Multi-omics analysis reveals the influence of genetic and environmental risk factors on developing gut microbiota in infants at risk of celiac disease

Maureen M Leonard, Hiren Karathia, Meritxell Pujolassos, Jacopo Troisi, Francesco Valitutti, Poorani Subramanian, Stephanie Camhi, Victoria Kenyon, Angelo Colucci, Gloria Serena, Salvatore Cucchiara, Monica Montuori, Basilio Malamisura, Ruggiero Francavilla, Luca Elli, Brian Fanelli, Rita Colwell, Nur Hasan, Ali R Zomorrodi, Alessio Fasano, CD-GEMM Team, Pasqua Piemontese, Angela Calvi, Mariella Baldassarre, Lorenzo Norsa, Chiara Maria Trovato, Celeste Lidia Raguseo, Tiziana Passaro, Paola Roggero, Marco Crocco, Annalisa Morelli, Michela Perrone, Marcello Chieppa, Giovanni Scala, Maria Elena Lionetti, Carlo Catassi, Adelaide Serretiello, Corrado Vecchi, Gemma Castillejo de Villsante, Maureen M Leonard, Hiren Karathia, Meritxell Pujolassos, Jacopo Troisi, Francesco Valitutti, Poorani Subramanian, Stephanie Camhi, Victoria Kenyon, Angelo Colucci, Gloria Serena, Salvatore Cucchiara, Monica Montuori, Basilio Malamisura, Ruggiero Francavilla, Luca Elli, Brian Fanelli, Rita Colwell, Nur Hasan, Ali R Zomorrodi, Alessio Fasano, CD-GEMM Team, Pasqua Piemontese, Angela Calvi, Mariella Baldassarre, Lorenzo Norsa, Chiara Maria Trovato, Celeste Lidia Raguseo, Tiziana Passaro, Paola Roggero, Marco Crocco, Annalisa Morelli, Michela Perrone, Marcello Chieppa, Giovanni Scala, Maria Elena Lionetti, Carlo Catassi, Adelaide Serretiello, Corrado Vecchi, Gemma Castillejo de Villsante

Abstract

Background: Celiac disease (CD) is an autoimmune digestive disorder that occurs in genetically susceptible individuals in response to ingesting gluten, a protein found in wheat, rye, and barley. Research shows that genetic predisposition and exposure to gluten are necessary but not sufficient to trigger the development of CD. This suggests that exposure to other environmental stimuli early in life, e.g., cesarean section delivery and exposure to antibiotics or formula feeding, may also play a key role in CD pathogenesis through yet unknown mechanisms. Here, we use multi-omics analysis to investigate how genetic and early environmental risk factors alter the development of the gut microbiota in infants at risk of CD.

Results: Toward this end, we selected 31 infants from a large-scale prospective birth cohort study of infants with a first-degree relative with CD. We then performed rigorous multivariate association, cross-sectional, and longitudinal analyses using metagenomic and metabolomic data collected at birth, 3 months and 6 months of age to explore the impact of genetic predisposition and environmental risk factors on the gut microbiota composition, function, and metabolome prior to the introduction of trigger (gluten). These analyses revealed several microbial species, functional pathways, and metabolites that are associated with each genetic and environmental risk factor or that are differentially abundant between environmentally exposed and non-exposed infants or between time points. Among our significant findings, we found that cesarean section delivery is associated with a decreased abundance of Bacteroides vulgatus and Bacteroides dorei and of folate biosynthesis pathway and with an increased abundance of hydroxyphenylacetic acid, alterations that are implicated in immune system dysfunction and inflammatory conditions. Additionally, longitudinal analysis revealed that, in infants not exposed to any environmental risk factor, the abundances of Bacteroides uniformis and of metabolite 3-3-hydroxyphenylproprionic acid increase over time, while those for lipoic acid and methane metabolism pathways decrease, patterns that are linked to beneficial immunomodulatory and anti-inflammatory effects.

Conclusions: Overall, our study provides unprecedented insights into major taxonomic and functional shifts in the developing gut microbiota of infants at risk of CD linking genetic and environmental risk factors to detrimental immunomodulatory and inflammatory effects. Video Abstract.

Trial registration: ClinicalTrials.gov NCT02061306.

Keywords: Celiac disease; Microbiota; Multi-omics analysis, gut microbiome.

