1
|
Salamah M, Sipos B, Schelz Z, Zupkó I, Kiricsi Á, Szalenkó-Tőkés Á, Rovó L, Katona G, Balogh GT, Csóka I. Development, in vitro and ex vivo characterization of lamotrigine-loaded bovine serum albumin nanoparticles using QbD approach. Drug Deliv 2025; 32:2460693. [PMID: 39901331 PMCID: PMC11795762 DOI: 10.1080/10717544.2025.2460693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/26/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025] Open
Abstract
The present study aimed to prepare and optimize lamotrigine-loaded bovine serum albumin nanoparticles (LAM-NP) using the Quality by Design (QbD) approach and to investigate both the in vitro and ex vivo effects of different cross-linking agents glutaraldehyde (GLUT), glucose (GLUC) and 1-(3-dimethylaminutesopropyl)-3-ethylcarbodiimide hydrochloride (EDC) on intranasal applicability. Cross-linked LAM-NP from EDC (NP-EDC-1) showed the lowest Z-average value (163.7 ± 1.9 nm) and drug encapsulation efficacy (EE%) of 97.31 ± 0.17%. The drug release of GLUC cross-linked LAM-NP (NP-GLUC-9), glutaraldehyde cross-linked LAM-NP (NP-GLUT-2), and NP-EDC-1 at blood circulation conditions was higher than the initial LAM. The results of the blood-brain barrier parallel artificial membrane permeability assay (BBB-PAMPA) showed an increase in the permeability of LAM through the BBB with NP-GLUC-9 and an increase in flux with all selected formulations. The ex vivo study showed that LAM diffusion from the selected formulations through the human nasal mucosa was higher than in case of initial LAM. The cytotoxicity study indicated that BSA-NP reduced LAM toxicity, and GLUC 9 mM and EDC 1 mg could be alternative cross-linking agents to avoid GLUT 2% v/v toxicity. Furthermore, permeability through Caco-2 cells showed that nasal epithelial transport/absorption of LAM was improved by using BSA-NPs. The use of BSA-NP may be a promising approach to enhance the solubility, permeability through BBB and decrease the frequency of dosing and adverse effects of LAM.
Collapse
Affiliation(s)
- Maryana Salamah
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Bence Sipos
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Schelz
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Ágnes Kiricsi
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, Szeged, Hungary
| | - Ágnes Szalenkó-Tőkés
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, Szeged, Hungary
| | - László Rovó
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, Szeged, Hungary
| | - Gábor Katona
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - György Tibor Balogh
- Department of Pharmaceutical Chemistry, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Ildikó Csóka
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| |
Collapse
|
2
|
Bruckschlegel C, Fleischmann V, Gajovic-Eichelmann N, Wongkaew N. Non-enzymatic electrochemical sensors for point-of-care testing: Current status, challenges, and future prospects. Talanta 2025; 291:127850. [PMID: 40049001 DOI: 10.1016/j.talanta.2025.127850] [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: 01/30/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Current electrochemical sensors in point-of-care (POC) testing devices rely mainly on enzyme-based sensors owing to superior sensitivity and selectivity. Nevertheless, the poor stability, high reagent cost, complex fabrication methods and requirement of specific operational conditions make their adaptability in real-world applications unfavorable. Non-enzymatic electrochemical sensors are thus developed as they are more robust and cost-effective strategies. The advancement in material science and nanotechnology enables the development of novel non-enzymatic electrodes with favorable analytical performance. However, the developments are yet far from being adopted as viable products. This review therefore aims to gain insight into the field and evaluate the current progress and challenges to eventually propose future research directions. Here, fabrication strategies based on traditional and emerging technology are discussed in the light of analytical performance and cost-effectiveness. Moreover, the discussion is given on the pros and cons of non-enzymatic sensors when they are employed with various kinds of sample matrices, i.e., clinical and non-clinical samples, which must be taken into consideration for sensor development. Furthermore, molecular imprinting technology in tackling the selectivity issue is introduced and current progress is provided. Finally, the promising strategies from literature for solving the remaining challenges are included which could facilitate further development of robust POC testing devices based non-enzymatic sensors. We believe that once researchers and technology developers have reached the point where most problems are solved, the non-enzymatic sensors are going to be the robust choice for POC testing in clinical diagnostic, ensuring food safety, monitoring contaminants in environment, and bioprocess control.
Collapse
Affiliation(s)
- Christoph Bruckschlegel
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany
| | - Vivien Fleischmann
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany
| | - Nenad Gajovic-Eichelmann
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses, Am Muehlenberg 13, 14476, Potsdam, Germany
| | - Nongnoot Wongkaew
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany.
| |
Collapse
|
3
|
Casanova MC, Zhao Y, Hutter S, Fil M, Do MH, Benech A, Muller TB, Curti C, Médebielle M, Vanelle P, Azas N, Broggi J. N,N'-bridged heterocyclic bis-iminium salts as potent antimalarial agents against multi-resistant Plasmodium falciparum. Eur J Med Chem 2025; 292:117633. [PMID: 40319575 DOI: 10.1016/j.ejmech.2025.117633] [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: 03/28/2025] [Accepted: 04/12/2025] [Indexed: 05/07/2025]
Abstract
The emergence of Plasmodium resistance to past and newly introduced antimalarial drugs reinforces the need for new antimalarial agents with innovative mechanisms of action. In this regard, the search for new redox-cycling compounds might offer an interesting approach to treat malaria since P. falciparum is sensitive to oxidative stress. Based on our experience in the design of powerful organic reducers, we undertook here the synthesis and evaluation of different families of N-heterocyclic iminium salts that could provide more selective redox-cycling candidates against the K1 multi-resistant Plasmodium falciparum strain (PfK1). Bis-aminopyridinium salts emerged as the most promising candidates, exhibiting nanomolar antiplasmodial activity comparable to that of methylene blue. The biological study also underlined the positive impact on the in vitro activity of electron-donating groups and bis-salt forms, likely due to simultaneous bivalent interactions with the target. Our investigations revealed differences in the mechanism of action according to the heterocycle nature, highlighting an original mechanism for bis-aminopyridinium derivatives, different from that of chloroquine.
Collapse
Affiliation(s)
- M C Casanova
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France; Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, 13005, Marseille, France
| | - Y Zhao
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117, Jinan, China
| | - S Hutter
- Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, 13005, Marseille, France
| | - M Fil
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France
| | - M H Do
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France; Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, 13005, Marseille, France
| | - A Benech
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France
| | - T-B Muller
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France
| | - C Curti
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France; Assistance Publique - Hôpitaux de Marseille (AP-HM), Service central de la qualité et de l'information pharmaceutiques (SCQIP), Pôle pharmacie, Hôpital de la Conception, 13005, Marseille, France
| | - M Médebielle
- Univ Lyon 1, CNRS, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires ICBMS-UMR 5246, 69622, Villeurbanne, France
| | - P Vanelle
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France; Assistance Publique - Hôpitaux de Marseille (AP-HM), Service central de la qualité et de l'information pharmaceutiques (SCQIP), Pôle pharmacie, Hôpital de la Conception, 13005, Marseille, France.
| | - N Azas
- Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, 13005, Marseille, France.
| | - J Broggi
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire ICR - UMR 7273, Faculté de Pharmacie, 13005, Marseille, France.
| |
Collapse
|
4
|
Aishwarya J, Das RP, Barik A, Kunwar A. Synthetic selenomelanin nanoparticles radio-sensitize non-melanocytic lung cancer (A549) cells by promoting G2/M arrest. Colloids Surf B Biointerfaces 2025; 252:114680. [PMID: 40245569 DOI: 10.1016/j.colsurfb.2025.114680] [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: 12/27/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Recent studies have postulated the natural existence of selenomelanin and its role in the radio-protection of healthy cells. The present study aimed to understand its radio-modulatory activity in non-melanocytic cancerous (A549) cells of lung origin. Briefly, selenomelanin was synthesized under laboratory conditions following the previously reported methodology. The various spectroscopic (electron paramagnetic resonance, X-ray photoelectron spectroscopy, atomic absorption spectroscopy, transmission electron microscopy and dynamic light scattering) analyses confirmed the formation of selenomelanin nanoparticles. The short-term (72 h) and long-term (14 days) toxicity profiling of selenomelanin by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) and clonogenic assays respectively revealed its half maximal inhibitory concentrations (IC50) of 72.03 ± 7.13 μg/ml and 0.85 ± 0.16 μg/ml respectively in A549 cells and of 81.56 ± 1.63 μg/ml and > 5 μg/ml respectively in healthy lung fibroblast (WI26) cells. Further, pre-treatment of selenomelanin (at concentrations non-toxic for WI26 cells) selectively augmented the radiosensitivity of A549 cells. Finally, mechanistic investigations in A549 cells revealed that selenomelanin increased the levels of reactive oxygen species, DNA damage and modulated the phospho-levels of CHK1 and CHK2 (effectors of cell cycle arrest) in the irradiated cells to favour G2/M arrest followed by cleavage of caspase 3 (effector of apoptosis). Together, the present study proposes the novel application of selenomelanin as a radiosensitizer to enhance the efficacy of radiotherapy in cancerous cells of lung origin.
Collapse
Affiliation(s)
- J Aishwarya
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay 400085, India; Homi Bhabha National Institute, Anushaktinagar 400094, India
| | - Ram Pada Das
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay 400085, India
| | - Atanu Barik
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay 400085, India; Homi Bhabha National Institute, Anushaktinagar 400094, India.
| | - Amit Kunwar
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay 400085, India; Homi Bhabha National Institute, Anushaktinagar 400094, India.
| |
Collapse
|
5
|
Khongmawloh E, Koner D, Snaitang R, Saha N. Nitric oxide-mediated induction of superoxide dismutase and catalase genes, and altered expression of glutathione-dependent genes to defend against the TiO 2 NP-induced oxidative stress in primary hepatocytes of air-breathing catfish, Clarias magur. Comp Biochem Physiol C Toxicol Pharmacol 2025; 294:110188. [PMID: 40068795 DOI: 10.1016/j.cbpc.2025.110188] [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: 09/19/2024] [Revised: 02/21/2025] [Accepted: 03/07/2025] [Indexed: 05/16/2025]
Abstract
The main objectives of the present investigation were to elucidate the possible induction of antioxidant genes under the TiO2 NP-induced oxidative stress and the potential involvement of endogenously produced nitric oxide (NO) in its antioxidant strategies in primary hepatocytes of air-breathing magur catfish (Clarias magur). As expected, exposure to TiO2 NPs led to (1) more ROS production as evidenced by a sharp rise of hydrogen peroxide (H2O2) and malonaldehyde (MDA) associated with cellular damage as evidenced by the increase of lactate dehydrogenase (LDH) leakage from hepatocytes, (2) induction of superoxide dismutase (SOD), catalase (CAT), followed by induction of different glutathione-related genes such as glutathione peroxidase (GPx), glutathione-S-transferase (GST), and thioredoxin glutathione reductase (TGR) with the induction of activities of corresponding enzymes, and (3) more production of NO associated with induction of inducible nitric oxide synthase (iNOS) activity and its corresponding gene. However, inhibition of NO production in primary hepatocytes using certain inhibitors in the presence of TiO2 NPs, resulted in (1) more generation of H2O2 and MDA, (2) inhibition of SOD and CAT genes expression in primary hepatocytes with more leakage of LDH leakage into the culture media. Thus, it can be contemplated that stimulation NO production plays a vital role in inducing the SOD-CAT system to handle the problems associated with enhanced TiO2 NP-induced ROS production and subsequent oxidative stress in magur catfish as a unique adaptational strategy. However, the NO-mediated induction of glutathione-related antioxidant genes under TiO2 NP-induced oxidative stress is yet to be established.
Collapse
Affiliation(s)
- Elvis Khongmawloh
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| | - Debaprasad Koner
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| | - Revelbornstar Snaitang
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| | - Nirmalendu Saha
- Biochemical Adaptation Laboratory, Department of Zoology, North-Eastern Hill University, Shillong 793022, India.
| |
Collapse
|
6
|
Baidya AT, Dante D, Das B, Wang L, Darreh-Shori T, Kumar R. Discovery and characterization of novel pyridone and furan substituted ligands of choline acetyltransferase. Eur J Pharmacol 2025; 998:177638. [PMID: 40252901 DOI: 10.1016/j.ejphar.2025.177638] [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: 01/17/2025] [Revised: 04/16/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
The key to the management of two devastating diseases, namely Alzheimer's Disease (AD) and Amyotrophic Lateral Sclerosis (ALS) lies in an early diagnosis, which is difficult due to its multifactorial nature. However, a common hallmark of AD and ALS is degeneration of cholinergic system. Choline acetyltransferase (ChAT) has been proposed as a potential target for development of cholinergic-specific biomarker. However, lack of selective, potent, brain permeable molecular probes of ChAT hinder development of ChAT biomarkers. In this study, we have successfully utilised structure-based virtual screening approach and identified two ChAT inhibitors from a database of 1.4 million compounds. The compounds were then subjected to rigorous in vitro characterization. Compound V6 showed Ki value of 11 μM and IC50 value of 21.73 μM, while V15 showed Ki and IC50 values of 4.5 and 9.42 μM, respectively for ChAT enzyme. V6 and V15 showed good solubility of 0.21 mg/mL and 0.17 mg/mL respectively and cytotoxicity analysis indicated no toxicity. We also performed a 200 ns molecular dynamics simulation, which revealed the intricate interaction dynamics for V6 and V15 with ChAT binding pocket. Moreover, the Tanimoto similarity analysis indicated the novelty and structural diversity of the hits. In conclusion, these validated hits provide a platform to develop potent, selective, blood-brain barrier permeable small molecules as chemical probes of ChAT or as Positron Emission Tomography tracer for early diagnosis and/or in vivo monitoring of the effect of new therapeutic candidates in spectrum of neurodegenerative disorders, in which cholinergic deficit is one of the hallmarks.
Collapse
Affiliation(s)
- Anurag Tk Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, U.P., India
| | - Davide Dante
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Bhanuranjan Das
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, U.P., India
| | - Lisha Wang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
| | - Taher Darreh-Shori
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, U.P., India.
| |
Collapse
|
7
|
de Moura CVR, Leite DBC, Muniz EC, Mendes AN, Filgueiras LA, de Abreu Júnior AR, Gonçalves JCR, Marques KKG, Sobral MV, Araujo PM, Dos Santos Rizzo M, de Amorim Carvalho FA, de Moraes Alves MM, Carvalho ALM, do Nascimento MO. Advanced amphotericin-B gel formulation: An efficient approach to combat cutaneous leishmaniasis. BIOMATERIALS ADVANCES 2025; 172:214220. [PMID: 40023082 DOI: 10.1016/j.bioadv.2025.214220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 03/04/2025]
Abstract
Several drug release studies are continuously carried out to minimize pharmacological limitations, such as insolubility in water and gastrointestinal irritability. This study focused on the development of gels for the administration of amphotericin-B (AmB) for the treatment of cutaneous leishmaniasis. Two gels were prepared from copolymers (polyglycerol and ε-caprolactone) incorporated with AmB. The gels were characterized by FTIR, NMR, TG, and DSC techniques. The incorporation of AmB into the copolymers showed that the medicine remained in the monomeric form, which is the least toxic. The AmB loading presented values of about 31 % (w:w) for TMP-HPG-PCL and for GLY-HPG-PCL, while the encapsulation efficiency was 93 % for both copolymers. According to the release profile, it is observed that, after 48 h, the percentage of AmB released for pure amphotericin, TMP-HPG-PCL-AmB, and GLY-HPG-PCL-AmB were 100 % ± 6 %, 100 % ± 1.5 %, and 47 % ± 13, respectively. Hemolytic study, Cytotoxic Evaluation showed that TMP-HPG-PCL and GLY-HPG-PCL were not toxic to both cell lines (HaCat and MCF-7). These results suggest that the copolymers are safe for in vivo applications being able to act as a drug carrier that have low water solubility corroborating their purpose of polymeric support in the transport of drugs. TMP-HPG-PCL-POL-AmB and for GLY-HPG-PCL-HPG-POL-AmB were used in female mice (BALB/c) infected with Leishmania major foremost for 40 days, and it was found that in animals treated with the gel/copolymer/AmB, there was a reduction in the number of parasites of around 70 %. Histopathological studies showed the presence of small intralobular granulomas and moderate Kupffer cell hypertrophy/hyperplasia. The results show that the TMP-HPG-PCL-POL-AmB and for GLY-HPG-PCL-HPG-POL-AmB efficiently combats cutaneous leishmaniasis.
