Human Breast Milk miRNA, Maternal Probiotic Supplementation and Atopic Dermatitis in Offspring

Melanie Rae Simpson, Gaute Brede, Jostein Johansen, Roar Johnsen, Ola Storrø, Pål Sætrom, Torbjørn Øien, Melanie Rae Simpson, Gaute Brede, Jostein Johansen, Roar Johnsen, Ola Storrø, Pål Sætrom, Torbjørn Øien

Abstract

Background: Perinatal probiotic ingestion has been shown to prevent atopic dermatitis (AD) in infancy in a number of randomised trials. The Probiotics in the Prevention of Allergy among Children in Trondheim (ProPACT) trial involved a probiotic supplementation regime given solely to mothers in the perinatal period and demonstrated a ~40% relative risk reduction in the cumulative incidence of AD at 2 years of age. However, the mechanisms behind this effect are incompletely understood. Micro-RNAs (miRNA) are abundant in mammalian milk and may influence the developing gastrointestinal and immune systems of newborn infants. The objectives of this study were to describe the miRNA profile of human breast milk, and to investigate breast milk miRNAs as possible mediators of the observed preventative effect of probiotics.

Methods: Small RNA sequencing was conducted on samples collected 3 months postpartum from 54 women participating in the ProPACT trial. Differential expression of miRNA was assessed for the probiotic vs placebo and AD vs non-AD groups. The results were further analysed using functional prediction techniques.

Results: Human breast milk samples contain a relatively stable core group of highly expressed miRNAs, including miR-148a-3p, miR-22-3p, miR-30d-5p, let-7b-5p and miR-200a-3p. Functional analysis of these miRNAs revealed enrichment in a broad range of biological processes and molecular functions. Although several miRNAs were found to be differentially expressed on comparison of the probiotic vs placebo and AD vs non-AD groups, none had an acceptable false discovery rate and their biological significance in the development of AD is not immediately apparent from their predicted functional consequences.

Conclusion: Whilst breast milk miRNAs have the potential to be active in a diverse range of tissues and biological process, individual miRNAs in breast milk 3 months postpartum are unlikely to play a major role in the prevention of atopic dermatitis in infancy by probiotics ingestion in the perinatal period.

Trial registration: ClinicalTrials.gov NCT00159523.

Conflict of interest statement

Competing Interests: TØ, OS and MRS participated in seminars sponsored by Tine BA. All other authors declare that they have no conflict of interest. No authors received funds, sponsorship or other financial, professional or personal gain from Siemens Medical Solutions diagnostics AS. The stated competing interests do not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Patient flow and sample selection…
Fig 1. Patient flow and sample selection for ProPACT trial and miRNA sequencing project.
aMissing 1 or more of the following biological samples collected at 3 months post-partum: breast milk and stool samples from mother, blood and stool samples from the infant.
Fig 2. Overview of RNA profile and…
Fig 2. Overview of RNA profile and relative abundance of highly expressed miRNAs from breast milk samples.
(A) Bioanalyzer 6000 Nano gel from a representative sample showing abundant short RNAs up to 1000nt; (B) closer review of small RNAs using Agilent’s Small RNA kit demonstrating peaks at 22-23nt, 29-30nt, 33-34nt, 53-54nt, 90nt and approximately 140nt and 170nt; (C) bar graph demonstrating average percentage of small RNA sequences aligned to different RNA species with 95% confidence intervals; (D) proportion of reads accounted for by the top 5, 10, 20 and 125 miRNAs along with the number of predicted target genes (excluding repeated target prediction for alternate transcripts of the same gene); (E) boxplot of counts per million (CPM) mature miRNA of the 20 most abundant miRNAs in the 54 samples.

References

    1. Argov-Argaman N, Smilowitz JT, Bricarello DA, Barboza M, Lerno L, Froehlich JW, et al. Lactosomes: Structural and Compositional Classification of Unique Nanometer-Sized Protein Lipid Particles of Human Milk. J Agric Food Chem. 2010. 10.1021/jf102495s
    1. Agarwal S, Karmaus W, Davis S, Gangur V. Immune markers in breast milk and fetal and maternal body fluids: a systematic review of perinatal concentrations. J Hum Lact. 2011;27(2):171–86.
    1. Matheson MC, Allen KJ, Tang MLK. Understanding the evidence for and against the role of breastfeeding in allergy prevention. Clin Exp Allergy. 2012;42(6):827–51. 10.1111/j.1365-2222.2011.03925.x
    1. Patelarou E, Girvalaki C, Brokalaki H, Patelarou A, Androulaki Z, Vardavas C. Current evidence on the associations of breastfeeding, infant formula, and cow's milk introduction with type 1 diabetes mellitus: a systematic review. Nutr Rev. 2012;70(9):509–19. 10.1111/j.1753-4887.2012.00513.x
    1. Davis MK. Breastfeeding and chronic disease in childhood and adolescence. Pediatr Clin North Am. 2001;48(1):125–41, ix.
    1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33. 10.1016/j.cell.2009.01.002
    1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.
    1. Webber J, Baxter D, Zhang S, Huang D, Huang KH, Lee M, et al. The microRNA spectrum in 12 body fluids. Clin Chem. 2010;56(11):9.
    1. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. PNAS. 2011;108(12):6.
    1. Kosaka N, Izumi H, Sekine K, Ochiya T. microRNA as a new immune-regulatory agent in breast milk. Silence. 2010;1(1):7 10.1186/1758-907x-1-7
    1. Zhou Q, Li M, Wang X, Li Q, Wang T, Zhu Q, et al. Immune-related microRNAs are abundant in breast milk exosomes. Int J Biol Sci. 2012;8(1):118–23.
    1. Munch E, Harris R, Mohammad M, Benham A, Perjerrey S, Showalter L, et al. Transcriptome profiling of microRNA by next-gen deep sequencing reveals known and novel miRNA species in the lipid fraction of human breast milk. PLoS One. 2013;8(2):e50564 10.1371/journal.pone.0050564
    1. Gu Y, Li M, Wang T, Liang Y, Zhong Z, Wang X, et al. Lactation-Related MicroRNA Expression Profiles of Porcine Breast Milk Exosomes. PLoS One. 2012;7(8):e43691 10.1371/journal.pone.0043691
    1. Baier S, Nguyen C, Xie F, Wood J, Zempleni J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononclear cells, HEK-293 kidney cell cultures, and mouse livers. J Nutr. 2014; 10.3945/jn.114.196436
    1. Dotterud CK, Storro O, Johnsen R, Oien T. Probiotics in pregnant women to prevent allergic disease: a randomized, double-blind trial. Br J Dermatol. 2010;163(3):616–23. 10.1111/j.1365-2133.2010.09889.x
    1. Panduru M, Panduru NM, Salavastru CM, Tiplica GS. Probiotics and primary prevention of atopic dermatitis: a meta-analysis of randomized controlled studies. J Eur Acad Dermatol Venereol. 2015;29(2):232–42. 10.1111/jdv.12496
    1. Pelucchi C, Chatenoud L, Turati F, Galeone C, Moja L, Bach JF, et al. Probiotics supplementation during pregnancy or infancy for the prevention of atopic dermatitis: a meta-analysis. Epidemiology. 2012;23(3):402–14. 10.1097/EDE.0b013e31824d5da2
    1. Williams H, Burney P, Hay R, Archer C, Shipley M, Hunter J, et al. The U.K. working party's diagnostic criteria for atopic dermatitis. I. Derivation of a minimum set of descriminators for atopic dermatitis. Br J Dermatol. 1994;13(3):383–96.
    1. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal. 2011;17(1):10–2. 10.14806/ej.17.1.200
    1. Dobin A, Davis C, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast univeral RNA-seq aligner. Bioinformatics. 2013;29(1):15–2. 10.1093/bioinformatics/bts635
    1. Liao Y, Smyth G, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2013;30(7):923–30. 10.1093/bioinformatics/btt656
    1. R Development Core Team. R: A language and environment for statistical computing R Foundataion for Statistical Computing; Vienna, Austria: 2008.
    1. Law C, Chen Y, Shi W, Smyth G. Voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15:R29 10.1186/gb-2014-15-2-r29
    1. Ritchie M, Phipson B, Wu D, Hu Y, Law C, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acid Res. 2015;43 (10.1093/nar/gkv007)
    1. Smyth G. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Molec. Biol. 2004;3(1):.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. 1995;B57:289–300.
    1. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):92–105. 10.1101/gr.082701.108
    1. Garcia DM, Baek D, Shin C, Bell GW, Grimson A, Bartel DP. Weak seed-pairing stability and high target-site abundance decrease the proficiency of lsy-6 and other microRNAs. Nat Struct Mol Biol. 2011;18(10):1139–46. 10.1038/nsmb.2115
    1. Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP. MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell. 2007;27(1):91–105. 10.1016/j.molcel.2007.06.017
    1. Agarwal V, Bell G, Nam J, Bartel D. Predicting effective microRNA target sites in mammalian mRNAs. Predicting effective microRNA target sites in mammalian mRNAs. 2015;4:e05005 10.7554/eLife.05005#.dpuf
    1. Huang D, Sherman B, Lempicki R. Systematic and integrative analysis of large gene lists using DAVID Bioinformatics Resources Nat. Protoc. 2009;4(1):44–57. 10.1038/nprot.2008.211
    1. Huang D, Sherman B, Lempicki R. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acid Res. 2009;37(1):1–13. 10.1093/nar/gkn923
    1. Modepalli V, Kumar A, Hinds LA, Sharp JA, Nicholas KR, Lefevre C. Differential temporal expression of milk miRNA during the lactation cycle of the marsupial tammar wallaby (Macropus eugenii). BMC Genomics. 2014;12:1012
    1. Izumi H, Kosaka N, Shimizu T, Sekine K, Ochiya T, Takase M. Bovine milk contains microRNA and messenger RNA that are stable under degradative conditions. J Dairy Sci. 2012;95(9):4831–41. 10.3168/jds.2012-5489
    1. van Deun J, Mestdagh P, Sormunen R, Cocquyt V, Vermaelen K, Vandesompele J, et al. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J Extracell Vesicles. 2014;3:24858 - 10.3402/jev.v3.
    1. Zonneveld M, Brisson A, van Herwijnen M, Tan S, van de Lest C, Redegeld F, et al. Recovery of extracellular vesicles form human breast milk is influenced by sample collection and vesicle isolation procedures. J Extracell Vesicles. 2014;3:24215 - 10.3402/jev.v3.
    1. Anderson RC, Dalziel JE, Gopal PK, Bassett S, Ellis A, Roy NC. The role of intestinal barrier function in early life in the development of colitits In: Fukata D, editor. Colitis. Available from: In Tech; 2012.
    1. Power ML, Schulkin J. Maternal regulation of offspring development in mammals is an ancient adaptation tied to lactation. Appl. Transl. Genomics. 2013;130:55–63. 10.1016/j.atg.2013.06.001
    1. Wang J, Chen L, Li P, Li X, Zhou H, Wang F, et al. Gene expression is altered in piglet small intestine by weaning and dietary glutamine supplementation. J Nutr. 2008;138(6):1025–32.
    1. Hu CH, Xiao K, Luan ZS, Song J. Early weaning increases intestinal permeability, alters expression of cytokines and tight junction proteins, and activates mitogen-activated protein kinases in pigs. J Anim Sci. 2013;91:1094–101. 10.2527/jas.2012-5796
    1. Chapkin RS, Zhao C, Ivanov I, Davidson LA, Goldsby JS, Lupton JR, et al. Noninvasive stool-based detection of infant gastrointestinal development using gene expression profiles from exfoliated epithelial cells. Am. J. Physiol. 2010;298:G582–9. 10.1152/ajpgi.00004.2010
    1. Walker WA, Iyengar RS. Breast milk, microbiota, and intestinal immune homeostasis. Pediatr Res. 2015;77(1):220–8. 10.1038/pr.2014.160
    1. Nata T, Fujiya M, Ueno N, Moriichi K, Konishi H, Tanabe H, et al. MicroRNA-146b improves intestinal injury in mouse colitis by activating nuclear factor-kappaB and improving epithelial barrier function. J Gene Med. 2013;15(6–7):249–60. 10.1002/jgm.2717
    1. Biton M, Levin A, Slyper M, Alkalay I, Horwitz E, Mor H, et al. Epithelial microRNAs regulate gut mucosal immunity via epithelium-T cell crosstalk. Nat Immunol. 2011;12(3):239–46. 10.1038/ni.1994
    1. Chassin C, Hempel C, Stockinger S, Dupont A, Kubler JF, Wedemeyer J, et al. MicroRNA-146a-mediated downregulation of IRAK1 protects mouse and human small intestine against ischemia/reperfusion injury. EMBO Mol Med. 2012;4(12):1308–19. 10.1002/emmm.201201298
    1. Chen T, Xi QY, Ye RS, Cheng X, Qi QE, Wang SB, et al. Exploration of microRNAs in porcine milk exosomes. BMC Genomics. 2014;15:100 10.1186/1471-2164-15-100
    1. Chen X, Gao C, Li H, Huang L, Sun Q, Dong Y, et al. Identification and characterization of microRNAs in raw milk during different periods of lactation, commercial fluid, and powdered milk products. Cell Res. 2010;20(10):1128–37. 10.1038/cr.2010.80
    1. Izumi H, Kosaka N, Shimizu T, Sekine K, Ochiya T, Takase M. Time-dependent expression profiles of microRNAs and mRNAs in rat milk whey. PLoS One. 2014;9(2):e88843 10.1371/journal.pone.0088843
    1. Lu TX, Rothenberg ME. Diagnostic, functional, and therapeutic roles of microRNA in allergic diseases. Clinical reviews in allergy and immunology. 2013;132(1):3–13. 10.1016/j.jaci.2013.04.039
    1. Dhahbi JM, Spindler SR, Atamna H, Yamakawa A, Boffelli D, Mote P, et al. 5' tRNA halves are present as abundant complexes in serum, concentrated in blood cells, and modulated by aging and calorie restriction. BMC Genomics. 2013;14(298).

Source: PubMed

3
Subscribe