Source of human milk (mother or donor) is more important than fortifier type (human or bovine) in shaping the preterm infant microbiome

Shreyas V Kumbhare, William-Diehl Jones, Sharla Fast, Christine Bonner, Geert 't Jong, Gary Van Domselaar, Morag Graham, Michael Narvey, Meghan B Azad, Shreyas V Kumbhare, William-Diehl Jones, Sharla Fast, Christine Bonner, Geert 't Jong, Gary Van Domselaar, Morag Graham, Michael Narvey, Meghan B Azad

Abstract

Milk fortifiers help meet the nutritional needs of preterm infants receiving their mother's own milk (MOM) or donor human milk. We conducted a randomized clinical trial (NCT03214822) in 30 very low birth weight premature neonates comparing bovine-derived human milk fortifier (BHMF) versus human-derived fortifier (H2MF). We found that fortifier type does not affect the overall microbiome, although H2MF infants were less often colonized by an unclassified member of Clostridiales Family XI. Secondary analyses show that MOM intake is strongly associated with weight gain and microbiota composition, including Bifidobacterium, Veillonella, and Propionibacterium enrichment. Finally, we show that while oxidative stress (urinary F2-isoprostanes) is not affected by fortifier type or MOM intake, fecal calprotectin is higher in H2MF infants and lower in those consuming more MOM. Overall, the source of human milk (mother versus donor) appears more important than the type of milk fortifier (human versus bovine) in shaping preterm infant gut microbiota.

Keywords: calprotectin; gut inflammation; gut microbiome; human donor milk; human milk fortifiers; mother’s own milk; oxidative stress; very low birth weight infants.

Conflict of interest statement

Declaration of interests M.B.A. has consulted for DSM Nutritional Products and serves on the Malaika Vx and Tiny Health scientific advisory boards. She has received honoraria for speaking at symposia sponsored by Medela, Prolacta Biosciences, and the Institute for Advancement of Breastfeeding and Lactation Education and has contributed without remuneration to online courses on breast milk and the infant microbiome produced by Microbiome Courses. S.V.K. is currently employed by Digbi Health (3T and AI Pvt. Ltd., India), a position taken up after concluding the research presented in this study. These entities had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the article; or decision to submit the article for publication.

Crown Copyright © 2022. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Flowchart and illustration of changes in gut bacterial diversity and community structure in VLBW infants over time (A) Participant flow chart: consolidated standards of reporting trials diagram. (B) Study design and sample collection time points (T1–T4). Infants born from 26 to 30 weeks were recruited into the study, received their assigned fortifier (BHMF or H2MF) until 33 weeks AGA, and were followed until 35 weeks AGA or hospital discharge. Therefore, the intervention (T1 to T3) and follow-up (T3 to T4) periods ranged in duration. (C) Alpha diversity indicating species diversity within samples (n = 30) across time. (D) Principal coordinate analysis based on Bray-Curtis dissimilarity (Jaccard distances in Figure S1 and phylum/genus level changes over time in Figures S2A and S2B) showing diversity between samples across four study time points (left) and with adjusted gestational age in days (right). T1, study day 0 (before fortification); T2, study day 7 (during fortification); T3, week 33 AGA (end of fortification); and T4, week 35 AGA (follow-up after fortification). Statistical significance: ∗∗∗p < 0.001 across all study time points (Kruskal-Wallis test) for alpha diversity or by permutational multivariate analysis of variance (PERMANOVA) for beta diversity and betadisper test (permutation test for homogeneity of multivariate dispersions; 99,999 permutations with strata by infant ID to account for repeated measures). The p values were adjusted for multiple comparisons using false discovery rate (FDR) correction. AGA, adjusted gestational age; BHMF, bovine-derived human milk fortifier; H2MF, human-derived human milk fortifier; VLBW, very low birth weight.
