1
|
Yadav M, Mallappa RH, Ambatipudi K. Human milk fat globule delivers entrapped probiotics to the infant's gut and acts synergistically to ameliorate oxidative and pathogenic stress. Food Chem 2025; 462:141030. [PMID: 39241685 DOI: 10.1016/j.foodchem.2024.141030] [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: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
The human milk fat globule membrane (hMFGM) and Lactobacillus modulate the infant's gut and benefit health. Hence, the current study assesses the probiotic potential of Lactiplantibacillus plantarum (MRK3), Limosilactobacillus ferementum (MK1) isolated from infant feces, and its interaction with hMFGM during conditions mimicking infant digestive tract. Both strains showed high tolerance to gastrointestinal conditions, cell surface hydrophobicity, and strong anti-pathogen activity against Staphylococcus aureus. During digestion, hMFGM significantly exhibited xanthine oxidase activity, membrane roughness, and surface topography. In the presence of hMFGM, survival of MRK3 was higher than MK1, and electron microscopic observation revealed successful entrapment of MRK3 in the membrane matrix throughout digestion. Interestingly, probiotic-membrane matrix interaction showed significant synergy to alleviate oxidative stress and damage induced by cell-free supernatant of Escherichia coli in Caco-2 cells. Our results show that a probiotic-encapsulated membrane matrix potentially opens the functional infant formula development pathway.
Collapse
Affiliation(s)
- Monica Yadav
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Rashmi Hogarehalli Mallappa
- Molecular Biology Unit, Dairy Microbiology Division, Indian Council of Agriculture Research-National Dairy Research Institute, Karnal 132001, India
| | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India.
| |
Collapse
|
2
|
Ashokkumar M, Palanisamy K, Ganesh Kumar A, Muthusamy C, Senthil Kumar KJ. Green synthesis of silver and copper nanoparticles and their composites using Ocimum sanctum leaf extract displayed enhanced antibacterial, antioxidant and anticancer potentials. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:438-448. [PMID: 39239690 DOI: 10.1080/21691401.2024.2399938] [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: 06/11/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Green-synthesized silver and copper nanoparticles (NPs), along with their composites, exhibit various biological activities. Ocimum sanctum (Holy basil), traditionally used as medicine in South Asia, treats respiratory disorders, digestive issues, skin diseases and inflammatory conditions. Modern scientific studies support these bioactivities; however, no studies have investigated their bioactivity in combination with NPs. In this study, silver and copper NPs were synthesized using AgNO3 and CuSO4·5H2O solutions, respectively, with Ocimum sanctum leaf extract, and their antibacterial, antioxidant and anticancer properties were examined. Spectroscopic analyses, including Fourier transform infra-red (FTIR), transmission electron microscopy (TEM) and X-ray diffraction (XRD), elucidated the physicochemical characteristics of the green-synthesized nanoparticles (Os-AgNPs and Os-CuNPs), revealing sizes of 11.7 and 13.1 nm, respectively. The Os-AgNPs:Os-CuNPs nano-composite with a 1:2 ratio exhibited a zone of inhibition ranging from 8 to 12 mm against tested bacterial pathogens. Additionally, the NPs and their composites demonstrated potent antioxidant activity, with notable 2-diphenyl-2-picrylhydrazyl (DPPH) scavenging activity observed in composites with ratios of 2:1 and 1:2. Furthermore, they displayed potential anticancer activity against human leukaemia (Jurkat) cancer cells. Although no distinct difference in anticancer property was observed among the NPs and their composites, our study highlights their well-defined nanostructure and significant biological activity, suggesting their potential as therapeutic agents in the pharmaceutical industry.
Collapse
Affiliation(s)
- M Ashokkumar
- Department of Physics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Thandalam, India
| | - K Palanisamy
- Department of Chemistry, Srinivasan College of Arts and Science(Affiliated to Bharathidasan University, Tiruchirappalli), Perambalur, India
| | - A Ganesh Kumar
- Department of Microbiology, Centre for Research and Development, Hindustan College of Arts & Science, Kelambakkam, India
| | - C Muthusamy
- Department of Biotechnology, Srinivasan College of Arts and Science (Affiliated to Bharathidasan University, Tiruchirappalli), Perambalur, India
| | - K J Senthil Kumar
- Center for General Education, National Chung Hsing University, Taichung, Taiwan
- Bachelor Program of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
3
|
Pacyga-Prus K, Sandström C, Šrůtková D, Schwarzer M, Górska S. Phosphorylation-dependent immunomodulatory properties of B.PAT polysaccharide isolated from Bifidobacterium animalis ssp. animalis CCDM 218. Carbohydr Polym 2024; 344:122518. [PMID: 39218543 DOI: 10.1016/j.carbpol.2024.122518] [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: 03/18/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024]
Abstract
A wide range of articles describe the role of different probiotics in the prevention or treatment of various diseases. However, currently, the focus is shifting from whole microorganisms to their easier-to-define components that can confer similar or stronger benefits on the host. Here, we aimed to describe polysaccharide B.PAT, which is a surface antigen isolated from Bifidobacterium animalis ssp. animalis CCDM 218 and to understand the relationship between its structure and function. For this reason, we determined its glycerol phosphate-substituted structure, which consists of glucose, galactose, and rhamnose residues creating the following repeating unit: To fully understand the role of glycerol phosphate substitution on the B.PAT function, we prepared the dephosphorylated counterpart (B.MAT) and tested their immunomodulatory properties. The results showed that the loss of glycerol phosphate increased the production of IL-6, IL-10, IL-12, and TNF-α in bone marrow dendritic cells alone and after treatment with Lacticaseibacillus rhamnosus GG. Further studies indicated that dephosphorylation can enhance B.PAT properties to suppress IL-1β-induced inflammatory response in Caco-2 and HT-29 cells. Thus, we suggest that further investigation of B.PAT and B.MAT may reveal distinct functionalities that can be exploited in the treatment of various diseases and may constitute an alternative to probiotics.
Collapse
Affiliation(s)
- Katarzyna Pacyga-Prus
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Corine Sandström
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Box 7015, SE-750 07 Uppsala, Sweden.
| | - Dagmar Šrůtková
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| |
Collapse
|
4
|
Wei LY, Lin YW, Luo JC, Li YX, Hu YT, Guo SY, Jiang Z, Zhao DD, Chen SB, Huang ZS. Design, synthesis and structure-activity relationship of novel 2-pyrimidinylindole derivatives as orally available anti-obesity agents. Eur J Med Chem 2024; 277:116773. [PMID: 39163779 DOI: 10.1016/j.ejmech.2024.116773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Due to the emerging global epidemic of obesity, developing safe and effective agents for anti-obesity is urgently needed. Our previous study found that 2-pyrimidinylindole derivative Wd3d exhibited potential anti-obesity activity. Herein, to further optimize the potential moiety, structural modifications were proceeded for two rounds in this study. Firstly, we designed, synthesized, and evaluated 36 new derivatives of 2-pyrimidinylindole scaffold with different substituents on the indole ring and pyrimidine ring to investigate their structure-activity relationship (SAR). Then, analogs with potent activity had the aldehyde group replaced with the acylhydrazone group to reduce cytotoxicity and improve metabolic stability. Detailed SAR studies and animal evaluation experiments led to the discovery of the compound 9ga, which significantly reduced TG accumulation with an EC50 value of 0.07 μM and showed relatively low cytotoxicity with an IC50 value of around 24 μM. Oral administration of 9ga effectively prevented the excessive growth of body weight and lessened fat mass as well as liver mass, decreased lipid accumulation in the liver and blood, and improved the heart injury parameter in the diet-induced obesity mouse model significantly better than Wd3d. A mechanism study showed that 9ga regulated the lipid metabolism during early adipogenesis by inhibiting PPARγ pathway. In conclusion, our study further highlights the anti-obesity potential of 2-pyrimidinylindole derivatives in diet-induced obesity.
Collapse
Affiliation(s)
- Li-Yuan Wei
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yu-Wei Lin
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jia-Chun Luo
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yi-Xian Li
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yu-Tao Hu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shi-Yao Guo
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhi Jiang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Dan-Dan Zhao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shuo-Bin Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Zhi-Shu Huang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
5
|
Janssens LK, Stove CP. The 'ABC' of split-nanoluciferase HIF heterodimerization bioassays: Applications, Benefits & Considerations. Biochem Pharmacol 2024; 229:116478. [PMID: 39128589 DOI: 10.1016/j.bcp.2024.116478] [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: 04/09/2024] [Revised: 07/10/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Hypoxia-inducible factors (HIF) are interesting targets for multiple therapeutic indications. While HIF activation is desired for the treatment of anemia-related and ischemic diseases, HIF inhibition is of tremendous interest to anti-cancer drug development. Different signaling events within the HIF pathway are being targeted by drug discovery programs, with a special interest in HIF-selective (possibly also HIF1/2 isoform-selective) compounds. In this study, we applied recently developed cell-based split-nanoluciferase HIF heterodimerization assays to study the effects of compounds, targeting HIF activity by various mechanisms of action. This study shows that the application of similar or diverse assay protocols allows to detect various influences on HIF heterodimerization as a key signaling event in the oxygen sensing pathway: increased HIF heterodimerization (roxadustat, MG-132), decreased HIF heterodimerization (PX-478, ibuprofen) and direct (HIF isoform-selective) heterodimerization inhibiting effects (PT-2385). Changes in treatment time and in the assay protocol allowed to assess direct and indirect effects on HIFα-HIFβ heterodimerization. In addition to the evaluation of applications of these new bioassays regarding pharmacological characterizations, benefits and considerations are discussed related to the use of cellular, luminescent-based bioassays. Briefly, benefits include the bidirectional nature of the biological readout, the upstream mechanism of detection, the differentiation between HIF1 and HIF2 effects and the simulation of various conditions. Specific and general considerations include cell-based, technical and disease/drug-related aspects (e.g., non-specific effects, color interference). In summary, the versatility of these bioassays offers benefits in widespread applications regarding drug discovery and pharmacological characterization of various therapeutics, applying either the same or optimized experimental protocols.
Collapse
Affiliation(s)
- Liesl K Janssens
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Christophe P Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
6
|
Wang Y, Zhao J, Xu Y, Miao J, Pan K, Li Y, Chen Y, Liu X, Zhao A, Qin J, Xu T, Fang M. Benzo(a)anthracene Targeting SLC1A5 to Synergistically Enhance PAH Mixture Toxicity. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:18619-18630. [PMID: 39373333 DOI: 10.1021/acs.est.4c07053] [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: 10/08/2024]
Abstract
Human exposure to polycyclic aromatic hydrocarbons (PAHs) as mutagenic and carcinogenic pollutants in the environment often occurs in the form of mixtures. Although the mixture effects of PAHs have been previously recognized, the toxicological mechanisms to explain them still remain quite unclear. This study combined metabolomics and chemical proteomics methods to comprehensively understand the mixture effects of a PAH mixture including benzo(a)anthracene (BaA), benzo(b)fluoranthene (BbF), benzo(a)pyrene (BaP), and chrysene (CHR). Among them, BaA has shown a strong synergistic effect with other PAHs. Interestingly, BaA alone is not a potent oxidative stress inducer in liver cells but dose-dependently amplifies oxidative damage caused by the PAH mixture. Global metabolomics analysis results revealed damage to the antioxidant glutathione synthesis, which was caused by the glutamine depletion caused by BaA in the mixture. Subsequently, the label-free chemical proteomics and cellular thermal shift analysis (CETSA) demonstrated that the PAH mixture altered the thermal shift of glutamine transporter SLC1A5. Furthermore, Western blotting and the isothermal titration calorimetry (ITC) interaction measurements showed nanomolar KD values between BaA and SLC1A5. Overall, this study showed that BaA synergistically contributed to PAH mixture induced oxidative damage by targeting SLC1A5 to inhibit glutamate transport into cells, resulting in the inhibition of glutathione synthesis.
Collapse
Affiliation(s)
- Yanwei Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Pharmacology, College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Jiahui Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yipeng Xu
- Department of Urology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Jing Miao
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Keyu Pan
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yihan Li
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Haining, Zhejiang 314400, P.R. China
| | - Yong Chen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xuesong Liu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ailin Zhao
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Jingyu Qin
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Tengfei Xu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Mingliang Fang
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| |
Collapse
|
7
|
Sallandt LL, Wolf CA, Schuster S, Enke H, Enke D, Wolber G, Niedermeyer THJ. Derivatization of Microcystins Can Increase Target Inhibition while Reducing Cellular Uptake. JOURNAL OF NATURAL PRODUCTS 2024. [PMID: 39427253 DOI: 10.1021/acs.jnatprod.4c00688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Microcystins, a large family of nonribosomal cyclic heptapeptides known for their hepatotoxicity, are among the best-studied cyanobacterial toxins. Recently, they have been discussed as leads for the development of anticancer drug substances. Their main mode-of-action is inhibition of the eukaryotic serine/threonine protein phosphatases 1 and 2A. Unlike many cytotoxins that can cross cell membranes by passive diffusion, microcystins depend on active uptake via organic anion transporting polypeptides 1B1 or 1B3. Both phosphatase inhibition and transportability strongly depend on the structure of the individual microcystin. Here, we present how chemical modification of positions 2 and 4 of the microcystin core structure can alter these two properties. Aiming to reduce transportability and increase phosphatase inhibition, we used pharmacophore modeling to investigate the phosphatase inhibition potential of microcystins derivatized with small molecules containing a variety of functional groups. The respective derivatives were synthesized using click chemistry. We discovered that some derivatized microcystins can address a yet undescribed subpocket of the protein phosphatase 1. The derivatized microcystins were tested for phosphatase 1 inhibition and cytotoxicity on transporter-expressing cell lines, revealing that target inhibition and transportability of microcystins can independently be influenced by the physicochemical properties, especially of the residue located in position 2 of the microcystin. Derivatization with small acids or amino acids resulted in microcystins with a favorable ratio of inhibition to transportability, making these derivatives potentially suitable for drug development.
Collapse
Affiliation(s)
- Laura L Sallandt
- Department of Pharmaceutical Biology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Clemens A Wolf
- Department of Pharmaceutical Chemistry (Molecular Drug Design), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | | | - Heike Enke
- Simris Biologics GmbH, 12489 Berlin, Germany
| | - Dan Enke
- Simris Biologics GmbH, 12489 Berlin, Germany
| | - Gerhard Wolber
- Department of Pharmaceutical Chemistry (Molecular Drug Design), Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Timo H J Niedermeyer
- Department of Pharmaceutical Biology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
8
|
Camargo-Ayala L, Prent-Peñaloza L, Osorio E, Camargo-Ayala PA, Jimenez CA, Zúñiga-Arbalti F, Brito I, Delgado GE, Gutiérrez M, Polo-Cuadrado E. Naphthyl-functionalized acetamide derivatives: Promising agents for cholinesterase inhibition and antioxidant therapy in Alzheimer's disease. Bioorg Chem 2024; 153:107896. [PMID: 39454497 DOI: 10.1016/j.bioorg.2024.107896] [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: 06/06/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
This study presents the synthesis and characterization of a series of 13 novel acetamides. These were subjected to Ellman's assay to determine the efficacy of the AChE and BChE inhibitors. Finally, we report their antioxidant activity as an alternative approach for the search for drugs to treat AD. These studies revealed that compounds 1a-1k and 2l-2m were obtained in moderate yield. Four amides (1h, 1j, 1k, and 2l) were selective for one of the enzymes (BChE); thus, those that inhibited BChE were more active than the positive control (galantamine) and showed better IC50 values (3.30-5.03 µM). The theoretical free binding energies calculated by MM-GBSA indicated that all inhibitors were more stable than rivastigmine, and the inhibition mechanisms involved the entire active site: peripheral anionic site, oxyanion hole, acyl-binding pockets, and catalytic site. We examined the cytotoxicity of compounds 1h, 1j, 1k, and 2l in human dermal cells and found that they did not exhibit any toxic effects under the tested conditions. Additionally, these compounds, which also inhibited BChE, displayed mixed inhibition and did not exhibit hemolytic effects on human erythrocytes. Furthermore, the ABTS and DPPH assays indicated that, although none of the compounds showed activity in the DPPH assay, the EC50 values for radical trapping by the ABTS method showed that compounds 1a, 1d, 1e, and 1g had EC50 values lower than 10 µg/mL, indicating their strong radical scavenging capacity. We also report the crystal structures of compounds 1c, 1d, 1f, and 1g, which are found in monoclinic crystal systems.
