Next Article in Journal
Physicochemical Quality and Fatty Acid Profile in the Meat of Goats Fed Forage Cactus as a Substitute for Tifton 85 Hay
Next Article in Special Issue
Molecular Survey and Genetic Characteristics of Vector-Borne Pathogens in Domestic Dogs from Four Regions of China
Previous Article in Journal
Development of an In Vivo Extended-Spectrum Cephalosporin-Resistant Escherichia coli Model in Post-Weaned Pigs and Its Use in Assessment of Dietary Interventions
Previous Article in Special Issue
Echocardiographic Assessment of the Pulmonary Vein to Pulmonary Artery Ratio in Canine Heartworm Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Giardia duodenalis Colonization Slightly Affects Gut Microbiota and Hematological Parameters in Clinically Healthy Dogs

1
Laboratory of Microbial Ecology and Genomics, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy
2
Laboratory of Histopathology, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy
3
Laboratory of Enhancement of Food Production, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy
4
Laboratory of Animal Medicine, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy
5
Laboratory of Parasitology, Mycology and Medical Entomology, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy
6
Corso Allevamento e Addestramento Cinofili, Guardia di Finanza, 06061 Castiglione del Lago, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Animals 2023, 13(6), 958; https://doi.org/10.3390/ani13060958
Submission received: 23 January 2023 / Revised: 27 February 2023 / Accepted: 28 February 2023 / Published: 7 March 2023
(This article belongs to the Special Issue Parasitic Diseases in Canines and Felines and Their Vectors)

Abstract

:

Simple Summary

Giardia is a worldwide cause of acute diarrheal disease both in humans and animals. Domestic dogs may be either asymptomatic or may show clinical signs. Here, we report a study comparing two groups of clinically healthy German shepherd dogs differing for G. duodenalis colonization. Gut microbiota and the hematological, biochemical, and fecal parameters related to intestinal function were investigated. The results display a scenario in which G. duodenalis exerts an effect upon the gut microbiota affecting the proportion of a few bacterial taxa known to be associated with improved lipids metabolism and protection from gut inflammation. This also suggests that the antiparasitic treatments that are usually administered to G. duodenalis-positive dogs might be avoided in clinically healthy subjects since the presence of G. duodenalis does not substantially modify the microbial ecology of the intestinal lumen nor the hematological markers of disease.

Abstract

Giardia duodenalis (Giardia) is a worldwide cause of acute diarrheal disease both in humans and animals. The primary aim of this study was to investigate possible variations in gut microbiota in a population of asymptomatic dogs (n = 31), naturally infected or not by Giardia. Gut microbiota and the hematological, biochemical, and fecal parameters related to intestinal function were investigated. Giardia infection was associated with a significant shift of beta diversity, showing a relevant reduction of Gammaproteobacteria and an increase of Fusobacteria in male-positive dogs if compared with negatives. A significant imbalance of different bacterial taxa, with particular reference to the Erysipelotrichales, Lactobacillales, Clostridiales, and Burkholderiales orders, was observed, with the first two being higher in Giardia-positive dogs. Giardia-positive males displayed significantly higher values of cCRP than negative males as well as positive females, supporting the presence of a pro-inflammatory state. Taken together, these results indicate that the presence of Giardia does not substantially modify the microbial ecology of the intestine nor the hematological markers of disease. Thus treatments against Giardia should be considered with caution in asymptomatic subjects.

1. Introduction

Giardia duodenalis (Giardia) is a common worldwide parasite of both humans and domestic animals, and it is currently recognized as the most prevalent gastrointestinal parasite in domestic dogs, closely followed by hookworms and coccidian [1,2,3,4,5]. The role of Giardia in causing a broad range of clinical manifestations, from asymptomatic to acute/chronic diarrheal disease, remains a matter of debate. Even though Giardia is frequently detected in diarrheic animals, particularly in puppies, many hosts remain asymptomatic despite shedding high numbers of environmentally resistant cysts [6]. The main sustained hypothesis on giardiosis and associated sequelae is that parasite attachment causes the loss of epithelial barrier function [7,8], favoring the penetration of intestinal bacteria into the inflamed intestinal wall, resulting in permanent damage to the intestinal epithelium [9]. Some authors postulated that changes in the resident intestinal microflora are responsible for the disease outcome of giardiosis [10]. However, the host–parasite interaction is not a one-way process, and changes in the host microbiome itself may favor the contact between parasites and host cells. Such changes may be caused by different stresses, such as nutritional or environmental changes, infections, or drug and antimicrobial treatments [6]. Thus, whether Giardia is a commensal or a parasite, it is conceivable that the perturbation of the host–parasite equilibrium may be the basis of some pathogenicity and may explain variations in symptoms both between hosts and within the same host over time [6]. If host microbiota may be primarily or secondarily involved in Giardia infection outcomes, many factors other than Giardia may affect the gut microbiome, potentially masking the Giardia microbiome relationship. The primary aim of this study was to investigate the impact of Giardia on gut microbiota in a homogeneous population of naturally infected dogs living in the same breeding facility. Moreover, the hematological, biochemical, and fecal parameters related to intestinal function were investigated.

2. Materials and Methods

2.1. Sample Description

Thirty-one German shepherd dogs living in the same breeding and training facility of the Italian Finance Police were included in the study. The dogs were housed in individual boxes and fed with the same commercial maintenance dry food (Crude protein: 29%, Crude fiber: 2%, Crude fat: 18%, Crude ash: 7.5%, Calcium: 1.3%). All dogs were annually vaccinated against the canine distemper virus (CDV), canine parvovirus (CPV), canine adenovirus (CAV), and Leptospirosis, regularly treated by anthelminthic drugs, and protected against ectoparasites using a slow-release insecticidal and repellent collar. The inclusion criteria were: (i) clinically healthy, without evidence of gastrointestinal disorders; (ii) no pharmacological therapy in the 2 months before the study; (iii) tested negative with a standard sedimentation-floatation coprological test. From each dog, the following samples were collected: a minimum of 10 g of fecal samples for parasitological and biochemical analysis; 2 independent biological replicates of rectal fecal swabs (FecalSwabTM, Copan Diagnostics Inc, Brescia, Italy) for microbial community analysis; 1 K3-EDTA and 1 plain tube of peripheral blood for hematological and biochemical investigations, respectively. Aseptic techniques and disposable equipment were used for each sample.

