1
|
Cruz-Gregorio A, Aranda-Rivera AK, Aparicio-Trejo OE, Medina-Campos ON, Sciutto E, Fragoso G, Pedraza-Chaverri J. α-Mangostin induces oxidative damage, mitochondrial dysfunction, and apoptosis in a triple-negative breast cancer model. Phytother Res 2023; 37:3394-3407. [PMID: 37012651 DOI: 10.1002/ptr.7812] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/21/2023] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
Triple-negative breast cancer (TNBC) does not express estrogen receptor, progesterone receptor, and human epidermal growth factor receptor; therefore, TNBC lacks targeted therapy, and chemotherapy is the only available treatment for this illness but causes side effects. A putative strategy for the treatment of TNBC could be the use of the polyphenols such as α-Mangostin (α-M), which has shown anticancerogenic effects in different cancer models and can modulate the inflammatory and prooxidant state in several pathological models. The redox state, oxidative stress (OS), and oxidative damage are highly related to cancer development and its treatment. Thus, this study aimed to evaluate the effects of α-M on redox state, mitochondrial metabolism, and apoptosis in 4T1 mammary carcinoma cells. We found that α-M decreases both protein levels and enzymatic activity of catalase, and increases reactive oxygen species, oxidized proteins and glutathione disulfide, which demonstrates that α-M induces oxidative damage. We also found that α-M promotes mitochondrial dysfunction by abating basal respiration, the respiration ligated to oxidative phosphorylation (OXPHOS), and the rate control of whole 4T1 cells. Additionally, α-M also decreases the levels of OXPHOS subunits of mitochondrial complexes I, II, III, and adenosine triphosphate synthase, the activity of mitochondrial complex I as well as the levels of peroxisome proliferator-activated receptor-gamma co-activator 1α, showing a mitochondrial mass reduction. Then, oxidative damage and mitochondrial dysfunction induced by α-M induce apoptosis of 4T1 cells, which is evidenced by B cell lymphoma 2 decrease and caspase 3 cleavage. Taken together, our results suggest that α-M induces OS and mitochondrial dysfunction, resulting in 4T1 cell death through apoptotic mechanisms.
Collapse
Affiliation(s)
- Alfredo Cruz-Gregorio
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología "Ignacio Chávez", CDMX, Mexico
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Ana Karina Aranda-Rivera
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, CDMX, Mexico
| | | | - Omar Noel Medina-Campos
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Edda Sciutto
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - José Pedraza-Chaverri
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, CDMX, Mexico
| |
Collapse
|
2
|
Costa-Machado LF, Garcia-Dominguez E, McIntyre RL, Lopez-Aceituno JL, Ballesteros-Gonzalez Á, Tapia-Gonzalez A, Fabregat-Safont D, Eisenberg T, Gomez J, Plaza A, Sierra-Ramirez A, Perez M, Villanueva-Bermejo D, Fornari T, Loza MI, Herradon G, Hofer SJ, Magnes C, Madeo F, Duerr JS, Pozo OJ, Galindo MI, Del Pino I, Houtkooper RH, Megias D, Viña J, Gomez-Cabrera MC, Fernandez-Marcos PJ. Peripheral modulation of antidepressant targets MAO-B and GABAAR by harmol induces mitohormesis and delays aging in preclinical models. Nat Commun 2023; 14:2779. [PMID: 37188705 PMCID: PMC10185515 DOI: 10.1038/s41467-023-38410-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/02/2023] [Indexed: 05/17/2023] Open
Abstract
Reversible and sub-lethal stresses to the mitochondria elicit a program of compensatory responses that ultimately improve mitochondrial function, a conserved anti-aging mechanism termed mitohormesis. Here, we show that harmol, a member of the beta-carbolines family with anti-depressant properties, improves mitochondrial function and metabolic parameters, and extends healthspan. Treatment with harmol induces a transient mitochondrial depolarization, a strong mitophagy response, and the AMPK compensatory pathway both in cultured C2C12 myotubes and in male mouse liver, brown adipose tissue and muscle, even though harmol crosses poorly the blood-brain barrier. Mechanistically, simultaneous modulation of the targets of harmol monoamine-oxidase B and GABA-A receptor reproduces harmol-induced mitochondrial improvements. Diet-induced pre-diabetic male mice improve their glucose tolerance, liver steatosis and insulin sensitivity after treatment with harmol. Harmol or a combination of monoamine oxidase B and GABA-A receptor modulators extend the lifespan of hermaphrodite Caenorhabditis elegans or female Drosophila melanogaster. Finally, two-year-old male and female mice treated with harmol exhibit delayed frailty onset with improved glycemia, exercise performance and strength. Our results reveal that peripheral targeting of monoamine oxidase B and GABA-A receptor, common antidepressant targets, extends healthspan through mitohormesis.
