Gut microbiota dysbiosis contributes to the development of hypertension

Jing Li, Fangqing Zhao, Yidan Wang, Junru Chen, Jie Tao, Gang Tian, Shouling Wu, Wenbin Liu, Qinghua Cui, Bin Geng, Weili Zhang, Ryan Weldon, Kelda Auguste, Lei Yang, Xiaoyan Liu, Li Chen, Xinchun Yang, Baoli Zhu, Jun Cai, Jing Li, Fangqing Zhao, Yidan Wang, Junru Chen, Jie Tao, Gang Tian, Shouling Wu, Wenbin Liu, Qinghua Cui, Bin Geng, Weili Zhang, Ryan Weldon, Kelda Auguste, Lei Yang, Xiaoyan Liu, Li Chen, Xinchun Yang, Baoli Zhu, Jun Cai

Abstract

Background: Recently, the potential role of gut microbiome in metabolic diseases has been revealed, especially in cardiovascular diseases. Hypertension is one of the most prevalent cardiovascular diseases worldwide, yet whether gut microbiota dysbiosis participates in the development of hypertension remains largely unknown. To investigate this issue, we carried out comprehensive metagenomic and metabolomic analyses in a cohort of 41 healthy controls, 56 subjects with pre-hypertension, 99 individuals with primary hypertension, and performed fecal microbiota transplantation from patients to germ-free mice.

Results: Compared to the healthy controls, we found dramatically decreased microbial richness and diversity, Prevotella-dominated gut enterotype, distinct metagenomic composition with reduced bacteria associated with healthy status and overgrowth of bacteria such as Prevotella and Klebsiella, and disease-linked microbial function in both pre-hypertensive and hypertensive populations. Unexpectedly, the microbiome characteristic in pre-hypertension group was quite similar to that in hypertension. The metabolism changes of host with pre-hypertension or hypertension were identified to be closely linked to gut microbiome dysbiosis. And a disease classifier based on microbiota and metabolites was constructed to discriminate pre-hypertensive and hypertensive individuals from controls accurately. Furthermore, by fecal transplantation from hypertensive human donors to germ-free mice, elevated blood pressure was observed to be transferrable through microbiota, and the direct influence of gut microbiota on blood pressure of the host was demonstrated.

Conclusions: Overall, our results describe a novel causal role of aberrant gut microbiota in contributing to the pathogenesis of hypertension. And the significance of early intervention for pre-hypertension was emphasized.

Keywords: Fecal transplant; Gut microbiota; Hypertension; Metabolism; Pre-hypertension.