Collapse
Affiliation(s)
| | - Diego Botelho Campelo Leite
- Department of Chemistry, Federal University of Piauí, Teresina, PI, Brazil; Federal Institute of Maranhão, São João dos Patos, MA, Brazil
| | - Edvani Curti Muniz
- Department of Chemistry, Federal University of Piauí, Teresina, PI, Brazil
| | | | - Lívia Alves Filgueiras
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina, PI, Brazil
| | | | - Juan Carlos Ramos Gonçalves
- Department of Pharmaceutical Sciences, Federal University of Paraíba, 58051-900 João Pessoa, PB, Brazil; Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, 58051-900 João Pessoa, PB, Brazil
| | - Karinne Kelly Gadelha Marques
- Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, 58051-900 João Pessoa, PB, Brazil
| | - Marianna Vieira Sobral
- Department of Pharmaceutical Sciences, Federal University of Paraíba, 58051-900 João Pessoa, PB, Brazil; Post Graduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, 58051-900 João Pessoa, PB, Brazil
| | - Paulo Monteiro Araujo
- Graduate Program in Pharmaceutical Sciences, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, PI, Brazil
| | - Marcia Dos Santos Rizzo
- Graduate Program in Pharmaceutical Sciences, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, PI, Brazil; Interdisciplinary Laboratory for Advanced Materials, Federal University of Piaui, Teresina, Brazil
| | - Fernando Aecio de Amorim Carvalho
- Antileishmanial Activity Laboratory, Medicinal Plants Research Center, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, PI, Brazil
| | - Michel Mualem de Moraes Alves
- Antileishmanial Activity Laboratory, Medicinal Plants Research Center, Department of Veterinary Morphophysiology - Federal University of Piauí, Teresina, PI, Brazil
| | - André Luis Menezes Carvalho
- Graduate Program in Pharmaceutical Sciences, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, PI, Brazil
| | - Matheus Oliveira do Nascimento
- Graduate Program in Pharmaceutical Sciences, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, PI, Brazil
| |
Collapse
|
8
|
Ilovaisky AI, Scherbakov AM, Miciurov D, Chernoburova EI, Merkulova VM, Bogdanov FB, Salnikova DI, Sorokin DV, Krasil'nikov MA, Bozhenko EI, Zavarzin IV, Terent'ev AO. Secosteroid - 1,3,4-oxadiazole hybrids: Synthesis and evaluation of their activity against hormone-dependent breast cancer cells. J Steroid Biochem Mol Biol 2025; 251:106745. [PMID: 40164235 DOI: 10.1016/j.jsbmb.2025.106745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
This study focused on the synthesis of secosteroids with good antiproliferative properties against hormone-dependent breast cancer. A straightforward and efficient method for synthesizing secosteroid - 1,3,4-oxadiazole hybrids was developed starting from 13α-hydroxy-3-methoxy-13,17-secoestra-1,3,5(10)-trien-17-oic acid hydrazide. The cyclization of hydrazide moiety with CS2 into 1,3,4-oxadiazole-2(3H)-thione fragment followed by sulfur alkylation resulted in the formation of various secosteroid - 2-mercapto-1,3,4-oxadiazole hybrids. These novel compounds were evaluated for their antiproliferative activity against the hormone-dependent human breast cancer cell line MCF-7. Among the synthesized hybrids, compounds 3i, 3o, and 3q displayed notable antiproliferative effects, with IC50 values ranging from 6.5 to 8.9 µM, comparable to the reference drug cisplatin. Furthermore, compound 3i showed minimal toxicity toward non-cancerous hFB-hTERT fibroblasts, indicating high selectivity. Compounds 3o and 3q exhibited antiestrogenic activity. Additionally, their effects on PARP and Bcl-2 suggest a pro-apoptotic mechanism of action. These findings highlight the potential of secosteroidal hybrids as promising candidates for the development of new anti-breast cancer agents targeting ERα and apoptosis pathways.
Collapse
Affiliation(s)
- Alexey I Ilovaisky
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander M Scherbakov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia; Gause Institute of New Antibiotics, Bol'shaya Pirogovskaya ulitsa 11, Moscow 119021, Russia
| | - Dumitru Miciurov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Elena I Chernoburova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Valentina M Merkulova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Fedor B Bogdanov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Diana I Salnikova
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Danila V Sorokin
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Mikhail A Krasil'nikov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Eugene I Bozhenko
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Igor V Zavarzin
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander O Terent'ev
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| |
Collapse
|
9
|
da Cruz ER, Corrêa DS, Miri JM, Bondan da Silva J, de Sousa JT, Farias IV, Reginatto FH, da Silva J, Grivicich I, de Barros Falcão Ferraz A, Picada JN. Mutagenic assessment and toxicological impact of bergenin in a phenolic-enriched extract from Endopleura uchi (Huber) Cuatrec bark, a medicinal plant from the Amazon rainforest. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:495-504. [PMID: 39929780 DOI: 10.1080/15287394.2025.2464920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2025]
Abstract
Endopleura uchi bark traditionally used in folk medicine attributed to its anti-inflammatory properties is due to the presence of bergenin. This study aimed to determine the toxicological parameters associated with exposure to a phenolic-enriched extract of E. uchi bark and bergenin a bioactive byproduct of this compound. The total phenolic and flavonoid contents were determined through spectrometric analyses, while phenolic compounds were identified using ultra-performance liquid chromatography coupled with high-resolution mass spectrometry (UPLC-MS). Antioxidant activity was assessed in vitro using the DPPH assay. Cytotoxicity and genotoxicity were assessed via the MTT assay and comet assay, respectively, whereas mutagenic activity was examined using Salmonella/microsome assay and micronucleus (MN) test. A high content of phenolic (732.22 ± 9.48 mg/g) GAE (gallic acid equivalence) and flavonoid 252.47 ± 5.7 mg/g QE (quercetin) compounds was found in bergenin the phenolic-enriched extract byproduct as well as isomers of gallic acid, epicatechin, isoquercitrin, castalagin, punicalin, and punicalagin. The DPPH value was 23.74 ± 0.45 μg/ml. In MTT assay, the extract exhibited an IC50 of 72.5 ± 2.6 µg/ml. Both extract and bergenin displayed genotoxic activity in L929 fibroblast cells at 50 µg/ml but not mutagenic effects in the Salmonella/microsome assay or MN test. Despite the genotoxic actions, E. uchi bark and bergenin extract did not induce gene or chromosomal mutations, suggesting a low risk of compromising genomic stability. The presence of bioactive compounds such as bergenin and punicalagin in E. uchi bark demonstrates a therapeutic potential of this native tree for treating inflammatory diseases.
Collapse
Affiliation(s)
- Elkejer Ribeiro da Cruz
- Laboratory of Genetic Toxicology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
- Center for Research in Product and Development (CEPPED), Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
- Federal Institute of Education, Science, and Technology of Piauí (IFPI), Teresina, PI, Brazil
| | - Dione Silva Corrêa
- Center for Research in Product and Development (CEPPED), Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | - Jéssica Machado Miri
- Laboratory of Cancer Biology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | | | - Jayne Torres de Sousa
- Laboratory of Genetic Toxicology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | - Ingrid Vicente Farias
- Pharmacognosy Laboratory, Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Flávio Henrique Reginatto
- Pharmacognosy Laboratory, Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Juliana da Silva
- Laboratory of Genetics Toxicology, La Salle University, Canoas, RS, Brazil
| | - Ivana Grivicich
- Laboratory of Cancer Biology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | - Alexandre de Barros Falcão Ferraz
- Pharmacognosy Laboratory, Department of Industrial Pharmaceutical, Health Sciences Center, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | | |
Collapse
|
10
|
Ghorab MM, Soliman AM, Habieb ME, Abdou FY. New acetamide-sulfonamide scaffolds with potential renal radiomodulatory effects: Insights into NF-κB pathway interactions. Bioorg Chem 2025; 160:108439. [PMID: 40209352 DOI: 10.1016/j.bioorg.2025.108439] [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: 02/23/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
With the increased use of radiation in cancer therapy, new effective antioxidants and anti-inflammatory agents are required to alleviate the negative impact caused by irradiation. A set of novel N-substituted-2-((2-oxo-2-((4-sulfamoylphenyl)amino)ethyl)thio) acetamide derivatives 3-14 was synthesized to act as possible radiation mitigators. The synthesized compounds were screened for their anti-inflammatory and antioxidant potential using selective COX-2 inhibitory activity and DPPH assays compared to celecoxib and ascorbic acid, respectively. Compound 9 was the most active in this series with selective COX-2 inhibitory activity (IC50 = 0.373 μM), and free radical scavenging properties (IC50 = 4.89 μM). In vitro and in vivo studies demonstrated that compound 9 exhibited a high safety profile, with low cytotoxicity on normal cells (IC50 > 800 μM) and a median lethal dose (LD50) of 300 mg/kg. The potential renal radiomodulatory effect of the promising candidate was investigated in mice exposed to gamma radiation (6 Gy). Compound 9 successfully reduced radiation-induced oxidative stress, as seen by lower levels of reactive oxygen species (ROS), malondialdehyde (MDA), and enhancement in the levels of reduced Glutathione (GSH) in kidney tissues. Moreover, compound 9 reduced kidney inflammatory markers; Nuclear factor kappa B (NF-κB) and Interleukin 6 (IL-6) in irradiated mice, while lowering serum urea and creatinine levels relative to untreated irradiated group. Compound 9 additionally modified the histological alterations caused by gamma irradiation-induced tubular epithelial cell necrosis. Accordingly, compound 9 can help to reduce the adverse effects of irradiation.
Collapse
Affiliation(s)
- Mostafa M Ghorab
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt..
| | - Aiten M Soliman
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt..
| | - Mahmoud E Habieb
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| | - Fatma Y Abdou
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| |
Collapse
|
11
|
Hameed A, Nawaz I, Alrokayan S, Hussain T, Iqbal J. Synthesis, structure activity relationship and biological evaluation of indole sulfonohydrazide derivatives as antagonists of P2Y1 and P2Y6 receptors. Bioorg Chem 2025; 160:108499. [PMID: 40280013 DOI: 10.1016/j.bioorg.2025.108499] [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: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
The P2Y receptors play a significant role in regulating various physiological functions, such as neurotransmission and inflammatory response. They are also considered promising therapeutic targets for the treatment and prevention of conditions like neurological disorders, pain, cardiovascular diseases, thrombosis, and cancer. Active research is ongoing to identify antagonists of P2Y receptor. Although extensive research has been conducted on P2Y receptors inhibitors, only a limited number of P2Y receptors antagonists have been identified and approved by regulatory authority. In the current research, new indole sulfonohydrazide derivatives (3a-3 k) were synthesized in good yield. Based on toxicity assays performed on h-1321 N1 astrocytoma cell line, these low molecular weight compound showed a safe toxicity profile. The synthesized derivatives were also screened against tP2Y1 and rP2Y6 receptors using a calcium mobilization assay. The results showed that compounds 3a, 3b, 3 h and 3 k were potent against tP2Y1 with IC50 values of 9.91 ± 1.01 μM, 3.49 ± 0.31 μM, 9.72 ± 0.82 μM, and 6.14 ± 0.17 μM, respectively. Additionally, three compounds, i.e., 3d, 3f, and 3 h, exhibited potency against rP2Y6 with IC50 value of 9.22 ± 1.10 μM, 16.25 ± 0.27 μM, and 1.89 ± 0.11 μM, respectively. Molecular docking study was conducted to support the in vitro analysis, which revealed that the tested compounds showed favorable interaction with the amino acids of the target P2Y1 receptor, including Phe62, Phe66, Leu102, Thr103, Pro105, Ala106, Phe119 and Met123. An in silico pharmacokinetic study was also performed, which revealed that the synthesized compounds met all the criteria for favorable gastrointestinal absorption, indicating potential for oral bioavailability. The stability and reactivity of compounds were determined by using the Guassian09 programme in which the density functional theory (DFT) calculations were performed by using the B3LYP/SVP level.
Collapse
Affiliation(s)
- Abdul Hameed
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan; Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Ismat Nawaz
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Salman Alrokayan
- Research Chair for Biomedical Application of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Tajamul Hussain
- Research Chair for Biomedical Application of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; Center of Excellence in Biotechnology Research, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan; Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan.
| |
Collapse
|
12
|
Zahran SS, El-Gazzar MGM, El-Gazzar MG, Ghorab MM. Novel chloropyridazine sulfonamides as aromatase inhibitors and apoptotic inducers in breast cancer. Bioorg Chem 2025; 160:108470. [PMID: 40239405 DOI: 10.1016/j.bioorg.2025.108470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/06/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
Aromatase catalyzes the rate-limiting and final step in the biosynthesis of estrogen. Inhibitors of this enzyme are effective targeted therapy for breast cancer. Molecular hybridization is a promising strategy in drug discovery that combines two or more biologically active moieties in a single structure. In this work, we aim at combining sulfonamide, chloropyridazine and pyrrole in a single design as potential aromatase inhibitors for breast cancer. The synthesized compounds were subjected to in vitro cytotoxic screening against MCF-7 breast cancer cell line, then were assessed for their ability to inhibit aromatase enzyme. Compound 10 exhibited a promising cytotoxic activity (IC50 1.83 μM) that nearly equal to the reference drug (doxorubicin, IC50 1.94 μM) on MCF-7 cells. Also, compound 10 was the most potent aromatase inhibitor with the lowest IC50 (0.06 μM) compared to letrozole (IC50 0.05 μM). Based on the promising results of compound 10, it was selected to investigate its apoptotic effect that disclosed a marked increase in Bax level to 5.42 folds and down-regulation in Bcl-2 expression to 0.34 folds in MCF-7 cells compared to letrozole. Moreover, compound 10 increased caspase 9 level by 4.84 folds. Also, compound 10 arrested the cell cycle at G1 phase and caused induction of early and late apoptosis in an AnnexinV-FITC assay. Compound 10 had been evaluated to study its synergistic effect with γ-radiation by evaluating the cytotoxicity against MCF-7 after exposure to gamma rays (8 Gy). In addition, compound 10 showed low toxicity against human normal breast (MCF-10 A) cell line. Docking study of compound 10 was performed and showed binding with the key amino acids in aromatase active site.
Collapse
Affiliation(s)
- Sally S Zahran
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| | - Mostafa G M El-Gazzar
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt
| | - Marwa G El-Gazzar
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt.
| | - Mostafa M Ghorab
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo 11787, Egypt.
| |
Collapse
|
13
|
Zhang H, Xu Z, Xu Z, Bian S, Qiao N, Wang X, Zhang M, Guo F, Cui Y, Zheng F, Liu J, Wu D, Xu H. The development of pyridazinone-based andrographolide derivatives as anti-cancer agents with the ability of inhibiting the TFAP4/Wnt/β-catenin signaling pathway. Bioorg Chem 2025; 160:108440. [PMID: 40215943 DOI: 10.1016/j.bioorg.2025.108440] [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: 02/08/2025] [Revised: 03/07/2025] [Accepted: 04/02/2025] [Indexed: 05/04/2025]
Abstract
Emerging evidence indicated that natural andrographolide and its derived compounds could exert anti-cancer effects on a broad range of cancer cells by several mechanisms of actions. However, the potent andrographolide derivatives with novel structures are needed and the comprehensive understanding of the underlying mechanisms of actions are still lacking. In this work, we reported the pyridazinone-based anti-cancer andrographolide derivative A61, which is superior to the widely-used anti-cancer drug 5-FU (around 5-fold more potent), it showed high potency to inhibit the growth and migration of a panel of cancer cells, in which the gastric cancer cells exhibited the highest drug sensitivity. Preliminary anti-cancer mechanistic studies indicated that A61 exerted its anti-gastric cancer effect by inducing cell apoptosis through intrinsic mitochondria-mediated pathways and arresting cell circle at S phase. Further exploration at the molecular level indicated that compound A61 may inhibit the transcriptional activity and nuclear localization of TFAP4 in gastric cancer cells by inhibiting the TFAP4/Wnt/β-catenin signaling pathway. For the first time, the TFAP4/Wnt/β-catenin signaling pathway was found to be responsible for the anti-cancer activity of andrographolide derivative. In addition, A61 was demonstrated to have significantly increased bioavailability in rats compared with andrographolide. This work provides pertinent information for the understanding of the anti-cancer mechanism by this class of compounds.
