key: cord-0849246-by2i8fag authors: Yuan, Lijuan; Hensley, Casey; Mahsoub, Hassan M.; Ramesh, Ashwin K.; Zhou, Peng title: Microbiota in viral infection and disease in humans and farm animals date: 2020-04-24 journal: Prog Mol Biol Transl Sci DOI: 10.1016/bs.pmbts.2020.04.005 sha: c503d80fc5c84fd80cf58116c604ccf56f5d6178 doc_id: 849246 cord_uid: by2i8fag Abstract The influence of the microbiota on viral infection susceptibility and disease outcome is undisputable although varies among viruses. The purpose of understanding the interactions between microbiota, virus, and host is to identify practical, effective, and safe approaches that target microbiota for the prevention and treatment of viral diseases in humans and animals, as currently there are few effective and reliable antiviral therapies available. The initial step for achieving this goal is to gather clinical evidences, focusing on the viral pathogens—from human and animal studies—that have already been shown to interact with microbiota. The subsequent step is to identify mechanisms, through experimental evidences, to support the development of translational applications that target microbiota. In this chapter, we review evidences of virus infections altering microbiota and of microbiota enhancing or suppressing infectivity, altering host susceptibility to certain viral diseases, and influencing vaccine immunogenicity in humans and farm animals. The human and animal body surface and cavities are inhabited by a large number of commensal microorganisms, collectively referred to as the microbiota. 1 This network of microbial communities recently has been revealed to have significant impacts on host health and immunity against infectious diseases. 1 The microbiota has come to the forefront of infection and immunity research, as there are mounting evidence suggesting that microbiota plays an important role in the development of host immune system and immunity. 2 This discovery has provided many research opportunities regarding the role of microbiota in viral diseases, in which virologists can explore how the microbiota suppresses or enhances the infectivity of viruses, the host susceptibility, and how to harness this information for targeted antiviral therapeutics. This new field of research on microbiomevirus-host interaction is made possible by the massive advances in the nucleotide sequencing approaches, metagenomic analysis and data mining, and bioinformatics analytical software. 3, 4 Most available data on the microbiome describe the gut microbiota; however, microbiota is pertinent not only to enteric viral diseases, but also to viral diseases that affect every organ and system in the body. Although the majority of the microbiota resides in the gut, there are large populations that also reside in the mouth, urinary tract and on the skin. [5] [6] [7] Most principles learned from the study of gut microbiome apply to microbial habitats throughout the body. The study of microbiota-virus-host interactions in each specific location of the body allows the understanding of localized immunity and susceptibility to invading viruses and how the local microbiota factor into these processes. In addition to bacteria, developments in sequencing techniques have led to the expansion of knowledge in the viral aspect of the microbiome, deemed the virome. The virome is comprised of all prokaryotic and eukaryotic viruses found in and on humans and animals. The role of the virome in health and disease is poorly understood at this time. Studies of the role of microbiome in viral diseases have focused almost exclusively on bacteria instead of viruses. In this chapter, we review and discuss how the microbiota (bacteria) enhances or suppresses infectivity and alters host susceptibility to certain viral diseases and also how virus infection alters microbiota in humans and farm animals, which include poultry, swine, and ruminant species. Because of the exceedingly fast-growing literature in this field, we only chose a few major viral diseases as examples for each species (Table 1) . We also provide brief background information on virome in general health in each species. Influenza is a common viral infection responsible for the seasonal pandemics that sweep the globe each year. Influenza viruses possess a segmented negative sense, single-stranded RNA genome and belong to the family Orthomyxoviridae. Influenza virus infection causes acute respiratory inflammation in humans with symptoms such as high fever, body aches, fatigue, and can result in death. Up to one-half million deaths occur annually, with close to five million reported cases. 45 The best defense thus far is the yearly strain-specific vaccine. The efficacy of this vaccine is low due to several factors, including continual antigenic drift (minor mutational changes that occur over time) and annual vaccine mismatches to circulating strains. 45 The low efficacy of the vaccine coupled with the ease of transmission and 12 ) Lactobacillus (Gosmann et al. 13 ), Lactobacillus crispatus (Fredricks et al. 11 ) Human norovirus (HuNoV) Escherichia coli (Nelson et al. 14 Ruminococcaceae, Faecalibacterium (Rodriguez-Diaz et al. 15 ) Gn pig Bacteroides, Lactobacillus (Lei et al. 16 ) - Human Bacteroidetes, Bacteroides, Prevotella (Harris et al. 17 ) Streptococcus bovis (Harris et al. 17 ), Clostridium cluster XI, Proteobacteria, Serratia, Escherichia coli (Harris et al. 18 ) Gn pig Proteobacteria (Twitchell et al. 19 ) Bacteroidetes (Twitchell et al. 19 ), Lactobacillus rhamnosus GG, Bifidobacterium lactis Bb12 (Vlasova et al. 20 ) Human Proteobacteria, Enterobacteriaceae, Veillonellaceae, Streptococcaceae (Wei et al. 21 ) Bacteroidetes (Wei et al. 21 ) Human hepatitis C (HCV) Human Bacteroidetes, Prevotella, Acinetobacter, Veillonella, Phascolarctobacterium (Aly et al. 22 ) severity of disease among the very young, very old, and other immunocompromised individuals has made alternative therapies attractive. 46 Lee et al. conducted a longitudinal study on the human nose/throat microbiome and the potential influence it may have on the susceptibility of the human host to influenza infection. 2 They demonstrated that the level of host susceptibility to influenza is associated with microbial community makeup within the nose/throat. 2 Nose/throat samples were assigned nasal/oropharyngeal (NOP) community state types (CST) and samples from study participants were assigned to one of five groups based on bacterial taxa present. All five of these groups differed significantly in their relative abundance of 15 different bacterial genera. 2 When looking at the implications of certain taxa within the NOP CSTs on susceptibility to influenza, results showed a negative association between Bacteroidetes and NOP CST 4, of which the highest relative abundance was both Streptococcus and Neisseria, on host susceptibility to influenza infection. A positive correlation was noted between Alloprevotella and Prevotella and infection susceptibility. 43 Lee et al. also demonstrated notable differences in the microbiome of people of different ages in regards to concurrent influenza infection. Of the NOP CSTs defined in the study, NOP CST 4, which was associated with a decreased vulnerability to influenza infection, was not as abundant or stable in children and showed a consistent increase in prevalence with increased age of a given participant. 2 This suggests the greater occurrence of influenza among young children can be attributed to immature and underdeveloped microbial networks in the nose/throat. 2 The microbiome of the nasopharynx may also play a role in severity of disease and the development of secondary bacterial infections related to influenza. Ding et al. showed that both influenza A and B infected individuals had a nasopharynx microbiome containing taxa such as Dolosigranulum and Staphylococcus. 8 These genera have been correlated with the development of pneumonia, a common sequela of influenza in elderly patients, and Dolosigranulum also has been identified as a causative agent in septicemia. [47] [48] [49] This study classified nasopharyngeal (NP) swabs from infected and healthy subjects into four different NP types. NP type A was most prevalent in influenza-infected patients, and consisted of the two bacteria mentioned above. 8 NP type B was significantly enriched in the healthy control group and was dominated by Corynebacterium, NP type C was dominated by Moraxella, and NP type D had a high prevalence of Staphylococcus. 8 Corynebacterium has been shown to hinder Streptococcus pneumoniae growth in vitro and has been demonstrated to be abundant in children lacking S. pneumoniae nasal colonization. 9 Corynebacterium has also been shown to have antimicrobial effects against S. aureus. 50 The discoveries made in the study conducted by Ding et al. support the previously mentioned study's findings, with NP type B being dominated by Corynebacterium with relatively low levels of Staphylococcus. 8 These results reiterate the need for more focused studies on the therapeutic potentials of certain bacteria among microbiota and how they can alter vulnerable patient outcomes. A recent study using a mouse model of influenza virus infection has shown that fermentable dietary fiber increased survival of influenza-infected mice by dampening airway inflammation through several immune modulating mechanisms. 10 Dietary fiber inulin altered the intestinal microbiota in mice and exhibited butyrogenic properties. In inulin-treated mice, the microbiome structure was significantly improved, and beneficial bacteria that produce short-chain fatty acids (SCFAs), including Bifidobacteriaceae, became dominant. Inulin or SCFAs intake prevented excessive neutrophil influx into the airways by blunting the levels of CXCL1 produced by lung monocytes and macrophages. In addition, inulin and SCFAs increased antiviral immunity through CD8 + T cell activation. 10 Another study in a mouse model of severe respiratory syncytial virus (RSV) reported similar findings that fermentable dietary fiber can reduce virus replication, protect the lungs against inflammation and pathological changes induced by RSV infection through microbiota-derived acetate in mice. 51 Since fermentable dietary fiber play a role in regulating immunity by promoting the growth of SCFA-producing bacteria, taking fermentable dietary fiber and reducing the excessive immune response and lung pathology caused by the viruses are a causal relationship, not just a correlation. To date, there are no human clinical studies on using fermentable dietary fiber to prevent influenza or other respiratory viral infection or to reduce the disease severity. This area of translational research warrants further investigation. According to the World Health Organization, 1.7 million new HIV cases occurred globally in 2018, and nearly 38 million people currently are living with the infection. 52 This disease typically is spread through sexual contact, and the disruption of cervicovaginal microbiota has been correlated with increased susceptibility to HIV infection. 53 Investigation of the cervicovaginal microbiota's influence on HIV infection susceptibility could lead to the development of therapeutic agents for preventing HIV in especially at-risk communities, such as those in sub-Saharan Africa, where approximately 24 million people are infected, and young women are eight times more likely than men to acquire the virus. 13 Though the majority of the human microbiota resides in the gut, the cervix and vagina maintain their own commensal microbial communities. 13 A cervicovaginal microbiota dominated by Lactobacillus, specifically L. crispatus, is associated with many health benefits, including reduced risk of contracting HIV. 52 When cervicovaginal dysbiosis occurs, Lactobacillus communities are disrupted and other resident microbes, such as Gardnerella or Mobiluncus, become the predominant species, resulting in bacterial vaginosis. 11 A study conducted by Fredricks et al. demonstrated increased bacterial diversity in women with bacterial vaginosis, and a relatively "normal" vaginal flora dominated by L. crispatus in women without the disease. 11 A HIV study by Gosmann et al. showed that none of the participants who contracted HIV had a cervicovaginal microbiota dominated by Lactobacillus prior to infection. 13 They also demonstrated that women who maintained the most diverse microbiome were significantly more likely to contract HIV than those with less diverse communities dominated by L. crispatus. 13 These results are consistent with the notion that a Lactobacillus dominated cervicovaginal microbiota for the prevention of HIV is critical. Several anaerobic bacteria also were implicated in the increased susceptibility to HIV infection in this study, including Prevotella and Sneathia. 13 These bacteria were shown to increase inflammation in the genital tract, further decreasing the participants' immune robustness against HIV infection. 13 Bacterial vaginosis and high diversity/low L. crispatus presence in the vagina has been associated with increased susceptibility to HIV infection, partly due to resulting genital inflammation and the vulnerability of cell types that dominate the inflamed genital tract. 12, 54 Genital inflammation has been shown to lead to disturbances in the vaginal epithelium, making women especially susceptible to HIV infection. 13, 54 Anahtar et al. found that a diverse cervicovaginal microbiota is strongly linked to genital inflammation, including an increase in proinflammatory cytokines. 12 Fusobacterium, Aerococcus, Sneathia, Gemella, Mobiluncus, and Prevotella were all found to have a significant association with an increase in inflammatory cytokines. 12 These results were confirmed in vitro when human epithelial cells co-cultured with these bacteria, specifically Prevotella amnii, Mobiluncus mulieris, Sneathia amnii, and Sneathia sanguinegens were shown to have a marked increase in cytokine secretion than those co-cultured with L. crispatus, which showed no detectable levels of cytokine secretion. 12 Women with increased proinflammatory cytokine levels attributed to highly diverse cervicovaginal microbiota also had higher levels of CD4+ T cells, one of HIV's main target cells, establishing a strong link between increased bacterial diversity and an increased risk of contracting HIV. 12 Along with cervicovaginal microbiota diversity and the implications it has on HIV status and pathogenesis, gut microbiota diversity has also been identified as playing a significant role in these areas in regards to HIV. Because HIV is known to replicate in gut lymphoid tissues, it is relevant to discuss the implications the disruption of the gut microbiota and subsequent inflammation may have on HIV pathogenesis and vice versa. 55 HIV has been previously associated with gut dysbiosis, and again represents a potential tool that can be used to mitigate the impact of HIV infection. 56 In a meta-analysis performed by Tuddenham et al., it was demonstrated that diversity of the microbiome in HIV-1 positive individuals was significantly decreased prior to controlling using demographic categories. 56 When controlling for sexual preferences and gender, this held true for men who have sex with women (MSW) as well as men. 56 Men who have sex with men (MSM) showed no significant association with microbiota diversity and HIV status; however, when controlling for MSM, HIV positive men were more likely to have a decreased diversity than HIV positive women. 56 This study highlights the complicated relationship between many factors involved in HIV infection, including diversity of the microbiota. In future studies, it will be beneficial to investigate the role the other factors play in diversity and how therapeutics can be tailored based on factors like gender and sexual preference. In a study by Qing et al., researchers demonstrated how diversity of specific short-chain fatty acid (SCFA) producing bacteria can influence HIV progression through intestinal injury. 57 Because SCFAs are known to contribute to immune homeostasis and the expression of antimicrobial peptides by epithelial cells in the gut, they have been implicated in the progression of HIV infection. 57 This study found that certain species in the Firmicutes and Bacteroidetes phyla, who produce various SCFAs such as butyric and valeric acid, were much less abundant in the HIV positive group. 57 Butyric acid has been shown to reduce colonic inflammation in patients with inflammatory bowel disease, and the reduction of bacterial producers of this metabolite may contribute to increased intestinal injury and permeability, ultimately leading to bacterial translocation from the gut resulting in systemic inflammation. [58] [59] [60] [61] [62] Several factors contribute to the enhancement of HIV infection, including cervicovaginal and gut dysbiosis. The resulting inflammatory and immune-related sequelae can have significant impact on the progression and outcome of the disease. The prevention and management of HIV infection in developed countries has improved dramatically since the first cases were reported in the 1980s; however, there are still millions of people in underdeveloped areas who would benefit from microbiota research in regards to HIV prevention. 52 The findings in the above studies show promise for the development of accessible and personalized medicine alternative to help these areas recover from a several decades long HIV epidemic. The majority of the world's acute gastroenteritis cases are caused by HuNoV, an enteric viral pathogen spread primarily via the fecal-oral route. 63, 64 HuNoVs are non-enveloped RNA viruses with a positive-sense singlestranded genome in the family Caliciviridae. 65 . Due to the ease of transmission, cases of HuNoV tend to be concentrated and typically are more problematic in children, especially in low-income regions. 63 In more developed countries, HuNoV is known to cause outbreaks of gastroenteritis in all age groups. 63 HuNoV is of great concern in public health practice, but due to the lack of reliable and well-established in vitro cell culture systems, the development of therapeutics has been slow. 66 Certain microbial populations can positively contribute to gut health and microbial balance, and one of the most intriguing benefits of these bacteria is their ability to influence outcomes of infectious diseases. 67 When the gut microbiota is disrupted, the protection potential is diminished and the host becomes susceptible to numerous infections. 14,67 Nelson et al. conducted a study whose results suggest that HuNoV infections are capable of causing significant alterations in human gut microbiota. 14 In this study, 20% of HuNoV-infected subjects showed significant changes in their gut microbiota, including an increase in Proteobacteria populations, especially Escherichia coli, and a decreased bacterial diversity. 14 Proteobacteria are associated with gut microbiota disruption, further suggesting the study participants had a disrupted microbiota at the time of sampling. 14 These patients were identified as being at risk for developing postinfection disease, such as irritable bowel syndrome. 68 infection Along with establishing the need for a stable gut microbiota as a natural defense against HuNoV infection, other studies have identified a potential link between human genetic signatures, such as histo-blood group antigen (HBGA), specifically secretor status, the gut microbiome, and their combined influence on immunity to HuNoV infection. Secretor status of an individual is determined by the functionality of the FUT2 gene, which is responsible for producing an enzyme critical to generating glycans present in saliva and mucus. 69 These glycans can serve as an infection initiating binding site of certain HuNoV strains. 69 When both alleles of this gene are altered and nonfunctional, a person is deemed a non-secretor. 69 Nonsecretors have been shown to be highly resistant to symptomatic HuNoV infections due to the mutation in the FUT2 gene, which diminishes the norovirus's ability to bind to receptors in the intestine, thus preventing HuNoV from entering cells and causing infection. 69 Several studies also have shown secretor status to be linked to microbiota composition. 15, 70 In a study by Rodriguez-Diaz et al., human patients were shown to have a correlation between their gut microbiota diversity, secretor status and susceptibility to HuNoV infection. 15 Host anti-HuNoV salivary IgA titers were obtained from the participants and were used as an indicator of previous infection susceptibility. 15 Though no significant differences in phyla composition of the gut (obtained through stool samples) and secretor status were found, significant family-level differences were present between nonsecretors and secretors; with significantly higher levels of Prevotellaceae and Paraprevotellaceae found in non-secretors. 15 The study then involved separating the obvious groupings into operational taxonomic groups (OTUs) and were then analyzed for diversity levels. Sixteen of the more than 5000 groups were found to have significant differences, and consisted of Firmicutes and Bacteroidetes phyla, found in non-secretors. There were also significant differences in richness of the sampled microbiota, though they were not significant in regards to secretors versus non-secretors. 15 Firmicutes, Bacteroidetes, Actinobacteria, and Verrucomicrobia were the most abundant phyla categorized from both secretors and non-secretors. This study also found anti-HuNoV IgA levels were significantly correlated with secretor versus non-secretor status, with secretors maintaining higher levels of the two groups. 15 The viral infectivity and host susceptibility to HuNoV was negatively correlated to the level of Ruminococcaceae and Faecalibacterium present in host samples. When these insights are combined with the negative correlation between anti-norovirus IgA titers and susceptibility to HuNoV infection, the notion that host microbiome, secretor status and susceptibility are all reliant on each other to some extent is apparent. 15 More thorough research is needed to evaluate other factors that simultaneously play a role in microbiome composition of all secretors and nonsecretors and how this influences host immunity. 15 This is especially important for the determination of the best strategies to protect those in low-income areas in which HuNoV is of greatest public health concern, and the microbiome is likely an effective target for future personalized therapies. 63 (Gn) pig model To study the role of microbiota in response to virus infection and vaccines, animal models that are without a preexisting microbiota and can be colonized with well-defined microbiota, (i.e., Gn animals) are an indispensable tool. Gn animals have played a critical role in the understanding of interactions among microbiota, viral pathogen or vaccine antigen, and the host. 71 The neonatal Gn pig model is well suited for the study of HuNoV and microbiome interaction because Gn pig model of HuNoV infection reflects HuNoV biology in terms of supporting the natural oral route of infection and results in diarrhea, transient viremia, and fecal virus shedding. 72, 73 Recently, we established a human gut microbiota (HGM) transplanted Gn pig model of HuNoV infection and disease, using a well-characterized stool sample from a healthy infant as HGM transplant and a HuNoV GII.4/2006b strain for virus inoculation. 16 Compared to germ-free Gn pigs, HuNoV inoculation in HGM transplanted Gn pigs resulted in increased HuNoV infection, characterized by significantly higher shedding titers and significantly longer mean duration of virus shedding. HuNoV infection also dramatically altered intestinal microbiota at the phylum and genus levels in HGM transplanted Gn pigs. At the phylum level, Proteobacteria (95.6% versus 56.5%) and Firmicutes (3.6% versus 0.5%) significantly decreased, while Bacteroidetes (0.1% versus 42.9%) significantly increased; at the genus level, Enterococcus, Bifidobacterium, Clostridium, Ruminococcus, and Anaerococcus significantly decreased, while Bacteroides and Lactobacillus significantly increased in HGM transplanted pigs infected with HuNoV compared to HGM transplanted pigs without HuNoV infection. In summary, enhanced GII.4 HuNoV infection was observed in the presence of HGM, and significant intestinal microbiota alterations were observed under HuNoV infection in HGM transplanted Gn pigs. 16 The mechanisms underlying the enhancing effect of HGM on HuNoV infection are probed using transcriptome analysis of the ileum tissues of the Gn pigs. One of the most important impact of HGM is the down regulation of interferon (IFN)-λ gene IFN-1L expression. IFN-λ is known to play a critical role in the host defense against enteric viruses, including rotavirus, reovirus 74 and norovirus. 75 Rotaviruses belong to the genus Rotavirus within the family Reoviridae, which are characterized by a segmented double-stranded RNA (dsRNA) genome. HRV is the leading cause of severe, dehydrating diarrhea among children under 5 years of age worldwide. 76 Annual mortality rates due to RV infections have declined from 528,000 in 2000 to 215,000 in 2013 76 owing to the implementation of HRV vaccines RotaTeq ® and Rotarix ® in the national immunization program in many countries. These two vaccine vaccines are highly efficacious (80-90%) in high income countries, but the efficacies are significantly reduced (40-60%) in low-income countries. Studies indicated that several factors, including microbiota dysbiosis, 77 concurrent uses of poliovirus oral vaccines, 78,79 enterovirus infections, 80 and malnutrition 81 contributed to the observed impaired efficacy of the oral RV vaccines, 79 likely by changing intestinal environments and homeostasis. 82, 83 Harris et al. 17 analyzed the serum IgA antibody responses to Rotarix ® in 78 Ghanaian infants and their fecal microbiome. Comparing between the 39 responder and non-responder pairs and to the Dutch infants, the overall microbiome composition was significantly different, with the responders more similar to healthy Dutch infants than nonresponders. Responses to Rotarix ® correlated with an increased abundance of Streptococcus bovis and the lack of response is correlated with the high abundance of Bacteroidetes phylum, especially several bacteria related to species from the Bacteroides and Prevotella genera. 17 Another study was reported for Pakistani infants. 18 IgA antibody responses to Rotarix ® correlated with higher abundance of bacteria belonging to Clostridium cluster XI and Proteobacteria, including bacteria related to Serratia and Escherichia coli. Abundance of these Proteobacteria was also significantly higher in Dutch infants when compared to non-responders in Pakistan. These clinical studies showed that the intestinal microbiota composition correlated with seroconversion rate in infants after vaccination and identified microbiome signatures of vaccine responder versus non-responder infants. Identification of key bacterial phylotypes that correlate with vaccine-induced responses could be used as indicator to predict which infants are at risk for vaccine failure and to design an intervention through modulating gut microbiota to improve rotavirus vaccine efficacy. 18, 77 A study of microbiome diversity in RataTeq vaccinated versus unvaccinated children in Spain did not find any differences. 84 2.4.2 Study of microbiota and immunogenicity of HRV vaccine in Gn pig model To clearly identify the role of microbiota in the immunogenicity of HRV vaccine, a Gn pig model of enteric dysbiosis was developed to evaluate the effects of different HGM on immune responses to HRV vaccination and protective efficacy. 19 Fecal samples collected from infants from Nicaragua were analyzed and classified as healthy (HHGM) or unhealthy (UHGM) based on enteropathy score and seroconversion status after vaccination with RotaTeq ® . Newborn Gn pigs were transplanted with the HHGM or UHGM, followed by oral vaccination with the live attenuated Wa HRV and challenged with virulent HRV. Prior to HRV challenge, HHGM in Gn pigs was predominantly composed of bacteria belonging to the phyla Firmicutes, Bacteroidetes, and Actinobacteria, whereas UHGM was composed of Proteobacteria and Firmicutes. When challenged, the severity of illness among vaccinated HHGM pigs was much lower compared to vaccinated UHGM pigs. Analysis of fecal microbiome post-challenge showed a significant increase in the abundance of Bacteroidetes in HHGM transplanted Gn pigs and a significant decrease in the abundance of Firmicutes, Proteobacteria, and Verrucomicrobia abundance. Stronger vaccine-induced immune responses (HRV-specific IgG, IgA and VN antibodies; and CD4 + IFN-γ + and CD8+ IFN-γ + T cells) were facilitated by HHGM, hence conferring a stronger protection against HRV disease among vaccinated HHGM pigs than UHGM pigs. 19 Similar results were also observed in Gn pigs colonized with Lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12, commensal bacteria commonly found as part of the microbiota of breast-fed infants. 20 Findings in Gn pigs are highly translatable to humans and as suggested by Dr. Harris "Correlate microbiome composition with vaccine effectiveness in appropriate experimental platforms will lead to the identification of safe, vaccine-supporting microbiota targets that are relevant to populations in need of improvement in vaccine-induced immunity." 18 Hepatitis B virus (HBV) and hepatitis C virus (HCV) are important public health issues that can lead to chronic infections and together are the most common causes of liver cirrhosis and hepatocellular carcinoma (HCC). Due to the well-documented crosstalk between the gut and liver, the microbiota and its immunomodulatory effects on HBV and HCV infections have recently become a topic of interest in the research community. 85,85a As was demonstrated by the previously discussed studies, certain phyla and families of beneficial bacteria appear to be underrepresented in patients with certain viral diseases. In a study by Wei et al., it was shown that patients with chronic HBV infection that had progressed to cirrhosis were lacking in Bacteroidetes composition in the gut, as well as showing increased Proteobacteria concentration, a phylum known to contain several pathogenic bacteria. 21 There was also marked prevalence of Enterobacteriaceae, Veillonellaceae, and Streptococcaceae, which made up for less than 1% of the gut microbiota sampled from healthy patients. 21 In two HCV studies by Aly et al. 22 and Bajaj 23 et al., low diversity of gut microbiome composition was associated with inflammation and disease. Interestingly, Aly et al.'s data showed chronic HCV patients had a significant increase in Bacteroidetes, as opposed to the previously discussed HBV patients, who had a marked decrease of this phylum in their composition. 21, 22 In the HCV patients, Prevotella was significantly increased, perhaps explaining the increase in Bacteroidetes phylum composition in this group. 22 This study also demonstrated a higher abundance of Proteobacteria in the healthy control group, dissimilar to the HBV study, which showed a higher abundance in the HBV group. 21, 22 In the HCV study, ill patients also displayed significant prevalence of Acinetobacter, Veillonella, and Phascolarctobacterium, as well, with no detection of Bifidobacterium genus, which was of relatively increased richness in two healthy patients. 22 Ruminococcus was also significantly enriched in healthy patients versus those with HCV, and this genus is considered to be largely beneficial in the gut. 22, 23 Though more research is needed in regards to the gut-liver axis and its role in immunity against HBV and HCV infections, the current literature has demonstrated a strong link between the gut microbiota and how the immune system functions in acute and chronic viral diseases. The disadvantage that remains is the microbiota's complexity, as is shown by the differences in compositions between patients with two different strains of the same virus. Fortunately, due to the development of vaccines, new hepatitis cases have dramatically decreased across the world, though chronic infections continue to pose a threat to those who do contract the virus. The gut microbiota may be a means to harness new therapies to relieve the increased mortality that comes with progression of chronic hepatitis infection. The human virome is constituted of viruses that infect our cells, virus-derived elements in our genome, and viruses that infect the broad array of other types of microorganisms that inhabit us. 86 The integration of endogenous retroviral DNA into the human genome accounts for 8% of the total genome, and is thought to promote development of certain diseases such as cancers and autoimmune diseases. 87 These sequences are also passed from mother to child, perpetuating the effects to offspring. 88 Another aspect of the virome is made up of prokaryotic viruses, or bacteriophages. Bacteriophages are the most numerous beings on Earth and have been found to promote health in humans, mostly through the development of phage therapy. Phage therapy has been utilized to combat antibiotic-resistant bacterial infections in humans. 89 Similar to the bacterial component of the microbiome, the virome contains certain makeup that varies between individuals. 90 Some common inhabitants of the human gut include Phycodnaviridae, Herpesviridae, Poxviridae, Mimiviridae, Iridoviridae, Adenoviridae, Anelloviridae, Astroviridae, Parvoviridae, Picornaviridae, Picobirnaviridae, and Reoviridae which have been identified in infant stool samples. [91] [92] [93] Viromes tend to stabilize over time as well, with humans establishing the individualized virome by around the age of 2 years, similar to the bacteriome. 94 Poultry contain the most economically important avian species because they provide a great source of animal protein. Poultry eggs and meat are crucial components to maintaining food security all over the globe. Fast-growing domestic poultry species are exposed to many environmental factors that can affect their health, performance, and productivity. To protect poultry from factors that may render them vulnerable to disease, scientists continuously investigate internal and external factors that may affect their health and productivity. The gastrointestinal tract has been a central part of these investigations as the gut health, function, and integrity are essential for overall wellbeing of birds. The gut microbiota, which contains tremendous numbers of bacterial species, has recently been extensively characterized in some avian species including chickens, turkeys, and ducks. [95] [96] [97] The influence of viral infections on the gut health and the homeostasis of commensal microbial populations of affected birds have been also investigated. Viral infections are also known risk factors for coinfection by other secondary, opportunistic pathogens as they cause damage to local and systemic immune defenses. 98 Here, we review the interplay between a number of viral pathogens and the gut microbiota of their respective natural avian hosts. The effect of LPAI infection on the homeostasis of the gut microbiota of poultry has been investigated very recently. Experimental infection of young chickens with H9N2 virus caused alterations in the intestinal microbiota composition. In infected chickens, the bacterial numbers of the phylum Proteobacteria were increased and the bacteria belonging to the genera Vampirovibrio, Pseudoflavonifractor, Ruminococcus, Clostridium cluster XIVb, and Isobaculum were enriched differentially. 24 Proteobacteria contains important gram-negative pathogenic bacteria belonging to the genera Escherichia, Shigella, Salmonella, and Helicobacter. 100 In uninfectedapparently healthy-chickens, differential enrichment of the genera Novosphingobium, Sphingomonas, Bradyrhizobium, and Bifidobacterium was observed. 24 Comparing the same chicken group before and after infection, Yitbarek et al. observed notable changes in the relative abundance of bacteria at various taxonomic levels. 24 Pre-infection, chickens were characterized by the increased abundance of two families and three genera of bacteria, while postinfection differential enrichment of members from one class, two orders, and one genus of bacteria were recorded. In a study by Li et al. 25 at the phylum level, the relative abundance of Proteobacteria was increased-as observed in the study above-while that of Firmicutes was decreased in H9N2 virus-infected young chickens. The relative abundance of endogenous bacterial genera within the family Enterobacteriaceae was increased; these genera included Escherichia-especially E. coli which colonized the ileum-, Clostridium, and Veillonella. On the other hand, a severe reduction in the probiotic organisms including the lactic acid-producing bacteria (i.e., Lactobacillus, Enterococcus, and Streptococcus), Candidatus Arthromitus, Bacteroides, Parabacteroides, and SMB53 was observed. 25 These findings clearly indicate that an influenza virus infection can disrupt the commensal microbial population in affected chickens. Waterfowl species are natural reservoir of IAV and infections in these birds, unlike chickens, are asymptomatic. The composition and diversity of cloacal microbiome and their relationship with the active status of IAV infection and affected host have been studied in five waterfowl species: A. acuta (northern pintail), A. americana (American wigeon), A. carolinensis (green-winged teal), A. clypeata (northern shoveler), and A. platyrhynchos (mallard). 101 Six bacterial phyla, namely Firmicutes, Proteobacteria, Bacteroidetes, Fusobacteria, Actinobacteria, and Tenericutes (ordered based on proportions of abundance) were consistently represented in IAVnegative, i.e., healthy, ducks of the above-mentioned five species. These phyla composed an average of 97.1% of the ducks' microbiota and thus were further investigated in the study. The relative abundance of bacterial phyla was drastically different within the various host species, with no unique patterns across species. However, green-winged teals and mallards showed the same pattern of abundance for all phyla, while northern shovelers showed the opposite pattern. The relationship among the various groups of bacterial taxa was evaluated using a microbiome diversity measurement, bacterial operational taxonomic unit (OTU) co-occurrence patterns. The observed OTUs in IAV-positive and -negative birds showed no consistent patterns among host species. Moreover, the number of observed OTUs by IAV infection status was only significant in two out of five duck species. 101 Findings from this study indicates that host genetics (and possibly other environmental factors, e.g., feed and habitat) may play a crucial role in the response of waterfowl to influenza virus infections. Another study on juvenile wild mallards (Anas platyrhynchos) only analyzed the differences in the cloacal microbiome of IAV-infected birds as compared to healthy ones. 26 Overall, the cloacal microbiome in healthy mallards was more rich, diverse, robust, and uniform than that in IAV-infected birds. Among 41 identified OTUs, IAV-infected mallards were found to have 12 versus 24 in healthy birds. Thus, IAV-negative mallards were richer in OTUs and had greater OTU diversity and evenness than those of IAVpositive group. The same six bacterial phyla listed above were also represented in both IAV-positive and -negative birds at similar levels of relative abundance. The higher differences were observed within the phyla Firmicutes, Proteobacteria, and Bacteroidetes. Members of the genus Streptococcus and species Veillonella dispar and Rothia mucilaginosa were major contributors to these differences. 26 In chickens with compromised microbiota, intestinal damage was caused by H9N2 virus infection, including reduced villus height, increased crypt depth, and reduced villus height:crypt depth ratio of the ileum, as well as lymphocytic infiltration of ileal mucosa. 25, 102 The breach of the intestinal wall integrity was consistent with downregulation in mRNA expression of the epithelial cell tight junction proteins (ZO-1, claudin 3, and occludin) and mucus layer proteins (trefoil factor 2 and mucin); the latter two have essential roles in preventing the inflammation and infection of the intestinal epithelium caused by invading bacteria. 25, 103, 104 Furthermore, the intestinal damage in H9N2 virus-infected chickens was associated by upregulation of mRNA expression of the proinflammatory cytokines IFN-γ, IL-22, IFN-α, and IL-17A by the intestinal epithelial cells. 25 The importance of balanced gut microbiota in modulating innate immune responses and protecting chickens against influenza virus infections was demonstrated by several studies. In intestinal microbiota-depleted chickens, H9N2 virus infection caused significant increase in oropharyngeal and cloacal virus shedding. Also, the mRNA expression of type I IFNs (i.e., INF-α and IFN-β) and IL-22 was suppressed in the respiratory and gastrointestinal tracts. 102 Newcastle disease virus (NDV, aka avian paramyxovirus 1) belongs to species Avian orthoavulavirus 1, genus Orthoavulavirus, family Paramyxoviridae, with a negative sense, single-stranded, monopartite RNA genome. 105 NDV affects a wide variety of avian species, among them the most susceptible and economically important are chickens and turkeys, 106 which can virtually be infected at all ages. Depending on the strain and tropism of the virus, NDV can cause a variety of symptoms involving the gastrointestinal tract, the nervous system, the respiratory system, and the reproductive system of affected birds, leading to up to 100% morbidity and mortality. 107 The adverse effect of NDV infection on the development of intestinal microbiota in chickens has been recently reported by Cui et al. 27 Although the study was performed in newly hatched chicks which were infected in ovo with NDV, it presented significant insight into the impact of such an infection on the intestinal microbial communities in chickens. In uninfected chicks, the duodenum had a richer (indicated by higher OTU counts and abundance indices) and more diverse microbiota than the cecum. In normal duodenum, the predominant bacterial phyla were Proteobacteria, Bacteroidetes, Firmicutes, and Actinobacteria, with six more less-dominant phyla. At the genus level, bacteria from Serratia, Escherichia, Chitinophagaceae unclassified, Saprospiraceae unclassified, and Lactobacillus were represented the most in the normal duodenum among other genera. In the cecum of uninfected chicks, the prevailing bacterial phyla were only Firmicutes and Proteobacteria, with five more marginally-represented phyla detected. At the genus level, Enterococcus, Paenibacillus, Erwinia, Escherichia, Lactobacillus, and Serratia constituted the great majority of the normal cecal microbiota. 27 Comparing NDV-infected with uninfected chicks, the richness and diversity of duodenal microbiota were lower in the former. The cecal microbiota was almost unaffected by the NDV infection. As for the microbial composition, both duodenal and cecal bacterial communities were adversely affected in a similar manner. An increase in Sinobacteraceae and Rhodoplanes family members was observed. Also, at the genus level, a massive loss was observed in members of the following families in both duodenum and cecum of NDVinfected chicks: Xanthomonadaceae, SC-I-84, Cytophagaceae, Bacteroidales, Chitinophagaceae, Gemm-1, Saprospiraceae, Ignavibacteriaceae, Rhizobiales, Sphingobacteriales, Ellin6067, Acidimicrobiales, and Pseudomonas. Moreover, a dramatic reduction was found in the abundance of Xanthomonadaceae and Cytophagaceae family members. 27 A significant reduction in the relative abundance of Serratia and Clostridium genera residing in the duodenum was recorded as opposed to greatly increased numbers in the ceca. This implied a possible translocation of these genera from duodenum to cecum of NDV-infected chicks. Moreover, authors proposed a translocation of several bacterial genera from the cecum to the duodenum of infected birds according to changes in their relative abundance. 27 Talebi et al. studied the effect of a mixture of probiotics (Lactobacillus acidophilus, Lactobacillus casei, Enterococcus faecium, and Bifidobacterium bifidium), administered in water, on the antibody response and production performance in NDV-vaccinated broiler chickens. 108 The study showed enhancement in birds' body weight and feed conversion rate; however, the humeral immune response to NDV vaccination was similar to that of untreated, vaccinated groups. Similar results were later revealed by Bautista-Garfias et al., 109 who studied the effect of oral administration of L. casei (ATCC7469 strain) on the humeral immune response to NDV vaccination and production performance in fighting roosters. Blood anti-NDV antibody levels in L. casei-treated group were comparable to those of the vaccine control birds. On the other hand, the natural mortality rate and body weight gain were significantly improved in the probiotic-treated group, compared with the untreated one. 109 The use of two (cecal and jejunal) bacterial isolates of L. reuteri PIA16 strain in feed, either alone or with a prebiotic (mannan oligosaccharide), demonstrated substantially enhanced humoral immune response to NDV vaccine in broiler chickens. The cell-mediated immune response; however, remained largely unaffected throughout the course of the experimental period. 28 Results from the probiotics studies indicate that these beneficial bacteria can boost the performance and overall health status in poultry. Although these studies showed generic positive impact on NDV-vaccinated chickens, the associated changes in the intestinal microbiota have not been examined. Moreover, whether the supplementation of these probiotics/prebiotics has an enhanced protective effect for NDV vaccines against the consequences of an NDV challenge is yet to be investigated. Infectious bursal disease virus (IBDV) belongs to the genus Avibirnavirus, family Birnaviridae, with a double-stranded, segmented RNA genome. 110 IBDV affects chickens at young ages and causes an acute, immunosuppressive disease, infectious bursal disease. IBDV infection causes depression of the local and humoral immune responses (due to a direct effect on B lymphocytes); and induces damages in the primary lymphoid organs, mainly the bursa of Fabricius, in addition to the spleen, thymus, and cecal tonsils. 111, 112 The role of IBDV in modulating the commensal microbiota in chickens has recently been studied. 29 Young chickens were infected with IBDV and the changes in the cecal bacterial communities were analyzed in comparison with uninfected, healthy birds. Regardless of the infection status, nine bacterial phyla were represented in the chicken cecal microbiota, with Firmicutes, Proteobacteria, Actinobacteria, and Bacteroidetes members forming more than 95% of total bacterial population in the cecum. At the family level, Lachnospiraceae and Ruminococcaceae composed the majority of cecal microbiota, with the relative abundance of Lachnospiraceae decreasing over time and that of Ruminococcaceae increasing over time for up to 7 weeks of age. At the genus level, an increase in the abundance of Faecalibacterium was observed till 4 weeks of age, and decreased thereafter. IBDV infection of young chickens caused immediate and obvious alteration in the composition of cecal microbiota. Changes were continuously seen over a three-week observation period postinfection, 29 though no certain pattern was observed. At the genus level, in comparison to uninfected group, IBDV-infected chickens had a lower abundance in the commensal Clostridium XlVa at day 3 postinfection, which became higher thereafter. On the other hand, after 7 days of IBDV infection chickens had a higher abundance of Faecalibacterium (Gram-positive bacteria) than uninfected ones, which became lower thereafter. A reduction in the abundance of Escherichia/Shigella (Proteobacteria) was observed in chickens until 3 weeks postinfection, which contributed to the overall decrease recorded for family Enterobacteriaceae. 29 These changes in the cecal microbiota were associated with IBDV-induced immunosuppression, the manifestations of which is described below. Another study showed that the infection of young chickens with IBDV or Campylobacter jejuni, either alone or combined, provoked significant modifications in the composition of gut microbiota, as compared with uninfected chickens. 30 Changes in the abundance of OTUs from the following bacterial genera were observed in the cecal contents of infected chickens: Campylobacter, Clostridium XIVa, Eubacterium, Faecalibacterium, Lachnospiracea incertae sedis, Lactobacillus, and Roseburia. The abundance of Campylobacter was increased in IBDV/C. jejuni-infected chickens and it was higher in birds inoculated with bacteria at 7 days versus 9 days postinfection. Obviously, the exposure time to a secondary pathogen following an immunosuppressive viral infection has a significant impact on its pathogenicity, colonization, and persistence, as is the case here with IBDV and C. jejuni. Similar observations have been reported for hemorrhagic enteritis of turkeys-a siadenovirus-based immunosuppressive disease-and secondary E. coli infections. 113 The particular effect of immunosuppression-inducing IBDV infection on the colonization and shedding of C. jejuni and Salmonella in the gastrointestinal tract of chickens has been investigated. Unlike C. jejuni, Salmonella is pathogenic in poultry; however, both are important pathogens in human and can cause severe food-borne gastroenteritis. 114, 115 Apart from C. jejuni and Salmonella infections, inoculation of young chickens with various strains of IBDV resulted in atrophy and pronounced lesions in the bursa of Fabricius, bursal B cell depletion, reduction or depletion in circulating B lymphocytes as well as those localized in the cecal lamina propria and cecal tonsils, and a reduced thickness of cecal mucosa. 27, 29, 32, 116, 117 Virus replication in the bursa, cecal tonsils, and cecum was detected for up to several weeks postinfection. 27, 29, 116 In IBDV-infected chickens with immunosuppression, unlike C. jejuni only-infected birds, the bacteria colonized the small intestines, and was detected at an earlier time point and at much higher titers in the cecum and cloaca (i.e., virus shedding). 27, 116 These results were consistent with the reduced B lymphocytes and the lack of C. jejunispecific IgG antibodies in the cecal lamina propria. 27 Likewise, IBDV infection of chickens exacerbated Salmonella multi-organ colonization, pathogenicity, fecal shedding, and persistence in chickens exposed to infections at early ages. 31, 32, 117 The highest Salmonella-mediated pathogenicity and mortality rates were recorded in chickens coinfected with IBDV and S. enteritidis, 31,32 but not S. typhimurium. 117 Both intestinal mucosal 41 and systemic (Salmonella-specific IgG) antibody responses were lower in chickens coinfected with IBDV and Salmonella, which coincided with the immunosuppressive effects described above. However, unlike C. jejuni, Salmonella clearance appeared not to be related to the antibody response. Numbers of localized T lymphocytes and cell-mediated immune responses did not seem to be affected by IBDV infections. 27,32 Turkey adenovirus 3, commonly known as hemorrhagic enteritis virus (HEV), belongs to genus Siadenovirus, family Adenoviridae, with a doublestranded, linear DNA genome. 118 HEV is present naturally in two strains; virulent, which causes an immune-mediated, acute disease characterized by bloody diarrhea, enteric hemorrhage, and death; and avirulent, which is widely used as a vaccine strain. Both strains cause transient period of immunosuppression and splenomegaly and target the IgM + B cells in the spleen and peripheral blood. In addition to turkeys, HEV causes immune-mediated diseases in pheasants and chickens characterized by pulmonary edema and splenomegaly. Lesions in several organs have been also reported in affected birds. 113 Changes in the abundance of microbial populations between HEVinfected and uninfected turkeys have recently been investigated. Analysis of OTUs detected in the gut of healthy turkeys revealed the presence of four abundant phyla: Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria. In healthy turkeys, Firmicutes was the predominant phylum (80-100%) in the three small intestinal sections, while in the cecum a more heterogeneous population containing representatives from Firmicutes, Bacteroidetes, and Proteobacteria was observed. 33 The diversity of cecal microbial population was also observed throughout the developmental stages of turkeys up to 16 weeks of age. The bacterial populations were also dissected at the genus level in the different gastrointestinal tract sections and Lactobacillus, Streptococcus, and Clostridium XI were the prevailing genera forming nearly 80% of the total microbiota. An interesting finding was that Campylobacter spp. was most abundant in the cecum of turkey poults at 10 weeks of age. 96 Interestingly, similar to turkeys, both chickens and ducks have a greater microbial diversity in the cecum as compared to the rest of the small intestine. 96, 119, 120 The abundance of jejunal bacterial populations at the family level was altered by natural infections of HEV in turkeys. Compared to uninfected turkeys, infected birds with clinical signs and HEV detected in the intestines-indicating an early-phase infection-had a lower abundance in Micrococcaceae and a higher abundance of Propionibacteriaceae, while those turkeys with no clinical signs and virus detected in the spleen-indicating a late-phase infection-had a much lower abundance in both families. The abundance of family Bacteroidaceae was extremely increased in HEV-infected turkeys as compared to uninfected controls. For the phyla Firmicutes, a substantial reduction in Lactobacillaceae and an increase in Clostridiaceae was observed in intestinally-positive turkeys with clinical signs. An opposite trend was recorded for turkeys with positive spleens and no clinical signs. However, both groups of infected turkeys showed an increased abundance of three other families belonging to Firmicutes. Family Campylobacteriaceae increased in abundance by 18-to 26-folds in HEV-infected turkeys. 33 HEV infection of turkeys has been associated with secondary E. coli infections during the transient immunosuppression period. 34 HEV is transmitted through fecal-oral route and eventually reaches the blood stream. In the spleen, the virus replicates rapidly in B lymphocytes and to a much lower extent in macrophages. Hyperplasia of the splenic white pulp occurs as a result of infiltration with high numbers of macrophages and lymphocytes, including CD4+ T cells. The interplay among these various types of immune cells within the site of viral replication leads to the production of very high levels of proinflammatory cytokines, e.g., interleukin 6 (IL-6) and tumor necrosis factor (TNF) as well as the anti-inflammatory cytokines of type I and II IFNs. The latter activates macrophages, inducing the production of the antiviral and immunosuppressive nitric oxide (NO). Most likely, IFN-I and NO play a role in clearing the virus later from the spleen. [121] [122] [123] Moreover, it was suggested that the intestinal hemorrhage in the jejunum area occurs through inflammation-induced "diapedesis." 124 This might be associated with a compromised intestinal epithelial cell barrier of the gut as noticed with other enteric viral infections. Viruses which can colonize a healthy poultry gut have been recently identified in chickens. Specific pathogen-free chickens were placed for a few days with age-matched peers in commercial and back yard farms with previous history of enteric and respiratory problems. Intestinal samples were then collected and the enteric virome and microbiome were analyzed. 125 The analysis revealed that the new viruses which colonized the gut of SPF birds belong to the families Picornaviridae, Picobirnaviridae, Reoviridae, and Astroviridae. This implies that members of these families are associated with enteric diseases in chickens. In these birds, alterations of the gut microbiome were also observed, specifically in the Lachnospiracea family and the Clostridium and Lactobacillus genera. Other virus families that were represented in the enteric virome of chickens included Adenoviridae, Birnaviridae, Caliciviridae, Coronaviridae, Leviviridae, Siphoviridae, and Retroviridae. 125 In another study, the fecal virome in healthy commercial chickens was analyzed by viral nucleic acid purification, illumina sequencing, and de novo assembly. 126 Analysis of the assembled viral genome sequences indicated the presence of viruses belonging to the families Adenoviridae, Caliciviridae, Circoviridae, Parvoviridae, Picobirnaviridae, Picornaviridae, and Reoviridae. 126 Additionally, novel, unclassified, viruses with circular single-stranded DNA genomes were identified. The fecal virome of wild waterfowls has been also recently analyzed in various species of migratory wild ducks. 127 The DNA and RNA viral genomes were isolated from purified viral particles, their nucleotide sequences obtained, and analyzed. The most abundant viral familied recognized in the duck's virome were Herpesviridae, Alloherpesviridae, Adenoviridae, Retroviridae, and Myoviridae. 127 In a more recent study on healthy waterfowl and shorebirds, the presence of 27 viral species was reported. These species represent the families Picobirnaviridae, Reoviridae, Astroviridae, Caliciviridae, Picornaviridae, Hepadnaviridae, and Parvoviridae. Variations in virus abundance and diversity were detected among the various species studied. 128 The results signify the potential of wild birds as reservoir of disease-causing poultry pathogens. Over 35% of agriculturally produced protein for consumption is produced in pigs globally. 129 China is the leading producer of pork generating over 54 million metric tons annually. The majority of the pork exported to the rest of the world comes from the European Union. 130 A constantly increasing human population demands for the availability of more animal-based protein for consumption around the world. This demand for more meat has led to the intensification of farming practices often causing overcrowding of animals allowing for rapid pathogen transmission. 129 Pork production and trade are primarily impacted by infectious diseases that affect swine. The domestication of pigs for the purpose of livestock and husbandry has increased their exposure to emerging and re-emerging viral diseases causing a significant impact on the worldwide pig populations. The recent advancement of technology and overcoming the requirement of culture-based species identification has allowed for extensive research initiatives that have investigated the role of microbiome and its influence on host immunity. [131] [132] [133] [134] This section will focus on different microbial (bacterial) communities that constitute the microbiome and their effects on viral infections in pigs that have a significant impact on domestic pig rearing. African swine fever (ASF) is a severe hemorrhagic disease, with a near 100% mortality rate. The disease is characterized by high fever, loss of appetite, ataxia and depression. 135 The causative agent African swine fever virus (ASFV) is a double-stranded DNA virus belonging to the Asfaviridae family. ASF is a reportable disease to the World Health Organization (WHO), and is often accompanied with regional, national and international trade restrictions with the capacity to impact the global pig meat economy. 136 Pigs of all ages are susceptible to virulent strains of ASF with the exception of African wild pigs. No vaccines or therapeutics are available, with the control of disease spread often carried out by culling large numbers of domestic pigs. Transmission between traditional ASFV hosts, warthogs (Phacochoerus aethiopicus) and bushpigs (Potamchoerus porcus), occurs through the bite of soft ticks belonging to the genus Ornithodoros, which are endemic to sub-Saharan Africa. 137 Ingestion of feed contaminated with biological fluid from wild boars and pigs, and fomites have been identified to be efficient modes of virus transmission in the absence of the arthropod vector. 135, 138 The oral route of infection is considered to be a major portal of entry for ASFV among pigs outside the natural territory of its arthropod host. Little work has been carried out in order to characterize the bacterial composition among different pigs, based on their susceptibility status to ASFV. Specific pathogen-free (SPF) pigs have been demonstrated to be highly susceptible to lethal infection by an attenuated strain of ASFV (E75CV1) that was previously tested to be safe for administering to domestic pigs as a vaccine. 139 Diversity analysis of bacterial communities belonging to SPF, and domestic pigs from the same breed, domestic pigs indigenous to Africa, and warthogs from Africa showed that SPF pigs had the lowest diversity among the different species of pigs analyzed. Firmicutes were found to be the most abundant phylum among all the animals. Bacteria belonging to the genera Plaudibacter, Anaeroplasma, Petrimonas, and Moraxella were found to be among the core communities in pig species that were naturally resistant to ASFV. 35 Naturally, pigs reared under strict SPF-conditions were observed to be the least diverse based on microbial composition which has been correlated with an impaired immune system development and the ASFV-resistant warthogs were identified to contain OTUs belonging to previously-uncharacterized bacterial genera. Porcine circovirus type 2 (PCV2) is a single-stranded circular DNA virus and is the primary causative agent of porcine circovirus associated disease. PCV2 belongs to the genus Circovirus within the family Circoviridae. 140 PCV2 targets the lymphoid organs leading to lymphoid depletion and tissue damage. 141 Direct contact with an infected pig has been known to be the most efficient route of virus transmission along with other routes such as contaminated vectors and fomites. 142 Barrows (pigs aged 27-40 days) infected with PCV2 were observed to show an increase in abundance of Prevotella spp. in their microbiota composition. 37 The presence of a more diverse microbiota and the presence of non-pathogenic E. coli in pigs have been associated with a reduced in clinical signs of PCV associated disease as well as improved growth in sub clinically infected pigs. Fecal microbiota transplant in 3-week old barrows reduced the number of pigs affected by PCV infection, including a reduction in viral load and increased viral-antigen specific antibodies. 38 Coinfections with porcine reproductive and respiratory syndrome virus (PRRSV) and PCV2 occur regularly among overcrowded pig farms. 36 Bacterial colonization dynamics at the time of viral infection, that have been associated with a reduced PCV2/PRRSV burden include the presence of low abundance of Methanobacteriaceae spp., and an increased abundance of the species of Ruminococcaceae, and Streptococcaceae. 36 PRRSV is a singlestranded, positive-sense RNA virus belonging to the family Arteriviridae of the order Nidovirales. 143 Characteristics of PRRSV infection in pigs include reproductive failure in sows, and respiratory disease in growing and finishing pigs. 144 Each year, the pig industry in the US loses $664 million due to the respiratory disease and reduced weight gain among growing pigs infected with PRRSV and coinfected with other pathogens, including PCV2. 38 Porcine epidemic diarrhea (PED) is a highly contagious and lethal enteric disease and is caused by PED virus (PEDV) which belongs to the Alphacoronavirus genus in the Coronaviridae family. Transmission occurs through the fecal-oral route with disease characterized by severe vomiting, diarrhea and dehydration. 145 Genomic sequencing of different PEDV isolates have shown the emergence of PEDV strains with insertion/deletions or multiple mutations in the spike (S) protein sequence allowing for the circulation PEDV among the global pig population. 146 The age of infected pigs, immune status and the virulence of the virus strain have all been attributed to the development of clinical signs of disease. 147 Pigs of all ages are susceptible to PEDV, with the highest mortality rate occurring among neonatal and suckling pigs. 148 A vaccine is available in the form of an adjuvanted attenuated virus that is administered to healthy pregnant sow. Antibodies generated by the sow during and after gestation have been successful to an extent but fail to induce sufficient protection in neonatal piglets farrowed from PEDV naïve sows. 149 Bacterial composition of the neonatal gastrointestinal tracts 35 often begins with the colonization of aerobic or facultatively aerobic bacteria belonging to Escherichia coli, Shigella flexneri, and Streptococcus spp. The consumption of oxygen within the GIT of the newborn piglets allows for the subsequent colonization of anaerobes such as Bacteroides, Clostridium, Oscillibacter, Bifidobacterium, Lactobacillus, and Ruminococcaceae spp. 150, 151 Analysis of the microbiota composition in the small and large intestines of PEDV infected piglets showed an increase in abundance of Escherichia-Shigella, Enterococcus, Fusobacterium, and Veillonella. 39 In parallel to this, a decrease in SCFA-producing bacteria such as Rikenellaceae (RC9 gut group), Butyricimonas, and Alistipes were also observed among infected piglets. 40 A reduction in SCFA production within the GIT has been linked with an increased state of dysbiosis in piglets implicating reasons for severity of PEDV infection. 152, 153 Huang et al. showed that bacteria belonging to Ruminococcaceae, Rikenellaceae, and Porphyromonadaceae, were associated with healthy piglets as compared to PEDV infected piglets. 39 Investigative studies on viruses outside their pathogenic interaction with their swine hosts have been limited. With an increased access to advanced sequencing technology, the influences of virome on swine health can be investigated in greater detail. Liver biopsies taken from healthy pigs revealed a diverse range of small ssDNA viral genome sequences that were closely related to viruses belonging to Anelloviridae, Circoviridae and Parvoviridae families. 154 Another study also reported a rich viral abundance and diversity among blood-fed mosquitoes collected from pig farms. Apart from the presence of viruses belonging to families that were known to cause infection among pigs, humans, and mosquitoes, a considerable number of nonclassified virus reads were also detected that were presumed to belong to unexplored novel virus species. 155 Microbiota diversity is an integral part of pig health. As identified by Ober and colleagues, an increased microbial diversity together with shifts in the presence of several bacterial families prior to infection allowed pigs to recover faster after the infection fully resolved. 36 The use of antimicrobials in regular farming practices have been attributed to the reduction in microbiota diversity increasing their susceptibility to infections. Considering alternative therapy such as fecal microbiome transplantation for enhancing swine health would enhance the overall economic benefits associated with pig farming. Rumen microbes play a critical role in the development of digestive system in neonatal, nursing calves. They also play important roles in the nutritional acquisition (feed digestion), and physiological and immunological functions of the host. Generally, they support each other to ferment plant structural and nonstructural carbohydrates and proteins. Along with the large-scale application of high throughout sequences and metagenome techniques, literatures in characterization and variation of microbiota in ruminant animals increased explosively. [156] [157] [158] [159] [160] The rumen microbiota is very complex, and the diversity of ruminal microorganisms can be affected by diet composition, genetics and environmental factors. There are approximately 7000 bacterial species and 1500 archaeal species in the rumen. 161 Rumen protozoa is present when animals are fed high-grain diets, and rumen fungi represents approximately 10% of the total rumen microbiota at any given time. 162 Plenty of studies have found differences of micro-biological development in the digestive system of calves in relation to variations in management practices, such as housing, feeding, and antimicrobial administration during the neonatal period. 163, 164 The following review focuses on the microbiota and how it changes and acts during virus infection in ruminants. Bovine rotavirus is the main pathogen associated with neonatal calf diarrhea. It infects young calves orally, replicates in the cytoplasm of intestinal epithelial cells, and destroys mature intestinal cells. 165 It has been reported that the incidence of rotavirus diarrhea was associated with a decrease in bacterial diversity in the gut, which is a well-known hallmark of dysbiosis in the early stages of life in the calves. 43 Alterations were found in the gut microbiota of neonatal diarrhea samples as a result of rotavirus infection. The relative abundances of Firmicutes and Bacteroidetes increased in healthy calves, while Proteobacteria was abundant in rotavirus diarrhea samples. 43 This was consistent with human rotavirus diarrhea cases that the phylum Proteobacteria is associated with gut dysbiosis and inflammation. 41 At the genus level, in rotavirus-infected calves, there was significant increase of the genera Escherichia, Clostridium, and Streptococcus. However, Subdoligranulum, Blautia, Bacteroides, and Coprococcus were decreased. Notably, Lactobacillus was significantly decreased in the rotavirus diarrhea calves. At the species level, Lactobacillus species such as L. gasseri and L. amylovorus were found to be relatively abundant in healthy calves. L. gasseri is known to prevent the severity of acute self-limiting diarrhea in adults. 42 In rotavirus-infected calves, the relative abundance of G. formicillis, B. glucerasea, B. coccoides, and B. obeum were significantly higher in the healthy group. Two species of C. perfringens and E. coli associated with the diarrheic calves were identified. 43 This study demonstrated that rotavirus infection changed the structure of the gut microbiota, which would affect the follow up diagnosis and administration. Bovine leukemia virus (BLV) is a retrovirus which causes enzootic bovine leukosis in cattle, and it is closely related to the human T-lymphotropic virus type 1. 166 BLV infection of domestic cattle is highly prevalent in several geographic regions. 167 Once BLV infects a cow, it cannot be eliminated. 168 Because the immune system of host cattle can be impaired during infection, BLV infection consistently results in the inability of cattle to maintain normal health. 169 Comparing to BLV infected dairy cows, Lachnospiraceae and Veillonellaceae families associated with ruminal fermentation were more abundant in the fecal microbiota of uninfected cows. 44 Higher titer BLV infection was associated with less diversity of microbiota in infected cows. Meanwhile, the virus propagation ability of BLV strains was negatively correlated with one taxon of Sanguibacteroides. Besides causing lymphoproliferation and leukemia, BLV infection may decrease energy production efficiency in the infected cows through modification of rumen and hindgut microbiota. 44 According to function speculation of the bacteria detected in the differential abundance analysis, energy production loss in the rumen and hindgut was assumed, which may explain the secondary negative effects such as increased susceptibility to other infections and decreased lifetime milk production and reproductive efficiency. [170] [171] [172] [173] Further studies need to systematically investigated rumen microbiota with rumen nutrition status, such as the level of volatile fatty acids together with BLV strain genetics. Escherichia coli strains comprise a group of bacteria with a huge genetic diversity that make them able to colonize different niches and to adapt as a component of intestinal commensal microbiota of animals and human beings. 174 E. coli are classified into eight different phylogenetic groups (A, B1, B2, C, D, E, F and Clado1). 175 Changes in compositions of commensal E. coli were observed during rotavirus and coronavirus intestinal infection in calves with diarrhea. 176 From this study with 30 calves each in the infected and control group, E. coli isolates identified in virus-infected calves were phylogenetically classified as B1 (70%), B2 (3.33%), C (3.33%), D (3.33%), E (13.33%) and unknown (6.7%), whereas E. coli isolates from the control group were classified only as B1 (83.3%), E (10%) and unknown (6.7%). B2, C and D groups were found only in samples from animals with diarrhea due to rotavirus and coronavirus infection. 176 According to Escobar-Paramo 177 and Clermont, 175 B2 and D are E. coli phylogenetic groups that comprise strains with pathogenic potential, being more commonly observed in extra-intestinal infections. In E. coli isolated from diarrheic calves, F5 and F18 Fimbriae were found. 176 Fimbriae are virulence factors that allow E. coli colonization mainly in the small intestine, avoiding bacteria elimination along with the feces. 178 F5 is one of the most frequent factors in enterotoxigenic E. coli pathotype isolated from calves with diarrhea. 179 F18 is more commonly associated with post-weaning diarrhea and edema disease in swine. 178, 180, 181 In summary, rotavirus and coronavirus infection changes the homeostasis of intestinal microbiota which involves the participation of E. coli in the pathological progress. E. coli was the only species that positively correlated with the number of neutrophils, which may explain the inflammation during diarrhea. 43 Lumpy skin disease (LSD) is a viral disease of cattle caused by lumpy skin disease virus (LSDV). LSDV is one of the most important animal poxviruses because of the serious economic consequences in cattle. The World Organization for Animal Health 182 categorizes LSD as a notifiable disease. 182 It is characterized by fever, reduced milk production and skin nodules. Mastitis, swelling of peripheral lymph nodes, loss of appetite, increased nasal discharge and watery eyes are also common. Temporary or permanent infertility occur among infected cows and bulls. The disease can cause high morbidity and low mortality. 183, 184 Secondary bacterial infection in the affected skin lesions can increase the severity and prolong the course of the disease. The analysis of prevalent bacterial communities in affected lesion were carried out using 16s rRNA gene sequencing. Up to 98 species were found, most of them belonging to the phyla of Proteobacteria, followed by Firmicutes, Actinobacteria, and Bacteroidetes. Many common mammalian pathogens are found in the Proteobacteria phylum. For example, the Brucella 185 and Rickettsia 186 genera belong to the Alphaproteobacteria class; Bordetella 187 and Neisseria 188 belong to the Betaproteobacteria class; and Escherichia, 189 Shigella and Salmonella belong to the Gammaproteobacteria class. All bacterial species found are known as opportunistic pathogens, but can withstand the inflammatory reaction. Ruminant virome is important in the nutrient and energy cycling, 190 development of immunity, 191 and a major source of genes through lysogenic conversion. 192 As metagenomic next-generation sequencing (mNGS) has been employed to identify uncommon and novel infectious etiologies, massive virus species have been identified in ruminants. 193 Besides the common diarrhea-causing viral pathogens rotavirus and coronavirus, torovirus, norovirus, nebovirus, astrovirus, kobuvirus and enterovirus have been detected from calves' feces with diarrhea by mNGS. 194 Viral genomes of pestivirus A, Ungulate erythroparvovirus 1, bosavirus, and hypothetical circular Rep-encoding single-stranded DNA viruses were also identified from calf serum with mucosal disease. 195 Most studies have involved epidemiologic investigations seeking to show association with diarrhea for each virus alone or in combination with potential pathogens. In bovine respiratory disease, 16 viruses were identified, of which bovine parvovirus 2 was the most prevalent (11.5%, 15/130) followed by ungulate tetraparvovirus 1 (UTPV1, 8.5%, 11/130) and bovine respiratory syncytial virus (BRSV, 8.5%, 11/130). 7 Unconventional viruses such as influenza D virus, bovine rhinitis A virus, bovine coronavirus and bovine rhinitis B virus had also been detected. 196, 197 Non-suppurative encephalitis is one of the most frequent pathological diagnosis in cattle with neurological disease. Six virus candidates: parainfluenza virus 5, bovine astrovirus CH13/NeuroS1 (BoAstV-CH13/ NeuroS1), bovine polyomavirus 2 (BoPV-2 SF), bovine herpesvirus 2, bovine herpesvirus 6 (BoHV-6) and a novel bovine betaretrovirus termed BoRV-CH15 had been detected in neurologically-diseased cows. BoAstV-CH13, BoPV-2 SF and BoHV-6 were significantly associated with the disease. These data expanded our knowledge on encephalitis-associated pathogens in cattle and point to the value of NGS in resolving complex infection scenarios in a clinical disease setting. 198 Most of the studies have focused on the variety of microbiome in ruminants. Limited work has been done to study the interaction between microbiota and ruminant viruses or the microbiota changes before and after different virus infection. Diarrhea due to rotavirus and coronavirus infection was associated with a decrease in bacterial diversity in the gut. 43, 176 Diarrhea of any etiology may result in a variety of complications as acid-base and electrolyte imbalance and increased lumen-fluid volume with consequent dehydration. 199, 200 These poor health condition encourages mass propagation of conditional pathogenic bacteria, which leads to the secondary bacterial infection. Meanwhile, the immunosuppression caused by BLV infection may decrease energy production efficiency in the cows through modification of rumen and hindgut microbiota. The mechanisms of interactions between microbiota and viruses in ruminant host in regard to pathological progress in viral diseases should be better explored in future research. Like the bacterial microbiome studies, the metagenome sequencing opens a door for us to peek into the virome structure in ruminant diseases. The diarrhea, respiratory diseases, and encephalitis are the main health threats to ruminants. The detection of virome components will fill in the knowledge gap in identifying the disease-associated pathogens. However, determining the contribution of these viruses to health and diseases in ruminants and other animal species would be challenging and much uncertainty still remains concerning their roles as primary pathogens, coinfection agents, or commensals. Microbiota is capable of modulating host immune systems and determining the susceptibility to various viral pathogens. The studies reviewed in this chapter have shown such significant implications of the microbiota on host immunity. The influence of the microbiota on viral infection susceptibility and viral disease outcome is undisputable although varies among viruses. These data are very important in terms of viral infection, as currently there are few effective and reliable antiviral therapies available. Further understanding of the interactions between viral pathogens, host species and their microbiota will eventually allow us to tailor the contents of the microorganisms to the extent that it is able to protect the host from certain life-threatening viral infections. More research is needed to establish concrete cause and effect relationships between the microbiota composition and viral infection, as many of these studies did not address other factors that also are known to influence microbiota composition, such as genetics, diet, and environment. It should also be noted that some of these studies focused on small cohorts of participants, and more work is needed to establish trends among larger populations. Despite the limitations of some of the presented works, the microbiota still prevails as a hopeful means of immunotherapy for prevention of viral infection in the future in both humans and farm animals. The commensal microbiota and viral infection: a comprehensive review The respiratory microbiome and susceptibility to influenza virus infection Chicken gut microbiota: importance and detection technology Cutaneous leishmaniasis induces a transmissible dysbiotic skin microbiota that promotes skin inflammation The landscape ecology and microbiota of the human nose, mouth, and throat Environment dominates over host genetics in shaping human gut microbiota Microbial composition of the human nasopharynx varies according to influenza virus type and vaccination status Corynebacterium accolens releases antipneumococcal free fatty acids from human nostril and skin surface triacylglycerols Dietary fiber confers protection against flu by shaping Ly6c(-) patrolling monocyte hematopoiesis and CD8(+) T cell metabolism Molecular identification of bacteria associated with bacterial vaginosis Cervicovaginal bacteria are a major modulator of host inflammatory responses in the female genital tract Lactobacillus-deficient cervicovaginal bacterial communities are associated with increased HIV acquisition in young South African women Disruption of the human gut microbiota following norovirus infection Relevance of secretor status genotype and microbiota composition in susceptibility to rotavirus and norovirus infections in humans Enhanced GII.4 human norovirus infection in gnotobiotic pigs transplanted with a human gut microbiota Significant correlation between the infant gut microbiome and rotavirus vaccine response in rural ghana Rotavirus vaccine response correlates with the infant gut microbiota composition in Pakistan Modeling human enteric dysbiosis and rotavirus immunity in gnotobiotic pigs Lactobacilli and bifidobacteria promote immune homeostasis by modulating innate immune responses to human rotavirus in neonatal gnotobiotic pigs Abnormal fecal microbiota community and functions in patients with hepatitis B liver cirrhosis as revealed by a metagenomic approach Gut microbiome alterations in patients with stage 4 hepatitis C HCV eradication does not impact gut dysbiosis or systemic inflammation in cirrhotic patients Influenza A virus subtype H9N2 infection disrupts the composition of intestinal microbiota of chickens Avian influenza virus subtype H9N2 affects intestinal microbiota, barrier structure injury, and inflammatory intestinal disease in the chicken ileum Community-level differences in the microbiome of healthy wild mallards and those infected by influenza A viruses Newcastle disease virus infection interferes with the formation of intestinal microflora in newly hatched specific-pathogen-free chicks Studies on immune response to Newcastle disease virus in broiler chickens fed with Lactobacillus reuteri PIA16 isolated from the gut of indigenous chicken of Assam Infectious bursal disease virus infection leads to changes in the gut associated-lymphoid tissue and the microbiota composition Infectious bursal disease virus inoculation infection modifies Campylobacter jejuni-host interaction in broilers Pathogenicity and persistence of Salmonella enteritidis and egg contamination in normal and infectious bursal disease virus-infected leghorn chicks Effect of infectious bursal disease (IBD) vaccine on Salmonella Enteritidis infected chickens Gastrointestinal microbial population of turkey (Meleagris gallopavo) affected by hemorrhagic enteritis virus Adenovirus infections Comparative analysis of the fecal microbiota from different species of domesticated and wild suids Increased microbiome diversity at the time of infection is associated with improved growth rates of pigs after co-infection with porcine reproductive and respiratory syndrome virus (PRRSV) and porcine circovirus type 2 (PCV2) Alteration of the pig intestinal microbiome when vaccinated against or inoculated with porcine circovirus 2 using a multivariate analysis model Fecal microbiota transplantation is associated with reduced morbidity and mortality in Porcine Circovirus associated disease Dynamic change of gut microbiota during porcine epidemic diarrhea virus infection in suckling piglets Altered gut microbiota profiles in sows and neonatal piglets associated with porcine epidemic diarrhea virus infection Intestinal microbial communities associated with acute enteric infections and disease recovery Therapeutic value of a Lactobacillus gasseri and Bifidobacterium longum fixed bacterium combination in acute diarrhea: a randomized, double-blind, controlled clinical trial Rotavirus-mediated alteration of gut microbiota and its correlation with physiological characteristics in neonatal calves Examination of the fecal microbiota in dairy cows infected with bovine leukemia virus Chasing seasonal influenza-the need for a universal influenza vaccine Estimates of global seasonal influenzaassociated respiratory mortality: a modelling study Advances in oral mucosal immunity and the microbiome The immunology of influenza virus-associated bacterial pneumonia Dolosigranulum pigrum causing nosocomial pneumonia and septicemia Staphylococcus aureus shifts toward commensalism in response to corynebacterium species Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response Biomedical prevention of HIV in women: challenges and approaches, with particular reference to the vaginal microbiome The influence of cervicovaginal microbiota on mucosal immunity and prophylaxis in the battle against HIV Genital inflammation, immune activation and risk of sexual HIV acquisition The gut microbiome and HIV-1 pathogenesis: a two-way street The impact of human immunodeficiency virus infection on gut microbiota alpha-diversity: an individual-level meta-analysis Gut microbiome, short-chain fatty acids, and mucosa injury in young adults with human immunodeficiency virus infection The Th17/Treg ratio, IL-1RA and sCD14 levels in primary HIV infection predict the T-cell activation set point in the absence of systemic microbial translocation Regulation of immune cell function by short-chain fatty acids Early impairment of gut function and gut flora supporting a role for alteration of gastrointestinal mucosa in human immunodeficiency virus pathogenesis Gut barrier structure, mucosal immunity and intestinal microbiota in the pathogenesis and treatment of HIV infection Butyrate inhibits NF-kappaB activation in lamina propria macrophages of patients with ulcerative colitis Global prevalence of norovirus in cases of gastroenteritis: a systematic review and meta-analysis Norovirus regulation by host and microbe Norovirus classification and proposed strain nomenclature High protective efficacy of probiotics and rice bran against human norovirus infection and diarrhea in gnotobiotic pigs The interplay between microbiome dynamics and pathogen dynamics in a murine model of Clostridium difficile infection Post-infectious irritable bowel syndrome-a review of the literature Susceptibility to winter vomiting disease: a sweet matter Faecal microbiota composition in adults is associated with the FUT2 gene determining the secretor status Utilizing gnotobiotic models to inform the role of the microbiome in vaccine response heterogeneity Median infectious dose of human norovirus GII.4 in gnotobiotic pigs is decreased by simvastatin treatment and increased by age Enterobacter cloacae inhibits human norovirus infectivity in gnotobiotic pigs IFN-lambda determines the intestinal epithelial antiviral host defense Interferon lambda (IFN-lambda) efficiently blocks norovirus transmission in a mouse model Global, regional, and national estimates of rotavirus mortality in children <5 years of age Effect of antibiotic-mediated microbiome modulation on rotavirus vaccine immunogenicity: a human, randomized-control proof-of-concept trial Rotavirus serum IgA immune response in children receiving rotarix coadministered with bOPV or IPV Causes of impaired oral vaccine efficacy in developing countries Impact of enterovirus and other enteric pathogens on oral polio and rotavirus vaccine performance in Bangladeshi infants The immune system in children with malnutrition-a systematic review Differences of rotavirus vaccine effectiveness by country: likely causes and contributing factors The mammalian intestinal microbiome: composition, interaction with the immune system, significance for vaccine efficacy, and potential for disease therapy. Pathogens Gut microbiota in children vaccinated with rotavirus vaccine Gut-liver axis, gut microbiota, and its modulation in the management of liver diseases: a review of the literature World Health Organization Hepatitis Fact Sheets The virome in mammalian physiology and disease Endogenous retroviruses in the human genome sequence Introduction to retroviridae Exploiting gut bacteriophages for human health Viruses in the faecal microbiota of monozygotic twins and their mothers Nearly constant shedding of diverse enteric viruses by two healthy infants Early life dynamics of the human gut virome and bacterial microbiome in infants Shared and distinct features of human milk and infant stool viromes Review article: the human intestinal virome in health and disease Spatial heterogeneity and stability of bacterial community in the gastrointestinal tracts of broiler chickens Characterization of the microbiome along the gastrointestinal tract of growing turkeys Characterization of gut microbiome dynamics in developing pekin ducks and impact of management system Current perspectives of the chicken gastrointestinal tract and its microbiome Diseases of Poultry Proteobacteria: a common factor in human diseases The cloacal microbiome of five wild duck species varies by species and influenza A virus infection status. mSphere Gut microbiota-mediated protection against influenza virus subtype H9N2 in chickens is associated with modulation of the innate responses Role of mucus layers in gut infection and inflammation Trefoil factor peptides and gastrointestinal function ICTV virus taxonomy profile: paramyxoviridae Experimental pathogenesis for chickens, turkeys, and pigeons of exotic Newcastle disease virus from an outbreak in California during Newcastle disease, other avian paramyxoviruses, and avian metapneumovirus infections Effects of a multi-strain probiotic (PrimaLac) on performance and antibody responses to Newcastle disease virus and infectious bursal disease virus vaccination in broiler chickens Comparative effect of Lactobacillus casei and a commercial mangosteen dietary supplement on body weight gain and antibody response to Newcastle disease virus vaccine in fighting roosters ICTV virus taxonomy profile: Birnaviridae The immunodepressive effect of infectious bursal disease virus in chickens Immunosuppression induced by infectious bursal disease virus Hemorrhagic enteritis and related infections Intestinal microbiome of poultry and its interaction with host and diet The long view: Salmonella-the last forty years Infectious bursal disease virus-induced immunosuppression exacerbates Campylobacter jejuni colonization and shedding in chickens Effect of a variant infectious bursal disease virus (E/Del) on Salmonella typhimurium infection in commercial broiler chickens The Springer Index of Viruses Impacts of novel duck reovirus infection on the composition of intestinal microbiota of Muscovy ducklings 16S rRNA genes Illumina sequencing revealed differential cecal microbiome in specific pathogen free chickens infected with different subgroup of avian leukosis viruses Hemorrhagic enteritis virus induced changes in the lymphocyte subpopulations in turkeys and the effect of experimental immunodeficiency on viral pathogenesis Pathogenesis of type II avian adenovirus infection in turkeys: in vivo immune cell tropism and tissue distribution of the virus Pathogenic avian adenovirus type II induces apoptosis in turkey spleen cells Haemorhagic enteritis virus infection in turkeys: a comparison of virulent and avirulent virus infections, and a proposed pathogenesis Comparative analysis of the intestinal bacterial and RNA viral communities from sentinel birds placed on selected broiler chicken farms Faecal virome of healthy chickens reveals a large diversity of the eukaryote viral community, including novel circular ssDNA viruses Fecal virome composition of migratory wild duck species Virome heterogeneity and connectivity in waterfowl and shorebird communities Global trends in infectious diseases of swine Top 10 Pork-Producing Countries Revised estimates for the number of human and bacteria cells in the body Developmental microbial ecology of the neonatal gastrointestinal tract Bacteria in the intestine, helpful residents or enemies from within? A human gut microbial gene catalog established by metagenomic sequencing Epidemiological considerations on African swine fever in Europe Gaps in African swine fever: analysis and priorities African swine fever virus infection in the soft tick, Ornithodoros (Alectorobius) puertoricensis (Acari: Argasidae) Transmission routes of African swine fever virus to domestic pigs: current knowledge and future research directions Expression library immunization can confer protection against lethal challenge with African swine fever virus Porcine circovirus type 2 (PCV2): pathogenesis and interaction with the immune system Environmental distribution of porcine circovirus type 2 (PCV2) in swine herds with natural infection Porcine reproductive and respiratory syndrome virus (PRRSV): pathogenesis and interaction with the immune system Improved vaccine against PRRSV: current progress and future perspective Porcine epidemic diarrhoea virus: a comprehensive review of molecular epidemiology, diagnosis, and vaccines Novel porcine epidemic diarrhea virus (PEDV) variants with large deletions in the spike (S) gene coexist with PEDV strains possessing an intact S gene in domestic pigs in Japan: a new disease situation Porcine epidemic diarrhea virus: current insights Deadly pig virus slips through US borders Pathogenicity and immunogenicity of attenuated porcine epidemic diarrhea virus PC22A strain in conventional weaned pigs Board invited review: the pig microbiota and the potential for harnessing the power of the microbiome to improve growth and health Early-life establishment of the swine gut microbiome and impact on host phenotypes Porcine epidemic diarrhea altered colonic microbiota communities in suckling piglets Changes in cecal microbiota community of suckling piglets infected with porcine epidemic diarrhea virus Liver virome of healthy pigs reveals diverse small ssDNA viral genomes A viral metagenomic analysis reveals rich viral abundance and diversity in mosquitoes from pig farms Fecal microbial diversity in pre-weaned dairy calves as described by pyrosequencing of metagenomic 16S rDNA. Associations of Faecalibacterium species with health and growth The composition and stability of the faecal microbiota of Merino sheep Development of an UPLC-MS/MS method for the analysis of mycotoxins in rumen fluid with and without maize silage emphasizes the importance of using matrix-matched calibration Revealing the impact of global mass bleaching on coral microbiome through 16S rRNA genebased metagenomic analysis Compendium of 4,941 rumen metagenome-assembled genomes for rumen microbiome biology and enzyme discovery The rumen microbiome: composition, abundance, diversity, and new investigative tools An Introduction to the Rumen Microbiome. Noble News and Views Development of colonic microflora as assessed by pyrosequencing in dairy calves fed waste milk An rRNAbased analysis for evaluating the effect of heat stress on the rumen microbial composition of Holstein heifers An overview of calf diarrhea-infectious etiology, diagnosis, and intervention Virus taxonomy: the database of the International Committee on Taxonomy of Viruses (ICTV) Mechanisms of pathogenesis induced by bovine leukemia virus as a model for human T-cell leukemia virus Bovine leukemia virus integration site selection in cattle that develop leukemia Bovine leukemia virus: a major silent threat to proper immune responses in cattle The implication of BLV infection in the productivity, reproductive capacity and survival rate of a dairy cow Lifetime effects of infection with bovine leukemia virus on longevity and milk production of dairy cows Effect of infection with bovine leukemia virus on milk production in Michigan dairy cows Epidemiology and genetic diversity of bovine leukemia virus The population genetics of commensal Escherichia coli The clermont Escherichia coli phylo-typing method revisited: improvement of specificity and detection of new phylo-groups Characterization of intestinal Escherichia coli isolated from calves with diarrhea due to rotavirus and coronavirus Identification of forces shaping the commensal Escherichia coli genetic structure by comparing animal and human isolates A systematic review and meta-analysis of the epidemiology of pathogenic Escherichia coli of calves and the role of calves as reservoirs for human pathogenic E. coli Enterotoxigenic Escherichia coli in veterinary medicine The F18 fimbrial adhesin FedF is highly conserved among F18 +Escherichia coli isolates Lumpy skin disease Lumpy skin disease of cattle: an emerging problem in the Sultanate of Oman Review: lumpy skin disease: an emerging threat to Europe, the Middle East and Asia Brucella-associated cervical bursitis in cattle Vector-borne bacteria in blood of camels in Iran: new data and literature review Recognition of mycobacterial antigens delivered by genetically detoxified Bordetella pertussis adenylate cyclase by T cells from cattle with bovine tuberculosis A new species of Neisseria from the dental plaque of the domestic cow, Neisseria dentiae sp Diagnostic studies of abortion in Danish cattle 2015-2017 Dietary energy drives the dynamic response of bovine rumen viral communities Gut DNA virome diversity and its association with host bacteria regulate inflammatory phenotype and neuronal immunotoxicity in experimental Gulf War illness Metagenomics of rumen bacteriophage from thirteen lactating dairy cattle Virus metagenomics in farm animals: a systematic review Viral enteritis in calves Evaluation of the serum virome in calves persistently infected with Pestivirus A, presenting or not presenting mucosal disease The pulmonary virome, bacteriological and histopathological findings in bovine respiratory disease from western Canada Respiratory viruses identified in western Canadian beef cattle by metagenomic sequencing and their association with bovine respiratory disease Exploring the virome of cattle with non-suppurative encephalitis of unknown etiology by metagenomics Characterization of the fecal bacterial microbiota of healthy and diarrheic dairy calves Clinical signs, profound acidemia, hypoglycemia, and hypernatremia are predictive of mortality in 1,400 critically ill neonatal calves with diarrhea