Collapse
Affiliation(s)
- Luis Filipe Costa-Machado
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
- Kaertor Foundation, EMPRENDIA Building, Floor 2, Office 4, Campus Vida, E-15706, Santiago de Compostela, Spain, E-15706, Santiago de Compostela, Spain
- BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Esther Garcia-Dominguez
- Freshage Research Group, Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Rebecca L McIntyre
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jose Luis Lopez-Aceituno
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
| | - Álvaro Ballesteros-Gonzalez
- Developmental Biology and Disease Models Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Andrea Tapia-Gonzalez
- Neural Plasticity Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - David Fabregat-Safont
- Applied Metabolomics Research Group, Hospital del Mar Medical Research Institute - (IMIM), Barcelona, Spain
- Environmental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water, University Jaume I, 12006, Castelló de la Plana, Castellón, Spain
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
- BioTechMed Graz, 8010, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Jesús Gomez
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Adrian Plaza
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
| | - Aranzazu Sierra-Ramirez
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain
| | - Manuel Perez
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - David Villanueva-Bermejo
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL UAM-CSIC), C/ Nicolás Cabrera, 9, P.O. Box. 28049, Madrid, Spain
| | - Tiziana Fornari
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL UAM-CSIC), C/ Nicolás Cabrera, 9, P.O. Box. 28049, Madrid, Spain
| | - María Isabel Loza
- Kaertor Foundation, EMPRENDIA Building, Floor 2, Office 4, Campus Vida, E-15706, Santiago de Compostela, Spain, E-15706, Santiago de Compostela, Spain
- BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Gonzalo Herradon
- Lab. Pharmacology, Faculty of Pharmacy, Universidad CEU San Pablo, Urb. Montepríncipe, 28668, Boadilla del Monte, Madrid, Spain
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
- BioTechMed Graz, 8010, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Christoph Magnes
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, 8010, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
- BioTechMed Graz, 8010, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Janet S Duerr
- Department of Biological Sciences, Ohio University, Athens, OH, 45701, USA
| | - Oscar J Pozo
- Applied Metabolomics Research Group, Hospital del Mar Medical Research Institute - (IMIM), Barcelona, Spain
| | - Maximo-Ibo Galindo
- Developmental Biology and Disease Models Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46022, Valencia, Spain
- UPV-CIPF Joint Research Unit "Disease Mechanisms and Nanomedicine". Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Isabel Del Pino
- Neural Plasticity Group, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550, Alicante, Spain
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Diego Megias
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Jose Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Mari Carmen Gomez-Cabrera
- Freshage Research Group, Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Pablo J Fernandez-Marcos
- Metabolic Syndrome Group - BIOPROMET. Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM + CSIC, E28049, Madrid, Spain.
| |
Collapse
|
3
|
Alam M, Rashid S, Fatima K, Adnan M, Shafie A, Akhtar MS, Ganie AH, Eldin SM, Islam A, Khan I, Hassan MI. Biochemical features and therapeutic potential of α-Mangostin: Mechanism of action, medicinal values, and health benefits. Biomed Pharmacother 2023; 163:114710. [PMID: 37141737 DOI: 10.1016/j.biopha.2023.114710] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023] Open
Abstract
α-Mangostin (α-MG) is a natural xanthone obtained from the pericarps of mangosteen. It exhibits excellent potential, including anti-cancer, neuroprotective, antimicrobial, antioxidant, and anti-inflammatory properties, and induces apoptosis. α-MG controls cell proliferation by modulating signaling molecules, thus implicated in cancer therapy. It possesses incredible pharmacological features and modulates crucial cellular and molecular factors. Due to its lesser water solubility and pitiable target selectivity, α-MG has limited clinical application. As a known antioxidant, α-MG has gained significant attention from the scientific community, increasing interest in extensive technical and biomedical applications. Nanoparticle-based drug delivery systems were designed to improve the pharmacological features and efficiency of α-MG. This review is focused on recent developments on the therapeutic potential of α-MG in managing cancer and neurological diseases, with a special focus on its mechanism of action. In addition, we highlighted biochemical and pharmacological features, metabolism, functions, anti-inflammatory, antioxidant effects and pre-clinical applications of α-MG.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, PO Box 173, Al-kharj 11942, Saudi Arabia
| | - Kisa Fatima
- Department of Biotechnology, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, PO Box 2440, Hail 2440, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Mohammad Salman Akhtar
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - A H Ganie
- Basic Sciences Department, College of Science and Theoretical Studies, Saudi Electronic University, Abha Male 61421, Saudi Arabia
| | - Sayed M Eldin
- Center of Research, Faculty of Engineering, Future University in Egypt, New Cairo 11835, Egypt
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ilyas Khan
- Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
4
|
Niu H, Li J, Liang H, Wu G, Chen M. Exogenous Hydrogen Sulfide Activates PI3K/Akt/eNOS Pathway to Improve Replicative Senescence in Human Umbilical Vein Endothelial Cells. Cardiol Res Pract 2023; 2023:7296874. [PMID: 37064727 PMCID: PMC10101749 DOI: 10.1155/2023/7296874] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 04/18/2023] Open
Abstract
Background Endothelial cell senescence is one of the key mechanistic factors in the pathogenesis of atherosclerosis. In terms of molecules, the phosphatidylinositol 3-kinase/protein kinase B/endothelial nitric oxide synthase (PI3K/Akt/eNOS) signaling plays an important role in the prevention and control of endothelial cell senescence, while hydrogen sulfide (H2S) improves the induced precocious senescence of endothelial cells through the PI3K/Akt/eNOS pathway. Comparatively, replicative senescence in endothelial cells is more in line with the actual physiological changes of human aging. This study aims to investigate the mechanism by which H2S improves endothelial cell replicative senescence and the involvement of the PI3K/Akt/eNOS pathway. Methods we established a model of replicative senescence in human umbilical vein endothelial cells (HUVECs) and explored the effect of 200 μmol/L sodium hydrosulfide (NaHS; a donor of H2S) on senescence, which was determined by cell morphology, the expression level of plasminogen activator inhibitor 1 (PAI-1), and the positive rate of senescence-associated β-galactosidase (SA-β-Gal) staining. Cell viability was detected by MTT assay to evaluate the effect of NaHS and the PI3K inhibitor, LY294002. Meanwhile, the protein expression of PI3K, Akt, p-Akt, and eNOS in endothelial cells of each group was detected by Western blot. Results the replicative senescence model was established in HUVECs at the passage of 16 cumulative cell population doubling values (CPDL). Treatment with NaHS not only significantly reduced the expression of PAI-1 and the positive rate of SA-β-Gal in HUVEC's replicative senescence model but also notably increased the expression of PI3K, p-Akt, p-eNOS, and the content of nitric oxide(NO). However, the effects of NaHS on the expression of the pathway and the content of NO in HUVECs were abolished when LY294002 specifically inhibited PI3K. Conclusion NaHS improves the replicative senescence of HUVECs with the contribution of the PI3K/Akt/eNOS pathway.