Conflict of interest statement

AF is a stockholder at Alba Therapeutics, serves as a consultant for Inova Diagnostics and Innovate Biopharmaceuticals, is an advisory board member for Axial Biotherapeutics and Ubiome, and has a speaker agreement with Mead Johnson Nutrition. MML serves as a consultant to HealthMode and Anokion, has a speaker agreement with Takeda Pharmaceuticals, and performs sponsored research with Glutenostics LLC. HK is a former employee, BF is a current employee, PS is a consultant, and RRC and NAH are stockholders at CosmosID Inc. Other authors have declared no competing interests exist.

Figures

Fig. 1
Fig. 1
Schematic representing the sample selection and study design. We selected 31 infants from the CDGEMM study [33] with fecal samples available at enrollment, 3 months, and 4–6 months after birth. The sample underwent metagenomic and metabolomic profiling and was next analyzed to identify associations between genetic and environmental risk factors and inter-subject and intra-subject variations
Fig. 2
Fig. 2
Analysis of associations between genetic and environmental risk factors and microbial species. We used MaAsLin [22], a widely used multivariate statistical framework, to identify statistically significant associations between each genetic and environmental risk factor and microbial species (p value < 0.01), No genetic risk, vaginal delivery, exclusive breastmilk feeding, and no exposure to antibiotics were taken as reference for genetic risk, delivery mode, feeding type, and antibiotic exposure, respectively. Microbial species were clustered based on Euclidean distance. Here, “u_s” denotes and unspecified species
Fig. 3
Fig. 3
Analysis of associations between genetic and environmental risk factors and functional pathways. We used MaAsLin [22] to identify statistically significant associations between each genetic and environmental risk factor and functional pathways (p value < 0.01), Pathways were clustered based on Euclidean distance. Additional File 8 for grouping of these pathways based on KEGG categorizations
Fig. 4
Fig. 4
Analysis of associations between genetic and environmental risk factors and metabolites. We used MaAsLin [22] to identify statistically significant associations between each genetic and environmental risk factor and metabolites (p value < 0.01). Metabolites were clustered based on Euclidean distance
Fig. 5
Fig. 5
Cross-sectional analysis of microbiota features for genetically predisposed infants. a functional pathways (p value < 0.05), and b metabolites that are differentially abundant between environmentally exposed and non-exposed infants according to Mann-Whitney U test (p value < 0.05). Additional File 8 for grouping of pathways based on KEGG categorizations. See Additional File 9 for boxplots showing altered abundances for these pathways and metabolites. Brackets show time points at which a significant difference between the exposed and non-exposed groups was observed
Fig. 6
Fig. 6
Longitudinal analysis of microbiota features for genetically predisposed infants a microbial species, b functional pathways, and c metabolites that are differentially abundant between each pair of time points (enrollment, 3 months, and 4–6 months) according to a paired Wilcoxon (Wilcoxon signed rank) test (p value < 0.05). Here, “Time1” denotes the earlier time point. In this figure, “u_s” denotes and unspecified species. Additional File 8 for grouping of pathways based on KEGG categorizations. See Additional File 9 for boxplots showing altered abundances for these pathways and metabolites

