Journal Home Online First Current Issue Archive For Authors Journal Information 中文版

Engineering >> 2022, Volume 15, Issue 8 doi: 10.1016/j.eng.2021.03.015

Reconstruction and Dynamics of the Human Intestinal Microbiome Observed In Situ

a CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
b University of Chinese Academy of Sciences, Beijing 100049, China
c Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
d Clinical Trial Center Office, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
e State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
f Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing 210011, China

# These authors contributed equally to this work.

Received: 2020-08-03 Revised: 2021-03-02 Accepted: 2022-03-16 Available online: 2021-05-06

Next Previous

Abstract

The human gut microbiome has primarily been studied through the use of fecal samples, a practice that has generated vital knowledge on the composition and functional capacities of gastrointestinal microbial communities. However, this reliance on fecal materials limits the investigation of microbial dynamics in other locations along the gastrointestinal tract (in situ), and the infrequent availability of fecal samples prevents analysis at finer temporal scales (e.g., hours). In our study, we utilized colonic transendoscopic enteral tubing, a technology originally developed for fecal microbiota transplantation, to sample the ileocecal microbiome twice daily; metagenomic and metatranscriptomic analyses were then conducted on these samples. A total of 43 ileocecal and 28 urine and fecal samples were collected from five healthy volunteers. The ileocecal and fecal microbiomes, as profiled in the five volunteers, were found to be similar in metagenomic profiling, yet their active genes (metatranscriptome) were found to be highly distinct. Both microbiomes were perturbed after laxative exposure; over time, they exhibited reduced dissimilarity to their pre-treatment state, thereby demonstrating resilience as an innate property of the gut microbiome, although they did not fully recover within our observation time window. Sampling of the ileocecal microbiome during the day and at night revealed the existence of diurnal rhythms in a series of bacterial species and functional pathways, particularly those related to short-chain fatty acid production, such as Propionibacterium acnes and coenzyme A biosynthesis II. Autocorrelation analysis and fluctuations decomposition further indicated the significant periodicity of the diurnal oscillations. Metabolomic profiling in the fecal and urine samples mirrored the perturbance and recovery in the gut microbiome, indicating the crucial contribution of the gut microbiome to many key metabolites involved in host health. This study provides novel insights into the human gut microbiome and its inner resilience and diurnal rhythms, as well as the potential consequences of these to the host.

SupplementaryMaterials

Figures

Fig. 1

Fig. 2

Fig. 3

Fig. 4

Fig. 5

Fig. 6

References

[ 1 ] Lynch JB, Hsiao EY. Microbiomes as sources of emergent host phenotypes. Science 2019;365(6460):1405–9. link1

[ 2 ] Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol 2016;14(1):20–32. link1

[ 3 ] Stearns JC, Lynch MDJ, Senadheera DB, Tenenbaum HC, Goldberg MB, Cvitkovitch DG, et al. Bacterial biogeography of the human digestive tract. Sci Rep 2011;1(1):170. link1

[ 4 ] Yasuda K, Oh K, Ren B, Tickle T, Franzosa E, Wachtman L, et al. Biogeography of the intestinal mucosal and lumenal microbiome in the rhesus macaque. Cell Host Microbe 2015;17(3):385–91. link1

[ 5 ] Zeng Y, Zeng D, Zhou Y, Niu L, Deng J, Li Y, et al. Microbial biogeography along the gastrointestinal tract of a red panda. Front Microbiol 2018;9:1411. link1

[ 6 ] Ahern PP, Maloy KJ. Understanding immune–microbiota interactions in the intestine. Immunology 2020;159(1):4–14. link1

[ 7 ] Manuzak JA, Zevin AS, Cheu R, Richardson B, Modesitt J, Hensley-McBain T, et al. Antibiotic-induced microbiome perturbations are associated with significant alterations to colonic mucosal immunity in rhesus macaques. Mucosal Immunol 2020;13(3):471–80. link1

[ 8 ] Tropini C, Moss EL, Merrill BD, Ng KM, Higginbottom SK, Casavant EP, et al. Transient osmotic perturbation causes long-term alteration to the gut microbiota. Cell 2018;173(7):1742–54.e17. link1

[ 9 ] Wang Y, Kuang Z, Yu X, Ruhn KA, Kubo M, Hooper LV. The intestinal microbiota regulates body composition through NFIL3 and the circadian clock. Science 2017;357(6354):912–6. link1