Collapse
Affiliation(s)
- Haiming Niu
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Jianwei Li
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Hongkai Liang
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Guishen Wu
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Miaolian Chen
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan 528400, China
| |
Collapse
|
5
|
Nunna S, Huang YP, Rasa M, Krepelova A, Annunziata F, Adam L, Käppel S, Hsu MH, Neri F. Characterization of Novel α-Mangostin and Paeonol Derivatives With Cancer-Selective Cytotoxicity. Mol Cancer Ther 2022; 21:257-270. [PMID: 34789561 PMCID: PMC9398122 DOI: 10.1158/1535-7163.mct-20-0787] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 04/22/2021] [Accepted: 11/03/2021] [Indexed: 01/07/2023]
Abstract
α-Mangostin (aMan) and Paeonol (Pae) have shown anticancer and anti-inflammatory properties. However, these two natural compounds have no clinical value because of their low solubility and low membrane permeability. In this study, we screened chemically synthesized derivatives from these two natural compounds as potential novel chemicals that increase cancer cell cytotoxicity over nontransformed human cells. We found that two derivative compounds, named α-Mangostin-1 (aMan1) and Paeonol-1 (Pae1) more efficiently and more specifically induced cytotoxicity in HCT116, HT29, and SW48 colorectal cancer cell lines than the parental compounds. Both aMan1 and Pae1 arrested HCT116 cells in the G1 phase and HT29 and SW48 cells in the G2-M phase of the cell cycle. Both aMan1 and Pae1 induced apoptosis in human colorectal cancer cells, through a caspase-dependent mechanism. aMan1 and Pae1 induced selective transcriptional responses in colorectal cancer cells involving genes related to metabolic stress and DNA damage response signaling pathways. Finally, experiments on primary colon organoids showed that both derivatives were able to kill cancer-derived organoids without affecting the viability of organoids derived from healthy tissue, where the parental compounds and the currently used chemotherapeutic drug irinotecan failed. In conclusion, our findings expand the knowledge of natural compound derivatives as anticancer agents and open new avenues of research in the derivation of lead compounds aimed at developing novel chemotherapeutic drugs for colorectal cancer treatment that selectively target cancer, but not healthy cells.
Collapse
Affiliation(s)
- Suneetha Nunna
- Leibniz-Institute on Ageing - Fritz-Lipmann-Institute (FLI), Jena, Germany
| | - Ying-Pei Huang
- Leibniz-Institute on Ageing - Fritz-Lipmann-Institute (FLI), Jena, Germany.,Nuclear Science & Technology Development Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Mahdi Rasa
- Leibniz-Institute on Ageing - Fritz-Lipmann-Institute (FLI), Jena, Germany
| | - Anna Krepelova
- Leibniz-Institute on Ageing - Fritz-Lipmann-Institute (FLI), Jena, Germany
| | | | - Lisa Adam
- Leibniz-Institute on Ageing - Fritz-Lipmann-Institute (FLI), Jena, Germany
| | - Sandra Käppel
- Leibniz-Institute on Ageing - Fritz-Lipmann-Institute (FLI), Jena, Germany
| | - Ming-Hua Hsu
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan, ROC.,Department of Medical and Applied Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Francesco Neri
- Leibniz-Institute on Ageing - Fritz-Lipmann-Institute (FLI), Jena, Germany.,Corresponding Author: Francesco Neri, Epigenetics group, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, 07745, Germany. E-mail:
| |
Collapse
|
6
|
Mitochondria as a potential target for the development of prophylactic and therapeutic drugs against Schistosoma mansoni infection. Antimicrob Agents Chemother 2021; 65:e0041821. [PMID: 34339272 DOI: 10.1128/aac.00418-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Emergence of parasites resistant to praziquantel, the only therapeutic agent, and its ineffectiveness as a prophylactic agent (inactive against the migratory/juvenile Schistosoma mansoni), makes the development of new antischistosomal drugs urgent. The parasite's mitochondrion is an attractive target for drug development because this organelle is essential for survival throughout the parasite's life cycle. We investigated the effects of 116 compounds against Schistosoma mansoni cercariae motility that have been reported to affect mitochondria-related processes in other organisms. Next, eight compounds plus two controls (mefloquine and praziquantel) were selected and assayed against motility of schistosomula (in vitro) and adults (ex vivo). Prophylactic and therapeutic assays were performed using infected mouse models. Inhibition of oxygen consumption rate (OCR) was assayed using Seahorse XFe24 Analyzer. All selected compounds showed excellent prophylactic activity, reducing the worm burden in the lungs to less than 15% that obtained in the vehicle control. Notably, ascofuranone showed the highest activity with a 98% reduction of the worm burden, suggesting the potential for development of ascofuranone as a prophylactic agent. The worm burden of infected mice with S. mansoni at the adult stage was reduced by more than 50% in mice treated with mefloquine, nitazoxanide, amiodarone, ascofuranone, pyrvinium pamoate, or plumbagin. Moreover, adult mitochondrial OCR was severely inhibited by ascofuranone, atovaquone, and nitazoxanide, while pyrvinium pamoate inhibited both mitochondrial and non-mitochondrial OCRs. These results demonstrate that the mitochondria of S. mansoni are feasible target for drug development.
