The intestinal microbiota and metabolites in patients with anorexia nervosa

Petra Prochazkova, Radka Roubalova, Jiri Dvorak, Jakub Kreisinger, Martin Hill, Helena Tlaskalova-Hogenova, Petra Tomasova, Helena Pelantova, Martina Cermakova, Marek Kuzma, Josef Bulant, Martin Bilej, Kvido Smitka, Alena Lambertova, Petra Holanova, Hana Papezova, Petra Prochazkova, Radka Roubalova, Jiri Dvorak, Jakub Kreisinger, Martin Hill, Helena Tlaskalova-Hogenova, Petra Tomasova, Helena Pelantova, Martina Cermakova, Marek Kuzma, Josef Bulant, Martin Bilej, Kvido Smitka, Alena Lambertova, Petra Holanova, Hana Papezova

Abstract

Brain-gut microbiota interactions are intensively studied in connection with various neurological and psychiatric diseases. While anorexia nervosa (AN) pathophysiology is not entirely clear, it is presumably linked to microbiome dysbiosis. We aimed to elucidate the gut microbiota contribution in AN disease pathophysiology. We analyzed the composition and diversity of the gut microbiome of patients with AN (bacteriome and mycobiome) from stool samples before and after renourishment, and compared them to healthy controls. Further, levels of assorted neurotransmitters and short-chain fatty acids (SCFA) were analyzed in stool samples by MS and NMR, respectively. Biochemical, anthropometric, and psychometric profiles were assessed. The bacterial alpha-diversity parameter analyses revealed only increased Chao 1 index in patients with AN before the realimentation, reflecting their interindividual variation. Subsequently, core microbiota depletion signs were observed in patients with AN. Overrepresented OTUs (operation taxonomic units) in patients with AN taxonomically belonged to Alistipes, Clostridiales, Christensenellaceae, and Ruminococcaceae. Underrepresented OTUs in patients with AN were Faecalibacterium, Agathobacter, Bacteroides, Blautia, and Lachnospira. Patients exhibited greater interindividual variation in the gut bacteriome, as well as in metagenome content compared to controls, suggesting altered bacteriome functions. Patients had decreased levels of serotonin, GABA, dopamine, butyrate, and acetate in their stool samples compared to controls. Mycobiome analysis did not reveal significant differences in alpha diversity and fungal profile composition between patients with AN and healthy controls, nor any correlation of the fungal composition with the bacterial profile. Our results show the changed profile of the gut microbiome and its metabolites in patients with severe AN. Although therapeutic partial renourishment led to increased body mass index and improved psychometric parameters, SCFA, and neurotransmitter profiles, as well as microbial community compositions, did not change substantially during the hospitalization period, which can be potentially caused by only partial weight recovery.

Keywords: BMI; EDE-Q; Microbiome; SCFA; bacteriome; dysbiosis; gut-brain-microbiota axis; mycobiome; neurotransmitter; renourishment.

Figures

Figure 1.
Figure 1.
Gut bacteriome alpha diversity variation between control individuals vs. AN1 vs. AN2 assessed based on A) Observed OTUs number, B) Total OTU richness predicted by Chao 1 index, and C) Shannon index. Significant differences between categories (p < 0.05 according to Tukey post-hoc tests) are indicated by different letters above the bars
Figure 2.
Figure 2.
Proportions of dominating bacterial classes (represented by >1% of reads) in the three studied groups
Figure 2.
Figure 2.
Proportions of dominating bacterial classes (represented by >1% of reads) in the three studied groups
Figure 3.
Figure 3.
PCoA showing variation in bacterial microbiota composition between controls vs. AN1 vs. AN2. Compositional variation was assessed based on A) relative abundance-based (Bray-Curtis) and B) prevalence-based (Jaccard) dissimilarities
Figure 3.
Figure 3.
PCoA showing variation in bacterial microbiota composition between controls vs. AN1 vs. AN2. Compositional variation was assessed based on A) relative abundance-based (Bray-Curtis) and B) prevalence-based (Jaccard) dissimilarities
Figure 4.
Figure 4.
Bacterial core microbiota in the studied groups. A) Proportion of reads corresponding to core bacterial OTUs (i.e. detected in >90% samples) in each studied group and B) heatmap showing their prevalences
Figure 4.
Figure 4.
Bacterial core microbiota in the studied groups. A) Proportion of reads corresponding to core bacterial OTUs (i.e. detected in >90% samples) in each studied group and B) heatmap showing their prevalences
Figure 5.
Figure 5.
Relative abundances of bacterial OTUs (squared-root transformed) that varied, according to DESeq2 analyses (FDR

