1
|
Mayyas A, Al-Samydai A, Al-Karablieh N, Zalloum WA, Al-Tawalbeh D, Al-Mamoori F, Amr RA, Al Nsairat H, Carradori S, Al-Halaseh LK, Aburjai T. A phytotherapeutic approach to hinder the resistance against clindamycin by MRSA: in vitro and in silico studies. Future Sci OA 2025; 11:2458438. [PMID: 39895160 PMCID: PMC11792796 DOI: 10.1080/20565623.2025.2458438] [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: 04/03/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
AIMS This study investigates the potential effects of essential oils (EOs) in enhancing the efficacy of clindamycin against Methicillin-resistant Staphylococcus aureus (MRSA) using in vitro and computer simulations. The research seeks to identify essential oils that exhibit synergistic activity with clindamycin and determine their potential key active components. MATERIALS AND METHODS Essential oils commonly used in traditional medicine were tested for their antimicrobial activity against MRSA. The minimum inhibitory concentration (MIC) was determined using in vitro microdilution assays. A synergistic test with clindamycin was performed, and molecular docking studies evaluated the interaction between a key compound (trans-cinnamaldehyde) and MRSA protein. RESULTS EOs from Cinnamomum verum, Rosmarinus officinalis, Salvia officinalis, and Thymus vulgaris demonstrated significant inhibitory and synergistic activities against MRSA, standard strain, and human clinical isolates. Gas Chromatography/Mass Spectroscopy identified trans-cinnamaldehyde, eucalyptol, and thymol as prominent antibacterial compounds. Molecular docking studies confirmed trans-cinnamaldehyde's strong binding to MRSA's AgrA protein, elucidating its enhanced efficacy. CONCLUSION The study underscores the potential of plant-based therapies to augment the effectiveness of conventional antibiotics like clindamycin in combating MRSA and addressing antibiotic resistance by integrating traditional plant remedies with modern medical approaches.
Collapse
Affiliation(s)
- Amal Mayyas
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Ali Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Nehaya Al-Karablieh
- Department of Plant Protection, School of Agriculture, The University of Jordan, Amman, Jordan
- Hamdi Mango Centre for Scientific Research, The University of Jordan, Amman, Jordan
| | - Waleed A Zalloum
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Deniz Al-Tawalbeh
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Farah Al-Mamoori
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Zarqa University, Zarqa, Jordan
| | - Rula A. Amr
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Hamdi Al Nsairat
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Simone Carradori
- Department of Pharmacy “G. d’Annunzio”, University of Chieti-Pescara, Chieti, SC, Italy
| | - Lidia Kamal Al-Halaseh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mutah University, Al-Karak, Jordan
| | - Talal Aburjai
- Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
2
|
Phong NV, Kim HS, Zhao Y, Yeom E, Yang SY. Indirubin-3'-oxime as a dual-action agent: mitigating heat-induced male infertility in Drosophila melanogaster and inhibiting soluble epoxide hydrolase. J Enzyme Inhib Med Chem 2025; 40:2447719. [PMID: 39840826 PMCID: PMC11755746 DOI: 10.1080/14756366.2024.2447719] [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: 09/04/2024] [Revised: 11/29/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
This study investigated the potential of the indirubin-3'-oxime (I3O) compound to mitigate temperature-induced male infertility in Drosophila melanogaster. Elevated temperatures significantly reduced egg-hatching rates, but I3O supplementation improved these rates, suggesting it can partially restore fertility under heat stress. Additionally, I3O was found to inhibit soluble epoxide hydrolase (sEH), an enzyme involved in the metabolism of epoxyeicosatrienoic acids, which are vital for reproductive health. I3O exhibited sEH inhibitions with an IC50 value of 59.74 ± 0.41 µM. Enzyme kinetics revealed that I3O acts as a non-competitive inhibitor of sEH with a Ki value of 78.88 µM. Molecular docking showed strong interactions between I3O and key residues in the allosteric regions within the sEH enzyme, with a binding affinity of -9.2 kcal/mol. These interactions were supported by 100 ns molecular dynamics simulations, which confirmed the stability of the sEH-I3O complex.
Collapse
Affiliation(s)
- Nguyen Viet Phong
- Department of Biology Education, Teachers College and Institute for Phylogenomics and Evolution, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo-Sung Kim
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- School of Life Sciences, College of Natural Sciences, KNU-G LAMP Project Group, KNU-Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Yan Zhao
- School of Pharmacy, Yantai University, Yantai, PR China
| | - Eunbyul Yeom
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- School of Life Sciences, College of Natural Sciences, KNU-G LAMP Project Group, KNU-Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Seo Young Yang
- Department of Biology Education, Teachers College and Institute for Phylogenomics and Evolution, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
3
|
Liang H, Chen Z, Zhu M, Zhong J, Lin S, Chen J, Yuan J, Jiang P, Zhao X, Xiao Y. Efficacy and potential pharmacological mechanism of Astragalus-Salvia miltiorrhiza combination in diabetic nephropathy: integrating meta-analysis, network pharmacology, molecular docking, and experimental validation. Ren Fail 2025; 47:2466116. [PMID: 40015687 PMCID: PMC11869347 DOI: 10.1080/0886022x.2025.2466116] [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: 06/17/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a diabetes mellitus (DM)-induced complication that poses high morbidity and mortality risks. The Astragalus and Salvia miltiorrhiza couplet medicines (AS) are commonly employed in DN clinical treatment in China, but their clinical efficacy and potential pharmacological mechanisms are yet to be evaluated. MATERIAL AND METHODS A meta-analysis of 15 studies involving 1,443 patients was conducted. Furthermore, network pharmacology predicted components and targets, which were verified by molecular docking and in vivo validation. RESULTS In our meta-analysis, AS notably elevated clinical outcomes and renal function among patients with DN. Meanwhile, when the treatment duration exceeds 12 weeks, AS demonstrated a significant reduction in fasting blood glucose levels, indicating a time-dependent effect. Moreover, based on network pharmacology results, AS likely enhanced clinical outcomes by interacting with vital signaling pathways, including PI3K/Akt, MAPK, and NF-kappa B. Molecular docking studies have confirmed that PTGS2, the key therapeutic target of AS, can be closely combined with bioactive components GLY, quercetin, apigenin, and daidzein. Additionally, in vivo experiments have corroborated that AS can ameliorate renal function, UACR, and biomarkers associated with iron metabolism, such as GPX4, PTGS2, FTH1, and FTL1. CONCLUSION Through rigorous experimental validation, our study demonstrates AS's significant clinical efficacy in managing DN. Specifically, AS has been shown to enhance renal function, ameliorate renal fibrosis, and positively influence iron metabolism. Despite these promising outcomes, future research with a larger sample size must be conducted to further substantiate these findings.
Collapse
Affiliation(s)
- Huiyu Liang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zedong Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Mingmin Zhu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jingying Zhong
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shufan Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jianfeng Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jing Yuan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pingping Jiang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ya Xiao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
4
|
Li QJ, Shao HH, Zheng LL, Liu Q, Huo CC, Yi DR, Feng T, Cen S. Thonningianin A disrupts pA104R-DNA binding and inhibits African swine fever virus replication. Emerg Microbes Infect 2025; 14:2482697. [PMID: 40138179 PMCID: PMC11966994 DOI: 10.1080/22221751.2025.2482697] [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: 12/04/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
African swine fever is a highly lethal disease caused by the African swine fever virus (ASFV), posing a significant threat to the global pig industry, wherease no approved treatments are currently available. The ASFV DNA-binding protein, pA104R, plays a critical role in viral genome packaging and replication, making it a key target for drug discovery. Through structure-based virtual screening, we identified a polyphenolic compound, thonningianin A, which disrupts the pA104R-DNA binding and significantly inhibits ASFV replication. Mechanistic study revealed that thonningianin A binds to the DNA-binding region of pA104R, forming strong hydrogen bonds with H100 and occupying the vital DNA-binding residues K92, R94, and K97. In addition, we resolved the high-resolution (1.8 Å) structure of pA104R (PDB ID 9JS5), providing valuable insights for future drug screening. Together, these results demonstrate that thonningianin A holds great potential for the development of anti-ASFV drug, as a herb extract with favourable pharmacokinetic properties and safety.
Collapse
Affiliation(s)
- Quan-jie Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Hui-han Shao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Lin-lin Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
| | - Qian Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Chen-chao Huo
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Dong-rong Yi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Tao Feng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- CAMS Key Laboratory of Antiviral Drug Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Ren K, Zou L, Yang J, Wang Y, Min L. The Role of Autophagy and Cell Communication in COPD Progression: Insights from Bioinformatics and scRNA-seq. COPD 2025; 22:2444663. [PMID: 39991824 DOI: 10.1080/15412555.2024.2444663] [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: 09/05/2024] [Revised: 12/07/2024] [Accepted: 12/14/2024] [Indexed: 02/25/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by restricted airflow that leads to significant respiratory difficulties. This progressive disease often results in diminished pulmonary function and the onset of additional respiratory conditions. Autophagy, a critical cellular homeostasis mechanism, plays a significant role in the exacerbation of COPD. In this study, we utilized various bioinformatics tools to identify autophagy-related genes activated by smoking in individuals with COPD. Furthermore, we explored the immune landscape of COPD through these genes, analyzing cell communication patterns using scRNA-seq data. This analysis focused on key pathways between epithelial cells and other cellular subpopulations with different autophagy scores, essential for understanding the initiation and progression of COPD.
Collapse
Affiliation(s)
- Kaiqi Ren
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Lu Zou
- Yzngzhou Municipal Health Commission, Yangzhou, China
| | - Jingjing Yang
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yuxiu Wang
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Lingfeng Min
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
6
|
Gafforov Y, Bekić S, Yarasheva M, Mišković J, Živanović N, Chen JJ, Petri E, Abdullaev B, Rapior S, Lim YW, Abdullaev I, Abbasi AM, Ghosh S, Wan-Mohtar WAAQI, Rašeta M. Bioactivity profiling of Sanghuangporus lonicerinus: antioxidant, hypoglycaemic, and anticancer potential via in-vitro and in-silico approaches. J Enzyme Inhib Med Chem 2025; 40:2461185. [PMID: 39992291 PMCID: PMC11852365 DOI: 10.1080/14756366.2025.2461185] [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: 10/09/2024] [Revised: 01/11/2025] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
This study investigates the mycochemical profile and biological activities of hydroethanolic (EtOH), chloroform (CHCl3), and hot water (H2O) extracts of Sanghuangporus lonicerinus from Uzbekistan. Antioxidant capacity was assessed using 2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2'-azino-bis-3-ethylbenzothiazoline-6-sulfonic acid (ABTS), NO, and FRAP assays, and in vitro hypoglycaemic effects were evaluated through α-amylase and α-glucosidase inhibition. Antiproliferative potential was explored by analysing the binding affinities of EtOH and H2O extracts to estrogen receptor α (ERα), ERβ, androgen receptor (AR), and glucocorticoid receptor (GR), with molecular docking providing structural insights. LC-MS/MS analysis revealed solvent-dependent phenolic profiles, with the EtOH extract containing the highest total phenolic content (143.15 ± 6.70 mg GAE/g d.w.) and the best antioxidant capacity. The EtOH extract showed significant hypoglycaemic effects, with 85.29 ± 5.58% inhibition of α-glucosidase and 41.21 ± 0.79% inhibition of α-amylase. Moderate ERβ binding suggests potential for estrogen-mediated cancer therapy, while strong AKR1C3 inhibition by the EtOH extract supports its therapeutic potential.
Collapse
Affiliation(s)
- Yusufjon Gafforov
- Central Asian Center of Development Studies, New Uzbekistan University, Tashkent, Uzbekistan
- Mycology Laboratory, Institute of Botany, Academy of Sciences of Republic of Uzbekistan, Tashkent, Uzbekistan
| | - Sofija Bekić
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Manzura Yarasheva
- Microbiology Laboratory, Navruz International Corp. LLC, Kibray, Uzbekistan
| | - Jovana Mišković
- Department of Biology and Ecology, Faculty of Sciences, ProFungi Laboratory, University of Novi Sad, Novi Sad, Serbia
| | - Nemanja Živanović
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Jia Jia Chen
- College of Landscape Architecture, Jiangsu Vocational College of Agriculture and Forestry, Zhenjiang, China
| | - Edward Petri
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Bekhzod Abdullaev
- Central Asian Center of Development Studies, New Uzbekistan University, Tashkent, Uzbekistan
| | - Sylvie Rapior
- CEFE, Univ Montpellier, CNRS, EPHE, IRD, Natural Substances and Chemical Mediation Team, Montpellier, France
- Laboratory of Botany, Phytochemistry and Mycology, Faculty of Pharmacy, Univ Montpellier, Montpellier, France
| | - Young Won Lim
- School of Biological Sciences and Institute of Biodiversity, Seoul National University, Seoul, Republic of Korea
| | | | - Arshad Mehmood Abbasi
- Department of Environmental Sciences, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Soumya Ghosh
- Natural and Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Wan Abd Al Qadr Imad Wan-Mohtar
- Functional Omics and Bioprocess Development Laboratory, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Milena Rašeta
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
- Department of Biology and Ecology, Faculty of Sciences, ProFungi Laboratory, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
7
|
Zhu J, Xu Z, Liu X. Chemical composition, antioxidant activities, and enzyme inhibitory effects of Lespedeza bicolour Turcz. essential oil. J Enzyme Inhib Med Chem 2025; 40:2460053. [PMID: 39912419 PMCID: PMC11803819 DOI: 10.1080/14756366.2025.2460053] [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: 08/12/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Lespedeza bicolour Turcz. is a traditional medicinal plant with a wide range of ethnomedicinal values. The main components of L. bicolour essential oil (EO) were β-pinene (15.41%), β-phellandrene (12.43%), and caryophyllene (7.79%). The EO of L. bicolour showed antioxidant activity against ABTS radical and DPPH radical with an IC50 value of 0.69 ± 0.03 mg/mL and 10.44 ± 2.09 mg/mL, respectively. The FRAP antioxidant value was 81.96 ± 6.17 μmol/g. The EO had activities against acetylcholinesterase, α-glucosidase, and β-lactamase with IC50 values of 309.30 ± 11.16 μg/mL, 360.47 ± 35.67 μg/mL, and 27.54 ± 1.21 μg/mL, respectively. Molecular docking showed methyl dehydroabietate docked well with all tested enzymes. Sclareol and (+)-borneol acetate showed the strongest binding affinity to α-glucosidase and β-lactamase, respectively. The present study provides a direction for searching enzyme inhibitors for three tested enzymes and shows L. bicolour EO possesses the potential to treat a series of diseases.