Collapse
Affiliation(s)
- Lorena Camargo-Ayala
- Doctorado en Ciencias Mención I + D de Productos Bioactivos, Instituto de Química de Recursos Naturales, Laboratorio de Síntesis Orgánica, Universidad de Talca, Casilla 747, Talca 3460000, Chile.
| | - Luis Prent-Peñaloza
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andrés Bello, Quillota 980, Viña del Mar, Chile
| | - Edison Osorio
- Facultad de Ciencias Naturales y Matemáticas, Universidad de Ibagué, Carrera 22, Calle 67, Ibagué 730001, Colombia
| | - Paola Andrea Camargo-Ayala
- Doctorado en Ciencias Biomédicas, Laboratorio de Patología Molecular, Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile
| | - Claudio A Jimenez
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción 4130000, Chile
| | - Felipe Zúñiga-Arbalti
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Víctor Lamas 1290, Concepción 4030000, Chile
| | - Iván Brito
- Departamento de Química, Facultad de Ciencias Básicas, Universidad de Antofagasta, Avda., Universidad de Antofagasta, Campus Coloso, Antofagasta 02800, Chile
| | - Gerzon E Delgado
- Departamento de Química, Facultad de Ciencias Básicas, Universidad de Antofagasta, Avda., Universidad de Antofagasta, Campus Coloso, Antofagasta 02800, Chile; Laboratorio de Cristalografía, Departamento de Química, Facultad de Ciencias, Universidad de Los Andes, Mérida 5101, Venezuela
| | - Margarita Gutiérrez
- Laboratorio de Síntesis Orgánica y Actividad Biológica (LSO-Act-Bio), Universidad de Talca, Casilla 747, Talca 3460000, Chile.
| | - Efraín Polo-Cuadrado
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción 4130000, Chile.
| |
Collapse
|
9
|
Blanco Carcache PJ, Castro-Dionicio IY, Mirtallo Ezzone NP, Salinas-Arrellano ED, Bahar J, Clinton SK, Kinghorn AD. Molecular Networking, Docking, and Biological Evaluation of Licarin A from Myristica fragrans as a Potential Cancer Chemopreventive Agent. Molecules 2024; 29:4919. [PMID: 39459287 PMCID: PMC11510329 DOI: 10.3390/molecules29204919] [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: 08/30/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Currently, clinically available cancer chemopreventive drug options are limited to mostly tamoxifen and its derivatives, such as raloxifene, and approved specifically for breast cancer. Thus, the availability of chemopreventive drug molecules for other types of malignant cancers would be desirable. In previous reports, the arils of Myristica fragrans (mace) have been found to exhibit cancer chemopreventive activity. Therefore, the purpose of the present study was to identify a natural product from this species with potential chemopreventive activity guided by chemoinformatic sample analysis via Global Natural Products Social (GNPS) molecular networking and molecular docking. The neolignan licarin A (1) was identified as a potential chemopreventive constituent, and subsequently submitted to several in vitro bioassays and a zebrafish toxicity evaluation. In this work, 1 afforded superior phosphoNF-κBp65 phosphorylation activity in DU-145 prostate cancer cells compared to isoliquiritigenin (2), which was used as a natural product chemopreventive control. Both 1 and 2 showed a longer-lasting reduction in cellular stress in a cell oxidative stress real-time dose-response assay than the positive control using Hepa1c1c7 mouse hepatoma cells. In addition, 1 displayed similar activities to 2, while also being less toxic to zebrafish (Danio rerio) than both this chalcone and the clinically used chemopreventive drug tamoxifen.
Collapse
Affiliation(s)
- Peter J. Blanco Carcache
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (I.Y.C.-D.); (N.P.M.E.); (E.D.S.-A.); (J.B.)
| | - Ines Y. Castro-Dionicio
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (I.Y.C.-D.); (N.P.M.E.); (E.D.S.-A.); (J.B.)
| | - Nathan P. Mirtallo Ezzone
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (I.Y.C.-D.); (N.P.M.E.); (E.D.S.-A.); (J.B.)
| | - Eric D. Salinas-Arrellano
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (I.Y.C.-D.); (N.P.M.E.); (E.D.S.-A.); (J.B.)
| | - Joshua Bahar
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (I.Y.C.-D.); (N.P.M.E.); (E.D.S.-A.); (J.B.)
| | - Steven K. Clinton
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
- Division of Medical Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - A. Douglas Kinghorn
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (I.Y.C.-D.); (N.P.M.E.); (E.D.S.-A.); (J.B.)
| |
Collapse
|
10
|
Saraf P, Bhardwaj B, Verma A, Siddiqui MA, Verma H, Kumar P, Srivastava S, Krishnamurthy S, Srikrishna S, Shrivastava SK. Design, synthesis, and evaluation of benzhydrylpiperazine-based novel dual COX-2/5-LOX inhibitors with anti-inflammatory and anti-cancer activity. RSC Med Chem 2024:d4md00471j. [PMID: 39430948 PMCID: PMC11487423 DOI: 10.1039/d4md00471j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024] Open
Abstract
Piperazine derivatives were screened using the ChEMBL database, paving the way for the design, synthesis, and evaluation of a novel series of dual COX-2/5-LOX inhibitors and identifying their role in mitigating cancer cell proliferation. Compound 9d with 4-Cl substitution at the terminal phenyl ring showed promising inhibition of COX-2 (IC50 = 0.25 ± 0.03 μM) and 5-LOX (IC50 = 7.87 ± 0.33 μM), outperforming the standards celecoxib (IC50 = 0.36 ± 0.023 μM) and zileuton (IC50 = 14.29 ± 0.173 μM), respectively. The two most active derivatives 9d and 9g indicated a significant anti-inflammatory response in a paw edema model by inhibiting PGE2, IL-6, and TNF-α and an increase in IL-10 concentrations. Interestingly, 9d effectively reduced pain by 55.78%, closely comparable to the 59.09% exhibited by the standard indomethacin, and was also devoid of GI, liver, kidney, and cardiac toxicity. Furthermore, 9d demonstrated anti-cancer potential against in vitro A549, COLO-205, and MIA-PA-CA-2 human cancer cell lines and an in vivo Drosophila cancer model. The pharmacokinetic investigations revealed that 9d has good oral absorption characteristics.
Collapse
Affiliation(s)
- Poorvi Saraf
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Bhagwati Bhardwaj
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Akash Verma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Mohammad Aquib Siddiqui
- Pharmacology Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India
| | - Himanshu Verma
- Pharmacology Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India
| | - Pradeep Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University Varanasi-221005 India
| | - Samridhi Srivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| | - Sairam Krishnamurthy
- Pharmacology Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India
| | - Saripella Srikrishna
- Department of Biochemistry, Institute of Science, Banaras Hindu University Varanasi-221005 India
| | - Sushant Kumar Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi-221005 India +91 945 2156 527
| |
Collapse
|
11
|
Piacente F, Guccione G, Scarano N, Lunaccio D, Miro C, Abbotto E, Salis A, Tasso B, Dentice M, Bruzzone S, Cichero E, Millo E. Discovery of Novel Thiazole-Based SIRT2 Inhibitors as Anticancer Agents: Molecular Modeling, Chemical Synthesis and Biological Assays. Int J Mol Sci 2024; 25:11084. [PMID: 39456864 PMCID: PMC11508362 DOI: 10.3390/ijms252011084] [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: 09/12/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The search and development of effective sirtuin small molecule inhibitors (SIRTIs) continues to draw great attention due to their wide range of pharmacological applications. Based on SIRTs' involvement in different biological pathways, their ligands were investigated for many diseases, such as cancer, neurodegenerative disorders, diabetes, cardiovascular diseases and autoimmune diseases. The elucidation of a substantial number of SIRT2-ligand complexes is steering the identification of novel and more selective modulators. Among them, SIRT2 in the presence of the SirReal2 analog series was the most studied. On this basis, we recently reported structure-based analyses leading to the discovery of thiazole-based compounds acting as SIRT2 inhibitors (T1, SIRT2 IC50 = 17.3 µM). Herein, ligand-based approaches followed by molecular docking simulations allowed us to evaluate in silico a novel small series of thiazoles (3a-3d and 5a, 5d) as putative SIRT2 inhibitors. Results from the computational studies revealed comparable molecular interaction fields (MIFs) and docking positionings of most of these compounds with respect to reference SIRT2Is. Biochemical and biological assays validated this study and pointed to compound 5a (SIRT2 IC50 = 9.0 µM) as the most interesting SIRT2I that was worthy of further development as an anticancer agent.
Collapse
Affiliation(s)
- Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (G.G.); (D.L.); (E.A.); (A.S.); (E.M.)
| | - Giorgia Guccione
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (G.G.); (D.L.); (E.A.); (A.S.); (E.M.)
| | - Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (B.T.)
| | - Dario Lunaccio
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (G.G.); (D.L.); (E.A.); (A.S.); (E.M.)
| | - Caterina Miro
- Department of Clinical Medicine & Surgery, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy; (C.M.); (M.D.)
| | - Elena Abbotto
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (G.G.); (D.L.); (E.A.); (A.S.); (E.M.)
| | - Annalisa Salis
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (G.G.); (D.L.); (E.A.); (A.S.); (E.M.)
| | - Bruno Tasso
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (B.T.)
| | - Monica Dentice
- Department of Clinical Medicine & Surgery, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy; (C.M.); (M.D.)
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (G.G.); (D.L.); (E.A.); (A.S.); (E.M.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (B.T.)
| | - Enrico Millo
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (G.G.); (D.L.); (E.A.); (A.S.); (E.M.)
| |
Collapse
|
12
|
Gomes Daré R, Beatriz Chieco Costa A, Silva Martins T, Lopes LB. Simvastatin and adenosine-co-loaded nanostructured lipid carriers for wound healing: Development, characterization and cell-based investigation. Eur J Pharm Biopharm 2024:114533. [PMID: 39414092 DOI: 10.1016/j.ejpb.2024.114533] [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: 08/13/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Chronic wounds represent a significant global health burden, characterized by delayed skin healing and associated comorbidities. The present study aimed to develop nanostructured lipid carriers (NLCs) as a topical delivery system for the co-administration of simvastatin and adenosine to address chronic wound management. The rationale behind the co-delivery approach was to mitigate the cytotoxicity associated with high-dose simvastatin, while preserving its therapeutic benefits through a potential synergistic or additive effect. A significant challenge in the development of these NLCs was the encapsulation of the highly hydrophilic adenosine within the hydrophobic lipid matrix. The NLCs were prepared using a hot homogenization-sonication method with a double emulsion technique and optimized through a series of formulation trials, employing various surfactants, solid and liquid lipids, to achieve efficient drug encapsulation, particularly for the hydrophilic adenosine. Optimized formulations F5- and F10-S/A 0.6 %/2 % (containing 0.6 % simvastatin and 2 % adenosine), exhibited promising physicochemical properties. The main difference was the liquid lipid used: F5 containing Miglyol 810 N, while F10 Capmul MCM C-8. Both formulations displayed a mean particle size below 230 nm, a polydispersity index (PDI) of approximately 0.2, and a zeta potential of around -22 mV. While simvastatin association efficiency (AE) was nearly 100 %, adenosine AE was higher for F10 (24 %), compared to F5 (13.5 %). F5 demonstrated superior stability compared to F10, maintaining consistent particle size and PDI over a 60-day period. Formulation F5 also demonstrated superior cell-based in vitro performance compared to F10, with higher cell viability (MTT assay), greater cell proliferation induction (SRB assay), and enhanced cell proliferation and migration in the wound-scratch assay. While F10 displayed higher adenosine AE, F5 excelled in terms of stability and biological activity. The slightly increase in intracellular reactive oxygen species levels observed with F5 may contribute to its enhanced proliferative effects. In-depth characterization revealed that F5 comprised spherical nanoparticles, and thermal analysis indicated no significant changes in the nanocarrier structure upon drug encapsulation. Additionally, ex vivo permeability study demonstrated superior skin retention of both simvastatin and adenosine for F5 compared to an emulsion control. Overall, the F5 nanocarrier demonstrated suitable physicochemical properties, cellular biocompatibility, induction of cell proliferation and migration events, and drug retention capacity in the skin layers, indicating its potential as a promising topical treatment for difficult-to-heal wounds.
Collapse
Affiliation(s)
- Regina Gomes Daré
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil.
| | - Ana Beatriz Chieco Costa
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil
| | - Tereza Silva Martins
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, 210 São Nicolau Street, 09913-030 Diadema, SP, Brazil
| | - Luciana B Lopes
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil.
| |
Collapse
|
13
|
Woods PS, Cetin-Atalay R, Meliton AY, Sun KA, Shamaa OR, Shin KWD, Tian Y, Haugen B, Hamanaka RB, Mutlu GM. HIF-1α regulates mitochondrial function in bone marrow-derived macrophages, but not in tissue-resident alveolar macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618294. [PMID: 39464148 PMCID: PMC11507697 DOI: 10.1101/2024.10.14.618294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
HIF-1α plays a critical role in shaping macrophage phenotype and effector function. We have previously shown that tissue-resident alveolar macrophages (TR-AMs) have extremely low glycolytic capacity at steady-state, but can shift toward glycolysis under hypoxic conditions. Here, using inducible HIF-1α knockout ( Hif1a -/- ) TR-AMs and bone marrow-derived macrophages (BMDMs) and show that TR-AM HIF-1α is required for the glycolytic shift under prolyl hydroxylase inhibition, but is dispensable at steady-state for inflammatory effector function. In contrast, HIF-1α deletion in BMDMs led to diminished glycolytic capacity at steady-state and reduced inflammatory capacity, but higher mitochondrial function. Gene set enrichment analysis revealed enhanced c-Myc transcriptional activity in Hif1a -/- BMDMs, and upregulation of gene pathways related to ribosomal biogenesis and cellular proliferation. The findings highlight the heterogeneity of HIF-1α function in distinct macrophage populations and provide new insight into how HIF-1α regulates gene expression, inflammation, and metabolism in macrophages.
