1
|
Mayyas A, Al-Samydai A, Al-Karablieh N, Zalloum WA, Al-Tawalbeh D, Al-Mamoori F, Amr RA, Al Nsairat H, Carradori S, Al-Halaseh LK, Aburjai T. A phytotherapeutic approach to hinder the resistance against clindamycin by MRSA: in vitro and in silico studies. Future Sci OA 2025; 11:2458438. [PMID: 39895160 PMCID: PMC11792796 DOI: 10.1080/20565623.2025.2458438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
AIMS This study investigates the potential effects of essential oils (EOs) in enhancing the efficacy of clindamycin against Methicillin-resistant Staphylococcus aureus (MRSA) using in vitro and computer simulations. The research seeks to identify essential oils that exhibit synergistic activity with clindamycin and determine their potential key active components. MATERIALS AND METHODS Essential oils commonly used in traditional medicine were tested for their antimicrobial activity against MRSA. The minimum inhibitory concentration (MIC) was determined using in vitro microdilution assays. A synergistic test with clindamycin was performed, and molecular docking studies evaluated the interaction between a key compound (trans-cinnamaldehyde) and MRSA protein. RESULTS EOs from Cinnamomum verum, Rosmarinus officinalis, Salvia officinalis, and Thymus vulgaris demonstrated significant inhibitory and synergistic activities against MRSA, standard strain, and human clinical isolates. Gas Chromatography/Mass Spectroscopy identified trans-cinnamaldehyde, eucalyptol, and thymol as prominent antibacterial compounds. Molecular docking studies confirmed trans-cinnamaldehyde's strong binding to MRSA's AgrA protein, elucidating its enhanced efficacy. CONCLUSION The study underscores the potential of plant-based therapies to augment the effectiveness of conventional antibiotics like clindamycin in combating MRSA and addressing antibiotic resistance by integrating traditional plant remedies with modern medical approaches.
Collapse
Affiliation(s)
- Amal Mayyas
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Ali Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Nehaya Al-Karablieh
- Department of Plant Protection, School of Agriculture, The University of Jordan, Amman, Jordan
- Hamdi Mango Centre for Scientific Research, The University of Jordan, Amman, Jordan
| | - Waleed A Zalloum
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Deniz Al-Tawalbeh
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Farah Al-Mamoori
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Zarqa University, Zarqa, Jordan
| | - Rula A. Amr
- Department of Pharmacy, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Hamdi Al Nsairat
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Simone Carradori
- Department of Pharmacy “G. d’Annunzio”, University of Chieti-Pescara, Chieti, SC, Italy
| | - Lidia Kamal Al-Halaseh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mutah University, Al-Karak, Jordan
| | - Talal Aburjai
- Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
2
|
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
|
3
|
Shao C, Xia W, Liu Y. Bioinformatic Analysis and Molecular Docking Identify Isorhamnetin Is a Candidate Compound in the Treatment of Pulmonary Artery Hypertension. Anatol J Cardiol 2025; 29:52-65. [PMID: 39605239 PMCID: PMC11793806 DOI: 10.14744/anatoljcardiol.2024.4723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The current study aims to identify the key pathways and potential therapeutic targets for pulmonary arterial hypertension (PAH) and to further evaluate the anti-PAH effects of isorhamnetin. METHODS The dataset of gene expression profiling for PAH (GSE113439) was downloaded from the gene expression omnibus (GEO) database. Isorhamnetin target genes were extracted from the comparative toxicogenomics database (CTD). Various bioinformatics methods were employed to identify the core pathways associated with PAH and potential intervention targets. Molecular docking was conducted between the interacting target and the candidate compound, isorhamnetin. RESULTS One thousand nine hundred sixty-two upregulated genes and 642 downregulated genes were identified. Molecular complex detection analyses revealed that the significant biological processes associated with upregulated genes included DNA damage response, mitotic cell cycle, and chromosome organization. In contrast, the signifi ant biological processes related to downregulated genes encompassed cellular response to growth factor stimulus, response to growth factor, and blood vessel development. Immune infilt ation analysis indicated that PAH is associated with signifi ant changes in the distribution of immune cells and differential expression of immune checkpoints. Furthermore, 58 isorhamnetin targets were extracted from the CTD, and we identified 1 interacting gene, NFE2L2, among the differentially expressed genes (DEGs), DEGs related to ferroptosis, and isorhamnetin targets. Isorhamnetin demonstrated strong affinities with vascular endothelial growth factor (VEGF) receptors and transcription factors (ATM and ZNF24) associated with VEGFs, as well as the ferroptosis protein NFE2L2. CONCLUSIONS Pulmonary arterial hypertension is characterized by a series of abnormalities in downstream molecular signaling pathways, including DNA damage, immune dysregulation, VEGF signaling deficienc , and the ferroptosis process. These may represent the core pathophysiological mechanisms of PAH. Ferroptosis-related genes, such as NFE2L2 and TF (ATM, ZNF24) associated with VEGFs, are potential therapeutic targets that contribute to the mechanisms mentioned above. Isorhamnetin is a promising candidate compound for the treatment of PAH.
Collapse
Affiliation(s)
- Chen Shao
- Department of Nursing Science, The Second People’s Hospital of Lianyungang, Jiangsu, China
| | - Wei Xia
- Department of Pharmacology, The Second People’s Hospital of Lianyungang, Jiangsu, China
| | - Yang Liu
- Department of Internal and Pediatrics, School of Clinical Medicine, Qilu Medical University, Zibo, Shandong, China
| |
Collapse
|
4
|
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
|
5
|
Khan T. An insight into in silico strategies used for exploration of medicinal utility and toxicology of nanomaterials. Comput Biol Chem 2025; 117:108435. [PMID: 40158237 DOI: 10.1016/j.compbiolchem.2025.108435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Nanomaterials (NMs) and the exploration of their comprehensive uses is an emerging research area of interest. They have improved physicochemical and biological properties and diverse functionality owing to their unique shape and size and therefore they are being explored for their enormous uses, particularly as medicinal and therapeutic agents. Nanoparticles (NPs) including metal and metal oxide-based NPs have received substantial consideration because of their biological applications. Computer-aided drug design (CADD) involving different strategies like homology modelling, molecular docking, virtual screening (VS), quantitative structure-activity relationship (QSAR) etc. and virtual screening hold significant importance in CADD used for lead identification and target identification. Despite holding importance, there are very few computational studies undertaken so far to explore their binding to the target proteins and macromolecules. Although the structural properties of nanomaterials are well documented, it is worthwhile to know how they interact with the target proteins making it a pragmatic issue for comprehension. This review discusses some important computational strategies like molecular docking and simulation, Nano-QSAR, quantum chemical calculations based on Density functional Theory (DFT) and computational nanotoxicology. Nano-QSAR modelling, based on semiempirical calculations and computational simulation can be useful for biomedical applications, whereas the DFT calculations make it possible to know about the behaviour of the material by calculations based on quantum mechanics, without the requirement of higher-order material properties. Other than the beneficial interactions, it is also important to know the hazardous consequences of engineered nanostructures and NPs can penetrate more deeply into the human body, and computational nanotoxicology has emerged as a potential strategy to predict the delirious effects of NMs. Although computational tools are helpful, yet more studies like in vitro assays are still required to get the complete picture, which is essential in the development of potent and safe drug entities.
Collapse
Affiliation(s)
- Tahmeena Khan
- Department of Chemistry, Integral University, Lucknow, U.P 226026, India.
| |
Collapse
|
6
|
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
|
7
|
Clayton J, Shi L, Robertson MJ, Skiniotis G, Michaelides M, Stavitskaya L, Shen J. A putative binding model of nitazene derivatives at the μ-opioid receptor. Neuropharmacology 2025; 273:110437. [PMID: 40185362 DOI: 10.1016/j.neuropharm.2025.110437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/09/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
Nitazenes are a class of novel synthetic opioids with exceptionally high potency. Currently, an experimental structure of μOR-opioid receptor (μOR) in complex with a nitazene is lacking. Here we used a suite of computational tools, including consensus docking, conventional molecular dynamics (MD) and metadynamics simulations, to investigate the μOR binding modes of nitro-containing meto-, eto-, proto-, buto-, and isotonitazenes and nitro-less analogs, metodes-, etodes-, and protodesnitazenes. Docking generated three binding modes, whereby the nitro-substituted or unsubstituted benzimidazole group extends into SP1 (subpocket 1 between transmembrane helix or TM 2 and 3), SP2 (subpocket 2 between TM1, TM2, and TM7) or SP3 (subpocket 3 between TM5 and TM6). Simulations suggest that etonitazene and likely also other nitazenes favor the SP2-binding mode. Comparison to the experimental structures of μOR in complex with BU72, fentanyl, and mitragynine pseudoindoxyl (MP) allows us to propose a putative model for μOR-ligand recognition in which ligand can access hydrophobic SP1 or hydrophilic SP2, mediated by the conformational change of Gln1242.60. Interestingly, in addition to water-mediated hydrogen bonds, the nitro group in nitazenes forms a π-hole interaction with the conserved Tyr751.39. Our computational analysis provides new insights into the mechanism of μOR-opioid recognition, paving the way for investigations of the structure-activity relationships of nitazenes.
Collapse
Affiliation(s)
- Joseph Clayton
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, 20993, USA; Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, Neuroimaging Research Branch, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Lidiya Stavitskaya
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, 20993, USA.
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA.
| |
Collapse
|
8
|
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
|
9
|
Yildiz MM, Ates I, Gurbuz HN, Unal MA, Nazir H, Uzunoglu A, Ozkan SA, Topal BD. Exploring efavirenz-DNA interactions: A multidisciplinary approach through electrochemical, toxicological, and in silico investigations. J Pharm Biomed Anal 2025; 259:116763. [PMID: 40024028 DOI: 10.1016/j.jpba.2025.116763] [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: 10/14/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Recently, there has been a growing approach that approved drugs have been tested for additional purposes. Efavirenz is a non-nucleoside reverse transcriptase inhibitor used to treat human immunodeficiency virus infection. In addition, it has selective cytotoxic effects against cancer cells. This study constructed an electrochemical dsDNA nanobiosensor to monitor Efavirenz-dsDNA interaction based on the amine-functionalized multi-walled carbon nanotubes. The experimental conditions of the nanobiosensor, such as dropping the volume of nanomaterial suspension, activation of the nanosensor, and dsDNA concentration, were optimized. The peak currents of dsDNA bases were enhanced, and the peak potentials of Efavirenz have shifted to the less positive potential thanks to the modified sensor with amine-functionalized multi-walled carbon nanotubes. The interaction mechanism was also evaluated in incubated solutions. Docking calculations showed that Efavirenz is active in the large cleft regions of DNA that suggest minor groove binding. The effect of efavirenz on the expression profile of particular stress and possible DNA genotoxicity was studied via examining gene polymorphisms in hepatic cells. These findings align with previously released research that shows Efavirenz-treated hepatic cells to have altered mitochondrial function and elevated ROS levels.
