Naturalization of the microbiota developmental trajectory of Cesarean-born neonates after vaginal seeding

Se Jin Song, Jincheng Wang, Cameron Martino, Lingjing Jiang, Wesley K Thompson, Liat Shenhav, Daniel McDonald, Clarisse Marotz, Paul R Harris, Caroll D Hernandez, Nora Henderson, Elizabeth Ackley, Deanna Nardella, Charles Gillihan, Valentina Montacuti, William Schweizer, Melanie Jay, Joan Combellick, Haipeng Sun, Izaskun Garcia-Mantrana, Fernando Gil Raga, Maria Carmen Collado, Juana I Rivera-Viñas, Maribel Campos-Rivera, Jean F Ruiz-Calderon, Rob Knight, Maria Gloria Dominguez-Bello, Se Jin Song, Jincheng Wang, Cameron Martino, Lingjing Jiang, Wesley K Thompson, Liat Shenhav, Daniel McDonald, Clarisse Marotz, Paul R Harris, Caroll D Hernandez, Nora Henderson, Elizabeth Ackley, Deanna Nardella, Charles Gillihan, Valentina Montacuti, William Schweizer, Melanie Jay, Joan Combellick, Haipeng Sun, Izaskun Garcia-Mantrana, Fernando Gil Raga, Maria Carmen Collado, Juana I Rivera-Viñas, Maribel Campos-Rivera, Jean F Ruiz-Calderon, Rob Knight, Maria Gloria Dominguez-Bello

Abstract

Background: Early microbiota perturbations are associated with disorders that involve immunological underpinnings. Cesarean section (CS)-born babies show altered microbiota development in relation to babies born vaginally. Here we present the first statistically powered longitudinal study to determine the effect of restoring exposure to maternal vaginal fluids after CS birth.

Methods: Using 16S rRNA gene sequencing, we followed the microbial trajectories of multiple body sites in 177 babies over the first year of life; 98 were born vaginally, and 79 were born by CS, of whom 30 were swabbed with a maternal vaginal gauze right after birth.

Findings: Compositional tensor factorization analysis confirmed that microbiota trajectories of exposed CS-born babies aligned more closely with that of vaginally born babies. Interestingly, the majority of amplicon sequence variants from maternal vaginal microbiomes on the day of birth were shared with other maternal sites, in contrast to non-pregnant women from the Human Microbiome Project (HMP) study.

Conclusions: The results of this observational study prompt urgent randomized clinical trials to test whether microbial restoration reduces the increased disease risk associated with CS birth and the underlying mechanisms. It also provides evidence of the pluripotential nature of maternal vaginal fluids to provide pioneer bacterial colonizers for the newborn body sites. This is the first study showing long-term naturalization of the microbiota of CS-born infants by restoring microbial exposure at birth.

Funding: C&D, Emch Fund, CIFAR, Chilean CONICYT and SOCHIPE, Norwegian Institute of Public Health, Emerald Foundation, NIH, National Institute of Justice, Janssen.

Keywords: Translation to humans.

Conflict of interest statement

Declaration of interests New York University has a U.S. patent (10357521) on behalf of M.G.D.-B., related to methods for restoring the microbiota of newborns.

Copyright © 2021. Published by Elsevier Inc.

