Paroxetine Administration Affects Microbiota and Bile Acid Levels in Mice

Frederik Dethloff, Fernando Vargas, Emmanuel Elijah, Robert Quinn, Dong Ik Park, David P Herzog, Marianne B Müller, Emily C Gentry, Rob Knight, Antonio Gonzalez, Pieter C Dorrestein, Christoph W Turck, Frederik Dethloff, Fernando Vargas, Emmanuel Elijah, Robert Quinn, Dong Ik Park, David P Herzog, Marianne B Müller, Emily C Gentry, Rob Knight, Antonio Gonzalez, Pieter C Dorrestein, Christoph W Turck

Abstract

Recent interest in the role of microbiota in health and disease has implicated gut microbiota dysbiosis in psychiatric disorders including major depressive disorder. Several antidepressant drugs that belong to the class of selective serotonin reuptake inhibitors have been found to display antimicrobial activities. In fact, one of the first antidepressants discovered serendipitously in the 1950s, the monoamine-oxidase inhibitor Iproniazid, was a drug used for the treatment of tuberculosis. In the current study we chronically treated DBA/2J mice for 2 weeks with paroxetine, a selective serotonin reuptake inhibitor, and collected fecal pellets as a proxy for the gut microbiota from the animals after 7 and 14 days. Behavioral testing with the forced swim test revealed significant differences between paroxetine- and vehicle-treated mice. Untargeted mass spectrometry and 16S rRNA profiling of fecal pellet extracts showed several primary and secondary bile acid level, and microbiota alpha diversity differences, respectively between paroxetine- and vehicle-treated mice, suggesting that microbiota functions are altered by the drug. In addition to their lipid absorbing activities bile acids have important signaling activities and have been associated with gastrointestinal diseases and colorectal cancer. Antidepressant drugs like paroxetine should therefore be used with caution to prevent undesirable side effects.

Keywords: antidepressant; bile acids; metabolomics; microbiome; paroxetine.

Copyright © 2020 Dethloff, Vargas, Elijah, Quinn, Park, Herzog, Müller, Gentry, Knight, Gonzalez, Dorrestein and Turck.

Figures

Figure 1
Figure 1
Paroxetine treatment revealed antidepressant-like and anxiolytic effects in DBA/2J mice. Chronic paroxetine (PARO) treatment for 14 days increased time of active coping (p = 1.5 E-7) in the Forced Swim Test (FST). Sample size was n = 10 for PARO group and n = 10 for Vehicle group. Bars represent mean ± SD, ***p

Figure 2

Fecal pellet alpha diversity is…

Figure 2

Fecal pellet alpha diversity is significantly different (p-value = 0.011402, q-value = 0.034207)…

Figure 2
Fecal pellet alpha diversity is significantly different (p-value = 0.011402, q-value = 0.034207) in paroxetine- compared to vehicle-treated mice. Alpha-diversity was compared between treatment and control groups using Faith's Phylogenetic Diversity (PMID: 19455206). Sample sizes were n=8 for Paroxetine 1 week group, n = 12 for Paroxetine 2 week group and n = 20 for Vehicle group.