Figure 6.

A) PCoA showing variation in…

Figure 6.

A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls…

Figure 6.
A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls vs. AN1 vs. AN2, and B) Relative abundances of bacterial metagenomic pathways (squared-root transformed) that varied, according to DESeq2 analyses (FDR

Figure 6.

A) PCoA showing variation in…

Figure 6.

A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls…

Figure 6.
A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls vs. AN1 vs. AN2, and B) Relative abundances of bacterial metagenomic pathways (squared-root transformed) that varied, according to DESeq2 analyses (FDR

Figure 7.

Fungal microbiota variation in the…

Figure 7.

Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions…

Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples

Figure 7.

Fungal microbiota variation in the…

Figure 7.

Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions…

Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples

Figure 8.

Significant associations between bacterial OTU…

Figure 8.

Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions…

Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown

Figure 8.

Significant associations between bacterial OTU…

Figure 8.

Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions…

Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown
All figures (14)
Similar articles
Cited by
References
    1. Association AP . Diagnostic and statistical manual of mental disorders, 5th ed. Washington (DC, USA): American Psychiatric Publishing, Inc.; 2013.
    1. Liang D, Leung RK, Guan W, Au WW.. Involvement of gut microbiome in human health and disease: brief overview, knowledge gaps and research opportunities. Gut Pathog. 2018;10:3. doi:10.1186/s13099-018-0230-4. - DOI - PMC - PubMed
    1. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017. April;17(4):219–25. doi:10.1038/nri.2017.7. - DOI - PubMed
    1. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome. Genome Med. 2016. April 27;8(1):51. doi:10.1186/s13073-016-0307-y. - DOI - PMC - PubMed
    1. Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC, Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA, et al. The gut mycobiome of the human microbiome project healthy cohort. Microbiome. 2017. November 25;5(1):153. doi:10.1186/s40168-017-0373-4. - DOI - PMC - PubMed
Show all 56 references
Publication types
MeSH terms
Substances
Related information
Grant support
This work was supported by the Ministry of Health of the Czech Republic [NV18-01-00040]; Ministry of Health of the Czech Republic [17-28905A].
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 6.
Figure 6.
A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls vs. AN1 vs. AN2, and B) Relative abundances of bacterial metagenomic pathways (squared-root transformed) that varied, according to DESeq2 analyses (FDR

Figure 6.

A) PCoA showing variation in…

Figure 6.

A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls…

Figure 6.
A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls vs. AN1 vs. AN2, and B) Relative abundances of bacterial metagenomic pathways (squared-root transformed) that varied, according to DESeq2 analyses (FDR

Figure 7.

Fungal microbiota variation in the…

Figure 7.

Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions…

Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples

Figure 7.

Fungal microbiota variation in the…

Figure 7.

Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions…

Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples

Figure 8.

Significant associations between bacterial OTU…

Figure 8.

Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions…

Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown

Figure 8.

Significant associations between bacterial OTU…

Figure 8.

Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions…

Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown
All figures (14)
Similar articles
Cited by
References
    1. Association AP . Diagnostic and statistical manual of mental disorders, 5th ed. Washington (DC, USA): American Psychiatric Publishing, Inc.; 2013.
    1. Liang D, Leung RK, Guan W, Au WW.. Involvement of gut microbiome in human health and disease: brief overview, knowledge gaps and research opportunities. Gut Pathog. 2018;10:3. doi:10.1186/s13099-018-0230-4. - DOI - PMC - PubMed
    1. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017. April;17(4):219–25. doi:10.1038/nri.2017.7. - DOI - PubMed
    1. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome. Genome Med. 2016. April 27;8(1):51. doi:10.1186/s13073-016-0307-y. - DOI - PMC - PubMed
    1. Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC, Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA, et al. The gut mycobiome of the human microbiome project healthy cohort. Microbiome. 2017. November 25;5(1):153. doi:10.1186/s40168-017-0373-4. - DOI - PMC - PubMed
Show all 56 references
Publication types
MeSH terms
Substances
Related information
Grant support
This work was supported by the Ministry of Health of the Czech Republic [NV18-01-00040]; Ministry of Health of the Czech Republic [17-28905A].
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6.
Figure 6.
A) PCoA showing variation in relative abundances of predicted metabolic pathways between controls vs. AN1 vs. AN2, and B) Relative abundances of bacterial metagenomic pathways (squared-root transformed) that varied, according to DESeq2 analyses (FDR

Figure 7.

Fungal microbiota variation in the…

Figure 7.

Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions…

Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples

Figure 7.

Fungal microbiota variation in the…

Figure 7.

Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions…

Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples

Figure 8.

Significant associations between bacterial OTU…

Figure 8.

Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions…

Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown

Figure 8.

Significant associations between bacterial OTU…

Figure 8.

Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions…

Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown
All figures (14)
Figure 7.
Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples
Figure 7.
Figure 7.
Fungal microbiota variation in the three studied groups. A) Dominating fungal class proportions (represented by >1% of reads); PCoA for B) Bray-Curtis and C) Jaccard dissimilarities between samples
Figure 8.
Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown
Figure 8.
Figure 8.
Significant associations between bacterial OTU abundances and concentrations of SCFAs or neurotransmitters. Predictions and 95% confidence intervals for negative binomial generalized linear mixed models (GLMMs) are shown

