101
|
Pereira CM, Shimizu JF, Cassani NM, Santos IA, Bittar C, Oliveira Cintra AC, Sampaio SV, Harris M, Rahal P, Gomes Jardim AC. Bothropstoxins I and II as potent phospholipase A2 molecules from Bothrops jararacussu to impair Hepatitis C virus infection. Biochimie 2025:S0300-9084(25)00081-1. [PMID: 40288437 DOI: 10.1016/j.biochi.2025.04.006] [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: 06/25/2024] [Revised: 03/26/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Hepatitis C is a hepatological disorder induced by the Hepacivirus hominis (Hepatitis C virus, HCV), with approximately 170 million individuals estimated to be presently affected globally. The current treatment for infected patients primarily relies on direct-acting antivirals (DAAs). However, this treatment is marked by its high cost, numerous side effects, and documented instances of antiviral resistance. These challenges underscore the imperative for developing novel therapeutic strategies. In this framework, naturally occurring compounds have exhibited considerable medical significance attributable to their biological functionalities. Compounds extracted from snake venoms have evidenced antiviral efficacy against a variety of viral pathogens including Orthoflavivirus denguei (DENV), Orthoflavivirus flavi (YFV), Orthoflavivirus zikaense (ZIKV), and HCV. Here, the activity of 10 proteins isolated from snakes' venom of Bothrops genus were evaluated against HCV replicative cycle. The full-length JFH-1 HCV system was used to infect the Huh-7.5 cell. Cell viability was measured simultaneously through MTT assay. Eight compounds inhibited up to 99% of HCV infection, being the most potent inhibitory rates observed in BthTX-I and BthTX-II, with an SI of 13.5 and 1736, respectively, being able to block 84.7% and 96% of HCV infectivity, in the same order. BthTX-II also demonstrated a protective effect in cells treated prior to HCV infection of approximately 86.7%. Molecular docking calculations suggest interactions between the two proteins with HCV E1-E2 glycoprotein complex. BthTX-II exhibited stronger interactions, indicated by 22 hydrophobic interactions. In conclusion, these compounds were shown to inhibit HCV infectivity by either acting on the virus particles or protecting the cells against infection.
Collapse
Affiliation(s)
- Carina Machado Pereira
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Jacqueline Farinha Shimizu
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil; Laboratory of Antiviral Research, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Natasha Marques Cassani
- Laboratory of Antiviral Research, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Igor Andrade Santos
- Laboratory of Antiviral Research, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Cintia Bittar
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | | | - Suely Vilela Sampaio
- Laboratory of Toxinology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Paula Rahal
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Ana Carolina Gomes Jardim
- Genomics Study Laboratory, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil; Laboratory of Antiviral Research, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
102
|
Abed T, Aly SH, Salim SA, Haikal RR, Shams-Eldin R, El-Moslamy SH, Abdelazim EB, Helmy MS, Ali AA, Eissa NG, Elnakady YA, Elsabahy M, Kamoun EA. Optimizing hydrogel performance composed of Japanese pagoda tree extract loaded-gelatin-sodium alginate-polyethylene oxide for biomedical applications: Influence incorporated calcium-based metal organic frameworks and zinc oxide NPs. Int J Biol Macromol 2025; 310:143526. [PMID: 40288718 DOI: 10.1016/j.ijbiomac.2025.143526] [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: 02/21/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Gelatin/sodium alginate/polyethylene oxide hydrogel was synthesized by self-gelation method and utilized to achieve enhanced mechanical strength. Japanese pagoda, tree extracts rich in flavonoids and isoflavonoids was incorporated to enhance tissue repair along with ZnO-NPs to provide the hydrogel with necessary antimicrobial and other desirable biological properties. The inclusion of Ca-BDC MOF enhanced hydrogel matrix by providing more surface area and the presence of calcium ion needed for tissue regeneration. Results revealed that composite hydrogel displayed suitable gelation time and swelling rate that was further improved by incorporating the plant extract, ZnO-NPs, and Ca-BDC MOF. The efficacy of the tested formulations to suppress or stop the growth of multi-drug-resistant human pathogens was assessed using both microbial turbidity and zone of inhibition assays. The optimal hydrogel loaded with (20 % plant extract, 0.25 % ZnO and 0.1 % Ca-BDC MOF) formula have stronger antimicrobial effects against Gram-positive bacteria as well as yeast cells than it did against Gram-negative bacteria. Statistically, this formula has recorded a higher percentage of biofilm inhibition on Bacillus cereus (93.45 ± 2.91 %), and Candida krusei (78.48 ± 6.44 %), followed by Klebsiella pneumoniae (76.93 ± 3.65 %). Composite hydrogel loaded with (Japanese pagoda tree extract, ZnO-NPs, Ca-BDC MOF) could be exploited as promising biocompatible antimicrobial biomaterial for biomedical applications.
Collapse
Affiliation(s)
- Tasneem Abed
- Badr University in Cairo Research Center, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Shaza H Aly
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Samar A Salim
- Polymeric and Biomaterials for Medical and Pharmaceutical Applications Research Group, Nanotechnology Research Center (NTRC), The British University in Egypt (BUE), El-Sherouk City, Cairo 11837, Egypt.
| | - Rana R Haikal
- Center for Materials Science, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Reham Shams-Eldin
- Center for Materials Science, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Shahira H El-Moslamy
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg Al-Arab City, 21934, Alexandria, Egypt
| | - Esraa B Abdelazim
- Badr University in Cairo Research Center, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Mohamed S Helmy
- Molecular Biology Department, National Research Centre, El-Tahrir St, Dokki, Giza, Egypt; Proteome Research Laboratory, Central Laboratories Network and Centres of Excellence, National Research Centre, El-Tahrir St, Dokki, Giza, Egypt
| | - Ahmed A Ali
- Molecular Biology Department, National Research Centre, El-Tahrir St, Dokki, Giza, Egypt
| | - Noura G Eissa
- Badr University in Cairo Research Center, Badr University in Cairo, Badr City, Cairo 11829, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Yasser A Elnakady
- Department of Zoology, College of Science, King Saud University, B.O. Box 2455, 11415 Riyadh, Saudi Arabia
| | - Mahmoud Elsabahy
- Badr University in Cairo Research Center, Badr University in Cairo, Badr City, Cairo 11829, Egypt; Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Elbadawy A Kamoun
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia..
| |
Collapse
|
103
|
Chao HWH, Chao WWJ, Chao HM. Catalpol Protects Against Retinal Ischemia Through Antioxidation, Anti-Ischemia, Downregulation of β-Catenin, VEGF, and Angiopoietin-2: In Vitro and In Vivo Studies. Int J Mol Sci 2025; 26:4019. [PMID: 40362263 PMCID: PMC12072090 DOI: 10.3390/ijms26094019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Retinal ischemic disorders present significant threats to vision, characterized by inadequate blood supply oxygen-glucose deprivation (OGD), oxidative stress, and cellular injury, often resulting in irreversible injury. Catalpol, an iridoid glycoside derived from Rehmannia glutinosa, has demonstrated antioxidative and neuroprotective effects. This study aimed at investigating the protective effects and mechanisms of catalpol against oxidative stress or OGD in vitro and retinal ischemia in vivo, focusing on the modulation of key biomarkers of retinal ischemia, including HIF-1α, vascular endothelial growth factor (VEGF), angiopoietin-2, MCP-1, and the Wnt/β-catenin pathway. Cellular viability was assessed using retinal ganglion cell-5 (RGC-5) cells cultured in DMEM; a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed. H2O2 (1 mM)/OGD was utilized. Vehicle or different catalpol concentrations were administered 15 min before the ischemic-like insults. The Wistar rat eyes' intraocular pressure was increased to 120 mmHg for 60 min to induce retinal ischemia. Intravitreous injections of catalpol (0.5 or 0.25 mM), Wnt inhibitor DKK1 (1 μg/4 μL), anti-VEGF Lucentis (40 μg/4 μL), or anti-VEGF Eylea (160 μg/4 μL) were administered to the rats' eyes 15 min before or after retinal ischemia. Electroretinogram (ERG), fluorogold retrograde labeling RGC, Western blotting, ELISA, RT-PCR, and TUNEL were utilized. In vitro, both H2O2 and OGD models significantly (p < 0.001/p < 0.001; H2O2 and OGD) induced oxidative stress/ischemic-like insults, decreasing RGC-5 cell viability (from 100% to 55.14 ± 2.19%/60.84 ± 4.57%). These injuries were insignificantly (53.85 ± 1.28% at 0.25 mM)/(63.46 ± 3.30% at 0.25 mM) and significantly (p = 0.003/p = 0.012; 64.15 ± 2.41%/77.63 ± 8.59% at 0.5 mM) altered by the pre-administration of catalpol, indicating a possible antioxidative and anti-ischemic effect of 0.5 mM catalpol. In vivo, catalpol had less effect at 0.25 mM for ERG amplitude ratio (median [Q1, Q3] 14.75% [12.64%, 20.48%]) and RGC viability (mean ± SE 63.74 ± 5.13%), whereas (p < 0.05 and p < 0.05) at 0.5 mM ERG's ratio (35.43% [24.35%, 43.08%]) and RGC's density (74.34 ± 5.10%) blunted the ischemia-associated significant (p < 0.05 and p < 0.01) reduction in ERG b-wave amplitude (6.89% [4.24%, 10.40%]) and RGC cell viability (45.64 ± 3.02%). Catalpol 0.5 mM also significantly protected against retinal ischemia supported by the increased amplitude ratio of ERG a-wave and oscillatory potential, along with recovering a delayed a-/b-wave response time ratio. When contrasted with DKK1 or Lucentis, catalpol exhibited similar protective effects against retinal ischemia via significantly (p < 0.05) blunting the ischemia-induced overexpression of β-catenin, VEGF, or angiopoietin-2. Moreover, ischemia-associated significant increases in apoptotic cells in the inner retina, inflammatory biomarker MCP-1, and ischemic indicator HIF-1α were significantly nullified by catalpol. Catalpol demonstrated antiapoptotic, anti-inflammatory, anti-ischemic (in vivo retinal ischemia or in vitro OGD), and antioxidative (in vitro) properties, counteracting retinal ischemia via suppressing upstream Wnt/β-catenin and inhibiting downstream HIF-1α, VEGF, and angiopoietin-2, together with its decreasing TUNEL apoptotic cell number and inflammatory MCP-1 concentration.
Collapse
Affiliation(s)
- Howard Wen-Haur Chao
- Department of Medicine, School of Medicine, Aston University, Birmingham B4 7ET, UK
- Department of Medical Education, Leeds University, Leeds LS2 9JT, UK
| | - Windsor Wen-Jin Chao
- Department of Medicine, School of Medicine, Aston University, Birmingham B4 7ET, UK
- Department of Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Hsiao-Ming Chao
- Department of Chinese Medicine, School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Institute of Pharmacology, Department of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Department of Ophthalmology, Shin Kong Hospital, Taipei 105, Taiwan
| |
Collapse
|
104
|
Yoo NH, Baek YS, Kim HK, Lee CO, Kim MJ. Antioxidant and Anti-Inflammatory Activities of Astilboides tabularis (Hemsl.) Engl. Root Extract. Molecules 2025; 30:1892. [PMID: 40363699 PMCID: PMC12073835 DOI: 10.3390/molecules30091892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Here, we examined the antioxidant and anti-inflammatory activities of the ethyl acetate (EtOAc) fraction of Astilboides tabularis (Hemsl.) Engl. root extracts, initially prepared from a 70% ethanol extraction. This EtOAc fraction exhibited significant scavenging activity against DPPH radicals (IC50: 11.38 ± 0.48 µg/mL) and ABTS radicals (IC50: 7.46 ± 0.58 µg/mL), and had a high total phenolic content (i.e., 407.02 ± 13.56 mg GAE/g). In addition, the EtOAc fraction demonstrated concentration-dependent protective effects in a RAW264.7 macrophage cell model subjected to oxidative stress. In lipopolysaccharide (LPS)-stimulated RAW264.7 cells, nitric oxide (NO) production and the expression of inflammatory mediators (iNOS, COX-2, TNF-α, IL-1β, IFN-β) were inhibited in a concentration-dependent manner. Western blot and real-time PCR (RT-PCR) analyses revealed that the EtOAc fraction also suppressed inflammatory mediator expression via inhibiting the activation of the NF-κB and MAPK signaling pathways. Finally, LC-QTOF-MS and LC-MS/MS analyses identified gallic acid and bergenin as compounds contributing to observed antioxidant and anti-inflammatory effects. In conclusion, the EtOAc fraction of A. tabularis root extracts exhibited strong anti-oxidant and anti-inflammatory properties, suggesting potential usage for treating various inflammatory diseases.
Collapse
Affiliation(s)
- Nam Ho Yoo
- Department of Bio-Resource Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.H.Y.); (Y.S.B.)
| | - Young Sun Baek
- Department of Bio-Resource Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.H.Y.); (Y.S.B.)
| | - Hee Kyu Kim
- Gangwondo Forest Science Institute, Chuncheon 24207, Republic of Korea; (H.K.K.); (C.O.L.)
| | - Chan Ok Lee
- Gangwondo Forest Science Institute, Chuncheon 24207, Republic of Korea; (H.K.K.); (C.O.L.)
| | - Myong Jo Kim
- Department of Bio-Resource Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.H.Y.); (Y.S.B.)
| |
Collapse
|
105
|
Zemskaya AS, Arutyunyan AF, Sherman DK, Yanvarev DV, Shuvalov AV, Kalnina LB, Kaluzhny DN, Novikov RA, Solyev PN, Valuev-Elliston VT. Isolation of recombinant HIV-1 Rev protein and investigation of a new class of benzimidazole inhibitors capability to disrupt Rev-RRE complex. Bioorg Chem 2025; 161:108487. [PMID: 40288010 DOI: 10.1016/j.bioorg.2025.108487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
In the present study, an efficient method for the expression and purification of recombinant HIV Rev protein with a C-terminal hexahistidine tag was proposed. Noteworthy, this method circumvents the precipitation of the protein into inclusion bodies and their subsequent aggregation during purification. It does not necessitate denaturing isolation conditions, in contrast to currently widely used protocols. As a result, protocols for HIV Rev isolation have been developed allowing the production of non-aggregated Rev protein in a good yield, high purity, and free of bacterial RNA impurities. This high-purity result became possible due to high salt extraction buffer usage. Complementary [α-32P]-labeled Rev response element (RRE) RNA has been synthesized and an inhibitor test system was developed based on Rev-RRE complex formation. We were able to reveal a novel class of potential Rev-RRE inhibitors based on dimeric benzimidazole derivatives and used those results to validate the testing system. The proposed protocols for screening and structure-activity relationship for new inhibitors of Rev binding to viral RNA broaden the scope of potential candidates for anti-HIV drug development.
Collapse
Affiliation(s)
- Anastasia S Zemskaya
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia.
| | - Albert F Arutyunyan
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Daria K Sherman
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Dmitry V Yanvarev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Alexey V Shuvalov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Lyudmila B Kalnina
- Ivanovsky Institute of Virology, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health, 18 Gamaleya St, 123098 Moscow, Russia
| | - Dmitry N Kaluzhny
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Roman A Novikov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| | - Pavel N Solyev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia.
| | - Vladimir T Valuev-Elliston
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St, 119991 Moscow, Russia
| |
Collapse
|
106
|
Ibrahim AIO, Moodley D, Maboza E, Olivier A, Petrik L. Zeolite-Y-Loaded Chitosan Nanoparticles as Endodontic Antimicrobial Agent: An In vitro Study. Eur J Dent 2025. [PMID: 40267957 DOI: 10.1055/s-0045-1802947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVES The objectives were to synthesize a bioactive nanocomposite as an endodontic antimicrobial agent by loading previously synthesized electrosprayed chitosan nanoparticles (Ch-Np) into Zeolite-Y as a carrier and compare its antimicrobial activity against two endodontic pathogens using the agar diffusion test. Additionally, the effect of tissue inhibitors (dentin powder and serum albumin) on the antimicrobial activity of the Ch-Np-Zeolite nanocomposite was studied. Finally, the possible cytotoxicity of the novel nanocomposite against Balb/c 3T3 mouse fibroblast cells was evaluated. MATERIALS AND METHODS A concentration of 3% (w/v) electrosprayed Ch-Np was mixed with Zeolite-Y in a concentration of 53.3 (w/v) and characterized using high-resolution scanning electron microscopy (HR-SEM) and energy-dispersive X-ray spectroscopy (EDS) analysis. The antimicrobial activity was evaluated against Streptococcus mutans and Enterococcus faecalis using the agar diffusion test, and the time-kill test was performed using the broth microdilution technique in the presence of tissue inhibitors. The cytotoxicity was evaluated against 3T3 mouse fibroblast cells using the standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. STATISTICAL ANALYSIS The difference between the antimicrobial activity of Ch-Np-Zeolite nanocomposite against S. mutans and E. faecalis was analyzed using the Mann-Whitney's U test. The effect of tissue inhibitors on the antimicrobial activity of Ch-Np-Zeolite nanocomposite was analyzed by comparing the mean of log colony-forming unit per milliliter over time. For the cytotoxicity assay, a statistically significant difference between each group and their control was made using a t-test with a probability value of p ≤ 0.05, considered a significant difference. RESULTS HR-SEM of the dried paste-like mixture Ch-Np-Zeolite revealed the typical crystal habit of the supporting zeolite, and EDS analysis confirmed that the zeolite parent material retained its elemental composition after loading with Ch-Np. The antimicrobial activity of Ch-Np-Zeolite was demonstrated by the mean diameter inhibition zones of 9.57 and 7.85 mm for S. mutans and E. faecalis, respectively. Streptococcus mutans and E. faecalis were completely eradicated in the presence of tissue inhibitors. The Ch-Np-Zeolite nanocomposite significantly promoted the growth of 3T3 fibroblast cells (p < 0.05), supporting its lack of cytotoxicity. CONCLUSION Zeolite-Y-loaded Ch-Np nanocomposite shows promising antimicrobial activity while maintaining its biocompatibility even in the presence of tissue inhibitors.
