1
|
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
|
2
|
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
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
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
|
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
|
Alhagri IA, Al-Hazmy SM, Alammari SS, Alminderej FM, Messaoudi S, Aroua LM. Synthesis and characterization of a solvatochromic urea-schiff base derivative: Investigating optical properties, hydrogen bonding effect, copper ion sensing, computational analysis, DNA and β-cyclodextrin interactions. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 340:126337. [PMID: 40344888 DOI: 10.1016/j.saa.2025.126337] [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: 02/28/2025] [Revised: 04/26/2025] [Accepted: 05/03/2025] [Indexed: 05/11/2025]
Abstract
This study investigates the fluorescence behavior of the synthesized compound (E)-1-(4-chloro-2-(((2-hydroxynaphthalen-1-yl) methylene) amino) phenyl)-3-(naphthalen-1-yl) urea (3DB) in various solvents. A significant increase in fluorescence intensity was observed when transitioning from ethanol to less polar solvents like CH2Cl2, CHCl3, and CCl4, indicating enhanced fluorescence due to reduced non-radiative processes. Emission wavelengths remained stable with minor shifts (5-6 nm), while significant blue shifts in absorption occurred in water due to strong hydrogen bonding. Fluorescence spectra showed red shifts (519 nm in water, 508 nm in glycerol, and 486 nm in ethylene glycol), highlighting the impact of hydrogen bonding on electronic transitions. Emission intensity in water was six times higher than in ethylene glycol, suggesting that strong hydrogen bonds stabilize the excited state. The study also revealed that 3DB exhibits a large Stokes shift, avoiding reabsorption of emitted light (inner filter effect). Fluorescence was completely quenched by low concentrations of copper ions, demonstrating 3DB's potential as a copper sensor. Density functional theory (DFT) and time-dependent DFT (TDDFT) calculations indicated that luminescence quenching in the Cu(II) complex is due to intramolecular charge transfer (ICT). Additionally, 3DB formed stable complexes with DNA and β-cyclodextrin (β-CD), with binding constants (Kb) of 1.30 × 103 M-1 and 1.89 × 103 M-1, respectively, and negative Gibbs free energy values, indicating spontaneous interactions. Fluorescence spectroscopy confirmed DNA binding, showing a 49.62 % increase in intensity and a 4 nm blue shift, consistent with groove-binding. Docking studies further supported favorable interactions with DNA. These results underscore 3DB's potential in sensing, imaging, environmental monitoring, and biological applications.
Collapse
Affiliation(s)
- Ibrahim A Alhagri
- Department of Chemistry, College of Science, Qassim University, Buraydah, Qassim 51452, Kingdom of Saudi Arabia
| | - Sadeq M Al-Hazmy
- Department of Chemistry, College of Science, Qassim University, Buraydah, Qassim 51452, Kingdom of Saudi Arabia
| | - Sarah S Alammari
- Department of Chemistry, College of Science, Qassim University, Buraydah, Qassim 51452, Kingdom of Saudi Arabia
| | - Fahd M Alminderej
- Department of Chemistry, College of Science, Qassim University, Buraydah, Qassim 51452, Kingdom of Saudi Arabia
| | - Sabri Messaoudi
- Department of Chemistry, College of Science, Qassim University, Buraydah, Qassim 51452, Kingdom of Saudi Arabia
| | - Lotfi M Aroua
- Department of Chemistry, College of Science, Qassim University, Buraydah, Qassim 51452, Kingdom of Saudi Arabia; Laboratory of (Bio)Organic, Structural and Polymer Chemistry (LR99ES14), Department of Chemistry, Faculty of Sciences of Tunis, Tunis El-Manar University, El-Manar, I 2092 Tunis, Tunisia; Carthage University, Faculty of Sciences of Bizerte, 7021 Jarzouna, Bizerte, Tunisia.
| |
Collapse
|
11
|
Han Y, Ding X, Tan J, Sun Y, Duan Y, Liu Z, Zheng G, Lu D. Sequence and taxonomic feature evaluation facilitated the discovery of alcohol oxidases. Synth Syst Biotechnol 2025; 10:907-915. [PMID: 40386440 PMCID: PMC12083922 DOI: 10.1016/j.synbio.2025.04.014] [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: 01/20/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Recent advancements in data technology offer immense opportunities for the discovery and development of new enzymes for the green synthesis of chemicals. Current protein databases predominantly prioritize overall sequence matches. The multi-scale features underpinning catalytic mechanisms and processes, which are scattered across various data sources, have not been sufficiently integrated to be effectively utilized in enzyme mining. In this study, we developed a sequence- and taxonomic-feature evaluation driven workflow to discover enzymes that can be expressed in E. coli and catalyze chemical reactions in vitro, using alcohol oxidase (AOX) for demonstration, which catalyzes the conversion of methanol to formaldehyde. A dataset of 21 reported AOXs was used to construct sequence scoring rules based on features, including sequence length, structural motifs, catalytic-related residues, binding residues, and overall structure. These scoring rules were applied to filter the results from HMM-based searches, yielding 357 candidate sequences of eukaryotic origin, which were categorized into six classes at 85 % sequence similarity. Experimental validation was conducted in two rounds on 31 selected sequences representing all classes. Among these selected sequences, 19 were expressed as soluble proteins in E. coli, and 18 of these soluble proteins exhibited AOX activity, as predicted. Notably, the most active recombinant AOX exhibited an activity of 8.65 ± 0.29 U/mg, approaching the highest activity of native eukaryotic enzymes. Compared to the established UniProt-annotation-based workflow, this feature-evaluation-based approach yielded a higher probability of highly active recombinant AOX (from 8.3 % to 19.4 %), demonstrating the efficiency and potential of this multi-dimensional feature evaluation method in accelerating the discovery of active enzymes.
Collapse
Affiliation(s)
- Yilei Han
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Xuwei Ding
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, Shanghai, 200237, China
| | - Junjian Tan
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yajuan Sun
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, Shanghai, 200237, China
| | - Yunjiang Duan
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Zheng Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Gaowei Zheng
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, Shanghai, 200237, China
| | - Diannan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
12
|
Correas NH, Martínez AR, Abellán A, Sánchez HP, Tejada L. Curing strategies and bioactive peptide generation in ham: In vitro digestion and in silico evaluation. Food Chem 2025; 484:144360. [PMID: 40252451 DOI: 10.1016/j.foodchem.2025.144360] [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: 01/21/2025] [Revised: 03/30/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
This study assessed the impact of curing salts and maturation times on peptide production and bioactivity in dry-cured hams using in vitro and in silico methods. Ninety-six hams underwent six curing treatments and two maturation stages (38 % and 42 % weight loss). Mass spectrometry identified bioactive peptides, while in silico tools predicted their bioactivities. Reduced sodium nitrifying salts (treatment IX) and 42 % weight loss showed the most significant results, enhancing low-molecular-weight peptides generation (EE, VG, VD) linked to high functionality. Antioxidant and antihypertensive activities were prominent in samples with 42 % weight loss. Peptides under 1.5 kDa were more abundant at advanced maturation stages. In silico analyses predicted ACE and DPP-IV inhibition and antioxidant effects. Dipeptides like DG, ES, and DV showed similarities to FDA-approved molecules, suggesting potential therapeutic uses. The study highlights those specific treatments boost biopeptides formation, requiring further research on their potential as functional foods or therapeutic agents.
