1
|
Czeczot ADM, Muniz MN, Perelló MA, Silva ÉED, Timmers LFSM, Berger A, Gonzalez LC, Arraché Gonçalves G, Moura S, Machado P, Bizarro CV, Basso LA. Crystal structure of dihydroneopterin aldolase from Mycobacterium tuberculosis associated with 8-mercaptoguanine, and development of novel S8-functionalized analogues as inhibitors: Synthesis, enzyme inhibition, in vitro toxicity and antitubercular activity. J Enzyme Inhib Med Chem 2024; 39:2388207. [PMID: 39140692 PMCID: PMC11328599 DOI: 10.1080/14756366.2024.2388207] [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: 04/14/2024] [Revised: 07/10/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
The crystallographic structure of the FolB enzyme from Mycobacterium tuberculosis (MtFolB), complexed with its inhibitor 8-mercaptoguanine (8-MG), was elucidated at a resolution of 1.95 Å. A novel series of S8-functionalized 8-MG derivatives were synthesised and evaluated as in vitro inhibitors of dihydroneopterin aldolase (DHNA, EC 4.1.2.25) activity of MtFolB. These compounds exhibited IC50 values in the submicromolar range. Evaluation of the activity for five compounds indicated their inhibition mode and inhibition constants. Molecular docking analyses were performed to determine the enzyme-inhibitor intermolecular interactions and ligand conformations upon complex formation. The inhibitory activities of all compounds against the M. tuberculosis H37Rv strain were evaluated. Compound 3e exhibited a minimum inhibitory concentration in the micromolar range. Finally, Compound 3e showed no apparent toxicity in both HepG2 and Vero cells. The findings presented herein will advance the quest for novel, specific inhibitors targeting MtFolB, an attractive molecular target for TB drug development.
Collapse
Affiliation(s)
- Alexia de Matos Czeczot
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mauro Neves Muniz
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcia Alberton Perelló
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Éverton Edésio Dinis Silva
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Andresa Berger
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Laura Calle Gonzalez
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Arraché Gonçalves
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sidnei Moura
- Laboratório de Biotecnologia de Produtos Naturais e Sintéticos, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Rio Grande do Sul, Brazil
| | - Pablo Machado
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristiano Valim Bizarro
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiz Augusto Basso
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
2
|
Viana AR, Poleze TC, da S Bruckmann F, Bottari NB, Peroza LR, Rosales I, Zago NS, Schetinger MRC, Krause LMF, Rhoden CRB, Mortari SR. Liposome preparation of alpha-arbutin: stability and toxicity assessment using mouse B16F10 melanoma cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:879-894. [PMID: 39221705 DOI: 10.1080/15287394.2024.2393308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Melanoma is the most aggressive type of skin cancer, with few therapeutic alternatives following metastasis development. In recent years, drug delivery-associated nanotechnology has shown promising targeted results with diminished adverse effects compared to conventional treatments. This study aimed to (1) examine the effects of plant-derived α-arbutin, a natural compound and (2) compare these findings with bioactively developed liposomes containing α-arbutin utilizing the B16-F10 murine melanoma cell line as a model. Liposomes were obtained through reversed-phase evaporation by applying a spray dryer to assess their stability. The following biologic assays were measured cytotoxicity/antiproliferative (MTT, Neutral Red, and dsDNA PicoGreen). In addition, the levels of melanin and purinergic enzymes were also measured. The production of reactive oxygen species (ROS) and nitric oxide (NO) was determined as a measure of oxidative state. Treatment with nano-liposome containing alpha-arbutin induced a significant 68.4% cytotoxicity, similar to the positive control, in the B16-F10 murine melanoma cell line at 72 hr. Further, arbutin and liposomes containing alpha-arbutin increased levels of ROS and nitrite formation at 72 hr at the highest concentration (100 and 300 µg/ml) of treatments. Arbutin and liposomes containing alpha-arbutin reduced melanin levels at all tested concentrations. In addition, arbutin and alpha-arbutin containing liposomes lowered nucleotides (AMP, ADP, and ATP) and nucleoside (adenosine) levels in melanoma cells. Evidence suggests that α-arbutin containing liposome can be considered as an alternative immunosuppressive agent stimulated in melanoma treatment.
Collapse
Affiliation(s)
- Altevir R Viana
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
| | - Thatyana C Poleze
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
| | - Franciele da S Bruckmann
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
- Laboratory of Nanostructured Magnetic Materials - LAMMAN, Franciscan University, Santa Maria, RS, Brazil
| | - Nathieli B Bottari
- Postgraduate Program in Toxicological Biochemistry, Federal University of Santa Maria-RS, Santa Maria, Brazil
| | - Luis R Peroza
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
| | - Ingrid Rosales
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
| | - Natalia S Zago
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
| | - Maria R C Schetinger
- Postgraduate Program in Toxicological Biochemistry, Federal University of Santa Maria-RS, Santa Maria, Brazil
| | - Luciana M F Krause
- Department of Morphology, Federal University of Santa Maria-RS, Santa Maria, Brazil
| | - Cristiano R B Rhoden
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
- Laboratory of Nanostructured Magnetic Materials - LAMMAN, Franciscan University, Santa Maria, RS, Brazil
| | - Sergio R Mortari
- Postgraduate Program in Nanosciences, Franciscan University-UFN,Santa Maria, RS, Brazil
| |
Collapse
|
3
|
Jochum K, Miccoli A, Sommersdorf C, Poetz O, Braeuning A, Tralau T, Marx-Stoelting P. Comparative case study on NAMs: towards enhancing specific target organ toxicity analysis. Arch Toxicol 2024; 98:3641-3658. [PMID: 39207506 PMCID: PMC11489238 DOI: 10.1007/s00204-024-03839-7] [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: 07/03/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Traditional risk assessment methodologies in toxicology have relied upon animal testing, despite concerns regarding interspecies consistency, reproducibility, costs, and ethics. New Approach Methodologies (NAMs), including cell culture and multi-level omics analyses, hold promise by providing mechanistic information rather than assessing organ pathology. However, NAMs face limitations, like lacking a whole organism and restricted toxicokinetic interactions. This is an inherent challenge when it comes to the use of omics data from in vitro studies for the prediction of organ toxicity in vivo. One solution in this context are comparative in vitro-in vivo studies as they allow for a more detailed assessment of the transferability of the respective NAM data. Hence, hepatotoxic and nephrotoxic pesticide active substances were tested in human cell lines and the results subsequently related to the biology underlying established effects in vivo. To this end, substances were tested in HepaRG and RPTEC/tERT1 cells at non-cytotoxic concentrations and analyzed for effects on the transcriptome and parts of the proteome using quantitative real-time PCR arrays and multiplexed microsphere-based sandwich immunoassays, respectively. Transcriptomics data were analyzed using three bioinformatics tools. Where possible, in vitro endpoints were connected to in vivo observations. Targeted protein analysis revealed various affected pathways, with generally fewer effects present in RPTEC/tERT1. The strongest transcriptional impact was observed for Chlorotoluron in HepaRG cells (increased CYP1A1 and CYP1A2 expression). A comprehensive comparison of early cellular responses with data from in vivo studies revealed that transcriptomics outperformed targeted protein analysis, correctly predicting up to 50% of in vivo effects.
Collapse
Affiliation(s)
- Kristina Jochum
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Andrea Miccoli
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
- Institute for Marine Biological Resources and Biotechnology (IRBIM), National Research Council, Ancona, Italy
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | | | - Oliver Poetz
- Signatope GmbH, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Tewes Tralau
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Philip Marx-Stoelting
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany.
| |
Collapse
|
4
|
Hermans D, Maertens B, Verstringe S, Pasmans F, Vervloesem J, Jacquier V, Boyen F, Verbrugghe E. Research Note: Curbing Salmonella Enteritidis in broiler chickens with palm-free medium-chain fatty acids. Poult Sci 2024; 103:104172. [PMID: 39178817 PMCID: PMC11385747 DOI: 10.1016/j.psj.2024.104172] [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: 06/13/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024] Open
Abstract
Salmonellosis is still one of the most reported zoonoses worldwide and poultry meat is a major source, as chickens are often persistent carriers of Salmonella. Medium-chain fatty acids (MCFA) are known for their strong antimicrobial activity. MCFAs used today in the animal feed industry, however, mainly originate from the palm oil industry, which is notorious for its negative impact on the climate. We investigated the effect of a specific blend of palm-free MCFAs (ranging from C6 to C9) on Salmonella Enteritidis (SE) colonization in broiler chickens and in vitro SE characteristics. Fifty Ross 308 broiler chickens were randomly divided in 2 treatment groups. Chickens received either un-supplemented feed or feed supplemented with 300 ppm MCFAs from D0 onwards. On D7, all chickens were orally inoculated with 1600 CFU of SE. Cloacal swabs (D11) and samples of liver and caeca (D12) of all animals were collected and SE was enumerated. Percentage of SE-positive caecum samples was significantly (P = 0.044) reduced in birds receiving MCFAs compared to those receiving unsupplemented feed (36% vs. 64%). In vitro work performed with the same SE strain showed that preincubating the Salmonella bacteria with MCFAs at a sub-minimal inhibitory concentration significantly (p < 0.05) reduced bacterial adhesion to and invasion in Caco-2 cells, which may explain the observed reduction in intestinal SE colonization in the in vivo trial. Together, these results show that the tested eco-friendly MCFA blend could be a promising tool in the control of Salmonella in broilers.
Collapse
Affiliation(s)
- D Hermans
- Nutrition Sciences NV, 9031 Drongen, Belgium.
| | | | | | - F Pasmans
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | | | - V Jacquier
- Nutrition Sciences NV, 9031 Drongen, Belgium
| | - F Boyen
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - E Verbrugghe
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| |
Collapse
|
5
|
Correia AC, Costa I, Silva R, Sampaio P, Moreira JN, Sousa Lobo JM, Silva AC. Design of experiment (DoE) of mucoadhesive valproic acid-loaded nanostructured lipid carriers (NLC) for potential nose-to-brain application. Int J Pharm 2024; 664:124631. [PMID: 39182742 DOI: 10.1016/j.ijpharm.2024.124631] [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: 05/01/2024] [Revised: 07/30/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Epilepsy is a highly prevalent neurological disease and valproic acid (VPA) is used as a first-line chronic treatment. However, this drug has poor oral bioavailability, which requires the administration of high doses, resulting in adverse effects. Alternative routes of VPA administration have therefore been investigated, such as the nose-to-brain route, which allows the drug to be transported directly from the nasal cavity to the brain. Here, the use of nanostructured lipid carriers (NLC) to encapsulate drugs administered in the nasal cavity has proved advantageous. The aim of this work was to optimise a mucoadhesive formulation of VPA-loaded NLC for intranasal administration to improve the treatment of epilepsy. The Design of Experiment (DoE) was used to optimise the formulation, starting with component optimisation using Mixture Design (MD), followed by optimisation of the manufacturing process parameters using Central Composite Design (CCD). The optimised VPA-loaded NLC had a particle size of 76.1 ± 2.8 nm, a polydispersity index of 0.190 ± 0.027, a zeta potential of 28.1 ± 2.0 mV and an encapsulation efficiency of 85.4 ± 0.8%. The in vitro release study showed VPA release from the NLC of 50 % after 6 h and 100 % after 24 h. The in vitro biocompatibility experiments in various cell lines have shown that the optimised VPA-loaded NLC formulation is safe up to 75 µg/mL, in neuronal (SH-SY5Y), nasal (RPMI 2650) and hepatic (HepG2) cells. Finally, the interaction of the optimised VPA-loaded NLC formulation with nasal mucus was investigated and mucoadhesive properties were observed. The results of this study suggest that the use of intranasal VPA-loaded NLC may be a promising alternative to promote VPA targeting to the brain, thereby improving bioavailability and minimising adverse effects.
Collapse
Affiliation(s)
- A C Correia
- UCIBIO, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - I Costa
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal; UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - R Silva
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal; UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - P Sampaio
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4200-135, Portugal; IBMC-Instituto de Biologia Celular e Molecular, Porto 4200-135, Portugal
| | - J N Moreira
- CNC - Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine (Pólo I), University of Coimbra, Coimbra 3004-531, Portugal; Faculty of Pharmacy, Univ Coimbra - University of Coimbra, CIBB, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal
| | - J M Sousa Lobo
- UCIBIO, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - A C Silva
- UCIBIO, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal; Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal; FP-I3ID (Instituto de Investigação, Inovação e Desenvolvimento), FP-BHS (Biomedical and Health Sciences Research Unit), Faculty of Health Sciences, University Fernando Pessoa, Porto 4249 004, Portugal.
| |
Collapse
|
6
|
Hashem MS, Sobh RA, Fahim AM, Elsayed GH. Alginate sulfonamide hydrogel beads for 5-fluorouracil delivery: antitumor activity, cytotoxicity assessment, and theoretical investigation. Int J Biol Macromol 2024; 282:136573. [PMID: 39426771 DOI: 10.1016/j.ijbiomac.2024.136573] [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: 05/11/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
This study focused on grafting a new monomer (E)-N-(4-(3-(4-bromophenyl) acryloyl) phenyl)-4-methyl benzene sulfonamide (Br-PS) onto sodium alginate (Alg) using a free radical polymerization method. The optimal parameters for the grafting polymerization reaction were investigated, including initiator and monomer concentrations, polymerization reaction duration, and temperature. Additionally, the conversion, graft, and solid content percentages were calculated. The resulting novel poly (Br-PS)-g-Alg was thoroughly analyzed using Fourier-transform infrared spectroscopy (FT-IR), proton nuclear magnetic resonance (1H NMR), and scanning electron microscopy (SEM). Moreover, poly (Br-PS)-g-Alg was tested for cytotoxicity and selectivity values on lung cancer cell line (A549), breast cancer cell line (MDA-MB-231), and a normal cell line (MDCK) using the neutral red uptake test. Poly (Br-PS)-g-Alg demonstrated more inhibitory impact (IC50 = 33.37 and 40.9 μg/mL) and high selectivity (selectivity index = 4.83 and 3.94) on the A549 and MDA-MB-231 cell lines, respectively. Furthermore, uniform beads of creative poly (Br-PS)-g-Alg were fabricated, and their swelling rate in various media was studied. These beads could potentially serve as drug carriers for 5-fluorouracil (5-FU). Release experiments in simulated gastric (SGF) and intestinal fluids (SIF) showed a slower 5-FU release pattern in SGF compared to SIF. The proposed structures of poly (Br-PS)-g-Alg were theoretically verified using density functional theory with DFT/B3LYP/6-31(G) basis set, revealing distinct interactions due to the presence of different functional groups. The findings of this study could significantly impact the development of new drug delivery systems.
Collapse
Affiliation(s)
- M S Hashem
- Polymers and Pigments Department, National Research Centre (NRC), Dokki, Giza 12622, Egypt.
| | - Rokaya A Sobh
- Polymers and Pigments Department, National Research Centre (NRC), Dokki, Giza 12622, Egypt
| | - Asmaa M Fahim
- Department of Green Chemistry, National Research Centre (NRC), Dokki, Giza 12622, Egypt.
| | - Ghada H Elsayed
- Hormones Department, National Research Centre (NRC), Dokki, Giza 12622, Egypt; Stem Cells Lab, Centre of Excellence for Advanced Sciences, National Research Centre (NRC), Dokki, Giza 12622, Egypt
| |
Collapse
|
7
|
Zagórska-Dziok M, Mokrzyńska A, Ziemlewska A, Nizioł-Łukaszewska Z, Sowa I, Feldo M, Wójciak M. Assessment of the Antioxidant and Photoprotective Properties of Cornus mas L. Extracts on HDF, HaCaT and A375 Cells Exposed to UVA Radiation. Int J Mol Sci 2024; 25:10993. [PMID: 39456776 PMCID: PMC11507244 DOI: 10.3390/ijms252010993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
The influence of UV radiation on skin discoloration, skin aging and the development of skin cancer is widely known. As a part of this study, the effect of extracts from three varieties of Cornus mas L. (C. mas L.) on skin cells exposed to UVA radiation was assessed. The analyses were performed on both normal and cancer skin cells. For this purpose, the potential photoprotective effects of the obtained extracts (aqueous and ethanolic) was assessed by performing two cytotoxicity tests (Alamar blue and Neutral red). Additionally, the antioxidant capacity was compared using three different assays. The 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) probe was used to evaluate the intracellular level of free radicals in cells exposed to the simultaneous action of UVA radiation and dogwood extracts. Additionally, the ability to inhibit excessive pigmentation was determined by assessing the inhibition of melanin formation and tyrosinase activity. The obtained results confirmed the strong antioxidant properties of dogwood extracts and their photoprotective effect on normal skin cells. The ability to inhibit the viability of melanoma cells was also observed. Additionally, a reduction in oxidative stress in skin cells exposed to UVA radiation and a strong inhibition of melanin formation and tyrosinase activity have been demonstrated. This study shows that dogwood extract could be a valuable cosmetic raw material that can play both a photoprotective and antihyperpigmentation role in cosmetic preparations.