Collapse
Affiliation(s)
- Manolya Mujgan Yildiz
- Ankara University, Graduate School of Health Science, Dışkapı, Ankara 06110, Turkiye; Lokman Hekim University, Faculty of Pharmacy, Department of Analytical Chemistry, Çankaya, Ankara 06510, Turkiye
| | - Ilker Ates
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ankara 06560, Turkiye
| | - Havva Nur Gurbuz
- Selcuk University, Department of Nanotechnology and Advanced Materials, Konya 42075, Turkiye
| | - Mehmet Altay Unal
- Ankara University, Stem Cell Institute, Balgat, Ankara 06520, Turkiye
| | - Hasan Nazir
- Ankara University, Faculty of Science, Department of Chemistry, Beşevler, Ankara 06100, Turkiye
| | - Aytekin Uzunoglu
- Istanbul Technical University, Faculty of Chemistry-Metallurgical Engineering, Department of Materials and Metallurgical Engineering, Istanbul 34467, Turkiye
| | - Sibel A Ozkan
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, Ankara 06560, Turkiye.
| | - Burcu Dogan Topal
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, Ankara 06560, Turkiye.
| |
Collapse
|
10
|
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
|
11
|
Khan NT, Hasan Akash MM, Sajib AA, Akhteruzzaman S. Allele-specific detection of isoniazid metabolism modulating variants of N-acetyltransferase 2 enzyme and their frequencies in the Bangladeshi population. Gene 2025; 957:149480. [PMID: 40204038 DOI: 10.1016/j.gene.2025.149480] [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: 10/11/2024] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Tuberculosis is one of the oldest diseases that still affects millions of people worldwide and remains a significant public health challenge. The N-acetyltransferase 2 (NAT2) enzyme metabolizes isoniazid (INH), a primary antibiotic in tuberculosis treatment. The single nucleotide polymorphisms (SNPs) of NAT2 affect the metabolism and function of isoniazid. The rs1801280 (T341C) and rs1208 (G803A) variants of NAT2 are associated with INH drug responses. Individuals with the slow-metabolizing rs1801280 variant of the NAT2 enzyme are at a higher risk of INH-induced liver damage and require lower doses or longer treatment regimens. At the same time, individuals with the fast-metabolizing rs1208 variant are at risk of treatment failure due to rapid drug metabolism. Genotyping of the NAT2 variants can help clinicians personalize tuberculosis treatment, optimize drug doses, and thus minimize adverse effects. Under this study, an allele-specific PCR (ASPCR) method was developed for genotyping the NAT2 variants, and the results were validated through targeted sequencing. The allele frequencies at the rs1801280 locus were 0.60 for the T allele and 0.40 for the C. For rs1208, the participants' allele frequencies were 0.27 for the G allele and 0.73 for the A allele. This ASPCR method is quick, affordable, and could be used in routine genotyping to personalize the treatment for tuberculosis patients, leading to more effective and safer treatments. We also used molecular docking to study how the rs1801280 and rs1208 variants affect the interaction between the NAT2 enzyme and drugs. A slight change was visible in the flexibility of the amino acid residues. However, those amino acids were not involved in the ligand binding mechanism.
Collapse
Affiliation(s)
- Nabiha Tasneem Khan
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh; Biotechnology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | | | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Sharif Akhteruzzaman
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Wang H, Peng LJ, Lu W, Li GR, Zhao PT, Lv X, Dong MQ, Liu ML. Acacetin reverses hypoxic pulmonary hypertension by inhibiting hypoxia-induced proliferation of pulmonary artery smooth muscle cells via SIRT1-HMGB1 pathway. Eur J Pharmacol 2025; 998:177650. [PMID: 40258398 DOI: 10.1016/j.ejphar.2025.177650] [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: 12/04/2024] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Hypoxic pulmonary hypertension (HPH) is characterized by sustained elevation of pulmonary arterial pressure and vascular remodeling. The present study is to investigate the efficacy of acacetin on HPH and its potential molecular mechanism. C57/BL6 mice were exposed to hypobaric hypoxia for six weeks. At 4th week of hypoxia exposure, mice were administrated with the water-soluble prodrug of acacetin (5, 10, 20 mg/kg) or equivalent normal saline for another two weeks. The haemodynamic and pathohistological assessment were performed. Primary pulmonary artery smooth muscle cells (PASMCs) were cultured to examine the anti-proliferation efficacy of acacetin (0.3-3 μM). The activity and expression of sirtuin1 (SIRT1) acetylation and distribution of high-mobility group box 1 (HMGB1) were determined in lungs and/or cultured PASMCs with or without RNA interference of SIRT1. Macromolecular docking and molecular dynamics simulation were done to explore the potential binding between acacetin and SIRT1. Results showed that acacetin prodrug significantly reversed the increased pulmonary pressure and vascular remodeling in HPH mice, which is associated with inhibiting the reduction in SIRT1 and the increase in HMGB1, and inhibiting the nucleocytoplasmic translocation of HMGB1. In cultured PASMCs, acacetin inhibited the hyper-proliferation induced by hypoxia, reversed the SIRT1 reduction and inhibited the nucleocytoplasmic translocation of HMGB1 and HMGB1 increase. Silencing SIRT1 abolished all the beneficial effects of acacetin. These results demonstrate that acacetin is very effective in reversing HPH by inhibiting PASMC hyper-proliferation via regulating SIRT1-HMGB1 signaling, suggesting that acacetin is likely a promising drug candidate for treating patients with HPH.
Collapse
Affiliation(s)
- Hui Wang
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611137, China
| | - Li-Jing Peng
- Department of Cardiology, 986th Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710054, China
| | - Wang Lu
- Endoscopic Center, Xi'an Chest Hospital, Xi'an, Shaanxi, 710100, China
| | - Gui-Rong Li
- Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu, 210032, China
| | - Peng-Tao Zhao
- School of Medicine, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Xing Lv
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Ming-Qing Dong
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China.
| | - Man-Ling Liu
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
15
|
Mahajan S, Kumar R, Singh A, Dhaka P, Pareek A, Kumar P, Tomar S. Targeting the host protein G3BP1 for the discovery of novel antiviral inhibitors against Chikungunya virus. Virology 2025; 608:110551. [PMID: 40306108 DOI: 10.1016/j.virol.2025.110551] [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: 12/25/2024] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
The molecular interactions between Chikungunya virus (CHIKV) non-structural protein 3 (nsP3) and the host GTPase Activating SH3 Domain Binding Protein 1 (G3BP1) are critical for CHIKV replication. The C-terminus hypervariable domain (HVD) of nsP3 protein binds to the nuclear transport factor 2 (NTF2)-like domain of G3BP1 through two tandem FGDF motifs, aiding in the disruption of stress granule (SG) formation. Given G3BP1's role in the antiviral response, it presents an attractive target for antiviral drug development. In this study, seven potential small molecules targeting the FGDF motif binding pocket of G3BP1 were identified using a structure-based virtual screening approach. The binding modes of these molecules were further investigated through molecular docking and simulations. Surface Plasmon Resonance (SPR) and Isothermal Titration Calorimetry (ITC) experiments confirmed their binding to purified G3BP1 with micromolar (μM) affinity. The antiviral efficacy of these molecules was assessed using in vitro cell culture-based assays, revealing that L-7, WIN, SB2, NAL, DHD, GSK, and FLU effectively inhibited CHIKV replication with EC50 values of 1.99, 0.40, 5.38, 1.52, 7.39, 3.66, and 0.61 μM, respectively. Additionally, CHIKV-infected cells treated with these compounds exhibited fewer virus-induced SGs compared to untreated controls without affecting SG formation under oxidative stress conditions. These findings indicate that identified inhibitors successfully block G3BP1-nsP3 interactions and suppress CHIKV replication. This is one of the first reports of small antiviral molecules targeting G3BP1, a host protein essential for stress granule formation in the antiviral cellular response and CHIKV replication.
Collapse
Affiliation(s)
- Supreeti Mahajan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Ravi Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Preeti Dhaka
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Akshay Pareek
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667.
| |
Collapse
|
16
|
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
|
17
|
Mustafa MH, Rehman FU, Ali M, Javed M, Ahmad N, Shafique T, Zidan A, Bahadur A, Iqbal S, Mahmood S, Farouk AE, Jafri I. In silico analysis of Moringaceae derived potential drug-like compounds against Newcastle disease virus. Steroids 2025; 219:109628. [PMID: 40349813 DOI: 10.1016/j.steroids.2025.109628] [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: 09/30/2024] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Newcastle disease virus (NDV) classified in the Avian avulavirus 1 [genus Orthoavulavirus, subfamily Avulavirinae, family Paramyxoviridae] constitutes a serious financial risk to the global poultry market. Available vaccines do not show good results in catering to the virus. Currently there is no FDA-approved drug to treat the disease. Nucleoprotein (NP) is a structural protein playing that constitutes a serious financial risk to the global poultry market.a valuable role in the virus replication process and encapsidation. This study is an effort to screen phytochemicals, from the plant family Moringaceae, as potential inhibitors of the N protein. ADMET (adsorption, distribution, metabolism, excretion and toxicity) analysis was performed to screen potential phytochemicals with drug likeliness. Molecular Docking was performed for the binding affinities. Gas Chromatography-Mass Spectrometry (GC-MS) and Density Function Theory (DFT) were performed to evaluate the phytochemicals bioavailability and reactivity, respectively. The stability of protein-ligand complexes was examined by 50 ns MD simulations and MM/PBSA values were calculated. Out of 128 phytochemicals, 22 phytochemicals were selected following ADMET screening. Based on the binding energies and the number of H bonding the following 10 phytochemicals were suggested as potential inhibitors to N protein of NDV - cis-11,14-eicosadienoic acid methyl ester, aurantiamide acetate, α-tocopherol, 4,8,12,16-tetramethylheptadecan-4-olide, 3,7,11,15-tetramethyl-2-hexadecen-1-ol, β-amyrin, β-sitosterol-3-O-β-d-galactopyranoside, α-amyrin, pterygospermin and sitogluside. Furthermore, DFT results showed that the 4 pytochemicals - Cis-11,14-eicosadienoic acid methyl ester, aurantiamide acetate, α-tocopherol, and 3,7,11,15-tetramethyl-2-hexadecen-1-ol were most reactive and thus could be used as potential inhibitors of NDV N protein. Further studies are required to validate the selected four phytochemicals as drug candidates against NDV.
Collapse
Affiliation(s)
- Muhammad Hammad Mustafa
- Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore 54770, Pakistan; Department of Chemistry, Faculty of Chemistry and Life Sciences, Government College University, Lahore, Pakistan
| | - Fayyaz-Ur Rehman
- Institute of Chemistry, University of Sargodha, Sargodha 40100, Pakistan
| | - Muhammad Ali
- Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore 54770, Pakistan
| | - Mohsin Javed
- Department of Chemistry, School of Science, University of Management and Technology (UMT), Lahore 54770, Pakistan
| | - Nazir Ahmad
- Department of Chemistry, Faculty of Chemistry and Life Sciences, Government College University, Lahore, Pakistan
| | - Tayyaba Shafique
- Department of Chemistry, School of Science, University of Management and Technology (UMT), Lahore 54770, Pakistan
| | - Ammar Zidan
- Biomedical Engineering Department, College of Engineering and Technologies, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Ali Bahadur
- Nanomaterials Research Center, Department of Chemistry, College of Science, Mathematics, and Technology, Wenzhou-Kean University, Wenzhou 325060 Zhejiang Province, China; Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, 1000 Morris Ave, Union, NJ 07083, USA.
| | - Shahid Iqbal
- Department of Chemistry, School of Natural Sciences (SNS), National University of Science and Technology (NUST), H-12, Islamabad 46000, Pakistan.
| | - Sajid Mahmood
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315100, China; Low Dimensional Materials Research Center at Khazar University, AZ1096 Baku, Azerbaijan
| | - Abd-ElAziem Farouk
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ibrahim Jafri
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
18
|
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
|
19
|
Rahaman KA, Mukim MSI, Hasan ML, Kim H, Pan CH, Kwon OS, Song DG, Han HS. Protein to biomaterials: Unraveling the antiviral and proangiogenic activities of Ac-Tβ 1-17 peptide, a thymosin β4 metabolite, and its implications in peptide-scaffold preparation. Bioact Mater 2025; 49:437-455. [PMID: 40177110 PMCID: PMC11964602 DOI: 10.1016/j.bioactmat.2025.02.008] [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: 07/28/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
Peptide metabolites are emerging biomolecules with numerous possibilities in biomaterial-based regenerative medicine due to their inherent bioactivities. These small, naturally occurring compounds are intermediates or byproducts of larger proteins and peptides, and they can have profound effects, such as antiviral therapeutics, proangiogenic agents, and regenerative medicinal applications. This study is among the first to focus on using thymosin β4 protein-derived metabolites to pioneer novel applications for peptide metabolites in biomaterials. This study found that the novel peptide metabolite acetyl-thymosin β4 (amino acid 1-17) (Ac-Tβ1-17) exhibited significant protease inhibition activity against SARS-CoV-2, surpassing its precursor protein. Additionally, Ac-Tβ1-17 demonstrated beneficial effects, such as cell proliferation, wound healing, and scavenging of reactive oxygen species (ROS) in human umbilical vein endothelial cells (HUVEC). Integrating Ac-Tβ1-17 into a peptide-based scaffold facilitated cell growth and angiogenesis inside the scaffold and through gradual release into the surrounding environment. The Ac-Tβ1-17 peptide treatment induced significant biochemical responses in HUVEC, increasing Akt, ERK, PI3K, MEK, and Bcl-2 gene expression and proangiogenic proteins. Ac-Tβ1-17 peptide treatment showed similar results in ex vivo by enhancing mouse fetal metatarsal growth and angiogenesis. These findings highlight the potential of natural protein metabolites to generate biologically active peptides, offering a novel strategy for enhancing biomaterial compatibility. This approach holds promise for developing therapeutic biomaterials using peptide metabolites, presenting exciting prospects for future research and applications.