Collapse
|
7
|
Hu YH, Han J, Wang L, Shi C, Li Y, Olatunji OJ, Wang X, Zuo J. α-Mangostin Alleviated Inflammation in Rats With Adjuvant-Induced Arthritis by Disrupting Adipocytes-Mediated Metabolism-Immune Feedback. Front Pharmacol 2021; 12:692806. [PMID: 34305602 PMCID: PMC8293671 DOI: 10.3389/fphar.2021.692806] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
A previously identified anti-rheumatic compound α-mangostin (MAN) possesses notable metabolism regulatory properties. In this study, we investigated the immune implication of MAN-altered fat metabolism on adjuvant-induced arthritis (AIA) in rats. Seven days after AIA induction, the rats received oral treatment of MAN at 50 mg/kg/day for 30 days. Metabolic indicators and basic clinical parameters were evaluated using samples collected on day 20 and 38 since immunization. Expression of nicotinamide phosphoribosyltransferase (NAMPT), sirtuin 1 (SIRT1), peroxisome proliferator activated receptor gamma (PPAR-γ), stearoyl-coa desaturase 1 (SCD-1), toll like receptor 4 (TLR4), prostaglandin-endoperoxide synthase 2 (COX-2), (p)-JNK, (p)-p65 and IL-1β were investigated by either RT-qPCR or immunobloting methods. In in vitro experiments, we treated (pre)-adipocytes with monocytes/macrophages and MAN, and investigated the changes of macrophages brought by pre-adipocytes co-culture. Generally, MAN restored the impaired fat anabolism in AIA rats, indicated by increased fat reservoir, leptin and adiponectin secretion, and PPAR-γ and SCD-1 expression. Meanwhile, it decreased circulating IL-1β and IL-6 levels, restored serological lipid profile changes, and relieved oxidative stresses, demonstrating potent therapeutic effects on AIA. AIA rats-derived monocytes inhibited mRNA PPAR-γ and SCD-1 expression in pre-adipocytes. Contrarily, MAN facilitated adipocyte differentiation in vitro, and increased free fatty acids production. It also significantly increased PPAR-γ and SCD-1 expression, which can be abrogated by PPAR-γ inhibitor T0070907. Similarly, lipopolysaccharide-primed macrophages inhibited PPAR-γ expression in the co-cultured pre-adipocytes, which was reversed by MAN. In the same co-culture system, lipopolysaccharide-induced inflammation was amplified by the co-existence of pre-adipocytes. More secretion of IL-1β and IL-6 and higher levels expression of COX-2, p-JNK, p-p65 and TLR4 were observed in lipopolysaccharide-treated macrophages when co-cultured by pre-adipocytes. The intensified inflammatory situation was eased by MAN. The treatment with pre-adipocytes culture medium achieved similar effects. Medium from lipopolysaccharide-treated adipocytes promoted IL-1β, IL-6 and MCP-1 production in separately cultured macrophages, and COX-2, p-JNK, p-p65 and TLR4 expression were increased at the meantime. MAN treatment on pre-adipocytes impaired these changes. It suggests that fat anabolism in AIA rats was deficient due to increased energy expenditure caused by inflammatory conditions. MAN restored fat metabolism homeostasis by up-regulating PPAR-γ, and reshaped secretion profile of adipocytes.
Collapse
Affiliation(s)
- Ying-Hao Hu
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, China
| | - Jun Han
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, China.,Drug Research and Development Center, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Lin Wang
- Department of Pharmacy, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Chao Shi
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, China
| | - Yan Li
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, China
| | | | - Xiu Wang
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, China
| | - Jian Zuo
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
| |
Collapse
|
8
|
A review on α-mangostin as a potential multi-target-directed ligand for Alzheimer's disease. Eur J Pharmacol 2021; 897:173950. [PMID: 33607107 DOI: 10.1016/j.ejphar.2021.173950] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive memory loss, declining language skills and other cognitive disorders. AD has brought great mental and economic burden to patients, families and society. However due to the complexity of AD's pathology, drugs developed for the treatment of AD often fail in clinical or experimental trials. The main problems of current anti-AD drugs are low efficacy due to mono-target method or side effects, especially high hepatotoxicity. To tackle these two main problems, multi-target-directed ligand (MTDL) based on "one molecule, multiple targets" has been studied. MTDLs can regulate multiple biological targets at the same time, so it has shown higher efficacy, better safety. As a natural active small molecule, α-mangostin (α-M) has shown potential multi-factor anti-AD activities in a series of studies, furthermore it also has a certain hepatoprotective effect. The good availability of α-M also provides support for its application in clinical research. In this work, multiple activities of α-M related to AD therapy were reviewed, which included anti-cholinesterase, anti-amyloid-cascade, anti-inflammation, anti-oxidative stress, low toxicity, hepatoprotective effects and drug formulation. It shows that α-M is a promising candidate for the treatment of AD.
Collapse
|
9
|
Hotarat W, Nutho B, Wolschann P, Rungrotmongkol T, Hannongbua S. Delivery of Alpha-Mangostin Using Cyclodextrins through a Biological Membrane: Molecular Dynamics Simulation. Molecules 2020; 25:molecules25112532. [PMID: 32485931 PMCID: PMC7321106 DOI: 10.3390/molecules25112532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
α-Mangostin (MGS) exhibits various pharmacological activities, including antioxidant, anticancer, antibacterial, and anti-inflammatory properties. However, its low water solubility is the major obstacle for its use in pharmaceutical applications. To increase the water solubility of MGS, complex formation with beta-cyclodextrins (βCDs), particularly with the native βCD and/or its derivative 2,6-dimethyl-β-CD (DMβCD) is a promising technique. Although there have been several reports on the adsorption of βCDs on the lipid bilayer, the release of the MGS/βCDs inclusion complex through the biological membrane remains unclear. In this present study, the release the MGS from the two different βCDs (βCD and DMβCD) across the lipid bilayer was investigated. Firstly, the adsorption of the free MGS, free βCDs, and inclusion complex formation was studied by conventional molecular dynamics simulation. The MGS in complex with those two βCDs was able to spontaneously release free MGS into the inner membrane. However, both MGS and DMβCD molecules potentially permeated into the deeper region of the interior membrane, whereas βCD only adsorbed at the outer membrane surface. The interaction between secondary rim of βCD and the 1-palmitoeyl-2-oleoyl-glycero-3-phosphocholine (POPC) phosphate groups showed the highest number of hydrogen bonds (up to 14) corresponding to the favorable location of βCD on the POPC membrane. Additionally, the findings suggested that electrostatic energy was the main driving force for βCD adsorption on the POPC membrane, while van der Waals interactions played a predominant role in DMβCD adsorption. The release profile of MGS from the βCDs pocket across the lipid bilayer exhibited two energy minima along the reaction coordinate associated with the permeation of the MGS molecule into the deeper region of the POPC membrane.