Collapse
|
14
|
Carlier B, Heymans SV, Collado-Lara G, Musetta L, Ingram M, Toumia Y, Paradossi G, Vos HJ, Roskams T, D'hooge J, Van Den Abeele K, Sterpin E, Himmelreich U. Phase-change ultrasound contrast agents for proton range verification: towards an in vivoapplication. Phys Med Biol 2024; 69:205014. [PMID: 39312948 DOI: 10.1088/1361-6560/ad7e76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
Abstract
Objective.In proton therapy, range uncertainties prevent optimal benefit from the superior depth-dose characteristics of proton beams over conventional photon-based radiotherapy. To reduce these uncertainties we recently proposed the use of phase-change ultrasound contrast agents as an affordable and effective range verification tool. In particular, superheated nanodroplets can convert into echogenic microbubbles upon proton irradiation, whereby the resulting ultrasound contrast relates to the proton range with high reproducibility. Here, we provide a firstin vivoproof-of-concept of this technology.Approach.First, thein vitrobiocompatibility of radiation-sensitive poly(vinyl alcohol) perfluorobutane nanodroplets was investigated using several colorimetric assays. Then,in vivoultrasound contrast was characterized using acoustic droplet vaporization (ADV) and later using proton beam irradiations at varying energies (49.7 MeV and 62 MeV) in healthy Sprague Dawley rats. A preliminary evaluation of thein vivobiocompatibility was performed using ADV and a combination of physiology monitoring and histology.Main results.Nanodroplets were non-toxic over a wide concentration range (<1 mM). In healthy rats, intravenously injected nanodroplets primarily accumulated in the organs of the reticuloendothelial system, where the lifetime of the generated ultrasound contrast (<30 min) was compatible with a typical radiotherapy fraction (<5 min). Spontaneous droplet vaporization did not result in significant background signals. Online ultrasound imaging of the liver of droplet-injected rats demonstrated an energy-dependent proton response, which can be tuned by varying the nanodroplet concentration. However, caution is warranted when deciding on the exact nanodroplet dose regimen as a mild physiological response (drop in cardiac rate, granuloma formation) was observed after ADV.Significance.These findings underline the potential of phase-change ultrasound contrast agents forin vivoproton range verification and provide the next step towards eventual clinical applications.
Collapse
Affiliation(s)
- Bram Carlier
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sophie V Heymans
- Department of Physics and Astronomy, KU Leuven KULAK, Kortrijk, Belgium
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | | | - Luigi Musetta
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Marcus Ingram
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Yosra Toumia
- Department of Chemical Sciences and Technology, Universita degli Studi di Roma Tor Vergata, Rome, Italy
- Istituto Nazionale di Fisica Nucleare (INFN) sez. di Roma Tor Vergata, Rome, Italy
| | - Gaio Paradossi
- Department of Chemical Sciences and Technology, Universita degli Studi di Roma Tor Vergata, Rome, Italy
| | - Hendrik J Vos
- Deparment of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tania Roskams
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jan D'hooge
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | | | - Edmond Sterpin
- Department of Oncology, KU Leuven, Leuven, Belgium
- Particle Therapy Interuniversity Center Leuven-PARTICLE, Leuven, Belgium
| | - Uwe Himmelreich
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Delgado-Waldo I, Dokudovskaya S, Loissell-Baltazar YA, Pérez-Arteaga E, Coronel-Hernández J, Martínez-Vázquez M, Pérez-Yépez EA, Lopez-Saavedra A, Jacobo-Herrera N, Pérez Plasencia C. Laherradurin Inhibits Colorectal Cancer Cell Growth by Induction of Mitochondrial Dysfunction and Autophagy Induction. Cells 2024; 13:1649. [PMID: 39404412 PMCID: PMC11475353 DOI: 10.3390/cells13191649] [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: 09/09/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
LAH, an acetogenin from the Annonaceae family, has demonstrated antitumor activity in several cancer cell lines and in vivo models, where it reduced the tumor size and induced programmed cell death. We focused on the effects of LAH on mitochondrial dynamics, mTOR signaling, autophagy, and apoptosis in colorectal cancer (CRC) cells to explore its anticancer potential. METHODS CRC cells were treated with LAH, and its effects on mitochondrial respiration and glycolysis were measured using Seahorse XF technology. The changes in mitochondrial dynamics were observed through fluorescent imaging, while Western blot analysis was used to examine key autophagy and apoptosis markers. RESULTS LAH significantly inhibited mitochondrial complex I activity, inducing ATP depletion and a compensatory increase in glycolysis. This disruption caused mitochondrial fragmentation, a trigger for autophagy, as shown by increased LC3-II expression and mTOR suppression. Apoptosis was also confirmed through the cleavage of caspase-3, contributing to reduced cancer cell viability. CONCLUSIONS LAH's anticancer effects in CRC cells are driven by its disruption of mitochondrial function, triggering both autophagy and apoptosis. These findings highlight its potential as a therapeutic compound for further exploration in cancer treatment.
Collapse
Affiliation(s)
- Izamary Delgado-Waldo
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico
| | - Svetlana Dokudovskaya
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (S.D.); (Y.A.L.-B.)
| | - Yahir A. Loissell-Baltazar
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (S.D.); (Y.A.L.-B.)
| | - Eduardo Pérez-Arteaga
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
| | - Jossimar Coronel-Hernández
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
| | - Mariano Martínez-Vázquez
- Instituto de Química, Universidad Nacional Autónoma de México, C. Exterior, C. Universitaria, Coyoacán, Ciudad de México 04510, Mexico;
| | - Eloy Andrés Pérez-Yépez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
| | - Alejandro Lopez-Saavedra
- Advanced Microscopy Applications Unit (ADMIRA), Instituto Nacional de Cancerología, San Fernando 22. Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey Ciudad de Mexico, C. Puente #222, Coapa, Arboledas del Sur, Tlalpan, Ciudad de Mexico 14380, Mexico
| | - Nadia Jacobo-Herrera
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
| | - Carlos Pérez Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México 54090, Mexico
| |
Collapse
|
16
|
Al-Ghanayem AA. In-vitro anti-acne activity of Teucrium oliverianum methanolic extract against Cutibacterium acnes. Front Pharmacol 2024; 15:1388625. [PMID: 39421673 PMCID: PMC11484032 DOI: 10.3389/fphar.2024.1388625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Background Acne vulgaris is a skin infection widely seen in adolescents between 10-19 years with males affected more than females. It mainly affects the face but may also affect the back and chest. The symptoms vary with mild acne manifesting as comedones and moderate acne as inflammatory lesions (papulopustular), nodules, and mild scarring while severe acne has the same symptoms that have not subsided within 6 months of treatment. Various treatments including topical medications containing different antibiotics are used to treat acne. Recently, herbal treatments have been shown as better alternatives to conventional treatment. Teucrium oliverianum Ging. ex Benth (Lamiaceae) is traditionally used for skin infections such as wound healing and biofilm formation. Methodology Methanolic extract of T. oliverianum was subjected to liquid chromatography-mass spectrometry (LC-MS) analysis, and its antibacterial effect against Cutibacterium acnes. The anti-acne, anti-inflammatory, and antioxidant effects were also assessed using HaCaT cells infected with C. acnes. The cytotoxicity of the extract was evaluated using a neutral red uptake assay, and anti-inflammatory effects were determined by measuring TNF-α, IL-1β, INF-γ, and COX2 inhibition. The antioxidant action was assessed by ROS generation in HaCaT cells infected with C. acnes. Results LC-MS analysis of the extract showed the presence of 16 different metabolites with L-carnitine, esculin sesquihydrate, and gamma-linoleic acid as major metabolites. The methanolic extract of T. oliverianum showed an antibacterial effect against C. acnes with an IC50 value of 263.2 μg/mL. The extract attenuated the cytotoxicity of C. acnes on the HaCaT cell and the IC50 was found to be 676.2 μg/mL. It also decreased dose-dependently the expression of TNF-α, IL-1β, INF-γ, and inhibited COX2 in the HaCaT cells infected with C. acnes. It also decreased the generation of reactive oxygen species. Conclusion The results support the use of T. oliverianum as an anti-acne agent but it possesses mild antibacterial action. It showed anti-inflammatory effects in HaCaT cells infected with C. acnes. It is also an effective antioxidant and decreased the generation of reactive oxygen species. Comparison of the anti-acne effects and adverse reactions of extract with other treatments will provide more insight into its clinical efficacy and toxicity.
Collapse
Affiliation(s)
- Abdullah A. Al-Ghanayem
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| |
Collapse
|
17
|
Kassem AF, Sediek AA, Omran MM, Foda DS, Al-Ashmawy AAK. Design, synthesis and in vitro anti-proliferative evaluation of new pyridine-2,3-dihydrothiazole/thiazolidin-4-one hybrids as dual CDK2/GSK3β kinase inhibitors. RSC Adv 2024; 14:31607-31623. [PMID: 39376524 PMCID: PMC11456921 DOI: 10.1039/d4ra06146b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
Herein, the molecular hybridization drug discovery approach was used in the design and synthesis of twelve novel pyridine-2,3-dihydrothiazole hybrids (2a,b-5a,b and 13a,b-14a,b) and fourteen pyridine-thiazolidin-4-one hybrids (6a,b-12a,b) as anti-proliferative analogues targeting CDK2 and GSK3β kinase inhibition. Almost all of the newly synthesized hybrids, including their precursors (1a,b), were evaluated for their anti-proliferative activity against three human cancer cell lines-MCF-7, HepG2 and HEp-2-as well as normal Vero cell lines. Both compounds 1a (pyridine-thiourea precursor) and 8a (pyridine-5-acetyl-thiazolidin-4-one hybrid) exhibited excellent anti-proliferative activity against HEp-2 (IC50 = 7.5 μg mL-1, 5.9 μg mL-1, respectively). Additionally, 13a (pyridine-5-(p-tolyldiazenyl-2,3-dihydrothiazole)) hybrid demonstrated excellent anti-proliferative activity against HepG2 (IC50 = 9.5 μg mL-1), with an acceptable safety profile against Vero (<45% inhibition at 100 μg mL-1) in the cases of 8a and 13a alone. The three promising anti-proliferative hybrids (1a, 8a, 13a) were selected for the assessment of their in vitro inhibitory kinase activity against CDK2/GSK3β using roscovitine (IC50 = 0.88 μg mL-1) and CHIR-99021 (IC50 = 0.07 μg mL-1) as references, respectively. Compound 13a was the most potent dual CDK2/GSK3β inhibitor (IC50 = 0.396 μg mL-1, 0.118 μg mL-1, respectively) followed by 8a (IC50 = 0.675 μg mL-1, 0.134 μg mL-1, respectively), and the weakest was 1a. To elucidate the mechanism of the most potent anti-proliferative 13a hybrid, further cell cycle analysis was performed revealing that it caused G1 cell cycle arrest and induced apoptosis. Moreover, it resulted in an increase in Bax and caspase-3 with a decrease in Bcl-2 levels in HepG2 cells compared with untreated cells. Finally, in silico drug likeness/ADME prediction for the three potent compounds as well as a molecular docking simulation study were conducted in order to explore the binding affinity and interactions in the binding site of each enzyme, which inspired their usage as anti-proliferative leads for further modification.
Collapse
Affiliation(s)
- Asmaa F Kassem
- Chemistry of Natural and Microbial Products Department, National Research Centre Dokki 12622 Cairo Egypt
| | - Ashraf A Sediek
- Chemical Industries Institute, National Research Centre Dokki 12622 Cairo Egypt
| | - Mervat M Omran
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University Cairo Egypt
| | - Doaa S Foda
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Cairo Egypt
| | - Aisha A K Al-Ashmawy
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Cairo Egypt
| |
Collapse
|
18
|
Sousa SM, Branco H, Avan A, Palmeira A, Morelli L, Santos LL, Giovannetti E, Vasconcelos MH, Xavier CPR. Darifenacin: a promising chitinase 3-like 1 inhibitor to tackle drug resistance in pancreatic ductal adenocarcinoma. Cancer Chemother Pharmacol 2024; 94:585-597. [PMID: 39225813 PMCID: PMC11438711 DOI: 10.1007/s00280-024-04712-1] [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: 04/23/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is among the most aggressive malignancies. Our previous work revealed Chitinase 3-like 1 (CHI3L1) involvement in PDAC resistance to gemcitabine, identifying it as a promising therapeutic target. Here, we aimed to identify putative CHI3L1 inhibitors and to investigate their chemosensitizing potential in PDAC. METHODS Docking analysis for CHI3L1 identified promising CHI3L1 inhibitors, including darifenacin (muscarinic receptor antagonist). PDAC cell lines (BxPC-3, PANC-1) and primary PDAC cells were used to evaluate darifenacin's effects on cell growth (Sulforhodamine B, SRB), alone or in combination with gemcitabine or gemcitabine plus paclitaxel. Cytotoxicity against normal immortalized pancreatic ductal cells (HPNE) was assessed. Recombinant protein was used to confirm the impact of darifenacin on CHI3L1-induced PDAC cellular resistance to therapy (SRB assay). Darifenacin's effect on Akt activation was analysed by ELISA. The association between cholinergic receptor muscarinic 3 (CHRM3) expression and therapeutic response was evaluated by immunohistochemistry of paraffin-embedded tissues from surgical resections of a 68 patients' cohort. RESULTS In silico screening revealed the ability of darifenacin to target CHI3L1 with high efficiency. Darifenacin inhibited PDAC cell growth, with a GI50 of 26 and 13.6 µM in BxPC-3 and PANC-1 cells, respectively. These results were confirmed in primary PDAC-3 cells, while darifenacin showed no cytotoxicity against HPNE cells. Importantly, darifenacin sensitized PDAC cells to standard chemotherapies, reverted CHI3L1-induced PDAC cellular resistance to therapy, and decreased Akt phosphorylation. Additionally, high CHMR3 expression was associated with low therapeutic response to gemcitabine. CONCLUSION This work highlights the potential of darifenacin as a chemosensitizer for PDAC treatment.
Collapse
Affiliation(s)
- Sofia M Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- LQOF - Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, 91886-17871, Iran
- Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 91886-17871, Iran
| | - Andreia Palmeira
- LQOF - Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros do Porto de Leixões, Matosinhos, 4450-208, Portugal
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, 56100, Italy
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics Research Group and Surgical Oncology Department, IPO-Instituto Português de Oncologia, Rua Dr. António Bernardino de Almeida 865, Porto, 4200-072, Portugal
- ICBAS-UP-School of Medicine and Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050- 313, Portugal
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, HV Amsterdam, 1081, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per La Scienza, San Giuliano, 56017, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal.
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, 4585-116, Portugal.
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, 4585-116, Portugal.
| |
Collapse
|
19
|
Geaneotes PJ, Janosko CP, Afeke C, Deiters A, Floreancig PE. Potent and Selective Oxidatively Labile Ether-Based Prodrugs through Late-Stage Boronate Incorporation. Angew Chem Int Ed Engl 2024; 63:e202409229. [PMID: 38986017 DOI: 10.1002/anie.202409229] [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: 05/15/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/12/2024]
Abstract
This manuscript describes a new strategy for prodrug synthesis in which a relatively inert ether group is introduced at an early stage in a synthetic sequence and functionalized in the final step to introduce a prodrug-activating group through a chemoselective process. Boryl allyloxy (BAO) ether groups are synthesized through several metal-mediated processes to form entities that are readily cleaved under oxidative conditions commonly found in cancer cells. The high cleavage propensity of the BAO group allows for ether cleavage, making these compounds substantially more hydrolytically stable in comparison to acyl-linked prodrugs while retaining the ability to release alcohols. We report the preparation of prodrug analogues of the natural products camptothecin and pederin from acetal precursors that serve as protecting groups in their synthetic sequences. The BAO acetal groups cleave in the presence of hydrogen peroxide to release the cytotoxic agents. The pederin-based prodrug shows dramatically greater cytotoxicity than negative controls and outstanding selectivity and potency toward cancer cell lines in comparison to non-cancerous cell lines. This late-stage functionalization approach to prodrug synthesis should be applicable to numerous systems that can be accessed through chemoselective processes.