Collapse
Affiliation(s)
- Martyna Zagórska-Dziok
- Department of Technology of Cosmetic and Pharmaceutical Products, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (M.Z.-D.); (A.M.); (A.Z.); (Z.N.-Ł.)
| | - Agnieszka Mokrzyńska
- Department of Technology of Cosmetic and Pharmaceutical Products, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (M.Z.-D.); (A.M.); (A.Z.); (Z.N.-Ł.)
| | - Aleksandra Ziemlewska
- Department of Technology of Cosmetic and Pharmaceutical Products, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (M.Z.-D.); (A.M.); (A.Z.); (Z.N.-Ł.)
| | - Zofia Nizioł-Łukaszewska
- Department of Technology of Cosmetic and Pharmaceutical Products, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (M.Z.-D.); (A.M.); (A.Z.); (Z.N.-Ł.)
| | - Ireneusz Sowa
- Department of Analytical Chemistry, Medical University of Lublin, Aleje Raclawickie 1, 20-059 Lublin, Poland;
| | - Marcin Feldo
- Department of Vascular Surgery, Medical University of Lublin, Staszica 11 St., 20-081 Lublin, Poland;
| | - Magdalena Wójciak
- Department of Analytical Chemistry, Medical University of Lublin, Aleje Raclawickie 1, 20-059 Lublin, Poland;
| |
Collapse
|
8
|
Singh J, Hussain Y, Meena A, Sinha RA, Luqman S. Asiatic acid impedes NSCLC progression by inhibiting COX-2 and modulating PI3K signaling. FEBS Lett 2024. [PMID: 39394402 DOI: 10.1002/1873-3468.15027] [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: 04/30/2024] [Revised: 07/16/2024] [Accepted: 08/06/2024] [Indexed: 10/13/2024]
Abstract
Non-small cell lung cancer comprises up to 85% of lung cancer cases and has a poor prognosis. At present, there are still no effective treatments for this illness. Evidence suggests that the prostaglandin [cyclooxygenase-2 (COX-2)] and leukotriene [lipoxygenase-5 (5-LOX)] pathways are involved in lung cancer carcinogenesis. Therefore, novel agents that target COX-2 and 5-LOX may have therapeutic potential. In the present study, we examined the role of asiatic acid (AA), a triterpenoid saponin, in targeting the protein kinases responsible for lung cancer proliferation and mobility. The experimental data revealed that AA inhibited the growth of lung cancer cells (> 50%) and it significantly impeded the proliferation of lung cancer cells by inhibiting COX-2, which results in downregulation of the phosphotidyl inositol-3 kinase/protein kinase B/mammalian target of rapamycin signaling pathway, leading to an induction of cytotoxic autophagy-mediated apoptosis. Mechanistically, the expression of mitogen-activated protein kinase/extracellular signal-regulated kinase, hypoxia-inducible factor-1 and vascular endothelial growth factor is downregulated by AA, thereby reducing cell mobility and invasion. It also shows negative osmotic fragility on healthy human erythrocytes. It is concluded that AA may be a viable therapeutic drug for non-small cell lung cancer treatment, which opens new opportunities for synthesizing analogues.
Collapse
Affiliation(s)
- Jyoti Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Yusuf Hussain
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
9
|
Kasprzak D, Gaweł-Bęben K, Kukula-Koch W, Strzępek-Gomółka M, Wawruszak A, Woźniak S, Chrzanowska M, Czech K, Borzyszkowska-Bukowska J, Głowniak K, Matosiuk D, Orihuela-Campos RC, Jodłowska-Jędrych B, Laskowski T, Meissner HO. Lepidium peruvianum as a Source of Compounds with Anticancer and Cosmetic Applications. Int J Mol Sci 2024; 25:10816. [PMID: 39409148 PMCID: PMC11476809 DOI: 10.3390/ijms251910816] [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: 09/05/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Lepidium peruvianum-an edible herbaceous biennial plant distributed in the Andes-has been used for centuries as food and as a natural medicine in treating hormonal disorders, as an antidepressant, and as an anti-osteoporotic agent. The presented study aims to prove its beneficial cosmetic and chemopreventive properties by testing the antiradical, whitening, cytotoxic, and anticancer properties of differently colored phenotypes that were extracted using three solvents: methanol, water, and chloroform, with the help of the chemometric approach to provide evidence on the impact of single glucosinolanes (seven identified compounds in the HPLC-ESI-QTOF-MS/MS analysis) on the biological activity of the total extracts. The tested extracts exhibited moderate antiradical activity, with the methanolic extract from yellow and grey maca phenotypes scavenging 49.9 ± 8.96% and 48.8% ± 0.44% of DPPH radical solution at a concentration of 1 mg/mL, respectively. Grey maca was the most active tyrosinase inhibitor, with 72.86 ± 3.42% of the enzyme activity calculated for the water extract and 75.66 ± 6.21% for the chloroform extract. The studies in cells showed no cytotoxicity towards the human keratinocyte line HaCaT in all studied extracts and a marked inhibition of cell viability towards the G361 melanoma cell line, which the presence of pent-4-enylglucosinolate, glucotropaeolin, and glucoalyssin in the samples could have caused. Given all biological activity tests combined, the three mentioned compounds were shown to be the most significant positive contributors to the results obtained, and the grey maca water extract was found to be the best source of the former compound among the tested samples.
Collapse
Affiliation(s)
- Dorota Kasprzak
- Department of Cosmetology, Faculty of Health Sciences, Wincenty Pol Academy of Applied Sciences in Lublin, Choiny 2 Street, 20-816 Lublin, Poland;
| | - Katarzyna Gaweł-Bęben
- Department of Cosmetology, The University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (K.G.-B.); (M.S.-G.); (K.C.); (K.G.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland
| | - Marcelina Strzępek-Gomółka
- Department of Cosmetology, The University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (K.G.-B.); (M.S.-G.); (K.C.); (K.G.)
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland
| | - Sylwia Woźniak
- Chair and Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, 4a Chodzki Str., 20-93 Lublin, Poland; (S.W.); (D.M.)
| | - Marcelina Chrzanowska
- Department of Pharmaceutical Technology and Biochemistry and BioTechMed Centre, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12 St., 80-233 Gdańsk, Poland; (M.C.); (J.B.-B.); (T.L.)
| | - Karolina Czech
- Department of Cosmetology, The University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (K.G.-B.); (M.S.-G.); (K.C.); (K.G.)
| | - Julia Borzyszkowska-Bukowska
- Department of Pharmaceutical Technology and Biochemistry and BioTechMed Centre, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12 St., 80-233 Gdańsk, Poland; (M.C.); (J.B.-B.); (T.L.)
| | - Kazimierz Głowniak
- Department of Cosmetology, The University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (K.G.-B.); (M.S.-G.); (K.C.); (K.G.)
| | - Dariusz Matosiuk
- Chair and Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, 4a Chodzki Str., 20-93 Lublin, Poland; (S.W.); (D.M.)
| | - Rita Cristina Orihuela-Campos
- Academic Department of Stomatology for Children and Adolescents, Integrated Faculties of Medicine, Stomatology and Nursing, Cayetano Heredia Peruvian University, Av. Honorio Delgado 430, Lima 15102, Peru;
| | - Barbara Jodłowska-Jędrych
- Department of Histology and Embryology, Medical University of Lublin, 11 Radziwiłłowska Str., 20-093 Lublin, Poland;
| | - Tomasz Laskowski
- Department of Pharmaceutical Technology and Biochemistry and BioTechMed Centre, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12 St., 80-233 Gdańsk, Poland; (M.C.); (J.B.-B.); (T.L.)
| | - Henry O. Meissner
- Therapeutic Research, TTD International Pty Ltd., 39 Leopard Ave., Elanora, Gold Coast, QLD 4221, Australia;
| |
Collapse
|
10
|
Mânica da Cruz IB, Chelotti ME, Turra BO, Cardoso de Afonso Bonotto N, Pulcinelli DF, Kerkhoff Escher AL, Klein C, de Azevedo Mello P, Bitencourt GR, Barbisan F. Achyrocline satureioides infusion, popularly prepared and consumed, has an in vitro protective effect on human neural cells exposed to rotenone. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118350. [PMID: 38763375 DOI: 10.1016/j.jep.2024.118350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional harvest of Achyrocline satureioides (AS) occurs at dawn on Good Friday in some South American countries. Inflorescences are traditionally used as infusions for several disorders, including neuropsychiatric disorders. Pillows and cushions are popularly filled with AS to attenuate the symptoms of depression, anxiety, and sleep disturbances. However, evidence for the potential beneficial effects of AS on human neural cells remains unclear. AIM OF THE STUDY An in vitro model of SH-SY5Y human neural cells was applied to evaluate the effect of AS infusion, prepared as commonly used, on cells exposed to rotenone and to investigate its potential for neuropsychiatric disorders. MATERIALS AND METHODS A hot aqueous extract was obtained from a traditionally prepared AS inflorescence infusion and chemically characterized by high-resolution mass spectrometry and spectrophotometric quantification of total polyphenols, tannins, and flavonoids. The SH-SY5Y cell cultures were treated with AS extract at concentrations of 1, 3, 5, 10, 50, 100, and 300 μL/mL to determine the potential cyto- and genotoxic effects of AS on neural cells using MTT, Neutral Red, and GEMO assays. Apoptosis modulation was assessed using flow cytometry and apoptosis-modulating genes were evaluated by qRT-PCR. The protective effect of AS on the neurotoxicity triggered by rotenone exposure (30 nM) was determined by analyzing cellular viability and oxidative markers such as lipid peroxidation and protein carbonylation, and DNA damage was assessed by micronucleus assay. RESULTS The AS extract, as traditionally prepared, had estimated concentrations of 409.973 ± 31.107 μg/mL, 0.1041 ± 0.0246 mg GAE/mL, and 63.309 ± 3.178 mg QE/mL of total tannins, total polyphenols, and flavonoids, respectively. At concentrations of 30 and 100 μl/mL, AS decreased apoptotic events, whereas the highest concentration (300 μl/mL) increased apoptosis compared to that in the control (p < 0.05). In cells exposed to rotenone, AS treatment induced cell proliferation, reduced DNA damage (as evaluated by micronuclei), and reduced lipid and protein oxidation. CONCLUSIONS The data indicate the non-cytotoxic and beneficial effects of AS extract on human neural cells by reducing cellular mortality and oxidative stress in neural cells triggered by rotenone exposure.
Collapse
Affiliation(s)
- Ivana Beatrice Mânica da Cruz
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Gerontologia, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Maria Eduarda Chelotti
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Barbara Osmarin Turra
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Nathália Cardoso de Afonso Bonotto
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Débora Felipetto Pulcinelli
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Ana Laura Kerkhoff Escher
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Caroline Klein
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Paola de Azevedo Mello
- Departamento de Química, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Química, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Gustavo Rossato Bitencourt
- Programa de Pós-Graduação em Química, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Fernanda Barbisan
- Laboratório Biogenômica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Gerontologia, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
11
|
Doan-Nguyen TP, Aunkham A, Preedanorawut P, Chanpong T, Schulte A, Crespy D, Suginta W. Nanodots functionalized with chitooligosaccharides for blocking chitoporins. Colloids Surf B Biointerfaces 2024; 245:114284. [PMID: 39395215 DOI: 10.1016/j.colsurfb.2024.114284] [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: 08/29/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
We report the synthesis of functionalized nanodots as potential powerful blockers of solute transport through a chitoporin. Ultrasmall silica nanocapsules with a diameter of ∼ 6 nm were coated with chitooligosaccharides to be used as a "lid" binding to the opening of the chitoporin VhChiP of Vibrio campbellii. Efficient blocking is attributed to the adequate size of the nanodots and their functionalization with oligochitosan, which has strong affinity towards the Vibrio chitoporin. This strategy paves the way towards the development of nanomaterials for blocking other porins.
Collapse
Affiliation(s)
- Thao P Doan-Nguyen
- Department of Materials Science and Engineering, School of Molecular Science and Engineering (MSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Anuwat Aunkham
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Patitta Preedanorawut
- Department of Materials Science and Engineering, School of Molecular Science and Engineering (MSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Thanida Chanpong
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Albert Schulte
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Daniel Crespy
- Department of Materials Science and Engineering, School of Molecular Science and Engineering (MSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand.
| | - Wipa Suginta
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand.
| |
Collapse
|
12
|
Kowalczyk K, Błauż A, Krawczyk K, Rychlik B, Plażuk D. Design and synthesis of ferrocenyl 1,4-dihydropyridines and their evaluation as kinesin-5 inhibitors. Dalton Trans 2024; 53:16038-16053. [PMID: 39291736 DOI: 10.1039/d4dt01853b] [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: 09/19/2024]
Abstract
Kinesin-5 inhibitors offer cancer cell-targeted approach, thus securing reduced systemic toxicity compared to other antimitotic agents. By modifying the 1,4-dihydropyridine-based kinesin-5 inhibitor CPUYL064 with a ferrocenyl moiety (Fc), we designed and prepared a series of organometallic hybrids that show high antiproliferative activity, with the best compounds exhibiting up to 19-fold increased activity. This enhanced activity can be attributed to the presence of the ferrocenyl moiety.
Collapse
Affiliation(s)
- Karolina Kowalczyk
- Laboratory of Molecular Spectroscopy, Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, ul. Tamka 12, 91-403 Łódź, Poland.
| | - Andrzej Błauż
- Cytometry Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Krzysztof Krawczyk
- Cytometry Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Błażej Rychlik
- Cytometry Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Damian Plażuk
- Laboratory of Molecular Spectroscopy, Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, ul. Tamka 12, 91-403 Łódź, Poland.
| |
Collapse
|
13
|
Wang Q, Farhadipour M, Thijs T, Ruilova Sosoranga E, Van der Schueren B, Ceulemans LJ, Deleus E, Lannoo M, Tack J, Depoortere I. Bitter-tasting drugs tune GDF15 and GLP-1 expression via bitter taste or motilin receptors in the intestine of patients with obesity. Mol Metab 2024; 88:102002. [PMID: 39111389 PMCID: PMC11380393 DOI: 10.1016/j.molmet.2024.102002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
OBJECTIVE Growth differentiation factor 15 (GDF15), a stress related cytokine, was recently identified as a novel satiety signal acting via the GFRAL receptor located in the hindbrain. Bitter compounds are known to induce satiety via the release of glucagon-like peptide 1 (GLP-1) through activation of bitter taste receptors (TAS2Rs, 25 subtypes) on enteroendocrine cells in the gut. This study aimed to investigate whether and how bitter compounds induce a stress response in intestinal epithelial cells to affect GDF15 expression in patients with obesity, thereby facilitating satiety signaling from the gut. METHODS The acute effect of oral intake of the bitter-containing medication Plaquenil (hydroxychloroquine sulfate) on plasma GDF15 levels was evaluated in a placebo-controlled, double-blind, randomized, two-visit crossover study in healthy volunteers. Primary crypts isolated from the jejunal mucosa from patients with obesity were stimulated with vehicle or bitter compounds, and the effect on GDF15 expression was evaluated using RT-qPCR or ELISA. Immunofluorescence colocalization studies were performed between GDF15, epithelial cell type markers and TAS2Rs. The role of TAS2Rs was tested by 1) pretreatment with a TAS2R antagonist, GIV3727; 2) determining TAS2R4/43 polymorphisms that affect taste sensitivity to TAS2R4/43 agonists. RESULTS Acute intake of hydroxychloroquine sulfate increased GDF15 plasma levels, which correlated with reduced hunger scores and plasma ghrelin levels in healthy volunteers. This effect was mimicked in primary jejunal cultures from patients with obesity. GDF15 was expressed in enteroendocrine and goblet cells with higher expression levels in patients with obesity. Various bitter-tasting compounds (medicinal, plant extracts, bacterial) either increased or decreased GDF15 expression, with some also affecting GLP-1. The effect was mediated by specific intestinal TAS2R subtypes and the unfolded protein response pathway. The bitter-induced effect on GDF15/GLP-1 expression was influenced by the existence of TAS2R4 amino acid polymorphisms and TAS2R43 deletion polymorphisms that may predict patient's therapeutic responsiveness. However, the effect of the bitter-tasting antibiotic azithromycin on GDF15 release was mediated via the motilin receptor, possibly explaining some of its aversive side effects. CONCLUSIONS Bitter chemosensory and pharmacological receptors regulate the release of GDF15 from human gut epithelial cells and represent potential targets for modulating metabolic disorders or cachexia.