Figures

Fig. 1
Fig. 1
Decreased diversity and shift of gut enterotypes in human adults with pHTN and HTN. a Rarefaction curves for gene number in control (n = 41), pHTN (n = 56), and HTN (n = 99) after 100 random sampling. The curve in each group is near smooth when the sequencing data are great enough with few new genes undetected. b, c Comparison of the microbial gene count and α diversity (as accessed by Shannon index) based on the genera profile in the three groups. C, control; P, pHTN; H, HTN. P = 0.024, C vs P; P = 0.04, C vs H; for gene count. P = 0.023, C vs P; P = 0.016, C vs H; for α diversity. P values are from Kruskal-Wallis test. d A total of 196 samples are clustered into enterotype 1 (blue) and enterotype 2 (red) by PCA of Jensen-Shannon divergence values at the genus level. The major contributor in the two enterotypes is Prevotella and Bacteroides, respectively. e Relative abundances of the top genera (Prevotella and Bacteroides) in each enterotype. P = 6.31e−31 and P = 2.09e−15, respectively; Wilcoxon rank sum test. f The percentage of control, pHTN and HTN samples distributed in two enterotypes. 26.83% normotensive controls, 48.21% pHTN, and 45.45% HTN are found in enterotype 1. P = 0.02, C vs P; P = 0.03, C vs H; Fisher’s exact test. Boxes represent the inter quartile ranges, the inside line or points represent the median, and circles are outliers
Fig. 2
Fig. 2
Genera strikingly different across groups. a Relative abundance of the top 44 most different genera across groups at the criteria of P value <0.1 by Wilcoxon rank sum test. C, control; P, pHTN; H, HTN. The abundance profiles are transformed into Z scores by subtracting the average abundance and dividing the standard deviation of all samples. Z score is negative (shown in blue) when the row abundance is lower than the mean. Genera at P value <0.01 are marked with dark green star, P value <0.05 with light green star, and P value ≥0.05 with gray circle. b The box plot shows the relative abundance of four genera enriched in pHTN and HTN patients, and 11 genera abundant in control. Genera are colored according to the phylum. Boxes represent the inter quartile ranges, lines inside the boxes denote medians, and circles are outliers
Fig. 3
Fig. 3
Comparative analysis of GM enrichment across groups based on CAGs. a CAGs are defined as a minimum of 50 linked genes, and the correlation network of CAGs differentially enriched in pHTN and the control group is performed by Spearman’s correlation based on the abundance. b The network of CAGs enriched in HTN is compared to controls. CAGs are colored according to the taxonomic assignment as labeled, and the node size is scaled with the number of genes within the CAG. Edges between nodes denote Spearman correlation >0.8 (red) or between 0.7 and 0.8 (gray)
Fig. 4
Fig. 4
Microbial gene functions annotation in pHTN and HTN. a PCA based on the relative abundance of KEGG orthology groups in 196 samples. Significant differences across groups are established at the first principal component (PC1) values, and shown in the box plots above. **P value <0.001, Wilcoxon rank sum test. b The average abundance of KEGG modules differentially enriched in control, pHTN, and HTN gut microbiome. Twenty nine modules enriched in control, and 11 modules overrepresented in both pHTN and HTN are shown in green and pink, respectively. The functional potential of KEGG modules are demonstrated on the right. c Heat map showing the abundance of 11 most significantly altered CAZy family in pHTN or HTN as compared to control
Fig. 5
Fig. 5