References

    1. Association AP . Diagnostic and statistical manual of mental disorders, 5th ed. Washington (DC, USA): American Psychiatric Publishing, Inc.; 2013.
    1. Liang D, Leung RK, Guan W, Au WW.. Involvement of gut microbiome in human health and disease: brief overview, knowledge gaps and research opportunities. Gut Pathog. 2018;10:3. doi:10.1186/s13099-018-0230-4.
    1. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017. April;17(4):219–25. doi:10.1038/nri.2017.7.
    1. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome. Genome Med. 2016. April 27;8(1):51. doi:10.1186/s13073-016-0307-y.
    1. Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC, Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA, et al. The gut mycobiome of the human microbiome project healthy cohort. Microbiome. 2017. November 25;5(1):153. doi:10.1186/s40168-017-0373-4.
    1. Chong CYL, Bloomfield FH, O’Sullivan JM. Factors affecting gastrointestinal microbiome development in neonates. Nutrients. 2018. February 28;10(3):274. doi:10.3390/nu10030274.
    1. Kleiman SC, Watson HJ, Bulik-Sullivan EC, Huh EY, Tarantino LM, Bulik CM, Carroll IM. The intestinal microbiota in acute anorexia nervosa and during renourishment: relationship to depression, anxiety, and eating disorder psychopathology. Psychosom Med. 2015. Nov-Dec;77(9):969–981. doi:10.1097/PSY.0000000000000247.
    1. Armougom F, Henry M, Vialettes B, Raccah D, Raoult D, Ratner AJ. Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients. PLoS One. 2009. September 23;4(9):e7125. doi:10.1371/journal.pone.0007125.
    1. Morita C, Tsuji H, Hata T, Gondo M, Takakura S, Kawai K, Yoshihara K, Ogata K, Nomoto K, Miyazaki K, et al. Gut dysbiosis in patients with anorexia nervosa. PLoS One. 2015;10(12):e0145274. doi:10.1371/journal.pone.0145274.
    1. Million M, Angelakis E, Maraninchi M, Henry M, Giorgi R, Valero R, Vialettes B, Raoult D. Correlation between body mass index and gut concentrations of Lactobacillus reuteri, Bifidobacterium animalis, Methanobrevibacter smithii and Escherichia coli. Int J Obes (Lond). 2013. November;37(11):1460–1466. doi:10.1038/ijo.2013.20.
    1. Borgo F, Riva A, Benetti A, Casiraghi MC, Bertelli S, Garbossa S, Anselmetti S, Scarone S, Pontiroli AE, Morace G, et al. Microbiota in anorexia nervosa: the triangle between bacterial species, metabolites and psychological tests. PLoS One. 2017;12(6):e0179739. doi:10.1371/journal.pone.0179739.
    1. Morkl S, Lackner S, Muller W, Gorkiewicz G, Kashofer K, Oberascher A, Painold A, Holl A, Holzer P, Meinitzer A, et al. Gut microbiota and body composition in anorexia nervosa inpatients in comparison to athletes, overweight, obese, and normal weight controls. Int J Eat Disord. 2017. December;50(12):1421–1431. doi:10.1002/eat.22801.
    1. Mack I, Cuntz U, Gramer C, Niedermaier S, Pohl C, Schwiertz A, Zimmermann K, Zipfel S, Enck P, Penders J. Weight gain in anorexia nervosa does not ameliorate the faecal microbiota, branched chain fatty acid profiles, and gastrointestinal complaints. Sci Rep. 2016. May;27(6):26752. doi:10.1038/srep26752.
    1. Hanachi M, Manichanh C, Schoenenberger A, Pascal V, Levenez F, Cournede N, Dore J, Melchior JC. Altered host-gut microbes symbiosis in severely malnourished anorexia nervosa (AN) patients undergoing enteral nutrition: an explicative factor of functional intestinal disorders? Clin Nutr. 2019. October;38(5):2304–2310. doi:10.1016/j.clnu.2018.10.004.
    1. Monteleone AM, Troisi J, Fasano A, Dalle Grave R, Marciello F, Serena G, Calugi S, Scala G, Corrivetti G, Cascino G, et al. Multi-omics data integration in anorexia nervosa patients before and after weight regain: a microbiome-metabolomics investigation. Clin Nutr. 2020. July 31;40(3):1137–1146. doi:10.1016/j.clnu.2020.07.021.
    1. Neuman H, Debelius JW, Knight R, Koren O. Microbial endocrinology: the interplay between the microbiota and the endocrine system. FEMS Microbiol Rev. 2015. July;39(4):509–521. doi:10.1093/femsre/fuu010.