Collapse
Affiliation(s)
- Qian Wang
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Mona Farhadipour
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Theo Thijs
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | | | - Bart Van der Schueren
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical and Experimental Endocrinology, University of Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Leuven Intestinal Failure and Transplantation (LIFT) Center, University Hospitals Leuven, Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Matthias Lannoo
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.
| |
Collapse
|
14
|
Gege C, Hahn F, Wangen C, Häge S, Herrmann A, Uhlig N, Eberlein V, Issmail L, Klopfleisch R, Grunwald T, Marschall M, Kohlhof H, Vitt D. Synthesis and Characterization of DHODH Inhibitors Based on the Vidofludimus Scaffold with Pronounced Anti-SARS-CoV-2 Activity. ChemMedChem 2024; 19:e202400292. [PMID: 38887198 DOI: 10.1002/cmdc.202400292] [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: 04/22/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024]
Abstract
New strategies for the rapid development of broad-spectrum antiviral therapies are urgently required for emerging and re-emerging viruses like the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Host-directed antivirals that target universal cellular metabolic pathways necessary for viral replication present a promising approach with broad-spectrum activity and low potential for development of viral resistance. Dihydroorotate dehydrogenase (DHODH) was identified as one of those universal host factors essential for the replication of many clinically relevant human pathogenic viruses. DHODH is the rate-limiting enzyme catalyzing the fourth step in the de novo pyrimidine synthesis. Therefore, it is also developed as a therapeutic target for many diseases relying on cellular pyrimidine resources, such as cancer, autoimmune diseases and viral or bacterial infection. Thus, several DHODH inhibitors, including vidofludimus calcium (VidoCa, IMU-838), are currently in development or have been investigated in clinical trials for the treatment of virus infections such as SARS-CoV-2-mediated coronavirus disease 19 (COVID-19). Here, we report the medicinal chemistry optimization of VidoCa that resulted in metabolically more stable derivatives with improved DHODH target inhibition in various mammalian species, which translated into improved efficacy against SARS-CoV-2.
Collapse
Affiliation(s)
- Christian Gege
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Sigrun Häge
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Nadja Uhlig
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Valentina Eberlein
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Leila Issmail
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163, Berlin, Germany
| | - Thomas Grunwald
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Hella Kohlhof
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| | - Daniel Vitt
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| |
Collapse
|
15
|
Iosageanu A, Stefan LM, Craciunescu O, Cimpean A. Anti-Inflammatory and Wound Healing Properties of Different Honey Varieties from Romania and Correlations to Their Composition. Life (Basel) 2024; 14:1187. [PMID: 39337969 PMCID: PMC11432766 DOI: 10.3390/life14091187] [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: 08/08/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
The complex composition of honey plays a crucial role in wound healing, exhibiting varying effects at different stages of the healing process. This study investigated seven honey varieties sourced from different regions of Romania using in vitro experimental models developed in macrophage-like, fibroblast, and keratinocyte cell lines to explore the mechanisms by which honey promoted the healing process. This study assessed the impact of honey on inflammatory cytokine production in macrophage-like cells, cell proliferation and collagen synthesis in fibroblasts, and cell proliferation and migration in keratinocytes. Additionally, correlation analysis was conducted to examine the relationship between honey composition and its biological properties. Honey varieties presented both anti- and pro-inflammatory effects. Moreover, they displayed dose-dependent pro-proliferative effects, stimulating collagen synthesis and cell migration, thereby enhancing the re-epithelialization process. The Pearson coefficient analysis indicated a strong positive correlation between biological activities and phenolic content. Additionally, there was a medium positive correlation with the ascorbic acid content and a medium negative correlation with the glucose content in the different honey varieties. Romanian honey varieties rich in phenolics showed potential in modulating inflammation, proliferation, collagen synthesis, and cell migration, suggesting their suitability for further evaluation and development of innovative dressings for skin tissue regeneration.
Collapse
Affiliation(s)
- Andreea Iosageanu
- Faculty of Biology, University of Bucharest, 91-95, Splaiul Independentei, 050095 Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, 060031 Bucharest, Romania
| | - Laura Mihaela Stefan
- National Institute of Research and Development for Biological Sciences, 060031 Bucharest, Romania
| | - Oana Craciunescu
- National Institute of Research and Development for Biological Sciences, 060031 Bucharest, Romania
| | - Anisoara Cimpean
- Faculty of Biology, University of Bucharest, 91-95, Splaiul Independentei, 050095 Bucharest, Romania
| |
Collapse
|
16
|
Herrmann A, Gege C, Wangen C, Wagner S, Kögler M, Cordsmeier A, Irrgang P, Ip WH, Weil T, Hunszinger V, Groß R, Heinen N, Pfaender S, Reuter S, Klopfleisch R, Uhlig N, Eberlein V, Issmail L, Grunwald T, Hietel B, Cynis H, Münch J, Sparrer KMJ, Ensser A, Tenbusch M, Dobner T, Vitt D, Kohlhof H, Hahn F. Orally bioavailable RORγ/DHODH dual host-targeting small molecules with broad-spectrum antiviral activity. Antiviral Res 2024; 231:106008. [PMID: 39306285 DOI: 10.1016/j.antiviral.2024.106008] [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] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
Host-directed antivirals (HDAs) represent an attractive treatment option and a strategy for pandemic preparedness, especially due to their potential broad-spectrum antiviral activity and high barrier to resistance development. Particularly, dual-targeting HDAs offer a promising approach for antiviral therapy by simultaneously disrupting multiple pathways essential for viral replication. Izumerogant (IMU-935) targets two host proteins, (i) the retinoic acid receptor-related orphan receptor γ isoform 1 (RORγ1), which modulates cellular cholesterol metabolism, and (ii) the enzyme dihydroorotate dehydrogenase (DHODH), which is involved in de novo pyrimidine synthesis. Here, we synthesized optimized derivatives of izumerogant and characterized their antiviral activity in comparison to a recently described structurally distinct RORγ/DHODH dual inhibitor. Cell culture-based infection models for enveloped and non-enveloped DNA and RNA viruses, as well as a retrovirus, demonstrated high potency and broad-spectrum activity against human viral pathogens for RORγ/DHODH dual inhibitors at nanomolar concentrations. Comparative analyses with equipotent single-target inhibitors in metabolite supplementation approaches revealed that the dual-targeting mode represents the mechanistic basis for the potent antiviral activity. For SARS-CoV-2, an optimized dual inhibitor completely blocked viral replication in human airway epithelial cells at 5 nM and displayed a synergistic drug interaction with the nucleoside analog molnupiravir. In a SARS-CoV-2 mouse model, treatment with a dual inhibitor alone, or in combination with molnupiravir, reduced the viral load by 7- and 58-fold, respectively. Considering the clinical safety, oral bioavailability, and tolerability of izumerogant in a recent Phase I study, izumerogant-like drugs represent potent dual-targeting antiviral HDAs with pronounced broad-spectrum activity for further clinical development.
Collapse
Affiliation(s)
| | | | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Sabrina Wagner
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Melanie Kögler
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Arne Cordsmeier
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Pascal Irrgang
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Wing-Hang Ip
- Leibniz Institute of Virology, Hamburg, Germany.
| | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Victoria Hunszinger
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Natalie Heinen
- Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, Germany.
| | - Stephanie Pfaender
- Leibniz Institute of Virology, Hamburg, Germany; Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, Germany; University of Luebeck, Department of Natural Sciences, Institute of Virology and Cell Biology, Lübeck, Germany.
| | - Sebastian Reuter
- University Hospital Essen - Ruhrlandklinik, Department of Pulmonary Medicine, Experimental Pneumology, Essen, Germany.
| | - Robert Klopfleisch
- Institute for Animal Pathology, Freie Universität Berlin, Berlin, Germany.
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Benjamin Hietel
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle, Germany.
| | - Holger Cynis
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle, Germany; Junior Research Group "Immunomodulation in Pathophysiological Processes", Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | | | - Armin Ensser
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Matthias Tenbusch
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
17
|
Piechowicz L, Kosznik-Kwaśnicka K, Jarzembowski T, Daca A, Necel A, Bonawenturczak A, Werbowy O, Stasiłojć M, Pałubicka A. Staphylococcus aureus Co-Infection in COVID-19 Patients: Virulence Genes and Their Influence on Respiratory Epithelial Cells in Light of Risk of Severe Secondary Infection. Int J Mol Sci 2024; 25:10050. [PMID: 39337536 PMCID: PMC11431965 DOI: 10.3390/ijms251810050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Pandemics from viral respiratory tract infections in the 20th and early 21st centuries were associated with high mortality, which was not always caused by a primary viral infection. It has been observed that severe course of infection, complications and mortality were often the result of co-infection with other pathogens, especially Staphylococcus aureus. During the COVID-19 pandemic, it was also noticed that patients infected with S. aureus had a significantly higher mortality rate (61.7%) compared to patients infected with SARS-CoV-2 alone. Our previous studies have shown that S. aureus strains isolated from patients with COVID-19 had a different protein profile than the strains in non-COVID-19 patients. Therefore, this study aims to analyze S. aureus strains isolated from COVID-19 patients in terms of their pathogenicity by analyzing their virulence genes, adhesion, cytotoxicity and penetration to the human pulmonary epithelial cell line A549. We have observed that half of the tested S. aureus strains isolated from patients with COVID-19 had a necrotizing effect on the A549 cells. The strains also showed greater variability in terms of their adhesion to the human cells than their non-COVID-19 counterparts.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Tomasz Jarzembowski
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Agnieszka Daca
- Department of Physiopathology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Ada Bonawenturczak
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland
| | - Anna Pałubicka
- Specialist Hospital in Koscierzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Koscierzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland
| |
Collapse
|
18
|
Clara da Silva Durigon M, Renata Caitano Visnheski B, Braz Júnior O, Christina Thomas J, Fogagnoli Simas F, Piovan L. Polyfunctionalized organoselenides: New synthetic approach from selenium-containing cyanohydrins and anti-melanoma activity. Bioorg Med Chem Lett 2024; 110:129860. [PMID: 38942128 DOI: 10.1016/j.bmcl.2024.129860] [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: 03/09/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
A series of seleno-containing polyfunctionalized compounds was synthesized exploring cyanohydrin chemistry, including α-hydroxy esters, α-hydroxy acids, 1,2-diols, and 1,2-diacetates, with yields ranging from 26 up to 99 %. The cytotoxicity of all synthesized compounds was then evaluated using a non-tumor cell line (BALB/3T3 murine fibroblasts), and those deemed non-cytotoxic had their anti-melanoma activity evaluated using B16-F10 murine melanoma cells. These assays identified two compounds with selective cytotoxic activity against the tested melanoma cell line, showing a potential anti-melanoma application.
Collapse
|
19
|
Goldar S, Gachumi G, Siciliano SD, Hogan NS. The role of efflux transporters in cytotoxicity and intracellular concentration of chlorpyrifos and chlorpyrifos oxon in human cell lines. Toxicol In Vitro 2024; 101:105942. [PMID: 39284535 DOI: 10.1016/j.tiv.2024.105942] [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: 12/19/2023] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024]
Abstract
In this study, we investigated the role of two efflux transporters, p-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), in the cytotoxicity and intracellular accumulation of the organophosphate pesticide chlorpyrifos (CPF) and its active metabolite, CPF-oxon (CPFO), in a human-derived liver cell line (HepG2) and kidney epithelial cell line (HK-2). The cytotoxicity to CPF and CPFO differed between cell lines where HK-2 had lower IC50 values which could be attributed to lower basal expression and inducibility of metabolizing enzymes, transporters, and nuclear receptors in HK-2 cells. In HepG2 cells, co-exposure of CPF with a specific inhibitor of either P-gp or BCRP enhanced the cytotoxicity of CPF while co-exposure of CPFO with VRP enhanced the cytotoxicity of CPFO, suggesting the role of these transporters in the elimination CPF and CPFO. Inhibition of efflux transporters did not affect the cytotoxicity of CPF and CPFO in HK-2 cells. Co-incubation of CPF with P-gp and BCRP inhibitors increased the intracellular concentration of CPF in HepG2 cells suggesting that both transporters play a role in limiting the cellular accumulation of CPF in HepG2 cells. Our results provide evidence that inhibition of efflux transporters can enhance CPF-induced toxicity through enhanced cellular accumulation and raises additional questions regarding how pesticide-transporter interactions may influence toxicity of mixtures containing pesticides and other environmental chemicals.
Collapse
Affiliation(s)
- Samira Goldar
- Toxicology Graduate Program, Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
| | - George Gachumi
- Department of Soil Science, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - Steven D Siciliano
- Department of Soil Science, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada; Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
| | - Natacha S Hogan
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada; Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada.
| |
Collapse
|
20
|
Beisl J, Jochum K, Chen Y, Varga E, Marko D. Combinatory Effects of Acrylamide and Deoxynivalenol on In Vitro Cell Viability and Cytochrome P450 Enzymes of Human HepaRG Cells. Toxins (Basel) 2024; 16:389. [PMID: 39330847 PMCID: PMC11436166 DOI: 10.3390/toxins16090389] [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: 08/05/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
Acrylamide (AA) can be formed during the thermal processing of carbohydrate-rich foods. Deoxynivalenol (DON), a mycotoxin produced by Fusarium spp., contaminates many cereal-based products. In addition to potential co-exposure through a mixed diet, co-occurrence of AA and DON in thermally processed cereal-based products is also likely, posing the question of combinatory toxicological effects. In the present study, the effects of AA (0.001-3 mM) and DON (0.1-30 µM) on the cytotoxicity, gene transcription, and expression of major cytochrome P450 (CYP) enzymes were investigated in differentiated human hepatic HepaRG cells. In the chosen ratios of AA-DON (10:1; 100:1), cytotoxicity was clearly driven by DON and no overadditive effects were observed. Using quantitative real-time PCR, about twofold enhanced transcript levels of CYP1A1 were observed at low DON concentrations (0.3 and 1 µM), reflected by an increase in CYP1A activity in the EROD assay. In contrast, CYP2E1 and CYP3A4 gene transcription decreased in a concentration-dependent manner after incubation with DON (0.01-0.3 µM). Nevertheless, confocal microscopy showed comparably constant protein levels. The present study provided no indication of an induction of CYP2E1 as a critical step in AA bioactivation by co-occurrence with DON. Taken together, the combination of AA and DON showed no clear physiologically relevant interaction in HepaRG cells.