Collapse
Affiliation(s)
- Khandoker Asiqur Rahaman
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Md Sofequl Islam Mukim
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Md Lemon Hasan
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Hyeok Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Cheol-Ho Pan
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Oh-Seung Kwon
- Doping Control Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Dae-Geun Song
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Hyung-Seop Han
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
- Research and Development Center, Elecell Corporation, Seoul, 02455, Republic of Korea
| |
Collapse
|
20
|
Li R, Wang Q, Gao R, Shen R, Wang Q, Cui X, Jiang Z, Zhang L, Fang J. Sepsis Important Genes Identification Through Biologically Informed Deep Learning and Transcriptomic Analysis. Clin Exp Pharmacol Physiol 2025; 52:e70031. [PMID: 40356040 DOI: 10.1111/1440-1681.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 05/15/2025]
Abstract
Sepsis is a life-threatening disease caused by the dysregulation of the immune response. It is important to identify influential genes modulating the immune response in sepsis. In this study, we used P-NET, a biologically informed explainable artificial intelligence model, to evaluate the gene importance for sepsis. About 688 important genes were identified, and these genes were enriched in pathways involved in inflammation and immune regulation, such as the PI3K-Akt signalling pathway, necroptosis and the NF-κB signalling pathway. We further selected differentially expressed genes both at bulk and single-cell levels and found TIMP1, GSTO1 and MYL6 exhibited significant different expressions in multiple cell types. Moreover, the expression levels of these 3 genes were correlated with the abundance of important immune cells, such as M-MDSC cells. Further analysis demonstrated that these three genes were highly expressed in sepsis patients with worse outcomes, such as severe, non-survived and shock sepsis patients. Using a drug repositioning strategy, we found navitoclax, curcumin and rotenone could down-regulate and bind to these genes. In conclusion, TIMP1, GSTO1 and MYL6 may serve as promising biomarkers and targets for sepsis treatment.
Collapse
Affiliation(s)
- Ruichen Li
- University of Shanghai for Science and Technology, Shanghai, China
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Qiushi Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Ru Gao
- University of Shanghai for Science and Technology, Shanghai, China
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Rutao Shen
- The National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Qihao Wang
- University of Shanghai for Science and Technology, Shanghai, China
| | - Xiuliang Cui
- The National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Zhiming Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Lijie Zhang
- Department of Information, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jingjing Fang
- Naval Medical Center, Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
He G, Wu Z, Yang X, Luo X, Zhang L, Du Z, Li S, Wan C. Design and synthesis of thiolutin derived PSMD14/HDAC dual-target inhibitors against esophageal squamous cell carcinoma. Bioorg Chem 2025; 161:108500. [PMID: 40311241 DOI: 10.1016/j.bioorg.2025.108500] [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: 03/14/2025] [Revised: 04/17/2025] [Accepted: 04/20/2025] [Indexed: 05/03/2025]
Abstract
Esophageal cancer is one of the most migratory, invasive, and lethal malignancies and has a poor prognosis, highlighting the urgent need to develop more effective drugs for its treatment. Given that PSMD14 and HDAC play an important role in the treatment of esophageal cancer, thiolutin is used as a lead compound to design and synthesize a series of dual-target PSMD14/HDAC small molecule inhibitors, aiming to discover more effective anti-esophageal cancer drugs. Through the in vitro screening of PSMD14/HDAC enzyme inhibitory activities of a series of thiolutin derivatives, it was found that compound 8b, with a linker length of 8 and a Zn2+-chelating group of 1,2-phenylenediamine, exhibited the most balanced inhibitory activity against PSMD14/HDAC.The impact of 8b on PSMD14/HDAC at the cellular level was evaluated, and its drug-like properties were further assessed in vivo. Compound 8b demonstrates balanced dual-target activity (PSMD14 IC50 = 238.7 ± 27 nM, HDAC1 IC50 = 141.2 ± 10.3 nM) and excellent cytotoxicity against esophageal cancer cells (IC50 = 30-250 nM), effectively reversing epithelial-mesenchymal transition in cancer cells. Moreover, 8b exhibited excellent pharmacokinetic characteristics. More importantly, in a nude mouse xenograft model with subcutaneous transplantation of KYSE 30 cells, compound 8b (0.8 mg/kg, BID, PO, TGI = 81 %; 0.8 mg/kg, Q3D, SC, TGI = 77 %) significantly inhibited tumor growth, outperforming single-agent or combination treatments, thereby highlighting the therapeutic advantages of dual-target inhibition. These findings highlight the potential of dual-target PSMD14/HDAC inhibitors as a promising strategy for developing anti-esophageal cancer drugs.
Collapse
Affiliation(s)
- Guoguo He
- Key Laboratory of Conservation and Utilization of Biological Resources in the Tarim Basin, Alaer, Xinjiang 843300, PR China; College of Life Science and Technology, Tarim University, Alaer, Xinjiang 843300, PR China
| | - Zhenhui Wu
- Key Laboratory of Conservation and Utilization of Biological Resources in the Tarim Basin, Alaer, Xinjiang 843300, PR China; College of Life Science and Technology, Tarim University, Alaer, Xinjiang 843300, PR China
| | - Xuan Yang
- College of Chemical Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xinrong Luo
- Key Laboratory of Conservation and Utilization of Biological Resources in the Tarim Basin, Alaer, Xinjiang 843300, PR China; College of Life Science and Technology, Tarim University, Alaer, Xinjiang 843300, PR China
| | - Lili Zhang
- Key Laboratory of Conservation and Utilization of Biological Resources in the Tarim Basin, Alaer, Xinjiang 843300, PR China; College of Life Science and Technology, Tarim University, Alaer, Xinjiang 843300, PR China
| | - Zhenting Du
- College of Chemical Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Shuwei Li
- Key Laboratory of Conservation and Utilization of Biological Resources in the Tarim Basin, Alaer, Xinjiang 843300, PR China; College of Life Science and Technology, Tarim University, Alaer, Xinjiang 843300, PR China.
| | - Chuanxing Wan
- Key Laboratory of Conservation and Utilization of Biological Resources in the Tarim Basin, Alaer, Xinjiang 843300, PR China; College of Life Science and Technology, Tarim University, Alaer, Xinjiang 843300, PR China.
| |
Collapse
|
22
|
Suzuki Y, Shuang L, Takemasa E, Takasaki Y, Yorozuya T, Mogi M. MRGPRX2-Mediated Mast Cell Degranulation by Monomethyl Methacrylate: Unveiling a Pathway in Bone Cement Implantation Syndrome. Clin Exp Pharmacol Physiol 2025; 52:e70046. [PMID: 40365957 DOI: 10.1111/1440-1681.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/31/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025]
Abstract
Bone cement implantation syndrome is a critical complication of orthopaedic surgery, characterised by hypotension and hypoxemia. This syndrome is hypothesised to result from obstruction caused by fat droplets and the biochemical release of histamine caused by bone cement components. This study aimed to elucidate the histamine release mechanism, focusing on Mas-related G protein-coupled receptor X2 expressed on mast cells, which is hypothesised to be activated by bone cement components. Using a mast cell-deficient mouse femur fracture model, we examined bone cement's effect on serum histamine. Rat basophil-like cells expressing Mas-related G protein-coupled receptor X2 were exposed to monomethyl methacrylate, a bone cement component, to assess degranulation via β-hexosaminidase release. Our findings demonstrated that histamine levels significantly increased in wild-type mice post-cement application, from 27.7 ± 11.1 to 35.3 ± 12.9 ng/mL (p = 0.016). Furthermore, Mas-related G protein-coupled receptor X2 expressing cells showed a marked increase in β-hexosaminidase release upon monomethyl methacrylate stimulation (p = 4.30 × 10-5). These results support the hypothesis that activating Mas-related G protein-coupled receptor X2 by monomethyl methacrylate contributes to bone cement implantation syndrome via histamine release. Bone cement implantation syndrome can manifest as a condition involving either peripheral vascular embolism, the release of chemical mediators, or a combination of both. Our research elucidates the role of chemical mediators, particularly histamine-induced vasodilation, in the pathophysiology of bone cement implantation syndrome, providing valuable insights that pave the way for targeted interventions to mitigate this severe complication during orthopaedic surgery.
Collapse
Affiliation(s)
- Yasuyuki Suzuki
- Department of Anaesthesiology, Saiseikai Matsuyama Hospital, Matsuyama City, Japan
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon City, Japan
- Research Division, Saiseikai Research Institute of Health Care and Welfare, Tokyo, Japan
| | - Liu Shuang
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon City, Japan
| | - Erika Takemasa
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon City, Japan
| | - Yasushi Takasaki
- Department of Anaesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Toon City, Japan
| | - Toshihiro Yorozuya
- Department of Anaesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Toon City, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon City, Japan
| |
Collapse
|
23
|
Guo B, Shi S, Xiong J, Guo Y, Wang B, Bai L, Qiu Y, Li S, Gao D, Dong Z, Tu Y. Identification of potential biomarkers in cardiovascular calcification based on bioinformatics combined with single-cell RNA-seq and multiple machine learning analysis. Cell Signal 2025; 131:111705. [PMID: 40024421 DOI: 10.1016/j.cellsig.2025.111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The molecular and genetic mechanisms underlying vascular calcification remain unclear. This study aimed to determine the differences in calcification marker-related gene expression in macrophages. METHODS The expression profiling datasets GSE104140 and GSE235995 were analysed to identify differentially expressed genes (DEGs) between fibroatheroma with calcification and diffuse intimal thickening. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, Weighted Gene Co-expression Network Analysis (WGCNA), and Gene Set Enrichment Analysis (GSEA) were performed to assess functional characteristics. Hub genes were identified through a protein-protein interaction (PPI) network and machine learning approaches. Single-cell RNA sequencing data (GSE159677) validated the expression of calcification-related genes in macrophages, while Mendelian randomization analysis explored their potential causal relationship with coronary calcification. Further validation was conducted using enzyme-linked immunosorbent assay (ELISA) on coronary calcification samples and immunohistochemistry in ApoE-/- mice. Intravascular ultrasound was performed to assess coronary calcification severity. RESULTS AND CONCLUSIONS Two key biomarkers, ITGAX and MYD88, were identified as diagnostic indicators of cardiovascular calcification. Both biomarkers were significantly upregulated in calcified samples and were strongly associated with immune processes. Single-cell RNA sequencing confirmed their high expression in multiple immune cell types. Additionally, molecular docking analysis revealed that retinoic acid interacted with both biomarkers, suggesting potential therapeutic relevance. Immunohistochemical and ELISA analyses further validated their elevated expression in calcified samples. These findings provide novel insights into the molecular mechanisms of vascular calcification and highlight potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Bingchen Guo
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China.
| | - Si Shi
- Harbin Medical University, Harbin, China; Department of Respirology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Jie Xiong
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yutong Guo
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Bo Wang
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Liyan Bai
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yi Qiu
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Shucheng Li
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Dianyu Gao
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Zengxiang Dong
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Yingfeng Tu
- Harbin Medical University, Harbin, China; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150000, China; Department of Cardiology, The Shanxi Provincial People's Hospital, Taiyuan 030000, China.
| |
Collapse
|
24
|
Zemskaya AS, Arutyunyan AF, Sherman DK, Yanvarev DV, Shuvalov AV, Kalnina LB, Kaluzhny DN, Novikov RA, Solyev PN, Valuev-Elliston VT. Isolation of recombinant HIV-1 Rev protein and investigation of a new class of benzimidazole inhibitors capability to disrupt Rev-RRE complex. Bioorg Chem 2025; 161:108487. [PMID: 40288010 DOI: 10.1016/j.bioorg.2025.108487] [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: 12/16/2024] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
In the present study, an efficient method for the expression and purification of recombinant HIV Rev protein with a C-terminal hexahistidine tag was proposed. Noteworthy, this method circumvents the precipitation of the protein into inclusion bodies and their subsequent aggregation during purification. It does not necessitate denaturing isolation conditions, in contrast to currently widely used protocols. As a result, protocols for HIV Rev isolation have been developed allowing the production of non-aggregated Rev protein in a good yield, high purity, and free of bacterial RNA impurities. This high-purity result became possible due to high salt extraction buffer usage. Complementary [α-32P]-labeled Rev response element (RRE) RNA has been synthesized and an inhibitor test system was developed based on Rev-RRE complex formation. We were able to reveal a novel class of potential Rev-RRE inhibitors based on dimeric benzimidazole derivatives and used those results to validate the testing system. The proposed protocols for screening and structure-activity relationship for new inhibitors of Rev binding to viral RNA broaden the scope of potential candidates for anti-HIV drug development.