Collapse
Affiliation(s)
- Noelia Hernández Correas
- Faculty of Pharmacy and Nutrition, Universidad Católica de Murcia-UCAM, Campus de los Jerónimos, 30107 Murcia, Spain.
| | - Alejandro Rodríguez Martínez
- Structural Bioinformatics and High Performance Computing (BIO-HPC) Research Group, Universidad Católica de Murcia-UCAM, Campus de los Jerónimos, 30107 Murcia, Spain
| | - Adela Abellán
- Faculty of Pharmacy and Nutrition, Universidad Católica de Murcia-UCAM, Campus de los Jerónimos, 30107 Murcia, Spain
| | - Horacio Pérez Sánchez
- Structural Bioinformatics and High Performance Computing (BIO-HPC) Research Group, Universidad Católica de Murcia-UCAM, Campus de los Jerónimos, 30107 Murcia, Spain.
| | - Luis Tejada
- Faculty of Pharmacy and Nutrition, Universidad Católica de Murcia-UCAM, Campus de los Jerónimos, 30107 Murcia, Spain
| |
Collapse
|
13
|
Purohit K, Pathak R, Hayes E, Sunna A. Novel bioactive peptides from ginger rhizome: Integrating in silico and in vitro analysis with mechanistic insights through molecular docking. Food Chem 2025; 484:144432. [PMID: 40279907 DOI: 10.1016/j.foodchem.2025.144432] [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: 11/26/2024] [Revised: 04/14/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Ginger (Zingiber officinale) is widely recognised for its functional benefits, primarily attributed to its diverse phytochemicals. However, its proteome remains largely unexplored. This study hypothesised that isolated peptides may exhibit different bioactivities or more targeted mechanisms of action and could be investigated at a molecular level. Proteins were enzymatically hydrolysed under five conditions, and peptides were identified using LC-MS/MS. In silico screening suggested antioxidant, ACE-inhibitory, and antibacterial properties, further assessed through molecular docking and in vitro validation. 41 potentially bioactive peptides were identified. In vitro assays confirmed these properties for selected peptides, P1 (GSPVWIIPEPT), P2 (FASYPVKK), P3 (GPEKIFYDGPYL), and P4 (IAISPSYPIK). Notably, P4 exhibited potent mixed-type ACE-inhibition and bacteriostatic effects. Molecular docking provided mechanistic insights into these interactions. These findings highlight ginger as a promising source of bioactive peptides while underscoring the need to complement AI tools with in vitro and in vivo validations due to observed discrepancies.
Collapse
Affiliation(s)
- Kruttika Purohit
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia; Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia's Bioactives (FAAB), Sydney, NSW 2109, Australia
| | - Rachana Pathak
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia; Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia's Bioactives (FAAB), Sydney, NSW 2109, Australia
| | - Evan Hayes
- Factors Group Australia, Sydney, NSW 2116, Australia
| | - Anwar Sunna
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia; Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia's Bioactives (FAAB), Sydney, NSW 2109, Australia.
| |
Collapse
|
14
|
Soda AK, Huang T, Zhou W, Chen H, Jiang H, Jadhav SB, Xing Z, Yu Y, Tian L, Wong DF, Perlmutter JS, Ni R, Benzinger TLS, Tu Z. Synthesis and in vivo biological characterization of six carbon-11 sigma-1 receptor radiotracers in rodent and nonhuman primate. Bioorg Med Chem 2025; 126:118218. [PMID: 40339216 DOI: 10.1016/j.bmc.2025.118218] [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: 02/05/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/10/2025]
Abstract
Six enantiomers of three racemic sigma-1 receptor (σ1R) ligands were resolved, and absolute configuration was determined. Their high σ1R potency and selectivity were determined through in vitro binding assays, further validated by molecular docking analysis. Central Nervous System Multiparameter Optimization algorithm (CNS MPO) predicts efficient brain penetration for these enantiomers. Six C-11 radiotracers were radiosynthesized successfully, ex vivo biodistribution in rats showed that (-)-[11C]7 had high brain uptake of ∼4.8-fold for 5 min versus 60 min. Mouse brain PET imaging studies showed (-)-[11C]7 and (-)-[11C]16 have in vivo binding specificity for σ1R. Macaque PET scans showed high brain uptake for all six radiotracers, with (-)-[11C]7 peaked at ∼45 min (SUV 2.5), possessing the best washout kinetics and highest cerebellum-to-white matter ratio (∼3.1), in agreement with in vitro or ex vivo measures of σ1R expression. Radiometabolite analysis showed that no newly formed radiometabolite was observed post-injection of (-)-[11C]7. Our data suggest that further evaluation is warranted to determine that (-)-[11C]7 is a suitable PET radiotracer for imaging σ1R in the brain of animal and human.
Collapse
Affiliation(s)
- Anil Kumar Soda
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tianyu Huang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wenjuan Zhou
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hong Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Jiang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sandip B Jadhav
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhimin Xing
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yanbo Yu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Linlin Tian
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dean F Wong
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Programs in Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
15
|
Bisericaru DM, Pérez-Jiménez M, Romero-Muñoz M, Caldera F, Bordignon S, Trotta F, Matencio A. Optimizing 6-benzylaminopurine for micropropagation: A cyclodextrin monomers and polymers approach. Carbohydr Polym 2025; 362:123658. [PMID: 40409815 DOI: 10.1016/j.carbpol.2025.123658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/03/2025] [Accepted: 04/24/2025] [Indexed: 05/25/2025]
Abstract
In the face of escalating climate change challenges, there is an imperative to enhance crop resilience and productivity. The plant growth regulator 6-benzylaminopurine (BA[P]) shows considerable potential in agriculture, yet its practical use is hampered by low water solubility and susceptibility to light and heat instability. This study addresses these limitations and addresses the transformative potential of complexing 6-BAP with cyclodextrins (CDs) or CD-based nanosponges (CD-NS). Calculations of inclusion constants of various CDs gave the opportunity to synthetize the best CD-NSs for BAP complexation, being β-CD the best one (KF = 289.50 ± 23.16 M-1). A general enhancing effect observed for CD-based polymers than monomeric ones: The complexation efficiency was determined and compared (67 % monomers vs average 89 % for the polymers), as well as their improved water solubility, controlled release (βNS-CDI > β-CD > βNS-CA), and increased bioavailability in vitro. Furthermore, CDs and CD-NS offer effective protection against thermal degradation, potentially extending the shelf life of 6-BAP formulations, which is shown by an enlarged effect in shoots of Prunus salicina LIndl, with applications in agriculture. Thus, shoots cultured in vitro in culture media supplemented with these complexes showed an improvement in the shoot production rate along with an increase in the endogenous BAP and riboside BAP. With the main objective to demonstrate the increase of capacity of BAP-based complexes, this research lays the foundation of the eco-friendly use of dextrin-based polymers for micropropagation improvement.
