Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Infections by human gastrointestinal helminths are associated with changes in faecal microbiota diversity and composition

  • Timothy P. Jenkins,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliation Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom

  • Yasara Rathnayaka,

    Roles Formal analysis, Investigation, Methodology

    Affiliation Department of Zoology, University of Peradeniya, Peradeniya, Sri Lanka

  • Piyumali K. Perera,

    Roles Formal analysis, Methodology

    Affiliation Department of Zoology, University of Peradeniya, Peradeniya, Sri Lanka

  • Laura E. Peachey,

    Roles Writing – review & editing

    Affiliation Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom

  • Matthew J. Nolan,

    Roles Methodology, Writing – review & editing

    Affiliation Royal Veterinary College, University of London, North Mymms, United Kingdom

  • Lutz Krause,

    Roles Data curation, Formal analysis, Methodology, Resources, Software, Writing – review & editing

    Affiliation The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Australia

  • Rupika S. Rajakaruna,

    Roles Formal analysis, Funding acquisition, Investigation, Methodology, Writing – review & editing

    Affiliation Department of Zoology, University of Peradeniya, Peradeniya, Sri Lanka

  • Cinzia Cantacessi

    Roles Conceptualization, Funding acquisition, Investigation, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing

    cc779@cam.ac.uk

    Current address: Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom

    Affiliation Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom

Abstract

Investigations of the impact that patent infections by soil-transmitted gastrointestinal nematode parasites exert on the composition of the host gut commensal flora are attracting growing interest by the scientific community. However, information collected to date varies across experiments, and further studies are needed to identify consistent relationships between parasites and commensal microbial species. Here, we explore the qualitative and quantitative differences between the microbial community profiles of cohorts of human volunteers from Sri Lanka with patent infection by one or more parasitic nematode species (H+), as well as that of uninfected subjects (H-) and of volunteers who had been subjected to regular prophylactic anthelmintic treatment (Ht). High-throughput sequencing of the bacterial 16S rRNA gene, followed by bioinformatics and biostatistical analyses of sequence data revealed no significant differences in alpha diversity (Shannon) and richness between groups (P = 0.65, P = 0.13 respectively); however, beta diversity was significantly increased in H+ and Ht when individually compared to H-volunteers (P = 0.04). Among others, bacteria of the families Verrucomicrobiaceae and Enterobacteriaceae showed a trend towards increased abundance in H+, whereas the Leuconostocaceae and Bacteroidaceae showed a relative increase in H- and Ht respectively. Our findings add valuable knowledge to the vast, and yet little explored, research field of parasite—microbiota interactions and will provide a basis for the elucidation of the role such interactions play in pathogenic and immune-modulatory properties of parasitic nematodes in both human and animal hosts.

Introduction

More than 1 billion people worldwide are estimated to be infected by gastrointestinal (GI) soil-transmitted helminths, including the roundworm Ascaris lumbricoides, the whipworm Trichuris trichiura and the hookworms Necator americanus and Ancylostoma duodenale [1]. Infections by these nematodes alone are estimated to cause the loss of 4.98 million disability-adjusted life years (DALYs) globally [2], mainly affecting areas of developing countries characterised by suboptimal standards of sanitation and hygiene [3]. Despite global efforts to control infections by these parasites via mass drug administration (MDA), repeated exposure to infective larvae and high re-infection rates in at-risk populations make interruption of the life cycles of these nematodes and their elimination difficult to achieve [4, 5]. These challenges, together with the realistic threat of emerging drug resistance [6] drive the continual search for new, integrated strategies to control these diseases, based on a thorough understanding of the fundamental biology and epidemiology of these pathogens and their interactions with the human host [7]. Recently, studies of the intimate mechanisms that regulate the relationships between GI soil-transmitted nematodes and their vertebrate hosts have involved investigations of the impact that patent parasite infections exert on the composition of the gut commensal flora and relative abundance of individual bacterial groups [810]. The increased attention towards detailed explorations of parasite-microbiota interactions stems from knowledge that the gut commensal flora plays several key essential roles in human health, including nutrient metabolism, protection against pathogens and regulation of both innate and adaptive immune responses [11, 12]. Therefore, given that GI nematodes and the gut microbial flora share the same environment within the human host, it is plausible that parasite-microbiota interactions impact substantially on the health and homeostasis of helminth-infected individuals [10]. For instance, GI nematodes and the microbiota compete for host nutrients while, in parallel, the known immune-modulatory properties of a range of parasites may translate into dramatic modifications of the mucosal and systemic immunity to the resident bacteria [10]. The effects that GI nematode infections exert on the gut commensal flora of vertebrate hosts have long been neglected; however, recent studies have contributed preliminary information to this little-known field of research, mainly driven by the need to better understand the factors that determine the immune-modulatory properties of selected species of parasitic nematodes [1317]. Our group has recently attempted to determine the impact that experimental infections by the human hookworm, N. americanus, exert on the composition of the gut microbiota of human volunteers [15, 17, 18]. While no shifts in the relative abundance of individual bacterial taxa were observed over the course of these studies, increases in microbial species richness and diversity were detected following experimental infections [15, 17, 18]. However, these studies, conducted under controlled experimental settings and with a known number of infective larvae, are unlikely to represent ‘real-world’ infections (caused by the simultaneous presence of multiple parasite species with varying infection burdens). Thus far, to the best of our knowledge, only two studies have evaluated differences in the composition of the gut microbiota of human subjects naturally infected by GI nematodes [8, 9], with contrasting results. Indeed, while in the first study, Cooper and colleagues [8] detected a reduction in faecal bacterial diversity in Ecuadorean school children naturally infected by T. trichiura and A. lumbricoides compared to uninfected children or children solely infected by the former, a study by Lee et al. [9], reported a greater richness in the faecal microbiota of a cohort of indigenous Malaysian volunteers infected by multiple GI nematodes (i.e. hookworms, whipworms and roundworms) when compared with that of a group of uninfected subjects from New York. This data highlights the need for additional explorations of the impact that natural patent infections exert on the gut microbiota of infected human subjects. In addition, in the studies by Cooper et al. [8] and Lee et al. [9], the microbial profiling of helminth-infected and uninfected subjects with that of volunteers subjected to regular anthelmintic treatment was not examined. Given the widespread use of MDA in parasite-endemic areas [5], the possible consequences of regular use of chemotherapeutics on the composition of the host gut commensal flora deserves further investigation. Amongst the developing countries in which MDA is in use, Sri Lanka provides a suitable setting for such a study, given a 29% prevalence estimate for soil-transmitted helminths (including GI nematodes) in school children [19] and the implementation of mass deworming programmes since 1994 [20]. Therefore, in this study, we explore the qualitative and quantitative differences between the microbial community profiles of human volunteers (from diverse Sri Lankan communities) infected by one or more GI nematode species, and compare the gut microbial profiles of these subjects with those from a cohort of volunteers from the same geographical area who had been subjected to regular prophylactic anthelmintic treatment.