2.2. Giardia and Cryptosporidium spp. Detection and Quantification

The detection of Giardia and Cryptosporidium spp. was performed in fecal samples using the commercially available immunofluorescence test according to manufacturer instructions (Merifluor® Cryptosporidium/Giardia, Meridian Bioscience, Cincinnati, OH, USA). Quantification of Giardia cysts and Cryptosporidium spp. oocysts was made by counting protozoa elements under the microscope and expressed as a number of (oo-) cysts per 1 g of fecal sample to a maximum of 50,000. Specimens with higher parasite (oo-) cysts were recorded in the report as >50,000.

2.3. Microbial Community Analyses

2.3.1. DNA Extraction

Total DNA for metataxonomic analysis was extracted using a column-based kit (QIAamp DNA Mini Kit, QIAGEN, Hilden, Germany) starting from 200 μL of fecal sample in swab’s buffer (Modified Cary Blair medium), following the manufacturer’s instruction. Thermal lysis was carried out at 56 °C for 2 h, and RNaseA (70 Kunitz units/mg protein) was added to each sample to ensure RNA-free preparation. Total DNA was resuspended in 200 μL of nuclease-free water and stored at −20 °C until library preparation for sequencing.

2.3.2. 16S rRNA Sequencing

Extracted DNA was used as a template in amplicon PCR to target the hypervariable V3 and V4 regions of the bacterial 16S rRNA gene. The amplification check was performed by 2% TAE agarose gel electrophoresis to identify a DNA fragment accounting for 550 bp length. The 16S library was prepared according to the Illumina 16S Metagenomic sequencing Library Preparation protocol, using the primers Bact341F and Bact785R (Fwd: CCTACGGGNGGCWGCAG and Rev:GACTACHVGGGTATCTAATCC) previously described by Klindworth A et al. [11] using the Nextera XT DNA Library Prep kit (Illumina, Milano, Italy). PCR clean-up was performed with Agencourt AMPure XP beads (Beckman Coulter Genomics, Indianapolis, IN, USA). Libraries were checked for both concentration and quality using Qubit and 2200 TapeStation (Agilent, Milano, Italy), respectively. Samples were equimolarity pooled, and sequencing was performed with an Illumina MiSeq platform using a MiSeq 600V3 cartridge (600 cycles, 2 × 300 bp, paired-end reads). Read sequences were deposited in the Sequence Read Archive (SRA) of the NCBI under the BioProject PRJNA736250.

2.3.3. Reads Preprocessing and OTU Table Construction

After sequencing, data underwent a quality control procedure using the FastQC tool [12]. Data were then cleaned by removing adapters and primers and performing the dereplication of sequences using an in-house bash script. In addition, data were filtered based on the quality and length of the reads so that only reads with a quality higher than a given threshold (QPhred ≥ 20) and longer than 100 bp were retained. All subsequent steps were performed using QIIME2 pipeline version 2020.2 [13]. Raw sequence data were screened, trimmed, and denoised with DADA2 [14] (parameters: p-trunc-len-f = 288; p-trunc-len-r = 264) and quality filtered based on q-score. Operational taxonomic units (OTUs) were defined as sequences with at least 97% similarity with the Greengenes database (last release May 2013, version 13.8) [15]. Samples were rarefied to 98197 sequences per sample. The rarefaction depth was based on the lowest read depth of samples.

2.4. Hematological and Biochemical Analysis

Blood EDTA samples were used for complete blood cell count (CBC) with the hematology analyzer XN-1000 V (Sysmex Europe GmbH, Norderstedt, Germany), equipped with veterinary software (Software of Automated hematology Analyzer for Animal XN-V series (Sysmex). All samples were analyzed within 8 h after blood collection. Blood plain tubes were centrifuged 3000× g for 10 min, and serum samples were collected for biochemical analysis: routine biochemical profile analysis was performed using an automated clinical chemistry analyzer (Cobas C501; Roche Diagnostics International AG, Rotkreuz, Switzerland); folate and cobalamin (B12) immunoassay analysis was performed with an automated Cobas e601 analyzer (Roche Diagnostics). Serum canine C reactive protein (cCRP) concentration (mg/L) was determined via a commercially available turbidimetric immunoassay kit (Turbovet canine CRP, Acuvet Biotech, Zaragoza, Spain) applied to a Cobas c501 analyzer, following the manufacturer’s instructions. Fecal calprotectin was determined via a species-specific ELISA kit (Canine Calprotectin CP, MyBioSource, San Diego, CA, USA), following the manufacturer’s instructions. Briefly, 10 mg of samples were homogenized in 100 μL of PBS and centrifuged for 20 min at 1500× g; the supernatant was carefully collected and stored at −80 °C until analysis. Fecal samples were then thawed and measured in a unique batch.

2.5. Statistical Analyses

The overall goal of the statistical analysis was to compare the gut microbiota and the investigated hematological and biochemical parameters with respect to Giardia infection (Giardia Positive dogs (GP) and Giardia Negative dogs (GN)). Supposing a possible effect of the dog gender, the above-mentioned analysis was performed also considering the data stratified by the gender of the dogs. Therefore, the subgroups Giardia-Positive females (GPF), Giardia-Negative females (GNF), Giardia-Positive males (GPM), and Giardia-Negative males (GNM) were also evaluated in the statistical analysis. Due to the limited number of dogs, an additional stratification for age was not carried out. Nevertheless, the information about dogs’ age was used to check whether the investigated groups had a different age distribution by the Wilcoxon test. R 4.0.5 software [16], QIIme2 [13], and MetaboAnalyst 5.0 web portal [17] were used to conduct the statistical analysis. p-value < 0.05 was considered significant.

2.5.1. Analysis of 16S rRNA Gene

Alpha diversity analysis was performed on the preprocessed count table and was measured by means of the Chao1 index to assess the richness. The Shannon index was used to assess the evenness and observed OTUs metrics to describe the community structure. The non-parametric Wilcoxon test was used to compare the alpha diversity and the OTUs number distribution between the GP and GN dogs. In addition, the non-parametric Kruskal–Wallis test was conducted to compare the GPF, GNF, GPM, and GNM groups. If significant, the pairwise comparison Wilcoxon test was performed by adjusting the p-value (padj-value) for multiple comparisons using the false discovery rate (FDR) method [18]. Beta diversity was evaluated with the phylogeny-based Unifrac distance metric. The PERMANOVA test (permutation number 999) was used to compare the beta diversity parameters among groups [19]. FDR correction for multiple testing was applied in pairwise comparisons between groups. The volcano plot [20] was drawn to identify the OTUs differing significantly between the groups of study. The features resulting in significance (p-value < 0.05) were selected and analyzed by partial least square discriminant analysis (PLS-DA) [21]. The most discriminating OTUs were shown in descending order of their coefficient scores. The ones with a coefficient score greater than 80 were identified and discussed.

