysis. An AD rat model was generated by infusing amyloid-β peptide (Aβ) (25-35) into the hippocampus to induce Aβ accumulation. The AD rats were fed either 0.5% dextrin (AD-Con) or 0.5% SCF (AD-SCF group) in a high-fat diet for seven weeks. The rats in the normal/control group received an Aβ (35-25) infusion (no Aβ deposition) and were fed a control diet (Normal-C). Aβ deposition, memory function, inflammation, and glucose/lipid metabolism were evaluated. RESULTS The network analysis revealed significant intersections between AD-related targets and bioactive SCF compounds, like gomisin A, schisandrin, and longikaurin A. Key AD genes prostaglandin-endoperoxide synthase-2 (PTGS2, cyclooxygenase-2) and acetylcholinesterase (AChE) were linked to SCF compounds. In the rats with AD induced by bilaterally infusing amyloid-β (25-35) into the hippocampus, the 0.5% SCF intake mitigated hippocampal amyloid-β deposition, neuroinflammation, memory deficits, and dysregulated glucose and lipid metabolism versus the AD controls. SCF reduced hippocampal AChE activity, inflammatory cytokine expression related to PTGS2, and malondialdehyde contents and preserved neuronal cell survival-related factors such as brain-derived neurotrophic factor and ciliary neurotrophic factor similar to normal rats. The neuroprotective effects validated the network analysis findings. CONCLUSIONS SCF could be a potential AD therapeutic agent by activating the parasympathetic nervous system to reduce hippocampal oxidative stress and inflammation, warranting further clinical investigations of its efficacy.
Collapse
Affiliation(s)
- Hye-Jeong Yang
- Food Functionality Research Division, Korea Food Research Institute, Wanju 55365, Republic of Korea; (H.-J.Y.); (M.-J.K.); (H.-J.H.)
| | - Ting Zhang
- Department of Food and Nutrition, Institute of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Republic of Korea; (T.Z.); (X.W.)
| | - Min-Jung Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju 55365, Republic of Korea; (H.-J.Y.); (M.-J.K.); (H.-J.H.)
| | - Haeng-Jeon Hur
- Food Functionality Research Division, Korea Food Research Institute, Wanju 55365, Republic of Korea; (H.-J.Y.); (M.-J.K.); (H.-J.H.)
| | - Xuangao Wu
- Department of Food and Nutrition, Institute of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Republic of Korea; (T.Z.); (X.W.)
| | - Dai-Ja Jang
- Food Functionality Research Division, Korea Food Research Institute, Wanju 55365, Republic of Korea; (H.-J.Y.); (M.-J.K.); (H.-J.H.)
| | - Sunmin Park
- Department of Food and Nutrition, Institute of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Republic of Korea; (T.Z.); (X.W.)
- Department of Bioconvergence, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
45
|
Khalil AM, Sabry OM, El-Askary HI, El Zalabani SM, Eltanany BM, Pont L, Benavente F, Mohamed AF, Fayek NM. Uncovering the therapeutic potential of green pea waste in breast cancer: a multi-target approach utilizing LC-MS/MS metabolomics, molecular networking, and network pharmacology. BMC Complement Med Ther 2024; 24:379. [PMID: 39482666 PMCID: PMC11526710 DOI: 10.1186/s12906-024-04669-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND PISUM SATIVUM: (PS) is a universal legume plant utilized for both human and animal consumption, particularly its seeds, known as green peas. The processing of PS in food industries and households produces a significant amount of waste that needs to be valorized. METHODS In this study, the metabolite profiles of the 70% ethanolic extracts of PS wastes, namely peels (PSP) and a combination of leaves and stems (PSLS), were investigated by liquid chromatography-electrospray ionization-quadrupole time-of-flight tandem mass spectrometry (LC-ESI-QTOF-MS/MS) followed by molecular networking. RESULTS Different classes of metabolites were identified, being flavonoids and their derivatives, along with phenolic acids, the most abundant categories. Additionally, a comprehensive network pharmacology strategy was applied to elucidate