Collapse
Affiliation(s)
- Amir Isam Omer Ibrahim
- Department of Restorative Dental Sciences, College of Dentistry, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Restorative Dentistry, Faculty of Dentistry, University of the Western Cape, Cape Town, South Africa
| | - Desigar Moodley
- Department of Restorative Dentistry, Faculty of Dentistry, University of the Western Cape, Cape Town, South Africa
- Edelweiss Dentistry Products Gmbh, Wolfurt, Austria
| | - Ernest Maboza
- WHO Collaborating Centre for Oral Health, Faculty of Dentistry, University of the Western Cape, Cape Town, South Africa
| | - Annette Olivier
- WHO Collaborating Centre for Oral Health, Faculty of Dentistry, University of the Western Cape, Cape Town, South Africa
| | - Leslie Petrik
- Department of Chemistry, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa
| |
Collapse
|
107
|
Franchin M, Saliba ASMC, Dos Santos Ramos A, Spada FP, Dos Santos DC, de Oliveira Leandro M, Da Campo Junqueira Gonçalves E, Bueno-Silva B, Xu Y, Wang K, Bennett J, Cunha TM, de Alencar SM, Granato D. Alginate-based microcapsules loaded with Brazilian green propolis decrease reactive oxygen species production, reduce inflammatory cytokines, and mitigate intestinal inflammation. Int J Biol Macromol 2025; 310:143357. [PMID: 40280527 DOI: 10.1016/j.ijbiomac.2025.143357] [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: 12/12/2024] [Revised: 03/31/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Ulcerative colitis causes intestinal inflammation, with treatments often limited in efficacy and safety. New technologies enable the controlled release of bioactive compounds, and Brazilian green propolis could benefit in managing inflammation. This study proposed developing alginate-based microcapsules loaded with ethanolic extract of green propolis (EEGP-MC), evaluating their effects on inflammatory cytokines, reactive oxygen species, and experimental colitis. The results demonstrated that the EEGP-MC reached peak release of phenolic compounds in the intestinal phase (IP) at 4 h (76.9 %) and 6 h (75.0 %). Similarly, Artepillin C peaked at 22.3 ± 1.2 mg/g at 4 h and 22.5 ± 1.1 mg/g at 6 h in the IP. In THP-1 cell cultures, pretreatment with EEGP-MC (1000 μg/mL) and IP (300 and 1000 μg/mL) reduced TNF-α and IL-6 levels and ROS production. Additionally, oral administration of EEGP-MC at 300 mg/kg demonstrated superior protective activity in the colonic mucosa, reducing lesions by 86.1 % compared to 54.9 % with EEGP alone. Finally, the treatment with EEGP-MC suppressed TNF-α, IL-6, and IL-1β cytokines in the intestinal tissue. No toxicity was observed for the EEGP-MC. These findings highlight EEGP-MC as an innovative technology with promising applications for managing chronic inflammation in the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Marcelo Franchin
- Bioactivity and Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, Limerick, Ireland; Graduate Program in Biological Sciences, Federal University of Alfenas (Unifal-MG), Alfenas, Brazil.
| | | | - Anderson Dos Santos Ramos
- Center for Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Fernanda Papa Spada
- Center for Nuclear Energy in Agriculture, University of São Paulo (USP), Piracicaba, SP, Brazil
| | - Daniela Coelho Dos Santos
- Center for Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Maísa de Oliveira Leandro
- Center for Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Bruno Bueno-Silva
- Departamento de Biociências, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, SP, Brasil
| | - Yandong Xu
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kai Wang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jason Bennett
- Bioactivity and Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, Limerick, Ireland
| | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Severino Matias de Alencar
- Center for Nuclear Energy in Agriculture, University of São Paulo (USP), Piracicaba, SP, Brazil; Agri-food Industry, Food and Nutrition Department, Luiz de Queiroz College of Agriculture, University of São Paulo, ESALQ/USP, Piracicaba, SP, Brazil
| | - Daniel Granato
- Bioactivity and Applications Laboratory, Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, Limerick, Ireland.
| |
Collapse
|
108
|
Demirhan HK, Omer Oglou E, Aksoy ZB, Kiran F. Evaluation of the anti-inflammatory, antioxidant and regenerative effects of microbiota-derived postbiotics in human periodontal ligament mesenchymal stromal cells. Clin Oral Investig 2025; 29:262. [PMID: 40263129 PMCID: PMC12014813 DOI: 10.1007/s00784-025-06341-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE This study investigates the regenerative and protective effects of postbiotics (cell-free supernatant) derived from the Lactiplantibacillus plantarum EIR/IF-1 strain on human periodontal ligament mesenchymal stromal cells (hPDL-MSCs). MATERIALS AND METHODS hPDL-MSCs were isolated from periodontal ligament tissues (PDL) of wisdom teeth using enzymatic digestion and subsequently characterized through immunophenotyping. The effect of postbiotics on the viability of hPDL-MSCs was assessed using the MTT assay and flow cytometry, while their impact on cell migration was evaluated via the scratch assay. Anti-inflammatory effects of postbiotics were investigated on lipopolysaccharide (LPS, derived from Porphyromonas gingivalis)-stimulated hPDL-MSCs through Enzyme-Linked Immunosorbent Assay (ELISA). Additionally, the antioxidant effects of postbiotics were analyzed in hydrogen peroxide (H₂O₂)-induced hPDL-MSCs by measuring reactive oxygen species (ROS) levels using flow cytometry. The expression of collagen type I (COL1A1) gene was further assessed by quantitative reverse transcription PCR and immunofluorescence staining. RESULTS Treatment with postbiotics (250 µg/mL) significantly increased the viability and migration capability of hPDL-MSCs, while enhancing collagen production for PDL repair. Treatment with postbiotics for 24 h resulted in a 54.53 ± 2.01% reduction in intracellular ROS levels compared to untreated H2O2-induced hPDL-MSCs. Furthermore, postbiotics significantly decreased the production of pro-inflammatory cytokines (IL-8, IL-6, and IL-1β), and increased the anti-inflammatory cytokine IL-10 (2.67-fold) compared to untreated LPS-stimulated hPDL-MSCs. CONCLUSION Our findings indicate that postbiotics exhibit biological activity throughout all stages of the healing process, beginning with the modulation of the inflammatory response to LPS stimulation, followed by the promotion of cell migration, proliferation, and collagen synthesis. Given the unmet need for safe and adjuvant therapeutic approaches that promote comprehensive periodontal regeneration in periodontal diseases, this study presents postbiotics as a promising candidate. CLINICAL RELEVANCE Postbiotics could be integrated into regenerative therapies as a novel bioactive material to improve the healing and regenerative outcomes in periodontal defects by both controlling inflammation and stimulating tissue repair processes.
Collapse
Affiliation(s)
- Hazal Kibar Demirhan
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara, 06100, Turkey
- Graduate School of Natural and Applied Sciences, Ankara University, Ankara, 06110, Turkey
| | - Emine Omer Oglou
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara, 06100, Turkey
- Graduate School of Natural and Applied Sciences, Ankara University, Ankara, 06110, Turkey
| | | | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara, 06100, Turkey.
| |
Collapse
|
109
|
Fiorani M, Buffi G, Bagherlou N, Canonico B, De Matteis R, Guidarelli A, Montanari M, Battistelli M, Papa S, Coppo L, Cerioni L, Spina A, Cantoni O. Clozapine blunts mitochondrial biogenesis in differentiating adipocytes: The increased ATP demand is met via stimulation of electron transport chain expression and activity in residual mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119967. [PMID: 40280335 DOI: 10.1016/j.bbamcr.2025.119967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/25/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Clozapine (CLZ), a second-generation antipsychotic, is associated with an elevated risk of metabolic syndrome, the underlying mechanism of which remains poorly understood. We recently showed that CLZ inhibits lipid accumulation and CAAT/enhancer-binding protein β and peroxisome proliferator-activated receptor γ expression in early differentiating SW872 liposarcoma cells. Additionally, while not affecting viability, CLZ disrupts the cellular redox state of these cells by inhibiting NADPH oxidase-dependent ROS formation, thereby leading to nuclear factor (erythroid-derived2)-like 2 downregulation, reduced antioxidant defence and increased mitochondrial ROS emission. We confirmed and extended these results by showing that, under the same conditions, CLZ reduces the size of the lipid droplets, inhibits the otherwise increased expression of transcription factors regulating mitochondrial biogenesis, as peroxisome proliferator-activated receptor γ coactivator 1-α, and prevents the increase in mitochondrial DNA and mass. Consistently, decreased expression of mitochondrial proteins as thioredoxin 2, 2-oxoglutarate/malate carrier, and translocase of outer mitochondrial membrane 20 was also observed. However, the expression of various components of the electron transport chain was unexpectedly increased, and this event was accompanied by enhanced mitochondrial dehydrogenase activity, coupled oxygen consumption, mitochondrial membrane potential, ATP synthesis and ROS production. Moreover, residual mitochondria appeared remarkably enlarged and functional, with dense and organized cristae and uniform electron density. Thus, early adipocytes differentiated with or without CLZ meet the increased ATP demand by switching from glycolysis to oxidative phosphorylation, respectively via enhanced mitochondrial biogenesis, and increased activity of residual mitochondria.
Collapse
Affiliation(s)
- Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - Gloria Buffi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Nazanin Bagherlou
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Rita De Matteis
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Lucia Coppo
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Liana Cerioni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Spina
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
110
|
Roma RR, Oliveira FSA, Fernandes DGS, Garcia W, Soares EN, Costa SL, Teixeira CS. ConA-glutamate interactions: New insights into its neuroprotective effect. Int J Biol Macromol 2025; 310:143463. [PMID: 40280512 DOI: 10.1016/j.ijbiomac.2025.143463] [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: 02/03/2025] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
L-Glutamate is the primary excitatory neurotransmitter in the brain; excessive levels induce L-glutamate-mediated excitotoxicity, linked to Alzheimer's and Parkinson's. Plant-derived molecules with antioxidant and anti-inflammatory properties that modulate this are of interest. Canavalia ensiformis lectin (ConA) serves as a model lectin for CNS studies. This study aimed to analyze in vitro and in silico the neuroprotective potential of ConA against glutamatergic excitotoxicity and identify the involved protein domain and mechanisms. Native and demetallized ConA were used for cytotoxicity and neuroprotection assays in PC12 cells. Molecular docking and fluorescence spectroscopy were also employed. ConA (1-50 mM) did not show cytotoxicity in PC12 cells and protected them from glutamatergic excitotoxicity at 15.6 μg/mL, significantly increasing cell viability from 80 % to over 90 %. Furthermore, affinity and binding assays indicated that the carbohydrate recognition domain was not involved in neuroprotection; instead, the amino acid-binding site played a crucial role. Our findings conclude that ConA possesses neuroprotective potential against glutamatergic excitotoxicity in PC12 cells via an L-glutamate sequestration mechanism mediated by the amino acid-binding site.
Collapse
Affiliation(s)
- Renato R Roma
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Fábio S A Oliveira
- Agrarian and Biodiversity Sciences Center, Federal University of Cariri, Crato, Ceará, Brazil
| | | | - Wanius Garcia
- Natural and Human Sciences Center, Federal University of ABC, Santo André, SP, Brazil
| | - Erica N Soares
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Claudener S Teixeira
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, CE, Brazil; Agrarian and Biodiversity Sciences Center, Federal University of Cariri, Crato, Ceará, Brazil.
| |
Collapse
|
111
|
Chai M, Li W, Wang B, Zhang X, Li D, Zhang Z, Wang Z, He X. Precursor-Directed Biosynthesis of Phenylpyrrole Analogues in Myrothecium verrucaria. JOURNAL OF NATURAL PRODUCTS 2025. [PMID: 40262037 DOI: 10.1021/acs.jnatprod.5c00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Supplementation of p-aminobenzoic acid and its derivatives into the culture medium of Myrothecium verrucaria HPU-ZMN, a fungus isolated from the Yellow River wetland, resulted in the production of 21 novel phenylpyrrole derivatives designated as myropherroles A-U (1-21). Structural elucidation of these compounds was achieved through a comprehensive analysis of NMR and HRESIMS data. Notably, the myropherroles exhibit more diverse structures of the pyrrole compared to naturally occurring phenylpyrrole derivatives. Compound 2 displayed an acetylcholinesterase (AChE) inhibitory effect with an IC50 value of 0.9 μM.
Collapse
Affiliation(s)
- Miaomiao Chai
- School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Wei Li
- School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Bing Wang
- School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Xiaomin Zhang
- Key Laboratory of Marine Drugs Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, People's Republic of China
| | - Dehai Li
- Key Laboratory of Marine Drugs Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, People's Republic of China
| | - Zhenzhen Zhang
- School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
- Jiaozuo Key Laboratory for Huaiyao Comprehensive Development, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Zhenhui Wang
- School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
- Jiaozuo Key Laboratory for Huaiyao Comprehensive Development, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Xueqian He
- School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
- Jiaozuo Key Laboratory for Huaiyao Comprehensive Development, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| |
Collapse
|
112
|
Stefanova D, Garip A, Mateev E, Kondeva-Burdina M, Yordanov Y, Tzankova D, Mateeva A, Valkova I, Georgieva M, Zlatkov A, Tzankova V. Antioxidant and Neuroprotective Properties of Selected Pyrrole-Containing Azomethine Compounds in Neurotoxicity Models In Vitro. Int J Mol Sci 2025; 26:3957. [PMID: 40362198 PMCID: PMC12072147 DOI: 10.3390/ijms26093957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Neurodegenerative diseases involve oxidative stress and enzyme dysregulation, necessitating novel neuroprotective agents. This study evaluates the neuroprotective and antioxidant potential of seven pyrrole-based compounds with predicted radical scavenging activity and inhibitory effects on monoamine oxidase B (MAO-B) and acetylcholinesterase (AChE). The compounds were tested in vitro using SH-SY5Y neuroblastoma cells and subcellular rat brain fractions, including synaptosomes, mitochondria, and microsomes. Neuroprotective and antioxidant effects were assessed in oxidative stress models, including H2O2-induced stress in SH-SY5Y cells, 6-hydroxydopamine toxicity in synaptosomes, tert-butyl hydroperoxide-induced stress in mitochondria, and non-enzyme lipid peroxidation in microsomes. In silico screening for lipophilicity, hydrogen bonding, total polar surface area (TPSA), and ionization properties, was performed to evaluate bioavailability. All compounds exhibited a weak neurotoxic effect on the subcellular fractions at a concentration of 100 µM. However, in oxidative stress models, they demonstrated significant neuroprotective and antioxidant effects at 100 µM. In SH-SY5Y cells, compounds 7, 9, 12, 14, and 15 exhibited low toxicity and strong protective effects at concentrations as low as 1 µM. In silico analysis prioritized compounds 1, 7, 9, 12, and 14 for further development based on their favorable bioavailability. The tested pyrrole-based compounds exhibit promising neuroprotective and antioxidant properties, with several candidates showing potential for further development based on both in vitro efficacy and predicted oral bioavailability.
Collapse
Affiliation(s)
- Denitsa Stefanova
- Department Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (A.G.); (M.K.-B.); (Y.Y.); (V.T.)
| | - Alime Garip
- Department Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (A.G.); (M.K.-B.); (Y.Y.); (V.T.)
| | - Emilio Mateev
- Department “Pharmaceutical Chemistry”, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (E.M.); (D.T.); (A.M.); (M.G.); (A.Z.)
| | - Magdalena Kondeva-Burdina
- Department Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (A.G.); (M.K.-B.); (Y.Y.); (V.T.)
| | - Yordan Yordanov
- Department Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (A.G.); (M.K.-B.); (Y.Y.); (V.T.)
| | - Diana Tzankova
- Department “Pharmaceutical Chemistry”, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (E.M.); (D.T.); (A.M.); (M.G.); (A.Z.)
| | - Alexandrina Mateeva
- Department “Pharmaceutical Chemistry”, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (E.M.); (D.T.); (A.M.); (M.G.); (A.Z.)
| | - Iva Valkova
- Department “Chemistry”, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria;
| | - Maya Georgieva
- Department “Pharmaceutical Chemistry”, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (E.M.); (D.T.); (A.M.); (M.G.); (A.Z.)
| | - Alexander Zlatkov
- Department “Pharmaceutical Chemistry”, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (E.M.); (D.T.); (A.M.); (M.G.); (A.Z.)
| | - Virginia Tzankova
- Department Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University-Sofia, 1431 Sofia, Bulgaria; (A.G.); (M.K.-B.); (Y.Y.); (V.T.)
| |
Collapse
|
113
|
Ingole VV, Mhaske PC, Shaikh SB, Katade SR. A new coumarin from Tridax procumbens L. as potential anticancer, antibacterial, antioxidant agent and its molecular docking study. Nat Prod Res 2025:1-7. [PMID: 40264251 DOI: 10.1080/14786419.2025.2493190] [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: 07/13/2023] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
A study investigated the phytochemical constituent of the methanol extract from the aerial part of T. procumbens L. The investigation resulted in the identification of a new coumarin. A novel coumarin compound was successfully isolated using the silica gel column chromatographic technique and various spectroscopic methods were employed to elucidate its structure. Isolated new coumarin was evaluated for its cytotoxicity, antimicrobial, and antioxidant activities. The isolated coumarin exhibited cytotoxicity IC50 values of 4.92 and 5.99 µg/mL against MCF-7 and MDA-MB-249 cell lines, respectively, in the MTT assay. Additionally, the compound demonstrated notable antioxidant activity, with an IC50 value of 1.36 µg/mL (p < 0.05). The antimicrobial evaluation revealed MIC values of 125 µg/mL against S. aureus and 31.5 µg/mL against P. aeruginosa. This activity is strongly supported by an in silico study. Further studies are required to determine its effectiveness as an antioxidant, anticancer, and antibacterial agent.