Collapse
Affiliation(s)
- Paul J Geaneotes
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| | - Chasity P Janosko
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| | - Cephas Afeke
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| | - Paul E Floreancig
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| |
Collapse
|
20
|
Lindsay CA, Tan CY, Krishnan D, Uchenik D, Eugenio GDA, Salinas ED, de Blanco EJC, Kinghorn AD, Rakot Ondraibe HL. Steroids and Epicoccarines from Penicillium aurantiancobrunneum. PHYTOCHEMISTRY LETTERS 2024; 63:79-86. [PMID: 39280884 PMCID: PMC11391924 DOI: 10.1016/j.phytol.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Lichens are symbiotic organisms comprised of mycobionts and photobiont partners. They are known to produce bioactive secondary metabolites and most of these are biosynthesized by mycobionts. Investigations of cultures of isolated lichen-associated fungi have shown promise for the discovery of cytotoxic compounds. Thus, the lichen-associated fungus Penicillium aurantiacobrunneum was studied for its potential to produce novel compounds and the new sterols (20S*)-hydroxy-24(28)-dehydrocampesterol (1), 7α-methoxy-8β-hydroxypaxisterol (2), 14-nor-epicoccarine A (3) and 14-nor-epicoccarine B (4), as well as the known compound PF1140 (5), were isolated. The structures of these compounds were elucidated using methods including nuclear magnetic resonance (NMR) spectroscopy and high-resolution electrospray ionization mass spectrometry (HRESIMS). Following cytotoxicity assays, compound 1 demonstrated activity against the pancreatic adenocarcinoma epithelial HPAC cell line at 17.76 ± 5.35 μM. Since the structures of compounds 3 and 4 were very similar to tetramic acid derivatives that were reported to be biosynthesized from a polyketide synthase- non-ribosomal peptide synthetase (PKS-NRPS) hybrid pathway, a plausible biosynthetic route for production in P. aurantiacobrunneum is proposed herein.
Collapse
Affiliation(s)
- Charmaine A Lindsay
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Choon Y Tan
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Deepa Krishnan
- Instrumentation Facility, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Dmitriy Uchenik
- Instrumentation Facility, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Gerardo D Anaya Eugenio
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Instrumentation Facility, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Eric D Salinas
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Instrumentation Facility, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Esperanza J Carcache de Blanco
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | | |
Collapse
|
21
|
Arabiyat S, Alzoubi A, Al-Daghistani H, Al-Hiari Y, Kasabri V, Alkhateeb R. Evaluation of Quinoline-Related Carboxylic Acid Derivatives as Prospective Differentially Antiproliferative, Antioxidative, and Anti-Inflammatory Agents. Chem Biol Drug Des 2024; 104:e14615. [PMID: 39358207 DOI: 10.1111/cbdd.14615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 07/27/2024] [Accepted: 08/17/2024] [Indexed: 10/04/2024]
Abstract
The higher prevalence of cancer and the unmet need for antioxidant/anti-inflammatory chemotherapeutic compounds with little side effect are of utmost importance. In addition, the increased likelihood of failure in clinical trials along with increasing development costs may have diminished the range of choices among newer drugs for clinical use. This has dictated the necessity to seek out novel medications by repurposing as it needs less time, effort, and resources to explore new uses of a current or unsuccessful medication. In this study, we examined the biological activity of 10 potential quinoline derivatives. Given the half-maximal inhibitory concentration (IC50 value) in lipopolysaccharide (LPS) induced inflammation of RAW264.7 mouse macrophages, all commercial FQs and selected quinolines (quinoline-4-carboxlic and quinoline-3-carboxylic acids) exerted impressively appreciable anti-inflammation affinities versus classical NSAID indomethacin without related cytotoxicities in inflamed macrophages. Conversely, all 14 tested compounds lacked antioxidative DPPH radical scavenging capacities as compared to ascorbic acid. Gemifloxacin, considerably unlike markets FQs, indomethacin and quinoline derivatives, exerted exceptional and differential antiproliferation propensities in colorectum SW480, HCT116, and CACO2, pancreatic PANC1, prostate PC3, mammary T47D, lung A375, and melanoma A549 adherent monolayers using the sulforhodamine B colorimetric method versus antineoplastic cisplatin. All quinoline derivatives and gemifloxacin alike, but not levofloxacin, ciprofloxacin, or indomethacin, displayed substantially selective viability reduction affinities in prolonged tumor incubations of cervical HELA and mammary MCF7 cells. Specifically kynurenic acid (hydrate), quinoline-2-carboxylic acid, quinoline-4-carboxylic acid, quinoline-3-carboxylic acid, and 1,2-dihydro-2-oxo-4-quinoline carboxylic acids possessed the most remarkable growth inhibition capacities against mammary MCF7 cell line, while quinoline-2-carboxylic acid was the only quinoline derivative with significant cytotoxicity on cervical HELA cancer cells. It is highly speculated that chelation with divalent metals via co-planarity with close proximity of the COOH and the N atom could have the potential molecular mechanism for optimally promising repurposed pharmacologies. Conclusively, this study revealed the considerably profound repurposed duality of cytotoxicity and anti-inflammation pharmacologies of quinoline derivatives. Activity-guided structural modifications of the present nuclear scaffolds can be inherently linked to the betterment and enhancement of their repurposed pharmacologies.
Collapse
Affiliation(s)
| | - Ahmad Alzoubi
- Department of Medical Laboratory Sciences, Faculty of Medical Allied Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Hala Al-Daghistani
- Department of Medical Laboratory Sciences, Faculty of Medical Allied Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | | | | | | |
Collapse
|
22
|
Hillis AL, Martin TD, Manchester HE, Högström J, Zhang N, Lecky E, Kozlova N, Lee J, Persky NS, Root DE, Brown M, Cichowski K, Elledge SJ, Muranen T, Fruman DA, Barry ST, Clohessy JG, Madsen RR, Toker A. Targeting Cholesterol Biosynthesis with Statins Synergizes with AKT Inhibitors in Triple-Negative Breast Cancer. Cancer Res 2024; 84:3250-3266. [PMID: 39024548 PMCID: PMC11443248 DOI: 10.1158/0008-5472.can-24-0970] [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: 03/22/2024] [Revised: 05/22/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Triple-negative breast cancer (TNBC) is responsible for a disproportionate number of breast cancer patient deaths due to extensive molecular heterogeneity, high recurrence rates, and lack of targeted therapies. Dysregulation of the phosphoinositide 3-kinase (PI3K)/AKT pathway occurs in approximately 50% of TNBC patients. Here, we performed a genome-wide CRISPR/Cas9 screen with PI3Kα and AKT inhibitors to find targetable synthetic lethalities in TNBC. Cholesterol homeostasis was identified as a collateral vulnerability with AKT inhibition. Disruption of cholesterol homeostasis with pitavastatin synergized with AKT inhibition to induce TNBC cytotoxicity in vitro in mouse TNBC xenografts and in patient-derived estrogen receptor (ER)-negative breast cancer organoids. Neither ER-positive breast cancer cell lines nor ER-positive organoids were sensitive to combined AKT inhibitor and pitavastatin. Mechanistically, TNBC cells showed impaired sterol regulatory element-binding protein 2 (SREBP-2) activation in response to single-agent or combination treatment with AKT inhibitor and pitavastatin, which was rescued by inhibition of the cholesterol-trafficking protein Niemann-Pick C1 (NPC1). NPC1 loss caused lysosomal cholesterol accumulation, decreased endoplasmic reticulum cholesterol levels, and promoted SREBP-2 activation. Taken together, these data identify a TNBC-specific vulnerability to the combination of AKT inhibitors and pitavastatin mediated by dysregulated cholesterol trafficking. These findings support combining AKT inhibitors with pitavastatin as a therapeutic modality in TNBC. Significance: Two FDA-approved compounds, AKT inhibitors and pitavastatin, synergize to induce cell death in triple-negative breast cancer, motivating evaluation of the efficacy of this combination in clinical trials.
Collapse
Affiliation(s)
- Alissandra L. Hillis
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | - Timothy D. Martin
- Division of Genetics, Department of Genetics, Brigham and Women’s Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts.
| | - Haley E. Manchester
- Genetics Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Jenny Högström
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | - Na Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Emmalyn Lecky
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | - Nina Kozlova
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | - Jonah Lee
- Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| | | | - David E. Root
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Karen Cichowski
- Genetics Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Stephen J. Elledge
- Division of Genetics, Department of Genetics, Brigham and Women’s Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts.
| | - Taru Muranen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | - David A. Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California.
| | - Simon T. Barry
- Bioscience, Discovery, Oncology Research and Development, AstraZeneca, Cambridge, Massachusetts.
| | - John G. Clohessy
- Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| | - Ralitsa R. Madsen
- MRC-Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom.
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
23
|
Jung Y, Park S, Lee SS, Kim SH. Centrifugation-Mediated Crystal Growth of Attractive Colloids for Band Edge Lasing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402887. [PMID: 38895964 DOI: 10.1002/smll.202402887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/22/2024] [Indexed: 06/21/2024]
Abstract
Attractive depletion interactions are utilized to organize colloidal particles into crystalline arrays with high crystallinity through spontaneous phase separation. However, uncontrolled nucleation frequently leads to the formation of crystalline grains with varied crystal orientations, which hampers the optical performance of photonic crystals. Here, colloidal crystals have been engineered with uniform orientation and high surface coverage by applying centrifugal force during the depletion-induced assembly of polystyrene particles. The centrifugal force encourages the particles to move toward the bottom surface, which fosters heterogeneous nucleation and supports rapid crystal growth, yielding densely-packed and uniformly-arranged crystal grains with high reflectivity. This study has observed that the nucleation and crystal growth behavior is significantly influenced by the salt concentration. Based on the pair potentials, the transition boundary has been quantitatively analyzed between fluid and crystal phases and identified the threshold for homogeneous nucleation. Utilizing the high-reflectivity colloidal crystals, band-edge lasing is achieved by dissolving the water-soluble dye into the aqueous suspensions. Upon optical excitation, a lasing emission characterized is observed by a narrow spectral width at the short-wavelength band edge. Notably, the laser wavelength can be adjusted by altering the salt concentration or particle diameter, offering a versatile approach to tuning the optical properties.
Collapse
Affiliation(s)
- Yongseok Jung
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sanghyuk Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sang Seok Lee
- Functional Composite Materials Research Center, Institute of Advanced Composite Materials Korea Institute of Science and Technology (KIST), Jeonbuk, 55324, Republic of Korea
| | - Shin-Hyun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
24
|
Silva A, Carpena M, Cassani L, Grosso C, Garcia-Oliveira P, Delerue-Matos C, Simal-Gandara J, Barroso MF, Prieto MA. Optimization and Bioactive Evaluation of Bifurcaria bifurcata Antioxidant-Rich Extracts for Functional Food and Pharmaceutical Applications. Antioxidants (Basel) 2024; 13:1189. [PMID: 39456443 PMCID: PMC11505410 DOI: 10.3390/antiox13101189] [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: 08/16/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
In recent years, consumers have been increasingly interested in natural, healthier, functional foods, with a focus on sea-based products such as algae. Bifurcaria bifurcata (BB) is a macroalga that belongs to the Phaeophyceae class. These brown algae are recognized as the source of bioactive molecules of great interest to the pharmaceutical and nutraceutical industries. The present work applied response surface methodology to optimize the microwave-assisted extraction of the poorly studied algae. The optimization variables were time, pressure, and solvent composition (ethanol/water) and the response parameters selected were yield, total phenolic and flavonoid content, and the antioxidant profile by evaluating DPPH•+, ABTS•+ scavenging activity, and β-carotene discoloration capacity. The results obtained reveal remarkable bioactivity of the crude extract of BB with positive results as an antioxidant and antimicrobial agent. Furthermore, the BB extract's capacity to inhibit enzymes related to neurodegenerative diseases and its anti-inflammatory and anti-proliferation activity open the possibility of future food or pharmaceutical applications.
Collapse
Affiliation(s)
- Aurora Silva
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)—CITEXVI, 36310 Vigo, Spain; (A.S.); (M.C.); (P.G.-O.); (J.S.-G.)
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal; (C.G.); (C.D.-M.)
| | - Maria Carpena
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)—CITEXVI, 36310 Vigo, Spain; (A.S.); (M.C.); (P.G.-O.); (J.S.-G.)
| | - Lucia Cassani
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)—CITEXVI, 36310 Vigo, Spain; (A.S.); (M.C.); (P.G.-O.); (J.S.-G.)
| | - Clara Grosso
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal; (C.G.); (C.D.-M.)
| | - Paula Garcia-Oliveira
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)—CITEXVI, 36310 Vigo, Spain; (A.S.); (M.C.); (P.G.-O.); (J.S.-G.)
| | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal; (C.G.); (C.D.-M.)
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)—CITEXVI, 36310 Vigo, Spain; (A.S.); (M.C.); (P.G.-O.); (J.S.-G.)
| | - Maria Fatima Barroso
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal; (C.G.); (C.D.-M.)
| | - Miguel A. Prieto
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)—CITEXVI, 36310 Vigo, Spain; (A.S.); (M.C.); (P.G.-O.); (J.S.-G.)
| |
Collapse
|
25
|
Tau S, Chamberlin MD, Yang H, Marotti JD, Roberts AM, Carmichael MM, Cressey L, Dragnev C, Demidenko E, Hampsch RA, Soucy SM, Kolling F, Samkoe KS, Alvarez JV, Kettenbach AN, Miller TW. Endocrine persistence in ER+ breast cancer is accompanied by metabolic vulnerability in oxidative phosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615177. [PMID: 39386444 PMCID: PMC11463551 DOI: 10.1101/2024.09.26.615177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Despite adjuvant treatment with endocrine therapies, estrogen receptor-positive (ER+) breast cancers recur in a significant proportion of patients. Recurrences are attributable to clinically undetectable endocrine-tolerant persister cancer cells that retain tumor-forming potential. Therefore, strategies targeting such persister cells may prevent recurrent disease. Using CRISPR-Cas9 genome-wide knockout screening in ER+ breast cancer cells, we identified a survival mechanism involving metabolic reprogramming with reliance upon mitochondrial respiration in endocrine-tolerant persister cells. Quantitative proteomic profiling showed reduced levels of glycolytic proteins in persisters. Metabolic tracing of glucose revealed an energy-depleted state in persisters where oxidative phosphorylation was required to generate ATP. A phase II clinical trial was conducted to evaluate changes in mitochondrial markers in primary ER+/HER2-breast tumors induced by neoadjuvant endocrine therapy ( NCT04568616 ). In an analysis of tumor specimens from 32 patients, tumors exhibiting residual cell proliferation after aromatase inhibitor-induced estrogen deprivation with letrozole showed increased mitochondrial content. Genetic profiling and barcode lineage tracing showed that endocrine-tolerant persistence occurred stochastically without genetic predisposition. Mice bearing cell line- and patient-derived xenografts were used to measure the anti-tumor effects of mitochondrial complex I inhibition in the context of endocrine therapy. Pharmacological inhibition of complex I suppressed the tumor-forming potential of persisters and synergized with the anti-estrogen fulvestrant to induce regression of patient-derived xenografts. These findings indicate that mitochondrial metabolism is essential in endocrine-tolerant persister ER+ breast cancer cells and warrant the development of treatment strategies to leverage this vulnerability in the context of endocrine-sensitive disease. Statement of Significance Endocrine-tolerant persister cancer cells that survive endocrine therapy can cause recurrent disease. Persister cells exhibit increased energetic dependence upon mitochondria for survival and tumor re-growth potential.