potentially active metabolites, key targets, and the pathways involved in cytotoxic activity against breast cancer. This cytotoxic activity was investigated in MCF-7 and MCF-10a cell lines. Results revealed that PSLS extract exhibited a potent cytotoxic activity with a good selectivity index (IC50 = 17.67 and selectivity index of 3.51), compared to the reference drug doxorubicin (IC50 = 2.69 µg/mL and selectivity index of 5.28). Whereas PSP extract appeared to be less potent and selective (IC50 = 32.92 µg/mL and selectivity index of 1.62). A similar performance was also observed for several polyphenolics isolated from the PSLS extract, including methyl cis p-coumarate, trans p-coumaric acid, and liquiritigenin/ 7-methyl liquiritigenin mixture. Methyl cis p-coumarate showed the most potent cytotoxic activity against MCF-7 cell line and the highest selectivity (IC50 = 1.18 µg/mL (6.91 µM) and selectivity index of 27.42). The network pharmacology study revealed that the isolated compounds could interact with several breast cancer-associated protein targets including carbonic anhydrases 1, 2, 4, 9, and 12, as well as aldo-keto reductase family 1 member B1, adenosine A3 receptor, protein tyrosine phosphatase non-receptor type 1, and estrogen receptor 2. CONCLUSION The uncovered therapeutic potential of PSLS and its metabolite constituents pave the way for an efficient and mindful PS waste valorization, calling for further in-vitro and in-vivo research.
Collapse
Affiliation(s)
- Asmaa M Khalil
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Omar M Sabry
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University, Cairo, 4645241, Egypt
| | - Hesham I El-Askary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Soheir M El Zalabani
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Basma M Eltanany
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Laura Pont
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA·UB), University of Barcelona, Barcelona, 08028, Spain
- Serra Húnter Program, Generalitat de Catalunya, Barcelona, 08007, Spain
| | - Fernando Benavente
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA·UB), University of Barcelona, Barcelona, 08028, Spain
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Faculty of Pharmacy, King Salman International University (KSIU), Ras Sedr, 46612, Egypt
| | - Nesrin M Fayek
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
46
|
Pan X, Jiang S, Zhang X, Wang Z, Wang X, Cao L, Xiao W. Recent strategies in target identification of natural products: Exploring applications in chronic inflammation and beyond. Br J Pharmacol 2024. [PMID: 39428703 DOI: 10.1111/bph.17356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/01/2024] [Accepted: 08/25/2024] [Indexed: 10/22/2024] Open
Abstract
Natural products are a treasure trove for drug discovery, especially in the areas of infection, inflammation and cancer, due to their diverse bioactivities and complex, and varied structures. Chronic inflammation is closely related to many diseases, including complex diseases such as cancer and neurodegeneration. Improving target identification for natural products contributes to elucidating their mechanism of action and clinical progress. It also facilitates the discovery of novel druggable targets and the elimination of undesirable ones, thereby significantly enhancing the productivity of drug discovery and development. Moreover, the rise of polypharmacological strategies, considered promising for the treatment of complex diseases, will further increase the demand for target deconvolution. This review underscores strategies for identifying natural product targets (NPs) in the context of chronic inflammation over the past 5 years. These strategies encompass computational methodologies for early target discovery and the anticipation of compound binding sites, proteomics-driven approaches for target delineation and experimental biology techniques for target validation and comprehensive mechanistic exploration.