Collapse
Affiliation(s)
- Anastasia S Zemskaya
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia.
| | - Albert F Arutyunyan
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Daria K Sherman
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Dmitry V Yanvarev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Alexey V Shuvalov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Lyudmila B Kalnina
- Ivanovsky Institute of Virology, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health, 18 Gamaleya St, 123098 Moscow, Russia
| | - Dmitry N Kaluzhny
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Roman A Novikov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Pavel N Solyev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia.
| | - Vladimir T Valuev-Elliston
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| |
Collapse
|
25
|
Sui Y, Peng C, Zhou P, Qiu L, Qu C, Li W, Wu C, Liu J. Insect odorant-binding protein modified biosensor for sensitive and specific electrochemical detection of alcohols. Biosens Bioelectron 2025; 278:117382. [PMID: 40101655 DOI: 10.1016/j.bios.2025.117382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Olfaction biosensors are playing crucial roles in detecting volatile organic compounds (VOCs) in various domains, while the response pattern of biosensors to different alcohols and the underlying reasons for the differences in response remain unclear. Herein, this study presents a sensitive electrochemical olfactory biosensor utilizing Drosophila odorant-binding protein (LUSH) as a sensing material for the detection of 11 alcohols with different molecular structures (alkyl chain lengths, hydroxyl group numbers, and cyclic alcohols) and phenol. The electrodes covalently immobilized with the LUSH proteins were characterized by atomic force microscopy (AFM), electrochemical impedance spectroscopy (EIS), and cyclic voltammetry (CV), and their ability to detect alcohols was investigated through EIS. Results showed that the LUSH modified biosensor exhibited ultrasensitive detection of multiple alcohols (detection limits: 10-100 fM), with linear ranges from 10-14 to 10-7 M and coefficients of determination (R2) of 0.948-0.992. In addition, the biosensor demonstrated high selectivity toward interfering compounds (selectivity coefficients <0.22), excellent reproducibility (relative standard deviation, RSD: 1.2%, n = 4 for parallel sensors), and good stability (response decreased by 10.2% on the 10th day). Notably, the sensitivity of the biosensor to alcohols showed alkyl chain-length dependence of n-alcohols and was influenced by the number of hydroxyl groups and the cyclic structure. More importantly, molecular docking revealed the binding modes, binding energies, and key amino acids involved in the LUSH-alcohol interaction and explained the response discrepancies.
Collapse
Affiliation(s)
- Yutong Sui
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Cong Peng
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Peng Zhou
- Beijing Environmental Engineering Technology CO., LTD, 101111, China
| | - Lina Qiu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Chen Qu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Wenhui Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Chuandong Wu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Jiemin Liu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China; Beijing Institute of Graphic Communication, Beijing, 102600, China.
| |
Collapse
|
26
|
Yu Y, Cui Z, Zhou T, Wang Y, Chen P, Wang S, Zhu Y, Liu J, Jiang S, Liu Y. Umami peptide synergy unveiled: A comprehensive study from molecular simulation to practical validation of sensing strategy. Biosens Bioelectron 2025; 278:117331. [PMID: 40043636 DOI: 10.1016/j.bios.2025.117331] [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: 10/29/2024] [Revised: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/30/2025]
Abstract
Synergistic effect is one of the main properties of umami substances, as a new natural umami agent, umami peptide synergy has not been systematically explored. Presently, conventional methods relying on human sensory evaluation and intelligent instrument analysis pose challenges due to their time-consuming and lack of high throughput. This research provides a detailed molecular-level understanding of multiple umami peptides interact with T1R1-VFT simultaneous, revealing that multiple umami peptides promotes stronger binding affinity and more effective receptor activation (from -7.3 kcal mol-1 to -11.19 kcal mol-1). The kinetic simulations demonstrated a significant reduction in the average fluctuation of protein amino acid residues during the binding process. Moreover, the hydrophobic regions on the protein surface were diminished following binding, and the resultant complex structure was more tightly packed, these phenomena may collectively represent the manifestation of synergistic effects. To validate the simulation results, biolayer interferometry sensing strategies were developed to measure the interaction process, indicating that umami peptides and T1R1-VFT could association and dissociation in solution without significant interactions with other proteins. When multiple umami peptides interacted with T1R1-VFT, the kinetic equilibrium constant decreased and affinity increased (from 1.2 e-6 M to 8.3 e-7 M), showing significant synergistic effect. Furthermore, the practical application ability of this sensing strategy was verified in a complex matrix with multiple real samples. Overall, this comprehensive study combined micro-molecular simulation and biological experiment verification, offering a deeper understanding of umami peptide synergy and paving the way for innovative approaches in flavor science and food product development.
Collapse
Affiliation(s)
- Yanyang Yu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhiyong Cui
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tianxing Zhou
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yueming Wang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Panpan Chen
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shengnan Wang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yiwen Zhu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jing Liu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China; School of Food Science and Engineering, Ningxia University, Yinchuan, 750021, China
| | - Shui Jiang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yuan Liu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China; School of Food Science and Engineering, Ningxia University, Yinchuan, 750021, China.
| |
Collapse
|
27
|
Terenteva O, Mostovaya O, Bukharov M, Mukhametzyanov T, Bikmukhametov A, Lyubina A, Voloshina A, Petrov K, Padnya P, Stoikov I. Peptidomimetics based on thiacalixarene with L-tyrosine moieties: Antibacterial activity against methicillin-resistant Staphylococcus aureus and degradation induced by binding to α-chymotrypsin. Bioorg Chem 2025; 160:108434. [PMID: 40187027 DOI: 10.1016/j.bioorg.2025.108434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The design of new antimicrobial agents is an important challenge due to the growing resistance of microorganisms to existing antibiotics. In recent years, the trend towards the development of compounds and materials with (bio)degradable properties has emerged. In this work, we propose and develop a method for the synthesis of new peptidomimetics, i.e., water-soluble macrocyclic quaternary ammonium salts containing L-tyrosine fragments based on p-tert-butylthiacalix[4]arene in various stereoisomeric forms (cone, partial cone, and 1,3-alternate). These compounds have low cytotoxicity (IC50 = 80-267 μM) and high antibacterial activity (MIC = 0.5-15.6 μM) against Gram-positive bacterial strains including methicillin-resistant Staphylococcus aureus (MRSA). The obtained peptidomimetics can bind α-chymotrypsin with the formation of supramolecular systems and their subsequent degradation. Our results demonstrate the first example of multi-action thiacalixarene derivatives with antibacterial activity, protein binding ability and degradation induced by binding to α-chymotrypsin. The obtained results open the possibility of creating multi-action peptidomimetic systems with antimicrobial and biodegradable effect.
Collapse
Affiliation(s)
- Olga Terenteva
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Olga Mostovaya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Mikhail Bukharov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Timur Mukhametzyanov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Azamat Bikmukhametov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Anna Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Alexandra Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Konstantin Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Pavel Padnya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| | - Ivan Stoikov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| |
Collapse
|
28
|
Zhou Y, Xu L, Yan Y, Li Y, Wang S, Ding X, Zhu P, Jiao J, Zhang L, Ma J, Chen L. Association and binding interaction between per- and polyfluoroalkyl substances and maternal thyroid hormones: A case study based on a prospective birth cohort in Wuxi, China. ENVIRONMENTAL RESEARCH 2025; 275:121439. [PMID: 40118311 DOI: 10.1016/j.envres.2025.121439] [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: 01/10/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND AND AIM The relationship between prenatal exposure to per- and polyfluoroalkyl substances (PFASs), a well-known endocrine disruptor, and thyroid hormones (THs) levels remains unclear. Therefore, this study aimed to investigate this relationship in a birth cohort during the second trimester. METHOD This prospective study included 562 pregnant women in the Wuxi Birth Cohort from 2019 to 2021 and quantified the serum concentrations of 23 PFASs and 5 THs. Multiple statistical models were used to assess the associations between individual or combined PFASs concentrations and THs, while molecular docking simulated the interactions between PFASs and four thyroid-related proteins. RESULTS The median concentration of ∑23PFASs was 71.91 ng/mL, with perfluorovaleric acid (PFPeA) (18.13 ng/mL) emerging as the predominant PFAS. Most PFASs were negatively associated with maternal free thyroxine (FT4) and thyroid-stimulating hormone (TSH) levels, whereas perfluorobutane sulfonate (PFBS) was positively correlated with TSH levels. A similar trend was observed in the weighted quantile sum (WQS) model, in which combined PFASs exposure was inversely associated with the FT4 and TSH levels. Molecular docking results showed that compared with TH natural ligand thyroxine (T4), perfluorooctanoic acid (PFOA), perfluorooctane sulfonate (PFOS), and 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl- PFESA) exhibited relatively high binding affinity with thyroid-related proteins (-6.6 to -9.8 kcal/mol vs. T4: -5.6 to -8.6 kcal/mol). Furthermore, PFASs with medium chain lengths and sulfonic acid groups exhibited enhanced protein-binding properties. CONCLUSION PFASs exposure may affect THs homeostasis during pregnancy. Moreover, different types and concentrations of PFASs have different effects on THs in the maternal serum.
Collapse
Affiliation(s)
- Yahui Zhou
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, Wuxi, 214023, Jiangsu, China
| | - Lingling Xu
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, China
| | - Yan Yan
- Department of Obstetrics and Gynecology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214000, Jiangsu, China
| | - Yao Li
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, China
| | - Shunan Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, China
| | - Xinliang Ding
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, China
| | - Pengfei Zhu
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, China
| | - Jiandong Jiao
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, China
| | - Le Zhang
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, Wuxi, 214023, Jiangsu, China
| | - Jinqi Ma
- Department of Obstetrics and Gynecology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214000, Jiangsu, China.
| | - Limei Chen
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, China.
| |
Collapse
|
29
|
Liu C, Yi X, Wang P, Wang P, Li Y, Xu H, Li L, Yang W, Tu Y. Tinosporine alleviates collagen-induced arthritis in rats via modulating the gut microbiota-metabolome-immunity axis. Int Immunopharmacol 2025; 157:114752. [PMID: 40300353 DOI: 10.1016/j.intimp.2025.114752] [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: 02/17/2025] [Revised: 04/08/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an inflammation-mediated autoimmune disease. Tinosporine (TIN), derived from Tinospora sinensis (Lour.) Merr. has significant anti-inflammatory and immunosuppressive effects. However, the anti-RA effect and mechanism of TIN have not been fully elucidated. OBJECTIVE This study elucidates the mechanism of TIN in alleviating collagen-induced arthritis (CIA) in rats based on the gut microbiome-metabolomic-immunity axis. MATERIALS AND METHODS We established CIA rat model to evaluate the efficacy of TIN. Based on 16S rRNA sequencing analysis, fecal metabolomics profiling and the concentrations of short-chain fatty acids determination to investigate the effect of TIN on gut microbiota composition and metabolome changes. Histopathology showed that TIN protected the intestinal barrier, then use ELISA and flow cytometry to analyze the mechanism of TIN, and qRT-PCR and WB were employed for verification. RESULTS TIN ameliorated joint damage and inflammation in CIA rats, histopathological observation confirmed that TIN had protective effect on intestinal barrier. 16S rRNA sequencing analysis and fecal metabolomics profiling confirmed that TIN intervention regulates the composition of gut microbiome and promote the propagation of probiotics, the abundance changes of 12 serum metabolites in the CIA group were reversed by TIN intervention. After intervention with TIN, propionic acid, butyric acid, isobutyric acid, valeric acid, isovaleric acid, and caproic acid rise significantly, but acetic acid cannot be reversed. Then ELISA and flow cytometry confirmed that TIN intervention could regulate the level of inflammatory factors, maintain the integrity of the intestinal barrier and restoring the imbalance of Th1/Th2 and Th17/Treg ratios in the colon of CIA rats. CONCLUSION TIN inhibited the inflammatory response of CIA rats by regulating the gut microbiome-metabolome-immunity axis, reversed the abnormalities of intestinal flora and differential metabolites, and maintained the integrity of the intestinal barrier.
