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

The Intestinal Eukaryotic Virome in Healthy and Diarrhoeic Neonatal Piglets

  • Oskar E. Karlsson ,

    Contributed equally to this work with: Oskar E. Karlsson, Jenny Larsson

    oskar.e.karlsson@slu.se

    Affiliations Department of Biomedical Sciences and Veterinary Public Health (BVF), Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden, SLU Global Bioinformatics Centre, Department of Animal Breeding and Genetics (HGEN), SLU, Uppsala, Sweden, The OIE Collaborating Centre for the Biotechnology-based Diagnosis of Infectious Diseases in Veterinary Medicine, Uppsala, Sweden

  • Jenny Larsson ,

    Contributed equally to this work with: Oskar E. Karlsson, Jenny Larsson

    Affiliation Department of Clinical Sciences (KV), Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden

  • Juliette Hayer,

    Affiliation SLU Global Bioinformatics Centre, Department of Animal Breeding and Genetics (HGEN), SLU, Uppsala, Sweden

  • Mikael Berg ,

    ‡ These authors also contributed equally to this work.

    Affiliations Department of Biomedical Sciences and Veterinary Public Health (BVF), Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden, The OIE Collaborating Centre for the Biotechnology-based Diagnosis of Infectious Diseases in Veterinary Medicine, Uppsala, Sweden

  • Magdalena Jacobson

    ‡ These authors also contributed equally to this work.

    Affiliation Department of Clinical Sciences (KV), Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden

Abstract

Neonatal porcine diarrhoea of uncertain aetiology has been reported from a number of European countries. The aim of the present study was to use viral metagenomics to examine a potential viral involvement in this diarrhoea and to describe the intestinal virome with focus on eukaryotic viruses. Samples from the distal jejunum of 50 diarrhoeic and 19 healthy piglets from 10 affected herds were analysed. The viral fraction of the samples was isolated and nucleic acids (RNA and DNA fractions) were subjected to sequence independent amplification. Samples from diarrhoeic piglets from the same herds were pooled whereas samples from healthy piglets were analysed individually. In total, 29 clinical samples, plus two negative controls and one positive control consisting of a mock metagenome were sequenced using the Ion Torrent platform. The resulting sequence data was subjected to taxonomic classification using Kraken, Diamond and HMMER. In the healthy specimens, eight different mammalian virus families were detected (Adenoviridae, Anelloviridae, Astroviridae, Caliciviridae, Circoviridae, Parvoviridae, Picornaviridae, and Reoviridae) compared to four in the pooled diarrhoeic samples (Anelloviridae, Circoviridae, Picornaviridae, and Reoviridae). It was not possible to associate a particular virus family with the investigated diarrhoea. In conclusion, this study does not support the hypothesis that the investigated diarrhoea was caused by known mammalian viruses. The results do, however, indicate that known mammalian viruses were present in the intestine as early as 24–48 hours after birth, indicating immediate infection post-partum or possibly transplacental infection.

Introduction

Neonatal porcine diarrhoea (NPD) is a common problem in pig farming worldwide and thus contributes to morbidity and mortality among piglets, particularly in intensive farming systems [1,2]. Hence, NPD has substantial adverse effects on both economical and animal welfare aspects of the pig industry [3]. The condition is associated with a number of well-known pathogens including bacterial agents, protozoa, and viruses such as rotavirus, transmissible gastroenteritis virus (TGEV), and porcine epidemic diarrhoea virus (PEDV) [2]. Additional viruses including astrovirus [4,5], bocaparvovirus [6], kobuvirus [7,8], sapovirus [9], and sapelovirus [10] have occasionally been associated with diarrhoea in piglets, although their clinical significance is yet to be established.

Reports from a number of countries, including Sweden, describe problems with NPD despite adequate vaccination- and management routines [1113]. This condition, referred to as new neonatal porcine diarrhoea syndrome (NNPDS), does not seem to be associated with well-known pathogens and it is hypothesized that unexpected or divergent viruses, evading routine diagnostic testing, may be a contributing factor [14]. To investigate a potential viral involvement in this disease syndrome, an unbiased exploratory approach such as that offered by viral metagenomics, would be desirable [15,16]. Metagenomics is the study of metagenomes, defined as the genomes of all organisms present in a sample and offers the possibility of studying the whole microbial composition.

With the advent of metagenomics studies, the knowledge of the different components and the complexity of the microbiome greatly expanded [17,18]. The intestine represents a complex environment with both eukaryotic, prokaryotic, archaeal, and viral elements [19]. Hitherto, most studies on the intestinal microbiome have addressed the prokaryotic component whereas much less is known about the virome. However, recent viral metagenomics studies show that the intestinal virome in healthy mammals can be divided into three parts—the prokaryotic (bacteriophages), the archaeal, and the eukaryotic virome, see Fig 1 [19,20]. The eukaryotic virome comprises viruses infecting the host, endogenous viral elements, and viruses associated with other eukaryotic components of the ingesta.

thumbnail
Fig 1. Schematic illustration of the intestinal virome.

The mammalian intestinal virome is proposed to have three parts—the eukaryotic, the prokaryotic, and the archaeal. The eukaryotic virome includes host-infecting viruses, viruses infecting other eukaryotes in the intestine, and feed-associated viruses.

https://doi.org/10.1371/journal.pone.0151481.g001

Investigations of the porcine intestinal virome are so far limited [2123]. Previous comparative analyses between healthy and diarrhoeic piglets are based on faecal samples and investigate piglets around weaning (19–30 days of age) [21,22] or at 6 weeks of age [23]. However, comparative analyses of the intestinal virome from healthy and diarrhoeic neonatal piglets have, to the best of the authors’ knowledge, not been made. Neither has the eukaryotic intestinal virome of healthy neonates been characterised in livestock.