Collapse
Affiliation(s)
- Daniel Mihai Bisericaru
- Department of Chemistry, Nis Interdepartmental Centre, University of Turin, Via P. Giuria 7, 10125 Turin, Italy
| | - Margarita Pérez-Jiménez
- Department of Plant Biotechnology, Genomics and Breeding, Murcian Institute of Agricultural and Environmental Research and Development (IMIDA), Spain
| | - Miriam Romero-Muñoz
- Department of Plant Biotechnology, Genomics and Breeding, Murcian Institute of Agricultural and Environmental Research and Development (IMIDA), Spain
| | - Fabrizio Caldera
- Department of Chemistry, Nis Interdepartmental Centre, University of Turin, Via P. Giuria 7, 10125 Turin, Italy
| | - Simone Bordignon
- Department of Chemistry, Nis Interdepartmental Centre, University of Turin, Via P. Giuria 7, 10125 Turin, Italy
| | - Francesco Trotta
- Department of Chemistry, Nis Interdepartmental Centre, University of Turin, Via P. Giuria 7, 10125 Turin, Italy
| | - Adrián Matencio
- Department of Chemistry, Nis Interdepartmental Centre, University of Turin, Via P. Giuria 7, 10125 Turin, Italy; Departamento de Bioquímica y Biología Molecular-A, Facultad de Biología, Universidad de Murcia-Regional Campus of International Excellence "Campus Mare Nostrum", E-30100 Murcia, Spain.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Yu H, Chen Q, Xu TC, Wang Y, Xu WF, Li M, Zhang X, Zhao C, Zhang DL, Jin PF, Li SY, Tang SA. Bioactive terpenoids and sterols from the fruiting bodies of Fomitopsis pinicola. PHYTOCHEMISTRY 2025; 236:114510. [PMID: 40258453 DOI: 10.1016/j.phytochem.2025.114510] [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: 10/25/2024] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
A chemical investigation of the fruiting bodies of Fomitopsis pinicola was conducted, resulting in the isolation of 51 compounds, which included 16 previously undescribed lanostane triterpenes (1-14, 16-17), 30 known analogs (15, 18-46) as well as two sterols (47-48) and three diterpenes (49-51). Comprehensive analyses utilizing 1D, 2D NMR, MS and IR spectroscopy and DFT calculations of 13C NMR chemical shifts have elucidated the structures of the compounds in question. Two undescribed compounds in addition to fomitosides F, G and K showed cytotoxicity against MCF-7 cell line with IC50 values ranging from 11.01 to 31.92 μM. Meanwhile, these isolates showed α-glucosidase inhibitory activity, and compound 7 showed the IC50 value of 9.22 μM.
Collapse
Affiliation(s)
- Huan Yu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Qian Chen
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Tian-Chi Xu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Yue Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Wen-Feng Xu
- Department of Pharmaceutical Science, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing, Beijing, 100730, PR China
| | - Min Li
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Xu Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Chuan Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, PR China
| | - Dong-Li Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Peng-Fei Jin
- Department of Pharmaceutical Science, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing, Beijing, 100730, PR China
| | - Shao-Yong Li
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China.
| | - Sheng-An Tang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China.
| |
Collapse
|
19
|
Hannig P, Gargallo R, Mazzini S, Borgonovo G, Zuccolo M, Táborská E, Táborský P. Interaction process behind the strong stabilization of G-quadruplexes by alkaloid fagaronine. Biophys Chem 2025; 323:107443. [PMID: 40252303 DOI: 10.1016/j.bpc.2025.107443] [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: 01/26/2025] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Benzo[c]phenanthridine alkaloids are known for their stabilizing effects on non-canonical DNA structures, particularly G-quadruplexes (G4s). In this study, the interaction of fagaronine, a rare benzo[c]phenanthridine alkaloid, with several DNA structures (including B-DNA, parallel, antiparallel and hybrid G4s) is studied using molecular fluorescence and circular dichroism (CD) spectroscopy. It has been found that fagaronine significantly enhances the stability of all tested G4 conformations. Furthermore, a study by NMR spectroscopy provided valuable information on the mechanism of interaction of the ligand with the parallel G4 structure adopted by Pu22T14T23, a sequence mutated with respect to that found within the promoter region of the c-myc gene. Remarkably, when compared with data reported in the literature, fagaronine appears to exhibit one of the strongest G4 thermal stabilization effects ever recorded for a small ligand.
Collapse
Affiliation(s)
- Pavel Hannig
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Raimundo Gargallo
- Department of Chemical Engineering and Analytical Chemistry, University of Barcelona, Marti i Franquès 1, 08028 Barcelona, Spain
| | - Stefania Mazzini
- Department of Food, Environmental and Nutritional Sciences (DEFENS), University of Milan, 20133 Milan, Italy
| | - Gigliola Borgonovo
- Department of Food, Environmental and Nutritional Sciences (DEFENS), University of Milan, 20133 Milan, Italy
| | - Marco Zuccolo
- Department of Agricultural and Environmental Sciences - Production, Landscape, Agroenergy (DiSAA), University of Milan, 20133 Milan, Italy
| | - Eva Táborská
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Petr Táborský
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic.
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Huang K, Chen X, Li S, Zhang X, Zhang Y, Zhang Y. Indole alkaloids from Uncaria rhynchophylla and their inhibitory activities against α-glucosidase. PHYTOCHEMISTRY 2025; 236:114490. [PMID: 40147593 DOI: 10.1016/j.phytochem.2025.114490] [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: 11/21/2024] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Sixteen indole alkaloids were isolated from the hook-bearing stems of Uncaria rhynchophylla (Rubiaceae family), including seven undescribed ones, uncarialines F-L (1-5, 7, and 8), and a naturally occurring alkaloid, 3-epicorynanthine (6). Among them, alkaloids 1 and 2 were identified as rare quaternary ammonium alkaloids, and alkaloid 7 exhibited an unprecedented indole alkaloid framework. Their structures were characterized by a comprehensive analysis of NMR, MS, ECD and single-crystal X-ray diffraction. Notably, alkaloid 5 demonstrate potent inhibitory activity against α-glucosidase, with an IC50 value of 18.45 ± 0.77 μM. Furthermore, the inhibitory kinetics of α-glucosidase revealed that alkaloid 5 belong to the mix inhibition type. Molecular docking analysis showed that alkaloid 5 possessed superior binding affinity with α-glucosidase (-10.7 kcal/mol).
Collapse
Affiliation(s)
- Kepu Huang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Xuelin Chen
- Key Laboratory of Tropical Plant Resource and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, 650223, China
| | - Sheng Li
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Xinjian Zhang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Yumei Zhang
- Key Laboratory of Tropical Plant Resource and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Yu Zhang
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|
26
|
El-Sofany WI, Alanezi TD, Latif S, Abdelhedi O, Hamden K. Prodigiosin As N-heterocyclic compound: Production optimization, bioactivity evaluation, and in-silico docking against key enzymes related to inflammation, obesity, diabetes, and the insulin signaling pathway. Enzyme Microb Technol 2025; 188:110639. [PMID: 40187164 DOI: 10.1016/j.enzmictec.2025.110639] [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: 01/15/2025] [Revised: 03/03/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Diabetes is known to cause severe pancreatic inflammation and reduce insulin levels, leading us to investigate the effects of prodigiosin (PG), a red, heterocyclic bacterial compound extracted from Serratia marcescens. The physicochemical and nutritional conditions, along with the extraction solvents for PG, have been optimized for efficient production. PG was produced through bacterial culture, purified by high-performance liquid chromatography (HPLC) and thin-layer chromatography (TLC), characterized by Fourier-transform infrared spectroscopy (FTIR) and ultraviolet (UV) spectroscopy. In vitro, PG effectively inhibited key inflammatory enzymes, such as phospholipase A2 (PLA2) and elastase (ELA), in a dose-dependent manner, achieving maximum inhibition rates of 85.3 and 91.4 % at concentrations of 320 µg/mL, with IC₅₀ values of 63 µg/mL and 54.7 µg/mL, respectively. PG also exhibited a maximum inhibition of 82.4 % for myeloperoxidase (MPO) at a concentration of 160 µg/mL, with an IC₅₀ value of 25.9 µg/mL. This indicates that PG is a good candidate for treating these two metabolic diseases. Moreover, PG shows a significant ability to activate insulin signaling through its capacity to stimulate protein tyrosine phosphatase 1B (PTP1B) and inhibit dipeptidyl peptidase-4 (DPP-4), with IC₅₀ values of 67 and 28 µg/mL, respectively, compared to the specific inhibitors CLM and STG (with IC₅₀ values of 19 and 27 µg/mL, respectively). These powerful affinities, stability, and the durability of PG inhibition of these enzymes are confirmed by the determination of binding energy, ligand efficiency, and estimated inhibition constant (Ki). Conclusion: PG benefits from sustainable, cost-effective biological production and exhibits potent anti-inflammatory, antioxidant, and anti-diabetic properties, positioning it as a promising candidate for pharmaceutical and food applications.