2.5.2. Hematological and BIOCHEMICAL Analyses

The volcano plot was built to identify the hematological and biochemical parameters that differ significantly between the groups of study. The significant variables (p < 0.05) were selected and described by means of a box plot.

3. Results

3.1. Sample Description

Overall, 19 female and 12 male German Shepherd dogs were included. The median age was 19 months (range: 15–85 months). Dogs ranged from 28 to 35 kg of body weight. All dogs were non-diarrhoeic and clinically healthy.

3.2. Giardia Detection and Quantification

Overall, 13 dogs were positive for Giardia (GP), of which 8 were females (GPF), and 5 were males (GPM). All positive dogs ranged from 15 to 24 months (Table S1), showing a significantly lower age distribution in Giardia-positive dogs than negatives (p-181 value = 0.0003). Equivalent conclusions were inferred considering the age distribution among females (padj-value = 0.021) and males (padj-value = 0.046). The same percentage of Giardia infection was observed among males and females (42%). The number of Giardia cysts per 1 g of fecal sample was higher than 50,000 in six dogs, between 10,000 and 50,000 in four dogs, and less than 10,000 in three dogs (Table S1). Cryptosporidium spp. was not detected in all the examined samples.

3.3. Effect of Giardia Infection on Gut Microbial Community Ecology

After data preprocessing, a total of 705,154 reads (n = 31, mean for sample = 22,746.9, SD = 7254.902) were retained for bioinformatic analyses. The number of identified OTUs ranged between 200 and 600 if considering the entire sample with the exception of dog number 9, presenting a number of identified OTUs higher than 1000. No significant differences were found in the number of OTUs between GP and GN. Focusing on gender, no significant differences were found between GPF and GNF and between GPM and GNM. Considering the positive dogs, no significant differences were found in the number of OTUs between GPF and GPM, while the number of OTUs was significantly higher in GNF than in GNM (padj-value = 0.011) (Figure 1). Alpha diversity was analyzed using Chao1 and Shannon indices. Chao1 indices were 470.11 ± 94.93 (GP), 596.93 ± 501.66 (GN), 495.80 ± 60.66 (GPF), 429.88 ± 129.96 (GPM), 765.38 ± 584.98 (GNF) and 332.22 ± 94.70 (GNM) while Shannon indices were 7.90 ± 0.73 (GP), 7.89 ± 0.78 (GN), 8.07 ± 0.45 (GPF), 7.65 ± 1.06 (GPM), 8.32 ± 0.5 (GNF), and 7.20 ± 0.64 (GNM). No statistically significant difference between GP and GN groups with respect to both Chao1 and Shannon indices was observed. The same result was found when comparing GPF and GNF and between GPM and GNM. Focusing on positive dogs, no statistically significant difference was observed between GPF and GPM for both tested indices, whereas comparing GNF and GNM, the alpha diversity indices 204 Chao1 and the Shannon were significantly higher in GNF than GNM (padj-value = 0.0057 and padj-value = 0.043, respectively) (Figure 1). The unique OTUs of different groups were summarized according to the result of the OTU clustering analysis in Table 1.
The OTUs table is reported in Table S2. The beta diversity index between the GP and GN groups was significantly different (p-value = 0.0025). The same results were found in the comparison between GNF and GPF (padj-value = 0.02), whereas no statistically significant difference was observed between GNM and GPM. Focusing on negative dogs, the beta diversity index was different between the GNF and GNM groups (padj-value = 0.02), while no statistically significant differences were found between GPF and GPM groups. To display the proportion of different taxa at the class level, the bar plot reported in Figure 2 was generated based on the relative abundance of taxa. The results showed that 90% of microorganisms in the fecal samples of the entire dataset belonged to seven classes (Bacilli, Bacteroida, Betaproteobacteria, Clostridia, Eryspelotrichi, Fusobacteria, Gammaproteobacteria) (Figure 2A). The comparison between GNF and GNM groups revealed the presence of three classes unevenly distributed: Gammaproteobacteria, Bacteroida, and Fusobacteria. The Gammaproteobacteria were found with a higher frequency in the GNM group than the GNF one; on the contrary, the two classes of Bacteroida and Fusobacteria displayed a higher frequency in GNF than in GNM (Figure 2B). A very similar distribution in the relative taxa frequency was found between the GPF and GPM groups (Figure 2C).
Figure 3 reports the results of volcano plots identifying the OTUs differing significantly (p < 0.05) between GN and GP (a), GNF and GPF (b), and GNM and GPM (c).
The outputs of the PLS-DA analysis, applied to the previously identified significant OTUs, were shown using the scores plots and the variable importance in projection plots (Figure 4).
Moreover, in the supplementary results, the volcano plots and the outputs of the PLS-DA applied to GPF and GPM (a) and GNF and GNM (b) are shown (Figures S1 and S2, respectively). All 228 plots displayed a good discriminatory power, explaining from more than 50% (Figure 4a) to about 80% (Figure S2b) of the total variance of the models. The most relevant variables, corresponding to coefficient values greater than 80 for each of the five pairwise comparisons, were reported in Table 2.
The analysis revealed significant differences in the relative abundance of specific taxa between positive and negative dogs with specific reference to Bacteroidales order that was found more abundant in Giardia-positive dogs. In addition, when the positive and negative groups were broken down by gender, significant differences emerged. Specifically, within the female group, GP dogs displayed more abundance of Erysipelotrichales and Bacteroidales orders than GN ones. Within the male group, GN dogs were mainly colonized by Clostridiales than GP. Comparing gender, GPM showed a higher abundance of Clostridiales and Lactobacillales with respect to GPF and a higher abundance of Erysipelotrichales was found in both GPM and GPF, probably referring to different families or genera. Regarding GNF, they were characterized by a higher abundance of Burkholderiales compared to the GNM group, which displayed a higher abundance of Clostridiales compared to GNF (Table 2).