Materials and methods

Ethics statement

This study was approved and carried out in strict accordance and compliance with the guidelines of the Institutional Ethical Review Committee, Faculty of Medicine, University of Peradeniya, Sri Lanka (Research Project No. 2015/EC/58). Written informed consent was obtained from all subjects enrolled in the study.

Study area and characteristics of the population

A total number of 76 subjects from nine villages in four districts of Sri Lanka were screened for the presence of patent infections by GI nematodes (Fig 1). Participants were distributed as follows:

  • 45 subjects from the Kandy district, villages of Rangala, Lolgama, Elagolla, Lunugala, Hanthana, Akurana;
  • 10 from the Jaffna district, Valalai village;
  • 15 from the Puttalam district, Kandakuliya village;
  • 6 from the Kegalle district, Mawanella village (Fig 1).
thumbnail
Fig 1. Sampling locations in Sri Lanka.

Numbers 1–9 represent the villages Rangala, Lolgama, Elagolla, Lunugala, Hanthana, Mawanella, Akurana (in Kandy District), Valalai (in Jaffna District) and Kandakuliya (in Puttalam District), respectively.

https://doi.org/10.1371/journal.pone.0184719.g001

Subjects were both men and women, of varying ages and social background, with no reported symptoms of GI disease or any other concomitant diseases, and who had not received antibiotic treatment over at least 6 months prior to the study (Table 1). Participants from the villages of Rangala, Lolgama, Elagolla, Lunugala and Hanthana were workers in tea estates, while those from Kandakuliya and Valalai were fishermen (cf. Fig 1). Subjects from Mawanella and Akurana were living in congested urban areas characterised by poor sanitary living conditions. All participants were interviewed directly by YR, PKP and RSR using a standardised, pre-tested questionnaire aimed to identify means of access to water, knowledge of sanitary and hygiene standards, availability of and access to health care facilities, awareness of risks of infection by GI helminths, and frequency of anthelmintic treatments. A copy of the questionnaire is provided in S1 Fig.

thumbnail
Table 1. Village, age (years), gender of helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects enrolled in this study.

The parasite species infecting H+ volunteers and compound administered to Ht volunteers are also indicated (NA = not applicable; NP = not provided).

https://doi.org/10.1371/journal.pone.0184719.t001

Sample collection and parasitological analyses

Each volunteer was asked to provide a fresh stool sample for parasitological analyses. The presence of nematode eggs/larvae in each sample was assessed using a modified sucrose flotation method described previously [21]. Stool samples were refrigerated and transported to the laboratory for processing. For each sample, approximately 3 g of faeces were measured, mixed with distilled water in a capped centrifuge tube to a final volume of 15 ml. The mixtures were stirred thoroughly using a wooden applicator and centrifuged at 2045 g for 20 mins at room temperature (~27°C). Following centrifugation, the supernatants were discarded and the resulting pellets were re-suspended in distilled water and centrifuged (twice) until clear supernatants were obtained. The pellets were then emulsified using saturated sucrose solution, mixed thoroughly, and centrifuged for 20 min at 2045 g. Approximately 5 ml of the top meniscus of the resulting suspensions were collected in a centrifuge tube, mixed with distilled water up to a final volume of 15 ml and centrifuged for 10 min at 1370 g. This procedure was repeated and 1 ml of each suspension with the pellet was transferred to 1.5 ml eppendorf® tubes using a Pasteur pipette. Distilled water was added to a final volume of 1.5 ml and the tubes were centrifuged at 1150 g for 10 min. The clear supernatants were decanted and microscope slides were prepared using the remaining 0.5 ml pellets and examined under a light microscope. Helminth eggs were identified using established morphological keys [22].

