Skip to main content

Worms and bugs of the gut: the search for diagnostic signatures using barcoding, and metagenomics–metabolomics

Abstract

Gastrointestinal (GI) helminth infections cause significant morbidity in both humans and animals worldwide. Specific and sensitive diagnosis is central to the surveillance of such infections and to determine the effectiveness of treatment strategies used to control them. In this article, we: (i) assess the strengths and limitations of existing methods applied to the diagnosis of GI helminth infections of humans and livestock; (ii) examine high-throughput sequencing approaches, such as targeted molecular barcoding and shotgun sequencing, as tools to define the taxonomic composition of helminth infections; and (iii) discuss the current understanding of the interactions between helminths and microbiota in the host gut. Stool-based diagnostics are likely to serve as an important tool well into the future; improved diagnostics of helminths and their environment in the gut may assist the identification of biomarkers with the potential to define the health/disease status of individuals and populations, and to identify existing or emerging anthelmintic resistance.

Graphical Abstract

Background

Gastrointestinal (GI) helminths cause significant disease in both humans and animals. In humans, soil-transmitted helminths (STHs; including Ascaris lumbricoides, Trichuris trichiura, Necator americanus, Ancylostoma duodenale, Ancylostoma ceylanicum and Strongyloides stercoralis), blood flukes (Schistosoma spp.) and food-borne liver flukes (e.g. Clonorchis and Opisthorchis spp.) afflict > 1.5 billion people globally, most of whom live in disadvantaged and neglected communities [1,2,3,4]. Children are particularly vulnerable to the clinical consequences of STH infections, which include stunted growth, malnutrition and/or anaemia [5], whilst fluke infections can ultimately lead to GI, hepatosplenic or urogenital diseases (Schistosoma spp.) [1] and/or cancers (Schistosoma haematobium, Clonorchis sinensis and Opisthorchis viverrini) [6]. These helminths contribute to a tangible reduction in quality of life, equating to > 5 million disability-adjusted life years (DALYs) for STH-infected persons [4] and > 3 million DALYs for adults infected with schistosomiasis [7]. In addition, species of GI helminths of livestock represent a major threat to food security and animal health. In Europe alone, the cost of infections by GI helminths (including the ‘barber’s pole worm’ Haemonchus contortus, the ‘brown stomach worm’ Teladorsagia circumcincta and the ‘liver fluke’ Fasciola hepatica) has been estimated at €1.8 billion due to production losses, the cost of treatment programmes and death [8].

Traditionally, the fight against parasitic helminths of both human and animal health importance has almost exclusively relied on the use of anthelmintics, and will continue to do so for some time, as no viable alternatives are yet available at the scale needed for effective prevention or control. For human-infective helminths, anthelmintics are distributed primarily via coordinated periodic mass drug administration (MDA) campaigns [5]. For GI helminths of livestock, large-scale and wide-reaching preventative treatment of animals is frequently undertaken; however, the management of anthelmintic use, in terms of drug class [9], dosage and route [10], and timing [11], can vary markedly between or within countries and regions [11,12,13]. Although anthelmintic treatment is relatively effective, some challenges exist, including: (i) variable efficacy against different worm species and developmental stages; (ii) reinfection soon after treatment [14,15,16]; and (iii) the evolution of drug resistance [8, 13, 17, 18]. Anthelmintic resistance has evolved as a consequence of the excessive use of single drug classes. In some nematode species of livestock, for example H. contortus, resistance is widespread to the point that farming has become unviable in some regions of the world [19, 20]. Although the nature and extent of resistance in human-infective helminths is less understood compared with that in helminths of veterinary significance [21,22,23], the extensive use of anthelmintics via MDA does put a significant selection pressure on human-infective helminth populations. Thus, the monitoring of drug efficacy should form part of an effective control programme, and improved management strategies need to be implemented to preserve drug efficacy and/or overcome resistance. Although considerable efforts have attempted to develop vaccines against helminths [24,25,26], no commercial vaccines are in current use against STHs of humans (e.g. [27, 28]), whilst only two vaccines are available for use against GI helminths of livestock, namely, Barbervax® and Wirevax® (Wormvax Australia Pty Ltd.) against H. contortus (with short-term protection) in Australia and South Africa, and one vaccine for use against a non-GI helminth, namely, Providean Hidatil EG 95® (Tecnovax) against cystic hydatidosis caused by Echinococcus granulosus in South America [28, 29].

Accurate diagnosis remains a critical component of monitoring parasite infections and their response to anthelmintic treatments. The diagnosis of GI helminth infections in humans and live animals usually relies on the microscopic detection/identification of eggs or larvae in faecal samples by faecal smear, faecal flotation and/or larval culture. These methods are used to assess the parasite species present and/or estimate the intensities of infections within individuals, groups or populations [30]. While routinely used, these conventional techniques can be relatively labour intensive and time consuming to perform (unless automated), and their diagnostic performance depends on sample preparation, the reproductive potential of a worm species and/or infection prevalence [31,32,33]. Molecular methods have been developed to attempt to overcome these limitations [34,35,36,37,38,39] but, to date, they have been used mainly to address academic research questions and there has been limited uptake in large-scale control programmes. The World Health Organisation (WHO) revised the neglected tropical disease (NTD) roadmap (2021–2030) [40], which sets out a strategic vision to control and ‘eliminate’ 20 NTDs afflicting humans, including STHs. As the control of STHs relies heavily on MDA, consideration must be given to which strategy is best suited to achieving a substantial, measurable and sustained reduction or elimination of transmission, and how novel diagnostics approaches can support sound surveillance strategies [34, 41].

In the present review, we: (i) examine the strengths and weaknesses of conventional methods currently available for the diagnosis of GI helminth infections of humans and animals; (ii) appraise high-throughput molecular barcoding and metagenomic sequencing as tools to determine the taxonomic composition of helminth infections; and (iii) review current knowledge of the interactions between helminths and microbiota in the host gut. The overall aim is to provide a basis to identify biomarkers with the potential to define the health/disease status of individuals and populations, as well as to detect existing or emerging anthelmintic resistance.

Conventional diagnostic methods

Coproscopic techniques

Most commonly, microscopy-based methods used to examine faecal samples for helminth eggs include: direct examination of stool smears on a glass slide, such as the Kato Katz technique commonly used to detect STHs and schistosome infections [42]; flotation using a counting chamber (e.g. McMaster), commonly used for detection of infections by GI helminths in livestock; and the morphological identification of larvae following coproculture [43]. Faecal flotation methods often include a centrifugation step to concentrate eggs from samples, such as with the FLOTAC/mini-FLOTAC technique [44, 45]. These methods have been reviewed extensively in previous articles [31, 46, 47].

Although these methods are affordable and widely used around the world, they are not without limitations. The enumeration of eggs per gram (i.e. faecal egg counts [FEC]) is often used to estimate infection intensity, but this relationship is only valid for parasites with high reproductive potential (e.g. hookworms and Haemonchus spp.) [46, 48]. This approach is useful in endemic areas and/or settings where the prevalence of infections is high, but evidence indicates that diagnostic sensitivity can be low (43–52% for direct microscopy, depending on STH species) [31], resulting in underestimations of prevalence in field-based studies (cf. [35, 37, 49,50,51,52]). Moreover, the specificity of the diagnosis is reliant on the knowledge and skills of the diagnostician, i.e. their ability to conduct the assay in a repeatable/reproducible manner and to accurately identify eggs to genus or species within a relatively short time-frame to overcome challenges linked to egg degradation or hatching. Approaches to automate the analysis of microscopy-based helminth egg detection, including egg concentration through filtration [53], are under development and becoming available, such as the digitized microscopy tool FECPAKG2 [54, 55] or Lab-On-A-Disk (LOD), the latter of which combines microfluidics-based separation of helminth eggs [56] with machine-learning for egg identification (cf. Trichuris eggs [57]). Whilst the FECPAKG2 potentially has a higher throughput than conventional microscopy and does not require a specialist, it necessitates cloud/internet access to make results immediately available [55]. Currently, this tool is used for the diagnosis of livestock helminth infections [55, 58], but the limited sensitivity of FECPAKG2–based diagnosis indicates that it will not be suitable for the detection of human STHs in geographical areas with medium or low prevalence of infections [51, 54]. Machine learning-based methods have also been evaluated and applied for the diagnosis of helminthiases (e.g. [59,60,61,62,63]). However, the utility of these recent technological developments for the diagnosis of human helminth infections will depend on their performance in large-scale field surveys and on their cost and benefits [56].

Coproantigen detection

Immunodiagnostic tools detect parasite antigens in faeces (coproantigens). Enzyme-linked immunosorbent assays (ELISAs) that target parasites affecting companion animals (Toxocara canis, Ancylostoma caninum and Trichuris vulpis) [64], the human STHs An. ceylanicum [65] and A. lumbricoides [66] and the cestode Taenia solium [67] have been developed; although, for the latter, cross-reactivity with other cestode species appears to be an issue [68]. ELISA kits are also commercially available; for example, for the detection of liver fluke (F. hepatica) antigens from faeces of both livestock [69] and humans [70]. Results achieved by ELISAs show a strong correlation between coproantigen and copro-DNA levels in stools from individuals with Ascaris infection [66], while the correlations between microscopy-based worm burden and real-time (quantitative PCR [qPCR]) are moderate for hookworm [35, 52] or strong for A. lumbricoides [35, 52] and T. trichiura [52]. It is important to consider that the performance of ELISAs for the detection of coproantigen can be affected by several components in stool samples, including salts, proteases, antibodies and organic compounds, and are dependent on sample preservation and storage; variations in any of these might interfere with antigen detection and lead to false positive or negative results [70]. This also applies to nucleic acid-based methods.