Collapse
Affiliation(s)
- Julia Beisl
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Kristina Jochum
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- German Federal Institute of Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Yi Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Elisabeth Varga
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Unit Food Hygiene and Technology, Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine, Veterinarplatz 1, 1210 Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
21
|
Ye G, Bu F, Pan R, Mendoza A, Saxena D, Zheng J, Perlman S, Liu B, Li F. Dual-role epitope on SARS-CoV-2 spike enhances and neutralizes viral entry across different variants. PLoS Pathog 2024; 20:e1012493. [PMID: 39236072 PMCID: PMC11407660 DOI: 10.1371/journal.ppat.1012493] [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: 04/10/2024] [Revised: 09/17/2024] [Accepted: 08/09/2024] [Indexed: 09/07/2024] Open
Abstract
Grasping the roles of epitopes in viral glycoproteins is essential for unraveling the structure and function of these proteins. Up to now, all identified epitopes have been found to either neutralize, have no effect on, or enhance viral entry into cells. Here, we used nanobodies (single-domain antibodies) as probes to investigate a unique epitope on the SARS-CoV-2 spike protein, located outside the protein's receptor-binding domain. Nanobody binding to this epitope enhances the cell entry of prototypic SARS-CoV-2, while neutralizing the cell entry of SARS-CoV-2 Omicron variant. Moreover, nanobody binding to this epitope promotes both receptor binding activity and post-attachment activity of prototypic spike, explaining the enhanced viral entry. The opposite occurs with Omicron spike, explaining the neutralized viral entry. This study reveals a unique epitope that can both enhance and neutralize viral entry across distinct viral variants, suggesting that epitopes may vary their roles depending on the viral context. Consequently, antibody therapies should be assessed across different viral variants to confirm their efficacy and safety.
Collapse
Affiliation(s)
- Gang Ye
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Fan Bu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ruangang Pan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Alise Mendoza
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Divyasha Saxena
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Jian Zheng
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, United States of America
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, United States of America
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Bin Liu
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
22
|
Ribeiro JLP, Costa I, Silva R, Lopes SMM, Saraiva L, Pinho E Melo TMVD. 3-Tetrazolyl-β-carboline derivatives as potential neuroprotective agents. Bioorg Med Chem 2024; 111:117841. [PMID: 39094526 DOI: 10.1016/j.bmc.2024.117841] [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: 04/12/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
3-Tetrazolyl-β-carbolines were prepared by the Pictet-Spengler approach using a tryptophan analogue as building block, in which the carboxylic acid was replaced by the bioisosteric tetrazole group. Knowing that β-carbolines are often associated with psychopharmacological effects, the study of the 3-tetrazolyl-β-carbolines as potential neuroprotective agents against Parkinson's disease was investigated. The evaluation of neuroprotective effects against 1-methyl-4-phenylpyridin-1-ium (MPP+)-induced cytotoxicity allowed to identify compounds with relevant neuroprotective activity. One derivative, 3-(1-benzyl-1H-tetrazol-5-yl)-1-(p-dimethylaminophenyl)-β-carboline, stood out for its low cytotoxicity and excellent performance, preventing cell death induced by this neurotoxin. The most promising compounds were also evaluated for their neuroprotective properties against iron (III)-induced cytotoxicity. However, only one 3-tetrazolyl-β-carboline derivative slightly reduced iron-induced cytotoxicity. Overall, the neuroprotective properties of 3-tetrazolyl-β-carbolines have been demonstrated and this finding may contribute to the development of new therapies for Parkinson's disease.
Collapse
Affiliation(s)
- João L P Ribeiro
- University of Coimbra, Coimbra Chemistry Centre-Institute of Molecular Sciences, and Department of Chemistry, 3004-535 Coimbra, Portugal
| | - Inês Costa
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - Renata Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - Susana M M Lopes
- University of Coimbra, Coimbra Chemistry Centre-Institute of Molecular Sciences, and Department of Chemistry, 3004-535 Coimbra, Portugal
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Teresa M V D Pinho E Melo
- University of Coimbra, Coimbra Chemistry Centre-Institute of Molecular Sciences, and Department of Chemistry, 3004-535 Coimbra, Portugal.
| |
Collapse
|
23
|
Raza W, Meena A, Luqman S. THF induces apoptosis by downregulating initiation, promotion, and progression phase biomarkers in skin and lung carcinoma. J Biochem Mol Toxicol 2024; 38:e23838. [PMID: 39243196 DOI: 10.1002/jbt.23838] [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: 05/06/2024] [Revised: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 09/09/2024]
Abstract
3,5,7-Trihydroxy-2-phenylchromen-4-one (THF) possesses a diverse range of pharmacological activities. Evidence suggests that THF exerts anticancer activity by distinct mechanisms of action. This study explores the anticancer potential of THF in human lung (A549) and skin (A431) cancer cells by employing different antiproliferative assays. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, neutral red uptake, sulphorhodamine B, and cell motility assays were used to confirm the anticancer potential of THF. Cell target-based and quantitative reverse transcription polymerase chain reaction (qRT-PCR) assays were used to explore the effect of THF on the initiation, promotion and progression phase biomarkers of carcinogenesis. THF suppresses the activity of lipoxygenase-5 up to ~40% in both A549 and A431 cells and up to ~50% hyaluronidase activity in A549 cells. qRT-PCR assay reveals that THF inhibits the activity of phosphatidyl inositol-3 kinase/protein kinase B/mammalian target of rapamycin in both cell lines, which is responsible for the initiation of cancer. It also arrests the G2/M phase of the cell cycle in A431 cells and increases the sub-diploid population in both A549 and A431 cell lines which leads to cell death. Annexin V-FITC assay confirmed that THF induces apoptosis and necrosis in A431 and A549 cell lines. Further investigation revealed that THF not only enhances reactive oxygen species production but also modulates mitochondrial membrane potential in both cell lines. It significantly inhibits S-180 tumour formation at 5 and 10 mg/kg bw, i.p. dose. An acute skin toxicity study on mice showed that erythema and edema scores are within the acceptable range, besides acceptable drug-likeness properties and non-toxic effects on human erythrocytes. Conclusively, THF showed potent anticancer activity on skin and lung carcinoma cell lines, suppressed the level of the biomarkers and inhibited tumour growth in mice.
Collapse
Affiliation(s)
- Waseem Raza
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
24
|
Ventura E, Gonçalves JM, Vilke JM, d'Errico G, Benedetti M, Regoli F, Bebianno MJ. Are mixtures of micro/nanoplastics more toxic than individual micro or nanoplastic contamination in the clam Ruditapes decussatus? MARINE POLLUTION BULLETIN 2024; 206:116697. [PMID: 39018822 DOI: 10.1016/j.marpolbul.2024.116697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/10/2024] [Accepted: 07/03/2024] [Indexed: 07/19/2024]
Abstract
The abundance of micro (MPs) and nano (NPs) sized plastic particles in the ocean is concerning due to their harmful effects on marine life. The interactions between MPs and NPs in the marine environment and their impact on marine biota remain not fully understood. This study contributes with new insights into the interaction between polystyrene NPs (PSNPs) and polyethylene MPs (PEMPs) on the clam Ruditapes decussatus. Results showed ingestion of MPs and NPs by clams, with PSNPs demonstrating higher toxicity in hemolymph. While no genotoxicity was observed, clams treated with MPs and the mixture showed increased acetylcolinesterase (AchE) activity over time. Additionally, the antioxidant defense system mitigated oxidative stress, suggesting effective neutralization of reactive oxygen species. Hazard assessment indicated the greatest impact on clam digestive glands after ten days of exposure, with an antagonistic interaction between MPs and NPs noted.
Collapse
Affiliation(s)
- Emma Ventura
- CIMA, Centre of Marine and Environmental Research\ARNET - Infrastructure Network in Aquatic Research, University of Algarve, Campus de Gambelas, 8000-139 Faro, Portugal; Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Joanna M Gonçalves
- CIMA, Centre of Marine and Environmental Research\ARNET - Infrastructure Network in Aquatic Research, University of Algarve, Campus de Gambelas, 8000-139 Faro, Portugal
| | - Juliano M Vilke
- CIMA, Centre of Marine and Environmental Research\ARNET - Infrastructure Network in Aquatic Research, University of Algarve, Campus de Gambelas, 8000-139 Faro, Portugal
| | - Giuseppe d'Errico
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Maura Benedetti
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy; National Future Biodiversity Centre (NFBC), Palermo, Italy
| | - Francesco Regoli
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy; National Future Biodiversity Centre (NFBC), Palermo, Italy
| | - Maria João Bebianno
- CIMA, Centre of Marine and Environmental Research\ARNET - Infrastructure Network in Aquatic Research, University of Algarve, Campus de Gambelas, 8000-139 Faro, Portugal.
| |
Collapse
|
25
|
Erdogan MK, Gundogdu R, Toy Y, Halil Gecibesler I, Yapar Y, Behcet L, Zengin G. Comparison of Anticancer, Antioxidant, Enzyme Inhibitory Effects and Phytochemical Contents Between Edible Lettuce (Lactuca sativa) and a New Wild Species (Lactuca anatolica). Chem Biodivers 2024; 21:e202400552. [PMID: 38958194 DOI: 10.1002/cbdv.202400552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024]
Abstract
In this study, the bioactive components, enzyme inhibitory, antioxidant and anticancer potentials of edible (L. sativa) and a new species (L. anatolica) of Lactuca were evaluated and compared. The quantitative analyzes of the bioactive components of L. sativa (LS) and L. anatolica (LA) were analyzed quantitatively by GC-MS and Orbitrab HPLC-HRMS. Antioxidant, enzyme inhibitory and anticancer properties were analyzed by various assays. In general, LA exhibited more stronger antioxidant properties compared to LS. The extracts showed similar inhibitory effects on these enzymes. It was determined that LS was dominant in terms of linoleic acid (23.71 %), while LA contained a high level of α-linolenic acid (31.70 %). LA and LS inhibited the viability of A549 and MCF-7 cells in a dose-dependent manner. IC50 values for LA, LS and cisplatin were determined as 120.3, 197.5, 4.3 μg/mL in A549 cell line and 286.2, 472.8, 7.2 μg/mL in MCF-7 cell line, respectively. It was revealed that LA and LS treatment at 50 μg/mL concentrations in A549 cells completely suppressed the colony forming capacity, and treatment with IC50 doses inhibited cell migration, and triggered apoptosis by regulating caspase-3, cPARP, p53 and p21. The findings of this study suggested that these species have significant pharmacological potential.
Collapse
Affiliation(s)
- Mehmet Kadir Erdogan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Ramazan Gundogdu
- Department of Pharmacy Services, Vocational School of Health Services, Bingol University, Bingol, Turkiye
- Current address: Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Yusuf Toy
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Ibrahim Halil Gecibesler
- Department of Occupational Health and Safety, Faculty of Health Science, Bingol University, Bingol, Turkiye
| | - Yakup Yapar
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Lutfi Behcet
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol, Turkiye
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkiye
| |
Collapse
|
26
|
Yang Y, Yao Z, Huo L. The Nf1-Q181X point mutation induces M2 macrophage polarization via the AKT/STAT pathway to promote smooth muscle cell proliferation and migration. Mol Biol Rep 2024; 51:946. [PMID: 39215899 DOI: 10.1007/s11033-024-09887-7] [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: 06/24/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Increased case reports have shown that patients with NF1 have an increased risk of extensive vascular vasculopathy. Previous studies demonstrated the presence of macrophages and smooth muscle cells in the neoplastic intima of carotid arteries after injury in Nf1+/- mice. However, whether NF1 gene mutations affect macrophage polarization and macrophage-smooth muscle cell interactions remains to be elucidated. METHODS Scratch assay and transwell assay were utilized to detect cell migration ability. The dye 2',7'dichlorofluorescin diacetate and neutral red stain were used to assess intracellular ROS production and cell phagocytosis function, respectively. Proteins and mRNA expression were determined by western blot, RT-qPCR, and immunofluorescence. Finally, the macrophage (MAC) and vascular smooth muscle cell (VSMC) co-culture system was used to detect cellular crosstalk. RESULTS Cell function assays confirmed that the Nf1-Q181X point mutation attenuated the phagocytosis of bone marrow-derived macrophages (BMDMs) and promoted the migration and ROS production of BMDMs. Moreover, we found that the Nf1-Q181X point mutation inhibited M1 but promoted M2 macrophage polarization by down-regulating p38, ERK, and JNK and up-regulating the Akt/STAT3 signaling pathway, respectively. Furthermore, in the MAC-VSMC co-culture system, we demonstrated that Nf1-Q181X point mutation-activated M2 BMDMs promoted proliferation and migration of VSMCs and induced the transformation of VSMCs from contractile phenotype to synthetic phenotype. CONCLUSION The findings suggest that the Nf1-Q181X point mutation can mediate macrophage polarization and promote smooth muscle cell proliferation and migration, providing clinical clues for the treatment of NF1-complicated vasculopathy.
Collapse
Affiliation(s)
- Yang Yang
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, 100038, China
| | - Zhichao Yao
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, 100038, China
| | - Lirong Huo
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, 100038, China.
| |
Collapse
|
27
|
Lebeau G, Paulo-Ramos A, Hoareau M, El Safadi D, Meilhac O, Krejbich-Trotot P, Roche M, Viranaicken W. Metabolic Dependency Shapes Bivalent Antiviral Response in Host Cells in Response to Poly:IC: The Role of Glutamine. Viruses 2024; 16:1391. [PMID: 39339867 PMCID: PMC11436187 DOI: 10.3390/v16091391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
The establishment of effective antiviral responses within host cells is intricately related to their metabolic status, shedding light on immunometabolism. In this study, we investigated the hypothesis that cellular reliance on glutamine metabolism contributes to the development of a potent antiviral response. We evaluated the antiviral response in the presence or absence of L-glutamine in the culture medium, revealing a bivalent response hinging on cellular metabolism. While certain interferon-stimulated genes (ISGs) exhibited higher expression in an oxidative phosphorylation (OXPHOS)-dependent manner, others were surprisingly upregulated in a glycolytic-dependent manner. This metabolic dichotomy was influenced in part by variations in interferon-β (IFN-β) expression. We initially demonstrated that the presence of L-glutamine induced an enhancement of OXPHOS in A549 cells. Furthermore, in cells either stimulated by poly:IC or infected with dengue virus and Zika virus, a marked increase in ISGs expression was observed in a dose-dependent manner with L-glutamine supplementation. Interestingly, our findings unveiled a metabolic dependency in the expression of specific ISGs. In particular, genes such as ISG54, ISG12 and ISG15 exhibited heightened expression in cells cultured with L-glutamine, corresponding to higher OXPHOS rates and IFN-β signaling. Conversely, the expression of viperin and 2'-5'-oligoadenylate synthetase 1 was inversely related to L-glutamine concentration, suggesting a glycolysis-dependent regulation, confirmed by inhibition experiments. This study highlights the intricate interplay between cellular metabolism, especially glutaminergic and glycolytic, and the establishment of the canonical antiviral response characterized by the expression of antiviral effectors, potentially paving the way for novel strategies to modulate antiviral responses through metabolic interventions.
Collapse
Affiliation(s)
- Grégorie Lebeau
- PIMIT—Processus Infectieux en Milieu Insulaire Tropical, INSERM UMR 1187, CNRS 9192, IRD 249, Plateforme CYROI, Université de La Réunion, 97490 Sainte-Clotilde, France
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, France
| | - Aurélie Paulo-Ramos
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, France
| | - Mathilde Hoareau
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, France
| | - Daed El Safadi
- PIMIT—Processus Infectieux en Milieu Insulaire Tropical, INSERM UMR 1187, CNRS 9192, IRD 249, Plateforme CYROI, Université de La Réunion, 97490 Sainte-Clotilde, France
| | - Olivier Meilhac
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, France
| | - Pascale Krejbich-Trotot
- PIMIT—Processus Infectieux en Milieu Insulaire Tropical, INSERM UMR 1187, CNRS 9192, IRD 249, Plateforme CYROI, Université de La Réunion, 97490 Sainte-Clotilde, France
| | - Marjolaine Roche
- PIMIT—Processus Infectieux en Milieu Insulaire Tropical, INSERM UMR 1187, CNRS 9192, IRD 249, Plateforme CYROI, Université de La Réunion, 97490 Sainte-Clotilde, France
| | - Wildriss Viranaicken
- PIMIT—Processus Infectieux en Milieu Insulaire Tropical, INSERM UMR 1187, CNRS 9192, IRD 249, Plateforme CYROI, Université de La Réunion, 97490 Sainte-Clotilde, France
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, France
| |
Collapse
|
28
|
de Almeida Roque A, Zablocki da Luz J, Filipak Neto F, Barjhoux I, Rioult D, de Oliveira Ribeiro CA. Low concentrations of complex mixtures of pesticides and metabolites are toxic to common Carp brain cells ( Cyprinus carpio carpio). Drug Chem Toxicol 2024:1-11. [PMID: 39210515 DOI: 10.1080/01480545.2024.2397432] [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: 06/07/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Pesticide use increases annually, and Brazil is the world's largest consumer. However, unlike the European Union (EU), there is no established limit value for pesticide mixtures in drinking water, and therefore the concentration of pesticides can reach 3354 times the EU limit. Thus, determining the risk of exposure to pesticide mixtures and their main metabolites is challenging and requires the use of alternative methods. In the present study, the Common Carp Brain (CCB) cell line was used to evaluate the in vitro toxicity of relevant pesticide mixtures (glyphosate, 2,4-D, atrazine, and mancozeb) and their main metabolites after 72 h of exposure. The tested concentrations were based on the Acceptable Daily Intake (ADI) defined by Brazilian legislation. The results showed that cells exposed to lower concentrations of the pesticide mixtures and the pesticide + metabolite mixtures were affected by a decrease in cell confluence, resazurin metabolism, and wound healing capacity. The IBR index showed that lower concentrations had more severe effects, suggesting the absence of safe concentrations of these pesticide and metabolite mixtures for the CCB cell line within the tested concentration range. These findings raise concerns about the effects of exposure to these substances on animal and human health.