The aim of the present study was to examine a potential involvement of previously described mammalian viruses in porcine neonatal diarrhoea of uncertain aetiology and to describe the intestinal eukaryotic virome in both healthy and diarrhoeic pigs. The current study presents comparative metagenomic data analysis of intestinal samples from 19 healthy and 50 diarrhoeic piglets from 10 herds in Sweden.

Material and Methods

Samples were collected from animals from ten conventional herds (designated A to J, see Table 1) located in the middle of Sweden. All sampling procedures were approved by the Ethics Committee for Animal Experimentation, Uppsala, Sweden (permission number: C120/11).

thumbnail
Table 1. Descriptive data on selected herds (n = 10, A-J) and piglets (n = 69).

https://doi.org/10.1371/journal.pone.0151481.t001

Recruitment of herds to the study was performed on a voluntary basis and all farmers gave their permission to conduct the study on their farms. This investigation is a part of a series of studies on NPD with uncertain aetiology. Pathological and bacteriological findings have been characterised previously [13,24] and are mentioned in the discussion.

Sample collection

All herds were affected by NPD, despite routine vaccination of sows and gilts against ETEC. Two herds (A and E) were satellites in the same sow pool; two others (I and J) were located in proximity to each other and partially shared the same personnel. A total of 50 diarrhoeic animals and 19 healthy controls were included in the study; there were five diarrhoeic animals and two healthy controls from each herd (except for herd I where only one control piglet was obtained). Piglets originated from 60 different litters (three to seven per herd). Pigs treated with antibiotics were excluded. The 45 females and 24 males were all cross-breeds. Diarrhoeic pigs ranged in age from 0.5 to 6 days (median age, 1 day) and the controls from 1 to 5 (median age, 2 days), see Table 1.

The animals were transported alive to the Section of Pathology, Swedish University of Agricultural Sciences (SLU) where they were anaesthetised with an intramuscular injection of tiletamine and zolazepam, 6 mg/kg (Zoletil ® 50, Virbac Animal Health, Carros Cedex, France) and euthanized by intracardiac injection of pentobarbital sodium, 100 mg/kg (Pentobarbital for Euthanasia 20% w/v solution for injection, Pharmasol Ltd, Andover, UK). Tissue specimens from the distal jejunum (40 cm proximal to the ileocaecal junction, including both intestinal tissue and contents) were collected immediately after euthanasia, snap frozen in liquid nitrogen and stored at -70°C.

Sample preparation

Samples were prepared according to [24] with modifications. In brief, a 2.5 mm transverse section of the distal jejunum from each pig was placed in 1 ml PBS with 5% bovine serum albumin (BSA, Sigma Aldrich, St. Louis, MO, USA). The samples were subjected to three consecutive cycles of freeze-thawing on dry ice and homogenisation by the Omni TH Motor (Omni International, Kennesaw, GA, USA) using soft tissue Omni Tip Plastic Homogenizer Probes (Omni International). After homogenisation, samples were centrifuged for 10 min at 600 x g. The supernatants were collected and filtered through a 0.45 μm sterile filter (Merck Millipore, Billerica, MA, USA) and split into three fractions—one for extraction of viral RNA, one for extraction of viral DNA and one to be stored untreated at -70°C. Samples were kept on ice/dry ice slurry throughout the process. The RNA and DNA samples were treated with a nuclease cocktail containing TURBO DNase (2 U/μL; Thermo Fisher Scientific, Waltham, MA, USA), RNase Cocktail Enzyme Mix (RNase A; 500 U/mL) and RNase T1 (20,000 U/mL; Thermo Fisher Scientific) in 1x TURBO DNase Buffer. RNA and DNA were extracted as previously described [25]. Extracted RNA and DNA were stored at -70°C. A process control was prepared together with the clinical samples. This control contained PBS/5% BSA and was treated the same way as the samples, yielding one control each for RNA and DNA extractions. These controls were used to evaluate possible contaminants from the laboratory environment and reagents during preparation of the clinical specimens.

Sequence Independent Single Primer Amplification

Sequence Independent Single Primer Amplification (SISPA) was performed according to Rosseel et al. 2013 [26]. Two mock metagenomes containing viruses from various families and genome types were used as positive controls—one for DNA viruses and one for RNA viruses. To check for contamination during the amplification, several non-template controls (NTCs) were included in every amplification set. The NTCs, process control, and positive controls, were processed next to the other samples as described below. In short, samples were tagged with two different SISPA tags (FR20RV-12N, 5’-GCCGGAGCTCTGCAGATATCNNNNNNNNNNNN-3’ and K-12N, 5’-GACCATCTAGCGACCTCCACNNNNNNNNNNNN-3’, respectively) in separate reactions. RNA samples were heated to 95°C for five minutes in the presence of the SISPA primer-tags and then put on ice. Samples were subsequently converted to cDNA using Superscript III (Thermo Fisher Scientific) according to manufacturer’s instructions. DNA samples were heated to 95°C for five minutes in the presence of the SISPA primer-tags and dNTPs, then put on ice. After 2 min, the Klenow Fragment (3’→ 5’ exo) (New England Biolabs, Ipswich, MA, USA) was added and samples were incubated according to manufacturer’s instructions. SISPA was performed in two separate reactions for all samples, one with the FR20RV primer (5’-GCCGGAGCTCTGCAGATATC-3’) and one with the K primer (5’-GACCATCTAGCGACCTCCAC-3’) as previously described [26]. However, the polymerase was the KAPA HIFI HotStart ready mix (KAPA Biosystems, Wilmington, MA, USA).