Collapse
Affiliation(s)
- Walaa I El-Sofany
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia; Medical and Diagnostic Research Center, University of Ha'il, Ha'il 55473, Saudi Arabia
| | - Tahani D Alanezi
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia; Medical and Diagnostic Research Center, University of Ha'il, Ha'il 55473, Saudi Arabia
| | - Salman Latif
- Department of Chemistry, College of Science, University of Ha'il, Ha'il 81451, Saudi Arabia; Medical and Diagnostic Research Center, University of Ha'il, Ha'il 55473, Saudi Arabia
| | - Ola Abdelhedi
- Institute of Biotechnology of Beja (ISBB), University of Jendouba, Beja, Tunisia
| | - Khaled Hamden
- Laboratory of Bioresources: Integrative Biology and Exploiting, Higher Institute of Biotechnology of Monastir, University of Monastir, Tunisia; Higher School of Health Sciences and Technology of Sfax, Sfax university, Tunisia.
| |
Collapse
|
27
|
Bhura N, Gupta J. Exploring phytochemical candidates against nephrolithiasis via similarity searching, network pharmacology, and docking studies. Biochem Biophys Res Commun 2025; 771:151975. [PMID: 40393161 DOI: 10.1016/j.bbrc.2025.151975] [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: 03/21/2025] [Revised: 04/19/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Nephrolithiasis (kidney stones) affects nearly 13 % of the global population, with a high recurrence rate posing significant challenges. Current remedies often fail to prevent recurrence, necessitating new therapeutic approaches. This study explores phytochemicals with potential protective effects against nephrolithiasis using similarity searching, network pharmacology, and molecular docking methods. Six anti-nephrolithiatic parent molecules were identified through a literature review. Structural similarity searches yielded 67 related compounds, which were screened using Lipinski' s rule of five, along with ADME (absorption, distribution, metabolism, excretion) parameters and toxicity profiles, and subsequently compared with standard drugs. The standard drug offered poor results; therefore, we aimed to overcome this limitation by searching for phytochemicals with favorable ADMET properties. Shortlisted compounds and nephrolithiasis-related targets were analyzed using Swiss Target Prediction and GeneCards. Hub genes were identified via Cytoscape' s Cytohubba plugin and subjected to Gene Ontology (GO) and KEGG pathway analysis. Molecular docking evaluated the binding of phytochemical ligands to disease proteins. Five phytochemicals namely 4- 4-Vinylguaiacol, anethole, isoeugenol, nadolol, and silibinin, were identified as potential candidates. Network pharmacology revealed 191 common targets between these compounds and nephrolithiasis. GO analysis highlighted key biological processes (response to organic substances, chemical stimuli), cellular components (nuclear lumen, nucleoplasm), and molecular functions (enzyme binding, catalytic activity). KEGG pathway analysis identified pathways in cancer, hepatitis B, and Kaposi sarcoma-associated herpesvirus infection as relevant to nephrolithiasis. Molecular docking demonstrated strong binding affinities between the shortlisted phytochemicals and disease targets. The multi-level screening and molecular docking findings highlight 4-Vinylguaiacol, anethole, isoeugenol, nadolol, and silibinin as promising therapeutic agents for nephrolithiasis. Among these, silibinin was tested for the in-vitro cytotoxicity assay based on the docking score. Importantly, in vitro validation using NRK-52E cells confirmed nephroprotective efficacy of silibinin. While calcium oxalate crystal exposure significantly reduced cell viability to below 50 %, co-treatment with silibinin improved cell survival to approximately 70 %, underscoring its potential to mitigate oxalate-induced cytotoxicity. Further experimental studies are required to validate these findings.
Collapse
Affiliation(s)
- Nancy Bhura
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
| |
Collapse
|
28
|
Chen J, Zhang H, Qiu M, Hu J, Lin L, Mai L, Huang G, Chen X, Li X, Qin X, Zhao H. Honokiol in the treatment of triple-negative breast cancer: a network pharmacology approach and experimental validation. Biochem Biophys Res Commun 2025; 771:152008. [PMID: 40398092 DOI: 10.1016/j.bbrc.2025.152008] [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: 01/08/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Triple-negative breast cancer (TNBC) is a rare and highly metastatic form of cancer. Honokiol (HNK), a biphenolic compound, has been utilized in TNBC treatment, though its specific targets remain unclear. This study aimed to elucidate the effects of HNK on TNBC by combining network pharmacology predictions and experimental validation to uncover its mechanisms. MDA-MB 231 and MDA-MB 468 cells were pre-treated with varying doses of HNK for 24 h. Cell viability, proliferation, and apoptosis were assessed using CCK8 and FACS assays, whereas a wound healing assay was used to evaluate cell migration. A tubule formation assay was used to assess blood vessel formation in HUVECs. Additionally, in vivo activity was confirmed using a zebrafish xenograft model. Network pharmacology and molecular docking predicted active ingredients, key targets, and potential mechanisms of HNK against TNBC. Results indicated that HNK induces apoptosis in MDA-MB 231 and MDA-MB 468 cells and inhibits their migration and proliferation. Furthermore, HNK suppressed blood vessel formation. Zebrafish xenograft experiments validated HNK's inhibitory effect on TNBC cells in vivo. Network pharmacology identified 36 potential HNK targets against TNBC, including HSP90AA1, AKT1, EGFR, ERBB2, HSP90AB1, PGR, MDM2, HDAC1, NR3C1, and MAPK14. Key signaling pathways such as PI3K-Akt, MAPK, Rap1, Ras, and FoxO were implicated in HNK's anti-TNBC mechanism. Molecular docking demonstrated spontaneous interactions between HNK and the targeted proteins. In conclusion, HNK may reduce angiogenesis by blocking the EGFR and HSP90AB1 pathways thereby decreasing proliferation and increasing apoptosis in TNBC cells.
Collapse
Affiliation(s)
- Jing Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China; Medical Research Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Haipeng Zhang
- Department of Blood Transfusion, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Min Qiu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Jiemei Hu
- Department of Gynecology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Lu Lin
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Liping Mai
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Guiping Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Xiuyun Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Xiaohong Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Xianyu Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, PR China.
| | - Haishan Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China.
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Anderson AM, O'Connor MS, Pipkin J, Malanga M, Sohajda T, Loftsson T, Szente L, García-Fandiño R, Piñeiro Á. A comprehensive nomenclature system for cyclodextrins. Carbohydr Polym 2025; 360:123600. [PMID: 40399013 DOI: 10.1016/j.carbpol.2025.123600] [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: 03/06/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 05/23/2025]
Abstract
Modified cyclodextrins (CDs) are cyclic oligosaccharides with many applications in drug delivery, catalysis, and as active pharmaceutical ingredients. In general, they exist as distributions of structurally diverse molecules rather than single-isomer compounds. Their performance depends on the number of glucopyranose units (GPUs), and the type, number, and position of chemical substitutions in their hydroxyl groups. Effectively targeting individual species within these distributions is essential for optimizing CDs for specific applications. Computational techniques can generate large datasets to AI-driven structural optimization, but the absence of a standardized nomenclature system for modified CDs presents a major barrier to progress in this direction. This lack of consensus limits effective communication, data sharing, automation, and collaboration. To address this, a clear and extensible nomenclature for modified CDs is proposed. In this framework, GPUs are treated like amino-acid residues, with unsubstituted GPUs as reference building-blocks and substituted ones considered as mutations. This approach precisely defines substitution types and patterns, resolves cyclic permutation ambiguities, and offers versatility for both simple and complex modifications, including chiral center alterations and covalently linked CD oligomers. By introducing this standardized nomenclature, we aim to enhance molecular design, improve reproducibility, and streamline both experimental and computational research in the CD field.
