Dietary Modulation of Gut Microbiota Contributes to Alleviation of Both Genetic and Simple Obesity in Children

Chenhong Zhang, Aihua Yin, Hongde Li, Ruirui Wang, Guojun Wu, Jian Shen, Menghui Zhang, Linghua Wang, Yaping Hou, Haimei Ouyang, Yan Zhang, Yinan Zheng, Jicheng Wang, Xiaofei Lv, Yulan Wang, Feng Zhang, Benhua Zeng, Wenxia Li, Feiyan Yan, Yufeng Zhao, Xiaoyan Pang, Xiaojun Zhang, Huaqing Fu, Feng Chen, Naisi Zhao, Bruce R Hamaker, Laura C Bridgewater, David Weinkove, Karine Clement, Joel Dore, Elaine Holmes, Huasheng Xiao, Guoping Zhao, Shengli Yang, Peer Bork, Jeremy K Nicholson, Hong Wei, Huiru Tang, Xiaozhuang Zhang, Liping Zhao, Chenhong Zhang, Aihua Yin, Hongde Li, Ruirui Wang, Guojun Wu, Jian Shen, Menghui Zhang, Linghua Wang, Yaping Hou, Haimei Ouyang, Yan Zhang, Yinan Zheng, Jicheng Wang, Xiaofei Lv, Yulan Wang, Feng Zhang, Benhua Zeng, Wenxia Li, Feiyan Yan, Yufeng Zhao, Xiaoyan Pang, Xiaojun Zhang, Huaqing Fu, Feng Chen, Naisi Zhao, Bruce R Hamaker, Laura C Bridgewater, David Weinkove, Karine Clement, Joel Dore, Elaine Holmes, Huasheng Xiao, Guoping Zhao, Shengli Yang, Peer Bork, Jeremy K Nicholson, Hong Wei, Huiru Tang, Xiaozhuang Zhang, Liping Zhao

Abstract

Gut microbiota has been implicated as a pivotal contributing factor in diet-related obesity; however, its role in development of disease phenotypes in human genetic obesity such as Prader-Willi syndrome (PWS) remains elusive. In this hospitalized intervention trial with PWS (n = 17) and simple obesity (n = 21) children, a diet rich in non-digestible carbohydrates induced significant weight loss and concomitant structural changes of the gut microbiota together with reduction of serum antigen load and alleviation of inflammation. Co-abundance network analysis of 161 prevalent bacterial draft genomes assembled directly from metagenomic datasets showed relative increase of functional genome groups for acetate production from carbohydrates fermentation. NMR-based metabolomic profiling of urine showed diet-induced overall changes of host metabotypes and identified significantly reduced trimethylamine N-oxide and indoxyl sulfate, host-bacteria co-metabolites known to induce metabolic deteriorations. Specific bacterial genomes that were correlated with urine levels of these detrimental co-metabolites were found to encode enzyme genes for production of their precursors by fermentation of choline or tryptophan in the gut. When transplanted into germ-free mice, the pre-intervention gut microbiota induced higher inflammation and larger adipocytes compared with the post-intervention microbiota from the same volunteer. Our multi-omics-based systems analysis indicates a significant etiological contribution of dysbiotic gut microbiota to both genetic and simple obesity in children, implicating a potentially effective target for alleviation.

Research in context: Poorly managed diet and genetic mutations are the two primary driving forces behind the devastating epidemic of obesity-related diseases. Lack of understanding of the molecular chain of causation between the driving forces and the disease endpoints retards progress in prevention and treatment of the diseases. We found that children genetically obese with Prader-Willi syndrome shared a similar dysbiosis in their gut microbiota with those having diet-related obesity. A diet rich in non-digestible but fermentable carbohydrates significantly promoted beneficial groups of bacteria and reduced toxin-producers, which contributes to the alleviation of metabolic deteriorations in obesity regardless of the primary driving forces.

Keywords: Genome interaction network; Gut microbiota; Metabolomics; Metagenomics; Obesity; Prader–Willi syndrome.

