1
|
Basiouni S, Abel N, Eisenreich W, May-Simera HL, Shehata AA. Structural Analysis of Cardanol and Its Biological Activities on Human Keratinocyte Cells. Metabolites 2025; 15:83. [PMID: 39997708 PMCID: PMC11857407 DOI: 10.3390/metabo15020083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Background/Objectives: Cashew nutshell liquid (CNSL) is obtained during the industrial processing of cashew nuts. It contains anacardic acid (2-hydroxy-6-n-pentadecylbenzoic acid) and cardanol (3-n-pentadecylphenol). Therefore, CNSL provides a rich source of phenolic lipids serving as natural antioxidants or precursors for industrial uses. Here, we have analyzed in detail a commercial sample of cardanol by nuclear magnetic resonance (NMR) spectroscopy and its biological activities in the human keratinocyte cell line (HaCaT cells). Methods: The cytotoxic effects, genotoxicity, cell proliferation, and healing properties on HaCaT cells were studied using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, comet assay, proliferation assay, and scratch assay, respectively. Additionally, the modulatory effect of cardanol on the cellular fatty acid profile of HaCaT cells was analyzed by gas chromatography. Results: NMR showed the structure of cardanol as a mixture of the 8'-monoene (42%), the 8',11'-diene (22%), and the 8',11',14'-triene (36%) for the pentadecyl side chain with all double bonds in Z configuration. The cytotoxic effects on HaCaT cells only occurred at high concentrations of cardanol (>10 µg/mL), which caused significant reductions in cell viability. Using the comet assay, a dose-dependent increase in DNA damage was found at concentrations above 10 µg/mL. Scratch assays revealed that cardanol achieved 99% wound closure of HaCaT cells treated with 1 µg/mL cardanol after 48 h. Cardanol at 1 and 0.1 µg/mL significantly enhanced HaCaT cell proliferation and promoted migration, contributing to accelerated wound healing processes. As shown by gas chromatography, 1 µg/mL cardanol increased the total amount of polyunsaturated fatty acids (PUFA), including ω-3, ω-6, and ω-9 fatty acids. Conclusions: Together, these findings suggest that concentrations of <10 µg/mL cardanol are safe and exhibit beneficial biological activities, particularly wound-healing effects on HaCaT cells. Further studies are necessary to explore additional potential applications of cardanol, to refine its formulations for clinical use, and to ensure its safety and action in other target cells and species.
Collapse
Affiliation(s)
- Shereen Basiouni
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany; (S.B.); (H.L.M.-S.)
| | - Nina Abel
- Structural Membrane Biochemistry, Bavarian NMR Center, Technical University of Munich (TUM), 85748 Garching, Germany; (N.A.); (A.A.S.)
| | - Wolfgang Eisenreich
- Structural Membrane Biochemistry, Bavarian NMR Center, Technical University of Munich (TUM), 85748 Garching, Germany; (N.A.); (A.A.S.)
| | - Helen L. May-Simera
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany; (S.B.); (H.L.M.-S.)
| | - Awad A. Shehata
- Structural Membrane Biochemistry, Bavarian NMR Center, Technical University of Munich (TUM), 85748 Garching, Germany; (N.A.); (A.A.S.)
| |
Collapse
|
2
|
Lu Y, Chen W, Xuan Y, Li X, Wu S, Wang H, Guo T, Wang C, Tian S, Li H, Lai D, Zhao W, Huang X, Zhao X, Wang B, Zhang X, Li H, Huang Y, Ma X. ATF4/NUPR1 axis promotes cancer cell survival and mediates immunosuppression in clear cell renal cell carcinoma. Discov Oncol 2024; 15:607. [PMID: 39480570 PMCID: PMC11528094 DOI: 10.1007/s12672-024-01485-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Cancer cells encounter unavoidable stress during tumor growth. The stress-induced transcription factor, activating transcription factor 4 (ATF4), has been reported to upregulate various adaptive genes involved in salvage pathways to alleviate stress and promote tumor progression. However, this effect is unknown in clear cell renal cell carcinoma (ccRCC). In this study, we found that ATF4 expression was remarkably upregulated in tumor tissues and associated with poor ccRCC outcomes. ATF4 depletion significantly impaired ccRCC cell proliferation, migration, and invasion in vitro and in vivo by inhibiting the AKT/mTOR and epithelial-mesenchymal transition (EMT)-related signaling pathway. RNA sequencing and functional studies identified nuclear protein 1 (NUPR1) as a key downstream target of ATF4 for repressing ferroptosis and promoting ccRCC cell survival. In addition, targeting ATF4 or pharmacological inhibition using NUPR1 inhibitor ZZW115 promoted antitumor immunity in syngeneic graft mouse models, represented by increased infiltration of CD4+ and CD8+ T cells. Furthermore, ZZW115 could improve the response to the PD-1 immune checkpoint blockade. The results demonstrate that the ATF4/NUPR1 signaling axis promotes ccRCC survival and facilitates tumor-mediated immunosuppression, providing a set of potential targets and prognostic indicators for ccRCC patients.
Collapse
Affiliation(s)
- Yongliang Lu
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Weihao Chen
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Yundong Xuan
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Xiubin Li
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Shengpan Wu
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Hanfeng Wang
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Tao Guo
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Chenfeng Wang
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Shuo Tian
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Huaikang Li
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Dong Lai
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Wenlei Zhao
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Xing Huang
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Xupeng Zhao
- School of Medicine, Nankai University, Weijin Road 94, Tianjin, 300071, China
| | - Baojun Wang
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Xu Zhang
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Hongzhao Li
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| | - Yan Huang
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| | - Xin Ma
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| |
Collapse
|
3
|
Elizondo-Luevano JH, Quintanilla-Licea R, Castillo-Hernández SL, Sánchez-García E, Bautista-Villarreal M, González-Meza GM, Gloria-Garza MA, Rodríguez-Luis OE, Kluz MI, Kačániová M. In Vitro Evaluation of Anti-Hemolytic and Cytotoxic Effects of Traditional Mexican Medicinal Plant Extracts on Human Erythrocytes and Cell Cultures. Life (Basel) 2024; 14:1176. [PMID: 39337959 PMCID: PMC11433374 DOI: 10.3390/life14091176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Plant extracts of fifteen plants of ethnomedicinal use in Mexico were analyzed to provide scientific knowledge of their medicinal properties through the evaluation of different biological activities such as anti-hemolytic, antioxidant, and cytotoxic effects in normal cells. Therefore, methanolic extracts were obtained from each of the plants by the Soxhlet extraction. The hemolytic activity in human erythrocytes was evaluated, as was their potential to protect the erythrocyte membrane against the 2,2'-azobis (2-methylpropionamidine) dihydrochloride (AAPH) and 1,1-diphenyl-2-picryl hydrazyl (DPPH) radicals. Finally, the toxicity of the extracts in normal cell cultures of African green monkey kidney cells (Vero) and peripheral blood mononuclear cells (PBMC) was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction method. Most of the extracts showed low hemolytic activity and high anti-hemolytic activity as well as high selectivity indices (SI) and antioxidant effects. Extracts of H. inuloides, J. dioica, and J. spicigera induced cell proliferation of the Vero cells. K. daigremontiana, A. adstringens, S. mexicanum, J. spicigera, L. tridentata, and M. tenuiflora extracts showed PBMC cell proliferation. In the present study, it was observed that the evaluated extracts did not present hemolytic activity, and some presented low toxicity when Vero and PBMC cell cultures were exposed. In conclusion, traditionally used plants possess beneficial health properties, and it is hoped that this study will serve as a basis for understanding the biological effects of traditionally used plants and may complement future studies.
Collapse
Affiliation(s)
- Joel H. Elizondo-Luevano
- Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Cd., San Nicolás de los Garza 66455, Nuevo León, Mexico; (R.Q.-L.); (S.L.C.-H.); (E.S.-G.); (M.B.-V.)
- Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain
| | - Ramiro Quintanilla-Licea
- Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Cd., San Nicolás de los Garza 66455, Nuevo León, Mexico; (R.Q.-L.); (S.L.C.-H.); (E.S.-G.); (M.B.-V.)
| | - Sandra L. Castillo-Hernández
- Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Cd., San Nicolás de los Garza 66455, Nuevo León, Mexico; (R.Q.-L.); (S.L.C.-H.); (E.S.-G.); (M.B.-V.)
| | - Eduardo Sánchez-García
- Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Cd., San Nicolás de los Garza 66455, Nuevo León, Mexico; (R.Q.-L.); (S.L.C.-H.); (E.S.-G.); (M.B.-V.)
| | - Minerva Bautista-Villarreal
- Faculty of Biological Sciences, Universidad Autónoma de Nuevo León, Cd., San Nicolás de los Garza 66455, Nuevo León, Mexico; (R.Q.-L.); (S.L.C.-H.); (E.S.-G.); (M.B.-V.)
| | - Georgia M. González-Meza
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey 64849, Nuevo León, Mexico;
| | - Marcela A. Gloria-Garza
- Faculty of Odontology, Universidad Autónoma de Nuevo León, Dr. Eduardo Aguirre Pequeño, Monterrey 64460, Nuevo León, Mexico; (M.A.G.-G.); (O.E.R.-L.)
| | - Osvelia E. Rodríguez-Luis
- Faculty of Odontology, Universidad Autónoma de Nuevo León, Dr. Eduardo Aguirre Pequeño, Monterrey 64460, Nuevo León, Mexico; (M.A.G.-G.); (O.E.R.-L.)
| | - Maciej Ireneusz Kluz
- School of Medical & Health Sciences, University of Economics and Human Sciences in Warsaw, Okopowa 59, 01 043 Warszawa, Poland;
| | - Miroslava Kačániová
- School of Medical & Health Sciences, University of Economics and Human Sciences in Warsaw, Okopowa 59, 01 043 Warszawa, Poland;
- Institute of Horticulture, Faculty of Horticulture and Landscape Engineering, Slovak University of Agriculture, Tr. A. Hlinku 2, 94976 Nitra, Slovakia
| |
Collapse
|
4
|
Oiram Filho F, Mitri MP, Zocolo GJ, Canuto KM, de Brito ES. Validation of a Method for Anacardic Acid Quantification in Cashew Peduncles via High-Performance Liquid Chromatography Coupled to a Diode-Array Detector. Foods 2023; 12:2759. [PMID: 37509851 PMCID: PMC10379927 DOI: 10.3390/foods12142759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The cashew peduncle has a high nutritional value and contains a wide variety of phenolic compounds. Among these, anacardic acids (AnAc) are biologically active components; however, they influence the cashew juice flavor and, consequently, its acceptance. This study validates a high-performance liquid chromatography method for quantifying the AnAc present in cashew peduncles, using a C18 reverse-phase column and a diode-array detector. The calibration curve obtained showed satisfactory precision for intraday (CV = 0.20%) and interday (CV = 0.29%) quantification, linearity (y = 2333.5x + 2956.2; r2 = 0.9979), repeatability with respect to retention time (CV = 0.45%) and area (CV = 0.30%), and selectivity, and possessed detection and quantification limits of 0.18 and 0.85 µg·mL-1, respectively. Different cashew clones containing AnAc were extracted and analyzed using the proposed method. A recovery of >90% was achieved using two sequential extractions. The total AnAc content ranged from 128.35 to 217.00 mg·100 g-1 in peduncle samples obtained from five different cashew clones.