Collapse
Affiliation(s)
- Jiadong Zhu
- SDU‐ANU Joint Science College, Shandong University, Weihai, China
| | - Ziyue Xu
- SDU‐ANU Joint Science College, Shandong University, Weihai, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Kowloon, China
| | - Xu Liu
- Marine College, Shandong University, Weihai, China
| |
Collapse
|
8
|
El-Wakil MH, Ghazala RA, El-Dershaby HA, Drozdowska D, Wróbel-Tałałaj A, Parzych C, Ratkiewicz A, Kolesińska B, Abd El-Razik HA, Soliman FSG. Rational design, synthesis, and molecular modelling insights of dual DNA binders/DHFR inhibitors bearing arylidene-hydrazinyl-1,3-thiazole scaffold with apoptotic and anti-migratory potential in breast MCF-7 cancer cells. J Enzyme Inhib Med Chem 2025; 40:2468353. [PMID: 40035286 PMCID: PMC11881662 DOI: 10.1080/14756366.2025.2468353] [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: 01/11/2025] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
In light of searching for new breast cancer therapies, DNA-targeted small molecules were rationally designed to simultaneously bind DNA and inhibit human dihydrofolate reductase (hDHFR). Fourteen new arylidene-hydrazinyl-1,3-thiazoles (5-18) were synthesised and their dual DNA groove binding potential and in vitro hDHFR inhibition were performed. Two compounds, 5 and 11, proved their dual efficacy. Molecular docking and molecular dynamics simulations were performed for those active derivatives to explore their mode of binding and stability of interactions inside DHFR active site. Anti-breast cancer activity was assessed for 5 and 11 on MCF-7 cells using MTX as reference. IC50 measurements revealed that both compounds were more potent and selective than MTX. Cytotoxicity was examined against normal skin fibroblasts to examine safety and selectivity Moreover, mechanistic studies including apoptosis induction and wound healing were performed. Further in silico ADMET assessment was conducted to determine their eligibility as drug leads suitable for future optimisation and development.
Collapse
Affiliation(s)
- Marwa H. El-Wakil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rasha A. Ghazala
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hadeel A. El-Dershaby
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Danuta Drozdowska
- Department of Organic Chemistry, Medical University of Bialystok, Bialystok, Poland
| | | | - Cezary Parzych
- Department of Physical Chemistry, University of Bialystok, Institute of Chemistry, Bialystok, Poland
| | - Artur Ratkiewicz
- Department of Physical Chemistry, University of Bialystok, Institute of Chemistry, Bialystok, Poland
| | - Beata Kolesińska
- Institute of Organic Chemistry, Lodz University of Technology, Lodz, Poland
| | - Heba A. Abd El-Razik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Farid S. G. Soliman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
9
|
Vijayan S, Margesan T. The potential of Abrus precatorius leaves in arthritis alleviation computational approaches through lC-MS analysis. Future Sci OA 2025; 11:2483131. [PMID: 40131221 PMCID: PMC11938972 DOI: 10.1080/20565623.2025.2483131] [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: 03/02/2024] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
AIM This study explores the therapeutic potential of Abrus precatorius leaves in arthritis treatment using computational methods and LC-MS analysis. METHODS The plant material was taxonomically authenticated, and phytochemical analysis identified bioactive compounds such as alkaloids, flavonoids, and triterpenoids. RESULTS Swiss ADME analysis confirmed that multiple compounds complied with Lipinski's Rule of Five, while OSIRIS software indicated minimal toxicity. PASS analysis predicted anti-inflammatory and antioxidant activities. Molecular docking simulations of Abrine with key rheumatoid arthritis (RA) targets revealed strong binding affinities, suggesting potential mechanisms for RA treatment. CONCLUSION This research highlights the medicinal potential of Abrus precatorius leaves and emphasizes the importance of computational tools in understanding their pharmacological properties for arthritis management.
Collapse
Affiliation(s)
- Sukanya Vijayan
- Department of Pharmacognosy, SRM College of Pharmacy, SRMIST, Chengalpattu, Tamil Nadu, India
| | - Thirumal Margesan
- Department of Pharmacognosy, SRM College of Pharmacy, SRMIST, Chengalpattu, Tamil Nadu, India
| |
Collapse
|
10
|
Lou J, Wu X, Ji W, Yu J, Xu Y, Xiao W, Lu W, Xin K, Chen T, Tang Q, Liang G, Gao Y, Wu D. N-Terminal random curl-tandam α-helical peptide 7W: A potent antibacterial and anti-inflammatory dual-effect agent through tryptophan substitution. Eur J Med Chem 2025; 292:117686. [PMID: 40319576 DOI: 10.1016/j.ejmech.2025.117686] [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: 01/10/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
This study investigates the impact of tryptophan substitution on the properties of the Medisin family peptide MS-PT. By substituting hydrophobic amino acids in MS-PT1 with tryptophan, a series of derivative peptides were synthesized. Among them, the 7W peptide stood out with its unique N-terminal random curl and α-helix structure. In vitro, 7W effectively inhibited the secretion of pro-inflammatory cytokines like IL-6 and TNF-α in LPS-induced Membrane-Proximal Macrophages (MPMs) by blocking the MAPK/NF-κB signaling pathway. It also exhibited stronger antimicrobial activity against Gram-positive bacteria compared to the parent peptide MS-PT1, with good safety as indicated by a low hemolysis rate. In vivo, in the CLP-induced sepsis mouse model, 7W alleviated lung and liver injury, suppressed the expression of inflammatory factors in serum and tissues, and had a relatively long plasma half-life of 46.8 h. Mechanistically, 7W interacted preferentially with bacterial mimic membranes and LPS, and its anti-inflammatory effect might be mediated by binding to TLR4. These findings not only clarify the role of tryptophan substitution in modulating peptide properties but also offer a new strategy for the development of multifunctional antimicrobial peptides, suggesting that 7W has great potential as a therapeutic agent for sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Jietao Lou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xinyi Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Department of Pharmacy, The First People's Hospital of Jiande, Hangzhou, 311600, China
| | - Wenwen Ji
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiaye Yu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yanyan Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wanyang Xiao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Weijie Lu
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Kaiyun Xin
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Tianbao Chen
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yitian Gao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Di Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
11
|
Alrehaili J, Anwer R, Qais FA. Nalidixic acid inhibits the aggregation of HSA: Utilizing the molecular simulations to uncover the detailed insights. Comput Biol Chem 2025; 117:108415. [PMID: 40031372 DOI: 10.1016/j.compbiolchem.2025.108415] [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: 01/22/2025] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
Neurodegenerative diseases such as Parkinson's and Alzheimer's lead to the gradual decline of the nervous system, resulting in cognitive and motor impairments. With an aging population, the prevalence and associated healthcare costs are anticipated to rise. Misfolded protein aggregates are central to these diseases, disrupting cellular function and causing neuronal death. Preventing these toxic aggregates could preserve neurons and slow disease progression. Understanding how to inhibit protein aggregation is crucial for developing effective treatments. We explored the effect of nalidixic acid (NA) on protein aggregation using human serum albumin (HSA) as model protein. In vitro assays demonstrated that NA significantly reduced ThT fluorescence by 47.10 % and decreased turbidity by 63.07 %. NA also protected the protein's hydrophobic surfaces. The α-helical content of HSA dropped from 56.23 % to 11.43 % but was restored to 38.53 % with NA. We then utilized advanced molecular simulations to understand the kinetics and mechanism of aggregation inhibition by NA. Binding studies showed that NA attaches to HSA's subdomain IIA with a binding energy of -7.8 kcal/mol through hydrogen bonds, Van der Waals forces, and hydrophobic interactions. Molecular simulations confirmed the stability of HSA-NA complex. Additionally, NA increased solvent accessibility of HSA282-292 oligomers, reduced hydrogen bonding, and prevented β-sheet formation. Compared to existing anti-aggregation strategies, NA offers a promising alternative with its potential therapeutic applications in neurodegenerative diseases by stabilizing protein structures and preventing misfolding. These findings highlight NA's potential as a candidate for inhibiting protein aggregation and offer insights for therapeutic approaches. Further experimental studies utilizing in vivo models are needed to validate the anti-aggregation potential of NA.
Collapse
Affiliation(s)
- Jihad Alrehaili
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 4233-13317, Saudi Arabia
| | - Razique Anwer
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 4233-13317, Saudi Arabia
| | - Faizan Abul Qais
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
12
|
Yoon D, Lee H. In silico discovery of novel compounds for FAK activation using virtual screening, AI-based prediction, and molecular dynamics. Comput Biol Chem 2025; 117:108420. [PMID: 40157227 DOI: 10.1016/j.compbiolchem.2025.108420] [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: 11/24/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 04/01/2025]
Abstract
Focal Adhesion Kinase (FAK) is a non-receptor tyrosine kinase that plays a crucial role in cell proliferation, migration, and signal transduction. FAK is overexpressed in metastatic and advanced-stage cancers, where it is considered a key kinase in cancer growth and metastasis. However, recent research has revealed that FAK activity decreases in various diseases. we aimed to identify compounds that could enhance FAK activity using structure-based virtual screening and artificial intelligence models from a vast chemical database. We began with an extensive chemical database containing over 10 million compounds and used our newly developed pipeline to screen candidate molecules. To select compounds structurally similar to ZINC40099027 (ZN27), a known FAK activator, we calculated Tanimoto Similarity scores and chose compounds with a score of at least 0.8. Clustering was performed using K-means based on the molecular properties. Subsequently, we utilized docking simulation, deep learning and SAScorer to evaluate and predict the protein-ligand docking affinity and physicochemical properties of the candidate compounds. The deep learning models were selected as state-of-the-art models: GLAM predicts the blood-brain barrier permeability of FAK, and elEmBERT predicts the potential toxicity of compound. The combined results were used to create an evaluation matrix. We selected 10 promising candidate compounds from the initial dataset of 10 million. To evaluate the stability of these top 10 candidate compounds in interaction with the FAK protein, we conducted Molecular Dynamics (MD) simulations. We performed a molecular dynamics simulation for a total of 50 ns and identified the top three promising candidate compounds.
Collapse
Affiliation(s)
- Deokhyeon Yoon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hyunsu Lee
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
13
|
Gupta AK, Sahu Y, Pal D, Kumar N, Jain SK. Assessing novel analogues of nilutamide as a human androgen receptor antagonist: A detailed investigation of drug design using a bioisosteric methodology including ADMET profiling, molecular docking studies and molecular dynamics simulation. Comput Biol Chem 2025; 117:108424. [PMID: 40112513 DOI: 10.1016/j.compbiolchem.2025.108424] [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: 12/31/2024] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Cancer is a significant health and economic concern worldwide. Prostate cancer (PC) ranks as the fourth leading cause of global death and is the second most prevalent malignancy in males. Androgens are essential for the progress and growth of the prostate gland. PC is caused by androgens binding to receptors, which activates genes that promotes the development of PC. Nilutamide (NLM) is an antiandrogen medicine used in the treatment of PC. However, throughout treatment, it induces various toxicities and leads to resistance in patients. The objective of the work was to designed and evaluated safer NLM analogues using computational approaches with optimized pharmacokinetic profiles and less toxicity. Newer bioisosteres of the designed NLM analogues and their ADMET scores were calculated using the MolOpt and ADMETlab 3.0 tools, respectively. We conducted docking investigations of the designed ligands using AutoDock Vina software. The MolOpt web server produces 1575 bioisosteres of NLM using the scaffold transformation method. The 47 bioisosteres were selected based on pharmacokinetic profiles, drug likeness (DL) and drug score (DS) prediction scores and were determined to be optimum to excellent in comparison to NLM. The analogues NLM28, NLM31, NLM34, NLM38, NLM40, NLM44, NLM45, and NLM47 exhibited favorable interactions and docking scores with the protein (PDB ID: 2AM9). The molecular dynamics (MD) simulation results revealed that the NLM34 and NLM40 complexes were found stable during the 100 ns run. The findings indicate that the NLM analogues, particularly NLM34 and NLM40 have the potential to be used as promising antiandrogen agents for PC therapy.
Collapse
Affiliation(s)
- Ajay Kumar Gupta
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India
| | - Yogita Sahu
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India
| | - Dipti Pal
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles' College of Pharmacy, Udaipur, Rajasthan 313001, India
| | - Sanmati Kumar Jain
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India.
| |
Collapse
|
14
|
Ortega-Vallbona R, Talavera-Cortés D, Carpio LE, Coto Palacio J, Roncaglioni A, Garcia De Lomana M, Gadaleta D, Benfenati E, Gozalbes R, Serrano-Candelas E. DockTox: Targeting molecular initiating events in organ toxicity through molecular docking. Toxicology 2025; 515:154155. [PMID: 40252946 DOI: 10.1016/j.tox.2025.154155] [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: 02/10/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Adverse Outcome Pathways (AOPs) in toxicology describe the sequence of key events from chemical exposure to adverse outcomes, facilitating the development of predictive models. The EU ONTOX project uses this framework to predict liver, developmental brain, and kidney toxicity without animal testing. Focusing on Molecular Initiating Events (MIEs), more concretely on the interaction of chemicals with key proteins, we have developed an automated workflow for docking small molecules onto over 20 pre-processed protein structures, implemented in the online tool DockTox. This tool generates conformers of small molecules, performs docking on MIE-associated proteins, and provides binding energy, interacting residues, and interaction maps. Additionally, it compares the interactions to a reference list of known ligands, producing an interaction fraction as an additional similarity measure. Evaluation of the docking workflow's predictive performance on Peroxisome Proliferator-Activated Receptor α (PPARα) showed that interaction fraction values are more informative than binding energy alone for distinguishing binders from non-binders. This unique feature enhances the understanding of target protein interactions. DockTox supports the virtual screening of small molecules targeting MIE-associated proteins, offering insights into binding energies and interaction profiles. It is a valuable tool for anticipating adverse outcomes from chemical exposure in a tiered risk assessment approach.
