1,740
Views
1
CrossRef citations to date
0
Altmetric
Review

Employing pigs to decipher the host genetic effect on gut microbiome: advantages, challenges, and perspectives

ORCID Icon & ORCID Icon
Article: 2205410 | Received 09 Nov 2022, Accepted 11 Apr 2023, Published online: 30 Apr 2023

ABSTRACT

The gut microbiota is a complex and diverse ecosystem comprised of trillions of microbes and plays an essential role in host’s immunity, metabolism, and even behaviors. Environmental and host factors drive the huge variations in the gut microbiome among individuals. Here, we summarize accumulated evidences about host genetic effect on the gut microbial compositions with emphases on the correlation between host genetic kinship and the similarity of microbial compositions, heritability estimates of microbial taxa, and identification of genomic variants associated with the gut microbiome in pigs as well as in humans. A proportion of bacterial taxa have been reported to be heritable, and numerous variants associated with the diversity of the gut microbiota or specific taxa have been identified in both humans and pigs. LCT and ABO gene have been replicated in multiple studies, and its mechanism have been elucidated clearly. We also discuss the main advantages and challenges using pigs as experimental animals in exploring host genetic effect on the gut microbial composition and provided our insights on the perspectives in this area.

Introduction

Pigs are not only important farm animals that provide a major source of meat for human consumption, but also serve as a biomedical model for the studies of human diseases. Just as that of other mammals, the gastrointestinal tract of pigs inhabits ~1014 of bacterial cells that have been estimated to be 2 to 10 times greater than the number of host cellsCitation1,Citation2. The collective genes of the bacterial population that resides in the pig gut have been estimated to be about 17.2 million in our previous studyCitation3, which is more than 680 times than the number of genes in the pig genome (~25,000 genes). Gut microbiota forms a complex and diverse community of ecosystem and plays an essential role in host physiological processes, such as immunity and diseasesCitation4, metabolismCitation5, and even behaviorsCitation6. Various environmental factors including diets, housing, stress, and the utilization of medicines, and host factors, such as host genetics, sex, and age can influence the diversity, composition, and functional capacities of the gut microbiotaCitation7,Citation8.

In recent years, more and more studies have been involved in the discussion whether and how host genetics affects the compositions of the gut microbiome. Several host genes have been reported to be associated with the compositions or diversity of gut microbial community in humans and pigsCitation9–11. However, the studies have also suggested that environmental factors dominate over host genetics in shaping human gut mcirobiomeCitation8,Citation12. It is difficult to determine how much is the contribution of host genetics in shaping the gut microbiota in humans because environmental factors, such as diets, medicine, and living conditions, that significantly affect the gut microbial compositions are difficult to control for the study cohorts.

Pigs are always raised in uniformed commercial farm houses with high density. Diets, the utilization of medicines, e.g., antibiotics, and farm conditions are artificially provided and easily controlled. More importantly, microbial samples can be obtained from different gut locations that can’t be achieved in humans. These provide an excellent opportunity to study the effect of host genetics on the gut microbiota by using pigs as experimental animals. In this review, we first describe the variations of microbial compositions of pigs in different gut locations and at different ages and then summarize environmental and host factors influencing pig gut microbiome. Especially, we discuss the recent advances in deciphering the effects of host genetics on the gut microbial compositions and envisage the advantages, challenges, and perspectives on using pigs as experimental animals for investigating host genetic effect on the gut microbial composition.

Gut microbiota compositions of pigs compared to that of humans

Gut microbial compositions markedly vary between individuals (inter-individual heterogeneity) and among gut locations and ages (gut location- and age-heterogeneity). Fully understanding the compositions of pig gut microbiome in different intestinal locations and at different ages is important to both pig health managements and production performance improvements by adjusting the gut microbiota. A recent study presented a collection of cultured bacteria from pig gastrointestinal tract and identified distinct taxonomic groups that we did not know before and showed the diversity of metabolic functionsCitation13. This suggested that a more comprehensive and deep understanding of pig gut microbiota are further needed. The dynamic changes of a healthy microbial ecosystem with ages and the spatial distribution of gut microbiota in pig gastrointestinal tract have yet to be qualitatively or quantitatively defined.

A meta-analysis using 20 publically available data sets from 16S rRNA gene sequencing studies of pig gut microbiota indicated that gut locations that were sampled had the largest effect on the composition and structure of the pig gut microbiotaCitation14. Zhang et al. (2018)Citation15 investigated the spatial heterogeneity of mucosal and luminal microbiome across intestinal tract and revealed the distinct differences in microbial compositions among ileum, cecum, and colon and between luminal content and mucosa. From the small intestine to the large intestine, the abundances of both luminal and mucosal Bacteroidota were increased, while luminal Firmicutes and Proteobacteria and mucosal-attached Proteobacteria were remarkably decreasedCitation15. The study on microbial compositions in five gut locations of 13 castrated male Iberian pigs found that Lactobacillus and Clostridium were the two most abundant genera in the small intestine, while Prevotella was predominant in the colonCitation16. In our previous studies, we determined the spatial heterogeneity of gut microbial compositions among five gut locations (ileum and cecum luminal content, ileal and cecal mucosa, and feces) at the age of 240 days in F6 and F7 pigs from a mosaic population by 16S rRNA gene sequencingCitation11. We observed the obvious variations in microbial compositions at different gut locations. For examples, at the family level, Leuconostocaceae were nearly specific to luminal content, while Pseudomonadaceae were specific to mucosaCitation11. Enterobacteriaceae and Clostridiaceae were at least ten times more abundant in the ileum than in the other locations. Christensenellaceae had nearly ten times more abundance in feces than in luminal content and mucosa samples. With the metagenomic sequencing data of six ileum contents, 20 cecum contents and 472 feces samples from healthy pigs, a significant difference was found in the relative abundances of the same bacterial species in different gut locations. There were seven species specifically having high relative abundance (Ranking in the top 20) in ileum contents, while three and six bacterial species particularly showed high abundances in cecum contents and feces samples, respectivelyCitation3. Yang et al. (2016)Citation17 also compared the microbial compositions at different gut locations and identified eight bacterial species that were enriched in the cecum content, five species had the higher abundance in the jejunum and ileum contents, and eight species were successively decreased their abundances from anterior to posterior of intestineCitation17. These reports indicated the variations of the gut microbial compositions in different gut locations () and suggested that limiting the analysis of the gut microbiota to feces are far from enough to understand its complexity and elucidate the factors that determine itCitation7.

Figure 1. Microbial compositions in different gut locations and the factors influencing gut microbial compositions in pigs. (a) the main bacterial species residing in the ileum, cecum, and feces. The data from Chen et al. (2021). (b) host and environmental factors influencing pig gut microbiota.

Figure 1. Microbial compositions in different gut locations and the factors influencing gut microbial compositions in pigs. (a) the main bacterial species residing in the ileum, cecum, and feces. The data from Chen et al. (2021). (b) host and environmental factors influencing pig gut microbiota.

Distinct gut microbial compositions at different ages/growth stages have been reported in many studiesCitation3,Citation11,Citation18. A meta-analysis with the 16S rRNA gene sequencing data from 16 studies observed dynamic changes in gut microbial compositions following the ages. The top three genera in relative abundances were Bacteroides, Escherichia, and Lactobacillus in lactating pigs; Prevotella, Lactobacillus, and Faecalibacterium in growing pigs; and Prevotella, Lactobacillus, and Streptococcus in finishing pigsCitation18. We previously also determined the gut microbial compositions at the ages of 25, 120, and 240 days in F6 and F7 pigs of a mosaic population by 16S rRNA gene sequencingCitation11,Citation19. The richness of gut microbiota was significantly increased from 25 to 240 days of age, and the topological features of phylogenetic co-occurrence networks were increased along with the agesCitation19. Odoribacteraceae and Rikenellaceae were at least ten times more abundant in feces at the age of 25 days than at any other agesCitation11. Wang et al. (2019) investigated the pig gut microbial composition and structure at 16 time points from birth to market with 18 pigs during lactation, nursery, growing, and finishing stages, identified 19 phyla that existed in pig gut microbiome of the lifetime with Firmicutes and Bacteroidota being the most abundant, and observed an overall increased alpha-diversity of pig gut microbial community and distinct shifts in microbiome structure along different growth stagesCitation20.

Because of the high similarity of omnivorous and gastrointestinal structure between human and pig, pigs have been treated as an important animal model for studying gut microbiome. A previous report identified that 78% of the KO functional pathways identified in the pig gut metagenome are present in the human gut metagenome, while 96% of the KOs found in the human gut metagenome are present in the pig gut metagenomeCitation21. We previously compared the gut microbial compositions between humans and pigs with the gene catalogs of human and pig gut microbiomeCitation3,Citation22 and found that the percentage of common bacterial genera shared between human and pig (87.24% for human gut microbiota) was higher than that shared between human and mouse (70.00% for human gut microbiota). The similar result was also observed in F6 and F7 pigs of the mosaic population by 16S rRNA gene sequencing, in which microbiota composition of pig feces was more similar to that of human than of mice fecesCitation11. Notably, the common bacterial genera should show different abundances in human and pig gut microbiome. For example, at the genus level, Bacteroides and Eubacterium were most abundant in human feces. However, the most abundant genera in pig feces were Lactobacillus and Clostridium3.