[10] Kuang Z, Wang Y, Li Y, Ye C, Ruhn KA, Behrendt CL, et al. The intestinal microbiota programs diurnal rhythms in host metabolism through histone deacetylase 3. Science 2019;365(6460):1428–34. link1

[11] Gasparrini AJ, Crofts TS, Gibson MK, Tarr PI, Warner BB, Dantas G. Antibiotic perturbation of the preterm infant gut microbiome and resistome. Gut Microbes 2016;7(5):443–9. link1

[12] David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505(7484):559–63. link1

[13] Johnson AJ, Vangay P, Al-Ghalith GA, Hillmann BM, Ward TL, Shields-Cutler RR, et al. Daily sampling reveals personalized diet–microbiome associations in humans. Cell Host Microbe 2019;25(6):789–802.e5. link1

[14] Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334 (6052):105–8. link1

[15] Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, Goodman AL. Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature 2019;570(7762):462–7. link1

[16] Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 2018;555(7698):623–8. link1

[17] Haak BW, Lankelma JM, Hugenholtz F, Belzer C, de Vos WM, Wiersinga WJ. Long-term impact of oral vancomycin, ciprofloxacin and metronidazole on the gut microbiota in healthy humans. J Antimicrob Chemother 2019;74(3):782–6. link1

[18] Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. LifeLines cohort study. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 2016;352 (6285):565–9. link1

[19] Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, et al. Populationlevel analysis of gut microbiome variation. Science 2016;352(6285):560–4. link1

[20] Becattini S, Taur Y, Pamer EG. Antibiotic-induced changes in the intestinal microbiota and disease. Trends Mol Med 2016;22(6):458–78. link1

[21] Li SS, Zhu A, Benes V, Costea PI, Hercog R, Hildebrand F, et al. Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Science 2016;352(6285):586–9. link1

[22] Bharucha AE, Lacy BE. Mechanisms, evaluation, and management of chronic constipation. Gastroenterology 2020;158(5):1232–49.e3. link1

[23] Luthra P, Camilleri M, Burr NE, Quigley EMM, Black CJ, Ford AC. Efficacy of drugs in chronic idiopathic constipation: a systematic review and network meta-analysis. Lancet Gastroenterol Hepatol 2019;4(11):831–44. link1

[24] Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature 2012;489(7415):220–30. link1

[25] Relman DA. The human microbiome: ecosystem resilience and health. Nutr Rev 2012;70(Suppl 1):S2–9. link1

[26] Peng Z, Xiang J, He Z, Zhang T, Xu L, Cui B, et al. Colonic transendoscopic enteral tubing: a novel way of transplanting fecal microbiota. Endosc Int Open 2016;4(6):E610–3. link1

[27] Ding X, Li Q, Li P, Zhang T, Cui B, Ji G, et al. Long-term safety and efficacy of fecal microbiota transplant in active ulcerative colitis. Drug Saf 2019;42 (7):869–80. link1

[28] Fecal Microbiota Transplantation-standardization Study Group. Nanjing consensus on methodology of washed microbiota transplantation. Chin Med J 2020;133(19):2330–2. link1

[29] Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30(15):2114–20. link1

[30] Franzosa EA, McIver LJ, Rahnavard G, Thompson LR, Schirmer M, Weingart G, et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods 2018;15(11):962–8. link1

[31] Segata N, Waldron L, Ballarini A, Narasimhan V, Jousson O, Huttenhower C. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat Methods 2012;9(8):811–4. link1

[32] Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012;9(4):357–9. link1

[33] Dunn WB, Broadhurst D, Begley P, Zelena E, Francis-McIntyre S, Anderson N, et al. Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat Protoc 2011;6(7):1060–83. link1

[34] Hou W, Zhong D, Zhang P, Li Y, Lin M, Liu G, et al. A strategy for the targeted metabolomics analysis of 11 gut microbiota–host co-metabolites in rat serum, urine and feces by ultra high performance liquid chromatography-tandem mass spectrometry. J Chromatogr A 2016;1429:207–17. link1

[35] Rohart F, Gautier B, Singh A, Lê Cao KA, Schneidman D. mixOmics: an R package for ‘omics feature selection and multiple data integration. PLOS Comput Biol 2017;13(11):e1005752. link1

[36] Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 2003;13(11):2498–504. link1

[37] Mehta RS, Abu-Ali GS, Drew DA, Lloyd-Price J, Subramanian A, Lochhead P, et al. Stability of the human faecal microbiome in a cohort of adult men. Nat Microbiol 2018;3(3):347–55. link1

