Combined prenatal Lactobacillus reuteri and ω-3 supplementation synergistically modulates DNA methylation in neonatal T helper cells

Johanna Huoman, David Martínez-Enguita, Elin Olsson, Jan Ernerudh, Lennart Nilsson, Karel Duchén, Mika Gustafsson, Maria C Jenmalm, Johanna Huoman, David Martínez-Enguita, Elin Olsson, Jan Ernerudh, Lennart Nilsson, Karel Duchén, Mika Gustafsson, Maria C Jenmalm

Abstract

Background: Environmental exposures may alter DNA methylation patterns of T helper cells. As T helper cells are instrumental for allergy development, changes in methylation patterns may constitute a mechanism of action for allergy preventive interventions. While epigenetic effects of separate perinatal probiotic or ω-3 fatty acid supplementation have been studied previously, the combined treatment has not been assessed. We aimed to investigate epigenome-wide DNA methylation patterns from a sub-group of children in an on-going randomised double-blind placebo-controlled allergy prevention trial using pre- and postnatal combined Lactobacillus reuteri and ω-3 fatty acid treatment. To this end, > 866000 CpG sites (MethylationEPIC 850K array) in cord blood CD4+ T cells were examined in samples from all four study arms (double-treatment: n = 18, single treatments: probiotics n = 16, ω-3 n = 15, and double placebo: n = 14). Statistical and bioinformatic analyses identified treatment-associated differentially methylated CpGs and genes, which were used to identify putatively treatment-induced network modules. Pathway analyses inferred biological relevance, and comparisons were made to an independent allergy data set.

Results: Comparing the active treatments to the double placebo group, most differentially methylated CpGs and genes were hypermethylated, possibly suggesting induction of transcriptional inhibition. The double-treated group showed the largest number of differentially methylated CpGs, of which many were unique, suggesting synergy between interventions. Clusters within the double-treated network module consisted of immune-related pathways, including T cell receptor signalling, and antigen processing and presentation, with similar pathways revealed for the single-treatment modules. CpGs derived from differential methylation and network module analyses were enriched in an independent allergy data set, particularly in the double-treatment group, proposing treatment-induced DNA methylation changes as relevant for allergy development.

Conclusion: Prenatal L. reuteri and/or ω-3 fatty acid treatment results in hypermethylation and affects immune- and allergy-related pathways in neonatal T helper cells, with potentially synergistic effects between the interventions and relevance for allergic disease. Further studies need to address these findings on a transcriptional level, and whether the results associate to allergy development in the children. Understanding the role of DNA methylation in regulating effects of perinatal probiotic and ω-3 interventions may provide essential knowledge in the development of efficacious allergy preventive strategies. Trial registration ClinicalTrials.gov, ClinicalTrials.gov-ID: NCT01542970. Registered 27th of February 2012-Retrospectively registered, https://ichgcp.net/clinical-trials-registry/NCT01542970 .