Effects of non-genetic factors on pig gut microbial compositions

As a complex ecosystem, immense diversity of the gut microbial composition in intra- and inter-individuals have been reported. At some extreme states, natural variations in the gut microbiota can deteriorate to dysbiosis when stress conditions rapidly decrease the diversity of the gut microbiota and promote the expansion of specific bacterial taxaCitation23,Citation24. Besides host ages and gut locations, the non-genetic factors influencing gut microbial compositions mainly include the diets, the use of antibiotics and probiotics, health state, housing environments, and host gender.

Diets

Diets have profound effects on the composition of the gut microbiota and are one of the most important factors that change the gut microbiota. Most of the studies about diet effect on the gut microbiota have been performed in humans and mice, especially about high-fiber or high-fat dietary patterns on the gut microbiotaCitation25–27. In pigs, the study using a reciprocal cross-fostering model revealed that the introduction of solid feed and weaning are more important determinants of gut bacterial succession in piglets than breed and nursing motherCitation28. More studies have focused on the effects of specific dietary nutrients on the composition of gut microbiota, such as proteinCitation29, fibersCitation30,Citation31, vitaminsCitation32,Citation33, sodium butyrateCitation33, and some specific nutrient factors, e.g., soy isoflavoneCitation32 and methylsulfonylmethaneCitation34. For example, low crude protein diet increased interaction network complexity, although it did not alter the microbial diversityCitation35. Generally, production performances had always been phenotyped when investigating the effects of dietary nutrients on the composition of gut microbiota because these studies were mainly designed to investigate the improvement of pig production performances by the additive of specific nutrient factors through regulating gut microbiome.

Utilization of antibiotics and probiotics

Over the past few decades, antibiotics had been administered to pigs broadly in the pig industry for disease treatment and growth promotion. A study investigated the effect of performance-enhancing antibiotics on the gut microbiome and found that the use of antibiotics increased the abundance of Escherichia coli in the ileum and Lachnobacterium spp. in all gut locations of the medicated pigsCitation36,Citation37. Metagenomic analysis indicated the increased abundances of microbial functional genes relating to energy production and conversion and the increased diversity and abundances of antibiotic resistance genes in the antibiotic-fed pigsCitation36. Another feeding experiment of antibiotics cocktail also found significant alteration of the microbial compositions in the ileum and feces with a reduced abundances of Bifidobacterium, Lactobacillus, and Ruminococcus, and an increased abundance of Escherichia coli Citation38.

In recent years, considering the residual effect and antibiotic resistance in bacteria in the use of antibiotics, as an alternative, probiotics has been getting increased attention in pig productionCitation39. Probiotics show a strong ability to modulate the gut microbiota composition in healthy individuals by significantly reducing disease-related bacterial taxaCitation40. Among probiotics used in pig production, Lactobacillus has been the most commonly used probiotics, improving growth performance and feed conversion efficiencyCitation39 and regulating immune system in pigs, such as beneficial effects of Lactobacillus plantarum on the modulation of gut microbiota and immune response in weaned pigsCitation41.

Health state

Host health state such as environmental stressors, diseases, and infections also affects the gut microbial composition. For example, the diversity, composition, and functional capacity of fecal microbial community were different in humans or animal models with Crohn’s diseaseCitation42,Citation43, ulcerative colitisCitation44, and acute diarrheaCitation45. The similar conditions were also observed in pigs. Gut microbiota dysbiosis was associated with post-weaning diarrhea in pigsCitation46. Porcine deltacoronavirus (PDCoV) infection reduced the diversity and richness of gut bacteria and significantly changed the structure and abundance of the gut microbiota from the phylum to genusCitation47. Heat stressed pigs showed the decrease in Lactobacillus johnsonii and Lactobacillus reuteri and the increase in Clostridium sensu stricto 1 in the colon microbiotaCitation48.

Housing environments

Environmental factors, such as farm conditions and hygiene, also have significant effects on the gut microbial compositions. In our previous study, we compared the gut microbiome between free-living wild boars and Duroc pigs from two farms. Distinct gut microbial compositions were observed between wild boars and two Duroc pig populationsCitation3. Although genetic differences existed between wild boars and Duroc pigs, distinct gut microbial compositions were also found between two Duroc pig populations fed with the similar formula diet from different farms. This suggested that many other factors including environmental exposures, hygiene, temperature, and certain other unknown factors potentially affected the gut microbiota. The study by Wen et al. (2021) also indicated that environmentally enriched housing conditions affected pig gut microbiota in early life, and the relative abundance of several bacterial species involved in the production of short chain fatty acids was increasedCitation49.

Host gender

An increasing number of studies have indicated profound effect of host gender on the gut microbiome. For examples, compared to normal male mice, sexually mature castrated male mice showed a similar gut microbial composition to female miceCitation50,Citation51. Our previous study indicated an obvious influence of host gender on the gut microbial composition. Veillonellaceae, Roseburia, Bulleidia, and Escherichia had higher abundance in boars, while Treponema and Bacteroides were over-represented in giltsCitation52. Castration significantly changed the fecal microbiota composition of the boars toward that of giltsCitation52. A bi-directional interaction exists between gut microbiota and sex hormones. Host sex hormones, e.g., estrogens, can influence gut bacterial gene expression and compositionCitation53, whereas the gut microbiota can reactivate estrogens from their inactive glucuronidesCitation54.

Host genetic effect on the gut microbiome

Besides the non-genetic factors mentioned above, to what extent host genetics contributes to the inter-individual variations in the gut microbial compositions attracts great interest to researchers working on gut microbiome. In recent years, more and more studies have focused on this subject in both humans and animals, especially in humans. However, compared to environmental factors, the extent of host genetics shaping the composition of gut microbiome has not been consensus. The studies have demonstrated that environmental factors dominate over host genetics in shaping human gut microbiota in both cohorts of 1,046 healthy individuals with several distinct ancestral originsCitation8 and 8,208 Dutch individualsCitation12. The overall microbiome heritability was only 1.9% ~ 8.1%Citation8 or only about 6.6% of taxa are heritableCitation12. On the other hand, since 2014, more than dozen of the studies have investigated the associations of host genetic variants on the gut microbiome in humans (), DrosophilaCitation55,Citation56, and miceCitation57 and reported numerous genomic loci/variants associated with the diversity of gut microbiota and the abundance of individual taxa. More recently, the study using 36 healthy black and white females with similar age, weight, habitual diets, and health status indicated that individuality and ethnicity account for roughly 70% to 88% and 2% to 10% of taxonomic variation, respectively, which dominated the effects of a short-term diet intervention in shaping gut and oral microbiomes and gut viromesCitation58. However, the urine and plasma metabolites showed the tendency to more strongly associate with dietsCitation58. Here, we summarize the existing evidences about host genetic effects on the gut microbiome in pigs with emphases on the correlations between host genetic kinship and the dissimilarity of gut microbial compositions, heritability estimates of specific taxa, and identification of genomic variants, or microbial quantitative locus (miQTL) associated with the gut microbial compositions by genome-wide association studies (GWAS). Because the higher similarity of gut microbial compositions between pigs and humans than between humans and miceCitation11,Citation21, this has made pigs as an important model to study the host genetic effect on the gut microbiome. To facilitate the comparison and discuss the progress in this subject between humans and pigs, the advances in human genetic effect on gut microbiome investigated by GWAS are also discussed in detailed as a benchmark because more promising advances have been made in the studies of humans.

Table 1. Summary of identification of host genetic variants associated with gut microbiome by genome-wide association study.

Correlation between host genetic kinship and the dissimilarity of gut microbial compositions

There have still been controversies about the correlation between host genetic kinship and the dissimilarity of gut microbial compositions. The study in humans found that memberships with the closer degree of relatedness in families showed a higher overall similarity in gut microbiomeCitation72. Microbiotas were more similar within twin pairs compared to unrelated individuals, and monozygotic twin pairs had slightly more similar microbial compositions compared to dizygotic twin pairs in the Twins UK populationCitation73. The variation of a given taxon abundance was more frequently explained by host genetic similarity, while the presence of a given taxon more depended on a shared home environmentCitation71. However, Rothschild et al. (2018) found no significant association between genetic kinship and microbiome compositionCitation8. This controversial result should be caused by the fact that environments dominate over host genetics in shaping human gut microbiotaCitation8. Compared to humans, livestock and experimental animals are more easily controlled to a uniform condition by raising in the same farm house and being provided the same diets. This could eliminate part of the influence of environmental factors on gut microbiome and highlight the effect of host genetics. For examples, in cattle, sire breed composition primarily explains the differences in gut microbiota structure among breed groups of preweaning calves that were maintained in the same pasture and accessed to an identical dietCitation64. In pigs, the diversity of gut microbiota was compared among full-sibs, half-sibs, and unrelated pigs in two pig populations of Erhualian and Bamaxiang and found that full-sibs and half-sibs had a higher similarity of microbial compositions than unrelated pigs in both Weighted and Unweighted Unifrac analysis in two populations raised in the same farmCitation57. The investigation on the relationship between genetic relatedness based on genome-wide SNP identity-by-state and gut microbiota dissimilarity based on Bray-Curtis distance found significantly negative correlations between genetic similarity and microbiota dissimilarity in the F6 and F7 generations of a mosaic populationCitation11. The analysis was restricted to full-sib littermates raised in the same environment, which avoided the interferences of environmental and maternal factors to the resultsCitation11. In mice, bacterial communities retained strain specificity in eight progenitor mouse strains of the collaborative cross, although it became more similar after cohabitation of different mouse strains, indicating the interaction of host genetics and environmental factors in shaping gut microbiotaCitation62. Notably, compared to mice, pigs are more suitable as a model for the studies on the role of host genetics in the gut microbiome because pigs showed a higher similarity of the gut microbial composition to humans than miceCitation11. The pairwise overlap of genes in the gut microbiome was significantly higher for pigs and humans (9.49%) than for mice and humans (1.50%). More similar gut microbiome and easily controlled environments have made pigs as an important model to investigate the relationship between host genetic kinship and the similarity of gut microbial compositions.

Heritability estimates

Heritability has been generally used to predict genetic influence on phenotype and always shows the proportion of phenotypic variation in a trait (such as body weight) that is measured across a population and can be explained by genetics rather than environmental effects. In the studies of gut microbiome, the degree to which host genotype versus environment influences the gut microbiome has become increasingly more interesting. Quantifying the heritability of the gut microbiome is to discern genetics from environmental factors that structure the microbiome and to potentially identify members of the gut microbial community influenced by host genotypes. There are two conventional methods to quantify heritability of microbial taxa. One method is twin-based studies, which estimates heritability (h2) of microbial taxa by comparing the variation in microbial taxon abundances owing to host genetics. The other method is standard statistical approaches, such as additive genetic model and unique environment (ACE) model. In both methods, the abundance of each gut microbe is treated as a continuously varying quantitative traitCitation61. Heritability predicts the host genetic effect on the abundance of a gut microbe. This heritability estimate has been performed more in the studies of human gut microbiome. For instance, Goodrich et al. (2014)Citation73 firstly identified heritable gut microbial taxa in the TwinsUK population and reported that 5.3% of the taxa showed a heritability estimate greater than 0.2 in stool samples of 416 pairs of dizygotic and monozygotic twins. This percentage was increased to 8.8% in their further study with larger sample size of 1,126 twin pairsCitation74. The most heritable taxon was the family Christensenellaceae, which was confirmed in different cohortsCitation55,Citation74,Citation75. Further study in Canadian Genetic Environmental Microbial Project cohort identified that more than half of the heritable taxa could be compared between TwinsUK population and Canadian Genetic Environmental Microbial Project cohortCitation55. Davenport et al. (2015)Citation76 identified 15 heritable taxa in stools from different sampling seasons in Hutterites. Most of these heritable bacteria belong to the phyla Proteobacteria or Firmicutes. In mice, the study in HMDP strain mice revealed that host genetics explained a substantial amount of the variation (up to 0.5 or more) for many common taxa when experimental mice were maintained under controlled conditionsCitation70. O’Connor et al. (2014)Citation77 observed the influence of host genetic background in both microbial community structure and individual taxa and identified a wide range of heritability for taxa at the genus level (0.26–0.86) in a collaborative cross. In pigs, Camarinha-Silva et al. (2017)Citation78 estimated the heritability of gut bacterial genera using 207 Pietrain pigs as experimental animals and identified 8 out of 49 bacterial genera with heritability ranging from 0.32 to 0.57. Unlike the twins’ study, pig populations are composed of complex pedigree. Visscher et al. (2008)Citation79 suggested that for samples from the cohorts with complex pedigree relationships, liner mixed model is more suitable for the heritability estimate. In our previous study, with a liner mixed model, we identified 81 taxa having h2>0.15 in fecal samples of Bamaxiang pigs and 67 taxa in cecum luminal samples of Erhualian pigs. Among them, 31 taxa had h2>0.15 in both types of samplesCitation57. In the F6 and F7 generations of a Mosaic population, we estimated the heritability of the abundances of individual taxa/OTUs and found that the total heritabilities of gut microbiota were ≤11.8%Citation11. The total heritability for fecal samples was higher than that for luminal content samples (10.2% vs. 5.7%)Citation11. Based on congruent estimates in F6, F7, and across F6 and F7, a total of 55 most likely heritable taxa/OTUs were identified, including OTUs belonging to Christensenellaceae and Blautia that were concordance with the identifications obtained in human studiesCitation11. Interestingly, some bacterial taxa, such as ChristensenellaceaeCitation11,Citation63,Citation73, LachnospiraceaeCitation57,Citation70,Citation76, RikenellaceaeCitation57,Citation70,OscillospiraCitation70,Citation73,Citation80, RuminocaceaeCitation11,Citation63,Citation70,Citation74, and BlautiaCitation11,Citation57,Citation74 were identified to be heritable in multiple host species in different studies.

It should be noted that conventional heritability measurements tend to consider the interaction between host and gut microbiome as unidirection, in which the host regulates microbe colonizationCitation61. However, more comprehensive views have thought that both host and microbiome play a role in heritability. A community heritability (H 2C) which treats the host as part of an ecosystem and measures the extent to which variation in “whole-community” phenotype is attributable to the host genetic variation has been recommended to predict host genetic influence on the gut microbiomeCitation60.

Identification of miQTL influencing gut microbial composition by GWAS

Identifying host genomic loci (genetic variants) or key genes associated with the gut microbiome directly by applying GWAS (most popular method to date) or quantitative trait locus (QTL) mapping using quantitative measures or presence/absence of individual taxa and diversity indices and functional pathways of the gut microbial community as the complex traitsCitation55,Citation58,Citation67,Citation70,Citation74,Citation76,Citation77 is another method evaluating the effect of host genetics on the gut microbiome. As a mainstream approach, GWASs have been performed broadly to evaluate the effect of host genetics on the gut microbiome in different host species, such as humans ()Citation9,Citation56,Citation74,Citation81,Citation82, pigsCitation11,Citation57,Citation65, cattleCitation63,Citation80, and poultryCitation83. Here, we focused on the advances in the studies of pig gut microbiome. As the benchmarks, the studies about microbial GWASs on human gut microbiome are also summarized in details because more promising advances have been made in human studies with large sample sizes from several thousands to tens of thousands of feces samples. Blekhman et al. (2015)Citation58 first performed the microbial GWAS in 93 individuals from the Human Microbiome Project and reported the association between the abundance of Bifidobacterium in feces and LCT locus. Davenport et al. (2015) performed a GWAS in humans to identify genomic loci associated with gut bacterial taxa in 127 samples collected from different seasons with 16S rRNA gene sequencing and identified 37 associations at genome-wide Q value <0.2Citation76. Since then, more than dozen studies have investigated host genetic effects on human gut microbiome (). More recently, a fecal microbial GWAS in 4,117 individuals from a single multiethnic cohort identified ethnicity-specific associations between host genomic variants and gut microbiotaCitation84. The study in human gut microbiome with the largest sample size so far (stool samples from 18,340 individuals in 24 cohorts) identified 31 genomic loci associated with the gut microbiome at the threshold of 5 × 10−8, and this study replicated the finding of LCT genes influencing the abundance of Bifidobacterium79. However, the studies in humans described above have relied on stool samples. Fecal microbiota is mostly from the colon and luminal microbesCitation85. The microbiota farther up the gastrointestinal tract may be rarely detectable in the stool.

Less studies about host genetic effect on the gut microbiome by GWAS have been reported in pigs, and the sample sizes used in each study were also significantly smaller (). To our knowledge, until now, only five studies have reported the GWAS for identifying genomic loci for pig gut microbial composition. Yang et al. used pigs from the F6 and F7 generations of a mosaic population that was constructed with four Western pig breeds and four Chinese indigenous pig breeds as experimental animals. Whole-genome resequencing of 1,430 pigs from two generations identified more than 30 million host genomic variants (i.e., one variation per 100bp on the genome). A total of 5,110 samples collected at different ages and from different gut locations, including feces at the ages of 25, 120, and 240 d, cecal and ileal contents at days 240, and cecal and ileal mucosal scrapings at days 240 (only for F7 pigs) from these experimental pigs were performed high throughput sequencing of the V3-V4 region of 16S rRNA gene. Based on the F6 and F7 experimental pig populations with high genetic variations and gut microbiota data, genome-wide association analysis was carried out on 8,490 bacterial taxa. A total of 1,527 miQTL that significantly affected the abundance or absence/presence of 846 bacterial taxa were identified at the threshold of 5 × 10−8. Among 1,527 genome-wide significant signals, 6 signals exceeded the experiment-wide significant threshold (P < 1.5 × 10−12)11. Another study from our laboratory collected 256 cecum content samples from Erhualian pigs and 244 stool samples from Bamaxiang pigs and acquired the 16S rRNA gene sequencing data from these samples. All these 500 pigs were genotyped with 1.4 million genome-wide SNPs. GWAS for 1,411 OTUs and 153 taxa in fecal samples and 1,177 OTUs and 168 taxa in cecum contents identified 40 and 34 significant associations, respectivelyCitation57. In both studies described above, core taxa (existed in >95% experimental pigs) were treated as quantitative traits, and taxa identified in 20–95% pigs were considered both quantitative and binary features. Another representative study used 1,205–1,283 feces samples per time point from Duroc × Yorkshire × Landrace or Duroc × Landrace × Yorkshire, performed the GWAS for the α-diversity indices and 334 OTUs associated with growth and fatness, and identified a total of 207 significant associations between host genetic variants and microbial taxaCitation65.