Figures

Fig. 1
Fig. 1
The flow diagram of participants.
Fig. 2
Fig. 2
Improved bioclinical parameters and inflammatory conditions after the intervention. (a) Anthropometric markers. (b) Hepatic function markers. (c) Plasma glucose homeostasis. (d) Plasma lipid homeostasis. (e) Inflammation related markers. Data are shown as mean ± s.e.m. Wilcoxon matched-pairs signed rank test (two-tailed) was used to analyze variation between each two-time points in PWS or SO children. *P < 0.05, **P < 0.01. For most of the bioclinical variables, PWS n = 17 and SO n = 21; For OGTT Glycaemia AUC and OGTT Insulinemia AUC, PWS n = 16 and SO n = 20; For CRP, W.B.C., SAA, AGP, Adiponectin and IL-6, PWS n = 16 and SO n = 19. BMI: body mass index; ALT: Alanine aminotransferase; AST: Aspartate aminotransferase; OGTT: Oral glucose tolerance test; LDL: low-density lipoprotein; CRP: C reactive protein; W.B.C.: White blood cell count; SAA: serum amyloid A protein; AGP: α-acid glycoprotein; LBP: Lipopolysaccharide binding protein.
Fig. 3
Fig. 3
Concordance of structural shifts of gut microbiota and the improvement of the host metabolic health. (a) PCoA based on Bray–Curtis distance of all the 376 bacterial CAGs during the dietary intervention. (b) Clustering of gut microbiota based on distances between different groups calculated with MANOVA test of first 23 PCs (accounting for 80% of total variations) of PCoA based on Bray–Curtis distance of all bacterial CAGs. (c) Genome interaction groups interaction network. Network plot highlights correlation relationships between 18 GIGs of 161 prevalent bacterial CAGs at all time points from the two cohorts. Node size indicates the average abundance of the species/strains. Lines between nodes represent correlations between the nodes they connect, with line width indicating the correlation magnitude, and red and blue colors indicating positive and negative correlations, respectively. For clarity, only lines corresponding to correlations whose magnitude is greater than 0.5 are drawn, and unconnected nodes are omitted. (d) Procrustes analysis combining PCoA of GIGs (end of lines with solid symbols) with PCA of bioclinical variables presented in Fig. 1 (end of lines without solid symbols). For PWS, n = 17 at Day 0, 30, 60, and 90; For SO, n = 21 at Day 0 and n = 20 at Day 30. (e) Group level abundance shifts of GIGs that changed significantly during dietary intervention. Data are mean ± s.e.m. Wilcoxon matched-pairs signed rank test (two-tailed) was used to analyze variation between each two-time points in PWS or SO children. *P < 0.05, **P < 0.01.
Fig. 4
Fig. 4
Functional shifts of the gut microbiome during the dietary intervention. (a) The PCA score plot of the KO Groups recognized with HUMAnN showing a significant shift of the KO profiles after the intervention (log-transformed). (b) Clustering of KO profiles based on distances between different groups calculated with MANOVA test of the first five PCs of PCA of KO. (c) Key pathways of gut microbiota responding to dietary intervention. The left histogram shows the LDA scores computed for features (on the pathway level) differentially abundant between all samples before and after the intervention. The heat map shows the abundance of the key pathways. The stacked bar chart shows relative contribution of GIGs to each pathway. For PWS, n = 17 at Day 0, 30, 60, and 90; For SO, n = 21 at Day 0 and n = 20 at Day 30. In (c) day 0 and day 30 data of PWS and SO combined together for this analysis.
Fig. 5
Fig. 5
Altered profiles of urinary metabolites during the dietary intervention. (a) PCA score plot of urinary metabolite profiles obtained from SO and PWS groups during the dietary intervention (left) and the metabolic trajectories generated from the PCA score plot. (b) Validated OPLS-DA coefficient plots showing the alterations of metabolic profiles in the urine caused by the 30-day intervention. The plot related to the discrimination between 1H NMR spectra of urine from Day 0 and Day 30 of SO (top) groups (n = 17, r > 0.468, P < 0.05). Plot related to the discrimination between 1H NMR spectra of urine from Day 0 and Day 30 of the PWS groups (n = 17, r > 0.468, P < 0.05). See Table S14 for the metabolite identification key. (c) Heat map showing significantly changed metabolites after the 30-day intervention in the SO cohort. (d) Heat map showing the significantly changed metabolites after 30-, 60- and 90-day interventions in the PWS cohort. The significance of each statistical comparison is shown in Fig. S31.
Fig. 6
Fig. 6
Changes of co-metabolism between host and gut microbiota. (a) Co-inertia analysis (CIA) of relationships between the metabolomics PCA (end of lines with empty symbol) and microbiota CAGs PCA (end of lines with solid symbol). (b) Bacterial CAGs significantly associated with the key metabolites modulated by the intervention. The bootstrapped Spearman correlation coefficient between the 161 prevalent bacterial CAGs and the key metabolites was more than 0.4 and FDR 

