ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Bile Acid Profiling and Quantification in Biofluids Using Ultra-Performance Liquid Chromatography Tandem Mass Spectrometry

View Author Information
Imperial College of London, Division of Computational Systems Medicine, Department of Surgery and Cancer, Sir Alexander Building, Exhibition Road, South Kensington, London SW7 2AZ, United Kingdom
Imperial College of London, MRC-NHR National Phenome Centre, Department of Surgery and Cancer, IRDB building, Du Cane Road, London W12 0NN, United Kingdom
§ Imperial College of London, Department of Hepatology, St. Mary’s Hospital, Paddington, London, United Kingdom
King’s College London, Institute of Liver Sciences, Hospital NHS Foundation Trust, Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, London, United Kingdom
Cite this: Anal. Chem. 2015, 87, 19, 9662–9670
Publication Date (Web):September 1, 2015
https://doi.org/10.1021/acs.analchem.5b01556
Copyright © 2015 American Chemical Society

    Article Views

    6410

    Altmetric

    -

    Citations

    LEARN ABOUT THESE METRICS
    Read OnlinePDF (2 MB)
    Supporting Info (1)»

    Abstract

    Abstract Image

    Bile acids are important end products of cholesterol metabolism. While they have been identified as key factors in lipid emulsification and absorption due to their detergent properties, bile acids have also been shown to act as signaling molecules and intermediates between the host and the gut microbiota. To further the investigation of bile acid functions in humans, an advanced platform for high throughput analysis is essential. Herein, we describe the development and application of a 15 min UPLC procedure for the separation of bile acid species from human biofluid samples requiring minimal sample preparation. High resolution time-of-flight mass spectrometry was applied for profiling applications, elucidating rich bile acid profiles in both normal and disease state plasma. In parallel, a second mode of detection was developed utilizing tandem mass spectrometry for sensitive and quantitative targeted analysis of 145 bile acid (BA) species including primary, secondary, and tertiary bile acids. The latter system was validated by testing the linearity (lower limit of quantification, LLOQ, 0.25–10 nM and upper limit of quantification, ULOQ, 2.5–5 μM), precision (≈6.5%), and accuracy (81.2–118.9%) on inter- and intraday analysis achieving good recovery of bile acids (serum/plasma 88% and urine 93%). The ultra performance liquid chromatography–mass spectrometry (UPLC-MS)/MS targeted method was successfully applied to plasma, serum, and urine samples in order to compare the bile acid pool compositional difference between preprandial and postprandial states, demonstrating the utility of such analysis on human biofluids.

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.analchem.5b01556.

    • Sulfation scheme; tables of UPLC-MS/MS settings for the bile acid standards; table of chromatographic gradients; table of percent yields of reactions; table of intra- and interday validation of accuracy and precision; table of quantitiative matrix effects; table of evaluated carry-over; figure of deuterated standards recovery; table of recoveries and standard deviations (SD) of internally labeled standards (PDF)

    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

    This article is cited by 141 publications.