Figures

Figure 1.. Fecal microbiota development during the…
Figure 1.. Fecal microbiota development during the first year of life in babies discordant to birth mode/exposure.
(a) Compositional Tensor Factorization (CTF) first principal component (Y-axis) of infant samples over age in days (X-axis). (b) Convex hull volume (Y-axis) on the first three Principal Coordinates (unweighted UniFrac distances) in mothers (purple) and infants by birth mode or exposure (X-axis). Infants show highest volumes in Cesarean born and lowest in Vaginally born, with Cesarean-seeded babies showing intermediate volumes; all pairwise comparisons are significant using Mann-Whitney test with Bonferroni corrections at 0.05 level (Table S3). (c-e) Songbird differentials shown for day 2, 30, and 180 after birth; ternary plots of the inverse additive log-ratio transform (inverse-ALR) of Songbird differentials give the estimated probability of a microbe being observed in Cesarean (left-axes; red), Vaginal (bottom-axes; blue), or Cesarean-seeded (right-axes; green). The color of the dots depicts the seeding effectiveness, with yellow color indicating effectively seeded/suppressed and black indicating not effectively seeded. Below each triangle, bar plots of top and bottom 20% Songbird differentials summarized at genus-level taxa between Cesarean-seeded and Cesarean born babies; a positive value indicates higher association with the Cesarean-seeded group, a negative value indicates higher with Cesarean. Bars are colored by the ASVs’ seeding effectiveness. The majority of taxa discordant overrepresented in the Cesarean-seeded group over the Cesarean group are yellow-orange, indicating ASVs effectively seeded in the Cesarean seeded group, and these are observed at all ages. See alsoFigure S4, Supplementary Methods S2–S7.
Figure 2.. Oral microbiota development during the…
Figure 2.. Oral microbiota development during the first year of life in babies discordant to birth mode/exposure.
(a) Compositional Tensor Factorization (CTF) first principal component (Y-axis) of infant samples over age in days (X-axis). (b) Convex hull volume (Y-axis) on the first three Principal Coordinates (unweighted UniFrac distances) in mothers (purple) and infants by birth mode or exposure (X-axis). Infants show highest volumes in Cesarean born and lowest in Vaginally born, with Cesarean-seeded babies showing intermediate volumes; all pairwise comparisons are significant using Mann-Whitney test with Bonferroni corrections at 0.05 level (Table S3). (c-e) Songbird differentials shown for day 2, 30, and 180 after birth; ternary plots of the inverse additive log-ratio transform (inverse-ALR) of Songbird differentials give the estimated probability of a microbe being observed in Cesarean (left-axes; red), Vaginal (bottom-axes; blue), or Cesarean-seeded (right-axes; green). The color of the dots depicts the seeding effectiveness, with yellow color indicating effectively seeded/suppressed and black indicating not effectively seeded. Below each triangle, bar plots of top and bottom 20% Songbird differentials summarized at genus-level taxa between Cesarean-seeded and Cesarean born babies; a positive value indicates higher association with the Cesarean-seeded group, a negative value indicates higher with Cesarean. Bars are colored by the ASVs’ seeding effectiveness. The majority of taxa discordant overrepresented in the Cesarean-seeded group over the Cesarean group are yellow–orange, indicating ASVs effectively seeded in the Cesarean seeded group, and these are observed at all ages. See alsoFigure S4, Supplementary Methods S2–S7.
Figure 3.. Skin microbiota development during the…
Figure 3.. Skin microbiota development during the first year of life in babies discordant to birth mode/exposure.
(a) Compositional Tensor Factorization (CTF) first principal component (Y-axis) of infant samples over age in days (X-axis). (b) Convex hull volume (Y-axis) on the first three Principal Coordinates (unweighted UniFrac distances) in mothers (purple) and infants by birth mode or exposure (X-axis). Infants show highest volumes in Cesarean born and lowest in Vaginally born, with Cesarean-seeded babies showing intermediate volumes; all but one pairwise comparison are significant using Mann-Whitney test with Bonferroni corrections at 0.05 level (Table S3). (c-e) Songbird differentials shown for day 2, 30, and 180 after birth; ternary plots of the inverse additive log-ratio transform (inverse-ALR) of Songbird differentials give the estimated probability of a microbe being observed in Cesarean (left-axes; red), Vaginal (bottom-axes; blue), or Cesarean-seeded (right-axes; green). The color of the dots depicts the seeding effectiveness, with yellow color indicating effectively seeded/suppressed and black indicating not effectively seeded. Below each triangle, bar plots of top and bottom 20% Songbird differentials summarized at genus-level taxa between Cesarean-seeded and Cesarean born babies; a positive value indicates higher association with the Cesarean-seeded group, a negative value indicates higher with Cesarean. Bars are colored by the ASVs’ seeding effectiveness. The majority of taxa discordant overrepresented in the Cesarean-seeded group over the Cesarean group are yellow–orange, indicating ASVs effectively seeded in the Cesarean seeded group, and these are observed at all ages. See alsoFigure S4, Supplementary Methods S2–S7.
Figure 4.. Microbial source tracking of the…
Figure 4.. Microbial source tracking of the neonate microbiome (first month) through fast expectation-maximization microbial source tracking (FEAST).
Contributions (y-axes) of various maternal sources (rows) to the infant microbial community (columns) are estimated across age in days (x-axes) for the first month of life, in 15 mother-baby pairs. Error bars show 95% confidence interval of the mean calculated by bootstrapping; Dunn test based on Kruskal-Wallis were performed on each time points by each maternal source for each baby sink, significant differences are marked by different letters in each panel. The vaginal source -prominent in day “0” for oral and skin in babies exposed to vaginal fluids (vaginal and CS-seeded; panel e, f)- as not prominent later in any baby site. Baby site specific communities resemble the corresponding maternal site (panels a, h, o), consistent with specific site selection of bacteria. The maternal right areola appears as a source for baby oral bacteria (panel k), which likely means that baby oral bacteria is transmitted to the mother’s areola during lactation.
Figure 5.. Proportions of bacterial vaginal ASVs…
Figure 5.. Proportions of bacterial vaginal ASVs shared with other body sites in the mothers of the current study, at the day of delivery (a) and in non-pregnant women (b).
(a) V4 sequences from vaginal swabs and gauzes obtained from 97 parturient mothers form this study at the day of birth. Current study data were sequenced by Illumina HiSeq and processed by QIIME2 using the same pipeline as for the HMP data. (b) HMP V4 data from vaginal swabs obtained from 105 non-pregnant women; ASVs included in the analyses were present in at least 10% of the samples in the respective body site. Roche 454 V3V5 sequences were trimmed to obtain the V4 region. See alsoFigure S2 b–c, Supplementary Methods S13–S14.

