Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Maternal IgA protects against the development of necrotizing enterocolitis in preterm infants

Abstract

Neonates are protected from colonizing bacteria by antibodies secreted into maternal milk. Necrotizing enterocolitis (NEC) is a disease of neonatal preterm infants with high morbidity and mortality that is associated with intestinal inflammation driven by the microbiota1,2,3. The incidence of NEC is substantially lower in infants fed with maternal milk, although the mechanisms that underlie this benefit are not clear4,5,6. Here we show that maternal immunoglobulin A (IgA) is an important factor for protection against NEC. Analysis of IgA binding to fecal bacteria from preterm infants indicated that maternal milk was the predominant source of IgA in the first month of life and that a relative decrease in IgA-bound bacteria is associated with the development of NEC. Sequencing of IgA-bound and unbound bacteria revealed that before the onset of disease, NEC was associated with increasing domination by Enterobacteriaceae in the IgA-unbound fraction of the microbiota. Furthermore, we confirmed that IgA is critical for preventing NEC in a mouse model, in which pups that are reared by IgA-deficient mothers are susceptible to disease despite exposure to maternal milk. Our findings show that maternal IgA shapes the host–microbiota relationship of preterm neonates and that IgA in maternal milk is a critical and necessary factor for the prevention of NEC.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: IgA binding to the intestinal bacteria of preterm infants is positively correlated with maternal milk feeding and negatively correlated with the development of NEC.
Fig. 2: Reduced intestinal bacterial diversity driven by increased IgA-unbound Enterobacteriaceae precedes the development of NEC.
Fig. 3: IgA is a necessary component of breast milk for the prevention of the development of experimental NEC.

Similar content being viewed by others

Data availability

Patient-related data not included in the paper were generated as part of clinical trials and may be subject to patient confidentiality. The human study protocol was approved by the Institutional Review Board (protocol numbers PRO16030078 and PRO09110437) of the University of Pittsburgh. All raw and analyzed sequencing data have been deposited in Sequence Read Archive (accession number PRJNA526906).

Code availability

The algorithm for deconvolution of IgSeq data is available on GitHub (https://github.com/handlab/IgA_Seq_Deconvolution).

References

  1. Neu, J. & Walker, W. A. Necrotizing enterocolitis. N. Engl. J. Med. 364, 255–264 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hackam, D. & Caplan, M. Necrotizing enterocolitis: pathophysiology from a historical context. Semin. Pediatr. Surg. 27, 11–18 (2018).

  3. Hintz, S. R. et al. Neurodevelopmental and growth outcomes of extremely low birth weight infants after necrotizing enterocolitis. Pediatrics 115, 696–703 (2005).

    Article  PubMed  Google Scholar 

  4. Cortez, J. et al. Maternal milk feedings reduce sepsis, necrotizing enterocolitis and improve outcomes of premature infants. J. Perinatol. 38, 71–74 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Le Doare, K., Holder, B., Bassett, A. & Pannaraj, P. S. Mother’s milk: a purposeful contribution to the development of the infant microbiota and immunity. Front. Immunol. 9, 361 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lucas, A. & Cole, T. J. Breast milk and neonatal necrotising enterocolitis. Lancet 336, 1519–1523 (1990).

    Article  CAS  PubMed  Google Scholar 

  7. Pammi, M. et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: a systematic review and meta-analysis. Microbiome 5, 31 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Brower-Sinning, R. et al. Mucosa-associated bacterial diversity in necrotizing enterocolitis. PloS ONE 9, e105046 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Good, M. et al. The human milk oligosaccharide 2′-fucosyllactose attenuates the severity of experimental necrotising enterocolitis by enhancing mesenteric perfusion in the neonatal intestine. Br. J. Nutr. 116, 1175–1187 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wisgrill, L. et al. Human lactoferrin attenuates the proinflammatory response of neonatal monocyte‐derived macrophages. Clin. Exp. Immunol. 192, 315–324 (2018).

  11. He, Y.-M. et al. Transitory presence of myeloid-derived suppressor cells in neonates is critical for control of inflammation. Nat. Med. 24, 224–231 (2018).