References

    1. Lionetti E, Gatti S, Pulvirenti A, Catassi C. Celiac disease from a global perspective. Best Pract Res Clin Gastroenterol. 2015;29(3):365–379.
    1. Schuppan D. Current concepts of celiac disease pathogenesis. Gastroenterology. 2000;119(1):234–242.
    1. Green PH, Cellier C. Celiac disease. N Engl J Med. 2007;357(17):1731–1743.
    1. Vecsei E, Steinwendner S, Kogler H, Innerhofer A, Hammer K, Haas OA, et al. Follow-up of pediatric celiac disease: value of antibodies in predicting mucosal healing, a prospective cohort study. BMC Gastroenterol. 2014;14:28.
    1. Leonard MM, Weir DC, DeGroote M, Mitchell PD, Singh P, Silvester JA, et al. Value of IgA tTG in predicting mucosal recovery in children with celiac disease on a gluten-free diet. J Pediatr Gastroenterol Nutr. 2017;64(2):286–291.
    1. Ciacci C, Cirillo M, Cavallaro R, Mazzacca G. Long-term follow-up of celiac adults on gluten-free diet: prevalence and correlates of intestinal damage. Digestion. 2002;66(3):178–185.
    1. Rubio-Tapia A, Rahim MW, See JA, Lahr BD, Wu TT, Murray JA. Mucosal recovery and mortality in adults with celiac disease after treatment with a gluten-free diet. Am J Gastroenterol. 2010;105(6):1412–1420.
    1. Valitutti F, Trovato CM, Montuori M, Cucchiara S. Pediatric celiac disease: follow-up in the spotlight. Adv Nutr. 2017;8(2):356–361.
    1. Valitutti F, Fasano A. Breaking down barriers: how understanding celiac disease pathogenesis informed the development of novel treatments. Dig Dis Sci. 2019;64(7):1748–1758.
    1. West J, Fleming KM, Tata LJ, Card TR, Crooks CJ. Incidence and prevalence of celiac disease and dermatitis herpetiformis in the UK over two decades: population-based study. Am J Gastroenterol. 2014;109(5):757.
    1. Catassi C, Kryszak D, Bhatti B, Sturgeon C, Helzlsouer K, Clipp SL, et al. Natural history of celiac disease autoimmunity in a USA cohort followed since 1974. Ann Med. 2010;42(7):530–538.
    1. Ricano-Ponce I, Wijmenga C, Gutierrez-Achury J. Genetics of celiac disease. Best Pract Res Clin Gastroenterol. 2015;29(3):399–412.
    1. Tamburini S, Shen N, Wu HC, Clemente JC. The microbiome in early life: implications for health outcomes. Nat Med. 2016;22(7):713–722.
    1. Decker E, Engelmann G, Findeisen A, Gerner P, Laass M, Ney D, et al. Cesarean delivery is associated with celiac disease but not inflammatory bowel disease in children. Pediatrics. 2010;125(6):e1433–e1440.
    1. Marild K, Stephansson O, Montgomery S, Murray JA, Ludvigsson JF. Pregnancy outcome and risk of celiac disease in offspring: a nationwide case-control study. Gastroenterology. 2012;142(1):39–45.
    1. Akobeng AK, Ramanan AV, Buchan I, Heller RF. Effect of breast feeding on risk of coeliac disease: a systematic review and meta-analysis of observational studies. Arch Dis Child. 2006;91(1):39–43.
    1. Szajewska H, Chmielewska A, Piescik-Lech M, Ivarsson A, Kolacek S, Koletzko S, et al. Systematic review: early infant feeding and the prevention of coeliac disease. Aliment Pharmacol Ther. 2012;36(7):607–618.
    1. Norris JM, Barriga K, Hoffenberg EJ, Taki I, Miao D, Haas JE, et al. Risk of celiac disease autoimmunity and timing of gluten introduction in the diet of infants at increased risk of disease. JAMA. 2005;293(19):2343–2351.
    1. Marild K, Ye W, Lebwohl B, Green PH, Blaser MJ, Card T, et al. Antibiotic exposure and the development of coeliac disease: a nationwide case-control study. BMC Gastroenterol. 2013;13:109.
    1. Lionetti E, Castellaneta S, Francavilla R, Pulvirenti A, Tonutti E, Amarri S, et al. Introduction of gluten, HLA status, and the risk of celiac disease in children. N Engl J Med. 2014;371(14):1295–1303.
    1. Vriezinga SL, Auricchio R, Bravi E, Castillejo G, Chmielewska A, Crespo Escobar P, et al. Randomized feeding intervention in infants at high risk for celiac disease. N Engl J Med. 2014;371(14):1304–1315.
    1. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13(9):R79.
    1. Stewart CJ, Ajami NJ, O’Brien JL, Hutchinson DS, Smith DP, Wong MC, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562(7728):583.