Nucleic acid detection

Owing to their high analytical sensitivity and specificity [35, 52], nucleic acid amplification techniques (NAATs) are increasingly being used for the copro-diagnosis of helminth infections of humans and animals [34, 36, 71, 72]. NAATs aim to specifically target helminth DNA or RNA in extracts from stool samples via the binding of complementary oligonucleotide primers and subsequent enzymatic amplification of target DNA/RNA. The application of NAATs is important for the detection of helminth infections in low-prevalence settings [34]. Following the WHO’s updated targets for eliminating NTDs (including STHs), qPCR, irrespective of target, is increasingly being recognized as an important diagnostic tool to support both elimination and surveillance efforts [34, 73].

Currently, most of the available PCR-derivative tools target only a small number of helminth species, require suitable laboratory infrastructure (with electricity) and equipment and are usually low throughput, although a few multiplexed attempts offer the advantage of a somewhat higher throughput [38, 74, 75]. These constraints make them less suited to field application settings [76]. To begin to address this issue, recombinase polymerase amplification (RPA) in portable battery-operated instruments or loop-mediated isothermal amplification (LAMP)-based tools have been established for field use. Examples include RPA of S. haematobium DNA from genomic DNA from urine [77] and LAMP detection of STHs from nucleic acids extracts from adult worms [78]. To date, most NAATs are based on the amplification of repetitive sequence elements in the genomes of helminths, such as species-specific ribosomal RNA (rRNA) genes [39, 74, 79] or genome-wide, tandem repeats [36, 80].

It appears that high analytical sensitivity and specificity can be achieved using such targets. However, it is not yet known to what extent variation in nucleotide sequence, and/or copy number of repetitive elements, might affect the diagnostic performance of these assays in different geographical regions. This requires critical evaluation.

Barcoding of helminths by high-throughput sequencing

DNA barcoding is a method for the identification of species using a relatively short DNA region(s) of a gene or genes [81]. For a barcode to be useful, two basic criteria need to be met: (i) the barcode itself must contain sequence variants that differentiate between species, and this variation need to be markedly and consistently higher than levels of variation within individual species, irrespective of the geographical origin of a sample/specimen [82], and (ii) the regions flanking a barcode should be sufficiently conserved between species to allow the design of oligonucleotide primers that consistently bind (irrespective of species) to enable efficient DNA amplification by PCR. Identifying truly universal barcoding targets and primers targeting all helminth species is an ambitious goal yet to be achieved.

For a range of animal groups, DNA barcodes derived from the mitochondrial cytochrome c oxidase subunit 1 (cox1) gene [83, 84] and nuclear rRNA genes (18S or 28S and variable regions within such genes, such as D4-D6 of 28S rDNA and the V4 or V9 domains of 18S rDNA) are commonly used [85,86,87,88,89]. However, it is often (incorrectly) assumed that DNA barcodes such as those described above are ‘reliable’ or ‘useful’, without prior validation for new groups of organisms. For example, mitochondrial DNA (mtDNA) sequences of parasitic nematodes (including cox1) display high levels of synonymous sequence variation within species that limits their utility as barcodes, despite high levels of protein sequence conservation [89, 90]. On the other hand, nuclear rRNA genes (while useful for phylogenetic studies) [91, 92] are usually too conserved in sequence for reliable species identification. The second internal transcribed spacer of nuclear ribosomal DNA (ITS-2) is the only barcode that has been rigorously evaluated as a species marker for selected nematodes (i.e. Strongylida and Ascaridida) [93, 94] as well as for some trematode species [95]; usually, most barcodes have not been critically assessed for widespread application to a broad range of taxonomic groups. The extensive, early taxonomic evaluations of ITS-2 for select nematode groups [94] has paid off, and ITS-2 barcoding by high-throughput sequencing (HTS), enabled by database resources and informatics [96], is now well established for strongylid nematodes to define ‘nemabiomes’ [97,98,99,100]. This is not the case for other barcodes, as they do not provide reliable/accurate species markers.

Defining an informative barcode(s) for a wide range of species can be challenging because of a lack of knowledge of the genomes of many species and their taxonomic/phylogenetic relationships. Thus, designing primers to amplify the same barcode from diverse taxa can be problematic [90]. The broader the target group (e.g. family, order and/or phylum), the greater the likelihood that primers designed to the barcode will hybridize and amplify non-specifically in non-target regions of non-target taxa. Thus, it is important that barcodes, associated reagents (particularly primers) and protocols for amplification and sequencing are critically evaluated experimentally prior to being used in large-scale studies, rather than simply relying on barcodes and primers that have been identified or designed in silico alone, as databases do not represent the broader biodiversity [101, 102].

Metabarcoding to capture helminth diversity

HTS and metabarcoding allow for the targeted amplification of multiple, homologous regions from phylogenetically diverse taxa simultaneously in a mixed sample, and can determine the relative abundance of these regions, as compared to traditional barcoding with its limited capacity to target one species per sample [103, 104]. Conventional barcoding approaches comprise expensive and laborious sequencing steps. However, the marked decrease in cost and increase in accessibility of HTS over the last decade has encouraged novel approaches for the diagnosis or analysis of helminth infections or populations [84, 86,87,88, 91, 92, 97,98,99,100, 105,106,107,108,109]. The sheer volume of sequence output from ‘metabarcoding’ accommodates the use of conserved primers to “capture” and sequence these DNA regions from a diverse range of organisms, with downstream bioinformatics allowing the characterization of species/taxon identity and sequence diversity (both within and among taxa) within a sample. Whilst metabarcoding may provide the relative proportions of species present in a complex sample, currently established pipelines do not yield absolute/measuring values that correspond with the species present. This may be a disadvantage relative to conventional tools, such as microscopy and qPCR, if metabarcoding is proposed to be a diagnostics alternative. In addition, due to its nature of amplifying multiple barcodes simultaneously, it is unclear as to whether the diverse barcodes present in a sample are being amplified and sequenced equally efficiently. Some research groups have accounted for this by including ‘mock parasite populations’ in sequencing runs to normalize read counts and correct species representation bias [97].

A number of target genes have been proposed and tested via metabarcoding approaches to characterize helminths. Based on in silico work, mitochondrial 12S and 16S rRNA genes have been proposed as promising candidates for the metabarcoding of parasitic nematodes [110]; however, one key species of STH, T. trichiura (whipworm), was not included in this study. Thus, the utility of 12S/16S rRNA genes as universal barcodes remains to be rigorously assessed and established. Nevertheless, the mainstay of metabarcoding has focused on adapting ribosomal barcoding primers, most commonly ITS-2. A recent comparative analysis of partial mitochondrial 12S and 16S rDNA regions versus partial mitochondrial cox1 regions, and of the nuclear genes ITS-1, ITS-2 and 18S rDNA revealed that, although both approaches allow the successful identification of a wide range of nematode species, ITS-2 is best suited to specific identification [110]. Importantly, sequence differences in ITS-2 between species is consistently higher than the low levels of nucleotide variation within species. For these reasons, a reference database of ITS-2 sequences has been established [96] to aid metabarcoding studies of strongylid nematodes.

Metabarcoding using nuclear 18S rDNA and/or ITS-2 has been successfully applied to the specific identification of nematodes from copro-DNA from livestock [97,98,99,100] and wildlife animals [86, 107, 111,112,113,114] and across a range of roundworm species, including Ascaridia, Heligmosomoides, Nippostrongylus and Strongyloides [86], Haemonchus, Cooperia and Trichostrongylus [100], and/or cestodes (e.g. Hymenolepis diminuta) in wildlife [86] and livestock animals [97,98,99,100]. Markers in the 28S rRNA gene have been applied to identify common nematodes of rodents (including Heligmosomoides, Nippostrongylus and Strongyloides) via metabarcoding using faecal DNA [86].

This same concept has been recently expanded to characterize drug-resistant genetic variants [115,116,117], and offers a promising proof-of-concept to detect new variants for resistance or other genetic markers of traits of interest.

‘Shotgun’ metagenomic sequencing

The composition of samples, such as those derived from faeces, can be very complex, containing nucleic acids from a wide range of organisms, spanning species represented in food, the individual’s genome and a wide range of viruses, microbes and pathogens, including parasites [118]. Rather than focusing on a discrete target sequence as described above, shotgun metagenomics aims to use HTS to detect all nucleotide sequences (DNA or RNA) present in a given sample.

Metagenomics tools, in particular when used in epidemiological contexts, might provide additional value over conventional diagnostic approaches. In addition to revealing polyparasitism, such approaches may also reveal genetic variation within and among samples; this information could be of value by inferring the relationships of pathogens/parasites/microbes and can be employed to explore population dynamics over time. Increasingly, metagenomics is being used in applications where the molecular signals for parasites are weak or difficult to resolve. For example, in archaeoparasitology, DNA recovered from ancient settlements, human remains and mummies has been analysed using short genomic regions of up to 400 bp [119, 120], from which mitogenomes have been assembled [121]. Other DNA-based, shotgun metagenomic studies of sediments, combined with gene-specific 18S rRNA gene metabarcoding, revealed Ascaris and Trichuris spp. eggs present in a cesspit of a nineteenth century palace [119]. Strikingly, whole-genome sequencing analysis has been used to resolve the parasite genomes of preserved T. trichiura eggs up to 1000 years old, the oldest eukaryotic pathogens sequenced to date [122].