  12. Brandtzaeg, P. The mucosal immune system and its integration with the mammary glands. J. Pediatr. 156, S8–S15 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Hapfelmeier, S. et al. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science 328, 1705–1709 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lindner, C. et al. Diversification of memory B cells drives the continuous adaptation of secretory antibodies to gut microbiota. Nat. Immunol. 16, 880–888 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Wilson, E. & Butcher, E. C. CCL28 controls immunoglobulin (Ig)A plasma cell accumulation in the lactating mammary gland and IgA antibody transfer to the neonate. J. Exp. Med. 200, 805–809 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Palm, N. W. et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Cullender, T. C. et al. Innate and adaptive immunity interact to quench microbiome flagellar motility in the gut. Cell Host Microbe 14, 571–581 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rognum, T. O., Thrane, P. S., Stoltenberg, L., Vege, Å. & Brandtzaeg, P. Development of intestinal mucosal immunity in fetal life and the first postnatal months. Pediatr. Res. 32, 145–148 (1992).

    Article  CAS  PubMed  Google Scholar 

  19. Bunker, J. J. et al. Innate and adaptive humoral responses coat distinct commensal bacteria with immunoglobulin A. Immunity 43, 541–553 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kubinak, J. L. et al. MyD88 signaling in T cells directs IgA-mediated control of the microbiota to promote health. Cell Host Microbe 17, 153–163 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yee, W. H. et al. Incidence and timing of presentation of necrotizing enterocolitis in preterm infants. Pediatrics 129, e298–e304 (2012).

    Article  PubMed  Google Scholar 

  22. Winter, S. E. et al. Host-derived nitrate boosts growth of E. coli in the inflamed gut. Science 339, 708–711 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Vandeputte, D. et al. Quantitative microbiome profiling links gut community variation to microbial load. Nature 551, 507–511 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Olm, M. R. et al. Necrotizing enterocolitis is preceded by increased gut bacterial replication, Klebsiella, and fimbriae-encoding bacteria that may stimulate TLR4 receptors. Preprint at https://www.biorxiv.org/content/10.1101/558676v1 (2019).

  25. Barlow, B. et al. An experimental study of acute neonatal enterocolitis—the importance of breast milk. J. Pediatr. Surg. 9, 587–595 (1974).

    Article  CAS  PubMed  Google Scholar 

  26. Jilling, T. et al. The roles of bacteria and TLR4 in rat and murine models of necrotizing enterocolitis. J. Immunol. 177, 3273–3282 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Rogier, E. W. et al. Secretory antibodies in breast milk promote long-term intestinal homeostasis by regulating the gut microbiota and host gene expression. Proc. Natl Acad. Sci. USA 111, 3074–3079 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Harris, N. L. et al. Mechanisms of neonatal mucosal antibody protection. J. Immunol. 177, 6256–6262 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Planer, J. D. et al. Development of the gut microbiota and mucosal IgA responses in twins and gnotobiotic mice. Nature 534, 263–266 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mirpuri, J. et al. Proteobacteria-specific IgA regulates maturation of the intestinal microbiota. Gut Microbes 5, 28–39 (2014).

    Article  PubMed  Google Scholar 

  31. Slack, E. et al. Innate and adaptive immunity cooperate flexibly to maintain host–microbiota mutualism. Science 325, 617–620 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kawamoto, S. et al. Foxp3+ T cells regulate immunoglobulin A selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity 41, 152–165 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Byndloss, M. X. et al. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 357, 570–575 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Peterson, D. A., McNulty, N. P., Guruge, J. L. & Gordon, J. I. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2, 328–339 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Moor, K. et al. High-avidity IgA protects the intestine by enchaining growing bacteria. Nature 544, 498–502 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Gauger, E. J. et al. Role of motility and the flhDC operon in Escherichia coli MG1655 colonization of the mouse intestine. Infect. Immun. 75, 3315–3324 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Leatham, M. P. et al. Mouse intestine selects nonmotile flhDC mutants of Escherichia coli MG1655 with increased colonizing ability and better utilization of carbon sources. Infect. Immun. 73, 8039–8049 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Eibl, M. M., Wolf, H. M., Fürnkranz, H. & Rosenkranz, A. Prevention of necrotizing enterocolitis in low-birth-weight infants by IgA–IgG feeding. N. Engl. J. Med. 319, 1–7 (1988).

    Article  CAS  PubMed  Google Scholar 

  39. Foster, J. P., Seth, R. & Cole, M. J. Oral immunoglobulin for preventing necrotizing enterocolitis in preterm and low birth weight neonates. Cochrane Database Syst. Rev. 4, CD001816 (2016).