Collapse
Affiliation(s)
| | | | - James Pipkin
- Ligand Pharmaceuticals Incorporated, 3911 Sorrento Valley Boulevard, San Diego, CA 92121, USA
| | - Milo Malanga
- CarboHyde, Budapest, Berlini u. 47-49, 1045, Hungary
| | - Tamas Sohajda
- CarboHyde, Budapest, Berlini u. 47-49, 1045, Hungary
| | - Thorsteinn Loftsson
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavik, Iceland
| | - Lajos Szente
- CycloLab Cyclodextrin R&D Laboratory Ltd., Illatos u. 7., Budapest H-1097, Hungary
| | - Rebeca García-Fandiño
- Department of Organic Chemistry, Center for Research in Biological Chemistry and Molecular Materials, University of Santiago de Compostela, CIQUS, Spain.
| | - Ángel Piñeiro
- Department of Applied Physics, Faculty of Physics, University of Santiago de Compostela, Spain.
| |
Collapse
|
32
|
Kamel EM, Abdelrheem DA, Salah B, Lamsabhi AM. A mechanistic experimental and computational exploration of aldose reductase inhibition by coumarins from Ruta chalepensis. Biochem Biophys Res Commun 2025; 769:151946. [PMID: 40347621 DOI: 10.1016/j.bbrc.2025.151946] [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: 01/21/2025] [Revised: 05/04/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Aldose reductase (AR), a key enzyme in the polyol pathway, contributes to diabetic complications through oxidative stress and cellular damage. We investigated the inhibitory potential of coumarins from Ruta chalepensis as AR inhibitors, combining experimental and computational approaches. Among studied coumarins, aegelinol demonstrated the strongest AR inhibition with a mixed inhibition mode (IC50 = 3.75 ± 0.11 μM, Kᵢ = 3.04 ± 0.82 μM), approaching the potency of the positive control quercetin (IC50 = 2.22 ± 0.27 μM). Marmesinin and gelseminic acid also showed notable inhibition (IC50 = 5.32 ± 0.15 μM, 5.82 ± 0.61 μM, respectively), with noncompetitive binding mechanisms (Kᵢ = 4.86 ± 0.35 μM and 4.46 ± 0.52 μM), suggesting allosteric binding. Docking highlighted key interactions with critical AR residues, while molecular dynamics simulations provided insights into the stability and dynamics of AR-ligand complexes. Various MD parameters confirmed structural integrity, with the aegelinol-AR complex exhibiting the highest stability. MM/PBSA calculations supported these findings, showing the most favorable binding free energy for aegelinol, followed by marmesinin and gelseminic acid. Potential energy landscape analyses showed that aegelinol forms the most stable AR complex with the deepest energy basin. ADMET analysis confirmed aegelinol as a promising lead due to its high gastrointestinal absorption, BBB permeability, and balanced lipophilicity. Thus, Aegelinol, marmesinin, and gelseminic acid show strong potential as AR inhibitors, with aegelinol as the leading candidate for therapeutic development. These findings highlight Ruta chalepensis as a promising source of bioactive coumarins for targeted therapies against diabetic complications.
Collapse
Affiliation(s)
- Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Doaa A Abdelrheem
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Bashir Salah
- Department of Industrial Engineering, College of Engineering, King Saud University, P.O. Box 800, Riyadh, 11421, Saudi Arabia
| | - Al Mokhtar Lamsabhi
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Campus de Excelencia UAM-CSIC Cantoblanco, 28049, Madrid, Spain; Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| |
Collapse
|
33
|
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
|
34
|
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
|
35
|
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
|
36
|
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
|
37
|
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
|
38
|
Zou DL, Liao WZ, Yang HG, Lin B, Xu HR, Hua HM, Li DH. Discovery of polycyclic polyprenylated acylphloroglucinols with antitumor activities from Garcinia pedunculata Roxb. fruits based on molecular networking. Bioorg Chem 2025; 161:108513. [PMID: 40311239 DOI: 10.1016/j.bioorg.2025.108513] [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: 02/28/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Garpedvinin A (1), a novel polycyclic polyprenylated acylphloroglucinol (PPAP) with a bicyclo[4,2,1]nonane core, 13 previously undescribed PPAPs, garpedvinins B-N (2-14) and 6 known analogs (15-20) were isolated from Garcinia pedunculata by various chromatographic methods combined with Global Natural Products Social Molecular Networking. The structures were identified by the analyses of spectral characteristics, computational chemistry calculations and single-crystal X-ray diffraction. A plausible biosynthetic pathway for garpedvinin A was suggested based on the isolated precursor, cambogin. Compounds 2-6, 8, 11, 13, 15-18 and 20 displayed cytotoxic effects on three cancer cell lines, HepG2, A549 and MCF-7. 6 showed the strong inhibitory effect on the proliferation of HepG2 cells in vitro, inducing cell apoptosis in a concentration-dependent manner and blocking the cell cycle at the S phase. Furthermore, 6 affected the expression of apoptosis-related proteins Bax, Bcl2 and pro-Caspase-3.
Collapse
Affiliation(s)
- De-Li Zou
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Wen-Zhuo Liao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Han-Gao Yang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Bin Lin
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Hua-Rong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Hui-Ming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| | - Da-Hong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| |
Collapse
|
39
|
Innocenti N, Tähtinen P, Spagnolli G, Perrucci C, Bellini M, Parolin E, Bonaldo V, Biasini E, Mancini I. Enantiomers of the prion protein degrader SM875: Production and configurational assignment, in silico analysis and in vitro evaluation. Bioorg Chem 2025; 161:108489. [PMID: 40286470 DOI: 10.1016/j.bioorg.2025.108489] [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: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Prion diseases are rare and fatal neurodegenerative conditions affecting humans and animals caused by the misfolding of the cellular prion protein (PrP). Recently, the molecule 1-(4-bromophenyl)-1,4,5,7-tetrahydro-4-(4-hydroxy-3)-6H-pyrazolo[3,4-b]pyridin-6-one, named SM875, was identified as a promising PrP degrader through a computational approach targeting folding intermediates. The racemic mixture of SM875 showed biological activity but also exhibited variable toxicity. In this study, we optimized the synthesis of racemic SM875 and achieved high-purity enantiomeric separation via chiral HPLC. The docking calculation data of each enantiomer with a simplified model of Chiralpak IA®, used as the chiral stationary phase, were in line with their relative elution time. The electronic circular dichroic (ECD) spectra acquired for each isomer compared with the TD-DFT calculated spectrum for (R)-SM875 allowed the assignment of their absolute configuration. The biological evaluation revealed that the (R)-enantiomer solely reduces PrP levels, with associated toxicity, while the (S)-enantiomer is inactive. Molecular dynamics simulations corroborate the (R)-enantiomer's stronger interaction with PrP. These findings provide a foundation for therapeutic development targeting prion diseases.
