1
|
Wang R, Zeng M, Zhang B, Zhang Q, Xie S, Hu Y, Fan R, Wang M, Yu X, Zhang Y, Zheng X, Feng W. Epimedium sagittatum Maxim ameliorates adriamycin-induced nephropathy by restraining inflammation and apoptosis via the PI3K/AKT signaling pathway. Immun Inflamm Dis 2023; 11:e904. [PMID: 37382268 PMCID: PMC10266158 DOI: 10.1002/iid3.904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Modern pharmacological studies show that Epimedium sagittatum Maxim (EPI) has antioxidant, antiapoptotic, anti-inflammatory effects. However, the effects of EPI on adriamycin-induced nephropathy are unclear. AIM The main purpose of this study is to investigate the effects of EPI on adriamycin-induced nephropathy in rats. METHODS The chemical composition of EPI was detected by high performance liquid chromatography. Network pharmacology was used to collect the effects of EPI on adriamycin nephropathy; renal histological changes, podocyte injury, inflammatory factors, oxidative stress levels, apoptosis levels, and the PI3K/AKT signaling pathway were examined. Moreover, analyze the effects of icariin (the representative component of EPI) on adriamycin-induced apoptosis and PI3K/AKT signaling pathway of NRK-52e cells. RESULTS Network pharmacological results suggested that EPI may ameliorate adriamycin-induced nephropathy by inhibiting inflammatory response and regulating the PI3K/AKT signaling pathway. The experimental results showed that EPI could improve pathological injury, renal function, podocyte injury, and inhibit inflammation, oxidative stress, apoptosis in adriamycin-induced nephropathy rats through the PI3K/AKT signaling pathway. Furthermore, icariin inhibited adriamycin-induced mitochondrial apoptosis in NRK-52e cells. CONCLUSION This study suggested that EPI ameliorates adriamycin-induced nephropathy by reducing inflammation and apoptosis through the PI3K/AKT signaling pathway, icariin may be the pharmacodynamic substance basis for this effect.
Collapse
Affiliation(s)
- Ru Wang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| | - Mengnan Zeng
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| | - Beibei Zhang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| | - Qinqin Zhang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| | - Shuangshuang Xie
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| | - Yingbo Hu
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Ruyi Fan
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Mengya Wang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Xiao Yu
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Yuhan Zhang
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| | - Xiaoke Zheng
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| | - Weisheng Feng
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
| |
Collapse
|
2
|
Wang R, Yang B, Zhang B, Zhang Q, Cao B, Jia J, Liu M, Guo P, Zhang Y, Li X, Zheng X, Feng W. A new amide alkaloid induces the apoptosis of human melanoma A375 cells viainhibition of the STAT3 signaling pathway. NEW J CHEM 2023; 47:120-130. [DOI: 10.1039/d2nj04384j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
A new amide alkaloid (ZYL-01) can inhibit the activity of A375 cells by inducing cell apoptosisviainhibiting STAT3 signaling.
Collapse
Affiliation(s)
- Ru Wang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Bo Yang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Beibei Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Qinqin Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Bing Cao
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Jufang Jia
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Meng Liu
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Pengli Guo
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Yuhan Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Xiaokun Li
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Xiaoke Zheng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Weisheng Feng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou, 450046, China
| |
Collapse
|
3
|
Mello GHD, D'Ávila CMDS, Viana AR, Krause LMF, Cadoná FC. Cocoa presents cytotoxicity against melanoma cancer cell lines (A-375 e B16-F10) and improves chemotherapy activity by increasing oxidative stress. J Food Biochem 2022; 46:e14512. [PMID: 36332189 DOI: 10.1111/jfbc.14512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/05/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Melanoma frequently presents a poor chemotherapy response. In this scenario, investigations for new therapies are essential. Thus, cocoa is highlighted in this area since it presents many biological properties. This study investigated the anticarcinogenic activity of cocoa in melanoma cell lines (A-375 and B16-F10). Melanoma and fibroblast (HFF-1) cell lines were exposed to different concentrations of cocoa seeds (30 to 2000 ug/ml) at 24 and 72 h. Cocoa was also associated with paclitaxel IC50. We conducted viability, proliferation, and oxidative stress analyses. Our findings suggested that cocoa isolated, at almost all concentrations tested, was able to reduce viability and proliferation of B16-F10 cells and proliferation of A-375 cells via oxidative stress increasing. Also, cocoa caused no damage in fibroblast cells. Moreover, cocoa increased paclitaxel activity on A-375 by reducing cell proliferation and increasing oxidative stress. Therefore, the results highlight cocoa as a potent selective adjuvant anticancer agent against melanoma. PRACTICAL APPLICATIONS: In conclusion, more studies should be performed to deeply explore this remarkable action of cocoa as a an promising adjuvant to enhance chemotherapy.