Although metagenomic-based tools yield substantially more sequence data per sample, a major limitation is that such approaches are relatively more expensive, both in terms of infrastructure, equipment and cost per sample, even when a pooling strategy is applied. Furthermore, high technical skills (i.e. for bioinformatics processing of sequence data) and computational capacity (e.g. cloud-based or physical computational capacity for performing analysis) are usually required, both of which require extensive expertise and further expense. In the short term, this may require outsourcing to centralized facilities or commercial companies; however, capacity is growing throughout the world, particularly in hubs in low- to middle- income countries and will likely enable the development of suitable technologies and expertise in endemic countries over time. One significant advantage is that, once established, these technologies (and particularly metabarcoding) have the capacity to scale to very high throughput, which will help markedly reduce the costs per sample. Further cost reductions could be achieved by performing low coverage “genome skimming”, which specifically targets high copy number regions of genomes, such as repetitive elements in nuclear DNA and in mitochondrial DNAs [123], which could allow metagenomics to be performed at scale. Examples of the application of metagenomics or metabarcoding for the identification of helminth nucleic acids in faecal extracts and/or faecal cultures from humans and animal species are summarized in Table 1.

Table 1 Examples of metabarcoding and metagenomics approaches for the detection of helminth species in faecal samples or cultures

In instances where shotgun metagenomics approaches are used for the genetic identification and characterization of parasites, the number of sequence reads derived from the parasites within a given dataset might be substantially smaller than those originating from host and microbiome within the gut. Dependent on the aim and application, parasite-derived DNA and sequencing reads may be enriched by: (i) filtering the sample to collect worm eggs prior to DNA extraction; (ii) targeting parasite DNA via selective/whole genome amplification [124]; and/or (iii) bioinformatic selection of parasite sequences prior to downstream analyses (via, for instance, mapping to reference genomes when available). Recent genomic investigations, databases and resources underpin metagenomic investigations. In particular, the 50 Helminth Genomes Project [125] has generated extensive genomic resources for many important helminths of human and veterinary importance, and open-access data repositories, such as WormBase ParaSite [126]. The Earth BioGenome Project [127], which aims to sequence all eukaryotic life on earth, also promises to improve resources for helminths to support metagenomic analyses in the future.

One important feature of sequence data generated using shotgun metagenomic approaches is that it may provide end-users with the opportunity to not only identify known or possible pathogens within faecal samples, but also to simultaneously genetically characterize the microbial communities from within the GI tract of the vertebrate host and to explore interactions among parasites, microbes and the host.

Metabarcoding and/or metagenomics to define microbial signatures that associate with helminth-infections

Over the last decade, evidence has emerged that infections by soil-transmitted and GI helminths are accompanied by profound qualitative and quantitative modifications in the composition of the faecal microbiome of their hosts, although, to date, studies have mainly focused on the host bacteriome [128,129,130], rather than the virome [131] or mycobiome [132]. Modifications of the bacterial composition of the host microbiome can be characterized using microbiota-targeting metabarcoding and/or metagenomic approaches. Metabarcoding sequencing of one or more hypervariable regions of the bacterial 16S rRNA gene has been applied often to characterize faecal microbial profiles of both humans and animals infected by parasitic helminths [129, 133,134,135], both under experimental [136,137,138] and natural conditions of infection [139,140,141,142,143], and before and after anthelmintic treatment [143, 144]. Examples include the characterization of the faecal microbiota of cohorts of human volunteers with coeliac disease or multiple sclerosis experimentally infected with N. americanus [137, 145], and individuals naturally infected by STHs [142,143,144,145,146]. In relation to nematode species of veterinary importance, metabarcoding, using the bacterial 16S rRNA gene, has been applied to, for example, the characterization of the faecal microbiota of H. contortus-infected goats [147], T. circumcincta-infected sheep [148], or Ostertagia ostertagi-infected cattle [149].

Information on specific qualitative and/or quantitative alterations of microbial taxa in faecal samples from helminth-infected humans or animals have been reviewed elsewhere [128,129,130]. Often, direct comparisons of faecal microbial profiles of helminth-infected humans and/or animals compared with corresponding uninfected hosts lead to contrasting results (reviewed in [129]), even between data sets generated from the same host–parasite pair under similar experimental conditions [148]. Such discrepancies are likely linked to variability in structure and composition of the vertebrate gut microbiota [130, 134, 150]. Furthermore, intrinsic and/or extrinsic factors, including, but not limited to, age, gender, diet and/or underlying health conditions, all exert profound effects on the composition of the vertebrate gut microbiota [128, 129, 143]. Such inter-individual variability makes the discovery of reliable microbial-based biomarkers of helminth infection a challenging task; nevertheless, in a recent study, specific biomarker-discovery algorithms were applied to the identification of unique populations of faecal bacteria associated with worm colonization in T. circumcincta-infected sheep in an experimental system [148]. Whilst this study represents a promising step forward in the discovery of microbiota/microbiome-targeting biomarkers of helminth infections in both humans and animals, individual investigations examining the impact of worm colonization on gut microbial communities are often compromised by insufficient sample size and, thus, low statistical power [129]. A possible solution is to combine available datasets generated from a wide range of host–parasite pairs [151] and, in turn, the number of faecal samples needed to achieve sufficient statistical power to underpin robust biomarker discovery studies. Understanding the interaction between helminth infections and the host microbiome could unveil microbial signatures associated with such infection(s) (or not). Some recent studies indicate that species-specific microbial signatures are associated with helminth infections and worm burden [142], whilst a study in Cameroon showed how bacterial signatures can predict polyparasitism with 81–82% accuracy for A. lumbricoides or T. trichiura [152]. The same study demonstrated how diet and lifestyle are associated with different microbial signatures. In turn, selected microbiome populations could confer resistance or susceptibility to infection [141, 146, 150], or even act as a means of prophylaxis and treatment (e.g. by means of probiotics). Clearly, faecal sample extracts are ideal specimens for non-invasive biomarker-based diagnosis of helminth infections, as they contain end-products of host, parasite and/or microbiome metabolisms, and/or of the crosstalk between them; these products could represent an untapped source of potential diagnostic markers.

The prospect of metabolites as biomarkers

In addition to nucleic acids, vertebrate faeces contain a vast array of molecules originating from metabolic and catabolic processes from the host, the gut microbiome and the helminths themselves. Such molecules, generated by individual organisms or through the interactions between organisms, represent a potential source of novel biomarkers of infection(s).

Metabolomics aims to detect and characterize small molecules (e.g. amino acids, peptides, carbohydrates and fatty acids) involved in metabolic pathways in a single biological specimen. This discipline takes advantage of a plethora of ‘omics’ tools (e.g. chemogenomics) and platforms (liquid/gas chromatography coupled with mass spectrometry) to facilitate metabolite discovery and characterization [153,154,155]. Thus far, application of metabolomics techniques to the diagnosis of infectious diseases from faecal extracts remains limited. One example has demonstrated that symptomatic infection by the bacterium Clostridioides difficile was correlated with high levels of 4-methylpentanoic acid, a product of Stickland metabolism, in faecal extracts [156].

Excretory/secretory products (ESPs) from parasitic helminths are also known to contain a large number of metabolites that might serve as useful diagnostic biomarkers. For example, a recent study aimed at characterizing the metabolic “footprint” of Echinococcus multilocularis revealed metabolites (e.g. acetate and alanine) secreted or excreted by the parasite in vitro that could be further explored as diagnostic target candidates [157]. However, their specificity remains to be established, as the same panel of metabolic markers may be excreted/secreted by more than one parasite. Indeed, the same metabolites (e.g. myristic acid, lauric acid) were detected in ESPs from T. muris-infected mice [158], as well as from dogs infected by A. caninum [159]. Based on this observation, these metabolites are likely to be unsuitable as species-specific diagnostic targets for helminth infections. In the same study [158], more than 30 other metabolites were identified in ESPs isolated from N. braziliensis-infected rats or T. muris-infected mice, but, again, the suitability of these metabolites as potential biomarkers of infection remains to be established. In contrast, ESPs isolated from the dog roundworm T. canis were enriched for talose, an uncommon carbohydrate, which may represent a biomarker [160].

Changes in the composition and function of the host gut microbiome can be exploited in studies aimed at discovering novel infection biomarkers in helminth-infected subjects. In a recent study, Jenkins et al. [136] conducted a metabolomics analysis of faecal samples from a cohort of human volunteers pre-diagnosed with S. stercoralis infection, and compared data with that obtained from a cohort of uninfected individuals from the same geographical area. In this study, faecal extracts from S. stercoralis-infected subjects displayed an increased abundance of selected amino acids (leucine, alanine and lysine) compared with uninfected individuals. Whilst the number of subjects enrolled in this study was limited, and targeted metabolomics techniques (in lieu of broad-spectrum techniques) were applied to the characterization of the set of metabolites in faecal extracts, data from this study appear to provide a sound starting point for the exploration of bacterial secondary metabolites as bystander signatures of helminth infection. In turn, the characterization of worm-associated changes in bacterial and/or host metabolism might lead to a better understanding of the pathophysiology of helminth infections, and thus to the discovery and development of novel and improved treatment strategies aimed at assisting the restoration of gut homeostasis. Whilst faecal metabolites currently represent an untapped source of potential biomarkers for helminth infections, other molecules, such as non-coding short RNAs (e.g. microRNAs) of worms show promise, as reported in recent serological studies of S. mansoni in humans from endemic areas, as well as in rats experimentally infected with S. japonicum [161].

Conclusions

Stool can be seen as a window into a complex ecosystem in the GI tract of animals. Infectious agents detectable in stool encompass eukaryotes (including parasitic helminths, protists and fungi), bacteria, viruses and archaea. Thus, stool is a critically important biological matrix to reveal the composition of, and interactions between, organisms colonizing the GI tract of both healthy and diseased individuals.