Collapse
|
26
|
Moreira S, Martins AD, Alves MG, Pastor LM, Seco-Rovira V, Oliveira PF, Pereira MDL. Aminocarb Exposure Induces Cytotoxicity and Endoplasmic Reticulum Stress-Mediated Apoptosis in Mouse Sustentacular Sertoli Cells: Implications for Male Infertility and Environmental Health. BIOLOGY 2024; 13:721. [PMID: 39336148 PMCID: PMC11429014 DOI: 10.3390/biology13090721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Exposure to pesticides, poses a significant threat to male fertility by compromising crucial cells involved in spermatogenesis. Aminocarb, is a widely used carbamate insecticide, although its detrimental effects on the male reproductive system, especially on sustentacular Sertoli cells, pivotal for spermatogenesis, remains poorly understood. In this study, we investigated the effects of escalating concentrations of aminocarb on a mouse Sertoli cell line, TM4. Assessments included cytotoxic analysis, mitochondrial biogenesis and membrane potential, expression of apoptotic proteins, caspase-3 activity, and oxidative stress evaluation. Our findings revealed a dose-dependent reduction in the proliferation and viability of TM4 cells following exposure to increasing concentrations of aminocarb. Notably, exposure to 5 μM of aminocarb induced depolarization of mitochondria membrane potential, and a significant decrease in the ratio of phosphorylated eIF2α to total eIF2α, suggesting heightened endoplasmic reticulum stress via the activation of the eIF2α pathway. Moreover, the same aminocarb concentration was demonstrated to increase both caspase-3 protein levels and activity, indicating an apoptotic induction. Collectively, our results demonstrate that aminocarb serves as an apoptotic inducer for mouse sustentacular Sertoli cells in vitro, suggesting its potential to modulate independent pathways of the apoptotic cascade. These findings underscore the deleterious impact of aminocarb on spermatogenic performance and male fertility, highlighting the urgent need for further investigation into its mechanisms of action and mitigation strategies to safeguard male fertility.
Collapse
Affiliation(s)
- Sílvia Moreira
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- CICECO-Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana D Martins
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marco G Alves
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Luis Miguel Pastor
- Departamento de Biología Celular e Histología, Faculdad de Medicina, IMIB-Arrixaca, Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia, 30120 Murcia, Spain
| | - Vicente Seco-Rovira
- Departamento de Biología Celular e Histología, Faculdad de Medicina, IMIB-Arrixaca, Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia, 30120 Murcia, Spain
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria de Lourdes Pereira
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- CICECO-Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
27
|
Ramadan FHJ, Koszegi B, Vantus VB, Fekete K, Kiss GN, Rizsanyi B, Bognar R, Gallyas F, Bognar Z. Comparison of Mitochondrial and Antineoplastic Effects of Amiodarone and Desethylamiodarone in MDA-MB-231 Cancer Line. Int J Mol Sci 2024; 25:9781. [PMID: 39337269 PMCID: PMC11432025 DOI: 10.3390/ijms25189781] [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: 07/22/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Previously, we have demonstrated that amiodarone (AM), a widely used antiarrhythmic drug, and its major metabolite desethylamiodarone (DEA) both affect several mitochondrial processes in isolated heart and liver mitochondria. Also, we have established DEA's antitumor properties in various cancer cell lines and in a rodent metastasis model. In the present study, we compared AM's and DEA's mitochondrial and antineoplastic effects in a human triple-negative breast cancer (TNBC) cell line. Both compounds reduced viability in monolayer and sphere cultures and the invasive growth of the MDA-MB-231 TNBC line by inducing apoptosis. They lowered mitochondrial trans-membrane potential, increased Ca2+ influx, induced mitochondrial permeability transition, and promoted mitochondrial fragmentation. In accordance with their mitochondrial effects, both substances massively decreased overall, and even to a greater extent, mitochondrial ATP production decreased, as determined using a Seahorse live cell respirometer. In all these effects, DEA was more effective than AM, indicating that DEA may have higher potential in the therapy of TNBC than its parent compound.
Collapse
Affiliation(s)
- Fadi H J Ramadan
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| | - Balazs Koszegi
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| | - Viola B Vantus
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| | - Gyongyi N Kiss
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| | - Balint Rizsanyi
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| | - Rita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary
| | - Zita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary
| |
Collapse
|
28
|
Tarasiuk O, Invernizzi C, Alberti P. In vitro neurotoxicity testing: lessons from chemotherapy-induced peripheral neurotoxicity. Expert Opin Drug Metab Toxicol 2024:1-16. [PMID: 39246127 DOI: 10.1080/17425255.2024.2401584] [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: 04/15/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Chemotherapy induced peripheral neurotoxicity (CIPN) is a long-lasting, or even permanent, late toxicity caused by largely used anticancer drugs. CIPN affects a growing population of cancer survivors and diminishes their quality of life since there is no curative/preventive treatment. Among several reasons for this unmet clinical need, there is an incomplete knowledge on mechanisms leading to CIPN. Therefore, bench side research is still greatly needed: in vitro studies are pivotal to both evaluate neurotoxicity mechanisms and potential neuroprotection strategies. AREAS COVERED Advantages and disadvantages of in vitro approaches are addressed with respect to their applicability to the CIPN field. Different cell cultures and techniques to assess neurotoxicity/neuroprotection are described. PubMed search-string: (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (in vitro) AND (((((model) OR SH-SY5Y) OR PC12) OR iPSC) OR DRG neurons); (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (model) AND (((neurite elongation) OR cell viability) OR morphology). No articles published before 1990 were selected. EXPERT OPINION CIPN is an ideal experimental setting to test axonal damage and, in general, peripheral nervous system mechanisms of disease and neuroprotection. Therefore, starting from robust preclinical data in this field, potentially, relevant biological rationale can be transferred to other human spontaneous diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Chiara Invernizzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- Neuroscience, School of Medicine and Surgery, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
- Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
29
|
Klomsakul P, Chalopagorn P. In Vitro α-Amylase and α-Glucosidase Inhibitory Potential of Green Banana Powder Extracts. ScientificWorldJournal 2024; 2024:5515855. [PMID: 39280721 PMCID: PMC11401666 DOI: 10.1155/2024/5515855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/28/2024] [Accepted: 08/16/2024] [Indexed: 09/18/2024] Open
Abstract
This study investigated the proximate composition and inhibitory potential of hot water and ethanolic extracts of the pulp, peel and whole fruit of green banana (Musa sapientum) on α-amylase and α-glucosidase. Bioactive compounds were identified using GC-MS analysis. In addition, the cytotoxic effect on human gingival fibroblast (hGF) was evaluated using the sulphorhodamine B (SRB) assay. The results showed that the peel of green banana had the highest amount of ash (10.05%), fat (2.83%), protein (3.64%) and total dietary fibre (36.62%). The carbohydrate content of the whole fruit (81.79%) and pulp (81.50%) was higher than that of the peel (71.90%). The moisture content of the pulp (13.08%) was higher than that of the peel (11.58%) and whole fruit (11.30%). The ethanolic green banana peel extract showed a good inhibitory effect of α-amylase and α-glucosidase with the concentration necessary for 50% inhibition (IC50) of 0.512 and 0.100 mg·mL-1, respectively. The α-glucosidase inhibitory effect of the ethanolic green banana peel extract and the hot water green banana peel extract was not significantly different from that of acarbose (IC50 0.108 mg·mL-1). GC-MS analysis of the ethanolic green banana peel extract revealed fatty acids and fatty acid ester (9-octadecenamide (Z), octadecanamide and other compounds). The ethanolic peel extract exhibits a significant noncytotoxicity effect on hGF cells at concentrations ranging from 0.0001 to 1.0 mg·mL-1.
Collapse
Affiliation(s)
- Pongsathorn Klomsakul
- Department of BiologyFaculty of Science and TechnologyPhranakhon Rajabhat University, Bangkok 10220, Thailand
| | - Pornchanok Chalopagorn
- Department of ChemistryFaculty of Science and TechnologyPhranakhon Rajabhat University, Bangkok 10220, Thailand
| |
Collapse
|
30
|
Santiago-O’Farrill JM, Blessing Bollu A, Yang H, Orellana V, Pina M, Zhang X, Liu J, Bast RC, Lu Z. Crizotinib Enhances PARP Inhibitor Efficacy in Ovarian Cancer Cells and Xenograft Models by Inducing Autophagy. Mol Cancer Res 2024; 22:840-851. [PMID: 38780897 PMCID: PMC11372360 DOI: 10.1158/1541-7786.mcr-23-0680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 04/02/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) can encounter resistance through various mechanisms, limiting their effectiveness. Our recent research showed that PARPi alone can induce drug resistance by promoting autophagy. Moreover, our studies have revealed that anaplastic lymphoma kinase (ALK) plays a role in regulating the survival of ovarian cancer cells undergoing autophagy. Here, we explored whether the ALK-inhibitor crizotinib could enhance the efficacy of PARPi by targeting drug-induced autophagic ovarian cancer cell and xenograft models. Our investigation demonstrates that crizotinib enhances the anti-tumor activity of PARPi across multiple ovarian cancer cells. Combination therapy with crizotinib and olaparib reduced cell viability and clonogenic growth in two-olaparib resistant cell lines. More importantly, this effect was consistently observed in patient-derived organoids. Furthermore, combined treatment with crizotinib and olaparib led to tumor regression in human ovarian xenograft models. Mechanistically, the combination resulted in increased levels of reactive oxygen species (ROS), induced DNA damage, and decreased the phosphorylation of AKT, mTOR, and ULK-1, contributing to increased olaparib-induced autophagy and apoptosis. Notably, pharmacologic, or genetic inhibition or autophagy reduced the sensitivity of ovarian cancer cell lines to olaparib and crizotinib treatment, underscoring the role of autophagy in cell death. Blocking ROS mitigated olaparib/crizotinib-induced autophagy and cell death while restoring levels of phosphorylated AKT, mTOR and ULK-1. These findings suggest that crizotinib can improve the therapeutic efficacy of olaparib by enhancing autophagy. Implications: The combination of crizotinib and PARPi presents a promising strategy, that could provide a novel approach to enhance outcomes for patients with ovarian cancer.
Collapse
Affiliation(s)
| | - Alicia Blessing Bollu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Hailing Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Vivian Orellana
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Marc Pina
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Xudong Zhang
- Department of Pathology/Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jinsong Liu
- Department of Pathology/Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Robert C. Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
31
|
Pinho SA, Oliveira PJ, Cunha-Oliveira T. Heterogeneous redox responses in NHDF cells primed to enhance mitochondrial bioenergetics. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167495. [PMID: 39241844 DOI: 10.1016/j.bbadis.2024.167495] [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: 06/12/2024] [Revised: 08/01/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Aging and lifestyle-related diseases, such as cardiovascular diseases, diabetes, cancer, and neurodegenerative disorders, are major global health challenges. These conditions are often linked to redox imbalances, where cells fail to regulate reactive redox species (RRS), leading to oxidative stress and cellular damage. Although antioxidants are known to neutralize harmful RRS, their clinical efficacy remains inconsistent. One reason for this inconsistency is the inadequacy of current in vitro models to accurately mimic in vivo redox conditions. This study addresses the gap in understanding the heterogeneity of redox responses in cells by using metabolically primed human dermal fibroblasts (NHDF), a model relevant for precision mitochondrial medicine. We investigated how metabolic priming, which enhances mitochondrial bioenergetics, influences redox responses to oxidative stress induced by hydrogen peroxide (H2O2) and tert-butyl hydroperoxide (tBHP). Specifically, we explored the impact of cell population density and cell cycle distribution on redox dynamics. Our findings indicate that NHDF cells cultured in oxidative phosphorylation-promoting medium (OXm) exhibit significantly larger variability in oxidative stress responses. This variability suggests that enhanced mitochondrial bioenergetics necessitates a constant regulation of the cellular redox machinery, potentially leading to heterogeneous responses. Additionally, cells grown in OXm showed increased mitochondrial polarization and a lower percentage of cells in the G2/M phase, contributing to the observed heterogeneity. Key factors influencing this variability included cell population density at the time of oxidant exposure and fluctuations in cell cycle distribution. Our results highlight the necessity of employing multiple oxidants in metabolic priming models to achieve a comprehensive understanding of oxidative stress responses and redox regulation mechanisms. Furthermore, the study emphasizes the need to refine in vitro models to better reflect in vivo conditions, which is crucial for the development of effective redox-based therapeutic strategies.
Collapse
Affiliation(s)
- Sónia A Pinho
- CNC - UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Portugal.
| | - Paulo J Oliveira
- CNC - UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| | - Teresa Cunha-Oliveira
- CNC - UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| |
Collapse
|
32
|
Decharuangsilp S, Arwon U, Hoarau M, Vanichtanankul J, Saeyang T, Jantra T, Rattanajak R, Thiabma R, Sooksai N, Kongkasuriyachai D, Kamchonwongpaisan S, Yuthavong Y. Flexible 2,4-diaminopyrimidine bearing a butyrolactone as Plasmodium falciparum dihydrofolate reductase inhibitors. Bioorg Chem 2024; 153:107789. [PMID: 39250850 DOI: 10.1016/j.bioorg.2024.107789] [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: 05/22/2024] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 09/11/2024]
Abstract
Recently, P218, a new flexible antifolate targeting Plasmodium falciparum dihydrofolate reductase (PfDHFR), has entered its clinical trial with good safety profile and effective Pf infection prevention. However, it carries a free carboxyl terminal, which is hydrophilic and prone to metabolic glucuronidation. Here, a new series of P218 analogues carrying butyrolactone has been synthesized with the purpose of enhancing lipophilicity and minimizing metabolic instability. The inhibition constants against the mutant PfDHFR enzymes are in sub-nanomolar level and the antimalarial activity against antifolate-resistant parasites are in the low micromolar range. The crystal structure of the most potent analogue LA1 bound enzyme complex indicates interaction with multiple residues, including Arg122 and Phe116 in the active site. In vitro log D7.4 and kinetic solubility confirmed a higher lipophilicity of this butyrolactone series as compared to P218. These outcomes suggest the possibility to further develop butyrolactone derivatives as non-carboxyl antiplasmodial antifolates.
Collapse
Affiliation(s)
- Sasithorn Decharuangsilp
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand.
| | - Uthai Arwon
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Marie Hoarau
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Jarunee Vanichtanankul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Thanaya Saeyang
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Tararat Jantra
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Roonglawan Rattanajak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Ratthiya Thiabma
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Nawarat Sooksai
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Darin Kongkasuriyachai
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Yongyuth Yuthavong
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| |
Collapse
|
33
|
Zárate-Pérez A, Cruz-Cázares AP, Ordaz-Rosado D, García-Quiroz J, León-Del-Rio A, Avila E, Milo-Rocha E, Díaz L, García-Becerra R. The vitamin D analog EB1089 sensitizes triple-negative breast cancer cells to the antiproliferative effects of antiestrogens. Adv Med Sci 2024; 69:398-406. [PMID: 39233278 DOI: 10.1016/j.advms.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/12/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE Patients bearing estrogen receptor (ER)α-negative breast cancer tumors confront poor prognosis and are typically unresponsive to hormone therapy. Previous studies have shown that calcitriol, the active vitamin D metabolite, can induce ERα expression in ERα-negative cells. EB1089, a calcitriol analog with reduced calcemic effects, exhibits greater potency than calcitriol in inhibiting cancer cell growth. However, the impact of EB1089 on ERα expression in triple-negative breast cancer (TNBC) cells remains unexplored. This study aims to investigate whether EB1089 could induce functional ERα expression in TNBC cell lines, potentially enabling the antiproliferative effects of antiestrogens. MATERIALS AND METHODS TNBC cell lines HCC1806 and HCC1937 were treated with EB1089, and ERα expression was analyzed using real-time PCR and Western blots. The transcriptional activity of induced ERα was evaluated through a luciferase reporter assay. The antiproliferative effects of tamoxifen and fulvestrant antiestrogens were assessed using the sulforhodamine B assay in the EB1089-treated cells. RESULTS Our findings indicated that EB1089 significantly induced ERα mRNA and protein expression in TNBC cells. Moreover, EB1089-induced ERα exhibited transcriptional activity and effectively restored the inhibitory effects of antiestrogens, thereby suppressing cell proliferation in TNBC cells. CONCLUSION EB1089 induced the expression of functional ERα in TNBC cells, restoring the antiproliferative effects of antiestrogens. These results highlight the potential of using EB1089 as a promising strategy for re-establishment of the antiproliferative effect of antiestrogens as a possible management for TNBC. This research lays the foundation for potential advancements in TNBC treatment, offering new avenues for targeted and effective interventions.
Collapse
Affiliation(s)
- Adriana Zárate-Pérez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Alitzin Pamela Cruz-Cázares
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - David Ordaz-Rosado
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Janice García-Quiroz
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Alfonso León-Del-Rio
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico; Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Euclides Avila
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Edgar Milo-Rocha
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Lorenza Díaz
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico.
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico; Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
34
|
Alhazmi A, Sidney LE, Hopkinson A, Elsheikha HM. Comparative cytotoxicity of Acanthamoeba castellanii-derived conditioned medium on human corneal epithelial and stromal cells. Acta Trop 2024; 257:107288. [PMID: 38901524 DOI: 10.1016/j.actatropica.2024.107288] [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: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Soluble factors in the secretome of Acanthamoeba castellanii play crucial roles in the pathogenesis of Acanthamoeba keratitis (AK). Investigating the pathological effects of A. castellanii-derived conditioned medium (ACCM) on ocular cells can provide insights into the damage inflicted during AK. This study examined ACCM-induced cytotoxicity in primary human corneal stromal cells (CSCs) and a human SV40 immortalized corneal epithelial cell line (ihCECs) at varying ACCM concentrations (25 %, 50 %, 75 %, and 100 %). MTT, AlamarBlue, Sulforhodamine B, lactate dehydrogenase, and Caspase-3/7 activation assays were used to assess the impact of ACCM on the cell viability, proliferation and apoptosis. Additionally, fluorescent staining was used to reveal actin cytoskeleton changes. ACCM exposure significantly decreased cell viability, increased apoptosis, and disrupted the actin cytoskeleton, particularly at higher concentrations and longer exposures. Proteases were found to mediate these cytopathogenic effects, highlighting the need for characterization of A. castellanii proteases as key virulence factors in AK pathogenesis.
Collapse
Affiliation(s)
- Abdullah Alhazmi
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham NG7 2UH, UK; Faculty of Public Health and Health Informatics, Umm Al Qura University, Makkah, Saudi Arabia
| | - Laura E Sidney
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham NG7 2UH, UK
| | - Andy Hopkinson
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham NG7 2UH, UK
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK.
| |
Collapse
|
35
|
Wang Y, Zhang Y, Li Y, Zhang Z, Lian XY. The necessity of eliminating the interference of panaxatriol saponins to maximize the preventive effect of panaxadiol saponins against Parkinson's disease in rats. J Ginseng Res 2024; 48:464-473. [PMID: 39263312 PMCID: PMC11385176 DOI: 10.1016/j.jgr.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 09/13/2024] Open
Abstract
Background The effects of individual panaxadiol saponin and panaxatriol saponin on rodent models of Parkinson's disease (PD) have been recognized. However, it is not clear whether purified total ginsenosides as an entirety has effect against PD in rat model. This study compared the protective effects of a purified panaxadiol saponin fraction (PDSF), a purified panaxatriol saponin fraction (PTSF), and their mixtures against the rotenone (ROT)-induced PD in rats. Methods Potential effects of PDSF, PTSF, and their mixtures against motor dysfunction and impairments of nigrostriatal dopaminergic neurons (DN), blood-brain barrier (BBB), cerebrovascular endothelial cells (CEC), and glial cells were measured in the models of ROT-induced PD rats and cell damage. Pro-inflammatory NF-kB p65 (p65) activation was localized in DN and other cells in the striatum. Results PDSF and PTSF had a dose-dependent effect against motor dysfunction with a larger effective dose range for PDSF. PDSF protected CEC, glial cells, and DN in models of PD rats and cell damage, while PTSF had no such protections. Chronic ROT exposure potently activated p65 in CEC with enhanced pro-inflammatory and decreased anti-inflammatory factors and impaired BBB in the striatum, PDSF almost completely blocked the ROT-induced p65 activation and maintained both anti- and pro-inflammatory factors at normal levels and BBB integrity, but PTSF aggravated the p65 activation with impaired BBB. Furthermore, PTSF nullified all the effects of PDSF when they were co-administrated. Conclusion PDSF had significant protective effect against the ROT-induced PD in rats by protecting CEC, glial cells, and DN, likely through inhibiting NF-κB p65 in CEC from triggering neuroinflammation, and also directly protecting glial cells and neurons against ROT-induced toxicity. PDSF has great potential for preventing and treating PD.
Collapse
Affiliation(s)
- Yanwei Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yufen Zhang
- Anhui University of Chinese Medicine, Hefei, China
| | - Yueyue Li
- Anhui University of Chinese Medicine, Hefei, China
| | - Zhizhen Zhang
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan, China
| | - Xiao-Yuan Lian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
36
|
Sultan NS, Shoukry AA, Rashidi FB, Elhakim HKA. Biological Applications, In Vitro Cytotoxicity, Cellular Uptake, and Apoptotic Pathway Studies Induced by Ternary Cu (II) Complexes Involving Triflupromazine with Biorelevant Ligands. Cell Biochem Biophys 2024; 82:2651-2671. [PMID: 39018004 DOI: 10.1007/s12013-024-01376-9] [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] [Accepted: 06/18/2024] [Indexed: 07/18/2024]
Abstract
The novel mixed-ligand complexes derived from the parent antidepressant phenothiazine drug triflupromazine (TFP) were synthesized along with the secondary ligands glycine and histidine. [Cu(TFP)(Gly)Cl]·2H2O (1) and [Cu(TFP)(His)Cl]·2H2O (2) were examined for their in vitro biological properties. Cyclic voltammetry was used to study the binding of both complexes to CT-DNA. The two complexes were examined for antiviral, antiparasite, and anti-inflammatory applications. An in vitro cytotoxicity study on two different cancer cell lines, MCF-7, HepG2, and a normal cell line, HSF, shows promising selective cytotoxicity for cancer cells. An investigation of the cell cycle and apoptosis rates was evaluated by flow cytometry with Annexin V-FITC/Propidium Iodide (PI) staining of the treated cells. Gene expression and western blotting were carried out to determine the expression levels of the pro-apoptotic markers and the anti-apoptotic marker Bcl2. The tested complexes decreased cell viability and triggered apoptosis in human tumor cell lines. Molecular docking was also used to simulate Bcl2 inhibition. Finally, complex (2) has potent antitumor effects on human tumor cells, especially against HepG2 cells, as seen in the cellular drug uptake assay. Consequently, complex (2) may prove useful against cancer, especially liver cancer. For further understanding, it needs to be explored in vivo.
Collapse
Affiliation(s)
- Nourhan S Sultan
- Biotechnology department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Azza A Shoukry
- Inorganic Chemistry Division, Chemistry Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Fatma B Rashidi
- Biochemistry Division, Chemistry Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Heba K A Elhakim
- Biochemistry Division, Chemistry Department, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
37
|
Erick NO, Montserrat EGA, Antonio EPE, Rocío SP, Eduardo LU, Verónica GC, Miriam RS, Imelda JA, Del Carmen BFJ, Cruz RCJ, Tzasna HDC, María GBA, Guillermo AAJ. Photoprotective effect of topical treatment with Lopezia racemosa extract against deleterious UVB irradiation effects in the skin of hairless mice. Photochem Photobiol 2024; 100:1489-1506. [PMID: 38445720 DOI: 10.1111/php.13926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 03/07/2024]
Abstract
Lopezia racemosa is known as a "mosquito flower or perlilla." It is commonly found in corn crops. In traditional Mexican medicine, this plant is used to treat stomach cancer and urinary tract infections. Likewise, compounds and extracts isolated from plants have shown cytotoxic and anti-inflammatory effects. The objective of this study was to evaluate the photochemoprotective effect of topical treatment with the methanolic extract of L. racemosa (MELR) as a photochemoprotective agent against the harmful effects of UV irradiation (UVR) on a bacterial model and hairless mice. The MELR components were separated and analyzed via HPLC-UV-ESI-MS. Antioxidant activity was evaluated by the ability of MERL to scavenge DPPH and ABTS free radicals and by its FRAP capacity. The toxicity of MELR was evaluated in keratinocyte cultures. The photoprotective capacity of MELR was assessed through challenge experiments using models with bacteria and hairless CD1 et/et mice; cytokines related to the damage caused by UVR were also measured. In the methanolic extract of L. racemosa, five metabolites were detected and identified: two isomers of quercetin 6-C glycoside, orientin, quercetin 3-(6″-acetylglycoside) and quercetin 3-(6″-galloylglycoside) 7-(2,3-dihydroxytetrahydro-2H-pyran-4-yl acetate). MELR exhibited DPPH and ABTS radical scavenging properties, in addition to Fe ion reducing activity. MELR showed a photoprotective effect against UVB radiation-induced death in Escherichia coli bacteria. At the histological level, topical treatment of CD-1 et/et mice with MERL reduced the damage caused by UVR. Quantification of interleukins in the blood of mice revealed that the expression of IL-12 was greater in the control group treated with ultraviolet radiation than in the group protected with MELR. The methanolic extract of L. racemosa has photochemoprotective properties.
Collapse
Affiliation(s)
- Nolasco Ontiveros Erick
- Laboratorio de Fitoquímica, UBIPRO, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Estudios de Posgrado, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | | | - Estrella Parra Edgar Antonio
- Laboratorio de Fitoquímica, UBIPRO, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| | - Serrano Parrales Rocío
- Laboratorio de Bioactividad de Productos Naturales, UBIPRO, Facultad de Estudios Superiores (FES)-Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla de Baz, Estado de Mexico, Mexico
| | - López Urrutia Eduardo
- Laboratorio de Genómica Funcional del Cáncer, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| | - García Castillo Verónica
- Laboratorio de Genómica Funcional del Cáncer, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| | - Rodríguez-Sosa Miriam
- Laboratorio de Inmunidad Innata, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| | - Juárez-Avelar Imelda
- Laboratorio de Inmunidad Innata, UBIMED, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| | - Benítez Flores José Del Carmen
- Laboratorio de Histología, UMF, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| | - Rivera Cabrera José Cruz
- Laboratorio de Cromatografía de Líquidos, Departamento de Farmacología, Escuela Militar de Medicina, Ciudad de Mexico, Mexico
| | - Hernández Delgado Claudia Tzasna
- Laboratorio de Bioactividad de Productos Naturales, UBIPRO, Facultad de Estudios Superiores (FES)-Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla de Baz, Estado de Mexico, Mexico
| | - García Bores Ana María
- Laboratorio de Fitoquímica, UBIPRO, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| | - Avila Acevedo José Guillermo
- Laboratorio de Fitoquímica, UBIPRO, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de Mexico, Mexico
| |
Collapse
|
38
|
de Carvalho Rodrigues V, Guterres IZ, Pereira Savi B, Fongaro G, Silva IT, Vitor Salmoria G. Additive manufacturing of TPU devices for genital herpes treatment with sustained acyclovir release. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-16. [PMID: 39190633 DOI: 10.1080/09205063.2024.2396221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
The treatment of recurrent genital herpes typically involves daily doses of acyclovir for extended periods. Additive manufacturing is an intriguing technique for creating personalised drug delivery systems, which can enhance the effectiveness of treatments for various diseases. The vaginal route offers a viable alternative for the systemic administration of drugs with low oral bioavailability. In this study, we produced different grades of thermoplastic polyurethane (TPU) filaments through hot-melt extrusion, with acyclovir concentrations of 0%, 10%, and 20% by weight. We used fused filament fabrication to manufacture matrix-based devices, including intrauterine devices and intravaginal rings. Our results, obtained through SEM, FTIR, and DSC analyses, confirm the successful incorporation of acyclovir into the matrix. Thermal analysis reveals that the manufacturing process alters the organization of the TPU chains, resulting in a slight reduction in crystallinity. In our in-vitro tests, we observed an initial burst release on the first day, followed by sustained release at reduced rates for up to 145 days, demonstrating their potential for long-term applications. Additionally, cytotoxicity analysis suggests the excellent biocompatibility of the printed devices, and biological assays show a remarkable 99% reduction in HSV-1 replication. In summary, TPU printed devices offer a promising alternative for long-term genital herpes treatment, with the results obtained potentially contributing to the advancement of pharmaceutical manufacturing.