Collapse
Affiliation(s)
- Francisco Oiram Filho
- Department of Chemical Engineering, Federal University of Ceará, Fortaleza 60440-900, CE, Brazil
| | - Morgana Pereira Mitri
- Department of Chemical Engineering, Federal University of Ceará, Fortaleza 60440-900, CE, Brazil
| | | | | | - Edy Sousa de Brito
- Embrapa Agroindústria Tropical, Pici, Fortaleza 60511-110, CE, Brazil
- Embrapa Alimentos e Territórios, Maceió 60020-050, AL, Brazil
| |
Collapse
|
5
|
Márton M, Bánhegyi G, Gyöngyösi N, Kálmán EÉ, Pettkó‐Szandtner A, Káldi K, Kapuy O. A systems biological analysis of the ATF4-GADD34-CHOP regulatory triangle upon endoplasmic reticulum stress. FEBS Open Bio 2022; 12:2065-2082. [PMID: 36097827 PMCID: PMC9623533 DOI: 10.1002/2211-5463.13484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 08/24/2022] [Accepted: 09/10/2022] [Indexed: 01/25/2023] Open
Abstract
Endoplasmic reticulum (ER) stress-dependent accumulation of incorrectly folded proteins leads to activation of the unfolded protein response. The role of the unfolded protein response (UPR) is to avoid cell damage and restore the homeostatic state by autophagy; however, excessive ER stress results in apoptosis. Here we investigated the ER stress-dependent feedback loops inside one of the UPR branches by focusing on PERK-induced ATF4 and its two targets, called CHOP and GADD34. Our goal was to qualitatively describe the dynamic behavior of the system by exploring the key regulatory motifs using both molecular and theoretical biological techniques. Using the HEK293T cell line as a model system, we confirmed that the life-or-death decision is strictly regulated. We investigated the dynamic characteristics of the crucial elements of the PERK pathway at both the RNA and protein level upon tolerable and excessive levels of ER stress. Of particular note, inhibition of GADD34 or CHOP resulted in various phenotypes upon high levels of ER stress. Our computer simulations suggest the existence of two new feedback loops inside the UPR. First, GADD34 seems to have a positive effect on ATF4 activity, while CHOP inhibits it. We claim that these newly described feedback loops ensure the fine-tuning of the ATF4-dependent stress response mechanism of the cell.
Collapse
Affiliation(s)
- Margita Márton
- Department of Molecular Biology at the Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Gábor Bánhegyi
- Department of Molecular Biology at the Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Norbert Gyöngyösi
- Department of Molecular Biology at the Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Eszter Éva Kálmán
- Department of Molecular Biology at the Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | | | - Krisztina Káldi
- Department of PhysiologySemmelweis UniversityBudapestHungary
| | - Orsolya Kapuy
- Department of Molecular Biology at the Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| |
Collapse
|
6
|
LncRNA H19 inhibits ER stress induced apoptosis and improves diabetic cardiomyopathy by regulating PI3K/AKT/mTOR axis. Aging (Albany NY) 2022; 14:6809-6828. [PMID: 36044268 PMCID: PMC9467416 DOI: 10.18632/aging.204256] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/17/2022] [Indexed: 02/06/2023]
Abstract
Objective: Extensive studies have shown that ERS may be implicated in the pathogenesis of DCM. We explored the therapeutic effects of lncRNAH19 on DCM and its effect on ERS-associated cardiomyocyte apoptosis. Methods: C57/BL-6j mice were randomly divided into 3 groups: non-DM group (controls), DM group (DCM), and lncRNAH19 overexpression group (DCM+H19 group). The effect of H19 on cardiac function was detected. The effect of H19 on cardiomyocyte apoptosis and cardiac fibrosis in DM was examined. Differentially expressed genes (DEGs) and activated pathways were examined by bioinformatics analysis. STRING database was applied to construct a PPI network using Cytoscape software. The expression of p-PERK, p-IRE1, ATF6, CHOP, cleaved caspase-3, -9, -12 and BAX proteins in cardiac tissue was used to determine the ERS-associated apoptotic indicators. We established the HG-stimulated inflammatory cell model. The expression of p-PERK and CHOP in HL-1 cells following HG was determined by immunofluorescence labeling. The effects of H19 on ERS and PI3K/AKT/mTOR pathway were also detected. Results: H19 improved left ventricular dysfunction in DM. H19 could reduce cardiomyocytes apoptosis and improve fibrosis in vivo. H19 could reduce the expression of p-PERK, p-IRE1α, ATF6, CHOP, cleaved caspase-3, cleaved caspase-9, cleaved caspase-12, and BAX proteins in cardiac tissues. Furthermore, H19 repressed oxidative stress, ERS and apoptosis in vitro. Moreover, the effect of H19 on ERS-associated apoptosis might be rescued by LY294002 (the specific inhibitor for PI3K and AKT). Conclusion: H19 attenuates DCM in DM and ROS, ERS-induced cardiomyocyte apoptosis, which is associated with the activation of PI3K/AKT/mTOR signaling pathway.
Collapse
|
7
|
Souza NDO, Cunha DA, Rodrigues NDS, Pereira AL, Medeiros EJT, Pinheiro ADA, de Vasconcelos MA, do Nascimento Neto LG, Bezerra TT, Mazzetto SE, Lomonaco D, Teixeira EH, Saboia VDPA. Cashew nut shell liquids: Antimicrobial compounds in prevention and control of the oral biofilms. Arch Oral Biol 2021; 133:105299. [PMID: 34735926 DOI: 10.1016/j.archoralbio.2021.105299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/23/2021] [Accepted: 10/24/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The aim was to evaluate the antibacterial and antibiofilm activity of natural (n-CNSL) and technical (t-CNSL) cashew nut shell liquid against streptococci and enterococci related to dental caries and chronic apical periodontitis, respectively. MATERIAL AND METHODS Minimum inhibitory concentrations (MIC) and minimal bactericidal concentration (MBC) were determined to assess the antimicrobial effect of both CNSLs (n-CSNL and t-CNSL) against S. oralis ATCC 10557, S. sobrinus ATCC 6715, S. parasanguinis ATCC 903, S. mutans UA 159 and E. faecalis ATCC 19433. The antibiofilm activity was evaluated by total biomass quantification, colony forming unit (CFU) counting and scanning electron microscopy (SEM). Furthermore, cytotoxic effect of the substances was evaluated on L929 and HaCat cell lines by MTS assay. RESULTS The n-CNSL and t-CNSL showed inhibitory and bactericidal effect against all strains tested in this study, with MIC and MBC values ranging from 1.5 to 25 μg/mL. Overall, both CNSLs showed significant reduction in biomass quantification and enumeration of biofilm-entrapped cells for the strains analyzed, in biofilm formation and preformed biofilms (p < 0.05). In biofilm inhibition assay, the t-CNSL and n-CNSL showed reduction in biomass and CFU number for all bacteria, except in cell viability of S. parasanguinis treated with t-CNSL (p > 0.05). Indeed, SEM images showed a reduction in the amount of biomass, bacterial cells and changes in cellular morphology of S. mutans. CONCLUSION In conclusion, both substances showed effective antibacterial and antibiofilm activity against the strains used in the study, except in viability of S. parasanguinis cells treated with t-CNSL.