From conventional molecular methods to more advanced methods of shotgun or deep amplicon sequencing, the field of helminthology makes use of a diverse array of tools for both the detection, identification and characterization of helminths in faeces and other matrices. However, the translation of these tools to the field of diagnostics operated by health workers and programme managers is challenging, particularly in low- and middle-income countries, and, thus far, has been mainly limited to veterinary applications. Although currently available in an academic research context, metabarcoding and metagenomics have a notable capacity to increase the scale and throughput in analytical and diagnostic investigations, and have major potential to enhance our knowledge and understanding of parasite–microbe interactions and relationships and of species diversity within host individuals and populations, and/or guide chemotherapeutic and parasite control. Such a systems-biology approach (cf. Fig. 1) might also be used to facilitate the monitoring of the dynamics of worm populations and the success of MDA programmes. Clearly, well-controlled studies are needed in the future to establish novel microbiome- or host-specific signatures that correlate with worm infections. As the field of metabolomics grows, curated databases containing published findings and data sets are necessary to facilitate the direct comparison of metabolic profiles and define useful biomarkers or signatures (cf. [154]). Coordinated efforts in this area to improve diagnostic and analytical tools could transform our knowledge of parasite-microbe interactions and contribute to better surveillance, treatment and control of GI helminths of humans and animals.

Fig. 1
figure 1

A proposed systems-biology approach for diagnosing and characterizing signatures of worm infection in stool. The gut is a complex environment hosting a plethora of micro- and macro-organisms. The application of metabarcoding and metagenomics-metabolomics tools to the identification and characterization of parasite populations (including their genetic variants), as well as of host- and gut microbiome by-products of worm colonization will offer unique opportunities to: (i) better define the diversity of helminth populations; (ii) discover and develop sensitive and specific diagnostic tools; and (iii) dissect the complex relationships between gut function and pathophysiology of helminth disease. Figure elements used license-free courtesy smart.servier.com.

Availability of data and materials

Not applicable.

References

  1. McManus DP, Dunne DW, Sacko M, Utzinger J, Vennervald BJ, Zhou X-N. Schistosomiasis. Nat Rev Dis Primers. 2018;4:13.

    PubMed  Google Scholar 

  2. Pakharukova MY, Mordvinov VA. The liver fluke Opisthorchis felineus: biology, epidemiology and carcinogenic potential. Trans R Soc Trop Med Hyg. 2016;110:28–36.

    PubMed  Google Scholar 

  3. Qian M-B, Utzinger J, Keiser J, Zhou X-N. Clonorchiasis. Lancet. 2016;387:800–10.

    PubMed  Google Scholar 

  4. Pullan RL, Smith JL, Jasrasaria R, Brooker SJ. Global numbers of infection and disease burden of soil transmitted helminth infections in 2010. Parasit Vectors. 2014;7:37.

    PubMed Central  PubMed  Google Scholar 

  5. World Health Organization. Soil-transmitted helminth infections. 2021. https://www.who.int/news-room/fact-sheets/detail/soil-transmitted-helminth-infections. Accessed 14 Dec 2021.

  6. Feng M, Cheng X. Parasite-associated cancers (blood flukes/liver flukes). Adv Exp Med Biol. 2017;1018:193–205.

    CAS  PubMed  Google Scholar 

  7. Murray CJL, Vos T, Lozano R, Naghavi M, Flaxman AD, Michaud C, et al. 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. 2012;380:2197–223.

    PubMed  Google Scholar 

  8. Charlier J, Rinaldi L, Musella V, Ploeger HW, Chartier C, Vineer HR, et al. Initial assessment of the economic burden of major parasitic helminth infections to the ruminant livestock industry in Europe. Prev Vet Med. 2020;182:105103.

    CAS  PubMed  Google Scholar 

  9. Easton S, Pinchbeck GL, Bartley DJ, Hodgkinson JE, Matthews JB. A survey of experiences of UK cattle and sheep farmers with anthelmintic prescribers; are best practice principles being deployed at farm level? Prev Vet Med. 2018;155:27–37.

    PubMed  Google Scholar 

  10. Lanusse C, Alvarez L, Lifschitz A. Pharmacological knowledge and sustainable anthelmintic therapy in ruminants. Vet Parasitol. 2014;204:18–33.

    CAS  PubMed  Google Scholar 

  11. Williams EG, Brophy PM, Williams HW, Davies N, Jones RA. Gastrointestinal nematode control practices in ewes: identification of factors associated with application of control methods known to influence anthelmintic resistance development. Vet Parasitol Reg Stud. 2021;24:100562.

    Google Scholar 

  12. McArthur MJ, Reinemeyer CR. Herding the U.S. cattle industry toward a paradigm shift in parasite control. Vet Parasitol. 2014;204:34–43.

    CAS  PubMed  Google Scholar 

  13. Vineer HR, Morgan ER, Hertzberg H, Bartley DJ, Bosco A, Charlier J, et al. Increasing importance of anthelmintic resistance in European livestock: creation and meta-analysis of an open database. Parasite. 2020;27:69.

    Google Scholar 

  14. Jia T-W, Melville S, Utzinger J, King CH, Zhou X-N. Soil-transmitted helminth reinfection after drug treatment: a systematic review and meta-analysis. PLoS Negl Trop Dis. 2012;6:e1621.

    CAS  PubMed Central  PubMed  Google Scholar 

  15. Dunn JC, Bettis AA, Wyine NY, Lwin AMM, Tun A, Maung NS, et al. Soil-transmitted helminth reinfection four and six months after mass drug administration: results from the delta region of Myanmar. PLoS Negl Trop Dis. 2019;13:e0006591.

    PubMed Central  PubMed  Google Scholar 

  16. Speich B, Moser W, Ali SM, Ame SM, Albonico M, Hattendorf J, et al. Efficacy and reinfection with soil-transmitted helminths 18-weeks post-treatment with albendazole-ivermectin, albendazole-mebendazole, albendazole-oxantel pamoate and mebendazole. Parasit Vectors. 2016;9:123.

    PubMed Central  PubMed  Google Scholar 

  17. Kaplan RM. Biology, epidemiology, diagnosis, and management of anthelmintic resistance in gastrointestinal nematodes of livestock. Vet Clin North Am Food Anim Pract. 2020;36:17–30.

    CAS  PubMed  Google Scholar 

  18. George MM, Vatta AF, Howell SB, Storey BE, McCoy CJ, Wolstenholme AJ, et al. Evaluation of changes in drug susceptibility and population genetic structure in Haemonchus contortus following worm replacement as a means to reverse the impact of multiple-anthelmintic resistance on a sheep farm. Int J Parasitol Drugs Drug Resist. 2021;15:134–43.

    PubMed Central  PubMed  Google Scholar 

  19. Morgan ER, Charlier J, Hendrickx G, Biggeri A, Catalan D, Von Samson-Himmelstjerna G, et al. Global change and helminth infections in grazing ruminants in Europe: impacts, trends and sustainable solutions. Agriculture. 2013;3:484–502.

    Google Scholar 

  20. Kelleher AC, Good B, de Waal T, Keane OM. Anthelmintic resistance among gastrointestinal nematodes of cattle on dairy calf to beef farms in Ireland. Ir Vet J. 2020;73:12.

    PubMed Central  PubMed  Google Scholar 

  21. Doyle SR, Cotton JA. Genome-wide approaches to investigate anthelmintic resistance. Trends Parasitol. 2019;35:289–301.

    CAS  PubMed  Google Scholar 

  22. Berger DJ, Crellen T, Lamberton PHL, Allan F, Tracey A, Noonan JD, et al. Whole-genome sequencing of Schistosoma mansoni reveals extensive diversity with limited selection despite mass drug administration. Nat Commun. 2021;12:4776.

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Doyle SR, Laing R, Bartley D, Morrison A, Holroyd N, Maitland K, et al. Genomic landscape of drug response reveals novel mediators of anthelmintic resistance. bioRxiv. 2021. https://doi.org/10.1101/2021.11.12.465712.

    Article  PubMed Central  PubMed  Google Scholar 

  24. Chapman PR, Webster R, Giacomin P, Llewellyn S, Becker L, Pearson MS, et al. Vaccination of human participants with attenuated Necator americanus hookworm larvae and human challenge in Australia: a dose-finding study and randomised, placebo-controlled, phase 1 trial. Lancet Infect Dis. 2021;21:1725–36.

    CAS  PubMed  Google Scholar 

  25. Ryan NM, Hess JA, de Villena FP-M, Leiby BE, Shimada A, Yu L, et al. Onchocerca volvulus bivalent subunit vaccine induces protective immunity in genetically diverse collaborative cross recombinant inbred intercross mice. NPJ Vaccines. 2021;6:1–12.

    Google Scholar 

  26. Keitel WA, Potter GE, Diemert D, Bethony J, El Sahly HM, Kennedy JK, et al. A phase 1 study of the safety, reactogenicity, and immunogenicity of a Schistosoma mansoni vaccine with or without glucopyranosyl lipid A aqueous formulation (GLA-AF) in healthy adults from a non-endemic area. Vaccine. 2019;37:6500–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Gazzinelli-Guimarães AC, Gazzinelli-Guimarães P, Weatherhead JE. A historical and systematic overview of Ascaris vaccine development. Parasitology. 2021;148:1795–805.

    PubMed  Google Scholar 

  28. Zawawi A, Else KJ. Soil-transmitted helminth vaccines: are we getting closer? Front Immunol. 2020;11:576748.

    CAS  PubMed Central  PubMed  Google Scholar 

  29. Claerebout E, Geldhof P. Helminth vaccines in ruminants: from development to application. Vet Clin North Am Food Anim Pract. 2020;36:159–71.

    PubMed Central  PubMed  Google Scholar 

  30. Charlier J, Morgan ER, Rinaldi L, van Dijk J, Demeler J, Höglund J, et al. Practices to optimise gastrointestinal nematode control on sheep, goat and cattle farms in Europe using targeted (selective) treatments. Vet Rec Open. 2014;175:250–5.