Collapse
Affiliation(s)
- Varsharani V Ingole
- Department of Chemistry, PES's Modern College of Arts, Science and Commerce, Pune, India
- Department of Chemistry, STES's Sinhgad College of Science, Pune, India
| | - Pravin C Mhaske
- Post-Graduate Department of Chemistry, S. P. Mandali's, Sir Parshurambhau College, Pune, India
| | - Siraj B Shaikh
- Department of Chemistry, M.C.E. Society's Abeda Inamdar College of Arts, Science and Commerce, Pune, India
| | - Sushma R Katade
- Department of Chemistry, PES's Modern College of Arts, Science and Commerce, Pune, India
| |
Collapse
|
114
|
Tokonami A, Kawanaka M, Ikeda H, Nishii S, Kamegawa T, Yamamoto Y, Sadanaga Y, Shiigi H. Monitoring the Metabolic Activity of a Single Bacterial Cell Based on Scattering Intensity. Anal Chem 2025; 97:8293-8300. [PMID: 40194273 PMCID: PMC12019773 DOI: 10.1021/acs.analchem.4c06314] [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: 11/21/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
Cell activity is evaluated using the number of colonies formed on a medium or the number of live cells in a suspension or by staining nuclei with fluorescent dyes to determine whether cells are dead. However, the culture methods generally require extended culturing times, and damage to the cell membranes observed using fluorescent dyes is not necessarily related to cell survival or activity. Hence, accurately determining the activities of individual cells is impossible. Therefore, we developed a method for quantitatively evaluating the metabolic activities of single cells by focusing on the optical and chemical properties of formazan dye, i.e., 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). The oxidized form of MTT is soluble and highly permeable to cell membranes, but it is reduced to insoluble MTT formazan upon reaction with intracellular metabolic products. Single-cell observation using dark-field microscopy revealed that insoluble formazan aggregates within the cells formed particles that emitted characteristic scattered light. The formazan-derived scattered light component extracted via peak fitting was related to metabolic activity, demonstrating its usefulness as a parameter indicating the activity of an individual cell. This method enables the real-time evaluation of the activities of single cells, which should lead to not only the acceleration of bacterial screening and microbial control but also the development of antibiotics and suppression of drug-resistant bacteria.
Collapse
Affiliation(s)
- Akira Tokonami
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| | - Miya Kawanaka
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| | - Hikaru Ikeda
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| | - Shigeki Nishii
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| | - Takashi Kamegawa
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| | - Yojiro Yamamoto
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| | - Yasuhiro Sadanaga
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| | - Hiroshi Shiigi
- Department
of Applied Chemistry, Osaka Metropolitan
University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
- Osaka
International Research Center for Infectious Diseases, Osaka Metropolitan University, 1-2 Gakuen, Naka, Sakai, Osaka 599-8570, Japan
| |
Collapse
|
115
|
Shukla A, Sharma A, Gupta S, Mishra A, Singh A. Antitumor potential of ivermectin against T-cell lymphoma-bearing hosts. Med Oncol 2025; 42:169. [PMID: 40257544 DOI: 10.1007/s12032-025-02726-0] [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: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Ivermectin, a broad-spectrum antiparasitic agent from the ivermectin family, has shown promising anticancer potential. Originally developed for veterinary and human use against parasitic infections, ivermectin demonstrated significant antitumor effects in our study against tumor cells (Dalton's lymphoma cells). A dose-dependent decrease in tumor cell viability was observed following 24-h treatment with ivermectin, with an IC₅₀ value calculated at 10.55 µg/mL. In comparison, the standard anticancer drug cisplatin exhibited a slightly higher cytotoxic potency, with an IC₅₀ of 8.32 µg/mL under the same treatment duration. Flow cytometric analysis revealed that ivermectin induced cell cycle arrest in the G0-G1 phase. Apoptotic tumor cell death was confirmed via Annexin V/PI staining, further supported by nuclear condensation, a hallmark of apoptosis, visualized through both confocal microscopy and flow cytometry. The apoptosis was determined to be mitochondrial-dependent, as evidenced by a decline in mitochondrial membrane potential (ΔΨm) observed through JC-1 assay. The treatment increased DAPI-positive cells and exhibited severe chromatin condensation. Additionally, cell death was validated using Acridine Orange and Propidium Iodide staining, which highlighted increased cell membrane rupture and death through apoptosis and necrosis. Mitochondrial dependent apoptosis further supported by increased ROS production upon ivermectin treatment. Moreover, In vivo, ivermectin treatment led to a substantial reduction in tumor size in tumor-bearing mice, along with normalization of spleen size, body weight, and improvement histopathology of liver. These findings collectively support the therapeutic potential of ivermectin as a repurposed anticancer agent, acting through multiple mechanisms including cell cycle arrest, ROS generation, mitochondrial dysfunction, and apoptosis.
Collapse
Affiliation(s)
- Alok Shukla
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Arpit Sharma
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
- Department of Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India
| | - Shivani Gupta
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Abha Mishra
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India.
| | - Amit Singh
- Department of Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India.
- Baba Kinaram Medical College, Chandauli, 232104, India.
| |
Collapse
|
116
|
Van NTH, Bach PC, Thuong VT, Tuyen TT, Vien TA, Thuy DTT, Quynh DT, Nghi DH, Quan PM, Xuan NM, Toan TQ, Minh PTH, Hung NH. Chemical Composition and Pesticidal Activities Against Three Vector Mosquito Species of Zanthoxylum armatum DC. Essential Oils. Chem Biodivers 2025:e202500648. [PMID: 40257289 DOI: 10.1002/cbdv.202500648] [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: 02/22/2025] [Revised: 04/12/2025] [Accepted: 04/21/2025] [Indexed: 04/22/2025]
Abstract
In this study, we investigated the extraction yield and chemical composition of essential oils from the fruit, leaf, and twig of Zanthoxylum armatum from Vietnam. The fruit essential oil (FEO) with high content, representing a chemotype, was evaluated for pesticidal activities against three mosquito species Aedes aegypti, Aedes albopictus, and Culex quinquefasciatus, according to the WHO guidelines with modifications. The results showed that the major constituents of FEO were limonene (30.50%), sabinene (15.16%), terpinen-4-ol (13.05%), and γ-terpinene (7.49%). The major constituents of leaf essential oil (LEO) were sabinene (20%), 1,8-cineole (16.97%), limonene (12.32%), and 2-undecanone (9.17%). The twig essential oil (TEO) was rich in ketones (73.84%), with the main constituents being 2-undecanone (47.33%) and 2-tridecanone (26.14%). FEO exhibited potential pesticidal activities: larvicidal activities with 24-h LC50 values ranging from 21.55 to 29.53 µg/mL inhibited the biting of A. aegypti adults with a protection time of 176.5 ± 40.20 min, and exhibited potent adulticidal activities against A. aegypti. In addition, FEO did not exhibit cytotoxicity against the normal Vero cell line. This study contributes an evidence base to support the future development and use of FEO as a promising biopesticide agent for the control of disease-transmitting mosquito species and altering synthetic pesticides.
Collapse
Affiliation(s)
- Nguyen Thi Hong Van
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Pham Cao Bach
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Vo Thanh Thuong
- Department of Pharmacy, Duy Tan University, Da Nang, Vietnam
| | - Tran Thi Tuyen
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Trinh Anh Vien
- Department of Examination and Quality Assurance of Education, Hanoi Medical University-Thanh Hoa Campus, Thanh Hoa, Vietnam
| | - Dinh Thi Thu Thuy
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Dang Thu Quynh
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Do Huu Nghi
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Pham Minh Quan
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Nguyen Mua Xuan
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Tran Quoc Toan
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Pham Thi Hong Minh
- Institute of Chemistry (ICH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Nguyen Huy Hung
- Department of Pharmacy, Duy Tan University, Da Nang, Vietnam
- Center for Advanced Chemistry, Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
117
|
Jangra B, Goyal A, Srivastava A, Jachak SM. Development of rapid UPLC-PDA method for analysis of polyphenols in Terminalia chebula Retz. fruits, method validation and evaluation of anti-inflammatory activity. Nat Prod Res 2025:1-10. [PMID: 40258077 DOI: 10.1080/14786419.2025.2490051] [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: 09/30/2024] [Revised: 03/11/2025] [Accepted: 04/03/2025] [Indexed: 04/23/2025]
Abstract
Terminalia chebula is an important medicinal plant in traditional medicine, renowned for its fruits that offer various health benefits. This study aimed to investigate metabolic variation in fruit pericarp of T. chebula collected from four regions of India (North, South, East, and West). Rapid and efficient ultra-performance liquid chromatography with photodiode array detection method was developed for separation and quantification of polyphenols viz. gallic acid, corilagin, chebulagic acid, ellagic acid, and chebulinic acid using BEH Shield RP-C18 column (2.1 × 100 mm, 1.7 µm) within 10 min. Method validation was performed in accordance with International Council for Harmonisation (ICH) Q2 (R1) guidelines. Results indicated no significant geographical influence on phytoconstituents' content in samples. Chebulagic acid was found highest in sample collected from North India region. Activity was determined in terms of their ability to inhibit nitric oxide release in LPS-stimulated macrophages. This study presents unique, economical, time-efficient method for quality control, standardisation of T. chebula-based formulations, confirming its consistent potency across India.
Collapse
Affiliation(s)
- Bhawana Jangra
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Punjab, India
| | - Alok Goyal
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Punjab, India
| | - Amit Srivastava
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Punjab, India
| | - Sanjay Madhukar Jachak
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Punjab, India
| |
Collapse
|
118
|
Baharetha HM, Abdul Majid AMS, Nazari MV, Samad NA, Al-Mansoub MA, Taleb Agha M, Dahham SS. Optimised formulation and characterisation of liposomes for enhanced stability and antiproliferative efficacy of Orthosiphon aristatus var. aristatus extract in lung cancer treatment. Nat Prod Res 2025:1-13. [PMID: 40258057 DOI: 10.1080/14786419.2025.2494638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
This study aimed to characterise and evaluate the antiproliferative potency of a standardised 50% ethanol extract of Orthosiphon aristatus var. aristatus leaves against human lung cancer cells in vitro. A liposomal drug delivery system was developed to enhance bioavailability and efficacy. Three formulations were designed with different extract-to-phospholipid ratios, phospholipid, and cholesterol content. NP2, formulated with a 1:1 extract-to-phosphatidylcholine ratio and 20% cholesterol, demonstrated optimal stability and efficacy. Liposomes were quantitatively and qualitatively characterised using light microscopy, TEM, SEM, zeta sizer, and zeta potential analysis. NP2 demonstrated better bioactive compounds release properties and better stability than the extract. The extract and liposomes inhibited the proliferation of human lung adenocarcinoma (A549) and normal endothelial cells (EA.hy926). The findings indicate that the extract possesses potent anti-lung cancer activity, with the NP2 liposomal formulation enhancing its therapeutic potential.
Collapse
Affiliation(s)
- Hussein M Baharetha
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Malaysia
- Department of Pharmacy, College of Medicine and Health Sciences, Hadhramout University, Mukalla, Yemen
| | | | | | - Nozlena Abdul Samad
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Majed Ahmed Al-Mansoub
- Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Kota Samarahan, Malaysia
| | - Mohamad Taleb Agha
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Malaysia
| | - Saad S Dahham
- Department of Science, University of Technology and Applied Sciences, Rustaq, Oman
| |
Collapse
|
119
|
El Deeb BA, Faheem GG, Bakhit MS. Biosynthesis of silver nanoparticles by Talaromyces funiculosus for therapeutic applications and safety evaluation. Sci Rep 2025; 15:13750. [PMID: 40258887 PMCID: PMC12012204 DOI: 10.1038/s41598-025-95899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
The study investigated the capacity of the endophytic fungus Talaromyces funiculosus to biosynthesize extracellular AgNPs and assess their safety. The fungus was identified through morphological and phylogenetic analyses. The biosynthesized AgNPs were spherical crystalline, stable (6 months), and mono-dispersed (PDI: 0.007), exhibiting SPR at 422.5 nm, average diameter of 34.32 nm, and Zeta potential of -18.41 mV. The optimal biosynthesis conditions are 1 mM AgNO3, 5 g biomass, pH 5.5, and a reaction temperature of 60 °C. Escherichia coli (bacterial strains) and Candida tropicalis (yeast strains) exhibited the highest susceptibility with inhibition zones of 26.3 mm and 22.3 mm, respectively, at 50 µg/mL of AgNPs, and MICs of 3.7 µg/mL and 6.3 µg/mL, respectively. AgNPs exhibited cytotoxicity with IC50 values of 48.11 ppm for HEK-293 and 35.88 ppm for Hep-G2 cells, showing selective toxicity toward cancer cells. They demonstrated antioxidant activity by increasing GSH (10.29 to 14.76 mmol/g) and reducing MDA (40.57 to 26.28 nmol/ml) at 48.11 ppm. AgNPs also enhanced IL-10 production (96.47 to 177.0 pg/mL) and reduced TNF-α levels (55.77 to 41.06 pg/mL), indicating their anti-inflammatory properties. These results support the safe use of low-dose AgNPs, however, further studies are needed to evaluate AgNPs for clinical uses.
Collapse
Affiliation(s)
- Bahig A El Deeb
- Department of Botany and Microbiology, Faculty of Science, Sohag University, Sohag, 82524, Egypt.
- Higher Technological Institute of Applied Health Science in Sohag, Ministry of Higher Education, Cairo, Egypt.
| | - Gerges G Faheem
- Department of Botany and Microbiology, Faculty of Science, Sohag University, Sohag, 82524, Egypt
| | - Mahmoud S Bakhit
- Department of Botany and Microbiology, Faculty of Science, Sohag University, Sohag, 82524, Egypt
| |
Collapse
|
120
|
Abuçafy MP, Ramin BBS, Graminha AE, Santos WG, Frem RCG, Netto AVG, Pereira JC, Ribeiro SJL. Core-Shell UCNP@MOF Nanoplatforms for Dual Stimuli-Responsive Doxorubicin Release. ACS APPLIED BIO MATERIALS 2025; 8:2954-2964. [PMID: 40202909 PMCID: PMC12015952 DOI: 10.1021/acsabm.4c01796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Nanocarrier systems with multifunctional capabilities hold great potential for targeted cancer therapy, particularly for breast cancer treatment. Metal-organic frameworks (MOFs) are notable for their high porosity and, in some cases, biocompatibility, with ZIF-8 being particularly advantageous due to its pH-sensitive degradability, enabling selective drug release in tumor environments. Additionally, lanthanide-doped upconversion nanoparticles (UCNPs) offer unique optical properties that enhance both imaging and therapeutic applications. In this study, NaYF4/Yb3+Er3+ UCNPs were synthesized via a hydrothermal method, subsequently coated with poly(acrylic acid) (PAA) and encapsulated within a ZIF-8 shell, forming of UCNP@ZIF-8 core-shell nanocomposites. This system was designed to leverage stimulation by a 980 nm laser and acidic pH to facilitate drug release. When exposed to this specific laser wavelength, the nanocomposites demonstrated significantly enhanced drug release, achieving up to 90% release of the incorporated antitumor drug, doxorubicin (DOX), in acidic environments. In vitro studies indicated selective cytotoxicity, with MCF-7 tumor cell viability decreasing from 85.7% to 20% following laser activation, while showing minimal toxicity toward healthy cells. These findings underscore the potential of the UCNP@ZIF-8 nanocarrier system as a pH and laser-responsive platform for improved cancer therapy, enabling precise control over drug delivery while minimizing side effects on surrounding healthy tissues.
Collapse
Affiliation(s)
- Marina P. Abuçafy
- Institute
of Chemistry, São Paulo State University, Araraquara, São Paulo 14800-060, Brazil
| | - Beatriz B. S. Ramin
- Institute
of Chemistry, São Paulo State University, Araraquara, São Paulo 14800-060, Brazil
| | - Angelica E. Graminha
- Institute
of Chemistry, São Paulo State University, Araraquara, São Paulo 14800-060, Brazil
| | - Willy G. Santos
- Institute
of Chemistry, São Paulo State University, Araraquara, São Paulo 14800-060, Brazil
- Federal
University of ABC, UFABC, Santo
André, São Paulo 09210-170, Brazil
| | - Regina C. G. Frem
- Institute
of Chemistry, São Paulo State University, Araraquara, São Paulo 14800-060, Brazil
| | - Adelino V. G. Netto
- Institute
of Chemistry, São Paulo State University, Araraquara, São Paulo 14800-060, Brazil
| | | | - Sidney J. L. Ribeiro
- Institute
of Chemistry, São Paulo State University, Araraquara, São Paulo 14800-060, Brazil
| |
Collapse
|
121
|
Ghariani O, Elleuch J, Ferretti AM, Econdi S, Bisio C, Michaud P, Fendri I, Guidotti M, Abdelkafi S. Toxicological Effects of Silver-Modified Bentonite Nanocomposites on Microalgae: Impact on Cell Growth, Antioxidant Enzymes, and Gene Expression. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:629. [PMID: 40278494 PMCID: PMC12029818 DOI: 10.3390/nano15080629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
The increasing use of nanostructured silver-containing inorganic materials raises concerns about their impact on aquatic organisms. This study assessed the toxicity of silver-modified bentonite composites on Chlamydomonas sp. Two materials were tested: silver-exchanged bentonite (Ben-Ag) and its reduced form (Ben-Ag (H2)).Microalgae were exposed to 0.5 IC50, 1.5 IC50, and 2 IC50. Ben-Ag showed higher toxicity than Ben-Ag (H2), which even promoted algal growth at low doses. Fluorescence microscopy revealed morphological shrinkage in treated cells. Increased phenol content, elevated malondialdehyde (MDA) levels, and altered antioxidant enzyme activities further confirmed Ben-Ag toxicity, along with reduced growth and photosynthetic pigments. Transcriptomic analysis revealed significant changes in gene expression under Ben-Ag exposure. Genes involved in photosynthesis (petB, psbL), caspase activity (casp), and carotenoid metabolism (Q2CHY) were down-regulated, indicating stress-induced damage. In contrast, genes encoding stress response enzymes (SOD, peroxidase), carbon metabolism enzymes (rbcL, PGQ1), and β-carotene biosynthesis (Q2BKT) were up-regulated, reflecting cellular defense mechanisms. Overall, the study highlights the high toxicity of Ben-Ag to Chlamydomonas sp., emphasizing the importance of evaluating environmental risks before using such materials in aquatic environments.