Aberrant metabolic patterns in pHTN and HTN. a PLS-DA score plots based on the metabolic profiles in serum samples from control, pHTN, and HTN group in ES+ and ES−. n = 30 for control, n = 31 for pHTN, and n = 63 for HTN. b Score scatter plots of OPLS-DA comparing the metabolic differences identify the separation between pHTN and control, HTN and control, respectively. c Metabolites significantly changed in pHTN or HTN as compared to control at VIP >1.5 and P value (t test) <0.05 are identified. Venn diagrams demonstrate the number of altered metabolites shared between pHTN (green) and HTN (red) by the overlap. d The relative amount of 26 endogenous compounds concurrently varied in both pHTN and HTN groups is transformed into Z scores in the heat map. There are six metabolites failed to be identified. e The relationship between 26 endogenous metabolites and the 44 top altered genera (Fig. 2a) in pHTN and HTN is estimated by Spearman’s correlation analysis. And those with low correlated (|r| <0.4) are not shown. Genera and metabolites are distinguished as abundant in control (green) or HTN (pink)
Fig. 6
Fig. 6
A classification to identify pHTN and HTN patients from controls. a, b Random forest models are constructed using explanatory variables of CAGs + species (red curve), CAGs + metabolites (green curve), metabolites (yellow curve), CAGs (blue curve), and species (purple curve). The AUC shows the classification of control versus HTN, or control versus pHTN as the numbers of variables increase. The CAGs + metabolites-based classification is more efficient as indicated by a higher AUC. c ROC of the random forest classifier using CAGs + metabolites based on the 1000 most important variables by ranking the variables by importance. AUC = 0.91 for control versus pHTN (n = 12, red curve), AUC = 0.89 for control versus HTN (n = 12, green curve), and AUC = 0.57 for pHTN versus HTN (n = 13, blue curve). d The top 50 different CAGs distinguish HTN from control based on the random forest model using explanatory variables of CAGs + metabolites. e The top 50 CAGs discriminate between pHTN and control using explanatory variables of CAGs + metabolites. The lengths of bar in the histogram represent mean decrease accuracy, which indicates the importance of the CAG for classification. The color denote the enrichment of CAG in control (blue), in HTN or pHTN (red) according to OR score
Fig. 7
Fig. 7
Post-transplanted intestinal microbial profiles and BP of recipient mice. a Schematic representation of fecal microbiota transplantation. GF mice (n = 5 for control, n = 10 for HTN) are orally inoculated with prepared fecal contents from two patients of HTN and one normotensive control, respectively. The gut microbial profiles are analyzed at 7 days, and BP is measured at 10 weeks post-transplantation. C, control; H, HTN. b Venn diagram comparing the shared genera number in gut microbiome of human donors (n = 1 for control, n = 2 for HTN) and recipient mice (n = 3 for control, n = 6 for HTN). c Shannon index of recipient mice at the genus level demonstrate significantly reduced α diversity in HTN group. P = 0.048 from t test. Boxes represent the inter quartile ranges, lines inside the boxes denote medians, and circles are outliers. d PCoA plots of human donors and recipient mice based on microbial genera separate HTN group from the controls. e Heat map comparing the abundance of altered genera between control and HTN mice. Red, more abundant; blue, less abundant. Genera present consistent trend with the metagenomic analysis are marked with green points, while inconsistent with gray points. f SBP, DBP, MBP, and HR of the recipient mice (n = 5 for control, n = 10 for HTN) are measured by tail-cuff method. Data are presented as mean ± s.e.m. P = 0.018, SBP; P = 0.019, DBP; P = 0.014, MBP; P = 0.11, HR; t test