    CAS  Google Scholar 

  31. Nikolay B, Brooker SJ, Pullan RL. Sensitivity of diagnostic tests for human soil-transmitted helminth infections: a meta-analysis in the absence of a true gold standard. Int J Parasitol. 2014;44:765–74.

    PubMed Central  PubMed  Google Scholar 

  32. Werkman M, Wright JE, Truscott JE, Easton AV, Oliveira RG, Toor J, et al. Testing for soil-transmitted helminth transmission elimination: analysing the impact of the sensitivity of different diagnostic tools. PLoS Negl Trop Dis. 2018;12:e0006114.

    PubMed Central  PubMed  Google Scholar 

  33. Mbong Ngwese M, Prince Manouana G, Nguema Moure PA, Ramharter M, Esen M, Adégnika AA. Diagnostic techniques of soil-transmitted helminths: impact on control measures. Trop Med Infect Dis. 2020;5:93.

    PubMed Central  Google Scholar 

  34. Papaiakovou M, Littlewood DTJ, Gasser RB, Anderson RM. How qPCR complements the WHO roadmap (2021–2030) for soil-transmitted helminths. Trends Parasitol. 2021;37:698–708.

    CAS  PubMed  Google Scholar 

  35. Easton AV, Oliveira RG, O’Connell EM, Kepha S, Mwandawiro CS, Njenga SM, et al. Multi-parallel qPCR provides increased sensitivity and diagnostic breadth for gastrointestinal parasites of humans: field-based inferences on the impact of mass deworming. Parasit Vectors. 2016;9:38.

    PubMed Central  PubMed  Google Scholar 

  36. Pilotte N, Papaiakovou M, Grant JR, Bierwert LA, Llewellyn S, McCarthy JS, et al. Improved PCR-based detection of soil transmitted helminth infections using a next-generation sequencing approach to assay design. PLoS Negl Trop Dis. 2016;10:e0004578.

    PubMed Central  PubMed  Google Scholar 

  37. Cimino RO, Jeun R, Juarez M, Cajal PS, Vargas P, Echazú A, et al. Identification of human intestinal parasites affecting an asymptomatic peri-urban Argentinian population using multi-parallel quantitative real-time polymerase chain reaction. Parasit Vectors. 2015;8:380.

    PubMed Central  PubMed  Google Scholar 

  38. Llewellyn S, Inpankaew T, Nery SV, Gray DJ, Verweij JJ, Clements ACA, et al. Application of a multiplex quantitative PCR to assess prevalence and intensity of intestinal parasite infections in a controlled clinical trial. PLoS Negl Trop Dis. 2016;10:e0004380.

    PubMed Central  PubMed  Google Scholar 

  39. Verweij JJ, Brienen EAT, Ziem J, Yelifari L, Polderman AM, Van Lieshout L. Simultaneous detection and quantification of Ancylostoma duodenale, Necator americanus, and Oesophagostomum bifurcum in fecal samples using multiplex real-time PCR. Am J Trop Med Hyg. 2007;77:685–90.

    CAS  PubMed  Google Scholar 

  40. World Health Organization. Ending the neglect to attain the sustainable development goals. 2020. https://www.who.int/neglected_diseases/Ending-the-neglect-to-attain-the-SDGs--NTD-Roadmap.pdf. Accessed 14 Dec 2021.

  41. Lim MD, Brooker SJ, Belizario VY, Gay-Andrieu F, Gilleard J, Levecke B, et al. Diagnostic tools for soil-transmitted helminths control and elimination programs: a pathway for diagnostic product development. PLoS Negl Trop Dis. 2018;12:e0006213.

    PubMed Central  PubMed  Google Scholar 

  42. Katz N, Chaves A, Pellegrino J. A simple device for quantitative stool thick-smear technique in schistosomiasis mansoni. Rev Inst Med Trop Sao Paulo. 1972;14:397–400.

    CAS  PubMed  Google Scholar 

  43. Roeber F, Kahn L. The specific diagnosis of gastrointestinal nematode infections in livestock: larval culture technique, its limitations and alternative DNA-based approaches. Vet Parasitol. 2014;205:619–28.

    CAS  PubMed  Google Scholar 

  44. Utzinger J, Rinaldi L, Lohourignon LK, Rohner F, Zimmermann MB, Tschannen AB, et al. FLOTAC: a new sensitive technique for the diagnosis of hookworm infections in humans. Trans R Soc Trop Med Hyg. 2008;102:84–90.

    PubMed  Google Scholar 

  45. Cringoli G, Maurelli MP, Levecke B, Bosco A, Vercruysse J, Utzinger J, et al. The Mini-FLOTAC technique for the diagnosis of helminth and protozoan infections in humans and animals. Nat Protoc. 2017;12:1723–32.

    CAS  PubMed  Google Scholar 

  46. Khurana S, Sethi S. Laboratory diagnosis of soil transmitted helminthiasis. Trop Parasitol. 2017;7:86–91.

    PubMed Central  PubMed  Google Scholar 

  47. Roeber F, Jex AR, Gasser RB. Advances in the diagnosis of key gastrointestinal nematode infections of livestock, with an emphasis on small ruminants. Biotechnol Adv. 2013;31:1135–52.

    PubMed Central  PubMed  Google Scholar 

  48. Coulibaly JT, Ouattara M, Becker SL, Lo NC, Keiser J, N’Goran EK, et al. Comparison of sensitivity and faecal egg counts of Mini-FLOTAC using fixed stool samples and Kato-Katz technique for the diagnosis of Schistosoma mansoni and soil-transmitted helminths. Acta Trop. 2016;164:107–16.

    PubMed  Google Scholar 

  49. Buonfrate D, Formenti F, Perandin F, Bisoffi Z. Novel approaches to the diagnosis of Strongyloides stercoralis infection. Clin Microbiol Infect. 2015;21:543–52.

    CAS  PubMed  Google Scholar 

  50. Cavalcanti M, Silva L, Macedo H, Peralta R, Igreja R, Barreto M, et al. Real-time PCR improves detection of active infection in human feces and treatment failure following multiple chemotherapeutic rounds in Schistosoma mansoni low-endemic area. Ann Clin Pathol. 2016;2:1092–100.

    Google Scholar 

  51. Cools P, Vlaminck J, Albonico M, Ame S, Ayana M, José Antonio BP, et al. Diagnostic performance of a single and duplicate Kato-Katz, Mini-FLOTAC, FECPAKG2 and qPCR for the detection and quantification of soil-transmitted helminths in three endemic countries. PLoS Negl Trop Dis. 2019;13:e0007446.

    PubMed Central  PubMed  Google Scholar 

  52. Dunn JC, Papaiakovou M, Han KT, Chooneea D, Bettis AA, Wyine NY, et al. The increased sensitivity of qPCR in comparison to Kato-Katz is required for the accurate assessment of the prevalence of soil-transmitted helminth infection in settings that have received multiple rounds of mass drug administration. Parasit Vectors. 2020;13:324.

    CAS  PubMed Central  PubMed  Google Scholar 

  53. Mes TH, Eysker M, Ploeger HW. A simple, robust and semi-automated parasite egg isolation protocol. Nat Protoc. 2007;2:486–9.

    CAS  PubMed  Google Scholar 

  54. Moser W, Bärenbold O, Mirams GJ, Cools P, Vlaminck J, Ali SM, et al. Diagnostic comparison between FECPAKG2 and the Kato-Katz method for analyzing soil-transmitted helminth eggs in stool. PLoS Negl Trop Dis. 2018;12:e0006562.

    PubMed Central  PubMed  Google Scholar 

  55. Tyson F, Dalesman S, Brophy PM, Morphew RM. Novel equine faecal egg diagnostics: validation of the FECPAKG2. Animals. 2020;10:1254.

    PubMed Central  Google Scholar 

  56. Sukas S, Van Dorst B, Kryj A, Lagatie O, De Malsche W, Stuyver LJ. Development of a lab-on-a-disk platform with digital imaging for identification and counting of parasite eggs in human and animal stool. Micromachines. 2019;10:E852.

    PubMed  Google Scholar 

  57. Dacal E, Bermejo-Peláez D, Lin L, Álamo E, Cuadrado D, Martínez Á, et al. Mobile microscopy and telemedicine platform assisted by deep learning for the quantification of Trichuris trichiura infection. PLoS Negl Trop Dis. 2021;15:e0009677.

    PubMed Central  PubMed  Google Scholar 

  58. Upjohn MM, Shipton K, Lerotholi T, Attwood G, Verheyen KLP. Coprological prevalence and intensity of helminth infection in working horses in Lesotho. Trop Anim Health Prod. 2010;42:1655–61.

    PubMed  Google Scholar 

  59. Nagamori Y, Sedlak RH, DeRosa A, Pullins A, Cree T, Loenser M, et al. Further evaluation and validation of the VETSCAN IMAGYST: in-clinic feline and canine fecal parasite detection system integrated with a deep learning algorithm. Parasit Vectors. 2021;14:89.

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Nagamori Y, Sedlak RH, DeRosa A, Pullins A, Cree T, Loenser M, et al. Evaluation of the VETSCAN IMAGYST: an in-clinic canine and feline fecal parasite detection system integrated with a deep learning algorithm. Parasit Vectors. 2020;13:346.

    PubMed Central  PubMed  Google Scholar 

  61. Kalwa U, Legner C, Wlezien E, Tylka G, Pandey S. New methods of removing debris and high-throughput counting of cyst nematode eggs extracted from field soil. PLoS ONE. 2019;14:e0223386.

    CAS  PubMed Central  PubMed  Google Scholar 

  62. Akintayo A, Tylka GL, Singh AK, Ganapathysubramanian B, Singh A, Sarkar S. A deep learning framework to discern and count microscopic nematode eggs. Sci Rep. 2018;8:9145.