Collapse
Affiliation(s)
- Aliciane de Almeida Roque
- Department of Cell Biology, Laboratory of Cell Toxicology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Jessica Zablocki da Luz
- Department of Cell Biology, Laboratory of Cell Toxicology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Francisco Filipak Neto
- Department of Cell Biology, Laboratory of Cell Toxicology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Iris Barjhoux
- UMR-I 02 INERIS-URCA-ULH SEBIO - Stress Environnementaux et BIOsurveillance des milieux aquatiques, Université de Reims Champagne-Ardenne, Reims, France
| | - Damien Rioult
- UMR-I 02 INERIS-URCA-ULH SEBIO - Stress Environnementaux et BIOsurveillance des milieux aquatiques, Université de Reims Champagne-Ardenne, Reims, France
- URCATech Plateau Technique Mobile de Cytométrie Environnementale URCATech- MOBICYTE, Université de Reims Champagne-Ardenne, Reims, France
| | | |
Collapse
|
29
|
Ali NA, Elsayed GH, Mohamed SH, Abd Elkarim AS, Aly MS, Elgamal AM, Elsayed WM, El-Newary SA. Chia Seed ( Salvia hispanica) Attenuates Chemically Induced Lung Carcinomas in Rats through Suppression of Proliferation and Angiogenesis. Pharmaceuticals (Basel) 2024; 17:1129. [PMID: 39338293 PMCID: PMC11435337 DOI: 10.3390/ph17091129] [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: 07/11/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 09/30/2024] Open
Abstract
In 2022, 2.5 million cases of lung cancer were diagnosed, resulting in 1.8 million deaths. These statistics have motivated us to introduce a new natural product which is feasible in lung cancer therapies. This comprehensive study was performed to study the effects of chia seed extracts (70% ethanol and petroleum ether) on lung cancer in vitro and in vivo models. The invitro cytotoxicity activity of the chia extracts was studied in lung cancer cell lines (A549 cells). After 48 h, chia alcohol and ether extracts showed more inhibitory influence (IC50, 16.08, and 14.8 µg/mL, respectively) on A549 cells compared to Dox (IC50, 13.6 µg/mL). In vivo, administration of chia alcohol and ether extracts (500 mg/kg/day, orally for 20 weeks) recovered 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung cancer, as a significant reduction in the lung cancer biomarkers, including the relative weight of the lung (20.0 and 13.33%), ICAM(31.73 and 15.66%), and c-MYC (80 and 96%) and MMP9(60 and 69%) expression genes, and improvement in these changes were observed by histopathological examinations of the lung tissues compared to the lung control. Chia seeds fought lung cancer via suppression of proliferation, angiogenesis, inflammation, and activation apoptosis. These activities may be attributed to the chemical composition of chia, which is identified by LC-Mass, such as caffeic acid, vanillic acid, kaempferol-3-O-glucuronide, and taxifolin. Finally, we can conclude that chia seeds have an anti-lung cancer effect with a good safety margin.
Collapse
Affiliation(s)
- Naglaa A. Ali
- Hormones Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt; (N.A.A.); (G.H.E.); (S.H.M.)
| | - Ghada H. Elsayed
- Hormones Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt; (N.A.A.); (G.H.E.); (S.H.M.)
- Stem Cells Lab, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza 12622, Egypt
| | - Safaa H. Mohamed
- Hormones Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt; (N.A.A.); (G.H.E.); (S.H.M.)
| | - Asmaa S. Abd Elkarim
- Chemistry of Tanning Materials and Leather Technology Department, National Research Centre, Giza 12622, Egypt;
| | - Mohamed S. Aly
- Department of Animal Reproduction and Artificial Insemination, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt;
| | - Abdelbaset M. Elgamal
- Department of Chemistry of Microbial and Natural Products, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt;
| | - Wael M. Elsayed
- Chemistry of Medicinal Plants Department, National Research Centre, Giza 12622, Egypt;
| | - Samah A. El-Newary
- Medicinal and Aromatic Plants Research Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt
| |
Collapse
|
30
|
Silva-Reis SC, Costa VM, da Silva DC, Pereira DM, Correia XC, García-Mera X, Rodríguez-Borges JE, Sampaio-Dias IE. Discovery of hybrid Glypromate conjugates with neuroprotective activity against paraquat-induced toxicity. RSC Med Chem 2024:d4md00584h. [PMID: 39290380 PMCID: PMC11403921 DOI: 10.1039/d4md00584h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
Neurodegenerative disorders comprise a series of heterogeneous conditions that affect millions of people worldwide, representing a significant health burden in both developed and developing countries. Without disease-modifying treatments currently available, the development of effective neurotherapeutics is a health priority. In this work, a new series of peptide-conjugates of the Glypromate neuropeptide is reported to determine the interplay of annular constriction and neuroprotective activity. To this end, (1R,3S,4S)-2-azanorbornane-3-carboxylic acid was used as an l-proline and l-pipecolic acid surrogate in addition to functionalization of the glutamate residue with relevant active pharmaceutical ingredients (APIs), namely amantadine, memantine, and (R)-1-aminoindane. Using non-differentiated SH-SY5Y cells, conjugates 14a and 15a, functionalized with amantadine, significantly reduced protein aggregation, with 15a outperforming both Glypromate (2-fold enhancement, p < 0.05) and an equimolar mixture of Glypromate and amantadine (p < 0.0001). On the other hand, in SH-SY5Y differentiated cells, conjugate 18c functionalized with (R)-1-aminoindane counteracted the toxicity elicited by paraquat (p < 0.0001), while Glypromate was found to exacerbate the neurotoxicity. Altogether, this work adds new insights into Glypromate research by demonstrating that chemical conjugation and annular constriction are effective strategies to tune neuroprotective responses against different neurotoxic stimuli, paving the way for the development of new neurotherapeutics.
Collapse
Affiliation(s)
- Sara C Silva-Reis
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
- UCIBIO/REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - Vera M Costa
- UCIBIO/REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - Daniela Correia da Silva
- LAQV/REQUIMTE, Laboratory of Pharmacognosy, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - David M Pereira
- LAQV/REQUIMTE, Laboratory of Pharmacognosy, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto 4050-313 Porto Portugal
| | - Xavier Cruz Correia
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
| | - Xerardo García-Mera
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela E-15782 Santiago de Compostela Spain
| | - José E Rodríguez-Borges
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
| | - Ivo E Sampaio-Dias
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto 4169-007 Porto Portugal
| |
Collapse
|
31
|
Raabe HA, Costin GE, Allen DG, Lowit A, Corvaro M, O'Dell L, Breeden-Alemi J, Page K, Perron M, Flint Silva T, Westerink W, Baker E, Sullivan K. Human relevance of in vivo and in vitro skin irritation tests for hazard classification of pesticides. Cutan Ocul Toxicol 2024:1-21. [PMID: 39180341 DOI: 10.1080/15569527.2024.2387596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/24/2024] [Indexed: 08/26/2024]
Abstract
Background: Test methods to inform hazard characterization and labeling of pesticides to protect human health are typically conducted using laboratory animals, and for skin irritation/corrosion the rabbit Draize test is currently required by many regulatory agencies. Although the Draize test is generally regarded to provide protective classifications for human health, new approach methodologies (NAMs) have been developed that offer more human relevant models that circumvent the uncertainty associated with species differences that exist between rabbits and humans. Despite wide applicability and use of these test methods across a broad range of chemicals, they have not been widely adopted for testing pesticides and pesticidal formulations. One of the barriers to adoption of these methods in this sector is low concordance with results from the Draize rabbit test, particularly for chemicals within the mild to moderate irritation spectrum. Methods: This review compares and contrasts the extent to which available models used in skin irritation testing mimic the anatomy and physiology of human skin, and how each aligns with the known key events leading to chemically-induced adverse skin irritation and corrosion. Doing so fully characterizes the human relevance of each method. Results: As alternatives to the rabbit Draize test, several protocols using ex vivo, in chemico, and in vitro skin models are available as internationally harmonized test guidelines. These methods rely on a variety of models of human skin, including excised rodent skin, synthetic biochemical models of barrier function, cell culture systems, and reconstructed human tissue models. We find these models exhibit biological and mechanistic relevance aligned with human skin irritation responses. Further, recent retrospective analyses have shown that the reproducibility of the Draize test is less than 50% for mild and moderate responses, with many of the replicate predictions spanning more than one category (e.g., a moderate response reported in one study followed by a non-irritant response reported in another study). Conclusions: Based on this comparative evaluation, we recommend top-down and bottom-up testing strategies that use the most human relevant in vitro test methods for skin irritation and corrosion classification of pesticides and pesticide formulations. To further discriminate among mild and non-irritant formulations, optimization of a cytokine release protocol and subsequent analyses of reference formulation test results is recommended.
Collapse
Affiliation(s)
- Hans A Raabe
- Institute for In Vitro Sciences, Gaithersburg, MD, USA
| | | | | | - Anna Lowit
- US Environmental Protection Agency, Washington, DC, USA
| | | | | | | | | | | | | | | | - Elizabeth Baker
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | | |
Collapse
|
32
|
Raheema Sharafudeen R, Abraham A. Hepatoprotective potential of coconut inflorescence sap against paracetamol induced toxicity in hep G2 cell lines. Food Chem Toxicol 2024; 193:114946. [PMID: 39181230 DOI: 10.1016/j.fct.2024.114946] [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: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Coconut Inflorescence Sap (CIS) is the sweet, oyster-white colored, non-fermented juice obtained from the immature inflorescence of the Coconut tree. Acetaminophen (N-acetyl-p-aminophenol, or paracetamol) is one of the most frequently used drugs worldwide as an antipyretic or analgesic. HepG2 cell lines were used as an experimental model for studying in vitro hepatotoxicity induced by Paracetamol. The present study aims to identify biologically active compounds of CIS using LCMS analysis and to elucidate the ameliorative potential of CIS in alleviating paracetamol-induced hepatotoxicity. LC-MS analysis revealed the presence of 17 bioactive compounds. HepG2 cells were pretreated with Paracetamol (20 mM) for inducing toxicity, and Silymarin at a concentration of 50 μg/ml was used as a standard drug. The morphological analysis and MTT assay showed effective recovery from toxicity in cells treated with CIS in a dose-dependent manner. CIS at 25 μg/ml potentially showed the highest percentage of inhibitory activity against the toxicity induced by paracetamol. The treatment with paracetamol significantly increased the indicators of liver toxicity - LDH, SGOT, SGPT, and Glut.S Transferase in the media.CIS administration also increased the total protein levels, SOD, and Catalase activity. The morphological analysis, MTT assay, cytocompatibility studies, determination of enzymatic activities, etc., confirms the significant hepatoprotective efficacy of CIS.
Collapse
Affiliation(s)
| | - Annie Abraham
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
33
|
Pozzoli C, Martinelli G, Fumagalli M, Di Lorenzo C, Maranta N, Colombo L, Piazza S, Dell’Agli M, Sangiovanni E. Castanea sativa Mill. By-Products: Investigation of Potential Anti-Inflammatory Effects in Human Intestinal Epithelial Cells. Molecules 2024; 29:3951. [PMID: 39203029 PMCID: PMC11357300 DOI: 10.3390/molecules29163951] [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: 07/23/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Castanea sativa Mill. (C. sativa) processing and pruning generate several by-products, including leaves, burs, and shells (inner and outer teguments), which are considered an important source of high-value phytochemicals. Ellagitannins from C. sativa leaf extracts have been described to impair H. pylori viability and inflammation in gastric cells. Furthermore, chestnut shells showed an important anti-inflammatory effect in gastric epithelial cells. Dietary polyphenols, including tannins, have been reported to interfere with targets of inflammation, including the nuclear factor κB (NF-κB). A promising role as a further therapeutical target for gut disorders has been recently proposed for the regulatory subunit of hypoxia-inducible factor (HIF-1α), as a potential stabilizer of intestinal barrier integrity. Therefore, the main objective of this work is the chemical characterization of several chestnut by-products (bud, spiny bur, wood, pericarp and episperm), together with the exploitation of their anti-inflammatory properties in intestinal cells, scavenging capacity, and stability following gastrointestinal digestion. The chemical characterization confirmed the presence of bioactive polyphenols in the extracts, including ellagitannins. In CaCo-2 cells stimulated by an IL-1β-IFN-γ cocktail, nearly all chestnut by-products (50 µg/mL) inhibited the release of proinflammatory mediators (CXCL-10, IL-8, MCP-1, ICAM), along with the NF-κB-driven transcription, and induced the HRE-driven transcription. The stability of the most promising extracts, identified through PCA and cluster analysis, was addressed by in vitro gastrointestinal digestion. Despite the significant reduction in total polyphenol index of chestnut bud and wood after gastric and intestinal digestion, the activity of these extracts on both scavenging and anti-inflammatory parameters remained promising. These data contribute to exploit the potential of chestnut by-products as sources of dietary polyphenols with anti-inflammatory properties at the intestinal level. Moreover, this study could represent an important step to encourage the recycling and valorization of chestnut by-products, promoting the circular economy and reducing the environmental impact related to the management of agriculture waste.
Collapse
Affiliation(s)
- Carola Pozzoli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Giulia Martinelli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Marco Fumagalli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Chiara Di Lorenzo
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Nicole Maranta
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Luca Colombo
- Consorzio Castanicoltori di Brinzio, Orino e Castello Cabiaglio, Società Cooperativa Agricola-Varese, 21100 Varese, Italy;
| | - Stefano Piazza
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Mario Dell’Agli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| | - Enrico Sangiovanni
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, 20133 Milan, Italy; (C.P.); (G.M.); (M.F.); (C.D.L.); (N.M.); (E.S.)
| |
Collapse
|
34
|
Salaroglio IC, Stefanova D, Teixeira RG, Oliveira NFB, Ahmed A, Fusi F, Tzankova V, Yordanov Y, Machuqueiro M, Saponara S, Valente A, Riganti C. A novel combinatory treatment against a CDDP-resistant non-small cell lung cancer based on a Ruthenium(II)-cyclopentadienyl compound. Pharmacol Res 2024; 208:107353. [PMID: 39159730 DOI: 10.1016/j.phrs.2024.107353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/15/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024]
Abstract
The therapeutic approach to many solid tumors, including non-small cell lung cancer (NSCLC), is mainly based on the use of platinum-containing anticancer agents and is often characterized by acquired or intrinsic resistance to the drug. Therefore, the search for safer and more effective drugs is still an open challenge. Two organometallic ruthenium(II)-cyclopentadienyl compounds [Ru(η5-C5H4CHO)(Me2bipy)(PPh3)]+ (RT150) and [Ru(η5-C5H4CH2OH)(Me2bipy)(PPh3)][CF3SO3] (RT151) were tested against a panel of cisplatin-resistant NSCLC cell lines and xenografts. They were more effective than cisplatin in inducing oxidative stress and DNA damage, affecting the cell cycle and causing apoptosis. Importantly, they were found to be inhibitors of drug efflux transporters. Due to this property, the compounds significantly increased the retention and cytotoxicity of cisplatin within NSCLC cells. Notably, they did not display high toxicity in vitro against non-transformed cells (red blood cells, fibroblasts, bronchial epithelial cells, cardiomyocytes, and endothelial cells). Both compounds induced vasorelaxation and reduced endothelial cell migration, suggesting potential anti-angiogenic properties. RT151 confirmed its efficacy against NSCLC xenografts resistant to cisplatin. Either alone or combined with low doses of cisplatin, RT151 showed a good biodistribution profile in the liver, kidney, spleen, lung, and tumor. Hematochemical analysis and post-mortem organ pathology confirmed the safety of the compound in vivo, also when combined with cisplatin. To sum up, we have confirmed the effectiveness of a novel class of drugs against cisplatin-resistant NSCLC. Additionally, the compounds have a good biocompatibility and safety profile.