Collapse
Affiliation(s)
- Hang Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhihao Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhengyu Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Shaopan Bian
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ning Qiao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaodi Wang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mingwei Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Fan Guo
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - You Cui
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Fanyu Zheng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jia Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Di Wu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Haiwei Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
14
|
Campos-Pereira FD, Gonçalves LR, Jardim RVH, Cagnoni LB, Moraes KCM, Marin-Morales MA. Genotoxicity of putrescine and its effects on gene expression in HepG2 cell line. Toxicol In Vitro 2025; 106:106048. [PMID: 40086645 DOI: 10.1016/j.tiv.2025.106048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Decomposing bodies release necro-leachate, a toxic fluid containing harmful compounds such as biogenic amines. This study investigated the genotoxic effects of the different concentrations (0.5, 1.4, 2.3, 3.2 mM) of bioamine putrescine on HepG2 cells using the comet assay, the micronucleus test, and gene expression analysis. The results were compared to negative control and indicated significant DNA damage in the comet assay highlighting tail DNA intensity that exhibited significant differences across all tested concentrations (0.5 = 192 %, 1.4 = 189 %, 2.3 = 208 %, 3.2 = 132 %). The micronucleus test revealed a significant increase in micronuclei for concentrations 0.5 (193 %), 1.4 (229 %), 2.3 (206 %); nuclear buds 3.2 (173 %); chromosomal bridges 3.2 (735 %). Furthermore, genes linked to oxidative stress and DNA damage exhibited statistically significant expression alterations. These findings suggest that putrescine has genotoxic potential in human-derived HepG2 cells, raising concerns about cemetery contaminants' occupational and environmental risks. This study is the first to assess putrescine's toxicity as an environmental pollutant, as previous research has mainly focused on its role in the food sector. These insights highlight the potential threats necro-leachate poses to environmental health, emphasizing the need for further research on cemetery pollution.
Collapse
Affiliation(s)
- Franco Dani Campos-Pereira
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Avenida 24-A, n° 1515, Bela Vista, CEP 13506-900 Rio Claro, São Paulo, Brazil
| | - Letícia Rocha Gonçalves
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Avenida 24-A, n° 1515, Bela Vista, CEP 13506-900 Rio Claro, São Paulo, Brazil
| | - Raquel Vaz Hara Jardim
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Avenida 24-A, n° 1515, Bela Vista, CEP 13506-900 Rio Claro, São Paulo, Brazil
| | - Letícia Bulascochi Cagnoni
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Avenida 24-A, n° 1515, Bela Vista, CEP 13506-900 Rio Claro, São Paulo, Brazil
| | - Karen C M Moraes
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Avenida 24-A, n° 1515, Bela Vista, CEP 13506-900 Rio Claro, São Paulo, Brazil
| | - Maria Aparecida Marin-Morales
- Universidade Estadual Paulista "Júlio de Mesquita Filho" - Campus Rio Claro, Instituto de Biociências, Departamento de Biologia Geral e Aplicada, Avenida 24-A, n° 1515, Bela Vista, CEP 13506-900 Rio Claro, São Paulo, Brazil.
| |
Collapse
|
15
|
Mosallam FM, Shafik MA, Abd Elmawgoud SA, El-Saied MA, Elshimy RM. In vitro and in vivo attenuation of Salmonella resistance using a novel synthesized chloramphenicol magnesium Nano-complex. Microb Pathog 2025; 203:107511. [PMID: 40147554 DOI: 10.1016/j.micpath.2025.107511] [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: 01/01/2025] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
In this study, Chloramphenicol-Mg-Nano-complex (CHL-Mg-NC) was synthesized as a novel antimicrobial agent to attenuate chloramphenicol resistant Salmonella clinical isolates in vitro and in vivo. The CHL-Mg-NC was prepared in presence of gamma radiation and validated by SEM, DLS, Zeta potential, and FTIR, that revealed typical CHL-Mg-NC characteristics. The Phenotypes, biochemical investigations and molecular identification assays defined Salmonella isolates and further detection of invA gene in S. Paratyphi A NCRR-CHR1, S. Enteritidis NCRR-CHR2 and S. Typhimurium NCRR-CHR3 were appraised. In vitro anti-Salmonella efficacy of CHL-Mg-NC was assessed against Salmonella isolates in addition to Typhimurium ATCC 700720. Gamma radiation improved CHL-Mg-NC synthesis in dose-depend manner up to 5 kGy. CHL-Mg-NC showed MIC at a range from 0.156 to 0.625 μg/mL and MBC from 0.3125 to 2.5 μg/mL with MBC/MIC ratio less than or equal to 4. CHL-Mg-NC inhibited biofilm formation in the range of 45.31 %-100 %. It also had bactericidal activity at 2MIC within the low time ranged from 2h to 4h. The in vivo efficacy of CHL-Mg-NC was observed by the reduction in the number of viable Salmonella recovered from feces in infected mice and showed evident improvement in CHL-Mg-NC treated groups.CHL-Mg-NC has no significant cytotoxic effects on normal cells and CC50 is 13.5 μg/mL against CACO2 cells. Acute toxicity of CHL-Mg-NC indicates that the CHL-Mg-NC is safe at high concentrations.
Collapse
Affiliation(s)
- Farag M Mosallam
- Drug Microbiology Lab, Drug Radiation Research Department, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Maha A Shafik
- Drug Microbiology Lab, Drug Radiation Research Department, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | | | - Mohamed A El-Saied
- Department of Pathology, Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Rana M Elshimy
- Microbiology and Immunology, Egyptian Drug Authority, Cairo, Egypt; Microbiology and Immunology, Pharmacy Collage, Al-Ahram Canadian University, 6 October, Egypt
| |
Collapse
|
16
|
Ravichandran N, Uvarajan D, Ravikumar M, Mahendhran K, Krishnamoorthy K, Vellingiri B, Govindasamy C, Narayanasamy A. Gracilaria edulis-mediated silver nanoparticles as a targeted strategy for cervical cancer with integrated toxicity evaluation in zebrafish. Bioorg Chem 2025; 159:108361. [PMID: 40088685 DOI: 10.1016/j.bioorg.2025.108361] [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: 12/13/2024] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Cervical cancer remains a critical global health concern, demanding the development of innovative therapies to address the limitations of conventional chemotherapeutics, including systemic toxicity and lack of specificity. Silver nanoparticles synthesized using Gracilaria edulis (GE-AgNPs) present a novel therapeutic strategy, exhibiting selective cytotoxicity against the HEK293 normal epithelial cell line and HeLa cervical cancer cell line. Phytochemical analysis of Gracilaria edulis identified bioactive compounds such as 4-Benzaldehyde and 1H-1,3-Benzimidazole-1-acetonitrile, both associated with potent anticancer activities. Comprehensive characterization of GE-AgNPs through spectroscopy and microscopy revealed distinctive physicochemical properties, including an absorption peak at 332 nm and a hexagonal crystalline structure. Cytotoxicity assays confirmed that GE-AgNPs induce apoptosis in HeLa cells exhibit a concentration-dependent response, with an IC50 value of 54.05 μg/mL, while GE-AgNPs exhibited no significant toxicity to HEK293 cells at the tested concentrations, as evidenced by a higher IC50 value of 83.6 μg/mL. The pro-apoptotic effect was mediated through the development of reactive oxygen species (ROS), validated using dual staining and Hoechst assays, which demonstrated chromatin condensation indicative of apoptosis. Molecular analysis further elucidated the mechanism of action, highlighting significant inhibition of the PI3K/AKT signaling pathway. This was evidenced by downregulation of PI3K, AKT, and mTOR genes alongside the upregulation of PTEN, a critical tumor suppressor. Zebrafish embryo toxicity assays provided insights into the biocompatibility of GE-AgNPs, revealing low toxicity at therapeutic concentrations but developmental abnormalities and neurotoxicity at higher doses. These findings underscore the promise of GE-AgNPs as a targeted therapy option for cervical cancer, effectively modulating the PI3K/AKT pathway while maintaining manageable toxicity profiles. Further investigations into optimizing dosing regimens and exploring synergistic effects with existing treatments could enhance their clinical applicability.
Collapse
Affiliation(s)
- Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Deenathayalan Uvarajan
- Department of Biochemistry, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Manish Ravikumar
- Department of Biochemistry, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Karthikeyan Mahendhran
- Department of Microbiology, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Kavithaa Krishnamoorthy
- Department of Biotechnology, Hindusthan College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Balachandar Vellingiri
- Neurobiology (Ageing and Pediatric) Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
17
|
Orellana AM, Port's NMS, de Sá Lima L, Leite JA, Andreotti DZ, Kinoshita PF, Cantanzaro AB, Neto JAM, Scavone C, Kawamoto EM. Ouabain increases neuronal differentiation of hippocampal neural precursor cells. CURRENT RESEARCH IN NEUROBIOLOGY 2025; 8:100147. [PMID: 40166632 PMCID: PMC11957680 DOI: 10.1016/j.crneur.2025.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 04/02/2025] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Natacha Medeiros S. Port's
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Jacqueline Alves Leite
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, 74045-155, Brazil
| | - Diana Zukas Andreotti
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Arthur B. Cantanzaro
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - João Agostinho M. Neto
- Laboratory of Cancer biology and Antineoplastic agents. Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Elisa M. Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| |
Collapse
|
18
|
Mthembu SXH, Mazibuko-Mbeje SE, Silvestri S, Orlando P, Nkambule BB, Muller CJF, Tiano L, Dludla PV. Prolonged exposure to simvastatin affects coenzyme Q 9/10 status leading to impaired mitochondrial respiratory capacity and reduced viability of cultured cardiac cells. Toxicol In Vitro 2025; 106:106052. [PMID: 40089196 DOI: 10.1016/j.tiv.2025.106052] [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: 12/13/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
This study investigates the effects of prolonged simvastatin exposure on coenzyme Q9/10 (CoQ9/10) levels, an essential component of antioxidant defense, in cultured cardiac cells. Statins, commonly used to manage dyslipidemia and reduce cardiovascular risk, may impair mitochondrial function, but their impact on CoQ10 depletion and oxidative stress is not well understood. We examined the influence of simvastatin on mitochondrial oxidative capacity, reactive oxygen species (ROS) production, and CoQ9/10 status at concentrations of 0.3, 0.6, 1.25, 2.5, 5, 10, and 20 μM, over durations of 24, 48, and 72 h. Using an in vitro model of cultured H9c2 cardiomyoblasts, our results showed that short-term exposure (24 h) at lower concentrations (<5 μM) enhanced cytosolic and mitochondrial ROS levels without affecting mitochondrial function or CoQ9/10 status. However, prolonged exposure to higher concentrations (≥10 μM for >48 h) resulted in impaired mitochondrial oxidative capacity, indicated by increased proton leak and elevated ROS levels, which were followed by significantly reduced cell viability. These findings suggest that prolonged, high-dose simvastatin exposure may disrupt the oxidative balance of CoQ9/10, leading to myocardial injury. This research addresses a gap in understanding the long-term effects of statins on mitochondrial health and underscores the need for further studies to optimize statin therapy and minimize adverse effects on myocardial function.
Collapse
Affiliation(s)
- Sinenhlanhla X H Mthembu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry, Mafikeng Campus, Northwest University, Mmabatho 2735, South Africa.
| | | | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Centre for Cardiometabolic Research Africa (CARMA), Division of Medical Physiology, Stellenbosch University, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Phiwayinkosi V Dludla
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| |
Collapse
|
19
|
Petsevа Y, Mladenova K, Ganeva M, Radeva V, Videv P, Doumanov J, Petrova S. Heterodimeric vipoxin and its individual monomeric subunits display a dynamic structure-function relationship on RPE cells. Arch Biochem Biophys 2025; 768:110351. [PMID: 39978617 DOI: 10.1016/j.abb.2025.110351] [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: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Known as some of the most toxic venom components, snake venom Phospholipases A2 (svPLA2) impress with their great arsenal of activities, based on catalytic specificity and a variety of non-catalytic "pharmacological" effects using complex molecular mechanisms that can affect different tissues and organs. Here, we aim to reveal the role of the intricate interactions between the monomeric subunits of the heterodimeric neurotoxic complex vipoxin in order to perform multiple differentiated and regulated biological activities in RPE cells. Vipoxin, isolated from the venom of Vipera ammodytes ssp. meridionalis, is composed of a basic and toxic secreted PLA2 enzyme subunit (GIIA sPLA2, vipoxin basic component, VBC) and an acidic, enzymatically inactive and non-toxic subunit (vipoxin acidic component, VAC). We established that vipoxin and its separated monomeric subunits affect integrity and viability of the cells of two RPE lines using a combination of catalytic and non-catalyticmechanisms. Individual monomeric subunits VBC and VAC induce cytotoxicity, cytoskeletal rearrangements, affect transepithelial resistance and cell monolayer integrity, trigger apoptosis, p38 MAPK signaling pathway and genotoxicity, provoking very elaborate cellular response in both cell lines. VBC uses its catalytic and pharmacological activities more pronounced in RPE-1 than in ARPE-19 cell line, triggering DNA double-strand damage as well as a higher degree of cytotoxicity (up to 45 %) in a concentration-dependent manner. In contrast, the non-catalyticVAC exhibited insignificant effect on the membrane integrity of both RPE cell lines but induced very high degree of DNA damage in ARPE-19 cells. Heterodimeric vipoxin displaced its complex and dynamic biological efficiency in both cell lines. In general, all the investigated activities of vipoxin and its individual monomeric subunits proved our assumption for the existence of complex dynamic conformational and structural interactions between the subunits providing an immediate selection of the enzymatic or "pharmacological" mode of action, strongly dependent on RPE cell membrane composition and their microenvironment.
Collapse
Affiliation(s)
- Yulia Petsevа
- Sofia University "St. Kliment Ohridski", Faculty of Biology, Department of Biochemistry, Sofia, Bulgaria
| | - Kirilka Mladenova
- Sofia University "St. Kliment Ohridski", Faculty of Biology, Department of Biochemistry, Sofia, Bulgaria
| | - Maria Ganeva
- Sofia University "St. Kliment Ohridski", Faculty of Biology, Department of Biochemistry, Sofia, Bulgaria
| | - Venelina Radeva
- Sofia University "St. Kliment Ohridski", Faculty of Biology, Department of Biochemistry, Sofia, Bulgaria
| | - Pavel Videv
- Sofia University "St. Kliment Ohridski", Faculty of Biology, Department of Biochemistry, Sofia, Bulgaria
| | - Jordan Doumanov
- Sofia University "St. Kliment Ohridski", Faculty of Biology, Department of Biochemistry, Sofia, Bulgaria
| | - Svetla Petrova
- Sofia University "St. Kliment Ohridski", Faculty of Biology, Department of Biochemistry, Sofia, Bulgaria.
| |
Collapse
|
20
|
Barreto JO, da Silva LA, Lima LDOE, Nogueira PL, Ferreira RC, Alexandre HMDS, Silva ALE, Gonçalves JCR, Sobral MV, de Castro RD, Nascimento YMD, Tavares JF, Rocha WPDS, Guerra FQS. Antifungal and antibiofilm activity of Lippia microphylla Cham. essential oil. (Verbenaceae) on Candida albicans and human cell cytotoxicity. Arch Oral Biol 2025; 174:106243. [PMID: 40209654 DOI: 10.1016/j.archoralbio.2025.106243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/12/2025]
Abstract
OBJECTIVE This study evaluated the antifungal activity of Lippia microphylla essential oil (LM-EO) on Candida albicans. DESIGN In vitro assays were conducted to test LM-EO for its anti-Candida effects, antibiofilm activity, effects on the fluorescence intensity of Candida albicans biofilms observed via confocal microscopy, probable mechanism of action, and toxicity on human keratinocytes. Statistical analysis was performed considering α = 5 %. RESULTS The minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) of LM-EO ranged from 128 µg/mL to 256 µg/mL and 256 µg/mL to 1024 µg/mL, respectively. MIC values did not change in the presence of sorbitol, whereas a 4-fold increase was observed in the presence of ergosterol, suggesting that LM-EO may act on the cell membrane. This was subsequently observed via confocal microscopy using propidium iodide markers. LM-EO reduced biofilm adherence by 66-86 % (p < 0.0001) at low concentrations (256-2560 µg/mL); however, it showed toxicity in human cells. CONCLUSIONS LM-EO exhibited fungicidal activity likely through a mechanism related to ergosterol complexation but demonstrated cytotoxicity to human keratinocytes.