Figure 3

(A) Fecal pellet bile acid…

Figure 3

(A) Fecal pellet bile acid level ratios following 1 week (1w) and (B)…
Figure 3
(A) Fecal pellet bile acid level ratios following 1 week (1w) and (B) 2 weeks of paroxetine (PARO) treatment, sample size n = 10 each. Bars represent mean log2 fold changes PARO *Vehicle-1. Significance was tested using Student's t-test; significance is indicated (p < 0.05 = *; p < 0.01= **). Several fecal pellet bile acid levels are increased following PARO treatment. (C) Several fecal bile acid levels correlate significantly with behavior, body weight gain, PARO metabolites, and other bile acid levels (indicated with white asterisk, p < 0.1). Correlogram displays the Pearson correlation coefficient values of treatment with PARO. Lower left represents the correlations after 1 week (1w) of treatment und upper right the correlations after 2 weeks (2w) of treatment. Size and color intensity reflect the absolute value as indicated by the color bar. Abbreviations in order of appearance: BWgain, body weight gain; FST, forced swim test; paroxetine, PARO; paroxetine metabolite I, PARO M I; paroxetine metabolite III, PARO M III; F-DCA, phenylalanodeoxycholic acid; F-CA, phenylalanocholic acid; GLCA, glycolithocholic acid; GDCA, glycodeoxycholic acid; DCA, deoxycholic acid.
Figure 2
Figure 2
Fecal pellet alpha diversity is significantly different (p-value = 0.011402, q-value = 0.034207) in paroxetine- compared to vehicle-treated mice. Alpha-diversity was compared between treatment and control groups using Faith's Phylogenetic Diversity (PMID: 19455206). Sample sizes were n=8 for Paroxetine 1 week group, n = 12 for Paroxetine 2 week group and n = 20 for Vehicle group.
Figure 3
Figure 3
(A) Fecal pellet bile acid level ratios following 1 week (1w) and (B) 2 weeks of paroxetine (PARO) treatment, sample size n = 10 each. Bars represent mean log2 fold changes PARO *Vehicle-1. Significance was tested using Student's t-test; significance is indicated (p < 0.05 = *; p < 0.01= **). Several fecal pellet bile acid levels are increased following PARO treatment. (C) Several fecal bile acid levels correlate significantly with behavior, body weight gain, PARO metabolites, and other bile acid levels (indicated with white asterisk, p < 0.1). Correlogram displays the Pearson correlation coefficient values of treatment with PARO. Lower left represents the correlations after 1 week (1w) of treatment und upper right the correlations after 2 weeks (2w) of treatment. Size and color intensity reflect the absolute value as indicated by the color bar. Abbreviations in order of appearance: BWgain, body weight gain; FST, forced swim test; paroxetine, PARO; paroxetine metabolite I, PARO M I; paroxetine metabolite III, PARO M III; F-DCA, phenylalanodeoxycholic acid; F-CA, phenylalanocholic acid; GLCA, glycolithocholic acid; GDCA, glycodeoxycholic acid; DCA, deoxycholic acid.

