Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
The Gut Microbiota Ellagic Acid-Derived Metabolite Urolithin A and Its Sulfate Conjugate Are Substrates for the Drug Efflux Transporter Breast Cancer Resistance Protein (ABCG2/BCRP)
My Activity

Figure 1Loading Img
    Article

    The Gut Microbiota Ellagic Acid-Derived Metabolite Urolithin A and Its Sulfate Conjugate Are Substrates for the Drug Efflux Transporter Breast Cancer Resistance Protein (ABCG2/BCRP)
    Click to copy article linkArticle link copied!

    View Author Information
    Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS-CSIC, P.O. Box 164, 30100 Campus de Espinardo, Murcia, Spain
    Department of Biomedical Sciences-Physiology, Campus Vegazana, University of León, 24071 León, Spain
    # Department of Bioorganic Chemistry, Institute of Chemical Research, CSIC−University of Seville, 41092 Seville, Spain
    *Phone: +34 968 396344. Fax. +34 968 396213. E-mail: [email protected]
    Other Access OptionsSupporting Information (1)

    Journal of Agricultural and Food Chemistry

    Cite this: J. Agric. Food Chem. 2013, 61, 18, 4352–4359
    Click to copy citationCitation copied!
    https://doi.org/10.1021/jf4007505
    Published April 15, 2013
    Copyright © 2013 American Chemical Society

    Abstract

    Click to copy section linkSection link copied!
    Abstract Image

    The breast cancer resistance protein (BCRP/ABCG2) is a drug efflux transporter that can affect the pharmacological and toxicological properties of many molecules. Urolithins, metabolites produced by the gut microbiota from ellagic acid (EA) and ellagitannins, have been acknowledged with in vivo anti-inflammatory and cancer chemopreventive properties. This study evaluated whether urolithins (Uro-A, -B, -C, and -D) and their main phase II metabolites Uro-A sulfate, Uro-A glucuronide, and Uro-B glucuronide as well as their precursor EA were substrates for ABCG2/BCRP. Parental and Bcrp1-transduced MDCKII cells were used for active transport assays. Uro-A and, to a lesser extent, Uro-A sulfate showed a significant increase in apically directed translocation in Bcrp1-transduced cells. Bcrp1 did not show affinity for the rest of the tested compounds. Data were confirmed for murine, human, bovine, and ovine BCRP-transduced subclones as well as with the use of the selective BCRP inhibitor Ko143. The transport inhibition by Uro-A was analyzed by flow cytometry compared to Ko143 using the antineoplastic agent mitoxantrone as a model substrate. Results showed that Uro-A was able to inhibit mitoxantrone transport in a dose-dependent manner. This study reports for the first time that Uro-A and its sulfate conjugate are ABCG2/BCRP substrates. The results suggest that physiologically relevant concentrations of these gut microbiota-derived metabolites could modulate ABCG2/BCRP-mediated transport processes and mechanisms of cancer drug resistance. Further in vivo investigations are warranted.

    Copyright © 2013 American Chemical Society

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. Add or change your institution or let them know you’d like them to include access.

    Supporting Information

    Click to copy section linkSection link copied!

    Experimental procedure for the synthesis of Uro-A glucuronide and Uro-A sulfate. This material is available free of charge via the Internet at http://pubs.acs.org.

    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

    Cited By

    Click to copy section linkSection link copied!

    This article is cited by 67 publications.