    PubMed Central  PubMed  Google Scholar 

  63. Holmström O, Linder N, Ngasala B, Mårtensson A, Linder E, Lundin M, et al. Point-of-care mobile digital microscopy and deep learning for the detection of soil-transmitted helminths and Schistosoma haematobium. Glob Health Action. 2017;10:1337325.

    PubMed Central  PubMed  Google Scholar 

  64. Elsemore DA, Geng J, Cote J, Hanna R, Lucio-Forster A, Bowman DD. Enzyme-linked immunosorbent assays for coproantigen detection of Ancylostoma caninum and Toxocara canis in dogs and Toxocara cati in cats. J Vet Diagn Invest. 2017;29:645–53.

    CAS  PubMed  Google Scholar 

  65. Bungiro RD, Cappello M. Detection of excretory/secretory coproantigens in experimental hookworm infection. Am J Trop Med Hyg. 2005;73:915–20.

    PubMed  Google Scholar 

  66. Lagatie O, Verheyen A, Van Hoof K, Lauwers D, Odiere MR, Vlaminck J, et al. Detection of Ascaris lumbricoides infection by ABA-1 coproantigen ELISA. PLoS Negl Trop Dis. 2020;14:e0008807.

    CAS  PubMed Central  PubMed  Google Scholar 

  67. Guezala M, Rodriguez S, Zamora H, García HH, Gonzalez A, Tembo A, et al. Development of a species-specific coproantigen ELISA for human Taenia solium taeniasis. Am J Trop Med Hyg. 2009;81:433–7.

    CAS  PubMed  Google Scholar 

  68. Ng-Nguyen D, Stevenson MA, Dorny P, Gabriël S, Vo TV, Nguyen V-AT, et al. Comparison of a new multiplex real-time PCR with the Kato Katz thick smear and copro-antigen ELISA for the detection and differentiation of Taenia spp. in human stools. PLoS Negl Trop Dis. 2017;11:e0005743.

    PubMed Central  PubMed  Google Scholar 

  69. Charlier J, De Meulemeester L, Claerebout E, Williams D, Vercruysse J. Qualitative and quantitative evaluation of coprological and serological techniques for the diagnosis of fasciolosis in cattle. Vet Parasitol. 2008;153:44–51.

    PubMed  Google Scholar 

  70. Ubeira FM, Muiño L, Valero MA, Periago MV, Pérez-Crespo I, Mezo M, et al. MM3-ELISA detection of Fasciola hepatica coproantigens in preserved human stool samples. Am J Trop Med Hyg. 2009;81:156–62.

    PubMed  Google Scholar 

  71. Papaiakovou M, Pilotte N, Grant JR, Traub RJ, Llewellyn S, McCarthy JS, et al. A novel, species-specific, real-time PCR assay for the detection of the emerging zoonotic parasite Ancylostoma ceylanicum in human stool. PLoS Negl Trop Dis. 2017;11:e0005734.

    PubMed Central  PubMed  Google Scholar 

  72. Charlier J, van der Voort M, Kenyon F, Skuce P, Vercruysse J. Chasing helminths and their economic impact on farmed ruminants. Trends Parasitol. 2014;30:361–7.

    PubMed  Google Scholar 

  73. Papaiakovou M, Pilotte N, Dunn J, Littlewood DT, Cimino RO, Krolewiecki A, et al. Confirmation of multi-parallel quantitative real-time PCR as the gold standard for detecting soil-transmitted helminths in stool. medRxiv. 2021. https://doi.org/10.1101/2021.12.09.21267271.

    Article  Google Scholar 

  74. Stracke K, Clarke N, Awburn CV, Vaz Nery S, Khieu V, Traub RJ, et al. Development and validation of a multiplexed-tandem qPCR tool for diagnostics of human soil-transmitted helminth infections. PLoS Negl Trop Dis. 2019;13:e0007363.

    PubMed Central  PubMed  Google Scholar 

  75. Massetti L, Colella V, Zendejas PA, Ng-Nguyen D, Harriott L, Marwedel L, et al. High-throughput multiplex qPCRs for the surveillance of zoonotic species of canine hookworms. PLoS Negl Trop Dis. 2020;14:e0008392.

    CAS  PubMed Central  PubMed  Google Scholar 

  76. Momčilović S, Cantacessi C, Arsić-Arsenijević V, Otranto D, Tasić-Otašević S. Rapid diagnosis of parasitic diseases: current scenario and future needs. Clin Microbiol Infect. 2019;25:290–309.

    PubMed  Google Scholar 

  77. Rosser A, Rollinson D, Forrest M, Webster BL. Isothermal recombinase polymerase amplification (RPA) of Schistosoma haematobium DNA and oligochromatographic lateral flow detection. Parasit Vectors. 2015;8:446.

    CAS  PubMed Central  PubMed  Google Scholar 

  78. Rashwan N, Diawara A, Scott ME, Prichard RK. Isothermal diagnostic assays for the detection of soil-transmitted helminths based on the SmartAmp2 method. Parasit Vectors. 2017;10:496.

    PubMed Central  PubMed  Google Scholar 

  79. Verweij JJ, Stensvold CR. Molecular testing for clinical diagnosis and epidemiological investigations of intestinal parasitic infections. Clin Microbiol Rev. 2014;27:371–418.

    PubMed Central  PubMed  Google Scholar 

  80. Pilotte N, Maasch JRMA, Easton AV, Dahlstrom E, Nutman TB, Williams SA. Targeting a highly repeated germline DNA sequence for improved real-time PCR-based detection of Ascaris infection in human stool. PLoS Negl Trop Dis. 2019;13:e0007593.

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Gaytán Á, Bergsten J, Canelo T, Pérez-Izquierdo C, Santoro M, Bonal R. DNA Barcoding and geographical scale effect: the problems of undersampling genetic diversity hotspots. Ecol Evol. 2020;10:10754–72.

    PubMed Central  PubMed  Google Scholar 

  82. Puillandre N, Lambert A, Brouillet S, Achaz G. ABGD, automatic barcode gap discovery for primary species delimitation. Mol Ecol. 2012;21:1864–77.

    CAS  PubMed  Google Scholar 

  83. Blaxter ML. The promise of a DNA taxonomy. Philos Trans R Soc Lond B Biol Sci. 2004;359:669–79.

    CAS  PubMed Central  PubMed  Google Scholar 

  84. Andújar C, Arribas P, Yu DW, Vogler AP, Emerson BC. Why the COI barcode should be the community DNA metabarcode for the Metazoa. Mol Ecol. 2018;27:3968–75.

    PubMed  Google Scholar 

  85. Savolainen V, Cowan RS, Vogler AP, Roderick GK, Lane R. Towards writing the encyclopaedia of life: an introduction to DNA barcoding. Philos Trans R Soc Lond B Biol Sci. 2005;360:1805–11.

    CAS  PubMed Central  PubMed  Google Scholar 

  86. Kounosu A, Murase K, Yoshida A, Maruyama H, Kikuchi T. Improved 18S and 28S rDNA primer sets for NGS-based parasite detection. Sci Rep. 2019;9:15789.

    PubMed Central  PubMed  Google Scholar 

  87. Mitsi K, Arroyo AS, Ruiz-Trillo I. A global metabarcoding analysis expands molecular diversity of Platyhelminthes and reveals novel early-branching clades. Biol Lett. 2019;15:20190182.

    PubMed Central  PubMed  Google Scholar 

  88. Tanaka R, Hino A, Tsai IJ, Palomares-Rius JE, Yoshida A, Ogura Y, et al. Assessment of helminth biodiversity in wild rats using 18S rDNA based metagenomics. PLoS ONE. 2014;9:e110769.

    PubMed Central  PubMed  Google Scholar 

  89. Gonçalves LT, Bianchi FM, Deprá M, Calegaro-Marques C. Barcoding a can of worms: testing cox1 performance as a DNA barcode of Nematoda. Genome. 2021;64:705–17.

    PubMed  Google Scholar 

  90. DeSalle R, Goldstein P. Review and interpretation of trends in DNA barcoding. Front Ecol Evol. 2019;7:302.

    Google Scholar 

  91. Holterman M, van der Wurff A, van den Elsen S, van Megen H, Bongers T, Holovachov O, et al. Phylum-wide analysis of SSU rDNA reveals deep phylogenetic relationships among nematodes and accelerated evolution toward crown Clades. Mol Biol Evol. 2006;23:1792–800.

    CAS  PubMed  Google Scholar 

  92. van Megen H, van den Elsen S, Holterman M, Karssen G, Mooyman P, Bongers T, et al. A phylogenetic tree of nematodes based on about 1200 full-length small subunit ribosomal DNA sequences. Nematology. 2009;11:927–50.

    Google Scholar 

  93. Gasser RB, Chilton NB, Hoste H, Beveridge I. Rapid sequencing of rDNA from single worms and eggs of parasitic helminths. Nucleic Acids Res. 1993;21:2525–6.

    CAS  PubMed Central  PubMed  Google Scholar 

  94. Gasser RB, Bott NJ, Chilton NB, Hunt P, Beveridge I. Toward practical, DNA-based diagnostic methods for parasitic nematodes of livestock–bionomic and biotechnological implications. Biotechnol Adv. 2008;26:325–34.

    CAS  PubMed  Google Scholar 

  95. Nolan MJ, Cribb TH. The use and implications of ribosomal DNA sequencing for the discrimination of digenean species. Adv Parasitol. 2005;60:101–63.

    PubMed  Google Scholar 

  96. Workentine ML, Chen R, Zhu S, Gavriliuc S, Shaw N, de Rijke J, et al. A database for ITS2 sequences from nematodes. BMC Genet. 2020;21:74.

