Intestinal microbiota in infants at high risk for allergy: Effects of prebiotics and role in eczema development  Harm Wopereis, BSc, Kathleen Sim, MRCPCH,

Slides:



Advertisements
Similar presentations
A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis  Magali Noval Rivas, PhD, Oliver T. Burton,
Advertisements

Effects of early-life exposure to allergens and bacteria on recurrent wheeze and atopy in urban children  Susan V. Lynch, PhD, Robert A. Wood, MD, Homer.
Vitamin D over the first decade and susceptibility to childhood allergy and asthma  Elysia M. Hollams, PhD, Shu Mei Teo, PhD, Merci Kusel, MBBS, PhD, Barbara.
Repeated low-dose intradermal allergen injection suppresses allergen-induced cutaneous late responses  Giuseppina Rotiroti, MD, Mohamed Shamji, PhD, Stephen.
The interferon gene signature is increased in patients with early treatment-naive rheumatoid arthritis and predicts a poorer response to initial therapy 
SQ-standardized sublingual grass immunotherapy: Confirmation of disease modification 2 years after 3 years of treatment in a randomized trial  Stephen.
Kyoung-Bok Min, MD, PhD, Jin-Young Min, PhD 
Serum cathelicidin, nasopharyngeal microbiota, and disease severity among infants hospitalized with bronchiolitis  Kohei Hasegawa, MD, MPH, Jonathan M.
Mauro Maniscalco, MD, PhD, Debora Paris, PhD, Dominique J
Transcriptome analysis of proton pump inhibitor–responsive esophageal eosinophilia reveals proton pump inhibitor–reversible allergic inflammation  Ting.
Eosinophilic airway inflammation in asthmatic patients is associated with an altered airway microbiome  Asger Sverrild, MD, PhD, Pia Kiilerich, MSc, PhD,
Inflammatory phenotypes in patients with severe asthma are associated with distinct airway microbiology  Steven L. Taylor, BSc, Lex E.X. Leong, PhD, Jocelyn.
Volume 14, Issue 6, Pages (February 2016)
Effects of early-life exposure to allergens and bacteria on recurrent wheeze and atopy in urban children  Susan V. Lynch, PhD, Robert A. Wood, MD, Homer.
Human seroreactivity to gut microbiota antigens
Volume 22, Issue 3, Pages (September 2015)
The interferon gene signature is increased in patients with early treatment-naive rheumatoid arthritis and predicts a poorer response to initial therapy 
Volume 22, Issue 2, Pages (August 2015)
Faecalibacterium prausnitzii subspecies–level dysbiosis in the human gut microbiome underlying atopic dermatitis  Han Song, PhD, Young Yoo, MD, PhD, Junghyun.
A genome-wide association study to identify genetic determinants of atopy in subjects from the United Kingdom  Yize I. Wan, BMedSci, David P. Strachan,
Effect of probiotics in prevention of atopic dermatitis is dependent on the intrinsic microbiota at early infancy  Ekaterina Avershina, PhD, Raul Cabrera.
Microbiota studies in the bile duct strongly suggest a role for Helicobacter pylori in extrahepatic cholangiocarcinoma  F. Avilés-Jiménez, A. Guitron,
Associations between infant fungal and bacterial dysbiosis and childhood atopic wheeze in a nonindustrialized setting  Marie-Claire Arrieta, PhD, Andrea.
Cesarean section changes neonatal gut colonization
A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis  Magali Noval Rivas, PhD, Oliver T. Burton,
Vitamin D over the first decade and susceptibility to childhood allergy and asthma  Elysia M. Hollams, PhD, Shu Mei Teo, PhD, Merci Kusel, MBBS, PhD, Barbara.
Vijay R. Ramakrishnan, MD, Leah J. Hauser, MD, Leah M
Correction Journal of Allergy and Clinical Immunology
Capsaicin-evoked cough responses in asthmatic patients: Evidence for airway neuronal dysfunction  Imran Satia, MD, Nikolaos Tsamandouras, PhD, Kimberley.
Blood DNA methylation biomarkers predict clinical reactivity in food-sensitized infants  David Martino, PhD, Thanh Dang, PhD, Alexandra Sexton-Oates, BSc,
Volume 47, Issue 2, Pages e4 (August 2017)
Martin Depner, PhD, Markus J. Ege, MD, Michael J
Majda Dzidic, MSc, Thomas R
Dynamics of the nasal microbiota in infancy: A prospective cohort study  Moana Mika, MSc, Ines Mack, MD, Insa Korten, MD, Weihong Qi, PhD, Suzanne Aebi,