    1. Robin H.J. Kemperman, Christopher D. Chouinard, Richard A. Yost. Characterization of Bile Acid Isomers and the Implementation of High-Resolution Demultiplexing with Ion Mobility-Mass Spectrometry. Journal of the American Society for Mass Spectrometry 2023, 34 (7) , 1477-1490. https://doi.org/10.1021/jasms.3c00143
    2. Yang Zhang, Yuxi Huang, Jingjing Fan, Meng Zhang, Aobulikasimu Hasan, Yang Yi, Rong Yu, Xujie Zhou, Min Ye, Xue Qiao. Expanding the Scope of Targeted Metabolomics by One-pot Microscale Synthesis and Tailored Metabolite Profiling: Investigation of Bile Acid–Amino Acid Conjugates. Analytical Chemistry 2022, 94 (48) , 16596-16603. https://doi.org/10.1021/acs.analchem.2c02086
    3. Yan Cao, Wei Li, Wei Chen, Xiaoya Niu, Nian Wu, Yitao Wang, Jun Li, Pengfei Tu, Jiao Zheng, Yuelin Song. Squared Energy-Resolved Mass Spectrometry Advances Quantitative Bile Acid Submetabolome Characterization. Analytical Chemistry 2022, 94 (44) , 15395-15404. https://doi.org/10.1021/acs.analchem.2c03269
    4. Yan Ma, Yang Cao, Xiaocui Song, Yuanying Zhang, Jing Li, Yankai Wang, Xiaoqing Wu, Xiangbing Qi. BAFinder: A Software for Unknown Bile Acid Identification Using Accurate Mass LC-MS/MS in Positive and Negative Modes. Analytical Chemistry 2022, 94 (16) , 6242-6250. https://doi.org/10.1021/acs.analchem.1c05648
    5. Quan-Fei Zhu, Yan-Zhen Wang, Na An, Jun-Di Hao, Peng-Cheng Mei, Ya-Li Bai, Yu-Ning Hu, Pei-Rong Bai, Yu-Qi Feng. Alternating Dual-Collision Energy Scanning Mass Spectrometry Approach: Discovery of Novel Microbial Bile-Acid Conjugates. Analytical Chemistry 2022, 94 (5) , 2655-2664. https://doi.org/10.1021/acs.analchem.1c05272
    6. Dewakar Sangaraju, Yao Shi, Michael Van Parys, Adam Ray, Abigail Walker, Rachel Caminiti, Dennis Milanowski, Allan Jaochico, Brian Dean, Xiaorong Liang. Robust and Comprehensive Targeted Metabolomics Method for Quantification of 50 Different Primary, Secondary, and Sulfated Bile Acids in Multiple Biological Species (Human, Monkey, Rabbit, Dog, and Rat) and Matrices (Plasma and Urine) Using Liquid Chromatography High Resolution Mass Spectrometry (LC-HRMS) Analysis. Journal of the American Society for Mass Spectrometry 2021, 32 (8) , 2033-2049. https://doi.org/10.1021/jasms.0c00435
    7. Hsin-Yu Liao, Chin-Yi Wang, Ching-Hua Lee, Hsien-Li Kao, Wei-Kai Wu, Ching-Hua Kuo. Development of an Efficient and Sensitive Chemical Derivatization-Based LC–MS/MS Method for Quantifying Gut Microbiota-Derived Metabolites in Human Plasma and Its Application in Studying Cardiovascular Disease. Journal of Proteome Research 2021, 20 (7) , 3508-3518. https://doi.org/10.1021/acs.jproteome.1c00147
    8. Jiamin Zheng, Lun Zhang, Mathew Johnson, Rupasri Mandal, David S. Wishart. Comprehensive Targeted Metabolomic Assay for Urine Analysis. Analytical Chemistry 2020, 92 (15) , 10627-10634. https://doi.org/10.1021/acs.analchem.0c01682
    9. Carolina González-Riano, Danuta Dudzik, Antonia Garcia, Alberto Gil-de-la-Fuente, Ana Gradillas, Joanna Godzien, Ángeles López-Gonzálvez, Fernanda Rey-Stolle, David Rojo, Francisco J. Ruperez, Jorge Saiz, Coral Barbas. Recent Developments along the Analytical Process for Metabolomics Workflows. Analytical Chemistry 2020, 92 (1) , 203-226. https://doi.org/10.1021/acs.analchem.9b04553
    10. Jashbir Singh, Rita Metrani, Siddanagouda R. Shivanagoudra, Guddadarangavvanahally K. Jayaprakasha, Bhimanagouda S. Patil. Review on Bile Acids: Effects of the Gut Microbiome, Interactions with Dietary Fiber, and Alterations in the Bioaccessibility of Bioactive Compounds. Journal of Agricultural and Food Chemistry 2019, 67 (33) , 9124-9138. https://doi.org/10.1021/acs.jafc.8b07306
    11. Christopher D. Chouinard, Gabe Nagy, Ian K. Webb, Sandilya V. B. Garimella, Erin S. Baker, Yehia M. Ibrahim, Richard D. Smith. Rapid Ion Mobility Separations of Bile Acid Isomers Using Cyclodextrin Adducts and Structures for Lossless Ion Manipulations. Analytical Chemistry 2018, 90 (18) , 11086-11091. https://doi.org/10.1021/acs.analchem.8b02990
    12. Patrick Brophy, Corey D. Broeckling, James Murphy, Jessica E. Prenni. Ion-neutral Clustering of Bile Acids in Electrospray Ionization Across UPLC Flow Regimes. Journal of the American Society for Mass Spectrometry 2018, 29 (4) , 651-662. https://doi.org/10.1007/s13361-017-1878-6
    13. Elena Chekmeneva, Gonçalo dos Santos Correia, Queenie Chan, Anisha Wijeyesekera, Adrienne Tin, Jeffery Hunter Young, Paul Elliott, Jeremy K. Nicholson, and Elaine Holmes . Optimization and Application of Direct Infusion Nanoelectrospray HRMS Method for Large-Scale Urinary Metabolic Phenotyping in Molecular Epidemiology. Journal of Proteome Research 2017, 16 (4) , 1646-1658. https://doi.org/10.1021/acs.jproteome.6b01003
    14. Nicola Gray, Rabiya Zia, Adam King, Vishal C. Patel, Julia Wendon, Mark J. W. McPhail, Muireann Coen, Robert S. Plumb, Ian D. Wilson, and Jeremy K. Nicholson . High-Speed Quantitative UPLC-MS Analysis of Multiple Amines in Human Plasma and Serum via Precolumn Derivatization with 6-Aminoquinolyl-N-hydroxysuccinimidyl Carbamate: Application to Acetaminophen-Induced Liver Failure. Analytical Chemistry 2017, 89 (4) , 2478-2487. https://doi.org/10.1021/acs.analchem.6b04623
