Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain-gut axis alterations

Marcel van de Wouw, Marcus Boehme, Joshua M Lyte, Niamh Wiley, Conall Strain, Orla O'Sullivan, Gerard Clarke, Catherine Stanton, Timothy G Dinan, John F Cryan, Marcel van de Wouw, Marcus Boehme, Joshua M Lyte, Niamh Wiley, Conall Strain, Orla O'Sullivan, Gerard Clarke, Catherine Stanton, Timothy G Dinan, John F Cryan

Abstract

Key points: Chronic (psychosocial) stress changes gut microbiota composition, as well as inducing behavioural and physiological deficits. The microbial metabolites short-chain fatty acids (SCFAs) have been implicated in gastrointestinal functional, (neuro)immune regulation and host metabolism, but their role in stress-induced behavioural and physiological alterations is poorly understood. Administration of SCFAs to mice undergoing psychosocial stress alleviates enduring alterations in anhedonia and heightened stress-responsiveness, as well as stress-induced increases in intestinal permeability. In contrast, chronic stress-induced alterations in body weight gain, faecal SCFAs and the gene expression of the SCFA receptors FFAR2 and FFAR3 remained unaffected by SCFA supplementation. These results present novel insights into mechanisms underpinning the influence of the gut microbiota on brain homeostasis, behaviour and host metabolism, informing the development of microbiota-targeted therapies for stress-related disorders.

Abstract: There is a growing recognition of the involvement of the gastrointestinal microbiota in the regulation of physiology and behaviour. Microbiota-derived metabolites play a central role in the communication between microbes and their host, with short-chain fatty acids (SCFAs) being perhaps the most studied. SCFAs are primarily derived from fermentation of dietary fibres and play a pivotal role in host gut, metabolic and immune function. All these factors have previously been demonstrated to be adversely affected by stress. Therefore, we sought to assess whether SCFA supplementation could counteract the enduring effects of chronic psychosocial stress. C57BL/6J male mice received oral supplementation of a mixture of the three principle SCFAs (acetate, propionate and butyrate). One week later, mice underwent 3 weeks of repeated psychosocial stress, followed by a comprehensive behavioural analysis. Finally, plasma corticosterone, faecal SCFAs and caecal microbiota composition were assessed. SCFA treatment alleviated psychosocial stress-induced alterations in reward-seeking behaviour, and increased responsiveness to an acute stressor and in vivo intestinal permeability. In addition, SCFAs exhibited behavioural test-specific antidepressant and anxiolytic effects, which were not present when mice had also undergone psychosocial stress. Stress-induced increases in body weight gain, faecal SCFAs and the colonic gene expression of the SCFA receptors free fatty acid receptors 2 and 3 remained unaffected by SCFA supplementation. Moreover, there were no collateral effects on caecal microbiota composition. Taken together, these data show that SCFA supplementation alleviates selective and enduring alterations induced by repeated psychosocial stress and these data may inform future research into microbiota-targeted therapies for stress-related disorders.

Keywords: Behaviour; Chronic Stress; Gut Microbiota.

© 2018 The Authors. The Journal of Physiology © 2018 The Physiological Society.

Figures

Figure 1. Experimental design of the short‐chain…
Figure 1. Experimental design of the short‐chain fatty acid treatment, psychosocial stress and behavioural assessment
After 1 week of short‐chain fatty acid (SCFA) treatment, animals underwent a 3‐week psychosocial stress protocol containing intermittent social defeat and overcrowding procedures. Mice subsequently underwent behavioural assessment, starting with the least stressful test to the most stressful test. The order of the behavioural tests was as follows. Week 4: social interaction test (SIT) and sucrose preference test (SPT); week 5: three‐chamber sociability test (3‐CT) and female urine sniffing test (FUST); week 6: open field (OF), novel object recognition test (NOR) and marble burying test (MB); week 7: elevated plus maze (EPM), stress‐induced hyperthermia test (SIH) and tail‐suspension test (TST); week 8: fear conditioning (FC) and hot plate test (HPT); week 9: FITC‐dextran intestinal permeability test (FITC); week 10: forced swim test (FST); week 11: sacrifice. SCFA treatment was for the entire duration. [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 2. Chronic stress induces aggressor‐specific social…
Figure 2. Chronic stress induces aggressor‐specific social avoidance