Collapse
Affiliation(s)
- Chuan Liu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China.
| | - Xiangrui Yi
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Ping Wang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Peng Wang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Yafan Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Huijuan Xu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Ling Li
- School of Great Health Management, Xihua University, Chengdu 610039, China
| | - WenYu Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Xihua University, Chengdu 610039, China
| | - Ya Tu
- Experimental research center, China Academy of Chinese Medicine sciences, Beijing 100700, China.
| |
Collapse
|
30
|
Zhang M, Qiu H, Han Z, Ma Y, Hou J, Yuan J, Jia H, Zhou M, Lu H, Wu Y. Topical transdermal administration of lenalidomide nanosuspensions-based hydrogels against melanoma: In vitro and in vivo studies. Int J Pharm X 2025; 9:100316. [PMID: 39898009 PMCID: PMC11787432 DOI: 10.1016/j.ijpx.2025.100316] [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: 08/27/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Percutaneous neoadjuvant therapy has proven effective in diminishing tumor size and the surgical intervention area, which couldeffectively mitigate the risk of tumor recurrence and enhance immunotherapy efficacy. Lenalidomide, an approved medication orally used to treat myeloma, was loaded into nanosuspensions-based hydrogels (Len-NBHs) for transdermal administration as a percutaneous neoadjuvant therapy. This study was designed to investigate the inhibitory effect and mechanism of Len-NBHs on melanoma. Network pharmacology and transcriptomic analyses identified key targets and signaling pathways. The effects of lenalidomide on melanoma were further verified through Western blotting, immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction,using both in vitro cell experiments and in vivo melanoma mouse models. Lenalidomide could induce melanoma cells apoptosis, disrupt cell cycle progression, impede cell migration and invasion, and modify tumor microenvironment (TME). Mechanistically, lenalidomide reversed the abnormal activation of the PI3K-AKT signaling pathway and the overexpression of CD93, while also recruiting CD8+ T cells, CD4+ T cells, and dendritic cells to infiltrate the tumor site. Transdermal administration of Len-NBHs represents a promising adjuvant therapy for the treatment of malignant melanoma. Preoperative administration of Len-NBHs can inhibit the outward spread of melanoma, reduce tumor size, thereby decreasing the surgical excision area and improving patient survival rates and prognosis.
Collapse
Affiliation(s)
| | | | - Zheyi Han
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yazhong Ma
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingjing Hou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingwei Yuan
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Haiyan Jia
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Menglu Zhou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Hongjie Lu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yan Wu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| |
Collapse
|
31
|
Chi X, Chen R, Chen R, Xu Y, Deng Y, Yang X, Pan Z, Xu X, Pan Y, Li Q, Zhou P, Huang W. Discovery and characterization of novel FAK inhibitors for breast cancer therapy via hybrid virtual screening, biological evaluation and molecular dynamics simulations. Bioorg Chem 2025; 159:108400. [PMID: 40163988 DOI: 10.1016/j.bioorg.2025.108400] [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: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Focal adhesion kinase (FAK) is a critical drug target implicated in various disease pathways, including hematological malignancies and breast cancer. Therefore, identifying FAK inhibitors with novel scaffolds could offer new opportunities for developing effective therapeutic compounds. Herein, we disclosed the discovery of a new backbone inhibitor of FAK using an "internal" database, employing a structure-based high-transparency permeability virtual screening (HTVS) and a DeepDock algorithm based on geometric deep learning. Subsequently, molecular docking was conducted at different precisions to identify 10 compounds for further evaluation of biological activity. Ultimately, compound 4, a pyrimidin-4-amine derivative, demonstrated inhibitory activity against FAK and breast cancer cells, further supporting its potential as a FAK inhibitor. Moreover, molecular dynamics simulations were carried out to gain more detailed insights into the binding mechanism between compound 4 and FAK to guide subsequent structural optimization.
Collapse
Affiliation(s)
- Xinglong Chi
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Runmei Chen
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; School of Pharmacy, Hangzhou Medical College, Hangzhou 310058, PR China
| | - Roufen Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Yingxuan Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Yaru Deng
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Xinle Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China; College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Zhichao Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiangwei Xu
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Youlu Pan
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Qin Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310058, PR China.
| | - Peng Zhou
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China.
| | - Wenhai Huang
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China.
| |
Collapse
|
32
|
da Rocha MN, de Sousa DS, da Silva Mendes FR, Dos Santos HS, Marinho GS, Marinho MM, Marinho ES. Ligand and structure-based virtual screening approaches in drug discovery: minireview. Mol Divers 2025; 29:2799-2809. [PMID: 39223358 DOI: 10.1007/s11030-024-10979-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The compilation of ligand and structure-based molecular modeling methods has become an important practice in virtual screening applied to drug discovery. This systematic review addresses and ranks various virtual screening strategies to drive the selection of the optimal method for studies that have as their starting point a multi-ligand investigation and investigation based on the protein structure of a therapeutic target. This study shows examples of applications and an evaluation based on the objective and problematic of a series of virtual screening studies present in the ScienceDirect® database. The results showed that the molecular docking technique is widely used in scientific production, indicating that approaches that use protein structure as a starting point are the most promising strategy for drug discovery that relies on virtual screening-based research.
Collapse
Affiliation(s)
- Matheus Nunes da Rocha
- Postgraduate Program in Natural Sciences, Sciences and Technology Center, State University of Ceará, Fortaleza, CE, Brazil.
| | - Damião Sampaio de Sousa
- Postgraduate Program in Natural Sciences, Sciences and Technology Center, State University of Ceará, Fortaleza, CE, Brazil
| | | | - Helcio Silva Dos Santos
- Postgraduate Program in Natural Sciences, Sciences and Technology Center, State University of Ceará, Fortaleza, CE, Brazil
- Chemistry Department, State University of Acaraú Valley, Sobral, CE, Brazil
| | - Gabrielle Silva Marinho
- Faculdade de Educação, Ciências e Letras de Iguatu, State University of Ceará, Fortaleza, CE, Brazil
| | | | - Emmanuel Silva Marinho
- Postgraduate Program in Natural Sciences, Sciences and Technology Center, State University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
33
|
Sarangi S, Mahapatra RK. Identification of therapeutics against PfPK6 protein of Plasmodium falciparum: Structure and Deep Learning approach. Exp Parasitol 2025; 273:108947. [PMID: 40288672 DOI: 10.1016/j.exppara.2025.108947] [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: 11/29/2024] [Revised: 04/05/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
The Plasmodium falciparum Protein Kinase 6 (PfPK6) is a serine/threonine protein kinase categorized under the CMGC group, displaying both cyclin-dependent kinases (CDKs) and mitogen-activated protein kinases (MAPKs) activity. Previous research has indicated that PfPK6 is expressed during the trophozoite and schizont stages of the Plasmodium falciparum asexual blood stage. Unlike typical cyclin-dependent kinases, PfPK6 demonstrates kinase activity independent of cyclin, making it a promising target for drug identification. In this study, we utilized a computational approach to identify a novel PfPK6 inhibitor through virtual screening of small inhibitor compounds from diverse datasets, employing a structure-based approach and a Deep Learning (DL) model. The most promising inhibitor molecule, TCMDC-132409 from the Tres Cantos Antimalarial Set, exhibited a binding affinity of -13.553 kcal/mol against PfPK6. Additionally, a 200ns molecular dynamics simulation study confirmed the stability of the binding mode, indicating the potential of TCMDC-132409 as an antiplasmodial inhibitor for further investigation.
Collapse
Affiliation(s)
- Sibasish Sarangi
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
| | | |
Collapse
|
34
|
Majumder A, Jyotisha, Nasim F, Qureshi IA. Comparative proteomics and structure-based approach to unravel the therapeutic drug target of Theileria species. J Genet Eng Biotechnol 2025; 23:100488. [PMID: 40390487 PMCID: PMC12008689 DOI: 10.1016/j.jgeb.2025.100488] [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: 01/15/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 05/21/2025]
Abstract
Theileriosis, caused by protozoan parasites of genus Theileria, primarily affects both domestic and wild ruminants. It can lead to significant economic losses in livestock farming due to decreased productivity and high mortality rates in susceptible animals, while treatment measures are not cost-effective. Since most of mechanisms of this disease remain unknown, this study investigates the differences in the mode of pathogenesis between transforming and non-transforming groups through an in silico comparative proteomics approach to recognize the key players involved in host cell transformation. Although the major biological processes and molecular functions are almost conserved between the two groups, PEST-motif containing secretory proteins of SfiI, SVSP, and Tash-AT gene families were identified as important candidates with the potential to transform infected host cells. Several members of PEST-motif containing proteins possess signal peptides, nuclear localization signals, and trans-membrane helices, further supporting their potential to transform host cells. Additionally, structural analysis helped in the identification of a parasitic protein (SfiIp) from SfiI family as a plausible drug target. Virtual screening revealed FDA-approved drugs (i.e. atogepant and rimegepant) as promising compounds, showing the highest affinity for SfiIp during molecular docking. Further studies, including molecular dynamics simulation, principal component analysis, and free energy landscape, suggested that these drug molecules exhibit the stable interaction with protein. Therefore, our research could facilitate the identification and targeting of novel drug candidates that may be further implemented to recognize effective therapeutics against Theileria infections.
Collapse
Affiliation(s)
- Anusha Majumder
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Hyderabad 500046, India
| | - Jyotisha
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Hyderabad 500046, India
| | - Fouzia Nasim
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Hyderabad 500046, India
| | - Insaf Ahmed Qureshi
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Hyderabad 500046, India.
| |
Collapse
|
35
|
Ávila-Avilés RD, Bahena-Culhuac E, Hernández-Hernández JM. (-)-Epicatechin metabolites as a GPER ligands: a theoretical perspective. Mol Divers 2025; 29:2099-2115. [PMID: 39153018 PMCID: PMC12081483 DOI: 10.1007/s11030-024-10968-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Diet habits and nutrition quality significantly impact health and disease. Here is delve into the intricate relationship between diet habits, nutrition quality, and their direct impact on health and homeostasis. Focusing on (-)-Epicatechin, a natural flavanol found in various foods like green tea and cocoa, known for its positive effects on cardiovascular health and diabetes prevention. The investigation encompasses the absorption, metabolism, and distribution of (-)-Epicatechin in the human body, revealing a diverse array of metabolites in the circulatory system. Notably, (-)-Epicatechin demonstrates an ability to activate nitric oxide synthase (eNOS) through the G protein-coupled estrogen receptor (GPER). While the precise role of GPER and its interaction with classical estrogen receptors (ERs) remains under scrutiny, the study employs computational methods, including density functional theory, molecular docking, and molecular dynamics simulations, to assess the physicochemical properties and binding affinities of key (-)-Epicatechin metabolites with GPER. DFT analysis revealed distinct physicochemical properties among metabolites, influencing their reactivity and stability. Rigid and flexible molecular docking demonstrated varying binding affinities, with some metabolites surpassing (-)-Epicatechin. Molecular dynamics simulations highlighted potential binding pose variations, while MMGBSA analysis provided insights into the energetics of GPER-metabolite interactions. The outcomes elucidate distinct interactions, providing insights into potential molecular mechanisms underlying the effects of (-)-Epicatechin across varied biological contexts.