References

    1. Karlsson FH, Fak F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, et al. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun. 2012;3:1245. doi: 10.1038/ncomms2266.
    1. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60. doi: 10.1038/nature11450.
    1. Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013;2:e01202. doi: 10.7554/eLife.01202.
    1. Qin N, Yang F, Li A, Prifti E, Chen Y, Shao L, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature. 2014;513:59–64. doi: 10.1038/nature13568.
    1. Zeller G, Tap J, Voigt AY, Sunagawa S, Kultima JR, Costea PI, et al. Potential of fecal microbiota for early-stage detection of colorectal cancer. Mol Syst Biol. 2014;10:766. doi: 10.15252/msb.20145645.
    1. Fei N, Zhao L. An opportunistic pathogen isolated from the gut of an obese human causes obesity in germfree mice. ISME J. 2013;7:880–884. doi: 10.1038/ismej.2012.153.
    1. Chang CJ, Lin CS, Lu CC, Martel J, Ko YF, Ojcius DM, et al. Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota. Nat Commun. 2015;6:7489. doi: 10.1038/ncomms8489.
    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 host's metabolism. Mol Psychiatry. 2016;21:786–796. doi: 10.1038/mp.2016.44.
    1. Schaubeck M, Clavel T, Calasan J, Lagkouvardos I, Haange SB, Jehmlich N, et al. Dysbiotic gut microbiota causes transmissible Crohn's disease-like ileitis independent of failure in antimicrobial defence. Gut. 2016;65:225–237. doi: 10.1136/gutjnl-2015-309333.
    1. Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut. 2013;62:1787–1794. doi: 10.1136/gutjnl-2012-303816.
    1. Llopis M, Cassard AM, Wrzosek L, Boschat L, Bruneau A, Ferrere G, et al. Intestinal microbiota contributes to individual susceptibility to alcoholic liver disease. Gut. 2016;65:830–839. doi: 10.1136/gutjnl-2015-310585.
    1. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–585. doi: 10.1038/nm.3145.
    1. Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163:1585–1595. doi: 10.1016/j.cell.2015.11.055.
    1. Hillege HL, Girbes AR, de Kam PJ, Boomsma F, de Zeeuw D, Charlesworth A, et al. Renal function, neurohormonal activation, and survival in patients with chronic heart failure. Circulation. 2000;102:203–210. doi: 10.1161/01.CIR.102.2.203.
    1. Pierdomenico SD, Di Nicola M, Esposito AL, Di Mascio R, Ballone E, Lapenna D, et al. Prognostic value of different indices of blood pressure variability in hypertensive patients. Am J Hypertens. 2009;22:842–847. doi: 10.1038/ajh.2009.103.
    1. He FJ, Li J, Macgregor GA. Effect of longer-term modest salt reduction on blood pressure. Cochrane Database Syst Rev. 2013, DOI: 10.1002/14651858.CD004937.pub2: CD004937.
    1. Kato N, Takeuchi F, Tabara Y, Kelly TN, Go MJ, Sim X, et al. Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet. 2011;43:531–538. doi: 10.1038/ng.834.
    1. Moghadamrad S, McCoy KD, Geuking MB, Sagesser H, Kirundi J, Macpherson AJ, et al. Attenuated portal hypertension in germ-free mice: function of bacterial flora on the development of mesenteric lymphatic and blood vessels. Hepatology. 2015;61:1685–1695. doi: 10.1002/hep.27698.
    1. Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, et al. Gut dysbiosis is linked to hypertension. Hypertension. 2015;65:1331–1340. doi: 10.1161/HYPERTENSIONAHA.115.05315.
    1. Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, et al. Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol Genomics. 2015;47:187–197. doi: 10.1152/physiolgenomics.00136.2014.
    1. Khalesi S, Sun J, Buys N, Jayasinghe R. Effect of probiotics on blood pressure: a systematic review and meta-analysis of randomized, controlled trials. Hypertension. 2014;64:897–903. doi: 10.1161/HYPERTENSIONAHA.114.03469.
    1. Gomez-Guzman M, Toral M, Romero M, Jimenez R, Galindo P, Sanchez M, et al. Antihypertensive effects of probiotics Lactobacillus strains in spontaneously hypertensive rats. Mol Nutr Food Res. 2015;59:2326–2336. doi: 10.1002/mnfr.201500290.
    1. Durgan DJ, Ganesh BP, Cope JL, Ajami NJ, Phillips SC, Petrosino JF, et al. Role of the gut microbiome in obstructive sleep apnea-induced hypertension. Hypertension. 2016;67:469–474.