Collapse
Affiliation(s)
- Oumayma Ghariani
- Enzymatic Engineering and Microbiology Laboratory, Algae Biotechnology Unit, National Engineering School of Sfax, University of Sfax, Sfax 3038, Tunisia; (O.G.); (J.E.); (S.A.)
- CNR-SCITEC Istituto di Scienze e Tecnologie Chimiche “G. Natta”, Via C. Golgi 19, 20133 Milano, Italy; (S.E.); (M.G.)
| | - Jihen Elleuch
- Enzymatic Engineering and Microbiology Laboratory, Algae Biotechnology Unit, National Engineering School of Sfax, University of Sfax, Sfax 3038, Tunisia; (O.G.); (J.E.); (S.A.)
| | - Anna Maria Ferretti
- CNR-SCITEC Istituto di Scienze e Tecnologie Chimiche “G. Natta”, Via G. Fantoli 16/15, 20138 Milano, Italy;
| | - Stefano Econdi
- CNR-SCITEC Istituto di Scienze e Tecnologie Chimiche “G. Natta”, Via C. Golgi 19, 20133 Milano, Italy; (S.E.); (M.G.)
| | - Chiara Bisio
- Department of Science and Technological Innovation, DISIT, University of Eastern Piedmont, Via T. Michel 11, 15121 Alessandria, Italy;
| | - Philippe Michaud
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, Institut Pascal, F-63000 Clermont-Ferrand, France
| | - Imen Fendri
- Laboratory of Plant Biotechnology Applied to Crop Improvement, Faculty of Science of Sfax, University of Sfax, Sfax 3029, Tunisia;
| | - Matteo Guidotti
- CNR-SCITEC Istituto di Scienze e Tecnologie Chimiche “G. Natta”, Via C. Golgi 19, 20133 Milano, Italy; (S.E.); (M.G.)
| | - Slim Abdelkafi
- Enzymatic Engineering and Microbiology Laboratory, Algae Biotechnology Unit, National Engineering School of Sfax, University of Sfax, Sfax 3038, Tunisia; (O.G.); (J.E.); (S.A.)
| |
Collapse
|
122
|
Anjomshoa M, Amirheidari B, Sahihi M, Janczak J, Forootanfar H, Farsinejad A, Abolhassani Y, Karami-Mohajeri S. In vitro cellular and molecular plus in silico studies of a substituted bipyridine-coordinated Zn(II) ion: cytotoxicity, ROS-induced apoptosis, anti-metastasis, and BAX/BCL2 genes expression. J Biol Inorg Chem 2025:10.1007/s00775-025-02114-z. [PMID: 40253669 DOI: 10.1007/s00775-025-02114-z] [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: 07/19/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025]
Abstract
A new dimethyl-substituted bipyridine-Zn(II) complex (2Mebpy-Zn) was synthesized and structurally characterized. Single-crystalline structure of the complex was elucidated as [Zn(2Mebpy)3](ClO4)2∙1.5(dioxane) by X-ray diffraction, where 2Mebpy is 4,4'-dimethyl-2,2'-bipyridine. The three-dimensional electrostatic potential maps (3D ESP) were plotted for [Zn(2Mebpy)3]2+ cation and [Zn(2Mebpy)3](ClO4)2 molecule. In vitro cytotoxicity studies indicated significant cytotoxicity of 2Mebpy-Zn against both breast (MCF-7) and glioblastoma (U-87) cancer cells relative to normal murine embryo cells (NIH/3T3). The results are indicative of a superior selectivity toward MCF-7 over the other cell lines as confirmed by IC50 value of 5.1 ± 0.5 µM after 48 h. Interestingly, MCF-7 and U-87 cells death induced by 2Mebpy-Zn mostly proceed through an apoptotic pathway which probably associates with the overproduction of reactive oxygen species (ROS). The Zn(II) complex suppressed the metastatic affinity of MCF-7 cells by blocking migration as well as formation of colonies. Also, the expression of two opponent apoptosis-relevant genes (BAX and BCL2) measured by real-time polymerase chain reaction (qPCR) experiments indicated that 2Mebpy-Zn could potentially trigger apoptotic cell death. Moreover, 2Mebpy-Zn could cleave hydrolytically the pUC19 DNA without the need to add any external agent. Finally, the binding affinity of two enantiomers of 2Mebpy-Zn toward cancer therapeutic targets, such as anti-apoptotic proteins, estrogen receptor α, tubulin, and topoisomerase II, was studied by in silico molecular docking. In conclusion, 2Mebpy-Zn can be introduced as a potential therapeutic agent in breast cancer and indicates that other metal complexes with bipyridine derivatives can also exhibit promising anticancer effects.
Collapse
Affiliation(s)
- Marzieh Anjomshoa
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran.
| | - Bagher Amirheidari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran.
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehdi Sahihi
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut de Chimie de Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Jan Janczak
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okólna 2 Str., 50-422, Wrocław, Poland
| | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Farsinejad
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasaman Abolhassani
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran
| | - Somayyeh Karami-Mohajeri
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
123
|
Gulyás KV, Zhou L, Salamonsen D, Prester A, Bartels K, Bosman R, Haffke P, Li J, Tamási V, Deufel F, Thoma J, Andersson Rasmussen A, Csala M, Schroder Leiros HK, Xu Z, Widersten M, Rohde H, Schulz EC, Zhu W, Erdélyi M. Dynamically chiral phosphonic acid-type metallo-β-lactamase inhibitors. Commun Chem 2025; 8:119. [PMID: 40253435 PMCID: PMC12009420 DOI: 10.1038/s42004-025-01510-5] [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: 09/08/2024] [Accepted: 04/02/2025] [Indexed: 04/21/2025] Open
Abstract
Antibiotic resistance is a growing global health threat that risks the lives of millions. Among the resistance mechanisms, that mediated by metallo-β-lactamases is of particular concern as these bacterial enzymes dismantle most β-lactam antibiotics, which are our widest applied and cheapest to produce antibiotic agents. So far, no clinically applicable metallo-β-lactamase inhibitors are available. Aiming to adapt to structural variations, we introduce the inhibitor concept: dynamically chiral phosphonic acids. We demonstrate that they are straightforward to synthesize, penetrate bacterial membranes, inhibit the metallo-β-lactamase enzymes NDM-1, VIM-2 and GIM-1, and are non-toxic to human cells. Mimicking the transition state of β-lactam hydrolysis, they target the Zn ions of the metallo-β-lactamase active site. As a unique feature, both of their stereoisomers bind metallo-β-lactamases, which provides them unparalleled adaptability to the structural diversity of these enzymes, and may allow them to hamper bacteria's ability for resistance development.
Collapse
Affiliation(s)
- Kinga Virág Gulyás
- Department of Chemistry - BMC, Organic Chemistry and the Uppsala Antibiotic Center; Uppsala University, Uppsala, Sweden
| | - Liping Zhou
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Daniel Salamonsen
- Department of Chemistry, Faculty of Science and Technology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Andreas Prester
- University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Kim Bartels
- University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Robert Bosman
- University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Paul Haffke
- University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jintian Li
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Viola Tamási
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Fritz Deufel
- Department of Chemistry - BMC, Organic Chemistry and the Uppsala Antibiotic Center; Uppsala University, Uppsala, Sweden
| | - Johannes Thoma
- Department of Chemistry & Molecular Biology, Center for Antibiotic Resistance Research, CARe, University of Gothenburg, Gothenburg, Sweden
| | | | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | | | - Zhijian Xu
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mikael Widersten
- Department of Chemistry - BMC, Organic Chemistry and the Uppsala Antibiotic Center; Uppsala University, Uppsala, Sweden
| | - Holger Rohde
- University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Eike C Schulz
- University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Max-Planck-Institute for Structure and Dynamics of Matter, Hamburg, Germany
- Institute for Nanostructure and Solid State Physics, Universität Hamburg, Hamburg, Germany
| | - Weiliang Zhu
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Máté Erdélyi
- Department of Chemistry - BMC, Organic Chemistry and the Uppsala Antibiotic Center; Uppsala University, Uppsala, Sweden.
- Center of Excellence for the Chemical Mechanisms of Life, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
124
|
Guo X, Lu Y, Zhang X, Wang X, Wang H, Zhang Z. A 4-phenylbutyric acid-conjugated platinum diimine complex: Photocytotoxicity, cell cycle arrest and apoptosis in vitro. J Inorg Biochem 2025; 270:112930. [PMID: 40262391 DOI: 10.1016/j.jinorgbio.2025.112930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
4-phenylbutyric acid (PBA) is a class I and II histone deacetylase inhibitor, which can decrease cell proliferation, enhance cell differentiation, and induce apoptosis and cell cycle arrest in various cancer cells. PBA-modified photoactive platinum diimine complex has been prepared and characterized to augment its photodynamic therapy efficacy. Its ability to generate singlet oxygen, behavior in the presence of esterase, photocytotoxicity, cell cycle distribution, and apoptosis-inducing ability in MCF-7 cells have also been studied. The results suggested that the PBA-modified platinum diimine complex could act as a good singlet oxygen producer, release PBA in the presence of esterase, induce potent photocytotoxicity in tumor cells with a IC50 value of 3.67 ± 0.67 μM, arrest cell cycle at S phase, and induce cell death via apoptosis with the percentage of total apoptotic cells being 34.65 %. The results also revealed that PBA might be able to modulate the mode of cell cycle arrest induced by the photosensitizer in tumor cells.
Collapse
Affiliation(s)
- Xuemei Guo
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Yuxin Lu
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xu Zhang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xin Wang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Hongfei Wang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Zhigang Zhang
- Institute of Molecular Science, Chemical Biology and Molecular Engineering Laboratory of Education Ministry, Shanxi University, Taiyuan, Shanxi 030006, China.
| |
Collapse
|
125
|
Sun Y, Wang H. Revealing propionate metabolism-related genes in glioblastoma and investigating their underlying mechanisms. Front Oncol 2025; 15:1529369. [PMID: 40313243 PMCID: PMC12043635 DOI: 10.3389/fonc.2025.1529369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Background Propionate metabolism may affect tumor growth and aggressiveness, but the role of propionate metabolism-related genes (PMRGs) in glioblastoma (GBM) remains poorly understood. Methods Differentially expressed PMRGs (DE-PMRGs) were identified by comparing differentially expressed genes (DEGs) between GBM and normal tissues using TCGA-GBM, GSE42669, GSE162631 datasets. Functional enrichment analysis of DE-PMRGs was performed, followed by univariate Cox regression and least absolute shrinkage with selection operator (LASSO) analysis to identify potential prognostic biomarkers. In addition, prognostic models were developed and validated using independent cohorts. Genomic enrichment analysis (GSEA) was used to assess immune-related pathways in different risk subgroups. Finally, biomarker expression was confirmed using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results Differential expression analysis identified a total of 180 DE-PMRGs, which were strongly associated with drug response and insulin signaling pathways. Six biomarkers (SARDH, ACHE, ADSL, PNPLA3, MAPK1 and SREBF2) were identified to be associated with prognosis. The accuracy of the prognostic model was confirmed using the GSE42669 dataset, with risk score and MGMT promoter status identified as independent prognostic factors. GSEA showed enrichment of immune response activation and cell cycle regulatory pathways. qRT-PCR validation showed up-regulation of PNPLA3 and SARDH, and down-regulation of ADSL, in tumor tissues. Conclusions This study identified six PMRGs (SARDH, ACHE, ADSL, PNPLA3, MAPK1 and SREBF2) as potential prognostic biomarkers for glioblastoma. These biomarkers reveal the role of propionate metabolism in the progression of glioblastoma and may serve as important indicators of patient prognosis and treatment strategies.
Collapse
Affiliation(s)
- Yuchen Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huijuan Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
126
|
Oliveira EGD, Lanza JS, Cojean S, Moreira POL, Tunes LG, Gomes ML, DeFreitas-Silva G, Silva VSD, Veltri ERP, Torres-Santos EC, Demicheli C, Pomel S, Loiseau PM, Frézard F, Fernandez-Prada C, Andrade-Neto VVD, Monte-Neto RLD. Re-sensitization of antimony-resistant Leishmania by highly potent Sb V-porphyrin through the involvement of ERG6-coding gene. Biomed Pharmacother 2025; 187:118059. [PMID: 40250113 DOI: 10.1016/j.biopha.2025.118059] [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: 12/26/2024] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Leishmaniasis chemotherapy faces significant challenges, including high costs, severe side effects, and the emergence of drug-resistant parasites, demanding global efforts to identify novel antileishmanial agents. Pentavalent antimony (SbV), the primary treatment for over 78 years, suffers from reduced efficacy and high toxicity, underscoring the urgent need for alternatives. We have synthesized metalloporphyrins with potent antileishmanial properties, including the SbV-porphyrin complex (SbVT4MPP). SbVT4MPP exhibited high potency against both Sb-sensitive and Sb-resistant Leishmania spp. with IC50 as low as 0.05 and 0.12 µM, respectively, for amastigotes and promastigotes; 170-fold more effective than SbV and with a 28-37-fold selectivity index, highlighting the importance of host cells on drug activity. Sterol profiling of L. infantum revealed that SbVT4MPP ablated ergosterol production while accumulating cholestane-based sterols (e.g., cholesta-5,7,22-trien-3β-ol). Additional sterols appeared exclusively under SbVT4MPP treatment, accompanied by upregulated erg6, encoding sterol-C-24 methyltransferase (SMT), a key enzyme in sterol biosynthesis. Overexpression of ERG6 reduced SbVT4MPP potency, increasing the IC50 by 2.5-fold, confirming ERG6's role in its mode of action. Disruption of ergosterol biosynthesis was indirectly confirmed through hypoosmotic shock assays, which indicated increased membrane fluidity in SbVT4MPP-treated Leishmania. In vivo studies revealed a 96 % reduction in parasite load, highlighting SbVT4MPP's efficacy in visceral leishmaniasis model. Since ERG6 is absent in mammals, it represents a selective and promising pharmacological target. Our findings position SbVT4MPP as a novel chemical entity with potent in vitro and in vivo antileishmanial efficacy, providing mechanistic insights and warranting further preclinical investigation as a promising drug candidate for leishmaniasis treatment.
Collapse
Affiliation(s)
- Ellen Gonçalves de Oliveira
- Biotecnologia Aplicada a Patógenos (BAP), Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| | - Juliane Sousa Lanza
- Faculté de Pharmacie, Chimiotérapie Antiparasitaire (PARACHEM), UMR 8076 CNRS BioCIS, Université Paris-Saclay, Orsay, France; Laboratório de Sistemas Nanoestruturados - LABnano - Departamento de Fisiologia e Biofísica - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Sandrine Cojean
- Faculté de Pharmacie, Chimiotérapie Antiparasitaire (PARACHEM), UMR 8076 CNRS BioCIS, Université Paris-Saclay, Orsay, France.
| | - Paulo Otávio L Moreira
- Biotecnologia Aplicada a Patógenos (BAP), Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| | - Luiza Guimarães Tunes
- Biotecnologia Aplicada a Patógenos (BAP), Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| | - Marcela Luísa Gomes
- Departamento de Química - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | | | - Vinícius Santos da Silva
- Centro de Ciências Exatas e Tecnológicas - Universidade Federal do Recôncavo da Bahia, Bahia, Brazil.
| | - Eduardo Raul Pereira Veltri
- Laboratório de Bioquímica de Tripanosomatídeos - Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| | - Eduardo Caio Torres-Santos
- Laboratório de Bioquímica de Tripanosomatídeos - Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| | - Cynthia Demicheli
- Departamento de Química - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Sébastien Pomel
- Faculté de Pharmacie, Chimiotérapie Antiparasitaire (PARACHEM), UMR 8076 CNRS BioCIS, Université Paris-Saclay, Orsay, France.
| | - Philippe M Loiseau
- Faculté de Pharmacie, Chimiotérapie Antiparasitaire (PARACHEM), UMR 8076 CNRS BioCIS, Université Paris-Saclay, Orsay, France.
| | - Frédéric Frézard
- Laboratório de Sistemas Nanoestruturados - LABnano - Departamento de Fisiologia e Biofísica - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | | | - Valter Viana de Andrade-Neto
- Laboratório de Bioquímica de Tripanosomatídeos - Instituto Oswaldo Cruz - Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| | - Rubens Lima do Monte-Neto
- Biotecnologia Aplicada a Patógenos (BAP), Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| |
Collapse
|
127
|
Teichenné J, Tobajas Y, Leonard K, Tchoumtchoua J, Escoté X. Sustainable Polyphenol-Rich Extracts from Agricultural By-Products: Infectivity Inhibition Potential for Human Coronavirus 229E. Molecules 2025; 30:1806. [PMID: 40333750 PMCID: PMC12029669 DOI: 10.3390/molecules30081806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Polyphenol-rich extracts derived from agricultural by-products exhibit promising antiviral properties. This study evaluated the antiviral potential of extracts from red onion peels, vineyard prunings, olive prunings and chicory leaves against human coronavirus HuCoV-229E. Subcritical water extraction and resin adsorption techniques were applied to produce the extracts. The extracts were further characterised for their bioactive content, and three out of four extracts showed a high polyphenol content (>200 mg/g). The antiviral activity was assessed through viral infectivity and replication inhibition assays in human MRC-5 host cells. The results indicate that chicory leaf and red onion peel extracts demonstrated significant antiviral effects, with effective concentrations (EC50) of 61.43 µg/mL and 10.1 µg/mL, respectively. Olive pruning extract exhibited moderate activity, while vineyard pruning extract showed limited efficacy. These findings suggest that polyphenol-rich agricultural by-products could serve as sustainable sources for antiviral agents, warranting further investigation into their mechanisms of action and potential applications against other coronaviruses, including SARS-CoV-2.
