Animal Models of Norovirus Infection


Chapter 3.4

Animal Models of Norovirus Infection



C.E. Wobus*

J.B. Cunha*

M.D. Elftman**

A.O. Kolawole*
*    Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
**    Department of Biomedical and Diagnostic Sciences, University of Detroit Mercy, School of Dentistry, Detroit, MI, United States


Abstract


Noroviruses are highly prevalent, positive-sense RNA viruses that are transmitted by the fecal–oral route and infect the gastrointestinal tract of their host. Human noroviruses cause the majority of acute nonbacterial gastroenteritis infections worldwide, resulting in significant morbidity, mortality, and economic losses. The study of human viruses in their native host has inherent limitations. Thus, studies in animal models are critical for gaining a greater understanding of viral pathogenesis as well as the development of vaccines and therapeutics. Here, we describe multiple models that exist to study human noroviruses in nonhuman hosts and related viruses in their native hosts. The strengths and limitations of each of these models will also be discussed. Together, knowledge gained from these different models has significantly increased our understanding of these important human pathogens.



Keywords


animal models of infection

positive sense RNA virus

calicivirus

norovirus

enteric virus pathogenesis


1. Introduction


Advances in the understanding of human noroviruses (HuNoVs) are challenging because, despite being such ubiquitous pathogens, these viruses cause species-specific infections and, until very recently (Jones et al., 2014), have not been successfully cultured in vitro. The use of animal models to address questions that are difficult or impossible to answer in human studies is a common approach to complement in vitro and epidemiological data for human viruses in general. Animal models are based either on studies of human viruses in animals, often requiring the use of immunocompromised hosts and/or host-adapted viruses, or the use of related viruses in their natural hosts. As outlined in this chapter, both approaches have been used to study HuNoVs.

Human volunteer-based studies and epidemiologic analyses have been invaluable in determining features of HuNoV infection and disease (Vashist et al., 2009Kaufman et al., 2014). These are: (1) short incubation time (average 1 day) (Lee et al., 2013); (2) quick (1–4 days) resolution of disease symptoms, characterized mainly by diarrhea and/or vomiting (Kaufman et al., 2014Atmar et al., 2008); (3) viral shedding for weeks to months with or without disease symptoms (Kaufman et al., 2014Teunis et al., 2014); (4) viral spread mainly by the fecal–oral route and person-to-person transmission (Bitler et al., 2013), (5) histo-blood group antigen (HBGA) expression and secretor status as genetic susceptibility factors (Lindesmith et al., 2003; Hutson et al., 2002 2005; Tan and Jiang, 2014). However, studies relying on human subjects have ethical limitations and require vast resources, therefore preventing them from being routinely conducted (Vashist et al., 2009). Therefore, animal models for HuNoV infection, as well as HuNoV surrogates, are valuable and necessary tools that can elucidate basic aspects of HuNoV infection and pathogenesis (Karst et al., 2014Kniel, 2014).

Ideally, the best HuNoV animal model would use HuNoV or a virus with high genetic similarity that closely mirrors the biology and clinical features of HuNoV infection in an animal host. However, HuNoVs are members of the genetically diverse Norovirus genus within the Caliciviridae family that exhibit very narrow species specificity. Thus, HuNoV infection in a nonnative host is limited to some closely related primates (chimpanzees, newborn pigtail macaques), immunocompromised animals (ie, mice), and gnotobiotic (Gn) pigs and calves, albeit with varying levels of clinical signs (Bok et al., 2011Cheetham et al., 2006Souza et al., 2008Taube et al., 2013). Alternatively, many studies have been carried out using related caliciviruses as HuNoV surrogate models. Historically, feline calicivirus (FCV) was the first surrogate for human caliciviruses due to the availability of an established cell culture system, and its study provided an early understanding of calicivirus biology (Radford et al., 2004). However, FCV causes upper respiratory tract infections in cats (Radford et al., 2007), and therefore has limited utility as a model for gastroenteritis and will not be discussed further. In contrast, porcine sapovirus (PoSaV) causes diarrhea in swine and can be propagated in culture in the presence of bile acids (Flynn and Saif, 1988), providing an animal model of calicivirus gastroenteritis. However, it was not until the discovery of murine norovirus 1 (MNV-1), a norovirus that infects a small animal species (Karst et al., 2003) and the subsequent development of a cell culture model (Wobus et al., 2004), that a HuNoV surrogate became widely available for both in vitro and in vivo studies (Wobus et al., 2006). MNV has been instrumental in elucidating many basic aspects of NoV infection and pathogenesis (Karst et al., 2003; Gonzalez-Hernandez et al., 2013 2014; Taube et al., 2012 2009; Chachu et al., 2008). More recently, Tulane virus (TV) was discovered as a causative agent of diarrhea in rhesus macaques (Sestak et al., 2012). Combined with the ability to readily grow in cell culture (Farkas et al., 2008) and availability of an infectious clone (Wei et al., 2008), TV is another important HuNoV surrogate (Farkas, 2015). In this chapter, the characteristics of HuNoV animal and surrogate models under investigation in the field will be discussed, including their advantages and disadvantages. We will also point out contributions of each model to our collective understanding of NoV biology and therapeutic approaches for HuNoVs.

2. Animal models of HuNoV infection


To date, a limited number of HuNoV strains was shown to infect mice deficient in the recombination activating gene (Rag) and the common gamma chain (γc) (Taube et al., 2013), gnotobiotic pigs (Souza et al., 2007a), gnotobiotic calves (Souza et al., 2008), macaques (Subekti et al., 2002), and chimpanzees (Bok et al., 2011). So far, no single animal model recapitulates all aspects of HuNoV infection. As outlined later, each model has strengths to allow the study of specific aspects of HuNoV biology (Table 3.4.1).


Table 3.4.1


Summary of the Discussed HuNoV Animal Models With Their Strengths and Limitations



































Host Virus Strengths Limitations
Balb/c Rag2/γc−/− mouse (Taube et al., 2013) HuNoV (GII.4)

Genetically tractable host


Relatively low cost


Immune defects in host


Transient infection


Not orally susceptible


Asymptomatic infection


Lack of fecal shedding


Not natural host

Chimpanzee (Bok et al., 2011) HuNoV (GI.1, untyped strain)

Genetically similar to human


Orally susceptible


Transmission between animals


Similar shedding and antibody responses to humans


Asymptomatic infection


High cost


Not natural host


Moratorium on use

Newborn pigtail macaques (Subekti et al., 2002) HuNoV (GII.3)

Orally susceptible


Symptomatic infection


Fecal shedding


Similar serum IgG responses to humans


Transmission between animals


High cost


Unknown IgA responses


Not natural host

Gnotobiotic pigs (Cheetham et al., 2006; Souza et al., 2007a; Bui et al., 2013; Takanashi et al., 2011; Cheetham et al., 2007) HuNoV (GII.4, GII.12)

Symptomatic and asymptomatic infections


Orally susceptible


Fecal shedding


Susceptibility partially mediated by HBGAs


Similar incubation period, duration of symptoms to humans


Mild villous atrophy


Similar cytokine, antibody responses to humans


High cost


Limited ability to passage


Lack of microbiota


Underdeveloped immune system


Not natural host

Gnotobiotic calves (Souza et al., 2008) HuNoV (GII.4)

Symptomatic infection


Orally susceptible


Fecal shedding


Villous atrophy


High cost


Lack of microbiota


Underdeveloped immune system


Not natural host





2.1. Mice as an Animal Model


The first small animal model to study HuNoV infection was described in 2013 (Taube et al., 2013). Balb/c mice deficient in recombination activating genes (Rag−/−) 1 or 2 and common gamma chain (γc−/−) (Rag−/−/γc−/−) engrafted with human CD34+ hematopoietic stem cells (humanized mice), nonengrafted siblings, and immunocompetent wild type controls were challenged with a pool of HuNoV strains (Fig. 3.4.1). Viral genome titers were measured by quantitative polymerase chain reaction preceded by reverse transcription (RT-qPCR) in intestinal and nonintestinal tissues. Infection was measured by detecting increased genome titers over input in both humanized and nonhumanized Balb/c Rag−/−/γc−/− mice. Viral genomes were present in all tissues analyzed, except the brain. However, infection was asymptomatic and transient (lasting 2 days). Immunocompetent wild type Balb/c or B6/B10 Rag−/−/γc−/− mice were not infected, suggesting both the immune deficiency and genetic background of the mouse as determinants of susceptibility in this model. Unlike natural HuNoV infection, mice were susceptible only via the intraperitoneal, not the oral route, and no shedding of virus in the stool was detected. The former is likely a result of the genetic defects of this mouse strain, which lacks Peyer’s patches and associated M cells, a known gateway for NoV entry into the host (Gonzalez-Hernandez et al., 2014). Expression of structural and/or nonstructural proteins in cells with macrophage-like morphology in the spleen, small intestinal lamina propria, and Kupffer cells in the liver of Balb/c Rag−/−/γc−/−, but not of mock-infected mice, verified replication of HuNoV.

image

Figure 3.4.1 Human Norovirus Mouse Model.
Balb/c mice lacking the recombination activation genes (Rag) 1 or 2 and common gamma chain (γc) (Rag−/−γc−/−) were humanized with CD34+ hematopoietic stem cells. Humanized and nonhumanized Balb/c Rag−/−γc−/−, wild-type (WT) Balb/c controls and B6/B10 Rag−/−γc−/− mice were challenged with a pool of HuNoV strains by intraperitoneal (i.p.) and/or oral (p.o.) routes. Viral infection was determined by detecting increased viral genome titers over input and viral protein expression by immunohistochemistry. Productive infection routes are indicated by green arrows and presence of a virion, while nonproductive infections are indicated by black arrows and crossed-out virions. (Source: Summary of data from Taube et al., 2013.)

A mouse model of HuNoV infection can facilitate translational and basic science applications, such as drug development and identification of viral and host factors mediating infection. The advantages of a mouse model as opposed to large animal models are manifold, including reduced cost, shorter gestation length, and the ability to genetically manipulate the host. However, the necessity for deleting components of the immune system to permit HuNoV to cross the species barrier represents an inherent caveat that limits the use of this model for certain applications. For example, vaccine testing is not possible due to the absence of T and B cells in Rag-deficient animals. Therefore, the identification of mouse strains with other immune deficiencies that permit HuNoV infection could expand the utility of the mouse model. Furthermore, the discovery of different permissive mouse strains that can be orally infected, shed virus in feces and have a longer duration of infection, which would improve this model. Some of the open questions to address are the minimum number of particles required to initiate an infection, the ability of different HuNoV genotypes to cause an infection, and the ability for serial passaging and adaptation of HuNoV in the murine host.

2.2. Chimpanzees as an Animal Model


The close genetic relatedness of chimpanzees to humans led to the use of chimpanzees as animal model for human-specific pathogens. HuNoV infections of chimpanzees were first performed in 1978 (Wyatt et al., 1978). Although none of the chimpanzees challenged orally with filtered HuNoV-containing fecal samples developed symptoms associated with HuNoV infection, infected animals developed NoV-specific antibody responses, and viral antigen was detected in feces. Importantly, HuNoV was serially passaged via feces to additional uninfected animals, demonstrating that chimpanzees support asymptomatic HuNoV infection. This finding was confirmed a few decades later when Bok et al. (2011) intravenously inoculated seronegative chimpanzees with HuNoV. No clinical signs of gastroenteritis were observed but the onset and duration of virus shedding in stool and serum antibody responses were similar to those observed in humans (Fig. 3.4.2). HuNoV capsid protein was detected in intestinal lamina propria cells from the B cell and dendritic cell lineages, and NoV RNA was found in intestinal and liver biopsies. Two infected chimpanzees were protected against re-infection of homologous HuNoV when challenged either at 4 and 24 months, or at 10 months after the first infection. In addition, chimpanzees vaccinated intramuscularly with Norwalk virus (GI) virus-like particles (VLPs), but not those derived from GII VLPs, were protected from homologous Norwalk virus infection 18 months after vaccination and developed antibodies that blocked VLP binding to HBGAs in a surrogate neutralization assay. Although this study did not use the natural route of infection, it provided important information regarding the ability of VLP-based vaccination to protect from homologous but not heterologous, cross-genogroup challenge. Protection from homologous challenge is also observed in human clinical trials (Atmar et al., 2011), while vaccine efficacy after heterologous challenge remains to be studied in humans. These data demonstrate the utility of chimpanzees as a valuable infection model for HuNoV but the NIH moratorium on the use of chimpanzees for biomedical research has halted any future NIH-funded studies in this HuNoV animal model.

image

Figure 3.4.2 Human Norovirus Chimpanzee Model.
Seronegative chimpanzees were intravenously (i.v.) inoculated with genogroup I (GI) HuNoV (Norwalk virus). Virus replication was measured by RT-qPCR, virus protein by IHC and serum antibody by EIA. Infected chimpanzees were protected against re-infection with homologous HuNoV when rechallenged after the first infection or vaccinated intramuscularly (i.m.) with GI (Norwalk virus) VLPs. GII VLPs and placebo did not protect from challenge with GI HuNoV, thus providing important information regarding the ability of VLP-based vaccination to protect from homologous but not heterologous, cross-genotype challenge. (Source: Schematic summary of data from Bok et al., 2011.)

2.3. Macaques as an Animal Model


Nonhuman primates other than chimpanzees may provide an alternative HuNoV animal model but only limited infections in a few species have been performed to date. The susceptibility of common marmosets, cotton top tamarins, cynomolgus, and rhesus macaques to oral HuNoV infection was tested in one study (Rockx et al., 2005). Only rhesus macaques were found susceptible to Norwalk virus infection, albeit asymptomatically, with virus shedding in some animals and development of NoV-specific IgG and IgM, but not IgA, antibodies. While the asymptomatic infection status and duration of shedding resembles that found in humans (Graham et al., 1994Rockx et al., 2002), the lack of IgA responses limits the use of this species as a vaccine model.

More promising results were observed in a study which showed newborn pigtail macaques (Macaca nemestrina) to be symptomatically infected with Toronto virus P2-A (a GII.3 HuNoV strain) (Subekti et al., 2002). Two of three monkeys developed diarrhea and dehydration. All animals developed IgG antibody responses and shed virus in stool for at least 3 weeks. Similar development of disease and shedding results were obtained following passage into additional newborn macaques. In addition, the mothers of the infected infant monkeys became symptomatically infected via natural infection, demonstrating the ability of HuNoV to be transmitted and passaged in this species of monkeys. This promising first study warrants further development of Macaca nemestrina as a HuNoV disease and transmission model.

2.4. Gnotobiotic Pigs and Calves as Animal Models


Gnotobiotic (Gn) pigs are models for enteric diseases because their gastrointestinal anatomy and physiology are similar to that of humans (Saif et al., 1996). Gn pigs support symptomatic HuNoV infection characterized by mild diarrhea, transient viremia, viral shedding in feces, and the presence of HuNoV structural and nonstructural proteins in enterocytes (Cheetham et al., 2006) (Fig. 3.4.3). In one study (Cheetham et al., 2006), Gn pigs were orally inoculated with fecal filtrates of the NoV/GII/4/HS66/2001/US strain, or with pig-passaged intestinal contents. Two thirds of the inoculated animals developed mild diarrhea, 57% seroconverted, but only 44% of pigs shed low levels of virus in their feces, suggesting disease symptoms can develop without fecal shedding or seroconversion. Using different virus strains [a GII.4 2006b variant (Bui et al., 2013) or the GII.g/GII.12 HS206 strain (Takanashi et al., 2011)], more infected animals shed virus than developed diarrhea, demonstrating asymptomatic infections can occur in Gn pigs. Increases in genome titers over input were detected in most (18/22) pigs (Bui et al., 2013), confirming productive infection. The short incubation period (24–48 h) and duration of disease symptoms (1–3 days) or asymptomatic infection resemble clinical features of HuNoV infection in humans (Lee et al., 2013Lindesmith et al., 2003Lopman et al., 2004). To date, only GII.4 and GII.12 strains have been reported to infect pigs, but further studies are needed to test susceptibility of Gn pigs to additional genogroups/genotypes.

image

Figure 3.4.3 Human Norovirus Gnotobiotic Pig Model.
Gnotobiotic pigs support symptomatic HuNoV infection characterized by mild diarrhea, transient viremia, viral shedding in feces, and detection of HuNoV structural and nonstructural proteins in enterocytes by immunofluorescence microscopy. (Source: Schematic representation of findings from Cheetham et al., 2006 and Souza et al., 2007a.)

Susceptibility factors of HuNoV infection in the human population are secretor status and HBGA type (Lindesmith et al., 2003Hutson et al., 2002). A similar correlation between infection outcome and HBGA expression was seen in Gn pigs, wherein increased incidence of diarrhea, fecal shedding, and seroconversion rate were observed in pigs with the A+/H+ genotype compared to the A−/H− genotype (Cheetham et al., 2007).

Histopathologic examination of HuNoV infected Gn pigs detected mild villous atrophy in the duodenum of one out of seven animals and an increased number of apoptotic cells in intestinal villi of all infected animals (Cheetham et al., 2006), features also reported during HuNoV infection (Troeger et al., 2009). Structural or nonstructural HuNoV antigens were detected in enterocytes of the proximal small intestine, suggesting HuNoV replication occurs in these cells, but whether additional cell types also support infection in this model remains unknown. The tropism for enterocytes differs from the recently described tropism of HuNoV for B cells in vitro (Jones et al., 2014), but the cellular tropism of HuNoV in infected human intestines remains undefined (Lay et al., 2010).

The power of animals models for pathogenesis studies is highlighted by a study where naïve Gn pigs were infected with the HuNoV HS66 GII.4 strain, and serum and intestinal contents were analyzed for intestinal and systemic humoral and cellular immune responses (Souza et al., 2007a). Such studies are difficult to perform in human volunteers with unknown and variable NoV-exposure history. Infection induced low antibody titers and antibody-secreting cell numbers, which may explain the incomplete seroconversion rate observed in pigs. However, the kinetics of IgA and IgG antibody-secreting cell responses and the greater induction of a Th1 compared to Th2 and proinflammatory IL-6 responses are also observed in HuNoV-infected volunteers (Lindesmith et al., 2003Ramani et al., 2015).

Animal models of disease are important for the development of therapeutics and Gn pigs have been evaluated as test models for both vaccines and antivirals. Similar to vaccine trials in humans (Atmar et al., 2011Bernstein et al., 2015Treanor et al., 2014), HuNoV VLPs are immunogenic in Gn pigs and induce homologous protection (Souza et al., 2007b). In an additional study, Gn pigs were vaccinated with VLPs and P particles (globular structures formed by multimerization of 12–36 copies of the P domain (Bereszczak et al., 2012)) and challenged with a closely related GII HuNoV. Primary NoV infection protected 83% of challenged animals against diarrhea and 57% against virus shedding. In contrast, VLP or P particles provided protection against diarrhea in 60% or 47%, respectively, and against virus shedding in 0% or 11%, respectively (Kocher et al., 2014). These studies suggest that a replicating vaccine (eg, attenuated virus) should be pursued as an alternative vaccination approach as it will likely confer superior protection to the recombinant subunit vaccine of VLPs or P particles.

The use of Gn pigs as an antiviral efficacy test model for HuNoV infection is only just beginning but will increase as more compounds move through the drug development pipeline (Arias et al., 2013Rocha-Pereira et al., 2014). One study (Jung et al., 2012) looked at interferon (IFN) α an important innate immune mediator with proven antiviral activity in humans (Feld and Hoofnagle, 2005). Treatment of Gn pigs with IFNα decreased viral shedding with a delayed onset and shorter duration of shedding, but viral shedding increased after treatment was discontinued (Jung et al., 2012). The inability of type I IFNs (IFNα/β) to clear NoV infection has also been seen in mice (Nice et al., 2014). Taking into account the known side effects of IFNα treatment in humans (Gota and Calabrese, 2003), IFNα will unlikely become a widely used treatment option for HuNoV infection in humans, and more specific anti-NoV drugs will need to be developed. Another area of future development will be studies regarding the utility of Gn pigs as a transmission model. Although the HS66 strain was serially passaged three times through Gn pigs with evidence of virus shedding at each passage and evidence of HuNoV-infected cells, viral titers and duration of fecal shedding decreased with each passage and no evidence of mutations was observed in the region analyzed (ie, ORF1/2 junction) (Cheetham et al., 2006). Thus, whether adaptive mutations occur in the HuNoV genome during infection in pigs remains to be addressed.

Additional Gn pig studies demonstrated that susceptibility to HuNoV infection changes with age, with lower median infectious dose (ID50) in neonates (4–5 days old) compared to older (33–34 days old) pigs (Bui et al., 2013). Whether susceptibility similarly changes with age in humans remains unknown. However, Gn pig studies (Bui et al., 2013Jung et al., 2012) did confirm earlier observations in humans that the use of statins (cholesterol lowering drugs) exacerbates HuNoV disease severity (Rondy et al., 2011). Simvastatin use in Gn pigs increased the incidence of diarrhea, and led to earlier onset and longer virus shedding (Bui et al., 2013Jung et al., 2012). Treated animals showed impaired Toll-like receptor (TLR) 3-dependent innate immune responses and lowered IFNα expression by alveolar macrophages and intestinal dendritic cells (Jung et al., 2012), but future studies are needed to determine whether similar mechanisms operate during NoV infection of humans.

In addition to Gn pigs, Gn calves were also investigated as an alternative large animal model (Souza et al., 2008), although to a limited extent. HuNoV infection of Gn calves with the GII.4 HS66 strain resulted in intestinal lesions (villous atrophy and loss of epithelial cells) in the small intestine, diarrhea and virus shedding in all analyzed animals. One of five calves also developed viremia. Viral capsid antigen was detected in small intestinal enterocytes and in lamina propria macrophage-like cells. HuNoV infection induced low levels of antibodies with only 67% (2/3) seroconversion but 100% (3/3) coproconversion (ie, antibodies in feces) in calves. Furthermore, early proinflammatory TNFα responses during diarrhea, high antiinflammatory IL-10 responses, and low to moderate Th1 (IL-12, IFNγ) and Th2 (IL-4) responses were observed. While more pronounced intestinal lesions were induced in Gn calves compared to Gn pigs, coinciding with increased numbers of IgA and IgG antibody secreting cells, most features of HuNoV infection in Gn calves are similar to those in Gn pigs.

In summary, both species of Gn animals are suitable models for the study of disease mechanisms, and thus represent important large animal models for preclinical, IND (investigational new drug)-enabling studies. However, the high cost of maintenance and required specialized care limit the broader use of this model in the scientific community. In addition, the lack of commensal bacteria in Gn animals, which promote development of the immune system (Sommer and Backhed, 2013) and of enteric viral infections (Kuss et al., 2011Uchiyama et al., 2014Kane et al., 2011), including NoV (Jones et al., 2014Miura et al., 2013), represent caveats for studies of viral pathogenesis and immune responses. However, recent studies of Gn animals colonized with human commensal bacteria (Kandasamy et al., 2014Vlasova et al., 2013) promise to overcome this limitation in future studies of HuNoV infection.

3. Human norovirus surrogates as animal models


The alternative approach to studying human viruses in a nonnative animal host is the study of surrogate viruses in their native hosts. The most widely used surrogate viruses for modeling HuNoV infection in vivo were PoSaV and are now MNV and recoviruses. All these viruses share the ability to readily grow in cell culture while sharing varying degrees of genetic relatedness and biological features with HuNoVs. Although no surrogate animal model recapitulates all aspects of HuNoV infection, each model has contributed important knowledge to our understanding of calicivirus biology as detailed later.

3.1. The Porcine Sapovirus Model


Sapoviruses (SaVs) are gastrointestinal pathogens transmitted by the fecal–oral route (Oka et al., 2015). Like HuNoVs, human SaVs (HuSaVs) are enteric viruses that cause outbreaks and sporadic cases of acute gastroenteritis in people of all ages around the globe (Oka et al., 2015). The clinical features of HuSaV disease are indistinguishable from HuNoV infections and laboratory diagnosis is required for accurate identification of these viruses (Oka et al., 2015). Unlike HuNoVs, HuSaVs do not bind HBGAs (Shirato-Horikoshi et al., 2007) but other cell attachment factors like sialic acid might be shared (Kim et al., 2014Rydell et al., 2009). No host genetic factor(s) of susceptibility has been identified for HuSaVs to date (Oka et al., 2015). PoSaVs are genetically closely related to HuSaVs (Scheuer et al., 2013), and the PoSaV Cowden strain is currently the only member of the Sapovirus genus with a tissue culture system that requires the addition of intestinal contents or bile acids during infection (Flynn and Saif, 1988Chang et al., 2004). This combined with the ability to genetically manipulate the viral genome (Chang et al., 2005) and development of acute gastroenteritis in PoSaV-infected pigs (Guo et al., 2001Flynn et al., 1988) led to the use of PoSaV as a surrogate for HuNoV and HuSaV infections.