Collapse
Affiliation(s)
- Nicole Innocenti
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology, University of Trento, via Sommarive 9, I-38123 Povo, Trento, Italy; Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, via Sommarive 14, I-38123, Povo, Trento, Italy
| | - Petri Tähtinen
- Department of Chemistry, University of Turku, Henrikinkatu 2, 20014 Turku, Finland
| | - Giovanni Spagnolli
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology, University of Trento, via Sommarive 9, I-38123 Povo, Trento, Italy
| | - Cecilia Perrucci
- Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, via Sommarive 14, I-38123, Povo, Trento, Italy
| | - Martina Bellini
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology, University of Trento, via Sommarive 9, I-38123 Povo, Trento, Italy; Department of Chemistry, University of Turku, Henrikinkatu 2, 20014 Turku, Finland
| | - Eleonora Parolin
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology, University of Trento, via Sommarive 9, I-38123 Povo, Trento, Italy; Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, via Sommarive 14, I-38123, Povo, Trento, Italy
| | - Valerio Bonaldo
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology, University of Trento, via Sommarive 9, I-38123 Povo, Trento, Italy
| | - Emiliano Biasini
- Dulbecco Telethon Laboratory of Prions and Amyloids, Department of Cellular, Computational and Integrative Biology, University of Trento, via Sommarive 9, I-38123 Povo, Trento, Italy.
| | - Ines Mancini
- Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, via Sommarive 14, I-38123, Povo, Trento, Italy.
| |
Collapse
|
40
|
Tillmanns J, Battisti V, Kicuntod J, Hahn F, Obergfäll D, Geiger P, Wagner S, Buschmann H, Lesch B, Lischka P, Sticht H, Langer T, Marschall M. The conserved core nuclear egress complex (NEC) as an antiherpesviral drug target: Pharmacophore-based identification of NEC-specific inhibitors. Antiviral Res 2025; 239:106168. [PMID: 40250630 DOI: 10.1016/j.antiviral.2025.106168] [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: 12/05/2024] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The nucleocytoplasmic capsid egress of herpesviruses is a uniquely regulated process. As well-established for the human cytomegalovirus (HCMV) core nuclear egress complex (NEC), the pUL50-pUL53 NEC heterodimer oligomerizes and builds hexameric lattices for the regulated nucleocytoplasmic release of viral capsids. Recently, we and others validated the NEC as a novel target for antiviral strategies. So far, the experimental targeting approaches included the development of NEC-directed small molecules, cell-penetrating peptides, NEC-specific mutagenesis, and the expression of NEC-interfering protein constructs. Our current postulate states that a small molecule-mediated interference with the assembly of the core NEC prevents NEC-dependent egress regulation and thereby strictly limits viral replication. Here, we present an experimental proof of this antiviral strategy, and the data provide evidence for the following points: (i) pharmacophore-based approaches demonstrated to be successful in the identification of NEC-specific inhibitory small molecules, (ii) already a low number of 36 analyzed small molecules yielded eight experimental hits with micromolar to submicromolar antiviral activity, (iii) their antiviral potency was asserted to the predicted NEC-interfering mode-of-action, (iv) two identified hit compounds presented a broad antiherpesviral activity, and (v) a further pharmacophore-assisted refinement of NEC-directed molecules may lead to the development of highly effective and even broadly acting antivirals. Combined, we strengthen the recently postulated potential of the NEC as a next-generation antiherpesviral drug target by identifying broadly active NEC inhibitors via a pharmacophore-based approach.
Collapse
Affiliation(s)
- Julia Tillmanns
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Verena Battisti
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Vienna, Vienna, Austria.
| | - Jintawee Kicuntod
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Debora Obergfäll
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Pia Geiger
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Sabrina Wagner
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | | | | | - Peter Lischka
- AiCuris Anti-Infective Cures AG, Wuppertal, Germany.
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, FAU, Erlangen, Germany.
| | - Thierry Langer
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Vienna, Vienna, Austria.
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
41
|
Mosalam EM, AboShabaan HS, Mahfouz MM, Sallam AS, Elhosary E, Allam A, Metwally EM, Shaldam MA, Ghoneim MES. Protective effect of empagliflozin against paracetamol-induced acute kidney injury through modulation of AMPK/SIRT1/PGC-1α pathway in experimental mice. Toxicol Appl Pharmacol 2025; 500:117382. [PMID: 40349789 DOI: 10.1016/j.taap.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: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Emerging evidences about paracetamol-induced kidney injury in clinical settings are concerning, especially when administered at high doses. Empagliflozin, an oral SGLT2 inhibitor, employed in the management of diabetes mellitus, exhibits antioxidant, anti-inflammatory, and anti-apoptotic attributes. Thus, the objective of this study is to investigate whether empagliflozin may alleviate paracetamol-triggered nephrotoxicity and unravel the mechanistic insights responsible for its protective impact. In this regard, male mice were assigned to four groups: normal, paracetamol, empagliflozin 10, and empagliflozin 20. Kidney function tests, histopathological examination, immunohistochemistry, oxidative stress biomarkers, inflammatory cytokines, and other molecular targets were detected. Our results showed that paracetamol administration impaired kidney functions along with causing aberrations in renal histoarchitecture. Additionally, paracetamol triggered oxidative stress, inflammation, and apoptosis via hindering the AMPK/SIRT1/PGC-1α cascade and Nrf2/HO-1 while activating the NF-κB hub. Nevertheless, pretreatment with empagliflozin markedly enhanced the kidney function tests and mitigated histopathological alterations caused by paracetamol. Additionally, empagliflozin suppressed the oxidative stress as confirmed by an upregulation of Nrf2, which subsequently increased HO-1, SOD, and GSH, while reducing the MDA level. Moreover, it inhibited the NF-κB-mediated inflammatory process by dampening NF-κB, IL-1β, and TNF-α expressions as well as lowering Bax expression-induced apoptosis. The observed safeguards effects were facilitated via boosting AMPK/SIRT1/PGC-1α signaling trajectory. Collectively, our study verified the enduring reno-protective potential of empagliflozin, particularly at high dose, in the context of paracetamol-induced renal injury by instigating the AMPK/SIRT1/PGC-1α hinge.
Collapse
Affiliation(s)
- Esraa M Mosalam
- Biochemistry Department, Faculty of Pharmacy, Menoufia University, 32511 Shebin EL-Kom, Menoufia, Egypt; Department of Pharm D, Faculty of Pharmacy, Jadara University, Irbid 21110, Jordan.
| | - Hind S AboShabaan
- Clinical Pathology Department, National Liver Institute Hospital, Menoufia University, Shebin El-Kom, Menoufia, Egypt.
| | - Marwa M Mahfouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia University, 32511 Shebin El-Kom, Menoufia, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia National University, Birket El-Sab, Menoufia, Egypt
| | - Amany Said Sallam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia University, 32511 Shebin El-Kom, Menoufia, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia National University, Birket El-Sab, Menoufia, Egypt.
| | - Enas Elhosary
- Department of Pathology, Faculty of Medicine, Helwan University, Cairo, Egypt.
| | - Albatoul Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), AL-Azhar University, Cairo, Egypt
| | - Ebtehal M Metwally
- Medical Physiology Department, Faculty of Medicine, Menoufia University, 32511 Shebin El-Kom, Menofia, Egypt.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt; Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy.
| | - Mai El-Sayed Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), 32897 Sadat City, Egypt.