Collapse
Affiliation(s)
- Wiparat Hotarat
- Center of Excellence in Computational Chemistry (CECC), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (W.H.); (B.N.)
| | - Bodee Nutho
- Center of Excellence in Computational Chemistry (CECC), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (W.H.); (B.N.)
| | - Peter Wolschann
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria;
- Institute of Theoretical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Thanyada Rungrotmongkol
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Molecular Sensory Science Center, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (T.R.); (S.H.); Tel.: +66(0)2218-5418 (T.R.); +66(0)2218-7603 (S.H.)
| | - Supot Hannongbua
- Center of Excellence in Computational Chemistry (CECC), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (W.H.); (B.N.)
- Correspondence: (T.R.); (S.H.); Tel.: +66(0)2218-5418 (T.R.); +66(0)2218-7603 (S.H.)
| |
Collapse
|
10
|
Chaeyklinthes T, Tiyao V, Roytrakul S, Phaonakrop N, Showpittapornchai U, Pradidarcheep W. Proteomics study of the antifibrotic effects of α-mangostin in a rat model of renal fibrosis. ASIAN BIOMED 2019. [DOI: 10.1515/abm-2019-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Background
Renal fibrosis is a consequence of a “faulty” wound-healing mechanism that results in the accumulation of extracellular matrix, which could lead to the impairment of renal functions. α-Mangostin (AM) may prevent the formation of liver fibrosis, but there has yet to be a conclusive investigation of its effect on renal fibrosis.
Objectives
To investigate the renoprotective effect of AM against thioacetamide (TAA)-induced renal fibrosis in rats at the morphological and proteomic levels.
Methods
We divided 18 male Wistar rats into 3 groups: a control group, a TAA-treated group, and a TAA + AM group. The various agents used to treat the rats were administered intraperitoneally over 8 weeks. Subsequently, the morphology of renal tissue was analyzed by histology using Sirius Red staining and the relative amount of stained collagen fibers quantified using ImageJ analysis. One-dimensional gel liquid chromatography with tandem mass spectrometry (GeLC-MS/MS) was used to track levels of protein expression. Proteomic bioinformatics tools including STITCH were used to correlate the levels of markers known to be involved in fibrosis with Sirius Red-stained collagen scoring.
Results
Histology revealed that AM could reduce the relative amount of collagen fibers significantly compared with the TAA group. Proteomic analysis revealed the levels of 4 proteins were modulated by AM, namely CASP8 and FADD-like apoptosis regulator (Cflar), Ragulator complex protein LAMTOR3 (Lamtor3), mitogen-activated protein kinase kinase kinase 14 (Map3k14), and C-Jun-amino-terminal kinase-interacting protein 3 (Mapk8ip3).
Conclusion
AM can attenuate renal fibrosis by the suppression of pathways involving Cflar, Lamtor3, Map3k14, and Mapk8ip3.
Collapse
Affiliation(s)
- Thana Chaeyklinthes
- Department of Science, Mahidol University International College, Mahidol University , Nakhon Pathom 73170 , Thailand
| | - Vilailak Tiyao
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University , Bangkok 10110 , Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency , Pathum Thani 12120 , Thailand
| | - Narumon Phaonakrop
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency , Pathum Thani 12120 , Thailand
| | | | - Wisuit Pradidarcheep
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University , Bangkok 10110 , Thailand
| |
Collapse
|
11
|
The Protective Effect of Alpha-Mangostin against Cisplatin-Induced Cell Death in LLC-PK1 Cells is Associated to Mitochondrial Function Preservation. Antioxidants (Basel) 2019; 8:antiox8050133. [PMID: 31096625 PMCID: PMC6562511 DOI: 10.3390/antiox8050133] [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: 04/12/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/28/2022] Open
Abstract
Cis-dichlorodiammineplatinum II (CDDP) is a chemotherapeutic agent that induces nephrotoxicity by different mechanisms, including oxidative stress, mitochondrial dysfunction, autophagy, and endoplasmic reticulum stress. This study aimed to evaluate if the protective effects of the antioxidant alpha-mangostin (αM) in CDDP-induced damage in proximal tubule Lilly laboratory culture porcine kidney (LLC-PK1) cells, are related to mitochondrial function preservation. It was found that αM co-incubation prevented CDDP-induced cell death. Furthermore, αM prevented the CDDP-induced decrease in cell respiratory states, in the maximum capacity of the electron transfer system (E) and in the respiration associated to oxidative phosphorylation (OXPHOS). CDDP also decreased the protein levels of voltage dependence anion channel (VDAC) and mitochondrial complex subunits, which together with the reduction in E, the mitofusin 2 decrease and the mitochondrial network fragmentation observed by MitoTracker Green, suggest the mitochondrial morphology alteration and the decrease in mitochondrial mass induced by CDDP. CDDP also induced the reduction in mitochondrial biogenesis observed by transcription factor A, mitochondria (TFAM) decreased protein-level and the increase in mitophagy. All these changes were prevented by αM. Taken together, our results imply that αM’s protective effects in CDDP-induced toxicity in LLC-PK1 cells are associated to mitochondrial function preservation.