Collapse
Affiliation(s)
| | | | - Laureano E Carpio
- ProtoQSAR SL, Parque Tecnológico de Valencia, Paterna, Spain; Moldrug AI Systems SL, Parque Tecnológico de Valencia, Paterna, Spain
| | | | - Alessandra Roncaglioni
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marina Garcia De Lomana
- Bayer AG, Machine Learning Research, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Domenico Gadaleta
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Rafael Gozalbes
- ProtoQSAR SL, Parque Tecnológico de Valencia, Paterna, Spain; Moldrug AI Systems SL, Parque Tecnológico de Valencia, Paterna, Spain
| | | |
Collapse
|
15
|
Awad-Igbaria Y, Sakas R, Milhem L, Fishboom T, Ben-Menashe A, Edelman D, Shamir A, Soustiel JF, Palzur E. Mitochondrial translocator-protein ligand etifoxine reduces pain symptoms and protects against motor dysfunction development following peripheral nerve injury in rats. Neuropharmacology 2025; 273:110456. [PMID: 40189017 DOI: 10.1016/j.neuropharm.2025.110456] [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: 02/14/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025]
Abstract
Peripheral nerve injury enhances mitochondrial translocator protein (TSPO) expression in the spinal cord and dorsal root ganglia (DRG), which is associated with neuroinflammation and mitochondrial dysfunction contributing to chronic pain development. Here, we investigate the effect of TSPO ligand Etifoxine, on the development of chronic pain and motor dysfunction following sciatic nerve injury. Mechanical and thermal sensitivity, as well as motor function, were measured in rats before and after sciatic nerve crush (SNC). Rats were treated with the Etifoxine (50 mg/kg, twice daily) for one week. At the end of the experiment, RT-PCR and immunohistochemistry (IHC) were performed to assess mitochondrial stress and neuroinflammation. Additionally, high-resolution respirometry (O2k) was used to evaluate mitochondrial function in the spinal cord following mitochondrial permeability transition pore (mPTP) induction by Ca2+. Etifoxine treatment post-SNC alleviated mechanical and thermal hypersensitivity, as well as motor dysfunction in rats. In addition, Etifoxine treatment modulates neuroinflammation and mitochondrial stress. Specifically, we found a significant reduction in microglia presence and the transcription of pro-inflammatory cytokines (TNFα, IL-6, IL-1β) in the DRG and spinal cord of the SNC/etifoxine-treated group. Furthermore, Etifoxine treatment prevent the decline in mitochondrial respiration, including non-phosphorylation, ATP-linked respiration, and maximal respiration, after mPTP induction by Ca2+. Our findings suggest that TSPO-ligand Etifoxine protects against motor dysfunction and the development of chronic pain by reducing neuroinflammation and apoptosis in the DRG and spinal cord. Importantly, the beneficial effects of TSPO-ligands are reflected in the restoration of the mitochondrial function under challenging conditions.
Collapse
Affiliation(s)
- Yaseen Awad-Igbaria
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel; Research Institute of Galilee Medical Center, Nahariya, Israel.
| | - Reem Sakas
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel; Research Institute of Galilee Medical Center, Nahariya, Israel
| | - Lama Milhem
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel; Research Institute of Galilee Medical Center, Nahariya, Israel
| | - Tom Fishboom
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel; Research Institute of Galilee Medical Center, Nahariya, Israel
| | - Aviv Ben-Menashe
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel; Research Institute of Galilee Medical Center, Nahariya, Israel
| | - Doron Edelman
- Department of Neurosurgery, Sourasky Medical Center, Tel-Aviv, Israel
| | - Alon Shamir
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Psychobiology Research Laboratory, Mazor Mental Health Center, Akko, Israel
| | - Jean F Soustiel
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel; Research Institute of Galilee Medical Center, Nahariya, Israel; Department of Neurosurgery, Galilee Medical Center, Nahariya, Israel
| | - Eilam Palzur
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel; Research Institute of Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
16
|
Xie J, Zhong S, Huang D, Shao W. PocketDTA: A pocket-based multimodal deep learning model for drug-target affinity prediction. Comput Biol Chem 2025; 117:108416. [PMID: 40073710 DOI: 10.1016/j.compbiolchem.2025.108416] [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: 11/05/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025]
Abstract
Drug-target affinity prediction is a fundamental task in the field of drug discovery. Extracting and integrating structural information from proteins effectively is crucial to enhance the accuracy and generalization of prediction, which remains a substantial challenge. This paper proposes a pocket-based multimodal deep learning model named PocketDTA for drug-target affinity prediction, based on the principle of "structure determines function". PocketDTA introduces the pocket graph structure that encodes protein residue features pretrained using a biological language model as nodes, while edges represent different protein sequences and spatial distances. This approach overcomes the limitations of lack of spatial information in traditional prediction models with only protein sequence input. Furthermore, PocketDTA employs relational graph convolutional networks at both atomic and residue levels to extract structural features from drugs and proteins. By integrating multimodal information through deep neural networks, PocketDTA combines sequence and structural data to improve affinity prediction accuracy. Experimental results demonstrate that PocketDTA outperforms state-of-the-art prediction models across multiple benchmark datasets by showing strong generalization under more realistic data splits and confirming the effectiveness of pocket-based methods for affinity prediction.
Collapse
Affiliation(s)
- Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Shengsheng Zhong
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Dingkai Huang
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Wei Shao
- Scientific Research Management Department, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
17
|
Zhao L, Zhang Z, Su H, Zhang W, Sun J, Li Y, Teng M. Molecular docking-QSAR-Kronecker-regularized least squares-based multiple machine learning for assessment and prediction of PFAS-protein binding interactions. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138069. [PMID: 40179788 DOI: 10.1016/j.jhazmat.2025.138069] [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: 10/02/2024] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Ubiquitous per- and poly-fluoroalkyl substances (PFAS) threaten human's health and attract worldwide attention. PFAS-mediated toxicity involves adverse effects of PFAS on proteins, and assessment of PFAS-protein binding interactions helps to explain PFAS' adverse effects on human health. In-silico modeling can generate information and decrease experimental costs. Accordingly, in this study, molecular docking was used to determine the binding affinities of 430 PFAS with human serum albumin (HSA), peroxisome proliferator-activated receptor gamma (PPARγ), and transthyretin (TTR). Specifically, analytic hierarchy process, fuzzy comprehensive evaluation, and quantitative structure-activity relationship model were used to assess and predict the binding affinities between PFAS and HSA, PPARγ, and TTR. The binding patterns were determined by defining "PEOE_RPC-, E_vdw, MNDO_LUMO, and vsurf features" as key factors related to charge, energy and shape characteristic of PFAS. Finally, Kronecker-regularized least squares (Kron-RLS) model was applied to predict the binding affinities between PFAS- and G protein-coupled receptor 40 (GPR40), as a new target for prediction. Results showed that the Kron-RLS model exhibited good performance and generated precise predictions (R2 = 0.94). In conclusion, this study demonstrated that computational simulations could be used to aid the scientific management of the growing number of PFAS, and could be broadened to include a wide range of environmental contaminations.
Collapse
Affiliation(s)
- Lihui Zhao
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China; College of geoexploration science and technology, Jilin University, Changchun 130026, China
| | - Zixuan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Hailei Su
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Wenjun Zhang
- Key Laboratory of Integrated Regulation and Resources Development on Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Jiaqi Sun
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yunxia Li
- College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Miaomiao Teng
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China.
| |
Collapse
|
18
|
Ijod G, Nawawi NIM, Qoms MS, Rashedi Ismail Fitry M, Rahim MHA, Charalampopoulos D, Sulaiman R, Adzahan NM, Azman EM. Synergistic effects of intermolecular copigmentation and high-pressure processing on stabilizing mangosteen pericarp anthocyanins. Food Chem 2025; 480:143888. [PMID: 40120312 DOI: 10.1016/j.foodchem.2025.143888] [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: 01/11/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
The mangosteen pericarp contains unstable non-acylated ACNs, rendering it prone to degradation. Therefore, intermolecular copigmentation of semi-purified ACNs (SPA) with tartaric acid (SPA-TA), sinapic acid (SPA-SA), catechin (SPA-CE), and sucrose (SPA-SU) in 1:5 and 1:10 M ratios were used to increase their stability during storage for 77 days at 25 ± 1 °C in pH 3 buffer solution. The SPA-TA1:5 complex showed the significant highest stability of total monomeric ACN content (TMAC) with a half-life (t1/2) = 56.9 days, cyanidin-3-O-sophoroside (C3S) with t1/2 = 48.1 days and color retention (69.80 %) compare to SPA with TMAC (t1/2 = 37.4), C3S (t1/2 = 13.3) and color retention (57.2 %) after 49 days (p < 0.05). The thermal stability of SPA-TA1:5 at 60 °C improved after high-pressure processing (HPP) at 300 and 500 MPa for 10 min. Fourier-transform infrared spectroscopy (FT-IR) and molecular docking indicate copigmentation interactions, including hydrogen bonding and π-π interactions. This study demonstrates a sustainable method to stabilize non-acylated ACNs, offering a natural alternative to synthetic dyes for food and beverage applications.
Collapse
Affiliation(s)
- Giroon Ijod
- Department of Food Technology, Faculty of Food Science and Technology, Jalan Universiti 1, 43400, Universiti Putra Malaysia, Selangor, Malaysia
| | - Nur Izzati Mohamed Nawawi
- Department of Food Technology, Faculty of Food Science and Technology, Jalan Universiti 1, 43400, Universiti Putra Malaysia, Selangor, Malaysia
| | - Mohammed S Qoms
- Department of Food Science, Faculty of Food Science and Technology, Jalan Universiti 1, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Mohammad Rashedi Ismail Fitry
- Department of Food Technology, Faculty of Food Science and Technology, Jalan Universiti 1, 43400, Universiti Putra Malaysia, Selangor, Malaysia
| | - Muhamad Hafiz Abd Rahim
- Department of Food Science, Faculty of Food Science and Technology, Jalan Universiti 1, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | | | - Rabiha Sulaiman
- Department of Food Technology, Faculty of Food Science and Technology, Jalan Universiti 1, 43400, Universiti Putra Malaysia, Selangor, Malaysia
| | - Noranizan Mohd Adzahan
- Department of Food Technology, Faculty of Food Science and Technology, Jalan Universiti 1, 43400, Universiti Putra Malaysia, Selangor, Malaysia
| | - Ezzat Mohamad Azman
- Department of Food Technology, Faculty of Food Science and Technology, Jalan Universiti 1, 43400, Universiti Putra Malaysia, Selangor, Malaysia.
| |
Collapse
|
19
|
Fu J, Wang Q, Fu H, Wang D, Mayard A, Pan W, Vincent SP. Design, synthesis, and discovery of cinnamoyl amide derivatives as potent NagZ inhibitors with antibacterial activity. Eur J Med Chem 2025; 291:117622. [PMID: 40249971 DOI: 10.1016/j.ejmech.2025.117622] [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: 03/04/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025]
Abstract
β-N-acetylglucosaminidase (NagZ) plays an important role in the bacterial cell wall biosynthetic pathway. Inhibiting its activity could potentially impede bacterial growth. We report a study on the design and synthesis of cinnamoyl amides derived from rosmarinic acid (RA), and their enzymatic, antibacterial activity against NagZ and Pseudomonas aeruginosa respectively. In vitro enzyme activity determination showed that the best synthetic RA analogues displayed higher inhibitory activity than that of parent RA, in the same range than the most potent NagZ inhibitors reported so far. Remarkably, compounds 11h and Br-6 displayed interesting binding affinity values with Ki=3.3 ± 0.5 and 3.5 ± 1.0 μM, respectively. Docking simulations evidenced significant binding interactions of cinnamoyl amide derivatives with the active site of NagZ. Moreover, kinetic evaluations indicated these compounds displayed competitive behavior. Additionally, MICs of 11h and Br-6 combined with two β-Lactam antibiotics (imipenem and ceftazidime) were evaluated against P. aeruginosa by microdilution checkerboard assay, establishing that antibacterial agents show synergistic effects. In vivo antibacterial efficacy assay using a full-thickness skin defect model with P. aeruginosa infection confirmed these observations.
Collapse
Affiliation(s)
- Jian Fu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine/National Engineering Technology Research Center for Miao Medicine/Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, Guiyang, 550025, China; Department of Chemistry, University of Namur, NARILIS (Namur Research Institute for Life Sciences), Rue de Bruxelles 61, 5000, Namur, Belgium
| | - Qingqing Wang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine/National Engineering Technology Research Center for Miao Medicine/Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, Guiyang, 550025, China
| | - Huixiao Fu
- The First People's Hospital of Guiyang, Guiyang, 550002, China
| | - Dan Wang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine/National Engineering Technology Research Center for Miao Medicine/Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, Guiyang, 550025, China
| | - Aurélie Mayard
- Research Unit in Biology of Microorganisms (URBM), NARILIS (Namur Research Institute for Life Sciences), University of Namur, Rue de Bruxelles 61, 5000, Namur, Belgium
| | - Weidong Pan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, China.
| | - Stéphane P Vincent
- Department of Chemistry, University of Namur, NARILIS (Namur Research Institute for Life Sciences), Rue de Bruxelles 61, 5000, Namur, Belgium.
| |
Collapse
|
20
|
Billen M, Reynders S, Claes S, Kleinboelting S, Rozenski J, Bulai RG, Rocca E, Homer NZM, Webster SP, Kaminski TP, Lescrinier E, Schols D, Verwilst P. Discovery and exploration of disubstituted [1,2,5]oxadiazolo-[3,4-b]pyrazines as novel C-C chemokine receptor type 5 signaling inhibitors targeting the intracellular allosteric binding pocket. Eur J Med Chem 2025; 291:117600. [PMID: 40222165 DOI: 10.1016/j.ejmech.2025.117600] [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] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
The C-C chemokine receptor type 5 is a G protein-coupled receptor expressed on various immune cells, playing a crucial role in inflammation and chemotaxis. Beyond its physiological functions, C-C chemokine receptor type 5 is implicated in numerous diseases, including cardiovascular, central nervous system, immune system, and infectious diseases, as well as in the progression of cancer. The therapeutic potential of C-C chemokine receptor type 5 inhibition has been demonstrated by antagonists targeting the extracellular domain, notably maraviroc, a Food and Drug Administration-approved Human Immunodeficiency Virus entry inhibitor. However, challenges such as suboptimal pharmacokinetics and efficacy necessitate new antagonists with unique modes of action. Recent advancements in G protein-coupled receptor structural characterization have identified a novel intracellular allosteric binding site in chemokine receptors. This study introduces a series of disubstituted [1,2,5]oxadiazolo-[3,4-b]pyrazines targeting the intracellular allosteric binding pocket of C-C chemokine receptor type 5. Among these, compound 3ad emerged as a promising C-C chemokine receptor type 5-selective allosteric antagonist with a half-maximal inhibitory concentration of 1.09 μM and an almost 30-fold selectivity over C-C chemokine receptor type 2. Molecular dynamics simulations and a competition assay with a Gαq11 mimetic were used to confirm the intracellular binding mode of these compounds. This novel class of C-C chemokine receptor type 5-selective intracellular antagonists offers a foundation for developing molecular tools and therapeutic agents, potentially overcoming the limitations of current extracellular C-C chemokine receptor type 5 antagonists.