Collapse
Affiliation(s)
- Jaqueline Oliveira Barreto
- Graduate Program in Dentistry (PPGO), Department of Clinic and Social Dentistry, Center for Health Sciences, Federal University of Paraiba, João Pessoa, PB 58051-900, Brazil
| | - Larissa Alves da Silva
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Luanna de Oliveira E Lima
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Paula Lima Nogueira
- Graduate Program in Dentistry (PPGO), Department of Clinic and Social Dentistry, Center for Health Sciences, Federal University of Paraiba, João Pessoa, PB 58051-900, Brazil
| | - Rafael Carlos Ferreira
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Heivila Monique da Silva Alexandre
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Anauara Lima E Silva
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Juan Carlos Ramos Gonçalves
- Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Marianna Vieira Sobral
- Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Ricardo Dias de Castro
- Department of Clinic and Social Dentistry, Center for Health Sciences, Federal University of Paraiba, João Pessoa, PB 58051-900, Brazil
| | - Yuri Mangueira do Nascimento
- Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Josean Fechine Tavares
- Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil
| | | | - Felipe Queiroga Sarmento Guerra
- Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa, PB 58051-900, Brazil.
| |
Collapse
|
21
|
Neves Rebello Alves L, Merigueti LP, Casotti MC, Cancian de Araújo B, Silva Dos Reis Trabach R, Batitucci MDCP, Meira DD, de Paula F, de Vargas Wolfgramm Dos Santos E, Louro ID. Glyphosate-based herbicide as a potential risk factor for breast cancer. Food Chem Toxicol 2025; 200:115404. [PMID: 40122508 DOI: 10.1016/j.fct.2025.115404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 03/09/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Breast cancer is the most common neoplasm in women worldwide, with both genetic and environmental factors playing a role in its development. Glyphosate, the active ingredient in widely used agricultural herbicides, is recognized as a potential carcinogen and endocrine disruptor, making it a candidate for inducing epigenetic modifications linked to breast cancer. This study investigates the effects of the glyphosate-based herbicide Roundup® on non-tumorigenic (MCF10A) and tumorigenic (MCF7 and MDA-MB-231) breast cell lines, focusing on the expression of key breast cancer-related genes. Additionally, the study examines the association with epigenetic modifications and the use of epidrugs to reverse potential alterations, aiming to understand the risks and mechanisms of herbicide action. Results indicate that Roundup® affects cells through a non-estrogenic mechanism, impacting both hormone-dependent and -independent cells with varying toxic and proliferative effects depending on dose and exposure time. Moreover, it altered the expression of breast cancer-related genes such as BRCA1 and BRCA2 at low doses. The use of epigenetic modulators was able to reverse some Roundup®-induced changes, suggesting the herbicide's role in epigenetic modifications. Overall, these findings highlight the importance of understanding glyphosate-based herbicide mechanisms in humans, which could enable personalized prevention strategies to mitigate breast cancer risks.
Collapse
Affiliation(s)
- Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular (NGHM), Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória, 29075-910, ES, Brazil; Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo (UFES), Vitória, 29047-105, ES, Brazil.
| | - Luiza Poppe Merigueti
- Núcleo de Genética Humana e Molecular (NGHM), Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória, 29075-910, ES, Brazil.
| | - Matheus Correia Casotti
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo (UFES), Vitória, 29047-105, ES, Brazil.
| | - Bruno Cancian de Araújo
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo (UFES), Vitória, 29047-105, ES, Brazil.
| | - Raquel Silva Dos Reis Trabach
- Núcleo de Genética Humana e Molecular (NGHM), Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória, 29075-910, ES, Brazil.
| | | | - Débora Dummer Meira
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo (UFES), Vitória, 29047-105, ES, Brazil.
| | - Flávia de Paula
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo (UFES), Vitória, 29047-105, ES, Brazil.
| | | | - Iúri Drumond Louro
- Núcleo de Genética Humana e Molecular (NGHM), Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória, 29075-910, ES, Brazil; Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo (UFES), Vitória, 29047-105, ES, Brazil.
| |
Collapse
|
22
|
Ismail A, Hassan NW, Saudi MN, Abdel-Ghany YS, Labib HF, El-Deeb NM, El-Hawash SA. Exploring Thieno/Furo[2,3-b]pyridines as new scaffolds for potential FAK inhibition: Design, synthesis, biological evaluation and in silico studies. Bioorg Chem 2025; 159:108392. [PMID: 40139117 DOI: 10.1016/j.bioorg.2025.108392] [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: 01/03/2025] [Revised: 03/02/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025]
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that plays a vital role in regulating cancer cell survival, proliferation, migration, and angiogenesis. Aiming to explore new potent inhibitors, a series of thieno/furo[2,3-b]pyridine derivatives was designed and synthesized. The newly synthesized compounds were evaluated for their in vitro anti-proliferative activity against human liver (HUH-7), lung (A549) and breast (MCF-7) cancer cell lines, in addition to their cytotoxic activity against normal lung cell line (WI-38) to predict their safety profile. Seven compounds (4a, 4c, 5, 6, 10c, 11 and 12) displayed significant anti-proliferative activity as well as high selectivity towards the tested cancer cell lines (SI > 2). Among them, two compounds (4a and 4c) potently inhibited FAK enzyme with IC50 values of 54.96 and 50.98 nM, respectively. Flow cytometric cell cycle analysis indicated that compounds 4a and 4c caused cell cycle arrest at G1 phase. Compound 4c also exhibited an increase in the expression level of caspase-3 enzyme. Moreover, molecular docking study of the most promising compounds into FAK's active site was performed to elucidate their possible binding modes and to provide a structural basis for the further structural guidance design of FAK inhibitors.
Collapse
Affiliation(s)
- Azza Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Nayera W Hassan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Manal N Saudi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Yasser S Abdel-Ghany
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Hala F Labib
- Department of Pharmaceutical Chemistry, College of Pharmacy, Arab Academy of Science Technology and Maritime Transport, Alexandria, Egypt
| | - Nehal M El-Deeb
- Pharmaceutical Bioproducts Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934, Alexandria, Egypt.; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8 M5, Canada
| | - Soad A El-Hawash
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
23
|
Bilgehan A, Şeker Z, Qaoud MT, Özhan G. In vitro investigation of the toxicological mechanisms of Fingolimod (S)-phosphate in HEPG2 cells. Toxicol Res (Camb) 2025; 14:tfaf064. [PMID: 40321693 PMCID: PMC12050031 DOI: 10.1093/toxres/tfaf064] [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: 01/14/2025] [Revised: 03/31/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Fingolimod (FTY720) was the first sphingosine-1-phosphate (S1P) receptor modulator approved by the US Food and Drug Administration for the treatment of multiple sclerosis. The active form, FTY720 (S)-P, acts as a potent agonist of the S1P receptor, leading to its downregulation on the cell surface, reduced activity, and termination of sphingosine-dependent intracellular signalling. Elevated hepatic enzyme levels, clinically significant liver injury, and acute liver failure have been observed in patients treated with FTY720 (S)-P, which requires additional monitoring. This is the first study to investigate the mechanisms underlying the hepatotoxicity of FTY720 (S)-P and represents an important contribution to elucidating its toxicity mechanisms in the human hepatocellular carcinoma cell line HepG2. Following a 72-h exposure, standard methods were used to evaluate specific targets, including cytotoxic effect potentials, mitochondrial parameters, and changes of the antioxidant enzyme levels. FTY720 (S)-P exposure resulted in time- and dose-dependent decreases in cell viability, mitochondrial membrane potential, and ATP levels, as well as the induction of oxidative stress. The complex toxic profile observed for FTY720 (S)-P is hypothesized to originate from its interaction with sirtuin proteins, particularly SIRT3 and SIRT5. It was also complemented with molecular docking simulations to assess the compound's targeting potential by analysing its interaction profile and binding pose within the active sites of both proteins. The results supported the proposed hypothesis, demonstrating an optimal fitting profile and favourable interaction behaviour within the binding pockets of the SIRT3 and SIRT5 enzymes.
Collapse
Affiliation(s)
- Ayşenur Bilgehan
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul University, Fatih, Istanbul 34116, Türkiye
- Institute of Graduate Studies in Health Sciences, Istanbul University, Fatih, Istanbul 34116, Türkiye
| | - Zehra Şeker
- Institute of Graduate Studies in Health Sciences, Istanbul University, Fatih, Istanbul 34116, Türkiye
| | - Mohammed T Qaoud
- Faculty of Pharmacy, Department of Pharmacy, Cyprus International University, Nicosia, Northern Cyprus 99258, Türkiye
| | - Gül Özhan
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul University, Fatih, Istanbul 34116, Türkiye
| |
Collapse
|
24
|
da Silva Gebara R, da Silva MS, Calixto SD, Simão TLBV, Zeraik AE, Lassounskaia E, Muzitano MF, Petretski JH, Gomes VM, de Oliveira Carvalho A. Antifungal, Antimycobacterial, Protease and α‒Amylase Inhibitory Activities of a Novel Serine Bifunctional Protease Inhibitor from Adenanthera pavonina L. Seeds. Probiotics Antimicrob Proteins 2025; 17:1320-1342. [PMID: 38117407 DOI: 10.1007/s12602-023-10194-z] [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] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Antifungal resistance poses a significant challenge to disease management, necessitating the development of novel drugs. Antimicrobial peptides offer potential solutions. This study focused on extraction and characterization of peptides from Adenanthera pavonina seeds with activity against Candida species, Mycobacterium tuberculosis, proteases, and α-amylases. Peptides were extracted in phosphate buffer and heated at 90°C for 10 min to create a peptide rich heated fraction (PRHF). After confirming antimicrobial activity and the presence of peptides, the PRHF underwent ion exchange chromatography, yielding retained and non-retained fractions. These fractions were evaluated for antimicrobial activity and cytotoxicity against murine macrophages. The least toxic and most active fraction underwent reversed-phase chromatography, resulting in ten fractions. These fractions were tested for peptides and antimicrobial activity. The most active fraction was rechromatographed on a reversed-phase column, resulting in two fractions that were assessed for antimicrobial activity. The most active fraction revealed a single band of approximately 6 kDa and was tested for inhibitory effects on proteases and α-amylases. Thermal stability experiments were conducted on the 6 kDa peptide at different temperatures followed by reassessment of antifungal activity and circular dichroism. The 6 kDa peptide inhibited yeasts, M. tuberculosis, human salivary and Tenebrio molitor larvae intestine α-amylases, and proteolytic activity from fungal extracts, and thus named ApPI. Remarkably, ApPI retained antifungal activity and conformation after heating and is primarily composed of α-helices. ApPI is a thermally stable serine protease/α-amylase inhibitor from A. pavonina seeds, offering promise as a foundational molecule for innovative therapeutic agents against fungal infections and tuberculosis.
Collapse
Affiliation(s)
- Rodrigo da Silva Gebara
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - Marciele Souza da Silva
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - Sanderson Dias Calixto
- Laboratório de Biologia do Reconhecer, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - Thatiana Lopes Biá Ventura Simão
- Laboratório de Biologia do Reconhecer, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - Ana Eliza Zeraik
- Laboratório de Química e Função de Proteinas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - Elena Lassounskaia
- Laboratório de Biologia do Reconhecer, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - Michelle Frazão Muzitano
- Laboratório de Produtos Bioativos, Universidade Federal do Rio de Janeiro, Macaé, 27933-378, RJ, Brazil
| | - Jorge Hudson Petretski
- Laboratório de Biologia do Reconhecer, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - Valdirene Moreira Gomes
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil
| | - André de Oliveira Carvalho
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, 28013-602, RJ, Brazil.
| |
Collapse
|
25
|
Yıldırım A, Aksoy T, Balcıoğlu İC. Comparative Assessment of colorimetric assays in evaluating intracellular drug susceptibility of Leishmania tropica against conventional antileishmanial drugs. Parasitol Int 2025; 106:103021. [PMID: 39719247 DOI: 10.1016/j.parint.2024.103021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/12/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
This study aims to identify the most sensitive colorimetric test for assessing intracellular drug susceptibility of Leishmania tropica to conventional antileishmanial drugs. To this end, the efficacy of four colorimetric methods-MTT, XTT, MTS, and WST-8-was compared using reference L. tropica promastigotes. The intracellular drug susceptibility was further evaluated using the test with the widest absorbance range on isolates from Türkiye CL patients: two responsive to a single course of meglumine antimoniate (MA) and two that showed no clinical improvement after two treatments. CL isolates were identified via real-time PCR targeting the ITS1 region. Promastigote suspensions at standardized densities (0.08 × 106 to 10 × 106 promastigotes/well) were prepared in both RPMI (phenol red-containing) and RPMIØRP (phenol red-free) media, then analyzed with ELISA-based MTT, XTT, MTS, and WST-8 to identify the method with the broadest specific absorbance range. Intracellular drug susceptibility of CL isolates was subsequently assessed in a macrophage/amastigote model by infecting THP-1 macrophages with promastigotes from both reference and patient isolates, followed by treatment with MA, sodium stibogluconate (SSG), miltefosine (MTF), pentamidine (PMD), and amphotericin B (AmB). Promastigotes obtained from parasite rescue and transformation assays were analyzed using the most sensitive colorimetric method to determine IC₅₀ values. Species identification confirmed all four CL isolates as L. tropica, and the XTT assay provided the widest absorbance range in RPMIØRP media. IC₅₀ values for both treatment-responsive and unresponsive isolates were similar to those of the reference isolate, showing susceptibility to all tested drugs without statistically significant differences. Expanding the isolate set is necessary to further evaluate the predictive value of SbV (pentavalent antimonials) susceptibility for treatment outcomes. The identification of XTT as the most sensitive method for intracellular antileishmanial susceptibility testing is expected to aid in standardizing laboratory models and provide valuable insights for researchers and clinicians managing treatment-unresponsive CL cases.
Collapse
Affiliation(s)
- Ahmet Yıldırım
- Manisa Celal Bayar University, Medical Faculty, Department of Parasitology, Manisa, Turkey
| | - Tülay Aksoy
- Manisa Celal Bayar University, Medical Faculty, Department of Parasitology, Manisa, Turkey.
| | | |
Collapse
|
26
|
Gandlevskiy N, Viana AR, Druzian GT, Oliveira DK, Schuch AP, Barge A, Cravotto G, Moraes Flores EM. Ultrasound-assisted green synthesis of silver nanoparticles using Ruta graveolens L. Extract and antitumor evaluation. ULTRASONICS SONOCHEMISTRY 2025; 117:107340. [PMID: 40263047 DOI: 10.1016/j.ultsonch.2025.107340] [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: 01/06/2025] [Revised: 02/17/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
This study aims to evaluate the ultrasound-assisted synthesis of silver nanoparticles (AgNPs) based on an aqueous extract of Ruta graveolens L. to reduce Ag+ to Ag0. In addition, the biological activity of the synthesized AgNPs was evaluated against tumor cells. The following parameters were evaluated for the synthesis: proportion between Ag+ solution (0.1 mol L-1 Ag+) and R. graveolens L. extract, pH of R. graveolens L. extract (5, 7, and 9), temperature of solution containing Ag+ and R. graveolens L. extract, ultrasound (US) type (bath and probes), and the parameters for US as frequency (37, and 80 kHz for bath and 20 kHz for probes), amplitude and time of application of US. In order to confirm the US effect, "silent" experiments (without US) were performed. Using the optimized conditions (US bath, proportion between Ag+ solution and R. graveolens L. of 1 + 5, v v-1, 80 kHz, 70 % amplitude, 70 °C, pH 9, and 25 min of sonication time) it was possible to obtain mean size, PI, and zeta potential of AgNPs of 30 nm, 0.129, -34.44 mV, respectively. For comparison of results, AgNPs synthesized in the "silent" condition presented mean size, PI, and zeta potential of 66 nm, 0.412, and -22.12 mV, respectively. The US synthesized AgNPs were lower, more uniform, and stable when compared with magnetic stirring. In addition, the morphology of AgNPs using US was predominantly spherical and monodisperse. The biological activity using cell lines HaCat (keratinocytes), L929 (fibroblasts), and B16-F10 (melanoma) against nanoparticles synthesized using US was evaluated against the different cell lines and the antioxidant activity of the AgNPs was measured by the 2,2-diphenyl-1-picrylhydrazyl (DPPH) method. A higher cytotoxic effect on the melanoma cell line (IC50 = 2.12 µg mL-1) compared to normal cells. A good result was found in the DPPH assay, with an IC50 of 234.3 µg mL-1 for free radical scavenging. Therefore, the US technology presents a promising and sustainable green method avoiding the use of toxic reagents and obtained AgNPs showed potent anticancer activity.