Keywords: Allergy prevention; CD4+ T cells; Combined intervention; Cord blood; DNA methylation; Lactobacillus reuteri; MethylationEPIC 850K; Postnatal; Prenatal; ω-3 fatty acids.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Graphical representations of multidimensional scaling (MDS) analyses are shown in panels AC for the double active (Lω) and single active treatment groups (LP, Pω), respectively, in comparison to the double placebo group (PP). Red dots represent the active treatment group, blue dots the placebo group. In panels D-F, volcano plots illustrate the distribution of DMCs when comparing each of the active treatment groups to the double placebo group (PP). DMCs are defined as having an MMD of > 5% along with an FDR-corrected p value of < 0.1, and are depicted as red dots. DMC—differentially methylated CpG, MDS—multidimensional scaling, MMD—mean methylation difference, L—Lactobacillus reuteri, ω—ω-3 fatty acids, P—placebo
Fig. 2
Fig. 2
A graphic representation of overlapping DMGs between the different comparisons. The sets represent the different comparisons, and the intersectional sizes show the number of overlapping genes between the comparisons. Dark blue represents the double-treated group, light blue the L. reuteri single-treated group and coral blue the ω-3 single-treated group. DMGs—differentially methylated genes, L—Lactobacillus reuteri, ω—ω-3 fatty acids, P—placebo
Fig. 3
Fig. 3
Visualisation of the consensus module created with the DMGs from the differential methylation comparison between the double-treated L.reuteri/ω-3 (Lω) and the double placebo (PP) group. Nodes represent genes and connecting lines represent protein–protein interactions (STRING combined score > 0.7) within the network. Red nodes illustrate hypermethylated genes, blue nodes indicate hypomethylated genes and both colours denote mixed methylation patterns. Black lines enclose biologically relevant clusters identified by bioinformatic pathway enrichment analyses. Nodes marked with * are also present in an independent allergy data set (described later in the paper). DMGs—differentially methylated genes, L—Lactobacillus reuteri, ω—ω-3 fatty acids, P—placebo
Fig. 4
Fig. 4
Venn diagrams illustrating the degree of overlap and uniqueness for the obtained genes from the three network modules for the double active treatment (Lω vs. PP) and the single active treatment (LP vs. PP, Pω vs. PP) comparisons. In A the number of common genes from the overlap are displayed for each comparison is shown, with corresponding gene names denoted in italics, and selection of related pathways in bold. No relevant pathways were revealed for the shared genes of the LP vs. PP and Pω vs. PP comparisons. In B the number of genes that are unique for each comparison are displayed along with a selection of corresponding related pathways. No pathways of relevance were revealed for the unique genes from the Pω vs. PP comparison. L—Lactobacillus reuteri, ω—ω-3 fatty acids, P—placebo
Fig. 5
Fig. 5
River plots illustrating the methylation status for overlapping genes being shared between the double-treated Lω group and A the single-treated probiotic (LP vs. PP), and B ω-3 treated (Pω vs. PP) group. Panel C illustrates comparisons between the single treated groups (LP and Pω vs. PP) and D shows the comparison between all treatment groups. Genes are represented as flows of size proportional to number of genes within their methylation status in a particular set, and differences between sets are shown as connections between flows. The respective gene names are denoted next to each flow. Red represents hypermethylation, blue hypomethylation and green a mixed methylation pattern. The number of genes in the respective flows are denoted in the graph. L—Lactobacillus reuteri, ω—ω-3 fatty acids, P—placebo
Fig. 6
Fig. 6
Enrichment analyses relating DMCs from A. differential DNA methylation analyses and B. network analyses for all three comparisons, to DMCs from a study investigating DNA methylation patterns in adults with seasonal allergic rhinitis. Panel A shows treatment DMC overlaps with allergy DMCs. Panel B shows consensus module gene associated DMC overlaps with allergy DMCs. The number of overlapping DMCs is shown at the bottom of the bars (n). A one-sided Fisher’s exact test was used for the enrichment analyses. Computed p values represent the enrichment significance, while the fold enrichment illustrates how over-represented the overlap is, compared to what would be expected by chance for the given background

References

    1. Blaser MJ. The theory of disappearing microbiota and the epidemics of chronic diseases. Nat Rev Immunol. 2017;17:461–463. doi: 10.1038/nri.2017.77.
    1. Lambrecht BN, Hammad H. The immunology of the allergy epidemic and the hygiene hypothesis. Nat Immunol. 2017;18:1076–1083. doi: 10.1038/ni.3829.
    1. Simon KA, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci. 2015;282:20143085.
    1. Gollwitzer ES, Marsland BJ. Impact of early-life exposures on immune maturation and susceptibility to disease. Trends Immunol. 2015;36:684–696. doi: 10.1016/j.it.2015.09.009.
    1. Jenmalm MC. The mother-offspring dyad: microbial transmission, immune interactions and allergy development. J Intern Med. 2017;282:484–495. doi: 10.1111/joim.12652.
    1. Macpherson AJ, de Agüero MG, Ganal-Vonarburg SC. How nutrition and the maternal microbiota shape the neonatal immune system. Nat Rev Immunol. 2017;17:508–517. doi: 10.1038/nri.2017.58.