References

    1. Gensollen T, Iyer SS, Kasper DL, and Blumberg RS (2016). How colonization by microbiota in early life shapes the immune system. Science (New York, N.Y.) 352, 539–544. 10.1126/science.aad9378.
    1. Al Nabhani Z, and Eberl G (2020). Imprinting of the immune system by the microbiota early in life. Mucosal Immunol 13, 183–189. 10.1038/s41385-020-0257-y.
    1. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, and Knight R (2010). Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A 107, 11971–11975. 10.1073/pnas.1002601107.
    1. Bokulich NA, Chung J, Battaglia T, Henderson N, Jay M, Li H, A DL, Wu F, Perez-Perez GI, Chen Y, et al. (2016). Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 8, 343ra382. 10.1126/scitranslmed.aad7121.
    1. Yassour M, Vatanen T, Siljander H, Hamalainen AM, Harkonen T, Ryhanen SJ, Franzosa EA, Vlamakis H, Huttenhower C, Gevers D, et al. (2016). Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci Transl Med 8, 343ra381. 10.1126/scitranslmed.aad0917.
    1. Shao Y, Forster SC, Tsaliki E, Vervier K, Strang A, Simpson N, Kumar N, Stares MD, Rodger A, Brocklehurst P, et al. (2019). Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature 574, 117–121. 10.1038/s41586-019-1560-1.
    1. Stewart CJ, Ajami NJ, O’Brien JL, Hutchinson DS, Smith DP, Wong MC, Ross MC, Lloyd RE, Doddapaneni H, Metcalf GA, et al. (2018). Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 562, 583–588. 10.1038/s41586-018-0617-x.
    1. Malamitsi-Puchner A, Protonotariou E, Boutsikou T, Makrakis E, Sarandakou A, and Creatsas G (2005). The influence of the mode of delivery on circulating cytokine concentrations in the perinatal period. Early Hum Dev 81, 387–392. 10.1016/j.earlhumdev.2004.10.017.
    1. Stokholm J, Blaser MJ, Thorsen J, Rasmussen MA, Waage J, Vinding RK, Schoos AM, Kunoe A, Fink NR, Chawes BL, et al. (2018). Maturation of the gut microbiome and risk of asthma in childhood. Nat Commun 9, 141. 10.1038/s41467-017-02573-2.
    1. Andersen V, Moller S, Jensen PB, Moller FT, and Green A (2020). Caesarean Delivery and Risk of Chronic Inflammatory Diseases (Inflammatory Bowel Disease, Rheumatoid Arthritis, Coeliac Disease, and Diabetes Mellitus): A Population Based Registry Study of 2,699,479 Births in Denmark During 1973–2016. Clin Epidemiol 12, 287–293. 10.2147/CLEP.S229056.
    1. Blustein J, Attina T, Liu M, Ryan AM, Cox LM, Blaser MJ, and Trasande L (2013). Association of caesarean delivery with child adiposity from age 6 weeks to 15 years. Int J Obes (Lond) 37, 900–906. 10.1038/ijo.2013.49.
    1. Ardic C, Usta O, Omar E, Yildiz C, and Memis E (2020). Caesarean delivery increases the risk of overweight or obesity in 2-year-old children. J Obstet Gynaecol, 1–6. 10.1080/01443615.2020.1803236.
    1. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, Kim SG, Li H, Gao Z, Mahana D, et al. (2014). Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 158, 705–721. 10.1016/j.cell.2014.05.052.
    1. Martinez KA 2nd, Devlin JC, Lacher CR, Yin Y, Cai Y, Wang J, and Dominguez-Bello MG (2017). Increased weight gain by C-section: Functional significance of the primordial microbiome. Sci Adv 3, eaao1874. 10.1126/sciadv.aao1874.