Figure 2
Figure 2
Impact of human milk fortifier type, bovine (BHMF, n = 14) or human (H2MF, n = 16), on gut bacterial composition in VLBW infants Bar plots depict the relative abundance of gut bacterial genera (left, individual infants; right, group means). T1, study day 0 (before fortification); T2, study day 7 (during fortification); T3, week 33 AGA (end of fortification); and T4, week 35 AGA (follow-up after fortification). AGA, adjusted gestational age; BHMF, bovine-derived human milk fortifier; H2MF, human-derived human milk fortifier; VLBW, very low birth weight. Figure S3 shows results at the phylum level. The prefix “Uncl_” indicates an unclassified genus of the particular bacterial family or order.
Figure 3
Figure 3
Impact of human milk fortifier type on the gut microbiome, gut inflammation, and oxidative stress in VLBW infants over time (A and B) Longitudinal trajectories of (A) Shannon index and (B) inverse Simpson index, indicating alpha (within-sample) diversity over time in infants receiving BHMF (n = 14) or H2MF (n = 16); p values are from trend comparison using splinectomeR (999 permutations). (C) PCoA depicts beta (between-sample) diversity based on Bray-Curtis dissimilarity (Jaccard distances in Figure S4A) at each time point; p values are from PERMANOVA (99,999 permutations). See Table S1A for betadisper test results (permutation test for homogeneity of multivariate dispersions). (D) Genera with >10% prevalence across all samples that differed in prevalence or relative abundance (centered log-ratio [CLR] transformed) between groups over time are shown here (complete data are shown in Tables S1B–S1D, and regression analysis is shown in Figure S5). For Erwinia, the distance between groups at T1 indicates a random difference at baseline. (E) Gut inflammation (fecal calprotectin) and oxidative stress (urinary F2-isoprostane). The p values are from Wilcoxon sum rank test (for boxplots) or using splinectomeR (for longitudinal trajectories, 999 permutations) or Fisher’s exact test (for prevalences) (∼p < 0.10, ∗p < 0.05, ∗∗p < 0.01). T1, study day 0 (before fortification); T2, study day 7 (during fortification); T3, week 33 AGA (end of fortification); and T4, week 35 AGA (follow-up after fortification). AGA, adjusted gestational age; BHMF, bovine-derived human milk fortifier; H2MF, human-derived human milk fortifier; PCoA, principal coordinate analysis; VLBW, very low birth weight.
Figure 4
Figure 4
Association of mother’s own milk (MOM) intake, fortifier type, and other factors with gut microbiome composition in VLBW infants (A) Microbiome variance is explained by various factors, modeled individually by EnvFit using Bray-Curtis dissimilarity. Fortifier type, BHMF versus H2MF; MOM group, high versus low intake; F2IsoP, F2-isoprostane levels; antibiotics, percentage of days on antibiotics until a particular study time point. FDR-adjusted p values are denoted by ∼p < 0.1, ∗p < 0.05, ∗∗p < 0.01. (B) PCoA depicting beta diversity between MOM groups (low-MOM versus high-MOM infants, see Figure S8 for details) over time, modeled from PERMANOVA (99,999 permutations) using the fortifier type and MOM group in a multivariable analysis. See Table S1A for betadisper test results (permutation test for homogeneity of multivariate dispersions). (C) Heatmap illustrating correlations between bacterial genera relative abundances (CLR transformed) and other continuous covariates at each time point (T1–T4). Dendrogram clustering was based on pairwise distances obtained from Spearman correlations, independently for each time point (horizontal axis). Correlations represented were not significant (p > 0.05, FDR corrected). Clusters 1 and 2 on the vertical axis depict the clustering of genera based on their correlations with covariates. MOM groups were determined based on % MOM volumes that represent the proportion of MOM used to prepare the feeds prior to fortification. T1, study day 0 (before fortification: BHMF, n = 14; H2MF, n = 16; low MOM, n = 10; high MOM, n = 20); T2, study day 7 (during fortification: BHMF, n = 14; H2MF, n = 16; low MOM, n = 8; high MOM, n = 22); T3, week 33 AGA (end of fortification: BHMF, n = 14; H2MF, n = 16; low MOM, n = 13; high MOM, n = 17); and T4, week 35 AGA (follow-up after fortification: BHMF, n = 14; H2MF, n = 16; low MOM, n = 14; high MOM, n = 16). BHMF, bovine-derived human milk fortifier; H2MF, human-derived human milk fortifier; PCoA, principal coordinate analysis; VLBW, very low birth weight; F2IsoP, F2-isoprostanes; AGA, adjusted gestational age; antibiotics, percentage of days on antibiotics until a specific study time point; TEV, total enteral volume. The prefix “Uncl_” indicates an unclassified genus of the named bacterial family or order.