Collapse
Affiliation(s)
- Nikita Gandlevskiy
- Department of Drug Science and Technology, Turin University, 10125, via P. Giulia 9, Turin, Italy
| | - Altevir Rossato Viana
- Department of Chemistry, Federal University of Santa Maria 97105-900 Santa Maria, Brazil
| | - Gabriel Toneto Druzian
- Department of Chemistry, Federal University of Santa Maria 97105-900 Santa Maria, Brazil
| | | | - Andre Passaglia Schuch
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria 97105-900 Santa Maria, Brazil
| | - Alessandro Barge
- Department of Drug Science and Technology, Turin University, 10125, via P. Giulia 9, Turin, Italy
| | - Giancarlo Cravotto
- Department of Drug Science and Technology, Turin University, 10125, via P. Giulia 9, Turin, Italy
| | | |
Collapse
|
27
|
Moreira Silva EZ, Rodrigues AC, Kohler AF, Cruz JV, Prado KB, Gradia DF, de Oliveira DP, Pestana CB, Leme DM. Evaluating dermal toxicity of the flame retardant aluminum diethylphosphinate by in silico-in vitro testing strategy. CHEMOSPHERE 2025; 379:144421. [PMID: 40286754 DOI: 10.1016/j.chemosphere.2025.144421] [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: 12/10/2024] [Revised: 03/28/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
Flame retardants (FRs) are a group of chemicals used in different products to improve fire safety; however, many of them negatively impact human health, encouraging the use of safer alternatives. The aluminum diethylphosphinate (AlPi) is one of the potential alternatives to harmful FRs; however, close data gaps on human toxicity and enhanced mechanistic understanding are still needed. This study evaluated the dermal toxicity potential of AlPi using in silico models (OECD QSAR Toolbox, Toxtree) and a multi-biomarkers approach with human keratinocyte models (HaCaT cell line and reconstructed human epidermis (RHE) model). Our findings revealed no significant increases in reactive oxygen species (ROS, H2DCFDA) or pro-inflammatory cytokines (IL-6, IL-8, IL-10, IL-1β, IL12p70, TNF) in HaCaT cells exposed to AlPi at non-cytotoxic concentrations (30, 60, 120 μg/ml). This suggests that AlPi does not induce oxidative stress or inflammatory responses in the skin. Additionally, in silico predictions and in vitro assays (HaCaT - IL-6; OECD TG 439 with SkinVitro-RHE) did not classify AlPi as a skin sensitizer or skin irritant. Regarding changes in DNA, AlPi-exposed HaCaT cells did not show significant levels of γ-H2AX; however, this FR increased the level of 5-hydroxymethylcytosine (5-hmC) and TET1 expression, which is a gene involved in the regulation of the DNA methylation. In summary, most biomarker responses indicated that AlPi poses minimal toxic effects on the skin; however, further research is needed to understand better the biological consequences of its effect on DNA methylation.
Collapse
Affiliation(s)
| | | | - Ana Flávia Kohler
- Department of Genetics, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Juliana Varella Cruz
- Department of Genetics, Federal University of Paraná (UFPR), Curitiba, PR, Brazil; School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Karin Braun Prado
- Department of Basic Pathology, Federal University of Parana (UFPR), Curitiba, PR, Brazil
| | - Daniela Fiori Gradia
- Department of Genetics, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Danielle Palma de Oliveira
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Institute of Chemistry, Araraquara, SP, Brazil
| | | | - Daniela Morais Leme
- Department of Genetics, Federal University of Paraná (UFPR), Curitiba, PR, Brazil; National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Institute of Chemistry, Araraquara, SP, Brazil.
| |
Collapse
|
28
|
Said AI, Ewes WA, Hamdi A, El-Rashedy AA, Ahmed M. New pyrrolo[3,4-d] isoxazolidines hybrid with furan as antitumor agents and multi-target enzyme inhibitors: Synthesis and in silico study. Bioorg Chem 2025; 159:108377. [PMID: 40121772 DOI: 10.1016/j.bioorg.2025.108377] [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: 12/22/2024] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Herein, new Pyrrolo[3,4-d] isoxazolidines hybrid with furan were synthesized by 1,3-dipolar cycloaddition reaction of nitrone 2 with N-substituted maleimides 3a-j. The synthesized compounds were screened in vitro cytotoxic assay against four cancer cell lines namely, HeLa, HEPG-2, HCT-116 and MCF-7 using doxorubicin (DOX) as a reference using MTT assay. The results demonstrated that compounds 4b and 4j exhibited the highest antitumor activity with IC50 =6.22-16.44 μM in comparable to DOX (IC50 = 4.17-5.57μM). The most active hybrids 4b and 4j were further subjected to multi-targeting assays against EGFR, VEGFR-2, and Topo II. They showed good to moderate inhibitory activities. In addition, flow cytometric analysis of 4b and 4j inhibited cell population of MCF-7 cells in the S phase. Compound 4b, and 4j were further evaluated using molecular docking and dynamics simulations (20 ns) and the EGFR, TOPII, or VEGFR-2 receptor protein. All the data sets accurately predict the strongest binding affinity for the selected compounds, as evidenced by the highest free binding energy from MM/GBSA calculations and significant amino acid steric interactions. Furthermore, the RMS/RMSF/Rg/SASA dynamics parameters show the formed complexes demonstrate satisfactory stability. The ADMET properties indicate that the selected new ligands have shown a promising drug-like profile and can be considered potential candidates for future anti-cancer therapies, with perspective validating their anticancer activity by in vitro studies.
Collapse
Affiliation(s)
- Awad I Said
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt.
| | - Wafaa A Ewes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura university, Mansoura 35516, Egypt.
| | - Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura university, Mansoura 35516, Egypt
| | - Ahmed A El-Rashedy
- Natural and Microbial Products Department, National Research Center (NRC), Egypt; Department Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Mostafa Ahmed
- Chemistry Department, Faculty of Science, New Valley University, El-Kharja, 72511, Egypt.
| |
Collapse
|
29
|
Sunagawa Y, Iwashimizu S, Ono M, Mochizuki S, Iwashita K, Sato R, Shimizu S, Funamoto M, Shimizu K, Hamabe-Horiike T, Katanasaka Y, Murakami A, Asakawa T, Inai M, Kan T, Komiyama M, Hawke P, Mori K, Arakawa Y, Hasegawa K, Sakamoto K, Kurokawa J, Morimoto T. The citrus flavonoid nobiletin prevents the development of doxorubicin-induced heart failure by inhibiting apoptosis. J Pharmacol Sci 2025; 158:84-94. [PMID: 40288827 DOI: 10.1016/j.jphs.2025.03.011] [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: 01/25/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The anthracycline anticancer drug doxorubicin (DOX) induces myocardial cell death and heart failure. The aim of the present study was to investigate whether nobiletin (NOB), a natural flavonoid isolated from citrus peel, has a protective effect against DOX-induced cardiotoxicity. METHODS AND RESULTS H9C2 cells were pretreated with 100 μM NOB and then treated with 1 μM DOX. An MTT assay revealed that NOB improved the decreased cell viability induced by DOX. A TUNEL assay showed that NOB treatment improved DOX-induced apoptosis in H9C2 cells. Western blotting indicated that DOX-induced increases in cleaved caspase-3 and -9 expression were significantly suppressed by NOB treatment. Motion field imaging of human iPS cell-derived cardiomyocyte sheets showed that NOB significantly suppressed a DOX-induced reduction of their contractile function. Next, to investigate the effect of NOB in vivo, DOX was intraperitoneally administered to mice. Echocardiography showed that oral administration of NOB reduced DOX-induced left ventricular systolic dysfunction, and a TUNEL assay showed that oral administration also inhibited apoptosis in the mouse heart. CONCLUSIONS These results indicate that NOB treatment suppressed DOX-induced cardiotoxicity by reducing apoptosis. Further study of the mechanism of this effect may lead to the development of a novel therapy for DOX-induced heart failure.
Collapse
Affiliation(s)
- Yoichi Sunagawa
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Shizuoka General Hospital, Shizuoka, 420-8527, Japan
| | - Sonoka Iwashimizu
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Masaya Ono
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Saho Mochizuki
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Kenshiro Iwashita
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Rina Sato
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Satoshi Shimizu
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Masafumi Funamoto
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan
| | - Kana Shimizu
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan
| | - Toshihide Hamabe-Horiike
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Shizuoka General Hospital, Shizuoka, 420-8527, Japan
| | - Yasufumi Katanasaka
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Shizuoka General Hospital, Shizuoka, 420-8527, Japan
| | - Akira Murakami
- Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Hyogo, Japan
| | - Tomohiro Asakawa
- Department of Synthetic Organic & Medicinal Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Marine Science and Technology, Tokai University, Shizuoka, 424-8610, Japan
| | - Makoto Inai
- Department of Synthetic Organic & Medicinal Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Toshiyuki Kan
- Department of Synthetic Organic & Medicinal Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Maki Komiyama
- Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan
| | - Philip Hawke
- Laboratory of Scientific English, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Kiyoshi Mori
- Shizuoka General Hospital, Shizuoka, 420-8527, Japan; Graduate School of Public Health, Shizuoka Graduate University of Public Health, Japan, Shizuoka, 420-0881, Japan; Division of Molecular and Clinical Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Koji Hasegawa
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan
| | - Kazuho Sakamoto
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Junko Kurokawa
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Tatsuya Morimoto
- Department of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan; Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, 612-8555, Japan; Shizuoka General Hospital, Shizuoka, 420-8527, Japan.
| |
Collapse
|
30
|
Pereira JC, de Sousa RWR, Conceição MLP, do Nascimento MLLB, de Almeida ATA, Dos Reis AC, de Sousa Cavalcante ML, Dos Reis Oliveira C, Martins IRR, Torres-Leal FL, Dittz D, de Castro E Sousa JM, Ferreira PMP, Carneiro da Silva FC. Buthionine sulfoximine acts synergistically with doxorubicin as a sensitizer molecule on different tumor cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:409-431. [PMID: 39815616 DOI: 10.1080/15287394.2024.2448663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The chemotherapeutic drug doxorubicin (DOX) has been widely used for treating solid tumors attributed to its antiproliferative effectiveness; however, its clinical use is limited due to side effects, including cardiotoxicity, myelosuppression, and drug resistance. Combining DOX with buthionine sulfoximine (BSO), a glutathione (GSH) synthesis inhibitor, showed promising results in overcoming these adverse effects, potentially reducing the required DOX dose while maintaining efficacy. The aim of the present study was to examine the effects of different concentrations of BSO and DOX, both individually and in combination, utilizing B16/F10 (murine melanoma), SNB-19 (human glioblastoma), S180 (murine sarcoma), and SVEC4-10 (murine endothelial) cell lines. Cell viability, migration, and clonogenicity were assessed using the following assays MTT, scratch, and colony formation. Antioxidant levels of GSH, as well as activities catalase (CAT), and superoxide dismutase (SOD) were measured. BSO alone exhibited minimal cytotoxic effects, while DOX alone reduced cell viability significantly. The combination of BSO+DOX decreased IC50 values for most cell lines, demonstrating a synergistic effect, especially in B16/F10, S180, and SVEC4-10 cells. BSO+DOX combination significantly inhibited cell migration and clonogenicity compared to DOX alone. While GSH levels were decreased with BSO+DOX treatment activities of CAT and SOD increased following DOX administration but remained unchanged by BSO. These results suggest that BSO may be considered a valuable tool to improve DOX therapeutic efficacy, particularly in cases of chemotherapy-resistant tumors, as BSO enhances DOX activity while potentially reducing systemic chemotherapeutic drug toxicity.
Collapse
Affiliation(s)
- Joedna Cavalcante Pereira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Rayran Walter Ramos de Sousa
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Micaely Lorrana Pereira Conceição
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | | | - Ana Tárcila Alves de Almeida
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Antonielly Campinho Dos Reis
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| | - Mickael Laudrup de Sousa Cavalcante
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Camila Dos Reis Oliveira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Italo Rossi Roseno Martins
- Academic Unit of Life Sciences, Teachers' Forming Center, Federal University of Campina Grande, Cajazeiras-PB, Brazil
| | - Francisco Leonardo Torres-Leal
- Metabolic Diseases, Exercise and Nutrition Research Group (Domen), Laboratory of Metabolic Diseases Glauto Tuquarre, Department of Biophysics and Physiology, Federal University of Piaui, Teresina-PI, Brazil
| | - Dalton Dittz
- Laboratory of Antineoplastic Pharmacology (Lafan), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| | - João Marcelo de Castro E Sousa
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Felipe Cavalcanti Carneiro da Silva
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| |
Collapse
|
31
|
Ke X, Wu Q, Cai S, Wang C, Lu T, Sun Z, Tian X, Wu X, Wang B, Sun B. Dl-3-n-Butylphthalide enhances the survival of rat bone marrow stem cells via a reactive oxygen species mediated Erk1/2 signaling pathway. Brain Res 2025; 1855:149551. [PMID: 40086743 DOI: 10.1016/j.brainres.2025.149551] [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: 12/30/2024] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Survival of bone marrow stem cells (BMSCs) is crucial for successful bone marrow transplantation. However, the underlying molecular mechanisms remain inadequately understood. Our previous research has demonstrated that dl-3-n-butylphthalide (NBP) can protect rat BMSCs (rBMSCs) from cell death via its antioxidative properties and by activating the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. The findings suggest that the PI3K/Akt pathway may be one of the primary targets through which NBP exert its protective effects. In this study, we explored an additional signaling pathway to further elucidate the molecular mechanisms involved in NBP-mediated protection against oxidative stress injury in rBMSCs. Oxidative stress was induced in rBMSCs using hydrogen peroxide (H2O2), imitating the cerebral ischemia microenvironment surrounding transplanted cells in vitro. The protective effects of NBP on rBMSCs against apoptosis were observed, achieving by decreasing the level of reduce reactive oxygen species (ROS) and malondialdehyde (MDA) while simultaneously increasing the concentration of superoxide dismutase (SOD). Notably, these protective effects were partially inhibited by U0126, an extracellular signal-regulated kinase1/2 (Erk1/2) inhibitor, which enhanced the suppression of NBP's antiapoptotic effects. Our results indicated that NBP could protect rBMSCs from apoptosis through modulation of ROS/Erk pathways. Further investigations are warranted to clarify the unknown mechanisms.
Collapse
Affiliation(s)
- Xianjin Ke
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, PR China
| | - Qianqian Wu
- Department of Electrophysiology Laboratory, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Shikun Cai
- Department of Neurology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Chengyun Wang
- Department of Neurology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Ting Lu
- Department of Neurology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Zhenjie Sun
- Department of Neurology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Xiangyang Tian
- Department of Neurology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Xian Wu
- Department of Stomatology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, PR China
| | - Bingjian Wang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China.
| | - Bo Sun
- Department of Neurology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, PR China; Department of Neurology, The Huaian Clinical College of Xuzhou Medical University, Huai'an, Jiangsu 223300, PR China.
| |
Collapse
|
32
|
El Mais AER, D'Anna B, Albinet A, Aït-Aïssa S. In vitro assessment of aryl hydrocarbon, estrogen, and androgen receptor-mediated activities of secondary organic aerosols formed from the oxidation of polycyclic aromatic hydrocarbons and furans. ENVIRONMENTAL RESEARCH 2025; 273:121220. [PMID: 40010426 DOI: 10.1016/j.envres.2025.121220] [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: 12/23/2024] [Revised: 02/05/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
Biomass burning constitutes a significant source of fine particulate matter (PM2.5) in the atmosphere, particularly during winter due to residential wood heating. This source also emits substantial quantities of volatile and semi-volatile organic compounds, leading through (photo-)chemical and physical processes, to the formation of secondary organic aerosols (SOAs), accounting for a significant fraction of PM2.5. The current understanding of the biological effects of SOA resulting from the oxidation of major gaseous precursors emitted by biomass burning (e.g., polycyclic aromatic hydrocarbons (PAHs), phenols, furans) is still limited. Mechanism-based in vitro cellular bioassays targeting toxicologically relevant modes of action have proven valuable in assessing and quantifying the overall biological activity of complex mixtures like SOA, thereby revealing the presence of toxicologically relevant compounds. The main objective of this study was to investigate, using a battery of in vitro mechanism-based cellular bioassays, the aryl hydrocarbon (AhR), estrogen (ER), and androgen receptor (AR)-mediated activities of laboratory-generated SOA resulting from the oxidation of four PAHs and three furans. SOA was produced using an oxidation flow reactor (OFR) under either daytime (OH radicals) or nighttime (NO3 radicals) conditions. Furan-derived SOA did not exhibit any biological activity with the targeted endpoints. PAH-derived SOA, formed from AhR weakly or inactive PAHs, showed significant AhR-mediated activities. Notably, SOA resulting from naphthalene and acenaphthylene + acenaphthene demonstrated the highest AhR activation potency, with greater activities observed for SOA formed through NO3 radical oxidation. No endocrine-disrupting activity was observed for the PAH-derived SOA, similar to the individual parent PAHs (with the exception of fluorene and phenanthrene PAHs which were weekly anti-androgenic). These findings underscore the substantial contribution of PAH-derived SOA to the AhR-mediated activities of PM.