3.4. Hematological and Biochemical Analysis

All the tested hematological and biochemical parameters were within the range of normal values in all dogs (Table S1). The volcano plot analysis showed no significant differences in hematological and biochemical parameters between GP and GN dogs. Focusing on gender subgroups, statistically significant higher levels of Triglycerides (TG) were observed among GNF if compared with GPF (Figure 5a, p-value = 0.0051), while, at the significance limit, GPM had higher levels of cCRP than GNM (Figure 5b, p-value = 0.0725). Considering the Giardia-negative dogs, no differences were observed in the hematological and biochemical parameters between males and females. Instead, among the Giardia-positive dogs, GPF had significantly higher levels of lipase than GPM (p251 value = 0.0429) and significantly lower levels of cCRP (p-value = 0.0204) (Figures S3 and S4).

4. Discussion

The primary aim of this study was to investigate possible variations in gut microbiota in a population of asymptomatic dogs, naturally infected or not by Giardia; secondarily, we investigated possible variations in some hematological, biochemical, and fecal parameters with respect to Giardia infection. Few previous studies aimed to investigate the relationship between Giardia and gut microbiota structure and composition in dogs [22,23,24,25]. However, the majority of these studies were affected by different possible confounders such as uncontrolled lifestyles (stray dogs), different origins or breeding of dogs, concurrent parasitic infections, different clinical signs, and anthelmintic treatments [22,23,25]. In order to minimize potential biases, this study was performed on a homogeneous population belonging to the same breed, housed in the same conditions, and fed with the same commercial maintenance dry food. Moreover, all dogs included in the present study were clinically healthy, no therapies against Giardia or bacteria were administered in the previous two months, and coprological investigations excluded co-infection with other intestinal parasites. In our survey, the overall percentage of Giardia infection was 42%, with no differences between male and female dogs. Prevalence rates of Giardia infection in dogs vary depending on the population under study and diagnostic method used and can be as high as 45% [26]. Similar prevalence in the shelter and commercial kennels were previously reported [4,5,27,28]. We found that infected dogs were younger than negative ones, and this was an expected finding since previous reports showed that younger dogs have a higher risk of Giardia infection than older ones [6,28,29,30,31,32]. Similarly, it has been demonstrated that children are more likely to suffer from clinical infections than adults [33]. Differences in susceptibility to Giardia infection among different age groups are likely due to age-related shifts in microbiota composition as well as the variability of the hosts’ immune factors [34]. Even though the precise mechanisms that undergo Giardia pathogenesis are incompletely understood, a pivotal role has been postulated for microbiota by recent research [35]. Functional and compositional modifications of gut microbiota have been demonstrated during the course of Giardia infection with particular reference to changes in microbial community biodiversity and altered species abundance [36]. In our investigation, some significant differences in terms of microbial diversity were observed among tested groups. Particularly Giardia infection was associated with a significant shift of beta diversity substantiated by a relevant reduction of Gammaproteobacteria and an increase of Fusobacteria in GPM if compared with GNM. This also highlights a reduction of the differences in terms of microbial abundance between males and females in the presence of the infection. The reduction of Gammaproteobacteria and the increase of Fusobacteria have been observed in other experimental settings with regard to positive asymptomatic dogs [24]. Moreover, PLS-DA analysis displayed a significant imbalance of different bacterial taxa with particular reference to OTUs referred to as the Erysipelotrichales, Lactobacillales, Clostridiales, and Burkholderiales orders, with the first two being present with a greater extent in Giardia-positive dogs. Both Erysipelotrichales and Lactobacillales orders are of particular interest in this specific context. The occurrence of Erysipelotrichales in Giardia-positive dogs could be strongly related to the progression of the infection. Indeed, many researchers have addressed the importance of Erysipelotrichaceae in inflammation-related disorders of the gastrointestinal tract so far, with particular reference to the human context. Erysipelotrichaceae abundance levels were found to be increased in the lumen of colorectal cancer patients as compared to healthy controls [37] and to be significantly higher in the tumor group of an animal model of 1, 2-dimethylhydrazine-induced colon cancer [38]. However, high inter-host variation has been observed so far [35], probably due to the inherent differences in species related to the gut microbiota (i.e., mice and humans) and/or differences in the immune responses upon sensing bacterial ligands [39,40]. Moreover, many studies have demonstrated the association between this bacterial order and host lipid metabolisms [37,38,41,42,43,44]. Regarding the order of Lactobacillales, their involvement in contrasting infection-induced gut oxidative stresses [45] and preventing the adherence of Giardia trophozoites to the mucosal surface are well known [46]. These findings draw a possible scenario in which the microbiota of Giardia-positive subjects, on the one hand, metabolically supports the persistence of Giardia by means of Erysipelotrichaceae abundance and on the other hand, counteracts the host’s inflammation caused by Giardia itself by means of Lactobacillales action. In our study, all the biochemical parameters were within the normal ranges, and significant differences were observed between Giardia-positive and negative dogs with respect to gender. Particularly, among Giardia-positive dogs, serum lipase was significantly higher in females than in males. Moreover, GPF showed significantly lower values of triglycerides than Giardia-negative females. These findings may be linked to Giardia’s limited ability to synthesize lipids for membranes and organelles biosynthesis, energy production, and growth [47]. Thus, lipids have to be provided by the host itself and/or its gut microbiota through the conversion of primary bile acids to secondary bile acids [48]. As a consequence, the host lipid metabolism during giardiosis might have a key role in keeping the parasite persistent in the intestinal lumen [24]. When a host ingests the cysts, these enter the digestive tract, where they are stimulated by the acidic milieu in the stomach and the presence of bile and trypsin in the duodenum to develop into motile trophozoites in the proximal small intestine. In the upper intestinal tract, the trophozoites proliferate and use their adhesive disc to attach to the intestinal villi [49]. As the parasite density increases and the trophozoites descend into the lower intestinal tract, they encounter decreased cholesterol, an increase in pH, and increased concentrations of bile and lactic acid [50]. These conditions promote trophozoite differentiation into infectious cysts that are released into the environment [50]. Moderately-enhanced bile acid concentration has been found to promote Giardia growth in vitro [48]. The mechanism by which bile stimulates parasite growth is unknown, but uptake of conjugated bile salt by Giardia could reduce intraluminal bile salt concentrations and possibly interfere with micellar solubilization of fat and Giardia–bile salt interactions in vitro and in vivo [51]. Moreover, Giardia infection is associated with malabsorption of fats due to mechanical mucosal damage, leading to intestinal steatosis and increased transit of lipids into the distal small intestine and colon [51]. In our study, we investigated different functional (cobalamin, folate) and biochemical (C-reactive protein, calprotectin, alkaline phosphatase) biomarkers that have been shown to be useful indicators of intestinal inflammation in dogs [52]. Interestingly, Giardia-positive males displayed significantly higher values of cCRP than negative males as well as positive females. Even though values remained within the reference range, this finding supports the presence of a pro-inflammatory state in these subjects. Differently, no significant differences among groups were observed in the values of the other investigated parameters. Taken together, these findings are coherent with the absence of clinical symptoms and allow us to exclude severe gastro-intestinal inflammation in the investigated dogs, reinforcing the possibility of a delicate Giardia–microbiome–host equilibrium that may prevent clinical manifestations.