Collapse
Affiliation(s)
- Xian Pan
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Shan Jiang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Xinzhuang Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Zhenzhong Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Liang Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Xiao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| |
Collapse
|
47
|
Miao Y, Liu W, Alsallameh SMS, Albekairi NA, Muhseen ZT, Butch CJ. Unraveling Cordia myxa's anti-malarial potential: integrative insights from network pharmacology, molecular modeling, and machine learning. BMC Infect Dis 2024; 24:1180. [PMID: 39427127 PMCID: PMC11490058 DOI: 10.1186/s12879-024-10078-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
Malaria is a potentially fatal infective illness caused due to parasites that belong to the Plasmodium genus, which are transferred to humans with the help of the stings of affected female Anopheles mosquitoes, and it persists as a serious public wellness problem worldwide. Cordia myxa is a medicinal plant that possesses various medicinal characteristics like antimicrobial, anti-inflammation, antioxidant, and antidiabetic activities, which makes it an important natural resource for the therapy of different maladies in traditional medicine. In this investigation, a certain network pharmacology method has been utilized to identify the potent active components, possible targets as well as signaling pathways present in C. myxa in relation to malaria therapy. The active compounds were submitted to molecular docking approaches to validate their successful activity against the potential targets. The study concluded that three constituents named cosmosiin, stigmastanol, robinetin, and quercetin were highly active and could regulate the expression of Interleukin 6 (IL6) and Cysteine-aspartic acid protease 3 (CASP3), which may act as a potential therapeutic target for malaria treatment. These analyses are validated by molecular dynamics simulation which reflects on the overall structural stability of the intermolecular conformation and interactions. These results can also be witnessed in simulation-based trajectories binding free energies, which concluded the significant role of electrostatic and van der Waals energies in total intermolecular interactions. Finally, we utilized machine learning to predict the anti-malarial activity of C. myxa compounds, comparing them with approved drugs. Using the Chemprop model and MAIP predictions, we assessed ten compounds, revealing their potential as lead anti-malarial agents. This study establishes a groundwork for comprehending the function of the anti-malaria action of C. myxa.
Collapse
Affiliation(s)
- Yufei Miao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Wenkang Liu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Sarah Mohammed Saeed Alsallameh
- Department of Medical Laboratories Techniques, College of Health and Medical Techniques, Gilgamesh Ahliya University Gau, Baghdad, Iraq
| | - Norah A Albekairi
- College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Ziyad Tariq Muhseen
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China.
- Department of Pharmacy, Al-Mustaqbal University, Hillah, Babylon, 51001, Iraq.
| | - Christopher J Butch
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China.
- State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
48
|
Al-Ajalein AA, Ibrahim N‘I, Fauzi MB, Mokhtar SA, Naina Mohamed I, Shuid AN, Mohamed N. Evaluating the Anti-Osteoporotic Potential of Mediterranean Medicinal Plants: A Review of Current Evidence. Pharmaceuticals (Basel) 2024; 17:1341. [PMID: 39458982 PMCID: PMC11510337 DOI: 10.3390/ph17101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Bones are biological reservoirs for minerals and cells, offering protection to the other organs and contributing to the structural form of the body. Osteoporosis is a prevalent bone condition that significantly impacts people's quality of life. Treatments utilizing natural products and medicinal plants have gained important attention in the management of osteoporosis and its associated implications, such as osteoporotic fractures. Even though thousands of plants grow in the Mediterranean region, the use of medicinal plants as an alternative therapy for osteoporosis is still limited. Methods: This article provides a comprehensive overview of seven Mediterranean medicinal plants that are used in osteoporosis and osteoporotic fractures in in vitro, in vivo, and clinical trials. The mechanism of action of the medicinal plants and their bioactive compounds against diseases are also briefly discussed. Results: The findings clearly indicate the ability of the seven medicinal plants (Ammi majus, Brassica oleracea, Ceratonia siliqua L., Foeniculum vulgare, Glycyrrhiza glabra, Salvia officinalis, and Silybum marianum) as anti-osteoporosis agents. Xanthotoxin, polyphenols, liquiritin, formononetin, silymarin, and silibinin/silybin were the main bioactive compounds that contributed to the action against osteoporosis and osteoporotic fractures. Conclusions: In this review, the Mediterranean medicinal plants prove their ability as an alternative agent for osteoporosis and osteoporotic fractures instead of conventional synthetic therapies. Thus, this can encourage researchers to delve deeper into this field and develop medicinal-plant-based drugs.