Figure 5
Figure 5
Association of mother’s own milk (MOM) intake with microbiome trajectories, gut inflammation, and oxidative stress in VLBW infants over time (A) Trajectories of bacterial relative abundance (CLR transformed) and prevalence over time in infants with high versus low MOM intake (see Figure S8 for details). Genera with >10% prevalence across all samples that differed in prevalence or relative abundance between groups over time are shown here (complete data are shown in Tables S1E–S1G, and regression analyses are shown in Figure S10). (B) Gut inflammation (fecal calprotectin) and oxidative stress (urinary F2-isoprostanes) over time. The p values are from Wilcoxon sum rank test (for boxplots) or using splinectomeR (for longitudinal trajectories, 999 permutations) or Fisher’s exact test (for prevalences) (∼p < 0.10, ∗p < 0.05, ∗∗p < 0.01). MOM groups were determined based on % MOM volumes that represent the proportion of MOM used to prepare the feeds prior to fortification. T1, study day 0 (before fortification: low MOM, n = 10; high MOM, n = 20); T2, study day 7 (during fortification: low MOM, n = 8; high MOM, n = 22); T3, week 33 AGA (end of fortification: low MOM, n = 13; high MOM, n = 17); and T4, week 35 AGA (follow-up after fortification: low MOM, n = 14; high MOM, n = 16).

References

    1. Arslanoglu S., Boquien C.-Y., King C., Lamireau D., Tonetto P., Barnett D., Bertino E., Gaya A., Gebauer C., Grovslien A., et al. Fortification of human milk for preterm infants: update and recommendations of the European milk bank association (EMBA) working group on human milk fortification. Front. Pediatr. 2019;7:76. doi: 10.3389/fped.2019.00076.
    1. Dutta S., Singh B., Chessell L., Wilson J., Janes M., McDonald K., Shahid S., Gardner V.A., Hjartarson A., Purcha M., et al. Guidelines for feeding very low birth weight infants. Nutrients. 2015;7:423–442. doi: 10.3390/nu7010423.
    1. Gregory K. Update on nutrition for preterm and full-term infants. J. Obstet. Gynecol. Neonatal Nurs. 2005;34:98–108. doi: 10.1177/0884217504272805.
    1. Martin C.R., Ling P.-R., Blackburn G.L. Review of infant feeding: key features of breast milk and infant formula. Nutrients. 2016;8:279. doi: 10.3390/nu8050279.
    1. Neu J. Mother’s own milk: how does it differ from donor milk for the baby. Breastfeed. Med. 2019;14:S3–S4. doi: 10.1089/bfm.2019.0036.
    1. Asbury M.R., Butcher J., Copeland J.K., Unger S., Bando N., Comelli E.M., Forte V., Kiss A., LeMay-Nedjelski L., Sherman P.M., et al. Mothers of preterm infants have individualized breast milk microbiota that changes temporally based on maternal characteristics. Cell Host Microbe. 2020;28:669–682.e4. doi: 10.1016/j.chom.2020.08.001.
    1. Shulhan J., Dicken B., Hartling L., Larsen B.M. Current knowledge of necrotizing enterocolitis in preterm infants and the impact of different types of enteral nutrition products. Adv. Nutr. 2017;8:80–91. doi: 10.3945/an.116.013193.
    1. Friel J.K., Diehl-Jones B., Cockell K.A., Chiu A., Rabanni R., Davies S.S., Roberts L.J., 2nd Evidence of oxidative stress in relation to feeding type during early life in premature infants. Pediatr. Res. 2011;69:160–164. doi: 10.1203/PDR.0b013e3182042a07.