Following PCR, the products were cleaned using the GeneJET PCR purification kit (Thermo Fisher Scientific) and subsequently subjected to cleavage using EcoRV (New England Biolabs). Cleaved products were repurified using the GeneJET PCR purification kit (Thermo Fisher Scientific) and quantified using the Qubit 2.0 Fluorometer (Thermo Fisher Scientific) and Qubit® dsDNA HS Assay Kit (Thermo Fisher Scientific). Fraction size was checked by gel electrophoresis [27]. Finally, SISPA products from the DNA and RNA fractions with K and FR20RV primers were pooled per piglet and control sample (the positive control with mock metagenomes, the process control, and the NTCs from the SISPA reaction). Thereafter, samples from diarrhoeic piglets within the same herd were pooled, resulting in three samples per herd.

Library preparation and sequencing

Sequencing was performed using the Ion Proton system (Thermo Fisher Scientific) at the National Genomics Infrastructure in Uppsala. 0.4 μg DNA from each sample was fragmented using the S2 system from Covaris (Covaris Inc., Woburn, USA). End repair and adaptor ligation were performed by the AB Library builder (Thermo Fisher Scientific). Samples were amplified according to the Ion Xpress Plus gDNA Fragment Library Preparation protocol and selected with a target size of 250–310 bp (Blue PippinTM, Sage Science, Beverly, MA, USA). Library size and concentration were assessed by a Bioanalyzer High Sensitivity Chip (Agilent Technologies, Santa Clara, CA, USA) and by the Fragment Analyzer system (Advanced Analytical, Ankeny, IA, USA). Samples were pooled together in sets of six, followed by template preparation using the Ion Proton Template OT2 200 v3 Kit on either the Ion OneTouch 2 system or the Ion Chef system (Thermo Fisher Scientific). Samples were then sequenced on the Ion Proton system using the Ion Proton Sequencing 200 v3 Kit on Ion PI v2 chips (200 bp read length, Thermo Fisher Scientific). The Ion Reporter system was running Torrent Suite 4.2.1 at the time of analysis.

Bioinformatics

Quality control and filtering.

Data in the form of de-multiplexed FASTQ files were acquired from the sequencing centre together with quality metrics for the sequencing runs. All FASTQ files were subjected to quality controls using PrinSeq version 0.20.4 [28]. For settings, a mean quality (Phred Quality score) [29] of 20 was used; sequences shorter than 110 bases were omitted. Following this, the datasets were mapped towards the host genome (Sus Scrofa domesticus genome, Genome assembly: Sscrofa10.2 (GCA_000003025.4) [30] using bowtie2 version 2.2.5 [31], to remove host sequences.

Taxonomic classification.

Taxonomic classification was performed on read level using Kraken version 0.10.5-beta [32] and the BLASTx equivalent of Diamond (version 0.7.10) using the non-redundant (nr) database from NCBI (version of 2016-01-20) [33]. The same reads were used for predicting protein sequences with FragGeneScan (FGS) 1.19, and the resulting sequences were scanned, using HMMER3, for the viral profiles Hidden Markov Models (HMM) of the vFam database (February 2014 release) [34,35]. The Kraken database used for the classification is an expanded in-house database covering all sequences from the viral (VRL) and phage (PHG) divisions of GenBank (NCBI), as well as the bacterial and archaeal sequences from RefSeq [36]. See Fig 2 for an overview of the data analysis pipeline. The results are only presented descriptively since the individual samples from the healthy animals were not directly comparable with the pooled diarrhoeic samples.

thumbnail
Fig 2. Schematic illustration of the bioinformatic pipeline.

Raw data from the sequencing platform, in FASTQ format, was processed by PrinSeq to select high quality reads. The fractions of high quality reads were then mapped towards the porcine genome, using Bowtie 2, to remove host sequences. The unmapped reads were then classified by three different methods, Kraken, Diamond and HMM/vFam. The resulting three datasets were grouped in two datasets, one representing reads classified on nucleotide and protein level and one representing reads classified only on protein level.

https://doi.org/10.1371/journal.pone.0151481.g002

Results

Sequencing data averaged 220 Mbases of data for the clinical samples with a mean read length of 185 bases; see Table 2 for the metrics of individual samples. The positive control generated 6.3 Gbases of high quality (52% good quality sequences) with a mean read length of 194 bases after filtering. In contrast, the NTC generated only 124 Mbases (21.74% good quality sequences) with a mean read length of 194 bases. In the positive control, all viruses included in the mock metagenome were detected whereas the NTC rendered no relevant hits. The process control contained 204 Mbases of good quality (26% good quality sequences) with a mean length of 180 bases; no mammalian viruses were detected. All clinical samples contained endogenous viral elements. Sequences classified as PERV were absent in the NTC and the process control. Data for the clinical samples are available at the European Nucleotide Archive, study accession number PRJEB11519.

thumbnail
Table 2. Overview of sequencing results and initial filtering and read classification.