References

    1. Fredman SJ, Fava M, Kienke AS, White CN, Nierenberg AA, Rosenbaum JF. Partial response, nonresponse, and relapse with selective serotonin reuptake inhibitors in major depression: a survey of current next-step practices. J Clin Psychiatry (2000) 61:403408. 10.4088/JCP.v61n0602
    1. Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry (2006) 163(1):28–40. 10.1176/appi.ajp.163.1.28
    1. Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun (2015) 48:186194. 10.1016/j.bbi.2015.03.016
    1. Wong ML, Inserra A, Lewis MD, Mastronardi CA, Leong L, Choo J, et al. Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition. Mol Psychiatry (2016) 21:797805. 10.1038/mp.2016.46
    1. Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the hosts metabolism. Mol Psychiatry (2016) 21:786796. 10.1038/mp.2016.44
    1. Dinan TG, Cryan JF. Mood by microbe: towards clinical translation. Genome Med (2016) 6:36. 10.1186/s13073-016-0292-1
    1. Yang C, Fujita Y, Ren Q, Ma M, Dong C, Hashimoto K. Bifidobacterium in the gut microbiota confer resilience to chronic social defeat stress in mice. Sci Rep (2017) 7:45942. 10.1038/srep45942
    1. Ho P, Ross DA. More than a gut feeling: the implications of the gut microbiota in psychiatry. Biol Psychiatry (2017) 81:e35e37. 10.1016/j.biopsych.2016.12.018
    1. Sharon G, Cruz NJ, Kang DW, Gandal MJ, Wang B, Kim YM, et al. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell (2019) 177(6):1600–1618.e17. 10.1016/j.cell.2019.05.004
    1. Macedo D, Filho AJMC, Soares de Sousa CN, Quevedo J, Barichello T, Júnior HVN, et al. Antidepressants, antimicrobials or both? Gut microbiota dysbiosis in depression and possible implications of the antimicrobial effects of antidepressant drugs for antidepressant effectiveness. J Affect Disord (2017) 208:2232. 10.1016/j.jad.2016.09.012
    1. Philpott HL, Nandurkar S, Lubel J, Gibson PR. Drug-induced gastrointestinal disorders. Frontline Gastroenterol (2014) 5(1):4957. 10.1136/postgradmedj-2013-100316rep
    1. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol (2014) 30:332–8. 10.1097/MOG.0000000000000057
    1. Cussotto S, Clarke G, Dinan TG, Cryan J. Psychotropics and the Microbiome: a Chamber of Secrets. Psychopharmacology (2019) 236:1411–32. 10.1007/s00213-019-5185-8
    1. Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. Bile acids as regulatory molecules. J Lipid Res (2009) 50:1509–20. 10.1194/jlr.R900007-JLR200
    1. Allen K, Jaeschke H, Copple BL. Bile acids induce inflammatory genes in hepatocytes: a novel mechanism of inflammation during obstructive cholestasis. Am J Pathol (2011) 178(1):17586. 10.1016/j.ajpath.2010.11.026
    1. Nguyen A, Bouscarel B. Bile acids and signal transduction: role in glucose homeostasis. Cell Signal (2008) 20(12):218097. 10.1016/j.cellsig.2008.06.014. mediated by FXR, pregnane X receptor, vitamin D receptor and TGR5 receptor.
    1. Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile-acid signalling for metabolic diseases. Nat Rev Drug Discov (2008) 7(8):67893. 10.1038/nrd2619
    1. Carrillo-Roa T, Labermaier C, Weber P, Herzog DP, Lareau C, Santarelli S, et al. Common genes associated with antidepressant response in mouse and man identify key role of glucocorticoid receptor sensitivity. PloS Biol (2017) 15(12):e2002690. 10.1371/journal.pbio.2002690
    1. Gonzalez A, Navas-Molina JA, Kosciolek T, McDonald D, Vazquez-Baeza Y, Ackermann G, et al. Qiita: rapid, web-enabled microbiome meta-analysis. Nat Methods (2018) 15:796–8. 10.1038/s41592-018-0141-9
    1. Lozupone C, Knight R. UniFrac: a New Phylogenetic Method for Comparing Microbial Communities. Appl Env Microbiol (2005) 71:8228–35. 10.1128/AEM.71.12.8228-8235.2005
    1. Chambers MC, MacLean B, Burke R, Amode D, Ruderman DL, Neumann S. A cross-platform toolkit for mass spectrometry and proteomics. Nat Biotechnol (2012) 30:918–20. 10.1038/nbt.2377
    1. Myers OD, Sumner SJ, Li S, Barnes S, Du X. One Step Forward for Reducing False Positive and False Negative Compound Identifications from Mass Spectrometry Metabolomics Data: New Algorithms for Constructing Extracted Ion Chromatograms and Detecting Chromatographic Peaks. Anal Chem (2017) 89(17):8696–703. 10.1021/acs.analchem.7b00947