[38] Schirmer M, Franzosa EA, Lloyd-Price J, McIver LJ, Schwager R, Poon TW, et al. Dynamics of metatranscription in the inflammatory bowel disease gut microbiome. Nat Microbiol 2018;3(3):337–46. link1

[39] Chong J, Wishart DS, Xia J. Using MetaboAnalyst 4.0 for comprehensive and integrative metabolomics data analysis. Curr Protoc Bioinformatics 2019;68 (1):e86. link1

[40] Jin M, Kalainy S, Baskota N, Chiang D, Deehan EC, McDougall C, et al. Faecal microbiota from patients with cirrhosis has a low capacity to ferment nondigestible carbohydrates into short-chain fatty acids. Liver Int 2019;39 (8):1437–47. link1

[41] Vital M, Howe A, Bergeron N, Krauss RM, Jansson JK, Tiedje JM. Metagenomic insights into the degradation of resistant starch by human gut microbiota. Appl Environ Microbiol 2018;84(23):e01562–18. link1

[42] Takahashi K, Nishida A, Fujimoto T, Fujii M, Shioya M, Imaeda H, et al. Reduced abundance of butyrate-producing bacteria species in the fecal microbial community in Crohn’s disease. Digestion 2016;93(1):59–65. link1

[43] Rosario D, Benfeitas R, Bidkhori G, Zhang C, Uhlen M, Shoaie S, et al. Understanding the representative gut microbiota dysbiosis in metformintreated type 2 diabetes patients using genome-scale metabolic modeling. Front Physiol 2018;9:775. link1

[44] Liu H, Han M, Li SC, Tan G, Sun S, Hu Z, et al. Resilience of human gut microbial communities for the long stay with multiple dietary shifts. Gut 2019;68 (12):2254–5. link1

[45] Ng KM, Aranda-Díaz A, Tropini C, Frankel MR, Van Treuren W, O’Loughlin CT, et al. Recovery of the gut microbiota after antibiotics depends on host diet, community context, and environmental reservoirs. Cell Host Microbe 2019;26 (5):650–65.e4. link1

[46] Liu C, Zhang J, Li M, Zhao L, Ji C, Ma Q. Alterations and structural resilience of the gut microbiota under dietary fat perturbations. J Nutr Biochem 2018;61:91–100. link1

[47] MacPherson CW, Mathieu O, Tremblay J, Champagne J, Nantel A, Girard SA, et al. Gut bacterial microbiota and its resistome rapidly recover to basal state levels after short-term amoxicillin-clavulanic acid treatment in healthy adults. Sci Rep 2018;8(1):11192. link1

[48] Goethel A, Turpin W, Rouquier S, Zanello G, Robertson SJ, Streutker CJ, et al. Nod2 influences microbial resilience and susceptibility to colitis following antibiotic exposure. Mucosal Immunol 2019;12(3):720–32. link1

[49] Mortensen MS, Hebbelstrup Jensen B, Williams J, Brejnrod AD, O’Brien Andersen L, Röser D, et al. Stability and resilience of the intestinal microbiota in children in daycare—a 12 month cohort study. BMC Microbiol 2018;18(1):223. link1

[50] Louis P, Flint HJ. Diversity, metabolism and microbial ecology of butyrateproducing bacteria from the human large intestine. FEMS Microbiol Lett 2009;294(1):1–8. link1

[51] Tahara Y, Yamazaki M, Sukigara H, Motohashi H, Sasaki H, Miyakawa H, et al. Gut microbiota-derived short chain fatty acids induce circadian clock entrainment in mouse peripheral tissue. Sci Rep 2018;8(1):1395. link1

[52] Segers A, Desmet L, Thijs T, Verbeke K, Tack J, Depoortere I. The circadian clock regulates the diurnal levels of microbial short-chain fatty acids and their rhythmic effects on colon contractility in mice. Acta Physiol 2019;225(3): e13193. link1

[53] Fellows R, Denizot J, Stellato C, Cuomo A, Jain P, Stoyanova E, et al. Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases. Nat Commun 2018;9(1):105. link1

[54] Waldecker M, Kautenburger T, Daumann H, Busch C, Schrenk D. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J Nutr Biochem 2008;19 (9):587–93. link1

[55] Zhang T, Long C, Cui B, Buch H, Wen Q, Li Q, et al. Colonic transendoscopic tube-delivered enteral therapy (with video): a prospective study. BMC Gastroenterol 2020;20(1):135. link1

Related Research