    1. Jose M. Villalgordo, Laura Trulli, Rocío García-Villalba, Victor García, Yusuf Althobaiti, Francisco A. Tomás-Barberán. Novel Regioselective Synthesis of Urolithin Glucuronides─Human Gut Microbiota Cometabolites of Ellagitannins and Ellagic Acid. Journal of Agricultural and Food Chemistry 2022, 70 (19) , 5819-5828. https://doi.org/10.1021/acs.jafc.2c00170
    2. Xiao Yang, Francisco A. Tomás-Barberán. Tea Is a Significant Dietary Source of Ellagitannins and Ellagic Acid. Journal of Agricultural and Food Chemistry 2019, 67 (19) , 5394-5404. https://doi.org/10.1021/acs.jafc.8b05010
    3. Gloria Mazzone, Marirosa Toscano, and Nino Russo . Density Functional Predictions of Antioxidant Activity and UV Spectral Features of Nasutin A, Isonasutin, Ellagic Acid, and One of Its Possible Derivatives. Journal of Agricultural and Food Chemistry 2013, 61 (40) , 9650-9657. https://doi.org/10.1021/jf403262k
    4. Hongyan Gou, Ruijie Zeng, Harry Cheuk Hay Lau, Jun Yu. Gut microbial metabolites: Shaping future diagnosis and treatment against gastrointestinal cancer. Pharmacological Research 2024, 208 , 107373. https://doi.org/10.1016/j.phrs.2024.107373
    5. Shiyu Li, Shuangli Zhu, Jun Yu. The role of gut microbiota and metabolites in cancer chemotherapy. Journal of Advanced Research 2024, 64 , 223-235. https://doi.org/10.1016/j.jare.2023.11.027
    6. Nevena Zelenović, Predrag Ristić, Natalija Polović, Tamara Todorović, Milica Kojadinović, Milica Popović. A Multi-Spectroscopic and Molecular Docking Analysis of the Biophysical Interaction between Food Polyphenols, Urolithins, and Human Serum Albumin. Molecules 2024, 29 (18) , 4474. https://doi.org/10.3390/molecules29184474
    7. Jinyi Wang, Tingting Zhou. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Medicinal Research Reviews 2024, 90 https://doi.org/10.1002/med.22077
    8. Noemí Aranda‐Merino, Antonio Marín‐Garrido, Cristina Román‐Hidalgo, María Ramos‐Payán, Nieves Abril, Rut Fernández‐Torres, Miguel Ángel Bello‐López. Bioavailability of flumequine and diclofenac in mice exposed to a metal‐drug chemical cocktail. Evaluation of the protective role of selenium.. British Journal of Pharmacology 2024, 181 (13) , 1935-1951. https://doi.org/10.1111/bph.16312
    9. Yuhang Zhou, Wenjie Han, Yun Feng, Yue Wang, Tao Sun, Junnan Xu. Microbial metabolites affect tumor progression, immunity and therapy prediction by reshaping the tumor microenvironment (Review). International Journal of Oncology 2024, 65 (1) https://doi.org/10.3892/ijo.2024.5661
    10. Laura Álvarez-Fernández, Esther Blanco-Paniagua, Gracia Merino. ABCG2 Transports the Flukicide Nitroxynil and Affects Its Biodistribution and Secretion into Milk. Pharmaceutics 2024, 16 (4) , 558. https://doi.org/10.3390/pharmaceutics16040558
    11. Bowen Zheng, Yuying Wang, Baian Zhou, Fengyuan Qian, Diya Liu, Danrong Ye, Xiqian Zhou, Lin Fang. Urolithin A inhibits breast cancer progression via activating TFEB-mediated mitophagy in tumor macrophages. Journal of Advanced Research 2024, 8 https://doi.org/10.1016/j.jare.2024.04.010
    12. Andrew A. Verdegaal, Andrew L. Goodman. Integrating the gut microbiome and pharmacology. Science Translational Medicine 2024, 16 (732) https://doi.org/10.1126/scitranslmed.adg8357
    13. Gopalsamy Rajiv Gandhi, Poovathumkal James Antony, Stanislaus Antony Ceasar, Alan Bruno Silva Vasconcelos, Monalisa Martins Montalvão, Mariana Nobre Farias de Franca, Ayane de Sá Resende, Chelankara Suresh Sharanya, Yi Liu, Govindasamy Hariharan, Ren-You Gan. Health functions and related molecular mechanisms of ellagitannin-derived urolithins. Critical Reviews in Food Science and Nutrition 2024, 64 (2) , 280-310. https://doi.org/10.1080/10408398.2022.2106179