    1. Pluskal T, Castillo S, Villar-Briones A, Orešič M. MZmine 2: Modular framework for processing, visualizing, and analyzing mass spectrometry-based molecular profile data. BMC Bioinf (2010) 11:395. 10.1186/1471-2105-11-395
    1. Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods (2016) 13(9):731–40. 10.1038/nmeth.3901
    1. Sillaber I, Panhuysen M, Henniger MSH, Ohl F, Kuhne C, Pütz B, et al. Profiling of behavioral changes and hippocampal gene expression in mice chronically treated with the SSRI paroxetine. Psychopharmacol (Berl.) (2008) 200:557572. 10.1007/s00213-008-1232-6
    1. Webhofer C, Gormanns P, Tolstikov V, Zieglgansberger W, Sillaber I, Holsboer F, et al. Metabolite profiling of antidepressant drug action reveals novel drug targets beyond monoamine elevation. Transl Psychiatry (2011) 1:e58. 10.1038/tp.2011.56
    1. Webhofer C, Gormanns P, Reckow S, Lebar M, Maccarrone G, Ludwig T, et al. Proteomic and metabolomic profiling reveals time-dependent changes in hippocampal metabolism upon paroxetine treatment and biomarker candidates. J Psychiatr Res (2013) 47:289298. 10.1016/j.jpsychires.2012.11.003
    1. Park DI, Dournes C, Sillaber I, Uhr M, Asara JM, Gassen NC, et al. Purine and pyrimidine metabolism: Convergent evidence on chronic antidepressant treatment response in mice and humans. Sci Rep (2016) 6:35317. 10.1038/srep35317
    1. Park DI, Dournes C, Sillaber I, Ising M, Asara JM, Webhofer C, et al. Delineation of molecular pathway activities of the chronic antidepressant treatment response suggests important roles for glutamatergic and ubiquitin-proteasome systems. Trans Psychiatry (2017) 7:e1078. 10.1038/tp.2017.39
    1. Alnouti Y. Bile acid sulfation: a pathway of bile acid elimination and detoxification. Toxicol Sci (2009) 108:225–46. 10.1093/toxsci/kfn268
    1. Hagey LR, Krasowski MD. Microbial biotransformations of bile acids as detected by electrospray mass spectrometry. Adv Nutr (2013) 4:29–35. 10.3945/an.112.003061
    1. Quinn RA, Vrbanac A, Melnik A, Patras KA, Christy M, Nelson AT, et al. Chemical Impacts of the Microbiome Across Scales Reveal Novel Conjugated Bile Acids. Nature (2020) 579:123–9. 10.1038/s41586-020-2047-9
    1. Behr C, Slopianka M, Haake V, Strauss V, Sperber S, Kamp H, et al. Analysis of metabolome changes in the bile acid pool in feces and plasma of antibiotic-treated rats. Toxicol Appl Pharmacol (2019) 363:7987. 10.1016/j.taap.2018.11.012
    1. Munoz-Bellido J, Munoz-Criado S, Garcia-Rodriguez J. Antimicrobial activity of psychotropic drugs. Int J Antimicrob Agents (2000) 14:177180. 10.1016/S0924-8579(99)00154-5
    1. Dior M, Delagreverie H, Duboc H, Jouet P, Coffin B, Brot L, et al. Interplay between bile acid metabolism and microbiota in irritable bowel syndrome. Neurogastroenterol Motil (2016) 28:1330–40. 10.1111/nmo.12829
    1. Duboc H, Rajca S, Rainteau D, Benarous D, Maubert MA, Quervain E, et al. Connecting dysbiosis, bile-acid dysmetabolism and Gut inflammation in inflammatory bowel diseases. Gut (2013) 62:531–9. 10.1136/gutjnl-2012-302578
    1. Pereira-Fantini M, Bines JE, Lapthorne S, Fouhy F, Scurr M, Cotter PD, et al. Short bowel syndrome (SBS)-associated alterations within the gut-liver axis evolve early and persist long-term in the piglet model of short bowel syndrome. J Gastroenterol Hepatol (2016) 31:1946–55. 10.1111/jgh.13383
    1. Theriot CM, Bowman A, Young VB. Antibiotic-Induced Alterations of the Gut Microbiota Alter Secondary Bile Acid Production and Allow for Clostridium difficile Spore Germination and Outgrowth in the Large Intestine. mSphere (2015) 1:e00045–15. 10.1128/mSphere.00045-15
    1. Anakk S, Watanabe M, Ochsner SA, McKenna NJ, Finegold MJ, Moore DD. Combined deletion of Fxr and Shp in mice induces Cyp17a1 and results in juvenile onset cholestasis. J Clin Invest (2011) 121:8695. 10.1172/JCI42846
    1. Fu T, Coulter S, Yoshihara E, Oh TG, Fang S, Cayabyab F. FXR Regulates Intestinal Cancer Stem Cell Proliferation. Cell (2019) 176:1098–112. 10.1016/j.cell.2019.01.036. 10981112.
    1. de Aguiar Vallim TQ, Tarling EJ, Edwards PA. Pleiotropic roles of bile acids in metabolism. Cell Metab (2013) 17:657–69. 10.1016/j.cmet.2013.03.013
    1. Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem (2003) 72:137–74. 10.1146/annurev.biochem.72.121801.161712