    1. Zhang X, Zhivaki D, Lo-Man R. Unique aspects of the perinatal immune system. Nat Rev Immunol. 2017;17:495–507. doi: 10.1038/nri.2017.54.
    1. Julia V, Macia L, Dombrowicz D. The impact of diet on asthma and allergic diseases. Nat Rev Immunol. 2015;15:308–322. doi: 10.1038/nri3830.
    1. Dzidic M, Boix-Amorós A, Selma-Royo M, Mira A, Collado M. Gut microbiota and mucosal immunity in the neonate. Med Sci (Basel, Switzerland) 2018;6:56.
    1. Renz H, Skevaki C. Early life microbial exposures and allergy risks: opportunities for prevention. Nat Rev Immunol. 2020;21(3):177–191. doi: 10.1038/s41577-020-00420-y.
    1. Miles EA, Calder PC. Can early omega-3 fatty acid exposure reduce risk of childhood allergic disease? Nutrients. 2017;9:784. doi: 10.3390/nu9070784.
    1. Blasbalg TL, Hibbeln JR, Ramsden CE, Majchrzak SF, Rawlings RR. Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century. Am J Clin Nutr. 2011;93:950–962. doi: 10.3945/ajcn.110.006643.
    1. Warstedt K, Duchén K. Increased linoleic acid/α-linolenic acid ratio in Swedish cord blood samples collected between 1985 and 2005. Eur J Nutr. 2012;52:659–665. doi: 10.1007/s00394-012-0369-6.
    1. Miyata J, Arita M. Role of omega-3 fatty acids and their metabolites in asthma and allergic diseases. Allergol Int. 2015;64:27–34. doi: 10.1016/j.alit.2014.08.003.
    1. Krauss-Etschmann S, Hartl D, Rzehak P, et al. Decreased cord blood IL-4, IL-13, and CCR4 and increased TGF-β levels after fish oil supplementation of pregnant women. J Allergy Clin Immunol. 2008;121:464–470. doi: 10.1016/j.jaci.2007.09.018.
    1. Bisgaard H, Stokholm J, Chawes BL, et al. Fish oil-derived fatty acids in pregnancy and wheeze and asthma in offspring. N Engl J Med. 2016;375:2530–2539. doi: 10.1056/NEJMoa1503734.
    1. Furuhjelm C, Warstedt K, Larsson J, et al. Fish oil supplementation in pregnancy and lactation may decrease the risk of infant allergy. Acta Paediatr. 2009;98:1461–1467. doi: 10.1111/j.1651-2227.2009.01355.x.
    1. Furuhjelm C, Warstedt K, Fagerås M, et al. Allergic disease in infants up to 2 years of age in relation to plasma omega-3 fatty acids and maternal fish oil supplementation in pregnancy and lactation. Pediatr Allergy Immunol. 2011;22:505–514. doi: 10.1111/j.1399-3038.2010.01096.x.
    1. Stokholm J, Blaser MJ, Thorsen J, et al. Maturation of the gut microbiome and risk of asthma in childhood. Nat Commun. 2018;9:141. doi: 10.1038/s41467-017-02573-2.
    1. Bisgaard H, Li N, Bonnelykke K, et al. Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J Allergy Clin Immunol. 2011;128:646–652. doi: 10.1016/j.jaci.2011.04.060.
    1. Sjögren YM, Jenmalm MC, Böttcher MF, Björkstén B, Sverremark-Ekström E. Altered early infant gut microbiota in children developing allergy up to 5 years of age. Clin Exp Allergy. 2009;39:518–526. doi: 10.1111/j.1365-2222.2008.03156.x.
    1. Chiu C-Y, Chan Y-L, Tsai M-H, Wang C-J, Chiang M-H, Chiu C-C. Gut microbial dysbiosis is associated with allergen-specific IgE responses in young children with airway allergies. World Allergy Org J. 2019;12:100021. doi: 10.1016/j.waojou.2019.100021.
    1. Ling Z, Li Z, Liu X, et al. Altered fecal microbiota composition associated with food allergy in infants. Appl Environ Microbiol. 2014;80:2546–2554. doi: 10.1128/AEM.00003-14.