    1. Sullivan S., Schanler R.J., Kim J.H., Patel A.L., Trawöger R., Kiechl-Kohlendorfer U., Chan G.M., Blanco C.L., Abrams S., Cotten C.M., et al. An exclusively human milk-based diet is associated with a lower rate of necrotizing enterocolitis than a diet of human milk and bovine milk-based products. J. Pediatr. 2010;156:562–567.e1. doi: 10.1016/j.jpeds.2009.10.040.
    1. Collado M.C., Cernada M., Neu J., Pérez-Martínez G., Gormaz M., Vento M. Factors influencing gastrointestinal tract and microbiota immune interaction in preterm infants. Pediatr. Res. 2015;77:726–731. doi: 10.1038/pr.2015.54.
    1. Torrazza R.M., Ukhanova M., Wang X., Sharma R., Hudak M.L., Neu J., Mai V. Intestinal Microbial Ecology and Environmental Factors Affecting Necrotizing Enterocolitis. PLoS One. 2013;8:e83304. doi: 10.1371/journal.pone.0083304.
    1. Goulet O. Potential role of the intestinal microbiota in programming health and disease. Nutr. Rev. 2015;73:32–40. doi: 10.1093/nutrit/nuv039.
    1. Cristofalo E.A., Schanler R.J., Blanco C.L., Sullivan S., Trawoeger R., Kiechl-Kohlendorfer U., Dudell G., Rechtman D.J., Lee M.L., Lucas A., Abrams S. Randomized trial of exclusive human milk versus preterm formula diets in extremely premature infants. J. Pediatr. 2013;163:1592–1595.e1. doi: 10.1016/j.jpeds.2013.07.011.
    1. Abrams S.A., Schanler R.J., Lee M.L., Rechtman D.J., the Prolacta Study Group Greater mortality and morbidity in extremely preterm infants fed a diet containing cow milk protein products. Breastfeed. Med. 2014;9:281–285. doi: 10.1089/bfm.2014.0024.
    1. Chen A.-C., Chung M.-Y., Chang J.H., Lin H.-C. Pathogenesis implication for necrotizing enterocolitis prevention in preterm very-low-birth-weight infants. J. Pediatr. Gastroenterol. Nutr. 2014;58:7–11. doi: 10.1097/MPG.0b013e3182a7dc74.
    1. Ghandehari H., Lee M.L., Rechtman D.J., H2MF Study Group An exclusive human milk-based diet in extremely premature infants reduces the probability of remaining on total parenteral nutrition: a reanalysis of the data. BMC Res. Notes. 2012;5:188. doi: 10.1186/1756-0500-5-188.
    1. O'Connor D.L., Kiss A., Tomlinson C., Bando N., Bayliss A., Campbell D.M., Daneman A., Francis J., Kotsopoulos K., Shah P.S., et al. Nutrient enrichment of human milk with human and bovine milk-based fortifiers for infants born weighing <1250 g: a randomized clinical trial. Am. J. Clin. Nutr. 2018;108:108–116. doi: 10.1093/ajcn/nqy067.
    1. Meier P.P., Patel A.L., Bigger H.R., Chen Y., Johnson T.J., Rossman B., Engstrom J.L. In: Diet and Nutrition in Critical Care. Rajendram R., Preedy V.R., Patel V.B., editors. Springer New York; 2015. Human milk feedings in the neonatal intensive care unit; pp. 807–822.
    1. Meier P., Patel A., Esquerra-Zwiers A. Donor human milk update: evidence, mechanisms, and priorities for research and practice. J. Pediatr. 2017;180:15–21. doi: 10.1016/j.jpeds.2016.09.027.
    1. Juncker H.G., Ruhé E.J.M., Burchell G.L., van den Akker C.H.P., Korosi A., van Goudoever J.B., van Keulen B.J. The effect of pasteurization on the antioxidant properties of human milk: a literature review. Antioxidants. 2021;10:1737. doi: 10.3390/antiox10111737.