    PubMed  Google Scholar 

  40. Caporaso, J. G. et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 6, 1621–1624 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Caporaso, J. G. et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc. Natl Acad. Sci. USA 108, 4516–4522 (2011).

    Article  PubMed  Google Scholar 

  42. Edgar, R. C. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat. Methods 10, 996–998 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Patel, C. B., Shanker, R., Gupta, V. K. & Upadhyay, R. S. q-PCR based culture-independent enumeration and detection of Enterobacter: an emerging environmental human pathogen in riverine systems and potable water. Front. Microbiol. 7, 172 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Good, M. et al. Lactobacillus rhamnosus HN001 decreases the severity of necrotizing enterocolitis in neonatal mice and preterm piglets: evidence in mice for a role of TLR9. Am. J. Physiol. Gastrointest. Liver Physiol. 306, G1021–G1032 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Good, M. et al. Breast milk protects against the development of necrotizing enterocolitis through inhibition of Toll-like receptor 4 in the intestinal epithelium via activation of the epidermal growth factor receptor. Mucosal Immunol. 8, 1166–1179 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Radulescu, A. et al. Heparin-binding epidermal growth factor-like growth factor overexpression in transgenic mice increases resistance to necrotizing enterocolitis. J. Pediatr. Surg. 45, 1933–1939 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the University of Pittsburgh In Situ Hybridization Laboratory for the preparation of tissue slides and the University of Pittsburgh Division of Laboratory Animal Research for assistance with animal husbandry; M. Band, C. Wright, T. Akraiko and the Roy J. Carver Biotechnology Center DNA Sequencing Center at the University of Illinois for assistance with NextGen Sequencing. Igha−/− mice were provided by Y. Belkaid. We thank A. Poholek, L. Konnikova, S. Canna and the members of the Hand and Morowitz laboratories for discussion and critical reading of the manuscript, N. Palm for advice on IgSeq analysis and D. Kostka for advice on deconvolution of IgSeq samples. This project was supported in part by the UPMC Children’s Hospital of Pittsburgh. M.G. is supported by K08DK101608, R03DK111473 and R01DK118568 from the National Institutes of Health, March of Dimes Foundation grant no. 5-FY17-79, the Children’s Discovery Institute of Washington University and St Louis Children’s Hospital. M.J.M. and the collection of samples are supported by R01AI092531. T.W.H. is supported by the Richard King Mellon Foundation Institute for Pediatric Research.

Author information

Authors and Affiliations

Authors

Contributions

K.P.G., M.J.M. and T.W.H. designed all of the experiments; K.P.G., B.A.F., J.T.T., J.J. and C.M. performed all of the experiments; C.M. and M.G. assisted with the implementation of the mouse NEC model; microbiome analyses was carried out by K.P.G., B.R.M., M.B.R., A.H.P.B. and T.W.H.; IgSeq analyses and development of deconvolution techniques were performed by K.P.G., M.B.R. and B.R.M.; R.B., M.J.M., M.G. and C.M. collected the preterm infant fecal samples; data analyses and synthesis were performed by K.P.G., B.R.M., M.J.M. and T.W.H.; K.P.G., B.R.M., M.J.M. and T.W.H. wrote the manuscript.

Corresponding author

Correspondence to Timothy W. Hand.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Joao Monteiro was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended Data

Extended Data Fig. 1 Maternal milk-derived antibodies binding to intestinal bacteria from preterm infants.

a,b, Representative flow plots show binding of intestinal bacteria derived from preterm infants to IgM (a; n = 20) and IgG (b; n = 12) relative to IgA. Numbers in quadrants show the mean percentage ± s.d. of data collected from the cohort in Table 1. c, Percentage IgA staining of intestinal bacteria of a single preterm infant fed exclusively with formula.

Source Data

Extended Data Fig. 2 Fraction of intestinal bacteria bound by IgA in preterm infants.

a, Percentage IgA-bound bacteria from longitudinally collected fecal samples from preterm infants in our study. The dotted red lines indicate the date of NEC diagnosis. b, Percentage IgA-bound intestinal bacteria from prospectively collected samples of patients that will develop NEC (n = 10, 39 samples combined) and controls (n = 13, 59 samples combined). Samples from multiple patients are pooled and represented in four-day windows post-delivery to increase the number of samples available for each time window. Box and whisker plots depict the mean (line) the 25th and 75th percentiles (box) and range (whiskers) for each time window. Two-way ANOVA.