Collapse
Affiliation(s)
- Abd El Rahman El Mais
- Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique Alata, Verneuil-en-Halatte, 60550, France; Aix Marseille Univ, CNRS, LCE, Marseille, France
| | | | - Alexandre Albinet
- Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique Alata, Verneuil-en-Halatte, 60550, France.
| | - Selim Aït-Aïssa
- Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique Alata, Verneuil-en-Halatte, 60550, France.
| |
Collapse
|
33
|
Saffar A, Bahrami AR, Sh Saljooghi A, Matin MM. ZIF-8/doxorubicin nanoparticles camouflaged with Cucurbita-derived exosomes for targeted prostate cancer therapy. J Mater Chem B 2025; 13:5705-5722. [PMID: 40261644 DOI: 10.1039/d5tb00086f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Development of biomimetic drug delivery systems (DDSs) holds great promise to overcome various nanoparticle-associated hindrances in cancer therapy. However, producing biomimetic nanoparticles camouflaged by animal cell-secreted exosomes presents several challenges, including low yield and some ethical considerations. Herein, we designed a biomimetic nanocarrier composed of zeolitic imidazolate framework-8 (ZIF-8) encapsulating doxorubicin (DOX) as the core and a shell of exosome-like nanoparticles (EXO) derived from Cucurbita moschata (CEXO). This design enhances safety and addresses some exosome limitations. The CEXO@ZIF-8/DOX platform was further functionalized with an epithelial cell adhesion molecule (EpCAM) aptamer (Apt-CEXO@ZIF-8/DOX) for targeted delivery to prostate cancer (PC) cells. After investigating the anticancer activity of CEXOs on PC-3 cells, the exosomes were utilized to coat ZIF-8/DOX. The immune evasion capability, cellular uptake, and anticancer effects of nanoplatforms were assessed. Moreover, the in vivo effectiveness of the targeted platform in inhibiting tumor growth and minimizing the adverse effects, was assessed using immunocompromised C57BL/6 mice bearing human PC-3 tumors. Cucurbita exosomes decreased cell viability and induced cell cycle arrest and apoptosis in PC-3 cells without affecting the normal cells. The biomimetic CEXO@ZIF-8/DOX improved immune escaping ability and hemocompatibility. The targeted nanocarrier, with augmented uptake and cellular toxicity in EpCAM-positive PC-3 cells, indicated active targeting efficacy mediated by the EpCAM aptamer. These results were supported by animal experiments that implied the effectiveness of Apt-CEXO@ZIF-8/DOX in inhibiting tumor growth without adverse side effects. This study introduces a novel functional nanocarrier that could potentially revolutionize DDSs by utilizing safer and more biocompatible plant exosomes.
Collapse
Affiliation(s)
- Adeleh Saffar
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
34
|
Cayero-Otero MD, Perez-Caballero L, Suarez-Pereira I, Hidalgo-Figueroa M, Delgado-Sequera A, Montesinos JM, Berrocoso E, Martín-Banderas L. Venlafaxine-PLGA nanoparticles provide a fast onset of action in an animal model of depression via nose-to-brain. Int J Pharm 2025; 678:125692. [PMID: 40339630 DOI: 10.1016/j.ijpharm.2025.125692] [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: 09/03/2024] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Current treatment of depression is hindered by the delayed onset of the action of antidepressant drugs, often resulting in treatment failure. Therefore, new therapeutic solutions are imperative. METHODOLOGY Venlafaxine-loaded poly(lactic-co-glycolic acid) nanoparticles were produced by a double emulsion-solvent evaporation method. Cellular safety assessment and internalization assays were carried out in vitro in human olfactory neuroepithelium cells. The antidepressant effect of intranasal (nose-to-brain) nanoparticle administration was assessed in animals submitted to an animal model of depression by behavioral tests, including open-field, sucrose preference test and tail suspension test. RESULTS The drug entrapment efficiency (55-65 %), particle size (190-210 nm), polydispersity index (<0.2), and zeta potential (-20 mV) of Venlafaxine-loaded poly(lactic-co-glycolic acid) nanoparticles were determined to be adequate. Nanoparticles did not show cytotoxic effects. Cell viability was more than 90 % for all formulations and concentrations assayed. The results of the quantitative and qualitative cell uptake assays were consistent, showing an evident internalization of the nanoparticles into the cells. Furthermore, venlafaxine-loaded nanoparticles administered for just 7 days were able to reverse the phenotype induced by a depressive-like model, showing a significant antidepressant-like effect compared to those treated with free venlafaxine. CONCLUSIONS These findings indicated that intranasal venlafaxine-loaded poly(lactic-coglycolic acid) nanoparticles could become a viable technique for improving venlafaxine brain uptake via nose-to-brain. It could also be a promising nanoplatform for enhancing the treatment of depression.
Collapse
Affiliation(s)
- M Dolores Cayero-Otero
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | - Laura Perez-Caballero
- Department of Psychobiology, University of Granada, Campus de Cartuja, Granada, Spain
| | - Irene Suarez-Pereira
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Department of Psychology, University of Cadiz, Cádiz, Spain
| | - María Hidalgo-Figueroa
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Department of Psychology, University of Cadiz, Cádiz, Spain; Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain
| | - Alejandra Delgado-Sequera
- Neuropsychopharmacology & Psychobiology Research Group, Department of Psychology, University of Cadiz, Cádiz, Spain; Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain
| | | | - Esther Berrocoso
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Department of Psychology, University of Cadiz, Cádiz, Spain; Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain
| | - Lucía Martín-Banderas
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS)-Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain.
| |
Collapse
|
35
|
Araújo PS, Caixeta MB, Qualhato G, Gonçalves BB, Gomes RS, Viali EDSN, Viali WR, Rocha TL. Genotoxicity, mutagenicity, and immunotoxicity assessment of green silver nanoparticles from Croton urucurana on neotropical snails Biomphalaria glabrata. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 377:126424. [PMID: 40373859 DOI: 10.1016/j.envpol.2025.126424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 05/05/2025] [Accepted: 05/10/2025] [Indexed: 05/17/2025]
Abstract
Green silver nanoparticles (G-Ag NPs) have been indicated as safer nanotechnology in nanomedicine, especially for controlling microorganisms, vectors, and gastropods of medical importance. However, their mechanisms of action and ecotoxicity in aquatic organisms remain unknown. Thus, the current study aimed to investigate the effects of G-Ag NPs synthesized through Croton urucurana leave aqueous extract on the neotropical freshwater snail Biomphalaria glabrata after in vivo exposure using multi-biomarker responses in hemocytes. Adult snails were exposed to G-Ag NPs (0.05 and 0.14 mg L-1) or leave aqueous extract (5.4 mg L-1) for 7 days. Genotoxicity (comet assay), mutagenicity (micronucleus test and nuclear abnormality assay), and immunotoxicity (phagocytosis assay by flow cytometry) were analyzed in hemocytes. Results showed that G-Ag NPs induced high mortality and genotoxic effects (DNA damage) compared to leave aqueous extract and negative control groups. G-Ag NPs also induced mutagenic effects (nuclear alterations), mainly notched and blebbed nuclei. Also, G-Ag NPs and extract did not change the phagocytic activity and cell viability of hemocytes. These findings contribute to understanding the toxicity of green nanoparticles in freshwater gastropods and represent a pioneering effort to evaluate the effects of G-Ag NPs and the C. urucurana aqueous extract in B. glabrata hemocytes. Overall, the immune system of gastropods is a target of green nanoparticle toxicity.
Collapse
Affiliation(s)
- Paula Sampaio Araújo
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil; Laboratory of Applied Materials and Nanomaterials, Goiano Federal Institute of Education, Science and Technology, Rio Verde, Goiás, Brazil
| | - Maxwell Batista Caixeta
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil; Science and Technology Laboratory, Department of Medicine, Institute of Biotechnology, Federal University of Catalão, Catalão, Goiás, Brazil
| | - Gabriel Qualhato
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Bruno Bastos Gonçalves
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Rodrigo Saar Gomes
- Natural Immunity Laboratory, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Eloiza da Silva Nunes Viali
- Laboratory of Applied Materials and Nanomaterials, Goiano Federal Institute of Education, Science and Technology, Rio Verde, Goiás, Brazil
| | - Wesley Renato Viali
- Laboratory of Applied Materials and Nanomaterials, Goiano Federal Institute of Education, Science and Technology, Rio Verde, Goiás, Brazil
| | - Thiago Lopes Rocha
- Laboratory of Environmental Biotechnology and Ecotoxicology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
36
|
El-Kattan N, Ibrahim MA, Emam AN, Metwally K, Youssef FS, Nassar NA, Mansour AS. Evaluation of the antimicrobial activity of chitosan- and curcumin-capped copper oxide nanostructures against multi-drug-resistant microorganisms. NANOSCALE ADVANCES 2025; 7:2988-3007. [PMID: 40182310 PMCID: PMC11962744 DOI: 10.1039/d4na00955j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
The emergence of multi-drug-resistant microorganisms presents a serious threat to infection control, for which new antimicrobial strategies are urgently needed. Herein, the antimicrobial activities of copper oxide nanoparticles capped with curcumin (Cur-CuO NPs) and copper oxide nanoparticles capped with chitosan (CS-CuO NPs) were investigated. They were prepared via the co-precipitation method. A total of 180 clinical ICU patients were found to have 70% Gram-negative and 30% Gram-positive isolates. Antimicrobial susceptibility testing indicated resistance of these isolates to 14 among the 21 tested antibiotics. Physicochemical properties of the curcumin-capped (Cur-CuO NPs) and chitosan-capped (CS-CuO NPs) copper oxide nanoparticles were identified using UV-vis spectroscopy, transmission electron microscopy (TEM), dynamic light scattering (DLS), zeta-potential (ζ), and Fourier transform infrared (FT-IR) spectroscopy. Cur-CuO- and CS-CuO-NPs exhibited potent antimicrobial efficacy, wherein CS-CuO NPs were found to possess a lower minimum inhibitory concentration (MIC) (3.9-15.6 μg mL-1) than Cur-CuO NPs (14.5-31.2 μg mL-1). Biocompatibility assay showed that Cur-CuO NPs were safer with an IC50 dose of 74.17 μg mL-1 than CS-CuO NPs with an IC50 dose of 41.01 μg mL-1. Results revealed that the Cur-CuO- and CS-CuO-NPs have the potential to be safely used as effective antimicrobial agents in clinical applications at low concentrations (6.25-12.5 μg mL-1).
Collapse
Affiliation(s)
- Noura El-Kattan
- Department of Microbiology, Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes Giza Egypt
| | - Mostafa A Ibrahim
- Production and R&D Unit, NanoFab Technology Company 6th October City Giza Egypt
| | - Ahmed N Emam
- Refractories, Ceramics and Building Materials Department, Advanced Materials Technology & Mineral Resources Research Institute, National Research Centre (NRC) El Bohouth St., Dokki 12622 Cairo Egypt
- Nanomedicine & Tissue Engineering Research Lab, Medical Research Centre of Excellence, National Research Centre (NRC) El Bohouth St., Dokki 12622 Cairo Egypt
| | - Khaled Metwally
- Genetics Department, Faculty of Agriculture, Ain Shams University P.O. Box 68, Hadayek Shoubra 11241 Cairo Egypt
| | - Fady Sayed Youssef
- Department of Pharmacology Faculty of Veterinary Medicine, Cairo University 12211 Giza Egypt
| | | | - Ahmed S Mansour
- Department of Laser Applications in Meteorology, Chemistry and Agriculture, National Institute of Laser Enhanced Sciences (NILES), Cairo University Cairo Egypt
- Faculty of Postgraduate Studies for Nanotechnology, Cairo University Zayed City Giza Egypt
| |
Collapse
|
37
|
Roussel S, Carrera Fragoso L, Grenier P, Bruxelles Q, Chénard V, Marcoux S, Greffard K, Fortin S, Vallières L, Bertrand N. Glycerol-Based Polymer to Improve the Cellular Uptake of Liposomes. Biomacromolecules 2025; 26:2811-2824. [PMID: 40258592 PMCID: PMC12077401 DOI: 10.1021/acs.biomac.4c01110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/23/2025]
Abstract
Nanomedicines modify the pharmacology of pharmaceutical ingredients, but most require cell internalization to deliver their payloads. Hence, modifying the surface properties of nanomedicines can improve their interactions with cells and modulate their pharmacology. Herein, we devised a polymer that increases how nanomedicines are internalized by cells. The alkylated poly(monoglycerol acrylate) (PMGA) polymer was synthesized by reversible addition-fragmentation chain-transfer (RAFT) polymerization with a terminal double 18-carbon moiety that allows its anchoring on the surface of liposomes. PMGA-decorated liposomes are internalized more efficiently in immune cells, compared to formulations without the polymer. Using inhibitors of internalization pathways, we established that PMGA promotes cell entry by the fast endophilin-mediated endocytosis (FEME). In comparison, noncoated control liposomes were mostly internalized by clathrin-mediated endocytosis. This work highlights the potential of PMGA to increase the internalization of nanomedicines by immune cells, and target a novel internalization pathway.
Collapse
Affiliation(s)
- Sabrina Roussel
- Faculty
of Pharmacy, Université Laval, 1050 ave de la Médecine, Québec G1V 0A6, Canada
- Endocrinology
and Nephrology Unit, Centre de recherche
du CHU de Québec—Université Laval, CHUL building, 2705 Laurier Blvd, Québec G1V 4G2, Canada
| | - Lucia Carrera Fragoso
- Faculty
of Pharmacy, Université Laval, 1050 ave de la Médecine, Québec G1V 0A6, Canada
- Endocrinology
and Nephrology Unit, Centre de recherche
du CHU de Québec—Université Laval, CHUL building, 2705 Laurier Blvd, Québec G1V 4G2, Canada
| | - Philippe Grenier
- Faculty
of Pharmacy, Université Laval, 1050 ave de la Médecine, Québec G1V 0A6, Canada
| | - Quentin Bruxelles
- Faculty
of Pharmacy, Université Laval, 1050 ave de la Médecine, Québec G1V 0A6, Canada
- Oncology
Unit, Centre de recherche du CHU de Québec—Université
Laval, SFA building,
10 rue de l’Espinay, Québec G1L 3L5, Canada
| | - Valérie Chénard
- Endocrinology
and Nephrology Unit, Centre de recherche
du CHU de Québec—Université Laval, CHUL building, 2705 Laurier Blvd, Québec G1V 4G2, Canada
| | - Sébastien Marcoux
- Neurosciences
Unit, Centre de recherche du CHU de Québec—Université
Laval, CHUL building,
2705 Laurier Blvd, Québec G1V 4G2, Canada
| | - Karine Greffard
- Endocrinology
and Nephrology Unit, Centre de recherche
du CHU de Québec—Université Laval, CHUL building, 2705 Laurier Blvd, Québec G1V 4G2, Canada
| | - Sébastien Fortin
- Faculty
of Pharmacy, Université Laval, 1050 ave de la Médecine, Québec G1V 0A6, Canada
- Oncology
Unit, Centre de recherche du CHU de Québec—Université
Laval, SFA building,
10 rue de l’Espinay, Québec G1L 3L5, Canada
| | - Luc Vallières
- Department
of Microbiology and Immunology, Faculty of Medecine, Université Laval, 1050 ave de la Médecine, Québec G1V 0A6, Canada
- Neurosciences
Unit, Centre de recherche du CHU de Québec—Université
Laval, CHUL building,
2705 Laurier Blvd, Québec G1V 4G2, Canada
| | - Nicolas Bertrand
- Faculty
of Pharmacy, Université Laval, 1050 ave de la Médecine, Québec G1V 0A6, Canada
- Endocrinology
and Nephrology Unit, Centre de recherche
du CHU de Québec—Université Laval, CHUL building, 2705 Laurier Blvd, Québec G1V 4G2, Canada
| |
Collapse
|
38
|
Griffin S, de Oliveira Mallia J, Psakis G, Attard J, Caruana M, Gatt R. Comparative analysis of N/TERT-1 and N/TERT-2G keratinocyte responses to oxidative stress and immune challenges. Cell Signal 2025; 132:111861. [PMID: 40355015 DOI: 10.1016/j.cellsig.2025.111861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/05/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
The responses of N/TERT-1 and N/TERT-2G keratinocyte cell lines to oxidative stress and immune challenges were investigated to assess their suitability for dermatological testing. The cell lines were exposed to various stimuli, including PAMPs, DAMPs, H₂O₂, and menadione, to assess cytokine production, oxidative stress markers, cell viability, apoptosis, and membrane integrity. IL-1α, IL-6, IL-8, TNF-α, and TGF-β levels significantly increased in N/TERT-1 cells following exposure to LPS, while N/TERT-2G cells remained unaffected. Both cell lines showed increased production of IL-1α, IL-1β, TNF-α, IL-6, and IL-8 in response to dsDNA and LMW and HMW Poly I:C, although TGF-β significantly decreased only in N/TERT-1 cells. In response to H₂O₂, a dose-dependent increase in cytokine levels was observed in N/TERT-2G, whereas N/TERT-1 did not exhibit a clear dose-dependent response. Markers of oxidative stress, including SOD and GSH, displayed similar patterns in both cell lines, with N/TERT-2G showing slightly higher sensitivity. Lipid peroxidation and mitochondrial membrane potential fluctuations were more pronounced in N/TERT-2G, suggesting greater oxidative stress sensitivity. The baseline GSH levels were higher in N/TERT-1 cells, which may contribute towards the enhanced resilience to oxidative stress. Despite decreased viability in MTT assays following H₂O₂ exposure, the lack of significant changes in cleaved Caspase-3 levels indicated that apoptosis was not the primary mechanism of cell death. These findings highlight the distinct characteristics of N/TERT-1 and N/TERT-2G cells, with N/TERT-1 showing higher baseline resilience to oxidative stress and N/TERT-2G displaying greater sensitivity, particularly to H₂O₂. The study underscores the importance of selecting the appropriate cell line for specific research applications in skin biology and disease modelling, considering the differences in their responses to oxidative and immune challenges.