    1. Collado M.C., Santaella M., Mira-Pascual L., Martínez-Arias E., Khodayar-Pardo P., Ros G., Martínez-Costa C. Longitudinal study of cytokine expression, lipid profile and neuronal growth factors in human breast milk from term and preterm deliveries. Nutrients. 2015;7:8577–8591. doi: 10.3390/nu7105415.
    1. Zanella A., Silveira R.C., Roesch L.F.W., Corso A.L., Dobbler P.T., Mai V., Procianoy R.S. Influence of own mother’s milk and different proportions of formula on intestinal microbiota of very preterm newbornsPLoS One. 2019;14:e0217296. doi: 10.1371/journal.pone.0217296.
    1. Ford S.L., Lohmann P., Preidis G.A., Gordon P.S., O'Donnell A., Hagan J., Venkatachalam A., Balderas M., Luna R.A., Hair A.B. Improved feeding tolerance and growth are linked to increased gut microbial community diversity in very-low-birth-weight infants fed mother’s own milk compared with donor breast milk. Am. J. Clin. Nutr. 2019;109:1088–1097. doi: 10.1093/ajcn/nqz006.
    1. Piñeiro-Ramos J.D., Parra-Llorca A., Ten-Doménech I., Gormaz M., Ramón-Beltrán A., Cernada M., Quintás G., Collado M.C., Kuligowski J., Vento M. Effect of donor human milk on host-gut microbiota and metabolic interactions in preterm infants. Clin. Nutr. 2021;40:1296–1309. doi: 10.1016/j.clnu.2020.08.013.
    1. Chou J.H., Roumiantsev S., Singh R. PediTools electronic growth chart calculators: applications in clinical care, research, and quality improvement. J. Med. Internet Res. 2020;22:e16204. doi: 10.2196/16204.
    1. Moore R.E., Townsend S.D. Temporal development of the infant gut microbiome. Open Biol. 2019;9:190128. doi: 10.1098/rsob.190128.
    1. Forbes J.D., Azad M.B., Vehling L., Tun H.M., Konya T.B., Guttman D.S., Field C.J., Lefebvre D., Sears M.R., Becker A.B., et al. Association of exposure to formula in the hospital and subsequent infant feeding practices with gut microbiota and risk of overweight in the first year of life. JAMA Pediatr. 2018;172:e181161. doi: 10.1001/jamapediatrics.2018.1161.
    1. Borewicz K., Gu F., Saccenti E., Hechler C., Beijers R., de Weerth C., van Leeuwen S.S., Schols H.A., Smidt H. The association between breast milk oligosaccharides and faecal microbiota in healthy breastfed infants at two, six, and twelve weeks of age. Sci. Rep. 2020;10:4270. doi: 10.1038/s41598-020-61024-z.
    1. Martín-Sosa S., Martín M.J., García-Pardo L.A., Hueso P. Sialyloligosaccharides in human and bovine milk and in infant formulas: variations with the progression of lactation. J. Dairy Sci. 2003;86:52–59. doi: 10.3168/jds.S0022-0302(03)73583-8.
    1. Robinson R.C. Structures and metabolic properties of bovine milk oligosaccharides and their potential in the development of novel therapeutics. Front. Nutr. 2019;6:50. doi: 10.3389/fnut.2019.00050.
    1. Vincent C., Stephens D.A., Loo V.G., Edens T.J., Behr M.A., Dewar K., Manges A.R. Reductions in intestinal Clostridiales precede the development of nosocomial Clostridium difficile infection. Microbiome. 2013;28:18. doi: 10.1186/2049-2618-1-18.
    1. Vincent C., Miller M.A., Edens T.J., Mehrotra S., Dewar K., Manges A.R. Bloom and bust: intestinal microbiota dynamics in response to hospital exposures and Clostridium difficile colonization or infection. Microbiome. 2016;14:12. doi: 10.1186/s40168-016-0156-3.
    1. Centers for Disease Control and Prevention . National Center for Emerging and Zoonotic Infectious Diseases, Division of Healthcare Quality and Promotion; Atlanta, GA: 2018. Clostridioides Difficile in Neonatal Intensive Care Unit Patients: A Systematic Review Centers for Disease Control and Prevention.
    1. Mathew O.P., Bhatia J.S., Richardson C.J. An outbreak of Clostridium difficile necrotizing enterocolitis. Pediatrics. 1984;73:265–266.
    1. Han V.K., Sayed H., Chance G.W., Brabyn D.G., Shaheed W.A. An outbreak of Clostridium difficile necrotizing enterocolitis: a case for oral vancomycin therapy? Pediatrics. 1983;71:935–941.
    1. Parra-Llorca A., Gormaz M., Alcántara C., Cernada M., Nuñez-Ramiro A., Vento M., Collado M.C. Preterm gut microbiome depending on feeding type: significance of donor human milk. Front. Microbiol. 2018;9:1376. doi: 10.3389/fmicb.2018.01376.
    1. Wang Z., Neupane A., Vo R., White J., Wang X., Marzano S.-Y.L. Comparing gut microbiome in mothers’ own breast milk- and formula-fed moderate-late preterm infants. Front. Microbiol. 2020;11:891. doi: 10.3389/fmicb.2020.00891.
    1. De la Cochetiere M.F., Piloquet H., des Robert C., Darmaun D., Galmiche J.P., Roze J.C. Early intestinal bacterial colonization and necrotizing enterocolitis in premature infants: the putative role of Clostridium. Pediatr. Res. 2004;56:366–370. doi: 10.1203/01.PDR.0000134251.45878.D5.
    1. Smith B., Bodé S., Skov T.H., Mirsepasi H., Greisen G., Krogfelt K.A. Investigation of the early intestinal microflora in premature infants with/without necrotizing enterocolitis using two different methods. Pediatr. Res. 2012;71:115–120. doi: 10.1038/pr.2011.1.
    1. Ríos-Covián D., Ruas-Madiedo P., Margolles A., Gueimonde M., de Los Reyes-Gavilán C.G., Salazar N. Intestinal short chain fatty acids and their link with diet and human health. Front. Microbiol. 2016;7:185. doi: 10.3389/fmicb.2016.00185.
    1. Montjaux-Régis N., Cristini C., Arnaud C., Glorieux I., Vanpee M., Casper C. Improved growth of preterm infants receiving mother's own raw milk compared with pasteurized donor milk. Acta Paediatr. 2011;100:1548–1554. doi: 10.1111/j.1651-2227.2011.02389.x.
    1. Hård A.L., Nilsson A.K., Lund A.M., Hansen-Pupp I., Smith L.E.H., Hellström A. Review shows that donor milk does not promote the growth and development of preterm infants as well as maternal milk. Acta Paediatr. 2019;108:998–1007. doi: 10.1111/apa.14702.
    1. Rai D., Adelman A.S., Zhuang W., Rai G.P., Boettcher J., Lönnerdal B. Longitudinal changes in lactoferrin concentrations in human milk: a global systematic review. Crit. Rev. Food Sci. Nutr. 2014;54:1539–1547. doi: 10.1080/10408398.2011.642422.
    1. Twigger A.-J., Hepworth A.R., Lai C.T., Chetwynd E., Stuebe A.M., Blancafort P., Hartmann P.E., Geddes D.T., Kakulas F. Gene expression in breastmilk cells is associated with maternal and infant characteristics. Sci. Rep. 2015;5:12933. doi: 10.1038/srep12933.
    1. Cai C., Zhang Z., Morales M., Wang Y., Khafipour E., Friel J. Feeding practice influences gut microbiome composition in very low birth weight preterm infants and the association with oxidative stress: a prospective cohort study. Free Radic. Biol. Med. 2019;142:146–154. doi: 10.1016/j.freeradbiomed.2019.02.032.
    1. Pergialiotis V., Konstantopoulos P., Karampetsou N., Koutaki D., Gkioka E., Perrea D.N., Papantoniou N. Calprotectin levels in necrotizing enterocolitis: a systematic review of the literature. Inflamm. Res. 2016;65:847–852. doi: 10.1007/s00011-016-0963-9.