    1. Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, Glickman JN, Siebert R, Baron RM, Kasper DL, and Blumberg RS (2012). Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489–493. 10.1126/science.1219328.
    1. Livanos AE, Greiner TU, Vangay P, Pathmasiri W, Stewart D, McRitchie S, Li H, Chung J, Sohn J, Kim S, et al. (2016). Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice. Nat Microbiol 1, 16140. 10.1038/nmicrobiol.2016.140.
    1. Moya-Perez A, Luczynski P, Renes IB, Wang S, Borre Y, Anthony Ryan C, Knol J, Stanton C, Dinan TG, and Cryan JF (2017). Intervention strategies for cesarean section-induced alterations in the microbiota-gut-brain axis. Nutr Rev 75, 225–240. 10.1093/nutrit/nuw069.
    1. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M, Korecka A, Bakocevic N, Ng LG, Kundu P, et al. (2014). The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 6, 263ra158. 10.1126/scitranslmed.3009759.
    1. Dominguez-Bello MG, De Jesus-Laboy KM, Shen N, Cox LM, Amir A, Gonzalez A, Bokulich NA, Song SJ, Hoashi M, Rivera-Vinas JI, et al. (2016). Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat Med 22, 250–253. 10.1038/nm.4039.
    1. Korpela K, Helve O, Kolho KL, Saisto T, Skogberg K, Dikareva E, Stefanovic V, Salonen A, Andersson S, and de Vos WM (2020). Maternal Fecal Microbiota Transplantation in Cesarean-Born Infants Rapidly Restores Normal Gut Microbial Development: A Proof-of-Concept Study. Cell 183, 324–334 e325. 10.1016/j.cell.2020.08.047.
    1. Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, Jernberg C, Bjorksten B, Engstrand L, and Andersson AF (2014). Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut 63, 559–566. 10.1136/gutjnl-2012-303249.
    1. Jiang L, Zhong Y, Elrod C, Natarajan L, Knight R, and Thompson WK (2020). BayesTime: Bayesian Functional Principal Components for Sparse Longitudinal Data. Arxiv.
    1. Martino C, Shenhav L, Marotz CA, Armstrong G, McDonald D, Vázquez-Baeza Y, Morton JT, Jiang L, Dominguez-Bello MG, Swafford AD, et al. (2020). Context-aware dimensionality reduction deconvolutes gut microbial community dynamics. Nature Biotechnology, 1–4. 10.1038/s41587-020-0660-7.
    1. Gloor GB, Macklaim JM, Pawlowsky-Glahn V, and Egozcue JJ (2017). Microbiome Datasets Are Compositional: And This Is Not Optional. Front Microbiol 8, 2224. 10.3389/fmicb.2017.02224.
    1. Morton JT, Marotz C, Washburne A, Silverman J, Zaramela LS, Edlund A, Zengler K, and Knight R (2019). Establishing microbial composition measurement standards with reference frames. Nat Commun 10, 2719. 10.1038/s41467-019-10656-5.
    1. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, Kurilshikov A, Bonder MJ, Valles-Colomer M, Vandeputte D, et al. (2016). Population-level analysis of gut microbiome variation. Science 352, 560–564. 10.1126/science.aad3503.
    1. Halfvarson J, Brislawn CJ, Lamendella R, Vazquez-Baeza Y, Walters WA, Bramer LM, D’Amato M, Bonfiglio F, McDonald D, Gonzalez A, et al. (2017). Dynamics of the human gut microbiome in inflammatory bowel disease. Nat Microbiol 2, 17004. 10.1038/nmicrobiol.2017.4.
    1. Zaneveld JR, McMinds R, and Vega Thurber R (2017). Stress and stability: applying the Anna Karenina principle to animal microbiomes. Nat Microbiol 2, 17121. 10.1038/nmicrobiol.2017.121.