Fig. 7

Impaired metabolism of gnotobiotic mice…

Fig. 7

Impaired metabolism of gnotobiotic mice transplanted with pre-intervention gut microbiota from a PWS…

Fig. 7
Impaired metabolism of gnotobiotic mice transplanted with pre-intervention gut microbiota from a PWS patient. (a) Body weight curves of gnotobiotic mice receiving the fecal microbiota from GD58 before (Day 0, red) and after (Day 90, green) the intervention. For mice receiving pre-intervention microbiota, 1 to 14 days after transplantation, n = 10, and 15 to 28 days, n = 5; For mice receiving post-intervention microbiota, 1 to 14 days after transplantation, n = 9, and 15 to 28 days, n = 4. (b) Adiposity index (%Fat mass/body weight) of gnotobiotic mice at 2 and 4 weeks after fecal transplantation. (c) Hematoxylin- and eosin-stained sections of epididymal fat pads (100 × magnification). Cell area of adipocyte in epididymal fat pad is shown as mean ± s.e.m. (d) RT-qPCR analysis of expression of Tnfα, Tlr4 and Il6 in the liver, ileum and colon. All mRNA quantification data were normalized to the housekeeping gene Glyceraldehyde-3-phosphate dehydrogenase (Gapdh). Gene expression levels are normalized to that of mice 2 weeks after inoculation with pre-intervention microbiota. The median of the data in each group is shown. Student t-test (two-tailed, in (a), (b) and (c)) or Mann–Whitney U test (two-tailed, in (d),) was used to analyze variation between gnotobiotic mice receiving pre- and post-intervention microbiota. *P < 0.05, **P < 0.01. In (b) to (d), for mice receiving pre-intervention microbiota, n = 5; for mice receiving post-intervention microbiota, n = 4.
All figures (7)
Fig. 7
Fig. 7
Impaired metabolism of gnotobiotic mice transplanted with pre-intervention gut microbiota from a PWS patient. (a) Body weight curves of gnotobiotic mice receiving the fecal microbiota from GD58 before (Day 0, red) and after (Day 90, green) the intervention. For mice receiving pre-intervention microbiota, 1 to 14 days after transplantation, n = 10, and 15 to 28 days, n = 5; For mice receiving post-intervention microbiota, 1 to 14 days after transplantation, n = 9, and 15 to 28 days, n = 4. (b) Adiposity index (%Fat mass/body weight) of gnotobiotic mice at 2 and 4 weeks after fecal transplantation. (c) Hematoxylin- and eosin-stained sections of epididymal fat pads (100 × magnification). Cell area of adipocyte in epididymal fat pad is shown as mean ± s.e.m. (d) RT-qPCR analysis of expression of Tnfα, Tlr4 and Il6 in the liver, ileum and colon. All mRNA quantification data were normalized to the housekeeping gene Glyceraldehyde-3-phosphate dehydrogenase (Gapdh). Gene expression levels are normalized to that of mice 2 weeks after inoculation with pre-intervention microbiota. The median of the data in each group is shown. Student t-test (two-tailed, in (a), (b) and (c)) or Mann–Whitney U test (two-tailed, in (d),) was used to analyze variation between gnotobiotic mice receiving pre- and post-intervention microbiota. *P < 0.05, **P < 0.01. In (b) to (d), for mice receiving pre-intervention microbiota, n = 5; for mice receiving post-intervention microbiota, n = 4.