    1. Jangi S, Gandhi R, Cox LM, Li N, Von Glehn F, Yan R, et al. Alterations of the human gut microbiome in multiple sclerosis. Nat Commun. 2016;7:12015.
    1. De Palma G, Capilla A, Nadal I, Nova E, Pozo T, Varea V, et al. Interplay between human leukocyte antigen genes and the microbial colonization process of the newborn intestine. Curr Issues Mol Biol. 2010;12(1):1–10.
    1. Olivares M, Neef A, Castillejo G, Palma GD, Varea V, Capilla A, et al. The HLA-DQ2 genotype selects for early intestinal microbiota composition in infants at high risk of developing coeliac disease. Gut. 2015;64(3):406–417.
    1. Sellitto M, Bai G, Serena G, Fricke WF, Sturgeon C, Gajer P, et al. Proof of concept of microbiome-metabolome analysis and delayed gluten exposure on celiac disease autoimmunity in genetically at-risk infants. PLoS One. 2012;7(3):e33387.
    1. Pozo-Rubio T, de Palma G, Mujico JR, Olivares M, Marcos A, Acuna MD, et al. Influence of early environmental factors on lymphocyte subsets and gut microbiota in infants at risk of celiac disease; the PROFICEL study. Nutr Hosp. 2013;28(2):464–473.
    1. Olivares M, Walker AW, Capilla A, Benitez-Paez A, Palau F, Parkhill J, et al. Gut microbiota trajectory in early life may predict development of celiac disease. Microbiome. 2018;6(1):36.
    1. Rintala A, Riikonen I, Toivonen A, Pietila S, Munukka E, Pursiheimo JP, et al. Early fecal microbiota composition in children who later develop celiac disease and associated autoimmunity. Scand J Gastroenterol. 2018:1–7.
    1. Torres J, Hu J, Seki A, Eisele C, Nair N, Huang R, et al. Infants born to mothers with IBD present with altered gut microbiome that transfers abnormalities of the adaptive immune system to germ-free mice. Gut. 2020;69(1):42–51.
    1. Vatanen T, Franzosa EA, Schwager R, Tripathi S, Arthur TD, Vehik K, et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature. 2018;562(7728):589–594.
    1. Leonard MM, Camhi S, Huedo-Medina TB, Fasano A. Celiac disease genomic, environmental, microbiome, and Metabolomic (CDGEMM) study design: approach to the future of personalized prevention of celiac disease. Nutrients. 2015;7(11):9325–9336.
    1. Hov JR, Zhong H, Qin B, Anmarkrud JA, Holm K, Franke A, Lie BA, Karlsen TH: The influence of the autoimmunity-associated ancestral HLA haplotype AH8. 1 on the human gut microbiota: a cross-sectional study. PLoS One 2015, 10(7):e0133804.
    1. Wei Y, Li Y, Yan L, Sun C, Miao Q, Wang Q, et al. Alterations of gut microbiome in autoimmune hepatitis. Gut. 2019.
    1. Ye Z, Zhang N, Wu C, Zhang X, Wang Q, Huang X, et al. A metagenomic study of the gut microbiome in Behcet’s disease. Microbiome. 2018;6(1):135.
    1. Cree BA, Spencer CM, Varrin-Doyer M, Baranzini SE, Zamvil SS. Gut microbiome analysis in neuromyelitis optica reveals overabundance of Clostridium perfringens. Ann Neurol. 2016;80(3):443–447.
    1. Shao Y, Forster SC, Tsaliki E, Vervier K, Strang A, Simpson N, et al. Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature. 2019:1–5.
    1. Bokulich NA, Chung J, Battaglia T, Henderson N, Jay M, Li H, Lieber AD, Wu F, Perez-Perez GI, Chen Y: Antibiotics, birth mode, and diet shape microbiome maturation during early life. Science translational medicine 2016, 8(343):343ra382-343ra382.
    1. Wampach L, Heintz-Buschart A, Fritz JV, Ramiro-Garcia J, Habier J, Herold M, et al. Birth mode is associated with earliest strain-conferred gut microbiome functions and immunostimulatory potential. Nat Commun. 2018;9(1):5091.
    1. Yoshida N, Emoto T, Yamashita T, Watanabe H, Hayashi T, Tabata T, et al. Bacteroides vulgatus and Bacteroides dorei reduce gut microbial lipopolysaccharide production and inhibit atherosclerosis. Circulation. 2018;138(22):2486–2498.
    1. Bayer AL, Fraker CA. The folate cycle as a cause of natural killer cell dysfunction and viral etiology in type 1 diabetes. Front Endocrinol (Lausanne) 2017;8:315.
    1. Sitkin SI, Tkachenko EI, Vakhitov T, Oreshko LS. Zhigalova TN: [serum metabolome by gas chromatography-mass spectrometry (GC-MS) in patients with ulcerative colitis and celiac disease] Eksp Klin Gastroenterol. 2013;12:44–57.