    1. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science. 2012. June 8;336(6086):1262–1267. doi:10.1126/science.1223813.
    1. Wong JM, De Souza R, Kendall CW, Emam A, Jenkins DJ. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol. 2006. March;40(3):235–243. doi:10.1097/00004836-200603000-00015.
    1. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015. Apr-Jun;28(2):203–209.
    1. Roubalova R, Prochazkova P, Papezova H, Smitka K, Bilej M, Tlaskalova-Hogenova H. Anorexia nervosa: gut microbiota-immune-brain interactions. Clin Nutr. 2020. March;39(3):676–684. doi:10.1016/j.clnu.2019.03.023.
    1. Shetty SA, Hugenholtz F, Lahti L, Smidt H, De Vos WM. Intestinal microbiome landscaping: insight in community assemblage and implications for microbial modulation strategies. FEMS Microbiol Rev. 2017. March 1;41(2):182–199. doi:10.1093/femsre/fuw045.
    1. Furtado M, Katzman MA. Neuroinflammatory pathways in anxiety, posttraumatic stress, and obsessive compulsive disorders. Psychiatry Res. 2015. September 30;229(1–2):37–48. doi:10.1016/j.psychres.2015.05.036.
    1. Wales J, Brewin N, Cashmore R, Haycraft E, Baggott J, Cooper A, Arcelus J. Predictors of positive treatment outcome in people with anorexia nervosa treated in a specialized inpatient unit: the role of early response to treatment. Eur Eat Disord Rev. 2016. September;24(5):417–424. doi:10.1002/erv.2443.
    1. Zaneveld JR, McMinds R, Vega Thurber R. Stress and stability: applying the Anna Karenina principle to animal microbiomes. Nat Microbiol. 2017. August;24(2):17121. doi:10.1038/nmicrobiol.2017.121.
    1. Parker BJ, Wearsch PA, Veloo ACM, Rodriguez-Palacios A. The genus Alistipes: gut bacteria with emerging implications to inflammation, cancer, and mental health. Front Immunol. 2020;11. doi:10.3389/fimmu.2020.00906.
    1. Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun. 2015. August;48:186–194. doi:10.1016/j.bbi.2015.03.016.
    1. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT, et al. Human genetics shape the gut microbiome. Cell. 2014. November 6;159(4):789–799. doi:10.1016/j.cell.2014.09.053.
    1. Ruppin H, Bar-Meir S, Soergel KH, Wood CM, Schmitt MG Jr.. Absorption of short-chain fatty acids by the colon. Gastroenterology. 1980. June;78(6):1500–1507. doi:10.1016/S0016-5085(19)30508-6.
    1. Gao J, Cahill CM, Huang X, Roffman JL, Lamon-Fava S, Fava M, Mischoulon D, Rogers JT. S-adenosyl methionine and transmethylation pathways in neuropsychiatric diseases throughout life. Neurotherapeutics. 2018. January;15(1):156–175.
    1. De Berardis D, Orsolini L, Serroni N, Girinelli G, Iasevoli F, Tomasetti C, De Bartolomeis A, Mazza M, Valchera A, Fornaro M, et al. A comprehensive review on the efficacy of S-adenosyl-L-methionine in major depressive disorder. CNS Neurol Disord Drug Targets. 2016;15(1):35–44. doi:10.2174/1871527314666150821103825.
    1. Hoffmann C, Dollive S, Grunberg S, Chen J, Li H, Wu GD, Lewis JD, Bushman FD. Archaea and fungi of the human gut microbiome: correlations with diet and bacterial residents. PLoS One. 2013;8(6):e66019. doi:10.1371/journal.pone.0066019.
    1. Gabaldon T, Martin T, Marcet-Houben M, Durrens P, Bolotin-Fukuhara M, Lespinet O, Arnaise S, Boisnard S, Aguileta G, Atanasova R, et al. Comparative genomics of emerging pathogens in the Candida glabrata clade. BMC Genomics. 2013. September;14(14):623. doi:10.1186/1471-2164-14-623.
    1. Prochazkova P, Roubalova R, Dvorak J, Tlaskalova-Hogenova H, Cermakova M, Tomasova P, Sediva B, Kuzma M, Bulant J, Bilej M, et al. Microbiota, microbial metabolites, and barrier function in a patient with anorexia nervosa after fecal microbiota transplantation. Microorganisms. 2019. September 10;7(9):338. doi:10.3390/microorganisms7090338.
    1. Kaye WH, Fudge JL, Paulus M. New insights into symptoms and neurocircuit function of anorexia nervosa. Nat Rev Neurosci. 2009. August;10(8):573–584. doi:10.1038/nrn2682.