References

    1. Abubucker S., Segata N., Goll J. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol. 2012;8(6):e1002358.
    1. Askree S.H., Hjelm L.N., Pervaiz M.A. Allelic dropout can cause false-positive results for Prader–Willi and Angelman syndrome testing. J. Mol. Diagn. 2011;13(1):108–112.
    1. Backhed F., Ding H., Wang T. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. U. S. A. 2004;101(44):15718–15723.
    1. Backhed F., Manchester J.K., Semenkovich C.F., Gordon J.I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. U. S. A. 2007;104(3):979–984.
    1. Barreto F.C., Barreto D.V., Liabeuf S. Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin. J. Am. Soc. Nephrol. 2009;4(10):1551–1558.
    1. Bonfig W., Dokoupil K., Schmidt H. A special, strict, fat-reduced, and carbohydrate-modified diet leads to marked weight reduction even in overweight adolescents with Prader–Willi syndrome (PWS) TheScientificWorldJOURNAL. 2009;9:934–939.
    1. Butler M.G. Prader–Willi syndrome: obesity due to genomic imprinting. Curr. Genomics. 2011;12(3):204–215.
    1. Cani P.D., Bibiloni R., Knauf C. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–1481.
    1. Cantarel B.L., Lombard V., Henrissat B. Complex carbohydrate utilization by the healthy human microbiome. PLoS One. 2012;7(6):e28742.
    1. Claesson M.J., Jeffery I.B., Conde S. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488(7410):178–184.
    1. Cloarec O., Dumas M.E., Craig A. Statistical total correlation spectroscopy: an exploratory approach for latent biomarker identification from metabolic 1H NMR data sets. Anal. Chem. 2005;77(5):1282–1289.
    1. Cotillard A., Kennedy S.P., Kong L.C. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500(7464):585–588.
    1. Craciun S., Balskus E.P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl. Acad. Sci. U. S. A. 2012;109(52):21307–21312.
    1. David L.A., Maurice C.F., Carmody R.N. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–563.
    1. Deeley M.C., Yanofsky C. Transcription initiation at the tryptophanase promoter of Escherichia coli K-12. J. Bacteriol. 1982;151(2):942–951.
    1. Dodt M., Roehr J.T., Ahmed R., Dieterich C. FLEXBAR-flexible barcode and adapter processing for next-generation sequencing platforms. Biology. 2012;1(3):895–905.
    1. Dumas M.E., Barton R.H., Toye A. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc. Natl. Acad. Sci. U. S. A. 2006;103(33):12511–12516.
    1. Dykens E.M., Maxwell M.A., Pantino E., Kossler R., Roof E. Assessment of hyperphagia in Prader–Willi syndrome. Obesity (Silver Spring) 2007;15(7):1816–1826.
    1. Ellison A.M., Bank M.S., Clinton B.D. Loss of foundation species: consequences for the structure and dynamics of forested ecosystems. Front. Ecol. Environ. 2005;3(9):479–486.
    1. Eriksson L., Trygg J., Wold S. CV-ANOVA for significance testing of PLS and OPLS (R) models. J. Chemom. 2008;22(11–12):594–600.
    1. Fagan A., Culhane A.C., Higgins D.G. A multivariate analysis approach to the integration of proteomic and gene expression data. Proteomics. 2007;7(13):2162–2171.
    1. Fei N., Zhao L. An opportunistic pathogen isolated from the gut of an obese human causes obesity in germfree mice. ISME J. 2013;7(4):880–884.
    1. Finn R.D., Clements J., Eddy S.R. HMMER web server: interactive sequence similarity searching. Nucleic Acids Res. 2011;39(Web Server issue):W29–W37.
    1. Flint H.J., Scott K.P., Duncan S.H., Louis P., Forano E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 2012;3(4):289–306.