    1. Savino F., Castagno E., Calabrese R., Viola S., Oggero R., Miniero R. High faecal calprotectin levels in healthy, exclusively breast-fed infants. Neonatology. 2010;97:299–304. doi: 10.1159/000255161.
    1. Kaczmarczyk M., Löber U., Adamek K., Węgrzyn D., Skonieczna-Żydecka K., Malinowski D., Łoniewski I., Markó L., Ulas T., Forslund S.K., Łoniewska B. The gut microbiota is associated with the small intestinal paracellular permeability and the development of the immune system in healthy children during the first two years of life. J. Transl. Med. 2021;19:177. doi: 10.1186/s12967-021-02839-w.
    1. Groer M., Ashmeade T., Louis-Jacques A., Beckstead J., Ji M. Relationships of feeding and mother's own milk with fecal calprotectin levels in preterm infants. Breastfeed. Med. 2016;11:207–212. doi: 10.1089/bfm.2015.0115.
    1. McMurdie P.J., Holmes S. Phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8:e61217. doi: 10.1371/journal.pone.0061217.
    1. Leo Lahti, Sudarshan Shetty . 2017. Tools for Microbiome Analysis in R.
    1. Shields-Cutler R.R., Al-Ghalith G.A., Yassour M., Knights D. SplinectomeR enables group comparisons in longitudinal microbiome studies. Front. Microbiol. 2018;9:785. doi: 10.3389/fmicb.2018.00785.
    1. Gu Z., Eils R., Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32:2847–2849. doi: 10.1093/bioinformatics/btw313.
    1. Mallick H, McIver LJ, Rahnavard A, Ma S, Zhang Y, Nguyen LH, Tickle TL, Weingart G, Ren B, Schwager E, et al. Multivariable association discovery in population-scale meta-omics studies.Preprint at bioRxiv. 10.1101/2021.01.20.427420.
    1. Ritari J., Salojärvi J., Lahti L., de Vos W.M. Improved taxonomic assignment of human intestinal 16S rRNA sequences by a dedicated reference database. BMC Genom. 2015;16:1056. doi: 10.1186/s12864-015-2265-y.
    1. Callahan B.J., McMurdie P.J., Rosen M.J., Han A.W., Johnson A.J.A., Holmes S.P. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods. 2016;13:581–583. doi: 10.1038/nmeth.3869.
    1. Davis N.M., Proctor D.M., Holmes S.P., Relman D.A., Callahan B.J. Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. Microbiome. 2018;6:226. doi: 10.1186/s40168-018-0605-2.
    1. McMurtry V.E., Gupta R.W., Tran L., Blanchard E.E., 4th, Penn D., Taylor C.M., Ferris M.J. Bacterial diversity and Clostridia abundance decrease with increasing severity of necrotizing enterocolitis. Microbiome. 2015;3:11. doi: 10.1186/s40168-015-0075-8.
    1. Caporaso J.G., Lauber C.L., Walters W.A., Berg-Lyons D., Huntley J., Fierer N., Owens S.M., Betley J., Fraser L., Bauer M., et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 2012;6:1621–1624. doi: 10.1038/ismej.2012.8.
    1. Ma E., Ingram K.H., Milne G.L., Garvey W.T. F2-Isoprostanes reflect oxidative stress correlated with lean mass and bone density but not insulin resistance. J. Endocr. Soc. 2017;1:436–448. doi: 10.1210/js.2017-00006.
    1. Callahan B.J., Sankaran K., Fukuyama J.A., McMurdie P.J., Holmes S.P. Bioconductor Workflow for Microbiome Data Analysis: from raw reads to community analyses [version 2; peer review: 3 approved] F1000Res. 2016;5:1492. doi: 10.12688/f1000research.8986.2.
    1. Oksanen J., Blanchet F.G., Kindt R., Legendre P., O'Hara R.B. 2013. Vegan: Community Ecology Package.
    1. Fenton T.R., Kim J.H. A systematic review and meta-analysis to revise the Fenton growth chart for preterm infants. BMC Pediatr. 2013;13:59. doi: 10.1186/1471-2431-13-59.

Source: PubMed

3
Suscribir