    15. Robert G. Nichols, Nicole E. Hume, Philip B. Smith, Jeffrey M. Peters, and Andrew D. Patterson . Omics Approaches To Probe Microbiota and Drug Metabolism Interactions. Chemical Research in Toxicology 2016, 29 (12) , 1987-1997. https://doi.org/10.1021/acs.chemrestox.6b00236
    16. Ke Lan, Mingming Su, Guoxiang Xie, Brian C. Ferslew, Kim L.R. Brouwer, Cynthia Rajani, Changxiao Liu, and Wei Jia . Key Role for the 12-Hydroxy Group in the Negative Ion Fragmentation of Unconjugated C24 Bile Acids. Analytical Chemistry 2016, 88 (14) , 7041-7048. https://doi.org/10.1021/acs.analchem.6b00573
    17. Laura Martinez-Gili, Alexandros Pechlivanis, Julie A.K. McDonald, Sofina Begum, Jonathan Badrock, Jessica K. Dyson, Rebecca Jones, Gideon Hirschfield, Stephen D. Ryder, Richard Sandford, Simon Rushbrook, Douglas Thorburn, Simon D. Taylor-Robinson, Mary M.E. Crossey, Julian R. Marchesi, George Mells, Elaine Holmes, David Jones. Bacterial and metabolic phenotypes associated with inadequate response to ursodeoxycholic acid treatment in primary biliary cholangitis. Gut Microbes 2023, 15 (1) https://doi.org/10.1080/19490976.2023.2208501
    18. Chen Yang, Zhenxia Xu, Qingde Huang, Xu Wang, Fenghong Huang, Qianchun Deng. Targeted microbiome metabolomics reveals flaxseed oil supplementation regulated the gut microbiota and farnesoid X receptor pathway in high-fat diet mice. Food Science and Human Wellness 2023, 12 (6) , 2324-2335. https://doi.org/10.1016/j.fshw.2023.03.036
    19. Yan Jin, Jinhua Chi, Kaelene LoMonaco, Alexandria Boon, Haiwei Gu. Recent review on selected xenobiotics and their impacts on gut microbiome and metabolome. TrAC Trends in Analytical Chemistry 2023, 166 , 117155. https://doi.org/10.1016/j.trac.2023.117155
    20. Victoria Ramos-Garcia, Isabel Ten-Doménech, Máximo Vento, Clara Bullich-Vilarrubias, Marina Romaní-Pérez, Yolanda Sanz, Angelica Nobili, Marika Falcone, Marina Di Stefano, Guillermo Quintás, Julia Kuligowski. Fast profiling of primary, secondary, conjugated, and sulfated bile acids in human urine and murine feces samples. Analytical and Bioanalytical Chemistry 2023, 415 (20) , 4961-4971. https://doi.org/10.1007/s00216-023-04802-8
    21. Bertrand Routy, John G. Lenehan, Wilson H. Miller, Rahima Jamal, Meriem Messaoudene, Brendan A. Daisley, Cecilia Hes, Kait F. Al, Laura Martinez-Gili, Michal Punčochář, Scott Ernst, Diane Logan, Karl Belanger, Khashayar Esfahani, Corentin Richard, Marina Ninkov, Gianmarco Piccinno, Federica Armanini, Federica Pinto, Mithunah Krishnamoorthy, Rene Figueredo, Pamela Thebault, Panteleimon Takis, Jamie Magrill, LeeAnn Ramsay, Lisa Derosa, Julian R. Marchesi, Seema Nair Parvathy, Arielle Elkrief, Ian R. Watson, Rejean Lapointe, Nicola Segata, S.M. Mansour Haeryfar, Benjamin H. Mullish, Michael S. Silverman, Jeremy P. Burton, Saman Maleki Vareki. Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: a phase I trial. Nature Medicine 2023, 29 (8) , 2121-2132. https://doi.org/10.1038/s41591-023-02453-x
    22. Jie Liu, Huaying Fang, Na Hong, Chaolan Lv, Qihua Zhu, Yinping Feng, Bo Wang, Jiashuang Tian, Yue Yu, . Gut Microbiome and Metabonomic Profile Predict Early Remission to Anti-Integrin Therapy in Patients with Moderate to Severe Ulcerative Colitis. Microbiology Spectrum 2023, 11 (3) https://doi.org/10.1128/spectrum.01457-23
    23. Victoria Ramos-Garcia, Isabel Ten-Doménech, Alba Moreno-Giménez, Laura Campos-Berga, Anna Parra-Llorca, Amparo Ramón-Beltrán, María J. Vaya, Fady Mohareb, Corentin Molitor, Paulo Refinetti, Andrei Silva, Luis A. Rodrigues, Serge Rezzi, Andrew C. C. Hodgson, Stéphane Canarelli, Eirini Bathrellou, Eirini Mamalaki, Melina Karipidou, Dimitrios Poulimeneas, Mary Yannakoulia, Christopher K. Akhgar, Andreas Schwaighofer, Bernhard Lendl, Jennifer Karrer, Davide Migliorelli, Silvia Generelli, María Gormaz, Miltiadis Vasileiadis, Julia Kuligowski, Máximo Vento. Fact-based nutrition for infants and lactating mothers—The NUTRISHIELD study. Frontiers in Pediatrics 2023, 11 https://doi.org/10.3389/fped.2023.1130179
    24. Sebastian Simstich, Günter Fauler. Bile Acids. 2023, 509-529. https://doi.org/10.1002/9783527836512.ch17
    25. Xuan ZHU, En-Qin XIA, Yu-Guo LIU, Wei-Qiao LI, Yan XIN, Xiao-Zhuan LIN, Pavel MAROZIK, Hong-Hui GUO. Characterization and quantification of representative bile acids in ileal contents and feces of diet-induced obese mice by UPLC-MS/MS. Chinese Journal of Analytical Chemistry 2023, 51 (3) , 100175. https://doi.org/10.1016/j.cjac.2022.100175
    26. James L. Alexander, Benjamin H. Mullish, Nathan P. Danckert, Zhigang Liu, Marton L. Olbei, Aamir Saifuddin, Melissa Torkizadeh, Hajir Ibraheim, Jesús Miguéns Blanco, Lauren A. Roberts, Claire M. Bewshea, Rachel Nice, Simeng Lin, Hemanth Prabhudev, Caroline Sands, Verena Horneffer-van der Sluis, Matthew Lewis, Shaji Sebastian, Charlie W. Lees, Julian P. Teare, Ailsa Hart, James R. Goodhand, Nicholas A. Kennedy, Tamas Korcsmaros, Julian R. Marchesi, Tariq Ahmad, Nick Powell. The gut microbiota and metabolome are associated with diminished COVID-19 vaccine-induced antibody responses in immunosuppressed inflammatory bowel disease patients. eBioMedicine 2023, 88 , 104430. https://doi.org/10.1016/j.ebiom.2022.104430