5. Conclusions

Investigating Giardia ecological milieu in relation to the host’s resident microbial community and its metabolic context is a very challenging task as it requires taking into consideration homogeneous cohorts that do not show evident signs of disease, such as diarrheal syndrome and intestinal malabsorption, known to compromise the luminal microbial environment and the serum biochemical profile. Here, we report for the first time a study comparing a group of Giardia-negative dogs with a group of Giardia-positive but asymptomatic subjects. The main strength of this study is the lack of confounders, such as different breeding and nutrition of dogs, concurrent parasitic infections, and clinical signs. On the other hand, a possible limit is represented by the low number of dogs considered, which may have prevented us from observing further differences in the study groups. Our results showed that the presence of Giardia exerts an effect upon the gut microbial communities, enriched in protective taxa against gut inflammation and depleted in lipid-producing taxa, potentially usable to limit Giardia infection and relieve host inflammation. Taking together the outcomes of our study suggests that treatments against Giardia should be considered with caution in asymptomatic subjects in order to save the Giardia–microbiome–host equilibrium.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ani13060958/s1, Table S1: complete analysed dataset with metadata; Figure S1: Volcano plot showing the OTUs that differ significantly between groups of study; Figure S2: Partial least squares discriminant analysis (PLS-DA) results showing the comparison between OTU data acquired; Figure S3: Volcano plot showing the hematological and biochemical parameters that differ significantly between groups GPF and GPM; Figure S4: Box plot of the significant hematological and biochemical parameters in positive dogs by gender.

Author Contributions

Conceptualization, M.V., C.L. and G.C.; methodology, M.V., C.L., M.O. and G.C.; formal analysis, M.O., A.M. and M.M.; investigation, A.P., S.P., P.D., M.M.S., R.S., A.C. and A.S.; resources, C.L., M.V. and G.C.; data curation, A.P.; writing—original draft preparation, M.V. and C.L.; writing—review and editing, M.V., C.L. and M.M.; visualization, M.O., A.P. and M.M.; supervision, C.L.; funding acquisition, G.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Italian Ministry of Health, Grant N. RC IZSVE 05/18.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the ethical committee of the Istituto Zooprofilattico Sperimentale delle Venezie (code RC-IZSVE 8/2019).

Data Availability Statement

The datasets supporting the conclusions of this article are available in the NCBI repository, BioProject n. PRJNA736250 (https://www.ncbi.nlm.nih.gov/bioproject/PRJNA736250).