    14. Jingwei Cai, Alexis Auster, Sungjoon Cho, Zijuan Lai. Dissecting the human gut microbiome to better decipher drug liability: A once-forgotten organ takes center stage. Journal of Advanced Research 2023, 52 , 171-201. https://doi.org/10.1016/j.jare.2023.07.002
    15. Mengwei Zhang, Shumao Cui, Bingyong Mao, Qiuxiang Zhang, Jianxin Zhao, Hao Zhang, Xin Tang, Wei Chen. Ellagic acid and intestinal microflora metabolite urolithin A: A review on its sources, metabolic distribution, health benefits, and biotransformation. Critical Reviews in Food Science and Nutrition 2023, 63 (24) , 6900-6922. https://doi.org/10.1080/10408398.2022.2036693
    16. Georg Aichinger, Maja Stevanoska, Karsten Beekmann, Shana J. Sturla. Physiologically‐Based Pharmacokinetic Modeling of the Postbiotic Supplement Urolithin A Predicts its Bioavailability Is Orders of Magnitude Lower than Concentrations that Induce Toxicity, but also Neuroprotective Effects. Molecular Nutrition & Food Research 2023, 67 (15) https://doi.org/10.1002/mnfr.202300009
    17. Mengchen Liu, Jiaqiang Xu, Qianna Jin, Yunqiao Li. Interaction of Intestinal Microbiota with Medications. Current Drug Metabolism 2023, 24 (8) , 554-567. https://doi.org/10.2174/1389200224666230418104540
    18. Meir Djaldetti. Ellagic Acid - A Dietary Polyphenol with Anticancer Activity that Deserves More Consideration. Recent Progress in Nutrition 2023, 03 (03) , 1-32. https://doi.org/10.21926/rpn.2303011
    19. Qiqing Yang, Bin Wang, Qinghui Zheng, Heyu Li, Xuli Meng, Fangfang Zhou, Long Zhang. A Review of Gut Microbiota‐Derived Metabolites in Tumor Progression and Cancer Therapy. Advanced Science 2023, 10 (15) https://doi.org/10.1002/advs.202207366
    20. Nevena Zelenovic, Milica Kojadinovic, Lidija Filipovic, Vesna Vucic, Milos Milcic, Aleksndra Arsic, Milica Popovic. Interactions of Different Urolithins With Bovine Serum Albumin. Natural Product Communications 2023, 18 (5) , 1934578X2311693. https://doi.org/10.1177/1934578X231169366
    21. Valentina Artusa, Luana Calabrone, Lorenzo Mortara, Francesco Peri, Antonino Bruno. Microbiota-Derived Natural Products Targeting Cancer Stem Cells: Inside the Gut Pharma Factory. International Journal of Molecular Sciences 2023, 24 (5) , 4997. https://doi.org/10.3390/ijms24054997
    22. Lixue Wang, Jinlong Chen, Qijun Chen, Hui Song, Ziqian Wang, Wen Xing, Siyao Jin, Xueying Song, Hua Yang, Wenhua Zhao, . The Gut Microbiota Metabolite Urolithin B Prevents Colorectal Carcinogenesis by Remodeling Microbiota and PD-L1/HLA-B. Oxidative Medicine and Cellular Longevity 2023, 2023 , 1-16. https://doi.org/10.1155/2023/6480848
    23. Aneela Gulnaz, Ji-Eun Chang, Han-Joo Maeng, Kwang-Hee Shin, Kyeong-Ryoon Lee, Yoon-Jee Chae. A mechanism-based understanding of altered drug pharmacokinetics by gut microbiota. Journal of Pharmaceutical Investigation 2023, 53 (1) , 73-92. https://doi.org/10.1007/s40005-022-00600-z
    24. Alessia Favaron, Laura E. McCoubrey, Moe Elbadawi, Abdul W. Basit, Mine Orlu. The Ageing Microbiome, Pharmaceutical Considerations, and Therapeutic Opportunities. 2023, 191-230. https://doi.org/10.1007/978-3-031-35811-1_8
    25. Than S. Kyaw, Peter J. Turnbaugh. Tiny Gatekeepers: Microbial Control of Host Drug Transporters. Clinical Pharmacology & Therapeutics 2022, 112 (3) , 443-445. https://doi.org/10.1002/cpt.2647