Collapse
Affiliation(s)
- Joan Teichenné
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (Y.T.); (X.E.)
| | - Yaiza Tobajas
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (Y.T.); (X.E.)
| | - Kevin Leonard
- Biomass Valorisation Platform, Extraction Department, CELABOR Srl, 4650 Herve, Belgium; (K.L.); (J.T.)
| | - Job Tchoumtchoua
- Biomass Valorisation Platform, Extraction Department, CELABOR Srl, 4650 Herve, Belgium; (K.L.); (J.T.)
| | - Xavier Escoté
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (Y.T.); (X.E.)
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
- CIBER in Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
128
|
Hassan MG, Abdel-Monem MO, Sleem ASMA, El Awady ME, Hamed AA. Antimicrobial, antibiofilm, cytotoxicity, and anti-DNA topoisomerase activity of Streptomyces sp. 22SH with ADME and in silico study. BMC Microbiol 2025; 25:219. [PMID: 40240972 PMCID: PMC12001559 DOI: 10.1186/s12866-025-03912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The genus Streptomyces has been recently proven to be a valuable and rich source of producing several bioactive compounds with substantial biological activity and applications in many fields such as medicine, environmental science, food industries, and agronomy. This study highlights the importance of Streptomyces as an antimicrobial, antibiofilm, and anticancer. Out of the 75 actinobacteria isolated from both marine and soil habitats, one isolate, HG2, was selected based on its potent antimicrobial activity. The isolate has been identified morphologically by studying colony and spore chain morphology using TEM and genetically by sequencing their 16 sr RNA gene as Streptomyces sp. 22SH with Accession number OK326829.1. Bioassay-guided fractionation of the Streptomyces sp. 22SH crude extract led to the isolation and purification of Cis-9-Octadecenoic. Biological evaluation including antimicrobial and antibiofilm activity of the crude and purified compound was performed on four clinical microbes (S. aureus ATCC6538-P, B. subtilis ATCC6633, and P. aeruginosa ATCC27853). The compound showed the ability to eradicate the biofilm formation by the tested pathogens. Additionally, the antitumor activity was assessed, and the compound showed a cytotoxic effect against liver carcinoma and breast cancer cells, with IC50 values of 17.48 ± 0.94 and 88.73 ± 4.78 µg/ml, respectively. While it displayed anti-topoisomerase activity with an IC50 of 0.65 ± 0.023 µg/ml. Furthermore, the compound's ADME-related physicochemical features and docking analysis were investigated.
Collapse
Affiliation(s)
- Mervat G Hassan
- Botany and Microbiology Department, Faculty of Science, Benha Univ., Benha, Egypt.
| | | | - Al Shaimaa M A Sleem
- Botany and Microbiology Department, Faculty of Science, Benha Univ., Benha, Egypt
| | - Mohamed E El Awady
- Microbial Biotechnology Department, National Research Centre, El-Buhouth St. 33, Dokki-Cairo, 12622, Egypt
| | - Ahmed A Hamed
- Microbial Chemistry Department, National Research Centre, El-Buhouth St. 33, Dokki-Cairo, 12622, Egypt.
| |
Collapse
|
129
|
de Araujo TS, da Costa AC, Dias Leite da Silva C, Ribeiro FDS, de Andrade RA, Paula Neto HA, Carvalho RS, Lima LMTR, Almeida MDS. Biochemical and Biophysical Divergences between Two Escherichia coli l-Asparaginase II Variants: Potential for Using EcA2-K12 as a Biosimilar. Biochemistry 2025. [PMID: 40237204 DOI: 10.1021/acs.biochem.4c00663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Escherichia coli l-asparaginase II (EcA2) is essential for treating Acute Lymphoblastic Leukemia, the most common childhood cancer. This enzyme catalyzes the hydrolysis of l-asparagine or l-glutamine to ammonia and l-aspartate or l-glutamate. The first FDA-approved EcA2 biopharmaceutical, Elspar, was introduced in 1978, followed by other biosimilars. Despite stringent approval criteria, variations in plasmatic activity and therapeutic efficacy persist across different EcA2 preparations, often leading to substandard product notifications. Many studies focus on the EcA2 from the E. coli K12 strain (EcA2-K12), which differs by four amino acids from reference biopharmaceuticals, including Elspar (EcA2-4M). Here, we show that EcA2-4 M has over twice the specific activity on both the hydrolysis of l-asparagine and on human lymphoblast cells compared to EcA2-K12. EcA2-K12 demonstrates 4-fold greater specificity for l-asparagine over l-glutamine, considering their kcat, but similar KM toward each amino acid. Interestingly, EcA2-K12 has 3-fold lower affinity for l-aspartate, linked to reduced stabilization of its N-terminal active site loop. Although both variants exhibit indistinguishable thermostability, EcA-K12 shows a higher tendency to oligomerize. We solved the 3D structures of both variants by X-ray crystallography, and normal-mode analysis revealed wider conformational changes in EcAK12's active site. Our data indicate that EcA2-K12 has lower activity due to the higher conformational dynamics of the N-terminal active site loop. Nevertheless, EcA2-K12 is a beneficial alternative or complement to existing therapeutic schemes with EcA2-4M, due to its higher specificity to l-asparagine, which is of fundamental importance since activity on l-glutamine is associated with harmful side effects.
Collapse
Affiliation(s)
- Talita Stelling de Araujo
- Protein Advanced Biochemistry (PAB), Institute of Medical Biochemistry (IBqM)-National Center for Structural Biology and Bioimaging (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Anna Catharinna da Costa
- Protein Advanced Biochemistry (PAB), Institute of Medical Biochemistry (IBqM)-National Center for Structural Biology and Bioimaging (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-Graduação em Química Biológica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Camila Dias Leite da Silva
- Protein Advanced Biochemistry (PAB), Institute of Medical Biochemistry (IBqM)-National Center for Structural Biology and Bioimaging (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Fernando de Sá Ribeiro
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-Graduação em Química Biológica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Rafael Alves de Andrade
- Protein Advanced Biochemistry (PAB), Institute of Medical Biochemistry (IBqM)-National Center for Structural Biology and Bioimaging (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Heitor Affonso Paula Neto
- Laboratório de Alvos Moleculares (LAM), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Renato Sampaio Carvalho
- Laboratório de Alvos Moleculares (LAM), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Luís Maurício T R Lima
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-Graduação em Química Biológica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-graduação em Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Marcius da Silva Almeida
- Protein Advanced Biochemistry (PAB), Institute of Medical Biochemistry (IBqM)-National Center for Structural Biology and Bioimaging (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-Graduação em Química Biológica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
130
|
Gómez-Guerrero E, Debray-García Y, Aztatzi-Aguilar OG, Colchero-Amateco FA, Amador-Muñoz O, Poblano-Bata J, Poblete-Naredo I, Albores A. A549 cells exposed to a marijuana smoke extract affect mono-layer integrity related to oxidative stress state. Toxicol In Vitro 2025; 107:106072. [PMID: 40250736 DOI: 10.1016/j.tiv.2025.106072] [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: 07/04/2024] [Revised: 03/14/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025]
Abstract
Marijuana smoke contains several toxic compounds that may induce dysregulation of oxidative mechanisms and barrier system in airway alveolar cells. This study aimed to assess whether marijuana smoke extract (MSE) modifies mono-layer integrity and antioxidant effects on the alveolar epithelial cells. A549 cells were exposed to MSE (0.1 to 5 μg/mL) or cannabinoid (+)-WIN 55,212-2 (WIN; 0.01 to 1 μM) for 24 h. Epithelial integrity and protein expression of claudin (Cl)-2, Cl-5, and occludin (Ocl) were evaluated by transepithelial electrical resistance (TEER), permeability assay, Western blot, immunofluorescence, and qPCR. TEER decreased after MSE or WIN exposure, whereas the monolayer permeability increased. Protein expression and localization of Cl-2 and Ocl were reduced after MSE treatment. However, WIN increased Cl-2 protein and decreased Cl-5 and Ocl. Differential gene expressions were observed between treatments. Malondialdehyde (MDA) production and advanced oxidation protein products (AOPP) determination showed that MSE increased AOPP, whereas WIN augmented the MDA production and decreased AOPP levels. The activity of antioxidant enzymes shows an increase in catalase, glutathione-S-transferase, γ-glutamyl transferase and arginase after MSE treatment and a decrease with WIN. Data indicates that MSE exposure alters epithelial integrity and the alveolar cells antioxidant response that, finally, may lead to lung damage.
Collapse
Affiliation(s)
- Elvira Gómez-Guerrero
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Yazmín Debray-García
- Departamento de Investigación de Toxicología y Medicina Ambiental, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Octavio Gamaliel Aztatzi-Aguilar
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Fanny Azuzena Colchero-Amateco
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Omar Amador-Muñoz
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Josefina Poblano-Bata
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Irais Poblete-Naredo
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Arnulfo Albores
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico.
| |
Collapse
|
131
|
Halmai M, Donkó-Tóth V, Keglevich P, Kánai K, Weber M, Dékány M, Abdallah EA, Bózsity N, Zupkó I, Nehr-Majoros A, Szőke É, Helyes Z, Hazai L. Synthesis and In Vitro Evaluation of the Anticancer Effect of Novel Phosphonium Vindoline Derivatives. Int J Mol Sci 2025; 26:3775. [PMID: 40332477 PMCID: PMC12028158 DOI: 10.3390/ijms26083775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
The Vinca alkaloid vindoline was coupled at position 17 with several trisubstituted phosphine derivatives and their in vitro anticancer activities on 60 human tumor cell lines (NCI60) were investigated. This phosphonium-type ionic side chain is beneficial because it allows therapeutic molecules to pass through the cell membrane. Thus, the candidates coupled to it can exert their activities in the mitochondria. The coupling of vindoline with the trisubstituted phosphines was achieved through flexible or rigid linkers. Instead of the ionic phosphonium structural part, a neutral moiety, namely the triphenylmethyl group, was also added to the side chain, being sterically similar but without a charge and phosphorus atom. In addition, the triphenylphosphine element was also built at position 10 of vindoline. Most of the derivatives showed low micromolar growth inhibition (GI50) values against most cell lines. Among them, conjugate 9e was outstanding: it exhibited nanomolar anticancer activity on the RPMI-8226 leukemia cell line (GI50 = 20.0 nM). Compound 9g elicited cell cycle disturbance and apoptosis on A2780 ovary cancer cells and inhibited their migration at subantiproliferative concentrations. The selectivity of the conjugates was determined by their effects on non-tumor Chinese hamster ovary (CHO) cells in the CellTiter-Glo Luminescent Cell Viability Assay. Compound 9e showed an estimated half-maximal inhibitory concentration (IC50) value of 1.36 µM, suggesting good selectivity on cancer cells. These results open new perspectives of novel phosphonium-based vindoline derivatives as anticancer compounds.
Collapse
Affiliation(s)
- Mónika Halmai
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Viktória Donkó-Tóth
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Péter Keglevich
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Károly Kánai
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Márton Weber
- Spectroscopic Research Department, Gedeon Richter Plc., Gyömrői út 19-21, H-1103 Budapest, Hungary
| | - Miklós Dékány
- Spectroscopic Research Department, Gedeon Richter Plc., Gyömrői út 19-21, H-1103 Budapest, Hungary
| | - Ejlal A. Abdallah
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Noémi Bózsity
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Andrea Nehr-Majoros
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- HUN-REN PTE Chronic Pain Research Group, Szigeti út 12, H-7624 Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- HUN-REN PTE Chronic Pain Research Group, Szigeti út 12, H-7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- HUN-REN PTE Chronic Pain Research Group, Szigeti út 12, H-7624 Pécs, Hungary
- PharmInVivo Ltd., Szondi Gy. u. 10, H-7629 Pécs, Hungary
| | - László Hazai
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| |
Collapse
|
132
|
Pfleger T, Ortmayr K, Steiner K, Zaher R, Seiser S, Elbe-Bürger A, Heiss E, Klang V. Radical scavenging effect of skin delivery systems using Korean red ginseng extract and assessment of their biocompatibility with human primary dermal fibroblasts and HaCaT keratinocytes. Int J Pharm 2025; 674:125477. [PMID: 40097056 DOI: 10.1016/j.ijpharm.2025.125477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Korean red ginseng (KRG) extract is proposed for cosmetic use, but no data on biological effects of KRG-loaded vehicles exist. The study aimed to optimize new multi- and monophase vehicles for KRG extract delivery, assess their biocompatibility and evaluate their radical scavenging effect in vitro. Storage stability of oil-in-water nanoemulsions (NEs) and hydroalcoholic gels (2 % w/w KRG) was assessed over twelve weeks using dynamic light scattering, rheology and pH measurements. Release profiles of ginsenosides Rb1 (more hydrophilic) and Rg1 (moderately lipophilic) through a cellulose membrane were also investigated employing Franz diffusion cells. Antioxidant potential and biocompatibility were assessed via 2,2-diphenyl-1-picrylhydrazyl (DPPH) and cell viability assays. Vehicles remained stable over twelve weeks at 8 °C (NEs Dh stable, gel viscosity + 3.5 %). Diffusion studies showed higher release of Rg1 vs. Rb1 (7.10 vs. 1.39 µg/cm-2 after 28 h). KRG-formulations demonstrated good biocompatibility with primary human dermal fibroblasts and HaCaT keratinocytes (72-94 % viability). Radical scavenging capacity of KRG extract did not differ between pure and incorporated form and was lower than that of a Hypericum extract or ascorbic acid. Results render KRG-formulations a potentially promising alternative to conventional antioxidants used in daily products.
Collapse
Affiliation(s)
- Tanja Pfleger
- University of Vienna, Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology and Biopharmaceutics, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Karin Ortmayr
- University of Vienna, Department of Pharmaceutical Sciences, Division of Pharmacognosy, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Katja Steiner
- University of Vienna, Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology and Biopharmaceutics, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Rawan Zaher
- University of Vienna, Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology and Biopharmaceutics, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Saskia Seiser
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Adelheid Elbe-Bürger
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Elke Heiss
- University of Vienna, Department of Pharmaceutical Sciences, Division of Pharmacognosy, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Victoria Klang
- University of Vienna, Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology and Biopharmaceutics, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| |
Collapse
|
133
|
da Silva Milhorini S, Rutckeviski R, Centa A, Ribeiro Smiderle F, Zavadinack M, Rosado FR, Iacomini M. Different molecular weight fucogalactans from Macrocybe titans mushroom promote distinct effect on breast cancer cell death. Carbohydr Polym 2025; 354:123318. [PMID: 39978922 DOI: 10.1016/j.carbpol.2025.123318] [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: 09/17/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
There is an incessant search for new therapies against cancer, able to unite effectiveness with specificity, leading to higher survival rates and lower deleterious effects. Herein, two fucogalactans (F-1 and F-2), isolated from Macrocybe titans, showed a (1 → 6)-linked α-D-Galp main chain partially substituted at O-2 by non-reducing end units of α-L-Fucp, with different Mw, being F-2 > 20 times higher than F-1. Both fucogalactans induced cell cycle arrest of MDA-MB-231 cells in G1 phase after 120 h of treatment. However, only F2 resulted in increased apoptosis and necrosis. For the MCF-7 cell line, no changes in the cell cycle and cell death were observed at 120 h. The non-tumoral cell line (VERO) was not affected. The results confirmed that polysaccharides with different Mw may have distinct effects and therefore this is an important feature to be considered on investigating anti-cancer treatments.
Collapse
Affiliation(s)
- Shayane da Silva Milhorini
- Department of Biochemistry and Molecular Biology, Federal University of Parana, CEP, 81531-980 Curitiba, PR, Brazil.
| | - Renata Rutckeviski
- Faculdades Pequeno Príncipe, CEP, 80230-020 Curitiba, PR, Brazil; Instituto de Pesquisa Pelé Pequeno Príncipe, CEP, 80240-020 Curitiba, PR, Brazil
| | - Ariana Centa
- Universidade Alto Vale do Rio do Peixe, CEP, 89500-000 Caçador, SC, Brazil
| | - Fhernanda Ribeiro Smiderle
- Faculdades Pequeno Príncipe, CEP, 80230-020 Curitiba, PR, Brazil; Instituto de Pesquisa Pelé Pequeno Príncipe, CEP, 80240-020 Curitiba, PR, Brazil
| | - Matheus Zavadinack
- Department of Biochemistry and Molecular Biology, Federal University of Parana, CEP, 81531-980 Curitiba, PR, Brazil
| | - Fábio Rogério Rosado
- Department of Biosciences, Federal University of Parana, CEP, 85950-000 Palotina, PR, Brazil
| | - Marcello Iacomini
- Department of Biochemistry and Molecular Biology, Federal University of Parana, CEP, 81531-980 Curitiba, PR, Brazil
| |
Collapse
|
134
|
Del Mármol C, Scalese G, Moreira R, Veiga N, Machado I, Faccio R, Lima A, Peralta RA, Pérez-Díaz L, Gambino D. Exploring a series of multifunctional Mn(I) tricarbonyls as prospective agents against trypanosomatid parasites: a comparative study with the Re(I) analogues. Dalton Trans 2025; 54:6495-6516. [PMID: 40138200 DOI: 10.1039/d5dt00241a] [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: 03/29/2025]
Abstract
Diseases caused by trypanosomatid parasites are among the most pressing neglected illnesses. Chagas disease, caused by Trypanosoma cruzi, and visceral Leishmaniasis, caused by Leishmania infantum, have a severe health impact in developing countries. Searching for prospective metal-based drugs against these diseases, five multifunctional fac-[Mn(CO)3(CTZ)(NN)](PF6) compounds, including four new derivatives, were synthesized and thoroughly characterized, featuring NN polypyridyl derivatives and Clotrimazole (CTZ) as bioactive ligands. The biological behavior was compared with that previously reported for the Re analogues. Mn compounds showed EC50 values in the low micromolar range against the infective trypomastigote form of Trypanosoma cruzi and the promastigote form of Leishmania infantum and moderate selectivity indexes. While their potency against T. cruzi was comparable to the Re analogues, their selectivity was lower. Key physicochemical properties relevant to drug development were assessed: Mn(I) compounds showed lower stability in relevant tested media compared with their Re(I) counterparts and higher lipophilicity than the free ligands and the Re analogues. To gain insight into the potential mechanisms of action, the interaction with DNA and the effects on ergosterol biosynthesis in T. cruzi and L. infantum were investigated. Minimal DNA association (<1%) and moderate interaction with this target discarded DNA binding as the primary mechanism of action. In contrast, inhibition of lanosterol 14-α-demethylase (CYP51), key enzyme involved in the parasites' ergosterol biosynthetic pathway, was experimentally confirmed. Metallomic study revealed an uptake by T. cruzi of the most promising compound, fac-[Mn(CO)3(CTZ)(tmp)](PF6), more than twice that of the Re(I) analogue and preferential association to soluble proteins. Proteomic analysis of T. cruzi epimastigotes treated with the Mn(I) and Re(I) analogues showed no change in CYP51 abundance, suggesting that reduced ergosterol levels may arise from post-translational modifications of the enzyme. Raman confocal microscopy allowed us to detect effects of the most promising Mn compound in treated T. cruzi. Furthermore, the photoinduced CO release properties of both Mn and Re analogues were examined, searching for an additional and yet non-studied potential mechanism of action of metal-tricarbonyls in these trypanosomatid parasites. Collectively, the results highlight the potential of Mn(I) tricarbonyls as promising candidates for further drug development.