Collapse
Affiliation(s)
- Nayara de Oliveira Souza
- Department of Restorative Dentistry, Federal University of Ceará, 949 Alexandre Baraúna St, Fortaleza, CE 60430-160, Brazil
| | - Diana Araújo Cunha
- Department of Restorative Dentistry, Federal University of Ceará, 949 Alexandre Baraúna St, Fortaleza, CE 60430-160, Brazil
| | - Nara de Sousa Rodrigues
- Department of Restorative Dentistry, University of Fortaleza, 1321 Washington Soares Ave., Fortaleza, CE 60811-905, Brazil
| | - Anna Luísa Pereira
- Department of Pathology and Forensic Medicine, Integrated Laboratory of Biomolecules, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil
| | - Elizabeth Jordanya Teixeira Medeiros
- Department of Pathology and Forensic Medicine, Integrated Laboratory of Biomolecules, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil
| | - Aryane de Azevedo Pinheiro
- Department of Pathology and Forensic Medicine, Integrated Laboratory of Biomolecules, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil
| | - Mayron Alves de Vasconcelos
- Department of Pathology and Forensic Medicine, Integrated Laboratory of Biomolecules, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil; Minas Gerais State University, Divinópolis, MG 35501-179, Brazil
| | - Luiz Gonzaga do Nascimento Neto
- Departamento do Núcleo Comum, Instituto Federal de Educação, Ciência e Tecnologia do Ceará, Campus Limoeiro do Norte, Limoeiro do Norte, CE 62930-000, Brazil
| | - Thayllan Teixeira Bezerra
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil
| | - Selma Elaine Mazzetto
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil
| | - Diego Lomonaco
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil
| | - Edson Holanda Teixeira
- Department of Pathology and Forensic Medicine, Integrated Laboratory of Biomolecules, Federal University of Ceará, 100 Five St., Fortaleza, CE 60355-636, Brazil.
| | - Vicente de Paulo Aragão Saboia
- Department of Restorative Dentistry, Federal University of Ceará, 949 Alexandre Baraúna St, Fortaleza, CE 60430-160, Brazil
| |
Collapse
|
8
|
Alfred N, Qian B, Qin X, Yin X, Prajapati M, Dou Y, Li Y, Zhang Z. Inhibition of eIF2α Phosphorylation by Peste des Petits Ruminant Virus Phosphoprotein Facilitates Viral Replication. Front Vet Sci 2021; 8:645571. [PMID: 34295932 PMCID: PMC8290123 DOI: 10.3389/fvets.2021.645571] [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: 12/23/2020] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Peste des petits ruminant virus (PPRV) causes a highly contagious disease in small ruminants. The molecular mechanism of PPRV replication and its interactions with hosts are poorly studied. In other paramyxoviruses, the viral phosphoprotein (P) has been associated with multiple functions for key biological processes such as the regulation of transcription, translation, and the control of cell cycle. Phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α) is an important process for gene regulation in host cells under stress, including viral infection. In the present study, molecular mechanisms associated with PPRV replication and viral interaction with host cells were investigated. We describe the ability of PPRV to dephosphorylate eIF2α and the potential of PPRV P protein to induce the host cellular growth arrest DNA damage protein (GADD34), which is known to be associated with eIF2α dephosphorylation. Furthermore, we observed that PPRV P protein alone could block PERK/eIF2α phosphorylation. We speculate that PPRV exploits eIF2α dephosphorylation to facilitate viral replication and that PPRV P protein is involved in this molecular mechanism. This work provides new insights into further understanding PPRV pathobiology and its viral/host interactions.
Collapse
Affiliation(s)
- Niyokwishimira Alfred
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Bang Qian
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiaodong Qin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangping Yin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Meera Prajapati
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Yongxi Dou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yanmin Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Zhidong Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| |
Collapse
|
9
|
Anacardic Acids from Amphipterygium adstringens Confer Cytoprotection against 5-Fluorouracil and Carboplatin Induced Blood Cell Toxicity While Increasing Antitumoral Activity and Survival in an Animal Model of Breast Cancer. Molecules 2021; 26:molecules26113241. [PMID: 34071241 PMCID: PMC8198955 DOI: 10.3390/molecules26113241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 11/17/2022] Open
Abstract
Amphipterygium adstringens (cuachalalate) contains anacardic acids (AAs) such as 6-pentadecyl salicylic acid (6SA) that show immunomodulatory and antitumor activity with minimal or no secondary adverse effects. By contrast, most chemotherapeutic agents, such as 5-fluorouracil (5-FU) and carboplatin (CbPt), induce myelosuppression and leukopenia. Here, we investigated the myeloprotective and antineoplastic potential of an AA extract or the 6SA as monotherapy or in combination with commonly used chemotherapeutic agents (5-FU and CbPt) to determine the cytoprotective action of 6SA on immune cells. Treatment of Balb/c breast tumor-bearing female mice with an AA mixture or 6SA did not induce the myelosuppression or leukopenia observed with 5-FU and CbPt. The co-administration of AA mixture or isolated 6SA with 5-FU or CbPt reduced the apoptosis of circulating blood cells and bone marrow cells. Treatment of 4T1 breast tumor-bearing mice with the AA mixture or 6SA reduced tumor growth and lung metastasis and increased the survival rate compared with monotherapies. An increased effect was observed in tumor reduction with the combination of 6SA and CbPt. In conclusion, AAs have important myeloprotective and antineoplastic effects, and they can improve the efficiency of chemotherapeutics, thereby protecting the organism against the toxic effects of drugs such as 5-FU and CbPt.
Collapse
|
10
|
Chen J, Zhou D, Kang J, Liu C, Huang R, Jiang Z, Liao Y, Liu A, Gao L, Song X, Zhao S, Chen Y, Wang H, Lan Z, Wang W, Guan H, Chen X, Huang J. ER stress modulates apoptosis in A431 cell subjected to EtNBSe-PDT via the PERK pathway. Photodiagnosis Photodyn Ther 2021; 34:102305. [PMID: 33901688 DOI: 10.1016/j.pdpdt.2021.102305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/18/2021] [Accepted: 04/19/2021] [Indexed: 01/01/2023]
Abstract
Photodynamic therapy (PDT) is a promising modality against various cancers including squamous cell carcinoma (SCC) with which the induction of apoptosis is an effective mechanism. Here, we initially describe the preclinical activity of 5-ethylamino-9-diethylaminobenzo [a] phenoselenazinium(EtNBSe)-mediated PDT treatment in SCC. Results of our studies suggest that EtNBSe-PDT provokes a cellular state of endoplasmic reticulum (ER) stress triggering the PERK/ eIF2α signaling pathway and induces the appearance of apoptosis in A431 cells at the meantime. With ER stress inhibitor 4-PBA or eIF2α inhibitor ISRIB, suppressing the EtNBSe-PDT induced ER stress substantially promotes apoptosis of A431 cells. Furthermore, we demonstrate that ATF4, whose expression is ER-stress-inducible and elevated in response to the PERK/eIF2α signaling pathway activation, contributes to cytoprotection against EtNBSe-PDT induced apoptosis. In a mouse model bearing A431 cells, EtNBSe shows intense phototoxicity and when associated with decreased ER stress, EtNBSe-PDT ameliorates tumor growth. Taken together, our study reveals an antagonistic activity of ER stress against EtNBSe-PDT treatment via inhibiting apoptosis in A431 cells. With further development, these results provide a proof-of-concept that downregulation of ER stress response has a therapeutic potential to improve EtNBSe-PDT sensitivity in SCC patients via the promotion of induced apoptosis.
Collapse
Affiliation(s)
- Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Dawei Zhou
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jian Kang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Chenxi Liu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Roujie Huang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhengqian Jiang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuxuan Liao
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - An Liu
- Department of Otorhinolaryngology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lihua Gao
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiangzhi Song
- College of Chemistry & Chemical Engineering, Central South University, Changsha, Hunan Province, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yihui Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hongyi Wang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zehao Lan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Weidong Wang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Haoyu Guan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jinhua Huang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
11
|
Acyl-Acyl Carrier Protein Desaturases and Plant Biotic Interactions. Cells 2021; 10:cells10030674. [PMID: 33803674 PMCID: PMC8002970 DOI: 10.3390/cells10030674] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 11/29/2022] Open
Abstract
Interactions between land plants and other organisms such as pathogens, pollinators, or symbionts usually involve a variety of specialized effectors participating in complex cross-talks between organisms. Fatty acids and their lipid derivatives play important roles in these biological interactions. While the transcriptional regulation of genes encoding acyl–acyl carrier protein (ACP) desaturases appears to be largely responsive to biotic stress, the different monounsaturated fatty acids produced by these enzymes were shown to take active part in plant biotic interactions and were assigned with specific functions intrinsically linked to the position of the carbon–carbon double bond within their acyl chain. For example, oleic acid, an omega-9 monounsaturated fatty acid produced by Δ9-stearoyl–ACP desaturases, participates in signal transduction pathways affecting plant immunity against pathogen infection. Myristoleic acid, an omega-5 monounsaturated fatty acid produced by Δ9-myristoyl–ACP desaturases, serves as a precursor for the biosynthesis of omega-5 anacardic acids that are active biocides against pests. Finally, different types of monounsaturated fatty acids synthesized in the labellum of orchids are used for the production of a variety of alkenes participating in the chemistry of sexual deception, hence favoring plant pollination by hymenopterans.
Collapse
|
12
|
Anacardic 6-pentadecyl salicylic acid induces apoptosis in breast cancer tumor cells, immunostimulation in the host and decreases blood toxic effects of taxol in an animal model. Toxicol Appl Pharmacol 2020; 410:115359. [PMID: 33290779 DOI: 10.1016/j.taap.2020.115359] [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] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 01/12/2023]
Abstract
Many antineoplastic agents induce myelosuppression and leukopenia as secondary effects in patients. The development of anticancer agents that simultaneously provoke antitumor immune response represents an important therapeutic advance. The administration of 6-pentadecyl salicylic acid (6SA) contributes to the antitumor immunity using 4T1 breast cancer cells in Balb/c female mice, with Taxol as a positive control and in cotreatment with 6SA (6SA + Taxol; CoT). Our results show that 6SA reduces tumor volume and size by inducing caspase-8-mediated apoptosis without reducing tumor infiltrated lymphocytes. Also, 6SA reduced lung metastasis and increased the proportion of immune cells in blood, lymph nodes and bone marrow; more evidently, in the proportion of tumor-infiltrated natural killer (NK) cells and cytotoxic T lymphocytes. Taxol reduces helper and cytotoxic lymphocytes causing systemic immunosuppression and myelosuppression in bone marrow, whereas 6SA does not decrease any immune cell subpopulations in circulating blood and lymph nodes. More importantly, the CoT decreased the Taxol-induced cytotoxicity in circulating T cells and bone marrow. Treatment with 6SA increases the secretion of IL-2, IL-12, GM-CSF, TNF-α and IFN-γ and significantly reduces IL-10 and IL-17 secretion, suggesting that the reduction of regulatory T cells and tumor-associated macrophages contribute to the host control of tumor development. Finally, 6SA has an effective antineoplastic activity against breast cancer cells in an immunocompetent animal, reduces the myelosuppression and leukopenia that Taxol produces, improves the antitumoral immunological microenvironment and increases the overall survival of the animals improving the quality of life of patients with cancer.