https://doi.org/10.1371/journal.pone.0151481.t002

Detection of mammalian viruses in porcine distal jejunum

Table 3 reports classification of viral sequences on nucleotide level (Kraken) and protein level (Diamond and HMM/vFam). Viral families detected on both nucleotide and protein level included sequences belonging to eight different virus families infecting mammals: Adenoviridae, Anelloviridae, Astroviridae, Caliciviridae, Circoviridae, Parvoviridae, Picornaviridae, and Reoviridae. Viral sequences in healthy samples included hits towards Adenoviridae (n = 3), Anelloviridae (n = 1), Astroviridae (n = 1), Caliciviridae (n = 1), Circoviridae (n = 8), Parvoviridae (n = 2), Picornaviridae (n = 10), and Reoviridae (n = 2). Viral sequences detected in pooled diarrhoeic samples were Anelloviridae (n = 1), Circoviridae (n = 1), Picornaviridae (n = 4), and Reoviridae (n = 2). More than one mammalian virus family was present in ten out of 19 samples from healthy piglets and in two out of 10 pooled samples from diarrhoeic piglets. In four samples from individual healthy piglets (2A, 1B, 2B, 1C, and 1G), sequences belonging to three different mammalian virus families were detected. See Table 4 for a summary of the virus families found in healthy and diarrhoeic samples respectively.

thumbnail
Table 3. Overview of samples and viruses detected at family level in the distal jejunum of neonatal piglets with and without diarrhoea.

https://doi.org/10.1371/journal.pone.0151481.t003

thumbnail
Table 4. Number of samples from healthy piglets (n = 19) and pooled diarrhoeic samples (n = 10) positive for virus families known to infect mammals.

https://doi.org/10.1371/journal.pone.0151481.t004

In several samples classification of reads to viral families occurred only on protein level. In five samples both Diamond and HMM/vFam detected Circoviridae. In addition, Diamond detected Circoviridae (n = 1), Picobirnaviridae (n = 1) and HMM/vFam detected Adenoviridae (n = 2), Circoviridae (n = 4), Caliciviridae (n = 7), Parvoviridae (n = 2), and Reoviridae (n = 1).

The most common virus family detected was Picornaviridae, which was present in 14 out of 29 samples followed by Circoviridae reported in 9 out of 29 samples. The majority of the Picornaviridae sequences were classified on nucleotide level as species Aichivirus C. Sequences classified as Circoviridae in samples (n = 9) were interestingly absent in all diarrhoeic samples but one (E) and were identified on a nucleotide level as po-circo-like virus 21, 22, 51, porcine stool-associated circular virus 5, and fur seal faeces associated circular DNA virus. In addition, some unassigned small circular ssDNA viruses were found in seven healthy piglets. Reoviridae sequences included hits on nucleotide level against the species Rotavirus A and Rotavirus C. Both Rotavirus A and C were detected in two samples (1B and pool B) and only Rotavirus A in one sample (Pool A). In three samples (1B, 2B, and 2E) 18, 20 and 5 reads, respectively, were classified on nucleotide level as the dsDNA virus immunodeficiency associated stool virus (IAS virus).

Discussion

To the best of our knowledge, this is the first comparative metagenomics study of the eukaryotic virome in the intestine of healthy and diarrhoeic neonatal piglets. Furthermore, it is the first study to describe the eukaryotic intestinal virome in healthy neonatal livestock. It was not possible to associate a particular virus family with NNPDS. Among the mammalian viruses found in diarrhoeic piglets—Anelloviridae, Circoviridae, Picornaviridae and Reoviridae—only Reoviridae includes well established viral enteropathogens causing piglet diarrhoea [2,37,38]. Rotavirus A and C were found in herd B (both in samples from healthy piglets and in the pooled diarrhoeic sample) and Rotavirus A in the pooled diarrhoeic sample from herd A. Previous investigations of the same animals have shown that common bacteriological enteropathogens were either absent (Clostridium perfringens type C), rare (enterotoxigenic Escherichia coli—only detected in two animals from herd A and E, respectively) or unrelated to the presence of diarrhoea (Clostridium perfringens type A and Clostridium difficile) [13]. However, 60% of the diarrhoeic animals in herd B, C, F, H, I, and J were found to be colonised in the small intestine by enteroadherent Enterococcus (E.) hirae [39]. Small intestinal villus atrophy was observed in five diarrhoeic piglets in total and small intestinal epithelial lesions 13. The majority of these animals were colonised by E. hirae (4 of 5 with villous atrophy and 10/13 with epithelial lesions). Although it is not possible to relate viral findings to individual piglets, two of the piglets with small intestinal atrophy originated from herd B (none from herd A) and it is hence possible that rotavirus contributed to the intestinal pathology seen in these animals. However, overall the results of this study do not show a major contribution of known viruses to the investigated diarrhoea.

The composition of mammalian viruses in the intestine of healthy and diarrhoeic neonatal piglets

A number of different virus families known to infect mammals were detected in the present study (Adenoviridae, Anelloviridae, Astroviridae, Caliciviridae, Circoviridae, Parvoviridae, Picornaviridae, and Reoviridae). These findings agree with recent studies on the porcine faecal virome indicating that the general composition of mammalian viruses in the porcine intestine is similar, despite differences in geographical location and age of the sampled pigs [2123,4042]. A notable difference, however, is the absence of the coronaviruses TGEV and PEDV in our study. This dissimilarity was expected since TGEV and PEDV have never been detected in Sweden.