    1. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol. 2012;129:434. doi: 10.1016/j.jaci.2011.10.025.
    1. Cuello-Garcia CA, Brozek JL, Fiocchi A, et al. Probiotics for the prevention of allergy: A systematic review and meta-analysis of randomized controlled trials. J Allergy Clin Immunol; 136.
    1. Wells JM. Immunomodulatory mechanisms of lactobacilli. Microb Cell Fact. 2011;10(Suppl 1):S17. doi: 10.1186/1475-2859-10-S1-S17.
    1. Abrahamsson TR, Sandberg Abelius M, Forsberg A, Björkstén B, Jenmalm MC. A Th1/Th2-associated chemokine imbalance during infancy in children developing eczema, wheeze and sensitization. Clin Exp Allergy. 2011;41:1729–1739. doi: 10.1111/j.1365-2222.2011.03827.x.
    1. Alhamwe B, Alhamdan F, Ruhl A, Potaczek DP, Renz H. The role of epigenetics in allergy and asthma development. Curr Opin Allergy Clin Immunol. 2020;20:48–55. doi: 10.1097/ACI.0000000000000598.
    1. Baribault C, Ehrlich KC, Ponnaluri VKCKC, Pradhan S, Lacey M, Ehrlich M. Developmentally linked human DNA hypermethylation is associated with down-modulation, repression, and upregulation of transcription. Epigenetics. 2018;13:275–289. doi: 10.1080/15592294.2018.1445900.
    1. Baubec T, Schübeler D. Genomic patterns and context specific interpretation of DNA methylation. Curr Opin Genet Dev. 2014;25:85–92. doi: 10.1016/j.gde.2013.11.015.
    1. Schmidl C, Delacher M, Huehn J, Feuerer M. Epigenetic mechanisms regulating T-cell responses. J Allergy Clin Immunol. 2018;142:728–743. doi: 10.1016/j.jaci.2018.07.014.
    1. Zhang H, Kaushal A, Merid S, et al. DNA methylation and allergic sensitizations, a genome-scale longitudinal study during adolescence. Allergy. 2019;74:1166–1175. doi: 10.1111/all.13746.
    1. Peng C, Meel ER, Cardenas A, et al. Epigenome-wide association study reveals methylation pathways associated with childhood allergic sensitization. Epigenetics. 2019;14(5):445–466. doi: 10.1080/15592294.2019.1590085.
    1. DeVries A, Wlasiuk G, Miller SJ, et al. Epigenome-wide analysis links SMAD3 methylation at birth to asthma in children of asthmatic mothers. J Allergy Clin Immunol. 2017;140:534–542. doi: 10.1016/j.jaci.2016.10.041.
    1. Lund RJ, Osmala M, Malonzo M, et al. Atopic asthma after rhinovirus-induced wheezing is associated with DNA methylation change in the SMAD3 gene promoter. Allergy. 2018;73:1735–1740. doi: 10.1111/all.13473.
    1. Martino D, Dang T, Sexton-Oates A, et al. Blood DNA methylation biomarkers predict clinical reactivity in food-sensitized infants. J Allergy Clin Immunol. 2015;135:1319–1328. doi: 10.1016/j.jaci.2014.12.1933.
    1. Martino D, Neeland M, Dang T, et al. Epigenetic dysregulation of naive CD4+ T-cell activation genes in childhood food allergy. Nat Commun. 2018;9:3308. doi: 10.1038/s41467-018-05608-4.
    1. Harb H, Irvine J, Amarasekera M, et al. The role of PKCζ in cord blood T-cell maturation towards Th1 cytokine profile and its epigenetic regulation by fish oil. Biosci Rep. 2017;37.
    1. Amarasekera M, Noakes P, Strickland D, Saffery R, Martino DJ, Prescott SL. Epigenome-wide analysis of neonatal CD4(+) T-cell DNA methylation sites potentially affected by maternal fish oil supplementation. Epigenetics. 2014;9:1570–1576. doi: 10.4161/15592294.2014.983366.