Acknowledgments

The authors wish to acknowledge Susanna Pasqualotto and Valentina Cagnin for their technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Palmer, C.S.; Traub, R.J.; Robertson, I.D.; Devlin, G.; Rees, R.; Thompson, R.A. Determining the zoonotic significance of Giardia and Cryptosporidium in Australian dogs and cats. Vet. Parasitol. 2008, 154, 142–147. [Google Scholar] [CrossRef] [PubMed]
  2. Epe, C.; Rehkter, G.; Schnieder, T.; Lorentzen, L.; Kreienbrock, L. Giardia in symptomatic dogs and cats in Europe—Results of a European study. Vet. Parasitol. 2010, 173, 32–38. [Google Scholar] [CrossRef] [PubMed]
  3. Halliez, M.C.M.; Buret, A.G. Extra-intestinal and long term consequences of Giardia duodenalis infections. World J. Gastro-enterol. 2013, 19, 8974–8985. [Google Scholar] [CrossRef] [PubMed]
  4. Capelli, G.; di Regalbono, A.F.; Iorio, R.; Pietrobelli, M.; Paoletti, B.; Giangaspero, A. Giardia species and other intestinal parasites in dogs in north-east and central Italy. Vet. Rec. 2006, 159, 422–424. [Google Scholar] [CrossRef]
  5. Paoletti, B.; Iorio, R.; Capelli, G.; Sparagano, O.A.E.; Giangaspero, A. Epidemiological Scenario of Giardiosis in Dogs from Central Italy. Ann. N. Y. Acad. Sci. 2008, 1149, 371–374. [Google Scholar] [CrossRef]
  6. Tysnes, K.R.; Skancke, E.; Robertson, L.J. Subclinical Giardia in dogs: A veterinary conundrum relevant to human infection. Trends Parasitol. 2014, 30, 520–527. [Google Scholar] [CrossRef]
  7. Cotton, J.A.; Beatty, J.K.; Buret, A.G. Host parasite interactions and pathophysiology in Giardia infections. Int. J. Parasitol. 2011, 41, 925–933. [Google Scholar] [CrossRef]
  8. Bartelt, L.A.; Roche, J.; Kolling, G.; Bolick, D.; Noronha, F.; Naylor, C.; Hoffman, P.; Warren, C.; Singer, S.; Guerrant, R. Persistent G. lamblia impairs growth in a murine malnutrition model. J. Clin. Investig. 2013, 123, 2672–2684. [Google Scholar] [CrossRef] [Green Version]
  9. Chen, T.-L.; Chen, S.; Wu, H.-W.; Lee, T.-C.; Lu, Y.-Z.; Wu, L.-L.; Ni, Y.-H.; Sun, C.-H.; Yu, W.-H.; Buret, A.G.; et al. Persistent gut barrier damage and commensal bacterial influx following eradication of Giardia infection in mice. Gut Pathog. 2013, 5, 26. [Google Scholar] [CrossRef] [Green Version]
  10. Singer, S.M.; Nash, T.E. The Role of Normal Flora in Giardia lamblia Infections in Mice. J. Infect. Dis. 2000, 181, 1510–1512. [Google Scholar] [CrossRef] [Green Version]
  11. Klindworth, A.; Pruesse, E.; Schweer, T.; Peplies, J.; Quast, C.; Horn, M.; Glöckner, F.O. Evaluation of General 16S Ribosomal RNA Gene PCR Primers for Classical and Next-Generation Sequencing-Based Diversity Studies. Nucleic Acids Res. 2013, 41, e1. [Google Scholar] [CrossRef] [PubMed]
  12. Andrews, S. FastQC: A Quality Control Tool for High Throughput Sequence Data. Babraham Bioinformatics; Babraham Institute: Cambridge, UK, 2010; Available online: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 9 June 2021).
  13. Bolyen, E.; Rideout, J.R.; Dillon, M.R.; Bokulich, N.A.; Abnet, C.C.; Al-Ghalith, G.A.; Alexander, H.; Alm, E.J.; Arumugam, M.; Asnicar, F.; et al. Reproducible, interactive, scalable and extensible microbiome data science us-ing QIIME 2. Nat. Biotechnol. 2019, 37, 852–857. [Google Scholar] [CrossRef] [PubMed]
  14. Callahan, B.J.; Mcmurdie, P.J.; Rosen, M.J.; Han, A.W.; Johnson, A.J.A.; Holmes, S.P. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods 2016, 13, 581–583. [Google Scholar] [CrossRef] [Green Version]
  15. DeSantis, T.Z.; Hugenholtz, P.; Larsen, N.; Rojas, M.; Brodie, E.L.; Keller, K.; Huber, T.; Dalevi, D.; Hu, P.; Andersen, G.L. Greengenes, 406 a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl. Environ. Microbiol. 2006, 72, 5069–5072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. R Core Team (2020)—European Environment Agency. Available online: https://www.eea.europa.eu/data-and410maps/indicators/oxygen-consuming-substances-in-rivers/r-development-core-team-2006 (accessed on 23 June 2021).
  17. Pang, Z.; Chong, J.; Zhou, G.; de Lima Morais, D.A.; Chang, L.; Barrette, M.; Gauthier, C.; Jacques, P.-É.; Li, S.; Xia, J. MetaboAnalyst 5.0: Narrowing the gap between raw spectra and functional insights. Nucleic Acids Res. 2021, 49, W388–W396. [Google Scholar] [CrossRef] [PubMed]
  18. Benjamini, Y.; Hochberg, Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J. R. Stat. Soc. Ser. B Methodol. 1995, 57, 289–300. [Google Scholar] [CrossRef]
  19. Estaki, M.; Jiang, L.; Bokulich, N.A.; McDonald, D.; González, A.; Kosciolek, T.; Martino, C.; Zhu, Q.; Birmingham, A.; Vázquez-Baeza, Y.; et al. QIIME 2 Enables Comprehensive End-to-End Analysis of Diverse Microbiome Data and Comparative Studies with Publicly Available Data. Curr. Protoc. Bioinform. 2020, 70, e100. [Google Scholar] [CrossRef] [PubMed]
  20. Li, W. Volcano plots in analyzing differential expressions with mrna microarrays. J. Bioinform. Comput. Biol. 2012, 10, 1231003. [Google Scholar] [CrossRef] [Green Version]
  21. Brereton, R.G.; Lloyd, G.R. Partial least squares discriminant analysis: Taking the magic away. J. Chemom. 2014, 28, 213–225. [Google Scholar] [CrossRef]
  22. Šlapeta, J.; Dowd, S.; Alanazi, A.D.; Westman, M.E.; Brown, G.K. Differences in the faecal microbiome of non-diarrhoeic clinically healthy dogs and cats associated with Giardia duodenalis infection: Impact of hookworms and coccidia. Int. J. Parasitol. 2015, 45, 585–594. [Google Scholar] [CrossRef]
  23. Lee, N.N.; A Bidot, W.; Ericsson, A.C.; Franklin, C.L. Effects of Giardia lamblia Colonization and Fenbendazole Treatment on Canine Fecal Microbiota. J. Am. Assoc. Lab. Anim. Sci. 2020, 59, 423–429. [Google Scholar] [CrossRef] [PubMed]
  24. Berry, A.S.F.; Johnson, K.; Martins, R.; Sullivan, M.C.; Amorim, C.F.; Putre, A.; Scott, A.; Wang, S.; Lindsay, B.; Baldassano, R.N.; et al. Natural Infection with Giardia Is Associated with Altered Community Structure of the Human and Canine Gut Microbiome. Msphere 2020, 5, e00670-20. [Google Scholar] [CrossRef]
  25. Fujishiro, M.A.; Lidbury, J.A.; Pilla, R.; Steiner, J.M.; Lappin, M.R.; Suchodolski, J.S. Evaluation of the effects of anthelmintic administration on the fecal microbiome of healthy dogs with and without subclinical Giardia spp. and Cryptosporidium canis infections. PLoS ONE 2020, 15, e0228145. [Google Scholar] [CrossRef] [PubMed]