    PubMed Central  PubMed  Google Scholar 

  97. Avramenko RW, Redman EM, Lewis R, Yazwinski TA, Wasmuth JD, Gilleard JS. Exploring the gastrointestinal “nemabiome”: deep amplicon sequencing to quantify the species composition of parasitic nematode communities. PLoS ONE. 2015;10:e0143559.

    PubMed Central  PubMed  Google Scholar 

  98. Queiroz C, Levy M, Avramenko R, Redman E, Kearns K, Swain L, et al. The use of ITS-2 rDNA nemabiome metabarcoding to enhance anthelmintic resistance diagnosis and surveillance of ovine gastrointestinal nematodes. Int J Parasitol Drugs Drug Resist. 2020;14:105–17.

    PubMed Central  PubMed  Google Scholar 

  99. Avramenko RW, Redman EM, Lewis R, Bichuette MA, Palmeira BM, Yazwinski TA, et al. The use of nemabiome metabarcoding to explore gastro-intestinal nematode species diversity and anthelmintic treatment effectiveness in beef calves. Int J Parasitol. 2017;47:893–902.

    PubMed  Google Scholar 

  100. Redman E, Queiroz C, Bartley DJ, Levy M, Avramenko RW, Gilleard JS. Validation of ITS-2 rDNA nemabiome sequencing for ovine gastrointestinal nematodes and its application to a large scale survey of UK sheep farms. Vet Parasitol. 2019;275:108933.

    CAS  PubMed  Google Scholar 

  101. Ekrem T, Willassen E, Stur E. A comprehensive DNA sequence library is essential for identification with DNA barcodes. Mol Phylogenet Evol. 2007;43:530–42.

    CAS  PubMed  Google Scholar 

  102. Schols R, Mudavanhu A, Carolus H, Hammoud C, Muzarabani KC, Barson M, et al. Exposing the barcoding void: an integrative approach to study snail-borne parasites in a One Health context. Front Vet Sci. 2020;7:1046.

    Google Scholar 

  103. Taberlet P, Coissac E, Pompanon F, Brochmann C, Willerslev E. Towards next-generation biodiversity assessment using DNA metabarcoding. Mol Ecol. 2012;21:2045–50.

    CAS  PubMed  Google Scholar 

  104. Deiner K, Bik HM, Mächler E, Seymour M, Lacoursière-Roussel A, Altermatt F, et al. Environmental DNA metabarcoding: transforming how we survey animal and plant communities. Mol Ecol. 2017;26:5872–95.

    PubMed  Google Scholar 

  105. Srivathsan A, Ang A, Vogler AP, Meier R. Fecal metagenomics for the simultaneous assessment of diet, parasites, and population genetics of an understudied primate. Front Zool. 2016;13:17.

    PubMed Central  PubMed  Google Scholar 

  106. Schneeberger PHH, Becker SL, Pothier JF, Duffy B, N’Goran EK, Beuret C, et al. Metagenomic diagnostics for the simultaneous detection of multiple pathogens in human stool specimens from Côte d’Ivoire: a proof-of-concept study. Infect Genet Evol. 2016;40:389–97.

    CAS  PubMed  Google Scholar 

  107. Aivelo T, Medlar A. Opportunities and challenges in metabarcoding approaches for helminth community identification in wild mammals. Parasitology. 2018;145:608–21.

    PubMed  Google Scholar 

  108. Avramenko RW, Bras A, Redman EM, Woodbury MR, Wagner B, Shury T, et al. High species diversity of trichostrongyle parasite communities within and between Western Canadian commercial and conservation bison herds revealed by nemabiome metabarcoding. Parasit Vectors. 2018;11:299.

    PubMed Central  PubMed  Google Scholar 

  109. Poissant J, Gavriliuc S, Bellaw J, Redman EM, Avramenko RW, Robinson D, et al. A repeatable and quantitative DNA metabarcoding assay to characterize mixed strongyle infections in horses. Int J Parasitol. 2021;51:183–92.

    CAS  PubMed  Google Scholar 

  110. Chan AHE, Chaisiri K, Morand S, Saralamba N, Thaenkham U. Evaluation and utility of mitochondrial ribosomal genes for molecular systematics of parasitic nematodes. Parasit Vectors. 2020;13:364.

    CAS  PubMed Central  PubMed  Google Scholar 

  111. Davey ML, Utaaker KS, Fossøy F. Characterizing parasitic nematode faunas in faeces and soil using DNA metabarcoding. Parasit Vectors. 2021;14:422.

    CAS  PubMed Central  PubMed  Google Scholar 

  112. Barone CD, Wit J, Hoberg EP, Gilleard JS, Zarlenga DS. Wild ruminants as reservoirs of domestic livestock gastrointestinal nematodes. Vet Parasitol. 2020;279:109041.

    CAS  PubMed  Google Scholar 

  113. Beaumelle C, Redman EM, de Rijke J, Wit J, Benabed S, Debias F, et al. Metabarcoding in two isolated populations of wild roe deer (Capreolus capreolus) reveals variation in gastrointestinal nematode community composition between regions and among age classes. Parasit Vectors. 2021;14:594.

    CAS  PubMed Central  PubMed  Google Scholar 

  114. Pafčo B, Čížková D, Kreisinger J, Hasegawa H, Vallo P, Shutt K, et al. Metabarcoding analysis of strongylid nematode diversity in two sympatric primate species. Sci Rep. 2018;8:5933.

    PubMed Central  PubMed  Google Scholar 

  115. Avramenko RW, Redman EM, Melville L, Bartley Y, Wit J, Queiroz C, et al. Deep amplicon sequencing as a powerful new tool to screen for sequence polymorphisms associated with anthelmintic resistance in parasitic nematode populations. Int J Parasitol. 2019;49:13–26.

    CAS  PubMed  Google Scholar 

  116. Evans MJ, Chaudhry UN, Costa-Júnior LM, Hamer K, Leeson SR, Sargison ND. A 4 year observation of gastrointestinal nematode egg counts, nemabiomes and the benzimidazole resistance genotypes of Teladorsagia circumcincta on a Scottish sheep farm. Int J Parasitol. 2021;51:393–403.

    CAS  PubMed  Google Scholar 

  117. Roose S, Avramenko RW, Pollo SMJ, Wasmuth JD, Ame S, Ayana M, et al. Characterization of the β-tubulin gene family in Ascaris lumbricoides and Ascaris suum and its implication for the molecular detection of benzimidazole resistance. PLoS Negl Trop Dis. 2021;15:e0009777.

    CAS  PubMed Central  PubMed  Google Scholar 

  118. Quince C, Walker AW, Simpson JT, Loman NJ, Segata N. Shotgun metagenomics, from sampling to analysis. Nat Biotechnol. 2017;35:833–44.

    CAS  PubMed  Google Scholar 

  119. Chessa D, Murgia M, Sias E, Deligios M, Mazzarello V, Fiamma M, et al. Metagenomics and microscope revealed T. trichiura and other intestinal parasites in a cesspit of an Italian nineteenth century aristocratic palace. Sci Rep. 2020;10:12656.

    CAS  PubMed Central  PubMed  Google Scholar 

  120. Khairat R, Ball M, Chang C-CH, Bianucci R, Nerlich AG, Trautmann M, et al. First insights into the metagenome of Egyptian mummies using next-generation sequencing. J Appl Genet. 2013;54:309–25.

    CAS  PubMed  Google Scholar 

  121. Søe MJ, Nejsum P, Seersholm FV, Fredensborg BL, Habraken R, Haase K, et al. Ancient DNA from latrines in Northern Europe and the Middle East (500 BC-1700 AD) reveals past parasites and diet. PLoS ONE. 2018;13:e0195481.

    PubMed Central  PubMed  Google Scholar 

  122. Doyle SR, Søe MJ, Nejsum P, Betson M, Cooper PJ, Peng L, et al. Population genomics of ancient and modern Trichuris trichiura. bioRxiv. 2021. https://doi.org/10.1101/2021.10.21.464505.

    Article  PubMed Central  PubMed  Google Scholar 

  123. Doyle SR, Sankaranarayanan G, Allan F, Berger D, Jimenez Castro PD, Collins JB, et al. Evaluation of DNA extraction methods on individual helminth egg and larval stages for whole-genome sequencing. Front Genet. 2019;10:826.

    CAS  PubMed Central  PubMed  Google Scholar 

  124. Oyola SO, Ariani CV, Hamilton WL, Kekre M, Amenga-Etego LN, Ghansah A, et al. Whole genome sequencing of Plasmodium falciparum from dried blood spots using selective whole genome amplification. Malar J. 2016;15:597.

    PubMed Central  PubMed  Google Scholar 

  125. International Helminth Genomes Consortium. Comparative genomics of the major parasitic worms. Nat Genet. 2019;51:163–74.

    CAS  Google Scholar 

  126. Howe KL, Bolt BJ, Shafie M, Kersey P, Berriman M. WormBase ParaSite—a comprehensive resource for helminth genomics. Mol Biochem Parasitol. 2017;215:2–10.

    CAS  PubMed Central  PubMed  Google Scholar 

  127. Lewin HA, Robinson GE, Kress WJ, Baker WJ, Coddington J, Crandall KA, et al. Earth BioGenome project: sequencing life for the future of life. Proc Natl Acad Sci USA. 2018;115:4325–33.

    CAS  PubMed Central  PubMed  Google Scholar 

  128. Kupritz J, Angelova A, Nutman TB, Gazzinelli-Guimaraes PH. Helminth-induced human gastrointestinal dysbiosis: a systematic review and meta-analysis reveals insights into altered taxon diversity and microbial gradient collapse. mBio. 2021;12:e02890-21.