Collapse
|
13
|
Mukherjee D, Bercz LS, Torok MA, Mace TA. Regulation of cellular immunity by activating transcription factor 4. Immunol Lett 2020; 228:24-34. [PMID: 33002512 DOI: 10.1016/j.imlet.2020.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/10/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Activating transcription factor 4 (ATF4) is a DNA binding transcription factor belonging to the family of basic Leucine zipper proteins. ATF4 can be activated in response to multiple cellular stress signals including endoplasmic reticulum stress in the event of improper protein folding or oxidative stress because of mitochondrial dysfunction as well as hypoxia. There are multiple downstream targets of ATF4 that can coordinate the regulation between survival and apoptosis of a cell based on time and exposure to stress. ATF4, therefore, has a broad range of control that results in the modulation of immune cells of the innate and adaptive responses leading to regulation of the cellular immunity. Studies provide evidence that ATF4 can regulate immune cells such as macrophages, T cells, B cells, NK cells and dendritic cells contributing to progression of disease. Immune cells can be exposed to stressed environment in the event of a pathogen attack, infection, inflammation, or in the tumor microenvironment leading to increased ATF4 activity to regulate these responses. ATF4 can further control differentiation and maturation of different immune cell types becoming a determinant of effective immune regulation. Additionally, ATF4 has been heavily implicated in rendering effector immune cells dysfunctional that are used to target tumorigenesis. Therefore, there is a need to evaluate where the literature stands in understanding the overall role of ATF4 in regulating cellular immunity to identify therapeutic targets and generalized mechanisms for different disease progressions.
Collapse
Affiliation(s)
- Debasmita Mukherjee
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Lena S Bercz
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Molly A Torok
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Thomas A Mace
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States; Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
14
|
Önal B, Özen D, Demir B, Gezen Ak D, Dursun E, Demir C, Akkan AG, Özyazgan S. The Anti-Inflammatory Effects of Anacardic Acid on a TNF-α - Induced Human Saphenous Vein Endothelial Cell Culture Model. Curr Pharm Biotechnol 2020; 21:710-719. [PMID: 31692436 DOI: 10.2174/1389201020666191105154619] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Coronary bypass operations are commonly performed for the treatment of ischemic heart diseases. Coronary artery bypass surgery with autologous human saphenous vein maintains its importance as a commonly used therapy for advanced atherosclerosis. Vascular inflammation-related intimal hyperplasia and atherosclerotic progress have major roles in the pathogenesis of saphenous vein graft disease. METHODS In our study, we investigated the effect of anacardic acid (AA), which is a bioactive phytochemical in the shell of Anacardium occidentale, on atherosclerosis considering its inhibitory effect on NF-κB. We observed relative ICAM-1 and NF-κB mRNA levels by qRT-PCR method in a TNF-α- induced inflammation model of saphenous vein endothelial cell culture after 0.1, 0.5, 1 and 5 μM of AA were applied to the cells. In addition, protein levels of ICAM-1 and NF-κB were evaluated by immunofluorescent staining. The results were compared between different concentrations of AA, and also with the control group. RESULTS It was found that 5 μM, 1 μM and 0.5 μM of AA had toxic effects, while cytotoxicity decreased when 0.1 μM of AA was applied both alone and with TNF-α. When AA was applied with TNF-α, there was a decrease and suppression in NF-κB expression compared with the TNF-α group. TNF-α-induced ICAM-1 expression was significantly reduced more in the AA-applied group than in the TNF-α group. CONCLUSION In accordance with our results, it can be said that AA has a protective role in the pathogenesis of atherosclerosis and hence in saphenous vein graft disease.
Collapse
Affiliation(s)
- Burak Önal
- Department of Medical Pharmacology, Medical Faculty, Biruni University, Istanbul, Turkey
| | - Deniz Özen
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Bülent Demir
- Department of Cardiology, Bakirkoy Dr Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Duygu Gezen Ak
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Erdinç Dursun
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Caner Demir
- Research and Development Department, Tayf Biotechnology, Istanbul, Turkey
| | - Ahmet Gökhan Akkan
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sibel Özyazgan
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
15
|
Gomes Júnior A, Islam MT, Nicolau LAD, de Souza LKM, Araújo TDS, Lopes de Oliveira GA, de Melo Nogueira K, da Silva Lopes L, Medeiros JVR, Mubarak MS, Melo-Cavalcante AAC. Anti-Inflammatory, Antinociceptive, and Antioxidant Properties of Anacardic Acid in Experimental Models. ACS OMEGA 2020; 5:19506-19515. [PMID: 32803044 PMCID: PMC7424580 DOI: 10.1021/acsomega.0c01775] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/09/2020] [Indexed: 06/11/2023]
Abstract
Anacardic acid (AA), a compound extracted from cashew nut liquid, exhibits numerous pharmacological activities. The aim of the current investigation was to assess the anti-inflammatory, antinociceptive, and antioxidant activities of AA in mouse models. For this, Swiss albino mice were pretreated with AA (10, 25, 50 mg/kg, intraperitoneally, ip) 30 min prior to the administration of carrageenan, as well as 25 mg/kg of prostaglandin E2, dextran, histamine, and compound 48/80. The antinociceptive activity was evaluated by formalin, abdominal, and hot plate tests, using antagonist of opioid receptors (naloxene, 3 mg/kg, ip) to identify antinociceptive mechanisms. Results from this study revealed that AA at 25 mg/kg inhibits carrageenan-induced edema. In addition, AA at 25 mg/kg reduced edema and leukocyte and neutrophilic migration to the intraperitoneal cavity, diminished myeloperoxidase activity and malondialdehyde concentration, and increased the levels of reduced glutathione. In nociceptive tests, it also decreased licking, abdominal writhing, and latency to thermal stimulation, possibly via interaction with opioid receptors. Taken together, these results indicate that AA exhibits anti-inflammatory and antinociceptive actions and also reduces oxidative stress in acute experimental models, suggesting AA as a promising compound in the pharmaceutical arena.
Collapse
Affiliation(s)
- Antonio
Luiz Gomes Júnior
- RENORBIO
- Post-Graduation Program in Biotechnology, Federal University of Piauí, Teresina 64049-550, Piauí, Brazil
- LAPNEX
- Laboratory of Research in Experimental Neurochemistry of Post-Graduation
Program in Pharmaceutical Sciences, Federal
University of Piauí, Teresina CEP 64049-550, Piauí, Brazil
- LAPGENIC
- Laboratory of Research in Genetic Toxicology of Post-Graduation
Program in Pharmaceutical Sciences, Federal
University of Piauí, Teresina 64049-550, Piauí, Brazil
| | - Muhammad Torequl Islam
- Laboratory
of Theoretical and Computational Biophysics, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
- Faculty
of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Lucas Antonio Duarte Nicolau
- LAFFEX -
Laboratory of Experimental Physiopharmacology in Biotechnology and
Biodiversity Center Research (BIOTEC), Federal
University of Piauí-CMRV, Parnaíba 64202-020, Piauí, Brazil
| | - Luan Kevin Miranda de Souza
- RENORBIO
- Post-Graduation Program in Biotechnology, Federal University of Piauí, Teresina 64049-550, Piauí, Brazil
- LAFFEX -
Laboratory of Experimental Physiopharmacology in Biotechnology and
Biodiversity Center Research (BIOTEC), Federal
University of Piauí-CMRV, Parnaíba 64202-020, Piauí, Brazil
| | - Tiago de Souza
Lopes Araújo
- RENORBIO
- Post-Graduation Program in Biotechnology, Federal University of Piauí, Teresina 64049-550, Piauí, Brazil
- LAFFEX -
Laboratory of Experimental Physiopharmacology in Biotechnology and
Biodiversity Center Research (BIOTEC), Federal
University of Piauí-CMRV, Parnaíba 64202-020, Piauí, Brazil
| | - Guilherme Antônio Lopes de Oliveira
- RENORBIO
- Post-Graduation Program in Biotechnology, Federal University of Piauí, Teresina 64049-550, Piauí, Brazil
- LAPNEX
- Laboratory of Research in Experimental Neurochemistry of Post-Graduation
Program in Pharmaceutical Sciences, Federal
University of Piauí, Teresina CEP 64049-550, Piauí, Brazil
- LAFFEX -
Laboratory of Experimental Physiopharmacology in Biotechnology and
Biodiversity Center Research (BIOTEC), Federal
University of Piauí-CMRV, Parnaíba 64202-020, Piauí, Brazil
| | - Kerolayne de Melo Nogueira
- LAFFEX -
Laboratory of Experimental Physiopharmacology in Biotechnology and
Biodiversity Center Research (BIOTEC), Federal
University of Piauí-CMRV, Parnaíba 64202-020, Piauí, Brazil
| | - Luciano da Silva Lopes
- LAPNEX
- Laboratory of Research in Experimental Neurochemistry of Post-Graduation
Program in Pharmaceutical Sciences, Federal
University of Piauí, Teresina CEP 64049-550, Piauí, Brazil
| | - Jand-Venes Rolim Medeiros
- RENORBIO
- Post-Graduation Program in Biotechnology, Federal University of Piauí, Teresina 64049-550, Piauí, Brazil
- LAFFEX -
Laboratory of Experimental Physiopharmacology in Biotechnology and
Biodiversity Center Research (BIOTEC), Federal
University of Piauí-CMRV, Parnaíba 64202-020, Piauí, Brazil
| | | | - Ana Amélia
de Carvalho Melo-Cavalcante
- RENORBIO
- Post-Graduation Program in Biotechnology, Federal University of Piauí, Teresina 64049-550, Piauí, Brazil
- LAPGENIC
- Laboratory of Research in Genetic Toxicology of Post-Graduation
Program in Pharmaceutical Sciences, Federal
University of Piauí, Teresina 64049-550, Piauí, Brazil
| |
Collapse
|
16
|
Osman SM, Abdel-Megied AM, Zain Eldain MH, Haleema S, Gopinath C, Sumalekshmy SA, Aboul-Enein HY. A highly sensitive GC-MS method for simultaneous determination of anacardic acids in cashew (Anacardium occidentale) nut shell oil in the presence of other phenolic lipid derivatives. Biomed Chromatogr 2019; 33:e4659. [PMID: 31325174 DOI: 10.1002/bmc.4659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 11/09/2022]
Abstract
The commercial value of cashew nut shell liquid (CNSL) has become a cornerstone of the agrowaste industry. It is the by-product of the cashew industry and has an 1/8 inch thickness of soft honeycomb structure. CNSL contains phenolic lipids with aliphatic chains such as anacardic acid, cardanol, cardol and methyl cardol, and their derivatives. The developed GC-MS method is rapid, accurate and selective using a selected derivatizing reagent, namely N-methyl-N-(trimethylsilyl)-trifluoroacetamide that was previously diluted 1:1% with anhydrous pyridine. The proposed GC-MS method was applied for the analysis of different CNSL samples. The results showed that all classes of CNSL compounds were detected. The four alkyl phenols were detected with their different alkyl sidechains without any interference. This method is also specified for the detection of fatty acids of saturated and unsaturated chains. Silylation did not cause any alteration in the chemical structure of CNSL compounds regardless of esterification action. Silylation is considered a safe derivatizing agent compatible with GC chromatography and specific for all volatile and nonvolatile polar and nonpolar CNSL compounds that could be detected in CNSL samples.