    1. Ma EH, Bantug G, Griss T, Condotta S, Johnson RM, Samborska B, et al. Serine is an essential metabolite for effector T cell expansion. Cell Metab. 2017;25(2):482.
    1. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17(5):690–703.
    1. Lundequist B, Nord CE, Winberg J. The composition of the faecal microflora in breastfed and bottle fed infants from birth to eight weeks. Acta Paediatr Scand. 1985;74(1):45–51.
    1. Chua HH, Chou HC, Tung YL, Chiang BL, Liao CC, Liu HH, et al. Intestinal dysbiosis featuring abundance of Ruminococcus gnavus associates with allergic diseases in infants. Gastroenterology. 2018;154(1):154–167.
    1. Kameyama K, Itoh K: Intestinal colonization by a Lachnospiraceae bacterium contributes to the development of diabetes in obese mice. Microbes and environments 2014:ME14054.
    1. Zeng H, Ishaq SL, Zhao F-Q, Wright A-DG. Colonic inflammation accompanies an increase of β-catenin signaling and Lachnospiraceae/Streptococcaceae bacteria in the hind gut of high-fat diet-fed mice. J Nutr Biochem. 2016;35:30–36.
    1. Cabral DJ, Penumutchu S, Reinhart EM, Zhang C, Korry BJ, Wurster JI, Nilson R, Guang A, Sano WH, Rowan-Nash AD: Microbial metabolism modulates antibiotic susceptibility within the murine gut microbiome. Cell metabolism 2019, 30(4):800-823. e807.
    1. Wilson C, Tiwana H, Ebringer A. Molecular mimicry between HLA-DR alleles associated with rheumatoid arthritis and Proteus mirabilis as the aetiological basis for autoimmunity. Microbes Infect. 2000;2(12):1489–1496.
    1. Sanchez E, De Palma G, Capilla A, Nova E, Pozo T, Castillejo G, et al. Influence of environmental and genetic factors linked to celiac disease risk on infant gut colonization by Bacteroides species. Appl Environ Microbiol. 2011;77(15):5316–5323.
    1. Cano PG, Santacruz A, Moya Á, Sanz Y. Bacteroides uniformis CECT 7771 ameliorates metabolic and immunological dysfunction in mice with high-fat-diet induced obesity. PLoS One. 2012;7(7):e41079.
    1. Orešič M, Simell S, Sysi-Aho M, Näntö-Salonen K, Seppänen-Laakso T, Parikka V, et al. Dysregulation of lipid and amino acid metabolism precedes islet autoimmunity in children who later progress to type 1 diabetes. J Exp Med. 2008;205(13):2975–2984.
    1. Brown CT, Davis-Richardson AG, Giongo A, Gano KA, Crabb DB, Mukherjee N, et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One. 2011;6(10):e25792.
    1. Liu W, Shi L-J, Li S-G. The immunomodulatory effect of alpha-lipoic acid in autoimmune diseases. BioMed Res Int. 2019;2019.
    1. Boros M, Ghyczy M, Érces D, Varga G, Tokés T, Kupai K, et al. The anti-inflammatory effects of methane. Crit Care Med. 2012;40(4):1269–1278.
    1. Zhang X, Li N, Shao H, Meng Y, Wang L, Wu Q, et al. Methane limit LPS-induced NF-κB/MAPKs signal in macrophages and suppress immune response in mice by enhancing PI3K/AKT/GSK-3β-mediated IL-10 expression. Sci Rep. 2016;6:29359.
    1. Agrawal S, Agrawal A, Said HM. Biotin deficiency enhances the inflammatory response of human dendritic cells. Am J Phys Cell Phys. 2016;311(3):C386–C391.
    1. Kung JT, Mackenzie CG, Talmage DW. The requirement for biotin and fatty acids in the cytotoxic T-cell response. Cell Immunol. 1979;48(1):100–110.
    1. Sedel F, Bernard D, Mock DM, Tourbah A. Targeting demyelination and virtual hypoxia with high-dose biotin as a treatment for progressive multiple sclerosis. Neuropharmacology. 2016;110:644–653.
    1. Abad-Lacruz A, Fernandez-Banares F, Cabre E, Gil A, Esteve M, Gonzalez-Huix F, et al. The effect of total enteral tube feeding on the vitamin status of malnourished patients with inflammatory bowel disease. International journal for vitamin and nutrition research Internationale Zeitschrift fur Vitamin-und Ernahrungsforschung Journal international de vitaminologie et de nutrition. 1988;58(4):428–435.
    1. Fernandez-Banares F, Abad-Lacruz A, Xiol X, Gine J, Dolz C, Cabre E, Esteve M, Gonzalez-Huix F, Gassull M. Vitamin status in patients with inflammatory bowel disease. Am J Gastroenterol. 1989:84(7).
    1. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, de Roos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–455.