DNA extraction and bacterial 16S rRNA Illumina sequencing

Genomic DNA was extracted directly from each sample, as well as from two negative (no-DNA template) controls, using the PowerSoil® DNA Isolation Kit (MO BIO Laboratories, Carlsbad, CA, USA), according to manufacturers’ instructions, within 1 month from collection. High-throughput sequencing of the V3-V4 hypervariable region of the bacterial 16S rRNA gene was performed on an Illumina MiSeq platform according to the standard protocols with minor adjustments. Briefly, the V3-V4 region was PCR-amplified using universal primers [23], that contained the Illumina adapter overhang nucleotide sequences, using the NEBNext hot start high-fidelity DNA polymerase (New England Biolabs) and the following thermocycling protocol, using DNA 2ng/μl: 2 min at 98°C, 20 cycles of 15 s at 98°C– 30 s at 63°C– 30 s at 72°C, and a final elongation of 5 min at 72°C. Amplicons were purified using AMPure XP beads (Beckman Coulter) and the NEBNext hot start high-fidelity DNA polymerase was used for the index PCR with Nextera XT index primers (Illumina) according to the following thermocycling protocol: 3 min at 95°C, 8 cycles of 30 s at 95°C– 30 s at 55°C– 30 s at 72°C, and 5 min at 72°C. The indexed samples were purified using AMPure XP beads, quantified using the Qubit dsDNA high sensitivity kit (Life Technologies), and equal quantities from each sample were pooled. The resulting pooled library was quantified using the NEBNext library quantification kit (New England Biolabs) and sequenced using the v3 chemistry (301 bp paired-end reads). Raw sequence data have been deposited in the European Nucleotide Archive database of EMBL-EBI under accession number PRJEB21999.

Bioinformatics and statistical analyses

Raw paired-end Illumina reads were trimmed for 16S rRNA gene primer sequences using Cutadapt (https://cutadapt.readthedocs.org/en/stable/). Pre-processed sequence data were processed using the Quantitative Insights Into Microbial Ecology (QIIME) software suite [24]. Successfully joined sequences were quality filtered in QIIME using default settings. Then, sequences were clustered into OTUs on the basis of similarity to known bacterial sequences available in the Greengenes database (v13.8; http://greengenes.secondgenome.com/; 97% sequence similarity cut-off) using the UCLUST software; sequences that could not be matched to references in the Greengenes database were clustered de novo based on pair-wise sequence identity (97% sequence similarity cut-off). The first selected cluster seed was considered as the representative sequence of each OTU. Then, representative sequences were assigned to taxonomy using the UCLUST software. Singleton OTUs and ‘contaminant’ sequences (from no-DNA control samples) were removed prior to downstream analyses. Total sum normalisation (TSS) was applied followed by cumulative-sum scaling (CSS) to correct biases introduced by TSS, and log2 transformation to account for the non-normal distribution of taxonomic counts data. Statistical analyses were executed using the Calypso software [25] (cgenome.net/calypso/); samples were clustered using supervised Canonical Correspondence Analysis (CCA) (including ‘infection status’ as explanatory variable). Differences in bacterial alpha diversity (Shannon diversity) and richness between groups, as well as in the abundance of individual taxa, were evaluated using paired t-test. Beta diversity was calculated using weighted UniFrac distances and differences in beta diversity were calculated using PERMDISP (Permutational Analysis of Multivariate Dispersions) through the betadisper function [26]. Differences in the composition of the faecal microbiota between groups were assessed using the LEfSe (Linear discriminant analysis Effect Size) workflow [27], by assigning infection/treatment ‘groupings’ as comparison class. Metagenome functional contents were analysed using the software package PICRUSt (v1.0.0) to predict gene contents and metagenomic functional information [28]. Sequences were aligned to data available in the Greengenes database v.13.5 and OTUs were assigned using a 97% identity cut-off. The resulting OTU table was then imported into PICRUSt and used to derive relative Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway abundance [28]. Differences in KEGG pathway abundance between groups were assessed using ANOVA embedded in the software suite STAMP [29]. To minimise the risk of other variables confounding the results, binomial logistic multiple-regression models were applied to the dataset. The infection status of each study participant was used as a dependent variable and other factors including age, gender, village, education and occupation, as independent variables, including interaction terms, to identify any risk factors associated with helminth infection (cf. S1 Table).

Results

Of 76 human volunteers enrolled in this study, 11 were positive for hookworms and/or roundworms and/or whipworms (H+) (Table 1), while 27 were negative despite no prior anthelmintic treatment (H-) (Table 1 and S1 Table). A total of 38 subjects had received regular anthelmintic treatment (Ht) with pyrantel pamoate (Table 1 and S1 Table). Therefore, 11 samples from each of H- and Ht cohorts were selected, based on available metadata, for high-throughput sequencing of bacterial 16S rRNA and subsequent comparative analyses with samples from the H+ group (Table 1). Logistic multiple-regression models were applied to these samples, but none of the assessed independent variables had a significant association with infection status of the study participants. From these 33 samples, a total of 17,576,532 paired-end reads were generated (not shown) and subjected to further processing. A total of 3,694,717 high-quality sequences (per sample mean 111,960 ± 40,845) were retained after quality control. Rarefaction curves generated following in silico subtraction of low-quality and contaminant sequences indicated that the majority of faecal bacterial communities were represented in the remaining sequence data, thus allowing us to undertake further analyses. These sequences were assigned to 11,371 OTUs and 12 bacterial phyla.