Candidate or causative genes influencing the gut microbial composition

About the genes influencing gut microbiome, many GWASs have discussed candidate genes isolated from the defined intervals around significant signals (variants). For examples, Blekhman et al. (2015)Citation58 first observed the significant correlation between SNPs in the LCT gene and the abundance of Bifidobacterium. Goodrich et al. (2016)Citation74 replicated the association between LCT and Bifidobacterium and identify another association between ALDH1L1 and SHA-98. Wang et al. (2016)Citation10 identified multiple genetic loci, including vitamin D receptor (VDR) gene associated with overall microbial variation and the abundance of individual taxa. Turpin et al. (2016)Citation55 identified the SNPs nearest to UBR3 associated with Rikenellaceae, the SNPs in CNTN6 associated with Faecalibacterium, the variants in DMRTB1 associated with Lachnospira, and the SNPs in SALL3 associated with Eubacterium. The study reported by Qin et al. (2022) not only replicated the association between the variants at the LCT locus and the abundance of Bifidobacterium, but also identified the correlations between the abundance of Faecalicatena lactaris and ABO gene and between the abundance of Enterococcus faecalis and the variants in the MED13L locus. Functional classifications have always been performed to annotate the functional categories of these isolated candidate genes near to strong signals from GWAS and suggested that these candidate genes are mainly associated with metabolism, gut homeostasis, immunity functions and response, signal transduction, and so on. In pigs, our previous study identified six signals exceeding the experiment-wide significant threshold (P < 1.5×10−12) in one cohort and the experiment-wide replication threshold(P < 0.007) in the other cohort. These six signals mapped within 3,037 bp from each other in the 272.8–273.1Mb interval on chromosome 1 and significantly affect the abundance or presence/absence of the OTUs from p-75-a5 or Erysipelotrichaceae as well as genus p-75-a5. Fine-mapping identified a 2.3-kb deletion in ABO gene is the causative mutation for this miQTLCitation11. Another study based on gene co-association network analysis in a Duroc pig population identified five regulator genes and dozen target genes regulating gut microbial communitiesCitation86.

Unfortunately, until now, the genes or genomic loci that could be replicated in multiple studies are few in both humans and pigs. To the best of our knowledge, only LCT and ABO locus have been found to be associated with gut microbes in different cohorts. Among them, LCT locus and its association with the abundance of Bifidobacterium has been most commonly replicated in multiple studies of human gut microbiomeCitation8,Citation9,Citation56,Citation58,Citation67,Citation74,Citation82. As for ABO gene, at least three studies with the large scale of sample sizes reported the significant associations of ABO gene with human or pig gut microbiomeCitation9,Citation82. But different from LCT locus, ABO gene has been reported to be correlated with different bacterial taxa in different studies. In 7,738 participants of the Dutch Microbiome Project, ABO locus was associated with Bifidobacterium bifidum, Collinsella aerofaciens, Collinsella, Coriobacteriaceae, and Coriobacteriales depending on the secretor status by host FUT2 genotypeCitation9. In the participants of the FR02 study, ABO gene was associated with Faecalicatena lactaris, which was also largely driven by secretor statusCitation82. In pigs, ABO gene was associated with the OTUs belonging to p-75-a5 or Erysipelotrichaceae, as well as the genus p-75-a5. However, no evidence showed that the associations in pigs were independent on the secretor statusCitation11.

Elucidating how host genes influence the gut microbial composition is important to regulate gut microbiome for host health or pig production performance. However, to our knowledge, until now, only the mechanisms of LCT and ABO genes affecting the abundance of gut bacterial taxa have been elucidated. LCT encodes lactase that hydrolyzes lactose, the main sugar in mammalian milk, to produce D-glucose and D-galactose. Mutations in the translated region of the LCT gene cause lactase deficiency that is a severe gastrointestinal disease characterized by watery diarrhea in infants fed with breast milk or other lactose-containing formulasCitation87. Dairy intake does not affect the abundance of Bifidobacterium in individuals producing lactase through adulthood. However, in lactase deficiency or intolerant individuals, dairy diet significantly increases the gut Bifidobacterium abundance, and heterozygote individuals had an intermediate increase in Bifidobacterium abundanceCitation67,Citation82. CAZyme profile analysis further revealed that, compared to Bifidobacterium spp. that are affected by LCT gene, Bifidobacterium dentium that is not affected by LCT harbored more genes encoding CAZyme families with preferred fiber/plant-related substrates, whereas Bifidobacterium spp. affected by LCT gene harbor more milk oligosaccharide-targeting CAZyme families (GH129 and GH112)Citation82.

ABO gene encodes the alpha 1–3-N-acetylgalactosaminyltransferase which adds N-acetylgalactosamine to glucoprotein of intestinal mucosa and blood cell. In pigs, the blood group A gene encodes A transferase, which transfers GalNAc to the galactose residue of the acceptor H substrates, including the heavily glycosylated mucins which can be used as carbon source by intestinal bacteriaCitation88, while the group O gene is nonfunctional due to a 2.3-kb deletion encompassing part of intron 7 and the whole exon 8. And H substrates in group O individuals remain without additional modificationsCitation89–91. Different mechanism of ABO gene affecting the abundance of gut bacterial taxa has been reported between humans and pigs. In humans, the effect of ABO gene on the gut microbes depends on secretor status (FUT2 genotypes)Citation9,Citation82, but was not associated with fiber intakeCitation82. In pigs, our study did not identify any evidences for an effect of FUT2 variants on the abundance of the bacterial taxa affected by ABO geneCitation11. By integrating multi-omics data of metabolome, cultureomics, metagenome, and transcriptome, we systematically illustrated how the ABO genotypes affect the abundance of Erysipelotrichaceae species:

The 2.3-kb deletion of the ABO gene (in type O individuals) results in the absence of N-acetylgalactoamine transferase activity of the encoded protein, and therefore, the inability to add N-acetylgalactoamine (GalNAc) to the highly glycosylated mucins in intestinal mucus. This led to the reduced intestinal concentrations of GalNAc in individuals with OO genotype, and thereby, reduced the growth of some Erysipelotrichaceae species used GalNAc as carbon source (). Furthermore, functional GalNAc import and catabolic pathway analyses found that, compared to bacterial taxa that were not affected by ABO locus, the bacterial taxa affected by ABO gene (Erysipelotrichaceae species) had more complete GalNAc import and catabolic pathways, and the genes involved in GalNAc import and catabolism tended to cluster together to form coregulated gene operons in these Erysipelotrichaceae speciesCitation11.

Figure 2. The mechanism of a 2.3-kb deletion in ABO gene affecting erysipelotrichaceae species.

Figure 2. The mechanism of a 2.3-kb deletion in ABO gene affecting erysipelotrichaceae species.

Gut microbiota conversely shapes the host epigenetics through its components and derived metabolites

The gut microbiota can regulate gene expressions of both local intestinal cells and peripheral tissues through its components and derived metabolites under a potent mechanism of epigenetic regulationCitation92. Microbial biosynthesis or metabolism influences the availability of chemical donors for DNA or histone modifications. DNA/histone methyltransferases and histone acetyltransferases generally depend on methyl and acetyl donors, respectively, for their catalytic activity. As an additional source, the gut microbiota can synthesize biological compounds that serve as epigenetic substrates, cofactors or regulators of epigenetic enzyme activityCitation93. For instance, the gut microbiota, such as Bifidobacterium and Lactobacillus species, generates folate and other B vitamins (B2, B12) that donate methyl groups for DNA/histone methylationCitation94,Citation95. The gut microbiota also regulates epigenetic modifying enzyme expression and/or activity. For example, short-chain fatty acids (SCFAs) that are exclusively produced by commensal microbes through fermentation of complex non-digestible carbohydrates and represent an important kind of epigenetically relevant molecules can inhibit deacetylase activity of histone deacetylases (HDACs) that is generally associated with increased expression of target genesCitation96. The researchers are also aware of the importance of the gut microbiome on the regulation of host epigenetics in pigs. The study in piglets found that early microbial colonization affects DNA methylation of genes related to intestinal immunity and metabolism in preterm pigsCitation97.