    1. Shenhav L, Thompson M, Joseph TA, Briscoe L, Furman O, Bogumil D, Mizrahi I, Pe’er I, and Halperin E (2019). FEAST: fast expectation-maximization for microbial source tracking. Nat Methods 16, 627–632. 10.1038/s41592-019-0431-x.
    1. Azad MB, Konya T, Maughan H, Guttman DS, Field CJ, Chari RS, Sears MR, Becker AB, Scott JA, Kozyrskyj AL, and Investigators CS (2013). Gut microbiota of healthy Canadian infants: profiles by mode of delivery and infant diet at 4 months. CMAJ 185, 385–394. 10.1503/cmaj.121189.
    1. Korpela K, Costea P, Coelho LP, Kandels-Lewis S, Willemsen G, Boomsma DI, Segata N, and Bork P (2018). Selective maternal seeding and environment shape the human gut microbiome. Genome Res 28, 561–568. 10.1101/gr.233940.117.
    1. Ferretti P, Pasolli E, Tett A, Asnicar F, Gorfer V, Fedi S, Armanini F, Truong DT, Manara S, Zolfo M, et al. (2018). Mother-to-Infant Microbial Transmission from Different Body Sites Shapes the Developing Infant Gut Microbiome. Cell Host Microbe 24, 133–145 e135. 10.1016/j.chom.2018.06.005.
    1. Helve O, Korpela K, Kolho K-L, Saisto T, Skogberg K, Dikareva E, Stefanovic V, Salonen A, de Vos WM, and Andersson S (2019). 2843. Maternal Fecal Transplantation to Infants Born by Cesarean Section: Safety and Feasibility. Open Forum Infect Dis 6, S68–S68. 10.1093/ofid/ofz359.148.
    1. Martinez I, Maldonado-Gomez MX, Gomes-Neto JC, Kittana H, Ding H, Schmaltz R, Joglekar P, Cardona RJ, Marsteller NL, Kembel SW, et al. (2018). Experimental evaluation of the importance of colonization history in early-life gut microbiota assembly. Elife 7. 10.7554/eLife.36521.
    1. Stout MJ, Zhou Y, Wylie KM, Tarr PI, Macones GA, and Tuuli MG (2017). Early pregnancy vaginal microbiome trends and preterm birth. Am J Obstet Gynecol 217, 356 e351–356 e318. 10.1016/j.ajog.2017.05.030.
    1. Rasmussen MA, Thorsen J, Dominguez-Bello MG, Blaser MJ, Mortensen MS, Brejnrod AD, Shah SA, Hjelmsø MH, Lehtimäki J, Trivedi U, et al. (2020). Ecological succession in the vaginal microbiota during pregnancy and birth. The ISME Journal 14, 2325–2335. 10.1038/s41396-020-0686-3.
    1. DiGiulio DB, Callahan BJ, McMurdie PJ, Costello EK, Lyell DJ, Robaczewska A, Sun CL, Goltsman DSA, Wong RJ, Shaw G, et al. (2015). Temporal and spatial variation of the human microbiota during pregnancy. Proceedings of the National Academy of Sciences 112, 11060. 10.1073/pnas.1502875112.
    1. Huh SY, Rifas-Shiman SL, Zera CA, Edwards JW, Oken E, Weiss ST, and Gillman MW (2012). Delivery by caesarean section and risk of obesity in preschool age children: a prospective cohort study. Arch Dis Child 97, 610–616. 10.1136/archdischild-2011-301141.
    1. Pistiner M, Gold DR, Abdulkerim H, Hoffman E, and Celedon JC (2008). Birth by cesarean section, allergic rhinitis, and allergic sensitization among children with a parental history of atopy. J Allergy Clin Immunol 122, 274–279. 10.1016/j.jaci.2008.05.007.
    1. Sevelsted A, Stokholm J, Bonnelykke K, and Bisgaard H (2015). Cesarean section and chronic immune disorders. Pediatrics 135, e92–98. 10.1542/peds.2014-0596.
    1. Thavagnanam S, Fleming J, Bromley A, Shields MD, and Cardwell CR (2008). A meta-analysis of the association between Caesarean section and childhood asthma. Clin Exp Allergy 38, 629–633. 10.1111/j.1365-2222.2007.02780.x.