Collapse
Affiliation(s)
- Samir M Osman
- Pharmacognosy Department, Faculty of Pharmacy, October 6 University, 6th October City, Egypt
| | - Ahmed M Abdel-Megied
- Pharmaceutical Analytical Chemistry Department., Faculty of Pharmacy, Kafrelshekh University, Kafrelsheikh City, Egypt
| | - Mohammed H Zain Eldain
- Pharmacognosy Department, Faculty of Pharmacy, October 6 University, 6th October City, Egypt
| | - Simimole Haleema
- Department of Chemistry, T.K.M. College of Arts and Science, Kollam, Kerala, India
| | - Chithra Gopinath
- Department of Chemistry, T.K.M. College of Arts and Science, Kollam, Kerala, India
| | | | - Hassan Y Aboul-Enein
- Pharmaceutical and Medicinal Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), Dokki, Cairo, Egypt
| |
Collapse
|
17
|
Oiram Filho F, Zocolo GJ, Canuto KM, Silva Junior IJD, Brito ES. Productivity of a preparative high‐performance liquid chromatography isolation of anacardic acids from cashew nut shell liquid. SEPARATION SCIENCE PLUS 2019. [DOI: 10.1002/sscp.201900014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
| | | | | | | | - Edy S. Brito
- Embrapa Agroindústria Tropical Fortaleza CE Brazil
| |
Collapse
|
18
|
Xia X, Huang C, Liao Y, Liu Y, He J, Guo Z, Jiang L, Wang X, Liu J, Huang H. Inhibition of USP14 enhances the sensitivity of breast cancer to enzalutamide. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:220. [PMID: 31126320 PMCID: PMC6534920 DOI: 10.1186/s13046-019-1227-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/13/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Androgen receptor (AR) is expressed in approximately 70% of breast tumors. Recent studies increasingly support AR as a potential therapeutic target of AR-positive breast cancer. We have previously reported that deubiquitinase USP14 stabilizes AR proteins by deubiquitination and USP14 inhibition results in inhibition of cell growth and tumor progression in AR-positive prostate cancer and breast cancer. The current study aims to explore the anticancer effect of a treatment combining AR antagonist enzalutamide with USP14 inhibition on breast cancer cells. METHODS The combining effects of enzalutamide and USP14 inhibition on breast cancer cell proliferation and apoptosis and associated cell signaling were evaluated in vitro and in vivo. RESULTS USP14 inhibition via administration of IU1 or USP14-specific siRNA/shRNA enhanced cell growth inhibition and apoptosis induction by enzalutamide in breast cancer cell lines in vitro and in vivo. Additionally, the combination of enzalutamide with USP14 inhibition/knockdown induced significant downregulation of AR proteins and suppression of AR-related signaling pathways, including Wnt/β-catenin and PI3K/AKT pathways. Moreover, AKT inhibition via MK2206 increased the antiproliferative and proapoptotic effects of enzalutamide+IU1 combined treatment. CONCLUSION Collectively, our data suggest that USP14 inhibition in combination with enzalutamide represents a potentially new therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Xiaohong Xia
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Chuyi Huang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yuning Liao
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yuan Liu
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Jinchan He
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Zhiqiang Guo
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Lili Jiang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, 57069, USA
| | - Jinbao Liu
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| | - Hongbiao Huang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| |
Collapse
|
19
|
Xie L, Chen Y, Chen J, Zhang H, Liao Y, Zhou Y, Zhou L, Qing C. Anti-tumor effects and mechanism of GA-13315, a novel gibberellin derivative, in human lung adenocarcinoma: an in vitro and in vivo study. Cell Mol Biol Lett 2019; 24:6. [PMID: 30651744 PMCID: PMC6327519 DOI: 10.1186/s11658-018-0126-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/03/2018] [Indexed: 12/11/2022] Open
Abstract
Objective To investigate the anti-tumor effects and the mechanism of the compound 13-chlorine-3, 15-dioxy-gibberellic acid methyl ester (GA-13315) in lung adenocarcinoma in vitro and in vivo. Methods The antiproliferative effect of GA-13313 on the A549 cell line was determined by MTT (3-[4, 5-dimethylthiazol-2-yl]-2, 5 diphenyl tetrazolium bromide) assay. A xenograft model of A549 was established to evaluate the anti-tumor effect and histopathological examination was performed to assess the toxicity of GA-13315. Apoptosis was detected by TUNEL staining in tissues and flow cytometry in cells; activation of caspase-3, caspase-8 and caspase-9 was evaluated by immunohistochemical analysis; protein levels of Bcl-2-associated X protein (Bax), B-cell lymphoma-2 (Bcl-2), caspase-4, activating transcription factor 4 (ATF4), glucose-regulated protein 78 (GRP78) and growth arrest and DNA damage-inducible gene 153 (GADD153) were determined by western blotting. Mitochondrial membrane potential (MMP) was measured by the JC-1 fluorescence probe. Results Our results showed that GA-13315 exhibited potent, dose- and time-dependent anti-proliferative activity, and the IC50 values were 37.43 ± 2.73, 28.08 ± 7.76 and 19.29 ± 7.61 μM at 24, 48, and 72 h, respectively. The xenograft experiment revealed that tumor weight and volume were significantly decreased after GA-13315 3 mg/kg and 9 mg/kg (P < 0.05) treatment, and GA-13315 had low toxicity in bone marrow, kidney and colon tissues. GA-13315 triggered remarkable apoptosis in A549 cells at the concentration of 25.6 μM and 32 μM (P < 0.05) and activated caspase-3, − 8 and − 9. Moreover, GA-13315 induced apoptosis through the mitochondrial apoptosis pathway by elevating the Bax/Bcl-2 ratio, releasing cytochrome c and activating caspase-9 in A549 cells. In the endoplasmic reticulum apoptosis pathway, the levels of caspase-4, ATF4, GRP78 and GADD153 were markedly upregulated. Conclusions This study suggests that GA-13315 can be considered as a promising chemotherapeutic agent with anticancer activity in treatment of lung cancer in future.
Collapse
Affiliation(s)
- Lin Xie
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Yajuan Chen
- 2School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Cancer Hospital of Yunnan Province, Kunming Medical University, NO.1168, West Chunrong Road, Chenggong Developing Area, Kunming, 650031 China
| | - Jingbo Chen
- 3Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Hongbin Zhang
- 3Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Yedan Liao
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Yonghong Zhou
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Ling Zhou
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Chen Qing
- 2School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Cancer Hospital of Yunnan Province, Kunming Medical University, NO.1168, West Chunrong Road, Chenggong Developing Area, Kunming, 650031 China
| |
Collapse
|
20
|
Al-Hrout A, Chaiboonchoe A, Khraiwesh B, Murali C, Baig B, El-Awady R, Tarazi H, Alzahmi A, Nelson DR, Greish YE, Ramadan W, Salehi-Ashtiani K, Amin A. Safranal induces DNA double-strand breakage and ER-stress-mediated cell death in hepatocellular carcinoma cells. Sci Rep 2018; 8:16951. [PMID: 30446676 PMCID: PMC6240095 DOI: 10.1038/s41598-018-34855-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Poor prognoses remain the most challenging aspect of hepatocellular carcinoma (HCC) therapy. Consequently, alternative therapeutics are essential to control HCC. This study investigated the anticancer effects of safranal against HCC using in vitro, in silico, and network analyses. Cell cycle and immunoblot analyses of key regulators of cell cycle, DNA damage repair and apoptosis demonstrated unique safranal-mediated cell cycle arrest at G2/M phase at 6 and 12 h, and at S-phase at 24 h, and a pronounced effect on DNA damage machinery. Safranal also showed pro-apoptotic effect through activation of both intrinsic and extrinsic initiator caspases; indicating ER stress-mediated apoptosis. Gene set enrichment analysis provided consistent findings where UPR is among the top terms of up-regulated genes in response to safranal treatment. Thus, proteins involved in ER stress were regulated through safranal treatment to induce UPR in HepG2 cells.
Collapse
Affiliation(s)
- Ala'a Al-Hrout
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE
| | - Amphun Chaiboonchoe
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Basel Khraiwesh
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
- Center for Genomics and Systems Biology (CGSB), Division of Science, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Chandraprabha Murali
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE
| | - Badriya Baig
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE
| | - Raafat El-Awady
- College of Pharmacy and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Hamadeh Tarazi
- College of Pharmacy and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Amnah Alzahmi
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - David R Nelson
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | | | - Wafaa Ramadan
- College of Pharmacy and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Kourosh Salehi-Ashtiani
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE.
- Center for Genomics and Systems Biology (CGSB), Division of Science, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE.
| | - Amr Amin
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE.