    1. Hootman KC, Trezzi J-P, Kraemer L, Burwell LS, Dong X, Guertin KA, et al. Erythritol is a pentose-phosphate pathway metabolite and associated with adiposity gain in young adults. Proc Natl Acad Sci. 2017;114(21):E4233–E4240.
    1. Fan FY, Sang LX, Jiang M. Catechins and their therapeutic benefits to inflammatory bowel disease. Molecules. 2017:22(3).
    1. Baron S, Turck D, Leplat C, Merle V, Gower-Rousseau C, Marti R, et al. Environmental risk factors in paediatric inflammatory bowel diseases: a population based case control study. Gut. 2005;54(3):357–363.
    1. Xu L, Lochhead P, Ko Y, Claggett B, Leong RW, Ananthakrishnan AN. Systematic review with meta-analysis: breastfeeding and the risk of Crohn’s disease and ulcerative colitis. Aliment Pharmacol Ther. 2017;46(9):780–789.
    1. Ungaro R, Bernstein CN, Gearry R, Hviid A, Kolho KL, Kronman MP, et al. Antibiotics associated with increased risk of new-onset Crohn’s disease but not ulcerative colitis: a meta-analysis. Am J Gastroenterol. 2014;109(11):1728–1738.
    1. Costenbader KH, Kim DJ, Peerzada J, Lockman S, Nobles-Knight D, Petri M, et al. Cigarette smoking and the risk of systemic lupus erythematosus: a meta-analysis. Arthritis Rheum. 2004;50(3):849–857.
    1. McCormic ZD, Khuder SS, Aryal BK, Ames AL, Khuder SA. Occupational silica exposure as a risk factor for scleroderma: a meta-analysis. Int Arch Occup Environ Health. 2010;83(7):763–769.
    1. Wilmanski T, Rappaport N, Earls JC, Magis AT, Manor O, Lovejoy J, et al. Blood metabolome predicts gut microbiome α-diversity in humans. Nat Biotechnol. 2019;37(10):1217–1228.
    1. Analysis of blood and fecal microbiome profile in patients with celiac disease. Human Microbiome Journal 2019, 11.
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput Biol. 2012;8(6):e1002358.
    1. Human Microbiome Project C Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–214.
    1. Sjogren YM, Jenmalm MC, Bottcher MF, Bjorksten B, Sverremark-Ekstrom E. Altered early infant gut microbiota in children developing allergy up to 5 years of age. Clin Exp Allergy. 2009;39(4):518–526.
    1. Ownby DR, Johnson CC, Peterson EL. Exposure to dogs and cats in the first year of life and risk of allergic sensitization at 6 to 7 years of age. JAMA. 2002;288(8):963–972.
    1. Virtanen SM, Takkinen HM, Nwaru BI, Kaila M, Ahonen S, Nevalainen J, et al. Microbial exposure in infancy and subsequent appearance of type 1 diabetes mellitus-associated autoantibodies: a cohort study. JAMA Pediatr. 2014;168(8):755–763.
    1. Ewels P, Magnusson M, Lundin S, Kaller M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics. 2016;32(19):3047–3048.
    1. Hasan NA, Young BA, Minard-Smith AT, Saeed K, Li H, Heizer EM, et al. Microbial community profiling of human saliva using shotgun metagenomic sequencing. PLoS One. 2014;9(5):e97699.
    1. Ponnusamy D, Kozlova EV, Sha J, Erova TE, Azar SR, Fitts EC, et al. Cross-talk among flesh-eating Aeromonas hydrophila strains in mixed infection leading to necrotizing fasciitis. Proc Natl Acad Sci U S A. 2016;113(3):722–727.
    1. Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19(5):455–477.
    1. Hyatt D, Chen G-L, LoCascio PF, Land ML, Larimer FW, Hauser LJ. Prodigal: prokaryotic gene recognition and translation initiation site identification. BMC bioinformatics. 2010;11(1):119.
    1. Jones P, Binns D, Chang H-Y, Fraser M, Li W, McAnulla C, et al. InterProScan 5: genome-scale protein function classification. Bioinformatics. 2014;30(9):1236–1240.
    1. Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.
    1. Zerbino DR, Birney E. Velvet: algorithms for de novo short read assembly using de Bruijn. Genome Res. 2004.
    1. Sumner LW, Amberg A, Barrett D, Beale MH, Beger R, Daykin CA, et al. Proposed minimum reporting standards for chemical analysis. Metabolomics. 2007;3(3):211–221.
    1. Chong J, Soufan O, Li C, Caraus I, Li S, Bourque G, et al. MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis. Nucleic Acids Res. 2018;46(W1):W486–W494.

Source: PubMed

3
Suscribir