Collapse
Affiliation(s)
- Margaux Billen
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000, Leuven, Belgium; University of Edinburgh, Mass Spectrometry Core, Centre for Cardiovascular Science, 47 Little France Crescent, EH16 4TJ, Edinburgh, United Kingdom
| | - Sten Reynders
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000, Leuven, Belgium
| | - Sandra Claes
- KU Leuven, Rega Institute for Medical Research, Molecular, Structural and Translational Virology, Herestraat 49 - Box 1041, 3000, Leuven, Belgium
| | | | - Jef Rozenski
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000, Leuven, Belgium
| | - Radu-George Bulai
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000, Leuven, Belgium
| | - Edoardo Rocca
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000, Leuven, Belgium
| | - Natalie Z M Homer
- University of Edinburgh, Mass Spectrometry Core, Centre for Cardiovascular Science, 47 Little France Crescent, EH16 4TJ, Edinburgh, United Kingdom
| | - Scott P Webster
- University of Edinburgh, Mass Spectrometry Core, Centre for Cardiovascular Science, 47 Little France Crescent, EH16 4TJ, Edinburgh, United Kingdom
| | - Tim P Kaminski
- InSingulo AB, Pepparedsleden 1, Mölndal, SE-43183, Sweden
| | - Eveline Lescrinier
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000, Leuven, Belgium
| | - Dominique Schols
- KU Leuven, Rega Institute for Medical Research, Molecular, Structural and Translational Virology, Herestraat 49 - Box 1041, 3000, Leuven, Belgium
| | - Peter Verwilst
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000, Leuven, Belgium.
| |
Collapse
|
21
|
Yang Y, Jian Y, He L. High performance persistent organic pollutants removal using stabilized enzyme aggregates over amino functionalized magnetic biochar. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:137868. [PMID: 40073570 DOI: 10.1016/j.jhazmat.2025.137868] [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: 01/22/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Herein, a highly efficient and recyclable biocatalyst was developed using stabilized enzyme aggregates on amino-functionalized magnetic biochar for removing persistent organic pollutants from water. The biochar derived from biomass featured abundant hydroxyl functional groups, after functionalization with amino functional groups and magnetic nanoparticles, it was employed for laccase immobilization via enzyme electrostatic adsorption, precipitation and cross-linking in a favorable orientation. This immobilized enzyme aggregates exhibited enhanced pH tolerance, thermal and storage stability than free enzyme. Complete removal of 20 mg/L bisphenol A was achieved within 60 min via C-C bond cleavage and hydroxylation. Notably, the removal efficiency remained at approximately 90 % even after six cycles. Furthermore, this biocatalyst was also successfully applied to efficiently remove other various persistent organic pollutants and demonstrated applicability in real environmental water samples. This study highlights the substantial potential of enzyme-based biocatalysts, presenting a sustainable and efficient approach for water purification and biomass resource recovery.
Collapse
Affiliation(s)
- Yadong Yang
- School of Environmental Science and Engineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Yangyang Jian
- Key laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lingzhi He
- Key laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
22
|
Utama K, Khamto N, Janthong A, Thiraphatchotiphum C, Roytrakul S, Kantapan J, Meepowpan P, Sangthong P. Discovery of cinnamoyl-flavonoid hybrid derivatives as inhibitors of SARS-CoV-2 M pro and anti-inflammatory agents: Experimental and in silico insights into their efficacy against lipopolysaccharide-induced lung injury. Eur J Pharmacol 2025; 998:177636. [PMID: 40252899 DOI: 10.1016/j.ejphar.2025.177636] [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: 11/06/2024] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
The chemical structures of the parental compounds of flavonoids from Boesenbergia rotunda were modified by conjugation with cinnamic acid to form cinnamoyl-flavonoid hybrid derivatives with enhanced anti-inflammatory and SARS-CoV-2 Mpro-inhibitory properties. Cinnamoyl-flavonoid hybrid derivatives 6 and 10 showed the potential to inhibit SARS-CoV-2 Mpro with IC50 values of 52.49 and 22.62 μM. Compounds 6 and 10 showed lower cytotoxicity in the human lung cell lines MRC-5 and A549 at concentrations greater than 50 μM. The effects of compounds 6 and 10 on cell viability were studied in a 3D co-culture model of A549 and MRC-5 treated with lipopolysaccharide (LPS) and observed through confocal microscopy. Compounds 6 and 10 downregulated p65 mRNA expression, resulting in a reduction of pro-inflammatory cytokines, including Interleukin 8 (IL-8) and Monocyte Chemoattractant Protein-1 (MCP-1/CCL2), leading to an anti-inflammatory response through Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signalling pathways. Compound 6 showed potential anti-inflammatory activity, downregulating Bcl-2 Associated X gene (BAX), which resulted in inhibition of apoptotic cell death when compared to compound 10. In silico molecular dynamic simulation shed light on how these cinnamoyl-flavonoid hybrid derivatives interact with myeloid differentiation factor 2 (MD-2), which is involved in the inflammatory response. Our findings suggest that cinnamoyl-flavonoid hybrid derivatives show potential as anti-inflammatory drugs and anti-SARS-CoV-2 drugs.
Collapse
Affiliation(s)
- Kraikrit Utama
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand; Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nopawit Khamto
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Atchara Janthong
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai, 50200, Thailand
| | | | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok, 12120, Thailand
| | - Jiraporn Kantapan
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Puttinan Meepowpan
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Materials Science and Technology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai, 50200, Thailand
| | - Padchanee Sangthong
- Center of Excellence in Materials Science and Technology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
23
|
Ciupak O, Demkowicz S, Rachon J, Biernacki K, Czubak P, Martyna A, Masłyk M, Kubiński K, Datta M, Rak J, Daśko M. Novel nonsteroidal steroid sulfatase inhibitors containing glutamic acid unit. Eur J Med Chem 2025; 291:117627. [PMID: 40245821 DOI: 10.1016/j.ejmech.2025.117627] [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: 02/12/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025]
Abstract
In the present work, we designed and successfully synthesized novel steroid sulfatase (STS) inhibitors based on coumarin, tyramine, triazole, and flavone cores with an additional glutamic acid residue in the structure. The molecular modeling studies revealed that designed derivatives have potential to bind to the molecular target active site, at least theoretically. The biological activity of synthesized compounds was evaluated under a two-step procedure including enzymatic assay and cellular studies using human choriocarcinoma JEG-3 cells. Among the synthesized compounds, the derivative 54E was the most active in both enzymatic and cellular experiments. This result agreed with the molecular modeling data, which indicated that derivative 54E demonstrates the highest affinity to the STS active site. In the enzymatic assay, the remaining STS activity values of 12.97, 17.58, and 20.52 % were observed at 10, 1, and 0.1 μM concentrations of compound 54E, respectively. The IC50 value of 22 nM determined in an experiment with JEG-3 cells for compound 54E was close to the IC50 value determined for the reference STS inhibitor Irosustat (2.7 nM). During the evaluation of the uptake mechanism of the compound 54E, we found that organic anion transporting polypeptides (OATPs) may be responsible for its internalization into the cells. Furthermore, the incubation of zebrafish larvae with the compound 54E revealed no detectable toxic effects in vivo indicating that the compound 54E is a very promising candidate for further preclinical investigations.
Collapse
Affiliation(s)
- Olga Ciupak
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233, Gdansk, Poland.
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233, Gdansk, Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233, Gdansk, Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233, Gdansk, Poland
| | - Paweł Czubak
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708, Lublin, Poland
| | - Aleksandra Martyna
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708, Lublin, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708, Lublin, Poland
| | - Konrad Kubiński
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Konstantynów 1i, 20-708, Lublin, Poland
| | - Magdalena Datta
- Laboratory of Biological Sensitizers, Department of Physical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Janusz Rak
- Laboratory of Biological Sensitizers, Department of Physical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233, Gdansk, Poland.
| |
Collapse
|
24
|
Chen Y, Zhang X, Zheng Z, Cao W, Qin X, Lin H, Chen Z, Zheng H, Zhu G, Gao J. In silico prospecting of ADH activating peptides from Pacific oyster (Crassostrea gigas) and protective effect on ethanol-induced damage in HepG2 cells. Food Chem 2025; 479:143777. [PMID: 40081072 DOI: 10.1016/j.foodchem.2025.143777] [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: 10/21/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Alcoholic liver disease (ALD) is becoming a major health threat in the world today. Alcohol dehydrogenase (ADH) plays an important role in alcohol metabolism. Pacific oyster (Crassostrea gigas) has been identified as a food-borne hepatoprotective agent. For the first time, we integrated in silico strategy, including simulated hydrolysis, bioinformatic prediction and molecular docking to screen ADH activating peptides from C. gigas. In vitro ADH activation activity and surface plasmon resonance (SPR) results showed that this strategy could stably screen ADH activating peptides. We selected six of them to further verify their protective effect on EtOH-induced HepG2 cells. Among them, peptide LQPPR (Leu-Gln-Pro-Pro-Arg) pretreatment increased cell viability, can effectively resist EtOH-induced cytotoxicity. And the transaminase (ALT, AST) in the cell supernatant decreased, indicating the cell damage was improved. The results also showed that the antioxidant capacity (SOD, GSH) of LQPPR pretreated cells increased, and the oxidative stress (MDA) decreased.
Collapse
Affiliation(s)
- Yajing Chen
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiuli Zhang
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhihong Zheng
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China.
| | - Wenhong Cao
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Xiaoming Qin
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Haisheng Lin
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Zhongqin Chen
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Huina Zheng
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Guoping Zhu
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Jialong Gao
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; National Research and Development Branch Center for Shellfish Processing (Zhanjiang), Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China.
| |
Collapse
|
25
|
Zhang Y, Fan YC, Zhang YC, Li Q, Su YY, Xu CS, Yu HL, Wang C, Zhang J, Liao ZX. Antitumor activity and mechanistic study of steroidal saponins from the rhizomes of Paris polyphylla var. yunnanensis. PHYTOCHEMISTRY 2025; 235:114455. [PMID: 40021107 DOI: 10.1016/j.phytochem.2025.114455] [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: 09/10/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Through phytochemical analysis of the large roots of Paris polyphylla var. yunnanensis, three previously undescribed steroidal saponins (Polypharsis A-C, 1-3) and four known steroidal saponins (4-7) were isolated. The structures of 1-3 were elucidated by 1D/2D NMR, HR-ESI-MS, acid hydrolysis and ECD calculations. The cytotoxic activity tests of seven compounds against three tumor cell lines (HepG2, MCF-7, and A549) were conducted. Among them, compounds 6 and 7 belong to the protodioscin class of compounds. Compound 6 exhibits significant inhibition of HepG2 cell growth with an IC50 value of 7.53 ± 2.07 μM, comparable to that of Doxorubicin (6.82 ± 1.59 μM). Compound 7 shows a strong inhibitory effect on the growth of the MCF-7 cell line with an IC50 value of 0.69 ± 0.26 μM, which is more potent than Doxorubicin (5.56 ± 0.08 μM). Subsequently, the antitumor efficacy of compound 6 against HepG2 cells and compound 7 against MCF-7 cells was investigated through in vitro experiments, and their respective mechanisms of action were further predicted using network pharmacology and molecular docking.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Yu-Chen Fan
- Nanjing Institute of Measurement and Testing Technology, Ma Qun Avenue No.10, Nanjing, 210049, China
| | - Yu-Chen Zhang
- Nanjing Institute of Measurement and Testing Technology, Ma Qun Avenue No.10, Nanjing, 210049, China
| | - Qing Li
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Yun-Yun Su
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Chen-Sen Xu
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Hao-Lin Yu
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Chao Wang
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Jing Zhang
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Zhi-Xin Liao
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
26
|
Li X, Zhou Y, Yue J, Sun M, Lei X, Li P, Li J, Sun D, Zeng Z. Enzyme mimics based on self-assembled peptide functionalized with graphene oxide for polyethylene terephthalate degradation. Colloids Surf B Biointerfaces 2025; 251:114588. [PMID: 40010083 DOI: 10.1016/j.colsurfb.2025.114588] [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: 11/06/2024] [Revised: 12/16/2024] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
The degradation of polyethylene terephthalate (PET) has garnered notable attention owing to its widespread accumulation and the challenges associated with its breakdown. Herein, the enzyme mimics with PET-hydrolytic activity were developed by combining peptide nanofibers with graphene oxide (GO). Inspired by native enzymes, we designed self-assembled peptides that included active amino acids (serine, histidine, aspartate and tryptophan) and different hydrophobic amino acids, with a 9-fluorenylmethoxycarbonyl group at the N-terminus. Our comparison of hydrophobic amino acids revealed that their content not only influenced the higher-order assembly of peptide but also affected molecular conformation and PET degradation ability. By co-assembling two peptides with catalytic and binding sites in a 1:1 ratio, a more effective active enzyme mimic was constructed which was owning to the cooperative interactions among the active amino acids; in addition, hydrogen bonds and π-π stacking interactions were the main forces in enhancing catalytic effects. To further improve PET-hydrolytic ability, the co-assembled enzyme mimic was functionalised with GO through π-π stacking. This GO-peptide nanofiber hybrid exhibited increased PET-hydrolytic, as GO provided a hydrophobic microenvironment for substrate attraction and abundant carbon for facilitating proton transfer. The GO-peptide nanofiber hybrid as enzyme mimics will be a promising material for PET degradation.
Collapse
Affiliation(s)
- Xia Li
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China; School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China
| | - Yaoling Zhou
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China
| | - Jingchao Yue
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China
| | - Mengyu Sun
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China
| | - Xiangmin Lei
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Healthy of Tianjin, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Piwu Li
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China; School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China
| | - Jianpeng Li
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | - Dengyue Sun
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China; School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China.
| | - Zhixiong Zeng
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha 410219, China.
| |
Collapse
|
27
|
Ilovaisky AI, Scherbakov AM, Miciurov D, Chernoburova EI, Merkulova VM, Bogdanov FB, Salnikova DI, Sorokin DV, Krasil'nikov MA, Bozhenko EI, Zavarzin IV, Terent'ev AO. Secosteroid - 1,3,4-oxadiazole hybrids: Synthesis and evaluation of their activity against hormone-dependent breast cancer cells. J Steroid Biochem Mol Biol 2025; 251:106745. [PMID: 40164235 DOI: 10.1016/j.jsbmb.2025.106745] [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: 01/20/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
This study focused on the synthesis of secosteroids with good antiproliferative properties against hormone-dependent breast cancer. A straightforward and efficient method for synthesizing secosteroid - 1,3,4-oxadiazole hybrids was developed starting from 13α-hydroxy-3-methoxy-13,17-secoestra-1,3,5(10)-trien-17-oic acid hydrazide. The cyclization of hydrazide moiety with CS2 into 1,3,4-oxadiazole-2(3H)-thione fragment followed by sulfur alkylation resulted in the formation of various secosteroid - 2-mercapto-1,3,4-oxadiazole hybrids. These novel compounds were evaluated for their antiproliferative activity against the hormone-dependent human breast cancer cell line MCF-7. Among the synthesized hybrids, compounds 3i, 3o, and 3q displayed notable antiproliferative effects, with IC50 values ranging from 6.5 to 8.9 µM, comparable to the reference drug cisplatin. Furthermore, compound 3i showed minimal toxicity toward non-cancerous hFB-hTERT fibroblasts, indicating high selectivity. Compounds 3o and 3q exhibited antiestrogenic activity. Additionally, their effects on PARP and Bcl-2 suggest a pro-apoptotic mechanism of action. These findings highlight the potential of secosteroidal hybrids as promising candidates for the development of new anti-breast cancer agents targeting ERα and apoptosis pathways.
Collapse
Affiliation(s)
- Alexey I Ilovaisky
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander M Scherbakov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia; Gause Institute of New Antibiotics, Bol'shaya Pirogovskaya ulitsa 11, Moscow 119021, Russia
| | - Dumitru Miciurov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Elena I Chernoburova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Valentina M Merkulova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Fedor B Bogdanov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Diana I Salnikova
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Danila V Sorokin
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Mikhail A Krasil'nikov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Eugene I Bozhenko
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Igor V Zavarzin
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander O Terent'ev
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| |
Collapse
|
28
|
Bai W, Wang Y, Ma J, Li G, Wang Y, Yang C, Zhang Q, Li Q, Zhang J, Zhang P. Histone deacetylase Hos1 promotes the homeostasis of Candida albicans cell wall and membrane and its specific inhibitor has an antifungal activity in vivo. Microbiol Res 2025; 296:128132. [PMID: 40112660 DOI: 10.1016/j.micres.2025.128132] [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: 09/28/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025]
Abstract
The rise of drug-resistant Candida albicans (C. albicans) has led to an urgent need for new therapeutic strategies. Histone deacetylases (HDACs) inhibition has been shown to limit fungal virulence while enhancing the efficacy of antifungal drugs against Candida. However, HDACs are highly conserved from yeast to humans, which has hindered the application of these inhibitors in the antifungal therapy. The aim of this study is to identify a suitable antifungal target and develop specific inhibitors targeting C. albicans HDACs. Based on sequence alignments, the HDAC Hos1 in C. albicans was proposed as a target for further investigation. We evaluated the impact of Hos1 on C. albicans pathogenicity using a murine model of disseminated candidiasis. Results showed that Hos1 null mutant caused less damage to mouse tissues. Additionally, we demonstrated that the reduced virulence was due to inhibition of cell wall O-mannan biosynthesis and altered metabolic flexibility, leading to decreased adaptability of C. albicans. Increased sensitivity of C. albicans to antifungal drugs was attributed to abnormal accumulation of ergosterol in the cell membrane. Furthermore, we identified Hos1 inhibitors from the ZINC database using molecular docking. These inhibitors exhibited highly specific inhibition of the deacetylation activity of C. albicans Hos1. Importantly, the inhibitors not only reduced colonization and invasion by C. albicans in vivo but also synergized with polyene drugs to combat C. albicans by causing abnormal accumulation of ergosterol. Our findings provide detailed insights into antifungal targets and a useful foundation for the discovery of antifungal drugs specifically targeting Candida.
Collapse
Affiliation(s)
- Wenhui Bai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanmei Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jia Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi' an, Shaanxi 710061, China
| | - Guanglin Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuchen Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Zonglian College, Xi'an Jiaotong University, Xi' an, Shaanxi 710061, China
| | - Chen Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qiyue Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qingqing Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Peipei Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
29
|
Guo B, Shi S, Xiong J, Guo Y, Wang B, Bai L, Qiu Y, Li S, Gao D, Dong Z, Tu Y. Identification of potential biomarkers in cardiovascular calcification based on bioinformatics combined with single-cell RNA-seq and multiple machine learning analysis. Cell Signal 2025; 131:111705. [PMID: 40024421 DOI: 10.1016/j.cellsig.2025.111705] [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: 01/13/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The molecular and genetic mechanisms underlying vascular calcification remain unclear. This study aimed to determine the differences in calcification marker-related gene expression in macrophages. METHODS The expression profiling datasets GSE104140 and GSE235995 were analysed to identify differentially expressed genes (DEGs) between fibroatheroma with calcification and diffuse intimal thickening. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, Weighted Gene Co-expression Network Analysis (WGCNA), and Gene Set Enrichment Analysis (GSEA) were performed to assess functional characteristics. Hub genes were identified through a protein-protein interaction (PPI) network and machine learning approaches. Single-cell RNA sequencing data (GSE159677) validated the expression of calcification-related genes in macrophages, while Mendelian randomization analysis explored their potential causal relationship with coronary calcification. Further validation was conducted using enzyme-linked immunosorbent assay (ELISA) on coronary calcification samples and immunohistochemistry in ApoE-/- mice. Intravascular ultrasound was performed to assess coronary calcification severity. RESULTS AND CONCLUSIONS Two key biomarkers, ITGAX and MYD88, were identified as diagnostic indicators of cardiovascular calcification. Both biomarkers were significantly upregulated in calcified samples and were strongly associated with immune processes. Single-cell RNA sequencing confirmed their high expression in multiple immune cell types. Additionally, molecular docking analysis revealed that retinoic acid interacted with both biomarkers, suggesting potential therapeutic relevance. Immunohistochemical and ELISA analyses further validated their elevated expression in calcified samples. These findings provide novel insights into the molecular mechanisms of vascular calcification and highlight potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Bingchen Guo
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China.
| | - Si Shi
- Harbin Medical University, Harbin, China; Department of Respirology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Jie Xiong
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yutong Guo
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Bo Wang
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Liyan Bai
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yi Qiu
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Shucheng Li
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Dianyu Gao
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Zengxiang Dong
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yingfeng Tu
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China; Department of Cardiology, The Shanxi Provincial People's Hospital, Taiyuan 030000, China.
| |
Collapse
|
30
|
D'Almeida CTDS, Sales ACDA, Xavier AAO, Mameri H, Ferreira MSL, Tavares GM. β-Lactoglobulin and sorghum phenolic compounds molecular binding: Interaction mechanism and thermal stability impact. Food Chem 2025; 478:143632. [PMID: 40058253 DOI: 10.1016/j.foodchem.2025.143632] [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: 09/19/2024] [Revised: 02/07/2025] [Accepted: 02/25/2025] [Indexed: 04/06/2025]
Abstract
The mechanism of molecular interaction between β-lactoglobulin (β-lg) and sorghum bran phenolic compounds from 4 genotypes was studied. Catechin (CA) and ferulic acid (FA) were used as model systems. Higher affinity for β-lg:FA interaction (Ksv ≈ 105 M-1) compared with β-lg:CA interaction (Ksv ≈ 104 M-1) was revealed, with different preferable binding sites identified through molecular docking. Nevertheless, regarding the molecular interaction between the proteins and the complex extracts of phenolic compounds, Ksv in the magnitude order of 104 M-1 were observed. Antioxidant capacity progressively increased after protein-phenolic interaction, indicating a potential synergistic effect. Concerning the thermal stability of the phenolic compounds, epimerization as the primary response of CA to thermal treatment (90 °C / 10 min) was identified, but the addition of β-lg exerted a protective effect against CA degradation (-7 % in β-lg:CA complexes); however, proteins were not able to protect complex phenolic matrices (e.g. sorghum extracts).
Collapse
Affiliation(s)
- Carolina Thomaz Dos Santos D'Almeida
- Laboratory of Bioactives, Food and Nutrition Graduate Program (PPGAN), Federal University of the State of Rio de Janeiro, UNIRIO, Brazil; Center of Innovation in Mass Spectrometry, Laboratory of Protein Biochemistry, UNIRIO, Rio de Janeiro 22290-240, Brazil.
| | | | - Ana Augusta O Xavier
- Department of Food Science and Nutrition, School of Food Engineering, University of Campinas, UNICAMP, Brazil.
| | - Hamza Mameri
- UMR 1208 IATE, Univ. Montpellier, INRAE, L'Institut-Agro Montpellier, F-34060 Montpellier, France.
| | - Mariana Simões Larraz Ferreira
- Laboratory of Bioactives, Food and Nutrition Graduate Program (PPGAN), Federal University of the State of Rio de Janeiro, UNIRIO, Brazil; Center of Innovation in Mass Spectrometry, Laboratory of Protein Biochemistry, UNIRIO, Rio de Janeiro 22290-240, Brazil.
| | - Guilherme M Tavares
- Department of Food Science and Nutrition, School of Food Engineering, University of Campinas, UNICAMP, Brazil.
| |
Collapse
|
31
|
Sui Y, Peng C, Zhou P, Qiu L, Qu C, Li W, Wu C, Liu J. Insect odorant-binding protein modified biosensor for sensitive and specific electrochemical detection of alcohols. Biosens Bioelectron 2025; 278:117382. [PMID: 40101655 DOI: 10.1016/j.bios.2025.117382] [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: 11/27/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Olfaction biosensors are playing crucial roles in detecting volatile organic compounds (VOCs) in various domains, while the response pattern of biosensors to different alcohols and the underlying reasons for the differences in response remain unclear. Herein, this study presents a sensitive electrochemical olfactory biosensor utilizing Drosophila odorant-binding protein (LUSH) as a sensing material for the detection of 11 alcohols with different molecular structures (alkyl chain lengths, hydroxyl group numbers, and cyclic alcohols) and phenol. The electrodes covalently immobilized with the LUSH proteins were characterized by atomic force microscopy (AFM), electrochemical impedance spectroscopy (EIS), and cyclic voltammetry (CV), and their ability to detect alcohols was investigated through EIS. Results showed that the LUSH modified biosensor exhibited ultrasensitive detection of multiple alcohols (detection limits: 10-100 fM), with linear ranges from 10-14 to 10-7 M and coefficients of determination (R2) of 0.948-0.992. In addition, the biosensor demonstrated high selectivity toward interfering compounds (selectivity coefficients <0.22), excellent reproducibility (relative standard deviation, RSD: 1.2%, n = 4 for parallel sensors), and good stability (response decreased by 10.2% on the 10th day). Notably, the sensitivity of the biosensor to alcohols showed alkyl chain-length dependence of n-alcohols and was influenced by the number of hydroxyl groups and the cyclic structure. More importantly, molecular docking revealed the binding modes, binding energies, and key amino acids involved in the LUSH-alcohol interaction and explained the response discrepancies.
Collapse
Affiliation(s)
- Yutong Sui
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Cong Peng
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Peng Zhou
- Beijing Environmental Engineering Technology CO., LTD, 101111, China
| | - Lina Qiu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Chen Qu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Wenhui Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Chuandong Wu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Jiemin Liu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China; Beijing Institute of Graphic Communication, Beijing, 102600, China.
| |
Collapse
|
32
|
Terenteva O, Mostovaya O, Bukharov M, Mukhametzyanov T, Bikmukhametov A, Lyubina A, Voloshina A, Petrov K, Padnya P, Stoikov I. Peptidomimetics based on thiacalixarene with L-tyrosine moieties: Antibacterial activity against methicillin-resistant Staphylococcus aureus and degradation induced by binding to α-chymotrypsin. Bioorg Chem 2025; 160:108434. [PMID: 40187027 DOI: 10.1016/j.bioorg.2025.108434] [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: 02/11/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The design of new antimicrobial agents is an important challenge due to the growing resistance of microorganisms to existing antibiotics. In recent years, the trend towards the development of compounds and materials with (bio)degradable properties has emerged. In this work, we propose and develop a method for the synthesis of new peptidomimetics, i.e., water-soluble macrocyclic quaternary ammonium salts containing L-tyrosine fragments based on p-tert-butylthiacalix[4]arene in various stereoisomeric forms (cone, partial cone, and 1,3-alternate). These compounds have low cytotoxicity (IC50 = 80-267 μM) and high antibacterial activity (MIC = 0.5-15.6 μM) against Gram-positive bacterial strains including methicillin-resistant Staphylococcus aureus (MRSA). The obtained peptidomimetics can bind α-chymotrypsin with the formation of supramolecular systems and their subsequent degradation. Our results demonstrate the first example of multi-action thiacalixarene derivatives with antibacterial activity, protein binding ability and degradation induced by binding to α-chymotrypsin. The obtained results open the possibility of creating multi-action peptidomimetic systems with antimicrobial and biodegradable effect.
Collapse
Affiliation(s)
- Olga Terenteva
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Olga Mostovaya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Mikhail Bukharov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Timur Mukhametzyanov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Azamat Bikmukhametov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Anna Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Alexandra Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Konstantin Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Pavel Padnya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| | - Ivan Stoikov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| |
Collapse
|
33
|
Gu J, You R, Zhao N, Xu X, Zhou J, Zhang X, Zhang Q, Jiang J, Jiang Y, Wen S. 2β-methoxy-2-deethoxyphantomolin synergistically enhances epirubicin effect against triple-negative breast cancer via targeted inhibition of AKT and HR pathways. Bioorg Chem 2025; 160:108453. [PMID: 40253761 DOI: 10.1016/j.bioorg.2025.108453] [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: 12/24/2024] [Revised: 03/01/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
Enhancing the efficacy of chemotherapy in treating triple-negative breast cancer (TNBC) remains crucial. Understanding the genes involved in cancer progression and targeted therapies may be beneficial for TNBC chemotherapy. Here, bioinformatics analysis revealed that AKT1 and the key gene of homologous recombination (HR), RAD51, were significantly upregulated in breast cancer. Meanwhile, we discovered that epirubicin (Epi) could activate AKT and HR pathways in MDA-MB-231 cells. Pharmacological inhibition of AKT or siAKT could reverse the activation of AKT and HR pathways and sensitize Epi. Hence, we sought natural products that could inhibit both AKT and HR pathways to enhance the sensitivity of Epi and expanded its therapeutic effects in TNBC. 2β-methoxy-2-deethoxyphantomolin (EM2) is a natural sesquiterpene lactone extracted from Elephantopus mollis with strong anticancer activity. We found that EM2 induced apoptosis of MDA-MB-231 cells by inhibiting AKT and HR pathways. Mechanistically, EM2 impaired transcription of AKT, directly bound to RAD51 protein, and facilitated the degradation of AKT and RAD51 in a caspase-dependent manner. Importantly, we determined that EM2 in combination with Epi treatment exhibited a synergistic anti-tumor effect in MDA-MB-231 cells. Moreover, overexpression of AKT could prevent EM2 from sensitizing Epi. The results of MDA-MB-231 xenograft tumor model confirmed that EM2 could reduce the dose of Epi and achieve anti-TNBC effect in vivo without toxicity to mice tissues. Overall, our work indicates EM2 in combination with Epi can greatly expand the therapeutic effect of Epi in TNBC, underscoring targeting AKT and RAD51 as a promising approach for TNBC chemotherapy sensitization.