- Zoology Department, Cairo University, Giza, Egypt.
| |
Collapse
|
21
|
Targeting proteasome-associated deubiquitinases as a novel strategy for the treatment of estrogen receptor-positive breast cancer. Oncogenesis 2018; 7:75. [PMID: 30250021 PMCID: PMC6155249 DOI: 10.1038/s41389-018-0086-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/05/2018] [Accepted: 08/31/2018] [Indexed: 12/21/2022] Open
Abstract
Estrogen receptor α (ERα) is expressed in ~67% of breast cancers and is critical to their proliferation and progression. The expression of ERα is regarded as a major prognostic marker, making it a meaningful target to treat breast cancer (BCa). However, hormone receptor-positive BCa was sometimes irresponsive or even resistant to classic anti-hormonal therapies (e.g., fulvestrant and tamoxifen). Hence, novel anti-endocrine therapies are urgent for ERα+ BCa. A phase II study suggested that bortezomib, an inhibitor blocking the activity of 20 S proteasomes, intervenes in cancer progression for anti-endocrine therapy in BCa. Here we report that proteasome-associated deubiquitinases (USP14 and UCHL5) inhibitors b-AP15 and platinum pyrithione (PtPT) induce growth inhibition in ERα+ BCa cells. Further studies show that these inhibitors induce cell cycle arrest and apoptosis associated with caspase activation, endoplasmic reticulum (ER) stress and the downregulation of ERα. Moreover, we suggest that b-AP15 and PtPT block ERα signaling via enhancing the ubiquitin-mediated degradation of ERα and inhibiting the transcription of ERα. Collectively, these findings demonstrate that proteasome-associated deubiquitinases inhibitors b-AP15 and PtPT may have the potential to treat BCa resistant to anti-hormonal therapy.
Collapse
|
22
|
Oiram Filho F, Alcântra DB, Rodrigues THS, Alexandre E Silva LM, de Oliveira Silva E, Zocolo GJ, de Brito ES. Development and Validation of a Reversed Phase HPLC Method for Determination of Anacardic Acids in Cashew (Anacardium occidentale) Nut Shell Liquid. J Chromatogr Sci 2018; 56:300-306. [PMID: 29300927 DOI: 10.1093/chromsci/bmx111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 12/06/2017] [Indexed: 11/12/2022]
Abstract
Cashew nut shell liquid (CNSL) contains phenolic lipids with aliphatic chains that are of commercial interest. In this work, a chromatographic method was developed to monitor and quantify anacardic acids (AnAc) in CNSL. Samples containing AnAc were analyzed on a high-performance liquid chromatograph coupled to a diode array detector, equipped with a reversed phase C18 (150 × 4.6 mm × 5 μm) column using acetonitrile and water as the mobile phase both acidified with acetic acid to pH 3.0 in an isocratic mode (80:20:1). The chromatographic method showed adequate selectivity, as it could clearly separate the different AnAc. To validate this method, AnAc triene was used as an external standard at seven different concentrations varying from 50 to 1,000 μg mL-1. The Student's t-test and F-test were applied to ensure high confidence for the obtained data from the analytical calibration curve. The results were satisfactory with respect to intra-day (relative standard deviation (RSD) = 0.60%) and inter-day (RSD = 0.67%) precision, linearity (y = 2,670.8x - 26,949, r2 > 0.9998), system suitability for retention time (RSD = 1.02%), area under the curve (RSD = 0.24%), selectivity and limits of detection (19.8 μg mg-1) and quantification (60.2 μg mg-1). The developed chromatographic method was applied for the analysis of different CNSL samples, and it was deemed suitable for the quantification of AnAc.
Collapse
Affiliation(s)
- Francisco Oiram Filho
- Department of Chemical Engineering, UFC, Federal University of Ceará, Campus do Pici, Bloco 709, 60455-760, Fortaleza, CE, Brazil
| | - Daniel Barbosa Alcântra
- Department of Chemistry, UFC, Federal University of Ceará, Campus do Pici, 60455-760, Fortaleza, CE, Brazil
| | | | | | | | - Guilherme Julião Zocolo
- Embrapa Agroindústria Tropical, R Dra Sara Mesquita, 2270, Pici, 60511-110, Fortaleza, CE, Brazil
| | - Edy Sousa de Brito
- Embrapa Agroindústria Tropical, R Dra Sara Mesquita, 2270, Pici, 60511-110, Fortaleza, CE, Brazil
| |
Collapse
|
23
|
Liao Y, Xia X, Liu N, Cai J, Guo Z, Li Y, Jiang L, Dou QP, Tang D, Huang H, Liu J. Growth arrest and apoptosis induction in androgen receptor-positive human breast cancer cells by inhibition of USP14-mediated androgen receptor deubiquitination. Oncogene 2018; 37:1896-1910. [PMID: 29353883 PMCID: PMC5886989 DOI: 10.1038/s41388-017-0069-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/23/2017] [Accepted: 09/19/2017] [Indexed: 11/10/2022]
Abstract
It has been well known that androgen receptor (AR) is critical to prostate cancer development and progression. It has also been documented that AR is expressed in more than 60% of breast tumors, which promotes the growth of estrogen receptor-negative (ER-)/AR-positive (AR+) breast cancer cells. Thus, AR might be a potential therapeutic target for AR-positive/ER-negative breast cancer patients. Previously we reported that in prostate cancer cells proteasome-associated deubiquitinase ubiquitin-specific protease 14 (USP14) stabilized AR protein level by removing its ubiquitin chain. In the current study, we studied the USP14-AR protein interaction and cell proliferation status after USP14 reduction or inhibition in breast cancer cells, and our results support the conclusion that targeting USP14 is a novel strategy for treating AR-responsive breast cancer. We found that inhibition of USP14 accelerated the K48-ubiquitination and proteasome-mediated degradation of AR protein. Additionally, both genetic and pharmacological inhibition of USP14 significantly suppressed cell proliferation in AR-responsive breast cancer cells by blocking G0/G1 to S phase transition and inducing apoptosis. Moreover, AR overexpression inhibited USP14 inhibition-induced events, suggesting that AR deubiquitination by USP14 is critical for breast cancer growth and USP14 inhibition is a possible strategy to treat AR-positive breast cancer.
Collapse
Affiliation(s)
- Yuning Liao
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohong Xia
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianyu Cai
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiqiang Guo
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanling Li
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lili Jiang
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Q Ping Dou
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
- Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Daolin Tang
- The Third Affiliated Hospital, Center for DAMP Biology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Protein Modification and Degradation Laboratory, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hongbiao Huang
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Jinbao Liu
- Affiliated Cancer Hospital of Guangzhou Medical University; Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
24
|
In Vitro Bioactivity of Methanolic Extracts from Amphipterygium adstringens (Schltdl.) Schiede ex Standl., Chenopodium ambrosioides L., Cirsium mexicanum DC., Eryngium carlinae F. Delaroche, and Pithecellobium dulce (Roxb.) Benth. Used in Traditional Medicine in Mexico. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3610364. [PMID: 29681972 PMCID: PMC5851410 DOI: 10.1155/2018/3610364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
Seven out of eight methanolic extracts from five plants native to Mexico were inactive against ten bacterial strains of clinical interest. The fruit extract of Chenopodium ambrosioides inhibited the bacteria Enterococcus faecalis (MIC = 4375 μg/ml), Escherichia coli (MIC = 1094 μg/ml), and Salmonella typhimurium (MIC = 137 μg/ml). The fruit extract of C. ambrosioides was with CC50 = 45 μg/ml most cytotoxic against the cell-line Caco-2, followed by the leaf extract from Pithecellobium dulce (CC50 = 126 μg/ml); interestingly, leaves of C. ambrosioides (CC50 = 563 μg/ml) and bark of P. dulce (CC50 = 347 μg/ml) extracts were much less cytotoxic. We describe for the first time the cytotoxic effect from extracts of the aerial parts and the flowers of Cirsium mexicanum (CC50 = 323 μg/ml and CC50 = 250 μg/ml, resp.). Phytochemical analysis demonstrated for both extracts high tannin and saponin and low flavonoid content, while terpenoids were found in the flowers. For the first time we report a cytotoxicological study on an extract of Eryngium carlinae (CC50 = 356 μg/ml) and likewise the bark extract from Amphipterygium adstringens (CC50 = 342 μg/ml). In conclusion the fruit extract of C. ambrosioides is a potential candidate for further biological studies.
Collapse
|
25
|
Wortel IMN, van der Meer LT, Kilberg MS, van Leeuwen FN. Surviving Stress: Modulation of ATF4-Mediated Stress Responses in Normal and Malignant Cells. Trends Endocrinol Metab 2017; 28:794-806. [PMID: 28797581 PMCID: PMC5951684 DOI: 10.1016/j.tem.2017.07.003] [Citation(s) in RCA: 413] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/06/2017] [Accepted: 07/10/2017] [Indexed: 01/14/2023]
Abstract
Activating transcription factor 4 (ATF4) is a stress-induced transcription factor that is frequently upregulated in cancer cells. ATF4 controls the expression of a wide range of adaptive genes that allow cells to endure periods of stress, such as hypoxia or amino acid limitation. However, under persistent stress conditions, ATF4 promotes the induction of apoptosis. Recent advances point to a role for post-translational modifications (PTMs) and epigenetic mechanisms in balancing these pro- and anti-survival effects of ATF4. We review here how PTMs and epigenetic modifiers associated with ATF4 may be exploited by cancer cells to cope with cellular stress conditions that are intrinsically associated with tumor growth. Identification of mechanisms that modulate ATF4-mediated transcription and its effects on cellular metabolism may uncover new targets for cancer treatment.
Collapse
Affiliation(s)
- Inge M N Wortel
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Science, Radboud university medical center, Nijmegen, The Netherlands; Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Laurens T van der Meer
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Science, Radboud university medical center, Nijmegen, The Netherlands
| | - Michael S Kilberg
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610-0245, USA.