Collapse
|
12
|
Qiu S, Granet R, Mbakidi JP, Brégier F, Pouget C, Micallef L, Sothea-Ouk T, Leger DY, Liagre B, Chaleix V, Sol V. Delivery of tanshinone IIA and α-mangostin from gold/PEI/cyclodextrin nanoparticle platform designed for prostate cancer chemotherapy. Bioorg Med Chem Lett 2016; 26:2503-2506. [PMID: 27040657 DOI: 10.1016/j.bmcl.2016.03.097] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/23/2016] [Accepted: 03/26/2016] [Indexed: 11/16/2022]
Abstract
A new anti-cancer drug delivery system, based on gold nanoparticles, has been designed for hydrophobic active compounds. The system is a conjugate of gold/polyethyleneimine (AuNPs/PEI) nanoparticles and sulphated β-cyclodextrin (CD). Anionic cyclodextrin was attached to the positively charged AuNPs/PEI nanoparticles by ionic bonds. Tanshinone IIA and α-mangostin were extracted, purified and encapsulated into the AuNPs/PEI/CD nanoparticles. In vitro preliminary cell viability assays against prostate cancer cell lines PC-3 and DU145 showed that encapsulation resulted in increased cytotoxicity.
Collapse
Affiliation(s)
- Shihong Qiu
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Robert Granet
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Jean-Pierre Mbakidi
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Frédérique Brégier
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Christelle Pouget
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Ludovic Micallef
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Tan Sothea-Ouk
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - David Y Leger
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Bertrand Liagre
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Vincent Chaleix
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| | - Vincent Sol
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, EA 1069, F-87000 Limoges, France
| |
Collapse
|
13
|
Curcumin Attenuates Gentamicin-Induced Kidney Mitochondrial Alterations: Possible Role of a Mitochondrial Biogenesis Mechanism. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:917435. [PMID: 26345660 PMCID: PMC4541007 DOI: 10.1155/2015/917435] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/03/2015] [Accepted: 07/15/2015] [Indexed: 11/18/2022]
Abstract
It has been shown that curcumin (CUR), a polyphenol derived from Curcuma longa, exerts a protective effect against gentamicin- (GM-) induced nephrotoxicity in rats, associated with a preservation of the antioxidant status. Although mitochondrial dysfunction is a hallmark in the GM-induced renal injury, the role of CUR in mitochondrial protection has not been studied. In this work, LLC-PK1 cells were preincubated 24 h with CUR and then coincubated 48 h with CUR and 8 mM GM. Treatment with CUR attenuated GM-induced drop in cell viability and led to an increase in nuclear factor (erythroid-2)-related factor 2 (Nrf2) nuclear accumulation and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) cell expression attenuating GM-induced losses in these proteins. In vivo, Wistar rats were injected subcutaneously with GM (75 mg/Kg/12 h) during 7 days to develop kidney mitochondrial alterations. CUR (400 mg/Kg/day) was administered orally 5 days before and during the GM exposure. The GM-induced mitochondrial alterations in ultrastructure and bioenergetics as well as decrease in activities of respiratory complexes I and IV and induction of calcium-dependent permeability transition were mostly attenuated by CUR. Protection of CUR against GM-induced nephrotoxicity could be in part mediated by maintenance of mitochondrial functions and biogenesis with some participation of the nuclear factor Nrf2.
Collapse
|
14
|
Synthesis and membrane-protective activity of novel derivatives of α-mangostin at the C-4 position. Bioorg Med Chem Lett 2015; 25:826-9. [DOI: 10.1016/j.bmcl.2014.12.075] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 11/20/2022]
|
15
|
Correa F, Buelna-Chontal M, Hernández-Reséndiz S, García-Niño WR, Roldán FJ, Soto V, Silva-Palacios A, Amador A, Pedraza-Chaverrí J, Tapia E, Zazueta C. Curcumin maintains cardiac and mitochondrial function in chronic kidney disease. Free Radic Biol Med 2013; 61:119-29. [PMID: 23548636 DOI: 10.1016/j.freeradbiomed.2013.03.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 03/21/2013] [Accepted: 03/22/2013] [Indexed: 01/14/2023]
Abstract
Curcumin, a natural pigment with antioxidant activity obtained from turmeric and largely used in traditional medicine, is currently being studied in the chemoprevention of several diseases for its pleiotropic effects and nontoxicity. In chronic renal failure, the pathogenic mechanisms leading to cardiovascular disorders have been associated with increased oxidative stress, a process inevitably linked with mitochondrial dysfunction. Thus, in this study we aimed at investigating if curcumin pretreatment exerts cardioprotective effects in a rat model of subtotal nephrectomy (5/6Nx) and its impact on mitochondrial homeostasis. Curcumin was orally administered (120mg/kg) to Wistar rats 7 days before nephrectomy and after surgery for 60 days (5/6Nx+curc). Renal dysfunction was detected a few days after nephrectomy, whereas changes in cardiac function were observed until the end of the protocol. Our results indicate that curcumin treatment protects against pathological remodeling, diminishes ischemic events, and preserves cardiac function in uremic rats. Cardioprotection was related to diminished reactive oxygen species production, decreased oxidative stress markers, increased antioxidant response, and diminution of active metalloproteinase-2. We also observed that curcumin's cardioprotective effects were related to maintaining mitochondrial function. Aconitase activity was significantly higher in the 5/6Nx + curc (408.5±68.7nmol/min/mg protein) than in the 5/6Nx group (104.4±52.3nmol/min/mg protein, P<0.05), and mitochondria from curcumin-treated rats showed enhanced oxidative phosphorylation capacities with both NADH-linked substrates and succinate plus rotenone (3.6±1 vs 1.1±0.9 and 3.1±0.7 vs 1.2±0.8, respectively, P<0.05). The mechanisms involved in cardioprotection included both direct antioxidant effects and indirect strategies that could be related to protein kinase C-activated downstream signaling.