    26. Reynaldo Moreno Uclés, Antonio González-Sarrías, Juan Carlos Espín, Francisco A. Tomás-Barberán, Marlene Janes, Henrique Cheng, John Finley, Frank Greenway, Jack N Losso. Effects of red raspberry polyphenols and metabolites on the biomarkers of inflammation and insulin resistance in type 2 diabetes: a pilot study. Food & Function 2022, 13 (9) , 5166-5176. https://doi.org/10.1039/D1FO02090K
    27. Rama Rao Malla, Rakshmitha Marni, Seema Kumari, Aninditha Chakraborty, Pappu Lalitha. Microbiome Assisted Tumor Microenvironment: Emerging Target of Breast Cancer. Clinical Breast Cancer 2022, 22 (3) , 200-211. https://doi.org/10.1016/j.clbc.2021.09.002
    28. Laura E. McCoubrey, Hannah Batchelor, Abdul W. Basit, Simon Gaisford, Mine Orlu. Impact of the Microbiome on Oral Biopharmaceutics. 2022, 277-295. https://doi.org/10.1002/9781119678366.ch17
    29. Seyyed Hossein Hasheminezhad, Motahareh Boozari, Mehrdad Iranshahi, Omid Yazarlu, Amirhossein Sahebkar, Maede Hasanpour, Milad Iranshahy. A mechanistic insight into the biological activities of urolithins as gut microbial metabolites of ellagitannins. Phytotherapy Research 2022, 36 (1) , 112-146. https://doi.org/10.1002/ptr.7290
    30. Laura E. McCoubrey, Simon Gaisford, Mine Orlu, Abdul W. Basit. Predicting drug-microbiome interactions with machine learning. Biotechnology Advances 2022, 54 , 107797. https://doi.org/10.1016/j.biotechadv.2021.107797
    31. Jiajia Zhou, Jian Ouyang, Zihao Gao, Haipeng Qin, Wu Jun, Tieliu Shi. MagMD: Database summarizing the metabolic action of gut microbiota to drugs. Computational and Structural Biotechnology Journal 2022, 20 , 6427-6430. https://doi.org/10.1016/j.csbj.2022.11.021
    32. Xintong Zhang, Ying Han, Wei Huang, Mingji Jin, Zhonggao Gao. The influence of the gut microbiota on the bioavailability of oral drugs. Acta Pharmaceutica Sinica B 2021, 11 (7) , 1789-1812. https://doi.org/10.1016/j.apsb.2020.09.013
    33. Ashutosh Gupta, Amit Kumar Singh, Ramesh Kumar, Sarah Jamieson, Abhay Kumar Pandey, Anupam Bishayee. Neuroprotective Potential of Ellagic Acid: A Critical Review. Advances in Nutrition 2021, 12 (4) , 1211-1238. https://doi.org/10.1093/advances/nmab007
    34. Jingjing Tu, Qiaoling Li, Benhong Zhou. The Tannins from Sanguisorba officinalis L. (Rosaceae): A Systematic Study on the Metabolites of Rats Based on HPLC–LTQ–Orbitrap MS2 Analysis. Molecules 2021, 26 (13) , 4053. https://doi.org/10.3390/molecules26134053
    35. J. Ansary, T.Y. Forbes-Hernandez, L. Regolo, D. Cianciosi, F. Giampieri. The anticancer activity of strawberry. Acta Horticulturae 2021, 38 (1309) , 925-932. https://doi.org/10.17660/ActaHortic.2021.1309.132
    36. Hitoshi Abe, Katsunori Itaya, Ishtiaq Jeelani. Total Synthesis of Urolithin C 3-Glucuronide. HETEROCYCLES 2021, 103 (2) , 1038. https://doi.org/10.3987/COM-20-S(K)51
    37. Jian Li, Yaqi Zhang, Shen Yang, Zhenhua Lu, Guiling Li, Shangyi Wu, Da-Ren Wu, Jingwen Liu, Bo Zhou, Hui-Min David Wang, , Shi-Ying Huang. The Beneficial Effects of Edible Kynurenic Acid from Marine Horseshoe Crab ( Tachypleus tridentatus ) on Obesity, Hyperlipidemia, and Gut Microbiota in High‐Fat Diet‐Fed Mice. Oxidative Medicine and Cellular Longevity 2021, 2021 (1) https://doi.org/10.1155/2021/8874503
    38. Claudia Ceci, Grazia Graziani, Isabella Faraoni, Ilaria Cacciotti. Strategies to improve ellagic acid bioavailability: from natural or semisynthetic derivatives to nanotechnological approaches based on innovative carriers. Nanotechnology 2020, 31 (38) , 382001. https://doi.org/10.1088/1361-6528/ab912c
    39. Jelena Tošović, Urban Bren. Antioxidative Action of Ellagic Acid—A Kinetic DFT Study. Antioxidants 2020, 9 (7) , 587. https://doi.org/10.3390/antiox9070587
    40. Juan Antonio Giménez-Bastida, María Ángeles Ávila-Gálvez, Juan Carlos Espín, Antonio González-Sarrías. The gut microbiota metabolite urolithin A, but not other relevant urolithins, induces p53-dependent cellular senescence in human colon cancer cells. Food and Chemical Toxicology 2020, 139 , 111260. https://doi.org/10.1016/j.fct.2020.111260
    41. Md Alauddin, Toshiyuki Okumura, Janet Rajaxavier, Shayan Khozooei, Simone Pöschel, Satoru Takeda, Yogesh Singh, Sara Y Brucker, Diethelm Wallwiener, André Koch, Madhuri S Salker. Gut Bacterial Metabolite Urolithin A Decreases Actin Polymerization and Migration in Cancer Cells. Molecular Nutrition & Food Research 2020, 64 (7) https://doi.org/10.1002/mnfr.201900390
    42. Min‐yi Lv, Chuan‐jian Shi, Fei‐fei Pan, Jiang Shao, Lu Feng, Guoqin Chen, Caiwen Ou, Jin‐fang Zhang, Wei‐ming Fu. Urolithin B suppresses tumor growth in hepatocellular carcinoma through inducing the inactivation of Wnt/β‐catenin signaling. Journal of Cellular Biochemistry 2019, 120 (10) , 17273-17282. https://doi.org/10.1002/jcb.28989
    43. Liming Chen, Jose E. Manautou, Theodore P. Rasmussen, Xiao-bo Zhong. Development of precision medicine approaches based on inter-individual variability of BCRP/ABCG2. Acta Pharmaceutica Sinica B 2019, 9 (4) , 659-674. https://doi.org/10.1016/j.apsb.2019.01.007
    44. M. A. Ávila-Gálvez, J. A. Giménez-Bastida, A. González-Sarrías, J. C. Espín. Tissue deconjugation of urolithin A glucuronide to free urolithin A in systemic inflammation. Food & Function 2019, 10 (6) , 3135-3141. https://doi.org/10.1039/C9FO00298G
    45. Kim Ohanna Pimenta Inada, Francisco A. Tomás-Barberán, Daniel Perrone, Mariana Monteiro. Metabolism of ellagitannins from jabuticaba (Myrciaria jaboticaba) in normoweight, overweight and obese Brazilians: Unexpected laxative effects influence urolithins urinary excretion and metabotype distribution. Journal of Functional Foods 2019, 57 , 299-308. https://doi.org/10.1016/j.jff.2019.04.025
    46. Liping Wang, Rongjin Sun, Qisong Zhang, Qing Luo, Sijing Zeng, Xiaoyan Li, Xia Gong, Yuhuan Li, Linlin Lu, Ming Hu, Zhongqiu Liu. An update on polyphenol disposition via coupled metabolic pathways. Expert Opinion on Drug Metabolism & Toxicology 2019, 15 (2) , 151-165. https://doi.org/10.1080/17425255.2019.1559815
    47. José M. Lorenzo, Paulo E. Munekata, Predrag Putnik, Danijela Bursać Kovačević, Voster Muchenje, Francisco J. Barba. Sources, Chemistry, and Biological Potential of Ellagitannins and Ellagic Acid Derivatives. 2019, 189-221. https://doi.org/10.1016/B978-0-444-64181-6.00006-1
    48. Dafne García-Mateos, Rocío García-Villalba, Jon A. Otero, José A. Marañón, Juan C. Espín, Ana I. Álvarez, Gracia Merino. An altered tissue distribution of flaxseed lignans and their metabolites in Abcg2 knockout mice. Food & Function 2018, 9 (1) , 636-642. https://doi.org/10.1039/C7FO01549F
    49. Milica Kojadinovic, Aleksandra Arsic, Gordana Petovic-Oggiano, Marija Gavrovic-Jankulovic, Marija Glibetic, Milica Popovic. Effect of urolithins on oxidative stress of colorectal adenocarcinomacells-Caco-2. International Journal of Food Sciences and Nutrition 2017, 68 (8) , 952-959. https://doi.org/10.1080/09637486.2017.1328665
    50. Ting Wu, Charlotte Grootaert, Stefan Voorspoels, Griet Jacobs, Judit Pitart, Senem Kamiloglu, Sam Possemiers, Marina Heinonen, Nevena Kardum, Maria Glibetic, Guy Smagghe, Katleen Raes, John Van Camp. Aronia ( Aronia melanocarpa ) phenolics bioavailability in a combined in vitro digestion/Caco-2 cell model is structure and colon region dependent. Journal of Functional Foods 2017, 38 , 128-139. https://doi.org/10.1016/j.jff.2017.09.008
    51. . Impact of Intestinal Efflux Transporters on Oral Absorption. 2017, 203-267. https://doi.org/10.1002/9781118916926.ch6
    52. Jakub P. Piwowarski, Iwona Stanisławska, Sebastian Granica, Joanna Stefańska, Anna K. Kiss. Phase II Conjugates of Urolithins Isolated from Human Urine and Potential Role of β -Glucuronidases in Their Disposition. Drug Metabolism and Disposition 2017, 45 (6) , 657-665. https://doi.org/10.1124/dmd.117.075200
    53. Antonio González-Sarrías, María Ángeles Núñez-Sánchez, Rocío García-Villalba, Francisco A. Tomás-Barberán, Juan Carlos Espín. Antiproliferative activity of the ellagic acid-derived gut microbiota isourolithin A and comparison with its urolithin A isomer: the role of cell metabolism. European Journal of Nutrition 2017, 56 (2) , 831-841. https://doi.org/10.1007/s00394-015-1131-7
    54. Francisco A. Tomás‐Barberán, Antonio González‐Sarrías, Rocío García‐Villalba, María A. Núñez‐Sánchez, María V. Selma, María T. García‐Conesa, Juan Carlos Espín. Urolithins, the rescue of “old” metabolites to understand a “new” concept: Metabotypes as a nexus among phenolic metabolism, microbiota dysbiosis, and host health status. Molecular Nutrition & Food Research 2017, 61 (1) https://doi.org/10.1002/mnfr.201500901
    55. E. Van Rymenant, C. Grootaert, K. Beerens, P. W. Needs, P. A. Kroon, A. Kerimi, G. Williamson, R. García-Villalba, A. González-Sarrías, F. Tomas-Barberan, J. Van Camp, J. Van de Voorde. Vasorelaxant activity of twenty-one physiologically relevant (poly)phenolic metabolites on isolated mouse arteries. Food & Function 2017, 8 (12) , 4331-4335. https://doi.org/10.1039/C7FO01273J
    56. María Ángeles Núñez-Sánchez, Ankur Karmokar, Antonio González-Sarrías, Rocío García-Villalba, Francisco A. Tomás-Barberán, María Teresa García-Conesa, Karen Brown, Juan Carlos Espín. In vivo relevant mixed urolithins and ellagic acid inhibit phenotypic and molecular colon cancer stem cell features: A new potentiality for ellagitannin metabolites against cancer. Food and Chemical Toxicology 2016, 92 , 8-16. https://doi.org/10.1016/j.fct.2016.03.011
    57. Rocío García-Villalba, Juan Carlos Espín, Francisco A. Tomás-Barberán. Chromatographic and spectroscopic characterization of urolithins for their determination in biological samples after the intake of foods containing ellagitannins and ellagic acid. Journal of Chromatography A 2016, 1428 , 162-175. https://doi.org/10.1016/j.chroma.2015.08.044
    58. Wylly Ramsés García-Niño, Cecilia Zazueta. Ellagic acid: Pharmacological activities and molecular mechanisms involved in liver protection. Pharmacological Research 2015, 97 , 84-103. https://doi.org/10.1016/j.phrs.2015.04.008
    59. Ana Ramírez de Molina, Teodoro Vargas, Susana Molina, Jenifer Sánchez, Jorge Martínez-Romero, Margarita González-Vallinas, Roberto Martín-Hernández, Ruth Sánchez-Martínez, Marta Gómez de Cedrón, Alberto Dávalos, Luca Calani, Daniele Del Rio, Antonio González-Sarrías, Juan Carlos Espín, Francisco A. Tomás-Barberán, Guillermo Reglero. The Ellagic Acid Derivative 4,4′-Di- O -Methylellagic Acid Efficiently Inhibits Colon Cancer Cell Growth through a Mechanism Involving WNT16. Journal of Pharmacology and Experimental Therapeutics 2015, 353 (2) , 433-444. https://doi.org/10.1124/jpet.114.221796