The phyla Firmicutes (mean of 50.9%) and Bacteroidetes (mean of 39.2%) were predominant in all samples analysed, followed by the phyla Proteobacteria (mean of 3.6%) and Actinobacteria (mean of 3.0%) (S2 Fig). At the family level, Prevotellaceae (mean of 26.4%), Ruminococcaceae (mean of 24.7%), Lachnospiraceae (mean of 13.0%) and Bacteroidaceae (mean of 8.1%) were most abundant (S2 Fig). Bacteroidaceae were highly abundant in some of the Ht (n = 3) and H+ (n = 2) subjects, but only in one H- study participant; the same samples also showed a significant reduction in Prevotellaceae (S2 Fig). The species Prevotella copri was abundant in the microbiota of >75% volunteers in this study and made up 7–69% (mean 17.3%) of all microbe species in these samples (S3 Fig).

Subject faecal microbial communities were ordinated by CCA, which separated samples by infection or treatment status (Fig 2) (P = 0.05). No significant differences in OTU alpha diversity (Shannon) and richness were recorded between groups (P = 0.65, P = 0.13) (S4 Fig).

thumbnail
Fig 2. Supervised Canonical Correspondence Analysis (CCA) displaying the compositional distribution of the faecal microbiota between helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects.

https://doi.org/10.1371/journal.pone.0184719.g002

However, the H+ and Ht microbiota was significantly more variable (i.e. characterised by increased beta diversity) compared with H- subjects (P = 0.04) (Fig 3), which indicated differences in overall heterogeneity of species composition between sample groups, rather than in overall species composition.

thumbnail
Fig 3. Permutational Analysis of Multivariate Dispersions indicating differences in global microbial community composition of study subjects.

(A) helminth-positive (H+) and helminth-negative (H-), (B) regularly treated (Ht) and H-, and (C) H+ and Ht subjects.

https://doi.org/10.1371/journal.pone.0184719.g003

Analysis by LEfSe, also supported by ANOVA, identified differences in abundance of individual taxa at the phylum, class, order, family, genus and species level between the three groups (Fig 4). In particular, Verrucomicrobiae (Class), Verrucomicrobiales (Order), Verrucomicrobiaceae and Enterobacteriaceae (Family), Lactococcus, Akkermansia and a genus belonging to the Enterobacteriaceae (Genus) and Akkermansia muciniphila (Species) showed a trend towards increased abundance in H+ compared to the other two groups (Fig 4). Compared to H+, Leuconostocaceae and Bacteroidaceae (Family) and Bacteroides (Genus), were increased in H- and Ht, respectively (Fig 4). Compared to H+, Leuconostocaceae and Bacteroidaceae (Family) and Bacteroides (Genus) were increased in H- and Ht, respectively (Fig 4).

thumbnail
Fig 4. Differentially abundant faecal bacteria in helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects, based on LDA Effect Size (LEfSe) analysis.

Phylum (A), Class (B), Order (C), Family (D), Genus (E), Species (F) and OTUs (G). Taxa highlighted in black/white/green indicate an overrepresentation in H+ /H- /Ht, respectively.

https://doi.org/10.1371/journal.pone.0184719.g004

The same microbial metabolic and functional KEGG pathways were inferred by PICRUSt analysis in all three groups (S5 Fig). However, ‘lipid metabolism’ (P = 0.003), ‘rig-like receptor signalling pathway’ (P = 0.024), and ‘apoptosis’ (P = 0.04), were down-regulated in H+ compared with H-/Ht subjects, while the ‘biotin pathway’ was upregulated in H+ compared with H-/Ht (P = 0.008) (Fig 5).

thumbnail
Fig 5. Differences in relative abundance of KEGG pathways encoded in the faecal microbiota of helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects.

Biotin metabolism (A), apoptosis (B), RIG-I-like receptor signalling (C) and ether lipid metabolism (D); significant differences (< 0.05) are indicated with asterisks (*).

https://doi.org/10.1371/journal.pone.0184719.g005

Discussion

In this study, we analysed the effects of patent infections by parasitic helminths, as well as of repeated prophylactic administrations of anthelmintics, on the composition of the faecal microbiota of human volunteers from endemic areas of Sri Lanka. Bacterial sequence data generated showed that the gut microbiota of subjects enrolled in our study was predominantly composed by species of the phylum Firmicutes, followed by those of the phyla Bacteroidetes, Proteobacteria and Actinobacteria, irrespective of infection and/or treatment status. This observation is in agreement with data from previous studies of the effects of natural or experimental helminth infections on the composition of the human gut microbiota [8, 9, 15, 17]. In particular, at the genus level, a significant proportion of most samples analysed were represented by Prevotella spp., which reflects the findings from a previous study conducted in a cohort of Ecuadorean children infected by roundworms [8]. Prevotella spp. are known to play a key role in carbohydrate metabolism [30] and thus their expansion is likely due to a high carbohydrate/fibre diet, which is common in Sri Lanka [31]. An abundance of bacteria of the genus Prevotella has been linked to a concomitant decrease of Bacteroides spp., and vice versa, likely as a consequence of the metabolic differences between these two genera [30]. Indeed, in this study, Bacteroides spp. were abundant in the gut microbiota of subjects from the fishing villages Valalai and Kandakuliya, whose diet is typically high in animal-derived proteins and fats [30]. Nevertheless, Bacteroides were also abundant in samples from volunteers from Hanthana (tea estate). However, since our questionnaire did not include detailed questions on individual dietary habits, it was not possible to speculate whether diet-related factors may have caused the observed differences in the composition of the gut microbiota of Hanthana villagers.