    CAS  PubMed Central  Google Scholar 

  129. Cortés A, Peachey LE, Jenkins TP, Scotti R, Cantacessi C. Helminths and microbes within the vertebrate gut—not all studies are created equal. Parasitology. 2019;146:1371–8.

    PubMed  Google Scholar 

  130. Cortés A, Peachey L, Scotti R, Jenkins TP, Cantacessi C. Helminth-microbiota cross-talk—a journey through the vertebrate digestive system. Mol Biochem Parasitol. 2019;233:111222.

    PubMed  Google Scholar 

  131. Veldhoen M, Heeney JL. A helminth-mediated viral awakening. Trends Immunol. 2014;35:452–3.

    CAS  PubMed  Google Scholar 

  132. Barelli C, Donati C, Albanese D, Pafčo B, Modrý D, Rovero F, et al. Interactions between parasitic helminths and gut microbiota in wild tropical primates from intact and fragmented habitats. Sci Rep. 2021;11:21569.

    CAS  PubMed Central  PubMed  Google Scholar 

  133. Cortés A, Rooney J, Bartley DJ, Nisbet AJ, Cantacessi C. Helminths, hosts, and their microbiota: new avenues for managing gastrointestinal helminthiases in ruminants. Expert Rev Anti Infect Ther. 2020;18:977–85.

    PubMed  Google Scholar 

  134. Jenkins TP, Brindley PJ, Gasser RB, Cantacessi C. Helminth microbiomes—a hidden treasure trove? Trends Parasitol. 2019;35:13–22.

    CAS  PubMed  Google Scholar 

  135. Zaiss MM, Harris NL. Interactions between the intestinal microbiome and helminth parasites. Parasite Immunol. 2016;38:5–11.

    CAS  PubMed  Google Scholar 

  136. Jenkins TP, Formenti F, Castro C, Piubelli C, Perandin F, Buonfrate D, et al. A comprehensive analysis of the faecal microbiome and metabolome of Strongyloides stercoralis infected volunteers from a non-endemic area. Sci Rep. 2018;8:15651.

    PubMed Central  PubMed  Google Scholar 

  137. Jenkins TP, Pritchard DI, Tanasescu R, Telford G, Papaiakovou M, Scotti R, et al. Experimental infection with the hookworm, Necator americanus, is associated with stable gut microbial diversity in human volunteers with relapsing multiple sclerosis. BMC Biol. 2021;19:74.

    CAS  PubMed Central  PubMed  Google Scholar 

  138. Rosa BA, Snowden C, Martin J, Fischer K, Kupritz J, Beshah E, et al. Whipworm-associated intestinal microbiome members consistent across both human and mouse hosts. Front Cell Infect Microbiol. 2021;11:637570.

    CAS  PubMed Central  PubMed  Google Scholar 

  139. 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:e76573.

    CAS  PubMed Central  PubMed  Google Scholar 

  140. Lee SC, Tang MS, Lim YAL, 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:e2880.

    PubMed Central  PubMed  Google Scholar 

  141. Jenkins TP, Peachey LE, Ajami NJ, MacDonald AS, Hsieh MH, Brindley PJ, et al. Schistosoma mansoni infection is associated with quantitative and qualitative modifications of the mammalian intestinal microbiota. Sci Rep. 2018;8:12072.

    PubMed Central  PubMed  Google Scholar 

  142. Rosa BA, Supali T, Gankpala L, Djuardi Y, Sartono E, Zhou Y, et al. Differential human gut microbiome assemblages during soil-transmitted helminth infections in Indonesia and Liberia. Microbiome. 2018;6:33.

    PubMed Central  PubMed  Google Scholar 

  143. Easton AV, Quiñones M, Vujkovic-Cvijin I, Oliveira RG, Kepha S, Odiere MR, et al. The impact of anthelmintic treatment on human gut microbiota based on cross-sectional and pre- and postdeworming comparisons in western Kenya. mBio. 2019;10:e00519-19.

    CAS  PubMed Central  PubMed  Google Scholar 

  144. Stracke K, Adisakwattana P, Phuanukoonnon S, Yoonuan T, Poodeepiyasawat A, Dekumyoy P, et al. Field evaluation of the gut microbiome composition of pre-school and school-aged children in Tha Song Yang, Thailand, following oral MDA for STH infections. PLoS Negl Trop Dis. 2021;15:e0009597.

    PubMed Central  PubMed  Google Scholar 

  145. 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.

    PubMed Central  PubMed  Google Scholar 

  146. Jenkins TP, Rathnayaka Y, Perera PK, Peachey LE, Nolan MJ, Krause L, et al. Infections by human gastrointestinal helminths are associated with changes in faecal microbiota diversity and composition. PLoS ONE. 2017;12:e0184719.

    PubMed Central  PubMed  Google Scholar 

  147. 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.

    CAS  PubMed Central  PubMed  Google Scholar 

  148. Rooney J, Cortés A, Scotti R, Price DRG, Bartley Y, Fairlie-Clarke K, et al. Vaccination against the brown stomach worm, Teladorsagia circumcincta, followed by parasite challenge, induces inconsistent modifications in gut microbiota composition of lambs. Parasit Vectors. 2021;14:189.

    CAS  PubMed Central  PubMed  Google Scholar 

  149. Li RW, Wu S, Li W, Huang Y, Gasbarre LC. Metagenome plasticity of the bovine abomasal microbiota in immune animals in response to Ostertagia ostertagi infection. PLoS ONE. 2011;6:e24417.

    CAS  PubMed Central  PubMed  Google Scholar 

  150. Glendinning L, Nausch N, Free A, Taylor DW, Mutapi F. The microbiota and helminths: sharing the same niche in the human host. Parasitology. 2014;141:1255–71.

    PubMed  Google Scholar 

  151. Scotti R, Southern S, Boinett C, Jenkins TP, Cortés A, Cantacessi C. MICHELINdb: a web-based tool for mining of helminth-microbiota interaction datasets, and a meta-analysis of current research. Microbiome. 2020;8:10.

    CAS  PubMed Central  PubMed  Google Scholar 

  152. Rubel MA, Abbas A, Taylor LJ, Connell A, Tanes C, Bittinger K, et al. Lifestyle and the presence of helminths is associated with gut microbiome composition in Cameroonians. Genome Biol. 2020;21:122.

    CAS  PubMed Central  PubMed  Google Scholar 

  153. Griffiths WJ, Koal T, Wang Y, Kohl M, Enot DP, Deigner H-P. Targeted metabolomics for biomarker discovery. Angew Chem Int Ed Engl. 2010;49:5426–45.

    CAS  PubMed  Google Scholar 

  154. Kokova D, Mayboroda OA. Twenty years on: metabolomics in helminth research. Trends Parasitol. 2019;35:282–8.

    PubMed  Google Scholar 

  155. Whitman JD, Sakanari JA, Mitreva M. Areas of metabolomic exploration for helminth infections. ACS Infect Dis. 2021;7:206–14.

    CAS  PubMed Central  PubMed  Google Scholar 

  156. Robinson JI, Weir WH, Crowley JR, Hink T, Reske KA, Kwon JH, et al. Metabolomic networks connect host-microbiome processes to human Clostridioides difficile infections. J Clin Invest. 2019;129:3792–806.

    PubMed Central  PubMed  Google Scholar 

  157. Ritler D, Rufener R, Li JV, Kämpfer U, Müller J, Bühr C, et al. In vitro metabolomic footprint of the Echinococcus multilocularis metacestode. Sci Rep. 2019;9:19438.

    PubMed Central  PubMed  Google Scholar 

  158. Wangchuk P, Kouremenos K, Eichenberger RM, Pearson M, Susianto A, Wishart DS, et al. Metabolomic profiling of the excretory-secretory products of hookworm and whipworm. Metabolomics. 2019;15:101.

    PubMed  Google Scholar 

  159. Gyawali P, Beale DJ, Ahmed W, Karpe AV, Magalhaes RJS, Morrison PD, et al. Determination of Ancylostoma caninum ova viability using metabolic profiling. Parasitol Res. 2016;115:3485–92.

    CAS  PubMed  Google Scholar 

  160. Wangchuk P, Lavers O, Wishart DS, Loukas A. Excretory/secretory metabolome of the zoonotic roundworm parasite Toxocara canis. Biomolecules. 2020;10:1157.

    CAS  PubMed Central  Google Scholar 

  161. Mu Y, McManus DP, Gordon CA, Cai P. Parasitic helminth-derived microRNAs and extracellular vesicle cargos as biomarkers for helminthic infections. Front Cell Infect Microbiol. 2021;11:708952.

    CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

MP is a grateful recipient of a Harding Distinguished Postgraduate Scholarship awarded by the University of Cambridge. SRD is supported by a UKRI Future Leaders Fellowship (MR/T020733/1) and the Wellcome Trust (206194). The laboratory of CC is funded by grants from the Isaac Newton Trust, the Biotechnology and Biological Sciences Research Council and the University of Cambridge. For the purpose of Open Access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission.

Author information

Authors and Affiliations

Authors

Contributions

MP and CC conceived the article. MP drafted the manuscript, with contributions from DTJL, SRD, RBG and CC. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Cinzia Cantacessi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

MP is a member of the WHO Diagnostics Technical Advisory Group (WHO DTAG) for Neglected Tropical Diseases since March 2021. The WHO played no role in producing the contents of this review. All other authors declare no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Papaiakovou, M., Littlewood, D.T.J., Doyle, S.R. et al. Worms and bugs of the gut: the search for diagnostic signatures using barcoding, and metagenomics–metabolomics. Parasites Vectors 15, 118 (2022). https://doi.org/10.1186/s13071-022-05225-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13071-022-05225-7