Collapse
Affiliation(s)
- Carolina Del Mármol
- Área Química Inorgánica, DEC, Facultad de Química, Universidad de la República, Uruguay.
- Programa de Posgrado de Facultad de Química, Universidad de la República, Uruguay
- PEDECIBA - Programa de Desarrollo de las Ciencias Básicas, Uruguay
| | - Gonzalo Scalese
- Área Química Inorgánica, DEC, Facultad de Química, Universidad de la República, Uruguay.
| | - Rodrigo Moreira
- Área Química Inorgánica, DEC, Facultad de Química, Universidad de la República, Uruguay.
- Programa de Posgrado de Facultad de Química, Universidad de la República, Uruguay
- PEDECIBA - Programa de Desarrollo de las Ciencias Básicas, Uruguay
| | - Nicolás Veiga
- Área Química Inorgánica, DEC, Facultad de Química, Universidad de la República, Uruguay.
| | - Ignacio Machado
- Área Química Analítica, DEC, Facultad de Química, Universidad de la República, Uruguay
| | - Ricardo Faccio
- Área Física, DETEMA, Facultad de Química, Universidad de la República, Uruguay
| | - Analía Lima
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo, Uruguay
- Instituto de Investigaciones Biológicas Clemente Estable, Uruguay
| | - Rosely A Peralta
- Departamento de Química, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Leticia Pérez-Díaz
- Sección Genómica Funcional, Facultad de Ciencias, Universidad de la República, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, DEC, Facultad de Química, Universidad de la República, Uruguay.
| |
Collapse
|
135
|
Vidović S, Ilić M, Nakomčić J, Nastić N, Kvrgić J, Song X, Jakimov D, Galović AJ, Jovanović NL, Zloh M. Mining bioactive components in agricultural crop and food production residue for sustainable solutions: In silico screening for skin anti-ageing properties. Int J Cosmet Sci 2025. [PMID: 40231485 DOI: 10.1111/ics.13059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/10/2025] [Indexed: 04/16/2025]
Abstract
Possible sustainable resources of beneficial compounds for various applications are agricultural crop and food production residues (ACFPR), which are supported by considerable efforts to characterize their compositions and biological activities. This knowledge can be utilized for the rational selection of agricultural crop residue extracts and their components and possible use in the development of value-added products, such as anti-ageing cosmetics. The appearance of wrinkles, pigmentation, and a reduction in skin elasticity are typical signs of ageing skin that are often alleviated by natural product-based preparations. Here, we use in silico approaches to identify natural compounds from agricultural crop and food production residues with the potential to alleviate symptoms of or reverse the skin ageing process. Target predictions combined with extensive database and literature searches were utilized to identify compounds present in ACR and proteins linked to skin ageing. The binding affinity of natural products to selected proteins using molecular docking and the respective intermolecular interaction analyses are predicted to provide an indicative measure of the compounds' potential for skin anti-ageing activity. A number of natural compounds with the potential to interact with protein targets such as collagenase, elastase, and hyaluronidase were identified. In addition to in silico findings, cytotoxicity assays were conducted using rose hip seed extracts against Hs294T (human metastatic melanoma, ATCC HTB-140) and MRC-5 (normal fetal lung fibroblasts, ATCC CCL 171), demonstrating selective cytotoxicity. ELISA assays revealed that rose hip seed extracts induced a significant increase in SIRT1 levels (160% of control) and a reduction in TGF-β levels (80% of control). These experimental results support the potential of agricultural crop residue extracts in modulating key proteins involved in skin ageing, reinforcing their viability as ingredients in anti-ageing cosmetic formulations. An analysis of the molecular relationships and pathways that organic substances from sustainable sources can affect offers the potential for developing formulations for skin rejuvenation with possible synergistic effects by utilizing the rational design of innovative skincare products and laying the framework for more effective screening of anti-ageing compounds for different applications.
Collapse
Affiliation(s)
- Senka Vidović
- Faculty of Technology Novi Sad, University of Novi Sad, Novi Sad, Serbia
| | - Milan Ilić
- Faculty of Pharmacy, Novi Sad, University of Business Academy in Novi Sad, Novi Sad, Serbia
| | - Jelena Nakomčić
- Faculty of Pharmacy, Novi Sad, University of Business Academy in Novi Sad, Novi Sad, Serbia
| | - Nataša Nastić
- Faculty of Technology Novi Sad, University of Novi Sad, Novi Sad, Serbia
| | - Jelena Kvrgić
- Faculty of Pharmacy, Novi Sad, University of Business Academy in Novi Sad, Novi Sad, Serbia
| | - Xuanpeng Song
- UCL School of Pharmacy, University College London, London, UK
| | - Dimitar Jakimov
- Faculty of Medicine, Oncology Institute of Vojvodina, University of Novi Sad, Sremska Kamenica, Serbia
| | | | | | - Mire Zloh
- Faculty of Pharmacy, Novi Sad, University of Business Academy in Novi Sad, Novi Sad, Serbia
- UCL School of Pharmacy, University College London, London, UK
| |
Collapse
|
136
|
Grenier D, Gelin M, Yang Y, Mularoni A, Guichou JF, Delcros JG, Krimm I. Binding-Site Switch for Protein Kinase CK2 Inhibitors. ChemMedChem 2025; 20:e202400868. [PMID: 39835439 DOI: 10.1002/cmdc.202400868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The serine/threonine protein kinase CK2, a tetramer composed of a regulatory dimer (CK2β2) bound to two catalytic subunits CK2α, is a well-established therapeutic target for various pathologies, including cancer and viral infections. Several types of CK2 inhibitors have been developed, including inhibitors that bind to the catalytic ATP-site, bivalent inhibitors that occupy both the CK2α ATP-site and the αD pocket, and inhibitors that target the CK2α/CK2β interface. Interestingly, the bivalent inhibitor AB668 shares a similar chemical structure with the interface inhibitor CCH507. In this study, we designed analogs of CCH507 using structure-based and fragment-based approaches. The ability of these analogs to bind the CK2α/CK2β interface was evaluated using biolayer interferometry and fluorescence anisotropy-based assays. Their potency to inhibit CK2 kinase activity was determined using the bioluminescent ADP-Glo assay. These experiments allowed us to investigate which chemical modifications prevent the binding of the compounds at the CK2α/CK2β interface. Seven out of sixteen compounds conserved the ability to bind at the protein-protein interface, among which three compounds exhibited better interface inhibition compared to CCH507.
Collapse
Affiliation(s)
- Dylan Grenier
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, 69373, France
| | - Muriel Gelin
- CNRS, INSERM, Univ. Montpellier, Centre de Biologie Structurale, Montpellier, 34090, France
| | - Yinshan Yang
- CNRS, INSERM, Univ. Montpellier, Centre de Biologie Structurale, Montpellier, 34090, France
| | - Angélique Mularoni
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, 69373, France
| | - Jean-François Guichou
- CNRS, INSERM, Univ. Montpellier, Centre de Biologie Structurale, Montpellier, 34090, France
| | - Jean-Guy Delcros
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, 69373, France
| | - Isabelle Krimm
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, 69373, France
| |
Collapse
|
137
|
Yusein-Myashkova S, Vladimirova D, Gospodinov A, Ugrinova I, Todorova J. Metformin Inhibits Cell Motility and Proliferation of Triple-Negative Breast Cancer Cells by Blocking HMGB1/RAGE Signaling. Cells 2025; 14:590. [PMID: 40277915 PMCID: PMC12025684 DOI: 10.3390/cells14080590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
High-mobility group box 1 (HMGB1) is a nuclear chromatin protein overexpressed in various cancers and linked to tumor progression. Outside the cell, HMGB1 binds to receptors such as the receptor for advanced glycation end products (RAGE), promoting metastasis. Targeting this signaling pathway may provide a new therapeutic strategy for aggressive cancers. Metformin, a well-established antidiabetic drug, directly interacts with HMGB1, inhibiting its pro-inflammatory functions. This study investigates metformin's effects on the HMGB1/RAGE signaling pathway in triple-negative breast cancer (TNBC) cells. Using wound-healing and colony formation assays, we demonstrate that metformin reduces HMGB1-induced cell migration and proliferation. Immunoblotting and immunofluorescence analyses reveal that metformin decreases RAGE stabilization on the cell membrane, disrupts NF-κB signaling, and reverses the epithelial-to-mesenchymal transition (EMT) by increasing E-cadherin, reducing vimentin, and stabilizing β-catenin at the cell membrane. Furthermore, metformin lowers HMGB1 and RAGE protein levels, disrupting the positive feedback loop that promotes cancer aggressiveness. These findings highlight metformin's potential as a therapeutic agent in TNBC by inhibiting HMGB1/RAGE-driven metastasis.
Collapse
Affiliation(s)
| | | | | | | | - Jordana Todorova
- Roumen Tsanev Institute of Molecular Biology, Bulgarian Academy of Sciences, Acad G. Bonchev Str., Bl. 21, 1113 Sofia, Bulgaria; (S.Y.-M.); (D.V.); (A.G.); (I.U.)
| |
Collapse
|
138
|
Temgire P, Arthur R, Upadhayay S, Arora S, Kapatia G, Kumar R, Navik US, Kumar P. Elucidating the neuroprotective potential of arbutin in 3-NPA induced HD-like pathology: Insights from in silico, in vitro, and in vivo models. Behav Brain Res 2025; 483:115475. [PMID: 39929340 DOI: 10.1016/j.bbr.2025.115475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/24/2024] [Accepted: 02/06/2025] [Indexed: 02/16/2025]
Abstract
Huntington's disease (HD) is an inherited, hyperkinetic condition manifested by a triad of motor abnormalities, progressive cognitive impairment, and psychiatric disturbances. Oxidative stress has been implicated among other cellular processes in the pathogenesis of HD. Arbutin, a hydroquinone antioxidant, is reportedly neuroprotective in several animal models of neurodegenerative diseases. Hence, this research aimed to investigate the neuroprotective effect of arbutin against HD using in silico, in vitro, and in vivo experimental approaches. Schrodinger software was used for the in-silico studies, while SH-SY5Y cells were used for in-vitro studies. In the in vivo studies, adult Wistar rats were divided into five groups and 3-nitro propionic acid (3-NPA) (10 mg/kg/day,i.p) alone, and with arbutin (50 and 100 mg/kg/day,i.p.) was administered for 21 days. The body weight and behavioral parameters, including locomotor activity and motor coordination, were assessed on the 1st, 7th, 14th & 21st days. On the 22nd day, animals were sacrificed; the striatum was harvested for biochemical, neurochemical, and histopathological assessment. In silico, results indicated that arbutin showed a good binding affinity for target proteins like Akt and Nrf2. Further, arbutin prevented cell death and oxidative stress in SH-SY5Y cells induced by 3-NPA. In addition, arbutin ameliorated the 3-NPA induced motor impairments, purine nucleoside imbalances (adenosine levels and its metabolites hypoxanthine, xanthine, adenine), oxidative stress, and histological alterations in the experimental animals. In conclusion, the present findings indicate that arbutin could be used as an adjuvant for the management of Huntington's disease.
Collapse
Affiliation(s)
- Pooja Temgire
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sahil Arora
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Ghudda, Bathinda 151401, India
| | - Gargi Kapatia
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
| | - Raj Kumar
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Ghudda, Bathinda 151401, India
| | - Uma Shanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India.
| |
Collapse
|
139
|
da Silva MOB, Silva ACD, Torreão BDC, Costa RMPB, Bezerra RP, Silva SDFD, Oliva MLV, Xavier LSS, Viana IF, Lins RD, Lorena VMD, Viana Marques DDA. Therapeutical Potential and Immunomodulatory Profile of Arthrospira platensis Compounds against Chagas Disease. ACS Infect Dis 2025; 11:1008-1017. [PMID: 40159082 PMCID: PMC11998005 DOI: 10.1021/acsinfecdis.4c01052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/14/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Arthrospira platensis, an ancient cyanobacterium, is rich in bioactive compounds with therapeutic potential, supporting its use in studies for various health conditions, including infectious and chronic diseases. This study aimed to evaluate the antiparasitic, cytotoxic, and immunomodulatory activities of A. platensis compounds against Trypanosoma cruzi. Peripheral Blood Mononuclear Cells (PBMC) and T. cruzi trypomastigotes were cultured for cytotoxic and antiparasitic analyses. Cytotoxicity was assessed in PBMC treated with different concentrations of crude extract, obtained by mechanical agitation in 0.1 M TRIS-HCl buffer (pH 7.2), and purified protein by DEAE-Sephadex A-50 chromatography and FPLC. Immune response was analyzed in infected and uninfected PBMC by measuring cytokines (IFN-γ, TNF, IL-2, IL-6, and IL-10) after treatment with purified protein and benznidazole. In vitro experiments showed that both crude extract and a 15 kDa purified protein were toxic to trypomastigotes in a dose-dependent manner, eliminating over 80% of the parasite at 1000 and 200 μg/mL, respectively. Both the extract and protein were nontoxic to PBMC, with the protein (SI: 20.7) being more selective than benznidazole (SI: 11.5). Results indicated that the purified protein modulated the immune response in T. cruzi-infected individuals, inducing a protective Th1 response while controlling an excessive inflammatory response with appropriate IL-10 levels. The anti-T. cruzi activity of this protein, alone or in combination with the commercial drug, suggests it could be a low-cost, safer, and more tolerable therapy for Chagas disease treatment.
Collapse
Affiliation(s)
- Maria
Rafaele Oliveira Bezerra da Silva
- Integrated
Multi-User Laboratory in Applied Biotechnology, Institute of Biological Sciences, University of Pernambuco, Santo Amaro, Recife, Pernambuco 50100-130, Brazil
- Biotechnology
and Parasitology Laboratory, University
of Pernambuco, Santo
Amaro, Recife, Pernambuco 50100-130, Brazil
- Department
of Immunology, Aggeu Magalhães Institute, Fiocruz Pernambuco, Recife, Pernambuco 50670420, Brazil
| | - Ana Carla da Silva
- Department
of Immunology, Aggeu Magalhães Institute, Fiocruz Pernambuco, Recife, Pernambuco 50670420, Brazil
| | - Byannca de Carvalho Torreão
- Department
of Immunology, Aggeu Magalhães Institute, Fiocruz Pernambuco, Recife, Pernambuco 50670420, Brazil
| | - Romero Marcos Pedrosa Brandão Costa
- Integrated
Multi-User Laboratory in Applied Biotechnology, Institute of Biological Sciences, University of Pernambuco, Santo Amaro, Recife, Pernambuco 50100-130, Brazil
| | - Raquel Pedrosa Bezerra
- Department
of Animal Morphology and Physiology, Federal
Rural of University of Pernambuco, Recife, Pernambuco 52171-900, Brazil
| | | | - Maria Luiza Vilela Oliva
- Department
of Biochemistry, Federal University of São
Paulo (UNIFESP), São
Paulo 04021-001, Brazil
| | | | - Isabelle F.T. Viana
- Departament
of Virology, Aggeu Magalhães Institute, Fiocruz Pernambuco, Recife, Pernambuco 50670420, Brazil
| | - Roberto Dias Lins
- Departament
of Virology, Aggeu Magalhães Institute, Fiocruz Pernambuco, Recife, Pernambuco 50670420, Brazil
| | | | - Daniela de Araújo Viana Marques
- Integrated
Multi-User Laboratory in Applied Biotechnology, Institute of Biological Sciences, University of Pernambuco, Santo Amaro, Recife, Pernambuco 50100-130, Brazil
- Biotechnology
and Parasitology Laboratory, University
of Pernambuco, Santo
Amaro, Recife, Pernambuco 50100-130, Brazil
- Department
of Immunology, Aggeu Magalhães Institute, Fiocruz Pernambuco, Recife, Pernambuco 50670420, Brazil
| |
Collapse
|
140
|
Nemčovičová I, Kóňa J, Poláková M, Klunda T, Bitala A, Benko M, Lenhartová S, Nemčovič M. Small glycomimetic antagonists of the cytomegalovirus glycoprotein UL141 prevent binding to TRAIL death receptor. J Biol Chem 2025; 301:108490. [PMID: 40216247 DOI: 10.1016/j.jbc.2025.108490] [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: 12/18/2024] [Revised: 03/24/2025] [Accepted: 04/03/2025] [Indexed: 05/13/2025] Open
Abstract
Human cytomegalovirus (HCMV) UL141 inhibits immune recognition of virally infected cells by natural killer cells and cytotoxic T cells through modulation of cellular receptors (e.g., TRAIL-R2/-R1, CD155, CD112). Recent findings suggest that UL141 is also a critical component of the HCMV virion, further emphasizing its significance. In this study, we aimed to develop a small synthetic compound as a UL141 antagonist. Building on our crystal structure analysis, we designed compounds to specifically bind viral UL141, thereby blocking its interaction with target receptors thus inhibiting its immunoevasive functions. We evaluated a small library of synthesized compounds composed of diverse saccharide units conjugated with nonsaccharide moieties, such as nonionic glycolipids, pyrrolidines, and "click" conjugates. An ELISA-like TMB-binding assay, coupled with dynabeads coating, was employed to assess the ability of these compounds to inhibit TRAIL-R2 binding in vitro. The most promising compounds capable of inhibiting complex formation were further analyzed using surface plasmon resonance. Compound 18 exhibited the strongest binding affinity to UL141, with KD of 2.93 μM. Molecular docking studies identified specific binding sites on UL141, and the fragmented molecular orbital method was applied to evaluate interaction energy patterns between the antagonist and the UL141 protein. Mutational analysis was conducted to validate the identified binding sites on UL141. Additionally, cellular cytotoxicity assays were performed to confirm the nontoxic properties of these compounds. Collectively, our findings suggest that synthetic glycomimetics represent promising candidates for targeting the viral glycoprotein HCMV UL141, thereby disrupting TRAIL death receptor signaling, thus mitigating viral activity.