Collapse
Affiliation(s)
- Iris C Salaroglio
- Department of Oncology and Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, 10126 Torino, Italy
| | - Denitsa Stefanova
- Medical University of Sofia, Faculty of Pharmacy, Department of Pharmacology, Pharmacotherapy and Toxicology, 2 Dunav Str., Sofia 1000, Bulgaria
| | - Ricardo G Teixeira
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal
| | - Nuno F B Oliveira
- BioISI - Instituto de Biossistemas e Ciências Integrativas, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa 1749-016, Portugal
| | - Amer Ahmed
- University of Siena, Department of Life Sciences, via Aldo Moro, 2, Siena 53100, Italy
| | - Fabio Fusi
- University of Siena, Department of Biotechnologies, Chemistry and Pharmacy, via Aldo Moro 2, Siena 53100, Italy
| | - Virginia Tzankova
- Medical University of Sofia, Faculty of Pharmacy, Department of Pharmacology, Pharmacotherapy and Toxicology, 2 Dunav Str., Sofia 1000, Bulgaria
| | - Yordan Yordanov
- Medical University of Sofia, Faculty of Pharmacy, Department of Pharmacology, Pharmacotherapy and Toxicology, 2 Dunav Str., Sofia 1000, Bulgaria
| | - Miguel Machuqueiro
- BioISI - Instituto de Biossistemas e Ciências Integrativas, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa 1749-016, Portugal
| | - Simona Saponara
- University of Siena, Department of Life Sciences, via Aldo Moro, 2, Siena 53100, Italy
| | - Andreia Valente
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal.
| | - Chiara Riganti
- Department of Oncology and Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, 10126 Torino, Italy.
| |
Collapse
|
35
|
Delenko J, Xue X, Chatterjee PK, Hyman N, Shih AJ, Adelson RP, Safaric Tepes P, Gregersen PK, Metz CN. Quercetin enhances decidualization through AKT-ERK-p53 signaling and supports a role for senescence in endometriosis. Reprod Biol Endocrinol 2024; 22:100. [PMID: 39118090 PMCID: PMC11308242 DOI: 10.1186/s12958-024-01265-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Patients with endometriosis suffer with chronic pelvic pain and infertility, and from the lack of pharmacologic therapies that consistently halt disease progression. Differences in the endometrium of patients with endometriosis vs. unaffected controls are well-documented. Specifically, shed endometrial tissues (delivered to the pelvic cavity via retrograde menstruation) reveal that a subset of stromal cells exhibiting pro-inflammatory, pro-fibrotic, and pro-senescence-like phenotypes is enhanced in endometriosis patients compared to controls. Additionally, cultured biopsy-derived endometrial stromal cells from endometriosis patients exhibit impaired decidualization, a defined differentiation process required for human embryo implantation and pregnancy. Quercetin, a senolytic agent, shows therapeutic potential for pulmonary fibrosis, a disorder attributed to senescent pulmonary fibroblasts. In rodent models of endometriosis, quercetin shows promise, and quercetin improves decidualization in vitro. However, the exact mechanisms are not completely understood. Therefore, we investigated the effects of quercetin on menstrual effluent-derived endometrial stromal cells from endometriosis patients and unaffected controls to define the signaling pathways underlying quercetin's effects on endometrial stromal cells. METHODS Menstrual effluent-derived endometrial stromal cells were collected and cultured from unaffected controls and endometriosis patients and then, low passage cells were treated with quercetin (25 µM) under basal or standard decidualization conditions. Decidualization responses were analyzed by measuring the production of IGFBP1 and PRL. Also, the effects of quercetin on intracellular cAMP levels and cellular oxidative stress responses were measured. Phosphokinase arrays, western blotting, and flow cytometry methods were performed to define the effects of quercetin on various signaling pathways and the potential mechanistic roles of quercetin. RESULTS Quercetin significantly promotes decidualization of control- and endometriosis-endometrial stromal cells. Quercetin substantially reduces the phosphorylation of multiple signaling molecules in the AKT and ERK1/2 pathways, while enhancing the phosphorylation of p53 and total p53 levels. Furthermore, p53 inhibition blocks decidualization while p53 activation promotes decidualization. Finally, we provide evidence that quercetin increases apoptosis of endometrial stromal cells with a senescent-like phenotype. CONCLUSIONS These data provide insight into the mechanisms of action of quercetin on endometrial stromal cells and warrant future clinical trials to test quercetin and other senolytics for treating endometriosis.
Collapse
Affiliation(s)
- Julia Delenko
- The Donald and Barbara Zucker School of Medicine, Hempstead, NY, 11549, USA
| | - Xiangying Xue
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Prodyot K Chatterjee
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Nathaniel Hyman
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Andrew J Shih
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Robert P Adelson
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Polona Safaric Tepes
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Peter K Gregersen
- The Donald and Barbara Zucker School of Medicine, Hempstead, NY, 11549, USA.
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
| | - Christine N Metz
- The Donald and Barbara Zucker School of Medicine, Hempstead, NY, 11549, USA.
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
| |
Collapse
|
36
|
Kennelly S, Sawyer JM, Payne AF, Ciota AT, Harki DA. Development of 3'-Deoxy-3',4'-didehydro-nucleoside Prodrug Inhibitors of West Nile and Zika Viruses. ACS Med Chem Lett 2024; 15:1334-1339. [PMID: 39140046 PMCID: PMC11318099 DOI: 10.1021/acsmedchemlett.4c00225] [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: 05/15/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 08/15/2024] Open
Abstract
The antiviral enzyme viperin catalyzes the formation of 3'-deoxy-3',4'-didehydro-cytidine-5'-triphosphate (ddhCTP). ddhCTP is incorporated into viral genomes and terminates genomic replication to confer broad-spectrum antiviral effects. We have previously utilized phosphoramidate pronucleotide (ProTide) technology to enable metabolic production of ddhCTP in cells from an exogenously dosed 3'-deoxy-3',4'-didehydro-cytidine ProTide, which confers inhibitory activity against West Nile virus (WNV) and Zika virus (ZIKV). Herein, we synthesized 3'-deoxy-3',4'-didehydro-nucleosides containing all native nucleobases (thymine, uracil, adenine, guanine, and hypoxanthine), elaborated each to a ProTide, and measured their activity for controlling WNV and ZIKV infection. In comparison to the ddhC ProTide, we found that the ProTides of 3'-deoxy-3',4'-didehydro-guanosine and 3'-deoxy-3',4'-didehydro-adenosine possess 2- and 4-fold greater antiviral effects against ZIKV, respectively. Collectively, this work advances the development of 3'-deoxy-3',4'-didehydro nucleosides as promising compounds for further development into broad-spectrum antiviral agents.
Collapse
Affiliation(s)
- Samantha
A. Kennelly
- Department
of Medicinal Chemistry, University of Minnesota, 2231 Sixth Street SE, Minneapolis, Minnesota 55455, United States
| | - Jacob M. Sawyer
- Department
of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Anne F. Payne
- Department
of Biomedical Sciences, State University
of New York at Albany School of Public Health, Albany, New York 12144, United States
- The
Arbovirus Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, New York 12201, United States
| | - Alexander T. Ciota
- Department
of Biomedical Sciences, State University
of New York at Albany School of Public Health, Albany, New York 12144, United States
- The
Arbovirus Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, New York 12201, United States
| | - Daniel A. Harki
- Department
of Medicinal Chemistry, University of Minnesota, 2231 Sixth Street SE, Minneapolis, Minnesota 55455, United States
- Department
of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
37
|
Rózga K, Błauż A, Moscoh Ayine-Tora D, Puścion E, Hartinger CG, Plażuk D, Rychlik B. Synthesis and Biological Properties of Ferrocenyl and Organic Methotrexate Derivatives. ACS OMEGA 2024; 9:33845-33856. [PMID: 39130602 PMCID: PMC11308014 DOI: 10.1021/acsomega.4c03602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/12/2024] [Accepted: 07/03/2024] [Indexed: 08/13/2024]
Abstract
Synthesis and biological activity of two series of modified side chain methotrexate (MTX) derivatives are presented, one with a ferrocenyl moiety inserted between the pteroyl and glutamate portions of the molecule and the other with glutamate substituted for short chain amino acids. Ferrocenyl derivatives of MTX turned out to be rather moderate inhibitors of dihydrofolate reductase (DHFR) although molecular modeling suggested more effective interactions between these compounds and the target enzyme. More interestingly, ferrocene-decorated MTX derivatives were able to impede the proliferation of four murine and human cell lines as well as their methotrexate-resistant counterparts, overcoming the multidrug resistance (MDR) barrier. They were also able to directly interact with Abcc1, an MDR protein. Of the amino acid pteroyl conjugates, the γ-aminobutyric acid derivative was an efficient inhibitor of DHFR but had no effect on cell proliferation in the concentration range studied while a taurine conjugate was a poor DHFR inhibitor but able to affect cell viability. We postulate that modification of the methotrexate side chain may be an efficient strategy to overcome efflux-dependent methotrexate resistance.
Collapse
Affiliation(s)
- Karolina Rózga
- Department
of Organic Chemistry, Faculty of Chemistry, University of Lodz, 12 Tamka, 91-403 Łódź, Poland
| | - Andrzej Błauż
- Cytometry
Lab, Department of Oncobiology and Epigenetics, Faculty of Biology
and Environmental Protection, University
of Lodz, 141/143 Pomorska, 90-236 Łódź, Poland
| | - Daniel Moscoh Ayine-Tora
- School
of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Department
of Chemistry, University of Ghana, LG 56 Legon-Accra, Ghana
| | - Ernest Puścion
- Cytometry
Lab, Department of Oncobiology and Epigenetics, Faculty of Biology
and Environmental Protection, University
of Lodz, 141/143 Pomorska, 90-236 Łódź, Poland
| | - Christian G. Hartinger
- School
of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Damian Plażuk
- Department
of Organic Chemistry, Faculty of Chemistry, University of Lodz, 12 Tamka, 91-403 Łódź, Poland
| | - Błażej Rychlik
- Cytometry
Lab, Department of Oncobiology and Epigenetics, Faculty of Biology
and Environmental Protection, University
of Lodz, 141/143 Pomorska, 90-236 Łódź, Poland
| |
Collapse
|
38
|
Lucia U, Bergandi L, Grisolia G, Fino D, Mareschi K, Marini E, Santa Banche Niclot AG, Tirtei E, Asaftei SD, Fagioli F, Ponzetto A, Silvagno F. The exposure to extremely low frequency electromagnetic-fields inhibits the growth and potentiates the sensitivity to chemotherapy of bidimensional and tridimensional human osteosarcoma models. Biomed Pharmacother 2024; 177:117162. [PMID: 39024997 DOI: 10.1016/j.biopha.2024.117162] [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: 04/11/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
We previously established a thermodynamical model to calculate the specific frequencies of extremely low frequency-electromagnetic field (ELF-EMF) able to arrest the growth of cancer cells. In the present study, for the first time, we investigated the efficacy of this technology on osteosarcoma, and we applied a precise frequency of the electromagnetic field on three human osteosarcoma cell lines, grown as adherent cells and spheroids. We evaluated the antitumour efficacy of irradiation in terms of response to chemotherapeutic treatments, which is usually poor in this type of cancer. Importantly, the results of this novel combinatorial approach revealed that the specific exposure can potentiate the efficacy of several chemotherapeutic drugs, both on bidimensional and tridimensional cancer models. The effectiveness of cisplatinum, methotrexate, ifosfamide and doxorubicin was greatly increased by the concomitant application of the specific ELF-EMF. Moreover, our experiments confirmed that ELF-EMF inhibited the proliferation and modulated the mitochondrial metabolism of all cancer models tested, whereas mesenchymal cells were not affected. The latter finding is extremely valuable, given the importance of preserving the cell reservoir necessary for tissue regeneration after chemotherapy. Altogether, this novel evidence opens new avenues to the clinical applications of ELF-EMF in oncology.
Collapse
Affiliation(s)
- Umberto Lucia
- Dipartimento Energia "Galileo Ferraris", Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy.
| | - Loredana Bergandi
- Department of Oncology, University of Torino, Via Santena 5 bis, Torino 10126, Italy
| | - Giulia Grisolia
- Dipartimento di Ingegneria dell'Ambiente, del Territorio e delle Infrastrutture, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Debora Fino
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Katia Mareschi
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Elena Marini
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy
| | | | - Elisa Tirtei
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Sebastian Dorin Asaftei
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Franca Fagioli
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Antonio Ponzetto
- Dipartimento di Scienze Mediche, Università di Torino, Corso Dogliotti 14, Torino 10126, Italy
| | - Francesca Silvagno
- Department of Oncology, University of Torino, Via Santena 5 bis, Torino 10126, Italy.
| |
Collapse
|
39
|
Fragelli BDL, Assis M, Rodolpho JMA, Godoy KF, Líbero LO, Anibal FF, Longo E. Modulation of cell death mechanisms via α-Ag 2WO 4 morphology-dependent factors. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 257:112947. [PMID: 38851043 DOI: 10.1016/j.jphotobiol.2024.112947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/09/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
The cytotoxic of α-Ag2WO4 synthesized in different morphologies (cuboidal (AW-C), hexagonal rod-like (AW-HRL) and nanometric rod-like (AW-NRL) was analyzed to understand the impact of morphological modulation on the toxicity of 3 T3 cell lines in the dark and when photoactivated by visible light. Pathways of toxicity were examined, such as parameters and electrostatic interaction, uptake, ion release and ROS production. Cytotoxicity was observed for all samples after reaching concentrations exceeding 7.8 μg/mL. Uptake tests demonstrated that the samples were not internalized by cells, likely due to their negative surface charge. AW-NRL exhibited autophagy in the absence of light and during photoactivation, primarily attributed to its ability to generate singlet oxygen. Analyzing intercellular ROS and RNS production, AW-HRL induced an increase in NO through exposure to photo-generated hydroxyl radicals, while AW-NRL showed increases only at non-photoactivated concentrations and AW-C did not exhibit increases. Interestingly, in the dark, these cells showed a low propensity for apoptosis, with late apoptosis and necrosis being more pronounced. When photoactivated, this behavior changed, revealing predominantly apoptotic and late apoptotic cell death. There is a need for an understanding of how morphology can alter the biological properties of α-Ag2WO4 to predict and optimize its effects on cellular responses.