In this study, CCA analysis clustered samples according to infection or treatment status. However, analyses of OTU richness (i.e. the number of species within a sample) and alpha diversity (i.e. a measure of sample richness and evenness, the latter being defined as the relative abundance of individual species within a sample) detected no significant differences between H+, H- and Ht individuals. While this observation supports the findings from a previous study carried out in a cohort of human volunteers experimentally infected with N. americanus [18], an investigation of the impact of naturally acquired helminth infections on the composition of the gut microbiota of a cohort of Ecuadorean children [8] resulted in contrasting results. Indeed, while sole infections by T. trichiura could not be associated with detectable changes in microbial richness and alpha diversity, the gut microbiota of subjects with concomitant infections by T. trichiura and A. lumbricoides displayed a notable decrease of the latter [8]. Nonetheless, a recent study conducted in a cohort of naturally helminth-infected indigenous Malaysians [9], as well as other studies in experimentally infected individuals [15, 17], indicated an increase in alpha diversity associated with parasite infections. It is plausible to hypothesise that these varying observations could be linked, for example, to differences in sample sizes, in the ‘baseline’ composition of the gut microbiota of subjects enrolled, in parasite species causing the infections (hookworms vs. whipworms vs. ascarids), and/or in the infection stage at which samples were collected (acute and chronic in case of experimental infections vs. ‘undefined’ in case of natural infections). Nevertheless, since a range of GI inflammatory diseases have been associated with a decrease in microbial diversity [3234], it has been hypothesised that a helminth-mediated increase in microbial alpha diversity may represent a potential mechanism by which helminths are able to suppress inflammation [9, 15, 17, 35]. This aspect deserves further investigation using pre-defined and standardised experimental set-ups.

Whilst we detected no differences in overall bacterial richness and alpha diversity between H+, H- and Ht groups, beta diversity was significantly increased in H+ and Ht subjects when each of these groups were individually compared to the uninfected H- cohort. Unlike alpha diversity, beta diversity provides a measure of the distance or dissimilarity between pairs or groups of samples [26]. An increased beta diversity has been previously observed in the gut microbiota of veterinary species, such as mice infected with Trichuris muris [36], rats infected with Hymenolepis diminuta [37], and goats infected with Haemonchus contortus [38]. In humans, the higher beta diversity observed in H+ individuals compared with the uninfected controls corroborates the hypothesis that helminth infections are accompanied by qualitative and quantitative changes in the composition of the host gut microbiota that are, based on data available thus far, inconsistent between individuals [15, 17, 18]. However, our data also suggests that anthelmintic treatment alone may be responsible for significant changes in the gut microbiota of human hosts. To date and to the best of our knowledge, no studies have shown a direct effect of anthelmintics on the composition the vertebrate gut microbiota. Nevertheless, pyrantel pamoate, the anthelmintic administered to subjects enrolled in our study, has been shown to affect, besides helminths, protozoans such as Giardia [39]. The microbiota profiling technique used in this study did not allow to investigate the effects that helminth infections or anthelmintic treatment exert on populations of commensal or pathogenic eukaryotes, and thus the hypothesis that repeated doses of pyrantel may have resulted in substantial modifications of such populations requires further testing.

Significant alterations of individual bacterial taxa were detected between H+, H- and Ht subjects. Amongst these, Akkermansia muciniphila (class Verrucomicrobiae) was significantly increased in H+ individuals when compared to uninfected subjects. A. muciniphila is an anaerobic bacterium commonly detected in the human gut microbiota, where it primarily degrades host mucins [40]. Both vertebrate and helminth mucins have been shown to play key roles in the complex network of interactions occurring at the helminth-host interface [41]. For instance, the surface coat of the infective larval stage of the canine roundworm Toxocara canis has been shown to express high levels of a mucin-like glycoprotein (TES-120) which is shed following its binding by host antibodies or immune cells, thus suggesting that these molecules play a major role in protecting the parasite from the host immune response [42]. On the other hand, a dramatically increased production of host mucins was observed in macaques experimentally infected with the whipworm T. trichiura [35], likely as a consequence of the onset of Th2-type immunity stimulated by the infection [43]. Therefore, it may be possible that our observation of increased populations of A. muciniphila may be a direct consequence of the surge in helminth- and host-derived mucins in infected subjects. Interestingly, previous studies have shown that A. muciniphila populations are reduced in individuals with severe appendicitis and inflammatory bowel disease (IBD), which led the authors to hypothesise an anti-inflammatory role for this bacterial species [4446]. Given the known anti-inflammatory properties of a range of helminth species [15, 17, 18, 35, 47, 48], the role of helminth-induced expansions in populations of mucin-degrading bacteria should be tested in future studies aimed at dissecting the causality of parasite-mediated suppression of inflammation.

Amongst the Firmicutes, the family Leuconostocaceae (order Lactobacillales) was significantly increased in H- subjects. These bacteria belong to the lactic acid bacteria, a major group of autochthonous microbes that reside in the gut of humans and animals and that exert immune-modulatory functions [49]. Lactic acid bacteria are known probiotics [49]; yet, recent investigations, by us and others, in humans experimentally or naturally infected by GI nematodes did not report significant associations between helminth infections and expanded populations of lactic acid bacteria [8, 9, 15, 17, 18]. Conversely, previous studies of murine models of nematode infections have shown a marked increase in populations of lactobacilli (belonging to the Lactobacillaceae, a family of lactic acid bacteria distinct from the Leuconostocaceae but with similar metabolic properties) in response to parasite establishment [13, 48, 50]. The specific groups of lactic acid bacteria shown to be associated with helminth infection are inconsistent between this and previous studies, which could be linked to host-specific responses to parasites; nevertheless, in the future, studies of parasite-microbiota interactions conducted on larger human and/or animal cohorts should particularly focus on this group of probiotics [49, 51].

In order to correlate data on the composition of the human gut microbiota in response to helminth infection and anthelmintic treatment to inferred changes in bacterial metabolism, we conducted a predictive metagenomics analysis using PICRUSt. Whilst inferred KEGG pathways were consistent across groups, as also shown in a similar investigation of the faecal microbiota of a helminth-infected community from Malaysia [9], ‘biotin metabolism’ was inferred to be upregulated in H+ individuals compared to H-. Biotin is a B-vitamin with key roles in gene expression, cell signalling and chromatin structure [52]; in particular, biotin dependent signalling pathways regulate the expression of genes with key biological functions, e.g. apoptosis and cell survival [52]. Indeed, a significant downregulation of the KEGG pathway ‘apoptosis’ was observed in H+ compared to H- volunteers. Overall, these findings emphasise that qualitative and quantitative compositional changes in helminth-infected individuals may be accompanied by significant alterations of the microbial metabolism which, in turn, may greatly impact host nutrition and immunity. Clearly, this data requires experimental validation using comprehensive metabolomics studies of the gut microbiota of helminth-infected hosts during acute and chronic infections.

Conclusion

Data from this study augments current knowledge of the effect that helminth infections and continued prophylactic treatment exert on the composition of the gut microbiota of the human host. However, inherent limitations may have impaired our ability to detect minor changes in populations of bacteria affected by parasites and/or anthelmintic treatment. Amongst these limitations, our relatively small sample size, dictated by the prevalence of helminth infections in the Sri Lankan community enrolled in this investigation, may have affected our statistical power; in addition, dietary variabilities, as well as differences in species of infecting helminths and parasite loads, while effectively representing a ‘real world’ scenario, may have introduced a range of confounding factors that, under the circumstances of this study, we were unable to fully evaluate. Nevertheless, we detected a significantly increased beta diversity in the microbiota of H+ compared with the H- counterpart, together with compositional changes in the gut microbiota of H+, H- and and Ht subjects, thus indicating a distinct effect of both helminth infection and continued prophylactic treatment on the host gut microbiota. In addition, we also identified potential microbial metabolic changes associated with helminth infections, which further emphasises the need for further investigations of the role/s that helminth-induced changes in bacterial metabolism play in the complex network of host-parasite interactions. Overall, our findings add valuable knowledge to the vast, and yet little explored, research field of parasite—microbiota interactions and will provide a basis for the elucidation of the role such interactions play in pathogenic and immune-modulatory properties of parasitic nematodes in both human and animal hosts.

Supporting information

S1 Fig. Questionnaire used for the collection of metadata from 76 human volunteers screened for the presence of patent infections by gastrointestinal nematodes.

https://doi.org/10.1371/journal.pone.0184719.s001

(EPS)

S2 Fig. Composition of the faecal microbiota of helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects at the Phylum (A) and Family (B) level.

https://doi.org/10.1371/journal.pone.0184719.s002

(EPS)

S3 Fig. Prevotella copri abundance in helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects.

https://doi.org/10.1371/journal.pone.0184719.s003

(EPS)

S4 Fig. Differences in overall taxonomic species richness (A) and diversity (B) between the faecal microbiota of helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects.

https://doi.org/10.1371/journal.pone.0184719.s004

(EPS)

S5 Fig. Relative abundances of KEGG pathways encoded in the gut microbiota of helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects, determined by inferred metagenomic analyses with PICRUSt.

https://doi.org/10.1371/journal.pone.0184719.s005

(EPS)

S1 Table. Metadata associated with helminth-positive (H+), helminth-negative (H-) and helminth-negative but regularly treated (Ht) subjects enrolled in the study.

https://doi.org/10.1371/journal.pone.0184719.s006

(XLSX)

Acknowledgments

We thank Drs Richard Dybowski and Piero Mastroeni (Department of Veterinary Medicine, University of Cambridge) for helpful discussions.