Collapse
Affiliation(s)
- Gabriela Haas de Mello
- Post-Graduate Program in Health and Life Sciences, Franciscan University, Santa Maria, Brazil
| | | | | | | | - Francine Carla Cadoná
- Post-Graduate Program in Health and Life Sciences, Franciscan University, Santa Maria, Brazil
| |
Collapse
|
4
|
Oliveira WN, Alencar EN, Rocha HAO, Amaral-Machado L, Egito EST. Nanostructured systems increase the in vitro cytotoxic effect of bullfrog oil in human melanoma cells (A2058). Biomed Pharmacother 2021; 145:112438. [PMID: 34861632 DOI: 10.1016/j.biopha.2021.112438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/06/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
The aim of this work was to investigate the in vitro cytotoxic effect of previously developed nanocapsules, nanoemulsion, and microemulsion based on bullfrog oil (BFO) against human melanoma cells (A2058). The nanosystems were produced as described in previous studies and characterized according to droplet/particle distribution and zeta potential. The biocompatibility was evaluated by the determination of the hemolytic potential against human erythrocytes. The cytotoxicity assessment was based on MTT and cell death assays, determination of Reactive Oxygen Species (ROS) levels, and cell uptake. The nanosystems were successfully reproduced and showed hemolytic potential smaller than 10% at all oil concentrations (50 and 100 µg.mL-1) (p < 0.05). The MTT assay revealed that the nanosystems decreased the mitochondrial activity up to 92 ± 2% (p < 0.05). The study showed that the free BFO induced cell apoptosis, while all the nanostructured systems caused cell death by necrosis associated with a ROS overproduction. This can be related to the increased ability of the nanostructured systems to deliver the BFO across all cellular compartments (membrane, cytoplasm, and nucleus). Finally, these results elucidate the in vitro BFO nanosystems cytotoxic effect against human melanoma cells (A2058), revealing the emulsified ones as the most cytotoxic systems. Overall, the findings suggest that the safety and antineoplastic activity of these systems can be further investigated by in vivo studies.
Collapse
Affiliation(s)
- W N Oliveira
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - E N Alencar
- Graduate Program in Pharmaceutical Nanotechnology, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - H A O Rocha
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - L Amaral-Machado
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - E S T Egito
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil; Graduate Program in Pharmaceutical Nanotechnology, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
5
|
Ferri-Liposomes: Preformulation and Selective Cytotoxicity against A549 Lung Cancer Cells. Pharmaceutics 2021; 13:pharmaceutics13050712. [PMID: 34068129 PMCID: PMC8152733 DOI: 10.3390/pharmaceutics13050712] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
Liposomes have become successful nanostructured systems used in clinical practices. These vesicles are able to carry important drug loadings with noteworthy stability. The aim of this work was to develop iron oxide-loaded stealth liposomes as a prospective alternative for the treatment of lung cancer. In this study, citric acid iron oxide nanoparticles (IONPs-Ac) were synthesized and encapsulated in stealth liposomes. Their cytotoxicity and selectivity against lung tumor cells were assessed. Stealth liposomal vesicles, with relevant content of IONPs-Ac, named ferri-liposomes (SL-IONPs-Ac), were produced with an average size of 200 nm. They displayed important cytotoxicity in a human lung cancer cells model (A549 cells), even at low concentrations, whereas free IONPs-Ac displayed adequate biocompatibility. Nevertheless, the treatment at the same concentration of ferri-liposomes against HEK-293 cells, a normal human cell lineage, was not significantly cytotoxic, revealing a probable lung tumor selectiveness of the fabricated formulation. Furthermore, from the flow cytometry studies, it was possible to infer that ferri-liposomes were able to induce A549 tumor cells death through apoptosis/ferroptosis processes, evidenced by a significant reduction of the mitochondrial membrane potential.