  26. Ballweber, L.R.; Xiao, L.; Bowman, D.D.; Kahn, G.; Cama, V.A. Giardiasis in dogs and cats: Update on epidemiology and public health signif-icance. Trends Parasitol. 2010, 26, 180–189. [Google Scholar] [CrossRef] [PubMed]
  27. Mundim, M.J.S.; Rosa, L.A.G.; Hortêncio, S.M.; Faria, E.; Rodrigues, R.; Cury, M. Prevalence of Giardia duodenalis and Cryptosporidium spp. in dogs from different living conditions in Uberlândia, Brazil. Vet. Parasitol. 2007, 144, 356–359. [Google Scholar] [CrossRef]
  28. Huber, F.; Bomfim, T.C.B.; Gomes, R. Comparison between natural infection by Cryptosporidium sp., Giardia sp. in dogs in two living situations in the West Zone of the municipality of Rio de Janeiro. Vet. Parasitol. 2005, 130, 69–72. [Google Scholar] [CrossRef]
  29. Uiterwijk, M.; Nijsse, R.; Kooyman, F.N.J.; Wagenaar, J.A.; Mughini-Gras, L.; Ploeger, H.W. Host factors associated with Giardia duodenalis infection in dogs across multiple diagnostic tests. Parasites Vectors 2019, 12, 556. [Google Scholar] [CrossRef] [Green Version]
  30. Claerebout, E.; Casaert, S.; Dalemans, A.-C.; De Wilde, N.; Levecke, B.; Vercruysse, J.; Geurden, T. Giardia and other intestinal parasites in different dog populations in Northern Belgium. Vet. Parasitol. 2009, 161, 41–46. [Google Scholar] [CrossRef]
  31. Bouzid, M.; Halai, K.; Jeffreys, D.; Hunter, P.R. The prevalence of Giardia infection in dogs and cats, a systematic review and meta-analysis of prevalence studies from stool samples. Vet. Parasitol. 2015, 207, 181–202. [Google Scholar] [CrossRef] [Green Version]
  32. Mircean, V.; Györke, A.; Cozma, V. Prevalence and risk factors of Giardia duodenalis in dogs from Romania. Vet. Parasitol. 2012, 184, 325–329. [Google Scholar] [CrossRef]
  33. Dhanabal, J.; Selvadoss, P.P.; Muthuswamy, K. Comparative Study of the Prevalence of Intestinal Parasites in Low Socioeconomic Areas from South Chennai, India. J. Parasitol. Res. 2014, 2014, 630968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Partida-Rodríguez, O.; Serrano-Vázquez, A.; Nieves-Ramírez, M.E.; Moran, P.; Rojas-Velázquez, L.; Portillo, T.; González, E.; Hernandez, E.G.; Finlay, B.B.; Ximenez, C. Human Intestinal Microbiota: Interaction Between Parasites and the Host Immune Response. Arch. Med. Res. 2017, 48, 690–700. [Google Scholar] [CrossRef] [PubMed]
  35. Fekete, E.; Allain, T.; Siddiq, A.; Sosnowski, O.; Buret, A.G. Giardia spp. and the Gut Microbiota: Dangerous Liaisons. Front. Microbiol. 2021, 11, 618106. [Google Scholar] [CrossRef] [PubMed]
  36. Barash, N.R.; Maloney, J.G.; Singer, S.M.; Dawson, S.C. Giardia Alters Commensal Microbial Diversity throughout the Murine Gut. Infect. Immun. 2017, 85, e00948-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Chen, W.; Liu, F.; Ling, Z.; Tong, X.; Xiang, C. Human Intestinal Lumen and Mucosa-Associated Microbiota in Patients with Colorectal Cancer. PLoS ONE 2012, 7, e39743. [Google Scholar] [CrossRef]
  38. Zhu, Q.; Jin, Z.; Wu, W.; Gao, R.; Guo, B.; Gao, Z.; Yang, Y.; Qin, H. Analysis of the Intestinal Lumen Microbiota in an Animal Model of Colorectal Cancer. PLoS ONE 2014, 9, e90849. [Google Scholar] [CrossRef]
  39. Zschaler, J.; Schlorke, D.; Arnhold, J. Differences in Innate Immune Response between Man and Mouse. Crit. Rev. Immunol. 2014, 34, 433–454. [Google Scholar] [CrossRef] [Green Version]
  40. Nguyen, T.L.A.; Vieira-Silva, S.; Liston, A.; Raes, J. How informative is the mouse for human gut microbiota research? Dis. Model. Mech. 2015, 8, 1–16. [Google Scholar] [CrossRef] [Green Version]
  41. Erysipelotrichaceae—An Overview|ScienceDirect Topics. Available online: https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular483biology/erysipelotrichaceae (accessed on 9 June 2021).
  42. Martínez, I.; Perdicaro, D.J.; Brown, A.W.; Hammons, S.; Carden, T.J.; Carr, T.P.; Eskridge, K.M.; Walter, J. Diet-Induced Alterations of Host Cholesterol Metabolism Are Likely To Affect the Gut Microbiota Composition in Hamsters. Appl. Environ. Microbiol. 2013, 79, 516–524. [Google Scholar] [CrossRef] [Green Version]
  43. Wu, J.; Liu, M.; Zhou, M.; Wu, L.; Yang, H.; Huang, L.; Chen, C. Isolation and genomic characterization of five novel strains of Erysipelotrichaceae from commercial pigs. BMC Microbiol. 2021, 21, 125. [Google Scholar] [CrossRef]
  44. Kaakoush, N.O. Insights into the Role of Erysipelotrichaceae in the Human Host. Front. Cell. Infect. Microbiol. 2015, 5, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kong, Y.; Olejar, K.J.; On, S.L.W.; Helikani, V. The potential of Lactobacillus spp. for modulating oxidative stress in the gas-trointestinal tract. Antioxidants 2020, 9, 610. [Google Scholar] [CrossRef] [PubMed]
  46. Shukla, G.; Devi, P.; Sehgal, R. Effect of Lactobacillus casei as a Probiotic on Modulation of Giardiasis. Dig. Dis. Sci. 2008, 53, 2671–2679. [Google Scholar] [CrossRef]
  47. Yichoy, M.; Duarte, T.T.; DE Chatterjee, A.; Mendez, T.L.; Aguilera, K.Y.; Roy, D.; Roychowdhury, S.; Aley, S.B.; DAS, S. Lipid metabolism in Giardia: A post497 genomic perspective. Parasitology 2021, 138, 267–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Riba, A.; Hassani, K.; Walker, A.; van Best, N.; von Zeschwitz, D.; Anslinger, T.; Sillner, N.; Rosenhain, S.; Eibach, D.; Maiga-Ascofaré, O.; et al. Disturbed gut microbiota and bile homeostasis in Giardia-infected mice contributes to metabolic dysregulation and growth impairment. Sci. Transl. Med. 2020, 12, eaay7019. [Google Scholar] [CrossRef] [PubMed]
  49. Lagunas-Rangel, F.A.; Yee, J.; Bermúdez-Cruz, R.M. An update on cell division of Giardia duodenalis trophozoites. Microbiol. Res. 2021, 250, 126807. [Google Scholar] [CrossRef]
  50. Halliday, C.E.W.; Inge, P.M.G.; Farthing, M.J.G. Characterization of bile salt uptake by Giardia lamblia. Int. J. Parasitol. 1995, 25, 1089–1097. [Google Scholar] [CrossRef]
  51. Farthing, M.J.G. Pathogenesis of giardiasis. Trans. R. Soc. Trop. Med. Hyg. 1993, 87, 17–21. [Google Scholar] [CrossRef]
  52. Heilmann, R.M.; Steiner, J.M. Clinical utility of currently available biomarkers in inflammatory enteropathies of dogs. J. Vet. Intern. Med. 2018, 32, 1495–1508. [Google Scholar] [CrossRef]