| |
Collapse
|
42
|
El-Shafey HW, Al-Sanea MM, Elnagar MR, Gendy AM, Serag MI, Almatary AM, Khalaf MA, Abdulla MH, Alhassan NS, Mohammed MAV, Eldehna WM, Hamdi A. Design and synthesis of novel 2-S-alkylated Quinazolinones as dual BRAF V600E and EGFR inhibitors in melanoma: Mechanistic insights from apoptosis and cell cycle modulation. Bioorg Chem 2025; 161:108526. [PMID: 40311244 DOI: 10.1016/j.bioorg.2025.108526] [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: 03/12/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Melanoma, an aggressive and highly metastatic form of skin cancer, remains challenging to treat due to its resistance to conventional therapies and frequent mutations in the BRAF signaling pathway. In this study, we report the design and synthesis of a novel series of thirteen quinazolinone derivatives, featuring a phenyl thiazole moiety linked via a triazole acetamide spacer. These compounds were developed as potential dual inhibitors of BRAFV600E and EGFR, which should offer a promising therapeutic strategy for melanoma treatment. The antiproliferative activity of these compounds was evaluated against the NCI-60 cell line panel, with six compounds advancing to a five-dose screening. Three compounds, 7k, 7l, and 7m, exhibited broad-spectrum anticancer activity, with mean growth inhibition (GI%) exceeding 100 %. Compound 7l demonstrated exceptional efficacy against melanoma subpanels (GI% = 152 %) and potent dual kinase inhibition, with IC50 values of 0.048 μM against B-RAFV600E and 0.037 μM against EGFR. In vitro studies of compound 7l revealed significant cytotoxicity against MALME-3 M (IC50 = 3.16 μM) and LOX-IMVI (IC50 = 2.50 μM) melanoma cell lines, with minimal toxicity towards normal Vero cells. Cell cycle analysis showed G1-phase arrest and disrupted DNA synthesis in melanoma cells, while apoptosis assays demonstrated a dramatic increase in early apoptotic cells from 7.28 % to 40.69 %. Compound 7l modulated key apoptotic markers, increasing the BAX/Bcl-2 ratio by 14.42-fold and elevating caspase 3 and 9 levels, indicating its potential to overcome drug resistance and enhance therapeutic efficacy in melanoma treatment.
Collapse
Affiliation(s)
- Hamed W El-Shafey
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Campus, Queensland, 4222, Australia
| | - Mohammad M Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72388, Saudi Arabia.
| | - Mohamed R Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11823, Egypt; Department of Pharmacology, College of Pharmacy, The Islamic University, Najaf 54001, Iraq
| | - Abdallah M Gendy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| | - Marwa I Serag
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Aya M Almatary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Mohamed A Khalaf
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Maha-Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Noura S Alhassan
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mansoor-Ali Vaali Mohammed
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria; Canal El Mahmoudia St., Alexandria 21648, Egypt
| | - Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
43
|
Guo J, Zhou LS, Lv X, Wang JF, Xu TZ, Wang ZH, Guo SJ, Guo A, Wang JX, Gao XM, Sun ZT, Wu QY. Mechanisms of LouDan LiFei granule attenuate inflammatory injury in AECOPD:A network pharmacology and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156719. [PMID: 40397998 DOI: 10.1016/j.phymed.2025.156719] [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: 09/26/2024] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Acute exacerbation of chronic obstructive pulmonary disease (AECOPD) significantly contributes to the high mortality rate associated with chronic obstructive pulmonary disease (COPD). It leads to a decline in health status, increased rates of readmission, and accelerated disease progression. LouDan LiFei granule (LDLF), a Traditional Chinese Medicine (TCM) granule, has demonstrated efficacy in treating AECOPD patients. However, its underlying regulatory mechanisms remain to be fully elucidated. PURPOSE To investigate the pharmacodynamic components and regulatory mechanisms of LDLF in the treatment of AECOPD, and to provide a scientific basis for the further clinical application of subsequent drugs. METHODS This study focused on identifying the pharmacodynamic components and regulatory mechanisms of LDLF. In vivo experiments, we established a rat model of AECOPD and systematically evaluated the therapeutic effect by monitoring the body weight, lung function and pathological changes in lung tissue. UPLC-MS/MS was employed to detect the components of LDLF medicated serum. Network pharmacology and molecular docking were utilized to determine the therapeutic targets and pathways of LDLF in AECOPD treatment. Finally, we further verified the research results by establishing AECOPD moded in rat and BEAS-2B. RESULTS Our study revealed that LDLF significantly reduced inflammation and pathological damage in lung tissue of AECOPD model rats and improved pulmonary function. UPLC-MS/MS analysis identified 1502 compounds in LDLF medicated serum, primarily comprising flavonoids, terpenoids, alkaloids, ketones, and aldehydes acids. Network pharmacology results suggested that LDLF may treat AECOPD by modulating Th17 cell differentiation and signaling pathways related to IL-17, T cell receptor, and NOD-like receptor. Molecular docking confirmed stable interactions between core compounds and their targets. In vivo validation showed that LDLF redused the proportion of Th17 cells and increased Treg cells proportion in peripheral blood of rat model. Protein expression levels of FOXP3 were elevated, and RORγ and STAT3 were reduced in lung tissue. Inflammatory markers (IL-6, IL-1β, IL-8, IL-17, TNF-α, IL-10, and TGF-β1) in serum and balf were improved after LDLF treatment. Additionally, LDLF significantly inhibited the NLRP3 inflammasome pathway at protein and mRNA levels in lung. In vitro experiments demonstrated that LDLF enhanced the BEAS-2B cells viability and inhibited inflammatory markers IL-6 and TNF-α as well as key targets of NLRP3, STAT3, CASP3, and AKT1. CONCLUSIONS This research highlights the potential for LDLF in the treatment of AECOPD and enhances our understanding of its pathogenic and therapeutic mechanisms.
Collapse
Affiliation(s)
- Jing Guo
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), YouDian Road, 54th, Shangcheng District, Hangzhou 310006, ZheJiang Province, China
| | - Lin-Shui Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), YouDian Road, 54th, Shangcheng District, Hangzhou 310006, ZheJiang Province, China
| | - Xin Lv
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), YouDian Road, 54th, Shangcheng District, Hangzhou 310006, ZheJiang Province, China
| | - Jian-Feng Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), YouDian Road, 54th, Shangcheng District, Hangzhou 310006, ZheJiang Province, China
| | - Ting-Zhen Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), YouDian Road, 54th, Shangcheng District, Hangzhou 310006, ZheJiang Province, China
| | - Zhong-Hai Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), YouDian Road, 54th, Shangcheng District, Hangzhou 310006, ZheJiang Province, China
| | - Si-Jia Guo
- The Second Affiliated Hospital of Tianjin University of Chinese Medicine, TianJin, 300000, China
| | - An Guo
- Tianjin University of Traditional Chinese Medicine. TianJin, 301600, China
| | - Jing-Xia Wang
- Beijing University of Traditional Chinese Medicine. Beijing, 100029, China
| | - Xue-Min Gao
- Beijing University of Traditional Chinese Medicine. Beijing, 100029, China
| | - Zeng-Tao Sun
- Tianjin University of Traditional Chinese Medicine. TianJin, 301600, China.
| | - Qing-Yuan Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), YouDian Road, 54th, Shangcheng District, Hangzhou 310006, ZheJiang Province, China.
