Skip to main content

The protective role of prebiotics and probiotics on diarrhea and gut damage in the rotavirus-infected piglets

Abstract

Rotavirus is one of the pathogenic causes that induce diarrhea in young animals, especially piglets, worldwide. However, nowadays, there is no specific drug available to treat the disease, and the related vaccines have no obvious efficiency in some countries. Via analyzing the pathogenesis of rotavirus, it inducing diarrhea is mainly due to disturb enteric nervous system, destroy gut mucosal integrity, induce intracellular electrolyte imbalance, and impair gut microbiota and immunity. Many studies have already proved that prebiotics and probiotics can mitigate the damage and diarrhea induced by rotavirus infection in hosts. Based on these, the current review summarizes and discusses the effects and mechanisms of prebiotics and probiotics on rotavirus-induced diarrhea in piglets. This information will highlight the basis for the swine production utilization of prebiotics and probiotics in the prevention or treatment of rotavirus infection in the future.

Introduction

Severe diarrhea is the primary cause of dehydration in the acute gastroenteritis, which can lead to some complications (even death) if leaving it untreated. Many viruses can induce acute gastroenteritis, such as rotavirus, norovirus, adenovirus, astrovirus, and calicivirus. Rotavirus is one of the main pathogens causing diarrhea in young animals (i.e., piglets). It mainly infects host duodenal and jejunal mucosal epithelial cells through the fecal–oral route, which will induce cell apoptosis and villi atrophy in gut, and then lead to malabsorption and diarrhea [1]. Piglets are extremely susceptible to rotavirus. During 1–4 weeks of age, the comprehensive infection rate is as high as 80%, and the mortality rate can reach up to 20% in piglets [2]. It has seriously impaired the health and growth of pigs all over the world, especially developing countries [3, 4].

Probiotics that are derived from Latin mean “for life”. Before probiotic microbes are discovered and utilized, their fermented products (such as beer, bread, wine, yogurt, mare’s milk wine, and cheese) have used for nutritional and therapeutic purposes [5]. Probiotics have a variety of beneficial effects on the body, including anti-inflammation, anti-oxidation, anti-cancer, anti-virus, and regulating intestinal flora and immune function [6,7,8,9,10,11,12]. Recently, prebiotics are developed as one of antibiotic substitutes [13]. This is mainly due that prebiotics may act as substrates for probiotics [14]. Moreover, the combination of probiotics and prebiotics as synbiotics can also regulate health of hosts [15]. Therefore, many studies have already focused on the curing and preventing effect of probiotics, prebiotics and synbiotics on the rotavirus-induced diarrhea via protecting gut health in piglets and some related models.

Rotavirus

Rotavirus is a member in the genus Rotavirus in the family Reoviridae, and its complete three-layered particles (inner, middle, and outer capsid layers) resemble a wheel with a smooth surface under electron microscopy [16]. The rotavirus genome is composed of 11 segments of double-stranded RNA encoding viral proteins (VP1–VP4, VP6 and VP7), and nonstructural proteins (NSP1–NSP5/6) [17,18,19,20]. According to the antigenicity of their VP6 protein, rotavirus are classified into ten different groups (A–J), whose A, B, C, E and H can only infect pigs; and rotavirus A is the highest prevalence, and one of the most obvious reasons of acute diarrhea of piglets [16].

Rotavirus pathogenesis

After rotavirus infects a juvenile host, it replicates primarily in epithelial cells of the middle and apex of the small intestine, inducing villi blunting and atrophy, thereby further impairing the intestinal absorptive surface area and hindering the absorption of essential nutrients such as carbohydrates, proteins, and fats [16]. In addition, right ventricular infection can lead to electrolyte imbalances and gut microbiome imbalances, which can further exacerbate intestinal damage. The combination of these factors leads to the occurrence of clinical symptoms associated with rotavirus infection (such as diarrhea, vomiting, fever, anorexia, and weight loss) [20]. In addition, some evidences have shown that rotavirus pathology can be systemic [21]. Rotavirus inducing diarrhea may have some mechanisms, as follows.

Rotavirus disturbs enteric nervous system

The intestinal motility is mainly regulated by the enteric nervous system, and 60% of cluster cells in the small intestine and 40% of cluster cells in the large intestine are in contact with nerve fibers [22]. Rotavirus infection down-regulates tyrosine hydroxylase in norepinephrine-sympathetic neurons, thereby reducing dopamine production [23]. This decreases norepinephrine production, and ultimately increases peristalsis in small intestine [23]. Rotavirus also activates intestinal glial cells through serotonin released by enterochromaffin cells, and then increases intestinal peristalsis [24]. These two effects can synergistically induce rapid intestinal peristalsis, causing host abdominal pain and diarrhea. Besides, rotavirus can activate vagus nerve afferents, and project into the medullary vomiting center of central nervous system, resulting to solitary nucleus excitation in the brainstem that dominates vomiting and diarrhea, and eventually form vomiting and diarrhea reflexes [25, 26]. Thus, rotavirus-induced diarrhea and some other symptoms are associated with abnormal stimulation of the enteric nervous system.

Rotavirus destroys gut mucosal integrity

Gut mucosa plays an important role in nutrient digestion and absorption, and barrier function. Rotavirus infection causes the damage of host intestinal mucosa (especially villi) through affecting the survival of intestinal epithelial cells [27]. Our studies have also shown that rotavirus could induce apoptosis and autophagy in the jejunum of piglets [28,29,30,31,32,33]. On the one hand, this leads to a deficiency in nutrient absorption, and water-electrolyte disturbances, which will induce subsequent malabsorptive diarrhea [34, 35]. On the other hand, rotavirus doing harm to gut mucosa will impair the expression and generation of tight-junction proteins (including zonula occludens 1, occludin and claudins) and mucins [28, 29, 31, 33, 36], which could induce the diarrhea via the secondary infection of other pathogens. In this process, it mainly exists three mechanisms, as follows.

Rotavirus induces oxidative stress

Redox balance is critical for the survival of cells (i.e., intestinal epithelial cells). Our recent studies in piglets found that rotavirus infection enhanced the level of reactive oxygen species in small intestine (data not published), and also showed that rotavirus challenge inhibited total antioxidant capacity and total superoxide dismutase activity, and increased malondialdehyde level in the small intestine of piglets [29, 31]. In the mouse model, Sodhi et al. [37] also got the similar results. In the rotavirus-infected IPEC-J2 cells, the mRNA levels of AMP-dependent protein kinase (AMPK), total nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream factors (including manganese superoxide dismutase, catalase, glutathione peroxidase, heme oxygenase 1 and quinone oxidoreductase 1) were down-regulated [36]. These studies proposed that rotavirus should impair intestinal mucosal integrity via disturbing antioxidant capacity, which was involved into AMPK-Nrf2 signaling pathway.

Besides, Cleary and Ashkenazi found that excess of peroxides and free radicals could stimulate intestinal glands, promote gland secretion, and induce host diarrhea, which indirectly illustrated that rotavirus impairing antioxidant capacity was also a potential mechanism of diarrhea [38].

Rotavirus stimulates some apoptosis- and autophagy-related pathways

In the epithelial cells of host small intestine, rotavirus also damages the intestinal epithelial mucosa by affecting autophagy- and apoptosis-related pathways. (1) Calcium/calmodulin dependent kinase 2 (CAMK2) and Ca2+-related adenosine 5´-monophosphate (AMP)-activated protein kinase (AMPK) pathways. The NSP4 protein of rotavirus can induce the release of endoplasmic reticulum calcium into the cytoplasm of infected cells, thereby activating CAMK2 and the Ca2+-related AMPK pathways to trigger autophagy, thereby facilitating rotavirus replication. Subsequently, rotavirus replication and maturation require the transfer of NSP4 to a new endoplasmic reticulum (ER) [39]. (2) LC3II-related pathway. The newly formed NSP4 and VP7 fuse with COPII vesicles and exit ER, then interact with the autophagy protein LC3II to induce cell autophagy and release new viruses [40]. (3) Protein kinase R-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2α (eIF2α) pathway. Rotavirus infection induces endoplasmic reticulum stress, thereby increasing PERK activity, leading to eIF2α phosphorylation, which selectively activates cAMP-dependent transcription factor 4 translation, and promotes of C/EBP homologous protein expression, and then inhibits B-cell lymphoma 2 expression, and induces of B-cell lymphoma 2-associated X transport from cytoplasm to mitochondria, thereby initiates cell apoptosis [30, 41, 42].

Rotavirus impairs endocrine in gut

The intestinal endocrine hormones play a vital role in the proliferation and survival of epithelial cells. Glucagon like peptide-2 (GLP-2), a 33-amino acid proglucagon-derived peptide, is generated and secreted by enteroendocrine cells of gut, which can promote intestinal growth and mucosal repair, increase epithelial cell proliferation, and inhibit enterocyte apoptosis [43]. Our previous study found that rotavirus challenge reduced GLP-2 level in the jejunal mucosa of piglets [29]. Thus, rotavirus destroying gut mucosa integrity is associated with the negative effect on gut endocrine.

Nonstructural protein 4 induces intracellular electrolyte imbalance

NSP4, a non-structural protein of rotavirus, is defined as a viral enterotoxin [44]. Except for impairing gut mucosal integrity, NSP4 may induce diarrhea by impairing intracellular electrolyte balance. (1) Via binding cell receptors, NSP4 boosts the production of 1,4,5-triphosphate-regulated inositol, and further activates the phosphatidyl inositol signaling pathway, which mobilizes extracellular Ca2+ inflow, in turn stimulates chloride (Cl) secretion pathways to increase the release of Cl into the lumen [45, 46]. (2) NSP4 synthesis inhibits the activity of the sodium (Na+) glucose co-transporter SGLT1 by activating TMEM16A channel, thereby inhibiting the intracellular transport of Na+ [47, 48]. (3) NSP4 can also activate the release of extracellular adenosine 5´-bisphosphate (ADP) of P2Y1 purinergic receptors on neighboring cells, which produces paracrine signals that may be manifested as intercellular Ca2+ waves, further amplifying electrolyte disorder of host cells and changing gastrointestinal physiology [49]. Intracellular electrolyte imbalance will lead to the decreased membrane stability and intracellular water loss, ultimately inducing diarrhea [50].

Rotavirus affects host immunity

As a pathogen, rotavirus can invade hosts (especially intestine), but not be cleaned by the anti-viral immunity. This is derived from some rotavirus-resistant mechanisms.

To affect the immune-related cells

Some studies have shown that rotavirus infection can interfere the role of immune-related cells in animals. (1) Dendritic cells. In mice, rotavirus infection increased the expression of major histocompatibility complex II, CD40, CD80, and CD86 in mesenteric lymph nodes dendritic cells, which synergistically regulate the maturation, differentiation, and function of dendritic cells. Dendritic cells are divided into CD11b+ and CD11b dendritic cells. CD11b+ dendritic cells promote antigen presentation, inflammatory response and regulating T cell activation. CD11b dendritic cells play an important role in antigen capture and processing, and immune regulation [51,52,53]. Rotavirus infection affecting those protein expression would increase the percentage of CD11c+CD11b+CD8a dendritic cells in the mesenteric lymph nodes and decrease the proportion of CD11c+CD11bCD8a+ dendritic cells, thereby downregulating immune response, leading to worsening of infection [54]. (2) Natural killer (NK) cells. Comstock et al. [55] found that rotavirus infection can reduce the number of NK cells in the mesenteric lymph nodes and ileal Peyer’s nodes of piglets. Vitamin A deficiency in sows infected with rotavirus had the decreasing number of NK cells [56]. In addition, it was found that rotavirus infection promotes immature NK cell activation, leading to cytotoxicity of bile duct cells and causing bile duct obstruction in mice [57]. Therefore, rotavirus infection can impair the number and activity of NK cells, and NK cell function, thereby lowering host immunity or causing other diseases. And this is related to the nutritional status of hosts. (3) Some other cells. In newborn piglets, rotavirus infection can reduce the total number of CD14 cells and monocytes in the intestinal lymph nodes, and also decrease the macrophage population in the mesenteric lymph nodes and ileal Peyer’s nodes [55]. González et al. [58] found that rotavirus infection reduces the activation of monocytes, thereby reducing uptake/binding with rotavirus-like particles. In addition, it was found in the rotavirus-infected mouse model that rotavirus infection increased the mRNA expression levels of macrophage inflammatory protein-1 and monocyte chemotactic protein-1 in the duodenum, jejunum, and ileum tissues [59]. In summary, rotavirus infection can reduce the number and activity of CD14 cells, monocytes and macrophages in host, and stimulate the expression of pro-inflammatory factors/chemokines secreted by these cells, thereby causing a decrease in immune function, and inducing inflammation.

To impair the pattern recognition receptors

Some components of rotavirus are a pathogen-associated molecular patterns, and it can be recognized by pattern recognition receptors (PRRs) in intestinal epithelial cells or immune cells [60]. However, rotavirus has 2 ways to escape from the related immune response. (1) NSP1, a non-structural protein of rotavirus, may play a role for hiding viral RNA, which prevents PRRs recognizing the virus, and reduces antiviral response of interferon (IFN) [61]. (2) NSP1 and VP3 (known as a viral protein) induce PRRs (such as retinoic acid-induced gene I, mitochondrial antiviral-signaling protein) degradation and/or decrease their formation, and degrade interferon regulatory factors (IRF), including IRF3, IRF5, IRF7, and IRF9 [62,63,64,65]. This will block the generation of host IFNs and pro-inflammatory cytokines, and decrease interferon-stimulated factor 3 formation, and eventually reduce the host innate antiviral immune activity [64, 66,67,68,69,70].

To regulate some immune-related pathways

Rotavirus can also impair the host’s immune response by regulating some immune related pathways. (1) Rotavirus-infected MA104 cells avoids the nuclear accumulation of activated signaling and transcriptional activating protein 1/2, and then effectively blocks the expression of type I and type II IFN-induced genes, which in turn reduces endogenous ISG mRNA levels [71]. (2) NSP1 in MA104 cells can inhibit the activation of NF-κB pathway by degrading E3 ubiquitin ligase complex, and inhibit the generation of host IFNs and pro-inflammatory cytokines [67, 72, 73]. (3) Rotavirus-infected piglet intestine and IPEC-J2 cells can also up-regulate microRNA-155-5p expression, and further targets and inhibits the mRNA levels of TBK1, IRF3 and IFN-β. This regulates TBK1/IRF3 signaling pathway [74].

To influence the immune factors

Adaptive immunity against rotavirus can be used for virus identification, clearance, and placement for reinfection [3]. In the early stage of rotavirus infection, a large amount of rotavirus-IgM antibodies are produced in host. However, these antibodies have few roles for virus neutralization, and are only used as a symptom of infection. After 5 d of rotavirus infection, the rotavirus-IgG antibodies will be generated by host, which are mainly used to neutralize virus. Our studies also found that rotavirus challenge could enhanced serum and/or intestinal rotavirus specific antibodies in piglets [28, 29, 33, 60, 70, 75]. Li et al. [76] reported that rotavirus infection increased the serum concentration of rotavirus specific IgM and IgG in piglets. In addition, Preidis et al. [77] got the similar results in mice.

Certainly, rotavirus infection also affects the innate immunity related factors. Zhao et al. [78] found that rotavirus infection increased serum levels of IL-2, IL-4 and IFN-β in weaned piglets, as well as increased the expression of interferon-stimulated gene 15 (ISG15). Chen et al. [75] reported that, compared with normal piglets, rotavirus-challenged piglets have the higher levels of serum IgA and IgG, and jejunal and ileal IL-2 levels. Moreover, in the rotavirus-infected mice, the generation of intestinal secreting immunoglobulin A, and serum interferon-γ (IFN-γ) and tumor necrosis factor-α was promoted [79].

However, some previous studies have shown that there are the suppression of antiviral immune response and cytokine generation in host during the early stage of rotavirus infection [80, 81]. Thus, the difference of these results in immune factors could be mainly derived from the infectious duration. And these also may explain why the symptom of rotavirus infection mainly occur in the early stage of infection, and gradually self-heal after 5 d of infection.

Rotavirus impairs intestinal microbiota

Gut ecosystem depends on the diversity and integrity of intestinal microflora, which is indispensable for many physiological processes, and plays a vital role in health (especially gut health) [82]. In the process of rotavirus-induced gastroenteritis, microbiota diversity was temporarily reduced, and microbiota composition was significantly altered [83]. Zhao et al. [84] determined the colon and rectum digesta of mice with rotavirus infection, and found that the relative abundance of Proteobacteria and Candidatus was increased, and the relative abundance of Firmicutes, Fusobacteria, Bacteroidetes, and Actinobacteria was decreased. Our study also reported that oral gavage of rotavirus resulted in a decrease in the total number of bacteria, lactobacilli, and bifidobacteria, and an increase in the population of Escherichia coli in the ileum and cecum digesta of piglets [28, 29, 33]. These results indicate that rotavirus infection induces a decrease in the total number of bacteria and beneficial bacteria in the host’s gut, as well as an enhancement in the number of harmful bacteria, which lead to the dysbiosis of gut microbiota and the increasing diarrhea duration. Thus, the pathogenicity of rotavirus is closely involved into the change of intestinal microbiota.

Probiotics and/or prebiotics alleviates the rotavirus-induced diarrhea

Due to the diversity and variability of rotavirus species, there is currently no specific drug available to treat rotavirus infection. In swine production, rotavirus can’t be eradicated. Most producers mainly use antibiotics to reduce mortality and diarrhea, and further minimize the impact of rotavirus infection on swine production [3, 4]. In addition, some vaccines to rotavirus have already researched and used [85,86,87,88,89]. However, because of the difference in age, sex, weight, environment, gut microbiome and genome, the application of these vaccines can’t achieve the expected results, especially in livestock [90]. Then, some dietary interventions ways (including nutrients, and functional materials) are used to prevent and cure the rotavirus-induced diarrhea [32, 60, 74, 91,92,93]. Probiotics, prebiotics and synbiotics have positive effects on gut health, which leads to their utilization of alleviating rotavirus-induced gastroenteritis in many studies of pigs and other animal models.

Probiotics

Nowadays, probiotics are internationally recognized as the live and non-pathogenic microorganisms that provide health benefits to hosts when administered in sufficient amounts [94]. Probiotics are mainly used to improve microbial balance in the gastrointestinal tract [8], which mainly contain Bifidobacterium, Lactobacillus and some other bacteria. Besides these, their roles may be played via food (for example, dairy products) fermentation and drug development. As an alternative to antibiotics, they have now become a research hotspot in animal production. There are a plenty of studies about the protective effects of probiotics on rotavirus infection (Table 1).

Table 1 The effect of probiotics on rotavirus infection in the in vivo and in vitro studies

Bifidobacterium

Bifidobacterium belongs to the phylum Actinomycetes, and is one of the first microorganisms to colonize the intestines of neonatal animals [133]. This bacterium has beneficial effects on hosts, such as preventing diarrhea, building the healthy microbiome, regulating the immune system, improving lactose intolerance, lowering cholesterol, and preventing cancer [134, 135]. In many studies of mouse and rat model, oral administration of different Bifidobacteria (including Bifidobacterium breve (YIT4064 and M-16V), Bifidobacterium bifidum, Bifidobacterium infantis, Bifidobacterium longum (subsp. Infantis CECT 7210, SPM1205 and SPM1206), Bifidobacterium animalis (subsp. Lactis Bb12), Bifidobacterium lactis (HN019), and Bifidobacterium thermophilus (RBL67)) may inhibit rotavirus infection and rotavirus-induced diarrhea, which is embodied on the decrease of diarrhea severity, incidence, and duration, and the shortened recovery period for diarrhea [95,96,97,98,99,100,101,102,103,104,105]. And dietary Bifidobacterium breve YIT4064 supplementation for female mice can efficiently protect their pups against rotavirus infection [95]. Rigo-Adrover et al. [136] fermented formula with Bifidobacterium breve and Streptococcus thermophilus, and this milk could alleviate the effect of rotavirus challenge on suckling rats.

Lactobacillus

The genus Lactobacillus belongs to the group of lactic acid bacteria (LAB). Lactobacilli are Gram-positive, catalase-negative, non-spore forming, rod-shaped, and facultative anaerobic, which mainly produce lactic acid by fermentation [137, 138]. Lactobacilli are widely found in food, feed, plants, animals, and humans, where fermentation may happen [139]. Generally, Lactobacillus is considered as one of the most abundant microorganisms in the intestine of hosts, and is closely linked to gut health [140]. Many studies in pigs, calves and mice have shown that the administration of lactobacilli (including Lactobacillus rhamnosus GG (LGG), Lactobacillus reuteri, Lactobacillus acidophilus, and Lactobacillus gasseri) may improve growth performance, and alleviate the rotavirus-induced diarrhea [9, 33, 54, 77, 109, 110, 113,114,115,116], and then could reduce the rotavirus-induced diarrhea in their pups [117]. And even, the products (i.e., exopolysaccharide) of lactobacilli had the positive effect on improving rotavirus-induced diarrhea and viral shedding, and prevented the disruption of intestinal epithelial integrity [141].

In addition, the administration of lactobacilli (such as Lactobacillus casei, Lactobacillus plantarum) relieves the infection and harm of some viruses (including transmissible gastroenteritis virus, porcine epidemic diarrhea virus) in the gut of hosts [142, 143].

Escherichia coli Nissle 1917

Escherichia coli Nissle 1917 (EcN) is a Gram-negative probiotic. EcN has the antagonistic effects on a variety of intestinal pathogens, and regulates the secretion of immune factors, which can enhance immunity in hosts [144]. Oral gavage of EcN attenuated the negative effect of rotavirus infection on gut functions and immunity, and reduced the mean peak titer of viral shedding and diarrhea in piglets [121, 122, 124,125,126]. And there was the synergetic effect on rotavirus infection when EcN was used with other materials (such as tryptophan) [123].

Some other bacteria

Except for Bifidobacterium and Lactobacillus, some other probiotics have also displayed the protective role for rotavirus infecting hosts. (1) Lacticaseibacillus rhamnosus GG. Via oral infusion, it might relieve the rotavirus infection, diarrhea, and gut damage, and improve immune function, cell autophagy and apoptosis in piglets and mice [118,119,120]. (2) Lactococcus lactis (subsp. Lactis JCM 5805). Oral administration of heat-killed Lactococcus lactis (subsp. Lactis JCM 5805) improved the retardation of body weight gain and fecal scores, and reduced rotavirus titer in the feces of mice [127]. (3) Pediococcus acidilactici. Oral gavage of Pediococcus acidilactici decreased the number of viral particles in the duodenum of specific pathogen-free mice [128]. (4) Lactiplantibacillus plantarum (299V). Leblanc et al. [129] found that the incubation of Lactiplantibacillus plantarum (299V) reduced the infection rate of rotavirus in IPEC-J2 cells. (5) Ligilactobacillus salivarius (FFIG35 and FFIG58). Indo et al. [130] reported that the incubation of Ligilactobacillus salivarius (FFIG35 and FFIG58) reduced the rotavirus replication in the porcine intestinal epithelial cells. (6) Segmented filamentous bacteria (SFB). SFB are some Gram-positive clostridial bacteria, mainly colonizes the surface of epithelial cells at the end of ileum, and plays an important role in preventing the pathogenic microbial infection, and the occurrence and development of autoimmune diseases [145, 146]. Shi et al. [131] and Ngo et al. [132] reported that oral infusion of SFB alleviated the damage of intestines, and inhibited the viral replication and infection in the rotavirus-challenge mice.

Prebiotics

In December 2016, the International Scientific Association for Probiotics and Prebiotics updated the definition of prebiotics as a class of substances that are selectively utilized by host microorganisms to confer health benefits on the host. Its broad definition may be involved into all substances that retain selectively microbiota-mediated mechanisms and must be beneficial for health [14]. At present, the main prebiotics include functional oligosaccharides, polysaccharides and natural plant extracts. The main purpose of prebiotics application is to stimulate the growth and activity of beneficial bacteria in the gastrointestinal tract, and has positive effects on host health via acting as a barrier to pathogens [13], and improving host immunity, antioxidant capacity and intestinal barrier [147].

Prebiotics also show the role for preventing rotavirus infection (Table 2). The studies of our lab have shown that supplementing pectic oligosaccharide, lentinan, or lactose in diets can efficiently improve intestinal microflora, gut barrier function and immunity, reduce diarrhea, and increase growth performance in the rotavirus-infected piglets [28, 29, 31, 70, 75]. Oral administration of β-glucan improved the health condition and reduced the piglet mortality from rotavirus associated diarrhea [148]. Supplementing human milk oligosaccharides or a mixture of short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides in formula reduced the duration of rotavirus-induced diarrhea in piglets partially by modulating colonic microbiota and immune response to rotavirus infection; and the effect of human milk oligosaccharides was better than that of the mixture [55, 76]. In the sows vaccinated with rotavirus, supplementing galacto-oligosaccharide in diets could reduce rotavirus infection in the neonatal piglets [149]; and oral administration of galacto-oligosaccharides reduced the incidence and severity of rotavirus-associated diarrhea in Lewis rats [150]. Rigo-Adrover et al. [136] reported that supplementing a mixture of short-chain galacto-oligosaccharides, long-chain fructo-oligosaccharides and pectin-derived acidic oligosaccharides in formula protected suckling rats from rotavirus gastroenteritis, and supplementing 2´-fucosyllactose and/or a mixture of short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides in formula reduced the clinical variables of rotavirus-induced diarrhea in newborn rats [151,152,153]. The treatment of polysaccharides from Portulaca oleracea L. could also reduce the expression of VP6, and viral titers during the internalization and replication phases of rotavirus in IPEC-J2 cells [154].

Table 2 The effect of prebiotics on rotavirus infection in the in vivo and in vitro studies

Additionally, in piglets and some models with rotavirus infection, the administration of many plant extracts could relieve diarrhea, and improved small intestinal lesion scores and fecal virus excretion. These plant extracts are involved into resveratrol, ginsenoside, saponin, quercetin, loureirin, baicalin, ziyuglycoside II, genipin, deoxyshikonin, steviol glycosides, bitter ginseng extract, red wine extract, Glycyrrhiza uralensis extract, Qiwei Baizhu powder, Macleaya cordata extracts, and Valeriana jatamansi Jones extracts [59, 155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170].

Synbiotics

Synbiotics are defined as “mixtures of probiotics and prebiotics that beneficially affects the host by improving the survival and implantation of live microbial dietary supplements” [171]. Synbiotics have the characteristics of probiotic and prebiotic, and were produced for overcoming some possible difficulties of probiotics survival in the gastrointestinal tract [172]. Therefore, an appropriate combination of both components should ensure a superior role, compared to the activity of probiotics or prebiotics alone [173]. From now on, there are several studies about the effect of synbiotics on the rotavirus-induced diarrhea (Table 3). Rigo-Adrover et al. showed that oral infusion of a mixture of short-chain galacto-oligosaccharides, long-chain fructo-oligosaccharides and Bifidobacterium breve (M-16V) was highly effective in modulating rotavirus-induced diarrhea for suckling rats [174, 175]. And when the probiotics were replaced by the milk fermented by Bifidobacterium breve and Streptococcus thermophilus, the incidence and severity of diarrhea were also reduced in suckling rats [176, 177].

Table 3 The effect of synbiotics on rotavirus infection in some studies

No many studies compare the efficiency of different probiotics and/or prebiotics treating and preventing rotavirus infection. Gagnon et al. [97] reported that some Bifidobacteria (such as Bifidobacterium thermacidophilum (RBL69, RBL70), Bifidobacterium longum (ATCC 15707) and Bifidobacterium pseudolongum (ATCC 25526)) were ineffective to rotavirus attachment and infection, but Bifidobacterium thermacidophilum (RBL67) could reduce rotavirus infection in mice. Rigo-Adrover et al. compared the efficiency of probiotics, prebiotics and synbiotics on rotavirus-infected rats, and found that the alleviating effect of synbiotics (mixture of short chain galacto-oligosaccharides, long chain fructo-oligosaccharides and Bifidobacterium breve (M-16V)) on rotavirus infection was better than that of single probiotics or prebiotics [174, 175].

However, the application of prebiotics and/or probiotics exists on the potential drawbacks or limitations, as following. (1) Individual differences. The composition of gut microbiota is different in each animal. This will lead to the various effects of prebiotics and/or probiotics. Some probiotic strains may not be beneficial, and even have negative effects to hosts [178]. (2) Dosage and usage. The optimal dosage and usage of most prebiotics and probiotics for animals are not yet clear. Different kinds of animals require different doses and usage cycles. Liu et al. [112] found that the low dose of Lactobacillus acidophilus (NCFM™) administration did not alleviate rotavirus infection in the gnotobiotic pigs. In addition, the use of prebiotics and probiotics may only be applicable to specific animals or specific health status [179]. (3) Product quality and cost. Nowadays, the quality and effectiveness of many probiotic and probiotic products are unstable for animals in the market. Some products may have good effects, but they is too expensive to use in animal breeding [180].

Mechanisms of probiotics and/or prebiotics against rotavirus

Rotavirus is one of the main causes of diarrhea in young animals, especially piglets, worldwide. It can invade hosts (especially intestine), thereby damaging host health and growth. According to the impairment of rotavirus on host (especially intestine) health, many studies analyzed the related indexes, and found that probiotics and/or prebiotics against rotavirus should be involved into several mechanisms, as follows (Fig. 1). (1) To increase immunity. Probiotics and/or prebiotics regulate the activity and function of immune cells (for example dendritic cells), and activate STAT1 through pattern recognition receptors and NF-κB signaling pathways, thereby increasing the production of type I interferon and interleukin, and activating ISG15 and immune response. In addition, probiotics and/or prebiotics can also increase the number of specific antibodies IgG in hosts. (2) To attenuate apoptosis. Probiotics and/or prebiotics might improve intestinal cell apoptosis induced by rotavirus infection through elevating antioxidant capacity and regulating some related signaling pathways (including MAPK/ERK/JNK, SIRT1/FoxO1/Rab7). (3) To enhance intestinal non-specific barrier. Probiotics and/or prebiotics improve the intestinal morphology, and the production of mucins and tight junction proteins. (4) To improve the absorption of water and glucose. Probiotics and/or prebiotics stimulate the mRNA expression and function of sodium/glucose cotransporter 1, thereby improving the water and glucose absorption of rotavirus-infected hosts. (5) To improve the intestinal microflora. Probiotics and/or prebiotics can maintain the balance of gut microbiota, and relieve the negative impact of rotavirus infection on intestinal microflora in hosts. (6) To directly interact with rotavirus. Probiotics and/or prebiotics can bind with VP4 and/or VP7 of rotavirus, thereby reducing the number of rotavirus-infected cells. In addition, probiotics and/or prebiotics can reduce NSP4 levels in the intestine of rotavirus-infected hosts.

Fig. 1
figure 1

The mechanisms of probiotics and/or prebiotics against rotavirus

To regulate intestinal microflora

The administration of probiotics and/or prebiotics could decline the negative effects of rotavirus infection on intestinal microflora in hosts. Preidis et al. [77] found that oral administration of Lactobacillus reuteri can increase the species richness and phylogenetic diversity of gut microbiota, and alleviate the impairment of rotavirus on gut microbiota in mice. In addition, adding breast milk oligosaccharides in formula can increase the level of Bacteroidetes in the colon of newborn piglets infected by rotavirus, thereby alleviating the imbalance of gut microbiota [76]. Our study also found that supplementing the diet with probiotics (i.e., LGG) or prebiotics (including pectic oligosaccharides, lactose, or shiitake polysaccharides) increased the number of Lactobacillus, Bifidobacterium, and total bacteria in the cecum of rotavirus-infected piglets, and reduced the number of Escherichia coli [28, 29, 33, 76]. This can be the main mechanism of probiotics and prebiotics improving host health.

To improve immunity

The administration of probiotics and/or prebiotics can effectively regulate the immune response of hosts. There are the different roles among different probiotics and prebiotics.

Innate immunity and inflammation

The innate immunity and inflammation are the important compounds of immune system of hosts. They mainly contain the activation and effect of immune-related cells, the production and effect of cytokines, and the related pathways. Following the invasion of pathogens, they immediately play a critical role of pathogen clearance in hosts [181]. Probiotics and prebiotics may influence these effects.

  1. (1)

    EcN. Kumar et al. [182] used Gram-positive and Gram-negative probiotics to incubate the rotavirus-infected cells, determined the pathways of innate immunity and inflammation by PCR arrays, and found that EcN activated innate immune (including cell movement of granulocytes, immune response of cells, activation of phagocytes, and activation of macrophages) and inflammatory responses (such as cytokine and chemokine mediated signaling pathway) to neutralize rotavirus infection. In the rotavirus-challenge piglets, oral infusion of EcN increased the function of NK cells and the production of a large number of plasmacyte-like dendritic cells, and reduced the monocytes expressing Toll-like receptor 4 and cytokine (for example IL-6) levels in the spleen, blood, and/or intestine [121,122,123, 125].

  2. (2)

    Lactobacillus, Lactococcus lactis and Ligilactobacillus salivarius. Oral administration of LGG reduced the percentage of CD11c+CD11b+CD8a dendritic cells and increased the percentage of CD11c+CD11bCD8a+ dendritic cells, thereby regulating immune cells against rotavirus invasion [54]. Liu et al. [108] found that LGG treatment might regulate the expression of Toll-like receptors (TLRs), and some cytokine responses in the rotavirus-infected IPEC-J2 cells. Our study showed that supplementing LGG in diet relieved the effect of rotavirus on IL-2 and IL-4 levels in the jejunum of piglets [33]. Dietary Lactobacillus reuteri and Lactobacillus acidophilus supplementation could enhanced the response and level of Th1 and Th2 cytokines (i.e., IL-12, IFN-γ, IL-4 and IL-10) in gnotobiotic pigs infected by rotavirus [114]. Oral administration of heat-killed Lactococcus lactis (subsp. Lactis JCM 5805) upregulated the expression levels of anti-viral factors induced by IFNs, such as Isg15, Mx1, Oasl2 and Viperin in the small intestinal epithelial cells of mice [127]. The treatment of Ligilactobacillus salivarius Ligilactobacillus salivarius (FFIG35 and FFIG58) stimulated the expression of IFN-β, IFN-λ and antiviral factors in the porcine intestinal epithelial cells infected by rotavirus, which was associated with modulating TLR signaling pathway [130].

  3. (3)

    Bifidobacterium. Bifidobacterium animalis (subsp. Lactis Bb12) treatment regulated the response of cytokines (such as IL-6, IL-10) via affecting the expression of TLRs and the number of immune cells in the ileal monocytes of piglets [103,104,105]. Oral administration of Bifidobacterium longum (SPM1205 and SPM1206) regulated IFN-mediated innate immunity in rotavirus-infected mice, which was involved into the increase of IFN-α and IFN-β levels and the mRNA expression of IFN receptor, STAT1 and IFN-induced antiviral effectors (myxovirus resistance A, and 20,50-oligoadenylate synthase) [101].

  4. (4)

    Prebiotics. Supplementing human milk oligosaccharides in formula increased systemic NK cells, memory effector T cells, and major histocompatibility complex II cells in rotavirus-infected pigs [55]. The studies of our lab have shown that the administration of prebiotics (including lentinan, pectic oligosaccharide, or lactose) could improve the concentrations and mRNA expression of cytokines and host defense peptides, and regulate the expression of pattern recognition receptors in the jejunum of rotavirus-challenge piglets [28, 70, 75]. The administration of Glycyrrhiza uralensis extract regulated the expression of cytokines via some signaling pathway (such as p38, JNK and NF-κB) in the small intestine and spleen of rotavirus-infected pigs [167]. In the studies of some animal models infected by rotavirus, the administration of prebiotics (such as 2´-fucosyllactose and/or a mixture of short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides, ziyuglycoside II, Macleaya cordata extracts, Valeriana jatamansi Jones extracts) could modulate the cytokine expression and generation via some signaling pathways (including pattern recognition receptors, TLR/NF-κB, JAK2/STAT3, PI3K/AKT) [151, 153, 161, 165, 170].

Antibody generation

After invading hosts, pathogens are recognized by antigen-presenting cells (such as phagocytes, macrophages, dendritic cells), and then immunocompetent cells are activated [80]. In humoral immunity, plasmacyte will generate the relative antibodies. The antibodies can inhibit pathogen attachment, invasion and proliferation, neutralize pathogens, and boost the pathogen clearance. Probiotics and prebiotics may promote the production of antibodies [183]. In some studies of mouse model, the administration of probiotics (including Bifidobacterium bifidum, Bifidobacterium infantis, Bifidobacterium thermophilus, Lactobacillus reuteri and Lactobacillus gasseri) increased fecal and serum rotavirus-specific IgA levels in the rotavirus-challenge mice, and even stimulated the humoral specific IgG and IgM responses in the rotavirus-challenge mice [77, 96, 97, 117]. Azevedo et al. [114] and Zhang et al. [115] found that the treatment of Lactobacillus reuteri and Lactobacillus acidophilus could enhance intestinal IgA-secreting cellular responses, serum IgM titers, and intestinal IgM and IgG titers in rotavirus-infected piglets. Oral gavage of EcN in the rotavirus-challenge gnotobiotic piglets increased the IgA level and response of small intestine [124]. Our studies also found that supplementing probiotics (i.e., LGG) or prebiotics (i.e., lentinan, pectic oligosaccharide, lactose) in diets increased the levels of IgA, IgG or rotavirus-specific antibody in the serum and/or intestine of rotavirus-infected piglets [28, 33, 70, 75]. In addition, many studies reported that probiotics and/or prebiotics administration might enhance the rotavirus-antibody titer and immune response in the hosts with rotavirus vaccine [105,106,107, 111, 112, 119, 122, 126, 149].

To maintain intestinal functions

The intestine has the function of digestion, absorption and barrier, which will play a critical for the growth and health of animals and human. The gut barrier encompasses some elements, such as mucosal integrity, and the intercellular junctions between epithelial cells [184]. LGG administration ameliorated morphology, mucin 1 and mucin 2 concentrations, and zonula occludens 1 and occludin expression in the jejunum of rotavirus-infected piglets [33], and increased mucin 3 level in the large intestine digesta of rotavirus-infected gnotobiotic pigs [9]. Oral gavage of Bifidobacterium bifidum (G9-1) increased the number of acidic mucin-positive goblet cells, and the mRNA expression of mucin 2, mucin 3, mucin 4, transforming growth factor β1, Trefoil factor 3, occludin, claudin-1 and villin-1 in the small intestine of rotavirus-infected mice [98]. Kumar et al. [182] analyzed the effect of EcN on rotavirus-infected cells through PCR arrays, and found that EcN upregulated intercellular contact and adhesion (especially formation of tight junctions). Our studies also found that prebiotics (i.e., lentinan, pectic oligosaccharide, lactose) administration for rotavirus-infected piglets improved morphology, the levels of mucins, and the expression of tight junction proteins in the jejunum [28, 29, 31]. Altogether, probiotics/prebiotics relieve the damage of rotavirus infection on the host intestinal mucosa by increasing the levels of mucins, tight junction proteins, and some other factors that affect cell growth and repair.

Except for barrier function, the function of digestion and absorption will be impaired by rotavirus infection. However, there are few studies about the effect of probiotics and/or prebiotics on digestion and absorption functions in hosts. Kawahara et al. [98] reported that oral infusion of Bifidobacterium bifidum (G9-1) stimulated the mRNA expression of sodium/glucose cotransporter 1 (SGLT1), thereby ameliorating water and glucose absorption in the rotavirus-infected mice. Song et al. [159] showed that baicalin administration might modulate the glucose metabolism, which mainly regulate the gluconeogenesis via affecting p-JNK/PDK1/AKT/SIK2 signaling pathway in the mice infected by rotavirus.

The gut functions are closely associated with the survival and activity of intestinal epithelial cells [185]. And probiotics and/or prebiotics might decrease cell apoptosis, and increase cell migration during rotavirus infection, which was due that probiotics and/or prebiotics elevated antioxidant capacity, and regulated some related signaling pathways (including MAPK/ERK/JNK, SIRT1/FoxO1/Rab7) [28, 29, 31, 118, 186,187,188].

Some other mechanisms

In the process of probiotics and/or prebiotics against rotavirus infection and rotavirus-induced diarrhea, there are some other ways. (1) To directly interact with external viral proteins. EcN or Bifidobacterium longum (SPM1205 and SPM1206) might bind VP4 and/or VP7 of rotavirus, and then reduce the number of virus-infected cells [101]. (2) To reduce NSP4. Our studies showed that the administration of probiotics (i.e., LGG) or prebiotics (i.e., lentinan, pectic oligosaccharide, lactose) decreased the NSP4 level in the intestine of rotavirus-challenge pigs [28, 29, 33, 70]. (3) To decrease Ca2+ outflow in cells. Some lactobacilli (i.e., LGG) and their metabolites downregulated the size of short-circuit current and the release of Ca2+ in the virus-infected cells [186], and some plant extract (i.e., red wine extract, loureirin C) could inhibit calcium-activated chloride channels in the rotavirus-infected mice [162, 166].

Conclusion

Rotavirus is one of the main causes of viral diarrhea in piglets worldwide. Probiotics and/or prebiotics have the capacity of promoting the health of pigs. They may, to some extent, prevent and cure rotavirus infection, and attenuate the negative effect of rotavirus infection on health and growth of hosts. In swine production, lactic acid bacteria and bifidobacteria have been widely applicated as the most effective probiotics, while prebiotics are mainly various plant extracts. Their functions are involved into restoring intestinal function, improving digestion and absorption capacity, reducing diarrhea symptoms, regulating gut microbiota, and enhancing immune function. Nowadays, there are many studies on probiotics and/or prebiotics healing and preventing rotavirus infection, which often get the good efficiency. This will supply the effective solution for the prevention of diarrhea (especially rotavirus-induced diarrhea) in the industry of swine breeding.

However, there are many studies needed to be done in future. Firstly, the mechanisms of probiotics and/or prebiotics protecting the hosts against rotavirus infection are not enough. In the second place, there are few studies on the comparation of the efficiency of different probiotics and/or prebiotics treating and preventing rotavirus infection. Last but not the least, during treating rotavirus infection, probiotics and/or prebiotics are mainly used as adjuvant therapy. The related dosages and usage methods should be profoundly researched.

Availability of data and materials

Not applicable.

Abbreviations

AMPK:

AMP-dependent protein kinase

CAMK2:

Calcium/calmodulin-dependent kinase II

EcN:

Escherichia coli Nissle 1917

ERK:

Extracellular regulated protein kinases

GLP-2:

Glucagon like peptide-2

IFN:

Interferon

Ig:

Immunoglobulin

IL:

Interleukin

IRF:

Interferon regulatory factors

ISG:

Interferon-stimulated genes

JNK:

JUN N-terminal kinases

LGG:

Lactobacillus rhamnosus GG

MAPK:

Mitogen-activated protein kinases

NF-κB:

Nuclear factor-kappa B

Nrf2:

Nuclear factor erythroid 2-related factor 2

NSP:

Viral non-structural proteins

PRRs:

Pattern recognition receptors

SFB:

Segmented filamentous bacteria

SGLT1:

Sodium/glucose cotransporter 1

TBK1:

TANK-binding kinase 1

VP:

Virus structure protein

References

  1. Estes MK, Kang G, Zeng CQ, Crawford SE, Ciarlet M. Pathogenesis of rotavirus gastroenteritis. Novartis Found Symp. 2001;238:82–100. https://doi.org/10.1002/0470846534.ch6.

    Article  CAS  PubMed  Google Scholar 

  2. Zhang H, Zhang Z, Wang Y, Wang X, Xia M, Wu H. Isolation, molecular characterization and evaluation of the pathogenicity of a porcine rotavirus isolated from Jiangsu Province. China Arch Virol. 2015;160:1333–8. https://doi.org/10.1007/s00705-015-2347-9.

    Article  CAS  PubMed  Google Scholar 

  3. Kumar D, Shepherd FK, Springer NL, Mwangi W, Marthaler DG. Rotavirus infection in swine: genotypic diversity, immune responses, and role of gut microbiome in rotavirus immunity. Pathogens (Basel, Switzerland). 2022;11:1078. https://doi.org/10.3390/pathogens11101078.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Vlasova AN, Amimo JO, Saif LJ. Porcine rotaviruses: epidemiology immune responses and control strategies. Viruses. 2017;18;9(3):48. https://doi.org/10.3390/v9030048.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Ragan MV, Wala SJ, Goodman SD, Bailey MT, Besner GE. Next-generation probiotic therapy to protect the intestines from injury. Front Cell Infect Microbiol. 2022;12:863949. https://doi.org/10.3389/fcimb.2022.863949.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Duffy LC, Zielezny MA, Riepenhoff-Talty M, Dryja D, Sayahtaheri-Altaie S, Griffiths E, et al. Effectiveness of Bifidobacterium bifidum in mediating the clinical course of murine rotavirus diarrhea. Pediatr Res. 1994;35:690–5. https://doi.org/10.1203/00006450-199406000-00014.

    Article  CAS  PubMed  Google Scholar 

  7. Araki K, Shinozaki T, Irie Y, Miyazawa Y. Trial of oral administration of Bifidobacterium breve for the prevention of rotavirus infections. Kansenshogaku Zasshi. 1999;73:305–10. https://doi.org/10.11150/Kansenshogakuzasshi1970.73.305.

    Article  CAS  PubMed  Google Scholar 

  8. Williams NT. Probiotics. Am J Health Syst Pharm. 2010;67:449–58. https://doi.org/10.2146/ajhp090168.

    Article  CAS  PubMed  Google Scholar 

  9. Liu F, Li G, Wen K, Wu S, Zhang Y, Bui T, et al. Lactobacillus rhamnosus GG on rotavirus-induced injury of ileal epithelium in gnotobiotic pigs. J Pediatr Gastroenterol Nutr. 2013;57:750–8. https://doi.org/10.1097/MPG.0b013e3182a356e1.

    Article  PubMed  Google Scholar 

  10. Tiptiri-Kourpeti A, Spyridopoulou K, Santarmaki V, Aindelis G, Tompoulidou E, Lamprianidou EE, et al. Lactobacillus casei exerts anti-proliferative effects accompanied by apoptotic cell death and up-regulation of TRAIL in colon carcinoma cells. PLoS ONE. 2016;11:e0147960. https://doi.org/10.1371/journal.pone.0147960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gonzalez-Ochoa G, Flores-Mendoza LK, Icedo-Garcia R, Gomez-Flores R, Tamez-Guerra P. Modulation of rotavirus severe gastroenteritis by the combination of probiotics and prebiotics. Arch Microbiol. 2017;199:953–61. https://doi.org/10.1007/s00203-017-1400-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tang J, Song X, Zhao M, Chen H, Wang Y, Zhao B, et al. Oral administration of live combined Bacillus subtilis and Enterococcus faecium alleviates colonic oxidative stress and inflammation in osteoarthritic rats by improving fecal microbiome metabolism and enhancing the colonic barrier. Front Microbiol. 2022;13:1005842. https://doi.org/10.3389/fmicb.2022.1005842.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Markowiak P, Śliżewska K. Effects of probiotics, prebiotics, and synbiotics on human health. Nutrients. 2017;9:1021. https://doi.org/10.3390/nu9091021.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Gibson GR, Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol. 2017;14:491–502. https://doi.org/10.1038/nrgastro.2017.75.

    Article  PubMed  Google Scholar 

  15. Nguyen T-T, Nguyen P-T, Pham M-N, Razafindralambo H, Hoang Q-K, Nguyen H-T. Synbiotics: a new route of self-production and applications to human and animal health. Probiotics Antimicrob Proteins. 2022;14:980–93. https://doi.org/10.1007/s12602-022-09960-2.

    Article  PubMed  Google Scholar 

  16. Zimmerman JJ, Karriker LA, Ramirez A, Schwartz KJ, Stevenson GW, Zhang JQ. Diseases of swine. 11th ed. Hoboken, NJ: Wiley-Blackwell/American Association of Swine Veterinarians; 2019.

  17. Caddy S, Papa G, Borodavka A, Desselberger U. Rotavirus research: 2014–2020. Virus Res. 2021;304:198499. https://doi.org/10.1016/j.virusres.2021.198499.

    Article  CAS  PubMed  Google Scholar 

  18. Hu L, Crawford SE, Hyser JM, Estes MK, Prasad BVV. Rotavirus non-structural proteins: structure and function. Curr Opin Virol. 2012;2:380–8. https://doi.org/10.1016/j.coviro.2012.06.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Desselberger U. Rotaviruses. Virus Res. 2014;190:75–96. https://doi.org/10.1016/j.virusres.2014.06.016.

    Article  CAS  PubMed  Google Scholar 

  20. Crawford SE, Ramani S, Tate JE, Parashar UD, Svensson L, Hagbom M, et al. Rotavirus infection. Nat Rev Dis Primers. 2017;3:17083. https://doi.org/10.1038/nrdp.2017.83.

    Article  PubMed  Google Scholar 

  21. Gómez-Rial J, Sánchez-Batán S, Rivero-Calle I, Pardo-Seco J, Martinón-Martínez JM, Salas A, et al. Rotavirus infection beyond the gut. Infect Drug Resist. 2019;12:55–64. https://doi.org/10.2147/IDR.S186404.

    Article  PubMed  Google Scholar 

  22. Cheng X, Voss U, Ekblad E. Tuft cells: distribution and connections with nerves and endocrine cells in mouse intestine. Exp Cell Res. 2018;369:105–11. https://doi.org/10.1016/j.yexcr.2018.05.011.

    Article  CAS  PubMed  Google Scholar 

  23. Hellysaz A, Svensson L, Hagbom M. Rotavirus downregulates tyrosine hydroxylase in the noradrenergic sympathetic nervous system in Ileum, early in infection and simultaneously with increased intestinal transit and altered brain activities. mBio. 2022;13:e0138722. https://doi.org/10.1128/mbio.01387-22.

    Article  CAS  PubMed  Google Scholar 

  24. Hagbom M, De Faria FM, Winberg ME, Westerberg S, Nordgren J, Sharma S, et al. Neurotrophic factors protect the intestinal barrier from rotavirus insult in mice. mBio. 2020;11(1):e02834-19. https://doi.org/10.1128/mBio.02834-19.

  25. Hagbom M, Istrate C, Engblom D, Karlsson T, Rodriguez-Diaz J, Buesa J, et al. Rotavirus stimulates release of serotonin (5-HT) from human enterochromaffin cells and activates brain structures involved in nausea and vomiting. PLoS Pathog. 2011;7:e1002115. https://doi.org/10.1371/journal.ppat.1002115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xue J, Askwith C, Javed NH, Cooke HJ. Autonomic nervous system and secretion across the intestinal mucosal surface. Auton Neurosci. 2007;133:55–63. https://doi.org/10.1016/j.autneu.2007.02.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Amimo JO, Raev SA, Chepngeno J, Mainga AO, Guo Y, Saif L, et al. Rotavirus interactions with host intestinal epithelial cells. Front Immunol. 2021;12:793841. https://doi.org/10.3389/fimmu.2021.793841.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yu W, Xiao X, Chen D, Yu B, He J, Zheng P, et al. Effect of dietary lactose supplementation on growth performance and intestinal epithelium functions in weaned pigs challenged by rotavirus. Animals (Basel). 2022;12(18):2336. https://doi.org/10.3390/ANI12182336.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Mao X, Xiao X, Chen D, Yu B, He J, Chen H, et al. Dietary apple pectic oligosaccharide improves gut barrier function of rotavirus-challenged weaned pigs by increasing antioxidant capacity of enterocytes. Oncotarget. 2017;8:92420–30. https://doi.org/10.18632/oncotarget.21367.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Zhao Y, Hu N, Jiang Q, Zhu L, Zhang M, Jiang J, et al. Protective effects of sodium butyrate on rotavirus inducing endoplasmic reticulum stress-mediated apoptosis via PERK-eIF2α signaling pathway in IPEC-J2 cells. J Anim Sci Biotechnol. 2021;12:69. https://doi.org/10.1186/s40104-021-00592-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mao X, Hu H, Xiao X, Chen D, Yu B, He J, et al. Lentinan administration relieves gut barrier dysfunction induced by rotavirus in a weaned piglet model. Food Funct. 2019;10:2094–101. https://doi.org/10.1039/c8fo01764f.

    Article  CAS  PubMed  Google Scholar 

  32. Tian G, Liang X, Chen D, Mao X, Yu J, Zheng P, et al. Vitamin D3 supplementation alleviates rotavirus infection in pigs and IPEC-J2 cells via regulating the autophagy signaling pathway. J Steroid Biochem Mol Biol. 2016;163:157–63. https://doi.org/10.1016/j.jsbmb.2016.05.004.

    Article  CAS  PubMed  Google Scholar 

  33. Mao X, Gu C, Hu H, Tang J, Chen D, Yu B, et al. Dietary Lactobacillus rhamnosus GG supplementation improves the mucosal barrier function in the intestine of weaned piglets challenged by porcine rotavirus. PLoS ONE. 2016;11:e0146312. https://doi.org/10.1371/journal.pone.0146312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Osborne MP, Haddon SJ, Spencer AJ, Collins J, Starkey WG, Wallis TS, et al. An electron microscopic investigation of time-related changes in the intestine of neonatal mice infected with murine rotavirus. J Pediatr Gastroenterol Nutr. 1988;7:236–48. https://doi.org/10.1097/00005176-198803000-00014.

    Article  CAS  PubMed  Google Scholar 

  35. Ramig RF. Pathogenesis of intestinal and systemic rotavirus infection. J Virol. 2004;78:10213–20. https://doi.org/10.1128/JVI.78.19.10213-10220.2004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Dong X, Wang Y, Zhu X, Shen L, Chen L, Niu L, et al. Sodium butyrate protects against rotavirus-induced intestinal epithelial barrier damage by activating AMPK-Nrf2 signaling pathway in IPEC-J2 cells. Int J Biol Macromol. 2022;228:186–96. https://doi.org/10.1016/j.ijbiomac.2022.12.219.

    Article  CAS  PubMed  Google Scholar 

  37. Sodhi CP, Katyal R, Rana SV, Attri S, Singh V. Study of oxidative-stress in rotavirus infected infant mice. Indian J Med Res. 1996;104:245–9.

    CAS  PubMed  Google Scholar 

  38. Cleary TG, Ashkenazi S. Intestinal electrolyte transport and diarrheal disease. N Engl J Med. 1990;322:1321–2. https://doi.org/10.1056/NEJM199005033221817.

    Article  CAS  PubMed  Google Scholar 

  39. Crawford SE, Estes MK. Viroporin-mediated calcium-activated autophagy. Autophagy. 2013;9:797–8. https://doi.org/10.4161/auto.23959.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Crawford SE, Criglar JM, Liu Z, Broughman JR, Estes MK. COPII vesicle transport is required for rotavirus NSP4 interaction with the autophagy protein LC3 II and trafficking to viroplasms. J Virol. 2019;94(1):e01341-19. https://doi.org/10.1128/JVI.01341-19.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Wang Q, Xin X, Wang T, Wan J, Ou Y, Yang Z, et al. Japanese encephalitis virus induces apoptosis and encephalitis by activating the PERK pathway. J Virol. 2019;93(17):e00887-19. https://doi.org/10.1128/JVI.00887-19.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Zhang Q, Xu Y, Chang R, Tong D, Xu X. Transmissible gastroenteritis virus N protein causes endoplasmic reticulum stress, up-regulates interleukin-8 expression and its subcellular localization in the porcine intestinal epithelial cell. Res Vet Sci. 2018;119:109–15. https://doi.org/10.1016/j.rvsc.2018.06.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Baldassano S, Amato A. GLP-2: what do we know? What are we going to discover? Regul Pept. 2014;194–195:6–10. https://doi.org/10.1016/j.regpep.2014.09.002.

    Article  CAS  PubMed  Google Scholar 

  44. Saurabh S, Sircar S, Kattoor JJ, Ghosh S, Kobayashi N, Banyai K, et al. Analysis of structure-function relationship in porcine rotavirus A enterotoxin gene. J Vet Sci. 2018;19:35–43. https://doi.org/10.4142/jvs.2018.19.1.35.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Dong Y, Zeng CQ, Ball JM, Estes MK, Morris AP. The rotavirus enterotoxin NSP4 mobilizes intracellular calcium in human intestinal cells by stimulating phospholipase C-mediated inositol 1,4,5-trisphosphate production. Proc Natl Acad Sci U S A. 1997;94:3960–5. https://doi.org/10.1073/pnas.94.8.3960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Morris AP, Estes MK. Microbes and microbial toxins: paradigms for microbial-mucosal interactions. VIII. Pathological consequences of rotavirus infection and its enterotoxin. Am J Physiol Gastrointest Liver Physiol. 2001;281:G303–10. https://doi.org/10.1152/ajpgi.2001.281.2.G303.

    Article  CAS  PubMed  Google Scholar 

  47. Ball JM, Tian P, Zeng CQ, Morris AP, Estes MK. Age-dependent diarrhea induced by a rotaviral nonstructural glycoprotein. Science. 1996;272:101–4. https://doi.org/10.1126/science.272.5258.101.

    Article  CAS  PubMed  Google Scholar 

  48. Ousingsawat J, Mirza M, Tian Y, Roussa E, Schreiber R, Cook DI, et al. Rotavirus toxin NSP4 induces diarrhea by activation of TMEM16A and inhibition of Na+ absorption. Pflugers Arch - Eur J Physiol. 2011;461:579–89. https://doi.org/10.1007/s00424-011-0947-0.

    Article  CAS  PubMed  Google Scholar 

  49. Chang-Graham AL, Perry JL, Engevik MA, Engevik KA, Scribano FJ, Gebert JT, et al. Rotavirus induces intercellular calcium waves through ADP signaling. Science. 2020;370(6519):eabc3621. https://doi.org/10.1126/science.abc3621.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Lundgren O, Svensson L. Pathogenesis of rotavirus diarrhea. Microbes Infect. 2001;3:1145–56. https://doi.org/10.1016/s1286-4579(01)01475-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Yin X, Chen S, Eisenbarth SC. Dendritic cell regulation of T helper cells. Annu Rev Immunol. 2021;39:759–90. https://doi.org/10.1146/annurev-immunol-101819-025146.

    Article  CAS  PubMed  Google Scholar 

  52. Macri C, Pang ES, Patton T, O’Keeffe M. Dendritic cell subsets. Semin Cell Dev Biol. 2018;84:11–21. https://doi.org/10.1016/j.semcdb.2017.12.009.

    Article  CAS  PubMed  Google Scholar 

  53. Leylek R, Idoyaga J. The versatile plasmacytoid dendritic cell: Function, heterogeneity, and plasticity. Int Rev Cell Mol Biol. 2019;349:177–211. https://doi.org/10.1016/bs.ircmb.2019.10.002.

    Article  CAS  PubMed  Google Scholar 

  54. Jiang Y, Ye L, Cui Y, Yang G, Yang W, Wang J, et al. Effects of Lactobacillus rhamnosus GG on the maturation and differentiation of dendritic cells in rotavirus-infected mice. Benef Microbes. 2017;8:645–56. https://doi.org/10.3920/BM2016.0157.

  55. Comstock SS, Li M, Wang M, Monaco MH, Kuhlenschmidt TB, Kuhlenschmidt MS, et al. Dietary human milk oligosaccharides but not prebiotic oligosaccharides increase circulating natural killer cell and mesenteric lymph node memory T cell populations in noninfected and rotavirus-infected neonatal piglets. J Nutr. 2017;147:1041–7. https://doi.org/10.3945/jn.116.243774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chepngeno J, Amimo JO, Michael H, Raev SA, Jung K, Lee MV, et al. Vitamin A deficiency and vitamin A supplementation affect innate and T cell immune responses to rotavirus A infection in a conventional sow model. Front Immunol. 2023;14:1188757. https://doi.org/10.3389/fimmu.2023.1188757.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Qiu Y, Yang J, Wang W, Zhao W, Peng F, Xiang Y, et al. HMGB1-promoted and TLR2/4-dependent NK cell maturation and activation take part in rotavirus-induced murine biliary atresia. PLoS Pathog. 2014;10:e1004011. https://doi.org/10.1371/journal.ppat.1004011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. González AM, Azevedo MSP, Jung K, Vlasova A, Zhang W, Saif LJ. Innate immune responses to human rotavirus in the neonatal gnotobiotic piglet disease model. Immunology. 2010;131:242–56. https://doi.org/10.1111/j.1365-2567.2010.03298.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Huang H, Liao D, Zhou G, Zhu Z, Cui Y, Pu R. Antiviral activities of resveratrol against rotavirus in vitro and in vivo. Phytomedicine. 2020;77:153230. https://doi.org/10.1016/j.phymed.2020.153230.

    Article  CAS  PubMed  Google Scholar 

  60. Mao X, Gu C, Ren M, Chen D, Yu B, He J, et al. L-Isoleucine administration alleviates rotavirus infection and immune response in the weaned piglet model. Front Immunol. 2018;9:1654. https://doi.org/10.3389/fimmu.2018.01654.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Guo Z, Chen L, Zeng H, Gomez JA, Plowden J, Fujita T, et al. NS1 protein of influenza A virus inhibits the function of intracytoplasmic pathogen sensor, RIG-I. Am J Respir Cell Mol Biol. 2007;36:263–9. https://doi.org/10.1165/rcmb.2006-0283RC.

    Article  CAS  PubMed  Google Scholar 

  62. Graff JW, Mitzel DN, Weisend CM, Flenniken ML, Hardy ME. Interferon regulatory factor 3 is a cellular partner of rotavirus NSP1. J Virol. 2002;76:9545–50. https://doi.org/10.1128/JVI.76.18.9545-9550.2002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Barro M, Patton JT. Rotavirus nonstructural protein 1 subverts innate immune response by inducing degradation of IFN regulatory factor 3. Proc Natl Acad Sci U S A. 2005;102:4114–9. https://doi.org/10.1073/pnas.0408376102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ding S, Zhu S, Ren L, Feng N, Song Y, Ge X, et al. Rotavirus VP3 targets MAVS for degradation to inhibit type III interferon expression in intestinal epithelial cells. Elife. 2018;7:e39494. https://doi.org/10.7554/eLife.39494.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Nandi S, Chanda S, Bagchi P, Nayak MK, Bhowmick R, Chawla-Sarkar M. MAVS protein is attenuated by rotavirus nonstructural protein 1. PLoS ONE. 2014;9:e92126. https://doi.org/10.1371/journal.pone.0092126.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Barro M, Patton JT. Rotavirus NSP1 inhibits expression of type I interferon by antagonizing the function of interferon regulatory factors IRF3, IRF5, and IRF7. J Virol. 2007;81:4473–81. https://doi.org/10.1128/JVI.02498-06.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Arnold MM, Patton JT. Diversity of interferon antagonist activities mediated by NSP1 proteins of different rotavirus strains. J Virol. 2011;85:1970–9. https://doi.org/10.1128/JVI.01801-10.

    Article  CAS  PubMed  Google Scholar 

  68. Arnold MM, Barro M, Patton JT. Rotavirus NSP1 mediates degradation of interferon regulatory factors through targeting of the dimerization domain. J Virol. 2013;87:9813–21. https://doi.org/10.1128/JVI.01146-13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hakim MS, Chen S, Ding S, Yin Y, Ikram A, Ma X-X, et al. Basal interferon signaling and therapeutic use of interferons in controlling rotavirus infection in human intestinal cells and organoids. Sci Rep. 2018;8:8341. https://doi.org/10.1038/s41598-018-26784-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Fan X, Hu H, Chen D, Yu B, He J, Yu J, et al. Lentinan administration alleviates diarrhea of rotavirus-infected weaned pigs via regulating intestinal immunity. J Anim Sci Biotechnol. 2021;12:43. https://doi.org/10.1186/s40104-021-00562-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Holloway G, Truong TT, Coulson BS. Rotavirus antagonizes cellular antiviral responses by inhibiting the nuclear accumulation of STAT1, STAT2, and NF-kappaB. J Virol. 2009;83:4942–51. https://doi.org/10.1128/JVI.01450-08.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Graff JW, Ettayebi K, Hardy ME. Rotavirus NSP1 inhibits NFkappaB activation by inducing proteasome-dependent degradation of beta-TrCP: a novel mechanism of IFN antagonism. PLoS Pathog. 2009;5:e1000280. https://doi.org/10.1371/journal.ppat.1000280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Morelli M, Dennis AF, Patton JT. Putative E3 ubiquitin ligase of human rotavirus inhibits NF-κB activation by using molecular mimicry to target β-TrCP. mBio. 2015;6(1):e02490-14. https://doi.org/10.1128/mBio.02490-14.

  74. Zhao Y, Ran Z, Jiang Q, Hu N, Yu B, Zhu L, et al. Vitamin D alleviates rotavirus infection through a microrna-155-5p mediated regulation of the TBK1/IRF3 signaling pathway in vivo and in vitro. Int J Mol Sci. 2019;20(14):3562. https://doi.org/10.3390/ijms20143562.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Chen H, Hu H, Chen D, Tang J, Yu B, Luo J, et al. Dietary pectic oligosaccharide administration improves growth performance and immunity in weaned pigs infected by rotavirus. J Agric Food Chem. 2017;65:2923–9. https://doi.org/10.1021/acs.jafc.7b00039.

    Article  CAS  PubMed  Google Scholar 

  76. Li M, Monaco MH, Wang M, Comstock SS, Kuhlenschmidt TB, Fahey GC, et al. Human milk oligosaccharides shorten rotavirus-induced diarrhea and modulate piglet mucosal immunity and colonic microbiota. ISME J. 2014;8:1609–20. https://doi.org/10.1038/ismej.2014.10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Preidis GA, Saulnier DM, Blutt SE, Mistretta T-A, Riehle KP, Major AM, et al. Host response to probiotics determined by nutritional status of rotavirus-infected neonatal mice. J Pediatr Gastroenterol Nutr. 2012;55:299–307. https://doi.org/10.1097/MPG.0b013e31824d2548.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Zhao Y, Yu B, Mao X, He J, Huang Z, Zheng P, et al. Dietary vitamin D supplementation attenuates immune responses of pigs challenged with rotavirus potentially through the retinoic acid-inducible gene I signalling pathway. Br J Nutr. 2014;112:381–9. https://doi.org/10.1017/S000711451400097X.

    Article  CAS  PubMed  Google Scholar 

  79. Zhang Z, Xiang Y, Li N, Wang B, Ai H, Wang X, et al. Protective effects of Lactobacillus rhamnosus GG against human rotavirus-induced diarrhoea in a neonatal mouse model. Pathog Dis. 2013;67:184–91. https://doi.org/10.1111/2049-632X.12030.

    Article  CAS  PubMed  Google Scholar 

  80. Arnold MM, Patton JT. Rotavirus antagonism of the innate immune response. Viruses. 2009;1:1035–56. https://doi.org/10.3390/v1031035.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Qin L, Ren L, Zhou Z, Lei X, Chen L, Xue Q, et al. Rotavirus nonstructural protein 1 antagonizes innate immune response by interacting with retinoic acid inducible gene I. Virol J. 2011;8:526. https://doi.org/10.1186/1743-422X-8-526.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Shi N, Li N, Duan X, Niu H. Interaction between the gut microbiome and mucosal immune system. Mil Med Res. 2017;4:14. https://doi.org/10.1186/s40779-017-0122-9.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Rodríguez-Díaz J, García-Mantrana I, Vila-Vicent S, Gozalbo-Rovira R, Buesa J, Monedero V, et al. Relevance of secretor status genotype and microbiota composition in susceptibility to rotavirus and norovirus infections in humans. Sci Rep. 2017;7:45559. https://doi.org/10.1038/srep45559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Zhao W, Yu ML, Tao X, Cheng MH, Liu CC, Liu Y, et al. Analysis of the intestinal microbial community altered during rotavirus infection in suckling mice. Virol J. 2021;18:254. https://doi.org/10.1186/s12985-021-01727-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. O’Ryan M. Rotarix (RIX4414): an oral human rotavirus vaccine. Expert Rev Vaccines. 2007;6:11–9. https://doi.org/10.1586/14760584.6.1.11.

    Article  CAS  PubMed  Google Scholar 

  86. O’Ryan M, Linhares AC. Update on Rotarix: an oral human rotavirus vaccine. Expert Rev Vaccines. 2009;8:1627–41. https://doi.org/10.1586/erv.09.136.

    Article  CAS  PubMed  Google Scholar 

  87. Taniguchi K. Human rotavirus vaccine. Uirusu. 2012;62:87–96. https://doi.org/10.2222/jsv.62.87.

    Article  CAS  PubMed  Google Scholar 

  88. Esona MD, Gautam R. Rotavirus. Clin Lab Med. 2015;35:363–91. https://doi.org/10.1016/j.cll.2015.02.012.

    Article  PubMed  Google Scholar 

  89. Gandhi GR, Santos VS, Denadai M, da Silva Calisto VK, de Souza Siqueira Quintans J, de Oliveira e Silva AM, et al. Cytokines in the management of rotavirus infection: A systematic review of in vivo studies. Cytokine. 2017;96:152–60. https://doi.org/10.1016/j.cyto.2017.04.013.

  90. Carvalho MF, Gill D. Rotavirus vaccine efficacy: current status and areas for improvement. Hum Vaccin Immunother. 2019;15:1237–50. https://doi.org/10.1080/21645515.2018.1520583.

    Article  PubMed  Google Scholar 

  91. Steele JC. Rotavirus. Clin Lab Med. 1999;19:691–703.

    Article  PubMed  Google Scholar 

  92. Mao X, Liu M, Tang J, Chen H, Chen D, Yu B, et al. Dietary leucine supplementation improves the mucin production in the jejunal mucosa of the weaned pigs challenged by porcine rotavirus. PLoS ONE. 2015;10:e0137380. https://doi.org/10.1371/journal.pone.0137380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhao Y, Yu B, Mao X, He J, Huang Z, Zheng P, et al. Effect of 25-hydroxyvitamin D3 on rotavirus replication and gene expressions of RIG-I signalling molecule in porcine rotavirus-infected IPEC-J2 cells. Arch Anim Nutr. 2015;69:227–35. https://doi.org/10.1080/1745039X.2015.1034522.

    Article  CAS  PubMed  Google Scholar 

  94. Capurso L. I probiotici [Probiotics]. Recenti Prog Med. 2016;107:267–77. doi: 10.1701/2296.24682.

    PubMed  Google Scholar 

  95. Yasui H, Kiyoshima J, Ushijima H. Passive protection against rotavirus-induced diarrhea of mouse pups born to and nursed by dams fed Bifidobacterium breve YIT4064. J Infect Dis. 1995;172:403–9. https://doi.org/10.1093/infdis/172.2.403.

  96. Qiao H, Duffy LC, Griffiths E, Dryja D, Leavens A, Rossman J, et al. Immune responses in rhesus rotavirus-challenged BALB/c mice treated with bifidobacteria and prebiotic supplements. Pediatr Res. 2002;51:750–5. https://doi.org/10.1203/00006450-200206000-00015.

    Article  PubMed  Google Scholar 

  97. Gagnon M, Vimont A, Darveau A, Fliss I, Jean J. Study of the ability of bifidobacteria of human origin to prevent and treat rotavirus infection using colonic cell and mouse models. PLoS ONE. 2016;11:e0164512. https://doi.org/10.1371/journal.pone.0164512.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kawahara T, Makizaki Y, Oikawa Y, Tanaka Y, Maeda A, Shimakawa M, et al. Oral administration of Bifidobacterium bifidum G9–1 alleviates rotavirus gastroenteritis through regulation of intestinal homeostasis by inducing mucosal protective factors. PLoS ONE. 2017;12:e0173979. https://doi.org/10.1371/journal.pone.0173979.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Shu Q, Qu F, Gill HS. Probiotic treatment using Bifidobacterium lactis HN019 reduces weanling diarrhea associated with rotavirus and Escherichia coli infection in a piglet model. J Pediatr Gastroenterol Nutr. 2001;33:171–7. https://doi.org/10.1097/00005176-200108000-00014.

    Article  CAS  PubMed  Google Scholar 

  100. Muñoz JAM, Chenoll E, Casinos B, Bataller E, Ramón D, Genovés S, et al. Novel probiotic Bifidobacterium longum subsp. infantis CECT 7210 strain active against rotavirus infections. Appl Environ Microbiol. 2011;77:8775–83.

    Article  PubMed  Google Scholar 

  101. Kang JY, Lee DK, Ha NJ, Shin HS. Antiviral effects of Lactobacillus ruminis SPM0211 and Bifidobacterium longum SPM1205 and SPM1206 on rotavirus-infected Caco-2 cells and a neonatal mouse model. J Microbiol. 2015;53:796–803. https://doi.org/10.1007/s12275-015-5302-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Azagra-Boronat I, Massot-Cladera M, Knipping K, Garssen J, Ben Amor K, Knol J, et al. Strain-specific probiotic properties of bifidobacteria and lactobacilli for the prevention of diarrhea caused by rotavirus in a preclinical model. Nutrients. 2020;12(2):498. https://doi.org/10.3390/nu12020498.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Vlasova AN, Chattha KS, Kandasamy S, Liu Z, Esseili M, Shao L, et al. Lactobacilli and bifidobacteria promote immune homeostasis by modulating innate immune responses to human rotavirus in neonatal gnotobiotic pigs. PLoS ONE. 2013;8:e76962. https://doi.org/10.1371/journal.pone.0076962.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Chattha KS, Vlasova AN, Kandasamy S, Rajashekara G, Saif LJ. Divergent immunomodulating effects of probiotics on T cell responses to oral attenuated human rotavirus vaccine and virulent human rotavirus infection in a neonatal gnotobiotic piglet disease model. J Immunol. 2013;191:2446–56. https://doi.org/10.4049/jimmunol.1300678.

    Article  CAS  PubMed  Google Scholar 

  105. Kandasamy S, Chattha KS, Vlasova AN, Rajashekara G, Saif LJ. Lactobacilli and Bifidobacteria enhance mucosal B cell responses and differentially modulate systemic antibody responses to an oral human rotavirus vaccine in a neonatal gnotobiotic pig disease model. Gut Microbes. 2014;5:639–51. https://doi.org/10.4161/19490976.2014.969972.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Wen K, Tin C, Wang H, Yang X, Li G, Giri-Rachman E, et al. Probiotic Lactobacillus rhamnosus GG enhanced Th1 cellular immunity but did not affect antibody responses in a human gut microbiota transplanted neonatal gnotobiotic pig model. PLoS ONE. 2014;9:e94504. https://doi.org/10.1371/journal.pone.0094504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Parreno V, Bai M, Liu F, Jing J, Olney E, Li G, et al. Probiotic as adjuvant significantly improves protection of the lanzhou trivalent rotavirus vaccine against heterologous challenge in a gnotobiotic pig model of human rotavirus infection and disease. Vaccines (Basel). 2022;10(9):1529. https://doi.org/10.3390/vaccines10091529.

    Article  PubMed  Google Scholar 

  108. Liu F, Li G, Wen K, Bui T, Cao D, Zhang Y, et al. Porcine small intestinal epithelial cell line (IPEC-J2) of rotavirus infection as a new model for the study of innate immune responses to rotaviruses and probiotics. Viral Immunol. 2010;23:135–49. https://doi.org/10.1089/vim.2009.0088.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Ventola H, Lehtoranta L, Madetoja M, Simonen-Tikka M-L, Maunula L, Roivainen M, et al. Effects of the viability of Lactobacillus rhamnosus GG on rotavirus infection in neonatal rats. World J Gastroenterol. 2012;18:5925–31. https://doi.org/10.3748/wjg.v18.i41.5925.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Günaydın G, Zhang R, Hammarström L, Marcotte H. Engineered Lactobacillus rhamnosus GG expressing IgG-binding domains of protein G: capture of hyperimmune bovine colostrum antibodies and protection against diarrhea in a mouse pup rotavirus infection model. Vaccine. 2014;32:470–7. https://doi.org/10.1016/j.vaccine.2013.11.057.

    Article  CAS  PubMed  Google Scholar 

  111. Zhang W, Azevedo MSP, Wen K, Gonzalez A, Saif LJ, Li G, et al. Probiotic Lactobacillus acidophilus enhances the immunogenicity of an oral rotavirus vaccine in gnotobiotic pigs. Vaccine. 2008;26:3655–61. https://doi.org/10.1016/j.vaccine.2008.04.070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Liu F, Wen K, Li G, Yang X, Kocher J, Bui T, et al. Dual functions of Lactobacillus acidophilus NCFM as protection against rotavirus diarrhea. J Pediatr Gastroenterol Nutr. 2014;58:169–76. https://doi.org/10.1097/MPG.0000000000000197.

    Article  PubMed  Google Scholar 

  113. Gandhar JS, De UK, Kala A, Malik YS, Yadav S, Paul BR, et al. Efficacy of microencapsulated probiotic as adjunct therapy on resolution of diarrhea, copper-zinc homeostasis, immunoglobulins, and inflammatory markers in serum of spontaneous rotavirus-infected diarrhoetic calves. Probiotics Antimicrob Proteins. 2022;14:1054–66. https://doi.org/10.1007/s12602-021-09862-9.

    Article  CAS  PubMed  Google Scholar 

  114. Azevedo MSP, Zhang W, Wen K, Gonzalez AM, Saif LJ, Yousef AE, et al. Lactobacillus acidophilus and Lactobacillus reuteri modulate cytokine responses in gnotobiotic pigs infected with human rotavirus. Benef Microbes. 2012;3:33–42. https://doi.org/10.3920/BM2011.0041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Zhang W, Azevedo MSP, Gonzalez AM, Saif LJ, van Nguyen T, Wen K, et al. Influence of probiotic Lactobacilli colonization on neonatal B cell responses in a gnotobiotic pig model of human rotavirus infection and disease. Vet Immunol Immunopathol. 2008;122:175–81. https://doi.org/10.1016/j.vetimm.2007.10.003.

    Article  CAS  PubMed  Google Scholar 

  116. Zhang W, Wen K, Azevedo MSP, Gonzalez A, Saif LJ, Li G, et al. Lactic acid bacterial colonization and human rotavirus infection influence distribution and frequencies of monocytes/macrophages and dendritic cells in neonatal gnotobiotic pigs. Vet Immunol Immunopathol. 2008;121:222–31. https://doi.org/10.1016/j.vetimm.2007.10.001.

    Article  CAS  PubMed  Google Scholar 

  117. Kadooka Y, Tominari K, Sakai F, Yasui H. Prevention of rotavirus-induced diarrhea by preferential secretion of IgA in breast milk via maternal administration of Lactobacillus gasseri SBT2055. J Pediatr Gastroenterol Nutr. 2012;55:66–71. https://doi.org/10.1097/MPG.0b013e3182533a2b.

    Article  CAS  PubMed  Google Scholar 

  118. Wu S, Yuan L, Zhang Y, Liu F, Li G, Wen K, et al. Probiotic Lactobacillus rhamnosus GG mono-association suppresses human rotavirus-induced autophagy in the gnotobiotic piglet intestine. Gut Pathog. 2013;5:22. https://doi.org/10.1186/1757-4749-5-22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Wen K, Liu F, Li G, Bai M, Kocher J, Yang X, et al. Lactobacillus rhamnosus GG dosage affects the adjuvanticity and protection against rotavirus diarrhea in gnotobiotic pigs. J Pediatr Gastroenterol Nutr. 2015;60:834–43. https://doi.org/10.1097/MPG.0000000000000694.

    Article  PubMed  Google Scholar 

  120. Pant N, Marcotte H, Brüssow H, Svensson L, Hammarström L. Effective prophylaxis against rotavirus diarrhea using a combination of Lactobacillus rhamnosus GG and antibodies. BMC Microbiol. 2007;7:86. https://doi.org/10.1186/1471-2180-7-86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Michael H, Paim FC, Miyazaki A, Langel SN, Fischer DD, Chepngeno J, et al. Escherichia coli Nissle 1917 administered as a dextranomar microsphere biofilm enhances immune responses against human rotavirus in a neonatal malnourished pig model colonized with human infant fecal microbiota. PLoS ONE. 2021;16:e0246193. https://doi.org/10.1371/journal.pone.0246193.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Michael H, Paim FC, Langel SN, Miyazaki A, Fischer DD, Chepngeno J, et al. Escherichia coli Nissle 1917 enhances innate and adaptive immune responses in a ciprofloxacin-treated defined-microbiota piglet model of human rotavirus infection. mSphere. 2021;6(2):e00074–21. https://doi.org/10.1128/mSphere.00074-21.

  123. Michael H, Srivastava V, Deblais L, Amimo JO, Chepngeno J, Saif LJ, et al. The combined Escherichia coli Nissle 1917 and tryptophan treatment modulates immune and metabolome responses to human rotavirus infection in a human infant fecal microbiota-transplanted malnourished gnotobiotic pig model. mSphere. 2022;7:0027022. https://doi.org/10.1128/msphere.00270-22.

    Article  CAS  Google Scholar 

  124. Kandasamy S, Vlasova AN, Fischer D, Kumar A, Chattha KS, Rauf A, et al. Differential effects of escherichia coli nissle and Lactobacillus rhamnosus strain GG on human rotavirus binding, infection, and B cell immunity. J Immunol. 2016;196:1780–9. https://doi.org/10.4049/jimmunol.1501705.

    Article  CAS  PubMed  Google Scholar 

  125. Vlasova AN, Shao L, Kandasamy S, Fischer DD, Rauf A, Langel SN, et al. Escherichia coli Nissle 1917 protects gnotobiotic pigs against human rotavirus by modulating pDC and NK-cell responses. Eur J Immunol. 2016;46:2426–37. https://doi.org/10.1002/eji.201646498.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Michael H, Miyazaki A, Langel SN, Amimo JO, Kick MK, Chepngeno J, et al. Escherichia coli Nissle 1917 enhances efficacy of oral attenuated human rotavirus vaccine in a gnotobiotic piglet model. Vaccines (Basel). 2022;10(1):83. https://doi.org/10.3390/vaccines10010083.

  127. Jounai K, Sugimura T, Morita Y, Ohshio K, Fujiwara D. Administration of Lactococcus lactis strain Plasma induces maturation of plasmacytoid dendritic cells and protection from rotavirus infection in suckling mice. Int Immunopharmacol. 2018;56:205–11. https://doi.org/10.1016/j.intimp.2018.01.034.

    Article  CAS  PubMed  Google Scholar 

  128. Niu T, Jiang Y, Fan S, Yang G, Shi C, Ye L, et al. Antiviral effects of Pediococcus acidilactici isolated from Tibetan mushroom and comparative genomic analysis. Front Microbiol. 2022;13:1069981. https://doi.org/10.3389/fmicb.2022.1069981.

  129. Leblanc D, Raymond Y, Lemay M-J, Champagne CP, Brassard J. Effect of probiotic bacteria on porcine rotavirus OSU infection of porcine intestinal epithelial IPEC-J2 cells. Arch Virol. 2022;167:1999–2010. https://doi.org/10.1007/s00705-022-05510-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Indo Y, Kitahara S, Tomokiyo M, Araki S, Islam MA, Zhou B, et al. Ligilactobacillus salivarius strains isolated from the porcine gut modulate innate immune responses in epithelial cells and improve protection against intestinal viral-bacterial superinfection. Front Immunol. 2021;12:652923. https://doi.org/10.3389/fimmu.2021.652923.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Shi Z, Zou J, Zhang Z, Zhao X, Noriega J, Zhang B, et al. Segmented filamentous bacteria prevent and cure rotavirus infection. Cell. 2019;179:644–658.e13. https://doi.org/10.1016/j.cell.2019.09.028.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Ngo VL, Shi Z, Jiang B, Gewirtz AT. Segmented filamentous bacteria impede rotavirus infection via retinoic acid receptor-mediated signaling. Gut Microbes. 2023;15:2174407. https://doi.org/10.1080/19490976.2023.2174407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Hidalgo-Cantabrana C, Delgado S, Ruiz L, Ruas-Madiedo P, Sánchez B, Margolles A. Bifidobacteria and their health-promoting effects. Microbiol Spectr. 2017;5(3). https://doi.org/10.1128/microbiolspec.BAD-0010-2016.

    Article  PubMed  Google Scholar 

  134. O’Callaghan A, van Sinderen D. Bifidobacteria and their role as members of the human gut microbiota. Front Microbiol. 2016;7:925. https://doi.org/10.3389/fmicb.2016.00925.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Alessandri G, Ossiprandi MC, MacSharry J, van Sinderen D, Ventura M. Bifidobacterial dialogue with its human host and consequent modulation of the immune system. Front Immunol. 2019;10:2348. https://doi.org/10.3389/fimmu.2019.02348.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Rigo-Adrover M, Pérez-Berezo T, Ramos-Romero S, van Limpt K, Knipping K, Garssen J, et al. A fermented milk concentrate and a combination of short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides/pectin-derived acidic oligosaccharides protect suckling rats from rotavirus gastroenteritis. Br J Nutr. 2017;117:209–17. https://doi.org/10.1017/S0007114516004566.

    Article  CAS  PubMed  Google Scholar 

  137. O’Callaghan J, O’Toole PW. Lactobacillus: host-microbe relationships. Curr Top Microbiol Immunol. 2013;358:119–54. https://doi.org/10.1007/82_2011_187.

    Article  CAS  PubMed  Google Scholar 

  138. Giraffa G, Chanishvili N, Widyastuti Y. Importance of lactobacilli in food and feed biotechnology. Res Microbiol. 2010;161:480–7. https://doi.org/10.1016/j.resmic.2010.03.001.

    Article  PubMed  Google Scholar 

  139. Duar RM, Lin XB, Zheng J, Martino ME, Grenier T, Pérez-Muñoz ME, et al. Lifestyles in transition: evolution and natural history of the genus Lactobacillus. FEMS Microbiol Rev. 2017;41:S27–48. https://doi.org/10.1093/femsre/fux030.

    Article  PubMed  Google Scholar 

  140. Heeney DD, Gareau MG, Marco ML. Intestinal Lactobacillus in health and disease, a driver or just along for the ride? Curr Opin Biotechnol. 2018;49:140–7. https://doi.org/10.1016/j.copbio.2017.08.004.

    Article  CAS  PubMed  Google Scholar 

  141. Kim K, Lee G, Thanh HD, Kim J-H, Konkit M, Yoon S, et al. Exopolysaccharide from Lactobacillus plantarum LRCC5310 offers protection against rotavirus-induced diarrhea and regulates inflammatory response. J Dairy Sci. 2018;101:5702–12. https://doi.org/10.3168/jds.2017-14151.

    Article  CAS  PubMed  Google Scholar 

  142. Huang S, Yu Q, Xie L, Ran L, Wang K, Yang Y, et al. Inhibitory effects of Lactobacillus plantarum metabolites on porcine epidemic diarrhea virus replication. Res Vet Sci. 2021;139:32–42. https://doi.org/10.1016/j.rvsc.2021.07.002.

    Article  CAS  PubMed  Google Scholar 

  143. Jiang X, Yu M, Qiao X, Liu M, Tang L, Jiang Y, et al. Up-regulation of MDP and tuftsin gene expression in Th1 and Th17 cells as an adjuvant for an oral Lactobacillus casei vaccine against anti-transmissible gastroenteritis virus. Appl Microbiol Biotechnol. 2014;98:8301–12. https://doi.org/10.1007/s00253-014-5893-2.

    Article  CAS  PubMed  Google Scholar 

  144. Zhao Z, Xu S, Zhang W, Wu D, Yang G. Probiotic Escherichia coli NISSLE 1917 for inflammatory bowel disease applications. Food Funct. 2022;13:5914–24. https://doi.org/10.1039/d2fo00226d.

    Article  CAS  PubMed  Google Scholar 

  145. Ericsson AC, Hagan CE, Davis DJ, Franklin CL. Segmented filamentous bacteria: commensal microbes with potential effects on research. Comp Med. 2014;64:90–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Flannigan KL, Denning TL. Segmented filamentous bacteria-induced immune responses: a balancing act between host protection and autoimmunity. Immunology. 2018;154:537–46. https://doi.org/10.1111/imm.12950.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Schley PD, Field CJ. The immune-enhancing effects of dietary fibres and prebiotics. Br J Nutr. 2002;87(Suppl 2):S221–30. https://doi.org/10.1079/BJNBJN/2002541.

    Article  CAS  PubMed  Google Scholar 

  148. Chethan GE, Garkhal J, Sircar S, Malik YPS, Mukherjee R, Sahoo NR, et al. Immunomodulatory potential of β-glucan as supportive treatment in porcine rotavirus enteritis. Vet Immunol Immunopathol. 2017;191:36–43. https://doi.org/10.1016/j.vetimm.2017.07.012.

    Article  CAS  PubMed  Google Scholar 

  149. Lee A, Liang L, Connerton PL, Connerton IF, Mellits KH. Galacto-oligosaccharides fed during gestation increase rotavirus A specific antibodies in sow colostrum, modulate the microbiome, and reduce infectivity in neonatal piglets in a commercial farm setting. Front Vet Sci. 2023;10:1118302. https://doi.org/10.3389/fvets.2023.1118302.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Massot-Cladera M, Del Rigo-Adrover MM, Herrero L, Franch À, Castell M, Vulevic J, et al. A galactooligosaccharide product decreases the rotavirus infection in suckling rats. Cells. 2022;11(10):1669. https://doi.org/10.3390/cells11101669.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Azagra-Boronat I, Massot-Cladera M, Knipping K, Van’t Land B, Tims S, Stahl B, et al. Oligosaccharides modulate rotavirus-associated dysbiosis and TLR gene expression in neonatal rats. Cells. 2019;8(8):876. https://doi.org/10.3390/cells8080876.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Sáez-Fuertes L, Azagra-Boronat I, Massot-Cladera M, Knipping K, Garssen J, Franch À, et al. Effect of rotavirus infection and 2’-fucosyllactose administration on rat intestinal gene expression. Nutrients. 2023;15(8):1996. https://doi.org/10.3390/nu15081996.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Azagra-Boronat I, Massot-Cladera M, Knipping K, Van’t Land B, Stahl B, Garssen J, et al. Supplementation with 2’-FL and scGOS/lcFOS ameliorates rotavirus-induced diarrhea in suckling rats. Front Cell Infect Microbiol. 2018;8:372. https://doi.org/10.3389/fcimb.2018.00372.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Zhou X, Li Y, Li T, Cao J, Guan Z, Xu T, et al. Portulaca oleracea L. polysaccharide inhibits porcine rotavirus in vitro. Animals (Basel). 2023;13(14):2306. https://doi.org/10.3390/ani13142306.

  155. Alfajaro MM, Rho M-C, Kim H-J, Park J-G, Kim D-S, Hosmillo M, et al. Anti-rotavirus effects by combination therapy of stevioside and Sophora flavescens extract. Res Vet Sci. 2014;96:567–75. https://doi.org/10.1016/j.rvsc.2014.03.011.

    Article  CAS  PubMed  Google Scholar 

  156. Cui Q, Fu Q, Zhao X, Song X, Yu J, Yang Y, et al. Protective effects and immunomodulation on piglets infected with rotavirus following resveratrol supplementation. PLoS ONE. 2018;13:e0192692. https://doi.org/10.1371/journal.pone.0192692.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Yu B, Jiang Y, Zhang B, Yang H, Ma T. Resveratrol dimer trans-ε-viniferin prevents rotaviral diarrhea in mice by inhibition of the intestinal calcium-activated chloride channel. Pharmacol Res. 2018;129:453–61. https://doi.org/10.1016/j.phrs.2017.11.016.

    Article  CAS  PubMed  Google Scholar 

  158. Banerjee S, Sarkar R, Mukherjee A, Miyoshi S-I, Kitahara K, Halder P, et al. Quercetin, a flavonoid, combats rotavirus infection by deactivating rotavirus-induced pro-survival NF-κB pathway. Front Microbiol. 2022;13:951716. https://doi.org/10.3389/fmicb.2022.951716.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Song L, Zhong P, Zhu X, Zhou R, Gao M, Lan Q, et al. The anti-rotavirus effect of baicalin via the gluconeogenesis-related p-JNK-PDK1-AKT-SIK2 signaling pathway. Eur J Pharmacol. 2021;897:173927. https://doi.org/10.1016/j.ejphar.2021.173927.

    Article  CAS  PubMed  Google Scholar 

  160. Kim J-H, Kim K, Kim W. Genipin inhibits rotavirus-induced diarrhea by suppressing viral replication and regulating inflammatory responses. Sci Rep. 2020;10:15836. https://doi.org/10.1038/s41598-020-72968-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Chen X, Liu L, Chen W, Qin F, Zhou F, Yang H. Ziyuglycoside II inhibits rotavirus induced diarrhea possibly via TLR4/NF-κB pathways. Biol Pharm Bull. 2020;43:932–7. https://doi.org/10.1248/bpb.b19-00771.

    Article  CAS  PubMed  Google Scholar 

  162. Guo SC, Yu B, Jia Q, Yan HY, Wang LQ, Sun FF, et al. Loureirin C extracted from Dracaena cochinchinensis S.C. Chen prevents rotaviral diarrhea in mice by inhibiting the intestinal Ca2+-activated Cl channels. J Ethnopharmacol. 2024;318:117077. https://doi.org/10.1016/j.jep.2023.117077.

    Article  CAS  PubMed  Google Scholar 

  163. Yang H, Oh K-H, Kim HJ, Cho YH, Yoo YC. Ginsenoside-Rb2 and 20(S)-Ginsenoside-Rg3 from Korean red ginseng prevent rotavirus infection in newborn mice. J Microbiol Biotechnol. 2018;28:391–6. https://doi.org/10.4014/jmb.1801.01006.

    Article  CAS  PubMed  Google Scholar 

  164. Huang H, Liao D, He B, Pu R, Cui Y, Zhou G. Deoxyshikonin inhibited rotavirus replication by regulating autophagy and oxidative stress through SIRT1/FoxO1/Rab7 axis. Microb Pathog. 2023;178:106065. https://doi.org/10.1016/j.micpath.2023.106065.

    Article  CAS  PubMed  Google Scholar 

  165. Zhang B, Wang Y, Jiang C, Wu C, Guo G, Chen X, et al. Valeriana jatamansi jones inhibits rotavirus-induced diarrhea via phosphatidylinositol 3-kinase/protein kinase B signaling pathway. J Microbiol Biotechnol. 2021;31:1115–22. https://doi.org/10.4014/jmb.2003.03006.

    Article  CAS  PubMed  Google Scholar 

  166. Ko E-A, Jin B-J, Namkung W, Ma T, Thiagarajah JR, Verkman AS. Chloride channel inhibition by a red wine extract and a synthetic small molecule prevents rotaviral secretory diarrhoea in neonatal mice. Gut. 2014;63:1120–9. https://doi.org/10.1136/gutjnl-2013-305663.

    Article  CAS  PubMed  Google Scholar 

  167. Alfajaro MM, Kim H-J, Park J-G, Ryu E-H, Kim J-Y, Jeong Y-J, et al. Anti-rotaviral effects of Glycyrrhiza uralensis extract in piglets with rotavirus diarrhea. Virol J. 2012;9:310. https://doi.org/10.1186/1743-422X-9-310.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Tam KI, Roner MR. Characterization of in vivo anti-rotavirus activities of saponin extracts from Quillaja saponaria Molina. Antiviral Res. 2011;90:231–41. https://doi.org/10.1016/j.antiviral.2011.04.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. He ST, He FZ, Wu CR, Li SX, Liu WX, Yang YF, et al. Treatment of rotaviral gastroenteritis with Qiwei Baizhu powder. World J Gastroenterol. 2001;7:735–40. https://doi.org/10.3748/wjg.v7.i5.735.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Jiang C, Yang H, Chen X, Qiu S, Wu C, Zhang B, et al. Macleaya cordata extracts exert antiviral effects in newborn mice with rotavirus-induced diarrhea via inhibiting the JAK2/STAT3 signaling pathway. Exp Ther Med. 2020;20:1137–44. https://doi.org/10.3892/etm.2020.8766.

  171. Swanson KS, Gibson GR, Hutkins R, Reimer RA, Reid G, Verbeke K, et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat Rev Gastroenterol Hepatol. 2020;17:687–701. https://doi.org/10.1038/s41575-020-0344-2.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Rioux KP, Madsen KL, Fedorak RN. The role of enteric microflora in inflammatory bowel disease: human and animal studies with probiotics and prebiotics. Gastroenterol Clin North Am. 2005;34(3):465-82, ix. https://doi.org/10.1016/j.gtc.2005.05.005.

    Article  PubMed  Google Scholar 

  173. Bengmark S. Bioecologic control of the gastrointestinal tract: the role of flora and supplemented probiotics and synbiotics. Gastroenterol Clin North Am. 2005;34:413–36, viii. https://doi.org/10.1016/j.gtc.2005.05.002.

    Article  PubMed  Google Scholar 

  174. Rigo-Adrover M, Saldaña-Ruíz S, van Limpt K, Knipping K, Garssen J, Knol J, et al. A combination of scGOS/lcFOS with Bifidobacterium breve M-16V protects suckling rats from rotavirus gastroenteritis. Eur J Nutr. 2017;56:1657–70. https://doi.org/10.1007/s00394-016-1213-1.

  175. Rigo-Adrover MM, van Limpt K, Knipping K, Garssen J, Knol J, Costabile A, et al. Preventive effect of a synbiotic combination of galacto- and fructooligosaccharides mixture with Bifidobacterium breve M-16V in a model of multiple rotavirus infections. Front Immunol. 2018;9:1318. https://doi.org/10.3389/fimmu.2018.01318.

  176. Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, et al. Preventive effect of a postbiotic and prebiotic mixture in a rat model of early life rotavirus induced-diarrhea. Nutrients. 2022;14(6):1163. https://doi.org/10.3390/nu14061163.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Rigo-Adrover MM, Knipping K, Garssen J, van Limpt K, Knol J, Franch À, et al. Prevention of rotavirus diarrhea in suckling rats by a specific fermented milk concentrate with prebiotic mixture. Nutrients. 2019;11(1):189. https://doi.org/10.3390/nu11010189.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Pandey KR, Naik SR, Vakil BV. Probiotics, prebiotics and synbiotics- a review. J Food Sci Technol. 2015;52:7577–87. https://doi.org/10.1007/s13197-015-1921-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Boyle RJ, Robins-Browne RM, Tang MLK. Probiotic use in clinical practice: what are the risks? Am J Clin Nutr. 2006;83:1256–64. https://doi.org/10.1093/ajcn/83.6.1256

    Article  CAS  PubMed  Google Scholar 

  180. de Vrese M, Schrezenmeir J. Probiotics, prebiotics, and synbiotics. In: Stahl U, Donalies UE, Nevoigt E, editors. Food Biotechnology. Advances in biochemical engineering/biotechnology, vol 111. Berlin, Heidelberg: Springer; 2008. https://doi.org/10.1007/10_2008_097.

    Article  PubMed  Google Scholar 

  181. Duan T, Du Y, Xing C, Wang HY, Wang R-F. Toll-like receptor signaling and its role in cell-mediated immunity. Front Immunol. 2022;13:812774. https://doi.org/10.3389/fimmu.2022.812774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Kumar A, Helmy YA, Fritts Z, Vlasova A, Saif LJ, Rajashekara G. Anti-rotavirus properties and mechanisms of selected gram-positive and gram-negative probiotics on polarized human colonic (HT-29) cells. Probiotics Antimicrob Proteins. 2023;15:107–28. https://doi.org/10.1007/s12602-021-09884-3.

    Article  CAS  PubMed  Google Scholar 

  183. Perez EE, Orange JS, Bonilla F, Chinen J, Chinn IK, Dorsey M, et al. Update on the use of immunoglobulin in human disease: a review of evidence. J Allergy Clin Immunol. 2017;139:S1–46. https://doi.org/10.1016/j.jaci.2016.09.023.

    Article  CAS  PubMed  Google Scholar 

  184. Mao X, Dou Y, Fan X, Yu B, He J, Zheng P, et al. The effect of dietary Yucca schidigera extract supplementation on productive performance, egg quality, and gut health in laying hens with Clostridium perfringens and coccidia challenge. Poult Sci. 2023;102:102822. https://doi.org/10.1016/j.psj.2023.102822.

  185. Chelakkot C, Ghim J, Ryu SH. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med. 2018;50:1–9. https://doi.org/10.1038/s12276-018-0126-x.

    Article  CAS  PubMed  Google Scholar 

  186. Buccigrossi V, Poeta M, Cioffi V, Terranova S, Nunziata F, Lo Vecchio A, et al. Lacticaseibacillus rhamnosus GG counteracts rotavirus-induced ion secretion and enterocyte damage by inhibiting oxidative stress and apoptosis through specific effects of living and postbiotic preparations. Front Cell Infect Microbiol. 2022;12:854989. https://doi.org/10.3389/fcimb.2022.854989.

  187. Bruno C, Paparo L, Pisapia L, Romano A, Cortese M, Punzo E, et al. Protective effects of the postbiotic deriving from cow’s milk fermentation with L. paracasei CBA L74 against Rotavirus infection in human enterocytes. Sci Rep. 2022;12:6268. https://doi.org/10.1038/s41598-022-10083-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Paparo L, Tripodi L, Bruno C, Pisapia L, Damiano C, Pastore L, et al. Protective action of Bacillus clausii probiotic strains in an in vitro model of Rotavirus infection. Sci Rep. 2020;10:12636. https://doi.org/10.1038/s41598-020-69533-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to all the members who edited and reviewed the manuscript.

Funding

This study was financially supported by 14th Five-Year for Breeding Research Project of Sichuan Provincial Department of Science and Technology (2021YFYZ0008).

Author information

Authors and Affiliations

Authors

Contributions

The authors’ responsibilities are as follows – all authors: contributed to the design and concept of this review; HY, XF, and XM: drafted the manuscript; all authors: critically reviewed the draft, and provided detailed commentary and changes; XM: had primary responsibility for final content. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Xiangbing Mao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that there are no conflicts of interest.

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

Yang, H., Fan, X., Mao, X. et al. The protective role of prebiotics and probiotics on diarrhea and gut damage in the rotavirus-infected piglets. J Animal Sci Biotechnol 15, 61 (2024). https://doi.org/10.1186/s40104-024-01018-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-024-01018-3

Keywords