Factors influencing the infant gut microbiome at age 3-6 months: Findings from the ethnically diverse Vitamin D Antenatal Asthma Reduction Trial (VDAART) 
The National Biome Initiative: An allergy perspective
Lower airway microbiota and mycobiota in children with severe asthma
Associations between infant fungal and bacterial dysbiosis and childhood atopic wheeze in a nonindustrialized setting  Marie-Claire Arrieta, PhD, Andrea.
Airway epithelial cells from asthmatic children differentially express proremodeling factors  Jesus M. Lopez-Guisa, PhD, Claire Powers, BA, Daniele File,
Furry pets modulate gut microbiota composition in infants at risk for allergic disease  Merja Nermes, MD, PhD, Akihito Endo, PhD, Jasmin Aarnio, BM, Seppo.
Early-life gut microbiome composition and milk allergy resolution
Bacterial diversity in Bercaea Cruentata gut described using high-throughput sequencing  L.L. Long, J.J. Guo, P. Li, Y.D. Guo  Forensic Science International:
Benjamin A. Turturice, BS, Diane R. Gold, MD, MPH, Augusto A
Volume 10, Issue 11, Pages (March 2015)
Collateral damage from oral ciprofloxacin versus nitrofurantoin in outpatients with urinary tract infections: a culture-free analysis of gut microbiota 
Peanut epitopes for IgE and IgG4 in peanut-sensitized children in relation to severity of peanut allergy  Annebeth E. Flinterman, MD, Edward F. Knol,
C. Kampmann, J. Dicksved, L. Engstrand, H. Rautelin 
The airway microbiome in patients with severe asthma: Associations with disease features and severity  Yvonne J. Huang, MD, Snehal Nariya, BS, Jeffrey.
Control lymphocyte subsets: Can one country's values serve for another's?  Wilson L. Mandala, PhD, Jintanat Ananworanich, MD, PhD, Tanakorn Apornpong,
IL-5 T-cell responses to house dust mite are associated with the development of allergen-specific IgE responses and asthma in the first 5 years of life 
FIGURE 1 α-Diversity comparisons of gut microbiota from DM and MOM infants by age and antibiotic exposure. Using ... FIGURE 1 α-Diversity comparisons of.
Jon Genuneit, MD, MSc  Journal of Allergy and Clinical Immunology 
Low diversity of the gut microbiota in infants with atopic eczema
Clinical correlates of lung ventilation defects in asthmatic children
Bifidobacterium pseudocatenulatum is associated with atopic eczema: A nested case- control study investigating the fecal microbiota of infants  Claudia.
Is eczema really on the increase worldwide?
Different IgE recognition of mite allergen components in asthmatic and nonasthmatic children  Yvonne Resch, MSc, Sven Michel, MSc, Michael Kabesch, MD,
Exacerbation of atopic dermatitis on grass pollen exposure in an environmental challenge chamber  Thomas Werfel, MD, Annice Heratizadeh, MD, Margarete.
Paul V. Licciardi, PhD, Anne Balloch, MSc, Fiona M
Baochen Shi, PhD, Nathanael J
Erik J. Saude, MD, PhD, Christopher D
Volume 7, Issue 3, Pages e7 (September 2018)
The Diversity of Gut Microbiome is Associated With Favorable Responses to Anti– Programmed Death 1 Immunotherapy in Chinese Patients With NSCLC  Yueping.
Peanut protein in household dust is related to household peanut consumption and is biologically active  Helen A. Brough, MRCPCH, MSc, Alexandra F. Santos,
Pradeep Reddy Marri, PhD, Debra A. Stern, MS, Anne L
Repeated low-dose intradermal allergen injection suppresses allergen-induced cutaneous late responses  Giuseppina Rotiroti, MD, Mohamed Shamji, PhD, Stephen.
Increased risk of eczema but reduced risk of early wheezy disorder from exclusive breast-feeding in high-risk infants  Charlotte Giwercman, MD, Liselotte.
Respiratory syncytial virus and rhinovirus severe bronchiolitis are associated with distinct nasopharyngeal microbiota  Jonathan M. Mansbach, MD, MPH,
Comparison of distal gut microbiota composition between Egyptian and U
Fig. 4 The abundance of Bifidobacterium in breast-fed infant guts is associated with FL transporter genes. The abundance of Bifidobacterium in breast-fed.
Presentation transcript:

Intestinal microbiota in infants at high risk for allergy: Effects of prebiotics and role in eczema development  Harm Wopereis, BSc, Kathleen Sim, MRCPCH, PhD, Alexander Shaw, PhD, John O. Warner, MD, FMedSci, Jan Knol, PhD, J. Simon Kroll, MA, FMedSci  Journal of Allergy and Clinical Immunology  Volume 141, Issue 4, Pages 1334-1342.e5 (April 2018) DOI: 10.1016/j.jaci.2017.05.054 Copyright © 2017 The Authors Terms and Conditions

Journal of Allergy and Clinical Immunology 2018 141, 1334-1342 Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions

Fig 1 Bar plots (means with 95% CIs) summarizing microbial intestinal richness (A) and phylogenetic diversity (B) at 4 and 26 weeks of age in breast-fed infants (B) and infants receiving pHF-OS (A) or control formula (C). Comparisons were done at a sequencing depth of 1636 reads per sample. Statistics were performed by using 1-way ANOVA with the Bonferroni multiple comparison correction. *Adjusted P < .05 and **adjusted P < .01. Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions

Fig 2 A, Bar plots (means with 95% CIs) summarizing weighted UniFrac distances at 4 and 26 weeks of age in infants receiving pHF-OS or control formula relative to breast-fed infants (A-B vs C-B, respectively). Distances are measured on a scale from 0 to 1, with 0 meaning 100% identical and 1 meaning 0% identical gut microbial composition. Statistical comparison of weighted UniFrac was performed by using a 1-way ANOVA with the Bonferroni multiple comparison correction. ****Adjusted P < .0001. B-D, Box plots summarizing log2-transformed levels of differential abundant genera present in more than 15% of samples and a mean relative abundance of greater than 0.1% when comparing pHF-OS with control formula at 26 weeks, namely Bifidobacterium species (Fig 2, B), Clostridium species (Fig 2, C), and an unassigned genus of Lachnospiraceae (Fig 2, D). Differential abundances were computed by using MetagenomeSeq, with correction for effects of ethnicity and having siblings and adjustment of significance values for multiple comparisons by using the Benjamini-Hochberg false discovery rate. **Adjusted P < .01 and ****adjusted P < .0001. Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions

Fig 3 A, PRCs showing changes in bacterial metabolites (levels of SCFAs and lactic acid) across time and the interaction with the different feeding groups. The horizontal axis represents time, and the vertical axis represents PRC score values. Fecal metabolite composition of infants receiving control formula (C) was used as a reference level and has zero PRC values; therefore its curve is over the horizontal axis. Changes for infants being breast-fed (B) or receiving pHF-OS (A) are shown as response curves relative to this reference. Metabolite response scores are shown on the separate vertical (1-dimensional) plot. The multiple of the PRC score with the response score provides a quantitative interpretation, as well as the direction of change at respective time points (4, 12, and 26 weeks). Significance of the interaction corrected for covariates was tested by using MCPT (P = .002, 499 permutations). B, Box plots summarizing stool pH at 4, 12, and 26 weeks of age for the different feeding groups. Statistical comparison was performed by using 1-way ANOVA with the Bonferroni multiple comparison correction comparing pHF-OS with control formula. *Adjusted P < .05, **adjusted P < .01, and ***adjusted P < .001. Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions

Fig 4 A, PRCs showing changes in bacterial metabolites (levels of SCFAs and lactic acid) across time and interaction with eczema development. The horizontal axis represents time, and the vertical axis represents PRC score values. The fecal metabolite composition of infants without eczema (NO ECZ) were used as the reference level and has zero PRC values. Change for infants having eczema in the first 18 months of life (ECZ) is shown as a response curve relative to this reference. Metabolite response scores are shown on the separate vertical (1-dimensional) plot. Significance of the interaction corrected for effects of feeding group, ethnicity, and having siblings was tested by using MCPT (P = .034, 499 permutations). B, Box plots summarizing stool pH at 4, 12, and 26 weeks of age in infants with eczema versus healthy infants. Statistical comparison was performed by using a 1-way ANOVA with the Bonferroni multiple comparison correction (not significantly different). Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions

Fig 5 A, Plot with bacterial taxa that change over time (dt = change from 4 to 26 weeks of age) in healthy infants and infants with eczema, as well as taxa that are differential over time in infants with eczema compared with healthy infants. Data are shown as fold changes with SDs of log2-transformed data. Taxa are detailed at the phylum (B, Bacteroidetes; F, Firmicutes; and P, Proteobacteria), family, and genus levels, respectively. The contrasts were computed by using MetagenomeSeq and corrected for the effects of feeding group, ethnicity, and having siblings. Adjustment of significance values for multiple comparisons was done by using the Benjamini-Hochberg false discovery rate. Differential taxa with an adjusted P value of less than .05 for all contrasts if present in more than 15% of the samples and a mean abundance of greater than 0.1% are shown. Taxa plotted on the dark gray background showed no significant (ns) temporal differences when comparing healthy infants with eczematous infants. Taxa plotted on light gray and white backgrounds were different, with adjusted P values of less than .1 and less than .05, respectively. B, Fecal concentration of D-lactic acid (D-LA) at 26 weeks in healthy infants (NOECZ) compared with infants with eczema (ECZ). C, Fecal concentration of L-lactic acid (L-LA) at 26 weeks of age in healthy subjects versus patients with eczema. D, Fecal concentration of butyrate (BA) at age 26 weeks in healthy subjects versus patients with eczema. Significance was tested with a Mann-Whitney 2-group comparison: *P < .05 and **P < .001. Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions

Fig E1 Bar plots (means with 95% CIs) summarizing the microbial intestinal richness (A) and phylogenetic diversity (B) at 4 and 26 weeks of age in infants with eczema (ECZ) compared with infants without eczema (NOECZ). Comparisons were done at a sequencing depth of 1636 reads per sample. Statistics were performed by using a 1-way ANOVA with the Bonferroni multiple comparison correction (not significantly different). Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions

Fig E2 Taxa identified with MetagenomeSeq that show differential relative abundances over time when comparing healthy infants with infants with eczema (adjusted P < .1). Levels for sample pairs are shown as log2-transformed data, as well as means and SDs at 4 and 26 weeks of age, and are shown for Enterobacter (A), an unassigned genus of Enterobacteriaceae (B), Parabacteroides (C), Eubacterium (D), and Anaerostipes (E) species, respectively. Left panels show data for infants without eczema (NO ECZ; n = 82 sample pairs), and right panels show data for infants with eczema (ECZ; n = 48 sample pairs), respectively. Journal of Allergy and Clinical Immunology 2018 141, 1334-1342.e5DOI: (10.1016/j.jaci.2017.05.054) Copyright © 2017 The Authors Terms and Conditions