    1. van Dijk SJ, Zhou J, Peters TJ, et al. Effect of prenatal DHA supplementation on the infant epigenome: results from a randomized controlled trial. Clin Epigenetics. 2016;8:114. doi: 10.1186/s13148-016-0281-7.
    1. Vähämiko S, Laiho A, Lund R, Isolauri E, Salminen S, Laitinen K. The impact of probiotic supplementation during pregnancy on DNA methylation of obesity-related genes in mothers and their children. Eur J Nutr. 2018:1–11.
    1. Forsberg A, Huoman J, Söderholm S, et al. Pre- and postnatal Lactobacillus reuteri treatment alters DNA methylation of infant T helper cells. Pediatric Allergy Immunol. 2020;31(5):544–553. doi: 10.1111/pai.13240.
    1. de Weerd HA, Badam TVS, Martínez-Enguita D, et al. MODifieR: an ensemble R package for inference of disease modules from transcriptomics networks. Bioinformatics (Oxford, England) 2020;36:3918–3919. doi: 10.1093/bioinformatics/btaa235.
    1. D’Aquila P, Carelli L, Rango F, Passarino G, Bellizzi D. Gut microbiota as important mediator between diet and DNA methylation and histone modifications in the host. Nutrients. 2020;12:597. doi: 10.3390/nu12030597.
    1. Ceccarelli V, Ronchetti S, Marchetti M, et al. Molecular mechanisms underlying eicosapentaenoic acid inhibition of HDAC1 and DNMT expression and activity in carcinoma cells. Biochim Biophys Acta. 2020;1863:194481. doi: 10.1016/j.bbagrm.2020.194481.
    1. González-Becerra K, Ramos-Lopez O, Barrón-Cabrera E, et al. Fatty acids, epigenetic mechanisms and chronic diseases: a systematic review. Lipids Health Dis. 2019;18:178. doi: 10.1186/s12944-019-1120-6.
    1. Salmond RJ, Filby A, Qureshi I, Caserta S, Zamoyska R. T-cell receptor proximal signaling via the Src-family kinases, Lck and Fyn, influences T-cell activation, differentiation, and tolerance. Immunol Rev. 2009;228:9–22. doi: 10.1111/j.1600-065X.2008.00745.x.
    1. Kaur S, Bansal Y, Kumar R, Bansal G. A panoramic review of IL-6: Structure, pathophysiological roles and inhibitors. Bioorgan Med Chem. 2020;28:115327. doi: 10.1016/j.bmc.2020.115327.
    1. Zhou X, Han X, Lyu S-C, et al. Targeted DNA methylation profiling reveals epigenetic signatures in peanut allergy. JCI Insight; 2021.
    1. Baccarelli A, Rusconi F, Bollati V, et al. Nasal cell DNA methylation, inflammation, lung function and wheezing in children with asthma. Epigenomics. 2012;4:91–100. doi: 10.2217/epi.11.106.
    1. Bruyns E, Marie-Cardine A, Kirchgessner H, et al. T cell receptor (TCR) interacting molecule (TRIM), a novel disulfide-linked dimer associated with the TCR–CD3–ζ complex, recruits intracellular signaling proteins to the plasma membrane. J Exp Med. 1998;188:561–575. doi: 10.1084/jem.188.3.561.
    1. Kirchgessner H, Dietrich J, Scherer J, et al. The transmembrane adaptor protein trim regulates t cell receptor (Tcr) expression and Tcr-mediated signaling via an association with the Tcr ζ chain. J Exp Med. 2001;193:1269–1284. doi: 10.1084/jem.193.11.1269.
    1. Mehta AK, Gracias DT, Croft M. TNF activity and T cells. Cytokine. 2018;101:14–18. doi: 10.1016/j.cyto.2016.08.003.
    1. Yamada A, Arakaki R, Saito M, Kudo Y, Ishimaru N. Dual role of fas/fasl-mediated signal in peripheral immune tolerance. Front Immunol. 2017;8:403. doi: 10.3389/fimmu.2017.00403.
    1. Demmelmair H, Koletzko B. Importance of fatty acids in the perinatal period. World Rev Nutr Diet. 2015;112:31–47. doi: 10.1159/000365427.