Collapse
Affiliation(s)
- Bruna D L Fragelli
- Center for Development of Functional Materials, Federal University of São Carlos (UFSCar), 13565-905 São Carlos, SP, Brazil.
| | - Marcelo Assis
- Department of Analytical and Physical Chemistry, University Jaume I (UJI), 12071 Castelló, Spain.
| | - Joice M A Rodolpho
- Laboratory of Inflammation and Infectious Diseases, Department of Morphology and Pathology, Federal University of São Carlos (UFSCar), 13565-905 São Carlos, SP, Brazil
| | - Krissia F Godoy
- Laboratory of Inflammation and Infectious Diseases, Department of Morphology and Pathology, Federal University of São Carlos (UFSCar), 13565-905 São Carlos, SP, Brazil
| | - Laura O Líbero
- Center for Development of Functional Materials, Federal University of São Carlos (UFSCar), 13565-905 São Carlos, SP, Brazil
| | - Fernanda F Anibal
- Laboratory of Inflammation and Infectious Diseases, Department of Morphology and Pathology, Federal University of São Carlos (UFSCar), 13565-905 São Carlos, SP, Brazil
| | - Elson Longo
- Center for Development of Functional Materials, Federal University of São Carlos (UFSCar), 13565-905 São Carlos, SP, Brazil
| |
Collapse
|
40
|
Coelho MM, Costa I, de Albuquerque ACF, Santos Junior FMD, Silva B, Silva R, Fernandes C, Remião F, Tiritan ME. Milligram scale enantioresolution of promethazine and its main metabolites, determination of their absolute configuration and assessment of enantioselective effects on human SY-SY5Y cells. J Pharm Biomed Anal 2024; 245:116152. [PMID: 38643704 DOI: 10.1016/j.jpba.2024.116152] [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: 03/03/2024] [Revised: 04/07/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
The misuse of pharmaceuticals has significantly increased in recent decades, becoming a major public health concern. The risks associated with medication misuse are particularly high in cases of overdose, especially when the active substances are chiral, as enantioselectivity plays an important role in toxicity. Promethazine (PMZ) is a chiral antihistamine marketed as a racemate and it is misused in "Purple Drank", a recreational drug beverage, that combines codeine and/or PMZ, with soda or alcohol leading to serious health consequences and fatalities in consumers around the world, particularly among teenagers. Information regarding the enantioselectivity in the toxicity of (R,S)-PMZ and its main metabolites, namely promethazine sulfoxide (PMZSO) and desmonomethyl promethazine (DMPMZ), is unknown. This work reported, for the first time, the enantioseparation, in milligram scale, of (R,S)-PMZ, (R,S)-DMPMZ, (R,S)- PMZSO and the determination of their absolute configurations by electronic circular dichroism (ECD). The enantioseparation of all the six enantiomers was accomplished in a homemade semi-preparative column with amylose tris-3,5-dimethylphenylcarbamate (AD) coated with aminopropyl Nucleosil silica. The enantiomeric purity was evaluated using the analytical Lux® 3 µm i-Amylose-3 column, yielding enantiomeric purity values ranging between 94.4% and 99.7%. The elution order of all the enantiomers was accomplished combining the ECD results with an optical rotation detector. The elution order of the enantiomers was influenced only by the chiral selector, rather than the mobile phase. The cytotoxicity of the racemates and the isolated enantiomers towards differentiated SH-SY5Y cells was evaluated. (R,S)-DMPMZ exhibited a significantly higher cytotoxicity than (R,S)-PMZ, suggesting the metabolic bioactivation of (R,S)-PMZ. Conversely, no significant cytotoxicity was found for (R,S)-PMZSO, underscoring a metabolic detoxification pathway. Remarkably, enantioselectivity was observed for the cytotoxicity of PMZ; (R)-PMZ was significantly more cytotoxic than (S)-PMZ. The results underscore the importance to isolate the enantiomers in their enantiomerically form and their correct identification for toxicity enantioselectivity studies, which are vital to understand the drug's behaviour and safety, especially in case of overdoses.
Collapse
Affiliation(s)
- Maria Miguel Coelho
- Laboratory of Organic Chemistry and Pharmaceuticals, Department of Chemical Sciences, Faculty of Pharmacy of the University of Porto, Porto 4050-313, Portugal; CIIMAR-Interdisciplinary Center of Marine and Environmental Research University of Porto, Porto de Leixões Cruise Terminal, Matosinhos 4450-208, Portugal; Associate Laboratory i4HB-Insitute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Inês Costa
- Associate Laboratory i4HB-Insitute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal; UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | | | | | - Bárbara Silva
- Associate Laboratory i4HB-Insitute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal; UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Renata Silva
- Associate Laboratory i4HB-Insitute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal; UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Carla Fernandes
- Laboratory of Organic Chemistry and Pharmaceuticals, Department of Chemical Sciences, Faculty of Pharmacy of the University of Porto, Porto 4050-313, Portugal; CIIMAR-Interdisciplinary Center of Marine and Environmental Research University of Porto, Porto de Leixões Cruise Terminal, Matosinhos 4450-208, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Insitute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal; UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Maria Elizabeth Tiritan
- Laboratory of Organic Chemistry and Pharmaceuticals, Department of Chemical Sciences, Faculty of Pharmacy of the University of Porto, Porto 4050-313, Portugal; CIIMAR-Interdisciplinary Center of Marine and Environmental Research University of Porto, Porto de Leixões Cruise Terminal, Matosinhos 4450-208, Portugal; 1H-TOXRUN - One Health Toxicology Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra 4585-116, Portugal.
| |
Collapse
|
41
|
Heydari M, Hosseinzadeh Colagar A, Sabour D, Khorasani HR. Evidence of Urtica dioica Agglutinin's Antiproliferative and Anti-migratory Potentials on the Hyaluronic Acid-Overexpressing Prostate Cancer Cells. PLANTA MEDICA 2024; 90:774-784. [PMID: 38942031 DOI: 10.1055/a-2324-2250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Hyaluronic acid is composed of repeating sugar units, glucuronic acid and N-acetylglucosamine, which are often associated with increased tumor progression. Urtica dioica agglutinin is a potential component that exhibits a high affinity for binding to N-acetylglucosamine. This study aimed to investigate U. dioica Agglutinin's potential to inhibit the proliferation and migration of prostate cancer cells with high expression of hyaluronic acid through molecular docking and in vitro studies. The expression of hyaluronan synthase genes in prostate tissue and cell lines was checked by an in silico study, and the interaction between hyaluronic acid with both CD44 transmembrane glycoprotein and U. dioica agglutinin was analyzed through molecular docking. U. dioica Agglutinin's effect on cell viability (neutral red uptake assay), migration (scratch wound healing assays), and both CD44 and Nanog expression (quantitative real-time polymerase chain reaction) were assessed in vitro. The results showed that in prostate cancer cell lines, the PC3 cell line has the highest expression of hyaluronan synthase genes. U. dioica agglutinin exhibits an interaction of six specific residues on CD44 compared to hyaluronic acid's singular residue. While U. dioica agglutinin alone effectively reduced cell viability and wound closer (≥ 150 µg/mL), combining it with hyaluronic acid significantly shifted the effective concentration to a higher dose (≥ 350 µg/mL). These results, together with low Nanog and high CD44 gene expression, suggest that U. dioica agglutinin may impair the CD44-HA pathway in PC3 cells. This possibility is supported by U. dioica Agglutinin's ability to compete with hyaluronic acid for binding to CD44. Based on this, U. dioica agglutinin as a plant lectin shows promise in inhibiting cancer proliferation and migration by targeting its dependence on hyaluronic acid.
Collapse
Affiliation(s)
- Mohammadkazem Heydari
- Department of Molecular and Cell Biology, Faculty of Science, University of Mazandaran, Babolsar, Mazandaran, Iran
| | - Abasalt Hosseinzadeh Colagar
- Department of Molecular and Cell Biology, Faculty of Science, University of Mazandaran, Babolsar, Mazandaran, Iran
| | - Davood Sabour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Babol, Iran
| | - Hamid Reza Khorasani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Babol, Iran
| |
Collapse
|
42
|
Carvalho LCF, Ferreira FM, Dias BV, Azevedo DCD, de Souza GHB, Milagre MM, de Lana M, Vieira PMDA, Carneiro CM, Paula-Gomes SD, Cangussu SD, Costa DC. Silymarin inhibits the lipogenic pathway and reduces worsening of non-alcoholic fatty liver disease (NAFLD) in mice. Arch Physiol Biochem 2024; 130:460-474. [PMID: 36328030 DOI: 10.1080/13813455.2022.2138445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022]
Abstract
CONTEXT The role of silymarin in hepatic lipid dysfunction and its possible mechanisms of action were investigated. OBJECTIVE To evaluate the effects of silymarin on hepatic and metabolic profiles in mice fed with 30% fructose for 8 weeks. METHODS We evaluated the antioxidant profile of silymarin; mice consumed 30% fructose and were treated with silymarin (120 mg/kg/day or 240 mg/kg/day). We performed biochemical, redox status, and histopathological assays. RT-qPCR was performed to detect ACC-1, ACC-2, FAS, and CS expression, and western blotting to detect PGC-1α levels. RESULTS Silymarin contains high levels of phenolic compounds and flavonoids and exhibited significant antioxidant capacity in vitro. In vivo, the fructose-fed groups showed increased levels of AST, ALT, SOD/CAT, TBARS, hepatic TG, and cholesterol, as well as hypertriglyceridaemia, hypercholesterolaemia, and increased ACC-1 and FAS. Silymarin treatment reduced these parameters and increased mRNA levels and activity of hepatic citrate synthase. CONCLUSIONS These results suggest that silymarin reduces worsening of NAFLD.
Collapse
Affiliation(s)
| | | | - Bruna Vidal Dias
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | | | - Matheus Marque Milagre
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Marta de Lana
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | | | | | - Sílvia de Paula-Gomes
- Laboratório de Bioquímica e Biologia Molecular, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Silvia Dantas Cangussu
- Laboratório de Fisiopatologia Experimental, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Daniela Caldeira Costa
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
43
|
de Souza TL, da Luz JZ, Roque ADA, Opuskevitch I, Ferreira FCADS, Ribeiro CADO, Neto FF. Exploring the endocrine disrupting potential of a complex mixture of PAHs in the estrogen pathway in Oreochromis niloticus hepatocytes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 273:107002. [PMID: 38936242 DOI: 10.1016/j.aquatox.2024.107002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024]
Abstract
This study aimed to investigate the toxicity and endocrine disrupting potential of a complex mixture of polycyclic aromatic hydrocarbons (PAHs) in the estrogen pathway using hepatocytes of Nile tilapia Oreochromis niloticus, the hepatocytes were exposed to various concentrations of the PAH mixture, and multiple endpoints were evaluated to assess their effects on cell viability, gene expression, oxidative stress markers, and efflux activity. The results revealed that the PAH mixture had limited effects on hepatocyte metabolism and cell adhesion, as indicated by the non-significant changes observed in MTT metabolism, neutral red retention, and crystal violet staining. However, significant alterations were observed in the expression of genes related to the estrogen pathway. Specifically, vitellogenin (vtg) exhibited a substantial increase of approximately 120% compared to the control group. Similarly, estrogen receptor 2 (esr2) showed a significant upregulation of approximately 90%. In contrast, no significant differences were observed in the expression of estrogen receptor 1 (esr1) and the G protein-coupled estrogen receptor 1 (gper1). Furthermore, the PAH mixture elicited complex responses in oxidative stress markers. While reactive oxygen species (ROS) and reactive nitrogen species (RNS) levels remained unchanged, the activity of catalase (Cat) was significantly reduced, whereas superoxide dismutase (Sod) activity, glutathione S-transferase (Gst) activity, and non-protein thiols levels were significantly elevated. In addition, the PAH mixture significantly influenced efflux activity, as evidenced by the increased efflux of rhodamine and calcein, indicating alterations in multixenobiotic resistance (MXR)-associated proteins. Overall, these findings, associated with bioinformatic analysis, highlight the potential of the PAH mixture to modulate the estrogen pathway and induce oxidative stress in O. niloticus hepatocytes. Understanding the mechanisms underlying these effects is crucial for assessing the ecological risks of PAH exposure and developing appropriate strategies to mitigate their adverse impacts on aquatic organisms.
Collapse
Affiliation(s)
- Tugstênio Lima de Souza
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil.
| | - Jessica Zablocki da Luz
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Aliciane de Almeida Roque
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Iracema Opuskevitch
- Copel GeT-SOS/DNGT - Rua José Izidoro Biazetto, no. 18, Bloco A, CEP 81200-240, Curitiba, PR, Brazil
| | | | - Ciro Alberto de Oliveira Ribeiro
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil
| | - Francisco Filipak Neto
- Laboratório de Toxicologia Celular, Departamento de Biologia Celular, Universidade Federal do Paraná, CEP 81.531-980, Curitiba, PR, Brazil.
| |
Collapse
|
44
|
Sikorska M, Ruzycka-Ayoush M, Rios-Mondragon I, Longhin EM, Meczynska-Wielgosz S, Wojewodzka M, Kowalczyk A, Kasprzak A, Nowakowska J, Sobczak K, Muszynska M, Cimpan MR, Runden-Pran E, Shaposhnikov S, Kruszewski M, Dusinska M, Nowicka AM, Grudzinski IP. Lack of cytotoxic and genotoxic effects of mPEG-silane coated iron(III) oxide nanoparticles doped with magnesium despite cellular uptake in cancerous and noncancerous lung cells. Toxicol In Vitro 2024; 99:105850. [PMID: 38801838 DOI: 10.1016/j.tiv.2024.105850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Cytotoxic and genotoxic effects of novel mPEG-silane coated iron(III) oxide nanoparticles doped with magnesium (Mg0.1-γ-Fe2O3(mPEG-silane)0.5) have been investigated on human adenocarcinomic alveolar basal epithelial (A549) and human normal bronchial epithelial (BEAS-2B) cells. In the studies several molecular and cellular targets addressing to cell membrane, cytoplasm organelles and nucleus components were served as toxicological endpoints. The as-synthesized nanoparticles were found to be stable in the cell culture media and were examined for different concentration and exposure times. No cytotoxicity of the tested nanoparticles was found although these nanoparticles slightly increased reactive oxygen species in both cell types studied. Mg0.1-γ-Fe2O3(mPEG-silane)0.5 nanoparticles did not produce any DNA strand breaks and oxidative DNA damages in A549 and BEAS-2B cells. Different concentration of Mg0.1-γ-Fe2O3(mPEG-silane)0.5 nanoparticles and different incubation time did not affect cell migration. The lung cancer cells' uptake of the nanoparticles was more effective than in normal lung cells. Altogether, the results evidence that mPEG-silane coated iron(III) oxide nanoparticles doped with magnesium do not elucidate any deleterious effects on human normal and cancerous lung cells despite cellular uptake of these nanoparticles. Therefore, it seems reasonable to conclude that these novel biocompatible nanoparticles are promising candidates for further development towards medical applications.