Collapse
Affiliation(s)
- Ivana Nemčovičová
- Biomedical Research Center (BMC), Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Juraj Kóňa
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia; Medical Vision, o. z., Bratislava, Slovakia
| | - Monika Poláková
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomáš Klunda
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Andrej Bitala
- Biomedical Research Center (BMC), Slovak Academy of Sciences, Bratislava, Slovakia
| | - Mário Benko
- Biomedical Research Center (BMC), Slovak Academy of Sciences, Bratislava, Slovakia
| | - Simona Lenhartová
- Biomedical Research Center (BMC), Slovak Academy of Sciences, Bratislava, Slovakia
| | - Marek Nemčovič
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
141
|
Teimury A, Khaledi EM, Hosseini ES. Antioxidant, anti-inflammatory, and anti-apoptotic activities of saffron and eryngium honey extracts. BMC Complement Med Ther 2025; 25:131. [PMID: 40211328 PMCID: PMC11987237 DOI: 10.1186/s12906-025-04867-1] [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: 12/09/2024] [Accepted: 03/19/2025] [Indexed: 04/13/2025] Open
Abstract
Saffron and Eryngium honeys are unique types of monofloral honeys collected from eastern and center of Iran. In present study, first, we investigated melissopalynological and physicochemical parameters in honey samples. Then we extracted the phenolic compounds and evaluated in vitro antioxidant by FRAP assay and anti-inflammatory activities by lipopolysaccharide induced RAW 264.7 macrophage cells. Phenolic extracts reduced nitric oxide and the secretion of the inflammatory cytokines IL-6 and TNFSF9 in the inflamed cells, and by reducing the NF-kB and increasing Nrf2 gene expression, showed an anti-inflammatory effect, but in general, Eryngium honey showed higher anti-inflammatory properties. Also, by studying the expression of Bcl-2, Bax and HO-1 genes, we showed that there is a relationship between inflammation and apoptosis in macrophage cells that the phenolic compounds showed their anti-apoptotic properties. Eryngium honey had a better performance in anti-apoptotic properties. Moreover, promising results indicate that Iranian honeys could be an important source of phenolic compounds presenting antioxidant and anti-inflammatory activity, which can have health-promoting properties if included in the diet and medicine.
Collapse
Affiliation(s)
- Azadeh Teimury
- Department of Cell and Molecular Biology, University of Kashan, Kashan, Iran
| | | | - Elahe Seyed Hosseini
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
142
|
Vágvölgyi M, Kocsis E, Tayeb BA, Zupkó I, Minorics R, Martins A, Hoyk Z, Ballai G, Szenti I, Kónya Z, Gáti T, Bogdán D, Tóth G, Hunyadi A. Ecdysteroid-Containing Squalenoylated Self-Assembling Nanoparticles Exert Tumor-Selective Sensitization to Reactive Oxygen Species (ROS)-Induced Oxidative Damage While Protecting Normal Cells: Implications for Selective Radiotherapy. J Med Chem 2025; 68:7197-7212. [PMID: 40152540 PMCID: PMC11997992 DOI: 10.1021/acs.jmedchem.4c02758] [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: 11/11/2024] [Revised: 03/04/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Central nervous system (CNS) tumors are exceptionally difficult to treat, and oxidative stress-inducing radiotherapy is an important treatment modality. In this study, we examined self-assembling pro-drug nanoconjugates of naturally derived antitumor ecdysteroids, which were designed to interfere with oxidative stress in brain tumor cells. Eight ecdysteroid-squalene conjugates were semi-synthesized and formulated into self-assembled aqueous nanosuspensions, which showed excellent stability for up to 16 weeks. The nanoassemblies demonstrated a strong dose-dependent sensitizing effect to tert-butyl hydroperoxide (tBHP)-induced oxidative damage in SH-SY5Y cells, while exerting a strong protective effect in MRC-5 fibroblast cells. In contrast, free ecdysteroids protected both cell lines from tBHP-induced damage. This suggests an important role for squalenoylation in the antitumor effect and indicates that our conjugates have potential as highly selective adjuvants in radiotherapy by sensitizing cancer cells and protecting surrounding tissues. Furthermore, our findings suggest a potential neuroprotective effect of nonconjugated ecdysteroids.
Collapse
Affiliation(s)
- Máté Vágvölgyi
- Institute
of Pharmacognosy, University of Szeged, Eötvös str. 6, Szeged H-6720, Hungary
| | - Endre Kocsis
- Institute
of Pharmacognosy, University of Szeged, Eötvös str. 6, Szeged H-6720, Hungary
| | - Bizhar A. Tayeb
- Institute
of Pharmacodynamics and Biopharmacy, University
of Szeged, Eötvös str. 6, Szeged H-6720, Hungary
| | - István Zupkó
- Institute
of Pharmacodynamics and Biopharmacy, University
of Szeged, Eötvös str. 6, Szeged H-6720, Hungary
| | - Renáta Minorics
- Institute
of Pharmacodynamics and Biopharmacy, University
of Szeged, Eötvös str. 6, Szeged H-6720, Hungary
| | - Ana Martins
- Institute
of Biophysics, HUN-REN Biological Research Centre, Temesvári blvd. 62, Szeged H-6726, Hungary
| | - Zsófia Hoyk
- Institute
of Biophysics, HUN-REN Biological Research Centre, Temesvári blvd. 62, Szeged H-6726, Hungary
| | - Gergő Ballai
- Department
of Applied and Environmental Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Rerrich Béla sq. 1, Szeged H-6720, Hungary
| | - Imre Szenti
- Department
of Applied and Environmental Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Rerrich Béla sq. 1, Szeged H-6720, Hungary
| | - Zoltán Kónya
- Department
of Applied and Environmental Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Rerrich Béla sq. 1, Szeged H-6720, Hungary
- HUN-REN-SZTE
Reaction Kinetics and Surface Chemistry Research Group, University of Szeged, Rerrich Béla sq. 1, Szeged H-6720, Hungary
| | - Tamás Gáti
- Servier
Research Institute of Medicinal Chemistry (SRIMC), Záhony str. 7, Budapest H-1031, Hungary
| | - Dóra Bogdán
- Department
of Organic Chemistry, Semmelweis University, Hőgyes Endre str. 7, Budapest H-1092, Hungary
| | - Gábor Tóth
- NMR
Group, Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Szt. Gellért sq. 4, Budapest H-1111, Hungary
| | - Attila Hunyadi
- Institute
of Pharmacognosy, University of Szeged, Eötvös str. 6, Szeged H-6720, Hungary
- HUN-REN-SZTE
Biologically Active Natural Products Research Group, Eötvös str. 6, H-6720 Szeged, Hungary
- Graduate
Institute of Natural Products, Shih-Chuan first Rd. 100, Kaohsiung 807, Taiwan
| |
Collapse
|
143
|
Marcolin E, Chemello C, Piovan A, Barbierato M, Morazzoni P, Ragazzi E, Zusso M. A Combination of 5-(3',4'-Dihydroxyphenyl)-γ-Valerolactone and Curcumin Synergistically Reduces Neuroinflammation in Cortical Microglia by Targeting the NLRP3 Inflammasome and the NOX2/Nrf2 Signaling Pathway. Nutrients 2025; 17:1316. [PMID: 40284180 PMCID: PMC12030566 DOI: 10.3390/nu17081316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: 5-(3',4'-dihydroxyphenyl)-γ-valerolactone (γ-VL), recently identified as a predominant microbial metabolite derived from proanthocyanidins, offers benefits such as reducing inflammation, oxidative stress, and supporting brain health. Its effects on neuroinflammation and microglial activation remain largely unexplored. Curcumin, a bioactive component isolated from Curcuma longa L., is well known for its ability to reduce microglial activation and pro-inflammatory mediator production and release. While the individual effects of γ-VL and curcumin are well documented, their potential combined effects remain unexplored. This research sought to investigate the possible synergistic effects of γ-VL and curcumin in reducing microglial activation. Methods: Primary rat cortical microglia were pre-treated with γ-VL and curcumin, alone or in combination, before stimulation with LPS. An MTT assay was used to evaluate cell viability, while pro-inflammatory mediators were assessed by real-time PCR and ELISA. Nitric oxide production was evaluated with the Griess assay. SynergyFinder Plus software analyzed potential synergistic effects. Results: The combination of low micromolar concentrations of γ-VL and curcumin synergistically reduced LPS-induced microglial activation. Specifically, the combination exhibited a significantly greater ability to inhibit the production and release of pro-inflammatory factors (such as IL-1β, TNF-α, and NO) compared to each compound individually. Mechanistically, the anti-inflammatory activity was attributed to the downregulation of NLRP3 expression, and the reduction in microglial activation was linked to the modulation of the NOX2/Nrf2 signaling pathway. Conclusions: The combination of low micromolar concentrations of γ-VL and curcumin produces synergistic anti-inflammatory effects in microglia by targeting key inflammatory pathways, indicating its potential utility as a treatment strategy for neurodegenerative diseases involving microglial activation.
Collapse
Affiliation(s)
- Emma Marcolin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (E.M.); (C.C.); (A.P.); (M.B.); (E.R.)
| | - Chiara Chemello
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (E.M.); (C.C.); (A.P.); (M.B.); (E.R.)
| | - Anna Piovan
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (E.M.); (C.C.); (A.P.); (M.B.); (E.R.)
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (E.M.); (C.C.); (A.P.); (M.B.); (E.R.)
| | - Paolo Morazzoni
- Nutraceutical Division, Distillerie Umberto Bonollo S.p.A., 35035 Mestrino, Italy;
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (E.M.); (C.C.); (A.P.); (M.B.); (E.R.)
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (E.M.); (C.C.); (A.P.); (M.B.); (E.R.)
| |
Collapse
|
144
|
Monteiro IDS, Fonseca ASA, dos Santos CR, de Carvalho JPS, da Silva SW, Veiga-Junior VF, Ribeiro R, Vieira IJC, Nogueira TSR, da Costa CAR, Machado GGL, Souza LR, Boas EVBV, Morais SS, Almeida JRGDS, Dutra LM, Santos VLDA, Silva AO, Sousa MH, Carneiro MLB, Joanitti GA. The Development and Characterization of an Andiroba Oil-Based Nanoemulsion ( Carapa guianensis, Aubl.): Insights into Its Physico-Chemical Features and In Vitro Potential Healing Effects. Pharmaceutics 2025; 17:498. [PMID: 40284494 PMCID: PMC12030521 DOI: 10.3390/pharmaceutics17040498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Andiroba oil, extracted from Carapa guianensis seeds, possesses therapeutic properties including anti-inflammatory and wound healing effects. This study aimed to develop and characterize a nanoemulsion formulation containing andiroba oil (NeAnd) and to evaluate its cytotoxicity and wound healing potential in vitro. Methods: The oil was evaluated for acidity, antioxidant activity, and fatty acid composition. NeAnd was produced by ultrasonication and characterized using FTIR (Fourier transform infrared spectroscopy), Raman spectroscopy, dynamic light scattering, and transmission electron microscopy. Results: NeAnd exhibited a spherical shape and stable physicochemical properties, with an average hydrodynamic diameter (HD) of 205.7 ± 3.9 nm, a polydispersity index (PdI) of 0.295 ± 0.05, a negative zeta potential of -4.16 ± 0.414 mV, and pH of approximately 6.5. These nanodroplets remained stable for 120 days when stored at 4 °C and maintained their parameters even under pH variations. FTIR and Raman analyses confirmed the presence of functional groups and the organization of fatty acid chains in NeAnd. Cell viability assays revealed no statistically significant differences in cytotoxicity at various concentrations (90-360 µg/mL) after 24 and 48 h. In scratch wound healing assays, NeAnd significantly enhanced wound closure (88.9%) compared to the PBS control (38%) and free andiroba oil (68.6%) in keratinocytes (p < 0.05). Conclusions: These promising findings indicate NeAnd as a potential nanophytomedicine for wound healing and tissue regeneration treatments.
Collapse
Affiliation(s)
- Isolda de Sousa Monteiro
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (I.d.S.M.); (A.S.A.F.); (C.R.d.S.); (S.S.M.); (M.L.B.C.)
- Post-Graduation Program in Sciences and Technologies in Health, Faculty of Ceilandia, University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (A.O.S.); (M.H.S.)
| | - Aimê Stefany Alves Fonseca
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (I.d.S.M.); (A.S.A.F.); (C.R.d.S.); (S.S.M.); (M.L.B.C.)
| | - Carolina Ramos dos Santos
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (I.d.S.M.); (A.S.A.F.); (C.R.d.S.); (S.S.M.); (M.L.B.C.)
| | - João Paulo Santos de Carvalho
- Laboratory of Optical Espectroscopy, Physics Institute, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasília 70910-900, DF, Brazil; (J.P.S.d.C.); (S.W.d.S.)
| | - Sebastião William da Silva
- Laboratory of Optical Espectroscopy, Physics Institute, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasília 70910-900, DF, Brazil; (J.P.S.d.C.); (S.W.d.S.)
| | - Valdir F. Veiga-Junior
- Chemistry Section, Military Institute of Engineering, Praça Gen. Tibúrcio, 80, Praia Vermelha, Rio de Janeiro 22290-270, RJ, Brazil; (V.F.V.-J.); (R.R.)
| | - Rayssa Ribeiro
- Chemistry Section, Military Institute of Engineering, Praça Gen. Tibúrcio, 80, Praia Vermelha, Rio de Janeiro 22290-270, RJ, Brazil; (V.F.V.-J.); (R.R.)
| | - Ivo José Curcino Vieira
- Laboratório de Ciências Químicas-LCQUI, Universidade Estadual do Norte Fluminense Darcy Ribeiro-UENF, Avenida Alberto Lamego 2000, Campos dos Goytacazes 28013-602, RJ, Brazil; (I.J.C.V.); (T.S.R.N.)
| | - Thalya Soares Ribeiro Nogueira
- Laboratório de Ciências Químicas-LCQUI, Universidade Estadual do Norte Fluminense Darcy Ribeiro-UENF, Avenida Alberto Lamego 2000, Campos dos Goytacazes 28013-602, RJ, Brazil; (I.J.C.V.); (T.S.R.N.)
| | - Carlos Alexandre Rocha da Costa
- Food Science Department—DCA, Federal University of Lavras—UFLA, Lavras CEP 37200-900, MG, Brazil; (C.A.R.d.C.); (G.G.L.M.); (L.R.S.); (E.V.B.V.B.)
| | - Gilson Gustavo Lucinda Machado
- Food Science Department—DCA, Federal University of Lavras—UFLA, Lavras CEP 37200-900, MG, Brazil; (C.A.R.d.C.); (G.G.L.M.); (L.R.S.); (E.V.B.V.B.)
| | - Lorrane Ribeiro Souza
- Food Science Department—DCA, Federal University of Lavras—UFLA, Lavras CEP 37200-900, MG, Brazil; (C.A.R.d.C.); (G.G.L.M.); (L.R.S.); (E.V.B.V.B.)
| | - Eduardo Valério Barros Vilas Boas
- Food Science Department—DCA, Federal University of Lavras—UFLA, Lavras CEP 37200-900, MG, Brazil; (C.A.R.d.C.); (G.G.L.M.); (L.R.S.); (E.V.B.V.B.)
| | - Samuel Silva Morais
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (I.d.S.M.); (A.S.A.F.); (C.R.d.S.); (S.S.M.); (M.L.B.C.)
| | - Jackson Roberto Guedes da Silva Almeida
- Núcleo de Estudos e Pesquisas de Plantas Medicinais (NEPLAME), Department of Pharmacy, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina 56304-917, PE, Brazil; (J.R.G.d.S.A.); (L.M.D.); (V.L.d.A.S.)
| | - Livia Macedo Dutra
- Núcleo de Estudos e Pesquisas de Plantas Medicinais (NEPLAME), Department of Pharmacy, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina 56304-917, PE, Brazil; (J.R.G.d.S.A.); (L.M.D.); (V.L.d.A.S.)
| | - Victória Laysna dos Anjos Santos
- Núcleo de Estudos e Pesquisas de Plantas Medicinais (NEPLAME), Department of Pharmacy, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina 56304-917, PE, Brazil; (J.R.G.d.S.A.); (L.M.D.); (V.L.d.A.S.)
| | - Atailson Oliveira Silva
- Post-Graduation Program in Sciences and Technologies in Health, Faculty of Ceilandia, University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (A.O.S.); (M.H.S.)