Social interaction with a CD1 mouse used in the social defeat procedure was assessed in the social interaction test 1 day after the last stressor (A). Mice were additionally assessed for social preference (B) and recognition (C) with a conspecific mouse in the three‐chamber sociability test 1 week post stress. The social interaction test was non‐parametrically distributed and analysed using the Kruskal‐Wallis test. Significant differences are depicted as: *P < 0.05; Control compared to Stress. In the three‐chamber sociability test, differences between ‘object’ versus ‘mouse’ and ‘familiar mouse’ versus ‘novel mouse’ were assessed using a Student's unpaired t test. Significant differences are depicted as: *P < 0.05, **P < 0.01 and ***P < 0.001. All data are expressed as means ± SEM (n = 9–10). [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 3. SCFAs decrease specific anxiety‐ and…
Figure 3. SCFAs decrease specific anxiety‐ and depressive‐like behaviours in control but not stressed animals
Anxiety‐like behaviour was assessed in the open field test 2 weeks post stress (A and B), whereas depressive‐like behaviour was determined using the forced swim test 6 weeks post stress (C). The open field test was non‐parametrically distributed and analysed using the Kruskal‐Wallis test, followed by the Mann‐Whitney test. The forced swim test was analysed using a two‐way ANOVA, followed by an LSD post hoc test. Significant differences are depicted as: *P < 0.05 and **P < 0.01; Control/Control compared to SCFA/Control, $$P < 0.01; SCFA/Control compared to SCFA/Stress. All data are expressed as means ± SEM (n = 9–10). Open circles on each graph represent individual animals. [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 4. Psychosocial stress induces long‐term anhedonia,…
Figure 4. Psychosocial stress induces long‐term anhedonia, which is absent after SCFA supplementation
A, hedonic and reward‐seeking behaviour was assessed using the female urine sniffing test 2 weeks post stress. B, the expression of genes involved in reward signalling was investigated in the striatum, which were the dopamine receptor D1a (DRD1a), dopamine receptor D2 (DRD2), brain‐derived neurotrophic factor (BDNF Exon IV) and tropomyosin receptor kinase B (TrkB). Data from the female urine sniffing test and BDNF gene expression was non‐parametrically distributed and analysed using the Kruskal‐Wallis test, followed by the Mann‐Whitney test. Gene expression data from all other genes were normally distributed and analysed using a two‐way ANOVA, followed by an LSD post hoc test. Significant differences are depicted as: **P < 0.01 and ***P < 0.001; Control/Control compared to SCFA/Control or Control/Stress, $P < 0.05; SCFA/Control compared to SCFA/Stress, #P < 0.05; Control/Stress compared to SCFA/Stress. All data are expressed as means ± SEM (n = 8–10). [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 5. Neither stress, nor SCFA treatment…
Figure 5. Neither stress, nor SCFA treatment affected the acquisition, retention and extinction phase data from the fear conditioning test
Cue‐ and context‐associative learning induced by foot shock was evaluated using fear conditioning 4 weeks post stress. At phase 1 (acquisition), mice were presented with a tone, followed by a foot chock. The context‐associative learning was assessed by measuring freezing behaviour in between tones (A), whereas cue‐associative learning was determined during the presentation of the tone (D). At phase 2 (retention), mice underwent the same protocol without the shocks to assess context‐ and cue‐associative retention (B and E, respectively). At phase 3 (extinction), mice were assessed for context‐ and cue‐associative extinction (C and F, respectively). All data are expressed as means ± SEM (n = 9–10). Open circles on each graph represent individual animals. [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 6. Psychosocial stress induces increased responsiveness…
Figure 6. Psychosocial stress induces increased responsiveness to acute stress, which was ameliorated by SCFAs
Stress responsiveness and HPA‐axis reactivity were assessed using the stress‐induced hyperthermia test 3 weeks after psychosocial stress (A), and by assessing the corticosterone levels in response to acute stress 6 weeks after psychosocial stress (B and C). The stressor used for the latter was the forced swim test. Hypothalamic genes involved in HPA‐axis signalling of which expression was investigated were corticotrophin‐releasing factor (CRF), mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) (D). For the hippocampus, these were corticotrophin‐releasing factor receptor 1 (CRFR1), MR and GR (E). All data were non‐parametrically distributed and analysed using the Kruskal‐Wallis test, followed by the Mann‐Whitney test. Significant differences are depicted as: *P < 0.05, **P < 0.01 and ***P < 0.001; Control/Control compared to SCFA/Control or Control/Stress, $P < 0.05, $$P < 0.01; SCFA/Control compared to SCFA/Stress, #P < 0.05 and ##P < 0.01; Control/Stress compared to SCFA/Stress. All data are expressed as means ± SEM (n = 8–10). [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 7. Stress induces intestinal permeability, which…