Collapse
Affiliation(s)
- Francisco Correa
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico; Department of Biochemistry, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | - Mabel Buelna-Chontal
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico; Department of Biochemistry, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | - Sauri Hernández-Reséndiz
- Department of Biochemistry, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | - Wylly R García-Niño
- Renal Pathophysiology Laboratory, Department of Nephrology, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | - Francisco J Roldán
- Department of Echocardiography, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | - Virgilia Soto
- Department of Pathology, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080, DF, Mexico
| | - Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | - Alejandra Amador
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | | | - Edilia Tapia
- Renal Pathophysiology Laboratory, Department of Nephrology, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico
| | - Cecilia Zazueta
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico; Department of Biochemistry, National Institute of Cardiology Ignacio Chavez, Mexico City, 14080 DF, Mexico.
| |
Collapse
|
16
|
Xu D, Nie Y, Liang X, Ji L, Hu S, You Q, Wang F, Ye H, Wang J. A Concise and Efficient Total Synthesis of α-Mangostin and β-Mangostin from Garcinia Mangostana. Nat Prod Commun 2013. [DOI: 10.1177/1934578x1300800818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The concise, efficient synthesis of α-mangostin is described in eight simple steps with 8.3% overall yield. Highlights include a practical approach to construct the isopentene groups and other diverse groups at C–2 and C–8 of the xanthene skeleton through Claisen rearrangement and Wittig reaction. Meanwhile the first total synthesis of β-mangostin is presented with a similar approach.
Collapse
Affiliation(s)
- Dandan Xu
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China 210009
| | - Ying Nie
- Pharmacy Experimental Center, China Pharmaceutical University, Nanjing, China 210009
| | - Xizhou Liang
- Simcere Dongyuan Pharmaceutical Co., Ltd., 10 Tianpu Road, Nanjing, China 211800
| | - Ling Ji
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China 210009
| | - Songyuan Hu
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China 210009
| | - Qidong You
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China 210009
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China 210009
| | - Fan Wang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China 210009
| | - Hongchun Ye
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China 210009
| | - Jinxin Wang
- Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China 210009
| |
Collapse
|
17
|
Márquez-Valadez B, Maldonado PD, Galván-Arzate S, Méndez-Cuesta LA, Pérez-De La Cruz V, Pedraza-Chaverrí J, Chánez-Cárdenas ME, Santamaría A. Alpha-mangostin induces changes in glutathione levels associated with glutathione peroxidase activity in rat brain synaptosomes. Nutr Neurosci 2013; 15:13-9. [DOI: 10.1179/147683012x13327575416400] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
18
|
Yuan J, Wu Y, Lu G. α-Mangostin suppresses lipopolysaccharide-induced invasion by inhibiting matrix metalloproteinase-2/9 and increasing E-cadherin expression through extracellular signal-regulated kinase signaling in pancreatic cancer cells. Oncol Lett 2013; 5:1958-1964. [PMID: 23833675 PMCID: PMC3700960 DOI: 10.3892/ol.2013.1290] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 03/14/2013] [Indexed: 12/31/2022] Open
Abstract
Invasion and metastasis are major factors in the poor prognosis of pancreatic cancer, which remains one of the most aggressive and lethal diseases worldwide. α-mangostin, a major xanthone compound identified in the pericarp of mangosteen (Garcinia mangostana, Linn; GML), possesses unique biological activities, including antioxidant, antitumor and anti-inflammatory effects. Whether α-mangostin is able to inhibit the invasive ability of pancreatic cancer cells has not been elucidated. In the present study, α-mangostin was shown to inhibit the invasive ability of the pancreatic cancer cell lines MIAPaCa-2 and BxPC-3. The results showed that α-mangostin inhibited the growth of the pancreatic cancer cells in a dose- and time-dependent manner. At concentrations of <5 μM, α-mangostin had no significant effects on cytotoxicity, but significantly inhibited the invasion and migration of pancreatic cancer cells and the expression of matrix metalloproteinase (MMP)-2 and MMP-9, while increasing the expression of E-cadherin. The present data also showed that α-mangostin exerted an inhibitory effect on the phosphorylation of extracellular-signal-regulated kinase (ERK). Furthermore, the reduction of ERK phosphorylation by small interfering RNA (siRNA) potentiated the effect of α-mangostin. Taken together, the data suggest that α-mangostin inhibited the invasion and metastasis of pancreatic cancer cells by reducing MMP-2 and MMP-9 expression, increasing E-cadherin expression and suppressing the ERK signaling pathway. The present study suggests that α-mangostin may be a promising agent against pancreatic cancer.
Collapse
Affiliation(s)
- Jiangtao Yuan
- Department of General Surgery, The Affiliated Hospital of Yan'an University, Yan'an, Shaanxi 716000
| | | | | |
Collapse
|
19
|
Koh JJ, Qiu S, Zou H, Lakshminarayanan R, Li J, Zhou X, Tang C, Saraswathi P, Verma C, Tan DTH, Tan AL, Liu S, Beuerman RW. Rapid bactericidal action of alpha-mangostin against MRSA as an outcome of membrane targeting. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:834-44. [PMID: 22982495 DOI: 10.1016/j.bbamem.2012.09.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 09/05/2012] [Accepted: 09/07/2012] [Indexed: 11/26/2022]
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) has created the need for better therapeutic options. In this study, five natural xanthones were extracted and purified from the fruit hull of Garcinia mangostana and their antimicrobial properties were investigated. α-Mangostin was identified as the most potent among them against Gram-positive pathogens (MIC=0.78-1.56 μg/mL) which included two MRSA isolates. α-Mangostin also exhibited rapid in vitro bactericidal activity (3-log reduction within 5 min). In a multistep (20 passage) resistance selection study using a MRSA isolated from the eye, no resistance against α-mangostin in the strains tested was observed. Biophysical studies using fluorescence probes for membrane potential and permeability, calcein encapsulated large unilamellar vesicles and scanning electron microscopy showed that α-mangostin rapidly disrupted the integrity of the cytoplasmic membrane leading to loss of intracellular components in a concentration-dependent manner. Molecular dynamic simulations revealed that isoprenyl groups were important to reduce the free energy for the burial of the hydrophobic phenyl ring of α-mangostin into the lipid bilayer of the membrane resulting in membrane breakdown and increased permeability. Thus, we suggest that direct interactions of α-mangostin with the bacterial membrane are responsible for the rapid concentration-dependent membrane disruption and bactericidal action.