    27. Lucy M.V. Gee, Ben Barron-Millar, Jack Leslie, Claire Richardson, Marco Y.W. Zaki, Saimir Luli, Rachel A. Burgoyne, Rainie I.T. Cameron, Graham R. Smith, John G. Brain, Barbara Innes, Laura Jopson, Jessica K. Dyson, Katherine R.C. McKay, Alexandros Pechlivanis, Elaine Holmes, Rolando Berlinguer-Palmini, Stella Victorelli, George F. Mells, Richard N. Sandford, Jeremy Palmer, John A. Kirby, Christos Kiourtis, Joao Mokochinski, Zoe Hall, Thomas G. Bird, Lee A. Borthwick, Christopher M. Morris, Peter S. Hanson, Diana Jurk, Elizabeth A. Stoll, Fiona E.N. LeBeau, David E.J. Jones, Fiona Oakley. Anti–Cholestatic Therapy with Obeticholic Acid Improves Short-Term Memory in Bile Duct–Ligated Mice. The American Journal of Pathology 2023, 193 (1) , 11-26. https://doi.org/10.1016/j.ajpath.2022.09.005
    28. Jiating Zheng, Xiaoru Peng, Taifeng Zhu, Shuyao Huang, Chao Chen, Guosheng Chen, Shuqin Liu, Gangfeng Ouyang. Detection of bile acids in small volume human bile samples via an amino metal-organic framework composite based solid-phase microextraction probe. Journal of Chromatography A 2022, 1685 , 463634. https://doi.org/10.1016/j.chroma.2022.463634
    29. Keith Z. Hazleton, Casey G. Martin, David J. Orlicky, Kathleen L. Arnolds, Nichole M. Nusbacher, Nancy Moreno-Huizar, Michael Armstrong, Nichole Reisdorph, Catherine A. Lozupone. Dietary fat promotes antibiotic-induced Clostridioides difficile mortality in mice. npj Biofilms and Microbiomes 2022, 8 (1) https://doi.org/10.1038/s41522-022-00276-1
    30. Iain Robert Louis Kean, Joseph Wagner, Anisha Wijeyesekera, Marcus De Goffau, Sarah Thurston, John A. Clark, Deborah K. White, Jenna Ridout, Shruti Agrawal, Riaz Kayani, Roddy O’Donnell, Padmanabhan Ramnarayan, Mark J. Peters, Nigel Klein, Elaine Holmes, Julian Parkhill, Stephen Baker, Nazima Pathan. Profiling gut microbiota and bile acid metabolism in critically ill children. Scientific Reports 2022, 12 (1) https://doi.org/10.1038/s41598-022-13640-0
    31. Nicholas C. Penney, Derek K. T. Yeung, Isabel Garcia-Perez, Joram M. Posma, Aleksandra Kopytek, Bethany Garratt, Hutan Ashrafian, Gary Frost, Julian R. Marchesi, Sanjay Purkayastha, Lesley Hoyles, Ara Darzi, Elaine Holmes. Multi-omic phenotyping reveals host-microbe responses to bariatric surgery, glycaemic control and obesity. Communications Medicine 2022, 2 (1) https://doi.org/10.1038/s43856-022-00185-6
    32. Benjamin H. Mullish, Laura Martinez‐Gili, Elena Chekmeneva, Gonçalo D. S. Correia, Matthew R. Lewis, Verena Horneffer‐Van Der Sluis, Lauren A. Roberts, Julie A. K. McDonald, Alexandros Pechlivanis, Julian R. F. Walters, Emma L. McClure, Julian R. Marchesi, Jessica R. Allegretti. Assessing the clinical value of faecal bile acid profiling to predict recurrence in primary Clostridioides difficile infection. Alimentary Pharmacology & Therapeutics 2022, 56 (11-12) , 1556-1569. https://doi.org/10.1111/apt.17247
    33. Zhipeng Wang, Hanglin Li, Yunlei Yun, Hongsen Wang, Bosu Meng, Yuhui Mu, Shouhong Gao, Xia Tao, Wansheng Chen. A dynamic multiple reaction monitoring strategy to develop and optimize targeted metabolomics methods: Analyzing bile acids in capecitabine-induced diarrhea. Journal of Pharmaceutical and Biomedical Analysis 2022, 219 , 114938. https://doi.org/10.1016/j.jpba.2022.114938
    34. Yan Cao, Wei Li, Xingcheng Gong, Xiaoya Niu, Jiao Zheng, Juan Yu, Jun Li, Pengfei Tu, Yuelin Song. Widely quasi-quantitative analysis enables temporal bile acids-targeted metabolomics in rat after oral administration of ursodeoxycholic acid. Analytica Chimica Acta 2022, 1212 , 339885. https://doi.org/10.1016/j.aca.2022.339885
    35. Pei-Chang Lee, Chi-Jung Wu, Ya-Wen Hung, Chieh Ju Lee, Chen-Ta Chi, I-Cheng Lee, Kuo Yu-Lun, Shih-Hsuan Chou, Jiing-Chyuan Luo, Ming-Chih Hou, Yi-Hsiang Huang. Gut microbiota and metabolites associate with outcomes of immune checkpoint inhibitor–treated unresectable hepatocellular carcinoma. Journal for ImmunoTherapy of Cancer 2022, 10 (6) , e004779. https://doi.org/10.1136/jitc-2022-004779
    36. Yu Wang, Haihong Hu, Jing Nie, Hui Zhou, Lushan Yu, Su Zeng. Inhibiting uptake activity of organic anion transporter 2 by bile acids. Drug Metabolism and Pharmacokinetics 2022, 43 , 100448. https://doi.org/10.1016/j.dmpk.2022.100448
    37. Xiang Zhao, Zitian Liu, Fuyun Sun, Lunjin Yao, Guangwei Yang, Kexin Wang. Bile Acid Detection Techniques and Bile Acid-Related Diseases. Frontiers in Physiology 2022, 13 https://doi.org/10.3389/fphys.2022.826740
    38. Matsepo Ramaboli, Lucky Nesengani, Leolin Katsidzira, Dirk Haller, James Kinross, Soeren Ocvirk, Stephen J.D. O’Keefe. Interactions between the environmental and human microbiota in the preservation of health and genesis of disease: symposium report. Current Opinion in Gastroenterology 2022, 38 (2) , 146-155. https://doi.org/10.1097/MOG.0000000000000817
    39. Yoshio Muguruma, Ryosuke Nagatomo, Shihori Kamatsuki, Katsuyuki Miyabe, Go Asano, Hiroyasu Akatsu, Koichi Inoue. Experimental design of a stable isotope labeling derivatized UHPLC–MS/MS method for the detection/quantification of primary/secondary bile acids in biofluids. Journal of Pharmaceutical and Biomedical Analysis 2022, 209 , 114485. https://doi.org/10.1016/j.jpba.2021.114485
    40. Zhipeng Wang, Hanglin Li, Yunlei Yun, Mengwei Zhang, Hongxia Yan, Dongyuan Lu, Yuehao Han, Hongsen Wang, Bosu Meng, Yuhui Mu, Shouhong Gao, Xia Tao, Wansheng Chen. A Dynamic Multiple Reaction Monitoring Strategy to Develop and Optimize Targeted Metabolomics Methods: An Example of Analyzing Bile Acids and its Application in Capecitabine-Induced Diarrhea. SSRN Electronic Journal 2022, 92 https://doi.org/10.2139/ssrn.4003377