    1. Weber MA, Schiffrin EL, White WB, Mann S, Lindholm LH, Kenerson JG, et al. Clinical practice guidelines for the management of hypertension in the community: a statement by the American Society of Hypertension and the International Society of Hypertension. J Clin Hypertens (Greenwich) 2014;16:14–26. doi: 10.1111/jch.12237.
    1. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528:262–266. doi: 10.1038/nature15766.
    1. Tanaka S, Yoshida M, Murakami Y, Ogiwara T, Shoji M, Kobayashi S, et al. The relationship of Prevotella intermedia, Prevotella nigrescens and Prevotella melaninogenica in the supragingival plaque of children, caries and oral malodor. J Clin Pediatr Dent. 2008;32:195–200. doi: 10.17796/jcpd.32.3.vp657177815618l1.
    1. Podschun R, Ullmann U. Klebsiella spp. as nosocomial pathogens: epidemiology, taxonomy, typing methods, and pathogenicity factors. Clin Microbiol Rev. 1998;11:589–603.
    1. Restuccia PA, Cunha BA. Klebsiella. Infect Control. 1984;5:343–347. doi: 10.1017/S0195941700060549.
    1. Wang X, Yue TL, Barone FC, White RF, Clark RK, Willette RN, et al. Discovery of adrenomedullin in rat ischemic cortex and evidence for its role in exacerbating focal brain ischemic damage. Proc Natl Acad Sci U S A. 1995;92:11480–11484. doi: 10.1073/pnas.92.25.11480.
    1. Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, et al. A decrease of the butyrate-producing species Roseburia hominis and Fecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63:1275–1283. doi: 10.1136/gutjnl-2013-304833.
    1. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux JJ, et al. Fecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008;105:16731–16736. doi: 10.1073/pnas.0804812105.
    1. Bryant MP, Small N. The anaerobic monotrichous butyric acid-producing curved rod-shaped bacteria of the rumen. J Bacteriol. 1956;72:16–21.
    1. Pinzone MR, Celesia BM, Di Rosa M, Cacopardo B, Nunnari G. Microbial translocation in chronic liver diseases. Int J Microbiol. 2012;2012:694629. doi: 10.1155/2012/694629.
    1. Greenblum S, Turnbaugh PJ, Borenstein E. Metagenomic systems biology of the human gut microbiome reveals topological shifts associated with obesity and inflammatory bowel disease. Proc Natl Acad Sci U S A. 2012;109:594–599. doi: 10.1073/pnas.1116053109.
    1. Nielsen HB, Almeida M, Juncker AS, Rasmussen S, Li J, Sunagawa S, et al. Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat Biotechnol. 2014;32:822–828. doi: 10.1038/nbt.2939.
    1. Aliyu SH, Marriott RK, Curran MD, Parmar S, Bentley N, Brown NM, et al. Real-time PCR investigation into the importance of Fusobacterium necrophorum as a cause of acute pharyngitis in general practice. J Med Microbiol. 2004;53:1029–1035. doi: 10.1099/jmm.0.45648-0.
    1. Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382–392. doi: 10.1016/j.chom.2014.02.005.
    1. Kanehisa M, Araki M, Goto S, Hattori M, Hirakawa M, Itoh M, et al. KEGG for linking genomes to life and the environment. Nucleic Acids Res. 2008;36:D480–484. doi: 10.1093/nar/gkm882.
    1. Feng Q, Liang S, Jia H, Stadlmayr A, Tang L, Lan Z, et al. Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nat Commun. 2015;6:6528. doi: 10.1038/ncomms7528.
    1. Zhang X, Zhang D, Jia H, Feng Q, Wang D, Liang D, et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med. 2015;21:895–905. doi: 10.1038/nm.3914.
    1. Tang XY, Zhu YQ, Wei B, Wang H. Expression and functional research of TLR4 in human colon carcinoma. Am J Med Sci. 2010;339:319–326. doi: 10.1097/MAJ.0b013e3181cef1b7.
    1. Cario E, Rosenberg IM, Brandwein SL, Beck PL, Reinecker HC, Podolsky DK. Lipopolysaccharide activates distinct signaling pathways in intestinal epithelial cell lines expressing Toll-like receptors. J Immunol. 2000;164:966–972. doi: 10.4049/jimmunol.164.2.966.
    1. Kumar S, Arya P, Mukherjee C, Singh BK, Singh N, Parmar VS, et al. Novel aromatic ester from Piper longum and its analogues inhibit expression of cell adhesion molecules on endothelial cells. Biochemistry. 2005;44:15944–15952. doi: 10.1021/bi050941u.