References

  1. 1. Brooker SJ, Nikolay B, Balabanova D, Pullan RL. Global feasibility assessment of interrupting the transmission of soil-transmitted helminths: a statistical modelling study. Lancet Infect Dis. 2015;15(8):941–50. pmid:25886799
  2. 2. Murray CJL, Vos T, Lozano R, AlMazroa MA, Memish ZA. Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2013;381(9867):628.
  3. 3. World Health Organization. Accelerating work to overcome the global impact of neglected tropical diseases—a roadmap for implementation. WHO 2012. http://www.emro.who.int/neglected-tropical-diseases/ntd-infocus/ntd-roadmap.html. Cited 22 March 2017.
  4. 4. Pearson MS, Tribolet L, Cantacessi C, Periago MV, Valero MA, Jariwala AR, et al. Molecular mechanisms of hookworm disease: stealth, virulence, and vaccines. J Allergy Clin Immunol. 2012;130(1):13–21. pmid:22742835
  5. 5. Truscott JE, Turner HC, Farrell SH, Anderson RM. Mathematical models of transmission, the impact of mass drug administration and transmission elimination criteria. Adv Parasitol. 2016;94:133–98. pmid:27756454
  6. 6. Albonico M, Engels D, Savioli L. Monitoring drug efficacy and early detection of drug resistance in human soil-transmitted nematodes: a pressing public health agenda for helminth control. Int J Parasitol. 2004;34(11):1205–10. pmid:15491582
  7. 7. Campbell SJ, Nery SV, McCarthy JS, Gray DJ, Soares Magalhaes RJ, Clements AC. A critical appraisal of control strategies for soil-transmitted helminths. Trends Parasitol. 2016;32(2):97–107. pmid:26795294.
  8. 8. Cooper P, Walker AW, Reyes J, Chico M, Salter SJ, Vaca M, et al. Patent human infections with the whipworm, Trichuris trichiura, are not associated with alterations in the faecal microbiota. PLoS One. 2013;8(10):e76573. pmid:24124574
  9. 9. Lee SC, Tang MS, Lim YA, Choy SH, Kurtz ZD, Cox LM, et al. Helminth colonization is associated with increased diversity of the gut microbiota. PLoS Negl Trop Dis. 2014;8(5):e2880. pmid:24851867
  10. 10. Glendinning L, Nausch N, Free A, Taylor DW, Mutapi F. The microbiota and helminths: sharing the same niche in the human host. Parasitology. 2014;141(10):1255–71. pmid:24901211
  11. 11. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012;336(6086):1268–73. pmid:22674334
  12. 12. Brown EM, Sadarangani M, Finlay BB. The role of the immune system in governing host-microbe interactions in the intestine. Nat Immunol. 2013;14(7):660–7. pmid:23778793
  13. 13. Reynolds LA, Smith KA, Filbey KJ, Harcus Y, Hewitson JP, Redpath SA, et al. Commensal-pathogen interactions in the intestinal tract: lactobacilli promote infection with, and are promoted by, helminth parasites. Gut Microbes. 2014;5(4):522–32. pmid:25144609
  14. 14. Zaiss MM, Rapin A, Lebon L, Dubey LK, Mosconi I, Sarter K, et al. The intestinal microbiota contributes to the ability of helminths to modulate allergic inflammation. Immunity. 2015;43(5):998–1010. pmid:26522986
  15. 15. Giacomin P, Zakrzewski M, Jenkins TP, Su X, Al-Hallaf R, Croese J, et al. Changes in duodenal tissue-associated microbiota following hookworm infection and consecutive gluten challenges in humans with coeliac disease. Sci Rep. 2016;6:36797. pmid:27827438
  16. 16. Giacomin P, Agha Z, Loukas A. Helminths and intestinal flora team up to improve gut health. Trends Parasitol. 2016;32(9):664–6. pmid:27234811
  17. 17. Giacomin P, Zakrzewski M, Croese J, Su X, Sotillo J, McCann L, et al. Experimental hookworm infection and escalating gluten challenges are associated with increased microbial richness in celiac subjects. Sci Rep. 2015;5:13797. pmid:26381211
  18. 18. Cantacessi C, Giacomin P, Croese J, Zakrzewski M, Sotillo J, McCann L, et al. Impact of experimental hookworm infection on the human gut microbiota. J Infect Dis. 2014;210(9):1431–4. pmid:24795483
  19. 19. Gunawardena K, Kumarendran B, Ebenezer R, Gunasingha MS, Pathmeswaran A, et al. Soil-transmitted helminth infections among plantation sector schoolchildren in Sri Lanka: Prevalence after ten years of preventive chemotherapy. PLoS Negl Trop Dis. 2011;5(9):e1341. pmid:21980549
  20. 20. IIsmail MM, Atukorala TMS, Naotunne TS, de Silva NR, Hettiarachchi I. Control of intestinal helminthiasis in pregnancy–the Sri Lankan experience. In: Crompton DWT, Montresor A, Nesheim MC, Savioli L, editors. Controlling disease due to helminth infections. Geneva: World Health Organization; 2003. pp. 127–133.
  21. 21. Perera P, Rajapakse R, Rajakaruna R. Gastrointestinal parasites of dogs in Hantana area in the Kandy District. JNSF. 2013;41(2).
  22. 22. Frank W, Lieder J. Parasitologie: transparente-Atlas. 1st ed. Hageman; 1987.
  23. 23. Klindworth A, Pruesse E, Schweer T, Peplies J, Quast C, Horn M, et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 2013;41(1):e1. pmid:22933715
  24. 24. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK. QIIME allows analysis of high-throughput community sequencing data. Nature Meth. 2010;7. pmid:20383131
  25. 25. Zakrzewski M, Proietti C, Ellis JJ, Hasan S, Brion MJ, Berger B, et al. Calypso: a user-friendly web-server for mining and visualizing microbiome-environment interactions. Bioinformatics. 2017;33(5):782–3. pmid:28025202