Collapse
Affiliation(s)
- Rodolfo Daniel Ávila-Avilés
- Laboratory of Epigenetics of Skeletal Muscle Regeneration, Department of Genetics and Molecular Biology, Centre for Research and Advanced Studies of IPN (CINVESTAV), Mexico City, Mexico
- Transdisciplinary Research for Drug Discovery, Sociedad Mexicana de Epigenética y Medicina Regenerativa A. C. (SMEYMER), Mexico City, Mexico
| | - Erick Bahena-Culhuac
- Laboratory of Epigenetics of Skeletal Muscle Regeneration, Department of Genetics and Molecular Biology, Centre for Research and Advanced Studies of IPN (CINVESTAV), Mexico City, Mexico
- Transdisciplinary Research for Drug Discovery, Sociedad Mexicana de Epigenética y Medicina Regenerativa A. C. (SMEYMER), Mexico City, Mexico
| | - J Manuel Hernández-Hernández
- Laboratory of Epigenetics of Skeletal Muscle Regeneration, Department of Genetics and Molecular Biology, Centre for Research and Advanced Studies of IPN (CINVESTAV), Mexico City, Mexico.
| |
Collapse
|
36
|
Feng M, Gong W, Zhu X, Zhu J, Hu J, Xu W, Ma Z, Fu S, Chen X. Covalent binding of 5-tetradecyloxy-2-furoic acid (TOFA) and c(RGDfK) and its co-delivery with Lipusu, a novel synergistic strategy to inhibit the proliferation of nasopharyngeal cancer. Eur J Pharm Sci 2025; 209:107092. [PMID: 40228725 DOI: 10.1016/j.ejps.2025.107092] [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: 01/27/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025]
Abstract
As the world's only commercially available paclitaxel liposome, Lipusu (Lip) has been clinically used in chemotherapy for >20 years, but the design concept of Lip remains largely unchanged since its initial development. Based on the study of Acetyl-CoA-carboxylase 1 (ACC1) in nasopharyngeal carcinoma (NPC), we proposed the concept of next-generation liposomes (NGL) utilizing lipid demand balance. In this study, we evaluated the feasibility of ACC1 and integrin αVβ3 as NPC targets, and designed 10 conjugates of 5-tetradecyloxy-2-furoic acid (TOFA) and c(RGDfK) that can bind to Lip. Considering the results of chemical parameter prediction, molecular docking, molecular dynamics simulation (MD) and other aspects, we finally selected and synthesized the compound F, and successfully constructed F-Lip by simple incubation method. Compared with Lip, F-Lip showed stronger toxicity in both HONE-1 cells and corresponding tumor-bearing mice. In conclusion, by regulating the balance of lipid demand, the toxicity of Lip can be improved so as to achieve the goal of inhibiting the proliferation of NPC. This study provides a new model for the future design and development of Lip.
Collapse
Affiliation(s)
- Min Feng
- College of Life Sciences and Pharmacy, Hainan University, Haikou, Hainan, PR China; Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Haikou, Hainan, PR China
| | - Wei Gong
- College of Life Sciences and Pharmacy, Hainan University, Haikou, Hainan, PR China; Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Haikou, Hainan, PR China
| | - Xin Zhu
- College of Life Sciences and Pharmacy, Hainan University, Haikou, Hainan, PR China
| | - Juan Zhu
- Department of Pathology, Yancheng City Dafeng People's Hospital, Yanchen, Jiangsu, PR China
| | - Junjie Hu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Haikou, Hainan, PR China
| | - Weihua Xu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Haikou, Hainan, PR China; Hainan Tropical Cancer Research Institute, Haikou, Hainan, PR China
| | - Zhichao Ma
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Haikou, Hainan, PR China
| | - Shengmiao Fu
- Hainan Lvtou Medical Laboratory Center, Haikou, Hainan, PR China.
| | - Xinping Chen
- College of Life Sciences and Pharmacy, Hainan University, Haikou, Hainan, PR China; Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Haikou, Hainan, PR China; Hainan Tropical Cancer Research Institute, Haikou, Hainan, PR China.
| |
Collapse
|
37
|
Dubey A, Alanazi AM, Bhardwaj R, Ragusa A. Identification of potential NUDT5 inhibitors from marine bacterial natural compounds via molecular dynamics and free energy landscape analysis. Mol Divers 2025; 29:1929-1944. [PMID: 39225905 PMCID: PMC12081488 DOI: 10.1007/s11030-024-10950-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
NUDIX hydrolase 5 (NUDT5) is an enzyme involved in the hydrolysis of nucleoside diphosphates linked to other moieties, such as ADP-ribose. This cofactor is vital in redox reactions and is essential for the activity of sirtuins and poly(ADP-ribose) polymerases, which are involved in DNA repair and genomic stability. It has been shown that NUDT5 activity can also influence NAD+ homeostasis, thereby affecting cancer cell metabolism and survival. In this regard, the discovery of NUDT5 inhibitors has emerged as a potential therapeutic approach in cancer treatment. In this study, we conducted a high-throughput virtual screening of marine bacterial compounds against the NUDT5 enzyme and four molecules were selected based on their docking scores. These compounds established strong interactions within the NUDT5 active site, with molecular analysis highlighting the key role of Trp28A and Trp46B residues. Molecular dynamics simulations over 200 ns indicated a stable behavior, in association with root mean square deviation values always below 3 Å, suggesting conformational stability. Free energy landscape analysis further supported their potential as NUDT5 inhibitors, offering avenues for novel therapeutic strategies against NUDT5-associated breast cancer.
Collapse
Affiliation(s)
- Amit Dubey
- Department of Pharmacology, Saveetha Dental College, and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nādu, 600077, India
- Department of Computational Chemistry and Drug Discovery Division, Quanta Calculus, Greater Noida, 201310, India
| | - Amer M Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rima Bhardwaj
- Department of Chemistry, Poona College, Savitribai Phule Pune University, Pune, India.
| | - Andrea Ragusa
- Institute of Nanotechnology, CNR-Nanotec, Via Monteroni, 73100, Lecce, Italy.
- Department of Life Sciences, Health and Health Professions, Link Campus University, Via del Casale Di San Pio V 44, 00165, Rome, Italy.
| |
Collapse
|
38
|
Shi XY, He YX, Ge MY, Liu P, Zheng P, Li ZH. Gastrodin promotes CNS myelinogenesis and alleviates demyelinating injury by activating the PI3K/AKT/mTOR signaling. Acta Pharmacol Sin 2025; 46:1610-1623. [PMID: 40011630 DOI: 10.1038/s41401-025-01492-z] [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: 08/20/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025]
Abstract
Demyelination is a common feature of numerous neurological disorders including multiple sclerosis and leukodystrophies. Although myelin can be regenerated spontaneously following injury, this process is often inadequate, potentially resulting in neurodegeneration and exacerbating neurological dysfunction. Several drugs aimed at promoting the differentiation of oligodendrocyte precursor cells (OPCs) have yielded unsatisfactory clinical effects. A recent study has shifted the strategy of pro-OPC differentiation towards enhancing myelinogenesis. In this study we identified the pro-myelinating drug using a zebrafish model. Five traditional Chinese medicine monomers including gastrodin, paeoniflorin, puerarin, salidroside and scutellarin were assessed by bath-application in Tg (MBP:eGFP-CAAX) transgenic line at 1-5 dpf. Among the 5 monomers, only gastrodin exhibited significant pro-myelination activity. We showed that gastrodin (10 µM) enhanced myelin sheath formation and oligodendrocyte (OL) maturation without affecting the number of OLs. Gastrodin markedly increased the phosphorylation levels of PI3K, AKT, and mTOR in primary cultured OLs via direct interaction with PI3K. Co-treatment with the PI3K inhibitor LY294002 (5 µM) mitigated gastrodin-induced OL maturation. Furthermore, injection of gastrodin (100 mg·kg-1·d-1, i.p.) effectively facilitated remyelination in a lysophosphatidylcholine-induced demyelinating mouse model and alleviated demyelination in the experimental autoimmune encephalomyelitis mice. These results identify gastrodin as a promising therapeutic agent for demyelinating diseases and highlight the potential of the zebrafish model for screening pro-myelinogenic pharmacotherapy.
Collapse
Affiliation(s)
- Xiao-Yu Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200030, China
| | - Yi-Xi He
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200030, China
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200030, China
| | - Man-Yue Ge
- Institute of Neuroscience, MOE Key Laboratory of Molecular Neurobiology, Naval Medical University, Shanghai, 200433, China
| | - Peng Liu
- Institute of Neuroscience, MOE Key Laboratory of Molecular Neurobiology, Naval Medical University, Shanghai, 200433, China.
| | - Ping Zheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200030, China.
| | - Zheng-Hao Li
- Institute of Neuroscience, MOE Key Laboratory of Molecular Neurobiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
39
|
Bin X, Pawelek PD. The enterobactin biosynthetic intermediate 2,3-dihydroxybenzoic acid is a competitive inhibitor of the Escherichia coli isochorismatase EntB. Protein Sci 2025; 34:e70160. [PMID: 40400396 PMCID: PMC12096016 DOI: 10.1002/pro.70160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/23/2025]
Abstract
The Escherichia coli enterobactin biosynthetic protein EntB is a bifunctional enzyme that catalyzes hydrolysis of isochorismate via its N-terminal isochorismatase (IC) domain, and then transfers phosphopantetheinylated 2,3-DHB to EntF via the EntB C-terminal aryl carrier protein (ArCP) domain. Here we used a fluorescence anisotropy binding assay to investigate the ability of 2,3-DHB to bind to enzymes in the DHB synthetic arm of the pathway. We found that 2,3-DHB binds to EntE as a natural substrate with high affinity (KD = 0.54 μM). Furthermore, apo-EntB was found to bind to 2,3-DHB with moderate affinity (KD = 8.95 μM), despite the fact that this intermediate is neither a substrate nor a product of EntB. Molecular docking simulations predicted a top-ranked ensemble in which 2,3-DHB is bound at the isochorismatase active site of apo-EntB. Steady-state coupled enzymatic assays revealed that 2,3-DHB is a competitive inhibitor of apo-EntB isochorismatase activity (Ki ~ 200 μM), consistent with modeling predictions. Monitoring the EntC-EntB coupled reaction in real time via isothermal titration microcalorimetry confirmed that EntB was required to drive the EntC reaction toward isochorismate formation. Furthermore, addition of 2,3-DHB to the ITC-monitored reaction resulted in a suppression of integrated reaction heats, consistent with our observation that the molecule acts as a competitive inhibitor of EntB. Finally, we found that 2,3-DHB lowered the efficiency of EntC-EntB isochorismate channeling by approximately 70%, consistent with steric blockage of the isochorismatase active site by bound 2,3-DHB. Given its inhibitory properties, we hypothesize that 2,3-DHB plays a regulatory role in feedback inhibition in order to maintain iron homeostasis upon intracellular accumulation of sufficient ferric enterobactin.