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Science, Radboud university medical center, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Chen X, Zhang X, Chen J, Yang Q, Yang L, Xu D, Zhang P, Wang X, Liu J. Hinokitiol copper complex inhibits proteasomal deubiquitination and induces paraptosis-like cell death in human cancer cells. Eur J Pharmacol 2017; 815:147-155. [PMID: 28887042 DOI: 10.1016/j.ejphar.2017.09.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 12/29/2022]
Abstract
The ubiquitin-proteasome system (UPS) plays a central role in the regulation of proteins that control cell growth and apoptosis and has therefore become an important target for anticancer therapy. Several constitutive subunits of the 19S proteasome display deubiquitinase (DUB) activity, suggesting that ubiquitin modification of proteins is dynamically regulated. Our study and others have shown that metal complexes, such as copper complexes, can induce cancer cell apoptosis through inhibiting 19S proteasome-associated DUBs and/or 20S proteasome activity. In this study, we found that (1) Hinokitiol copper complex (HK-Cu) induces striking accumulation of ubiquitinated proteins in A549 and K562 cells (2) HK-Cu potently inhibits the activity of the 19S proteasomal DUBs much more effectively than it does to the chymotrypsin-like activity of the 20S proteasome (3) HK-Cu effectively induces caspase-independent and paraptosis-like cell death in A549 and K562 cells, and (4) HK-Cu-induced cell death depends on ATF4-assosiated ER stress but is apparently not related to ROS generation. Altogether, these data indicate that HK-Cu can inhibit the activity of the 19S proteasomal DUBs and induce paraptosis-like cell death, representing a new drug candidate for cancer treatment.
Collapse
Affiliation(s)
- Xin Chen
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaolan Zhang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinghong Chen
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China; Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qianqian Yang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Yang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dacai Xu
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peiquan Zhang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuejun Wang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China; Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA
| | - Jinbao Liu
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
27
|
Cai J, Xia X, Liao Y, Liu N, Guo Z, Chen J, Yang L, Long H, Yang Q, Zhang X, Xiao L, Wang X, Huang H, Liu J. A novel deubiquitinase inhibitor b-AP15 triggers apoptosis in both androgen receptor-dependent and -independent prostate cancers. Oncotarget 2017; 8:63232-63246. [PMID: 28968984 PMCID: PMC5609916 DOI: 10.18632/oncotarget.18774] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer (PCa) remains a leading cause of cancer-related death in men. Especially, a subset of patients will eventually progress to the metastatic castrate-resistant prostate cancer (CRPC) which is currently incurable. Deubiquitinases (DUBs) associated with the 19S proteasome regulatory particle are increasingly emerging as significant therapeutic targets in numerous cancers. Recently, a novel small molecule b-AP15 is identified as an inhibitor of the USP14/UCHL5 (DUBs) of the 19S proteasome, resulting in cell growth inhibition and apoptosis in several human cancer cell lines. Here, we studied the therapeutic effect of b-AP15 in PCa, and our results indicate that (i) b-AP15 decreases viability, proliferation and triggers cytotoxicity to both androgen receptor-dependent and -independent PCa cells in vitro and in vivo, associated with caspase activation, inhibition of mitochondria function, increased reactive oxygen species (ROS) generation and endoplasmic reticulum (ER) stress; (ii) pan-caspase inhibitor z-VAD-FMK and ROS scavenger N-acetyl-L-cysteine (NAC) efficiently block apoptosis but not proteasome inhibition induced by exposure of b-AP15; (iii) treatment with b-AP15 in androgen-dependent prostate cancer (ADPC) cells down-regulates the expression of androgen receptor (AR), which is degraded via the ubiquitin proteasome system. Hence, the potent anti-tumor effect of b-AP15 on both androgen receptor-dependent and -independent PCa cells identifies a new promising therapeutic strategy for prostate cancer.
Collapse
Affiliation(s)
- Jianyu Cai
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Xiaohong Xia
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Yuning Liao
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Ningning Liu
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China.,Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Zhiqiang Guo
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Jinghong Chen
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Li Yang
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Huidan Long
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Qianqian Yang
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Xiaolan Zhang
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Lu Xiao
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Xuejun Wang
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China.,Division of Basic Biomedical Sciences, Sanford School of Medicine of The University of South Dakota, Vermillion, South Dakota 57069, USA
| | - Hongbiao Huang
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| | - Jinbao Liu
- Protein Modification and Degradation Lab, SKLRD, School of Basic Medical Sciences, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangdong 511436, China
| |
Collapse
|
28
|
Huang H, Liu N, Liao Y, Liu N, Cai J, Xia X, Guo Z, Li Y, Wen Q, Yin Q, Liu Y, Wu Q, Rajakumar D, Sheng X, Liu J. Platinum-containing compound platinum pyrithione suppresses ovarian tumor proliferation through proteasome inhibition. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:79. [PMID: 28619062 PMCID: PMC5471884 DOI: 10.1186/s13046-017-0547-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/31/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Ovarian carcinoma is one of the most aggressive gynecological malignant neoplasms and makes up 25-30% of all cancer cases of the female genital tract. Currently, resistance to traditional chemotherapy is a great challenge for patients with Epithelial ovarian cancer (EOC). Therefore, identifying novel agents for EOC treatment is essential and urgent. METHOD MTS assay was used to analyze the cell viability and proliferation of cancer cells. Flow cytometry was employed to analyze cell cycle distribution and cell apoptosis. Protein signaling pathways were detected by western blot and immunohistochemical staining. Nude mouse experiment was performed to test the in vivo effect of platinum pyrithione (PtPT). RESULTS PtPT is a chemically well-characterized synthetic complex of platinum that potently inhibits proteasome-associated deubiquitinases USP14 and UCHL5 activity and shows selective cytotoxicity to multiple cancer cells without damaging DNA. We found that PtPT significantly accumulated ubquitinated-proteins and suppressed the proliferation of multiple EOC cells. Additionally, PtPT induced G2 phase arrest and apoptosis in both A2780 and SKOV3 cells. More importantly, animal experiments showed that PtPT dramatically suppressed the growth of EOC xenografts without obvious side effects. CONCLUSION These results suggest that through proteasome inhibition, PtPT significantly suppressed the proliferation of EOC in vitro and in vivo and could be developed as a novel agent for EOC treatment in the future.
Collapse
Affiliation(s)
- Hongbiao Huang
- Key Laboratory of Protein Modification and Degradation, Department of Obsterics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, 510510, China.,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Ni Liu
- Key Laboratory of Protein Modification and Degradation, Department of Obsterics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, 510510, China.,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Yuning Liao
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Ningning Liu
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.,Guangzhou Institute of Cardiovascular Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Jianyu Cai
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Xiaohong Xia
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Zhiqiang Guo
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Yanling Li
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Qirong Wen
- Key Laboratory of Protein Modification and Degradation, Department of Obsterics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, 510510, China
| | - Qi Yin
- Key Laboratory of Protein Modification and Degradation, Department of Obsterics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, 510510, China
| | - Yan Liu
- Key Laboratory of Protein Modification and Degradation, Department of Obsterics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, 510510, China
| | - Qingxia Wu
- Key Laboratory of Protein Modification and Degradation, Department of Obsterics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, 510510, China
| | - Dhivya Rajakumar
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Xiujie Sheng
- Key Laboratory of Protein Modification and Degradation, Department of Obsterics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, 510510, China.
| | - Jinbao Liu
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences and Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China.
| |
Collapse
|
29
|
Liao Y, Liu N, Hua X, Cai J, Xia X, Wang X, Huang H, Liu J. Proteasome-associated deubiquitinase ubiquitin-specific protease 14 regulates prostate cancer proliferation by deubiquitinating and stabilizing androgen receptor. Cell Death Dis 2017; 8:e2585. [PMID: 28151478 PMCID: PMC5386460 DOI: 10.1038/cddis.2016.477] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 12/14/2022]
Abstract
Androgen receptor (AR) is frequently over-expressed and plays a critical role in the growth and progression of human prostate cancer. The therapy attempting to target AR signalling was established in decades ago but the treatment of prostate cancer is far from being satisfactory. The assignable cause is that our understanding of the mechanism of AR regulation and re-activation remains incomplete. Increasing evidence suggests that deubiquitinases are involved in the regulation of cancer development and progression but the specific underlying mechanism often is not elucidated. In the current study, we have identified ubiquitin-specific protease 14 (USP14) as a novel regulator of AR, inhibiting the degradation of AR via deubiquitinating this oncoprotein in the androgen-responsive prostate cancer cells. We found that (i) USP14 could bind to AR, and additionally, both genetic and pharmacological inhibition of USP14 accelerated the ubiquitination and degradation of AR; (ii) downregulation or inhibition of USP14 suppressed cell proliferation and colony formation of LNcap cells and, conversely, overexpression of USP14 promoted the proliferation; and (iii) reduction or inhibition of USP14 induced G0/G1 phase arrest in LNcap prostate cancer cells. Hence, we conclude that USP14 promotes prostate cancer progression likely through stabilization of AR, suggesting that USP14 could be a promising therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Yuning Liao
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Ningning Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China.,Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xianliang Hua
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Jianyu Cai
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xiaohong Xia
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Xuejun Wang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China.,Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, USA
| | - Hongbiao Huang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Jinbao Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| |
Collapse
|
30
|
Jiang ZQ, Ma YX, Li MH, Zhan XQ, Zhang X, Wang MY. 5-Hydroxymethylfurfural protects against ER stress-induced apoptosis in GalN/TNF-α-injured L02 hepatocytes through regulating the PERK-eIF2α signaling pathway. Chin J Nat Med 2016; 13:896-905. [PMID: 26721708 DOI: 10.1016/s1875-5364(15)30095-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Indexed: 01/12/2023]
Abstract
5-Hydroxymethylfurfural (5-HMF), a water-soluble compound extracted from wine-processed Fructus corni, is a novel hepatic protectant for treating acute liver injury. The present study was designed to investigate the protective effect of 5-HMF in human L02 hepatocytes injured by D-galactosamine (GalN) and tumor necrosis factor-α (TNF-α) in vitro and to explore the underlying mechanisms of action. Our results showed that 5-HMF caused significant increase in the viability of L02 cells injured by GalN/TNF-α, in accordance with a dose-dependent decrease in apoptotic cell death confirmed by morphological and flow cytometric analyses. Based on immunofluorescence and Western blot assays, we found that GalN/TNF-α induced ER stress in the cells, as indicated by the disturbance of intracellular Ca(2+) concentration, the activation of protein kinase RNA (PKR)-like ER kinase (PERK), phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α), and expression of ATF4 and CHOP proteins, which was reversed by 5-HMF pre-treatment in a dose-dependent manner. The anti-apoptotic effect of 5-HMF was further evidenced by balancing the expression of Bcl-2 family members. In addition, the knockdown of PERK suppressed the expression of phospho-PERK, phospho-eIF2α, ATF4, and CHOP, resulting in a significant decrease in cell apoptosis after the treatment with GalN/TNF-α. 5-HMF could enhance the effects of PERK knockdown, protecting the cells against the GalN/TNF-α insult. In conclusion, these findings demonstrate that 5-HMF can effectively protect GalN/TNF-α-injured L02 hepatocytes against ER stress-induced apoptosis through the regulation of the PERK-eIF2α signaling pathway, suggesting that it is a possible candidate for liver disease therapy.