Collapse
Affiliation(s)
- Alhareth Abdulraheem Al-Ajalein
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| | - Nurul ‘Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Sabarul Afian Mokhtar
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| | - Ahmad Nazrun Shuid
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi Mara (UITM), Jalan Hospital, Sungai Buloh 47000, Malaysia;
| | - Norazlina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| |
Collapse
|
49
|
Chen Y, Tang Y, Li Y, Rui Y, Zhang P. Enhancing the Efficacy of Active Pharmaceutical Ingredients in Medicinal Plants through Nanoformulations: A Promising Field. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1598. [PMID: 39404324 PMCID: PMC11478102 DOI: 10.3390/nano14191598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
This article explores the emerging field of nanomedicine as a drug delivery system, aimed at enhancing the therapeutic efficacy of active pharmaceutical ingredients in medicinal plants. The traditional methods of applying medicinal plants present several limitations, such as low bioavailability, poor solubility, challenges in accurately controlling drug dosage, and inadequate targeting. Nanoformulations represent an innovative approach in drug preparation that employs nanotechnology to produce nanoscale particles or carriers, which are designed to overcome these limitations. Nanoformulations offer distinct advantages, significantly enhancing the solubility and bioavailability of drugs, particularly for the poorly soluble components of medicinal plants. These formulations effectively enhance solubility, thereby facilitating better absorption and utilization by the human body, which in turn improves drug efficacy. Furthermore, nanomedicine enables targeted drug delivery, ensuring precise administration to the lesion site and minimizing side effects on healthy tissues. Additionally, nanoformulations can regulate drug release rates, extend the duration of therapeutic action, and enhance the stability of treatment effects. However, nanoformulations present certain limitations and potential risks; their stability and safety require further investigation, particularly regarding the potential toxicity with long-term use. Nevertheless, nanomaterials demonstrate substantial potential in augmenting the efficacy of active pharmaceutical ingredients in medicinal plants, offering novel approaches and methodologies for their development and application.
Collapse
Affiliation(s)
- Yuhao Chen
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuying Tang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuanbo Li
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yukui Rui
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
- Tangshan Jinhai New Material Co., Ltd., Tangshan 063000, China
- Faculty of Resources and Environment, China Agricultural University, Shanghe County Baiqiao Town Science and Technology Courtyard, Jinan 250100, China
| | - Peng Zhang
- Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
50
|
Sun Z, Guo X, Li C, Ling J, Chang A, Zhao H, Zhuo X. Exploring the therapeutic mechanisms of resveratrol for treating arecoline-induced malignant transformation in oral epithelial cells: insights into hub targets. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8290-8305. [PMID: 38934557 DOI: 10.1002/jsfa.13664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Betel nut chewing is a significant risk factor for oral cancer due to arecoline, its primary active component. Resveratrol, a non-flavonoid polyphenol, possesses anti-cancer properties. It has been shown to inhibit arecoline-induced oral malignant cells in preliminary experiments but the underlying mechanism remains unclear. This research therefore aimed to explore the potential therapeutic targets of resveratrol in treating arecoline-induced oral cancer. METHODS Data mining identified common targets and hub targets of resveratrol in arecoline-induced oral cancer. Gene set variation analysis (GSVA) was used to score and validate the expression and clinical significance of these hub targets in head and neck cancer (HNC) tissues. Molecular docking analysis was conducted on the hub targets. The effect of resveratrol intervention on hub targets was verified by experiments. RESULTS Sixty-one common targets and 15 hub targets were identified. Hub targets were highly expressed in HNC and were associated with unfavorable prognoses. They played a role in HNC metastasis, epithelial-mesenchymal transition, and invasion. Their expression also affected immune cell infiltration and correlated negatively with sensitivity to chemotherapeutic agents such as bleomycin and docetaxel. Experiments demonstrated that resveratrol down-regulated the expression of the hub targets, inhibited their proliferation and invasion, and induced apoptosis. CONCLUSION Resveratrol inhibits the arecoline-induced malignant phenotype of oral epithelial cells by regulating the expression of some target genes, suggesting that resveratrol may be used not only as an adjuvant treatment for oral cancer, but also as an adjuvant for oral cancer prevention due to its low toxicity and high efficacy. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhen Sun
- Department of otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaopeng Guo
- Department of otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Changya Li
- Department of otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Junjun Ling
- Department of otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Aoshuang Chang
- Department of otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Houyu Zhao
- Department of otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xianlu Zhuo
- Department of otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|