    1. Navarro-Tapia E, Sebastiani G, Sailer S, et al. Probiotic supplementation during the perinatal and infant period: effects on gut dysbiosis and disease. Nutrients. 2020;12:2243. doi: 10.3390/nu12082243.
    1. Davis EC, Dinsmoor AM, Wang M, Donovan SM. Microbiome composition in pediatric populations from birth to adolescence: impact of diet and prebiotic and probiotic interventions. Dig Dis Sci. 2020;65:706–722. doi: 10.1007/s10620-020-06092-x.
    1. Murphy R, Morgan XC, Wang XY, et al. Eczema-protective probiotic alters infant gut microbiome functional capacity but not composition: sub-sample analysis from a RCT. Beneficial Microbes. 2019;10:5–17. doi: 10.3920/BM2017.0191.
    1. Dotterud CK, Avershina E, Sekelja M, et al. Does maternal perinatal probiotic supplementation alter the intestinal microbiota of mother and child? J Pediatr Gastroenterol Nutr. 2015;61:200–207. doi: 10.1097/MPG.0000000000000781.
    1. Watson H, Mitra S, Croden FC, et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut. 2017;67:1974–1983. doi: 10.1136/gutjnl-2017-314968.
    1. Forsberg A, West CE, Prescott SL, Jenmalm MC. Pre- and probiotics for allergy prevention: time to revisit recommendations? Clin Exp Allergy. 2016;46:1506–1521. doi: 10.1111/cea.12838.
    1. Jenmalm MC, Duchén K. Timing of allergy-preventive and immunomodulatory dietary interventions - are prenatal, perinatal or postnatal strategies optimal? Clin Exp Allergy. 2013;43:273–278. doi: 10.1111/cea.12003.
    1. Abrahamsson TR, Jakobsson T, Böttcher MF, et al. Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2007;119:1174–1180. doi: 10.1016/j.jaci.2007.01.007.
    1. Solomon O, MacIsaac J, Quach H, et al. Comparison of DNA methylation measured by Illumina 450K and EPIC BeadChips in blood of newborns and 14-year-old children. Epigenetics. 2018;13(6):655–664. doi: 10.1080/15592294.2018.1497386.
    1. Martino D, Maksimovic J, Joo JH, Prescott SL, Saffery R. Genome-scale profiling reveals a subset of genes regulated by DNA methylation that program somatic T-cell phenotypes in humans. Genes immunity. 2012;13(5):388–398. doi: 10.1038/gene.2012.7.
    1. Tian Y, Morris TJ, Webster AP, et al. ChAMP: updated methylation analysis pipeline for Illumina BeadChips. Bioinformatics (Oxford, England) 2017;33:3982–3984. doi: 10.1093/bioinformatics/btx513.
    1. Teschendorff AE, Marabita F, Lechner M, et al. A beta-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450 k DNA methylation data. Bioinformatics (Oxford, England) 2013;29:189–196. doi: 10.1093/bioinformatics/bts680.
    1. Morin AM, Gatev E, McEwen LM, et al. Maternal blood contamination of collected cord blood can be identified using DNA methylation at three CpGs. Clin Epigenetics. 2017;9:75. doi: 10.1186/s13148-017-0370-2.
    1. Houseman EA, Accomando WP, Koestler DC, et al. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinform. 2012;13:86. doi: 10.1186/1471-2105-13-86.
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics (Oxford, England) 2007;8:118–127. doi: 10.1093/biostatistics/kxj037.
    1. Leek JT, Johnson EW, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–883. doi: 10.1093/bioinformatics/bts034.
    1. van Iterson M, van Zwet EW, Consortium B, Heijmans BT. Controlling bias and inflation in epigenome- and transcriptome-wide association studies using the empirical null distribution. Genome Biol. 2017;18:19. doi: 10.1186/s13059-016-1131-9.
    1. Nestor CE, Barrenas F, Wang H, et al. DNA methylation changes separate allergic patients from healthy controls and may reflect altered CD4+ T-cell population structure. PLoS Genet. 2014;10:e1004059. doi: 10.1371/journal.pgen.1004059.

Source: PubMed

3
購読する