Collapse
Affiliation(s)
| | - Iara Zanella Guterres
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Beatriz Pereira Savi
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Gislaine Fongaro
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Izabella Thaís Silva
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
- Laboratory of Pharmacognosy, Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Gean Vitor Salmoria
- Nimma, Department of Mechanical Engineering, Federal University of Santa Catarina, Florianópolis, Brazil
- Biomechanics Engineering Laboratory, University Hospital (HU), Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
39
|
Sonkar AB, Verma A, Yadav S, Singh J, Kumar R, Keshari AK, Kumar A, Kumar D, Shrivastava NK, Rani S, Rastogi S, Alamoudi MK, Nazam Ansari M, Saeedan AS, Kaithwas G, Saha S. Antiproliferative, apoptotic and anti-inflammatory potential of 5H-benzo[h]thiazolo[2,3-b]quinazoline analogues: Novel series of anticancer compounds. Int Immunopharmacol 2024; 137:112496. [PMID: 38901240 DOI: 10.1016/j.intimp.2024.112496] [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: 01/23/2024] [Revised: 05/30/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
Lung cancer (LC) is the most common cancer in males. As per GLOBOCAN 2020, 8.1 % of deaths and 5.9 % of cases of LC were reported in India. Our laboratory has previously reported the significant anticancer potential of 5H-benzo[h]thiazolo[2,3-b]quinazoline analogues. In this study, we have explored the anticancer potential of 7A {4-(6,7-dihydro-5H-benzo[h]thiazolo[2,3-b]quinazolin-7-yl)phenol} and 9A {7-(4-chlorophenyl)-9-methyl-6,7-dihydro-5H-benzo[h]thiazolo[2,3-b]quinazoline}by using in-vitro and in-vivo models of LC. In this study, we investigated the antiproliferative potential of quinazoline analogues using A549 cell line to identify the best compound of the series. The in-vitro and molecular docking studies revealed 7A and 9A compounds as potential analogues. We also performed acute toxicity study to determine the dose. After that, in-vivo studies using urethane-induced LC in male albino Wistar rats carried out further physiological, biochemical, and morphological evaluation (SEM and H&E) of the lung tissue. We have also evaluated the antioxidant level, inflammatory, and apoptotic marker expressions. 7A and 9A did not demonstrate any signs of acute toxicity. Animals treated with urethane showed a significant upregulation of oxidative stress. However, treatment with 7A and 9A restored antioxidant markers near-normal levels. SEM and H&E staining of the lung tissue demonstrated recovered architecture after treatment with 7A and 9A. Both analogues significantly restore inflammatory markers to normal level and upregulate the intrinsic apoptosis protein expression in the lung tissue. These experimental findings demonstrated the antiproliferative potential of the synthetic analogues 7A and 9A, potentially due to their anti-inflammatory and apoptotic properties.
Collapse
Affiliation(s)
- Archana Bharti Sonkar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Abhishek Verma
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Sneha Yadav
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Jyoti Singh
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Rohit Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Amit K Keshari
- Amity Institute of Pharmacy, Amity University, Lucknow Campus, Lucknow 226028, Uttar Pradesh, India
| | - Anurag Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Dharmendra Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Neeraj Kumar Shrivastava
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Soniya Rani
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Shubham Rastogi
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| | - Mariam K Alamoudi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohd Nazam Ansari
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Abdulaziz S Saeedan
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Gaurav Kaithwas
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India.
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226 025, Uttar Pradesh, India
| |
Collapse
|
40
|
Miranda CA, Mansano JRD, Mingatto FE. Ivermectin-induced toxicity in HepG2 cells and the protective effect of tetrahydrocurcumin and vitamin C. Drug Chem Toxicol 2024:1-11. [PMID: 39155661 DOI: 10.1080/01480545.2024.2389954] [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: 05/25/2024] [Revised: 07/31/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024]
Abstract
Ivermectin (IVM) is a semi-synthetic antiparasitic derived from abamectin, one of the natural avermectins. The liver promotes metabolism and excretion of IVM, representing a risk of toxicity to this organ. The use of antioxidants to alleviate damage caused by chemicals has been increasingly studied. Thus, the aim of this study was to evaluate the effects of IVM on HepG2 cells to elucidate the mechanisms related to its toxicity and the possible protection provided by tetrahydrocurcumin (THC) and vitamin C. HepG2 cells were treated with IVM (1-25 μM) for 24 and 48 h. IVM was cytotoxic to HepG2 cells, denoted by a dose-dependent decrease in cell proliferation and metabolic activity. In addition, IVM induced damage to the cell membrane at all tested concentrations and for both incubation times. IVM significantly decreased the mitochondrial membrane potential from concentrations of 5 μM (24 h) and 1 μM (48 h). Additionally, IVM showed a time- and dose-dependent decrease in cellular adenosine triphosphate levels. The levels of reduced glutathione were decreased in a time- and dose-dependent manner, while IVM stimulated the production of reactive oxygen and nitrogen species (RONS) at all tested doses, reaching rates above 50% following treatment at 7.5 μM (24 h) or 5 μM (48 h). Treatment with THC (50 μM) and vitamin C (50 μM) protected against IVM-induced cytotoxicity and RONS production. These results suggest that oxidative damage is involved in IVM-induced toxicity in HepG2 cells, and that THC and vitamin C can mitigate the toxic effects caused by the compound.
Collapse
Affiliation(s)
- Camila Araújo Miranda
- College of Agricultural and Technological Sciences, Department of Animal Science, São Paulo State University (UNESP), Dracena, Brazil
| | - João Rodolfo Domingues Mansano
- College of Agricultural and Technological Sciences, Department of Animal Science, São Paulo State University (UNESP), Dracena, Brazil
- Faculdades de Dracena, Medical School, Unifadra, Dracena, Brazil
| | - Fábio Erminio Mingatto
- College of Agricultural and Technological Sciences, Department of Animal Science, São Paulo State University (UNESP), Dracena, Brazil
| |
Collapse
|
41
|
Wilson W, Lowman D, Puthumana J, Kuriakose R, Singh ISB, Philip R. Biocompatible melanin from the marine black yeast Hortaea werneckii R23 with antioxidant and photoprotection property. Braz J Microbiol 2024:10.1007/s42770-024-01483-y. [PMID: 39155342 DOI: 10.1007/s42770-024-01483-y] [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: 05/23/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024] Open
Abstract
Pigments from diverse sources have a great deal of interest due to its multifaceted applications. Hence, this study reports the physicochemical and functional characterization of the black pigment melanin from the marine black yeast Hortaea werneckii R23. In the present study, Hortaea werneckii R23, produced a black pigment in the yeast biomass. The pigment was extracted from the harvested yeast biomass and followed by pigment purification, characterization and identification was done. Physicochemical characterization of the pigment showed acid precipitation, alkali solubilization, insolubility in most organic solvents and water. The black pigment was confirmed as melanin based on ultraviolet-visible spectroscopy, Fourier-transform infrared, and Nuclear magnetic resonance spectroscopy analyses. Furthermore, the analyses of the elemental composition indicated that the pigment possessed a moderately high percentage of nitrogen and also detectable proportion of sulfur. All these Physicochemical properties indicated that H. werneckii melanin (HwM) mostly consisted of eumelanin. HwM exhibited strong antioxidant potential as reactive oxygen species (ROS) scavenger by in vitro DPPH (1,1-diphenyl-2-picryl hydrazyl) and ABTS (2,2-azinobis-3-ethyl-benzothiozoline-6-sulphonic acid) radical scavenging assay, and lipid peroxidation assay. The photoprotectant role of HwM on UV-irradiated human epithelial cells (HEp-2) revealed its potential effect in photoprotection. In addition, cytotoxicity study by XTT and SRB assay confirmed its biocompatibility with HEp-2 cells. From these findings, it is evident that the HwM from the marine black yeast possesses strong antioxidant and photoprotectant activity, moreover, it is biocompatible to human epithelial cells. So HwM could be used as a protective agent against oxidative stress associated disorders in an environment-friendly perspective.
Collapse
Affiliation(s)
- Wilsy Wilson
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Kochi, 682016, Kerala, India
- St. Joseph's College, Irinjalakuda, 680121, University of Calicut, Kerala, India
| | - Douglas Lowman
- AppRidge International, LLC, Telford, TN, 37690-2235, USA
| | - Jayesh Puthumana
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, 682016, Kerala, India
| | - Reema Kuriakose
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Kochi, 682016, Kerala, India
| | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, 682016, Kerala, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Kochi, 682016, Kerala, India.
| |
Collapse
|
42
|
Ortega-Vidal J, Mut-Salud N, de la Torre JM, Altarejos J, Salido S. Chemical Characterization of Pruning Wood Extracts from Six Japanese Plum ( Prunus salicina Lindl.) Cultivars and Their Antitumor Activity. Molecules 2024; 29:3887. [PMID: 39202966 PMCID: PMC11357068 DOI: 10.3390/molecules29163887] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
The Japanese plum tree (Prunus salicina Lindl.) is mainly cultivated in temperate areas of China and some European countries. Certain amounts of wood (from pruning works) are generated every year from this crop of worldwide commercial significance. The main objective of this work was to value this agricultural woody residue, for which the chemical composition of pruning wood extracts from six Japanese plum cultivars was investigated, and the antiproliferative activity of extracts and pure phenolics present in those extracts was measured. For the chemical characterization, total phenolic content and DPPH radical-scavenging assays and HPLC‒DAD/ESI‒MS analyses were performed, with the procyanidin (-)-ent-epicatechin-(2α→O→7,4α→8)-epicatechin (5) and the propelargonidin (+)-epiafzelechin-(2β→O→7,4β→8)-epicatechin (7) being the major components of the wood extracts. Some quantitative differences were found among plum cultivars, and the content of proanthocyanidins ranged from 1.50 (cv. 'Fortune') to 4.44 (cv. 'Showtime') mg/g of dry wood. Regarding the antitumoral activity, eight wood extracts and four phenolic compounds were evaluated in MCF-7 cells after 48 h of induction, showing the wood extract from cv. 'Songold' and (‒)-annphenone (3), the best antiproliferative activity (IC50: 424 μg/mL and 405 μg/mL, respectively).
Collapse
Affiliation(s)
- Juan Ortega-Vidal
- Department of Inorganic and Organic Chemistry, Faculty of Experimental Sciences, Campus of International Excellence in Agri-Food (ceiA3), University of Jaén, 23071 Jaén, Spain; (J.O.-V.); (J.M.d.l.T.); (S.S.)
| | - Nuria Mut-Salud
- Institute of Biopathology and Regenerative Medicine, University of Granada, 18071 Granada, Spain;
| | - José M. de la Torre
- Department of Inorganic and Organic Chemistry, Faculty of Experimental Sciences, Campus of International Excellence in Agri-Food (ceiA3), University of Jaén, 23071 Jaén, Spain; (J.O.-V.); (J.M.d.l.T.); (S.S.)
| | - Joaquín Altarejos
- Department of Inorganic and Organic Chemistry, Faculty of Experimental Sciences, Campus of International Excellence in Agri-Food (ceiA3), University of Jaén, 23071 Jaén, Spain; (J.O.-V.); (J.M.d.l.T.); (S.S.)
| | - Sofía Salido
- Department of Inorganic and Organic Chemistry, Faculty of Experimental Sciences, Campus of International Excellence in Agri-Food (ceiA3), University of Jaén, 23071 Jaén, Spain; (J.O.-V.); (J.M.d.l.T.); (S.S.)
| |
Collapse
|
43
|
Reddy CN, Nuthakki VK, Sharma A, Malik S, Tabassum M, Kumar R, Choudhary S, Iqbal F, Tufail Z, Mondhe DM, Kumar A, Bharate SB. Synthesis and Biological Evaluation of Colchicine─Aryl/Alkyl Amine Hybrids as Potential Noncytotoxic Cholinesterase Inhibitors: Identification of SBN-284 as a Dual Inhibitor of Cholinesterases and NLRP3 Inflammasome. ACS Chem Neurosci 2024; 15:2779-2794. [PMID: 39056181 DOI: 10.1021/acschemneuro.4c00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024] Open
Abstract
Colchicine, one of the oldest anti-inflammatory natural products still used clinically, inhibits NF-κB signaling and NLRP3 inflammasome activation. Despite its cytotoxicity and narrow therapeutic range, colchicine continues to intrigue medicinal chemists exploring its anti-inflammatory potential. This study aimed to investigate the colchicine scaffold for its role in Alzheimer's disease by targeting neuroinflammation and cholinesterases. Molecular docking revealed that colchicine's hydrophobic trimethoxyphenyl framework can potentially bind to the peripheral anionic site of cholinesterases. Hybrid structures combining colchicine with aryl/alkyl amines were designed to bind both peripheral and catalytic sites of cholinesterases. We describe here the design, synthesis, and in vitro cytotoxicity evaluation of these colchicine-aryl/alkyl amine hybrids, along with their in silico interactions with the cholinesterase active site gorge. Nontoxic analogs demonstrating strong cholinesterase binding affinity were further evaluated for their anticholinesterase and antineuroinflammatory activities. The colchicine-donepezil hybrid, SBN-284 (3x), inhibited both acetylcholinesterase and butyrylcholinesterase as well as the NLRP3 inflammasome complex at low micromolar concentrations. It achieved this through noncompetitive inhibition, occupying the active site gorge and interacting with both peripheral and catalytic anionic sites of cholinesterases. Analog 3x was shown to cross the blood-brain barrier and exhibited no toxicity to neuronal cells, primary macrophages, or epithelial fR2 cells. These findings highlight the potential of this lead compound for further preclinical investigation as a promising anti-Alzheimer agent.
Collapse
Affiliation(s)
- Chilakala Nagarjuna Reddy
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vijay K Nuthakki
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ankita Sharma
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumera Malik
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Misbah Tabassum
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Rajesh Kumar
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Sushil Choudhary
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Fiza Iqbal
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Ziya Tufail
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Dilip M Mondhe
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Ajay Kumar
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Sandip B Bharate
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India
| |
Collapse
|
44
|
Chen M, Zhai L, Schønning K, Alpízar-Alpízar W, Larum O, Andersen LP, Holck S, Friis-Møller A. Protective Effects of Huo Xiang Zheng Qi Liquid on Clostridioides difficile Infection on C57BL/6 Mice. Microorganisms 2024; 12:1602. [PMID: 39203444 PMCID: PMC11356083 DOI: 10.3390/microorganisms12081602] [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: 06/25/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Clostridioides difficile-associated disease (CDAD) is a major healthcare-associated infection. New treatment options for CDAD are needed. A traditional Chinese medicinal formula, Huo Xiang Zheng Qi (HXZQ), was chosen to test against CDAD in a mouse model. METHODS C57BL/6 mice were challenged with C difficile (ATCC 43255) orally; then received saline; vancomycin 25 mg/kg; or HXZQ in two different concentrations twice daily for 5 days. The animals' body weight; clinical signs; and survival rates were registered daily. Fecal pellets from each animal were taken for PCR analysis as a control of infection. RESULTS 50% of the mice receiving saline died; 85.7% of the mice receiving vancomycin survived; 75% of the mice receiving HXZQ survived; and 87.5% of the mice receiving a 1:1 saline dilution of HXZQ survived. The HXZQ-treated groups were C. difficile PCR positive with loads less than that of the untreated mice. The weight loss in the vancomycin plus HXZQ 1:1 treated group; the vancomycin-treated group; and the untreated group were 3.08%, 4.06%, and 9.62%, respectively. CONCLUSIONS our results showed that HXZQ can protect mice from CDAD-related death as effectively as vancomycin and the combination of vancomycin and HXZQ may give even better protection.