  26. 26. Anderson MJ, Ellingsen KE, McArdle BH. Multivariate dispersion as a measure of beta diversity. Ecol Lett. 2006;9(6):683–93. pmid:16706913
  27. 27. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12. pmid:21702898
  28. 28. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotech. 2013;31(9):814–21. pmid:23975157
  29. 29. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics. 2014;30(21):3123–4. pmid:25061070
  30. 30. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science. 2011;334(6052):105–8. pmid:21885731
  31. 31. Jayawardena R, Byrne NM, Soares MJ, Katulanda P, Hills AP. Food consumption of Sri Lankan adults: an appraisal of serving characteristics. Public Health Nutr. 2013;16(4):653–8. pmid:22784794
  32. 32. Sepehri S, Kotlowski R, Bernstein CN, Krause DO. Microbial diversity of inflamed and noninflamed gut biopsy tissues in inflammatory bowel disease. Inflamm Bowel Dis. 2007;13(6):675–83. pmid:17262808
  33. 33. Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, Pelletier E, Frangeul L, et al. Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach. Gut. 2006;55(2):205–11. pmid:16188921
  34. 34. Ott SJ, Musfeldt M, Wenderoth DF, Hampe J, Brant O, Folsch UR, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53(5):685–93. pmid:15082587
  35. 35. Broadhurst MJ, Ardeshir A, Kanwar B, Mirpuri J, Gundra UM, Leung JM, et al. Therapeutic helminth infection of macaques with idiopathic chronic diarrhea alters the inflammatory signature and mucosal microbiota of the colon. PLoS Pathog. 2012;8(11):e1003000. pmid:23166490
  36. 36. Holm JB, Sorobetea D, Kiilerich P, Ramayo-Caldas Y, Estelle J, Ma T, et al. Chronic Trichuris muris infection decreases diversity of the intestinal microbiota and concomitantly increases the abundance of lactobacilli. PLoS One. 2015;10(5):e0125495. pmid:25942314
  37. 37. McKenney EA, Williamson L, Yoder AD, Rawls JF, Bilbo SD, Parker W. Alteration of the rat cecal microbiome during colonization with the helminth Hymenolepis diminuta. Gut Microbes. 2015;6(3):182–93. pmid:25942385
  38. 38. Li RW, Li W, Sun J, Yu P, Baldwin RL, Urban JF. The effect of helminth infection on the microbial composition and structure of the caprine abomasal microbiome. Sci Rep. 2016;6:20606. pmid:26853110
  39. 39. Hausen MA, Menna-Barreto RF, Lira DC, de Carvalho L, Barbosa HS. Synergic effect of metronidazole and pyrantel pamoate on Giardia lamblia. Parasitol Int. 2011;60(1):54–8. pmid:20946968
  40. 40. Derrien M, Collado MC, Ben-Amor K, Salminen S, de Vos WM. The Mucin degrader Akkermansia muciniphila is an abundant resident of the human intestinal tract. Appl Environ Microbiol. 2008;74(5):1646–8. pmid:18083887
  41. 41. Theodoropoulos G, Hicks SJ, Corfield AP, Miller BG, Carrington SD. The role of mucins in host-parasite interactions: Part II—helminth parasites. Trends Parasitol. 2001;17(3):130–5. http://dx.doi.org/10.1016/S1471-4922(00)01775-X pmid:11286796
  42. 42. Gems D, Maizels RM. An abundantly expressed mucin-like protein from Toxocara canis infective larvae: the precursor of the larval surface coat glycoproteins. Proc Natl Acad Sci U S A. 1996;93(4):1665–70. pmid:8643687
  43. 43. Wolff MJ, Broadhurst MJ, Loke P. Helminthic therapy: improving mucosal barrier function. Trends Parasitol. 2012;28(5):187–94. pmid:22464690
  44. 44. Swidsinski A, Dorffel Y, Loening-Baucke V, Theissig F, Ruckert JC, Ismail M, et al. Acute appendicitis is characterised by local invasion with Fusobacterium nucleatum/necrophorum. Gut. 2011;60(1):34–40. pmid:19926616
  45. 45. Caesar R, Tremaroli V, Kovatcheva-Datchary P, Cani PD, Backhed F. Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signaling. Cell Metab. 2015;22(4):658–68. pmid:26321659
  46. 46. Jeffery IB, Quigley EM, Ohman L, Simren M, O'Toole PW. The microbiota link to irritable bowel syndrome: an emerging story. Gut Microbes. 2012;3(6):572–6. pmid:22895081
  47. 47. Ramanan D, Bowcutt R, Lee SC, Tang MS, Kurtz ZD, Ding Y, et al. Helminth infection promotes colonization resistance via type 2 immunity. Science. 2016;352(6285):608–12. pmid:27080105
  48. 48. Walk ST, Blum AM, Ewing SA, Weinstock JV, Young VB. Alteration of the murine gut microbiota during infection with the parasitic helminth Heligmosomoides polygyrus. Inflamm Bowel Dis. 2010;16(11):1841–9. pmid:20848461
  49. 49. Kekkonen RA, Kajasto E, Miettinen M, Veckman V, Korpela R, Julkunen I. Probiotic Leuconostoc mesenteroides ssp. cremoris and Streptococcus thermophilus induce IL-12 and IFN-gamma production. World J Gastroenterol. 2008;14(8):1192–203.
  50. 50. Rausch S, Held J, Fischer A, Heimesaat MM, Kuhl AA, Bereswill S, et al. Small intestinal nematode infection of mice is associated with increased enterobacterial loads alongside the intestinal tract. PLoS One. 2013;8(9):e74026. pmid:24040152
  51. 51. Yoo JY, Kim SS. Probiotics and prebiotics: Present status and future perspectives on metabolic disorders. Nutrients. 2016;8(3):173. pmid:26999199
  52. 52. Zempleni J. Uptake, localization, and noncarboxylase roles of biotin. Annu Rev Nutr. 2005;25:175–96. pmid:16011464