    41. Tudor Mocan, Dong Wook Kang, Billy J. Molloy, Hyeonho Jeon, Zeno A. Spârchez, Diren Beyoğlu, Jeffrey R. Idle. Plasma fetal bile acids 7α-hydroxy-3-oxochol-4-en-24-oic acid and 3-oxachola-4,6-dien-24-oic acid indicate severity of liver cirrhosis. Scientific Reports 2021, 11 (1) https://doi.org/10.1038/s41598-021-87921-5
    42. Thomas S. Webberley, Giulia Masetti, Laura M. Baker, Jordanna Dally, Timothy R. Hughes, Julian R. Marchesi, Alison A. Jack, Sue F. Plummer, Guru Ramanathan, Paul D. Facey, Daryn R. Michael. The Impact of Lab4 Probiotic Supplementation in a 90-Day Study in Wistar Rats. Frontiers in Nutrition 2021, 8 https://doi.org/10.3389/fnut.2021.778289
    43. Katharina Habler, Bernhard Koeppl, Franz Bracher, Michael Vogeser. Targeted profiling of 24 sulfated and non-sulfated bile acids in urine using two-dimensional isotope dilution UHPLC-MS/MS. Clinical Chemistry and Laboratory Medicine (CCLM) 2021, Article ASAP.
    44. Ting Hu, Han Li, Benshan Xu, Ping Du, Lihong Liu, Zhuoling An. Parallel derivatization strategy for comprehensive profiling of unconjugated and glycine-conjugated bile acids using Ultra-high performance liquid chromatography-tandem mass spectrometry. The Journal of Steroid Biochemistry and Molecular Biology 2021, 214 , 105986. https://doi.org/10.1016/j.jsbmb.2021.105986
    45. Qi Yan Ang, Diana L Alba, Vaibhav Upadhyay, Jordan E Bisanz, Jingwei Cai, Ho Lim Lee, Eliseo Barajas, Grace Wei, Cecilia Noecker, Andrew D Patterson, Suneil K Koliwad, Peter J Turnbaugh. The East Asian gut microbiome is distinct from colocalized White subjects and connected to metabolic health. eLife 2021, 10 https://doi.org/10.7554/eLife.70349
    46. Lina Zhou, Di Yu, Sijia Zheng, Runze Ouyang, Yuting Wang, Guowang Xu. Gut microbiota-related metabolome analysis based on chromatography-mass spectrometry. TrAC Trends in Analytical Chemistry 2021, 143 , 116375. https://doi.org/10.1016/j.trac.2021.116375
    47. Jinxiu Lyu, Haijuan Li, Dengyang Yin, Meng Zhao, Qiang Sun, Mengzhe Guo. Analysis of eight bile acids in urine of gastric cancer patients based on covalent organic framework enrichment coupled with liquid chromatography-tandem mass spectrometry. Journal of Chromatography A 2021, 1653 , 462422. https://doi.org/10.1016/j.chroma.2021.462422
    48. Christoph G.W. Gertzen, Holger Gohlke, Dieter Häussinger, Diran Herebian, Verena Keitel, Ralf Kubitz, Ertan Mayatepek, Lutz Schmitt. The many facets of bile acids in the physiology and pathophysiology of the human liver. Biological Chemistry 2021, 402 (9) , 1047-1062. https://doi.org/10.1515/hsz-2021-0156
    49. Jessica R Allegretti, Colleen R Kelly, Ari Grinspan, Benjamin H Mullish, Jonathan Hurtado, Madeline Carrellas, Jenna Marcus, Julian R Marchesi, Julie A K McDonald, Ylaine Gerardin, Michael Silverstein, Alexandros Pechlivanis, Grace F Barker, Jesus Miguens Blanco, James L Alexander, Kate I Gallagher, Will Pettee, Emmalee Phelps, Sara Nemes, Sashidhar V Sagi, Matthew Bohm, Zain Kassam, Monika Fischer. Inflammatory Bowel Disease Outcomes Following Fecal Microbiota Transplantation for Recurrent C. difficile Infection. Inflammatory Bowel Diseases 2021, 27 (9) , 1371-1378. https://doi.org/10.1093/ibd/izaa283
    50. Kathryn R Koller, Annette Wilson, Daniel P Normolle, Jeremy K Nicholson, Jia V Li, James Kinross, Flora R Lee, Christie A Flanagan, Zoe T Merculieff, Priya Iyer, Daniela L Lammers, Timothy K Thomas, Stephen J D O'Keefe. Dietary fibre to reduce colon cancer risk in Alaska Native people: the Alaska FIRST randomised clinical trial protocol. BMJ Open 2021, 11 (8) , e047162. https://doi.org/10.1136/bmjopen-2020-047162
    51. Reiner Jumpertz von Schwartzenberg, Jordan E. Bisanz, Svetlana Lyalina, Peter Spanogiannopoulos, Qi Yan Ang, Jingwei Cai, Sophia Dickmann, Marie Friedrich, Su-Yang Liu, Stephanie L. Collins, Danielle Ingebrigtsen, Steve Miller, Jessie A. Turnbaugh, Andrew D. Patterson, Katherine S. Pollard, Knut Mai, Joachim Spranger, Peter J. Turnbaugh. Caloric restriction disrupts the microbiota and colonization resistance. Nature 2021, 595 (7866) , 272-277. https://doi.org/10.1038/s41586-021-03663-4
    52. Astia Rizki-Safitri, Fumiya Tokito, Masaki Nishikawa, Minoru Tanaka, Kazuya Maeda, Hiroyuki Kusuhara, Yasuyuki Sakai. Prospect of in vitro Bile Fluids Collection in Improving Cell-Based Assay of Liver Function. Frontiers in Toxicology 2021, 3 https://doi.org/10.3389/ftox.2021.657432
    53. Amy N. W. Schnelle, Luke T. Richardson, Michael E. Pettit, Sharon DeMorrow, Touradj Solouki. Trihydroxycholanoyl‐taurine in brains of rodents with hepatic encephalopathy. Journal of Mass Spectrometry 2021, 56 (6) https://doi.org/10.1002/jms.4729
    54. Yongjuan Jia, Xingli Liu, Li Xu, Junjun Ni, Liang Sun, Changkun Li. Rapid determination of 20 bile acids in human serum by ultra performance liquid chromatography–tandem mass spectrometry. Biomedical Chromatography 2021, 35 (5) https://doi.org/10.1002/bmc.5063
    55. Houguo Xu, Qingzhu Bi, Elena Pribytkova, Yuliang Wei, Bo Sun, Linlin Jia, Mengqing Liang. Different lipid scenarios in three lean marine teleosts having different lipid storage patterns. Aquaculture 2021, 536 , 736448. https://doi.org/10.1016/j.aquaculture.2021.736448