Collapse
Affiliation(s)
- Ming Chen
- Department of Clinical Microbiology, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Lin Zhai
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Kristian Schønning
- Department of Clinical Microbiology, University Hospital of Copenhagen, Hvidovre, 2650 Hvidovre, Denmark
| | - Warner Alpízar-Alpízar
- The Finsen Laboratory, The Finsen Center, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Ole Larum
- The Finsen Laboratory, The Finsen Center, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Leif Percival Andersen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Susanne Holck
- Department of Pathology, University Hospital of Copenhagen, Hvidovre, 2650 Hvidovre, Denmark
| | - Alice Friis-Møller
- Department of Clinical Microbiology, University Hospital of Copenhagen, Hvidovre, 2650 Hvidovre, Denmark
| |
Collapse
|
45
|
Ahmad E, Athar A, Nimisha, Zia Q, Sharma AK, Sajid M, Bharadwaj M, Ansari MA, Saluja SS. Harnessing nature's potential: Alpinia galanga methanolic extract mediated green synthesis of silver nanoparticle, characterization and evaluation of anti-neoplastic activity. Bioprocess Biosyst Eng 2024; 47:1183-1196. [PMID: 38509420 DOI: 10.1007/s00449-024-02993-7] [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: 10/27/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
With the advent of nanotechnology, the treatment of cancer is changing from a conventional to a nanoparticle-based approach. Thus, developing nanoparticles to treat cancer is an area of immense importance. We prepared silver nanoparticles (AgNPs) from methanolic extract of Alpinia galanga rhizome and characterized them by UV-Vis spectrophotometry, Fourier transform Infrared (FTIR) spectroscopy, Zetasizer, and Transmission electron Microscopy (TEM). UV-Vis spectrophotometry absorption spectrum showed surface plasmon between 400 and 480 nm. FTIR spectrum analysis implies that various phytochemicals/secondary metabolites are involved in the reduction, caping, and stabilization of AgNPs. The Zetasier result suggests that the particles formed are small in size with a low polydispersity index (PDI), suggesting a narrow range of particle distribution. The TEM image suggests that the particles formed are mostly of spherical morphology with nearly 20-25 nm. Further, the selected area electron diffraction (SAED) image showed five electron diffraction rings, suggesting the polycrystalline nature of the particles. The nanoparticles showed high anticancer efficacy against cervical cancer (SiHa) cell lines. The nanostructures showed dose-dependent inhibition with 40% killing observed at 6.25 µg/mL dose. The study showed an eco-friendly and cost-effective approach to the synthesis of AgNPs and provided insight into the development of antioxidant and anticancer agents.
Collapse
Affiliation(s)
- Ejaj Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi-110002, India
| | - Alina Athar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi-110002, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi-110002, India
| | - Qamar Zia
- Department of Medical Laboratory Sciences, Majmaah University, Majmaah, Saudi Arabia
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi-110002, India
| | - Mohammed Sajid
- Division of Molecular Genetics & Biochemistry, Molecular Biology Group, ICMR-National Institute of Cancer Prevention & Research, Noida, Uttar Pradesh, India
| | - Mausumi Bharadwaj
- Division of Molecular Genetics & Biochemistry, Molecular Biology Group, ICMR-National Institute of Cancer Prevention & Research, Noida, Uttar Pradesh, India
| | | | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi-110002, India.
- Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, 110002, India.
| |
Collapse
|
46
|
Coverdale JPC, Bedford RA, Carter OWL, Cao S, Wills M, Sadler PJ. In-cell Catalysis by Tethered Organo-Osmium Complexes Generates Selectivity for Breast Cancer Cells. Chembiochem 2024; 25:e202400374. [PMID: 38785030 DOI: 10.1002/cbic.202400374] [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: 04/23/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Anticancer agents that exhibit catalytic mechanisms of action offer a unique multi-targeting strategy to overcome drug resistance. Nonetheless, many in-cell catalysts in development are hindered by deactivation by endogenous nucleophiles. We have synthesised a highly potent, stable Os-based 16-electron half-sandwich ('piano stool') catalyst by introducing a permanent covalent tether between the arene and chelated diamine ligand. This catalyst exhibits antiproliferative activity comparable to the clinical drug cisplatin towards triple-negative breast cancer cells and can overcome tamoxifen resistance. Speciation experiments revealed Os to be almost exclusively albumin-bound in the extracellular medium, while cellular accumulation studies identified an energy-dependent, protein-mediated Os accumulation pathway, consistent with albumin-mediated uptake. Importantly, the tethered Os complex was active for in-cell transfer hydrogenation catalysis, initiated by co-administration of a non-toxic dose of sodium formate as a source of hydride, indicating that the Os catalyst is delivered to the cytosol of cancer cells intact. The mechanism of action involves the generation of reactive oxygen species (ROS), thus exploiting the inherent redox vulnerability of cancer cells, accompanied by selectivity for cancerous cells over non-tumorigenic cells.
Collapse
Affiliation(s)
- J P C Coverdale
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - R A Bedford
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - O W L Carter
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - S Cao
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - M Wills
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - P J Sadler
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
47
|
Liz Belli Cassa Domingues E, Gonçalves-Santos E, Santana Caldas I, Vilela Gonçalves R, Caetano-da-Silva JE, Cardoso Santos E, Mól Pelinsari S, Figueiredo Diniz L, Dias Novaes R. Identification of host antioxidant effectors as thioridazine targets: Impact on cardiomyocytes infection and Trypanosoma cruzi-induced acute myocarditis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167264. [PMID: 38806073 DOI: 10.1016/j.bbadis.2024.167264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Phenothiazines inhibit antioxidant enzymes in trypanosomatids. However, potential interferences with host cell antioxidant defenses are central concerns in using these drugs to treat Trypanosoma cruzi-induced infectious myocarditis. Thus, the interaction of thioridazine (TDZ) with T. cruzi and cardiomyocytes antioxidant enzymes, and its impact on cardiomyocytes and cardiac infection was investigated in vitro and in vivo. Cardiomyocytes and trypomastigotes in culture, and mice treated with TDZ and benznidazole (Bz, reference antiparasitic drug) were submitted to microstructural, biochemical and molecular analyses. TDZ was more cytotoxic and less selective against T. cruzi than Bz in vitro. TDZ-pretreated cardiomyocytes developed increased infection rate, reactive oxygen species (ROS) production, lipid and protein oxidation; similar catalase (CAT) and superoxide dismutase (SOD) activity, and reduced glutathione's (peroxidase - GPx, S-transferase - GST, and reductase - GR) activity than infected untreated cells. TDZ attenuated trypanothione reductase activity in T. cruzi, and protein antioxidant capacity in cardiomyocytes, making these cells more susceptible to H2O2-based oxidative challenge. In vivo, TDZ potentiated heart parasitism, total ROS production, myocarditis, lipid and protein oxidation; as well as reduced GPx, GR, and GST activities compared to untreated mice. Benznidazole decreased heart parasitism, total ROS production, heart inflammation, lipid and protein oxidation in T. cruzi-infected mice. Our findings indicate that TDZ simultaneously interact with enzymatic antioxidant targets in cardiomyocytes and T. cruzi, potentiating the infection by inducing antioxidant fragility and increasing cardiomyocytes and heart susceptibility to parasitism, inflammation and oxidative damage.
Collapse
Affiliation(s)
- Elisa Liz Belli Cassa Domingues
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Ivo Santana Caldas
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Reggiani Vilela Gonçalves
- Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa, 36570-900, Minas Gerais, Brazil; Programa de Pós-Graduação em Biologia Celular e Estrutural, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - José Edson Caetano-da-Silva
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Eliziária Cardoso Santos
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina 39100-000, Minas Gerais, Brazil
| | - Silvania Mól Pelinsari
- Programa de Pós-Graduação em Biologia Celular e Estrutural, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Lívia Figueiredo Diniz
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Rômulo Dias Novaes
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa, 36570-900, Minas Gerais, Brazil.
| |
Collapse
|
48
|
Chen JH, Wei CM, Lin QY, Wang Z, Zhang FM, Shi MN, Lan WJ, Sun CG, Lin WJ, Ma WZ. Notopterygium Incisum Extract Promotes Apoptosis by Preventing the Degradation of BIM in Colorectal Cancer. Curr Med Sci 2024; 44:833-840. [PMID: 38967889 DOI: 10.1007/s11596-024-2883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/09/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Colorectal cancer (CRC), a prevalent malignancy worldwide, has prompted extensive research into anticancer drugs. Traditional Chinese medicinal materials offer promising avenues for cancer management due to their diverse pharmacological activities. This study investigated the effects of Notopterygium incisum, a traditional Chinese medicine named Qianghuo (QH), on CRC cells and the underlying mechanism. METHODS The sulforhodamine B assay and colony formation assay were employed to assess the effect of QH extract on the proliferation of CRC cell lines HCT116 and Caco-2. Propidium iodide (PI) staining was utilized to detect cell cycle progression, and PE Annexin V staining to detect apoptosis. Western blotting was conducted to examine the levels of apoptotic proteins, including B-cell lymphoma 2-interacting mediator of cell death (BIM), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (BAX) and cleaved caspase-3, as well as BIM stability after treatment with the protein synthesis inhibitor cycloheximide. The expression of BAX was suppressed using lentivirus-mediated shRNA to validate the involvement of the BIM/BAX axis in QH-induced apoptosis. The in vivo effects of QH extract on tumor growth were observed using a xenograft model. Lastly, APCMin+ mice were used to study the effects of QH extract on primary intestinal tumors. RESULTS QH extract exhibited significant in vitro anti-CRC activities evidenced by the inhibition of cell proliferation, perturbation of cell cycle progression, and induction of apoptosis. Mechanistically, QH extract significantly increased the stability of BIM proteins, which undergo rapid degradation under unstressed conditions. Knockdown of BAX, the downstream effector of BIM, significantly rescued QH-induced apoptosis. Furthermore, the in vitro effect of QH extract was recapitulated in vivo. QH extract significantly inhibited the tumor growth of HCT116 xenografts in nude mice and decreased the number of intestinal polyps in the APCMin+ mice. CONCLUSION QH extract promotes the apoptosis of CRC cells by preventing the degradation of BIM.
Collapse
Affiliation(s)
- Jun-He Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Cheng-Ming Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Qian-Yu Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Zi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Fu-Ming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Mei-Na Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chang-Gang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261053, China
| | - Wan-Jun Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Wen-Zhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
| |
Collapse
|
49
|
Silva A, Cassani L, Carpena M, Lourenço-Lopes C, Grosso C, Chamorro F, García-Pérez P, Carvalho A, Domingues VF, Barroso MF, Simal-Gandara J, Prieto MA. Exploring the Potential of Invasive Species Sargassum muticum: Microwave-Assisted Extraction Optimization and Bioactivity Profiling. Mar Drugs 2024; 22:352. [PMID: 39195468 DOI: 10.3390/md22080352] [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: 05/29/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Sargassum muticum (SM) poses a serious environmental issue since it is a fast-expanding invasive species occupying key areas of the European shoreline, disrupting the autochthonous algae species, and disturbing the ecosystem. This problem has concerned the general population and the scientific community. Nevertheless, as macroalgae are recognized as a source of bioactive molecules, the abundance of SM presents an opportunity as a raw material. In this work, response surface methodology (RSM) was applied as a tool for the optimization of the extraction of bioactive compounds from SM by microwave-assisted extraction (MAE). Five different parameters were used as target functions: yield, total phenolic content (TPC); and the antioxidant measurements of 2,2-diphenyl-1-picrylhydrazyl radical scavenging activity (DPPH), 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (ABTS), and β-carotene bleaching (BC). After the optimal extraction conditions were determined (time = 14.00 min; pressure = 11.03 bar; ethanol = 33.31%), the chemical composition and bioactivity of the optimum extract was evaluated to appraise its antioxidant capability to scavenge reactive species and as a potential antibacterial, antidiabetic, antiproliferation, and neuroprotective agent. The results lead to the conclusion that MAE crude extract has bioactive properties, being especially active as an antiproliferation agent and as a nitric oxide and superoxide radical scavenger.
Collapse
Affiliation(s)
- Aurora Silva
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr António Bernardino de Almeida 431, 4249-072 Porto, Portugal
| | - Lucia Cassani
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
| | - Maria Carpena
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
| | - Catarina Lourenço-Lopes
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
| | - Clara Grosso
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr António Bernardino de Almeida 431, 4249-072 Porto, Portugal
| | - Franklin Chamorro
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
| | - Pascual García-Pérez
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Via Emilia Parmense 84, 29122 Piacenza, Italy
| | - Ana Carvalho
- Escola Superior de Biotecnologia, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Valentina F Domingues
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr António Bernardino de Almeida 431, 4249-072 Porto, Portugal
| | - M Fátima Barroso
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Rua Dr António Bernardino de Almeida 431, 4249-072 Porto, Portugal
| | - Jesus Simal-Gandara
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
| | - Miguel A Prieto
- Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
50
|
Hublikar M, Kadu V, Edake N, Raut D, Shirame S, Ahmed MZ, Makam P, Ahmad MS, Meshram RJ, Bhosale R. Design, Synthesis, Anti-Cancer, Anti-inflammatory and In Silico Studies of 3-Substituted-2-Oxindole Derivatives. Chem Biodivers 2024:e202400844. [PMID: 39078869 DOI: 10.1002/cbdv.202400844] [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: 04/07/2024] [Accepted: 07/29/2024] [Indexed: 09/25/2024]
Abstract
This study focuses on the design and synthesis of 3-substituted-2-oxindole derivatives aimed at developing dual-active molecules with anti-cancer and anti-inflammatory properties. The molecules were designed with diverse structural and functional features while adhering to Lipinski, Veber, and Leeson criteria. Physicochemical properties were assessed using SWISSADME to ensure drug-likeness and favourable pharmacokinetics. Multistep synthetic procedures were employed for molecule synthesis. In vitro evaluations confirmed the dual activity of the derivatives, with specific emphasis on the significance of dialkyl aminomethyl substitutions for potency against various cell lines. 4 a exhibited GI50 value 3.00E-05 against MDA-MB-231, 4 b has shown GI50 value 2E-05 against MDA-MB-231, 4 c has shown GI50 value 6E-05 against VERO, 4 d has shown GI50 value 8E-05 each against both the MDA-MB-231 and MCF-7 and 4 e has shown GI50 values 2E-05 and 5E-05 each against both the MCF-7 and VERO. The analysis indicates that compounds 3 c (71.19 %), 3 e (66.84 %), and 3 g (63.04 %) exhibited significant anti-inflammatory activity. Additionally, in silico binding free energy analysis and interaction studies revealed significant correlations between in vitro and computational data, identifying compounds 4 d, 4 e, 3 b, 3 i, and 3 e as promising candidates. Key residues such as Glu917, Cys919, Lys920, Glu850, Lys838, and Asp1046 were found to play critical roles in ligand binding and kinase inhibition, providing valuable insights for designing potent VEGFR2 inhibitors. The Quantum Mechanics-based Independent Gradient Model analysis further highlighted the electronic interaction landscape, showing larger attractive peaks and higher electron density gradients for compounds 4 d and 4 e compared to Sunitinib, suggesting stronger and more diverse attractive forces. These findings support the potential of these compounds for further development and optimization in anticancer drug design.
Collapse
Affiliation(s)
- Mahesh Hublikar
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Vikas Kadu
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Nagesh Edake
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Dattatraya Raut
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Sachin Shirame
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| | - Mahammad Z Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Parameshwar Makam
- Department of Chemistry, School of Applied and Life Sciences, Uttaranchal University, Arcadia Grant, P.O. Chandanwari, Premnagar, Dehradun, Uttarakhand, 248007, India
| | - Md Sibgatullah Ahmad
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, Maharashtra, 248007, India
| | - Rohan J Meshram
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, Maharashtra, 248007, India
| | - Raghunath Bhosale
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur, Maharashtra, 413255, India
| |
Collapse
|