Comparisons of the faecal virome between healthy and diarrhoeic pigs in a larger number of animals have so far been limited to two studies investigating piglets at an age of 19–30 days [21,22]. Similar to the present study, putative co-infections were common. Zhang et al. (2014) [21] reported a mean of 2 viral families in the faeces of healthy pigs (n = 29) compared to 3.8 viral families in diarrhoeic pigs (n = 27). Similarly, Shan et al. (2011) [22] found an average of 2.75 viral families in faecal samples from healthy pigs (n = 24) and 3.7 in diarrhoeic pigs (n = 12). When comparing our results with these previous studies, it should be noted that we used intestinal tissue instead of faeces. In addition, the diarrhoeic samples were pooled in the present study (each pool containing samples from five pigs) and thus the number of viral families in individual diarrhoeic piglets is unknown. Interestingly, compared to previous studies, we found an overall smaller number of different virus families both in samples from healthy individual piglets (0–3, mean = 1.5) and in pooled samples from diarrhoeic piglets (0–2, mean = 0.8). This difference is likely due to the lower age of the piglets in the present study (median age 1 day for diarrhoeic piglets and median age 2 days for controls). An additional factor that could have contributed to the low number of viruses in neonatal piglets is the passive immunity received through colostrum.

Another contradiction with previous studies is that we found a greater number of different virus families in healthy piglets compared to the pooled diarrhoeic samples. Two families or more were detected in 2/10 pooled diarrhoeic samples compared to 10/19 from healthy piglets. This could be related to age, as healthy piglets were slightly older. Alternatively, the diarrhoea could have led to a decreased complexity of the intestinal virome. These results call for further studies including analysis of individual diarrhoeic pigs.

Detection of Aichivirus C, eukaryotic small circular DNA viruses, and IAS virus

The most common findings were sequences belonging to Picornaviridae and eukaryotic circular single-stranded DNA viruses classified as Circoviridae. The majority of Picornaviridae-sequences—detected in 10/19 healthy piglets and in 4/10 pooled diarrhoeic samples—were classified at species level as Aichivirus C. A number of studies have reported a high prevalence of Aichivirus C in faeces from both healthy and diarrhoeic piglets [4345] and an association with diarrhoea is hence controversial. The transmission route of Aichivirus C is thought to be faecal-oral, but the virus has also been detected in sera only three days after birth, demonstrating its capacity to escape the gastrointestinal tract [46]. This, together with the present detection of Aichivirus C in the intestine within 24–48 h from birth, calls for further investigations of possible transplacental or lactogenic transmission routes.

Several sequences in the present study were identified as unclassified Circoviridae (po-circo-like virus 21, 22, 51 and fur seal faeces associated circular DNA virus) or as unassigned small circular ssDNA viruses. These newly discovered unassigned small circular ssDNA viruses, are highly divergent viruses encoding a well-conserved replication initiator protein (Rep) involved in rolling circle replication [47]. Recently, circular Rep -encoding ssDNA (CRESS-DNA) viruses have been identified in a variety of samples including insects, plants, sewage and mammalian faeces, but the origin and host of these viruses are presently unclear [4750]. Interestingly, we found sequences classified as CRESS-DNA viruses in only clinically healthy piglets. These results agree with the hypothesis that these viruses might be of dietary origin or possibly a commensal part of the eukaryotic virome [49,51].

Interestingly, a small number of sequences from three healthy piglets shared similarities with immunodeficiency -associated stool virus (IASV). This large dsDNA virus has previously been reported in only one study on diarrhoea in humans with advanced-stage HIV infection [52]. The IAS virus is highly divergent from previously characterised viruses and further characterisation of our sequences is required to establish if this in fact is an eukaryotic virus.

Considerations for the use of viral metagenomics in the analysis of clinical samples

As this study relied on a viral metagenomics approach for investigating the virome, the quality of the sequence reads is highly important. The present study generated an average of 220 Mbases of good-quality data per clinical sample, with a mean read length of 185 bases. The NTC and process control followed the same pattern as the clinical samples, but at an analysis level, reported no significant hits towards known viral families. However, the positive control generated 6.3 Gbases of data.

Endogenous viral elements (EVE) were detected within all samples. However, no porcine EVE was detected within the process control and the NTC. In the positive control, porcine EVE sequences were detected which is probably due to the use of porcine cell lines for culturing viruses.

All samples were subjected to three analytical methods suited for detecting sequences in metagenomics datasets, Kraken, Diamond and HMM. As Kraken is only detecting similar sequences at a nucleotide level, two additional methods were used for detecting more distant homologues; the BLASTx equivalent of Diamond and HMMER3 for scanning predicted protein sequences (using FGS) for viral profiles (vFam). Together all three methods enable not only detection of viruses on species level but also a broader detection on family level. As a consequence, some additional viral families were detected only by protein homology. Diamond and HMM/vFam detected putative Circoviridae sequences in five different samples. By classification with Diamond, Picobirnaviridae homologues were classified on species level to porcine picobirnavirus, however this concerns only four reads in one diarrhoeic sample. Porcine picobirnavirus has frequently been detected in pigs but its association to diarrhoea is uncertain [53,54]. Moreover, classifications with only HMM/vFam included additional hits towards Caliciviridae, Adenoviridae, Reoviridae, Parvoviridae and Circoviridae, indicating the presence of proteins homologous to proteins from viruses belonging to these families. However, the results did not show any consistent pattern in relation to the piglets’ health status.

The mammalian intestinal virome in neonates

The present study showed that viral infections were present as early as 24–48 h after birth. The presence of several different eukaryotic viruses in the intestine of healthy piglets suggests that some of the viruses may be regular constituents of the porcine intestinal virome, whereas others may be capable of causing disease under certain circumstances. As an increasing number of eukaryotic viruses are being detected in mammalian tissues without any overt signs of disease in the host, the concept of commensal virus infections has been proposed [19,20]. It has recently also been suggested that eukaryotic viruses may be important for gut homeostasis as the presence of a common enteric RNA virus was able to replace the beneficial functions of commensal bacteria in mice [55].