    1. Wang W, Zheng JP, Zhu SX, Guan WJ, Chen M, Zhong NS. Carbocisteine attenuates hydrogen peroxide-induced inflammatory injury in A549 cells via NF-kappaB and ERK1/2 MAPK pathways. Int Immunopharmacol. 2015;24:306–313. doi: 10.1016/j.intimp.2014.12.018.
    1. Lu Y, AJ, Wang G, Hao H, Huang Q, Yan B, et al. Gas chromatography/time-of-flight mass spectrometry based metabonomic approach to differentiating hypertension- and age-related metabolic variation in spontaneously hypertensive rats. Rapid Commun Mass Spectrom. 2008;22:2882–2888. doi: 10.1002/rcm.3670.
    1. De Deyn PP, Robitaille P, Vanasse M, Qureshi IA, Marescau B. Serum guanidino compound levels in uremic pediatric patients treated with hemodialysis or continuous cycle peritoneal dialysis. Correlations between nerve conduction velocities and altered guanidino compound concentrations. Nephron. 1995;69:411–417. doi: 10.1159/000188511.
    1. Qi Y, Aranda JM, Rodriguez V, Raizada MK, Pepine CJ. Impact of antibiotics on arterial blood pressure in a patient with resistant hypertension—a case report. Int J Cardiol. 2015;201:157–158. doi: 10.1016/j.ijcard.2015.07.078.
    1. Baez S, Gordon HA. Tone and reactivity of vascular smooth muscle in germfree rat mesentery. J Exp Med. 1971;134:846–856. doi: 10.1084/jem.134.4.846.
    1. Hofer U. Microbiome: anelloviridae go viral. Nat Rev Microbiol. 2014;12:4–5. doi: 10.1038/nrmicro3192.
    1. Kohn FR, Kung AH. Role of endotoxin in acute inflammation induced by gram-negative bacteria: specific inhibition of lipopolysaccharide-mediated responses with an amino-terminal fragment of bactericidal/permeability-increasing protein. Infect Immun. 1995;63:333–339.
    1. Cybulsky MI, Chan MK, Movat HZ. Acute inflammation and microthrombosis induced by endotoxin, interleukin-1, and tumor necrosis factor and their implication in gram-negative infection. Lab Invest. 1988;58:365–378.
    1. Karbach SH, Schönfelder T, Brandão I, Wilms E, Hörmann N, Jäckel S, et al. Gut microbiota promote angiotensin II-induced arterial hypertension and vascular dysfunction. J Am Heart Assoc. 2016;5(9). doi:10.1161/JAHA.116.003698.
    1. Brindle JT, Nicholson JK, Schofield PM, Grainger DJ, Holmes E. Application of chemometrics to 1H NMR spectroscopic data to investigate a relationship between human serum metabolic profiles and hypertension. Analyst. 2003;128:32–36. doi: 10.1039/b209155k.
    1. Lim SS, Vos T, Flaxman AD, Danaei G, Shibuya K, Adair-Rohani H, et al. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380:2224–2260. doi: 10.1016/S0140-6736(12)61766-8.
    1. Group SR, Wright JT, Jr, Williamson JD, Whelton PK, Snyder JK, Sink KM, et al. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med. 2015;373:2103–2116. doi: 10.1056/NEJMoa1511939.
    1. Li R, Yu C, Li Y, Lam TW, Yiu SM, Kristiansen K, et al. SOAP2: an improved ultrafast tool for short read alignment. Bioinformatics. 2009;25:1966–1967. doi: 10.1093/bioinformatics/btp336.
    1. Luo R, Liu B, Xie Y, Li Z, Huang W, Yuan J, et al. SOAPdenovo2: an empirically improved memory-efficient short-read de novo assembler. Gigascience. 2012;1:18. doi: 10.1186/2047-217X-1-18.
    1. Li W, Godzik A. Cd-hit: a fast program for clustering and comparing large sets of protein or nucleotide sequences. Bioinformatics. 2006;22:1658–1659. doi: 10.1093/bioinformatics/btl158.
    1. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65. doi: 10.1038/nature08821.
    1. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature. 2011;473:174–180. doi: 10.1038/nature09944.
    1. Buchfink B, Xie C, Huson DH. Fast and sensitive protein alignment using DIAMOND. Nat Methods. 2015;12:59–60. doi: 10.1038/nmeth.3176.
    1. Huson DH, Auch AF, Qi J, Schuster SC. MEGAN analysis of metagenomic data. Genome Res. 2007;17:377–386. doi: 10.1101/gr.5969107.
    1. Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17:690–703. doi: 10.1016/j.chom.2015.04.004.
    1. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 2013;10:996–998. doi: 10.1038/nmeth.2604.
    1. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol. 2006;72:5069–5072. doi: 10.1128/AEM.03006-05.

Source: PubMed

3
Abonner