Source Data

Extended Data Fig. 3 Linear discriminant analysis of the microbiota of infants that will develop NEC and controls.

Linear discriminant effect size analysis comparing pooled samples from controls or infants that will develop NEC before the diagnosis of disease. OTUs that discriminate between patients that will progress to NEC (n = 10, 39 samples combined) and controls (n = 13, 59 samples combined) before disease onset are ranked by the effect size (represented by the linear discriminant analysis score).

Source Data

Extended Data Fig. 4 Deconvolution method to decrease the effect of contamination in IgSeq.

a, Representative staining of the abundance of IgA-bound bacteria before (unsorted) or after magnetic separation (IgA+ and IgA) measured by flow cytometry of all samples (NEC samples, n = 39; control samples, n = 59). b, Graphical depiction of our deconvolution methodology. In this schematic, size represents IgA binding (large, IgA+; small, IgA), which can be measured on intact cells, and colors represent two different taxa, which can only be measured by sequencing. c, Percentage of IgA+ bacteria (measured by flow cytometry of unsorted fecal samples) compared to percentage of 16S rRNA reads found in the IgA+ sample (IgA+/IgA+ + IgA). Each dot represents a paired sample derived from the same fecal sample (n = 140). d, The same analysis as in c was carried out on samples after deconvolution (n = 140). c,d, Data were analyzed by linear regression with Pearson’s correlation coefficient and contain samples (n = 42) that were excluded from our prospective NEC analysis as they were collected after the NEC diagnosis, after DOL 40 or complicated by non-NEC illness and/or treatment.

Source Data

Extended Data Fig. 5 Longitudinal analysis of the intestinal microbiota of preterm infants.

Stacked bar charts depict the relative abundances of OTUs (unsorted sample, deconvolved IgA+ and IgA fractions) at the family level, from all patients in our study, at all time points analyzed. Patients will develop NEC, n = 10, 39 samples combined; controls, n = 13, 59 samples combined.

Source Data

Extended Data Fig. 6 Ratio of IgA to IgA+ reads for low-abundance taxa.

ac, Ratio of reads (IgA/IgA+; log2-transformed values) from paired IgA+ and IgA samples shown for each analyzed DOL. a, Staphylococcaceae reads. b, Streptococcaceae reads. c, Enterococcaceae reads. For all graphs, each patient is shown in a different color and the R2 value is based on a linear regression and Pearson’s correlation coefficient. The number of samples used in each graph varies because instances in which the IgA+ and/or IgA samples had zero reads were excluded. a, NEC samples, n = 10, 30 samples combined; control samples, n = 13, 53 samples combined. b, NEC samples, n = 10, 19 samples combined; control samples, n = 13, 32 samples combined. c, NEC samples, n = 10, 30 samples combined; control samples, n = 13, 51 samples combined.

Source Data

Extended Data Fig. 7 Absolute number of bacteria and number of bacteria associated with the dominant taxa in preterm infants.

a, Total number of bacteria in each fecal sample determined by bead-based flow cytometry analysis. Black circles represent controls (n = 8, 43 samples combined); red triangles represent infants that went on to develop NEC (n = 5, 24 samples combined). b, Total number of Enterobacteriaceae in patients that will develop NEC (right) and controls (left). Semi-log non-linear regression. c, Total number of anaerobes in patients that will develop NEC (right) and controls (left). Semi-log non-linear regression.

Source Data

Extended Data Fig. 8 Enterobacter spp. is enriched in the IgA+ fraction of breast-fed mouse pups.

Magnetically sorted IgA+ (left) and IgA (right) samples from fecal samples of day 12 (day 5 of NEC protocol) pups (n = 4). Frequency of Enterobacter spp. in each sample is shown as measured by qPCR for Enterobacter spp. (23S rRNA expression) normalized to the relative number of bacteria in each sample (as measured by 16S rRNA expression). AU, arbitrary units. *P = 0.0379 by paired Student’s t-test.

Source Data

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gopalakrishna, K.P., Macadangdang, B.R., Rogers, M.B. et al. Maternal IgA protects against the development of necrotizing enterocolitis in preterm infants. Nat Med 25, 1110–1115 (2019). https://doi.org/10.1038/s41591-019-0480-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0480-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing