PT - JOURNAL ARTICLE AU - Weil, Patrick Philipp AU - Reincke, Susanna AU - Hirsch, Christian Alexander AU - Giachero, Federica AU - Aydin, Malik AU - Scholz, Jonas AU - Jönsson, Franziska AU - Hagedorn, Claudia AU - Nguyen, Duc Ninh AU - Thymann, Thomas AU - Pembaur, Anton AU - Orth, Valerie AU - Wünsche, Victoria AU - Jiang, Ping-Ping AU - Wirth, Stefan AU - Jenke, Andreas C. W. AU - Sangild, Per Torp AU - Kreppel, Florian AU - Postberg, Jan TI - Uncovering the gastrointestinal passage, intestinal epithelial cellular uptake and AGO2 loading of milk miRNAs in neonates using xenomiRs as tracers AID - 10.1101/2021.08.24.21262525 DP - 2022 Jan 01 TA - medRxiv PG - 2021.08.24.21262525 4099 - http://medrxiv.org/content/early/2022/10/06/2021.08.24.21262525.short 4100 - http://medrxiv.org/content/early/2022/10/06/2021.08.24.21262525.full AB - Background Human breast milk has a high miRNA content in comparison to other body fluids. It remains unknown whether and how milk miRNAs might affect intestinal gene regulation and homeostasis of the developing microbiome after initiation of enteral nutrition. However, this requires that milk miRNAs survive gastrointestinal passage at relevant concentrations, are taken up by cells, and become available to the RNAi machinery. It seems important to dissect the fate of these miRNAs after oral ingestion and gastrointestinal passage.Objective Our goal was to analyze the potential transmissibility of milk miRNAs via the gastrointestinal system in neonate humans and a porcine model in vivo. Our data contribute to the discussion whether milk miRNAs could influence gene regulation in neonates and thus might vertically transmit developmental relevant signals.Design We performed cross-species profiling of miRNAs via deep-sequencing and utilized dietary xenobiotic taxon-specific milk miRNA (xenomiRs) as tracers in human and porcine neonates, followed by functional studies in primary human fetal intestinal epithelial cells (HIEC-6) using Ad5-mediated miRNA-gene transfer.Results We show that mammals have in common many milk miRNAs yet exhibit taxon-specific miRNA fingerprints. We traced intact bovine-specific miRNAs from formula-nutrition in human preterm stool and 9 days after the onset of enteral feeding in intestinal cells of preterm piglets. Thereafter, several xenomiRs were enriched in the intestinal cells. Moreover, few hours after introducing enteral feeding in preterm piglets with supplemented reporter miRNAs (cel-miR-39-5p/-3p), we observed enrichment of these in blood serum and in AGO2-immunocomplexes from intestinal biopsies.Conclusions Taken together, our data point to a transmissibility of milk miRNAs. Thus transmissibility might impact on intestinal maturation, since we demonstrated that vector-mediated miRNA-gene transfer into HIEC-6 revealed putative targets of the corresponding miRNAs. Results suggest that milk miRNAs could influence gene expression in intestinal epithelia of neonates under special conditions.Graphical abstractBreastfeeding is undoubtedly the best source of nutrition for neonates. Human breast milk has a high miRNA content in comparison to other body fluids. However, it is controversially discussed whether such milk miRNAs could impact on gene regulation in neonates. Of note, if true, this would could mean a vertical transmission of relevant developmental signals. In our present manuscript we contribute to solve this puzzle: Mammals have in common a significant number of milk miRNAs yet exhibit taxon-specific fingerprints. These differences allowed us to exploit foreign species’ (xenobiotic) milk miRNAs (xenomiRs) as tracers in human preterm neonates, who routinely received formula-supplemented nutrition in addition to breast milk. Tracer miRNAs survived the gastrointestinal (GI) transit in human preterm neonates. In a preterm piglet model the reporter miRNA did not only survive GI transit, but accumulated in the bloodstream few hours post-feeding. Moreover, we show that, cel-mir-39-5p and cel-miR-39-3p became loaded to AGO2 in intestinal tissue samples, which is prerequisite for milk microRNAs’ biological relevance. Adenovirus type 5-based miRNA-gene transfer into HIEC-6 allowed us to examine predicted bovine milk miRNA targets on the protein and transcriptome levels.Competing Interest StatementThe authors have declared no competing interest.Clinical TrialNot relevantFunding StatementThis study was funded by the HELIOS Research Centre (JP) and the NEOMUNE research program (PTS) of the Danish Research Councils. University of Copenhagen has filed a patent concerning the use of bovine colostrum for groups of paediatric patients. The authors have no other relevant affiliations or financial involvement with any organisation or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilised in the production of this manuscript.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:Ethics approval and consent to participate This study was conducted with approval of the Witten/Herdecke University Ethics board (No. 41/2018). All animal procedures were approved by the National Council on Animal Experimentation in Denmark (protocol number 2012-15-293400193). The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations. In addition, for investigations involving human subjects, informed consent has been obtained from all participants.I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesThe datasets generated and/or analysed during the current study are available in the NCBI BioProject repository as fastq.gz files of microRNA-seq and mRNA-seq (SubmissionIDs: SUB7584111/ SUB9897357/BioProject ID: PRJNA740153/URL: https://www.ncbi.nlm.nih.gov/bioproject/740153). https://www.ncbi.nlm.nih.gov/bioproject/740153 (Ad5)Adenovirus-type 5(GI passage)gastrointestinal passage(HIEC)human intestinal epithelial cells(NEC)necrotizing enterocolitis(PTGS)post-transcriptional gene silencing(RNAi)RNA interference(TGS)transcriptional gene silencing