The composition of the human microbiome is considered a major source of interindividual variation in immunity and, by extension, susceptibility to diseases. Intestinal bacteria have been the major focus of research. However, diverse communities of viruses that infect microbes and the animal host cohabitate the gastrointestinal tract and collectively constitute the gut virome. Although viruses are typically investigated as pathogens, recent studies highlight a relationship between the host and animal viruses in the gut that is more akin to host–microbiome interactions and includes both beneficial and detrimental outcomes for the host. These viruses are likely sources of immune variation, both locally and extraintestinally. In this review, we describe the components of the gut virome, in particular mammalian viruses, and their ability to modulate host responses during homeostasis and disease.

The human body harbors diverse populations of infectious entities, collectively known as the microbiome, which interact with each other and with the host to influence health and disease. Although the most commonly studied are the bacterial members of the microbiome, there are vast numbers of viruses present in the human body. Together, these viruses form the virome. Comprehensive annotation of the human virome is confounded by the staggering diversity of viruses detected at multiple anatomical sites that can have ssRNA, dsRNA, ssDNA, or dsDNA genomes. Despite this challenge, recent advances in sequencing and analysis of metagenomic data have facilitated the discovery of new viruses and improved our ability to catalog viral communities in an unbiased manner (1, 2). These pioneering efforts reveal substantial intestinal virome diversity between individuals likely due to differences in bacterial composition and diet (3, 4). Studies comparing the virome between individuals have contributed to the growing evidence that differential exposure to viruses influences host physiology, either to the detriment or benefit of the host, much like the bacterial microbiome.

Although the study of virology has generally focused on disease-causing animal viruses, a large fraction of the virome is composed of bacteriophages and endogenous retroviral elements. Approximately 1015 bacteriophages exist in the human intestine (5). A gram of human stool contains 108–109 virus-like particles (6). The majority of these contain a DNA genome. Among the DNA viruses that can be matched to an annotated genome, 99% are bacteriophages and the remaining 1% are animal viruses such as anellovirus, parvovirus, adenovirus, and papillomavirus (6). Intrapersonal bacteriophage abundance is mostly stable over time but does show rapid sequence diversification (7). The predominant classifiable bacteriophage species in the gut are the dsDNA Caudovirales and ssDNA Microviridae (8). However, one uncharacterized dsDNA bacteriophage known as crAssphage is present in 73% of fecal metagenomes and predicted to infect Bacteroides species that are prevalent in the human gut (9, 10). In addition to directly influencing microbiome population dynamics by killing their bacterial hosts during lytic release of viral particles, bacteriophages that integrate into bacterial genomes contribute to the coding potential of the microbiome to indirectly influence the physiology of the animal host (8).

Endogenous retroviruses (ERVs) resemble present-day exogenous retroviruses but are integrated in the host genome and transferred vertically between generations. They are estimated to make up 8% of the human genome (11). The syncytin proteins that mediate placental development are derived from ERV env genes, and ERVs have dispersed IFN-inducible enhancer elements throughout mammalian genomes, suggesting that retroviral integration played a substantial role in mammalian evolution (12). Although most ERVs have accumulated many changes to their sequence over time that have rendered them defective, there are a limited number of ERVs with the potential to produce viral products that activate immune response or promote tumorigenesis (1215). ERVs can also facilitate insertional mutagenesis and chromosomal rearrangements that affect cellular gene expression (16). The virome can also include plant viruses, likely introduced through food, and viruses that infect archaea and eukaryotic members of the microbiome such as fungi (mycobiome). One study showed that 97% of virus-like particles from the healthy human gut that harbor an RNA genome represent pathogenic plant viruses, with the remaining 3% belonging to animal viruses (17). How these viruses affect animal hosts is unknown.

The remainder of the virome consists of RNA and DNA animal viruses that are not integrated into the germ line. At any given time, an individual human harbors multiple animal viruses, many of which establish chronic infections (1820). The prevalence of animal viruses that cause transient infections, also considered part of the virome, can be more difficult to investigate, especially if the infection is asymptomatic. In contrast to serological methods that capture the infectious history of an individual (21), metagenomic studies may miss the contribution of a virus that is no longer present in a patient or diseased tissue. Additionally, chronic infections are often difficult to detect because certain viruses can exist in a quiescent state (latency), and the immune system may restrict replication to levels that are undetectable by conventional methods. In one of the few longitudinal virome studies performed to date, fecal samples from healthy human infants were shown to harbor RNA and DNA animal viruses belonging to 16 distinct families during the first 24 mo of life (22). By adulthood, a typical individual will have been infected by at least 10 different viruses, with some individuals showing evidence of infection by 50–100 viral species (21).

The traditional paradigm of host–pathogen interactions, in which infection by an individual agent directly produces immediate disease, fails to fully capture our relationship with many of these animal viruses. The necessity of the host machinery for their life cycle suggests that these viruses are unlikely to be silent passengers. Recent studies highlight how the gastrointestinal tract is an important site for virus–microbiome and virus–host interactions that likely contribute to interindividual variation in immunity and disease susceptibility (Fig. 1). Therefore, in this article, we will focus on the impact of intestinal animal viruses as modifiers of the immune system. We will first review the pathways involved in recognition and responding to intestinal infection and provide evidence of functional interactions between animal viruses and the bacterial microbiome. We will next discuss the beneficial and detrimental impact of intestinal infections by viruses beyond their role as pathogens. At the end, we use examples of how knowledge gained from the study of viral infections at nonintestinal sites can guide future research into the gut virome.

Unlike bacteria, viruses need to infect host cells within the gastrointestinal tract to support their propagation. Target cells include the one-layer thick epithelial cells that serve the dual function of facilitating nutrient exchange and a physical barrier against invasion (23). Dendritic cells (DCs) and macrophages within the lamina propria (tissue underlying the epithelium) and GALT (such as Peyer patches) also commonly encounter viruses (24). Nucleic acid derived from enteric viruses are sensed by these cells through many of the same pattern recognition receptors (PRRs) that are important at other sites. These include endosomal TLRs that signal through MYD88 and TRIF and the cytosolic sensors retinoic acid inducible gene I (RIG-I) and melanoma differentiation–associated protein 5 (MDA5) that signal through MAVS to stimulate the expression of type I IFN (IFN-I) and type III IFN (IFN-III) (24). Both RIG-I and MDA5 are necessary for optimal antiviral responses to rotavirus, a dsRNA virus that infects the small intestinal epithelium to cause diarrheal disease in children (25). Sensing of ssRNA noroviruses, which also cause gastroenteritis in humans, can occur through MDA5, TLR7, and TLR3 in myeloid cells (26, 27). Although best known for responding to bacteria, recent studies suggest that cytosolic Nod-like receptors (NLRs) have an intestine-specific role in restricting viruses. Mice deficient in NLRP6, which can serve as a cofactor for RNA helicase DHX15 to signal through MAVS, display a blunted IFN-I/III response and are susceptible to oral infection by encephalomyocarditis virus and murine norovirus (MNV) (28). The multiple pathways involved in norovirus recognition may reflect interstrain differences or their ability to infect broad cell types, including myeloid cells, lymphocytes, and the specialized sensory epithelial cells known as tuft cells (2932). Inhibiting the ability of MNV to engage these rare tuft cells prevents infection, highlighting the importance of an exquisitely specific cell tropism and remarkable adaptation of enteric viruses (30). Rotavirus RNA is sensed by NLRP9b and another RNA helicase DHX9 in the intestinal epithelium and is essential for inflammasome-mediated cell death (pyroptosis). Given that rotaviruses antagonize IFN signaling, this pathway may explain why other PRRs are inadequate for controlling this virus (33).

Despite the ability of certain viruses to evade IFN responses, numerous studies highlight the essential role of these cytokines during intestinal infection. IFN-I (IFN-α and IFN-β) binds the IFN-α/β receptor (IFNAR1 and IFNAR2 complex), whereas IFN-III (IFN-λ) binds the IFN-λ receptor (IFNLR1 and IL-10R2 complex) to induce an antiviral gene expression program. Although the exact effector mechanisms in the gut remain obscure, they are presumably similar to other sites of infection and involve IFN-stimulated genes (ISGs) that reduce viral replication (e.g., modification of viral RNA) and induce a refractory state in neighboring uninfected cells. IFNAR1 is broadly expressed on a variety of cell types and is necessary for preventing systemic dissemination of MNV, rotavirus, and reovirus (34). In contrast, the relatively restricted expression of IFNLR1 to epithelial cells suggests it has a more defined role in controlling mucosal viral replication (35). IFN-λ, in combination with IL-22 production by group 3 innate lymphoid cells (ILC3s), effectively controls intestinal rotavirus infection (36). IFN-λ signaling in epithelial cells is also required to regulate fecal shedding and viral replication in mice infected with MNV and reovirus (37). IFNs also promote adaptive immune responses (38), which are critical for control of enteric viruses (3942). Suboptimal CD8+ T cell responses and avoidance of CD8+ T cell detection are associated with MNV persistence (43, 44). Furthermore, successful vaccination against rotavirus directly correlates with IgA production (45). Although we emphasize the role of cytokines downstream of PRRs in subsequent sections as a common means by which viruses affect host physiology, understanding how adaptive immunity functions or fails to control enteric viruses remains an important topic with direct relevance to vaccine efforts.

Boosting innate immunity may be an effective strategy for overcoming insufficient antiviral immunity in the gut. Treatment with bacterially derived flagellin prevents and cures chronic rotavirus infection of mice by triggering IL-22 and IL-18 production through TLR5 and NLRC4, respectively (46). For noroviruses, effective antivirals and vaccines currently do not exist, and there is increased concern that persistent norovirus infection may be contributing to morbidity in immunocompromised individuals or facilitating transmission (47, 48). Remarkably, administration of rIFN-λ is sufficient to clear infection of a persistent strain of MNV independently of the adaptive immune response (29, 49). As we discuss throughout this article, the induction of IFNs is a hallmark of viral infection and likely mediates many of the consequences of enteric viruses on host physiology.

Depletion of intestinal bacteria often reduces the replication of enteric viruses, as the bacterial microbiome is known to facilitate viral infection and modify antiviral immune responses (50, 51). Optimal poliovirus infectivity is dependent on the stabilization of virions upon binding to bacterial surface polysaccharides (52, 53). Binding to bacteria also promotes poliovirus attachment to target cells, which can enhance viral fitness by facilitating coinfection with multiple virions and genetic recombination events between viral strains (54). For similar reasons, intestinal titers and signs of inflammation are reduced following infection of antibiotic-treated Ifnar1−/− mice with reovirus (52). During vertical transmission of mouse mammary tumor virus through maternal milk, LPS bound to virions from the mother stimulates the production of the immunosuppressive cytokine IL-10 in the pups to allow the establishment of infection (55). Treatment of mice with antibiotics also decreases intestinal MNV replication (32, 56, 57). Here, bacteria reduce the efficacy of IFN-λ–mediated viral clearance and enhance infection of B cells (32, 56). During rotavirus infection, bacteria reduce systemic and intestinal anti-rotavirus IgA titers to support increased rotavirus replication (58). These studies indicate that animal viruses that infect the gastrointestinal tract have adapted to the presence of bacteria.

The importance of the microbiome to the intestinal environment is apparent in germ-free mice, which display numerous intestinal and immune abnormalities due to the lack of microbial communities (59). Germ-free mice and mice treated with antibiotics also show increased susceptibility to models of intestinal damage, peanut allergy, allergic asthma, and bacterial infections (6064). Although in some cases intestinal bacteria are sufficient to modulate these responses, in many models a role for the virome cannot be ruled out, especially given that antiviral signaling has a prominent role in diseases involving the gut. Nonhematopoietic expression of MAVS is required to protect mice against colitis following intestinal injury by dextran sodium sulfate (DSS) (65). Similarly, IFN-I signaling following stimulation of the MAVS and RIG-I pathways improves intestinal barrier function and protects mice from graft-versus-host disease (GVHD), a complication that occurs following allogeneic stem cell transplantation (66). In another example, administration of a TLR7 agonist enhances colonization resistance to vancomycin-resistant Enterococcus (VRE), a common hospital-acquired opportunistic pathogen, by stimulating DCs that induce IL-22 production by ILC3s (27). Another example in which intestinal viruses potentially promote colonization resistance was observed during fecal microbiome transplantation (FMT) in patients harboring Clostridium difficile. Surprisingly, filtrated feces (to remove the bacterial component and retain viruses) have the same efficacy as nonfiltrated feces in treating the patients (67). It is possible that the active component of the FMT consists of bacteriophages because patients with C. difficile infection had altered bacteriophage abundance and richness compared with healthy controls, and successful transplantation was associated with transfer of Caudovirales species (68). These and other observations indirectly suggest that enteric viral infections fortify the intestinal barrier in certain situations via triggering beneficial immune responses or influencing the bacterial microbiome.

Mechanistic experiments in mice, especially with MNV infections, provide formal evidence that viruses can function as a subset of the microbiome in a manner analogous to intestinal bacteria (Table I). Many MNV strains establish persistent infection in the intestine, frequently in the absence of obvious symptoms. Infection by a persistent strain of MNV compensates for the absence of bacteria in germ-free mice by restoring intestinal morphology and promoting lymphocyte differentiation (57). In addition, MNV protects antibiotic-treated mice from DSS-induced intestinal injury in a manner dependent on IFNAR1 (57). MNV can also protect antibiotic-treated mice from inflammatory lesions during superinfection with the intestinal bacterial pathogen Citrobacter rodentium and reduces colonization by VRE (27, 57). The recent discovery that MNV infects tuft cells, which coordinate type 2 immune responses and mucus production, suggests that infection by this virus could directly influence the function of the intestinal epithelium and warrants further investigation (30). It is possible that these effects extend beyond the gastrointestinal tract because MNV infection protects mice from lung injury following infection with Pseudomonas aeruginosa (69), and MNV restores serum Ig in germ-free mice to levels observed in conventional mice (57). The observation that norovirus RNA is detected in up to 16% of healthy humans reinforces the need to understand how enteric viral infections impact host biology when they are not causing diarrheal disease (70).

Other animal viruses may also promote intestinal homeostasis. Treatment of mice with a mixture of antivirals increases the severity of DSS-induced colitis, whereas treatment with inactivated rotavirus or TLR3/7 agonists reduces disease (71). In this case, the protective effect of TLR ligation was attributed to IFN-I expression by plasmacytoid DCs. This response to viruses may be conserved in humans because TLR3 and TLR7 gene variants are associated with increased severity of inflammatory bowel disease (IBD) in patients (71). Murine CMV, a herpesvirus that chronically infects a variety of tissues and cell types, promotes turnover of the epithelium in multiple organs including the intestine. This effect was attributed to epithelial proliferation induced by the ISG Apol9a/b expressed by macrophages downstream of elevated IFN-I and was shown to enhance intestinal wound healing (72). When taken together, these studies show that IFN-I and other antiviral responses induce factors that promote intestinal epithelial health in addition to those that inhibit viral replication. Whether IFNs in the gut are beneficial to the host may be context-specific and not without controversy (24). A major future direction is to elucidate the specific mechanisms of action downstream of IFN signaling in the models described above.

Excess IFN-I production and other antiviral responses in the gut can potentiate disease (Table I). Until recently, IFN-I in combination with antiviral drugs was standard treatment for chronic hepatitis C virus infection and was associated with significant toxicity, including gastrointestinal illness. There is also evidence from case studies to suggest that IFN-I therapy may potentiate the development of celiac disease, an autoimmune disorder that mainly occurs in individuals harboring HLA-DQ2 or DQ8 alleles in which inappropriate responses to gluten leads to intestinal damage (73). This side effect of antiviral therapy is consistent with the observation that patients with celiac disease display increased levels of IFN-I production by intestinal DCs that promote Th1 responses in the gut (74). It is therefore unsurprising that virus infections have long been suspected to be involved in the development of celiac disease (75). A compelling recent study demonstrated that celiac disease may be caused by reoviruses, dsRNA viruses that commonly infect humans and are typically associated with mild or undetectable disease (76). In an animal model, reovirus infection blocked the differentiation of peripheral regulatory T cells through IFN-I and enhanced dietary Ag-specific Th1 responses through the transcription factor IFN regulatory factor 1 (IRF1) (76). Patients with celiac disease were also more likely to have higher anti-reovirus Ab titers, which was associated with higher expression of IRF1 in the small intestinal mucosa (76). Therefore, enteric viruses that are otherwise tolerated may induce serious intestinal disease in susceptible individuals.

The paradigm of virus plus susceptibility gene interaction was initially demonstrated in experiments with MNV and reinforces the concept that viruses function as members of the gut microbiome. A common variant of ATG16L1, a gene that mediates the cellular degradative pathway of autophagy, is associated with increased susceptibility to a form of IBD known as Crohn disease. IBD is widely considered a disorder originating from a perturbed microbiome (77). Atg16L1 mutant mice and Crohn disease patients harboring the ATG16L1 risk allele display morphological defects in Paneth cells (78), antimicrobial epithelial cells in the small intestine that are essential for preventing inflammation (79). In the Atg16L1 mutant mice, the Paneth cell defects and other inflammatory pathologies were dependent on infection by MNV (78, 80). In this model, loss of Atg16L1 in the intestinal epithelium sensitizes Paneth cells to necroptosis mediated by TNF-α produced in response to viral infection (81). MNV also accelerates the onset of intestinal inflammation in mice deficient in the toxin transporter MDR1a that are colonized by Helicobacter bilis (82) and IL-10–deficient mice (83). Thus, immune responses to an otherwise beneficial or innocuous virus can contribute to intestinal disease when combined with genetic susceptibility.

Although host responses to MNV and Paneth cell properties are likely conserved between mice and humans, further evidence is required to support the role of IFN-I or the virome in Crohn disease. A number of other viruses, including enterovirus, have been linked to Crohn disease (84, 85). In a virome study, expansion of the Caudovirales bacteriophages in the gut was observed in Crohn disease patients (86). In this article, bacteriophage expansion was associated with decreased bacterial diversity suggesting that virus–microbiome interactions contribute to disease pathogenesis (86). A similar expansion of bacteriophage diversity is observed in patients with colorectal cancer, and specific bacteriophage signatures can delineate patients in early or late stage and those with reduced survival (87). Also, a gut virome analysis of patients displaying GVHD with intestinal involvement revealed a marked increase in animal viruses with a DNA genome and bacteriophage richness (88). In particular, the presence of a dsRNA Picobirnaviridae species is predictive of a severe enteric disease (88). The combined approach of metagenomics through deep sequencing and targeted investigation of specific viral agents (like reovirus and celiac disease) may be necessary to explore the contribution of viruses to Crohn disease, GVHD, and other complex inflammatory disorders that are likely influenced by multiple infectious and genetic factors.

The impact of the intestinal virome may extend beyond the gastrointestinal tract to influence autoimmune diseases (Table I). One explanation for such extraintestinal effects of viruses is the spread of infectious particles or viral RNA/DNA from the intestine to other body sites. Indeed, the transit of Ags between the gastrointestinal tract and pancreatic lymph nodes has been suggested as a mechanism for the effects of environmental agents on type 1 diabetes (T1D), an autoimmune disease where insulin-producing β cells are destroyed by pancreas-infiltrating autoreactive lymphocytes (89). Polymorphisms in MDA5 (IFIH1) and an IFN-I gene expression signature are associated with disease onset, supporting a role for viruses in disease progression (90, 91). Also, the appearance of autoantibodies in patients correlates with infection by coxsackievirus B (CVB) 1, a positive-sense ssRNA virus that belongs to a diverse and prevalent group of picornaviruses that transmit fecal–orally (92). Infection with CVB3 and CVB6, which may provide cross-protection against CVB1, reduces the risk of T1D development in prediabetic children (92). Similar protection against virus-induced T1D is induced by CVB vaccination of mice (93). Although direct causation has not been established, CVB is believed to induce T1D by chronically infecting the pancreas and altering the local immune response (91). Rotavirus infection is also associated with progression to T1D (94). In the NOD mouse model of T1D, oral infection of adult mice with rotavirus leads to IFN-I–dependent bystander activation of lymphocytes in the pancreatic lymph nodes and acceleration of T1D onset, likely because of the spread of infectious virus to the mesenteric and pancreatic lymph nodes (9598). In contrast, neonatal infection of NOD mice with rotavirus or reovirus delays the onset of disease, suggesting that timing of virus infections impacts the course of autoimmunity (99, 100).

A recent prospective study of infants at risk for T1D performed a longitudinal virome analysis and showed that increased bacteriophage diversity predicts a lack of progression to disease (101). Increased bacteriophage diversity correlated with changes in abundance of specific bacterial taxa, which may be related to the extensive literature using NOD mice demonstrating that the composition of the bacterial microbiome is an important factor in disease development (102). The same study also found an enrichment of sequences belonging to Circoviridae in the controls compared with individuals who develop T1D, raising the possibility that these group of poorly characterized small ssDNA animal viruses are protective (101). Although these studies implicate multiple viruses in disease pathogenesis, an exact mechanistic role for viruses in humans has yet to be established and requires additional research.

Metagenomic studies of HIV-positive individuals have been particularly informative in that they reveal the presence of a dynamic gut virome in a disease state. Low peripheral CD4 cell counts lead to the development of AIDS, which is marked by increased susceptibility to secondary infection and other immunopathologies. The gut is a major site of HIV replication, HIV-specific immune responses, and damage (103). Enteric adenoviruses and anelloviruses are increased in HIV-positive patients with low peripheral CD4+ T cell counts (104). Similar expansion of the virome is observed in primates infected with SIV, with intestinal adenovirus associated with increased enteritis and parvovirus viremia associated with increased progression to AIDS (105). It is possible that this increased presence of viruses contributes to AIDS in a manner similar to the proposed role of the bacterial microbiome, where depletion of T cells in the gut disrupt the barrier, leading to the systemic dissemination of bacterial products that fuel chronic and pathological immune activation (106). Given that the majority of humans by the time they reach adulthood become transiently or chronically infected by the animal viruses discussed in this section (Anelloviridae, Adenoviridae, and Picornaviridae), careful analysis of the gut virome in other immune-related disorders is warranted.

When considering how the enteric virome might influence interindividual variation, it is worth examining the known mechanisms by which viruses at other sites of the body alter the immune system. Detailed immunological studies in mice have supported a role for the IFN response but also highlight other pathways (Table II). Viruses that are traditionally considered to display localized infection and disease can provoke systemic responses, such as influenza A virus inducing hepatitis and intestinal damage without local infection (107, 108). Liver damage induced by influenza A virus is caused by the accumulation of pathogen-specific CD8+ T cells, and intestinal damage is the result of an altered bacterial microbiome and increased IL-17a (107, 108). Therefore, an important possibility to examine is whether enteric viruses select for T cells that exert pathological outcomes once they migrate to other sites.

Latent viral infections may be a particularly potent modulator of host responses. Gammaherpesvirus 68 (γHV-68) or CMV infection in mice enhances macrophage and NK cell activation and improves the outcome of secondary infection with Listeria monocytogenes, Yersinia pestis, and influenza A (109111). In an animal model of primary immune deficiency, latent γHV-68 infection rescues survival following L. monocytogenes infection by inducing an inflammatory reaction that compensates for inadequate cytokine levels, suggesting that deleterious mutations can be masked by an individual’s virome (112). γHV-68 also protects against allergic asthma by altering the composition of macrophage subsets in the lung (113). In this case, the effect of viral infection was not dependent on latent infection and occurred during a developmental window. These findings in animal models are supported by elegant human cohort studies taking advantage of monozygotic twins with discrepancies in their history of exposure to infectious agents. CMV infection was identified as a particularly significant environmental variable that influences a broad range of immune parameters (114). Further, young adults previously exposed to CMV show a superior Ab and CD8+ T cell response to influenza A vaccination (110). Thus, exposure to viruses can explain interindividual heterogeneity when other factors fail to provide an adequate explanation.

The blood virome of healthy individuals include herpesviruses, anelloviruses, papillomaviruses, polyomaviruses, adenoviruses, parvoviruses, and pegivirus (115, 116). Although it is unclear whether the presence of these viruses in the blood is consequential, the inverse relationship between pegivirus and HIV disease progression suggests that deeper investigation of the blood virome will be fruitful (117). In contrast, there are a wealth of examples demonstrating that respiratory viruses cause or exacerbate chronic lung diseases. In cystic fibrosis patients who display altered mucus production because of mutations in a chloride channel, disease is associated with the presence of a core group of bacteriophages that infect bacterial species persistent in lungs (118). Cystic fibrosis patients also show increased susceptibility to infection with rhinoviruses, which is linked to poor recovery of lung function following flares (119121). Respiratory viruses are also linked to asthma (122). Patients have impaired IFN-I responses following rhinovirus infection, and rhinovirus C in particular is detectable in a significant proportion of children with moderate to severe asthma (122, 123). Consistent with this observation, the Y529 variant of cadherin-related family member 3 (CDHR3) that leads to increased binding of the virus to the lung epithelium confers susceptibility to rhinovirus C–associated asthma (124, 125). As gene variants such as the loss of function allele of fucosyltransferase 2 (FUT2) can determine whether intestinal viruses bind cells (126, 127), an important area of research will be to examine how heritable factors affect the gut virome.

To summarize, virus–host interactions at extraintestinal sites inform areas of future enteric virome research in the following ways. First, a number of these studies suggest that viruses that cause local infections such as in the lung have long range immunomodulatory effects. Therefore, examining the presence of viruses in affected tissues may not be sufficient, and a role for the gut virome should be considered. Second, the effect of a virus is not always apparent directly subsequent to virus exposure. This is clearly an important consideration for blood transfusions as viruses not routinely screened prior to blood donation could be transmitted to patients and have consequences for future disease pathogenesis. Perhaps similar concerns apply to FMTs that are routinely performed for C. difficile treatment and are being considered for many other conditions. Finally, studies with CMV highlight the effect of viruses on the lymphocyte compartment and how adaptive immunity to subsequent Ags (i.e., not the original virus) may be altered. Molecular mimicry and bystander effects have been discussed extensively in other contexts (18) but are rarely considered downstream possibilities of intestinal virus infections in healthy individuals.

Significant progress has been made in the last decade toward understanding enteric viruses beyond their role as pathogens. Although we continue to perform essential research into the pathogenic role of viruses and develop antivirals and vaccines, we can no longer ignore the possibility that they function as components of the microbiome. Like bacteria, the effects that viruses have are critically dependent on their tissue location, microenvironment, and host. These factors will directly influence whether the virus acts beneficially, acts detrimentally, or remains neutral for the host. With recent advances in metagenomics coupled with techniques that enrich in sensitivity (1, 2, 128), we can now perform large human studies with the aim of linking changes in specific viral populations with disease pathogenesis. These studies can then support the development of more defined animal and cell culture studies that address the mechanisms that individual viruses use to contribute to these phenotypes. This research will certainly lead to the discovery of novel ways in which viruses interact with the host that we can potentially harness for disease prevention and therapies. It may even be possible to engineer enteric viruses with desirable traits, much like current attempts at administering oncolytic viruses as adjuvants for cancer therapy (129131).

This work was supported by National Institutes of Health Grants HL123340, DK093668, DK103788, and AI121244, a Faculty Scholar grant from the Howard Hughes Medical Institute, the Stony Wold–Herbert Fund, the Merieux Institute, the Rainin Foundation, a Burroughs Wellcome Fund Investigator in the Pathogenesis of Infectious Diseases award (to K.C.), a Sir Keith Murdoch Fellowship, and a Vilcek Fellowship (to J.A.N.).

Abbreviations used in this article:

CVB

coxsackievirus B

DC

dendritic cell

DSS

dextran sodium sulfate

ERV

endogenous retrovirus

FMT

fecal microbiome transplantation

GVHD

graft-versus-host disease

γHV-68

gammaherpesvirus 68

IBD

inflammatory bowel disease

IFN-I

type I IFN

ILC3

group 3 innate lymphoid cell

IRF1

IFN regulatory factor 1

MDA5

melanoma differentiation–associated protein 5

MNV

murine norovirus

NLR

Nod-like receptor

PRR

pattern recognition receptor

RIG-I

retinoic acid inducible gene I

T1D

type 1 diabetes

VRE

vancomycin-resistant Enterococcus.

1
Krishnamurthy
,
S. R.
,
D.
Wang
.
2017
.
Origins and challenges of viral dark matter.
Virus Res.
239
:
136
142
.
2
Zhang
,
Y. Z.
,
M.
Shi
,
E. C.
Holmes
.
2018
.
Using metagenomics to characterize an expanding virosphere.
Cell
172
:
1168
1172
.
3
Minot
,
S.
,
R.
Sinha
,
J.
Chen
,
H.
Li
,
S. A.
Keilbaugh
,
G. D.
Wu
,
J. D.
Lewis
,
F. D.
Bushman
.
2011
.
The human gut virome: inter-individual variation and dynamic response to diet.
Genome Res.
21
:
1616
1625
.
4
Reyes
,
A.
,
M.
Haynes
,
N.
Hanson
,
F. E.
Angly
,
A. C.
Heath
,
F.
Rohwer
,
J. I.
Gordon
.
2010
.
Viruses in the faecal microbiota of monozygotic twins and their mothers.
Nature
466
:
334
338
.
5
Carding
,
S. R.
,
N.
Davis
,
L.
Hoyles
.
2017
.
Review article: the human intestinal virome in health and disease.
Aliment. Pharmacol. Ther.
46
:
800
815
.
6
Kim
,
M. S.
,
E. J.
Park
,
S. W.
Roh
,
J. W.
Bae
.
2011
.
Diversity and abundance of single-stranded DNA viruses in human feces.
Appl. Environ. Microbiol.
77
:
8062
8070
.
7
Minot
,
S.
,
A.
Bryson
,
C.
Chehoud
,
G. D.
Wu
,
J. D.
Lewis
,
F. D.
Bushman
.
2013
.
Rapid evolution of the human gut virome.
Proc. Natl. Acad. Sci. USA
110
:
12450
12455
.
8
Manrique
,
P.
,
M.
Dills
,
M. J.
Young
.
2017
.
The human gut phage community and its implications for health and disease.
Viruses
9
:
E141
.
9
Dutilh
,
B. E.
,
N.
Cassman
,
K.
McNair
,
S. E.
Sanchez
,
G. G.
Silva
,
L.
Boling
,
J. J.
Barr
,
D. R.
Speth
,
V.
Seguritan
,
R. K.
Aziz
, et al
.
2014
.
A highly abundant bacteriophage discovered in the unknown sequences of human faecal metagenomes.
Nat. Commun.
5
:
4498
.
10
Yutin
,
N.
,
K. S.
Makarova
,
A. B.
Gussow
,
M.
Krupovic
,
A.
Segall
,
R. A.
Edwards
,
E. V.
Koonin
.
2018
.
Discovery of an expansive bacteriophage family that includes the most abundant viruses from the human gut.
Nat. Microbiol.
3
:
38
46
.
11
Ryan
,
F. P.
2004
.
Human endogenous retroviruses in health and disease: a symbiotic perspective.
J. R. Soc. Med.
97
:
560
565
.
12
Chuong
,
E. B.
,
N. C.
Elde
,
C.
Feschotte
.
2016
.
Regulatory evolution of innate immunity through co-option of endogenous retroviruses.
Science
351
:
1083
1087
.
13
Hurst
,
T. P.
,
G.
Magiorkinis
.
2015
.
Activation of the innate immune response by endogenous retroviruses.
J. Gen. Virol.
96
:
1207
1218
.
14
Stauffer
,
Y.
,
S.
Marguerat
,
F.
Meylan
,
C.
Ucla
,
N.
Sutkowski
,
B.
Huber
,
T.
Pelet
,
B.
Conrad
.
2001
.
Interferon-alpha-induced endogenous superantigen. a model linking environment and autoimmunity.
Immunity
15
:
591
601
.
15
Goff
,
S. P.
1996
.
Operating under a Gag order: a block against incoming virus by the Fv1 gene.
Cell
86
:
691
693
.
16
Kassiotis
,
G.
,
J. P.
Stoye
.
2017
.
Making a virtue of necessity: the pleiotropic role of human endogenous retroviruses in cancer.
Philos. Trans. R. Soc. Lond. B Biol. Sci.
372
:
20160277
.
17
Zhang
,
T.
,
M.
Breitbart
,
W. H.
Lee
,
J. Q.
Run
,
C. L.
Wei
,
S. W.
Soh
,
M. L.
Hibberd
,
E. T.
Liu
,
F.
Rohwer
,
Y.
Ruan
.
2006
.
RNA viral community in human feces: prevalence of plant pathogenic viruses.
PLoS Biol.
4
:
e3
.
18
Cadwell
,
K.
2015
.
The virome in host health and disease.
Immunity
42
:
805
813
.
19
Virgin
,
H. W.
,
E. J.
Wherry
,
R.
Ahmed
.
2009
.
Redefining chronic viral infection.
Cell
138
:
30
50
.
20
Popgeorgiev
,
N.
,
S.
Temmam
,
D.
Raoult
,
C.
Desnues
.
2013
.
Describing the silent human virome with an emphasis on giant viruses.
Intervirology
56
:
395
412
.
21
Xu
,
G. J.
,
T.
Kula
,
Q.
Xu
,
M. Z.
Li
,
S. D.
Vernon
,
T.
Ndung’u
,
K.
Ruxrungtham
,
J.
Sanchez
,
C.
Brander
,
R. T.
Chung
, et al
.
2015
.
Viral immunology. Comprehensive serological profiling of human populations using a synthetic human virome.
Science
348
:
aaa0698
.
22
Lim
,
E. S.
,
Y.
Zhou
,
G.
Zhao
,
I. K.
Bauer
,
L.
Droit
,
I. M.
Ndao
,
B. B.
Warner
,
P. I.
Tarr
,
D.
Wang
,
L. R.
Holtz
.
2015
.
Early life dynamics of the human gut virome and bacterial microbiome in infants.
Nat. Med.
21
:
1228
1234
.
23
Ramanan
,
D.
,
K.
Cadwell
.
2016
.
Intrinsic defense mechanisms of the intestinal epithelium.
Cell Host Microbe
19
:
434
441
.
24
Metzger
,
R. N.
,
A. B.
Krug
,
K.
Eisenächer
.
2018
.
Enteric virome sensing-its role in intestinal homeostasis and immunity.
Viruses
10
:
146
.
25
Broquet
,
A. H.
,
Y.
Hirata
,
C. S.
McAllister
,
M. F.
Kagnoff
.
2011
.
RIG-I/MDA5/MAVS are required to signal a protective IFN response in rotavirus-infected intestinal epithelium.
J. Immunol.
186
:
1618
1626
.
26
McCartney
,
S. A.
,
L. B.
Thackray
,
L.
Gitlin
,
S.
Gilfillan
,
H. W.
Virgin
,
M.
Colonna
.
2008
.
MDA-5 recognition of a murine norovirus. [Published erratum appears in 2008 PLoS Pathog. 4. DOI: 10.1371/annotation/3ce83911-9ccf-4452-a690-2816d0e94c10]
PLoS Pathog.
4
:
e1000108
.
27
Abt
,
M. C.
,
C. G.
Buffie
,
B.
Sušac
,
S.
Becattini
,
R. A.
Carter
,
I.
Leiner
,
J. W.
Keith
,
D.
Artis
,
L. C.
Osborne
,
E. G.
Pamer
.
2016
.
TLR-7 activation enhances IL-22-mediated colonization resistance against vancomycin-resistant enterococcus.
Sci. Transl. Med.
8
:
327ra25
.
28
Wang
,
P.
,
S.
Zhu
,
L.
Yang
,
S.
Cui
,
W.
Pan
,
R.
Jackson
,
Y.
Zheng
,
A.
Rongvaux
,
Q.
Sun
,
G.
Yang
, et al
.
2015
.
Nlrp6 regulates intestinal antiviral innate immunity.
Science
350
:
826
830
.
29
Lee
,
S.
,
C. B.
Wilen
,
A.
Orvedahl
,
B. T.
McCune
,
K. W.
Kim
,
R. C.
Orchard
,
S. T.
Peterson
,
T. J.
Nice
,
M. T.
Baldridge
,
H. W.
Virgin
.
2017
.
Norovirus cell tropism is determined by combinatorial action of a viral non-structural protein and host cytokine.
Cell Host Microbe
22
:
449
459.e4
.
30
Wilen
,
C. B.
,
S.
Lee
,
L. L.
Hsieh
,
R. C.
Orchard
,
C.
Desai
,
B. L.
Hykes
Jr.
,
M. R.
McAllaster
,
D. R.
Balce
,
T.
Feehley
,
J. R.
Brestoff
, et al
.
2018
.
Tropism for tuft cells determines immune promotion of norovirus pathogenesis.
Science
360
:
204
208
.
31
Grau
,
K. R.
,
A. N.
Roth
,
S.
Zhu
,
A.
Hernandez
,
N.
Colliou
,
B. B.
DiVita
,
D. T.
Philip
,
C.
Riffe
,
B.
Giasson
,
S. M.
Wallet
, et al
.
2017
.
The major targets of acute norovirus infection are immune cells in the gut-associated lymphoid tissue.
Nat. Microbiol.
2
:
1586
1591
.
32
Jones
,
M. K.
,
M.
Watanabe
,
S.
Zhu
,
C. L.
Graves
,
L. R.
Keyes
,
K. R.
Grau
,
M. B.
Gonzalez-Hernandez
,
N. M.
Iovine
,
C. E.
Wobus
,
J.
Vinjé
, et al
.
2014
.
Enteric bacteria promote human and mouse norovirus infection of B cells.
Science
346
:
755
759
.
33
Zhu
,
S.
,
S.
Ding
,
P.
Wang
,
Z.
Wei
,
W.
Pan
,
N. W.
Palm
,
Y.
Yang
,
H.
Yu
,
H. B.
Li
,
G.
Wang
, et al
.
2017
.
Nlrp9b inflammasome restricts rotavirus infection in intestinal epithelial cells.
Nature
546
:
667
670
.
34
Ingle
,
H.
,
S. T.
Peterson
,
M. T.
Baldridge
.
2018
.
Distinct effects of type I and III interferons on enteric viruses.
Viruses
10
:
46
.
35
Pott
,
J.
,
S.
Stockinger
.
2017
.
Type I and III interferon in the gut: tight balance between host protection and immunopathology.
Front. Immunol.
8
:
258
.
36
Hernández
,
P. P.
,
T.
Mahlakoiv
,
I.
Yang
,
V.
Schwierzeck
,
N.
Nguyen
,
F.
Guendel
,
K.
Gronke
,
B.
Ryffel
,
C.
Hoelscher
,
L.
Dumoutier
, et al
.
2015
.
Interferon-λ and interleukin 22 act synergistically for the induction of interferon-stimulated genes and control of rotavirus infection.
Nat. Immunol.
16
:
698
707
.
37
Baldridge
,
M. T.
,
S.
Lee
,
J. J.
Brown
,
N.
McAllister
,
K.
Urbanek
,
T. S.
Dermody
,
T. J.
Nice
,
H. W.
Virgin
.
2017
.
Expression of Ifnlr1 on intestinal epithelial cells is critical to the antiviral effects of interferon lambda against norovirus and reovirus.
J. Virol.
91
:
e02079-16
.
38
Iwasaki
,
A.
,
R.
Medzhitov
.
2015
.
Control of adaptive immunity by the innate immune system.
Nat. Immunol.
16
:
343
353
.
39
Chachu
,
K. A.
,
A. D.
LoBue
,
D. W.
Strong
,
R. S.
Baric
,
H. W.
Virgin
.
2008
.
Immune mechanisms responsible for vaccination against and clearance of mucosal and lymphatic norovirus infection.
PLoS Pathog.
4
:
e1000236
.
40
Chachu
,
K. A.
,
D. W.
Strong
,
A. D.
LoBue
,
C. E.
Wobus
,
R. S.
Baric
,
H. W.
Virgin
IV
.
2008
.
Antibody is critical for the clearance of murine norovirus infection.
J. Virol.
82
:
6610
6617
.
41
Yokoyama
,
C. C.
,
J.
Loh
,
G.
Zhao
,
T. S.
Stappenbeck
,
D.
Wang
,
H. V.
Huang
,
H. W.
Virgin
,
L. B.
Thackray
.
2012
.
Adaptive immunity restricts replication of novel murine astroviruses.
J. Virol.
86
:
12262
12270
.
42
Franco
,
M. A.
,
H. B.
Greenberg
.
1999
.
Immunity to rotavirus infection in mice.
J. Infect. Dis.
179
(
Suppl. 3
):
S466
S469
.
43
Tomov
,
V. T.
,
L. C.
Osborne
,
D. V.
Dolfi
,
G. F.
Sonnenberg
,
L. A.
Monticelli
,
K.
Mansfield
,
H. W.
Virgin
,
D.
Artis
,
E. J.
Wherry
.
2013
.
Persistent enteric murine norovirus infection is associated with functionally suboptimal virus-specific CD8 T cell responses.
J. Virol.
87
:
7015
7031
.
44
Tomov
,
V. T.
,
O.
Palko
,
C. W.
Lau
,
A.
Pattekar
,
Y.
Sun
,
R.
Tacheva
,
B.
Bengsch
,
S.
Manne
,
G. L.
Cosma
,
L. C.
Eisenlohr
, et al
.
2017
.
Differentiation and protective capacity of virus-specific CD8(+) T cells suggest murine norovirus persistence in an immune-privileged enteric niche.
Immunity
47
:
723
738.e5
.
45
Westerman
,
L. E.
,
H. M.
McClure
,
B.
Jiang
,
J. W.
Almond
,
R. I.
Glass
.
2005
.
Serum IgG mediates mucosal immunity against rotavirus infection.
Proc. Natl. Acad. Sci. USA
102
:
7268
7273
.
46
Zhang
,
B.
,
B.
Chassaing
,
Z.
Shi
,
R.
Uchiyama
,
Z.
Zhang
,
T. L.
Denning
,
S. E.
Crawford
,
A. J.
Pruijssers
,
J. A.
Iskarpatyoti
,
M. K.
Estes
, et al
.
2014
.
Viral infection. Prevention and cure of rotavirus infection via TLR5/NLRC4-mediated production of IL-22 and IL-18.
Science
346
:
861
865
.
47
Bok
,
K.
,
K. Y.
Green
.
2012
.
Norovirus gastroenteritis in immunocompromised patients.
N. Engl. J. Med.
367
:
2126
2132
.
48
Schwartz
,
S.
,
M.
Vergoulidou
,
E.
Schreier
,
C.
Loddenkemper
,
M.
Reinwald
,
M.
Schmidt-Hieber
,
W. A.
Flegel
,
E.
Thiel
,
T.
Schneider
.
2011
.
Norovirus gastroenteritis causes severe and lethal complications after chemotherapy and hematopoietic stem cell transplantation.
Blood
117
:
5850
5856
.
49
Nice
,
T. J.
,
M. T.
Baldridge
,
B. T.
McCune
,
J. M.
Norman
,
H. M.
Lazear
,
M.
Artyomov
,
M. S.
Diamond
,
H. W.
Virgin
.
2015
.
Interferon-λ cures persistent murine norovirus infection in the absence of adaptive immunity.
Science
347
:
269
273
.
50
Shi
,
Z.
,
A. T.
Gewirtz
.
2018
.
Together forever: bacterial-viral interactions in infection and immunity.
Viruses
10
:
E122
.
51
Karst
,
S. M.
2016
.
The influence of commensal bacteria on infection with enteric viruses.
Nat. Rev. Microbiol.
14
:
197
204
.
52
Kuss
,
S. K.
,
G. T.
Best
,
C. A.
Etheredge
,
A. J.
Pruijssers
,
J. M.
Frierson
,
L. V.
Hooper
,
T. S.
Dermody
,
J. K.
Pfeiffer
.
2011
.
Intestinal microbiota promote enteric virus replication and systemic pathogenesis.
Science
334
:
249
252
.
53
Robinson
,
C. M.
,
P. R.
Jesudhasan
,
J. K.
Pfeiffer
.
2014
.
Bacterial lipopolysaccharide binding enhances virion stability and promotes environmental fitness of an enteric virus.
Cell Host Microbe
15
:
36
46
.
54
Erickson
,
A. K.
,
P. R.
Jesudhasan
,
M. J.
Mayer
,
A.
Narbad
,
S. E.
Winter
,
J. K.
Pfeiffer
.
2018
.
Bacteria facilitate enteric virus co-infection of mammalian cells and promote genetic recombination.
Cell Host Microbe
23
:
77
88.e5
.
55
Kane
,
M.
,
L. K.
Case
,
K.
Kopaskie
,
A.
Kozlova
,
C.
MacDearmid
,
A. V.
Chervonsky
,
T. V.
Golovkina
.
2011
.
Successful transmission of a retrovirus depends on the commensal microbiota.
Science
334
:
245
249
.
56
Baldridge
,
M. T.
,
T. J.
Nice
,
B. T.
McCune
,
C. C.
Yokoyama
,
A.
Kambal
,
M.
Wheadon
,
M. S.
Diamond
,
Y.
Ivanova
,
M.
Artyomov
,
H. W.
Virgin
.
2015
.
Commensal microbes and interferon-λ determine persistence of enteric murine norovirus infection.
Science
347
:
266
269
.
57
Kernbauer
,
E.
,
Y.
Ding
,
K.
Cadwell
.
2014
.
An enteric virus can replace the beneficial function of commensal bacteria.
Nature
516
:
94
98
.
58
Uchiyama
,
R.
,
B.
Chassaing
,
B.
Zhang
,
A. T.
Gewirtz
.
2014
.
Antibiotic treatment suppresses rotavirus infection and enhances specific humoral immunity.
J. Infect. Dis.
210
:
171
182
.
59
Round
,
J. L.
,
S. K.
Mazmanian
.
2009
.
The gut microbiota shapes intestinal immune responses during health and disease. [Published erratum appears in 2009 Nat. Rev. Immunol. 9: 600.]
Nat. Rev. Immunol.
9
:
313
323
.
60
Hernández-Chirlaque
,
C.
,
C. J.
Aranda
,
B.
Ocón
,
F.
Capitán-Cañadas
,
M.
Ortega-González
,
J. J.
Carrero
,
M. D.
Suárez
,
A.
Zarzuelo
,
F.
Sánchez de Medina
,
O.
Martínez-Augustin
.
2016
.
Germ-free and antibiotic-treated mice are highly susceptible to epithelial injury in DSS colitis.
J. Crohn’s Colitis
10
:
1324
1335
.
61
Stefka
,
A. T.
,
T.
Feehley
,
P.
Tripathi
,
J.
Qiu
,
K.
McCoy
,
S. K.
Mazmanian
,
M. Y.
Tjota
,
G. Y.
Seo
,
S.
Cao
,
B. R.
Theriault
, et al
.
2014
.
Commensal bacteria protect against food allergen sensitization.
Proc. Natl. Acad. Sci. USA
111
:
13145
13150
.
62
Herbst
,
T.
,
A.
Sichelstiel
,
C.
Schär
,
K.
Yadava
,
K.
Bürki
,
J.
Cahenzli
,
K.
McCoy
,
B. J.
Marsland
,
N. L.
Harris
.
2011
.
Dysregulation of allergic airway inflammation in the absence of microbial colonization.
Am. J. Respir. Crit. Care Med.
184
:
198
205
.
63
Kamada
,
N.
,
G. Y.
Chen
,
N.
Inohara
,
G.
Núñez
.
2013
.
Control of pathogens and pathobionts by the gut microbiota.
Nat. Immunol.
14
:
685
690
.
64
Rakoff-Nahoum
,
S.
,
J.
Paglino
,
F.
Eslami-Varzaneh
,
S.
Edberg
,
R.
Medzhitov
.
2004
.
Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis.
Cell
118
:
229
241
.
65
Li
,
X. D.
,
Y. H.
Chiu
,
A. S.
Ismail
,
C. L.
Behrendt
,
M.
Wight-Carter
,
L. V.
Hooper
,
Z. J.
Chen
.
2011
.
Mitochondrial antiviral signaling protein (MAVS) monitors commensal bacteria and induces an immune response that prevents experimental colitis.
Proc. Natl. Acad. Sci. USA
108
:
17390
17395
.
66
Fischer
,
J. C.
,
M.
Bscheider
,
G.
Eisenkolb
,
C. C.
Lin
,
A.
Wintges
,
V.
Otten
,
C. A.
Lindemans
,
S.
Heidegger
,
M.
Rudelius
,
S.
Monette
, et al
.
2017
.
RIG-I/MAVS and STING signaling promote gut integrity during irradiation- and immune-mediated tissue injury.
Sci Transl Med
9
:
eaag2513
.
67
Ott
,
S. J.
,
G. H.
Waetzig
,
A.
Rehman
,
J.
Moltzau-Anderson
,
R.
Bharti
,
J. A.
Grasis
,
L.
Cassidy
,
A.
Tholey
,
H.
Fickenscher
,
D.
Seegert
, et al
.
2017
.
Efficacy of sterile fecal filtrate transfer for treating patients with clostridium difficile infection.
Gastroenterology
152
:
799
811.e7
.
68
Zuo
,
T.
,
S. H.
Wong
,
K.
Lam
,
R.
Lui
,
K.
Cheung
,
W.
Tang
,
J. Y. L.
Ching
,
P. K. S.
Chan
,
M. C. W.
Chan
,
J. C. Y.
Wu
, et al
.
2018
.
Bacteriophage transfer during faecal microbiota transplantation in Clostridium difficile infection is associated with treatment outcome.
Gut
67
:
634
643
.
69
Thépaut
,
M.
,
T.
Grandjean
,
D.
Hober
,
P. E.
Lobert
,
P.
Bortolotti
,
K.
Faure
,
R.
Dessein
,
E.
Kipnis
,
B.
Guery
.
2015
.
Protective role of murine norovirus against Pseudomonas aeruginosa acute pneumonia.
Vet. Res.
46
:
91
.
70
Phillips
,
G.
,
C. C.
Tam
,
L. C.
Rodrigues
,
B.
Lopman
.
2010
.
Prevalence and characteristics of asymptomatic norovirus infection in the community in England.
Epidemiol. Infect.
138
:
1454
1458
.
71
Yang
,
J. Y.
,
M. S.
Kim
,
E.
Kim
,
J. H.
Cheon
,
Y. S.
Lee
,
Y.
Kim
,
S. H.
Lee
,
S. U.
Seo
,
S. H.
Shin
,
S. S.
Choi
, et al
.
2016
.
Enteric viruses ameliorate gut inflammation via Toll-like receptor 3 and Toll-like receptor 7-mediated interferon-β production.
Immunity
44
:
889
900
.
72
Sun
,
L.
,
H.
Miyoshi
,
S.
Origanti
,
T. J.
Nice
,
A. C.
Barger
,
N. A.
Manieri
,
L. A.
Fogel
,
A. R.
French
,
D.
Piwnica-Worms
,
H.
Piwnica-Worms
, et al
.
2015
.
Type I interferons link viral infection to enhanced epithelial turnover and repair.
Cell Host Microbe
17
:
85
97
.
73
Cammarota
,
G.
,
L.
Cuoco
,
R.
Cianci
,
F.
Pandolfi
,
G.
Gasbarrini
.
2000
.
Onset of coeliac disease during treatment with interferon for chronic hepatitis C.
Lancet
356
:
1494
1495
.
74
Di Sabatino
,
A.
,
K. M.
Pickard
,
J. N.
Gordon
,
V.
Salvati
,
G.
Mazzarella
,
R. M.
Beattie
,
A.
Vossenkaemper
,
L.
Rovedatti
,
N. A.
Leakey
,
N. M.
Croft
, et al
.
2007
.
Evidence for the role of interferon-alfa production by dendritic cells in the Th1 response in celiac disease.
Gastroenterology
133
:
1175
1187
.
75
Lerner
,
A.
,
M.
Arleevskaya
,
A.
Schmiedl
,
T.
Matthias
.
2017
.
Microbes and viruses are bugging the gut in celiac disease. Are they friends or foes?
Front. Microbiol.
8
:
1392
.
76
Bouziat
,
R.
,
R.
Hinterleitner
,
J. J.
Brown
,
J. E.
Stencel-Baerenwald
,
M.
Ikizler
,
T.
Mayassi
,
M.
Meisel
,
S. M.
Kim
,
V.
Discepolo
,
A. J.
Pruijssers
, et al
.
2017
.
Reovirus infection triggers inflammatory responses to dietary antigens and development of celiac disease.
Science
356
:
44
50
.
77
Huttenhower
,
C.
,
A. D.
Kostic
,
R. J.
Xavier
.
2014
.
Inflammatory bowel disease as a model for translating the microbiome.
Immunity
40
:
843
854
.
78
Cadwell
,
K.
,
J. Y.
Liu
,
S. L.
Brown
,
H.
Miyoshi
,
J.
Loh
,
J. K.
Lennerz
,
C.
Kishi
,
W.
Kc
,
J. A.
Carrero
,
S.
Hunt
, et al
.
2008
.
A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells.
Nature
456
:
259
263
.
79
Adolph
,
T. E.
,
M. F.
Tomczak
,
L.
Niederreiter
,
H. J.
Ko
,
J.
Böck
,
E.
Martinez-Naves
,
J. N.
Glickman
,
M.
Tschurtschenthaler
,
J.
Hartwig
,
S.
Hosomi
, et al
.
2013
.
Paneth cells as a site of origin for intestinal inflammation.
Nature
503
:
272
276
.
80
Cadwell
,
K.
,
K. K.
Patel
,
N. S.
Maloney
,
T. C.
Liu
,
A. C.
Ng
,
C. E.
Storer
,
R. D.
Head
,
R.
Xavier
,
T. S.
Stappenbeck
,
H. W.
Virgin
.
2010
.
Virus-plus-susceptibility gene interaction determines Crohn’s disease gene Atg16L1 phenotypes in intestine.
Cell
141
:
1135
1145
.
81
Matsuzawa-Ishimoto
,
Y.
,
Y.
Shono
,
L. E.
Gomez
,
V. M.
Hubbard-Lucey
,
M.
Cammer
,
J.
Neil
,
M. Z.
Dewan
,
S. R.
Lieberman
,
A.
Lazrak
,
J. M.
Marinis
, et al
.
2017
.
Autophagy protein ATG16L1 prevents necroptosis in the intestinal epithelium.
J. Exp. Med.
214
:
3687
3705
.
82
Lencioni
,
K. C.
,
A.
Seamons
,
P. M.
Treuting
,
L.
Maggio-Price
,
T.
Brabb
.
2008
.
Murine norovirus: an intercurrent variable in a mouse model of bacteria-induced inflammatory bowel disease.
Comp. Med.
58
:
522
533
.
83
Basic
,
M.
,
L. M.
Keubler
,
M.
Buettner
,
M.
Achard
,
G.
Breves
,
B.
Schröder
,
A.
Smoczek
,
A.
Jörns
,
D.
Wedekind
,
N. H.
Zschemisch
, et al
.
2014
.
Norovirus triggered microbiota-driven mucosal inflammation in interleukin 10-deficient mice.
Inflamm. Bowel Dis.
20
:
431
443
.
84
Hubbard
,
V. M.
,
K.
Cadwell
.
2011
.
Viruses, autophagy genes, and Crohn’s disease.
Viruses
3
:
1281
1311
.
85
Nyström
,
N.
,
T.
Berg
,
E.
Lundin
,
O.
Skog
,
I.
Hansson
,
G.
Frisk
,
I.
Juko-Pecirep
,
M.
Nilsson
,
U.
Gyllensten
,
Y.
Finkel
, et al
.
2013
.
Human enterovirus species B in ileocecal Crohn’s disease.
Clin. Transl. Gastroenterol.
4
:
e38
.
86
Norman
,
J. M.
,
S. A.
Handley
,
M. T.
Baldridge
,
L.
Droit
,
C. Y.
Liu
,
B. C.
Keller
,
A.
Kambal
,
C. L.
Monaco
,
G.
Zhao
,
P.
Fleshner
, et al
.
2015
.
Disease-specific alterations in the enteric virome in inflammatory bowel disease.
Cell
160
:
447
460
.
87
Nakatsu
,
G.
,
H.
Zhou
,
W. K. K.
Wu
,
S. H.
Wong
,
O. O.
Coker
,
Z.
Dai
,
X.
Li
,
C. H.
Szeto
,
N.
Sugimura
,
T.
Yuen-Tung Lam
, et al
.
2018
.
Alterations in enteric virome are associated with colorectal cancer and survival outcomes.
Gastroenterology
. DOI: .
88
Legoff
,
J.
,
M.
Resche-Rigon
,
J.
Bouquet
,
M.
Robin
,
S. N.
Naccache
,
S.
Mercier-Delarue
,
S.
Federman
,
E.
Samayoa
,
C.
Rousseau
,
P.
Piron
, et al
.
2017
.
The eukaryotic gut virome in hematopoietic stem cell transplantation: new clues in enteric graft-versus-host disease.
Nat. Med.
23
:
1080
1085
.
89
Turley
,
S. J.
,
J. W.
Lee
,
N.
Dutton-Swain
,
D.
Mathis
,
C.
Benoist
.
2005
.
Endocrine self and gut non-self intersect in the pancreatic lymph nodes.
Proc. Natl. Acad. Sci. USA
102
:
17729
17733
.
90
Smyth
,
D. J.
,
J. D.
Cooper
,
R.
Bailey
,
S.
Field
,
O.
Burren
,
L. J.
Smink
,
C.
Guja
,
C.
Ionescu-Tirgoviste
,
B.
Widmer
,
D. B.
Dunger
, et al
.
2006
.
A genome-wide association study of nonsynonymous SNPs identifies a type 1 diabetes locus in the interferon-induced helicase (IFIH1) region.
Nat. Genet.
38
:
617
619
.
91
Jean-Baptiste
,
V. S. E.
,
C. Q.
Xia
,
M. J.
Clare-Salzler
,
M. S.
Horwitz
.
2017
.
Type 1 diabetes and type 1 interferonopathies: localization of a type 1 common thread of virus infection in the pancreas.
EBioMedicine
22
:
10
17
.
92
Laitinen
,
O. H.
,
H.
Honkanen
,
O.
Pakkanen
,
S.
Oikarinen
,
M. M.
Hankaniemi
,
H.
Huhtala
,
T.
Ruokoranta
,
V.
Lecouturier
,
P.
André
,
R.
Harju
, et al
.
2014
.
Coxsackievirus B1 is associated with induction of β-cell autoimmunity that portends type 1 diabetes.
Diabetes
63
:
446
455
.
93
Stone
,
V. M.
,
M. M.
Hankaniemi
,
E.
Svedin
,
A.
Sioofy-Khojine
,
S.
Oikarinen
,
H.
Hyöty
,
O. H.
Laitinen
,
V. P.
Hytönen
,
M.
Flodström-Tullberg
.
2018
.
A Coxsackievirus B vaccine protects against virus-induced diabetes in an experimental mouse model of type 1 diabetes.
Diabetologia
61
:
476
481
.
94
Honeyman
,
M. C.
,
B. S.
Coulson
,
N. L.
Stone
,
S. A.
Gellert
,
P. N.
Goldwater
,
C. E.
Steele
,
J. J.
Couper
,
B. D.
Tait
,
P. G.
Colman
,
L. C.
Harrison
.
2000
.
Association between rotavirus infection and pancreatic islet autoimmunity in children at risk of developing type 1 diabetes.
Diabetes
49
:
1319
1324
.
95
Graham
,
K. L.
,
N.
Sanders
,
Y.
Tan
,
J.
Allison
,
T. W.
Kay
,
B. S.
Coulson
.
2008
.
Rotavirus infection accelerates type 1 diabetes in mice with established insulitis.
J. Virol.
82
:
6139
6149
.
96
Pane
,
J. A.
,
N. L.
Webster
,
B. S.
Coulson
.
2014
.
Rotavirus activates lymphocytes from non-obese diabetic mice by triggering toll-like receptor 7 signaling and interferon production in plasmacytoid dendritic cells.
PLoS Pathog.
10
:
e1003998
.
97
Pane
,
J. A.
,
F. E.
Fleming
,
K. L.
Graham
,
H. E.
Thomas
,
T. W.
Kay
,
B. S.
Coulson
.
2016
.
Rotavirus acceleration of type 1 diabetes in non-obese diabetic mice depends on type I interferon signalling.
Sci. Rep.
6
:
29697
.
98
Pane
,
J. A.
,
N. L.
Webster
,
K. L.
Graham
,
G.
Holloway
,
C.
Zufferey
,
B. S.
Coulson
.
2013
.
Rotavirus acceleration of murine type 1 diabetes is associated with a T helper 1-dependent specific serum antibody response and virus effects in regional lymph nodes.
Diabetologia
56
:
573
582
.
99
Graham
,
K. L.
,
J. A.
O’Donnell
,
Y.
Tan
,
N.
Sanders
,
E. M.
Carrington
,
J.
Allison
,
B. S.
Coulson
.
2007
.
Rotavirus infection of infant and young adult nonobese diabetic mice involves extraintestinal spread and delays diabetes onset.
J. Virol.
81
:
6446
6458
.
100
Wetzel
,
J. D.
,
E. S.
Barton
,
J. D.
Chappell
,
G. S.
Baer
,
M.
Mochow-Grundy
,
S. E.
Rodgers
,
Y.
Shyr
,
A. C.
Powers
,
J. W.
Thomas
,
T. S.
Dermody
.
2006
.
Reovirus delays diabetes onset but does not prevent insulitis in nonobese diabetic mice.
J. Virol.
80
:
3078
3082
.
101
Zhao
,
G.
,
T.
Vatanen
,
L.
Droit
,
A.
Park
,
A. D.
Kostic
,
T. W.
Poon
,
H.
Vlamakis
,
H.
Siljander
,
T.
Härkönen
,
A. M.
Hämäläinen
, et al
.
2017
.
Intestinal virome changes precede autoimmunity in type I diabetes-susceptible children.
Proc. Natl. Acad. Sci. USA
114
:
E6166
E6175
.
102
Pearson
,
J. A.
,
F. S.
Wong
,
L.
Wen
.
2016
.
The importance of the non obese diabetic (NOD) mouse model in autoimmune diabetes.
J. Autoimmun.
66
:
76
88
.
103
Brenchley
,
J. M.
,
D. C.
Douek
.
2008
.
HIV infection and the gastrointestinal immune system.
Mucosal Immunol.
1
:
23
30
.
104
Monaco
,
C. L.
,
D. B.
Gootenberg
,
G.
Zhao
,
S. A.
Handley
,
M. S.
Ghebremichael
,
E. S.
Lim
,
A.
Lankowski
,
M. T.
Baldridge
,
C. B.
Wilen
,
M.
Flagg
, et al
.
2016
.
Altered virome and bacterial microbiome in human immunodeficiency virus-associated acquired immunodeficiency syndrome.
Cell Host Microbe
19
:
311
322
.
105
Handley
,
S. A.
,
L. B.
Thackray
,
G.
Zhao
,
R.
Presti
,
A. D.
Miller
,
L.
Droit
,
P.
Abbink
,
L. F.
Maxfield
,
A.
Kambal
,
E.
Duan
, et al
.
2012
.
Pathogenic simian immunodeficiency virus infection is associated with expansion of the enteric virome.
Cell
151
:
253
266
.
106
Brenchley
,
J. M.
,
D. A.
Price
,
T. W.
Schacker
,
T. E.
Asher
,
G.
Silvestri
,
S.
Rao
,
Z.
Kazzaz
,
E.
Bornstein
,
O.
Lambotte
,
D.
Altmann
, et al
.
2006
.
Microbial translocation is a cause of systemic immune activation in chronic HIV infection.
Nat. Med.
12
:
1365
1371
.
107
Polakos
,
N. K.
,
J. C.
Cornejo
,
D. A.
Murray
,
K. O.
Wright
,
J. J.
Treanor
,
I. N.
Crispe
,
D. J.
Topham
,
R. H.
Pierce
.
2006
.
Kupffer cell-dependent hepatitis occurs during influenza infection.
Am. J. Pathol.
168
:
1169
1178
, quiz 1404–1405.
108
Wang
,
J.
,
F.
Li
,
H.
Wei
,
Z. X.
Lian
,
R.
Sun
