Household triclosan and triclocarban effects on the infant and maternal microbiome

Jessica V Ribado, Catherine Ley, Thomas D Haggerty, Ekaterina Tkachenko, Ami S Bhatt, Julie Parsonnet, Jessica V Ribado, Catherine Ley, Thomas D Haggerty, Ekaterina Tkachenko, Ami S Bhatt, Julie Parsonnet

Abstract

In 2016, the US Food and Drug Administration banned the use of specific microbicides in some household and personal wash products due to concerns that these chemicals might induce antibiotic resistance or disrupt human microbial communities. Triclosan and triclocarban (referred to as TCs) are the most common antimicrobials in household and personal care products, but the extent to which TC exposure perturbs microbial communities in humans, particularly during infant development, was unknown. We conducted a randomized intervention of TC-containing household and personal care products during the first year following birth to characterize whether TC exposure from wash products perturbs microbial communities in mothers and their infants. Longitudinal survey of the gut microbiota using 16S ribosomal RNA amplicon sequencing showed that TC exposure from wash products did not induce global reconstruction or loss of microbial diversity of either infant or maternal gut microbiotas. Broadly antibiotic-resistant species from the phylum Proteobacteria, however, were enriched in stool samples from mothers in TC households after the introduction of triclosan-containing toothpaste. When compared by urinary triclosan level, agnostic to treatment arm, infants with higher triclosan levels also showed an enrichment of Proteobacteria species. Despite the minimal effects of TC exposure from wash products on the gut microbial community of infants and adults, detected taxonomic differences highlight the need for consumer safety testing of antimicrobial self-care products on the human microbiome and on antibiotic resistance.

Keywords: antibiotic; microbiome; resistance; triclosan.

© 2017 The Authors. Published under the terms of the CC BY 4.0 license.

Figures

Figure 1. Urinary triclosan levels are elevated…
Figure 1. Urinary triclosan levels are elevated in TC mothers following 6 months of exposure
The concentrations of triclosan in picograms/microliter (pg/μl) are plotted as a log10 transformation of the absolute concentration +1, for visual clarity. The median value is represented as a black line, the interquartile range is represented by the box plot. The violin plot represents the full range of values obtained from the source data, where the width of the blue or red colored region represents the probability density of sample values at that level. The black dots represent outliers. Urinary triclosan measurements are available for 38 mothers (17 TC, 21 nTC) and 33 infants (15 TC, 18 nTC). The ranges for the violin plots are found in the source data. The P‐value for the differences in urinary triclosan levels between treatment arms in the mothers (represented by ***) is P = 5.66e‐5 (Mann–Whitney U‐test).Source data are available online for this figure.
Figure 2. Mother and infants have distinct…
Figure 2. Mother and infants have distinct microbiome compositions not driven by household TC exposure
  1. PCoA of Bray–Curtis dissimilarity for all (n = 221) samples shows that gut communities cluster by mothers and infants.

  2. PCoA separated by time and treatment.

Source data are available online for this figure.
Figure EV1. Individuals in mother and infant…
Figure EV1. Individuals in mother and infant groups have similar relative microbiome compositions at the phylum level throughout the first year of life independent of TC exposure
TC households (n = 17) and nTC (n = 22) households have stable relative abundance of phyla at 2‐, 6‐, and 10‐month visits. Phyla present in < 2% abundance are condensed for visual clarity.
Figure EV2. Maternal samples within households are…
Figure EV2. Maternal samples within households are more similar than between households
  1. Hierarchical clustering of Canberra distances shows that maternal samples from a given individual throughout the first year of life are more self‐similar than other household mothers at the same visit. The colors are representative of a single household, labeled by household and visit (B1 = 2 months, B2 = 6 months, B3 = 10 months), and node colors reflect TC grouping (red = nTC, blue = TC).

  2. PCoA of Canberra distances shows 9.3% of the variance is explained by the first two principal components and the absence of TC treatment clustering for any visit among mothers.

Figure EV3. Infant intestinal microbiome variability is…
Figure EV3. Infant intestinal microbiome variability is minimally driven by some known external factors by 2 months of age (n = 34)
  1. A–D

    PCoA with Bray–Curtis dissimilarity of the infants at 2 months of age suggests factors known to impact the microbiome, such as (A) birth method (PERMANOVA R2 = 0.0683, P = 0.011) and (B) formula (R2 = 0.128, P = 0.041) drive minimal variation. However, (C) maternal ethnicity (R2 = 0.0274, P = 0.505) and (D) pets (R2 = 0.0310, P = 0.393) in the household are not sufficient to distinguish the infants at 2 months of age.

Figure 3. TC randomization does not decrease…
Figure 3. TC randomization does not decrease gut microbial diversity in infants or mothers
Shannon diversity measures are plotted as the interquartile range with median for each TC exposure class and time‐point grouping for infants and mothers. The upper and lower whisker for box plots indicate ± 1.5× interquartile range. The points plotted beyond these whiskers are considered outliers and are plotted individually. The P‐value represented for the increase in infant gut microbiota diversity over time is ***P = 4.4e‐4 (Mann–Whitney U‐test).Source data are available online for this figure.
Figure EV4. Microbial diversity does not correlate…
Figure EV4. Microbial diversity does not correlate with urinary triclosan levels at 6 months for mothers or infants
Correlations were determined using a linear regression model (Shannon diversity ~ Urinary triclosan levels) on mother and infant samples separately.
Figure 4. Enrichment of Proteobacteria is observed…
Figure 4. Enrichment of Proteobacteria is observed in the mothers of TC households
  1. A

    Differentially abundant taxa between nTC and TC households. Values left of the gray line indicate an enrichment in nTC household and values to the right indicate an enrichment in TC households. Analyses are separated by mothers and infants for all samples across the three time points (FDR‐adjusted P‐value < 0.01).

  2. B, C

    Differentially abundant taxa are displayed for (B) infants and (C) mothers at per visit (FDR‐adjusted P‐value < 0.05).

Source data are available online for this figure.
Figure EV5. Triclosan resistance gene abundances do…
Figure EV5. Triclosan resistance gene abundances do not distinguish TC and nTC maternal samples following 6 months of exposure
Euclidean distance was calculated between samples of whole shotgun sequencing reads that aligned to CARD (approximately 0.03% or 6,000 reads for each sample in both intervention arms), then clustered with a hierarchical agglomeration method.

References

    1. Bengtsson‐Palme J, Angelin M, Huss M, Kjellqvist S, Kristiansson E, Palmgren H, Larsson DGJ, Johansson A (2015) The human gut microbiome as a transporter of antibiotic resistance genes between continents. Antimicrob Agents Chemother 59: 6551–6560
    1. Braoudaki M, Hilton AC (2004) Low level of cross‐resistance between triclosan and antibiotics in Escherichia coli K‐12 and E. coli O55 compared to E. coli O157. FEMS Microbiol Lett 235: 305–309
    1. Callahan BJ, Mcmurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP (2016) DADA2: high‐resolution sample inference from Illumina amplicon data. Nat Methods 13: 581
    1. Carey DE, McNamara PJ (2014) The impact of triclosan on the spread of antibiotic resistance in the environment. Front Microbiol 5: 780
    1. Cho I, Blaser MJ (2012) The human microbiome: at the interface of health and disease. Nat Rev Genet 13: 260–270
    1. Chu DM, Ma J, Prince AL, Antony KM, Seferovic MD, Aagaard KM (2017) Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat Med 23: 314–326
    1. Ciusa ML, Furi L, Knight D, Decorosi F, Fondi M, Raggi C, Coelho JR, Aragones L, Moce L, Visa P et al (2012) A novel resistance mechanism to triclosan that suggests horizontal gene transfer and demonstrates a potential selective pressure for reduced biocide susceptibility in clinical strains of Staphylococcus aureus . Int J Antimicrob Agents 40: 210–220
    1. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, Kim SG, Li H, Gao Z, Mahana D et al (2014) Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 158: 705–721
    1. Dixon P (2003) VEGAN, a package of R functions for community ecology. J Veg Sci 14: 927–930
    1. Forbes S, Knight CG, Cowley NL, Amézquita A, McClure P, Humphreys G, McBain AJ (2016) Variable effects of exposure to formulated microbicides on antibiotic susceptibility in firmicutes and proteobacteria. Appl Environ Microbiol 82: 3591–3598
    1. Gaulke CA, Barton CL, Proffitt S, Tanguay RL, Sharpton TJ (2016) Triclosan exposure is associated with rapid restructuring of the microbiome in adult zebrafish. PLoS One 11: e0154632
    1. Halden RU (2016) Lessons learned from probing for impacts of triclosan and triclocarban on human microbiomes. mSphere 1: e00089–16
    1. Hampton T (2011) Research provides new insights on how hygiene affects asthma and allergies. JAMA 305: 1400
    1. Han C, Lim YH, Hong YC (2016) Ten‐year trends in urinary concentrations of triclosan and benzophenone‐3 in the general U.S. population from 2003 to 2012. Environ Pollut 208: 803–810
    1. Hartmann EM, Hickey R, Hsu T, Betancourt Román CM, Chen J, Schwager R, Kline J, Brown GZ, Halden RU, Huttenhower C et al (2016) Antimicrobial chemicals are associated with elevated antibiotic resistance genes in the indoor dust microbiome. Environ Sci Technol 50: 9807–9815
    1. Hu J, Raikhel V, Gopalakrishnan K, Fernandez‐Hernandez H, Lambertini L, Manservisi F, Falcioni L, Bua L, Belpoggi F, L Teitelbaum S et al (2016) Effect of postnatal low‐dose exposure to environmental chemicals on the gut microbiome in a rodent model. Microbiome 4: 26
    1. Kennedy RC, Fling RR, Robeson MS, Saxton AM, Donnell RL, Darcy JL, Bemis DA, Liu J, Zhao L, Chen J (2016) Temporal development of gut microbiota in triclocarban exposed pregnant and neonatal rats. Sci Rep 6: 33430
    1. Khan R, Kong HG, Jung Y, Choi J, Baek K (2016) Triclosan resistome from metagenome reveals diverse enoyl acyl carrier protein reductases and selective enrichment of triclosan resistance genes. Sci Rep 6: 32322
    1. Ley C, Sanchez M de la L, Mathur A, Yang S, Sundaram V, Parsonnet J (2016) Stanford's Outcomes Research in Kids (STORK): a prospective study of healthy pregnant women and their babies in Northern California. BMJ Open 6: e010810
    1. Love MI, Huber W, Anders S (2014) Moderated estimation of fold change and dispersion for RNA‐seq data with DESeq2. Genome Biol 15: 550
    1. Martin R, Makino H, Cetinyurek Yavuz A, Ben‐Amor K, Roelofs M, Ishikawa E, Kubota H, Swinkels S, Sakai T, Oishi K et al (2016) Early‐life events, including mode of delivery and type of feeding, siblings and gender, shape the developing gut microbiota. PLoS One 11: e0158498
    1. Mazmanian SK, Cui HL, Tzianabos AO, Kasper DL (2005) An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122: 107–118
    1. Mazmanian SK, Round JL, Kasper DL (2008) A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453: 620–625
    1. McArthur AG, Waglechner N, Nizam F, Yan A, Azad MA, Baylay AJ, Bhullar K, Canova MJ, De Pascale G, Ejim L et al (2013) The comprehensive antibiotic resistance database. Antimicrob Agents Chemother 57: 3348–3357
    1. McMurdie PJ, Holmes S (2013) phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One 8: e61217
    1. McMurdie PJ, Holmes S (2014) Waste not, want not: why rarefying microbiome data is inadmissible. PLoS Comput Biol 10: e1003531
    1. Narrowe AB, Albuthi‐Lantz M, Smith EP, Bower KJ, Roane TM, Vajda AM, Miller CS (2015) Perturbation and restoration of the fathead minnow gut microbiome after low‐level triclosan exposure. Microbiome 3: 6
    1. Nobel YR, Cox LM, Kirigin FF, Bokulich NA, Yamanishi S, Teitler I, Chung J, Sohn J, Barber CM, Goldfarb DS et al (2015) Metabolic and metagenomic outcomes from early‐life pulsed antibiotic treatment. Nat Commun 6: 7486
    1. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO (2007) Development of the human infant intestinal microbiota. PLoS Biol 5: 1556–1573
    1. Poole AC, Pischel L, Ley C, Suh G, Goodrich JK, Haggerty TD, Ley RE, Parsonnet J (2016) Crossover control study of the effect of personal care products containing triclosan on the microbiome. mSphere 1: e00056–15
    1. Raymond F, Ouameur AA, Déraspe M, Iqbal N, Gingras H, Dridi B, Leprohon P, Plante P‐L, Giroux R, Bérubé È et al (2016) The initial state of the human gut microbiome determines its reshaping by antibiotics. ISME J 10: 707–720
    1. Savage JH, Matsui EC, Wood RA, Keet CA (2012) Urinary levels of triclosan and parabens are associated with aeroallergen and food sensitization. J Allergy Clin Immunol 130: 453–460.e7
    1. Seymour GJ, Palmer JE, Leishman SJ, Do HL, Westerman B, Carle AD, Faddy MJ, West MJ, Cullinan MP (2017) Influence of a triclosan toothpaste on periodontopathic bacteria and periodontitis progression in cardiovascular patients: a randomized controlled trial. J Periodontal Res 52: 61–73
    1. Shin N, Whon TW, Bae J (2015) Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol 33: 496–503
    1. Suller MT, Russell AD (2000) Triclosan and antibiotic resistance in Staphylococcus aureus . J Antimicrob Chemother 46: 11–18
    1. Tobar S, Tordesillas L, Berin MC (2016) Triclosan promotes epicutaneous sensitization to peanut in mice. Clin Transl Allergy 6: 4–7
    1. Voelker R (2016) Say goodbye to some antibacterials. JAMA 316: 1538
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR (2007) Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol 73: 5261–5267
    1. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez‐Bello MG, Contreras M , Magris M, Hidalgo G, Baldassano RN, Anokhin AP et al (2012) Human gut microbiome viewed across age and geography. Nature 486: 222
    1. Yee AL, Gilbert JA, Calafat AM, Ye X, Wong L‐Y, Reidy JA, Needham LL, Marty J, Hawj F, Hoffman JA et al (2016) MICROBIOME. Is triclosan harming your microbiome? Science 353: 348–349

Source: PubMed

3
Prenumerera