Collapse
Affiliation(s)
- Jianyi Gu
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou 510632, China
| | - Ronger You
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou 510632, China
| | - Na Zhao
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou 510632, China
| | - Xinwen Xu
- Department of Breast Surgery, The Second People' s Hospital of Foshan, Foshan, Guangdong 528000, China; Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China
| | - Junzhen Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China
| | - Xiaoying Zhang
- Department of Pathology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, Guangdong 511400, China
| | - Qing Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China
| | - Jianwei Jiang
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou 510632, China.
| | - Yue Jiang
- Department of Thyroid and Breast Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong 528303, China.
| | - Shunqian Wen
- Department of Hepatobiliary Surgery, The Second People' s Hospital of Foshan, Foshan, Guangdong 528000, China.
| |
Collapse
|
34
|
Ashraf R, Khalid Z, Qin QP, Iqbal MA, Taskin-Tok T, Bayil İ, Quah CK, Daud NAM, Alqahtany FZ, Amin MA, El-Bahy SM. Synthesis of N-heterocyclic carbene‑selenium complexes modulating apoptosis and autophagy in cancer cells: Probing the interactions with biomolecules and enzymes. Bioorg Chem 2025; 160:108435. [PMID: 40199010 DOI: 10.1016/j.bioorg.2025.108435] [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: 02/14/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Growing cancer resistance is a global threat that calls for development of newer chemotherapeutic analogues especially targeted based therapy to enhance efficacy and selectivity. In this contribution, herein, we report synthesis of selenium incorporated N-heterocyclic carbene (NHC) compounds to explore their potential cytotoxicity against HeLa cells. Test compounds were assured for suitability as drug candidates through physiochemical properties that showed lipophilicity logP 0.85-1.45 for C1-C3 and found stable in biological media (DMEM), whereas, least reactive with N-acetyl cysteine (NAC) and L-glutathione. All the studied compounds showed good cytotoxicity against various cancer strains while compound C1 [3,3-(hexane-1,6-diyl)bis(1-phenethyl-1H-imidazole-2(3H)-selenone)] and C2 [3,3-(hexane-1,6-diyl)bis(1-decyl-1H-imidazole-2(3H)-selenone)] showed promising results with IC50 values of 14.65 ± 0.66 and 8.05 ± 0.35 μg/mL respectively as compared to positive control 21.5 ± 0.05 μg/mL against HeLa cell lines. These compounds showed six-fold higher apoptosis than control with higher accumulation of Ca+ ions intracellularly that alters the expression level of autophagy proteins and increased capase-9 activity. Cell cycle analysis indicated an arrest of cycle in G1 phase of HeLa cells when treated with C1 & C2. Test compounds showed prominent affinity for binding with DNA and inhibiting thioredoxin reductase enzymes in time dependent manners. These findings indicate that Selenium NHC compounds are promising drug candidates to induce cytotoxicity via apoptosis, autophagy and mitochondrial membrane disruptions to manage tumor growth.
Collapse
Affiliation(s)
- Rizwan Ashraf
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan.
| | - Zohra Khalid
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, School of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China
| | - Muhammad Adnan Iqbal
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan; Organometallic and Coordination Chemistry Laboratory, Department of Chemistry, University of Agriculture Faisalabad, 38000, Pakistan.
| | - Tugba Taskin-Tok
- Faculty of Arts and Sciences, Department of Chemistry, Gaziantep University, +9027310, Gaziantep, Turkiye; Institute of Health Sciences, Department of Bioinformatics and Computational Biology, Gaziantep University, +9027310, Gaziantep, Turkiye
| | - İmren Bayil
- Institute of Health Sciences, Department of Bioinformatics and Computational Biology, Gaziantep University, +9027310, Gaziantep, Turkiye
| | - Ching Kheng Quah
- X-ray Crystallography Unit, School of Physics, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Nur Aisyah Mohamad Daud
- X-ray Crystallography Unit, School of Physics, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Mohammed A Amin
- Department of chemistry, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Salah M El-Bahy
- Department of chemistry, Turabah University college, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
35
|
Sasan SP, Martoliya Y, Vashishtha S, Kaur H, Kundu D, Gourinath S, Lynn A, Prasad R, Kundu B, Mondal AK. Structural Insight Into the Conversion of DhNik1, A Hybrid Histidine Kinase From Debaryomyces hansenii to a Cytotoxic Phosphatase Conformation for Novel Antifungal Agent. J Mol Biol 2025; 437:169116. [PMID: 40174667 DOI: 10.1016/j.jmb.2025.169116] [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: 01/07/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Group III hybrid histidine kinase (HHK3) is one of the most interesting signalling molecules and a novel drug target in fungi. HHK3 is converted into a cytotoxic phosphatase form in vivo by the action of a widely used agricultural fungicide indicating that HHK3 also exhibits dual functionality by acting as kinases and phosphatases towards their substrates like many bacterial sensor histidine kinases. However, this cytotoxic form of HHK3 remained elusive for further scientific exploitation. In this study, we have isolated a cytotoxic phosphatase LOCK-IN mutant of DhNik1, a prototype HHK3, and provided structural and functional insight into this form for the first time. The mutant DhNik1CT had an in-frame deletion in the poly-HAMP domain. The fusion of the poly-HAMP domain of DhNik1CT with the histidine kinase and receiver domain of another fungal hybrid histidine kinase also created a hybrid that was cytotoxic to the fungal cell. We generated the structural model of wild-type DhNik1 and DhNik1CT using AlphaFold multimer which highlighted the differences in HAMP domain arrangement and conformation between DhNik1 and DhNik1CT. MD simulation of the modelled structure revealed crucial role of ATP lid opening and closing in regulating the activity of DhNik1 and DhNik1CT. The structure of DhNik1CT was used for virtual screening to identify a small molecule which modulates the activity of DhNik1 towards cytotoxicity. Taken together, present study shows that the conversion of HHK3 to a toxic conformation by a small molecule is a feasible approach for discovering novel antifungal drug.
Collapse
Affiliation(s)
- Soorya Partap Sasan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Yogita Martoliya
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Shubham Vashishtha
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi Hauz Khas, New Delhi 110016, India
| | - Harbinder Kaur
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Debasree Kundu
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Samudrala Gourinath
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Andrew Lynn
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology, Amity University, Haryana 122413, India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi Hauz Khas, New Delhi 110016, India
| | - Alok Kumar Mondal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
36
|
Eldehna WM, Tawfik HO, Nafie MS, Al Kamaly O, El-Hamaky AA, El Hassab MA, Elsayed ZM, Elnaggar YSR, Al-Karmalawy AA, di Giacomo V, Balaha M. Novel benzofuran-conjugated indolin-2-ones as anticancer agents; design, synthesis, biological assessments, and molecular modeling insights. Bioorg Chem 2025; 160:108494. [PMID: 40286528 DOI: 10.1016/j.bioorg.2025.108494] [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: 01/28/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have been authorized for the treatment of breast cancer (BC) and prostate cancer (PC). Recent studies suggest that inhibiting angiogenesis through the vascular endothelial growth factor receptor (VEGFR) enhances cellular sensitivity to PARP inhibitors. This study presents the design, synthesis and full characterization of dual VEGFR-2 and PARP-1 inhibitors obtained by conjugating a PARP-1 inhibitor with VEGFR-2 inhibitor fragments (indole, benzofuran, and piperazine). Four compounds exhibited significant inhibitory activities against human prostate cancer cell lines (PC3) and breast cancer cell lines (MCF7) at 48 h. These compounds were identified as dual VEGFR-2 and PARP-1 inhibitors with low or sub-micromolar ranges, especially 12f, with IC50 values of 0.43 μM and 1.10 μM, respectively. Moreover, the potent compound 12f markedly decreased scratch wound closure and colony formation. Moreover, compound 12f significantly induced apoptosis in PC3 cells and arrested cells at the S phase. The dual inhibition of VEGFR-2 and PARP-1 protein kinase was further validated using western blotting. Applying molecular docking and dynamics determined the target compound's binding mechanism.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt.
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Omkulthom Al Kamaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Anwar A El-Hamaky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mahmoud A El Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Egypt
| | - Zainab M Elsayed
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt; Head of Research & International Publishing Administration (RIPA), Pharos University in Alexandria, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Viviana di Giacomo
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marwa Balaha
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt; Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
37
|
Mukherjee S, Tripathi A. Role of quercetin as a promising antiviral, therapeutic and immunomodulatory mediator against dengue virus induced robust infection in in-vivo Balb/C mice model. Eur J Med Chem 2025; 290:117536. [PMID: 40132497 DOI: 10.1016/j.ejmech.2025.117536] [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: 11/16/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Currently, there are no clinically approved antiviral agents against dengue-virus (DENV). This study aimed to determine the prophylactic, antiviral, and therapeutic potential of quercetin by its pre-treatment, co-treatment, and post-treatment [24, 48, and 72 h-post-infection (HPI)] of DENV-infected Balb/C mice through both oral and intraperitoneal (I.P) route, respectively. 80 mg/kg/day and 16 mg/kg/day of quercetin were non-toxic for oral and I.P administration, respectively. I.P. was found to be more effective than oral administration which significantly reduced DENV copy-number in co-treatment group (from day 1, p < 0.01); post-treatment (24hpi),and pretreatment groups (day 3 onwards, p < 0.05). Molecular-docking experiments indicated quercetin could act as a double-edged sword by strongly interacting with DENV envelope-glycoprotein (-8.1 kcal/mol) and NS5-RdRp domain (-8.0 kcal/mol), which are crucial for viral-attachment and replication. MD-simulation of docked complexes indicated their stability defined by low RMSD, RMSF, and stable H-bond with active-site residues. Significant reduction (p < 0.001) in TNF-α, IL-6, ROS-production, and vascular leakage was observed among pre-, co-, and post-treatment (24 and 48 HPI) groups with promising hepatic and renal-protective effects. Pharmacological and functional-molecular interaction networks indicate a significant effect of quercetin on vascular integrity byVEGF-KDR signaling pathway (via PI3K-Akt and Ras signalling), oxygen homeostasis through HIF-1 signalling, and the anti-inflammatory response via PI3K-Akt, IL-6 and its receptor signalling (PPI enrichment P = 3.19e-10).Thus, it can be concluded that I.P. co- and post-treatment (24hpi) of quercetin to DENV-infected mice could effectively reduce viral-titer, pro-inflammatory cytokines, ROS-response, and vascular permeability. Taken together this demonstrates quercetin as an important antiviral candidate against dengue.
Collapse
Affiliation(s)
- Saikat Mukherjee
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical, Medicine, Kolkata, West Bengal, India
| | - Anusri Tripathi
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical, Medicine, Kolkata, West Bengal, India.
| |
Collapse
|
38
|
Abdel-Maksoud MS, Alatawi RA, Albalawi SSA, Alrashidi MN, Abo-Dya NE, Elsherbiny N, Ragab YM, Awaji AA, El-Sherbiny M, Elfadil H, Abd-Alhaseeb MM. Diacerein's antiproliferative effects alone and with 5-fluorouracil in an Ehrlich solid tumour model: Molecular docking, molecular dynamics Simulation studies, and experimental Verification. Eur J Pharmacol 2025; 996:177564. [PMID: 40157706 DOI: 10.1016/j.ejphar.2025.177564] [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: 12/15/2024] [Revised: 03/16/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The current study used an experimental model of mammary gland carcinoma to assess the chemo-sensitizing effectiveness of the combined administration of diacerein and 5-Fluorouracil (5-FU). With docking scores of -8.1, -7.6, and -9.2 kcal/mol, respectively, the molecular docking experiments showed that diacerein exhibits significant binding affinities to Caspase-3, NF-κB, and AKT1. Molecular dynamics Simulations revealed that diacerein has favourable binding free energy (ΔGbind) of -26.7 kcal/mol for Caspase-3, -24.2 kcal/mol for NF-κB, and -39.9 kcal/mol for AKT1, combined with low root mean square deviation (RMSD) values of 3.1 Å, 1.6 Å, and 2.1 Å for the three targets respectively. To validate these findings in vivo, Ehrlich solid tumor (EST) was induced in female Swiss mice. Four groups of animals were randomly assigned: EST + vehicle, EST + 5-FU, EST + diacerein, and EST + combination. Diacerein and 5-FU combination treatment increased EST mice's life span and reduced the solid tumor's weight and volume. Furthermore, diacerein and 5-FU combination significantly suppressed oxidative stress, inhibited AKT phosphorylation, decreased downstream inflammation (NF-κB, TNF-α, IL-1β), and increased apoptosis by modulating Bax, Bcl2, P53, and caspase-3 levels in tumor tissues. In conclusion, by inhibiting the AKT/NF-κB axis, diacerein and 5-FU combination showed possible antiproliferative effectiveness in the EST model.
Collapse
Affiliation(s)
- Mohamed S Abdel-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | | | | | | | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia.
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia.
| | - Yasser M Ragab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, North Sinai, Egypt; Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Aeshah A Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, 71491, Saudi Arabia.
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia.
| | - Hassabelrasoul Elfadil
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad M Abd-Alhaseeb
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt; Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
39
|
Chukwuka AV, Adegboyegun AD, Adeogun AO. Algal bloom-mediated microplastic dispersion in coastal areas of West Africa: Integrated insights and risk projections from molecular models and remote-sensed evaluations. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137590. [PMID: 39954431 DOI: 10.1016/j.jhazmat.2025.137590] [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: 11/04/2024] [Revised: 01/01/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Algal blooms along the West African coast threaten ecosystems and human health due to nutrient enrichment and rising temperatures. This remote-sensing study examined the relationships between chlorophyll-a concentrations, environmental variables, and the potential for microplastic retention in blooms using molecular docking models for predictive insights. Correlation analyses revealed region-specific associations, with moderate positive correlations between chlorophyll and temperature along the southwest Nigeria-Togo coastline and near Liberia and Sierra Leone (r = 0.2-0.4) and strong correlations with particulate carbon across most regions (r = 0.6-0.8). Chlorophyll fluorescence correlations were generally low (r = 0.2), except for higher correlations in the Senegal-Gabon and Côte d'Ivoire-Ghana stretches, indicating that localized factors influence bloom dynamics. Molecular docking results predict that polycarbonate microplastics have the strongest binding affinities with algal proteins, particularly flagellin (-11.3 kcal/mol), suggesting significant retention potential within bloom matrices. In contrast, ethylene plastics displayed weaker interactions (up to -2.2 kcal/mol) and a high dissociation constant (Kd = 0.079 M), indicating minimal retention potential. The low Kd values for polycarbonateprotein interactions (e.g., 5.15e09 M for flagellin) predict a concerning scenario where microplastics become increasingly integrated into algal biomass, increasing exposure risks for marine life. Warm, nutrient-rich conditions along the West African coast, especially from southwest Nigeria to Togo and Côte d'Ivoire to Sierra Leone, are expected to increase the frequency and severity of algal blooms. This proliferation disrupts biodiversity and water quality while straining local fisheries by altering marine food webs. To mitigate microplastic entrapment from algal blooms and protect vulnerable marine ecosystems, targeted monitoring and intervention strategies are essential.