Studies on the neonatal intestinal virome in mammals younger than a week of age are limited to one recently published study in humans [20]. The number of eukaryotic viruses was low in the first week of life (similar to our study) and thereafter the complexity increases over time. This is in agreement with the development of the porcine intestinal virome, based on the present and previous studies [21,22].

Conclusions

Given previous knowledge of gastrointestinal viruses and their load in faeces as well as the intestinal mucosa during clinical disease, it is highly unlikely that a known virus would have remained undetected [56,57]. Our data did not support the hypothesis that NNPDS is caused by known mammalian viruses. However, the metagenomics analyses resulted in a preliminary list of viruses present in diarrhoeic and healthy neonatal piglets in Sweden, providing a starting point for future investigations. The study also concludes that viruses are present in the intestine as early on as 24–48 h after birth, indicating immediate infection post-partum, alternatively, transplacental infection. Nevertheless, the findings do require in vitro validation as well as full genomic characterisation to fully establish the diversity of the porcine neonatal virome. Further, the smaller number of different viruses found in this study on neonatal pigs compared to previous studies on pigs around weaning [21,22] support recent findings in humans demonstrating a progressive establishment of the intestinal virome [20]. These findings warrant further longitudinal studies on the early-life dynamics of the developing intestinal virome.

Acknowledgments

The authors would like to acknowledge Rodrigo Grandon, Department of Biomedical Sciences and Veterinary Public Health, SLU, Uppsala, Sweden for help during sample collection.

The authors would like to acknowledge support of the National Genomics Infrastructure (NGI) / Uppsala Genome Center and UPPMAX for providing assistance in massive parallel sequencing and computational infrastructure.

Support for the analyses of this project was provided by the SLU Global Bioinformatics Centre, part of the infrastructure at faculty Veterinary Medicine and Animal Science, SLU, Uppsala, Sweden.

The authors would also like to acknowledge the support from all animal owners.

Author Contributions

Conceived and designed the experiments: OEK JL MJ MB. Performed the experiments: OEK JL. Analyzed the data: OEK JL JH. Contributed reagents/materials/analysis tools: MJ MB JH. Wrote the paper: OEK JL JH MB MJ.