Collapse
Affiliation(s)
- Xue Bin
- Department of Chemistry and BiochemistryConcordia UniversityMontrealQuebecCanada
| | - Peter D. Pawelek
- Department of Chemistry and BiochemistryConcordia UniversityMontrealQuebecCanada
| |
Collapse
|
40
|
Menezes Dantas DD, Macêdo NS, Sousa Silveira ZD, Santos Barbosa CRD, Muniz DF, Bezerra AH, Sousa JTD, Alencar GG, Morais Oliveira-Tintino CDD, Tintino SR, da Rocha MN, Marinho ES, Marinho MM, Dos Santos HS, Melo Coutinho HD, Cunha FABD. Naringenin as potentiator of norfloxacin efficacy through inhibition of the NorA efflux pump in Staphylococcus aureus. Microb Pathog 2025; 203:107504. [PMID: 40154849 DOI: 10.1016/j.micpath.2025.107504] [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: 10/07/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Bacterial resistance is a major challenge in the treatment of Staphylococcus aureus infections, with efflux mechanisms highlighted as reducing the efficacy of antibiotics. In this study, we investigated the potential of naringenin, a natural flavonoid, as an antibacterial agent and efflux pump inhibitor in S. aureus strains 1199 and 1199B. The studies used minimum inhibitory concentration (MIC) assays, ethidium bromide (EtBr) fluorescence emission enhancement assays, cell membrane permeability assays, and in silico molecular docking and ADME prediction assays. Naringenin showed no relevant antibacterial activity (MIC ≥1024 μg/mL). However, it potentiated the effect of norfloxacin and EtBr, reducing their MICs and increasing the fluorescence emission of EtBr, suggesting a possible inhibition of the NorA efflux pump. Bacterial membrane permeability was not significantly affected. Molecular docking assays indicated that naringenin interacts with the chlorpromazine binding site and has more favorable affinity energy than the chlorpromazine-NorA complex. ADME prediction showed favorable physicochemical properties, good oral absorption, metabolic stability and central nervous system safety. Therefore, naringenin demonstrates the potential to reverse the efficacy of norfloxacin in S. aureus by associating with efflux inhibition through effective interactions with the NorA protein, suggesting its therapeutic potential against bacterial resistance.
Collapse
Affiliation(s)
- Debora de Menezes Dantas
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil.
| | - Nair Silva Macêdo
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Zildene de Sousa Silveira
- Graduate Program in Biological Sciences- PPGCB, Federal University of Pernambuco - UFPE, Recife, Pernambuco, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Cristina Rodrigues Dos Santos Barbosa
- Postdoctoral Intern at the Semiarid Bioprospecting Laboratory (LABSEMA), Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Débora Feitosa Muniz
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Antônio Henrique Bezerra
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, CE, Brazil; Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Josivânia Teixeira de Sousa
- Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Gabriel Gonçalves Alencar
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil
| | - Cícera Datiane de Morais Oliveira-Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Postdoctoral Intern at the Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Matheus Nunes da Rocha
- Ceará State University, Postgraduate Program in Natural Sciences, Laboratory of Chemistry of Natural Products, Fortaleza, CE, Brazil
| | - Emmanuel Silva Marinho
- Ceará State University, Postgraduate Program in Natural Sciences, Laboratory of Chemistry of Natural Products, Fortaleza, CE, Brazil
| | | | - Hélcio Silva Dos Santos
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Henrique Douglas Melo Coutinho
- Laboratory of Microbiology and Molecular Biology (LMBM), Regional University of Cariri - URCA, Crato, 63105-000, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| | - Francisco Assis Bezerra da Cunha
- Semi-arid Bioprospecting Laboratory and Alternative Methods, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil; Biological Chemistry Department, Pimenta Campus, Regional University of Cariri - URCA, Crato, CE, Brazil
| |
Collapse
|
41
|
Dai G, Yang Y, Mu W, Li Y, Tu Y. Integrating thermal proteome profiling and virtual screening to reveal the mechanism of Bletilla striata against osteoclast-driven osteoporosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156735. [PMID: 40220420 DOI: 10.1016/j.phymed.2025.156735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/30/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Osteoclast (OC) overactivation and increased bone resorption are central factors contributing to osteoporosis (OP). Bletilla striata (Thunb.) Reichb. f. is known for its hemostatic properties, however, its therapeutic potential and underlying mechanisms in OC-dependent OP remain unclear. PURPOSE This study aimed to evaluate the anti-OP effects of Bletilla striata extract (BSE) and elucidate its mechanisms of action (MoAs) through an innovative integrated strategy combining isothermal thermal proteome profiling (TPP) and structure-based virtual screening (SBVS). RESULTS BSE treatment significantly decreased bone loss in ovariectomized rats, as evidenced by micro-CT and pathological analyses. This effect was attributed to inhibition of OC formation by BSE. BSE markedly blocked RANKL-induced OC differentiation, inhibited F-actin ring formation, and decreased the expression of marker genes and proteins including NFATc1, CTSK, and MMP-9. Using an isothermal TPP strategy, 110 proteins were identified as the direct targets of BSE for OC inhibition based on proteome-level thermal stability analysis (fold change > 2.0). Subsequently, using label-free quantitative proteomics, 159 proteins were identified as indirect response proteins resulting from the interaction between BSE and direct targets. The SBVS approach was employed to establish a one-to-one correspondence between the chemical ingredients in BSE and the direct targets revealed by isothermal TPP. Based on these data, an "active ingredient-direct target-indirect response signal" interaction network was constructed, which revealed the key targets (ATM, PIK3R1, BUB1B, and NR3C1) and active ingredients (phenanthrene dimers and steroids) of BSE responsible for inhibiting OC formation. CONCLUSION This study not only highlights the therapeutic potential of BSE for treating OP but also provides a novel methodological framework for interpreting the MoAs of complex Traditional Chinese Medicine (TCM) extracts. By integrating isothermal TPP and SBVS, this study offers new insights into the mechanism of TCM.
Collapse
Affiliation(s)
- Guiyu Dai
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ying Yang
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China
| | - Wen Mu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yanfang Li
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China.
| | - Yanbei Tu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
42
|
Lin M, Li K, Zhang Y, Pan F, Wu W, Zhang J. DisDock: A Deep Learning Method for Metal Ion-Protein Redocking. Proteins 2025; 93:1171-1180. [PMID: 39838957 DOI: 10.1002/prot.26791] [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: 01/30/2024] [Revised: 09/10/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025]
Abstract
The structures of metalloproteins are essential for comprehending their functions and interactions. The breakthrough of AlphaFold has made it possible to predict protein structures with experimental accuracy. However, the type of metal ion that a metalloprotein binds and the binding structure are still not readily available, even with the predicted protein structure. In this study, we present DisDock, a deep learning method for predicting protein-metal docking. DisDock takes distogram of randomly initialized protein-ligand configuration as input and outputs the distogram of the predicted binding complex. It combines the U-net architecture with self-attention modules to enhance model performance. Taking inspiration from the physical principle that atoms in closer proximity display a stronger mutual attraction, this predictor capitalizes on geometric information to uncover latent characteristics indicative of atom interactions. To train our model, we employ a high-quality metalloprotein dataset sourced from the Mother of All Databases (MOAD). Experimental results demonstrate that our approach outperforms other existing methods in prediction accuracy for various types of metal ions.
Collapse
Affiliation(s)
- Menghan Lin
- Department of Statistics, Florida State University, Tallahassee, Florida, USA
| | - Keqiao Li
- Department of Statistics, Florida State University, Tallahassee, Florida, USA
| | - Yuan Zhang
- Department of Statistics, Florida State University, Tallahassee, Florida, USA
| | - Feng Pan
- Department of Statistics, Florida State University, Tallahassee, Florida, USA
| | - Wei Wu
- Department of Statistics, Florida State University, Tallahassee, Florida, USA
| | - Jinfeng Zhang
- Department of Statistics, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
43
|
Yao L, Huang Q, Wang H, Feng T, Yu C, Xie K, Liu H, Kang W, Sun M, Yue H. Unlocking novel biopeptides hidden in Camellia seed cake fermented by Bacillus subtilis through in silico and cellular model approaches. Food Chem 2025; 476:143342. [PMID: 39978006 DOI: 10.1016/j.foodchem.2025.143342] [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: 10/11/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025]
Abstract
In this study, Bacillus subtilis was used to ferment the CSC and produce hydrolysates (CSCH), from which novel bioactive peptides were identified. The ultrafiltration fraction of CSCH under 3 kDa (CSCH-3) revealed the most efficient in vitro antioxidant and anti-tyrosinase activity. The peptide profile of CSCH-3 was further characterized using LC-MS/MS, and novel biopeptides were screened through in silico analysis and molecular docking methods. Four peptides (LPFR, WGFKPK, PFDLR, and FPGEL) were recognized as the most promising antioxidant and anti-tyrosinase peptides based on their better binding affinities (< 5 kcal/mol) with the tested receptors. Cell antioxidant assay revealed that the four peptides exhibited significant (P < 0.05) antioxidant activity against AAPH-induced oxidative damage. Meanwhile, B16F10 cell model tests revealed that tyrosinase activity was significantly (P < 0.05) inhibited by LPFR (44.62 %), WGFKPK (32.12 %), PFDLR (34.06 %), and FPGEL (33.66 %) compared to the control. The docking results suggested that the four peptides were tightly bound to antioxidant related receptors (DPPH, ABTS, CAT, SOD, and Keap1) and tyrosinase, suggesting that each peptide could exhibit multiple bioactivities via various structure-activity linkages.
Collapse
Affiliation(s)
- Lingyun Yao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Qingru Huang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Huatian Wang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Tao Feng
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Chuang Yu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Kun Xie
- Bio-Tide Research Institute of GuangDong Co., Ltd, Guangzhou 510806, China
| | - Hao Liu
- Bio-Tide Research Institute of GuangDong Co., Ltd, Guangzhou 510806, China
| | - Wencui Kang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Min Sun
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Heng Yue
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China.
| |
Collapse
|
44
|
Chatziefthymiou SD, Kuzikov M, Afandi S, Kovacs G, Srivastava S, Zaliani A, Gruzinov A, Pompidor G, Lunelli M, Ahmed GR, Labahn J, Hakanpää J, Windshügel B, Kolbe M. Identification, validation, and characterization of approved and investigational drugs interfering with the SARS-CoV-2 endoribonuclease Nsp15. Protein Sci 2025; 34:e70156. [PMID: 40371758 PMCID: PMC12079475 DOI: 10.1002/pro.70156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025]
Abstract
Since the emergence of SARS-CoV-2 at the end of 2019, the virus has caused significant global health and economic disruptions. Despite the rapid development of antiviral vaccines and some approved treatments such as remdesivir and paxlovid, effective antiviral pharmacological treatments for COVID-19 patients remain limited. This study explores Nsp15, a 3'-uridylate-specific RNA endonuclease, which has a critical role in immune system evasion and hence in escaping the innate immune sensors. We conducted a comprehensive drug repurposing screen and identified 44 compounds that showed more than 55% inhibition of Nsp15 activity in a real-time fluorescence assay. A validation pipeline was employed to exclude unspecific interactions, and dose-response assays confirmed 29 compounds with an IC50 below 10 μM. Structural studies, including molecular docking and x-ray crystallography, revealed key interactions of identified inhibitors, such as TAS-103 and YM-155, with the Nsp15 active site and other critical regions. Our findings show that the identified compounds, particularly those retaining potency under different assay conditions, could serve as promising hits for developing Nsp15 inhibitors. Additionally, the study emphasizes the potential of combination therapies targeting multiple viral processes to enhance treatment efficacy and reduce the risk of drug resistance. This research contributes to the ongoing efforts to develop effective antiviral therapies for SARS-CoV-2 and possibly other coronaviruses.