Collapse
Affiliation(s)
- Jun-Jie Koh
- Singapore Eye Research Institute, 11 Third Hospital Avenue, 168751, Singapore.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Taher M, Susanti D, Rezali MF, Zohri FSA, Ichwan SJA, Alkhamaiseh SI, Ahmad F. Apoptosis, antimicrobial and antioxidant activities of phytochemicals from Garcinia malaccensis Hk.f. ASIAN PAC J TROP MED 2012; 5:136-41. [DOI: 10.1016/s1995-7645(12)60012-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 10/22/2011] [Accepted: 11/15/2011] [Indexed: 11/28/2022] Open
|
21
|
Molina-Jijón E, Tapia E, Zazueta C, El Hafidi M, Zatarain-Barrón ZL, Hernández-Pando R, Medina-Campos ON, Zarco-Márquez G, Torres I, Pedraza-Chaverri J. Curcumin prevents Cr(VI)-induced renal oxidant damage by a mitochondrial pathway. Free Radic Biol Med 2011; 51:1543-57. [PMID: 21839166 DOI: 10.1016/j.freeradbiomed.2011.07.018] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Revised: 07/11/2011] [Accepted: 07/21/2011] [Indexed: 10/17/2022]
Abstract
We report the role of mitochondria in the protective effects of curcumin, a well-known direct and indirect antioxidant, against the renal oxidant damage induced by the hexavalent chromium [Cr(VI)] compound potassium dichromate (K(2)Cr(2)O(7)) in rats. Curcumin was given daily by gavage using three different schemes: (1) complete treatment (100, 200, and 400 mg/kg bw 10 days before and 2 days after K(2)Cr(2)O(7) injection), (2) pretreatment (400 mg/kg bw for 10 days before K(2)Cr(2)O(7) injection), and (3) posttreatment (400 mg/kg bw 2 days after K(2)Cr(2)O(7) injection). Rats were sacrificed 48 h later after a single K(2)Cr(2)O(7) injection (15 mg/kg, sc) to evaluate renal and mitochondrial function and oxidant stress. Curcumin treatment (schemes 1 and 2) attenuated K(2)Cr(2)O(7)-induced renal dysfunction, histological damage, oxidant stress, and the decrease in antioxidant enzyme activity both in kidney tissue and in mitochondria. Curcumin pretreatment attenuated K(2)Cr(2)O(7)-induced mitochondrial dysfunction (alterations in oxygen consumption, ATP content, calcium retention, and mitochondrial membrane potential and decreased activity of complexes I, II, II-III, and V) but was unable to modify renal and mitochondrial Cr(VI) content or to chelate chromium. Curcumin posttreatment was unable to prevent K(2)Cr(2)O(7)-induced renal dysfunction. In further experiments performed in curcumin (400 mg/kg)-pretreated rats it was found that this antioxidant accumulated in kidney and activated Nrf2 at the time when K(2)Cr(2)O(7) was injected, suggesting that both direct and indirect antioxidant effects are involved in the protective effects of curcumin. These findings suggest that the preservation of mitochondrial function plays a key role in the protective effects of curcumin pretreatment against K(2)Cr(2)O(7)-induced renal oxidant damage.
Collapse
Affiliation(s)
- Eduardo Molina-Jijón
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510 University City, DF, Mexico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Gaona-Gaona L, Molina-Jijón E, Tapia E, Zazueta C, Hernández-Pando R, Calderón-Oliver M, Zarco-Márquez G, Pinzón E, Pedraza-Chaverri J. Protective effect of sulforaphane pretreatment against cisplatin-induced liver and mitochondrial oxidant damage in rats. Toxicology 2011; 286:20-7. [PMID: 21575670 DOI: 10.1016/j.tox.2011.04.014] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/21/2011] [Accepted: 04/27/2011] [Indexed: 02/07/2023]
Abstract
In the present work was analyzed whether sulforaphane (SFN) may protect against cisplatin (CIS)-induced hepatic damage, oxidant stress and mitochondrial dysfunction. Four groups of male Wistar rats were studied: control, CIS, CIS+SFN and SFN. SFN was given i.p. (500 μg/kg/d × 3 days) before CIS administration (single i.p. injection, 10mg/kg). Rats were sacrificed 3 days after CIS injection to evaluate hepatic damage (histological analysis, liver/body weight ratio and serum activity of aspartate aminotransferase and alanine aminotransferase), oxidant stress (lipid peroxidation and protein carbonyl and glutathione content), antioxidant enzymes (catalase, glutathione reductase, glutathione peroxidase, glutathione-S-transferase and superoxide dismutase) in liver homogenates and isolated mitochondria and mitochondrial function (oxygen consumption using either malate/glutamate or succinate as substrates and the activity of mitochondrial complex I, II, II-III, IV and V). Furthermore it was evaluated if SFN is able to scavenge some reactive oxygen species in vitro. It was found that SFN prevents CIS-induced (a) hepatic damage, (b) oxidant stress and decreased activity of antioxidant enzymes in liver and mitochondria and (c) mitochondrial alterations in oxygen consumption and decreased activity of mitochondrial complex I. It was also found that the scavenging ability of SFN for peroxynitrite anion, superoxide anion, singlet oxygen, peroxyl radicals, hydrogen peroxide and hydroxyl radicals was very low or negligible. The hepatoprotective effect of SFN was associated to the preservation of mitochondrial function, antioxidant enzymes and prevention of liver and mitochondrial oxidant stress.
Collapse
|
23
|
Protective effect of sulforaphane against cisplatin-induced mitochondrial alterations and impairment in the activity of NAD(P)H: Quinone oxidoreductase 1 and γ glutamyl cysteine ligase: Studies in mitochondria isolated from rat kidney and in LLC-PK1 cells. Toxicol Lett 2010; 199:80-92. [DOI: 10.1016/j.toxlet.2010.08.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 08/12/2010] [Accepted: 08/13/2010] [Indexed: 12/12/2022]
|