    1. Gibson G.R., Wang X. Regulatory effects of bifidobacteria on the growth of other colonic bacteria. J. Appl. Bacteriol. 1994;77(4):412–420.
    1. Godon J.J., Zumstein E., Dabert P., Habouzit F., Moletta R. Molecular microbial diversity of an anaerobic digestor as determined by small-subunit rDNA sequence analysis. Appl. Environ. Microbiol. 1997;63(7):2802–2813.
    1. Jiang L., Huang J., Wang Y., Tang H. Eliminating the dication-induced intersample chemical-shift variations for NMR-based biofluid metabonomic analysis. Analyst. 2012;137(18):4209–4219.
    1. Kanehisa M., Goto S., Sato Y., Furumichi M., Tanabe M. KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 2012;40(Database issue):D109–D114.
    1. Kubota T., Das S., Christian S.L., Baylin S.B., Herman J.G., Ledbetter D.H. Methylation-specific PCR simplifies imprinting analysis. Nat. Genet. 1997;16(1):16–17.
    1. Kurtz S., Phillippy A., Delcher A.L. Versatile and open software for comparing large genomes. Genome Biol. 2004;5(2):R12.
    1. Labruna G., Pasanisi F., Nardelli C. High leptin/adiponectin ratio and serum triglycerides are associated with an “at-risk” phenotype in young severely obese patients. Obesity. 2011;19(7):1492–1496.
    1. Lacroix D., Moutel S., Coupaye M. Metabolic and adipose-tissue signatures in adults with Prader–Willi syndrome, a model of extreme adiposity. J. Clin. Endocrinol. Metab. 2014:jc20143127.
    1. Langmead B., Salzberg S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods. 2012;9(4):357–359.
    1. Le Chatelier E., Nielsen T., Qin J. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500(7464):541–546.
    1. Ley R.E., Backhed F., Turnbaugh P., Lozupone C.A., Knight R.D., Gordon J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. U. S. A. 2005;102(31):11070–11075.
    1. Li W., Godzik A. Cd-hit: a fast program for clustering and comparing large sets of protein or nucleotide sequences. Bioinformatics. 2006;22(13):1658–1659.
    1. Li M., Wang B., Zhang M. Symbiotic gut microbes modulate human metabolic phenotypes. Proc. Natl. Acad. Sci. U. S. A. 2008;105(6):2117–2122.
    1. Li R., Yu C., Li Y. SOAP2: an improved ultrafast tool for short read alignment. Bioinformatics. 2009;25(15):1966–1967.
    1. Li H., Handsaker B., Wysoker A. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25(16):2078–2079.
    1. Mende D.R., Sunagawa S., Zeller G., Bork P. Accurate and universal delineation of prokaryotic species. Nat. Methods. 2013;10(9):881–884.
    1. Miller J.L., Lynn C.H., Shuster J., Driscoll D.J. A reduced-energy intake, well-balanced diet improves weight control in children with Prader–Willi syndrome. J. Hum. Nutr. Diet. 2013;26(1):2–9.
    1. Nielsen H.B., Almeida M., Juncker A.S. Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat. Biotechnol. 2014;32(8):822–828.
    1. Overbeek R., Olson R., Pusch G.D. The SEED and the rapid annotation of microbial genomes using subsystems technology (RAST) Nucleic Acids Res. 2014;42(D1):D206–D214.
    1. Papavramidis S.T., Kotidis E.V., Gamvros O. Prader–Willi syndrome-associated obesity treated by biliopancreatic diversion with duodenal switch. Case report and literature review. J. Pediatr. Surg. 2006;41(6):1153–1158.
    1. Peng Y., Leung H.C., Yiu S.M., Chin F.Y. IDBA-UD: a de novo assembler for single-cell and metagenomic sequencing data with highly uneven depth. Bioinformatics. 2012;28(11):1420–1428.
    1. Pokusaeva K., Fitzgerald G.F., van Sinderen D. Carbohydrate metabolism in Bifidobacteria. Genes Nutr. 2011;6(3):285–306.