- Green Nanotechnology Group, University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil
| | - Marcelo Henrique Sousa
- Post-Graduation Program in Sciences and Technologies in Health, Faculty of Ceilandia, University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (A.O.S.); (M.H.S.)
- Green Nanotechnology Group, University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil
| | - Marcella Lemos Brettas Carneiro
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (I.d.S.M.); (A.S.A.F.); (C.R.d.S.); (S.S.M.); (M.L.B.C.)
| | - Graziella Anselmo Joanitti
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (I.d.S.M.); (A.S.A.F.); (C.R.d.S.); (S.S.M.); (M.L.B.C.)
- Post-Graduation Program in Sciences and Technologies in Health, Faculty of Ceilandia, University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasília 72220-275, DF, Brazil; (A.O.S.); (M.H.S.)
| |
Collapse
|
145
|
Alsahafi T, Bouback T, Albeshri A, Alnhhas S, Ali M, Moatasim Y, Kutkat O, Gaballah M, Alfasi F, Mater EH, Al-Sarraj F, Badierah R, Alotibi IA, Almulaiky YQ. Antiviral potential of Melissa officinalis extracts against influenza and emerging coronaviruses. Sci Rep 2025; 15:12118. [PMID: 40204903 PMCID: PMC11982357 DOI: 10.1038/s41598-025-96417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Melissa officinalis is a perennial medicinal plant traditionally used for its diverse biological activities, including antiviral properties. This study investigates the antiviral efficacy of various extracts, including water, acetone, alkaloid, non-alkaloid, ethanol, and methanol extracts, against influenza A (H1N1), SARS-CoV-2, and MERS-CoV. The water extract demonstrated significant inhibitory effects on SARS-CoV-2 (IC50 = 421.9 µg/mL) and MERS-CoV (IC50 = 222.1 µg/mL) in Vero E6 cells (an African green monkey kidney cell line), with a CC50 of 4221 µg/mL, indicating a favorable selectivity index. Additionally, it exhibited strong activity against H1N1 in Madin-Darby canine kidney cell line (MDCK cells) (IC50 = 57.30 µg/mL, CC50 = 3073 µg/mL). Among all the extracts, the methanol extract showed the highest antiviral activity. It has IC50 = 2.549 µg/ml and selectivity index (SI) = 230 against H1N1.While it showed IC50 = 10.83 µg/ml against SARS-CoV-2 and 9.82 µg/ml against MERS-CoV with SI values of 54.2 and 59.77, respectively. Molecular docking studies revealed that 5-Methyl-5 H-naphtho[2,3-c]carbazole,7 H-Dibenzo[b, g]carbazole, 7-methyl, hesperidin, luteolin-7-glucoside-3'-glucuronide, Melitric acid A, and other compounds exhibited high binding affinities to the receptor-binding domains (RBDs) of SARS-CoV-2 and MERS-CoV spike glycoproteins, suggesting their potential to interfere with viral entry. Furthermore, GC-MS-identified bioactive compounds, including docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), paromomycin, and phenolic acids, demonstrated additional antiviral potential. These results underscore the potential of M. officinalis extracts as natural antiviral agents, offering a foundation for further in vitro and in vivo validation and potential therapeutic applications against respiratory viral infections, including coronaviruses and influenza viruses.
Collapse
Affiliation(s)
- Tasneem Alsahafi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thamer Bouback
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
- Princess Dr. Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdul-Aziz University, Jeddah, 21589, Saudi Arabia.
| | - Abdulaziz Albeshri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sara Alnhhas
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed Ali
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo, 12622, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo, 12622, Egypt
| | - Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo, 12622, Egypt
| | - Mohamed Gaballah
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo, 12622, Egypt
| | - Fahad Alfasi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ehab H Mater
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Faisal Al-Sarraj
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Raied Badierah
- Medical Laboratory, King Abdulaziz University Hospital, King Abdul-Aziz University, Jeddah, 21589, Saudi Arabia
| | - Ibrahim A Alotibi
- Medical Laboratory, King Abdulaziz University Hospital, King Abdul-Aziz University, Jeddah, 21589, Saudi Arabia
| | - Yaaser Q Almulaiky
- The Applied College, University of Jeddah, Jeddah, Saudi Arabia.
- Chemistry Department, Faculty of Applied Science, Taiz University, Taiz, Yemen.
| |
Collapse
|
146
|
Bastone AL, John-Neek P, Dziadek V, Mansel F, Hagedorn M, Fleischauer J, Weigel B, Paul G, Schambach A, Rothe M. Meta-Analysis and Optimization of the In Vitro Immortalization Assay for Safety Assessment of Retroviral Vectors in Gene Therapy. Hum Gene Ther 2025. [PMID: 40200886 DOI: 10.1089/hum.2024.221] [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: 04/10/2025] Open
Abstract
The underlying risk of retroviral vector-induced insertional oncogenesis in gene therapies requires a reliable preclinical safety assessment. Dysregulation of genes neighboring the vector's integration sites has triggered hematopoietic malignancies in patients treated with different vector genera and designs. With ca. 18 years in practical use, the in vitro immortalization (IVIM) assay can quantify this mutagenic potential and is actively requested by regulatory authorities during preclinical stages. Here, we present a thorough meta-analysis of IVIM data alongside a step-by-step cell culture protocol. On this basis, we propose clonal outgrowth as the single indicator of mutagenicity, simplifying the IVIM assay cost- and time-wise.
Collapse
Affiliation(s)
- Antonella L Bastone
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Philipp John-Neek
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Violetta Dziadek
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Friederike Mansel
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Maike Hagedorn
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Jenni Fleischauer
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Bettina Weigel
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Gabi Paul
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
147
|
Fajardo J, Vianna MH, Ferreira TG, de O.Lemos AS, Souza TDF, Campos LM, Paula PDL, Andrade NB, Gamarano LR, Queiroz LS, Oliveira BDA, da Silva AD, Chedier LM, Denadai ÂL, Tavares GD, Barradas TN, Fabri RL. Enhanced Antitumor and Antibacterial Activities of Ursolic Acid through β-Cyclodextrin Inclusion Complexation. ACS OMEGA 2025; 10:12906-12916. [PMID: 40224443 PMCID: PMC11983339 DOI: 10.1021/acsomega.4c08337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/13/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
Ursolic acid (UA) is a pentacyclic triterpenoid known for its wide range of biological activities, including anticancer and antimicrobial effects. However, its poor solubility in water limits its therapeutic potential. Therefore, this work aims to evaluate the physicochemical properties of the ursolic acid/β-cyclodextrin inclusion complex (UA/βCD IC) and investigate the enhancement of the in vitro antitumor and antibacterial activities of UA when complexed with βCD. Molecular docking simulation showed that the carbonyl group of UA binds to the internal cavity of βCD, forming a hydrogen bond with the glucosidic residues of βCD. FTIR analysis revealed significant changes in the absorption peaks of UA/βCD IC, indicating interaction between the compounds, such as the reduction in intensity of the C=O and ν(O-H) bands. These results were supported by thermal analysis, as the degradation temperature of UA (233°C) and βCD (294°C) was suppressed in UA/βCD IC (191°C) compared to the free components. In addition, NMR analysis revealed significant changes in the chemical shift of the H located on the anomeric carbon (C1) of the glucose units in β-CD for the IC spectra (Δδ: 0.0041 ppm) compared to βCD, which are related to perturbations in the atomic electronic density. The colloidal characterization results also showed that UA/βCD IC has more stable colloidal properties with higher zeta potential values compared to free UA. As shown by the solubility assay, the interaction between UA and βCD formed stable inclusion complexes that increased the aqueous solubility of UA by approximately 35.85% (AUC: UA = 12.72, βCD = 6.78, UA/βCD = 17.28, p < 0.05). Scanning electron microscopy images revealed that IC was also associated with significant changes in particle shape and size. In addition, the UA/βCD IC showed greater antitumor activity than free UA, particularly in the MDA (71.95 ± 4.88%) and MCF-7 (73.40 ± 1.55%) cell lines. It showed similar efficacy to etoposide in HL60 (86.9 ± 0.84%) and JURKAT (85.35 ± 4.03%) cells. The UA/βCD IC significantly reduced the MIC values, improving the antibacterial activity particularly against E. faecalis (UA MIC: 31.3 μg/mL; UA/βCD MIC: 7.8 μg/mL), followed by S. aureus, B. cereus, and K. pneumoniae (UA MIC: 31.3 μg/mL; UA/βCD MIC: 15.6 μg/mL). Therefore, the UA/βCD IC significantly modifies the physicochemical properties of UA, resulting in enhanced aqueous solubility and biological properties, as confirmed by the improved antitumor and antibacterial activities.
Collapse
Affiliation(s)
- Júlia
B. Fajardo
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Mariana H. Vianna
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Thayná G. Ferreira
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Ari S. de O.Lemos
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Thalita de F. Souza
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Lara M. Campos
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Priscila de L. Paula
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Nubia B. Andrade
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Lívia R. Gamarano
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Lucas S. Queiroz
- Research
Group for Food Production Engineering, National Food Institute, Technical University of Denmark, Ørsteds Plads, Kongens
Lyngby 2800, Denmark
| | - Bruno de A. Oliveira
- Department
of Chemistry, Institute of Exact Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Adilson D. da Silva
- Department
of Chemistry, Institute of Exact Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Luciana M. Chedier
- Department
of Botany, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Ângelo
M. L. Denadai
- Department
of Pharmacy, Institute of Life Sciences, Federal University of Juiz de Fora, Campus Governador Valadares, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| | - Guilherme D. Tavares
- Department
of Pharmacy, Faculty of Pharmacy, Federal
University of Juiz de Fora, Juiz
de Fora, Minas Gerais CEP
36036-900, Brazil
| | - Thaís N. Barradas
- Department
of Pharmacy, Faculty of Pharmacy, Federal
University of Juiz de Fora, Juiz
de Fora, Minas Gerais CEP
36036-900, Brazil
| | - Rodrigo L. Fabri
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Juiz de Fora, Minas Gerais CEP 36036-900, Brazil
| |
Collapse
|
148
|
El-Banna AA, Eltamany EE, Yassen ASA, Lotfy A, El-Tanahy AHH, Badr JM, Algandaby MM, Murshid SS, Elhady SS, Abdelhameed RFA. Integrated Network Pharmacology, Molecular Modeling, LC-MS Profiling, and Semisynthetic Approach for the Roots of Rubia tinctorum L. Metabolites in Cancer Treatment. ACS OMEGA 2025; 10:13027-13045. [PMID: 40224436 PMCID: PMC11983213 DOI: 10.1021/acsomega.4c09853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025]
Abstract
Rubia tinctorum L. is one of the most widely used plants in folk medicine, with many reported pharmacological activities. One of these valuable activities is its anticancer efficacy. The aim of this study is to explore the multilevel mechanisms of R. tinctorum metabolites in cancer treatment using network pharmacology, together with molecular docking and in vitro studies. The network pharmacology analysis enabled us to reveal the hit anticancer R. tinctorum constituents, which were found to be acacetin, alizarin, anthragallol, 2-hydroxyanthraquinone, and xanthopurpurin. The most enriched cancer-linked target genes were PLCG1, BCL2, CYP1B1, NSD2, and ESR2. The pathways that were mostly involved in the anticancer mechanism of R. tinctorum metabolites were found to be metabolic pathways as well as pathways in cancer and apoptosis. Molecular docking of the identified hit anticancer constituents on the active sites of the most enriched genes unveiled that acacetin and alizarin possessed the lowest binding energies on the active sites of NSD2 and BCL2, respectively. While anthragallol showed the most stabilized interaction on the active sites of PLCG1, CYP1B1, and ESR2. Consequently, R. tinctorum extracts were evaluated for their in vitro cytotoxicity on a panel of cancerous cells. Among the tested R. tinctorum extracts, the chloroform extract was the strongest one with an IC50 = 3.987 μg/mL on the MCF-7 breast cancer cell line. Consequently, it was subjected to chromatographic separation and purification to isolate its major components with reported anticancer activity (scopoletin, rubiadin, chrysophanic acid, alizarin, purpurin, nor-damnacanthal, emodin, and rutin). Alizarin and purpurin constituted the main anthraquinones in R. tinctorum . Thus, they were quantified using LC/MS analysis. Moreover, a semisynthetic approach of alizarin toward the enhancement of its anticancer effect on the tested cancer cells was attained. Among the synthesized compounds, 2-methyl alizarin was the most active one with an IC50 = 8.878 μg/mL against the HepG2 cell line. This study provides deep insights into the anticancer mechanisms of R. tinctorum metabolites for the first time using network pharmacology and valorizes their significance as valuable anticancer agents.
Collapse
Affiliation(s)
- Alaa A. El-Banna
- Department
of Pharmacognosy, Faculty of Pharmacy, Alexandria
University, Alexandria 21521, Egypt
- Department
of Pharmacognosy, College of Pharmacy, Najran
University, Najran 66454, Saudi Arabia
| | - Enas E. Eltamany
- Department
of Pharmacognosy, Faculty of Pharmacy, Suez
Canal University, Ismailia 41522, Egypt
| | - Asmaa S. A. Yassen
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt
- Department
of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Ahmed Lotfy
- Egyptian
Liver Research Institute and Hospital (ELRIAH), Mansoura 35111, Egypt
- Department
of Surgery, Medical University of South
Carolina, Charleston, South Carolina 29425, United States
| | - Aya H. H. El-Tanahy
- Department
of Pharmacognosy, Faculty of Pharmacy, Delta
University for Science and Technology, Gamasa 7730103, Egypt
| | - Jihan M. Badr
- Department
of Pharmacognosy, Faculty of Pharmacy, Suez
Canal University, Ismailia 41522, Egypt
| | - Mardi M. Algandaby
- Department
of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Samar S. Murshid
- Department
of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sameh S. Elhady
- Department
of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Reda F. A. Abdelhameed
- Department
of Pharmacognosy, Faculty of Pharmacy, Suez
Canal University, Ismailia 41522, Egypt
- Department of Pharmacognosy, Faculty of
Pharmacy, Galala University, New Galala 43713, Egypt
| |
Collapse
|
149
|
Kaczmarczyk O, Augustyniak D, Żak A. Imaging of Hydrated and Living Cells in Transmission Electron Microscope: Summary, Challenges, and Perspectives. ACS NANO 2025; 19:12710-12733. [PMID: 40156542 PMCID: PMC11984313 DOI: 10.1021/acsnano.5c00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025]
Abstract
Transmission electron microscopy (TEM) is well-known for performing in situ studies in the nanoscale. Hence, scientists took this opportunity to explore the subtle processes occurring in living organisms. Nevertheless, such observations are complex─they require delicate samples kept in the liquid phase, low electron dose, and proper cell viability verification methods. Despite being highly demanding, so-called "live-cell" experiments have seen some degree of success. The presented review consists of an exhaustive literature review on reported "live-cell" studies and associated subjects, including liquid phase imaging, electron radiation interactions with liquids, and methods for cell viability testing. The challenges of modern, reliable research on living organisms are widely explained and discussed, and future perspectives for developing these techniques are presented.
Collapse
Affiliation(s)
- Olga Kaczmarczyk
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, 50-370 Wroclaw, Poland
| | - Daria Augustyniak
- Department
of Pathogen Biology and Immunology, Faculty of Biological Sciences, University of Wroclaw, 51-148 Wroclaw, Poland
| | - Andrzej Żak
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, 50-370 Wroclaw, Poland
- Department
of Material Science and Engineering, Massachusetts
Institute of Science and Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
150
|
Angeli LRADE, Santos GBD, Ferreira JRM, Serafim BLC, Lima TZ, Lima LGCADE, Bueno DF, Guarniero R. LOCAL INJECTION OF HUMAN DENTAL PULP STEM CELLS FOR TREATMENT OF JUVENILE AVASCULAR NECROSIS OF THE FEMORAL HEAD: PRELIMINARY RESULTS IN IMMATURE PIGS. ACTA ORTOPEDICA BRASILEIRA 2025; 33:e283445. [PMID: 40206451 PMCID: PMC11978309 DOI: 10.1590/1413-785220243201e283445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/23/2024] [Indexed: 04/11/2025]
Abstract
Introduction Legg-Calvé-Perthes disease is a major cause of hip joint deformities in children. Currently, experimental research is directed at investigating biological therapies, including the use of human dental pulp stem cells (hDPSC), which have not yet been studied for this purpose in swine models. This study aimed to evaluate whether local injection of hDPSC induces bone mineralization in the proximal femoral epiphysis in an experimental model of avascular necrosis of the femoral head in immature pigs. Methods Ten immature pigs underwent surgery to induce osteonecrosis of the proximal femoral epiphysis on the right side. In the intervention group (IG), hDPSC injections were performed immediately after osteonecrosis induction, and in the control group (CG), no additional procedure was performed. Left hips were used as controls. After 8 weeks, all animals were euthanized, and macroscopic, radiographic, and histological evaluations were performed. Results Bone mineralization was greater in the right hips of the IG compared to the CG (p = 0.0356), with an average mineralization index increase of 77.78% after hDPSC injection. Radiographic evaluation of the epiphyseal index showed a greater collapse in the right IG hips compared to the right CG hips (p < 0.001) and macroscopic evaluation showed a higher chance of the femoral head being flat (p = 0,049). Conclusion The injection of hDPSC into the proximal femoral epiphysis with induced osteonecrosis increases bone mineralization in immature pigs, but these treated hips show more deformity compared to the untreated hips. Level of Evidence IV, Case Series .
Collapse
Affiliation(s)
- Luiz Renato Agrizzi DE Angeli
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| | - Gustavo Bispo Dos Santos
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| | | | - Bárbara Lívia Corrêa Serafim
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| | | | | | | | - Roberto Guarniero
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| |
Collapse
|