    1. Kitahara M, Takamine F, Imamura T, Benno Y. Assignment of Eubacterium sp. VPI 12708 and related strains with high bile acid 7alphadehydroxylating activity to Clostridium scindens and proposal of Clostridium hylemonae sp. nov., isolated from human faeces. Int J Syst Evol Microbiol (2000) 50:971978. 10.1099/00207713-50-3-971
    1. Kitahara M, Takamine F, Imamura T, Benno Y. Clostridium hiranonis sp. nov., a human intestinal bacterium with bile acid 7alpha-dehydroxylating activity. Int J Syst Evol Microbiol (2001) 51:3944. 10.1099/00207713-51-1-39
    1. Ridlon JM, Kang DJ, Hylemon PB. The human gut sterolbiome: bile acid-microbiome endocrine aspects and therapeutics. Acta Pharm Sin B (2006) 5:99105. 10.1016/j.apsb.2015.01.006
    1. Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol (2012) 10:735–42. 10.1038/nrmicro2876
    1. Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci (2012) 13:701–12. 10.1038/nrn3346
    1. Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K. Gut microbes and the brain: paradigm shift in neuroscience. J Neurosci (2014) 34:15490–6. 10.1523/JNEUROSCI.3299-14.2014
    1. Sampson TR, Mazmanian SK. Control of brain development, function, and behavior by the microbiome. Cell Host Microbe (2015) 17:565–76. 10.1016/j.chom.2015.04.011
    1. Vuong HE, Yano JM, Fung TC, Hsiao EY. The microbiome and host behavior. Annu Rev Neurosci (2017) 40:21–49. 10.1146/annurev-neuro-072116-031347
    1. Chu C, Murdock MH, Jing D, Won TH, Chung H, Kressel AM, et al. The Microbiota regulate neuronal function and fear extinction learning. Nature (2019) 574:543–8. 10.1038/s41586-019-1644-y
    1. Dinan TG, Cryan JF. Melancholic microbes: a link between gut microbiota and depression? Neurogastroenterol Motil (2013) 25:713–9. 10.1111/nmo.12198
    1. Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF, Dinan TG. Minireview: gut microbiota: the neglected endocrine organ. Mol Endocrinol (2014) 28:1221–38. 10.1210/me.2014-1108
    1. El Aidy S, Dinan TG, Cryan JF. Immune modulation of the brain-gut microbe axis. Front Microbiol (2014) 5:146. 10.3389/fmicb.2014.00146
    1. Forsythe P, Bienenstock J, Kunze WA. Vagal pathways for microbiome-brain-gut axis communication. Adv Exp Med Biol (2014) 817:115–33. 10.1007/978-1-4939-0897-4_5
    1. Lyte M. Microbial endocrinology in the microbiome-gut-brain axis: how bacterial production and utilization of neurochemicals influence behavior. PloS Pathog (2013) 9:e1003726. 10.1371/journal.ppat.1003726
    1. Selkrig J, Wong P, Zhang X, Pettersson S. Metabolic tinkering by the gut microbiome: implications for brain development and function. Gut Microbes (2014) 5:369–80. 10.4161/gmic.28681
    1. Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. Genes Brain Behav (2014) 13:69–86. 10.1111/gbb.12109
    1. Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil (2011) 23:255–64. 10.1111/j.1365-2982.2010.01620
    1. Aizawa E, Tsuji H, Asahara T, Takahashi T, Teraishi T, Yoshida S, et al. Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J Affect Disord (2016) 202:254–7. 10.1016/j.jad.2016.05.038
    1. Schmidtner AK, Slattery DA, Glasner J, Hiergeist A, Gryksa K, Malik VA, et al. Minocycline alters behavior, microglia and the gut microbiome in a trait-anxiety-dependent manner. Transl Psychiatry (2019) 9:223. 10.1038/s41398-019-0556-9
    1. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, et al. Population-level analysis of gut microbiome variation. Science. (2016) 352:560–4. 10.1126/science.aad3503
    1. Camilleri M. Advances in understanding of bile acid diarrhea. Expert Rev Gastroenterol Hepatol (2014) 8:49– 61. 10.1586/17474124.2014.851599
    1. Jones ML, Martoni CJ, Ganopolsky JG, Labbé A, Prakash S. The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention. Expert Opin Biol Ther (2014) 14:467– 482. 10.1517/14712598.2014.880420
    1. Nieuwdorp M, Gilijamse PW, Pai N, Kaplan LM. Role of the microbiome in energy regulation and metabolism. Gastroenterology. (2014) 146:1525– 1533. 10.1053/j.gastro.2014.02.008
    1. Swann JR, Want EJ, Geier FM, Spagou K, Wilson ID, Sidaway JE, et al. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc Natl Acad Sci U S A (2011) 108(Suppl 1):4523–4530. 10.1073/pnas.1006734107

Source: PubMed

3
Tilaa