Collapse
Affiliation(s)
- Sholeem Griffin
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta.
| | - Jefferson de Oliveira Mallia
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| | - Georgios Psakis
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| | - Juan Attard
- Department of Food Sciences and Nutrition, Faculty of Health Sciences, University of Malta, Msida MSD2080, Malta
| | - Matthias Caruana
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta
| | - Ruben Gatt
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| |
Collapse
|
39
|
Dos Santos MG, Demonceaux M, Schimith LE, Goux M, Solleux C, Muccillo-Baisch AL, Arbo BD, Andre-Miral C, Hort MA. Pharmacokinetic Prediction and Cytotoxicity of New Quercetin Derivatives. Chem Biodivers 2025:e202500119. [PMID: 40345208 DOI: 10.1002/cbdv.202500119] [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: 01/10/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Quercetin (QUE) possesses various pharmacological properties; however, its low bioavailability and solubility hinder its beneficial effects. Enzymatic glycosylation has been explored to improve these aspects. In the present study, we used a sucrose phosphorylase variant to catalyze the regioselective transglucosylation of QUE, predicted the pharmacokinetic properties and toxicity of these molecules using in silico tools, and evaluated their cytotoxicity compared to the original molecule and a β-glucosylated derivative of QUE. Three α-glucosylated derivatives were obtained, which demonstrated improved pharmacokinetics, including a higher volume of distribution and lower clearance rate, with minimal likelihood of cytochrome P450 enzyme inhibition compared to QUE. QUE and the β-glucosylated derivative exhibited cytotoxicity in both cell types evaluated, whereas their α-glucosylated derivatives were nontoxic. The results presented provide an insight into the predicted behavior of these molecules in the body and, combined with cytotoxicity evaluation, will serve as a foundation for investigating the biological effects and mechanisms of action of these new molecules.
Collapse
Affiliation(s)
- Michele Goulart Dos Santos
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Marie Demonceaux
- Unit at the Biological Sciences at Biotechnologies, Nantes University, Nantes, France
| | - Lucia Emanueli Schimith
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Marine Goux
- Unit at the Biological Sciences at Biotechnologies, Nantes University, Nantes, France
| | - Claude Solleux
- Unit at the Biological Sciences at Biotechnologies, Nantes University, Nantes, France
| | - Ana Luiza Muccillo-Baisch
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Bruno Dutra Arbo
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Corinne Andre-Miral
- Unit at the Biological Sciences at Biotechnologies, Nantes University, Nantes, France
| | - Mariana Appel Hort
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| |
Collapse
|
40
|
Wun SJ, Tan L, Lonhienne TG, Low YS, Josh P, Kuo A, Smith MT, Gao Y, Pierens GK, Guddat LW, West NP. Florasulam Is a Potent Inhibitor of Mycobacterium tuberculosis Acetohydroxyacid Synthase and Possesses In Vivo Antituberculosis Activity. ACS Infect Dis 2025; 11:1180-1189. [PMID: 40214257 DOI: 10.1021/acsinfecdis.4c01028] [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] [Indexed: 05/10/2025]
Abstract
Tuberculosis (TB) remains as a leading cause of morbidity and mortality, accounting for ∼1.3 million fatalities worldwide per year. There are two major concerns: (i) the rise in the number of multi- and extensively drug-resistant strains of TB and (ii) the significant side-effects related to the use of many of the current therapies to treat drug-resistant and drug-sensitive TB alike. Thus, there is an ongoing need to discover new drugs and drug targets to combat this disease. Here, acetohydroxyacid synthase (AHAS), the first enzyme in the branched-chain amino acids (BCAAs) biosynthesis pathway, is comprehensively investigated as such a drug target. All five chemical classes of plant AHAS inhibitors, established as commercial herbicides, were assessed as leads. Members of the triazolopyrimidine family (e.g., metosulam, penoxsulam, and florasulam) are the most potent inhibitors of Mycobacterium tuberculosis AHAS (MtbAHAS) with Ki values as low as 20 nM. These compounds also exhibit the property of accumulative time-dependent inhibition, a feature that appears to be crucial for herbicidal activity and more generally for biocidal activity. Of these, the anti-TB activity of florasulam was the most effective, with an MIC of 500 nM against virulent Mtb grown in culture. This compound is also effective in killing intramacrophage Mtb and reduces bacterial load, as compared to vehicle-only by 13-fold in the lungs of mice infected with Mtb. Thus, triazolopyrimidines as AHAS inhibitors, and in-particular florasulam, represents a promising new class of leads for anti-TB drug development.
Collapse
Affiliation(s)
- Shun Jie Wun
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Thierry G Lonhienne
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Yu Shang Low
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Peter Josh
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Andy Kuo
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Maree T Smith
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Yanhua Gao
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Gregory K Pierens
- UQ Centre for Advanced Imaging, St Lucia, Queensland 4072, Australia
| | - Luke W Guddat
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas P West
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
41
|
Fadaly WAA, Nemr MTM, Abd El-Hameed AM, Mohamed FEA, Zidan TH. Design and Synthesis of New pyrazole Hybrids Linked to Oxime and Nitrate Moieties as COX-2, EGFR L858R/T790M Inhibitors and Nitric Oxide Donors with dual Anti-inflammatory/Anti-proliferative Activities. Bioorg Chem 2025; 161:108563. [PMID: 40349531 DOI: 10.1016/j.bioorg.2025.108563] [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: 01/08/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Two new series of pyrazole derivatives 14a-l and 17a-c with oxime/nitrate moieties as EGFRWT, mutant (EGFRL858R/T790M) and COX-2 inhibitors were synthesized and evaluated for anti-proliferative and anti-inflammatory activities. Compounds 14c, 14e, 14 g, 14i-l, 17b and 17c exhibited COX-2 selectivity in the range of (S.I. = 17-42) when compared to celecoxib (S.I. = 20.43). Concerning anti-neoplastic activity, screening was carried out against 60 human cancer cell lines by (NCI); Nine compounds (14c, 14e, 14 g, 14i-l, 17b and 17c) showed excellent inhibitory activity against all cancer cell lines especially non-small cell lung cancer (NSCLC). Further cytotoxicity testing of compounds 14c, 14e, 14 g, 14i-l, 17b and 17c was conducted on established EGFRT790M/L858R-resistant NSCLC (H1975), all tested compounds except 14 l exhibited potent activity (IC50 = 3.02-27.32 μM) which is higher than that of osimertinib (IC50 = 37.29 μM). It was noted that compound 17c, showed cell cycle arrest at G0/G1 phase of NSCLC (H1975) cells. In addition, compounds 14c, 14e, 14 g, 14i-l, 17b and 17c induced improved selective inhibitory activity against double mutant EGFRL858R/T790M tyrosine kinases with IC50 in the range of (0.031-0.076 μM, with selectivity index range S.I. of 2.5-14.58) which was comparable to that of osimertinib (IC50 = 0.037 μM, with S.I. of 1.89). The most potent anti-cancer compounds 14c, 14e, 14 g, 14i-l, 17b and 17c released NO in a slow rate of (1.45-3.37 %). Finally, applying covalent docking, we identified the covalent binding of 14 g, 14 k, and 17c with Cys797, providing insights into their potential as irreversible inhibitors targeting EGFRL858R/T790M protein.
Collapse
Affiliation(s)
- Wael A A Fadaly
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini Street, 11562 Cairo, Egypt.
| | - Abeer M Abd El-Hameed
- Chemistry Department, Faculty of Science, Taibah University, P.O. BOX 30002, Al-Madinah, Al-Munawarah 14177, Saudi Arabia
| | - Fatma E A Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Taha H Zidan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
42
|
Costa ÍB, Cruz FG, Boness HVM, Marques E, Borges JMP, de Faria Lopes GP, da Silva VDA, Estrela-Lima A, Dos Santos El-Bachá R. Coumarin MAMMEA A/BB cytotoxicity inhibits the chemoresistance and migration of glioblastoma cells in vitro. Fitoterapia 2025; 184:106607. [PMID: 40348030 DOI: 10.1016/j.fitote.2025.106607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
High-grade gliomas are the most aggressive brain tumors, which have no effective treatment. This work investigated a new anti-glioma strategy using mammea A/BB in vitro, a 4-phenylcoumarin isolated from the roots of Kielmeyera argentea. This work evaluated the cytotoxicity of mammea A/BB to human glioblastoma (U251), rat glioma (C6) cells and rat astrocytes in primary culture, comparing to temozolomide (TMZ) by MTT test. Cell migration assay, morphological analysis of DAPI-labeled nuclei and immunofluorescence for P-glycoprotein (P-gp) were also performed. After 72 h, the mammea A/BB significantly induced cytotoxicity in a concentration-dependent manner in U251 and C6 cells, with the EC50 27 ± 2 μM and 57 ± 14 μM, respectively. The natural compound was not cytotoxic to astrocytes in primary culture up to 200 μM. It was possible to observe a significant inhibition of tumoral cell migration in treatments with 10 mM mammea A/BB. Both cell lines were resistant to TMZ, but significantly sensitive to mammea A/BB. The percentage of picnotic nuclei of cells treated with 30 mM mammea A/BB was higher than the control. Besides, the treatment with mammea A/BB showed no significant difference in P-gp expression, but it was increased in TMZ treatment after 72 h. Even with cell lines presenting different molecular profiles, the results indicate that mammea A/BB is a promising candidate as a new antitumor drug against glioma cells in vitro.
Collapse
Affiliation(s)
- Ísis Barbosa Costa
- Postgraduate in Animal Science in the Tropics, Federal University of Bahia (UFBA), Salvador, BA, 40110-902, Brazil
| | - Frederico Guaré Cruz
- Institute of Chemistry, Federal University of Bahia (UFBA), Salvador, BA 40110-902, Brazil
| | | | - Edson Marques
- State University of Bahia (UNEB), Salvador, BA 41150-000, Brazil
| | - Julita Maria Pereira Borges
- Department of Sciences of Health, Southwestern Bahia State University (UESB), Vitória da Conquista, BA 45083-900, Brazil
| | | | - Victor Diogenes Amaral da Silva
- Department of Biochemistry and Biophysics; Neurochemistry and Cell Biology Laboratory (LabNq), Institute of Sciences of Health, Federal University of Bahia (UFBA), Salvador, BA 40110-902, Brazil
| | - Alessandra Estrela-Lima
- Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia (UFBA), Salvador, BA 40170-110, Brazil.
| | - Ramon Dos Santos El-Bachá
- Department of Biochemistry and Biophysics; Neurochemistry and Cell Biology Laboratory (LabNq), Institute of Sciences of Health, Federal University of Bahia (UFBA), Salvador, BA 40110-902, Brazil
| |
Collapse
|
43
|
Bortnevskaya YS, Zakharov NS, Senkov VS, Gradova MA, Karpechenko NY, Nikolskaya ED, Mollaeva MR, Yabbarov NG, Novikov AS, Bragina NA, Zhdanova KA. Synthesis, Molecular Docking, and Biological Activity of New EGFR-Targeted Photosensitizers Based on Cationic Porphyrins Encapsulated into Pluronic F127 Micelles. Mol Pharm 2025. [PMID: 40340474 DOI: 10.1021/acs.molpharmaceut.5c00247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The development of new effective photosensitizers (PS) for photodynamic therapy (PDT) is one of the important tasks in medical and organic chemistry. PSs inhibiting epidermal growth factor receptors (EGFR) overexpressed in cancer cells are of particular importance. In this work, we proposed the design and molecular docking of novel hybrid photosensitizers based on meso-aryl-substituted porphyrins and the Erlotinib molecule, a clinically approved tyrosine kinase inhibitor. The spacer length between the macrocycles and Erlotinib, hydrophilicity, and hydrophobicity of the porphyrin ring substituents were varied in the obtained compounds to evaluate structure-activity relationships (SAR). Photophysical and photochemical characteristics were studied for all of the received compounds in the presence of solubilizers suitable for the creation of dosage forms. Nanomicelles based on Pluronic F127 were obtained and characterized for the received compounds. In vitro biological tests on three cancer cell lines, MCF-7 (breast carcinoma), A431 (epidermoid carcinoma), MDA-MB-231 (breast adenocarcinoma), and normal NKE cells (human kidney epithelial cells) were performed, which showed low dark toxicity as well as light-induced activity of conjugates in the nanomolar range. Confocal microscopy experiments showed preferred accumulation of UB-2 and a lower accumulation of UB-3 PSs. In the case of UB-3, we observed a pronounced colocalization with early endosome antigen (EEA1). Also, cell apoptosis and inhibition of phosphorylation of EGFR were demonstrated for the UB-3 compound. Thus, the proposed design of targeting PS containing cationic pyridyl moieties and a linker between the porphyrin macrocycle and Erlotinib can contribute to antitumor PDT.
Collapse
Affiliation(s)
- Yulia S Bortnevskaya
- MIREA - Russian Technological University, Institute of Fine Chemical Technology, Vernadsky pr., 86, 119571 Moscow, Russian Federation
| | - Nikita S Zakharov
- MIREA - Russian Technological University, Institute of Fine Chemical Technology, Vernadsky pr., 86, 119571 Moscow, Russian Federation
| | - Vadim S Senkov
- MIREA - Russian Technological University, Institute of Fine Chemical Technology, Vernadsky pr., 86, 119571 Moscow, Russian Federation
| | - Margarita A Gradova
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Kosygin Street 4, 119991 Moscow, Russia
| | - Natalia Yu Karpechenko
- MIREA - Russian Technological University, Institute of Fine Chemical Technology, Vernadsky pr., 86, 119571 Moscow, Russian Federation
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe Highway, 24, 115522 Moscow, Russia
- Pirogov National Research Medical University, Ministry of Health of Russia, 1 Ostrovityanova St., 117997 Moscow, Russia
| | - Elena D Nikolskaya
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygina Street, 4, 119334 Moscow, Russia
| | - Mariia R Mollaeva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygina Street, 4, 119334 Moscow, Russia
| | - Nikita G Yabbarov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygina Street, 4, 119334 Moscow, Russia
| | - Alexander S Novikov
- Institute of Chemistry, Saint Petersburg State University, Universitetskaya nab. 7-9, 199034 Saint Petersburg, Russia
- Research Institute of Chemistry, Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya Street, 6, 117198 Moscow, Russia
| | - Natal'ya A Bragina
- MIREA - Russian Technological University, Institute of Fine Chemical Technology, Vernadsky pr., 86, 119571 Moscow, Russian Federation
| | - Kseniya A Zhdanova
- MIREA - Russian Technological University, Institute of Fine Chemical Technology, Vernadsky pr., 86, 119571 Moscow, Russian Federation
| |
Collapse
|
44
|
Stanková K, Ondrejkovičová G, Martinková M, Pilátová MB, Kuchár J, Litecká M. A simple approach to orthogonally protected 2,3-diaminopropanols: intermediates for l- and d-Dap-containing peptidomimetics. Carbohydr Res 2025; 553:109512. [PMID: 40367840 DOI: 10.1016/j.carres.2025.109512] [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: 04/04/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025]
Abstract
A simple approach to orthogonally protected 2,3-diaminopropanols from easily available d- and l-erythrofuranose has been accomplished. The pivotal step involved a cascade Overman rearrangement to install two novel C-N bonds and create the desired diamino alcohol motif, while a one-pot ozonolysis/reductive workup accompanied by the rational execution of suitable functional group transformations allowed the carbon skeleton of the target compounds to be completed. A series of the synthesised diaminopropanols was evaluated regarding their capacity to alter proliferation of selected cancer cell lines.