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Li Y, Gao S, Zhang X, Cao Z, Guo Y, Zhao R, Liu Y, Li X, Lin H, Qin Q, Yi B, Zhao G. Joint technique "parallel peptide synthesis & de novo sequencing" development for the structure verification and high-throughput activity screening of biological peptides from sea cucumber (Stichopus japonicus) intestinal hydrolysate. Food Res Int 2025; 212:116475. [PMID: 40382048 DOI: 10.1016/j.foodres.2025.116475] [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: 12/12/2024] [Revised: 03/19/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025]
Abstract
This study introduces a novel joint technique combining "parallel peptide synthesis" and "de novo sequencing", which facilitates the high-throughput screening and structure validation of biological peptides derived from food and traditional Chinese medicine (TCM). Sea cucumber (Stichopus japonicus) was used as a model organism, undergoing simulated gastrointestinal digestion followed by de novo sequencing to predict potential peptides. Subsequently, cost-effective filter pipette tips were innovatively employed as parallel reaction vessels, enabling efficient peptide synthesis through the microfluidic flow of amino acid solutions over the loaded resin. After high-throughput biological activity screening, peptide YPGQLT was identified as the most potent antioxidant and acetylcholinesterase (AChE) inhibitor. It was then subjected to a molecular docking study to further explore its potential ligand-receptor interactions. This synergistic effect highlights peptide YPGQLT as a promising candidate for Alzheimer's disease (AD) treatment, thereby enhancing the potential biomedical application of sea cucumber. Notably, de novo predicted sequences can be further verified by comparing their characteristics, such as retention time and MS/MS spectrum, with those from the standard reference synthesized using this parallel synthesis protocol.
Collapse
Affiliation(s)
- Yimeng Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shengfang Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaohui Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhuo Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Youyou Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Runkun Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yonggang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinxin Li
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongying Lin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qi Qin
- Department of Neurology & Innovation center for neurological disorders, Xuanwu Hospital, Capital Medical University, Beijing 100029, China
| | - Bingqing Yi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Guodong Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Engineering Research Center for Pharmaceutics of Chinese Materia Medica and New Drug Development, Ministry of Education, Beijing 100029, China.
| |
Collapse
|
47
|
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
|
48
|
Varadharajan V, Balu AK, Sinclair BJ, Perinbarajan GK, Jenifer A D, Ganesan Sudha H, Ramaswamy A, Venkidasamy B, Thiruvengadam M. Comprehensive analysis of Syzygium cumini L. pomace extract as an α-amylase inhibitor: In vitro inhibition, kinetics, and computational studies. Bioorg Chem 2025; 161:108498. [PMID: 40339502 DOI: 10.1016/j.bioorg.2025.108498] [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: 12/05/2024] [Revised: 04/12/2025] [Accepted: 04/19/2025] [Indexed: 05/10/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a widespread metabolic disorder characterized by impaired regulation of blood glucose levels. Jamun (Syzygium cumini L.) fruits and seeds have been traditionally used in Ayurveda to manage diabetes. While fruit and seed extracts have been extensively studied for their anti-α-amylase properties, pomace, a byproduct of juice extraction, remains under explored. This study investigated the α-amylase inhibitory potential of jamun pomace (JP) extract by using in vitro and in silico methods. Enzyme inhibition assays revealed an half-maximal inhibitory concentration (IC₅₀) value of 85.68 ± 5.22 μg/mL for the JP extract, comparable to acarbose (64.28 ± 7.15 μg/mL). The extract exhibited mixed-mode inhibition, whereas acarbose showed competitive mode inhibition. At 10 μg/mL, the Vmax of JP extract was half that of acarbose, demonstrating significant inhibition. GC-MS analysis identified 11 volatile compounds (R1-R11) in the JP extract. Density Functional Theory (DFT) and ADMET analyses confirmed the chemical reactivity of the volatiles, drug-like properties, and low toxicity. Molecular docking revealed a high binding score for R11 (-8.0 kcal/mol), similar to acarbose (-8.2 kcal/mol). Molecular dynamics simulations further demonstrated the stability of α-amylase complexes with R11, R3, and R8, with R11 showing the lowest binding energy (-28.75 ± 6.25 kcal/mol). These findings suggest that R11 and JP extracts hold promise as anti-diabetic agents. Utilizing JP extract as a nutraceutical offers the dual benefit of diabetes management and sustainable waste valorization in jamun juice production.
Collapse
Affiliation(s)
| | - Ashwath Kumar Balu
- Department of Biotechnology, PSG College of Technology, Peelamedu, Coimbatore, India; Department of Biotechnology, Indian Institute of Technology, Hyderabad, India
| | - Bruce Joshua Sinclair
- Departmet of Electronics and Communication Engineering, PSG College of Technology, Coimbatore, India
| | - Gopi Krishna Perinbarajan
- Departmet of Electronics and Communication Engineering, PSG College of Technology, Coimbatore, India
| | - Dharshini Jenifer A
- Department of Chemical Engineering, National Institute of Technology, Surathkal, Karnataka, India
| | | | - Arulvel Ramaswamy
- Department of Biotechnology, K S Rangasamy College of Technology, Tiruchengode, Namakkal District, Tamil Nadu 637 215, India
| | - Baskar Venkidasamy
- Centre for Biosciences and Biotechnology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India.
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Science, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
49
|
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
|
50
|
Zhai J, Yan H, Liu M, Jiang C, Jin M, Xie B, Ma C, Cong B, Wen D. Decoding gelsenicine-induced neurotoxicity in mice via metabolomics and network toxicology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156753. [PMID: 40250031 DOI: 10.1016/j.phymed.2025.156753] [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: 02/13/2025] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Gelsenicine, the most toxic constituent of Gelsemium elegans Benth., is known for its diverse pharmacological activities alongside potent neurotoxicity, frequently leading to poisoning incidents following mistaken ingestion. However, its molecular mechanisms remain largely unexplored. PURPOSE This study aimed to elucidate the key mechanistic network underlying gelsenicine-induced neurotoxicity by employing a comprehensive strategy that integrated metabolomics, network toxicology, molecular docking, and experimental validation. METHODS Acute oral toxicity tests were conducted in C57BL/6J mice to assess toxic symptoms, determine the median lethal dose (LD50), and evaluate histopathological changes. Untargeted metabolomics was performed to identify differential metabolites and associated pathways in serum, hippocampus (HIP), and medulla oblongata (MO). Integration of network toxicology pinpointed core targets and pathways, which were further validated through molecular docking and RT-qPCR. A core "compound-target-metabolite-pathway" network involved in gelsenicine-induced neurotoxicity was established. RESULTS Gelsenicine exhibited an oral LD50 of approximately 1.82 mg/kg and induced neurotoxic damage in the HIP and MO. Two untargeted metabolomic approaches detected a broad range of metabolites, revealing that gelsenicine markedly altered the metabolic profiles of serum, HIP, and MO. Network toxicology analysis identified 187 key targets associated with gelsenicine neurotoxicity. Integrated analyses with the predicted targets of differential metabolites indicated that gelsenicine primarily interferes with the energy metabolism network centered on the malate-aspartate shuttle (MAS), affecting pathways such as carbon metabolism, amino acid metabolism, TCA cycle, and PPAR signaling pathway. Malate, glutamate, and aspartate were identified as core metabolites and potential biomarkers of gelsenicine poisoning. RT-qPCR validation revealed that gelsenicine interfered with the expression of core targets, including GLUD1, MDH, GOT and ME, all of which exhibited good binding energy with gelsenicine. CONCLUSION This study unveiled a novel mechanistic insight into gelsenicine-induced neurotoxicity, demonstrating its capacity to perturb multiple energy metabolism pathways associated with MAS. These findings could enhance the theoretical understanding of gelsenicine's neurotoxic effects and highlight potential applications in clinical diagnosis and forensic identification.
Collapse
Affiliation(s)
- Jinxiao Zhai
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China; College of Forensic Medicine, Jining Medical University, Jining 272067, China
| | - Hui Yan
- Department of Forensic Toxicology, Shanghai Key Laboratory of Forensic Medicine, Academy of Forensic Science, 1347 West Guangfu Road, Shanghai 200063, China
| | - Minghao Liu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Chen Jiang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Mingyang Jin
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China.
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, China.
| |
Collapse
|