Figure 1. Box plot of the number of identified OTUs and Alpha-diversity (Chao1 and Shannon indices) by groups of study: GP and GN; GPF, GPM, GNF, and GNM. The box plot synthesize the data, providing the principal measures of central tendency and dispersion. Specifically, the diagram comprises a box with horizontal limits defining the upper and lower quantiles representing the interquartile range, with the median marked by a horizontal line within the box. The whiskers are vertical lines extending from the box as low as the 2.5th percentile and as high as the 97.5th percentile. Extreme values are indicated by dots.
Figure 1. Box plot of the number of identified OTUs and Alpha-diversity (Chao1 and Shannon indices) by groups of study: GP and GN; GPF, GPM, GNF, and GNM. The box plot synthesize the data, providing the principal measures of central tendency and dispersion. Specifically, the diagram comprises a box with horizontal limits defining the upper and lower quantiles representing the interquartile range, with the median marked by a horizontal line within the box. The whiskers are vertical lines extending from the box as low as the 2.5th percentile and as high as the 97.5th percentile. Extreme values are indicated by dots.
Animals 13 00958 g001
Figure 2. Barplots showing the average abundance of bacterial taxa at the order level between (A) GN and GP dogs, (B) GNM and GNF, and (C) GPM and GPF.
Figure 2. Barplots showing the average abundance of bacterial taxa at the order level between (A) GN and GP dogs, (B) GNM and GNF, and (C) GPM and GPF.
Animals 13 00958 g002
Figure 3. Volcano plot showing the OTUs that differ significantly between groups of study: (a) GP and GN, (b) GPF and GNF, and (c) GPM and GNM. The pink points indicate variables of interest that display both large-magnitude fold changes (x-axis) as well as high statistical significance (−log10 of p-value Wilcoxon test, y-axis). The horizontal line shows the p-value cut-off (p-value = 0.10), with points above the line having a p-value < 0.10 and points below the line having a p-value > 0.1. The vertical lines show 2-fold changes.
Figure 3. Volcano plot showing the OTUs that differ significantly between groups of study: (a) GP and GN, (b) GPF and GNF, and (c) GPM and GNM. The pink points indicate variables of interest that display both large-magnitude fold changes (x-axis) as well as high statistical significance (−log10 of p-value Wilcoxon test, y-axis). The horizontal line shows the p-value cut-off (p-value = 0.10), with points above the line having a p-value < 0.10 and points below the line having a p-value > 0.1. The vertical lines show 2-fold changes.
Animals 13 00958 g003
Figure 4. Partial least squares discriminant analysis (PLS-DA) results showing the comparison between OTU data acquired for (a) GP vs. GN, (b) GPF vs. GNF, and (c) GPM vs. GNM. On the left, the 2-D PLS-DA scores plots; on the right, the variable importance in projection plots. The most discriminating OTUs are shown in descending order of their coefficient scores. The color boxes indicate whether OTU is increased (red) or decreased (blue) in positive (1) vs. negative (0).
Figure 4. Partial least squares discriminant analysis (PLS-DA) results showing the comparison between OTU data acquired for (a) GP vs. GN, (b) GPF vs. GNF, and (c) GPM vs. GNM. On the left, the 2-D PLS-DA scores plots; on the right, the variable importance in projection plots. The most discriminating OTUs are shown in descending order of their coefficient scores. The color boxes indicate whether OTU is increased (red) or decreased (blue) in positive (1) vs. negative (0).
Animals 13 00958 g004
Figure 5. Box plot of the significant hematological and biochemical parameters in (a) female and (b) male dogs. TG is expressed in mmol/L, CRP in mg/L.
Figure 5. Box plot of the significant hematological and biochemical parameters in (a) female and (b) male dogs. TG is expressed in mmol/L, CRP in mg/L.
Animals 13 00958 g005
Table 1. Unique OTUs per group.
Table 1. Unique OTUs per group.
GroupsGPGNGPFGPMGNFGNM
N.OTUs211248637546523462640
Table 2. Differential abundance of microbial taxa among groups. In the “Group” column are reported the groups with higher abundance. GP = Giardia-positive; GN = Giardia-Negative; GPF = Giardia-Positive Females; GPM = Giardia-Postive Males; GNF = Giardia-Negative Females; GNM = Giardia-Negative Males.
Table 2. Differential abundance of microbial taxa among groups. In the “Group” column are reported the groups with higher abundance. GP = Giardia-positive; GN = Giardia-Negative; GPF = Giardia-Positive Females; GPM = Giardia-Postive Males; GNF = Giardia-Negative Females; GNM = Giardia-Negative Males.
IDPhylumClassOrderGroup
GN vs. GP
158BacteroidetesBacteroidiaBacteroidalesGP
443BacteroidetesBacteroidiaBacteroidalesGP
GNF vs. GPF
147FirmicutesErysipelotrichiErysipelotrichalesGPF
463BacteroidetesBacteroidiaBacteroidalesGPF
GNM vs. GPM
56FirmicutesClostridiaClostridialesGNM
GNF vs. GNM
45ProteobacteriaBetaproteobacteriaBurkholderialesGNF
52ProteobacteriaBetaproteobacteriaBurkholderialesGNF
56FirmicutesClostridiaClostridialesGNM
GPF vs. GPM
885FirmicutesClostridiaClostridialesGPM
124FirmicutesErysipelotrichiErysipelotrichalesGPM
201FirmicutesClostridiaClostridialesGPM
238FirmicutesClostridiaClostridialesGPM
105FirmicutesBacilliLactobacillalesGPM
613FirmicutesErysipelotrichiErysipelotrichalesGPF
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Peruzzo, A.; Vascellari, M.; Massaro, A.; Mancin, M.; Stefani, A.; Orsini, M.; Danesi, P.; Petrin, S.; Carminato, A.; Santoro, M.M.; et al. Giardia duodenalis Colonization Slightly Affects Gut Microbiota and Hematological Parameters in Clinically Healthy Dogs. Animals 2023, 13, 958. https://doi.org/10.3390/ani13060958

AMA Style

Peruzzo A, Vascellari M, Massaro A, Mancin M, Stefani A, Orsini M, Danesi P, Petrin S, Carminato A, Santoro MM, et al. Giardia duodenalis Colonization Slightly Affects Gut Microbiota and Hematological Parameters in Clinically Healthy Dogs. Animals. 2023; 13(6):958. https://doi.org/10.3390/ani13060958

Chicago/Turabian Style

Peruzzo, Arianna, Marta Vascellari, Andrea Massaro, Marzia Mancin, Annalisa Stefani, Massimiliano Orsini, Patrizia Danesi, Sara Petrin, Antonio Carminato, Michele Matteo Santoro, and et al. 2023. "Giardia duodenalis Colonization Slightly Affects Gut Microbiota and Hematological Parameters in Clinically Healthy Dogs" Animals 13, no. 6: 958. https://doi.org/10.3390/ani13060958

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop