Elsevier

Chemosphere

Volume 112, October 2014, Pages 1-8
Chemosphere

Metagenomic profiles and antibiotic resistance genes in gut microbiota of mice exposed to arsenic and iron

https://doi.org/10.1016/j.chemosphere.2014.03.068Get rights and content

Highlights

  • Changes of mice gut microbiota under As and/or Fe exposure were analyzed.

  • Co-exposure of As and Fe mitigated effects on gut microbial community in mice.

  • Exposure of As and/or Fe changed types and abundance of ARGs.

  • Changes of gut microbiota influenced host metabolic profiles.

  • Gut microbiota should be considered during risk assessment of As and/or Fe.

Abstract

Iron (Fe) has been widely applied to treat arsenic (As)-contaminated water, and Fe could influence bioavailability and toxicity of As. However, little is known about the impact of As and/or Fe on gut microbiota, which plays important roles in host health. In this study, high-throughput sequencing and quantitative real time PCR were applied to analyze the impact of As and Fe on mouse gut microbiota. Co-exposure of As and Fe mitigated effects on microbial community to a certain extent. Correlation analysis showed the shifts in gut microbiota caused by As and/or Fe exposure might be important reason of changes in metabolic profiles of mouse. For antibiotic resistance genes (ARGs), co-exposure of As and Fe increased types and abundance of ARGs. But for high abundance ARGs, such as tetQ, tetO and tetM, co-exposure of As and Fe mitigated effects on their abundances compared to exposure to As and Fe alone. No obvious relationship between ARGs and mobile genetic elements were found. The changes in ARGs caused by metal exposure might be due to the alteration of gut microbial diversity. Our results show that changes of gut microbial community caused by As and/or Fe can influence host metabolisms and abundances of ARGs in gut, indicating that changes of gut microbiota should be considered during the risk assessment of As and/or Fe.

Introduction

Gut harbors diverse microbes that play a key role in well-being of their host. The gut microbiota acts in a concerted manner to achieve metabolic communication with the host, and many different bacterial genera and species are involved in metabolite production (Wikoff et al., 2009, Mestdagh et al., 2012, Martinez et al., 2013). Changes in gut microbiota are linked with inflammatory and metabolic disorders (Nicholson et al., 2012). Many researches have showed that metal exposure could change the gut microbiota (Dostal et al., 2012). On the other hand, the gut microbiota could change transportation and metabolism of metals (Wiele et al., 2010). Thus, it is necessary to identify impacts of metal on the gut microbiota under oral metal exposure. Arsenic (As) as ubiquitous metalloid has been paid much attention due to its high toxicity. Consumption of drinking water is the main source of As exposure. Iron (Fe) coagulation/flocculation has been widely applied in the actual treatment of As-contaminated water due to its low cost and high efficiency (Mohan and Pittman, 2007). Our previous study demonstrated that combined exposure of As and Fe in mouse could significantly reduce hepatic toxicity of As (Liu et al., 2013). In addition, some altered host-gut co-metabolites in serum and urine were identified, indicating the possible changes of gut microbiota. Thus, it is necessary to explore the impacts of As and/or Fe on gut microbiota to better understand their combined effects.

Gut microbiota is an important antibiotic resistance genes (ARGs) reservoir, playing important roles in host health. Recently, it has been suggested that environmental pollution could affect abundance of resistance traits. In fact, there are various known mechanisms by which the resistance traits may be retained or propagated in the presence of metals (Baker-Austin et al., 2006, Stepanauskas et al., 2006). Bacterial resistance mechanisms exist to mitigate toxicological effects of excessive bioavailable metals as part of their stress response strategy. Defense-associated metals are often closely associated with those responsible for antibiotic resistance on mobile genetic elements (MGEs) (Beaber et al., 2004). These genes can encode for generic detoxifying mechanisms (e.g. efflux pumps), which non-specifically reduce intracellular concentrations of both metals and antibiotics (cross resistance) (Berg et al., 2010, Knapp et al., 2011). Thus, constant exposure to metals can increase ARGs’ frequency in the gene pool in environmental or gut bacteria. There are many reports on correlation between tolerance to metals (including As and Fe) and antibiotic resistance in environment (Tuckfield and McArthur, 2008, Kaur et al., 2011, Ji et al., 2012). However, effects of As and/or Fe on the ARGs in gut microbiota are still unknown.

In the present study, we exposed pure water, As alone, Fe alone and As + Fe to male mice for 90 d, respectively. After exposure, gut microbiota were analyzed by high-throughput sequencing. Relationships between gut microbiota and mouse metabolic profiles were characterized by correlation analysis. The ARGs in gut microbiota were determined based on high throughput sequencing and verified by quantitative real-time PCR (qRT-PCR). This study firstly provides the effects of As and/or Fe exposure on gut microbiota and ARGs. Combined with results on metabolic profiles of mouse serum and urine, this study might be very useful for understanding of toxicological effects and mechanism of actions of As and/or Fe exposure.

Section snippets

Animal treatment

Five-week-old male mice (Mus musculus, ICR) were purchased from the experimental animal center of Academy of Military Medical Science of China. Forty mice (about 18 g) were randomly assigned to four groups (ten mice in one group). The mice in four groups were exposed to pure water, 3 mg L−1 As, 5 mg L−1 Fe and 3 mg −1 As + 5 mg L−1 Fe under ambient conditions (25 ± 3 °C, 50 ± 5% relative humidity, and a 12/12 h light/dark cycle) for 90 d, respectively. The concentrations selected for As and Fe were based on our

Histopathological maps

Representative images of H&E-stained histologic sections of mouse intestine are shown in Fig. 1. Compared with control group, As alone exposure induced small-bowel mucosal edema of intestine. Fe alone exposure caused the necrosis of intestine. For co-exposure of As and Fe, little damage was observed. We deduce that co-exposure of As and Fe has antagonistic effects on mouse intestine. The results are similar with our previous report, in which the biological effects of co-exposure of As and Fe

Conclusions

Exposure of As alone and Fe alone could alter the diversity and functions of gut microbiota and abundance of ARGs and MGEs. However, co-exposure of As and Fe had antagonistic effects on microbial community and ARGs to a certain extent. The alterations of gut microbiota might be an important reason of changes in metabolic profiles of mice serum and urine. The changes of gut microbiota should be considered during the risk assessment of As and/or Fe.

Acknowledgements

This research was supported by grants from the Natural Science Foundation of Jiangsu Province (SBK201320987), Foundation of State Key Laboratory of Pollution Control and Resource Reuse, Science Foundation of Nanjing University and National Natural Science Foundation of China (51208250). The authors declare no conflict of interest.

References (47)

  • J. Berg et al.

    Cu exposure under field conditions coselects for antibiotic resistance as determined by a novel cultivation-independent bacterial community tolerance assay

    Environ. Sci. Technol.

    (2010)
  • P. Boerlin et al.

    Antimicrobial resistance: its emergence and transmission

    Anim. Health Res. Rev.

    (2008)
  • I. Cho et al.

    Antibiotics in early life alter the murine colonic microbiome and adiposity

    Nature

    (2012)
  • I. Chopra et al.

    Tetracycline antibiotics: mode of action, applications, molecular biology, and epidemiology of bacterial resistance

    Microbiol. Mol. Biol. Rev.

    (2001)
  • S.P. Claus et al.

    Colonization-induced host-gut microbial metabolic interaction

    mBio

    (2011)
  • D.H. Huson et al.

    MEGAN analysis of metagenomic data

    Genome Res.

    (2007)
  • S. Kaur et al.

    Role of arsenic and its resistance in nature

    Can. J. Microbiol.

    (2011)
  • C.W. Knapp et al.

    Indirect evidence of transposon-mediated selection of antibiotic resistance genes in aquatic systems at low-level oxytetracycline exposures

    Environ. Sci. Technol.

    (2008)
  • C.W. Knapp et al.

    Antibiotic resistance gene abundances correlate with metal and geochemical conditions in archived Scottish soils

    PLoS One

    (2011)
  • J.E. Koenig et al.

    Succession of microbial consortia in the developing infant gut microbiome

    Proc. Natl. Acad. Sci. USA

    (2011)
  • E. Kristiansson et al.

    Pyrosequencing of antibiotic-contaminated river sediments reveals high levels of resistance and gene transfer elements

    PLoS One

    (2011)
  • R.E. Ley et al.

    Obesity alters gut microbial ecology

    Proc. Natl. Acad. Sci. USA

    (2005)
  • B. Liu et al.

    ARDB–antibiotic resistance genes database

    Nucl. Acids Res.

    (2009)
  • Cited by (0)

    View full text