Collapse
Affiliation(s)
- Spyros D. Chatziefthymiou
- Photon Science, DESYHamburgGermany
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Maria Kuzikov
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPortHamburgGermany
- School of Science, Constructor UniversityBremenGermany
| | - Sara Afandi
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Greta Kovacs
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Sukrit Srivastava
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPortHamburgGermany
| | | | | | - Michele Lunelli
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Golam Rizvee Ahmed
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Jörg Labahn
- Forschungszentrum Jülich, Institute of Complex Systems (ICS‐6)JülichGermany
| | | | - Björn Windshügel
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPortHamburgGermany
- School of Science, Constructor UniversityBremenGermany
| | - Michael Kolbe
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
- Faculty of Mathematics, Informatics and Natural SciencesUniversität HamburgHamburgGermany
| |
Collapse
|
45
|
Shen J, Wu SY, Lin P, Jiang X, Hou Y. Identification and optimization of volatile organic compounds to enhance bait attractiveness for red imported fire ants (Solenopsis invicta Buren). PEST MANAGEMENT SCIENCE 2025; 81:3240-3249. [PMID: 39906913 DOI: 10.1002/ps.8696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND The red imported fire ant (RIFA, Solenopsis invicta), a highly destructive invasive pest, has rapidly spread through human trade, posing significant threats to agricultural and forest ecosystems. Due to its preference for high-fat and high-protein foods, ham sausage is commonly used as bait to monitor RIFA populations in invaded areas. However, the presence of volatile organic compounds (VOCs) in such baits may affect their effectiveness because VOCs can act as either attractants or repellents. Identifying VOCs that specifically attract RIFA is essential to improve bait efficacy. RESULTS This study aimed to identify attractant compounds within bait VOCs for RIFA, leveraging the highly-expressed antennal odorant-binding protein 1 (OBP1) and reverse chemical ecology approach. Additionally, we examined the effects of mixtures of these attractants on RIFA behavior. Our findings revealed that anethole, 1S-(-)-β-pinene, and β-caryophyllene individually attracted RIFA at 0.1 μg/μL. Notably, a combination of anethole and 1S-(-)-β-pinene enhanced behavioral activity more than individual compounds, suggesting synergistic effects. Conversely, the addition of β-caryophyllene to anethole significantly reduced RIFA activity. These results provide a theoretical basis for developing behavioral regulators targeting RIFA. CONCLUSION This study demonstrates that the integration of OBP-based in vitro assays with computational simulations can effectively identify behaviorally active compounds for RIFA. Additionally, it clarifies the optimal ratios of active VOCs in baits, offering valuable theoretical guidance for enhancing RIFA population monitoring efforts. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiacheng Shen
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Sheng-Yen Wu
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Peng Lin
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xinyi Jiang
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Youming Hou
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
46
|
Wang X, Shi B, Xia C, Hou M, Wang J, Tian A, Shi C, Ma C. Poly-L-lysine functionalized silica membrane-enhanced colorimetric loop-mediated isothermal amplification for sensitive and rapid detection of Vibrio parahaemolyticus. Talanta 2025; 288:127744. [PMID: 39961248 DOI: 10.1016/j.talanta.2025.127744] [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: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 03/05/2025]
Abstract
Traditional detection of foodborne pathogen relies on advanced analyzers, which is inadequate for the rapid control of infections, particularly in resource-limited regions, highlighting the necessity of developing detection systems for point-of-care testing (POCT). Herein, taking Vibrio parahaemolyticus as a detecting target, we reported poly-L-lysine functionalized silica membrane (PL-SM) based loop-mediated isothermal amplification (pLAMP) platform for sensitive on-site detection. This platform utilized PL-SM for DNA capture driven by the electrostatic attraction between protonated amine groups of poly-L-lysine and negatively charged phosphate groups of DNA, followed by introducing a colorimetric indicator calcein for LAMP amplification. After optimization, the colorimetric mode of pLAMP allowed the screening of V. parahaemolyticus with the visual limit of detection (vLOD) of 1 CFU/mL in 50 min, 1000-fold lower than methods based on commercial kits. Validation was performed using 174 seafoods, which was 97 % concordant to those of real-time PCR. Furthermore, an image processing approach was developed based on the analysis of the RGB under UV light. Paired with a smartphone, the objective analytical method could be readily conducted in the field. Thus, we propose a sensitive and visual detection platform, which may play a crucial role in improving testing efficiency and accuracy in food safety, medical diagnostics, and environmental monitoring.
Collapse
Affiliation(s)
- Xiujuan Wang
- Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Binghui Shi
- Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Cengceng Xia
- Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Mengnan Hou
- Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Jingying Wang
- Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Anning Tian
- Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Chao Shi
- Qingdao Nucleic Acid Rapid Testing International Science and Technology Cooperation Base, College of Life Sciences, Department of Pathogenic Biology, School of Basic Medicine, and Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, PR China
| | - Cuiping Ma
- Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| |
Collapse
|
47
|
Feng S, Gui J, Qin B, Ye J, Zhao Q, Guo A, Sang M, Sun X. Resveratrol Inhibits VDAC1-Mediated Mitochondrial Dysfunction to Mitigate Pathological Progression in Parkinson's Disease Model. Mol Neurobiol 2025; 62:6636-6654. [PMID: 38819635 DOI: 10.1007/s12035-024-04234-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
An increase in α-synuclein (α-syn) levels and mutations in proteins associated with mitochondria contribute to the development of familial Parkinson's disease (PD); however, the involvement of α-syn and mitochondria in idiopathic PD remains incompletely understood. The voltage-dependent anion channel I (VDAC1) protein, which serves as a crucial regulator of mitochondrial function and a gatekeeper, plays a pivotal role in governing cellular destiny through the control of ion and respiratory metabolite flux. The ability of resveratrol (RES), which is a potent phytoalexin with antioxidant and anti-inflammatory properties, to regulate VDAC1 in PD is unknown. The objective of this study was to evaluate the role of VDAC1 in the pathological process of PD and to explore the mechanism by which resveratrol protects dopaminergic neurons by regulating VDAC1 to maintain the mitochondrial permeability transition pore (mPTP) and calcium ion balance. The effects of RES on the motor and cognitive abilities of A53T mice were evaluated by using small animal behavioral tests. Various techniques, including immunofluorescence staining, transmission electron microscopy, enzyme-linked immunoadsorption, quantitative polymerase chain reaction (PCR), and Western blotting, among others, were employed to assess the therapeutic impact of RES on neuropathy associated with PD and its potential in regulating mitochondrial VDAC1. The findings showed that RES significantly improved motor and cognitive dysfunction and restored mitochondrial function, thus reducing oxidative stress levels in A53T mice. A significant positive correlation was observed between the protein expression level of VDAC1 and mitochondrial α-syn expression, as well as disease progression, whereas no such correlation was found in VDAC2 and VDAC3. Administration of RES resulted in a significant decrease in the protein expression of VDAC1 and in the protein expression of α-syn both in vivo and in vitro. In addition, we found that RES prevents excessive opening of the mPTP in dopaminergic neurons. This may prevent the abnormal aggregation of α-syn in mitochondria and the release of mitochondrial apoptosis signals. Furthermore, the activation of VDAC1 reversed the resveratrol-induced decrease in the accumulation of α-syn in the mitochondria. These findings highlight the potential of VDAC1 as a therapeutic target for PD and identify the mechanism by which resveratrol alleviates PD-related pathology by modulating mitochondrial VDAC1.
Collapse
Affiliation(s)
- Shenglan Feng
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Jianjun Gui
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Bingqing Qin
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Junjie Ye
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
- Department of Clinical Laboratory, Wuhan Asia Heart Hospital, Wuhan, 430022, Hubei, China
| | - Qiang Zhao
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Ai Guo
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Ming Sang
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China.
| | - Xiaodong Sun
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
48
|
Rashid MHO, Ezzikouri S, Soliman AM, Akter L, Momohara K, Hifumi T, Miyoshi N, Hishiki T, Abdel-Moneim AS, Kohara M, Tsukiyama-Kohara K. Drug repositioning: Identification of potent inhibitors of NS3 protease and NS5 RdRp for control of DENV infection. Biomed Pharmacother 2025; 187:118104. [PMID: 40300391 DOI: 10.1016/j.biopha.2025.118104] [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: 01/29/2025] [Revised: 04/13/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
Dengue virus (DENV) threatens global health; specific antiviral drugs are required to combat it. Such anti-DENV therapeutics can be rapidly developed by repositioning the drugs approved for other indications. This study investigated six medications of different classes drawn from a library of molecules. In silico analyses were performed to determine potential binding affinity for the DENV non-structural protein NS3 protease and NS5 RNA-dependent RNA polymerase (RdRp). Of the six candidates, galidesivir and tadalafil showed the highest binding affinities for the DENV NS3 protease and NS5 RdRp, with tadalafil demonstrating the highest binding affinity. Galidesivir and tadalafil substantially suppressed viral replication in DENV replicon cells without inducing cytotoxicity and showed half-maximal inhibitory concentrations of 10 μM and 2.56 μM, respectively. Both galidesivir and tadalafil effectively suppress DENV infection in human hepatoma and baby hamster kidney cells, and tadalafil demonstrates protease-inhibitory activity. In an AG129 mouse model of DENV infection, both galidesivir and tadalafil reduced viral loads in the serum, with tadalafil producing a notable reduction by day four. Both drugs markedly suppressed DENV replication in the hepatic tissue. Histopathologically, both galidesivir- and tadalafil-treated mice showed alleviation of DENV-induced lesions in the spleen and liver, indicating the potential therapeutic effects of these drugs. These findings highlight the potential of repositioning galidesivir and tadalafil as effective anti-DENV therapies with low cytotoxicity, meeting the urgent global need for new therapeutic agents against this pathogen.
Collapse
Affiliation(s)
- Md Haroon Or Rashid
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Ahmed M Soliman
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan; Biotechnology department, Animal Health Research Institute, Agricultural Research Center, Dokki, Giza 12618, Egypt
| | - Lipi Akter
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Kenki Momohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Tatsuro Hifumi
- Department of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Noriaki Miyoshi
- Department of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Takayuki Hishiki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | | | - Michinori Kohara
- Department of Microbiology and Cell Biology, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
49
|
Mondal A, Dutta S. Design and development of a bright NIR fluorescent probe for selective HSA detection in human blood serum and urine. Bioorg Chem 2025; 159:108356. [PMID: 40086185 DOI: 10.1016/j.bioorg.2025.108356] [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: 01/14/2025] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Human serum albumin (HSA), an important human blood protein, plays a critical role in maintaining osmotic pressure and facilitating the transport of various substances. Abnormal HSA levels are associated with diseases like kidney disease, heart problems, diabetes, and liver damage, necessitating the development of accurate methods for HSA detection. This paper describes the design, synthesis, and evaluation of four BODIPY-based near-infrared (NIR) fluorescent probes (BD1-BD4) for the selective detection of HSA. Among the synthesized probes, BD1 demonstrated exceptional sensitivity and specificity, exhibiting a 147-fold fluorescence enhancement at 660 nm (λex = 600 nm) with a Stokes shift of 60 nm. The probe achieved a low detection limit of 9.5 nM, enabling the effective quantification of HSA in complex biological samples such as human blood serum and artificial urine. Competitive binding studies using ibuprofen confirmed that BD1 binds selectively to binding site II of HSA, which was further supported by a molecular docking study. Additionally, BD1 demonstrated HSA detection with a high recovery rate in artificial urine.
Collapse
Affiliation(s)
- Arindam Mondal
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India
| | - Subrata Dutta
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India.
| |
Collapse
|
50
|
Marunouchi T, Kyono M, Kikuchi N, Tanonaka K. Gemfibrozil mitigates caspase-11-driven myocardial pyroptosis in ischemia/reperfusion injury in mice. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 12:100292. [PMID: 40134584 PMCID: PMC11932663 DOI: 10.1016/j.jmccpl.2025.100292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025]
Abstract
The size of the infarct area following acute myocardial infarction (AMI) is a critical prognostic factor. Caspase-11-dependent pyroptosis has been implicated as a key mechanism driving cardiomyocyte death after AMI. However, no therapeutic agents have been developed to inhibit myocardial cell death by targeting caspase-11. This study investigates the effects of gemfibrozil, a potential caspase-11 inhibitor, on ischemia/reperfusion-induced myocardial pyroptosis in mice. To model AMI, the left coronary artery of C57BL/6 N mice was ligated for 1 h, followed by reperfusion. Levels of cleaved caspase-11 and the N-terminal fragment of gasdermin D (GSDMD-N) in ischemic myocardial tissue increased progressively over time after ischemia/reperfusion. Gemfibrozil treatment during reperfusion significantly attenuated these increases in cleaved caspase-11 and GSDMD-N levels. Moreover, gemfibrozil reduced the extent of myocardial infarct size during reperfusion. In cultured cardiomyocytes isolated from adult mice, hypoxia/reoxygenation-induced increases in caspase-11 and GSDMD cleavage were similarly mitigated by gemfibrozil, which concurrently prevented necrotic cell death. These findings demonstrate the involvement of caspase-11-dependent pyroptosis in myocardial cell death following ischemia/reperfusion and suggest that gemfibrozil holds promise as a therapeutic agent for reducing myocardial infarct size after AMI.
Collapse
Affiliation(s)
- Tetsuro Marunouchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Mayu Kyono
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Naoko Kikuchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Kouichi Tanonaka
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| |
Collapse
|