    1. Mueller NT, Dominguez-Bello MG, Appel LJ, and Hourigan SK (2019). ‘Vaginal seeding’ after a caesarean section provides benefits to newborn children: FOR: Does exposing caesarean-delivered newborns to the vaginal microbiome affect their chronic disease risk? The critical need for trials of ‘vaginal seeding’ during caesarean section. BJOG. 10.1111/1471-0528.15979.
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, Owens SM, Betley J, Fraser L, Bauer M, et al. (2012). Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J 6, 1621–1624. 10.1038/ismej.2012.8.
    1. Thompson LR, Sanders JG, McDonald D, Amir A, Ladau J, Locey KJ, Prill RJ, Tripathi A, Gibbons SM, Ackermann G, et al. (2017). A communal catalogue reveals Earth’s multiscale microbial diversity. Nature 551, 457–463. 10.1038/nature24621.
    1. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, Alexander H, Alm EJ, Arumugam M, Asnicar F, et al. (2019). Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol 37, 852–857. 10.1038/s41587-019-0209-9.
    1. Amir A, McDonald D, Navas-Molina JA, Kopylova E, Morton JT, Zech Xu Z, Kightley EP, Thompson LR, Hyde ER, Gonzalez A, and Knight R (2017). Deblur Rapidly Resolves Single-Nucleotide Community Sequence Patterns. mSystems 2. 10.1128/mSystems.00191-16.
    1. Janssen S, McDonald D, Gonzalez A, Navas-Molina JA, Jiang L, Xu ZZ, Winker K, Kado DM, Orwoll E, Manary M, et al. (2018). Phylogenetic Placement of Exact Amplicon Sequences Improves Associations with Clinical Information. mSystems 3. 10.1128/mSystems.00021-18.
    1. McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ, Probst A, Andersen GL, Knight R, and Hugenholtz P (2012). An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J 6, 610–618. 10.1038/ismej.2011.139.
    1. Schiffer L, Azhar R, Shepherd L, Ramos M, Geistlinger L, Huttenhower C, Dowd JB, Segata N, and Waldron L (2019). HMP16SData: Efficient Access to the Human Microbiome Project Through Bioconductor. Am J Epidemiol 188, 1023–1026. 10.1093/aje/kwz006.
    1. James GM, Hastie TJ, and Sugar CA (2000). Principal component models for sparse functional data. Biometrika 87, 587–602. DOI 10.1093/biomet/87.3.587.
    1. Lozupone C, and Knight R (2005). UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol 71, 8228–8235. 10.1128/AEM.71.12.8228-8235.2005.
    1. Virtanen P, Gommers R, Oliphant TE, Haberland M, Reddy T, Cournapeau D, Burovski E, Peterson P, Weckesser W, Bright J, et al. (2020). SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat Methods 17, 261–272. 10.1038/s41592-019-0686-2.
    1. Anderson MJ (2017). Permutational Multivariate Analysis of Variance (PERMANOVA). In Wiley StatsRef: Statistics Reference Online, pp. 1–15. 10.1002/9781118445112.stat07841.
    1. Aitchison J (1983). Principal Component Analysis of Compositional Data. Biometrika 70, 57–65.
    1. Jari O, Blanchet FG, Michael F, Roeland K, Pierre L, Dan M, Peter RM, Hara RBO, Gavin LS, Peter S, et al. (2019). vegan: Community Ecology Package.
    1. Aitchison J (1982). The Statistical-Analysis of Compositional Data. J Roy Stat Soc B Met 44, 139–177.

Source: PubMed

3
Abonnere