Collapse
Affiliation(s)
- Kvetoslava Stanková
- Institute of Chemical Sciences, Department of Organic Chemistry, P.J. Šafárik University, Moyzesova 11, 040 01, Košice, Slovak Republic
| | - Gabriela Ondrejkovičová
- Institute of Chemical Sciences, Department of Organic Chemistry, P.J. Šafárik University, Moyzesova 11, 040 01, Košice, Slovak Republic
| | - Miroslava Martinková
- Institute of Chemical Sciences, Department of Organic Chemistry, P.J. Šafárik University, Moyzesova 11, 040 01, Košice, Slovak Republic.
| | - Martina Bago Pilátová
- Institute of Pharmacology, Faculty of Medicine, P.J. Šafárik University, SNP 1, 040 66, Košice, Slovak Republic
| | - Juraj Kuchár
- Institute of Chemical Sciences, Department of Inorganic Chemistry, P.J. Šafárik University, Moyzesova 11, 040 01, Košice, Slovak Republic
| | - Miroslava Litecká
- Institute of Inorganic Chemistry of the Czech Academy of Sciences, Husinec-Řež 1001, 25068, Řež, Czech Republic
| |
Collapse
|
45
|
Islam M, Barakat A, Alayyaf AMA, Haukka M, Verma VP, Abu-Serie MM, El-Yazbi AF, Shehat MG, Alseqely M, Teleb M. Synthesis of Marine-Inspired Multifaceted DNA Damaging Spirooxindoles Combating NSCLC and Associated Bacterial Infection. ACS Med Chem Lett 2025; 16:819-828. [PMID: 40365419 PMCID: PMC12067117 DOI: 10.1021/acsmedchemlett.5c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/03/2025] Open
Abstract
Targeted therapeutics have gained prominence in combating non-small cell lung carcinoma (NSCLC) and opportunistic bacterial infections like Staphylococcus aureus (S. aureus). This study explores dual-acting marine-inspired spirooxindoles to limit NSCLC and opportunistic bacteria. Pharmacophoric motifs from antitumor and antibacterial marine products were merged into a new series of pyrazole-clubbed spirooxindoles via a stereoselective [3 + 2] cycloaddition reaction. MTT screening identified 4e, 4i, and 4p-4s as potent cytotoxic agents, with 4p showing exceptional activity (IC50 = 0.042 μM) and tumor selectivity (SI = 58.28). 4p exhibited antibacterial efficacy against S. aureus (MIC = 25 μg/mL). DNA damage studies using a terbium(III) chloride biosensor revealed 4p's ability to damage both calf thymus and S. aureus DNA at low concentrations. Docking simulations presumed that 4p binds between DNA strands, while apoptosis studies indicated it induced G1/S phase cell cycle arrest and increased A549 apoptosis by 33.65%. These findings highlight 4p as a promising lead for further studies.
Collapse
Affiliation(s)
- Mohammad
Shahidul Islam
- Department
of Chemistry, College of Science, King Saud
University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Assem Barakat
- Department
of Chemistry, College of Science, King Saud
University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | | | - Matti Haukka
- Department
of Chemistry, University of Jyväskylä, P. O. Box 35, FI-40014 Jyväskylä, Finland
| | - Ved Prakash Verma
- Department
of Education in Science Mathematics, RIE, Bhubaneswar 751022, India
| | - Marwa M. Abu-Serie
- Medical
Biotechnology Department, Genetic Engineering and Biotechnology Research
Institute, City of Scientific Research and
Technological Applications (SRTA-City), Alexandria 21934, Egypt
| | - Amira F. El-Yazbi
- Department
of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Michael G. Shehat
- Department
of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, 21521 Alexandria, Egypt
| | - Mustafa Alseqely
- Department
of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, 21521 Alexandria, Egypt
| | - Mohamed Teleb
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Alamein City 5060310, Egypt
| |
Collapse
|
46
|
Bunnell AA, Marshall EM, Estes SK, Deadmond MC, Loesgen S, Strother JA. Embryonic Zebrafish Irritant-evoked Hyperlocomotion (EZIH) as a high-throughput behavioral model for nociception. Behav Brain Res 2025; 485:115526. [PMID: 40057202 DOI: 10.1016/j.bbr.2025.115526] [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: 11/01/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Behavioral models have served a key role in understanding nociception, the sensory system by which animals detect noxious stimuli in their environment. Developing zebrafish (Danio rerio) are a powerful study organism for examining nociceptive pathways, given the vast array of genetic, developmental, and neuroscience tools available for these animals. However, at present there are few widely-adopted behavioral models for nociception in developing zebrafish. This study examines the locomotor response of hatching-stage zebrafish embryos to dilute solutions of the noxious chemical and TRPA1 agonist allyl isothiocyanate (AITC). At this developmental stage, AITC exposure induces a robust uniphasic hyperlocomotion response. This behavior was thoroughly characterized by determining the effects of pre-treatment with an array of pharmacological agents, including anesthetics, TRPA1 agonists/antagonists, opioids, NSAIDs, benzodiazepines, SSRIs, and SNRIs. Anesthetics suppressed the response to AITC, pre-treatment with TRPA1 agonists induced hyperlocomotion and blunted the response to subsequent AITC exposures, and TRPA1 antagonists and the opioid buprenorphine tended to reduce the response to AITC. The behavioral responses of zebrafish embryos to a noxious chemical were minimally affected by the other pharmacological agents examined. The feasibility of using this behavioral model as a screening platform for drug discovery efforts was then evaluated by assaying a library of natural product mixtures from microbial extracts and fractions. Overall, our results indicate that irritant-evoked locomotion in embryonic zebrafish is a robust behavioral model for nociception with substantial potential for examining the molecular and cellular pathways associated with nociception and for drug discovery efforts.
Collapse
Affiliation(s)
- Amelia A Bunnell
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, United States
| | - Erin M Marshall
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, United States
| | | | - Monica C Deadmond
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, United States
| | - Sandra Loesgen
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, United States
| | - James A Strother
- Whitney Laboratory for Marine Bioscience, University of Florida, Saint Augustine, FL, United States; Oregon State University, Corvallis, OR, United States.
| |
Collapse
|
47
|
Nai S, Gaslonde T, Krystyańczuk J, Amand S, Picot L, Veloso CAG, Buisson D, Lallemand MC, de Oliveira Junior RG. New bis-norditerpene by biotransformation of favelines from Cnidoscolus quercifolius using Umbelopsis isabellina as biocatalyst. Fitoterapia 2025; 184:106605. [PMID: 40348031 DOI: 10.1016/j.fitote.2025.106605] [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: 01/28/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Favelines, tricyclic benzylcycloheptene diterpenes predominantly found in Cnidoscolus quercifolius, have demonstrated significant cytotoxic activity against human cancer cell lines, including melanoma. In this study, we employed a combinatorial approach to screen microbial strains for biotransformation, focusing on regioselective oxidation of the cyclohexyl ring in the faveline core structure. This strategy, applied directly to the crude plant extract, enabled the rapid identification of Umbelopsis isabellina as the active strain. Subsequent preparative biotransformation yielded a novel bis-norditerpene, 14-hydroxy-isofavelol (3). The new compound exhibited moderate cytotoxic activity (IC₅₀ = 68.8 μM) against chemoresistant human melanoma A2058 cells, which are resistant to dacarbazine but sensitive to vemurafenib. Compound 3 enhanced the cytotoxic effect of vemurafenib, reducing its IC₅₀ from 24.8 to 9.63 μM. This work demonstrates the utility of microbial biotransformation for generating new bioactive natural product derivatives and highlights the potential of 14-hydroxy-isofavelol as a chemosensitizing agent in melanoma therapy.
Collapse
Affiliation(s)
- Sovannychloé Nai
- CiTCoM UMR 8038 CNRS, Faculté de Pharmacie, Université Paris Cité, 75006 Paris, France
| | - Thomas Gaslonde
- CiTCoM UMR 8038 CNRS, Faculté de Pharmacie, Université Paris Cité, 75006 Paris, France
| | - Julia Krystyańczuk
- CiTCoM UMR 8038 CNRS, Faculté de Pharmacie, Université Paris Cité, 75006 Paris, France
| | - Séverine Amand
- MCAM UMR 7245 CNRS, Muséum National d'Histoire Naturelle (MNHN), 75005 Paris, France
| | - Laurent Picot
- LIENSs UMRi 7266 CNRS, La Rochelle Université, 17000 La Rochelle, France
| | | | - Didier Buisson
- MCAM UMR 7245 CNRS, Muséum National d'Histoire Naturelle (MNHN), 75005 Paris, France
| | | | | |
Collapse
|
48
|
Bakhet S, Mardosaitė R, Ahmed Baba M, Tamulevičienė A, Abakevičienė B, Klinavičius T, Dagilis K, Račkauskas S, Tamulevičius S, Lelešius R, Zienius D, Šalomskas A, Šmits K, Tamulevičius T. Virucidal Efficacy of Laser-Generated Copper Nanoparticle Coatings against Model Coronavirus and Herpesvirus. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26431-26444. [PMID: 40263124 PMCID: PMC12067380 DOI: 10.1021/acsami.5c03330] [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] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
High-efficiency antiviral surfaces can be an effective means of fighting viral diseases, such as the recent COVID-19 pandemic. Copper and copper oxides, their nanoparticles (NPs) (CuNPs), and coatings are among the effective antiviral materials having internal and external biocidal effects on viruses. In this work, CuNP colloids were produced via femtosecond laser ablation of the metal target in water, a photophysical, cost-effective green synthesis alternative utilizing sodium citrate surfactant stabilizing the NPs. Raman spectroscopy and X-ray diffraction studies confirmed that the 32 nm mean size CuNPs are mixtures of mainly metallic copper and copper(I) oxide. Polyvinyl butyral was utilized as the binding agent for the CuNPs deposited via high-throughput spray-coating technology. The virucidal efficacy of such coatings containing Cu content ranging from 2.9 to 11.2 atom % was confirmed against animal-origin coronavirus containing ribonucleic acid, the agent of avian infectious bronchitis (IBV), and herpesvirus containing DNA, the agent of bovine herpesvirus (BoHV-1) infection. It was demonstrated that after a short time of exposure, the Cu NP-based coatings do not have a toxic effect on the cell cultures while demonstrating a negative effect on the biological activity of both model viruses that was confirmed by quantification of the viruses via the determination of tissue culture infectious dose (TCID50) virus titer and their viral nucleic acids via determination of threshold cycle (Ct) employing real-time polymerase chain reaction analysis. The assays showed that the decrease in TCID50 virus titer and increase in Ct values correlated with Cu content in Cu NP-based coatings for both investigated viruses. Contact with coatings decreased IBV and BoHV-1 numbers from 99.42% to 100.00% and from 98.65% to 99.96%, respectively. These findings suggest that CuNPs show inhibitory effects leading to the inactivation of viruses and their nuclei regardless of the presence of a viral envelope.
Collapse
Affiliation(s)
- Shahd Bakhet
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
| | - Rasa Mardosaitė
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
| | - Mohamed Ahmed Baba
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
| | - Asta Tamulevičienė
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
- Department
of Physics, Kaunas University of Technology, Studentų Street 50, LT-51368 Kaunas, Lithuania
| | - Brigita Abakevičienė
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
- Department
of Physics, Kaunas University of Technology, Studentų Street 50, LT-51368 Kaunas, Lithuania
| | - Tomas Klinavičius
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
| | - Kristupas Dagilis
- Department
of Physics, Kaunas University of Technology, Studentų Street 50, LT-51368 Kaunas, Lithuania
| | - Simas Račkauskas
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
| | - Sigitas Tamulevičius
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
- Department
of Physics, Kaunas University of Technology, Studentų Street 50, LT-51368 Kaunas, Lithuania
| | - Raimundas Lelešius
- Department
of Veterinary Pathobiology, Lithuanian University
of Health Sciences, Tilžės Street 18, LT-47181 Kaunas, Lithuania
- Institute
of Microbiology and Virology, Lithuanian
University of Health Sciences, Tilžės Street 18, LT-47181 Kaunas, Lithuania
| | - Dainius Zienius
- Department
of Veterinary Pathobiology, Lithuanian University
of Health Sciences, Tilžės Street 18, LT-47181 Kaunas, Lithuania
- Institute
of Microbiology and Virology, Lithuanian
University of Health Sciences, Tilžės Street 18, LT-47181 Kaunas, Lithuania
| | - Algirdas Šalomskas
- Department
of Veterinary Pathobiology, Lithuanian University
of Health Sciences, Tilžės Street 18, LT-47181 Kaunas, Lithuania
| | - Krišja̅nis Šmits
- Institute
of Solid State Physics, University of Latvia, 8 Kengaraga Street, LV-1063 Riga, Latvia
| | - Tomas Tamulevičius
- Institute
of Materials Science of Kaunas University of Technology, K. Baršausko Street 59, LT-51423 Kaunas, Lithuania
- Department
of Physics, Kaunas University of Technology, Studentų Street 50, LT-51368 Kaunas, Lithuania
| |
Collapse
|
49
|
Jena S, Subham K, Kalra H, Jha S. Multimeric interacting interface of biologically synthesized zinc oxide nanoparticle corona efficiently sequesters α-synuclein against protein fibrillation. Biomater Sci 2025. [PMID: 40332135 DOI: 10.1039/d5bm00143a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons along with the accumulation of amyloid plaques with alpha-synuclein (αS) as the major constituent. αS is an intrinsically disordered protein with the potential to undergo a cascade of structural transitions from a soluble disordered conformation to ordered cross-β-sheet-rich insoluble amyloid fibrils. Small molecules like polyphenols and peptides with anti-amyloidogenic potential can mitigate fibrillation in vitro but fail in vivo owing to poor bioavailability. To overcome this problem, a platform that simultaneously enhances the bioavailability of the mitigators and efficiently sequesters αS monomers against amyloidosis is needed. Accordingly, herein, the sequestering potential of surface-moderated zinc oxide nanoparticles was explored; in silico and in vitro studies showed that the moderated nano-interfaces efficiently sequestered αS in amorphous aggregates, which were termed as flocs. Moreover, GC-MS-based analysis of the bio-nano corona highlighted the rationale for efficient sequestering of αS monomers against amyloidosis by the biologically synthesized zinc oxide nanoparticle compared with other nanoparticle surfaces. Thus, this work exemplifies the multimeric interacting interface as a platform to efficiently sequester the αS protein and simultaneously enhance the bioavailability of the phytochemicals.
Collapse
Affiliation(s)
- Sonali Jena
- Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India.
| | - Kumari Subham
- Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India.
| | - Harshit Kalra
- Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India.
| | - Suman Jha
- Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India.
| |
Collapse
|
50
|
Sena-Júnior AS, Andrade CNS, Moura PHM, dos Santos JHC, Trancoso CT, Silva EED, Silva DMRR, Telles ÊP, Silva LAS, Teles ILD, de Almeida SFM, de Souza DA, Santos JF, Martins FJA, Silva AMDOE, Lauton-Santos S, de Araujo GRS, Correa CB, Nunes RDS, Borges LP, Lira AAM. A formulation containing Cymbopogon flexuosus essential oil: improvement of biochemical parameters and oxidative stress in diabetic rats. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:617-636. [PMID: 40356885 PMCID: PMC12067095 DOI: 10.3762/bjnano.16.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/09/2025] [Indexed: 05/15/2025]
Abstract
Diabetes mellitus (DM) is a highly prevalent public health problem characterized by hyperglycemia that causes complications due to the generation of reactive oxygen species and oxidative damage. Studies have shown that essential oils containing citral, such as lemongrass, have various biological activities, including bactericidal, antiviral, antifungal, antioxidant, and hypoglycemic effects. Therefore, this study aims to obtain a microemulsified formulation containing the essential oil of Cymbopogon flexuosus (EOCF) and to evaluate its antioxidant and antidiabetic activity in diabetic rats. The microemulsion (ME) was obtained after consulting the corresponding pseudoternary phase diagram and showed stability, isotropy, Newtonian behavior, nanometric size (15.2 nm), and pH 4.2. Both EOCF and the ME showed high antioxidant activity, but the ME resulted in greater antioxidant activity, potentiating the activity of isolated EOCF. Finally, male Wistar rats (3 months old, 200-250 g) with streptozotocin-induced type-1 DM were supplemented with EOCF and ME (32 mg/kg) for 21 days. Both EOCF and ME supplementation resulted in reduced blood glucose levels and improved lipid profiles when compared to the control group. Additionally, the ME was able to provide additional benefits, such as reduced liver damage, improved renal function, reduced systemic inflammation, and increased high-density lipoprotein levels. Overall, the results show that EOCF was efficiently incorporated into the microemulsion, improving its antioxidant activity and showing promising results for use in the treatment of DM via the oral route.
Collapse
Affiliation(s)
- Ailton Santos Sena-Júnior
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | | | - Pedro Henrique Macedo Moura
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | | | - Cauãn Torres Trancoso
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | | | | | - Ênio Pereira Telles
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Luiz André Santos Silva
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Isabella Lima Dantas Teles
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | | | - Daniel Alves de Souza
- Department of Physiology, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Jileno Ferreira Santos
- Department of Morphology, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Felipe José Aidar Martins
- Department of Physical Education, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | | | - Sandra Lauton-Santos
- Department of Physiology, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | | | - Cristiane Bani Correa
- Department of Morphology, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Rogéria De Souza Nunes
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Lysandro Pinto Borges
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Ana Amélia Moreira Lira
- Department of Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| |
Collapse
|