Advantages and challenges of employing pigs to decipher the effect of host genetics on gut microbiota composition

Advantages

Intensive farming has been popularly used in the pig industry, which reduces the number of pig farms and expands the scale of farming. It has also achieved the industrialization of pig farming through transforming production methods from quantity expansion to quality-benefit. The efficiency of pig production has been greatly improved. The indoor intensive housing for pig farming always keeps pigs of similar ages in groups in common environments. The similar diets (formula feeds in general) are supplied in place for pig populations, which mix a variety of different feed ingredients according to a certain proportion, and meet the energy and nutrition requirements for each growth stage of pigs. Ventilation systems have always been used to regulate moisture and temperature of farm house. This kind of rearing model would benefit for the study investigating the host genetic effect on the gut microbiome in pigs because most environmental factors that also influence the gut microbial compositions are shared in common and can be controlled easily by man-made factors. More importantly, the formula feeds provided to all pigs are similar and meet the energy and nutrition requirements according to pig nutrition requirement criterion. This significantly reduces the diet effects on the gut microbiome. Furthermore, the healthy status of pigs and the use of medicines (e.g., antibiotics and probiotics) can be registered easily by workers. All these proceedings significantly reduce the variates influencing the gut microbial compositions. As an example, in our previous studyCitation11, all F6 and F7 pigs used in the experiments were reared under standard and uniform housing and feeding conditions and fed twice per day with formula diets containing 16% crude protein and 3,100 kJ digestible energy.

Another advantage using pigs as experimental animals is that the pedigrees of experimental pig cohorts can be recorded clearly for all pigs, even in multiple generations. Pedigree information is important to evaluate the host genetic effect on the gut microbiome, especially in the heritability estimation. Moreover, pigs consisting of large pedigrees with existing rich phenotypes should be important resource populations for high-throughput sequencing in the search of the causative variants influencing complex traitsCitation59. And the large pedigree design is also particularly useful in the presence of locus heterogeneity among familiesCitation66.

In addition, litter size of sows is large, which facilitates to adjust the maternal effect on the gut microbial compositions in the studies evaluating the genetic effect on the gut microbiome. At birth or during the nursing, vertical transmission of maternal microbiota to offspring supports postnatal growth and developmentCitation98. The nursing mother influences the gut microbial composition of offspring during early development via breastfeedingCitation99, which plays important roles for health of the animals at later stages of lifeCitation100. Sows also can significantly affect the gut microbiome of piglets as observed in other mammals. As a multiparous mammal, more than ten offspring at one birth of a given sow provide excellent materials for evaluating host genetic effect on the gut microbiome, e.g., heritability estimates. It can circumvent the challenge to distinguish between microbes that have been transmitted vertically (maternal effects) or selected genetically because the rates of microbial vertical transmission are not expected to differ among piglets in a farrow and the environments are identical, differences in microbial similarity are attributed to genetic factors alone.

To our knowledge, all existing studies in humans evaluating host genetic effect on the gut microbiome or identifying genetic variants associated with gut microbial taxa by GWAS were carried out with feces samples and also in most of the studies of pigs (). As mentioned above, distinct gut microbial compositions in different gut locations have been reported in many studiesCitation15–17, and limiting the analysis of the gut microbiota to feces are far from enough to understand its complexity and elucidate the factors that determine itCitation7. The conditions are similar to the gut microbial compositions at different agesCitation18,Citation19, and the samples collected at different ages were needed to systematically and deeply evaluate host genetic effect on the dynamic changes of gut microbes. The heterogeneity of miQTL effect should exist for the samples from different gut locations and ages. For example, the miQTL identified at the ABO locus affects Erysipelotrichaceae species in cecum content and mucosa samples in day 120 and 240 feces, but not in day 25 feces and in ileum contents at day 240Citation11. As an advantage using pigs as experimental animals, microbiota samples can be obtained from the same population at different ages, and lumen content and mucosa samples also can be obtained from different gut locations when pigs are slaughtered.

Elucidating the mechanisms of host variants affecting microbial taxa is important for the regulation of gut microbial compositions. To our knowledge, except LCT gene in humans and a 2.3-kb deletion of ABO gene in pigs, no other host variants have been elucidated its mechanism affecting the gut microbes. Multi-omics method that generally includes metagenome of the gut microbiome, metabolome of feces, intestinal contents, and other target tissues, transcriptome of microbes, intestinal tissues and other target tissues, and culturomics are useful to elucidate the mechanism of causative mutations affecting the gut microbes. Using pig as experimental animals, many different tissue types, e.g., intestinal biopsy, muscle, blood, and so on can be harvested from the same individuals that would facilitate the elucidations of mechanism of host genes in affecting the gut microbial compositions.

Main challenges

Compared to that in humans, less GWASs have been performed to identify host genetic variants associated with the diversity of gut microbiota and the abundance of specific taxa in pigs. Several studies have identified some significant associations in various populationsCitation11,Citation65,Citation86 (). However, similar to the conditions observed in humans, the replication of significant associations is low between studies, although the standard and uniform housing and feeding conditions significantly reduce the variates influencing the gut microbial compositions compared with that in humans, which should benefit the identification of genomic loci associated with gut microbial taxa in a study. The significant differences in farming conditions, managements, and workers provided to experimental pigs have still existed across the studies from different laboratories and groups, which led to the heterogeneity of microbial compositions and high interindividual variability across pig populations. Furthermore, to avoid the effect of maternal and early colonization on the gut microbial compositions, it should be best that experimental pigs should be from the same farm. However, huge expense and manpower are required when large sample size is used, especially, when microbial samples are collected from different gut locations, experimental pigs would need to be slaughtered for sample collection.

Another challenge, we think, resulting in the low replication between studies is the bias introduced in quantifying the abundances of bacterial taxa in samples by 16S rRNA gene sequencing (and also by shotgun metagenomic sequencing,Citation101 which has been met commonly in earlier studies of microbial community structure, including sample processing pipeline, DNA extraction (kit and protocol), PCR amplification (different efficiency of amplifications for bacterial taxa)Citation102, the choice of which hypervariable region(s) of 16S rRNA gene to sequenceCitation14, sequencing technology, and data processingCitation103,Citation104. As an example of the effect of technique bias on microbial GWAS, Qin et al. (2022) reported that the association of LCT locus with Bifidobacterium in a single cohort of 5,959 individuals was stronger than that in an integrative data of 18,473 individuals from 28 different cohorts, suggesting the importance of standardized methodologyCitation56,Citation82.

Perspectives

As we have discussed above, there are only a few studies about host genetic effect on the gut microbiome in pigs, and a small number of genomic variants associated with pig gut microbes have been reported. Compared to the current studies in humansCitation9,Citation56,Citation82, the sample sizes used in the studies of pigs are small (). As indicated by Lopera-Maya et al.Citation9 (2022), more than 50,000 individuals are required to identify an effect size similar to that of LCT and ABO for bacterial taxa present in >20% of the samples in humans. Therefore, in the future studies, we should take advantage of easily obtaining large sample sizes in pigs even from one farm under uniformed conditions. Furthermore, the protocols of experiments, such as DNA extraction and sequencing, and bioinformatic analyses to obtain the gut microbial composition data should be standard to facilitate to combine the datasets from different studies (meta-analysis of GWAS). In addition, following the decrease of sequencing cost, instead of 16S rRNA gene sequencing, metagenomic sequencing data has begun to be used in the GWASs of gut microbiome just like two current studies in humansCitation9,Citation82 and the previous study by Bonder et al. (2016)Citation67.

Obtaining the associations between genetic variants and microbial taxa is far from enough. To those genomic loci that are significantly associated with gut microbes with large effect size (e.g., achieving study-wide significance level), further fine-mapping should be performed to identify causative genes and mutations, and the mechanisms will also need to be elucidated using multi-omics data. Based on this, we can further investigate the effect of the interaction between host gene and gut microbiota on complex traits or pig production performances. Another method evaluating and verifying the effect of host gene or genomic locus on the gut microbiome is to inactivate the gene(s) of interest through genetic modification and define its effect on the gut microbiome. With the rapid development of new revolutionary technologies of genetic modification, such as CRISPR-Cas9 and transcription activator-like effector nuclease (TALEN), it is possible for the first time to precisely modify the porcine genome as never before. However, to our knowledge, there is no relevant report about investigating host genetic effects on the gut microbiota using these technologies. In mice, the knock-out of genes at least 30 genomic loci has been shown to produce distinct gut microbiomes or altered bacterial colonization engraftment compared to wild-type miceCitation105.

Finally, the gut microbiota community is a complex ecosystem. Beside bacteria, there are large numbers of viruses, fungi, and archaea. It is interesting to identify host genes/variants associated with viruses, fungi, and archaea. However, to our knowledge, there are few studies about host genetic effects on these microbes because bigger challenges exist for these kinds of microbes compared to bacteria owning to the low coverage of sequences in tested samples and the lack of universal marker genes (except fungi). Large sample size and deep sequencing are required for further GWAS analysis.

Acknowledgments

We are grateful to Hui Yang, Jinyan Wu and the colleagues in the State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, and Professor Michel Georges and Dr. Carole Charlier in the Unit of Animal Genomics, GIGA-Institute and Faculty of Veterinary Medicine, University of Liege for working together on the host genetic effects on pig gut microbiome.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was supported by The National Natural Science Foundation of China (32272831, 31790410, 31772579) and National pig industry technology system (CARS-35).