    1. Cryan JF, Kaupmann K. Don’t worry ‘B’ happy!: a role for GABA(B) receptors in anxiety and depression. Trends Pharmacol Sci. 2005. January;26(1):36–43. doi:10.1016/j.tips.2004.11.004.
    1. Den Besten G, Van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013. September;54(9):2325–2340. doi:10.1194/jlr.R036012.
    1. Fairburn CG. Eating disorders: the transdiagnostic view and the cognitive behavioral theory. In: Fairburn CG, editor. Cognitive behavior therapy and eating disorders. New York, USA: Guilford Press; 2008. p. 7–22.
    1. Fairburn CG, Beglin SJ. Assessment of eating disorders: interview or self-report questionnaire? Int J Eat Disord. 1994. December;16(4):363–370.
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, Fierer N, Knight R. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci U S A. 2011. March 15;108(Suppl 1):4516–4522. doi:10.1073/pnas.1000080107.
    1. Ihrmark K, Bodeker IT, Cruz-Martinez K, Friberg H, Kubartova A, Schenck J, Strid Y, Stenlid J, Brandstrom-Durling M, Clemmensen KE, et al. New primers to amplify the fungal ITS2 region–evaluation by 454-sequencing of artificial and natural communities. FEMS Microbiol Ecol. 2012. December;82(3):666–677. doi:10.1111/j.1574-6941.2012.01437.x.
    1. Jiang H, Lei R, Ding SW, Zhu S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinform. 2014. June;12(15):182. doi:10.1186/1471-2105-15-182.
    1. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods. 2016. July;13(7):581–583. doi:10.1038/nmeth.3869.
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics. 2011. August 15;27(16):2194–2200. doi:10.1093/bioinformatics/btr381.
    1. Nilsson RH, Larsson KH, Taylor AFS, Bengtsson-Palme J, Jeppesen TS, Schigel D, Kennedy P, Picard K, Glockner FO, Tedersoo L, et al. The UNITE database for molecular identification of fungi: handling dark taxa and parallel taxonomic classifications. Nucleic Acids Res. 2019. January 8;47(D1):D259–D264. doi:10.1093/nar/gky1022.
    1. Rognes T, Flouri T, Nichols B, Quince C, Mahe F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4:e2584. doi:10.7717/peerj.2584.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007. August;73(16):5261–5267. doi:10.1128/AEM.00062-07.
    1. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glockner FO. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013. January;41(D1):D590–6. doi:10.1093/nar/gks1219.
    1. Stamatakis A. RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics. 2014. May 1;30(9):1312–1313. doi:10.1093/bioinformatics/btu033.
    1. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol. 2013. April;30(4):772–780. doi:10.1093/molbev/mst010.
    1. McMurdie PJ, Holmes S, Watson M. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8(4):e61217. doi:10.1371/journal.pone.0061217.
    1. Douglas GM, Maffei VJ, Zanevel J, Yurgel SN, Brown JR, Christopher M, Taylor CM, Huttenhower C, Langille MGI. PICRUSt2: an improved and customizable approach for metagenome inference. BioRxiv. 2020. doi:10.1101/672295
    1. Ye Y, Doak TG, Ouzounis CA. A parsimony approach to biological pathway reconstruction/inference for genomes and metagenomes. PLoS Comput Biol. 2009. August;5(8):e1000465. doi:10.1371/journal.pcbi.1000465.
    1. Ondov BD, Bergman NH, Phillippy AM. Interactive metagenomic visualization in a Web browser. BMC Bioinform. 2011. September;30(12):385. doi:10.1186/1471-2105-12-385.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. doi:10.1186/s13059-014-0550-8.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate - a practical and powerful approach to multiple testing. J R Stat Soc Ser B-Stat Methodol. 1995;57:289–300.
    1. Dieterle F, Ross A, Schlotterbeck G, Senn H. Probabilistic quotient normalization as robust method to account for dilution of complex biological mixtures. Application in 1H NMR metabonomics. Anal Chem. 2006. July 1;78(13):4281–4290. doi:10.1021/ac051632c.

Source: PubMed

3
订阅