Collapse
|
6
|
Aranha ESP, Portilho AJDS, Bentes de Sousa L, da Silva EL, Mesquita FP, Rocha WC, Araújo da Silva FM, Lima ES, Alves APNN, Koolen HHF, Montenegro RC, Vasconcellos MCD. 22β-hydroxytingenone induces apoptosis and suppresses invasiveness of melanoma cells by inhibiting MMP-9 activity and MAPK signaling. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113605. [PMID: 33232779 DOI: 10.1016/j.jep.2020.113605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/11/2020] [Accepted: 11/17/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 22β-hydroxytingenone (22-HTG) is a quinonemethide triterpene isolated from Salacia impressifolia (Miers) A. C. Smith (family Celastraceae), which has been used in traditional medicine to treat a variety of diseases, including dengue, renal infections, rheumatism and cancer. However, the anticancer effects of 22-HTG and the underlying molecular mechanisms in melanoma cells have not yet been elucidated. AIM OF THE STUDY The present study investigated apoptosis induction and antimetastatic potencial of 22-HTG in SK-MEL-28 human melanoma cells. MATERIALS AND METHODS First, the in vitro cytotoxic activity of 22-HTG in cultured cancer cells was evaluated. Then, cell viability was determined using the trypan blue assay in melanoma cells (SK-MEL-28), which was followed by cell cycle, annexin V-FITC/propidium iodide assays (Annexin/PI), as well as assays to evaluate mitochondrial membrane potential, production of reactive oxygen species (ROS) using flow cytometry. Fluorescence microscopy using acridine orange/ethidium bromide (AO/BE) staining was also performed. RT-qPCR was carried out to evaluate the expression of BRAF, NRAS, and KRAS genes. The anti-invasiveness potential of 22-HTG was evaluated in a three-dimensional (3D) model of reconstructed human skin. RESULTS 22-HTG reduced viability of SK-MEL-28 cells and caused morphological changes, as cell shrinkage, chromatin condensation, and nuclear fragmentation. Furthermore, 22-HTG caused apoptosis, which was demonstrated by increased staining with AO/BE and Annexin/PI. The apoptosis may have been caused by mitochondrial instability without the involvement of ROS production. The expression of BRAF, NRAS, and KRAS, which are important biomarkers in melanoma development, was reduced by the 22-HTG treatment. In the reconstructed skin model, 22-HTG was able to decrease the invasion capacity of melanoma cells in the dermis. CONCLUSIONS Our data indicate that 22-HTG has anti-tumorigenic properties against melanoma cells through the induction of cell cycle arrest, apoptosis and inhibition of invasiveness potential, as observed in the 3D model. As such, the results provide new insights for future work on the utilization of 22-HTG in malignant melanoma treatment.
Collapse
Affiliation(s)
- Elenn Suzany Pereira Aranha
- Faculty of Pharmaceutical Sciences, Post Graduate Program in Biodiversity and Biotechnology of the Amazon (Bionorte), Federal University of Amazonas, Manaus, Amazonas, 69080-900, Brazil.
| | | | - Leilane Bentes de Sousa
- Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, 69080-900, Brazil.
| | - Emerson Lucena da Silva
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, 60430-275, Brazil.
| | - Felipe Pantoja Mesquita
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, 60430-275, Brazil.
| | - Waldireny C Rocha
- Health and Biotechnology Institute, Federal University of Amazonas, Coari, Amazonas, 69460-000, Brazil.
| | | | - Emerson Silva Lima
- Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, 69080-900, Brazil.
| | | | | | - Raquel Carvalho Montenegro
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, 60430-275, Brazil.
| | | |
Collapse
|
7
|
Microemulsion systems: from the design and architecture to the building of a new delivery system for multiple-route drug delivery. Drug Deliv Transl Res 2020; 11:2108-2133. [PMID: 33164165 DOI: 10.1007/s13346-020-00872-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2020] [Indexed: 02/04/2023]
Abstract
Poorly soluble active pharmaceutical ingredients (APIs) create major problems in drug dosage form formulation resulting in significant delays in drug pharmaceutical screening, impairing the drug dosage form production. Aiming to minimize the use of excipients for increasing drug apparent solubility and, as a result, its bioavailability, exploration of innovative approaches is an earnest need. Microemulsion is an alternative drug delivery system that emerged as a valuable tool to achieve safe formulations for insoluble compounds and to improve their biopharmaceutical properties and pharmacokinetics. This review aims to present the state of the art of microemulsion systems, bringing an overview about their origin and how they can be properly produced and thoroughly characterized by different approaches. Furthermore, comments on regulatory issues regarding stability assessment and toxicity evaluation are discussed. The review concludes with a current opinion on microemulsion systems. The overall objective of this work was to describe all the potentialities of microemulsion systems as a drug carrier for therapeutic purposes, highlighting the unique features of this nanotechnological platform. Display Image.
Collapse
|
8
|
Remedying the Mitochondria to Cure Human Diseases by Natural Products. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5232614. [PMID: 32733635 PMCID: PMC7376439 DOI: 10.1155/2020/5232614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria are the ‘engine' of cells. Mitochondrial dysfunction is an important mechanism in many human diseases. Many natural products could remedy the mitochondria to alleviate mitochondria-involved diseases. In this review, we summarized the current knowledge of the relationship between the mitochondria and human diseases and the regulation of natural products to the mitochondria. We proposed that the development of mitochondrial regulators/nutrients from natural products to remedy mitochondrial dysfunction represents an attractive strategy for a mitochondria-involved disorder therapy. Moreover, investigating the mitochondrial regulation of natural products can potentiate the in-depth comprehension of the mechanism of action of natural products.
Collapse
|