Figure 7. Stress induces intestinal permeability, which was rescued by SCFA treatment
A, in vivo intestinal permeability was assessed 5 weeks post stress. B, colonic genes involved in intestinal permeability of which gene expression was investigated were claudin‐1 (Cldn1), tight junction protein 1 (Tjp1), occludin (Ocln) and mucus 2 (Muc2). C, gene expression analysis of genes involved glucocorticoid signalling were corticotrophin‐releasing factor receptors 1 and 2 (CRFR1 and CRFR2 respectively), mineralocorticoid receptor (MR) and glucocorticoid receptor (GR). All data were non‐parametrically distributed and analysed using the Kruskal‐Wallis test, followed by the Mann‐Whitney test. Significant differences are depicted as: *P < 0.05, **P < 0.01 and ***P < 0.001; Control/Control compared to SCFA/Control or Control/Stress, $P < 0.05 and $$P < 0.01; SCFA/Control compared to SCFA/Stress, #P < 0.05 and ##P < 0.01; Control/Stress compared to SCFA/Stress. All data are expressed as means ± SEM (n = 8–10). Open circles on each graph represent individual animals. [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 8. Neither stress nor SCFAs affected…
Figure 8. Neither stress nor SCFAs affected caecal microbiota diversity
A, microbial alpha‐diversity was determined using Chao1, Simpson, Shannon metrics, as well as the number of species detected. B, beta‐diversity of the overall composition was depicted using principle component analysis. Alpha‐diversity metrics were non‐parametrically distributed and analysed using the Kruskal‐Wallis test. Data are depicted as median with IQR and minimum/maximum values as error bars (n = 9–10). [Color figure can be viewed at http://wileyonlinelibrary.com]
Figure 9. Subtle differences in caecum microbiota…
Figure 9. Subtle differences in caecum microbiota at family, genus and phylum levels were detected
The 16S sequencing revealed few significant differences in microbiota composition on phylum, family and genus level. Data are organised on stress‐effect (A), concurrent SCFA‐ and stress‐effect (B), and combined SCFA and stress effect (C). Data were non‐parametrically distributed and analysed using the Kruskal‐Wallis test, followed by the Mann‐Whitney test. Significant differences are depicted as: *P < 0.05, **P < 0.01 and ***P < 0.001; Control/Control compared to SCFA/Control or Control/Stress, $P < 0.05, $$P < 0.01 and $$$P < 0.001; SCFA/Control compared to SCFA/Stress, #P < 0.05 and ##P < 0.01; Control/Stress compared to SCFA/Stress. Data are depicted as median with IQR and minimum/maximum values as error bars (n = 9–10). [Color figure can be viewed at http://wileyonlinelibrary.com]

References

    1. Arnoldussen IAC, Wiesmann M, Pelgrim CE, Wielemaker EM, van Duyvenvoorde W, Amaral‐Santos PL, Verschuren L, Keijser BJF, Heerschap A, Kleemann R, Wielinga PY & Kiliaan AJ (2017). Butyrate restores HFD‐induced adaptations in brain function and metabolism in mid‐adult obese mice. Int J Obes 41, 935–944.
    1. Bailey MT, Dowd SE, Galley JD, Hufnagle AR, Allen RG & Lyte M (2011). Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor‐induced immunomodulation. Brain Behav Immun 25, 397–407.
    1. Bernstein CN ( 2017). The brain‐gut axis and stress in inflammatory bowel disease. Gastroenterol Clin North Am 46, 839–846.
    1. Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ, Graham D, Tsankova NM, Bolanos CA, Rios M, Monteggia LM, Self DW & Nestler EJ (2006). Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311, 864–868.
    1. Bharwani A, Mian MF, Foster JA, Surette MG, Bienenstock J & Forsythe P (2016). Structural and functional consequences of chronic psychosocial stress on the microbiome & host. Psychoneuroendocrinology 63, 217–227.
    1. Bharwani A, Mian MF, Surette MG, Bienenstock J & Forsythe P (2017). Oral treatment with Lactobacillus rhamnosus attenuates behavioural deficits and immune changes in chronic social stress. BMC Med 15, 7.
    1. Boets E, Gomand SV, Deroover L, Preston T, Vermeulen K, De Preter V, Hamer HM, Van den Mooter G, De Vuyst L, Courtin CM, Annaert P, Delcour JA & Verbeke KA (2017). Systemic availability and metabolism of colonic‐derived short‐chain fatty acids in healthy subjects: a stable isotope study. J Physiol 595, 541–555.
    1. Burokas A, Arboleya S, Moloney RD, Peterson VL, Murphy K, Clarke G, Stanton C, Dinan TG & Cryan JF (2017). Targeting the microbiota‐gut‐brain axis: prebiotics have anxiolytic and antidepressant‐like effects and reverse the impact of chronic stress in mice. Biol Psychiatry 82, 472–487.