References

  • Berg RD. The indigenous gastrointestinal microflora. Trends Microbiol. 1996;4(11):430–20. doi:10.1016/0966-842X(96)10057-3.
  • Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164(3):337–340. doi:10.1016/j.cell.2016.01.013.
  • Chen C, Zhou Y, Fu H, Xiong X, Fang S, Jiang H, Wu J, Yang H, Gao J, Huang L. Expanded catalog of microbial genes and metagenome-assembled genomes from the pig gut microbiome. Nat Commun. 2021;12(1):1106. doi:10.1038/s41467-021-21295-0.
  • Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535(7610):65–74. doi:10.1038/nature18847.
  • Tremaroli V, Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature. 2012;489(7415):242–249. doi:10.1038/nature11552.
  • Sylvia KE, Demas GE. A gut feeling: microbiome-brain-immune interactions modulate social and affective behaviors. Horm Behav. 2018;99:41–49. doi:10.1016/j.yhbeh.2018.02.001.
  • Benson AK, Kelly SA, Legge R, Ma F, Low SJ, Kim J, Zhang M, Oh PL, Nehrenberg D, Hua K, et al. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc Natl Acad Sci U S A. 2010;107(44):18933–18938. doi:10.1073/pnas.1007028107.
  • Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, Costea PI, Godneva A, Kalka IN, Bar N, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature. 2018;555(7695):210–215. doi:10.1038/nature25973.
  • Lopera-Maya EA, Kurilshikov A, van der Graaf A, Hu S, Andreu-Sanchez S, Chen L, Vila AV, Gacesa R, Sinha T, Collij V, et al. Effect of host genetics on the gut microbiome in 7,738 participants of the Dutch microbiome project. Nat Genet. 2022;54(2):143–151. doi:10.1038/s41588-021-00992-y.
  • Wang J, Thingholm LB, Skieceviciene J, Rausch P, Kummen M, Hov JR, Degenhardt F, Heinsen FA, Rühlemann MC, Szymczak S, et al. Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat Genet. 2016;48(11):1396–1406. doi:10.1038/ng.3695.
  • Yang H, Wu J, Huang X, Zhou Y, Zhang Y, Liu M, Liu Q, Ke S, He M, Fu H, et al. ABO genotype alters the gut microbiota by regulating GalNAc levels in pigs. Nature. 2022;606(7913):358–367. doi:10.1038/s41586-022-04769-z.
  • Gacesa R, Kurilshikov A, Vich Vila A, Sinha T, Klaassen MAY, Bolte LA, Andreu-Sánchez S, Chen L, Collij V, Hu S, et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature. 2022;604(7907):732–739. doi:10.1038/s41586-022-04567-7.
  • Wylensek D, Hitch TCA, Riedel T, Afrizal A, Kumar N, Wortmann E, Liu T, Devendran S, Lesker TR, Hernández SB, et al. A collection of bacterial isolates from the pig intestine reveals functional and taxonomic diversity. Nat Commun. 2020;11(1):6389. doi:10.1038/s41467-020-19929-w.
  • Holman DB, Brunelle BW, Trachsel J, Allen HK, Bik H. Meta-analysis to define a core microbiota in the swine gut. mSystems. 2017;2017(3):2. doi:10.1128/mSystems.00004-17.
  • Zhang L, Wu W, Lee YK, Xie J, Zhang H. Spatial heterogeneity and co-occurrence of mucosal and luminal microbiome across swine intestinal tract. Front Microbiol. 2018;9:48. doi:10.3389/fmicb.2018.00048.
  • Crespo-Piazuelo D, Estelle J, Revilla M, Criado-Mesas L, Ramayo-Caldas Y, Ovilo C, Fernández AI, Ballester M, Folch JM. Characterization of bacterial microbiota compositions along the intestinal tract in pigs and their interactions and functions. Sci Rep. 2018;8(1):12727. doi:10.1038/s41598-018-30932-6.
  • Yang H, Huang X, Fang S, Xin W, Huang L, Chen C. Uncovering the composition of microbial community structure and metagenomics among three gut locations in pigs with distinct fatness. Sci Rep. 2016;6(1):27427. doi:10.1038/srep27427.
  • Luo Y, Ren W, Smidt H, Wright AG, Yu B, Schyns G, McCormack UM, Cowieson AJ, Yu J, He J, et al. Dynamic distribution of gut microbiota in pigs at different growth stages: composition and contribution. Microbiol Spectr. 2022;10(3):e0068821. doi:10.1128/spectrum.00688-21.
  • Ke S, Fang S, He M, Huang X, Yang H, Yang B, Chen C, Huang L. Age-based dynamic changes of phylogenetic composition and interaction networks of health pig gut microbiome feeding in a uniformed condition. BMC Vet Res. 2019;15(1):172. doi:10.1186/s12917-019-1918-5.
  • Wang X, Tsai T, Deng F, Wei X, Chai J, Knapp J, Apple J, Maxwell CV, Lee JA, Li Y, et al. Longitudinal investigation of the swine gut microbiome from birth to market reveals stage and growth performance associated bacteria. Microbiome. 2019;7(1):109. doi:10.1186/s40168-019-0721-7.
  • Xiao L, Estelle J, Kiilerich P, Ramayo-Caldas Y, Xia Z, Feng Q, Liang S, Pedersen AØ, Kjeldsen NJ, Liu C, et al. A reference gene catalogue of the pig gut microbiome. Nat Microbiol. 2016;1(12):16161. doi:10.1038/nmicrobiol.2016.161.
  • Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, Arumugam M, Kultima JR, Prifti E, Nielsen T, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32(8):834–841. doi:10.1038/nbt.2942.
  • Fassarella M, Blaak EE, Penders J, Nauta A, Smidt H, Zoetendal EG. Gut microbiome stability and resilience: elucidating the response to perturbations in order to modulate gut health. Gut. 2021;70(3):595–605. doi:10.1136/gutjnl-2020-321747.
  • Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489(7415):220–230. doi:10.1038/nature11550.
  • De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, Collini S, Pieraccini G, Lionetti P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107(33):14691–14696. doi:10.1073/pnas.1005963107.
  • Fragiadakis GK, Smits SA, Sonnenburg ED, Van Treuren W, Reid G, Knight R, Manjurano A, Changalucha J, Dominguez-Bello MG, Leach J, et al. Links between environment, diet, and the hunter-gatherer microbiome. Gut Microbes. 2019;10(2):216–227. doi:10.1080/19490976.2018.1494103.
  • Daniel H, Gholami AM, Berry D, Desmarchelier C, Hahne H, Loh G, Mondot S, Lepage P, Rothballer M, Walker A, et al. High-fat diet alters gut microbiota physiology in mice. Isme J. 2014;8(2):295–308. doi:10.1038/ismej.2013.155.
  • Bian G, Ma S, Zhu Z, Su Y, Zoetendal EG, Mackie R, Liu J, Mu C, Huang R, Smidt H, et al. Age, introduction of solid feed and weaning are more important determinants of gut bacterial succession in piglets than breed and nursing mother as revealed by a reciprocal cross-fostering model. Environ Microbiol. 2016;18(5):1566–1577. doi:10.1111/1462-2920.13272.
  • Wu S, Bhat ZF, Gounder RS, Mohamed Ahmed IA, Al-Juhaimi FY, Ding Y, Bekhit AEDA. Effect of dietary protein and processing on gut microbiota-a systematic review. Nutrients. 2022;14(3):14. doi:10.3390/nu14030453.
  • Luo Y, Liu Y, Li H, Zhao Y, Wright AG, Cai J, Tian G, Mao X. Differential effect of dietary fibers in intestinal health of growing pigs: outcomes in the gut microbiota and immune-related indexes. Front Microbiol. 2022;13:843045.
  • Wu G, Tang X, Fan C, Wang L, Shen W, Ren S, Zhang L, Zhang Y. Gastrointestinal tract and dietary fiber driven alterations of gut microbiota and metabolites in durco x bamei crossbred pigs. Front Nutr. 2021;8:806646. doi:10.3389/fnut.2021.806646.
  • Zhu C, Yang J, Nie X, Wu Q, Wang L, Jiang Z. Influences of dietary vitamin e, selenium-enriched yeast, and soy isoflavone supplementation on growth performance, antioxidant capacity, carcass traits, meat quality and gut microbiota in finishing pigs. Antioxid (Basel). 2022;11(8):11. doi:10.3390/antiox11081510.
  • Zhen R, Liu C, Wei C, Luo Y, Hu X, Liu G, Yi H, Huang Y. Effect of different dosages of sodium butyrate and niacin on growth, faecal microbiota and vitamin B metabolism in weaned piglets. J Appl Microbiol. 2022;132(6):4466–4475. doi:10.1111/jam.15545.
  • Guo D, Zhang L, Zhang L, Han S, Yang K, Lin X, Wen C, Tong A, Zhang M, Yin Y, et al. Effect of dietary methylsulfonylmethane supplementation on growth performance, hair quality, fecal microbiota, and metabolome in ragdoll kittens. Front Microbiol. 2022;13:838164. doi:10.3389/fmicb.2022.838164.
  • Sarri L, Costa-Roura S, Balcells J, Seradj AR, de la Fuente G. The impact of genetics on gut microbiota of growing and fattening pigs under moderate N restriction. Animals (Basel): Open Access J MDPI. 2021;11(10):11. doi:10.3390/ani11102846.
  • Looft T, Johnson TA, Allen HK, Bayles DO, Alt DP, Stedtfeld RD, Sul WJ, Stedtfeld TM, Chai B, Cole JR, et al. In-feed antibiotic effects on the swine intestinal microbiome. Proc Natl Acad Sci U S A. 2012;109(5):1691–1696. doi:10.1073/pnas.1120238109.
  • Looft T, Allen HK, Cantarel BL, Levine UY, Bayles DO, Alt DP, Henrissat B, Stanton TB. Bacteria, phages and pigs: the effects of in-feed antibiotics on the microbiome at different gut locations. Isme J. 2014;8(8):1566–1576. doi:10.1038/ismej.2014.12.
  • Pi Y, Gao K, Peng Y, Mu CL, Zhu WY. Antibiotic-induced alterations of the gut microbiota and microbial fermentation in protein parallel the changes in host nitrogen metabolism of growing pigs. Animal. 2019;13(2):262–272. doi:10.1017/S1751731118001416.
  • Dowarah R, Verma AK, Agarwal N. The use of lactobacillus as an alternative of antibiotic growth promoters in pigs: a review. Anim Nutr. 2017;3(1):1–6. doi:10.1016/j.aninu.2016.11.002.
  • Toscano M, De Grandi R, Miniello VL, Mattina R, Drago L. Ability of lactobacillus kefiri LKF01 (DSM32079) to colonize the intestinal environment and modify the gut microbiota composition of healthy individuals. Dig Liver Dis. 2017;49(3):261–267. doi:10.1016/j.dld.2016.11.011.
  • Shin D, Chang SY, Bogere P, Won K, Choi JY, Choi YJ, Lee HK, Hur J, Park B-Y, Kim Y, et al. Beneficial roles of probiotics on the modulation of gut microbiota and immune response in pigs. Plos One. 2019;14(8):e0220843. doi:10.1371/journal.pone.0220843.
  • Dicksved J, Halfvarson J, Rosenquist M, Jarnerot G, Tysk C, Apajalahti J, Engstrand L, Jansson JK. Molecular analysis of the gut microbiota of identical twins with Crohn’s disease. Isme J. 2008;2(7):716–727. doi:10.1038/ismej.2008.37.
  • Zhang Y, Si X, Yang L, Wang H, Sun Y, Liu N. Association between intestinal microbiota and inflammatory bowel disease. Animal Model Exp Med. 2022;5(4):311–322. doi:10.1002/ame2.12255.
  • Gu W, Zhang L, Han T, Huang H, Chen J. Dynamic changes in gut microbiome of ulcerative colitis: initial study from animal model. J Inflamm Res. 2022;15:2631–2647. doi:10.2147/JIR.S358807.
  • Chung the H, Le SH. Dynamic of the human gut microbiome under infectious diarrhea. Curr Opin Microbiol. 2022;66:79–85.
  • Gresse R, Chaucheyras-Durand F, Fleury MA, Van de Wiele T, Forano E, Blanquet-Diot S. Gut microbiota dysbiosis in postweaning piglets: understanding the keys to health. Trends Microbiol. 2017;25(10):851–873. doi:10.1016/j.tim.2017.05.004.
  • Li J, Zhou J, Zhao S, Guo R, Zhong C, Xue T, Peng Q, Zhang B, Fan B, Liu C, et al. Pathogenicity, infective dose and altered gut microbiota in piglets infected with porcine deltacoronavirus. Virology. 2022;567:26–33.
  • Hu C, Niu X, Chen S, Wen J, Bao M, Mohyuddin SG, Yong Y, Liu X, Wu L, Yu Z, et al. A comprehensive analysis of the colonic flora diversity, short chain fatty acid metabolism, transcripts, and biochemical indexes in heat-stressed pigs. Front Immunol. 2021;12:717723.
  • Wen C, van Dixhoorn I, Schokker D, Woelders H, Stockhofe-Zurwieden N, Rebel JMJ, Smidt H. Environmentally enriched housing conditions affect pig welfare, immune system and gut microbiota in early life. Anim Microbiome. 2021;3(1):52. doi:10.1186/s42523-021-00115-2.
  • Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ, Danska JS, et al. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 2013;339(6123):1084–1088. doi:10.1126/science.1233521.
  • Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, Chervonsky A. Gender bias in autoimmunity is influenced by microbiota. Immunity. 2013;39:400–412.
  • He M, Gao J, Wu J, Zhou Y, Fu H, Ke S, Yang H, Chen C, Huang L. Host gender and androgen levels regulate gut bacterial taxa in pigs leading to sex-biased serum metabolite profiles. Front Microbiol. 2019;10:1359. doi:10.3389/fmicb.2019.01359.
  • Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: physiological and clinical implications. Maturitas. 2017;103:45–53.
  • Ervin SM, Li H, Lim L, Roberts LR, Liang X, Mani S, Redinbo MR. Gut microbial beta-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J Biol Chem. 2019;294(49):18586–18599. doi:10.1074/jbc.RA119.010950.
  • Turpin W, Espin-Garcia O, Xu W, Silverberg MS, Kevans D, Smith MI, Guttman DS, Griffiths A, Panaccione R, Otley A, et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat Genet. 2016;48(11):1413–1417. doi:10.1038/ng.3693.
  • Kurilshikov A, Medina-Gomez C, Bacigalupe R, Radjabzadeh D, Wang J, Demirkan A, Le Roy CI, Raygoza Garay JA, Finnicum CT, Liu X, et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat Genet. 2021;53:156–165.
  • Chen C, Huang X, Fang S, Yang H, He M, Zhao Y, Huang L. Contribution of host genetics to the variation of microbial composition of cecum lumen and feces in pigs. Front Microbiol. 2018;9:2626.
  • Blekhman R, Goodrich JK, Huang K, Sun Q, Bukowski R, Bell JT, Spector TD, Keinan A, Ley RE, Gevers D, et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 2015;16(1):191. doi:10.1186/s13059-015-0759-1.
  • Wijsman EM. The role of large pedigrees in an era of high-throughput sequencing. Hum Genet. 2012;131:1555–1563.
  • Whitham TG, Bailey JK, Schweitzer JA, Shuster SM, Bangert RK, LeRoy CJ, Lonsdorf EV, Allan GJ, DiFazio SP, Potts BM, et al. A framework for community and ecosystem genetics: from genes to ecosystems. Nat Rev Genet. 2006;7(7):510–523. doi:10.1038/nrg1877.
  • van Opstal EJ, Bordenstein SR. Rethinking heritability of the microbiome. Science. 2015;349(6253):1172–1173. doi:10.1126/science.aab3958.
  • Campbell JH, Foster CM, Vishnivetskaya T, Campbell AG, Yang ZK, Wymore A, Palumbo AV, Chesler EJ, Podar M. Host genetic and environmental effects on mouse intestinal microbiota. Isme J. 2012;6(11):2033–2044. doi:10.1038/ismej.2012.54.
  • Li F, Li C, Chen Y, Liu J, Zhang C, Irving B, Fitzsimmons C, Plastow G, Guan LL. Host genetics influence the rumen microbiota and heritable rumen microbial features associate with feed efficiency in cattle. Microbiome. 2019;7(1):92. doi:10.1186/s40168-019-0699-1.
  • Fan P, Bian B, Teng L, Nelson CD, Driver J, Elzo MA, Jeong KC. Host genetic effects upon the early gut microbiota in a bovine model with graduated spectrum of genetic variation. Isme J. 2020;14(1):302–317. doi:10.1038/s41396-019-0529-2.
  • Bergamaschi M, Maltecca C, Schillebeeckx C, McNulty NP, Schwab C, Shull C, Fix J, Tiezzi F. Heritability and genome-wide association of swine gut microbiome features with growth and fatness parameters. Sci Rep. 2020;10:10134.
  • Simpson CL, Justice CM, Krishnan M, Wojciechowski R, Sung H, Cai J, Green T, Lewis D, Behneman D, Wilson AF, et al. Old lessons learned anew: family-based methods for detecting genes responsible for quantitative and qualitative traits in the genetic analysis workshop 17 mini-exome sequence data. BMC Proc. 2011;5(Suppl 9):S83.
  • Bonder MJ, Kurilshikov A, Tigchelaar EF, Mujagic Z, Imhann F, Vila AV, Deelen P, Vatanen T, Schirmer M, Smeekens SP, et al. The effect of host genetics on the gut microbiome. Nat Genet. 2016;48(11):1407–1412. doi:10.1038/ng.3663.
  • Dobson AJ, Chaston JM, Newell PD, Donahue L, Hermann SL, Sannino DR, Westmiller S, Wong ACN, Clark AG, Lazzaro BP, et al. Host genetic determinants of microbiota-dependent nutrition revealed by genome-wide analysis of drosophila melanogaster. Nat Commun. 2015;6(1):6312. doi:10.1038/ncomms7312.
  • Chaston JM, Dobson AJ, Newell PD, Douglas AE, Drake HL. Host genetic control of the microbiota mediates the drosophila nutritional phenotype. Appl Environ Microbiol. 2016;82(2):671–679. doi:10.1128/AEM.03301-15.
  • Org E, Parks BW, Joo JW, Emert B, Schwartzman W, Kang EY, Mehrabian M, Pan C, Knight R, Gunsalus R, et al. Genetic and environmental control of host-gut microbiota interactions. Genome Res. 2015;25(10):1558–1569. doi:10.1101/gr.194118.115.
  • Li J, George Markowitz RH, Brooks AW, Mallott EK, Leigh BA, Olszewski T, Zare H, Bagheri M, Smith HM, Friese KA, et al. Individuality and ethnicity eclipse a short-term dietary intervention in shaping microbiomes and viromes. PLoS Biol. 2022;20(8):e3001758. doi:10.1371/journal.pbio.3001758.
  • Davenport ER. Elucidating the role of the host genome in shaping microbiome composition. Gut Microbes. 2016;7(2):178–184. doi:10.1080/19490976.2016.1155022.
  • Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell J, et al. Human genetics shape the gut microbiome. Cell. 2014;159(4):789–799. doi:10.1016/j.cell.2014.09.053.
  • Goodrich JK, Davenport ER, Beaumont M, Jackson MA, Knight R, Ober C, Spector TD, Bell JT, Clark AG, Ley RE, et al. Genetic determinants of the gut microbiome in UK twins. Cell Host & Microbe. 2016;19(5):731–743. doi:10.1016/j.chom.2016.04.017.
  • Lim MY, You HJ, Yoon HS, Kwon B, Lee JY, Lee S, Song Y-M, Lee K, Sung J, Ko G, et al. The effect of heritability and host genetics on the gut microbiota and metabolic syndrome. Gut. 2017;66(6):1031–1038. doi:10.1136/gutjnl-2015-311326.
  • Davenport ER, Cusanovich DA, Michelini K, Barreiro LB, Ober C, Gilad Y, White BA. Genome-wide association studies of the human gut microbiota. Plos One. 2015;10(11):e0140301. doi:10.1371/journal.pone.0140301.
  • O’Connor A, Quizon PM, Albright JE, Lin FT, Bennett BJ. Responsiveness of cardiometabolic-related microbiota to diet is influenced by host genetics. Mamm Genome. 2014;25(11–12):583–599. doi:10.1007/s00335-014-9540-0.
  • Camarinha-Silva A, Maushammer M, Wellmann R, Vital M, Preuss S, Bennewitz J. Host genome influence on gut microbial composition and microbial prediction of complex traits in pigs. Genetics. 2017;206(3):1637–1644. doi:10.1534/genetics.117.200782.
  • Visscher PM, Hill WG, Wray NR. Heritability in the genomics era–concepts and misconceptions. Nat Rev Genet. 2008;9(4):255–266. doi:10.1038/nrg2322.
  • Fan P, Nelson CD, Driver JD, Elzo MA, Penagaricano F, Jeong KC. Host genetics exerts lifelong effects upon hindgut microbiota and its association with bovine growth and immunity. Isme J. 2021;15(8):2306–2321. doi:10.1038/s41396-021-00925-x.
  • Hughes DA, Bacigalupe R, Wang J, Ruhlemann MC, Tito RY, Falony G, Joossens M, Vieira-Silva S, Henckaerts L, Rymenans L, et al. Genome-wide associations of human gut microbiome variation and implications for causal inference analyses. Nat Microbiol. 2020;5(9):1079–1087. doi:10.1038/s41564-020-0743-8.
  • Qin Y, Havulinna AS, Liu Y, Jousilahti P, Ritchie SC, Tokolyi A, Sanders JG, Valsta L, Brożyńska M, Zhu Q, et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat Genet. 2022;54:134–142.
  • Ji J, Luo CL, Zou X, Lv XH, Xu YB, Shu DM, Qu H. Association of host genetics with intestinal microbial relevant to body weight in a chicken F2 resource population. Poult Sci. 2019;98:4084–4093.
  • Boulund U, Bastos DM, Ferwerda B, van den Born BJ, Pinto-Sietsma SJ, Galenkamp H, Levin E, Groen AK, Zwinderman AH, Nieuwdorp M., et al. Gut microbiome associations with host genotype vary across ethnicities and potentially influence cardiometabolic traits. Cell Host & Microbe. 2022;30(10):1464–1480.e6.
  • Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman D. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638.
  • Reverter A, Ballester M, Alexandre PA, Marmol-Sanchez E, Dalmau A, Quintanilla R, Ramayo-Caldas Y. A gene co-association network regulating gut microbial communities in a duroc pig population. Microbiome. 2021;9:52.
  • Kuokkanen M, Kokkonen J, Enattah NS, Ylisaukko-Oja T, Komu H, Varilo T, Peltonen L, Savilahti E, Järvelä I. Mutations in the translated region of the lactase gene (LCT) underlie congenital lactase deficiency. Am J Hum Genet. 2006;78:339–344.
  • Ravcheev DA, Thiele I. Comparative genomic analysis of the human gut microbiome reveals a broad distribution of metabolic pathways for the degradation of host-synthetized mucin glycans and utilization of mucin-derived monosaccharides. Front Genet. 2017;8:111.
  • Yamamoto F, Yamamoto M. Molecular genetic basis of porcine histo-blood group AO system. Blood. 2001;97:3308–3310.
  • Nguyen DT, Choi H, Jo H, Kim JH, Dirisala VR, Lee KT, Kim TH, Park KK, Seo K, Park C. Molecular characterization of the human ABO blood group orthologus system in pigs. Anim Genet. 2011;42:325–328.
  • Choi MK, Le MT, Cho H, Yum J, Kang M, Song H, Kim JH, Chung HJ, Hong K, Park C. Determination of complete sequence information of the human ABO blood group orthologous gene in pigs and breed difference in blood type frequencies. Gene. 2018;640:1–5.
  • Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14:141–153.
  • Miro-Blanch J, Yanes O. Epigenetic regulation at the interplay netween gut microbiota and host metabolism. Front Genet. 2019;10:638.
  • LeBlanc JG, Laino JE, Del Valle MJ, Vannini V, van Sinderen D, Taranto MP, de Valdez GF, de Giori GS, Sesma F. B-group vitamin production by lactic acid bacteria–current knowledge and potential applications. J Appl Microbiol. 2011;111:1297–1309.
  • Rossi M, Amaretti A, Raimondi S. Folate production by probiotic bacteria. Nutrients. 2011;3:118–134.
  • Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett-Wilt GA, Rabaglia ME, Keller MP, Attie AD, Rey FE, Denu JM. Diet-microbiota interactions mediate global epigenetic programming in multiple host tissues. Mol Cell. 2016;64:982–992.
  • Pan X, Gong D, Nguyen DN, Zhang X, Hu Q, Lu H, Fredholm M, Sangild PT, Gao F. Early microbial colonization affects DNA methylation of genes related to intestinal immunity and metabolism in preterm pigs. DNA Res. 2018;25:287–296.
  • Funkhouser LJ, Bordenstein SR. Mom knows best: the universality of maternal microbial transmission. PLoS Biol. 2013;11:e1001631.
  • Jost T, Lacroix C, Braegger CP, Rochat F, Chassard C. Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ Microbiol. 2014;16:2891–2904.
  • Treichel NS, Prevorsek Z, Mrak V, Kostric M, Vestergaard G, Foesel B, Pfeiffer S, Stres B, Schöler A, Schloter M. Effect of the nursing mother on the gut microbiome of the offspring during early mouse development. Microb Ecol. 2019;78:517–527.
  • Meyer F, Fritz A, Deng ZL, Koslicki D, Lesker TR, Gurevich A, Robertson G, Alser M, Antipov D, Beghini F, et al. Critical assessment of metagenome interpretation: the second round of challenges. Nat Methods. 2022;19:429–440.
  • Klindworth A, Pruesse E, Schweer T, Peplies J, Quast C, Horn M, Glöckner FO. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 2013;41:e1.
  • Gohl DM, Vangay P, Garbe J, MacLean A, Hauge A, Becker A, Gould TJ, Clayton JB, Johnson TJ, Hunter R, et al. Systematic improvement of amplicon marker gene methods for increased accuracy in microbiome studies. Nat Biotechnol. 2016;34:942–949.
  • Brooks JP, Edwards DJ, Harwich MD Jr., Rivera MC, Fettweis JM, Serrano MG, Reris RA, Sheth NU, Huang B, Girerd P, et al. The truth about metagenomics: quantifying and counteracting bias in 16S rRNA studies. BMC Microbiol. 2015;15:66.
  • Bubier JA, Chesler EJ, Weinstock GM. Host genetic control of gut microbiome composition. Mamm Genome. 2021;32:263–281.