References

  1. 1. Svensmark B, Jorsal SE, Nielsen K, Willeberg P. Epidemiological studies of piglet diarrhoea in intensively managed Danish sow herds. I. Pre-weaning diarrhoea. Acta Vet Scand. 1989;30: 43–53. Available: http://europepmc.org/abstract/med/2782232 pmid:2782232
  2. 2. Zimmerman J, Karriker L, Ramirez A, Schwartz K, Stevenson G. Digestive System. In: Diseases of Swine. 10th ed. Hoboken N.J.: John Wiley & Sons; 2012. pp. 199–226.
  3. 3. Sjölund M, Zoric, M., Wallgren P. Financial impact of disease on pig production. Part III. Gastrointestinal disorders. Proceedings of the 6th European Symposium of Porcine Health Management. Sorrento, Italy; 2014. p. 111.
  4. 4. Shimizu M, Shirai J, Narita M, Yamane T. Cytopathic astrovirus isolated from porcine acute gastroenteritis in an established cell line derived from porcine embryonic kidney. J Clin Microbiol. 1990;28: 201–206. pmid:2107200
  5. 5. Mor SK, Chander Y, Marthaler D, Patnayak DP, Goyal SM. Detection and molecular characterization of Porcine astrovirus strains associated with swine diarrhea. J Vet Diagnostic Investig. 2012;24: 1064–1067.
  6. 6. Li B, Ma J, Xiao S, Fang L, Zeng S, Wen L, et al. Complete Genome Sequence of a Novel Species of Porcine Bocavirus, PBoV5. J Virol. 2012;86: 1286–1287. pmid:22205722
  7. 7. Park SJ, Kim HK, Moon HJ, Song DS, Rho SM, Han JY, et al. Molecular detection of porcine kobuviruses in pigs in Korea and their association with diarrhea. Arch Virol. 2010;155: 1803–11. pmid:20820823
  8. 8. Khamrin P, Maneekarn N, Kongkaew A, Kongkaew S, Okitsu S, Ushijima H. Porcine Kobuvirus in Piglets, Thailand. Emerg Infect Dis. 2009;15: 2075–2077. pmid:19961712
  9. 9. Zhang W, Shen Q, Hua X, Cui L, Liu J, Yang S. The first Chinese porcine sapovirus strain that contributed to an outbreak of gastroenteritis in piglets. J Virol. 2008;82: 8239–40. pmid:18508889
  10. 10. Lan D, Ji W, Yang S, Cui L, Yang Z, Yuan C, et al. Isolation and characterization of the first Chinese porcine sapelovirus strain. Arch Virol. 2011;156: 1567–74. pmid:21618029
  11. 11. Kongsted H, Jonach B, Haugegaard S, Angen O, Jorsal SE, Kokotovic B, et al. Microbiological, pathological and histological findings in four Danish pig herds affected by a new neonatal diarrhoea syndrome. BMC Vet Res. 2013;9: 206. pmid:24119974
  12. 12. Gin T, Le Guennec, J., Morvan, H., Martineau G-P. Clinical and laboratory investigations in 10 French pig herds dealing with enzootic neonatal diarrhoeatle. Procedings of the 21st international pig veterinary society (IPVS) congress. Vancouver, Canada; 2010. p. 758. Available: http://orbit.dtu.dk/fedora/objects/orbit:63423/datastreams/file_5574380/content
  13. 13. Larsson J, Aspán A, Lindberg R, Grandon R, Båverud V, Fall N, et al. Pathological and bacteriological characterization of neonatal porcine diarrhoea of uncertain aetiology. J Med Microbiol. Microbiology Society; 2015;64: 916–26.
  14. 14. P. Wallgren, S.Mattsson MM. New neonatal porcine diarrhoea: aspects on etiology. Procedings of the 22nd international pig veterinary society (IPVS) congress. Jeju, Korea; 2012. p. 76. Available: http://www.sva.se/globalassets/redesign2011/pdf/djurhalsa/gris/kongressrapporter/6_ipvs2012_jeju_sydkorea.pdf
  15. 15. Belák S, Karlsson OE, Blomström A-LL, Berg M, Granberg F. New viruses in veterinary medicine, detected by metagenomic approaches. Vet Microbiol. 2013;165: 95–101. pmid:23428379
  16. 16. Delwart EL. Viral metagenomics. Reviews in Medical Virology. 2007. pp. 115–131. pmid:17295196
  17. 17. Hugenholtz P, Tyson GW. Metagenomics. Nature. 2008;455: 481–483. pmid:18818648
  18. 18. Gevers D, Knight R, Petrosino JF, Huang K, McGuire AL, Birren BW, et al. The Human Microbiome Project: A Community Resource for the Healthy Human Microbiome. PLoS Biol. 2012;10.
  19. 19. Virgin HW. The virome in mammalian physiology and disease. Cell. 2014. pp. 142–150. pmid:24679532
  20. 20. Lim ES, Zhou Y, Zhao G, Bauer IK, Droit L, Ndao IM, et al. Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med. 2015;21: 1228–1234. pmid:26366711
  21. 21. Zhang B, Tang C, Yue H, Ren Y, Song Z. Viral metagenomics analysis demonstrates the diversity of viral flora in piglet diarrhoeic faeces in China. J Gen Virol. 2014;95: 1603–1611. pmid:24718833
  22. 22. Shan T, Li L, Simmonds P, Wang C, Moeser A, Delwart E. The fecal virome of pigs on a high-density farm. J Virol. 2011;85: 11697–708. pmid:21900163
  23. 23. Lager KM, Ng TF, Bayles DO, Alt DP, Delwart EL, Cheung AK. Diversity of viruses detected by deep sequencing in pigs from a common background. Journal of Veterinary Diagnostic Investigation. 2012. pp. 1177–1179. pmid:23051826
  24. 24. Daly GM, Bexfield N, Heaney J, Stubbs S, Mayer AP, Palser A, et al. A viral discovery methodology for clinical biopsy samples utilising massively parallel next generation sequencing. PLoS One. 2011;6.
  25. 25. Blomström AL, Widén F, Hammer AS, Belák S, Berg M. Detection of a novel astrovirus in brain tissue of mink suffering from shaking mink syndrome by use of viral metagenomics. J Clin Microbiol. 2010;48: 4392–4396. pmid:20926705
  26. 26. Rosseel T, Van Borm S, Vandenbussche F, Hoffmann B, van den Berg T, Beer M, et al. The Origin of Biased Sequence Depth in Sequence-Independent Nucleic Acid Amplification and Optimization for Efficient Massive Parallel Sequencing. PLoS One. 2013;8.
  27. 27. Granberg F, Karlsson OE, Belák S. Metagenomic approaches to disclose disease-associated pathogens: detection of viral pathogens in honeybees. Methods Mol Biol. 2015;1247: 491–511. pmid:25399116
  28. 28. Schmieder R, Edwards R. Quality control and preprocessing of metagenomic datasets. Bioinformatics. 2011;27: 863–864. pmid:21278185