    60. Antonio González-Sarrías, Joao Tomé-Carneiro, Andrea Bellesia, Francisco A. Tomás-Barberán, Juan Carlos Espín. The ellagic acid-derived gut microbiota metabolite, urolithin A, potentiates the anticancer effects of 5-fluorouracil chemotherapy on human colon cancer cells. Food & Function 2015, 6 (5) , 1460-1469. https://doi.org/10.1039/C5FO00120J
    61. Ana Rodriguez-Mateos, David Vauzour, Christian G. Krueger, Dhanansayan Shanmuganayagam, Jess Reed, Luca Calani, Pedro Mena, Daniele Del Rio, Alan Crozier. Bioavailability, bioactivity and impact on health of dietary flavonoids and related compounds: an update. Archives of Toxicology 2014, 88 (10) , 1803-1853. https://doi.org/10.1007/s00204-014-1330-7
    62. Guillermo Moreno-Sanz, Borja Barrera, Andrea Armirotti, Sine M. Bertozzi, Rita Scarpelli, Tiziano Bandiera, Julio G. Prieto, Andrea Duranti, Giorgio Tarzia, Gracia Merino, Daniele Piomelli. Structural determinants of peripheral O-arylcarbamate FAAH inhibitors render them dual substrates for Abcb1 and Abcg2 and restrict their access to the brain. Pharmacological Research 2014, 87 , 87-93. https://doi.org/10.1016/j.phrs.2014.06.004
    63. David M. Klein, Stephen H. Wright, Nathan J. Cherrington. Xenobiotic transporter expression along the male genital tract. Reproductive Toxicology 2014, 47 , 1-8. https://doi.org/10.1016/j.reprotox.2014.04.009
    64. María A. Nuñez-Sánchez, Rocío García-Villalba, Tamara Monedero-Saiz, Noelia V. García-Talavera, María B. Gómez-Sánchez, Carmen Sánchez-Álvarez, Ana M. García-Albert, Francisco J. Rodríguez-Gil, Miguel Ruiz-Marín, Francisco A. Pastor-Quirante, Francisco Martínez-Díaz, María J. Yáñez-Gascón, Antonio González-Sarrías, Francisco A. Tomás-Barberán, Juan C. Espín. Targeted metabolic profiling of pomegranate polyphenols and urolithins in plasma, urine and colon tissues from colorectal cancer patients. Molecular Nutrition & Food Research 2014, 58 (6) , 1199-1211. https://doi.org/10.1002/mnfr.201300931
    65. Antonio González-Sarrías, Juan Antonio Giménez-Bastida, María Ángeles Núñez-Sánchez, Mar Larrosa, María Teresa García-Conesa, Francisco A. Tomás-Barberán, Juan Carlos Espín. Phase-II metabolism limits the antiproliferative activity of urolithins in human colon cancer cells. European Journal of Nutrition 2014, 53 (3) , 853-864. https://doi.org/10.1007/s00394-013-0589-4
    66. W. Elaine Hardman. Diet components can suppress inflammation and reduce cancer risk. Nutrition Research and Practice 2014, 8 (3) , 233. https://doi.org/10.4162/nrp.2014.8.3.233
    67. Luca Dellafiora, Pedro Mena, Pietro Cozzini, Furio Brighenti, Daniele Del Rio. Modelling the possible bioactivity of ellagitannin-derived metabolites. In silico tools to evaluate their potential xenoestrogenic behavior. Food & Function 2013, 4 (10) , 1442. https://doi.org/10.1039/c3fo60117j

    Journal of Agricultural and Food Chemistry

    Cite this: J. Agric. Food Chem. 2013, 61, 18, 4352–4359
    Click to copy citationCitation copied!
    https://doi.org/10.1021/jf4007505
    Published April 15, 2013
    Copyright © 2013 American Chemical Society

    Article Views

    1890

    Altmetric

    -

    Citations

    Learn about these metrics

    Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

    Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

    The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.