Collapse
Affiliation(s)
- Azubuike Victor Chukwuka
- Environmental Quality Control Department, National Environmental Standards and Regulations Enforcement Agency (NESREA), Nigeria.
| | - Ayotunde Daniel Adegboyegun
- Environmental Quality Control Department, National Environmental Standards and Regulations Enforcement Agency (NESREA), Nigeria
| | - Aina O Adeogun
- Hydrobiology and Fisheries Unit, Department of Zoology, University of Ibadan, Nigeria.
| |
Collapse
|
40
|
Rossato Viana A, Aytar EC, Pippi N, Santos D, Rhoden CRB, Stefanello Vizzotto B, Flores EMM, Passaglia Schuch A, Krause LMF. Chemical composition, in vitro and in silico activity of the methanolic extract derived from Vassobia breviflora against clinically relevant bacteria. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:463-478. [PMID: 39849319 DOI: 10.1080/15287394.2025.2453858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
This study aimed to identify chemical compounds derived from Vassobia breviflora methanolic extract using ESI-ToF-MS and their antioxidant potential activity utilizing the following methods: total phenols, DPPH, and ABTS•+. The MTT assay measured cytotoxic activity, while DCFH-DA and nitric oxide assays were employed to determine reactive oxygen species (ROS) and reactive nitrogen species (RNS) levels using African green monkey kidney (VERO) and human keratinocyte (HaCat) cell lines. The minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) were assessed in seven clinical isolates and nine ATCC strains. Biofilm inhibition was tested against four biofilm-forming strains. The antioxidant properties of the methanolic extract were identified as follows: 35.74 mg GAE/g (gallic acid equivalents)/g for total phenols, 10.5 µg/ml for DPPH, and 50.68 µmol trolox/µg for ABTS•+. The mean inhibitory concentration (IC50) values were 622.86 µg/ml (VERO) and 784.33 µg/ml (HaCat). These concentrations did not markedly alter levels of ROS and RNS. Conversely, Bacillus cereus β-hemolytic displayed higher sensitivity to the extract, with MIC of 64 µg/ml and MBC of 128 µg/ml. Enterococcus faecium exhibited the lowest biofilm formation among the tested bacteria. The studied plant exhibited activity against all bacterial strains at concentrations lower than the IC50 VERO and HaCat cells, suggesting potential for future studies. Data present a comprehensive molecular docking analysis against the HlyIIR protein (PDB ID: 2FX0) and determined antimicrobial and endocrine-modulating potentials. Notably, lancifodilactone I and nicandrin B demonstrated the strongest binding affinities, with binding energies of -9.8 kcal/mol and -8.3 kcal/mol, respectively, and demonstrated significant antimicrobial effects against B. cereus. In addition, several compounds showed potential interactions with nuclear receptors, indicating potential endocrine-modulating effects. These findings provide insights into developing target-specific antimicrobial therapies and endocrine-modulating agents.
Collapse
Affiliation(s)
- Altevir Rossato Viana
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Brazil
| | - Erdi Can Aytar
- Faculty of Agriculture, Department of Horticulture Uşak, Usak University, Türkiye
| | - Nickolas Pippi
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Brazil
| | - Daniel Santos
- Department of Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | | | | | | | - André Passaglia Schuch
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Brazil
| | | |
Collapse
|
41
|
Zoheir KMA, Ali NI, Ashour AE, Kishta MS, Othman SI, Rudayni HA, Rashad AA, Allam AA. Lipoic acid improves wound healing through its immunomodulatory and anti-inflammatory effects in a diabetic mouse model. J Diabetes Metab Disord 2025; 24:56. [PMID: 39868353 PMCID: PMC11759746 DOI: 10.1007/s40200-025-01559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/28/2024] [Indexed: 01/28/2025]
Abstract
Objectives Diabetes mellitus is a chronic disease that has become more prevalent worldwide because of lifestyle changes. It leads to serious complications, including increased atherosclerosis, protein glycosylation, endothelial dysfunction, and vascular denervation. These complications impair neovascularization and wound healing, resulting in delayed recovery from injuries and an elevated risk of infections. The present study aimed to investigate the effect of lipoic acid (LA) on the key mediators involved in the wound healing process, specifically CD4 + CD25 + T cell subsets, CD4 + CD25 + Foxp3 + regulatory T (Treg) cells, T-helper-17 (Th17) cells that generate IL-17 A, glucocorticoid-induced tumor necrosis factor receptor (GITR) expressing cells, as well as cytokines such as IL-2, IL-1β, IL-6, and TNF-α and IFN-γ. These mediators play crucial roles in epidermal and dermal proliferation, hypertrophy, and cell migration. Methods We divided mice into 5 groups: the non-diabetic (normal control; NC), wounded non-diabetic mice (N + W), wounded diabetic mice (D + W), wounded diabetic mice treated with 50 mg/kg lipoic acid (D + W + L50) for 14 days, and wounded diabetic mice treated with 100 mg/kg lipoic acid (D + W + L100) for 14 days. Results Flow cytometric analysis indicated that lipoic acid-treated mice exhibited a significant decrease in the frequency of intracellular cytokines (IL-17 A, TNF-α and IFN-γ) in CD4 + T cells, as well as a reduction in the number of GITR-expressing cells. Conversely, a significant upregulation in the number CD4+, CD25+, FOXp3 + and CD4 + CD25 + Foxp3 + regulatory T (Treg) cells was observed in this group compared to both the normal + wounded (N + W) and diabetic + wounded (D + W) groups. Additionally, the mRNA Levels of inflammatory mediators (IL-2, IL-1β, IL-6, and TNF-α) were downregulated in lipoic acid-treated mice compared to other groups. T thereby he histological findings of diabetic skin wounds treated with lipoic acid showed well-healed surgical wounds. Conclusions These findings support the beneficial role of lipoic acid in fine-tuning the balance between anti-inflammatory and pro-inflammatory cytokines, influencing both their release and gene expression.
Collapse
Affiliation(s)
- Khairy M. A. Zoheir
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza 12622 Egypt
| | - Neama I. Ali
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza 12622 Egypt
| | - Abdelkader E. Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Salman International University, Ras Sudr, South Sinai Egypt
| | - Mohamed S. Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, and Stem Cell Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622 Egypt
| | - Sarah I. Othman
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, 11671 Riyadh, Saudi Arabia
| | - Hassan A. Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623 Saudi Arabia
| | - Ahmed A. Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829 Egypt
| | - Ahmed A. Allam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623 Saudi Arabia
| |
Collapse
|
42
|
Wang ZZ, Xiao CX, Huang WL, Hu Y, Zhang HT, Liu Z, Peng SH, Wei Z. Discovery of Flavonol derivatives as porcine reproductive and respiratory syndrome virus inhibitors. Bioorg Med Chem Lett 2025; 121:130162. [PMID: 40057134 DOI: 10.1016/j.bmcl.2025.130162] [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: 09/30/2024] [Revised: 01/14/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) causes serious threat to the global pig industry, and there was still no efficient treatment for porcine reproductive and respiratory syndrome (PRRS). Flavonol compounds were reported to show antiviral activity against a series of different virus. In this study, we designed a series of flavonol derivatives as promising lead structure for PRRSV inhibitors. A flavonol derivative database with diverse structures was first generated, and their anti-PRRSV activity were test. Among these compounds, compound 4s showed promising anti-PRRSV activity with EC50 values of 0.45 μM. In addition, it exhibited low cytotoxicity with CC50 higher than 100 μM. We also found that compound 4s inhibited PRRSV might be by repressing the activity of nsp4 protease. Molecular modeling study revealed that compound 4s binding to nsp4 mainly relies on a salt bridge and hydrophobic interaction. Our results might provide a new way for the development of PRRSV inhibitors.
Collapse
Affiliation(s)
- Zhi-Zheng Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China
| | - Chen-Xu Xiao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China
| | - Wen-Li Huang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China
| | - Yang Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China
| | - Hui-Ting Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China
| | - Zhang Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China
| | - Sheng-Hao Peng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China
| | - Zigong Wei
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China; Hubei Jiangxia Laboratory, Wuhan, Hubei, PR China.
| |
Collapse
|
43
|
Cong L, Li SC, Wei ZT, Yu SJ, Huang ZH, Cui YY, Yang QQ, Ding LL, Pan Q, Liu L, Li Y, Ran C. Identification and functional characterization of UDP-glycosyltransferase genes involved in cyetpyrafen resistance in Panonychus citri (McGregor). PEST MANAGEMENT SCIENCE 2025; 81:3212-3219. [PMID: 39887928 DOI: 10.1002/ps.8691] [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: 11/13/2024] [Revised: 12/15/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Cyetpyrafen is a newly developed acaricide. The citrus red mite, Panonychus citri (McGregor), is an important citrus pest that has developed resistance to cyetpyrafen. Uridine diphosphate-glycosyltransferases (UGTs) have been widely reported to be associated with resistance to multiple acaricides. However, it has been rarely documented that UGT genes participate in cyetpyrafen resistance in P. citri. RESULTS In this study, a significantly upregulated UGT gene, PcUGT201E1, was identified in P. citri using transcriptome analysis. Expression of PcUGT201E1 was significantly upregulated at all stages in the cyetpyrafen-resistant strain and silencing PcUGT201E1 significantly increased the susceptibility of P. citri to cyetpyrafen. Molecular docking of PcUGT201E1 with uridine diphosphate glucose (UDPG) and cyetpyrafen indicated that UDPG and cyetpyrafen can interact with PcUGT202E1 via hydrogen bonds. Heterologous expression and in vitro functional assays revealed that enzyme activity could be inhibited by cyetpyrafen and that recombinant PcUGT201E1 can deplete cyetpyrafen. CONCLUSION These results indicated that PcUGT201E1 participates in cyetpyrafen resistance in P. citri by sequestration, and provided a molecular foundation for understanding cyetpyrafen resistance in P. citri. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Lin Cong
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Si-Chen Li
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Zhi-Tang Wei
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Shi-Jiang Yu
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Ze-Hao Huang
- College of Horticulture, Sichuan Agricultural University, Chengdu, China
| | - Yang-Yang Cui
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Qi-Qi Yang
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Li-Li Ding
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Qi Pan
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Liu Liu
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Yang Li
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| | - Chun Ran
- Citrus Research Institute, Southwest University, National Citrus Engineering Research Center, Chongqing, China
| |
Collapse
|
44
|
Yasmine J, Sola P, Rymbai E, Dutta BJ, Buragohain S. Computational phytochemical screening for Parkinson's disease therapeutics: c-Abl and beyond. Comput Biol Chem 2025; 116:108370. [PMID: 39952103 DOI: 10.1016/j.compbiolchem.2025.108370] [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: 11/25/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
Parkinson's disease (PD), a rapidly growing neurodegenerative disorder, is characterized by intracellular α-synuclein aggregates. The tyrosine kinase c-Abl plays a critical role in PD pathogenesis. This study aimed to identify novel c-Abl inhibitors from natural products using molecular docking and dynamics simulations. We explored phytochemicals from Indian Medicinal Plants, Phytochemistry and Therapeutics (IMPPAT) database and employed molecular docking and molecular dynamics to discover c-Abl inhibitors. Three potential hits: IMPHY008934, IMPHY009589, and IMPHY006310 were identified. These compounds demonstrated comparable binding affinity to Nilotinib, a comparison drug. Toxicity predictions revealed IMPHY008934 and IMPHY009589 exhibited lower toxicity than Nilotinib. Molecular dynamics simulations confirmed the stability of IMPHY009589 and IMPHY008934 with c-Abl. Density functional theory (DFT) analysis showed that IMPHY006310 and IMPHY008934 displayed enhanced reactivity and polarizability. Our findings suggest these natural compounds may target c-Abl in PD pathogenesis and possibly downregulate the overexpressed α-synuclein and may serve as promising leads for PD therapy.
Collapse
Affiliation(s)
- Jesmina Yasmine
- Department of Pharmacology, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Assam 781125, India
| | - Piyong Sola
- Department of Pharmacology, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Assam 781125, India.
| | - Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, (A constituent College of JSS Academy of Higher Education & Research), Ooty, India
| | - Bhaskar Jyoti Dutta
- Department of Pharmacology, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Assam 781125, India
| | - Sankarkishor Buragohain
- Department of Pharmacology, NETES Institute of Pharmaceutical Science, NEMCARE Group of Institutions, Mirza, Assam 781125, India
| |
Collapse
|
45
|
Embaby EM, Megahed A, Mostafa SA, Samy A, Yousef EH, Dawood AF, Eldesoqui M. L-Citrulline Alleviates Testicular Ischemia/Reperfusion Injury in Rats by Modulating eNOS/iNOS Induced Nitric Oxide Production, Inflammation, and Apoptosis. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2025; 343:590-607. [PMID: 40059720 DOI: 10.1002/jez.2913] [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: 01/20/2025] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 05/07/2025]
Abstract
Testicular ischemia/reperfusion injury (TI/RI) is a significant clinical contributor to subfertility and infertility resulting from testicular torsion and subsequent detortion. Insufficient nitric oxide (NO) synthesis in TI/RI can result in endothelial dysfunction, as the vascular endothelium fails to produce sufficient NO to sustain appropriate vasodilation and blood perfusion. Many studies have found that NO plays an important role in the I/RI and its increase or decrease can affect the progression and outcome of I/RI. However, the role of NO in I/RI is controversial and complicated. NO derived by endothelial NO synthase (eNOS) shows a protective role in I/RI, while excessive NO derived by inducible NO synthase (iNOS) accelerates inflammation and increases oxidative stress, further aggravating I/RI. Nevertheless, the overexpression of eNOS may exacerbate I/RI. Here we try to investigate the new progress in the understanding of the roles of NO during I/RI. This study examined the interplay between cytotoxic and cytoprotective mechanisms underpinning NO produced from L-citrulline (L-Cit) on TI/R injured rats. Thirty-two adult Sprague-Dawley albino rats were equally randomized into the following groups: normal control group, sham group, TI/R group (3 h/4 h), and TI/R + L-Cit group (600 mg/kg) orally at 1 h before reperfusion. Compared to the TI/R-operated group, the injection of L-Cit markedly enhanced serum concentrations of reproductive hormones (p < 0.05). Elevated SOD, CAT, and GPx activity, along with reduced MDA and NO concentrations, indicated a diminished oxidative stress. The testicular levels of TNF-α, IL-1β, caspase-3, BAX, eNOS, iNOS, and NF-κB p65 were markedly reduced. Histopathological analysis corroborated the protective effect of L-Cit. The findings confirmed molecular models, demonstrating that L-Cit inhibited eNOS, iNOS, and IKKβ. The results showed that giving torsioned rats NO made from L-Cit protected them against hormonal imbalance, oxidative stress, inflammation, and apoptosis in I/RI. This makes L-Cit even more important for protecting against tissue I/RI during surgery. L-Cit not only promoted NO synthesis through eNOS activation, but it also facilitated the neutralization of iNOS production and its pathogenic NO levels during the reperfusion phase in I/R-injured rats.