  29. 29. Ewing B, Green P. Base-calling of automated sequencer traces using phred. II. Error probabilities. Genome Res. 1998;8: 186–194. pmid:9521922
  30. 30. Groenen MA, Archibald AL, Uenishi H, Tuggle CK, Takeuchi Y, Rothschild MF, et al. Analyses of pig genomes provide insight into porcine demography and evolution. Nature. 2012;491: 393–398. pmid:23151582
  31. 31. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nature Methods. 2012. pp. 357–359. pmid:22388286
  32. 32. Wood DE, Salzberg SL. Kraken: ultrafast metagenomic sequence classification using exact alignments. Genome Biol. 2014;15: R46. pmid:24580807
  33. 33. Buchfink B, Xie C, Huson DH. Fast and sensitive protein alignment using DIAMOND. Nat Methods. 2014;12: 59–60. pmid:25402007
  34. 34. Rho M, Tang H, Ye Y. FragGeneScan: Predicting genes in short and error-prone reads. Nucleic Acids Res. 2010;38: 1–12.
  35. 35. Skewes-Cox P, Sharpton TJ, Pollard KS, DeRisi JL. Profile hidden Markov models for the detection of viruses within metagenomic sequence data. PLoS One. 2014;9: e105067. pmid:25140992
  36. 36. Pruitt K, Brown G, Tatusova T, Maglott D. The Reference Sequence (RefSeq) Project. In: The NCBI handbook. 2nd ed. Bethesda: National Center for Biotechnology Information; 2002. Available: http://www.ncbi.nlm.nih.gov/books/NBK21091/
  37. 37. Morin M, Magar R, Robinson Y. Porcine group C rotavirus as a cause of neonatal diarrhea in a Quebec swine herd. Can J Vet Res. 1990;54: 385–389. pmid:2165848
  38. 38. Will L a, Paul PS, Proescholdt T a, Aktar SN, Flaming KP, Janke BH, et al. Evaluation of rotavirus infection and diarrhea in Iowa commercial pigs based on an epidemiologic study of a population represented by diagnostic laboratory cases. J Vet Diagn Invest. 1994;6: 416–22. pmid:7858020
  39. 39. Larsson J, Lindberg R, Aspán A, Grandon R, Westergren E, Jacobson M. Neonatal Piglet Diarrhoea Associated with Enteroadherent Enterococcus hirae. J Comp Pathol. 2014;151: 137–147. pmid:24915885
  40. 40. Sachsenröder J, Twardziok S, Hammerl JA, Janczyk P, Wrede P, Hertwig S, et al. Simultaneous identification of DNA and RNA viruses present in pig faeces using process-controlled deep sequencing. PLoS One. 2012;7: e34631. pmid:22514648
  41. 41. Sachsenröder J, Twardziok SO, Scheuch M, Johne R. The general composition of the faecal virome of pigs depends on age, but not on feeding with a probiotic bacterium. PLoS One. Public Library of Science; 2014;9: e88888.
  42. 42. Dumarest M, Muth E, Cheval J, Gratigny M, Hébert C, Gagnieur L, et al. Viral diversity in swine intestinal mucus used for the manufacture of heparin as analyzed by high-throughput sequencing. Biologicals. 2015;43: 31–6. pmid:25466699
  43. 43. Verma H, Mor SK, Abdel-Glil MY, Goyal SM. Identification and molecular characterization of porcine kobuvirus in U. S. swine. Virus Genes. 2013;46: 551–553. pmid:23334473
  44. 44. Di Bartolo I, Angeloni G, Tofani S, Monini M, Ruggeri FM. Infection of farmed pigs with porcine kobuviruses in Italy. Arch Virol. 2015;160: 1533–6. pmid:25809018
  45. 45. An DJ, Jeoung HY, Jeong W, Lee HS, Park JY, Kim B. Porcine kobuvirus from pig stool in Korea. Virus Genes. 2011;42: 208–211. pmid:21161358
  46. 46. Barry AF, Ribeiro J, Alfieri AF, van der Poel WHM, Alfieri AA. First detection of kobuvirus in farm animals in Brazil and the Netherlands. Infect Genet Evol. 2011;11: 1811–1814. pmid:21763785
  47. 47. Rosario K, Duffy S, Breitbart M. A field guide to eukaryotic circular single-stranded DNA viruses: Insights gained from metagenomics. Arch Virol. 2012;157: 1851–1871. pmid:22760663
  48. 48. Kraberger S, Argüello-Astorga GR, Greenfield LG, Galilee C, Law D, Martin DP, et al. Characterisation of a diverse range of circular replication-associated protein encoding DNA viruses recovered from a sewage treatment oxidation pond. Infect Genet Evol. 2015;31: 73–86. pmid:25583447
  49. 49. Delwart E, Li L. Rapidly expanding genetic diversity and host range of the Circoviridae viral family and other Rep encoding small circular ssDNA genomes. Virus Res. 2012;164: 114–121. pmid:22155583
  50. 50. Rosario K, Dayaram A, Marinov M, Ware J, Kraberger S, Stainton D, et al. Diverse circular ssDNA viruses discovered in dragonflies (Odonata: Epiprocta). J Gen Virol. 2012;93: 2668–81. pmid:22915694
  51. 51. Phan TG, Mori D, Deng X, Rajindrajith S, Ranawaka U, Fan Ng TF, et al. Small circular single stranded DNA viral genomes in unexplained cases of human encephalitis, diarrhea, and in untreated sewage. Virology. 2015;482: 98–104. pmid:25839169
  52. 52. Oude Munnink BB, Canuti M, Deijs M, de Vries M, Jebbink MF, Rebers S, et al. Unexplained diarrhoea in HIV-1 infected individuals. BMC Infect Dis. 2014;14: 22. pmid:24410947
  53. 53. Ganesh B, Banyai K, Kanungo S, Sur D, Malik YS, Kobayashi N, et al. Detection and Molecular Characterization of Porcine Picobirnavirus in Feces of Domestic Pigs from Kolkata, India. Indian J Virol. 2012;23: 387–391. pmid:24293831
  54. 54. Martínez LC, Masachessi G, Carruyo G, Ferreyra LJ, Barril PA, Isa MB, et al. Picobirnavirus causes persistent infection in pigs. Infect Genet Evol. 2010;10: 984–988. pmid:20601172
  55. 55. Kernbauer E, Ding Y, Cadwell K. An enteric virus can replace the beneficial function of commensal bacteria. Nature. 2014;516: 94–8. pmid:25409145
  56. 56. Karlsson OE, Belák S, Granberg F. The effect of preprocessing by sequence-independent, single-primer amplification (SISPA) on metagenomic detection of viruses. Biosecur Bioterror. 2013;11 Suppl 1: S227–34. pmid:23971810
  57. 57. Cheval J, Sauvage V, Frangeul L, Dacheux L, Guigon G, Dumey N, et al. Evaluation of high-throughput sequencing for identifying known and unknown viruses in biological samples. J Clin Microbiol. 2011;49: 3268–3275. pmid:21715589