    1. Prevey J.S., Germino M.J., Huntly N.J. Loss of foundation species increases population growth of exotic forbs in sagebrush steppe. Ecol. Appl. 2010;20(7):1890–1902.
    1. Quinlan A.R., Hall I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26(6):841–842.
    1. Ridaura V.K., Faith J.J., Rey F.E. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341(6150):1241214.
    1. Russell W.R., Gratz S.W., Duncan S.H. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am. J. Clin. Nutr. 2011;93(5):1062–1072.
    1. Scott K.P., Gratz S.W., Sheridan P.O., Flint H.J., Duncan S.H. The influence of diet on the gut microbiota. Pharmacol. Res. 2013;69(1):52–60.
    1. Segata N., Izard J., Waldron L. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60.
    1. Turnbaugh P.J., Ley R.E., Mahowald M.A., Magrini V., Mardis E.R., Gordon J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031.
    1. Turnbaugh P.J., Backhed F., Fulton L., Gordon J.I. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3(4):213–223.
    1. Vijay-Kumar M., Aitken J.D., Carvalho F.A. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. 2010;328(5975):228–231.
    1. Vrieze A., Van Nood E., Holleman F. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143(4):913–916. e7.
    1. Wang Z., Klipfell E., Bennett B.J. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63.
    1. Wang Z., Tang W.H., Buffa J.A. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur. Heart J. 2014;35(14):904–910.
    1. Wu J., An Y., Yao J., Wang Y., Tang H. An optimised sample preparation method for NMR-based faecal metabonomic analysis. Analyst. 2010;135(5):1023–1030.
    1. Wu G.D., Chen J., Hoffmann C. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–108.
    1. Xiao C., Hao F., Qin X., Wang Y., Tang H. An optimized buffer system for NMR-based urinary metabonomics with effective pH control, chemical shift consistency and dilution minimization. Analyst. 2009;134(5):916–925.
    1. Xiao S., Fei N., Pang X. A gut microbiota-targeted dietary intervention for amelioration of chronic inflammation underlying metabolic syndrome. FEMS Microbiol. Ecol. 2014;87(2):357–367.
    1. Xu Z., Hao B.L. CVTree update: a newly designed phylogenetic study platform using composition vectors and whole genomes. Nucleic Acids Res. 2009;37:W174–W178.
    1. Yang Y.X., Wang G.Y., C PX. Peking University Medical Press; Beijing: 2002. China Food Composition 2002.
    1. Zerbino D.R., Birney E. Velvet: algorithms for de novo short read assembly using de Bruijn graphs. Genome Res. 2008;18(5):821–829.
    1. Zhang C., Zhang M., Wang S. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J. 2010;4(2):232–241.
    1. Zhang C., Zhang M., Pang X., Zhao Y., Wang L., Zhao L. Structural resilience of the gut microbiota in adult mice under high-fat dietary perturbations. ISME J. 2012;6(10):1848–1857.
    1. Zhao L. The gut microbiota and obesity: from correlation to causality. Nat. Rev. Microbiol. 2013;11(9):639–647.
    1. Zhou Y., Gao H., Mihindukulasuriya K.A. Biogeography of the ecosystems of the healthy human body. Genome Biol. 2013;14(1):R1.
    1. Zhu W., Lomsadze A., Borodovsky M. Ab initio gene identification in metagenomic sequences. Nucleic Acids Res. 2010;38(12):e132.
    1. Zweigner J., Schumann R.R., Weber J.R. The role of lipopolysaccharide-binding protein in modulating the innate immune response. Microb. Inf./Instit. Pasteur. 2006;8(3):946–952.

Source: PubMed

3
Subskrybuj