Collapse
Affiliation(s)
- Malgorzata Sikorska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, PL-02-097 Warsaw, Poland.
| | - Monika Ruzycka-Ayoush
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, PL-02-097 Warsaw, Poland
| | - Ivan Rios-Mondragon
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien. 19, Bergen 5009, Norway
| | - Eleonora Marta Longhin
- Health Effects Laboratory, Department of Environmental Chemistry, Norwegian Institute for Air Research, 2007 Kjeller, Norway
| | - Sylwia Meczynska-Wielgosz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna Str. 16, PL-03-195, Warsaw, Poland
| | - Maria Wojewodzka
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna Str. 16, PL-03-195, Warsaw, Poland
| | - Agata Kowalczyk
- Department of Inorganic and Analytical Chemistry, Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL-02-093 Warsaw, Poland
| | - Artur Kasprzak
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego Str. 3, PL-00-664 Warsaw, Poland
| | - Julita Nowakowska
- Laboratory of Electron and Confocal Microscopy, Faculty of Biology, University of Warsaw, Miecznikowa Str.1, PL-02-096 Warsaw, Poland
| | - Kamil Sobczak
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101 Str., PL 02-089 Warsaw, Poland
| | - Magdalena Muszynska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101 Str., PL 02-089 Warsaw, Poland; Pro-Environment Poland Sp. z o. o., Zwirki i Wigury Str. 101, PL 02-098 Warsaw, Poland
| | - Mihaela Roxana Cimpan
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien. 19, Bergen 5009, Norway
| | - Elise Runden-Pran
- Health Effects Laboratory, Department of Environmental Chemistry, Norwegian Institute for Air Research, 2007 Kjeller, Norway
| | | | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna Str. 16, PL-03-195, Warsaw, Poland; Department of Medical Biology and Translational Research, Institute of Rural Health,Jaczewskiego Str. 2, PL-20-090 Lublin, Poland
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry, Norwegian Institute for Air Research, 2007 Kjeller, Norway
| | - Anna M Nowicka
- Department of Inorganic and Analytical Chemistry, Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL-02-093 Warsaw, Poland
| | - Ireneusz P Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, PL-02-097 Warsaw, Poland
| |
Collapse
|
45
|
Kononenko V, Joukhan A, Bele T, Križaj I, Kralj S, Turk T, Drobne D. Gelatin nanoparticles loaded with 3-alkylpyridinium salt APS7, an analog of marine toxin, are a promising support in human lung cancer therapy. Biomed Pharmacother 2024; 177:117007. [PMID: 38906020 DOI: 10.1016/j.biopha.2024.117007] [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: 04/19/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024] Open
Abstract
This study demonstrates the potential of gelatin nanoparticles as a nanodelivery system for antagonists of nicotinic acetylcholine receptors (nAChRs) to improve chemotherapy efficacy and reduce off-target effects. Too often, chemotherapy for lung cancer does not lead to satisfactory results. Therefore, new approaches directed at multiple pharmacological targets in cancer therapy are being developed. Following the activation of nAChRs (e.g. by nicotine), cancer cells begin to proliferate and become more resistant to chemotherapy-induced apoptosis. This work shows that the 3-alkylpyridinium salt, APS7, a synthetic analog of a toxin from the marine sponge Haliclona (Rhizoneira) sarai, acts as an nAChR antagonist that inhibits the pro-proliferative and anti-apoptotic effects of nicotine on A549 human lung adenocarcinoma cells. In this study, gelatin-based nanoparticles filled with APS7 (APS7-GNPs) were prepared and their effects on A549 cells were compared with that of free APS7. Both APS7 and APS7-GNPs inhibited Ca2+ influx and increased the efficacy of cisplatin chemotherapy in nicotine-stimulated A549 cells. However, significant benefits from APS7-GNPs were observed - a stronger reduction in the proliferation of A549 lung cancer cells and a much higher selectivity in cytotoxicity towards cancer cells compared with non-tumorigenic lung epithelial BEAS-2B cells.
Collapse
Affiliation(s)
- Veno Kononenko
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia.
| | - Ahmad Joukhan
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia; Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana SI-1000, Slovenia
| | - Tadeja Bele
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia; Faculty of medicine, University of Ljubljana, Vrazov trg 2, Ljubljana SI-1000, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Slavko Kralj
- Department for Materials Synthesis, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Tom Turk
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia
| | - Damjana Drobne
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia.
| |
Collapse
|
46
|
Mehravanfar H, Farhadian N, Abnous K. Indocyanine green-loaded N-doped carbon quantum dot nanoparticles for effective photodynamic therapy and cell imaging of melanoma cancer: in vitro, ex vivo and in vivo study. J Drug Target 2024; 32:820-837. [PMID: 38779708 DOI: 10.1080/1061186x.2024.2358511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Indocyanine Green (ICG) as an agent for photodynamic therapy (PDT) of melanoma cancer has low quantum yield, short circulation half-life, poor photo-stability, and tendency to aggregation. PURPOSE N-doped carbon quantum dot (CQD) nanoparticle was applied to encapsulate ICG and overcome ICG obstacle in PDT with simultaneous cell imaging property. METHODS CQD was prepared using hydrothermal method. Cell culture study and In vivo assessments on C57BL/6 mice containing melanoma cancer cells was performed. RESULTS Results showed that CQD size slightly enhanced from 24.55 nm to 42.67 nm after ICG loading. Detection of reactive oxygen species (ROS) demonstrated that CQD improved ICG photo-stability and ROS generation capacity upon laser irradiation. Cell culture study illustrated that ICG@CQD could decrease survival rate of melanoma cancer cells of B16F10 cell line from 48% for pure ICG to 28% for ICG@CQD. Confocal microscopy images approved more cellular uptake and more qualified cell imaging ability of ICG@CQD. In vivo assessments displayed obvious inhibitory effect of tumor growth for ICG@CQD in comparison to free ICG on the C57BL/6 mice. In vivo fluorescence images confirmed that ICG@CQD accumulates remarkably more than free ICG in tumor region. Finally, ICG@CQD was proposed as an innovative nanocarrier for PDT and diagnosis.
Collapse
Affiliation(s)
- Hadiseh Mehravanfar
- Chemical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Nafiseh Farhadian
- Chemical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
47
|
Tillmanns J, Kicuntod J, Ehring A, Elbasani E, Borst EM, Obergfäll D, Müller R, Hahn F, Marschall M. Establishment of a Luciferase-Based Reporter System to Study Aspects of Human Cytomegalovirus Infection, Replication Characteristics, and Antiviral Drug Efficacy. Pathogens 2024; 13:645. [PMID: 39204245 PMCID: PMC11356942 DOI: 10.3390/pathogens13080645] [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: 05/28/2024] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Human cytomegalovirus (HCMV) represents a highly medically important pathogen which has constantly been the subject of both molecular and clinical investigations. HCMV infections, especially those in high-risk patients, still raise many unanswered questions, so current investigations are focused on viral pathogenesis, vaccine development, and options for antiviral drug targeting. To this end, the use of suitable viral strains as well as recombinant reporter constructs in cultured cells and model systems has specific significance. We previously reported on the application of various herpesviruses that express green, red, or related fluorescent proteins, especially in the fields of virus-host interaction and antiviral research. Here, we characterized a recombinant version of the clinically relevant and cell type-adaptable HCMV strain TB40, which expresses firefly luciferase as a quantitative reporter of viral replication (TB40-FLuc). The data provide evidence for five main conclusions. First, HCMV TB40-FLuc is employable in multiple settings in primary human cells. Second, viral reporter signals are easily quantifiable, even at early time points within viral replication. Third, the FLuc reporter reflects the kinetics of viral intracellular replication, cascade-like viral IE-E-L protein production, and progeny release. Fourth, as relates to specific applications of the TB40-FLuc system, we demonstrated the reliability of quantitative antiviral compound determination in multi-well formats and its independence from fluorescence-based measurements in the case of autofluorescent inhibitors. Finally, we illustrated increased reporter sensitivity in comparison to other recombinant HCMVs. In essence, recombinant HCMV TB40-FLuc combines several molecular properties that are considered beneficial in studies on viral host tropism, replication efficiency, and antiviral drug assessment.
Collapse
Affiliation(s)
- Julia Tillmanns
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.T.); (J.K.); (A.E.); (D.O.); (R.M.); (F.H.)
| | - Jintawee Kicuntod
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.T.); (J.K.); (A.E.); (D.O.); (R.M.); (F.H.)
| | - Antonia Ehring
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.T.); (J.K.); (A.E.); (D.O.); (R.M.); (F.H.)
| | - Endrit Elbasani
- Institute of Virology, Hannover Medical School (MHH), 30625 Hannover, Germany; (E.E.); (E.M.B.)
| | - Eva Maria Borst
- Institute of Virology, Hannover Medical School (MHH), 30625 Hannover, Germany; (E.E.); (E.M.B.)
| | - Debora Obergfäll
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.T.); (J.K.); (A.E.); (D.O.); (R.M.); (F.H.)
| | - Regina Müller
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.T.); (J.K.); (A.E.); (D.O.); (R.M.); (F.H.)
| | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.T.); (J.K.); (A.E.); (D.O.); (R.M.); (F.H.)
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (J.T.); (J.K.); (A.E.); (D.O.); (R.M.); (F.H.)
| |
Collapse
|
48
|
Neves C, Paz JD, Abbadi BL, Rambo RS, Czeczot AM, Sperotto NDM, Dadda AS, Silva RBM, Perelló MA, Gonçalves GA, González LC, Bizarro CV, Machado P, Basso LA. 5-Fluoroindole Reduces the Bacterial Burden in a Murine Model of Mycobacterium tuberculosis Infection. ACS OMEGA 2024; 9:32969-32979. [PMID: 39100312 PMCID: PMC11292626 DOI: 10.1021/acsomega.4c03981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/14/2024] [Accepted: 07/10/2024] [Indexed: 08/06/2024]
Abstract
Tuberculosis is a disease caused by a single pathogen that leads to a death toll estimated to be more than a million per year. Mycobacterium tuberculosis (Mtb), which affects mainly the lungs, spreads by airborne transmission when infectious respiratory particles from an infected human enter the respiratory tract of another person. Despite diagnosis and treatment being well established, the rise of cases of patients infected with Mtb strains with multidrug resistance to the antibiotics used in the regimen against the disease is alarming. Indole used as a core molecule has been described as a promising structure to treat several diseases. 5-Fluoroindole (5-FI) compound, evaluated in the free base and in the hydrochloride (5-FI.HCl) forms, inhibited the growth of pan-sensitive Mtb H37Rv strain in the same range (4.7-29.1 μM) of clinical isolates that have resistance to at least two first-line drugs. Although 5-FI showed no cytotoxicity in Vero and HepG2 cells, high permeability (2.4.10-6 cm/s) in the PAMPA assay, and high metabolic stability (Clint 9.0 mL/min/kg) in rat liver microsomes, limited solubility at plasmatic and intestinal pH values prompted formation and employment of its salt form (5-FI.HCl). Although the 5-FI.HCl compound showed increased solubility at pH values of 7.4 and 9.1 and increased stability in aqueous solutions, data for intrinsic clearance (Clint = 48 mL/min/kg) and a half-life (t 1/2 = 12 min) showed decreased metabolic stability. As 5-FI.HCl showed both good absorption and ability to reach the systemic circulation of animals without the need to use vehicles containing cosolvents or surfactants, it was chosen to evaluate its effectiveness in the model of tuberculosis in mice. The in vivo results showed the concentration of the compound in plasma increasing within 30 min in the systemic circulation and the capacity of reducing the Mtb burden in the lungs at the concentration of 200 μmol/kg after 21 days of infection, with no toxicity in mice.
Collapse
Affiliation(s)
- Christiano
E. Neves
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do
Rio Grande do Sul, 90616-900 Porto Alegre, Rio
Grande do Sul, Brazil
| | - Josiane D. Paz
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do
Rio Grande do Sul, 90616-900 Porto Alegre, Rio
Grande do Sul, Brazil
| | - Bruno L. Abbadi
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
| | - Raoní S. Rambo
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
| | - Alexia M. Czeczot
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do
Rio Grande do Sul, 90616-900 Porto Alegre, Rio
Grande do Sul, Brazil
| | - Nathalia D. M. Sperotto
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
| | - Adilio S. Dadda
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
| | - Rodrigo B. M. Silva
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
| | - Marcia A. Perelló
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
| | - Guilherme A. Gonçalves
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Medicina e Ciências
da Saúde, Pontifícia Universidade
Católica do Rio Grande do Sul, 90616-900 Porto Alegre, Rio Grande do Sul, Brazil
| | - Laura C. González
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do
Rio Grande do Sul, 90616-900 Porto Alegre, Rio
Grande do Sul, Brazil
| | - Cristiano V. Bizarro
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do
Rio Grande do Sul, 90616-900 Porto Alegre, Rio
Grande do Sul, Brazil
| | - Pablo Machado
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do
Rio Grande do Sul, 90616-900 Porto Alegre, Rio
Grande do Sul, Brazil
- Programa
de Pós-Graduação em Medicina e Ciências
da Saúde, Pontifícia Universidade
Católica do Rio Grande do Sul, 90616-900 Porto Alegre, Rio Grande do Sul, Brazil
| | - Luiz A. Basso
- Instituto
Nacional de Ciência e Tecnologia em Tuberculose, Centro de
Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, 90616-900 Porto
Alegre, Rio Grande do Sul, Brazil
- Programa
de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do
Rio Grande do Sul, 90616-900 Porto Alegre, Rio
Grande do Sul, Brazil
- Programa
de Pós-Graduação em Medicina e Ciências
da Saúde, Pontifícia Universidade
Católica do Rio Grande do Sul, 90616-900 Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
49
|
Adriana M, Aleksandra M, Denise B, Kinga G, Joanna W, Aleksandra H, Robert W, Agnieszka MW, Magdalena RŻ. Zn(II) and Cu(II) Coordination Enhances the Antimicrobial Activity of Piscidin 3, but Not That of Piscidins 1 and 2. Inorg Chem 2024; 63:12958-12968. [PMID: 38946498 PMCID: PMC11256756 DOI: 10.1021/acs.inorgchem.4c01659] [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: 04/23/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024]
Abstract
Piscidins, antimicrobial peptides isolated from fish, are potent against a variety of human pathogens; they show minimum inhibitory concentration values comparable to those of commercially used antimicrobials. Piscidins 1 and 2 are generally more effective than piscidin 3 when applied alone; the contrary is observed for their metal complexes: Zn(II) and Cu(II) coordination does not enhance the efficacy of piscidins 1 and 2, while a moderate enhancement is observed for piscidin 3. All three piscidins bind Cu(II) in a so-called albumin-like binding mode, while for Zn(II) complexes, two coordination modes are observed: piscidins 1 and 2 bind Zn(II) by imidazole nitrogens from His4, His11, and His17 side chains; piscidin 3 coordinates Zn(II) by His3, His4, and His11 imidazole nitrogens and additionally supports the interaction, formed by carbonyl oxygen from His4. Most likely, the high antimicrobial activity of piscidin complexes is due to neither the stability of their complexes nor the change in their secondary structure. Copper(II) complexes with piscidins 1 and 2 can form hydroxyl radicals, which could be responsible for the antimicrobial membrane damaging activity of these complexes. Clearly, a different mechanism (most likely an intercellular targeted one) is observed for piscidin 3 metal complexes; in most cases, the coordination of Cu(II) and Zn(II) enhances the antimicrobial potency of piscidin 3, showing that not only piscidin 3 alone but also its metal complexes have a different mode of action than piscidins 1 and 2.
Collapse
Affiliation(s)
- Miller Adriana
- Faculty
of Chemistry, University of Wroclaw, ul. F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Mikołajczyk Aleksandra
- Screening
of Biological Activity Assays and Collection of Biological Material
Laboratory, Wroclaw Medical University Biobank, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211a, 50-556 Wroclaw, Poland
| | - Bellotti Denise
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Garstka Kinga
- Faculty
of Chemistry, University of Wroclaw, ul. F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Wątły Joanna
- Faculty
of Chemistry, University of Wroclaw, ul. F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Hecel Aleksandra
- Faculty
of Chemistry, University of Wroclaw, ul. F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Wieczorek Robert
- Faculty
of Chemistry, University of Wroclaw, ul. F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Matera-Witkiewicz Agnieszka
- Screening
of Biological Activity Assays and Collection of Biological Material
Laboratory, Wroclaw Medical University Biobank, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211a, 50-556 Wroclaw, Poland
| | | |
Collapse
|
50
|
Ango PY, Ghoda AW, Abega DF, Mapitse R, Kuete V, Ngadjui BT, Fotso GW, Kapche DWFG. Aframomumlabdane, a new bislabdane diterpenoid from Aframomum arundinaceum (zingiberaceae). Nat Prod Res 2024:1-9. [PMID: 38976503 DOI: 10.1080/14786419.2024.2372847] [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: 02/06/2024] [Accepted: 06/23/2024] [Indexed: 07/10/2024]
Abstract
A previously undescribed bislabdane diterpenoid namely aframomumlabdane (1), was isolated from the seed of Aframomum arundinaceum together with seven known compounds (2 - 8). Their structures were established based on a comprehensive analysis of HR-ESI-MS, in conjunction with their 1D and 2D-NMR data. Compound 1 was evaluated for its cytotoxic activity against four cancer cell lines: A549, HepG2, SPC212 and DLD-1. The best activity was observed against SPC212 lung cancer cell line with an IC50 value of 0.52 μM.
Collapse
Affiliation(s)
- Patrick Y Ango
- Department of Pharmacotoxicology, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Arlette W Ghoda
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Destaing F Abega
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Renameditswe Mapitse
- Department of Chemistry, Faculty of Science, University of Botswana, Gaborone, Botswana
| | - Victor Kuete
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Bonaventure T Ngadjui
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Ghislain W Fotso
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Deccaux W F G Kapche
- Department of Chemistry, Higher Teacher Training College, University of Yaoundé I, Yaoundé, Cameroon
| |
Collapse
|