,
Z.
Tian
.
2014
.
Respiratory influenza virus infection induces intestinal immune injury via microbiota-mediated Th17 cell-dependent inflammation. [Published errata appear in 2014 J. Exp. Med. 211: 2396–2397 and 2014 J. Exp. Med. 211: 2683.]
J. Exp. Med.
211
:
2397
2410
.
109
Barton
,
E. S.
,
D. W.
White
,
J. S.
Cathelyn
,
K. A.
Brett-McClellan
,
M.
Engle
,
M. S.
Diamond
,
V. L.
Miller
,
H. W.
Virgin
IV
.
2007
.
Herpesvirus latency confers symbiotic protection from bacterial infection.
Nature
447
:
326
329
.
110
Furman
,
D.
,
V.
Jojic
,
S.
Sharma
,
S. S.
Shen-Orr
,
C. J.
Angel
,
S.
Onengut-Gumuscu
,
B. A.
Kidd
,
H. T.
Maecker
,
P.
Concannon
,
C. L.
Dekker
, et al
.
2015
.
Cytomegalovirus infection enhances the immune response to influenza.
Sci. Transl. Med.
7
:
281ra43
.
111
White
,
D. W.
,
C. R.
Keppel
,
S. E.
Schneider
,
T. A.
Reese
,
J.
Coder
,
J. E.
Payton
,
T. J.
Ley
,
H. W.
Virgin
,
T. A.
Fehniger
.
2010
.
Latent herpesvirus infection arms NK cells.
Blood
115
:
4377
4383
.
112
MacDuff
,
D. A.
,
T. A.
Reese
,
J. M.
Kimmey
,
L. A.
Weiss
,
C.
Song
,
X.
Zhang
,
A.
Kambal
,
E.
Duan
,
J. A.
Carrero
,
B.
Boisson
, et al
.
2015
.
Phenotypic complementation of genetic immunodeficiency by chronic herpesvirus infection.
Elife
4
:
e04494
.
113
Machiels
,
B.
,
M.
Dourcy
,
X.
Xiao
,
J.
Javaux
,
C.
Mesnil
,
C.
Sabatel
,
D.
Desmecht
,
F.
Lallemand
,
P.
Martinive
,
H.
Hammad
, et al
.
2017
.
A gammaherpesvirus provides protection against allergic asthma by inducing the replacement of resident alveolar macrophages with regulatory monocytes. [Published erratum appears in 2018 Nat. Immunol. DOI: 10.1038/s41590-017-0024-8.]
Nat. Immunol.
18
:
1310
1320
.
114
Brodin
,
P.
,
V.
Jojic
,
T.
Gao
,
S.
Bhattacharya
,
C. J.
Angel
,
D.
Furman
,
S.
Shen-Orr
,
C. L.
Dekker
,
G. E.
Swan
,
A. J.
Butte
, et al
.
2015
.
Variation in the human immune system is largely driven by non-heritable influences.
Cell
160
:
37
47
.
115
Moustafa
,
A.
,
C.
Xie
,
E.
Kirkness
,
W.
Biggs
,
E.
Wong
,
Y.
Turpaz
,
K.
Bloom
,
E.
Delwart
,
K. E.
Nelson
,
J. C.
Venter
,
A.
Telenti
.
2017
.
The blood DNA virome in 8,000 humans.
PLoS Pathog.
13
:
e1006292
.
116
Bernardin
,
F.
,
E.
Operskalski
,
M.
Busch
,
E.
Delwart
.
2010
.
Transfusion transmission of highly prevalent commensal human viruses.
Transfusion
50
:
2474
2483
.
117
Williams
,
C. F.
,
D.
Klinzman
,
T. E.
Yamashita
,
J.
Xiang
,
P. M.
Polgreen
,
C.
Rinaldo
,
C.
Liu
,
J.
Phair
,
J. B.
Margolick
,
D.
Zdunek
, et al
.
2004
.
Persistent GB virus C infection and survival in HIV-infected men.
N. Engl. J. Med.
350
:
981
990
.
118
Willner
,
D.
,
M.
Furlan
,
M.
Haynes
,
R.
Schmieder
,
F. E.
Angly
,
J.
Silva
,
S.
Tammadoni
,
B.
Nosrat
,
D.
Conrad
,
F.
Rohwer
.
2009
.
Metagenomic analysis of respiratory tract DNA viral communities in cystic fibrosis and non-cystic fibrosis individuals.
PLoS One
4
:
e7370
.
119
Goffard
,
A.
,
V.
Lambert
,
J.
Salleron
,
S.
Herwegh
,
I.
Engelmann
,
C.
Pinel
,
I.
Pin
,
T.
Perrez
,
A.
Prévotat
,
A.
Dewilde
,
L.
Delhaes
.
2014
.
Virus and cystic fibrosis: rhinoviruses are associated with exacerbations in adult patients.
J. Clin. Virol.
60
:
147
153
.
120
Dijkema
,
J. S.
,
B. E.
van Ewijk
,
B.
Wilbrink
,
T. F.
Wolfs
,
J. L.
Kimpen
,
C. K.
van der Ent
.
2016
.
Frequency and duration of rhinovirus infections in children with cystic fibrosis and healthy controls: a longitudinal cohort study.
Pediatr. Infect. Dis. J.
35
:
379
383
.
121
Cousin
,
M.
,
N.
Molinari
,
V.
Foulongne
,
D.
Caimmi
,
I.
Vachier
,
M.
Abely
,
R.
Chiron
.
2016
.
Rhinovirus-associated pulmonary exacerbations show a lack of FEV1 improvement in children with cystic fibrosis.
Influenza Other Respir. Viruses
10
:
109
112
.
122
Edwards
,
M. R.
,
K.
Strong
,
A.
Cameron
,
R. P.
Walton
,
D. J.
Jackson
,
S. L.
Johnston
.
2017
.
Viral infections in allergy and immunology: how allergic inflammation influences viral infections and illness.
J. Allergy Clin. Immunol.
140
:
909
920
.
123
Bizzintino
,
J.
,
W. M.
Lee
,
I. A.
Laing
,
F.
Vang
,
T.
Pappas
,
G.
Zhang
,
A. C.
Martin
,
S. K.
Khoo
,
D. W.
Cox
,
G. C.
Geelhoed
, et al
.
2011
.
Association between human rhinovirus C and severity of acute asthma in children.
Eur. Respir. J.
37
:
1037
1042
.
124
Bochkov
,
Y. A.
,
K.
Watters
,
S.
Ashraf
,
T. F.
Griggs
,
M. K.
Devries
,
D. J.
Jackson
,
A. C.
Palmenberg
,
J. E.
Gern
.
2015
.
Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication.
Proc. Natl. Acad. Sci. USA
112
:
5485
5490
.
125
Palmenberg
,
A. C.
2017
.
Rhinovirus C, asthma, and cell surface expression of virus receptor CDHR3.
J. Virol.
91
:
e00072-17
.
126
Currier
,
R. L.
,
D. C.
Payne
,
M. A.
Staat
,
R.
Selvarangan
,
S. H.
Shirley
,
N.
Halasa
,
J. A.
Boom
,
J. A.
Englund
,
P. G.
Szilagyi
,
C. J.
Harrison
, et al
.
2015
.
Innate susceptibility to norovirus infections influenced by FUT2 genotype in a United States pediatric population.
Clin. Infect. Dis.
60
:
1631
1638
.
127
Lopman
,
B. A.
,
T.
Trivedi
,
Y.
Vicuña
,
V.
Costantini
,
N.
Collins
,
N.
Gregoricus
,
U.
Parashar
,
C.
Sandoval
,
N.
Broncano
,
M.
Vaca
, et al
.
2015
.
Norovirus infection and disease in an ecuadorian birth cohort: association of certain norovirus genotypes with host FUT2 secretor status.
J. Infect. Dis.
211
:
1813
1821
.
128
Briese
,
T.
,
A.
Kapoor
,
N.
Mishra
,
K.
Jain
,
A.
Kumar
,
O. J.
Jabado
,
W. I.
Lipkin
.
2015
.
Virome capture sequencing enables sensitive viral diagnosis and comprehensive virome analysis. [Published erratum appears in 2017 MBio. 8: e00615-17.]
MBio.
6
:
e01491
e15
.
129
Kaufman
,
H. L.
,
F. J.
Kohlhapp
,
A.
Zloza
.
2015
.
Oncolytic viruses: a new class of immunotherapy drugs. [Published erratum appears in 2016 Nat. Rev. Drug Discov. 15: 143.]
Nat. Rev. Drug Discov.
14
:
642
662
.
130
Samson
,
A.
,
K. J.
Scott
,
D.
Taggart
,
E. J.
West
,
E.
Wilson
,
G. J.
Nuovo
,
S.
Thomson
,
R.
Corns
,
R. K.
Mathew
,
M. J.
Fuller
, et al
.
2018
.
Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade.
Sci Transl Med
10
:
eaam7577
.
131
Bourgeois-Daigneault
,
M. C.
,
D. G.
Roy
,
A. S.
Aitken
,
N.
El Sayes
,
N. T.
Martin
,
O.
Varette
,
T.
Falls
,
L. E.
St-Germain
,
A.
Pelin
,
B. D.
Lichty
, et al
.
2018
.
Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy.
Sci Transl Med
10
:
eaao1641
.

The authors have no financial conflicts of interest.