    1. Byrne CS, Chambers ES, Alhabeeb H, Chhina N, Morrison DJ, Preston T, Tedford C, Fitzpatrick J, Irani C, Busza A, Garcia‐Perez I, Fountana S, Holmes E, Goldstone AP & Frost GS (2016). Increased colonic propionate reduces anticipatory reward responses in the human striatum to high‐energy foods. Am J Clin Nutr 104, 5–14.
    1. Byrne CS, Chambers ES, Morrison DJ & Frost G (2015). The role of short chain fatty acids in appetite regulation and energy homeostasis. Int J Obes 39, 1331–1338.
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Pena AG, Goodrich JK, Gordon JI, Huttley GA, Kelley ST, Knights D, Koenig JE, Ley RE, Lozupone CA, McDonald D, Muegge BD, Pirrung M, Reeder J, Sevinsky JR, Turnbaugh PJ, Walters WA, Widmann J, Yatsunenko T, Zaneveld J & Knight R (2010). QIIME allows analysis of high‐throughput community sequencing data. Nat Methods 7, 335–336.
    1. Chambers ES, Viardot A, Psichas A, Morrison DJ, Murphy KG, Zac‐Varghese SE, MacDougall K, Preston T, Tedford C, Finlayson GS, Blundell JE, Bell JD, Thomas EL, Mt‐Isa S, Ashby D, Gibson GR, Kolida S, Dhillo WS, Bloom SR, Morley W, Clegg S & Frost G (2015). Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 64, 1744–1754.
    1. Cryan JF & Mombereau C (2004). In search of a depressed mouse: utility of models for studying depression‐related behavior in genetically modified mice. Mol Psychiatry 9, 326–357.
    1. De Vadder F, Kovatcheva‐Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, Backhed F & Mithieux G (2014). Microbiota‐generated metabolites promote metabolic benefits via gut‐brain neural circuits. Cell 156, 84–96.
    1. Desbonnet L, Clarke G, Shanahan F, Dinan TG & Cryan JF (2014). Microbiota is essential for social development in the mouse. Mol Psychiatry 19, 146–148.
    1. Dunphy‐Doherty F, O'Mahony SM, Peterson VL, O'Sullivan O, Crispie F, Cotter PD, Wigmore P, King MV, Cryan JF & Fone KCF (2018). Post‐weaning social isolation of rats leads to long‐term disruption of the gut microbiota‐immune‐brain axis. Brain Behav Immun 68, 261–273.
    1. Edgar RC (2010). Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461.
    1. Egorin MJ, Yuan ZM, Sentz DL, Plaisance K & Eiseman JL (1999). Plasma pharmacokinetics of butyrate after intravenous administration of sodium butyrate or oral administration of tributyrin or sodium butyrate to mice and rats. Cancer Chemother Pharmacol 43, 445–453.
    1. Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren‐Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermohlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I & Prinz M (2015). Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18, 965–977.
    1. Erny D, Hrabe de Angelis AL & Prinz M (2017). Communicating systems in the body: how microbiota and microglia cooperate. Immunology 150, 7–15.
    1. Fanous S, Hammer RP Jr & Nikulina EM (2010). Short‐ and long‐term effects of intermittent social defeat stress on brain‐derived neurotrophic factor expression in mesocorticolimbic brain regions. Neuroscience 167, 598–607.
    1. Farmer AD, Mohammed SD, Dukes GE, Scott SM & Hobson AR (2014). Caecal pH is a biomarker of excessive colonic fermentation. World J Gastroenterol 20, 5000–5007.
    1. Fernandes J, Su W, Rahat‐Rozenbloom S, Wolever TM & Comelli EM (2014). Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans. Nutr Diabetes 4, e121.
    1. Finger BC, Dinan TG & Cryan JF (2011). High‐fat diet selectively protects against the effects of chronic social stress in the mouse. Neuroscience 192, 351–360.
    1. Finger BC, Dinan TG & Cryan JF (2012). The temporal impact of chronic intermittent psychosocial stress on high‐fat diet‐induced alterations in body weight. Psychoneuroendocrinology 37, 729–741.
    1. Foster JA, Rinaman L & Cryan JF (2017). Stress and the gut‐brain axis: Regulation by the microbiome. Neurobiol Stress 7, 124–136.
    1. Fouhy F, Deane J, Rea MC, O'Sullivan O, Ross RP, O'Callaghan G, Plant BJ & Stanton C (2015). The effects of freezing on faecal microbiota as determined using MiSeq sequencing and culture‐based investigations. PLoS One 10, e0119355.