    56. Wiley Barton, Owen Cronin, Isabel Garcia‐Perez, Ronan Whiston, Elaine Holmes, Trevor Woods, Catherine B. Molloy, Michael G. Molloy, Fergus Shanahan, Paul D. Cotter, Orla O’Sullivan. The effects of sustained fitness improvement on the gut microbiome: A longitudinal, repeated measures case‐study approach. Translational Sports Medicine 2021, 4 (2) , 174-192. https://doi.org/10.1002/tsm2.215
    57. Rachel Y. Gao, Colin T. Shearn, David J. Orlicky, Kayla D. Battista, Erica E. Alexeev, Ian M. Cartwright, Jordi M. Lanis, Rachael E. Kostelecky, Cynthia Ju, Sean P. Colgan, Blair P. Fennimore. Bile acids modulate colonic MAdCAM-1 expression in a murine model of combined cholestasis and colitis. Mucosal Immunology 2021, 14 (2) , 479-490. https://doi.org/10.1038/s41385-020-00347-6
    58. Jenessa A. Winston, Alissa Rivera, Jingwei Cai, Andrew D. Patterson, Casey M. Theriot, . Secondary bile acid ursodeoxycholic acid alters weight, the gut microbiota, and the bile acid pool in conventional mice. PLOS ONE 2021, 16 (2) , e0246161. https://doi.org/10.1371/journal.pone.0246161
    59. Marine P.M. Letertre, Antonis Myridakis, Luke Whiley, Stéphane Camuzeaux, Matthew R. Lewis, Katie E. Chappell, Annie Thaikkatil, Marc-Emmanuel Dumas, Jeremy K. Nicholson, Jonathan R. Swann, Ian D. Wilson. A targeted ultra performance liquid chromatography – Tandem mass spectrometric assay for tyrosine and metabolites in urine and plasma: Application to the effects of antibiotics on mice. Journal of Chromatography B 2021, 1164 , 122511. https://doi.org/10.1016/j.jchromb.2020.122511
    60. Gaëlle Magliocco, Jules Desmeules, Marija Bosilkovska, Aurélien Thomas, Youssef Daali. The 1β-Hydroxy-Deoxycholic Acid to Deoxycholic Acid Urinary Metabolic Ratio: Toward a Phenotyping of CYP3A Using an Endogenous Marker?. Journal of Personalized Medicine 2021, 11 (2) , 150. https://doi.org/10.3390/jpm11020150
    61. Georgios Theodoridis, Alexandros Pechlivanis, Nikolaos Thomaidis, Apostolos Spyros, Constantinos Georgiou, Triantafyllos Albanis, Ioannis Skoufos, Stavros Kalogiannis, George Tsangaris, Athanasios Stasinakis, Ioannis Konstantinou, Alexander Triantafyllidis, Konstantinos Gkagkavouzis, Anastasia Kritikou, Marilena Dasenaki, Helen Gika, Christina Virgiliou, Dritan Kodra, Nikolaos Nenadis, Ioannis Sampsonidis, Georgios Arsenos, Maria Halabalaki, Emmanuel Mikros, . FoodOmicsGR_RI: A Consortium for Comprehensive Molecular Characterisation of Food Products. Metabolites 2021, 11 (2) , 74. https://doi.org/10.3390/metabo11020074
    62. Anqi Zhao, Liyun Zhang, Xuhuiqun Zhang, Indika Edirisinghe, Britt M. Burton-Freeman, Amandeep K. Sandhu. Comprehensive Characterization of Bile Acids in Human Biological Samples and Effect of 4-Week Strawberry Intake on Bile Acid Composition in Human Plasma. Metabolites 2021, 11 (2) , 99. https://doi.org/10.3390/metabo11020099
    63. Věra Dosedělová, Petra Itterheimová, Petr Kubáň. Analysis of bile acids in human biological samples by microcolumn separation techniques: A review. ELECTROPHORESIS 2021, 42 (1-2) , 68-85. https://doi.org/10.1002/elps.202000139
    64. Yuan Tian, Jingwei Cai, Erik L. Allman, Philip B. Smith, Andrew D. Patterson. Quantitative Analysis of Bile Acid with UHPLC-MS/MS. 2021, 291-300. https://doi.org/10.1007/978-1-0716-0849-4_15
    65. John A. Masucci, Feng Liang, Kerem Bingol, Vince Windisch, Gary W. Caldwell. Simultaneously Assessing Concentration Changes in 17 Biochemical Pathways as a Result of Drug Dosing and Cytochrome P450 and Non-cytochrome P450-Mediated Metabolism: A Quasi-Untargeted Metabolomics LC/MS Assay. 2021, 341-357. https://doi.org/10.1007/978-1-0716-1542-3_21
    66. Sylvère Durand, Claudia Grajeda-Iglesias, Fanny Aprahamian, Nitharsshini Nirmalathasan, Oliver Kepp, Guido Kroemer. The intracellular metabolome of starving cells. 2021, 137-156. https://doi.org/10.1016/bs.mcb.2021.04.001
    67. Florian Seyfried, Jutarop Phetcharaburanin, Maria Glymenaki, Arno Nordbeck, Mohammed Hankir, Jeremy K Nicholson, Elaine Holmes, Julian R. Marchesi, Jia V. Li. Roux-en-Y gastric bypass surgery in Zucker rats induces bacterial and systemic metabolic changes independent of caloric restriction-induced weight loss. Gut Microbes 2021, 13 (1) https://doi.org/10.1080/19490976.2021.1875108
    68. Miao Lin, Xiong Chen, Zhe Wang, Dongmei Wang, Jin-Lan Zhang. Global profiling and identification of bile acids by multi-dimensional data mining to reveal a way of eliminating abnormal bile acids. Analytica Chimica Acta 2020, 1132 , 74-82. https://doi.org/10.1016/j.aca.2020.07.067
    69. N S Ding, J A K McDonald, A Perdones-Montero, Douglas N Rees, S O Adegbola, R Misra, P Hendy, L Penez, J R Marchesi, E Holmes, M H Sarafian, A L Hart. Metabonomics and the Gut Microbiome Associated With Primary Response to Anti-TNF Therapy in Crohn’s Disease. Journal of Crohn's and Colitis 2020, 14 (8) , 1090-1102. https://doi.org/10.1093/ecco-jcc/jjaa039
    70. Jose Rodríguez-Morató, Nirupa R. Matthan. Nutrition and Gastrointestinal Microbiota, Microbial-Derived Secondary Bile Acids, and Cardiovascular Disease. Current Atherosclerosis Reports 2020, 22 (9) https://doi.org/10.1007/s11883-020-00863-7
