Skip to main content

Colonization and development of the gut microbiome in calves

Abstract

Colonization and development of the gut microbiome are crucial for the growth and health of calves. In this review, we summarized the colonization, beneficial nutrition, immune function of gut microbiota, function of the gut barrier, and the evolution of core microbiota in the gut of calves of different ages. Homeostasis of gut microbiome is beneficial for nutritional and immune system development of calves. Disruption of the gut microbiome leads to digestive diseases in calves, such as diarrhea and intestinal inflammation. Microbiota already exists in the gut of calf fetuses, and the colonization of microbiota continues to change dynamically under the influence of various factors, which include probiotics, diet, age, and genotype. Colonization depends on the interaction between the gut microbiota and the immune system of calves. The abundance and diversity of these commensal microbiota stabilize and play a critical role in the health of calves.

Introduction

Microbiota in the gut is involved in the health and development of ruminants [1], and it is crucial for the development of calves [2]. The rumen microbiome of calves forms rapidly after birth and begins to establish during early life. With the development of the gastrointestinal tract (GIT), the gut microbiome of calves changes gradually, especially during weaning [3, 4]. These changes in the gut microbiome, along with nutritional and immune functions, have potential effects on host metabolism. Therefore, to optimize the health and production efficiency of ruminants, it is important to understand the factors that affect the development of the GIT. The development of the gut microbiome is mainly manipulated through probiotics, diet, age, and genotype of the calf [5,6,7]. Nevertheless, there is a dynamic balance among the gut microbiome, host physiology, and diet. This dynamic balance of the gut microbiome directly influences the initial acquisition, continued development, and final stability of rumen and intestinal ecosystems [8].

Application of metabolomics, metatranscriptomics, and single-cell RNA sequencing have enabled a more accurate understanding of the composition of the GIT microbiome at different stages of calf growth along with further exploration of their functional analysis through mapping in KEGG databases. These sequencing results suggested that complex microbial communities colonized the GIT of newborn calves. The early settlement of gut microbiota influenced the performance and lifelong health of animals [9,10,11]. There are interactions between the gut microbiome and calf growth, and these interactions are vital to the development, growth, and health of the calf. Thus, the causal relationship between the gut microbiome and calf health and the core microbiota that links host genetics and phenotypes needs to be explored. Many investigators have focused on temporal changes in the gut microbiome of newborn ruminants, particularly after weaning [12]. However, there is still a lack of understanding of the changes and development of the gut microbiome in young ruminants. This review summarizes recent developments in our knowledge of the gut microbiome of young ruminants and aims to understand the patterns of microbiome development in young ruminants and to provide novel insights to improve gut health and production of ruminants.

Nutritional and immune function of the gut microbiome

Many studies on ruminants have focused on how the microbiome affected host nutrition and immunity, which ultimately enhanced host performance and health (Fig. 1) [13,14,15,16]. Gut microbiota provides nutrition for the host through fermentation. The rumen microbiome preserves the health of its host by destroying harmful byproducts of fermentation. If the structure of the rumen microbiota is disordered, the health of ruminants is threatened. For example, when ruminant diets contained a high proportion of concentrate, lactic acid bacteria increased rapidly to produce lactic acid, which led to the death of Gram-negative bacteria; this released lipopolysaccharides and caused gastrointestinal diseases in calves [17]. Rumen microbiota was indispensable for the degradation of plant fibers [18]. Among them, hemicellulose was degraded mainly by Prevotella spp., whereas cellulose was degraded mainly by Clostridium spp. In addition, certain dominant bacteria in the rumen were related to feed efficiency [19]. In calf rumen, Lachnospiraceae, Lactobacillaceae, and Veillonellaceae were related negatively with high feed utilization, whereas Methanomassiliicoccales was related positively to feed utilization [20], suggesting that the feed efficiency may be related to specific genes of carbohydrate-active enzymes in host.

Fig. 1
figure 1

The function of the gut microbiome in calves. Created with BioRender.com

GIT microbiota influences GIT development and function by producing short-chain fatty acids (SCFAs), amino acids (AAs) or their derivatives, and bacteriocins. SCFAs were produced by the rapid fermentation of carbohydrates and were the main energy-supply substances in the epithelial cells of the GIT. Acetate coordinated interactions between epithelial and immune cells by inducing B cells to produce T-cell-dependent immunoglobulin A (IgA), which altered bacterial localization within the colon [21]. Propionate participated directly in gluconeogenesis to provide energy to calves. Supplementation with propionate induced mRNA expression of genes involved in gluconeogenesis in immortalized bovine intestinal epithelial cells [22]. In the rumen, 80% of butyrate was converted into ketone bodies, which provided > 80% of the energy for the growth of rumen epithelial cells [23]. In the intestine, butyrate provided 70% of the energy for intestinal epithelial cells through β-oxidation [24]. Butyrate increased the expression of cell cycle-related genes and decreased the expression of apoptosis-related genes, thereby regulating the proliferation of rumen epithelial cells [25]. In colonic epithelial cells, butyrate arrested the cell cycle at the G1 phase. SCFAs also regulated mitogen-activated protein kinases, sphingolipids, insulin, oxytocin, calcium, cell proliferation, and apoptosis by inhibiting histone deacetylases and activating G-protein-coupled receptors. Moreover, butyrate increased plasma GLP-2 concentration, total tract dry matter, and organic matter digestibility in lactating dairy cows [26]. Valerate increased the TEER of Caco-2 cells and reduced the paracellular permeability [27]. In rodents, valerate was correlated negatively with allobaculum, and serum valerate was potentially harmful to the health of rats [28]. In weaned piglets, with increased levels of valerate after the supplement of Saccharomyces boulardii, the feed conversion ratio increased and diarrhea was decreased [29]. However, no studies have focused mainly on the effects of valerate on calf development, therefore, further exploration is required.

Not all proteins were absorbed fully and utilized in the mammalian small intestine, and the remaining proteins passed through the hindgut of animals as proteins or peptides after preliminary digestion [30]. These proteins and AAs were transformed into other forms of AAs and their derivatives after fermentation by hindgut microbiota. Microbiota was involved in the metabolism of branched-chain amino acids (BCAAs). BCAAs were important for calf performance; supplementation with BCAAs (2 g/kg body weight/d; 1:1:1 of valine, leucine, and isoleucine) during nursing increased average daily weight gain in calves significantly [31]. In addition, the content of BCAAs in feces was correlated positively with calf diarrhea; however, the BCAA content in serum was correlated negatively with diarrhea, which indicated that diarrhea impaired the ability to absorb BCAAs [32].

A similar phenomenon was observed for other AAs (e.g., alanine, glycine, arginine, ornithine, and glutamic acid) [32]. The content of BCAAs and other AAs, such as plasma diamine oxidase, were biomarkers of calf diarrhea due to the negative correlation between the AA status in plasma and calf diarrhea in other studies [33, 34]. Mechanistically, angiotensin I-converting enzyme 2 was linked to AAs, microbial ecology, and intestinal inflammation by activating the mTOR signaling pathway. In the hindgut, microbiota degraded proteins and produced bioactive AA derivatives, such as tryptamine, histamine, dopamine, phenylacetylglutamine, and phenylacetylglycine [30]. In addition, tryptophan derivatives, such as indole, indoleamine-2,3-dioxygenase 1 and 2, and tryptophan-2,3-dioxygenase, were involved in the kynurenine pathway and had important implications for intestinal homeostasis [35]. Mechanistically, tryptophan derivatives acted mainly on aryl hydrocarbon receptors to participate in pro-inflammatory and tolerance responses, which meant that the remaining bacteria, such as Limosilactobacillus reuteri and Lactobacillus, exerted nutritional and immune effects on the host by altering tryptophan metabolism [36,37,38].

Bacteriocins are bactericidal proteins or polypeptides encoded by bacterial or archaeal genes that are synthesized by ribosomes. Bacteriocin-producing bacteria are immune to their own bacteriocins. Bacteriocins have received substantial attention because of their ability to protect the host gut against harmful bacteria [39, 40]. Lactobacillus frumenti prevented early weaned piglet diarrhea by secreting gassericin A, which is a class of bacteriocins [41]. In addition, when the concentration of this bacteriocin was sufficient, Lacticin 3147 prevented staphylococcal mastitis infections in cows [42].

Gut microbiota also interacts with bile acids in the host. In recent years, limited research has been conducted on the effect of bile acids on the intestinal health of calves, while the studies of bile acids have been very popular in other animals [43,44,45,46]. However, a recent study showed that gut microbial-derived ursodeoxybile acid ameliorated diarrhea effectively and improved the growth performance of calves [47]. Ursodeoxybile acid isolated from calves that were successfully treated with fecal microbiota transplantation (FMT) reduced colitis-induced Escherichia (E.) coli infection and hindgut microbial damage in a mouse model [47]. This study showed that the bile acids were associated with calf diarrhea, which provided pioneering insights into the development of bile acids and microbial stability in calf intestines. Overall, microbiota in the calf GIT plays a nutritional and immune role through its own metabolites, and additional, yet unknown, metabolites and their mechanisms are expected to be discovered in the future.

Role of the microbiome on the gut barrier in calves

The protective barrier of the gut consists of mechanical, chemical, and immune components. Microbiota is necessary for the development and differentiation of the intestinal epithelium and immune system and regulates the innate and acquired immune systems in the intestine. A healthy structure of the gut microbial community is critical for host health. Under normal physiological conditions, the microbial structure in the intestine remains relatively stable, and it forms an intestinal biological barrier that inhibits the colonization of harmful bacteria and ensures calf health. Changes in gut acetate concentration also altered the responsiveness of the IgA pool to various types of bacteria [21]. IgA uncoupled and manipulated colonization during pathogenesis to promote homeostasis by neutralizing bacterial toxins or by enhancing the growth of targeted bacterial species [21, 48,49,50,51]. Intestinal bacteria, especially Gram-negative bacteria, activated intestinal dendritic cells, thereby further stimulating intestinal mucosal plasma cells to secrete IgA [52]. Moreover, intestinal commensal bacteria used their components or secretions to induce intestinal Paneth cells to synthesize antibacterial peptides through pattern recognition receptors. The resulting interactions of the microbe-related molecular pattern activated multiple signaling pathways to improve the intestinal mucosal barrier function and to promote the secretion of IgA, mucus glycoproteins, and antibacterial peptides [53], which contributed to the formation and protection of the gut barrier. For example, nisin that was obtained by calves in colostrum formed pores in the mycobacterial cell wall that reduced membrane integrity to kill M. paratuberculosis [54].

Intestinal commensal bacteria regulate the activity of their respective cytokines. For example, the level of IL-6 revealed the alterations in the inflammatory status of calves and cows when they suffered from GIT diseases [55, 56]. These bacteria regulated innate lymphocytes, which responded quickly to cytokine signals produced by the epithelium, such as Foxp3+ regulatory T cells [19, 57]. Microbial cell wall peptidoglycans maintained the structure and function of tight junctions through Toll-like receptor 2 [58]. The Gram-negative bacterium Akkermansia muciniphila increased the endocannabinoid content, which regulated the function of the intestinal barrier by reducing metabolic endotoxins [59]. Gut microbiota promoted the development of intestinal microvasculature by inducing angiopoietin-3 transcription factors [60]. As a result, the gut microbiota plays an important role in maintaining the structural and functional integrity of the epithelium.

The disruption of the gut barrier was accompanied by the disorder of microbial structure [61,62,63,64]. Supplementation with probiotics modulated the microbial structure through interactions with other microbiota to protect the gut barrier in newborn calves [65]. However, not all microbiota in the gut is beneficial to host health. Some genus or species of microbiota were the key biomarkers to determine the disorder of the gut microbial structure. Clostridioides difficile and Clostridium perfringens, which produced toxins, had a stonger colonization after the disorder of microbial structure [66, 67]. Key biomarkers in gut microbiota were also used to predict calf health, such as Ruminococcaceae, Lachnospiraceae, and Phocaeicola, Bacteroides, Prevotella, Faecalibacterium, and Butyricicoccus [68]. However, Enterococcus, Ligilactobacillus, Lactobacilus, Gallibacterium streptococcus, and Escherichia/Shigella were more abundant in diarrheic calves than those in healthy calves [68]. At the species level, Eggerthella lenta, Bifidobacterium longum, and Collinsella aerofaciens were associated with healthy status of calves, but E. coli and Lactobacillus species were associated with GIT diseases [69, 70].

Some viruses also impair the gut barrier and health of calves. Rotavirus is one of the main pathogens that cause diarrhea in calves. This infection not only reduced the richness and diversity of the intestinal microbiota significantly, but it also further disturbed the physiological homeostasis of calves [71,72,73]. These results suggested that the colonization of harmful microbiota interfered seriously with the stability of the host immune system, whereas beneficial microbiota improved gut health and immunity of the host.

Dynamics of the colonization process of microbiota in calf guts

Colonization of the gut microbiota in calves is a dynamic, gradual transition from colonization to stability. The colonization of GIT microbes of calves is influenced or changed by various factors, such as dams, environment, diet, and feed supplements [74]. The process of microbiota colonization generally exhibits a certain regularity. Four processes of gut microbiota assembly have been defined in early life: dispersal, selection, drift, and diversification, which determined the priority of the infant colonization process [75]. First, high levels of aerobes and facultative anaerobes (e.g., Lactobacillus and Bifidobacteria) appeared in the GIT of the calf that consumed oxygen in the form of separate molecules, which created hypoxic conditions [76]. Subsequently, strict anaerobes (e.g., Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria) gradually colonized and stabilized the gut.

The time at which microbiota colonized the GIT of ruminants has been a controversial research question. However, the colonization of gut microbiota was observed in ruminants at birth, and recent studies have shown that gut microbiota was already present in the gut of ruminants from the fetal period; microbiota were detected in the rumen, caecum, meconium, and amniotic fluid of calves at 5 months of gestation [77, 78]. The gut microbiota of newborn calves that were not licked by the dam was highly similar to the maternal oral microbes rather than the microbiota of the dam’s vagina, which indicated that the hindgut of newborn calves may rely on the placenta to obtain the dam’s oral or proximal GIT microbiota [79]. These studies illustrated that diverse microbiota colonized the gut before birth and changed rapidly in the early life of the calf [79,80,81,82,83].

Escherichia, Salmonella, Catellicoccus, Pseudomonas, and Phagocytophilum are the primary bacteria that colonized the fetuses of ruminants [84]. The intestinal bacteria of calves mainly consisted of Firmicutes, Proteobacteria, Bacteroidetes, and a small number of Actinomycetes. Numerous studies have shown that the proportion of Proteobacteria was high soon after the birth, but gradually decreased with an increase in Firmicutes and Bacteroidetes (Fig. 2) [8, 86, 87]. This may be because most Proteobacteria are facultative anaerobes, while Firmicutes and Bacteroidetes are strict anaerobes. Approximately 8 h after birth, E. coli and Streptococci colonized all GIT regions (i.e., stomach, small intestine, and cecum) of the calf, and Lactobacillus was detected later, and Clostridium perfringens was detected in the cecum; however, colonization in the other parts of the intestine was not detected at 18 h after birth [88]. Bacteria were only observed in the cecum and feces on the 2nd day after birth, and in the 1st week, Lactobacillus was dominated throughout the GIT. Faecalibacterium was one of the bacteria with the highest content in 1-week-old calves (21.7%); however, Faecalibacterium decreased with an increase in calf age. The fecal microbiota of 3-week-old calves was dominated by Bacteroides, Prevotella, Coccus-Useriella, and Faebacillus [89]. Bacteroides (15.3% ± 1.0%), Prevotella (21.6% ± 1.4%), and Faecalibacterium (10.3% ± 0.3%) were found in the colons of calves at the same age; however, the relative abundance of Clostridium XIV was only 1.6% ± 1% compared with that of the above three genera [90]. Lactococcus flavus and cellulolytic bacteria appeared only 5 weeks after birth; however, Streptococcus and Lactococcus were not detected [89]. Bacteroides prevotella, Clostridium coccoides, and Eubacterium rectale constituted the major fraction of the microbiota within 12 weeks after birth. These observations suggested that the fecal microbial community had the greatest similarity to the bacterial community in the colon; however, it did not represent the composition of the entire GIT. Bifidobacteria and Lactobacilli were able to enter the small intestine from the stomach more easily than E. coli, and these beneficial bacteria had a high density in the GIT of 20-week-old calves [76]. In addition, the diversity metrics (i.e., observed species, Faith’s PD, Shannon index, and evenness) were changed rapidly from 1 to 9 days of age in calves, and diarrheic status did not affect these metrics [91]. This evidence indicated that the gut microbiota changed with age.

Fig. 2
figure 2

Different levels of core microbiota at different growth stages of calves. ("f_" represents family levels and "g_" represents genus levels). Adapted from previous reports [8, 12, 77, 79, 85]. Created with BioRender.com

Rumen has rich microbiota that plays a crucial role in digestion and fermentation, hence, colonization of the rumen has been the subject of most investigations on ruminant microbiota. Newborn calves are thought to be pseudo-monogastric creatures as the rumen is still in development. Gut microbes play a more important role in the early stages of life than later in the rumen. Interestingly, there are significant differences in microbial structure in different regions of the GIT of ruminants [92]. Prevotella and Fibrobacter were predominant in the stomach, while Bacteroides, Clostridium, Alistipes, and Ruminococcus were prevalent in the large intestine, and E. coli was high abundant in the small intestine [93]. Presumably, this difference was due to the structural characteristics and functional differences of each region of the GIT. The intestine is important for feed digestion, nutrient absorption, and the immune system of young ruminants. The main bacteria in the feces of adult cows were Firmicutes (63.7%), Proteobacteria (18.3%), Bacteroides (7.6%), and Actinomycetes (6.8%) [94].

Compared with the dominant bacteria in the rumen, species of Firmicutes were more abundant in the intestine (i.e., duodenum, jejunum, ileum, cecum, colon, and feces), while Spirochaetes and Bacteroidetes were less abundant, and Tenericutes were lower only in the large intestine (i.e., cecum, colon, and feces) [92]. At the family level, Firmicutes and Proteobacteria were more abundant in the small intestine than in other GIT compartments, and bacteria such as Fibrobacter, Treponema, and Methanobrevibacte were present in the rumen but not in feces [92]. However, Methanobrevibacte was much more abundant in the small intestine (3.7%) than in the stomach (0.71%) or large intestine (1.1%) [93]. This indicated that even if bacteria were present in the rumen, the microbiotic difference between the intestine and rumen would make bacterial colonization difficult and influence bacterial abundance.

The expression pattern of Toll-like receptor (TLR) may be one of the reasons for regional differences in gut bacteria [90, 95]. The expression of TLR1 was highest in the ileum, followed by the jejunum, and lowest in the gastric mucosa of 3-week-old calves [95]. The hindgut of young ruminants is the main site of microbial fermentation in early life. Pioneer colonization by microbiota in the hindgut may be of maternal origin, which provides the basis for a newborn calf to utilize nutrients in milk [96]. Firmicutes, Bacteroidetes, and Proteobacteria are the dominant microbial phyla in the hindgut. The relative abundances of microbial genes involved in AA metabolism, carbohydrate metabolism, and energy metabolism were enriched in these microbiota, which indicated the importance of hindgut microbiota in fermentation during the pre-weaning period [97]. As neonatal calves have an underdeveloped rumen during this period, they rely on hindgut microbial fermentation to decompose undigested dietary components. This results in the production of key metabolites such as SCFAs, AAs, and vitamins, which may be absorbed by the hindgut to promote calf growth and development.

In fact, individual differences in the hindgut microbiome affected the growth status of calves [98]. The mucosa-adherent microbiota were significantly different from the microbiota in the lumen of human and rodent guts [99,100,101], which was also true for ruminants [102]. The distribution of microbiota in mucosa and lumen is also different. Bifidobacteria and Lactobacilli were the most abundant microbiota that adhered to the mucous membranes in the GIT of calves (9–11 months old) and sheep (6–9 months old) [103]. At 3 weeks after birth, different bacteria have adhered to the gut mucosa of the calves [90, 104]. The bacterial abundance in the ileal mucosa was higher than that in other regions of calves [104], although most mucus-attached microbiota cannot be classified at the genus level. Above all, the dominant microbiota in different regions of the calf GIT is different at the phylum and genus levels, which is due to niche exclusion and the difference in the morphological structure of each region. Therefore, an in-depth understanding of the early colonization of gut microbes and the factors that influence the establishment of microbiota can provide a basis for reasonable control of calf gut health.

Factors that affect the developmental process of microbiota in calf GIT

The change in the type and quantity of microbiota is a complex process, which is attributed to the interactions between the microbiome and the host, probiotics, diet, and age. A systematic study of microbial development in the GIT of young ruminants could improve feeding strategies, health, and calf production (Table 1).

Table 1 Factors that influence the development of gut microbiota of ruminants

Probiotics

Probiotics have been used widely to improve rumen fermentation and to prevent pathogen colonization in calves. Administering probiotics to calves promotes the establishment of a beneficial gut microbiota, maintains microbiota stability, and inhibits pathogen growth. Moreover, these effects are important before weaning, which indicates that probiotic supplements are more effective in newborn ruminants than in mature gastrointestinal environments. The colonization of E. coli O157:H7 in the intestines of pre-weaned calves decreased with probiotic feeding [105]. The activity of carboxymethyl cellulase and xylanase in the rumen of North American cattle increased with the feeding of anaerobic bacteria, which improved the digestibility of crude protein and cellulose, daily weight gain, and feed efficiency [119]. Active dry yeast increased the abundance of Vibrio spp. in the rumen of calves during the first 28 days of life, which resulted in an elevated butyrate concentration [120]. Supplementation with yeast cultures also increased bacterial diversity in calf rumens, and this effect was especially significant in diets with high fiber levels [121]. Yeast cultures also increased the total number of bacteria, fungi, and protozoa in an artificial rumen [122], which suggested that yeast cultures promoted the multiplication of fiber-degrading bacteria in the rumen and laid the foundation to start feeding calves a fiber-based diet [123].

Commensal E. coli utilized epithelial-derived nitrates, which have an advantage when competing for nitrates with Salmonella; these bacteria invaded the niche of Salmonella to provide resistance to colonization [124]. This demonstrates the concept of niche pre-emption and the priority effect of the infant bacteria. As an action mechanism, probiotics can prevent the colonization of pathogens by pre-empting the space in the GIT. That is, they compete with harmful bacteria for nutrients or produce antibacterial substances, which reduced rumen acidity, improved milk yield, and reduced E. coli excretion in dairy cows [125,126,127]. Based on these theories, we can artificially intervene in and modulate the gut microbiota of newborn calves through probiotic supplementation. A negative correlation between E. coli and Shigella was found in the feces of healthy calves, which was further confirmed in vitro [128]. This suggested that Lactobacillus supplementation inhibited pathogen colonization to reduce calf diarrhea [129, 130].

Compound probiotics, a supplement contains multiple strains of live bacteria rather than just only one or two, promoted the gut bacterial communities of calves and made their microbial community composition more similar [65]. In addition, a high concentration of compound probiotics improved the immunity of calves by increasing the concentration of total serum proteins and immunoglobulin [131]. Compound probiotics also improved the fermentation capacity of calf rumens by producing SCFAs. The average daily weight gain of calves that were fed probiotics also increased significantly; however, with an increase in calf age, the effect of adding probiotics gradually decreased, presumably due to the preferential effect of pioneer microbiota [132]. However, there is still a lack of information regarding the effects of probiotics on the composition, metabolism, and immune function of the gut microbiota. Therefore, further exploration of probiotics is necessary to develop novel, valuable, and safe methods for improving the gut health of young ruminants.

Diet

Weaning is the most stressful and important transitional period in the life of calves. At this time, multiple microorganisms have attached to the feed and colonized the calf GIT. The developing guts of the calves before weaning contained similar dominant phyla (Bacteroidetes, Firmicutes, and Proteobacteria). The intake of solid feed accelerated the initiation of rumen fermentation and substantially changed gut microbial components. In particular, fecal microbiota was richer and more uniform after weaning than the rumen microbiota [133]. Changes in dietary and feeding patterns before weaning had significant and lasting effects on the composition of the gastrointestinal microbiome of young ruminants [134,135,136]. Therefore, in terms of establishing the microbiota and cultivating the fermentation capacity of the rumen microbiome, diet management before weaning is important for young ruminants. Adding oat hay did not change the diversity of rumen microbes in calves; however, it changed the proportion of different microbial populations and affected the rumen pH indirectly [137]. Molasses beet pulp increased the concentration of VFA more than corn grain and promoted hindgut development in lactating cows; however, its application in calves requires further verification [138]. These studies have shown that changes in dietary composition greatly affected the composition of the gut microbiome in calves.

Age

The diversity and stability of gut microbes increase gradually with increased age of a calf [70]. In the rumen, the proportions of Bacteroidetes, Firmicutes, and Proteobacteria varied greatly with calf age [113]. In feces, anaerobic species of bacteria increased with calf age, whereas aerobic species decreased gradually in the GIT of calves [8]. The expression of TLRs in the gastrointestinal tract of calves decreased significantly with age, which reduced the secretion of AMPs in the gut to facilitate colonization by various microorganisms [95]. However, when the gut microbial structure has not yet been stabilized, it facilitated invasion by pathogens.

Calf rumen microbiota showed age-related changes in different classifications and functions [113]. The abundance of Bacteroidetes in the rumen contents of calves increased from 18.1% at 2 weeks to 74.8% at 6 weeks of age, and this age-related difference was even more significant at the bacterial genus level [87]. Prevotella (33.1%) dominated the calf at 2 weeks of age; however, the proportion decreased significantly to 5.1% at 6 weeks of age [87]. These results suggested that the bacterial composition of the rumen in the immediate postnatal period was markedly heterogeneous; however, the composition of gastrointestinal microbes converged in similarity with maturation [90, 134, 139]. The increase in the number of anaerobes in 3-day-old calves indicated the emergence of a new anaerobic environmental niche in the early life of calves [8]. It is certain that these gastrointestinal microbiome changes and their physiological functions mature with the increase of calf age. In the future, the variation in rumen microbiota by age, the origin of these gut microbes, and the route of transfer to a newborn calf need to be studied further.

Genotype

Genotype might also be an important factor that influenced changes in the rumen microbial community in ruminants [140, 141]. Differences in genotypes may influence the selection of certain species by affecting individual metabolic or physiological mechanisms. The effect of genotype on intestinal bacteria has recently been confirmed in pigs. The genotype significantly affected the abundance of Erysipelotrichaceae in the gut of pigs by regulating the concentration of N-acetyl-galactosamine, which indicated that the composition and abundance of intestinal bacteria were inherited to a certain extent [142]. In addition, the microbial community structure of twin calves under the same feeding conditions was the same according to metagenomics, and this structure was determined by the genotype of the host [115]. The diversity and community structure of intestinal anaerobic fungi in ruminants and non-ruminants were different, which indicated that different host genotypes have different microbial communities [116].

Interactions between the gut microbiome and host immunity

A large number of microbiota in the intestine have an important impact on calf growth. The richness and activity of these gut microbiota are influenced by various factors, which include the intestinal environment, nutritional level, and health conditions. Therefore, there is an interaction exists between the gut microbiome and calf health.

The gut microbiome can maintain normal physiological activity and functions of the immune system in animals, which also affect the central nervous system. As discussed above, the dominant microbiomes in different GIT regions are significantly different, which leads to regional differences in the microbiome that may in turn affect immune function in different regions of the GIT that include the development and maturation of the mucosal immune system [140]. IgA, particularly secretory IgA (SIgA) is an important component of the intestinal mucosal immune system in animals. Most pathogenic bacteria release toxins by adhering to luminal epithelial cells, and SIgA can screen bacteria that are beneficial to the host by coating bacteria or eliminating harmful bacteria. However, owing to the special placental structure of ruminants, IgA cannot pass directly from the mother to the fetus, except through the colostrum. This means that newborn calves acquires IgA by consuming colostrum, and then IgA selects beneficial microbiota to colonize the gut, which is a host behavior for selecting for intestinal microbiota. However, when the host immune system has matured over time and produced sufficient IgA, the production of IgA and the ability to coat bacteria were regulated by bacterial metabolites and cytokines, such as acetate and TGF-β [21, 143].

In addition to screening colonizers using IgA, the host also achieves similar effects through the release of miRNAs, a group of small endogenous RNA molecules that are indispensable for regulating rumen development in newborn calves. They bind to coding RNA, block the translation process to regulate gene expression, and influence the integrity of the host immune function. miRNAs related to bacterial density were regulators of lymphoid tissue development that regulated the maturation of dendritic cells and development of immune cells [144]. Specific miRNAs exist in the calf GIT. Specifically, the expression levels of miR-l5/16, miR-29, and miR-196 were correlated positively with the 16S rRNA gene copy numbers in Bifidobacterium and Lactobacillus [144]. High concentrations of miRNAs were found in colostrum, which indicated that miRNAs also acted as signaling molecules that were transmitted from mothers to calves to stimulate intestinal epithelial cell proliferation, stem cell activity, and development of the immune system [145,146,147]. These results suggested that the gut microbiome mediated gene expression by regulating the expression level of miRNAs and then regulated the development of immune function in the early life of the calves.

The immune system of the calves can actively select the microbiota colonized in the intestine, and stimulation of the exogenous microbiota is essential for the development of the immune system of newborn calves. In particular, FMT is an effective treatment for diarrhea in young animals [41]. Owing to the different protocols, results may also be different. FMT increased haptoglobin and paraoxonase levels in the serum of calves; however, another study showed that FMT aggravated gastrointestinal diseases in calves [69, 148]. Therefore, screening high-quality donor feces and standardizing FMT procedures may be the key to the success of FMT. Evidence for the remission of diarrhea in calves by FMT suggested that successful FMT alleviated diarrheic symptoms by altering the composition and metabolites of microbiota, some of which were used as biomarkers to evaluate the effect of FMT, such as Sporobacter and Selenomonas in donors and Lactobacillus in recipients [32, 47, 149]. However, it remains unclear which microbial species play a decisive role in the remission of symptoms in calves with diarrhea and their specific mechanisms.

Conclusions and further perspectives

The colonization of the gut microbiota (e.g., bacteria, archaea, fungi, protozoa, and viruses) in the early life of calves has attracted much attention. The gut microbiota, with nutritional and immune functions, plays a vital role in the behavior, immune system, growth, and rumen fermentation of calves. The composition, diversity, and richness of gut microbiota vary with age, species, diet, probiotics, sampling location (i.e., contents, mucous membrane, and feces), and gut segment (i.e., rumen and large and small intestines) of calves. There is an interaction between the gut microbiome (metabolites) and the calves (i.e., breed, immune system, and development). However, the causal relationship between the gut microbiome and biological health, their contribution to phenotypic variations, and the short- and long-term effects of microbial regulation remain unclear and require further study. The core gut microbiota that links host genetics (breeds) and phenotypes (e.g., methane emissions, gastrointestinal development, feed conversion rate, and milk production efficiency) also requires further exploration.

Availability of data and materials

Not applicable.

Abbreviations

AA:

Amino acid

BCAA:

Branched-chain amino acid

FMT:

Fecal microbiota transplantation

GIT:

Gastrointestinal tract

IgA:

Immunoglobulin A

PUFA:

Polyunsaturated fatty acid

SCFAs:

Short-chain fatty acids

SIgA:

Secretory immunoglobulin A

TLR:

Toll-like receptor

References

  1. Myer PR, Freetly HC, Wells JE, Smith TPL, Kuehn LA. Analysis of the gut bacterial communities in beef cattle and their association with feed intake, growth, and efficiency. J Anim Sci. 2017;95(7):3215–24. https://doi.org/10.2527/jas.2016.1059.

    Article  CAS  PubMed  Google Scholar 

  2. Malmuthuge N, Guan LL. Understanding the gut microbiome of dairy calves: Opportunities to improve early-life gut health. J Dairy Sci. 2017;100(7):5996–6005. https://doi.org/10.3168/jds.2016-12239.

    Article  CAS  PubMed  Google Scholar 

  3. Bergstrom A, Skov TH, Bahl MI, Roager HM, Christensen LB, Ejlerskov KT, et al. Establishment of intestinal microbiota during early life: a longitudinal, explorative study of a large cohort of danish infants. Appl Environ Microbiol. 2014;80(9):2889–900. https://doi.org/10.1128/aem.00342-14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Tanaka M, Nakayama J. Development of the gut microbiota in infancy and its impact on health in later life. Allergol Int. 2017;66(4):515–22. https://doi.org/10.1016/j.alit.2017.07.010

  5. Xie B, Huang WQ, Zhang CX, Diao QY, Cui K, Chai JM, et al. Influences of starter ndf level on growth performance and rumen development in lambs fed isocaloric and isonitrogenous diets. J Anim Sci. 2020;98(4):skaa093. https://doi.org/10.1093/jas/skaa093.

  6. Eckert E, Brown HE, Leslie KE, DeVries TJ, Steele MA. Weaning age affects growth, feed intake, gastrointestinal development, and behavior in holstein calves fed an elevated plane of nutrition during the preweaning stage. J Dairy Sci. 2015;98(9):6315–26. https://doi.org/10.3168/jds.2014-9062.

    Article  CAS  PubMed  Google Scholar 

  7. Lv XK, Chai JM, Diao QY, Huang WQ, Zhuang YM, Zhang NF. The signature microbiota drive rumen function shifts in goat kids introduced to solid diet regimes. Microorganisms. 2019;7(11):516. https://doi.org/10.3390/microorganisms7110516.

  8. Jami E, Israel A, Kotser A, Mizrahi I. Exploring the bovine rumen bacterial community from birth to adulthood. ISME J. 2013;7(6):1069–79. https://doi.org/10.1038/ismej.2013.2.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Bi YL, Cox MS, Zhang F, Suen G, Zhang NF, Tu Y, et al. Feeding modes shape the acquisition and structure of the initial gut microbiota in newborn lambs. Environ Microbiol. 2019;21(7):2333–46. https://doi.org/10.1111/1462-2920.14614.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Buford TW. (Dis)Trust your gut: the gut microbiome in age-related inflammation, health, and disease. Microbiome. 2017;5:80. https://doi.org/10.1186/s40168-017-0296-0.

  11. Wallace RJ, Sasson G, Garnsworthy PC, Tapio I, Gregson E, Bani P, et al. A heritable subset of the core rumen microbiome dictates dairy cow productivity and emissions. Sci Adv. 2019;5(7):eaav8391. https://doi.org/10.1126/sciadv.aav8391.

  12. Dias J, Marcondes MI, de Souza SM, Silva B, Noronha MF, Resende RT, et al. Bacterial community dynamics across the gastrointestinal tracts of dairy calves during preweaning development. Appl Environ Microbiol. 2018;84(9):02675-17. https://doi.org/10.1128/aem.02675-17.

  13. Troscher-Mussotter J, Saenz JS, Grindler S, Meyer J, Kononov SU, Mezger B, et al. Microbiome clusters disclose physiologic variances in dairy cows challenged by calving and lipopolysaccharides. Msystems. 2021;6(5):e00856-21. https://doi.org/10.1128/mSystems.00856-21.

  14. Welch CB, Ryman VE, Pringle TD, Lourenco JM. Utilizing the gastrointestinal microbiota to modulate cattle health through the microbiome-gut-organ axes. Microorganisms. 2022;10(7):1391. https://doi.org/10.3390/microorganisms10071391.

  15. Bronzo V, Lopreiato V, Riva F, Amadori M, Curone G, Addis MF, et al. The role of innate immune response and microbiome in resilience of dairy cattle to disease: the mastitis model. Animals. 2020;10(8):1397. https://doi.org/10.3390/ani10081397.

  16. Scarsella E, Zecconi A, Cintio M, Stefanon B. Characterization of microbiome on feces, blood and milk in dairy cows with different milk leucocyte pattern. Animals. 2021;11(5):1463. https://doi.org/10.3390/ani11051463.

  17. Malekkhahi M, Tahmasbi AM, Naserian AA, Danesh-Mesgaran M, Kleen JL, AlZahal O, et al. Effects of supplementation of active dried yeast and malate during sub-acute ruminal acidosis on rumen fermentation, microbial population, selected blood metabolites, and milk production in dairy cows. Anim Feed Sci Technol. 2016;213:29–43. https://doi.org/10.1016/j.anifeedsci.2015.12.018.

    Article  CAS  Google Scholar 

  18. Rubino F, Carberry C, Waters SM, Kenny D, McCabe MS, Creevey CJ. Divergent functional isoforms drive niche specialisation for nutrient acquisition and use in rumen microbiome. ISME J. 2017;11(4):932–44. https://doi.org/10.1038/ismej.2016.172.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Jewell KA, McCormick CA, Odt CL, Weimer PJ, Suen G. Ruminal bacterial community composition in dairy cows is dynamic over the course of two lactations and correlates with feed efficiency. Appl Environ Microbiol. 2015;81(14):4697–710. https://doi.org/10.1128/aem.00720-15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Li FY, Guan LL. Metatranscriptomic profiling reveals linkages between the active rumen microbiome and feed efficiency in beef cattle. Appl Environ Microbiol. 2017;83(9):e00061-17. https://doi.org/10.1128/aem.00061-17.

  21. Takeuchi T, Miyauchi E, Kanaya T, Kato T, Nakanishi Y, Watanabe T, et al. Acetate differentially regulates iga reactivity to commensal bacteria. Nature. 2021;595:560–4. https://doi.org/10.1038/s41586-021-03727-5.

    Article  CAS  PubMed  Google Scholar 

  22. Zhan K, Yang TY, Chen YY, Jiang MC, Zhao GQ. Propionate enhances the expression of key genes involved in the gluconeogenic pathway in bovine intestinal epithelial cells. J Dairy Sci. 2020;103(6):5514–24. https://doi.org/10.3168/jds.2019-17309.

    Article  CAS  PubMed  Google Scholar 

  23. Giesecke D, Beck U, Wiesmayr S, Stangassinger M. The effect of rumen epithelial development on metabolic activities and ketogenesis by the tissue in vitro. Comp Biochem Physiol B Biochem Mol Biol. 1979;62(4):459–63. https://doi.org/10.1016/0305-0491(79)90118-4.

    Article  CAS  Google Scholar 

  24. Liu JH, Xu TT, Zhu WY, Mao SY. High-grain feeding alters caecal bacterial microbiota composition and fermentation and results in caecal mucosal injury in goats. Br J Nutr. 2014;112(3):416–27. https://doi.org/10.1017/s0007114514000993.

    Article  CAS  PubMed  Google Scholar 

  25. Foditsch C, Pereira RV, Ganda EK, Gomez MS, Marques EC, Santin T, et al. Oral administration of faecalibacterium prausnitzii decreased the incidence of severe diarrhea and related mortality rate and increased weight gain in preweaned dairy heifers. PLos One. 2015;10(12):e0145485. https://doi.org/10.1371/journal.pone.0145485.

  26. Fukumori R, Izumi K, Oikawa S, Oba M. Effects of butyrate supplementation on blood glucagon-like peptide-2 concentration and gastrointestinal function in lactating dairy cows fed diets differing in starch content. J Dairy Sci. 2019;102:212. https://doi.org/10.3168/jds.2019-17677.

    Article  CAS  Google Scholar 

  27. Gao GZ, Zhou JR, Wang HQ, Ding YA, Zhou JW, Chong PH, et al. Effects of valerate on intestinal barrier function in cultured caco-2 epithelial cell monolayers. Mol Biol Rep. 2022;49(3):1817–25. https://doi.org/10.1007/s11033-021-06991-w.

    Article  CAS  PubMed  Google Scholar 

  28. Mei FF, Duan ZW, Chen MX, Lu JF, Zhao MH, Li LH, et al. Effect of a high-collagen peptide diet on the gut microbiota and short-chain fatty acid metabolism. J Funct Foods. 2020;75:104278. https://doi.org/10.1016/j.jff.2020.104278.

  29. Zhang WX, Bao CL, Wang J, Zang JJ, Cao YH. Administration of saccharomyces boulardii mafic-1701 improves feed conversion ratio, promotes antioxidant capacity, alleviates intestinal inflammation and modulates gut microbiota in weaned piglets. J Anim Sci Biotechnol. 2020;11:112. https://doi.org/10.1186/s40104-020-00516-4.

  30. Krautkramer KA, Fan J, Backhed F. Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol. 2021;19(2):77–94. https://doi.org/10.1038/s41579-020-0438-4.

    Article  CAS  PubMed  Google Scholar 

  31. Li JY, Suzuki K, Koike Y, Chen DS, Yonezawa T, Nishihara M, et al. Effects of dietary supplementation with branched-chain amino acids (bcaas) during nursing on plasma bcaa levels and subsequent growth in cattle. Asian-australas J Anim Sci. 2005;18(10):1440–4. https://doi.org/10.5713/ajas.2005.1440.

    Article  CAS  Google Scholar 

  32. Kim HS, Whon TW, Sung H, Jeong YS, Jung ES, Shin NR, et al. Longitudinal evaluation of fecal microbiota transplantation for ameliorating calf diarrhea and improving growth performance. Nat Commun. 2021;12:161. https://doi.org/10.1038/s41467-020-20389-5.

  33. Tsukano K, Suzuki K. Plasma amino acid abnormalities in calves with diarrhea. J Vet Med Sci. 2019;81(4):517–21. https://doi.org/10.1292/jvms.18-0645.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tsukano K, Lakritz J, Suzuki K. Plasma amino acid status is useful for understanding intestinal mucosal damage in calves with cryptosporidiosis. Amino Acids. 2020;52(10):1459–64. https://doi.org/10.1007/s00726-020-02904-6.

    Article  CAS  PubMed  Google Scholar 

  35. Platten M, Nollen EAA, Rohrig UF, Fallarino F, Opitz CA. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov. 2019;18(5):379–401. https://doi.org/10.1038/s41573-019-0016-5.

    Article  CAS  PubMed  Google Scholar 

  36. Lamas B, Natividad JM, Sokol H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal Immunol. 2018;11(4):1024–38. https://doi.org/10.1038/s41385-018-0019-2.

    Article  CAS  PubMed  Google Scholar 

  37. Fang ZF, Pan T, Wang HC, Zhu JL, Zhang H, Zhao JX, et al. Limosilactobacillus reuteri attenuates atopic dermatitis via changes in gut bacteria and indole derivatives from tryptophan metabolism. Int J Mol Sci. 2022;23(14):7735. https://doi.org/10.3390/ijms23147735.

  38. Zhang ZJ, Mu XH, Cao QN, Shi Y, Hu XS, Zheng H. Honeybee gut lactobacillus modulates host learning and memory behaviors via regulating tryptophan metabolism. Nat Commun. 2022;13:2037. https://doi.org/10.1038/s41467-022-29760-0.

  39. Hegarty JW, Guinane CM, Ross RP, Hill C, Cotter PD. Bacteriocin production: A relatively unharnessed probiotic trait? F1000Research. 2016;5:2587. https://doi.org/10.12688/f1000research.9615.1

  40. Soltani S, Hammami R, Cotter PD, Rebuffat S, Ben Said L, Gaudreau H, et al. Bacteriocins as a new generation of antimicrobials: toxicity aspects and regulations. FEMS Microbiol Rev. 2021;45(1):fuaa039. https://doi.org/10.1093/femsre/fuaa039.

  41. Hu J, Ma LB, Nie YF, Chen JW, Zheng WY, Wang XK, et al. A microbiota-derived bacteriocin targets the host to confer diarrhea resistance in early-weaned piglets. Cell Host Microbe. 2018;24(6):817–32. https://doi.org/10.1016/j.chom.2018.11.006.

    Article  CAS  PubMed  Google Scholar 

  42. Twomey DP, Wheelock AI, Flynn J, Meaney WJ, Hill C, Ross RP. Protection against staphylococcus aureus mastitis in dairy cows using a bismuth-based teat seal containing the bacteriocin, lacticin 3147. J Dairy Sci. 2000;83(9):1981–8. https://doi.org/10.3168/jds.S0022-0302(00)75075-2.

    Article  CAS  PubMed  Google Scholar 

  43. Mohammed AD, Mohammed Z, Roland MM, Chatzistamou I, Jolly A, Schoettmer LM, et al. Defective humoral immunity disrupts bile acid homeostasis which promotes inflammatory disease of the small bowel. Nat Commun. 2022;13:525. https://doi.org/10.1038/s41467-022-28126-w.

  44. Ramirez-Perez O, Cruz-Ramon V, Chinchilla-Lopez P, Mendez-Sanchez N. The role of the gut microbiota in bile acid metabolism. Ann Hepatol. 2017;16(Suppl. 1):S15–20. https://doi.org/10.5604/01.3001.0010.5494

  45. Zhang Y, Xie HB, Wang LR, Hu JH, Wang L, Zhang SP. Effect of weaning at 21 days of age on the content of bile acids in chyme of cecum. Animals. 2022;12(16):2138. https://doi.org/10.3390/ani12162138.

  46. Apajalahti J, Vienola K, Raatikainen K, Kettunen H, Vuorenmaa J. Distribution, metabolism, and recovery of resin acids in the intestine and tissues of broiler chickens in a feeding trial with tall oil fatty acid-supplemented diets. Front Vet Sci. 2020;7:437. https://doi.org/10.3389/fvets.2020.00437.

  47. He ZY, Ma YL, Yang SR, Zhang SY, Liu S, Xiao JX, et al. Gut microbiota-derived ursodeoxycholic acid from neonatal dairy calves improves intestinal homeostasis and colitis to attenuate extended-spectrum beta-lactamase-producing enteroaggregative Escherichia coli infection. Microbiome. 2022;10:79. https://doi.org/10.1186/s40168-022-01269-0.

  48. Ost KS, O’Meara TR, Stephens WZ, Chiaro T, Zhou HY, Penman J, et al. Adaptive immunity induces mutualism between commensal eukaryotes. Nature. 2021;596:114–8. https://doi.org/10.1038/s41586-021-03722-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gopalakrishna KP, Macadangdang BR, Rogers MB, Tometich JT, Firek BA, Baker R, et al. Maternal iga protects against the development of necrotizing enterocolitis in preterm infants. Nat Med. 2019;25(7):1110–U233. https://doi.org/10.1038/s41591-019-0480-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Melo-Gonzalez F, Kammoun H, Evren E, Dutton EE, Papadopoulou M, Bradford BM, et al. Antigen-presenting ilc3 regulate t cell-dependent iga responses to colonic mucosal bacteria. J Exp Med. 2019;216(4):728–42. https://doi.org/10.1084/jem.20180871.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Macpherson AJ, Koller Y, McCoy KD. The bilateral responsiveness between intestinal microbes and iga. Trends Immunol. 2015;36(8): 460–70. https://doi.org/10.1016/j.it.2015.06.006

  52. He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J, et al. Intestinal bacteria trigger t cell-independent immunoglobulin a(2) class switching by inducing epithelial-cell secretion of the cytokine april. Immunity. 2007;26(6):812–26. https://doi.org/10.1016/j.immuni.2007.04.014.

    Article  CAS  PubMed  Google Scholar 

  53. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Reddy DN. Role of the normal gut microbiota. World J Gastroenterol. 2015;21(29):8787–803. https://doi.org/10.3748/wjg.v21.i29.8787.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ali ZI, Saudi AM, Albrecht R, Talaat AM. The inhibitory effect of nisin on Mycobacterium avium ssp. Paratuberculosis and its effect on mycobacterial cell wall. J Dairy Sci. 2019;102(6):4935–44. https://doi.org/10.3168/jds.2018-16106.

    Article  CAS  PubMed  Google Scholar 

  55. Osorio JS, Trevisi E, Ji P, Drackley JK, Luchini D, Bertoni G, et al. Biomarkers of inflammation, metabolism, and oxidative stress in blood, liver, and milk reveal a better immunometabolic status in peripartal cows supplemented with smartamine m or metasmart. J Dairy Sci. 2014;97(12):7437–50. https://doi.org/10.3168/jds.2013-7679.

    Article  CAS  PubMed  Google Scholar 

  56. Rosa F, Busato S, Avaroma FC, Linville K, Trevisi E, Osorio JS, et al. Transcriptional changes detected in fecal rna of neonatal dairy calves undergoing a mild diarrhea are associated with inflammatory biomarkers. PLos One. 2018;13(1):e0191599. https://doi.org/10.1371/journal.pone.0191599.

  57. Spits H, Di Santo JP. The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nat Immunol. 2011;12(1):21–7. https://doi.org/10.1038/ni.1962.

    Article  CAS  PubMed  Google Scholar 

  58. Cario E, Gerken G, Podolsky DK. Toll-like receptor 2 controls mucosal inflammation by regulating epithelial barrier function. Gastroenterology. 2007;132(4):1359–74. https://doi.org/10.1053/j.gastro.2007.02.056.

    Article  CAS  PubMed  Google Scholar 

  59. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving glp-2-driven improvement of gut permeability. Gut. 2009;58(8):1091–103. https://doi.org/10.1136/gut.2008.165886.

    Article  CAS  PubMed  Google Scholar 

  60. Stappenbeck TS, Hooper LV, Gordon JI. Developmental regulation of intestinal angiogenesis by indigenous microbes via paneth cells. Proc Natl Acad Sci USA. 2002;99(24):15451–5. https://doi.org/10.1073/pnas.202604299.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Martel J, Chang SH, Ko YF, Hwang TL, Young JD, Ojcius DM. Gut barrier disruption and chronic disease. Trends Endocrinol Metab. 2022;33(4):247–65. https://doi.org/10.3920/BM2012.0065.

    Article  CAS  PubMed  Google Scholar 

  62. Paone P, Cani PD. Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut. 2020;69(12):2232–43. https://doi.org/10.1136/gutjnl-2020-322260.

    Article  CAS  PubMed  Google Scholar 

  63. Schoultz I, Keita ÃV. The intestinal barrier and current techniques for the assessment of gut permeability. Cells. 2020;9(8):1909. https://doi.org/10.3390/cells9081909.

  64. Geurts L, Neyrinck AM, Delzenne NM, Knauf C, Cani PD. Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and interventions using prebiotics. Benef Microbes. 2014;5(1):3–17. https://doi.org/10.3920/bm2012.0065.

    Article  CAS  PubMed  Google Scholar 

  65. Liu B, Wang CJ, Huasai S, Han A, Zhang J, He LN, et al. Compound probiotics improve the diarrhea rate and intestinal microbiota of newborn calves. Animals. 2022;12(3):322. https://doi.org/10.3390/ani12030322.

  66. Redding LE, Berry AS, Indugu N, Huang E, Beiting DP, Pitta D. Gut microbiota features associated with clostridioides difficile colonization in dairy calves. PLos One. 2021;16(12):e0251999. https://doi.org/10.1371/journal.pone.0251999.

  67. Cho YI, Yoon KJ. An overview of calf diarrhea - infectious etiology, diagnosis, and intervention. J Vet Sci. 2014;15(1):1–17. https://doi.org/10.4142/jvs.2014.15.1.1.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Gomez DE, Li L, Goetz H, MacNicol J, Gamsjaeger L, Renaud DL. Calf diarrhea is associated with a shift from obligated to facultative anaerobes and expansion of lactate-producing bacteria. Front Vet Sci. 2022;9:846383. https://doi.org/10.3389/fvets.2022.846383.

  69. Slanzon GS, Ridenhour BJ, Moore DA, Sischo WM, Parrish LM, Trombetta SC, et al. Fecal microbiome profiles of neonatal dairy calves with varying severities of gastrointestinal disease. PLos One. 2022;17(1):e0262317. https://doi.org/10.1371/journal.pone.0262317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ma T, Villot C, Renaud D, Skidmore A, Chevaux E, Steele M, et al. Linking perturbations to temporal changes in diversity, stability, and compositions of neonatal calf gut microbiota: prediction of diarrhea. ISME J. 2020;14(9):2223–35. https://doi.org/10.1038/s41396-020-0678-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Jang JY, Kim S, Kwon MS, Lee J, Yu DH, Song RH, et al. Rotavirus-mediated alteration of gut microbiota and its correlation with physiological characteristics in neonatal calves. J Microbiol. 2019;57(2):113–21. https://doi.org/10.1007/s12275-019-8549-1.

    Article  CAS  PubMed  Google Scholar 

  72. Bristol LS, Duhamel GE, Zinckgraf JW, Crabb JH, Nydam DV. Effect of passive antibodies derived from rotavirus-like particles on neonatal calf diarrhea caused by rotavirus in an oral challenge model. J Dairy Sci. 2021;104(11):11922–30. https://doi.org/10.3168/jds.2020-19834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Viidu DA, Motus K. Implementation of a pre-calving vaccination programme against rotavirus, coronavirus and enterotoxigenic Escherichia coli (F5) and association with dairy calf survival. BMC Vet Res. 2022;18:59. https://doi.org/10.1186/s12917-022-03154-2.

  74. Amin N, Seifert J. Dynamic progression of the calf’s microbiome and its influence on host health. Comput Struct Biotechnol. 2021;19:989–1001. https://doi.org/10.1016/j.csbj.2021.01.035.

    Article  CAS  Google Scholar 

  75. Sprockett D, Fukami T, Relman DA. Role of priority effects in the early-life assembly of the gut microbiota. Nat Rev Gastroenterol Hepatol. 2018;15(4):197–205. https://doi.org/10.1038/nrgastro.2017.173.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Vlkova E, Trojanova I, Rada V. Distribution of bifidobacteria in the gastrointestinal tract of calves. Folia Microbiol (Praha). 2006;51(4):325–8. https://doi.org/10.1007/bf02931825.

    Article  CAS  PubMed  Google Scholar 

  77. Bi YL, Tu Y, Zhang NF, Wang SQ, Zhang F, Suen G, et al. Multiomics analysis reveals the presence of a microbiome in the gut of fetal lambs. Gut. 2021;70(5):853–64. https://doi.org/10.1136/gutjnl-2020-320951.

    Article  CAS  PubMed  Google Scholar 

  78. Guzman CE, Wood JL, Egidi E, White-Monsant AC, Semenec L, Grommen SVH, et al. A pioneer calf foetus microbiome. Sci Rep. 2020;10(1):17712. https://doi.org/10.1038/s41598-020-74677-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Alipour MJ, Jalanka J, Pessa-Morikawa T, Kokkonen T, Satokari R, Hynonen U, et al. The composition of the perinatal intestinal microbiota in cattle. Sci Rep. 2018;8(1):10437. https://doi.org/10.1038/s41598-018-28733-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Elolimy A, Alharthi A, Zeineldin M, Parys C, Helmbrecht A, Loor JJ. Supply of methionine during late-pregnancy alters fecal microbiota and metabolome in neonatal dairy calves without changes in daily feed intake. Front Microbiol. 2019;10:2159. https://doi.org/10.3389/fmicb.2019.02159.

  81. Seferovic MD, Pace RM, Carroll M, Belfort B, Major AM, Chu DM, et al. Visualization of microbes by 16s in situ hybridization in term and preterm placentas without intraamniotic infection. Am J Obstet Gynecol. 2019;221(2):146.E1–23. https://doi.org/10.1016/j.ajog.2019.04.036.

  82. Malmuthuge N, Liang GX, Guan LL. Regulation of rumen development in neonatal ruminants through microbial metagenomes and host transcriptomes. Genome Biol. 2019;20:172. https://doi.org/10.1186/s13059-019-1786-0.

  83. Theis KR, Romero R, Winters AD, Greenberg JM, Gomez-Lopez N, Alhousseini A, et al. Does the human placenta delivered at term have a microbiota? Results of cultivation, quantitative real-time pcr, 16s rrna gene sequencing, and metagenomics. Am J Obstet Gynecol. 2019;220(3):267.E1–39. https://doi.org/10.1016/j.ajog.2018.10.018.

  84. Arshad MA, Hassan FU, Rehman MS, Huws SA, Cheng YF, Din AU. Gut microbiome colonization and development in neonatal ruminants: strategies, prospects, and opportunities. Anim Nutr. 2021;7(3):883–95. https://doi.org/10.1016/j.aninu.2021.03.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kim ET, Lee SJ, Kim TY, Lee HG, Atikur RM, Gu BH, et al. Dynamic changes in fecal microbial communities of neonatal dairy calves by aging and diarrhea. Animals. 2021;11(4):1113. https://doi.org/10.3390/ani11041113.

  86. Jami E, Mizrahi I. Composition and similarity of bovine rumen microbiota across individual animals. PLoS ONE. 2012;7(3):e0033306. https://doi.org/10.1371/journal.pone.0033306.

  87. Malmuthuge N, Griebel PJ, Guan L. The gut microbiome and its potential role in the development and function of newborn calf gastrointestinal tract. Front Vet Sci. 2015;2:36. https://doi.org/10.3389/fvets.2015.00036.

  88. Schaedler RW, Dubos R, Costello R. The development of the bacterial flora in the gastrointestinal tract of mice. J Exp Med. 1965;122(1):59–66. https://doi.org/10.1084/jem.122.1.59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Uyeno Y, Sekiguchi Y, Kamagata Y. Rrna-based analysis to monitor succession of faecal bacterial communities in holstein calves. Lett Appl Microbiol. 2010;51(5):570–7. https://doi.org/10.1111/j.1472-765X.2010.02937.x.

    Article  CAS  PubMed  Google Scholar 

  90. Malmuthuge N, Griebel PJ, Guan LL. Taxonomic identification of commensal bacteria associated with the mucosa and digesta throughout the gastrointestinal tracts of preweaned calves. Appl Environ Microbiol. 2014;80(6):2021–8. https://doi.org/10.1128/aem.03864-13.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Chen H, Liu Y, Huang K, Yang B, Zhang Y, Yu Z, et al. Fecal microbiota dynamics and its relationship to diarrhea and health in dairy calves. J Anim Sci Biotechnol. 2022;13(1):132. https://doi.org/10.1186/s40104-022-00758-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. de Oliveira MNV, Jewell KA, Freitas FS, Benjamin LA, Totola MR, Borges AC, et al. Characterizing the microbiota across the gastrointestinal tract of a brazilian nelore steer. Vet Microbiol. 2013;164(3–4):307–14. https://doi.org/10.1016/j.vetmic.2013.02.013.

  93. Xie F, Jin W, Si HZ, Yuan Y, Tao Y, Liu JH, et al. An integrated gene catalog and over 10,000 metagenome-assembled genomes from the gastrointestinal microbiome of ruminants. Microbiome. 2021;9:137. https://doi.org/10.1186/s40168-021-01078-x.

  94. Mao SY, Zhang RY, Wang DS, Zhu WY. The diversity of the fecal bacterial community and its relationship with the concentration of volatile fatty acids in the feces during subacute rumen acidosis in dairy cows. BMC Vet Res. 2012;8:237. https://doi.org/10.1186/1746-6148-8-237.

  95. Malmuthuge N, Li MJ, Fries P, Griebel PJ, Guan LL. Regional and age dependent changes in gene expression of toll-like receptors and key antimicrobial defence molecules throughout the gastrointestinal tract of dairy calves. Vet Immunol Immunopathol. 2012;146(1):18–26. https://doi.org/10.1016/j.vetimm.2012.01.010.

    Article  CAS  PubMed  Google Scholar 

  96. Yang M, Zhu H, Gao W, Qu Y. Study on maternal transmission characteristics of hindgut microorganism in newborn holstein calves. Chin J Anim Nutr. 2021;33(11):6445–51.

    CAS  Google Scholar 

  97. Song Y, Malmuthuge N, Steele MA, Guan LL. Shift of hindgut microbiota and microbial short chain fatty acids profiles in dairy calves from birth to pre-weaning. FEMS Microbiol Ecol. 2018;94(3):fix179. https://doi.org/10.1093/femsec/fix179.

  98. Elolimy A, Alharthi A, Zeineldin M, Parys C, Loor JJ. Residual feed intake divergence during the preweaning period is associated with unique hindgut microbiome and metabolome profiles in neonatal holstein heifer calves. J Anim Sci Biotechnol. 2020;11:13. https://doi.org/10.1186/s40104-019-0406-x.

    Article  CAS  Google Scholar 

  99. Chen WG, Liu FL, Ling ZX, Tong XJ, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLos One. 2012;7(6):e0039743. https://doi.org/10.1371/journal.pone.0039743.

  100. Pei LY, Ke YS, Zhao HH, Wang L, Jia C, Liu WZ, et al. Role of colonic microbiota in the pathogenesis of ulcerative colitis. BMC Gastroenterol. 2019;19(1):10. https://doi.org/10.1186/s12876-019-0930-3.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Romano-Keeler J, Moore DJ, Wang C, Brucker RM, Fonnesbeck C, Slaughter JC, et al. Early life establishment of site-specific microbial communities in the gut. Gut Microbes. 2014;5(2):192–201. https://doi.org/10.4161/gmic.28442.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Malmuthuge N, Li MJ, Goonewardene LA, Oba M, Guan LL. Effect of calf starter feeding on gut microbial diversity and expression of genes involved in host immune responses and tight junctions in dairy calves during weaning transition. J Dairy Sci. 2013;96(5):3189–200. https://doi.org/10.3168/jds.2012-6200.

    Article  CAS  PubMed  Google Scholar 

  103. Collado MC, Sanz Y. Quantification of mucosa-adhered microbiota of lambs and calves by the use of culture methods and fluorescent in situ hybridization coupled with flow cytometry techniques. Vet Microbiol. 2007;121(3–4):299–306. https://doi.org/10.1016/j.vetmic.2006.12.006.

    Article  CAS  PubMed  Google Scholar 

  104. Malmuthuge N, Li MJ, Chen YH, Fries P, Griebel PJ, Baurhoo B, et al. Distinct commensal bacteria associated with ingesta and mucosal epithelium in the gastrointestinal tracts of calves and chickens. FEMS Microbiol Ecol. 2012;79(2):337–47. https://doi.org/10.1111/j.1574-6941.2011.01220.x.

    Article  CAS  PubMed  Google Scholar 

  105. Zhao T, Doyle MP, Harmon BG, Brown CA, Mueller POE, Parks AH. Reduction of carriage of enterohemorrhagic Escherichia coli O157:H7 in cattle by inoculation with probiotic bacteria. J Clin Microbiol. 1998;36(3):641–7. https://doi.org/10.1128/jcm.36.3.641-647.1998.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Abe F, Ishibashi N, Shimamura S. Effect of administration of bifidobacteria and lactic acid bacteria to newborn calves and piglets. J Dairy Sci. 1995;78(12):2838–46. https://doi.org/10.3168/jds.S0022-0302(95)76914-4.

    Article  CAS  PubMed  Google Scholar 

  107. Lee YE, Kang IJ, Yu EA, Kim S, Lee HJ. Effect of feeding the combination with lactobacillus plantarum and bacillus subtilis on fecal microflora and diarrhea incidence of korean native calves. Korean J Vet Serv. 2012;354. https://doi.org/10.7853/kjvs.2012.35.4.343.

  108. Khademi AR, Hashemzadeh F, Khorvash M, Mahdavi AH, Pazoki A, Ghaffari MH. Use of exogenous fibrolytic enzymes and probiotic in finely ground starters to improve calf performance. Sci Rep. 2022;12(1):11942. https://doi.org/10.1038/s41598-022-16070-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Fernando SC, Purvis HT, Najar FZ, Sukharnikov LO, Krehbiel CR, Nagaraja TG, et al. Rumen microbial population dynamics during adaptation to a high-grain diet. Appl Environ Microbiol. 2010;76(22):7482–90. https://doi.org/10.1128/aem.00388-10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Thoetkiattikul H, Mhuantong W, Laothanachareon T, Tangphatsornruang S, Pattarajinda V, Eurwilaichitr L, et al. Comparative analysis of microbial profiles in cow rumen fed with different dietary fiber by tagged 16S rRNA gene pyrosequencing. Curr Microbiol. 2013;67(2):130–7. https://doi.org/10.1007/s00284-013-0336-3.

    Article  CAS  PubMed  Google Scholar 

  111. Rico DE, Preston SH, Risser JM, Harvatine KJ. Rapid changes in key ruminal microbial populations during the induction of and recovery from diet-induced milk fat depression in dairy cows. Br J Nutr. 2015;114(3):358–67. https://doi.org/10.1017/s0007114515001865.

    Article  CAS  PubMed  Google Scholar 

  112. Auffret MD, Dewhurst RJ, Duthie CA, Rooke JA, Wallace RJ, Freeman TC, et al. The rumen microbiome as a reservoir of antimicrobial resistance and pathogenicity genes is directly affected by diet in beef cattle. Microbiome. 2017;5:159. https://doi.org/10.1186/s40168-017-0378-z.

  113. Li RW, Connor EE, Li CJ, Baldwin RL, Sparks ME. Characterization of the rumen microbiota of pre-ruminant calves using metagenomic tools. Environ Microbiol. 2012;14(1):129–39. https://doi.org/10.1111/j.1462-2920.2011.02543.x.

    Article  CAS  PubMed  Google Scholar 

  114. Oikonomou G, Teixeira AGV, Foditsch C, Bicalho ML, Machado VS, Bicalho RC. Fecal microbial diversity in pre-weaned dairy calves as described by pyrosequencing of metagenomic 16s rdna. Associations of faecalibacterium species with health and growth. PLos One. 2013;8(4):E0063157. https://doi.org/10.1371/journal.pone.0063157.

  115. Mayer M, Abenthum A, Matthes JM, Kleeberger D, Ege MJ, Holzel C, et al. Development and genetic influence of the rectal bacterial flora of newborn calves. Vet Microbiol. 2012;161(1–2):179–85. https://doi.org/10.1016/j.vetmic.2012.07.023.

  116. Liggenstoffer AS, Youssef NH, Couger MB, Elshahed MS. Phylogenetic diversity and community structure of anaerobic gut fungi (phylum neocallimastigomycota) in ruminant and non-ruminant herbivores. ISME J. 2010;4(10):1225–35. https://doi.org/10.1038/ismej.2010.49.

    Article  PubMed  Google Scholar 

  117. Chaudhary PP, Sirohi SK, Saxena J. Diversity analysis of methanogens in rumen of bubalus bubalis by 16s riboprinting and sequence analysis. Gene. 2012;493(1):13–7. https://doi.org/10.1016/j.gene.2011.11.041.

    Article  CAS  PubMed  Google Scholar 

  118. Paz HA, Anderson CL, Muller MJ, Kononoff PJ, Fernando SC. Rumen bacterial community composition in holstein and jersey cows is different under same dietary condition and is not affected by sampling method. Front Microbiol. 2016;7:1206. https://doi.org/10.3389/fmicb.2016.01206.

  119. Tripathi VK, Sehgal JP, Puniya AK, Singh K. Effect of administration of anaerobic fungi isolated from cattle and wild blue bull (boselaphus tragocamelus) on growth rate and fibre utilization in buffalo calves. Arch Anim Nutr. 2007;61(5):416–23. https://doi.org/10.1080/17450390701556759.

    Article  CAS  PubMed  Google Scholar 

  120. Chaucheyras-Durand F, Walker ND, Bach A. Effects of active dry yeasts on the rumen microbial ecosystem: past, present and future. Anim Feed Sci Technol. 2008;145(1–4):5–26. https://doi.org/10.1016/j.anifeedsci.2007.04.019.

    Article  CAS  Google Scholar 

  121. Sousa DO, Oliveira CA, Velasquez AV, Souza JM, Chevaux E, Mari LJ, et al. Live yeast supplementation improves rumen fibre degradation in cattle grazing tropical pastures throughout the year. Anim Feed Sci Technol. 2018;236:149–58. https://doi.org/10.1016/j.anifeedsci.2017.12.015.

    Article  CAS  Google Scholar 

  122. Ding GZ, Chang Y, Zhao LP, Zhou ZM, Ren LP, Meng QX. Effect of saccharomyces cerevisiae on alfalfa nutrient degradation characteristics and rumen microbial populations of steers fed diets with different concentrate-to-forage ratios. J Anim Sci Biotechnol. 2014;5:24. https://doi.org/10.1186/2049-1891-5-24.

  123. Callaway ES, Martin SA. Effects of a saccharomyces cerevisiae culture on ruminal bacteria that utilize lactate and digest cellulose. J Dairy Sci. 1997;80(9):2035–44. https://doi.org/10.3168/jds.S0022-0302(97)76148-4.

    Article  CAS  PubMed  Google Scholar 

  124. Liou MJ, Miller BM, Litvak Y, Nguyen H, Natwick DE, Savage HP, et al. Host cells subdivide nutrient niches into discrete biogeographical microhabitats for gut microbes. Cell Host Microbe. 2022;30(6):836–47.e6. https://doi.org/10.1016/j.chom.2022.04.012.

  125. Liu JX, Taft DH, Maldonado-Gomez MX, Johnson D, Treiber ML, LemayQ DG, et al. The fecal resistome of dairy cattle is associated with diet during nursing. Nat Commun. 2019;10:4406. https://doi.org/10.1038/s41467-019-12111-x.

  126. Al-Qumber M, Tagg JR. Commensal bacilli inhibitory to mastitis pathogens isolated from the udder microbiota of healthy cows. J Appl Microbiol. 2006;101(5):1152–60. https://doi.org/10.1111/j.1365-2672.2006.03004.x.

    Article  CAS  PubMed  Google Scholar 

  127. Hibbing ME, Fuqua C, Parsek MR, Peterson SB. Bacterial competition: surviving and thriving in the microbial jungle. Nat Rev Microbiol. 2010;8(1):15–25. https://doi.org/10.1038/nrmicro2259.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Fan P, Kim M, Liu G, Zhai Y, Liu T, Driver JD, et al. The gut microbiota of neworn calves and influence of potential probiotics on reducing diarrheic disease by inhibition of pathogen colonization. Front Microbiol. 2021;12:772863. https://doi.org/10.3389/fmicb.2021.772863.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Fu Y, Diao Q, Tu Y, Wang J, Xu X. Effects of different combinations of probiotics on growth performance and serum biochemical parameters in dairy calves aged from 0 to 8 weeks. Chin J Anim Nutr. 2012;24(4):753–61.

    CAS  Google Scholar 

  130. Bayatkouhsar J, Tahmasebi AM, Naserian AA, Mokarram RR, Valizadeh R. Effects of supplementation of lactic acid bacteria on growth performance, blood metabolites and fecal coliform and lactobacilli of young dairy calves. Anim Feed Sci Technol. 2013;186(1–2):1–11. https://doi.org/10.1016/j.anifeedsci.2013.04.015.

    Article  CAS  Google Scholar 

  131. Wang HB, Yu ZT, Gao ZB, Li QW, Qiu XJ, Wu F, et al. Effects of compound probiotics on growth performance, rumen fermentation, blood parameters, and health status of neonatal holstein calves. J Dairy Sci. 2022;105(3):2190–200. https://doi.org/10.3168/jds.2021-20721.

    Article  CAS  PubMed  Google Scholar 

  132. Alawneh JI, Barreto MO, Moore RJ, Soust M, Al-harbi H, James AS, et al. Systematic review of an intervention: the use of probiotics to improve health and productivity of calves. Prev Vet Med. 2020;183:105147. https://doi.org/10.1016/j.prevetmed.2020.105147.

  133. Meale SJ, Li S, Azevedo P, Derakhshani H, Plaizier JC, Khafipour E, et al. Development of ruminal and fecal microbiomes are affected by weaning but not weaning strategy in dairy calves. Front Microbiol. 2016;7:582. https://doi.org/10.3389/fmicb.2016.00582.

    Article  PubMed  PubMed Central  Google Scholar 

  134. De Barbieri I, Hegarty RS, Silveira C, Gulino LM, Oddy VH, Gilbert RA, et al. Programming rumen bacterial communities in newborn merino lambs. Small Rumin Res. 2015;129:48–59. https://doi.org/10.1016/j.smallrumres.2015.05.015.

    Article  Google Scholar 

  135. Abecia L, Waddams KE, Martinez-Fernandez G, Martin-Garcia AI, Ramos-Morales E, Newbold CJ et al. An antimethanogenic nutritional intervention in early life of ruminants modifies ruminal colonization by archaea. Archaea. 2014;2014:841463. https://doi.org/10.1155/2014/841463

  136. Abecia L, Martin-Garcia AI, Martinez G, Newbold CJ, Yanez-Ruiz DR. Nutritional intervention in early life to manipulate rumen microbial colonization and methane output by kid goats postweaning. J Anim Sci. 2013;91(10):4832–40. https://doi.org/10.2527/jas.2012-6142.

    Article  CAS  PubMed  Google Scholar 

  137. Lin XY, Wang J, Hou QL, Wang Y, Hu ZY, Shi KR, et al. Effect of hay supplementation timing on rumen microbiota in suckling calves. Microbiologyopen. 2018;7(1):e00430. https://doi.org/10.1002/mbo3.430.

  138. Petri RM, Munnich M, Zebeli Q, Klevenhusen F. Graded replacement of corn grain with molassed sugar beet pulp modulates the fecal microbial community and hindgut fermentation profile in lactating dairy cows. J Dairy Sci. 2019;102(6):5019–30. https://doi.org/10.3168/jds.2018-15704.

    Article  CAS  PubMed  Google Scholar 

  139. Rey M, Enjalbert F, Combes S, Cauquil L, Bouchez O, Monteils V. Establishment of ruminal bacterial community in dairy calves from birth to weaning is sequential. J Appl Microbiol. 2014;116(2):245–57. https://doi.org/10.1111/jam.12405.

    Article  CAS  PubMed  Google Scholar 

  140. Taschuk R, Griebel PJ. Commensal microbiome effects on mucosal immune system development in the ruminant gastrointestinal tract. Anim Health Res Rev. 2012;13(1):129–41. https://doi.org/10.1017/s1466252312000096.

    Article  PubMed  Google Scholar 

  141. Zhou M, Chen YH, Griebel PJ, Guan LL. Methanogen prevalence throughout the gastrointestinal tract of pre-weaned dairy calves. Gut Microbes. 2014;5(5):628–38. https://doi.org/10.4161/19490976.2014.969649.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Yang H, Wu JY, Huang XC, Zhou YY, Zhang YF, Liu M, et al. Abo genotype alters the gut microbiota by regulating galnac levels in pigs. Nature. 2022;606:358–67. https://doi.org/10.1038/s41586-022-04769-z

  143. Thurnheer MC, Zuercher AW, Cebra JJ, Bos NA. B1 cells contribute to serum igm, but not to intestinal iga, production in gnotobiotic ig allotype chimeric mice. J Immunol. 2003;170(9):4564–71. https://doi.org/10.4049/jimmunol.170.9.4564.

    Article  CAS  PubMed  Google Scholar 

  144. Liang GX, Malmuthuge N, McFadden TB, Bao H, Griebel PJ, Stothard P, et al. Potential regulatory role of micrornas in the development of bovine gastrointestinal tract during early life. PLos One. 2014;9(3):e0092592. https://doi.org/10.1371/journal.pone.0092592.

  145. Van Hese I, Goossens K, Vandaele L, Opsomer G. Invited review: Micrornas in bovine colostrum-focus on their origin and potential health benefits for the calf. J Dairy Sci. 2020;103(1):1–15. https://doi.org/10.3168/jds.2019-16959.

    Article  CAS  PubMed  Google Scholar 

  146. Alsaweed M, Hartmann PE, Geddes DT, Kakulas F. Micrornas in breastmilk and the lactating breast: potential immunoprotectors and developmental regulators for the infant and the mother. Int J Env Res Public Health. 2015;12(11):13981–4020. https://doi.org/10.3390/ijerph121113981.

    Article  CAS  Google Scholar 

  147. Alsaweed M, Lai CT, Hartmann PE, Geddes DT, Kakulas F. Human milk mirnas primarily originate from the mammary gland resulting in unique mirna profiles of fractionated milk. Sci Rep. 2016;6:20680. https://doi.org/10.1038/srep20680.

  148. Rosa F, Michelotti TC, St-Pierre B, Trevisi E, Osorio JS. Early life fecal microbiota transplantation in neonatal dairy calves promotes growth performance and alleviates inflammation and oxidative stress during weaning. Animals. 2021;11(9):2704. https://doi.org/10.3390/ani11092704.

  149. Islam J, Tanimizu M, Shimizu Y, Goto Y, Ohtani N, Sugiyama K, et al. Development of a rational framework for the therapeutic efficacy of fecal microbiota transplantation for calf diarrhea treatment. Microbiome. 2022;10:31. https://doi.org/10.1186/s40168-021-01217-4.

Download references

Acknowledgements

We would like to thank Thomas A. Gavin, Professor Emeritus, Cornell University, for English language editing of this manuscript.

Funding

This work was funded by the grants from the National Key R&D Program of China (No.2022YFD1301004), National Natural Science Foundation of China (No. 31601962), Fundamental Research Funds for the Central Universities (2662019QD021) and Key Laboratory of Molecular Animal Nutrition of Zhejiang University (KLMAN202101 and KLMAN202205).

Author information

Authors and Affiliations

Authors

Contributions

QX, YD, and YG conceptualized the review. YD and YG co-wrote this manuscript. MH, JH, LY, XW, WD, JL, and QX revised the main manuscript. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Qingbiao Xu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Du, Y., Gao, Y., Hu, M. et al. Colonization and development of the gut microbiome in calves. J Animal Sci Biotechnol 14, 46 (2023). https://doi.org/10.1186/s40104-023-00856-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-023-00856-x

Keywords