    1. Galley JD, Mackos AR, Varaljay VA & Bailey MT (2017). Stressor exposure has prolonged effects on colonic microbial community structure in Citrobacter rodentium‐challenged mice. Sci Rep 7, 45012.
    1. Gao X, Cao Q, Cheng Y, Zhao D, Wang Z, Yang H, Wu Q, You L, Wang Y, Lin Y, Li X, Wang Y, Bian JS, Sun D, Kong L, Birnbaumer L & Yang Y (2018). Chronic stress promotes colitis by disturbing the gut microbiota and triggering immune system response. Proc Natl Acad Sci U S A 115, E2960–E2969.
    1. Golubeva AV, Joyce SA, Moloney G, Burokas A, Sherwin E, Arboleya S, Flynn I, Khochanskiy D, Moya‐Perez A, Peterson V, Rea K, Murphy K, Makarova O, Buravkov S, Hyland NP, Stanton C, Clarke G, Gahan CGM, Dinan TG & Cryan JF (2017). Microbiota‐related changes in bile acid & tryptophan metabolism are associated with gastrointestinal dysfunction in a mouse model of autism. EBioMedicine 24, 166–178.
    1. Gronier B, Savignac HM, Di Miceli M, Idriss SM, Tzortzis G, Anthony D & Burnet PWJ (2018). Increased cortical neuronal responses to NMDA and improved attentional set‐shifting performance in rats following prebiotic (B‐GOS((R))) ingestion. Eur Neuropsychopharmacol 28, 211–224.
    1. Grundy D (2015). Principles and standards for reporting animal experiments in The Journal of Physiology and Experimental Physiology . J Physiol 593, 2547–2549.
    1. Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC & McArthur S (2018). Microbiome‐host systems interactions: protective effects of propionate upon the blood‐brain barrier. Microbiome 6, 55.
    1. Iniguez SD, Riggs LM, Nieto SJ, Dayrit G, Zamora NN, Shawhan KL, Cruz B & Warren BL (2014). Social defeat stress induces a depression‐like phenotype in adolescent male c57BL/6 mice. Stress 17, 247–255.
    1. Johansson ME, Ambort D, Pelaseyed T, Schutte A, Gustafsson JK, Ermund A, Subramani DB, Holmen‐Larsson JM, Thomsson KA, Bergstrom JH, van der Post S, Rodriguez‐Pineiro AM, Sjovall H, Backstrom M & Hansson GC (2011). Composition and functional role of the mucus layers in the intestine. Cell Mol Life Sci 68, 3635–3641.
    1. Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, Weir TL, Ehrentraut SF, Pickel C, Kuhn KA, Lanis JM, Nguyen V, Taylor CT & Colgan SP (2015). Crosstalk between microbiota‐derived short‐chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe 17, 662–671.
    1. Kelly JR, Borre Y, C OB, Patterson E, El Aidy S, Deane J, Kennedy PJ, Beers S, Scott K, Moloney G, Hoban AE, Scott L, Fitzgerald P, Ross P, Stanton C, Clarke G, Cryan JF & Dinan TG (2016). Transferring the blues: Depression‐associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res 82, 109–118.
    1. Kim EJ, Pellman B & Kim JJ (2015). Stress effects on the hippocampus: a critical review. Learn Mem 22, 411–416.
    1. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M, Hirasawa A & Tsujimoto G (2011). Short‐chain fatty acids and ketones directly regulate sympathetic nervous system via G protein‐coupled receptor 41 (GPR41). Proc Natl Acad Sci U S A 108, 8030–8035.
    1. Klooker TK, Braak B, Painter RC, de Rooij SR, van Elburg RM, van den Wijngaard RM, Roseboom TJ & Boeckxstaens GE (2009). Exposure to severe wartime conditions in early life is associated with an increased risk of irritable bowel syndrome: a population‐based cohort study. Am J Gastroenterol 104, 2250–2256.
    1. Koh A, De Vadder F, Kovatcheva‐Datchary P & Backhed F (2016). From dietary fiber to host physiology: short‐chain fatty acids as key bacterial metabolites. Cell 165, 1332–1345.
    1. Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett‐Wilt GA, Rabaglia ME, Keller MP, Attie AD, Rey FE & Denu JM (2016). Diet‐microbiota interactions mediate global epigenetic programming in multiple host tissues. Mol Cell 64, 982–992.
    1. Li Z, Yi CX, Katiraei S, Kooijman S, Zhou E, Chung CK, Gao Y, van den Heuvel JK, Meijer OC, Berbee JFP, Heijink M, Giera M, Willems van Dijk K, Groen AK, Rensen PCN & Wang Y (2017). Butyrate reduces appetite and activates brown adipose tissue via the gut‐brain neural circuit. Gut 67, 1269–1279.
    1. Lin HV, Frassetto A, Kowalik EJ, Jr. , Nawrocki AR, Lu MM, Kosinski JR, Hubert JA, Szeto D, Yao X, Forrest G & Marsh DJ (2012). Butyrate and propionate protect against diet‐induced obesity and regulate gut hormones via free fatty acid receptor 3‐independent mechanisms. PLoS One 7, e35240.
    1. Liu J, Sun J, Wang F, Yu X, Ling Z, Li H, Zhang H, Jin J, Chen W, Pang M, Yu J, He Y & Xu J (2015). Neuroprotective effects of clostridium butyricum against vascular dementia in mice via metabolic butyrate. BioMed Res Int 2015, 412946.
    1. Lozupone C, Lladser ME, Knights D, Stombaugh J & Knight R (2011). UniFrac: an effective distance metric for microbial community comparison. ISME J 5, 169–172.
    1. Lu Y, Fan C, Li P, Lu Y, Chang X & Qi K (2016). Short chain fatty acids prevent high‐fat‐diet‐induced obesity in mice by regulating G protein‐coupled receptors and gut microbiota. Sci Rep 6, 37589.
    1. McKim DB, Patterson JM, Wohleb ES, Jarrett BL, Reader BF, Godbout JP & Sheridan JF (2016). Sympathetic release of splenic monocytes promotes recurring anxiety following repeated social defeat. Biol Psychiatry 79, 803–813.
    1. Magoc T & Salzberg SL (2011). FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics 27, 2957–2963.
    1. Marin IA, Goertz JE, Ren T, Rich SS, Onengut‐Gumuscu S, Farber E, Wu M, Overall CC, Kipnis J & Gaultier A (2017). Microbiota alteration is associated with the development of stress‐induced despair behavior. Sci Rep 7, 43859.
    1. Milligan G, Shimpukade B, Ulven T & Hudson BD (2017). Complex pharmacology of free fatty acid receptors. Chem Rev 117, 67–110.
    1. Miranda PM, De Palma G, Serkis V, Lu J, Louis‐Auguste MP, McCarville JL, Verdu EF, Collins SM & Bercik P (2018). High salt diet exacerbates colitis in mice by decreasing Lactobacillus levels and butyrate production. Microbiome 6, 57.
    1. Moloney RD, Johnson AC, O'Mahony SM, Dinan TG, Greenwood‐Van Meerveld B & Cryan JF (2016). Stress and the microbiota‐gut‐brain axis in visceral pain: relevance to irritable bowel syndrome. CNS Neurosci Ther 22, 102–117.
    1. Murphy K, Curley D, O'Callaghan TF, O'Shea CA, Dempsey EM, O'Toole PW, Ross RP, Ryan CA & Stanton C (2017). The composition of human milk and infant faecal microbiota over the first three months of life: a pilot study. Sci Rep 7, 40597.
    1. Nohr MK, Egerod KL, Christiansen SH, Gille A, Offermanns S, Schwartz TW & Moller M (2015). Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience 290, 126–137.
    1. O'Leary OF, Felice D, Galimberti S, Savignac HM, Bravo JA, Crowley T, El Yacoubi M, Vaugeois JM, Gassmann M, Bettler B, Dinan TG & Cryan JF (2014). GABAB(1) receptor subunit isoforms differentially regulate stress resilience. Proc Natl Acad Sci U S A 111, 15232–15237.
    1. Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL, Petersen KF, Kibbey RG, Goodman AL & Shulman GI (2016). Acetate mediates a microbiome‐brain‐beta‐cell axis to promote metabolic syndrome. Nature 534, 213–217.
    1. Pluznick JL (2017). Microbial short‐chain fatty acids and blood pressure regulation. Curr Hypertens Rep 19, 25.
    1. Pouteau E, Nguyen P, Ballevre O & Krempf M (2003). Production rates and metabolism of short‐chain fatty acids in the colon and whole body using stable isotopes. Proc Nutr Soc 62, 87–93.
    1. Pusceddu MM, El Aidy S, Crispie F, O'Sullivan O, Cotter P, Stanton C, Kelly P, Cryan JF & Dinan TG (2015a). N‐3 Polyunsaturated fatty acids (PUFAs) reverse the impact of early‐life stress on the gut microbiota. PLoS One 10, e0139721.
    1. Pusceddu MM, Kelly P, Ariffin N, Cryan JF, Clarke G & Dinan TG (2015b). N‐3 PUFAs have beneficial effects on anxiety and cognition in female rats: Effects of early life stress. Psychoneuroendocrinology 58, 79–90.
    1. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J & Glockner FO (2013). The SILVA ribosomal RNA gene database project: improved data processing and web‐based tools. Nucleic Acids Res 41, D590–596.
    1. Rahat‐Rozenbloom S, Fernandes J, Gloor GB & Wolever TM (2014). Evidence for greater production of colonic short‐chain fatty acids in overweight than lean humans. Int J Obes 38, 1525–1531.
    1. Ramirez K, Fornaguera‐Trias J & Sheridan JF (2017). Stress‐induced microglia activation and monocyte trafficking to the brain underlie the development of anxiety and depression. Curr Top Behav Neurosci 31, 155–172.
    1. Razzoli M & Bartolomucci A (2016). The dichotomous effect of chronic stress on obesity. Trends Endocrinol Metab 27, 504–515.
    1. Razzoli M, Pearson C, Crow S & Bartolomucci A (2017). Stress, overeating, and obesity: insights from human studies and preclinical models. Neurosci Biobehav Rev 76, 154–162.
    1. Rea K, Dinan TG & Cryan JF (2017). The brain‐gut axis contributes to neuroprogression in stress‐related disorders. Mod Trends Pharmacopsychiatry 31, 152–161.
    1. Reigstad CS, Salmonson CE, Rainey JF 3rd, Szurszewski JH, Linden DR, Sonnenburg JL, Farrugia G & Kashyap PC (2015). Gut microbes promote colonic serotonin production through an effect of short‐chain fatty acids on enterochromaffin cells. FASEB J 29, 1395–1403.
    1. Ringel‐Kulka T, Choi CH, Temas D, Kim A, Maier DM, Scott K, Galanko JA & Ringel Y (2015). Altered colonic bacterial fermentation as a potential pathophysiological factor in irritable bowel syndrome. Am J Gastroenterol 110, 1339–1346.
    1. Rodino‐Janeiro BK, Alonso‐Cotoner C, Pigrau M, Lobo B, Vicario M & Santos J (2015). Role of corticotropin‐releasing factor in gastrointestinal permeability. J Neurogastroenterol Motil 21, 33–50.
    1. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V, Chesselet MF, Keshavarzian A, Shannon KM, Krajmalnik‐Brown R, Wittung‐Stafshede P, Knight R & Mazmanian SK (2016). Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease. Cell 167, 1469–1480 e1412.
    1. Schellekens H, Clarke G, Jeffery IB, Dinan TG & Cryan JF (2012). Dynamic 5‐HT2C receptor editing in a mouse model of obesity. PLoS One 7, e32266.
    1. Schonfeld P & Wojtczak L (2016). Short‐ and medium‐chain fatty acids in energy metabolism: the cellular perspective. J Lipid Res 57, 943–954.
    1. Schwiertz A, Taras D, Schafer K, Beijer S, Bos NA, Donus C & Hardt PD (2010). Microbiota and SCFA in lean and overweight healthy subjects. Obesity 18, 190–195.
    1. Scott KA, Ida M, Peterson VL, Prenderville JA, Moloney GM, Izumo T, Murphy K, Murphy A, Ross RP, Stanton C, Dinan TG & Cryan JF (2017). Revisiting metchnikoff: age‐related alterations in microbiota‐gut‐brain axis in the mouse. Brain Behav Immun 65, 20–32.
    1. Shultz SR, Aziz NA, Yang L, Sun M, MacFabe DF & O'Brien TJ (2015). Intracerebroventricular injection of propionic acid, an enteric metabolite implicated in autism, induces social abnormalities that do not differ between seizure‐prone (FAST) and seizure‐resistant (SLOW) rats. Behav Brain Res 278, 542–548.
    1. Simeoli R, Mattace Raso G, Pirozzi C, Lama A, Santoro A, Russo R, Montero‐Melendez T, Berni Canani R, Calignano A, Perretti M & Meli R (2017). An orally administered butyrate‐releasing derivative reduces neutrophil recruitment and inflammation in dextran sulphate sodium‐induced murine colitis. Br J Pharmacol 174, 1484–1496.
    1. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN & Garrett WS (2013). The microbial metabolites, short‐chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573.
    1. Stam R, Akkermans LM & Wiegant VM (1997). Trauma and the gut: interactions between stressful experience and intestinal function. Gut 40, 704–709.
    1. Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG & Cryan JF (2016). The neuropharmacology of butyrate: the bread and butter of the microbiota‐gut‐brain axis? Neurochem Int 99, 110–132.
    1. Szyszkowicz JK, Wong A, Anisman H, Merali Z & Audet MC (2017). Implications of the gut microbiota in vulnerability to the social avoidance effects of chronic social defeat in male mice. Brain Behav Immun 66, 45–55.
    1. Tahara Y, Yamazaki M, Sukigara H, Motohashi H, Sasaki H, Miyakawa H, Haraguchi A, Ikeda Y, Fukuda S & Shibata S (2018). Gut microbiota‐derived short chain fatty acids induce circadian clock entrainment in mouse peripheral tissue. Sci Rep 8, 1395.
    1. Tong LC, Wang Y, Wang ZB, Liu WY, Sun S, Li L, Su DF & Zhang LC (2016). Propionate ameliorates dextran sodium sulfate‐induced colitis by improving intestinal barrier function and reducing inflammation and oxidative stress. Front Pharmacol 7, 253.
    1. Tramullas M, Dinan TG & Cryan JF (2012). Chronic psychosocial stress induces visceral hyperalgesia in mice. Stress 15, 281–292.
    1. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER & Gordon JI (2006). An obesity‐associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031.
    1. Van Bokhoven P, Oomen CA, Hoogendijk WJ, Smit AB, Lucassen PJ & Spijker S (2011). Reduction in hippocampal neurogenesis after social defeat is long‐lasting and responsive to late antidepressant treatment. Eur J Neurosci 33, 1833–1840.
    1. van de Wouw M, Schellekens H, Dinan TG & Cryan JF (2017). Microbiota‐gut‐brain axis: modulator of host metabolism and appetite. J Nutr 147, 727–745.
    1. van der Beek CM, Bloemen JG, van den Broek MA, Lenaerts K, Venema K, Buurman WA & Dejong CH (2015). Hepatic uptake of rectally administered butyrate prevents an increase in systemic butyrate concentrations in humans. J Nutr 145, 2019–2024.
    1. Vancampfort D, Stubbs B, Mitchell AJ, De Hert M, Wampers M, Ward PB, Rosenbaum S & Correll CU (2015). Risk of metabolic syndrome and its components in people with schizophrenia and related psychotic disorders, bipolar disorder and major depressive disorder: a systematic review and meta‐analysis. World Psychiatry 14, 339–347.
    1. Vazquez‐Baeza Y, Pirrung M, Gonzalez A & Knight R (2013). EMPeror: a tool for visualizing high‐throughput microbial community data. Gigascience 2, 16.
    1. Veeraiah P, Noronha JM, Maitra S, Bagga P, Khandelwal N, Chakravarty S, Kumar A & Patel AB (2014). Dysfunctional glutamatergic and gamma‐aminobutyric acidergic activities in prefrontal cortex of mice in social defeat model of depression. Biol Psychiatry 76, 231–238.
    1. Ver Hoeve ES, Kelly G, Luz S, Ghanshani S & Bhatnagar S (2013). Short‐term and long‐term effects of repeated social defeat during adolescence or adulthood in female rats. Neuroscience 249, 63–73.
    1. Vinolo MA, Rodrigues HG, Festuccia WT, Crisma AR, Alves VS, Martins AR, Amaral CL, Fiamoncini J, Hirabara SM, Sato FT, Fock RA, Malheiros G, dos Santos MF & Curi R (2012). Tributyrin attenuates obesity‐associated inflammation and insulin resistance in high‐fat‐fed mice. Am J Physiol Endocrinol Metab 303, E272–E282.
    1. Wiley NC, Dinan TG, Ross RP, Stanton C, Clarke G & Cryan JF (2017). The microbiota‐gut‐brain axis as a key regulator of neural function and the stress response: Implications for human and animal health. J Anim Sci 95, 3225–3246.
    1. Wohleb ES & Delpech JC (2016). Dynamic cross‐talk between microglia and peripheral monocytes underlies stress‐induced neuroinflammation and behavioral consequences. Prog Neuropsychopharmacol Biol Psychiatry 79, 40–48.
    1. Wohleb ES, McKim DB, Shea DT, Powell ND, Tarr AJ, Sheridan JF & Godbout JP (2014). Re‐establishment of anxiety in stress‐sensitized mice is caused by monocyte trafficking from the spleen to the brain. Biol Psychiatry 75, 970–981.
    1. Yang L, Zhao Y, Wang Y, Liu L, Zhang X, Li B & Cui R (2015). The effects of psychological stress on depression. Curr Neuropharmacol 13, 494–504.
    1. Zhao G, Nyman M & Jonsson JA (2006). Rapid determination of short‐chain fatty acids in colonic contents and faeces of humans and rats by acidified water‐extraction and direct‐injection gas chromatography. Biomed Chromatogr 20, 674–682.
    1. Zihni C, Mills C, Matter K & Balda MS (2016). Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol 17, 564–580.

Source: PubMed

3
구독하다