    71. Ruben A.T. Mars, Yi Yang, Tonya Ward, Mo Houtti, Sambhawa Priya, Heather R. Lekatz, Xiaojia Tang, Zhifu Sun, Krishna R. Kalari, Tal Korem, Yogesh Bhattarai, Tenghao Zheng, Noam Bar, Gary Frost, Abigail J. Johnson, Will van Treuren, Shuo Han, Tamas Ordog, Madhusudan Grover, Justin Sonnenburg, Mauro D’Amato, Michael Camilleri, Eran Elinav, Eran Segal, Ran Blekhman, Gianrico Farrugia, Jonathan R. Swann, Dan Knights, Purna C. Kashyap. Longitudinal Multi-omics Reveals Subset-Specific Mechanisms Underlying Irritable Bowel Syndrome. Cell 2020, 182 (6) , 1460-1473.e17. https://doi.org/10.1016/j.cell.2020.08.007
    72. Jan Klouda, Karel Nesměrák, Pavel Kočovský, Jiří Barek, Karolina Schwarzová-Pecková. A novel voltammetric approach to the detection of primary bile acids in serum samples. Bioelectrochemistry 2020, 134 , 107539. https://doi.org/10.1016/j.bioelechem.2020.107539
    73. Ming Li, Qian Wang, Yong Li, Shengtian Cao, Yingjun Zhang, Zhongqing Wang, Guozhu Liu, Jing Li, Baohua Gu. Apical sodium-dependent bile acid transporter, drug target for bile acid related diseases and delivery target for prodrugs: Current and future challenges. Pharmacology & Therapeutics 2020, 212 , 107539. https://doi.org/10.1016/j.pharmthera.2020.107539
    74. Joselyn N. Allen, Adwitia Dey, Jingwei Cai, Jingtao Zhang, Yuan Tian, Mary Kennett, Yanling Ma, T. Jake Liang, Andrew D. Patterson, Pamela A. Hankey-Giblin. Metabolic Profiling Reveals Aggravated Non-Alcoholic Steatohepatitis in High-Fat High-Cholesterol Diet-Fed Apolipoprotein E-Deficient Mice Lacking Ron Receptor Signaling. Metabolites 2020, 10 (8) , 326. https://doi.org/10.3390/metabo10080326
    75. Yuan Tian, Wei Gui, Imhoi Koo, Philip B. Smith, Erik L. Allman, Robert G. Nichols, Bipin Rimal, Jingwei Cai, Qing Liu, Andrew D. Patterson. The microbiome modulating activity of bile acids. Gut Microbes 2020, 11 (4) , 979-996. https://doi.org/10.1080/19490976.2020.1732268
    76. Nicolas Christinat, Armand Valsesia, Mojgan Masoodi. Untargeted Profiling of Bile Acids and Lysophospholipids Identifies the Lipid Signature Associated with Glycemic Outcome in an Obese Non-Diabetic Clinical Cohort. Biomolecules 2020, 10 (7) , 1049. https://doi.org/10.3390/biom10071049
    77. Elisa Danese, Davide Negrini, Mairi Pucci, Simone De Nitto, Davide Ambrogi, Simone Donzelli, Patricia M.-J. Lievens, Gian Luca Salvagno, Giuseppe Lippi. Bile Acids Quantification by Liquid Chromatography–Tandem Mass Spectrometry: Method Validation, Reference Range, and Interference Study. Diagnostics 2020, 10 (7) , 462. https://doi.org/10.3390/diagnostics10070462
    78. Cristina Gómez, Simon Stücheli, Denise V. Kratschmar, Jamal Bouitbir, Alex Odermatt. Development and Validation of a Highly Sensitive LC-MS/MS Method for the Analysis of Bile Acids in Serum, Plasma, and Liver Tissue Samples. Metabolites 2020, 10 (7) , 282. https://doi.org/10.3390/metabo10070282
    79. Noémie Péan, Aurelie Le Lay, Francois Brial, Jessica Wasserscheid, Claude Rouch, Mylène Vincent, Antonis Myridakis, Lyamine Hedjazi, Marc-Emmanuel Dumas, Elin Grundberg, Mark Lathrop, Christophe Magnan, Ken Dewar, Dominique Gauguier. Dominant gut Prevotella copri in gastrectomised non-obese diabetic Goto–Kakizaki rats improves glucose homeostasis through enhanced FXR signalling. Diabetologia 2020, 63 (6) , 1223-1235. https://doi.org/10.1007/s00125-020-05122-7
    80. Qi Yan Ang, Margaret Alexander, John C. Newman, Yuan Tian, Jingwei Cai, Vaibhav Upadhyay, Jessie A. Turnbaugh, Eric Verdin, Kevin D. Hall, Rudolph L. Leibel, Eric Ravussin, Michael Rosenbaum, Andrew D. Patterson, Peter J. Turnbaugh. Ketogenic Diets Alter the Gut Microbiome Resulting in Decreased Intestinal Th17 Cells. Cell 2020, 181 (6) , 1263-1275.e16. https://doi.org/10.1016/j.cell.2020.04.027
    81. Sandi L. Navarro, Lisa Levy, Keith R. Curtis, Isaac Elkon, Orsalem J. Kahsai, Hamza S. Ammar, Timothy W. Randolph, Natalie N. Hong, Fausto Carnevale Neto, Daniel Raftery, Robert S. Chapkin, Johanna W. Lampe, Meredith A. J. Hullar. Effect of a Flaxseed Lignan Intervention on Circulating Bile Acids in a Placebo-Controlled Randomized, Crossover Trial. Nutrients 2020, 12 (6) , 1837. https://doi.org/10.3390/nu12061837
    82. Jenessa A. Winston, Alissa J. Rivera, Jingwei Cai, Rajani Thanissery, Stephanie A. Montgomery, Andrew D. Patterson, Casey M. Theriot, . Ursodeoxycholic Acid (UDCA) Mitigates the Host Inflammatory Response during Clostridioides difficile Infection by Altering Gut Bile Acids. Infection and Immunity 2020, 88 (6) https://doi.org/10.1128/IAI.00045-20
    83. Xia Gong, Qisong Zhang, Yanjiao Ruan, Ming Hu, Zhongqiu Liu, Lingzhi Gong. Chronic Alcohol Consumption Increased Bile Acid Levels in Enterohepatic Circulation and Reduced Efficacy of Irinotecan. Alcohol and Alcoholism 2020, 55 (3) , 264-277. https://doi.org/10.1093/alcalc/agaa005
    84. Jessica R. Allegretti, Zain Kassam, Benjamin H. Mullish, Austin Chiang, Madeline Carrellas, Jonathan Hurtado, Julian R. Marchesi, Julie A.K. McDonald, Alexandros Pechlivanis, Grace F. Barker, Jesús Miguéns Blanco, Isabel Garcia-Perez, Wing Fei Wong, Ylaine Gerardin, Michael Silverstein, Kevin Kennedy, Christopher Thompson. Effects of Fecal Microbiota Transplantation With Oral Capsules in Obese Patients. Clinical Gastroenterology and Hepatology 2020, 18 (4) , 855-863.e2. https://doi.org/10.1016/j.cgh.2019.07.006
    85. Jordi Mayneris‐Perxachs, José‐Manuel Fernández‐Real. Exploration of the microbiota and metabolites within body fluids could pinpoint novel disease mechanisms. The FEBS Journal 2020, 287 (5) , 856-865. https://doi.org/10.1111/febs.15130