Collapse
Affiliation(s)
- Eman M Embaby
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Aya Megahed
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Sally Abdallah Mostafa
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Alaa Samy
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, Egypt
| | - Eman H Yousef
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, Damietta, Egypt
| | - Amal F Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
46
|
Barve PR, Barvkar VT, Giri AP, Kotkar HM. High levels of sinigrin trigger synthesis of fatty acids in Plutella xylostella (L.). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101424. [PMID: 39854962 DOI: 10.1016/j.cbd.2025.101424] [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: 10/07/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Diamondback moth (Lepidoptera: Plutellidae; Plutella xylostella L.) is a specialist insect of the Brassicaceae family, damaging economically important crops, such as cabbage and cauliflower. Glucosinolates, also known as 'mustard oil bombs' are present in all Brassicaceae members, of which sinigrin (allyl-glucosinolate or 2-propenyl-glucosinolate) is a major aliphatic compound. During herbivory, glucosinolates are converted to toxic isothiocyanates that deter insect pests. P. xylostella possesses glucosinolate sulfatases that desulfate them. Such a conversion renders them unfit for degradation to toxic products. Changes in the larval performance prompted us for RNA sequencing to understand probable adaptation mechanism under sinigrin stress. Differentially expressed genes were found to be related to larval cuticle proteins. Further, gene ontology and KEGG (Kyoto Encyclopedia of Genes and Genomes) analyses depict genes belonging to the categories, integral component of membrane, cellular processes and those involved in biosynthesis of fatty acids. Upregulation of cuticular genes viz. larval cuticle protein-17 (LCP-17), cuticular protein-19 (2CP-19) and ATP binding cassette transporter C7 (ABCC7), ABCC16 was validated by qRT-PCR. Liquid chromatography quadrupole time of flight mass spectrometry analysis of whole larvae feeding on sinigrin and their separated cuticle, depicted abundance of fatty acids. Changes in the topography of the larval cuticle were evident by scanning electron microscopy. Expression of PxABCH1 was corroborated to its role in the transport of cuticular lipids. Notably, molecular docking of PxABCH1 with cuticular fatty acids showed favorable binding interactions. To summarize, integrated transcriptomic and metabolomic analyses suggest that in response to a diet containing a high dose of sinigrin, P. xylostella re-programs metabolic pathways related to fatty acid biosynthesis that directly influence insect development.
Collapse
Affiliation(s)
- Pranoti R Barve
- Department of Botany, Savitribai Phule Pune University, Ganeshkhind, Pune- 411 007, Maharashtra, India
| | - Vitthal T Barvkar
- Department of Botany, Savitribai Phule Pune University, Ganeshkhind, Pune- 411 007, Maharashtra, India
| | - Ashok P Giri
- Plant Molecular Biology Unit, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Hemlata M Kotkar
- Department of Botany, Savitribai Phule Pune University, Ganeshkhind, Pune- 411 007, Maharashtra, India.
| |
Collapse
|
47
|
Ávila-Avilés RD, Bahena-Culhuac E, Hernández-Hernández JM. (-)-Epicatechin metabolites as a GPER ligands: a theoretical perspective. Mol Divers 2025; 29:2099-2115. [PMID: 39153018 DOI: 10.1007/s11030-024-10968-9] [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: 05/02/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Diet habits and nutrition quality significantly impact health and disease. Here is delve into the intricate relationship between diet habits, nutrition quality, and their direct impact on health and homeostasis. Focusing on (-)-Epicatechin, a natural flavanol found in various foods like green tea and cocoa, known for its positive effects on cardiovascular health and diabetes prevention. The investigation encompasses the absorption, metabolism, and distribution of (-)-Epicatechin in the human body, revealing a diverse array of metabolites in the circulatory system. Notably, (-)-Epicatechin demonstrates an ability to activate nitric oxide synthase (eNOS) through the G protein-coupled estrogen receptor (GPER). While the precise role of GPER and its interaction with classical estrogen receptors (ERs) remains under scrutiny, the study employs computational methods, including density functional theory, molecular docking, and molecular dynamics simulations, to assess the physicochemical properties and binding affinities of key (-)-Epicatechin metabolites with GPER. DFT analysis revealed distinct physicochemical properties among metabolites, influencing their reactivity and stability. Rigid and flexible molecular docking demonstrated varying binding affinities, with some metabolites surpassing (-)-Epicatechin. Molecular dynamics simulations highlighted potential binding pose variations, while MMGBSA analysis provided insights into the energetics of GPER-metabolite interactions. The outcomes elucidate distinct interactions, providing insights into potential molecular mechanisms underlying the effects of (-)-Epicatechin across varied biological contexts.
Collapse
Affiliation(s)
- Rodolfo Daniel Ávila-Avilés
- Laboratory of Epigenetics of Skeletal Muscle Regeneration, Department of Genetics and Molecular Biology, Centre for Research and Advanced Studies of IPN (CINVESTAV), Mexico City, Mexico
- Transdisciplinary Research for Drug Discovery, Sociedad Mexicana de Epigenética y Medicina Regenerativa A. C. (SMEYMER), Mexico City, Mexico
| | - Erick Bahena-Culhuac
- Laboratory of Epigenetics of Skeletal Muscle Regeneration, Department of Genetics and Molecular Biology, Centre for Research and Advanced Studies of IPN (CINVESTAV), Mexico City, Mexico
- Transdisciplinary Research for Drug Discovery, Sociedad Mexicana de Epigenética y Medicina Regenerativa A. C. (SMEYMER), Mexico City, Mexico
| | - J Manuel Hernández-Hernández
- Laboratory of Epigenetics of Skeletal Muscle Regeneration, Department of Genetics and Molecular Biology, Centre for Research and Advanced Studies of IPN (CINVESTAV), Mexico City, Mexico.
| |
Collapse
|
48
|
Alshammari SO, Alshammari QA. Natural product-derived ALK inhibitors for treating ALK-driven lung cancers: an in silico study. Mol Divers 2025; 29:1969-1982. [PMID: 39115579 DOI: 10.1007/s11030-024-10953-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/29/2024] [Indexed: 05/16/2025]
Abstract
Anaplastic lymphoma kinase (ALK)-driven lung cancer represents a critical therapeutic target, demanding innovative approaches for the identification of effective inhibitors. Anaplastic lymphoma kinase (ALK), a key protein involved in the pathogenesis of ALK-driven lung cancers, has been the focus of extensive drug discovery efforts. This study employed a comprehensive computational drug discovery approach, integrating virtual screening with the Lipinski filter, re-docking, molecular dynamics (MD) simulations, and free energy calculations to identify potential inhibitors from a natural compound library. Utilizing the MTiOpenScreen web server, we screened for compounds that exhibit favorable interactions with ALK, resulting in 1227 compounds with virtual screening scores ranging from - 10.2 to - 3.7 kcal/mol. Subsequent re-docking of three selected compounds (ZINC000059779788, ZINC000043552589, and ZINC000003594862) and one reference compound against ALK yielded docking scores - 10.4, - 10.2, - 10.2, and - 10.1 kcal/mol, respectively. These compounds demonstrated promising interactions with ALK, suggesting potential inhibitory effects. Advanced analyses, including MD simulation and binding free energy calculations, further supported the potential efficacy of these compounds. MD simulations, particularly the root mean square deviation (RMSD) and root mean square fluctuation (RMSF) analyses, revealed that compounds ZINC000059779788 and ZINC000003594862 achieved better stability compared to compound ZINC000043552589. These stable conformations suggest effective binding over time. Free energy calculations using the MM/GBSA method showed that ZINC000059779788 had the most favorable binding energy, indicating a strong and stable interaction with the ALK protein. The promising computational findings from this study emphasize the necessity for additional experimental testing to verify the therapeutic efficacy of these natural compounds for treating lung cancers.
Collapse
Affiliation(s)
- Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, College of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia.
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
49
|
Almeida-Bezerra JW, da Costa Silva JT, Morais-Braga MFB, da Cruz RP, Alencar GG, Alves DS, de Sousa Rodrigues EY, de Sousa SG, de Menezes IRA, Rocha JE, Filho JMB, Leite dos Santos CA, Costa AR, Domiciano CB, de Lima LR, Coutinho HDM. ADME/Tox study and the effect of β-Caryophyllene on the resistant strain of Staphylococcus aureus carrying the QacA/B efflux pump gene. Toxicol Rep 2025; 14:101929. [PMID: 39968054 PMCID: PMC11833615 DOI: 10.1016/j.toxrep.2025.101929] [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: 12/03/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/20/2025] Open
Abstract
The Gram-positive bacterium Staphylococcus aureus is responsible for causing both community-acquired and healthcare-associated infections, and it exhibits high antibiotic resistance due to the presence of efflux pumps. These pumps, such as QacA and QacB, are proteins that expel toxic substances, including antibiotics, making infection treatment more difficult. Among the alternatives to combat this resistance are terpenes, like β-caryophyllene, which have the potential to inhibit these efflux pumps due to their nonpolar nature. Considering this, the objective of this work is to investigate the ability of the mentioned terpene to act as an inhibitor of the QacA/B pump in S. aureus, as well as to analyze its pharmacokinetic and toxicological properties in silico. Initially, a molecular docking simulation was performed using the CryoEM structure of the QacA protein with the software AutoDock VINA to evaluate the interactions between β-caryophyllene and the target protein. Subsequently, in vitro assays were conducted to determine the Minimum Inhibitory Concentration (MIC) of β-caryophyllene and its ability to inhibit the efflux pump in combination with ampicillin in resistant strains of S. aureus. Additionally, in silico ADMET predictions were performed using the SwissADME platform. The results showed that the terpene enhanced the action of ampicillin, reducing the minimum inhibitory concentration (MIC) by 50 %. However, it was not able to reduce the MIC of ethidium bromide. The in silico analysis indicated that β-caryophyllene has good bioavailability and drug-likeness characteristics, but with limitations in its gastrointestinal absorption and brain permeability. The study concludes that β-caryophyllene is a promising candidate as an adjuvant in the treatment of antibiotic-resistant infections, especially due to its ability to partially inhibit efflux pumps.
Collapse
Affiliation(s)
| | | | | | - Rafael Pereira da Cruz
- Department of Biological Sciences, Cariri Regional University — URCA, Crato, CE 63105-000, Brazil
| | | | - Daniel Sampaio Alves
- Department of Biological Sciences, Cariri Regional University — URCA, Crato, CE 63105-000, Brazil
| | | | - Simone Galdino de Sousa
- Department of Biological Sciences, Cariri Regional University — URCA, Crato, CE 63105-000, Brazil
| | | | - Janaína Esmeraldo Rocha
- Center of Science and Technology CCT - State University of Ceara - UECE, Fortaleza, CE 63100-000, Brazil
| | | | | | - Adrielle Rodrigues Costa
- Center for Agrarian Sciences and Biodiversity, Federal University of Cariri - UFCA, Crato, CE 63133-610, Brazil
| | | | - Lucia Raquel de Lima
- Department of Biological Chemistry, Regional University of Cariri – URCA, Crato, Ceará 63105–000, Brazil
| | | |
Collapse
|
50
|
Wang S, Du R, Liu J, Zhong W, Zhang C, Jiang X, Wang X, Wu Q, Tong G, Luo L. Multi-approach analysis reveals the mechanism by which Shugan Xiaozhi decoction protects against metabolic dysfunction-associated steatohepatitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156712. [PMID: 40220418 DOI: 10.1016/j.phymed.2025.156712] [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: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is a human health-threatening hepatic disease with limited treatment strategies. As a clinical Traditional Chinese Medicine compound for MASH, Shugan Xiaozhi (SGXZ) decoction has a definite effect, but its mechanism in treating MASH is still not very clear. PURPOSE Exploring the potential mechanism of SGXZ decoction in treating MASH through multiomics and animal experimental validation. METHODS UPLC-ESI-MS method was used to identify the main components of SGXZ decoction. Periodic acid-schiff (PAS), picrosirius red (PSR), and oil red o staining were used to assess the effect of SGXZ decoction on MCD-induced MASH mouse model. The mechanism of SGXZ decoction on MASH was analyzed using multiomics techniques. TUNEL staining, western blot (WB), immunohistochemistry (IHC), kits, transmission electron microscopy (TEM), and immunofluorescence (IF) were used to validate the mechanism of SGXZ decoction on MASH. Finally, molecular docking and molecular dynamics simulation were used to verify the targeting between key components of SGXZ decoction and important targets for intervention. RESULTS Through UPLC-ESI-MS analysis, 30 main active ingredients were obtained from SGXZ decoction. SGXZ decoction improved MASH, as evidenced by the improvement in histopathology, hepatic function indexes, lipid and fibrosis indicators. Both proteomic and transcriptomic results suggested an important role for ferroptosis in SGXZ decoction intervention in MASH, ferroptosis-related pathways were the main significant pathways obtained from these analyses. In addition, SGXZ decoction treatment reduced cell death, inflammation, and oxidative stress levels and restored impaired mitochondrial morphology in MCD-induced MASH mice. Furthermore, Mechanism experiments proved that SGXZ decoction treatment improved iron metabolism and lipid peroxidation imbalance and activated the Xc- system in MASH mice. CONCLUSION SGXZ decoction does have a therapeutic effect on MASH, and its mechanism may be related to its regulation of p53/ SLC7A11/GPX4 pathway to reduce ferroptosis.
Collapse
Affiliation(s)
- Shuai Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Ruili Du
- The First Clinical Medical College of Henan University of Chinese Medicine, No. 19, Renmin Road, Jinshui District, Henan, 450003, PR China
| | - Jiahui Liu
- Department of Nephrology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong 518033, PR China
| | - Weichao Zhong
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Chunmei Zhang
- School of Basic Medical Science of Luoyang Polytechnic, No. 6 Keji Avenue, Yibin District, Henan, 471099, PR China
| | - Xia Jiang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, PR China
| | - Xiaohui Wang
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao ln-Depth Cooperation Zone in Hengqin, 519000, PR China.
| | - Guangdong Tong
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| | - Lidan Luo
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| |
Collapse
|