Collapse
Affiliation(s)
- Ze-Qun Jiang
- Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing 210023, China
| | - Yan-Xia Ma
- Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing 210023, China
| | - Mu-Han Li
- Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing 210023, China
| | - Xiu-Qin Zhan
- Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing 210023, China
| | - Xu Zhang
- Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing 210023, China.
| | - Ming-Yan Wang
- Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing 210023, China
| |
Collapse
|
31
|
Aracelli DSL, Md. TI, Antonio LGJ, Joao MDCES, Marcus VOBDA, Marcia FCJP, Hercilia MLR, Maria DGFDM, Ana ADCMC, Jose ADL. Pharmacological properties of cashew (Anacardium occidentale). ACTA ACUST UNITED AC 2016. [DOI: 10.5897/ajb2015.15051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
32
|
The Natural Occurring Compounds Targeting Endoplasmic Reticulum Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:7831282. [PMID: 27563337 PMCID: PMC4987485 DOI: 10.1155/2016/7831282] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/04/2016] [Indexed: 12/14/2022]
Abstract
ER stress has been implicated in pathophysiological development of many diseases. Persistent overwhelming stimuli trigger ER stress to initiate apoptosis, autophagy, and cell death. IRE1-JNK and eIF2α-CHOP signaling pathways are the two important players of ER stress, which is also modulated by ROS production, calcium disturbance, and inflammatory factors. ER stress has been developed as a novel strategy for diseases management. Recently, a vast of research focuses on the natural occurring compounds targeting ER stress, which results in medical benefits to human diseases. These small reported molecules mainly include polyphenols, alkaloids, and saponins. Many of them have been developed for use in clinical applications. To better understand the pharmacological mechanism of these molecules in ER stress in diseases, efforts have been made to discover and deliver medical merits. In this paper, we will summarize the natural occurring compounds targeting ER stress.
Collapse
|
33
|
Dong X, Liao Y, Liu N, Hua X, Cai J, Liu J, Huang H. Combined therapeutic effects of bortezomib and anacardic acid on multiple myeloma cells via activation of the endoplasmic reticulum stress response. Mol Med Rep 2016; 14:2679-84. [DOI: 10.3892/mmr.2016.5533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 06/17/2016] [Indexed: 11/05/2022] Open
|
34
|
Anacardic acid induces apoptosis-like cell death in the rice blast fungus Magnaporthe oryzae. Appl Microbiol Biotechnol 2015; 100:323-35. [DOI: 10.1007/s00253-015-6915-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/28/2015] [Accepted: 08/03/2015] [Indexed: 11/26/2022]
|
35
|
Leite ADS, Dantas AF, Oliveira GLDS, Gomes Júnior AL, de Lima SG, Citó AMDGL, de Freitas RM, Melo-Cavalcante AADC, Dantas Lopes JA. Evaluation of toxic, cytotoxic, mutagenic, and antimutagenic activities of natural and technical cashew nut shell liquids using the Allium cepa and Artemia salina bioassays. BIOMED RESEARCH INTERNATIONAL 2015; 2015:626835. [PMID: 25861638 PMCID: PMC4377390 DOI: 10.1155/2015/626835] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/21/2015] [Accepted: 01/21/2015] [Indexed: 01/11/2023]
Abstract
The cashew nut releases a substance that is known as cashew nut shell liquid (CNSL). There are both natural (iCNSL) and technical (tCNSL) cashew nut shell liquids. This study used an Artemia salina bioassay to evaluate the toxic effects of iCNSL and tCNSL cashew nut shell liquids. It also evaluated the toxicity, cytotoxicity, and mutagenicity of CNSL and its effects on the damage induced by copper sulfate (CuSO4·5H2O) on the meristems' root of Allium cepa. Effects of the damage induced by CuSO4·5H2O were evaluated before (pre-), during (co-), and after (post-) treatments. The iCNSL contained 94.5% anacardic acid, and the tCNSL contained 91.3% cardanol. The liquids were toxic to A. salina. Toxicity, cytotoxicity, and mutagenicity were observed with iCNSL compared with the negative control. Similarly, iCNSL failed to inhibit the toxicity and cytotoxicity of CuSO4·5H2O. The tCNSL was not toxic, cytotoxic, or mutagenic in any of the concentrations. However, the lowest iCNSL concentrations and all of the tCNSL concentrations had preventive, antimutagenic, and reparative effects on micronuclei and on chromosomal aberrations in the A. cepa. Therefore, protective, modulating, and reparative effects may be observed in the A. cepa, depending on the concentration and type of CNSL used.
Collapse
Affiliation(s)
- Aracelli de Sousa Leite
- Laboratório de Pesquisa em Genética Toxicológica de Pós-Graduação em Ciências Farmacêuticas da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
- Programa de Pós-Graduação em Biotecnologia (RENORBIO) da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
| | - Alisson Ferreira Dantas
- Programa de Pós-Graduação em Biologia Animal, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, 70910-900 Brasília, DF, Brazil
| | - George Laylson da Silva Oliveira
- Laboratório de Pesquisa em Neuroquímica Experimental do Programa de Pós-Graduação em Ciências Farmacêuticas da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
| | - Antonio L. Gomes Júnior
- Laboratório de Pesquisa em Genética Toxicológica de Pós-Graduação em Ciências Farmacêuticas da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
| | - Sidney Gonçalo de Lima
- Departamento de Química, CCN, Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
| | | | - Rivelilson M. de Freitas
- Laboratório de Pesquisa em Neuroquímica Experimental do Programa de Pós-Graduação em Ciências Farmacêuticas da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
| | - Ana Amélia de C. Melo-Cavalcante
- Laboratório de Pesquisa em Genética Toxicológica de Pós-Graduação em Ciências Farmacêuticas da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
- Programa de Pós-Graduação em Biotecnologia (RENORBIO) da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
| | - José Arimateia Dantas Lopes
- Programa de Pós-Graduação em Biotecnologia (RENORBIO) da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
- Laboratório de Pesquisa em Neuroquímica Experimental do Programa de Pós-Graduação em Ciências Farmacêuticas da Universidade Federal do Piauí, 6409-550 Teresina, PI, Brazil
| |
Collapse
|
36
|
Min KJ, Jung KJ, Kwon TK. Carnosic Acid Induces Apoptosis Through Reactive Oxygen Species-mediated Endoplasmic Reticulum Stress Induction in Human Renal Carcinoma Caki Cells. J Cancer Prev 2014; 19:170-8. [PMID: 25337586 PMCID: PMC4189511 DOI: 10.15430/jcp.2014.19.3.170] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/05/2014] [Accepted: 09/05/2014] [Indexed: 01/16/2023] Open
Abstract
Background: Carnosic acid, which is one of extract components of rosemary, has anti-inflammatory, anti-oxidant, and anti-cancer effects. However, the anti-cancer effect of carnosic acid in human renal carcinoma cells is unknown. Methods: Flow cytometry analysis was used to examine the effects of carnosic acid on apoptosis, and Asp-Glu-Val-Asp-ase activity assay kit was used to investigate the involvement of caspase activation. To determine protein expression of apoptotic and endoplasmic reticulum (ER) stress-related proteins, we used Western blotting. Intracellular accumulation of reactive oxygen species (ROS) was determined using the fluorescent probes 2’, 7’-dichlorodihydrofluorescein diacetate (H2DCFDA). Results: Carnosic acid induced sub-diploid DNA content, sub-G1, population and poly (ADP-ribose) polymerase (PARP) cleavage and activated caspase-3. A pan-caspase inhibitor, a benzyloxycarbonylvalyl-alanyl-aspartyl fluoromethyl ketone, markedly reduced apoptosis in carnosic acid-treated cells. Carnosic acid promoted intracellular ROS production, and pretreatment with the ROS scavengers (N-acetyl-L-cysteine and glutathione ethyl ester) inhibited carnosic acid-induced apoptosis. Furthermore, carnosic acid also induced expression of ER stress marker proteins, including activating transcription factor 4 (ATF4) and CCAAT/enhancer-binding protein-homologous protein (CHOP), in a dose- and time-dependent manner. Down-regulation of ATF4 and CHOP by small interfering RNA (siRNA) markedly reduced carnosic acid-induced sub-G1 population and PARP cleavage. In addition, carnosic acid induced apoptosis in human breast carcinoma MDA-MB-361 and human hepatocellular carcinoma SK-HEP1 cells, but not in normal human skin fibroblast cells and normal mouse kidney epithelial TMCK-1 cells. Conclusion: Carnosic acid induced apoptosis through production of ROS and induction of ER stress in human renal carcinoma Caki cells.
Collapse
Affiliation(s)
- Kyoung-Jin Min
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Korea
| | - Kyong-Jin Jung
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Korea
| |
Collapse
|