    86. Ibrahim Choucair, Ina Nemet, Lin Li, Margaret A. Cole, Sarah M. Skye, Jennifer D. Kirsop, Michael A. Fischbach, Valentin Gogonea, J. Mark Brown, W. H. Wilson Tang, Stanley L. Hazen. Quantification of bile acids: a mass spectrometry platform for studying gut microbe connection to metabolic diseases. Journal of Lipid Research 2020, 61 (2) , 159-177. https://doi.org/10.1194/jlr.RA119000311
    87. Ting Hu, Zhuoling An, Chen Shi, Pengfei Li, Lihong Liu. A sensitive and efficient method for simultaneous profiling of bile acids and fatty acids by UPLC-MS/MS. Journal of Pharmaceutical and Biomedical Analysis 2020, 178 , 112815. https://doi.org/10.1016/j.jpba.2019.112815
    88. Xian Cheng, Jian-Liang Zhou, Zi-Qi Shi, Ying-Hao Yin, Jian-Qun Liu, Han-Xiang Chen, Ping Li, Li-Fang Liu, Gui-Zhong Xin. Ammonium fluoride-induced stabilization for anion attachment mass spectrometry: Facilitating the pseudotargeted profiling of bile acids submetabolome. Analytica Chimica Acta 2019, 1081 , 120-130. https://doi.org/10.1016/j.aca.2019.07.006
    89. Benjamin H Mullish, Julie A K McDonald, Alexandros Pechlivanis, Jessica R Allegretti, Dina Kao, Grace F Barker, Diya Kapila, Elaine O Petrof, Susan A Joyce, Cormac G M Gahan, Izabela Glegola-Madejska, Horace R T Williams, Elaine Holmes, Thomas B Clarke, Mark R Thursz, Julian R Marchesi. Microbial bile salt hydrolases mediate the efficacy of faecal microbiota transplant in the treatment of recurrent Clostridioides difficile infection. Gut 2019, 68 (10) , 1791-1800. https://doi.org/10.1136/gutjnl-2018-317842
    90. Wenjing Liu, Qingqing Song, Yan Cao, Yanan Zhao, Huixia Huo, Yitao Wang, Yuelin Song, Jun Li, Pengfei Tu. Advanced liquid chromatography-mass spectrometry enables merging widely targeted metabolomics and proteomics. Analytica Chimica Acta 2019, 1069 , 89-97. https://doi.org/10.1016/j.aca.2019.04.013
    91. Anisha Wijeyesekera, Josef Wagner, Marcus De Goffau, Sarah Thurston, Adilson Rodrigues Sabino, Sara Zaher, Deborah White, Jenna Ridout, Mark J. Peters, Padmanabhan Ramnarayan, Ricardo G. Branco, M. Estee Torok, Frederic Valla, Rosan Meyer, Nigel Klein, Gary Frost, Julian Parkhill, Elaine Holmes, Nazima Pathan. Multi-Compartment Profiling of Bacterial and Host Metabolites Identifies Intestinal Dysbiosis and Its Functional Consequences in the Critically Ill Child. Critical Care Medicine 2019, 47 (9) , e727-e734. https://doi.org/10.1097/CCM.0000000000003841
    92. Xueyun Zheng, Francesca B. Smith, Noor A. Aly, Jingwei Cai, Richard D. Smith, Andrew D. Patterson, Erin S. Baker. Evaluating the structural complexity of isomeric bile acids with ion mobility spectrometry. Analytical and Bioanalytical Chemistry 2019, 411 (19) , 4673-4682. https://doi.org/10.1007/s00216-019-01869-0
    93. Mainak Dutta, Jingwei Cai, Wei Gui, Andrew D. Patterson. A review of analytical platforms for accurate bile acid measurement. Analytical and Bioanalytical Chemistry 2019, 411 (19) , 4541-4549. https://doi.org/10.1007/s00216-019-01890-3
    94. Samuel Mach, Alexandr Jegorov, Zdeněk Šimek. Metabolism of obeticholic acid in brown bullhead (Ameiurus nebulosus). Environmental Science and Pollution Research 2019, 26 (20) , 20316-20324. https://doi.org/10.1007/s11356-019-05398-2
    95. Jessica R. Allegretti, Zain Kassam, Madeline Carrellas, Benjamin H. Mullish, Julian R. Marchesi, Alexandros Pechlivanis, Mark Smith, Ylaine Gerardin, Sonia Timberlake, Daniel S. Pratt, Joshua R. Korzenik. Fecal Microbiota Transplantation in Patients With Primary Sclerosing Cholangitis: A Pilot Clinical Trial. American Journal of Gastroenterology 2019, 114 (7) , 1071-1079. https://doi.org/10.14309/ajg.0000000000000115
    96. Vinod S. Hegade, Alexandros Pechlivanis, Julie A. K. McDonald, Douglas Rees, Margaret Corrigan, Gideon M. Hirschfield, Simon D. Taylor‐Robinson, Elaine Holmes, Julian R. Marchesi, Stuart Kendrick, David E. Jones. Autotaxin, bile acid profile and effect of ileal bile acid transporter inhibition in primary biliary cholangitis patients with pruritus. Liver International 2019, 39 (5) , 967-975. https://doi.org/10.1111/liv.14069
    97. Emma Rose McGlone, Stephen R Bloom. Bile acids and the metabolic syndrome. Annals of Clinical Biochemistry: International Journal of Laboratory Medicine 2019, 56 (3) , 326-337. https://doi.org/10.1177/0004563218817798
    98. Jordi Mayneris-Perxachs, Jonathan R. Swann. Metabolic phenotyping of malnutrition during the first 1000 days of life. European Journal of Nutrition 2019, 58 (3) , 909-930. https://doi.org/10.1007/s00394-018-1679-0
    99. Jing Zhao, Qing-li Zhang, Jian-hua Shen, Kai Wang, Jia Liu. Magnesium lithospermate B improves the gut microbiome and bile acid metabolic profiles in a mouse model of diabetic nephropathy. Acta Pharmacologica Sinica 2019, 40 (4) , 507-513. https://doi.org/10.1038/s41401-018-0029-3
    100. Tanya Monaghan, Benjamin H Mullish, Jordan Patterson, Gane KS Wong, Julian R Marchesi, Huiping Xu, Tahseen Jilani, Dina Kao. Effective fecal microbiota transplantation for recurrent Clostridioides difficile infection in humans is associated with increased signalling in the bile acid-farnesoid X receptor-fibroblast growth factor pathway. Gut Microbes 2019, 10 (2) , 142-148. https://doi.org/10.1080/19490976.2018.1506667
    Load all citations

    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