Gnotobiotic pigs orally infected with the PoSaV prototype strain Cowden develop mild to severe diarrhea, shed virus in the feces, and some develop viremia (Guo et al., 2001). Histological analysis of small intestinal segments from PoSaV-infected pigs showed villous atrophy, villous shortening and blunting, villous fusion, crypt hyperplasia, and cytosolic vacuolization of enterocytes. PoSaV antigen detection indicates that the major site of PoSaV replication is in villous enterocytes in the proximal small intestine. Therefore, the clinical and pathological features of PoSaV infection resemble those found in HuSaV (Oka et al., 2015) and HuNoV (Green, 2013) infections in humans. However, whether the cellular tropism of PoSaV mirrors the in vivo tropism of HuNoVs and HuSaVs remains to be determined. Importantly, this model demonstrated that limited amino acid changes in a virus strain following serial passaging in cell culture resulted in an attenuated PoSaV phenotype in vivo (Guo et al., 2001). Given the large genetic variability of HuNoVs and HuSaVs, this suggests a wide range of phenotypes for these human viruses.

In summary, the PoSaV experimental infection model using Gn pigs is a valuable tool that has and will undoubtedly contribute to a greater understanding of SaV pathogenesis and disease. Future studies using the PoSaV model also have the potential to advance translational studies towards vaccine and antiviral development for HuSaVs. However, the use of PoSaV as a surrogate for HuNoV is less ideal given the availability of MNV and TV, which are more closely related to HuNoV and for which the molecular tools and infection or disease models are available that are needed to perform detailed mechanistic and translational studies.

3.2. The Mouse Norovirus Model


Murine norovirus (MNV) is a natural pathogen of wild rodents and laboratory mice (Henderson, 2008Farkas et al., 2012Tsunesumi et al., 2012Smith et al., 2012). To this date, MNV is the only NoV that has a small animal model and can be efficiently cultivated in tissue culture (Wobus et al., 2006). Since its discovery in 2003 (Karst et al., 2003), the MNV model has enabled significant advances in the understanding of norovirus-host interactions on the molecular, cellular, and organismal levels and has provided valuable perspectives on host–pathogen interactions in general (reviewed, eg, in Karst et al., 2014Wobus et al., 2006). The utility of MNV as a model system for other NoVs is based on the following main features: (1) MNV can readily be cultivated in primary cells and stable cell lines derived from its natural host (Wobus et al., 2004), enabling a thorough analysis of the entire virus life cycle from attachment to release at the cellular and molecular levels. (2) The genetic tractability of mice has allowed researchers to identify diverse host factors that are important for survival and clearance of MNV, providing insights into host defenses that are likely to be important for protection during HuNoV infection (reviewed in Karst et al., 2014 2015Karst, 2010). (3) The availability of several reverse-genetics systems (Ward et al., 2007Yunus et al., 2010Chaudhry et al., 2007Sandoval-Jaime et al., 2015) permits a detailed understanding of viral determinants of NoV infections, including receptor utilization, tissue tropism and viral persistence (eg, Taube et al., 2012Nice et al., 2013Strong et al., 2012). While MNV does not recapitulate clinical symptoms of HuNoV infection in immunocompetent hosts, it provides novel perspectives toward understanding interactions between intestinal viruses and their natural, coevolved host (Jones et al., 2014Cadwell, 2015Kernbauer et al., 2014Cadwell et al., 2010).

Sequence analysis of the MNV genome shows that these viruses are homologous to other viruses in the NoV genus, including HuNoV, but form a separate genogroup (GV) (Karst et al., 2003Smith et al., 2012). Three of MNV’s four open reading frames (ORFs) encoding the nonstructural proteins (ORF1), major and minor capsid proteins (ORF2 and ORF3, respectively) are shared with all other NoVs, while an additional fourth ORF is only present within GV (McFadden et al., 2011Thackray et al., 2007). Studies of NoV proteins have suggested that products of the ORF1 polyprotein of MNV and HuNoV have common functional properties, such as the rearrangement of cellular membranes during virus replication or during translation initiation (reviewed in Thorne and Goodfellow, 2014). In addition, MNV shares with HuNoVs the use of carbohydrates for cell attachment, although the specific carbohydrates only partially overlap (Tan and Jiang, 2014Taube et al., 2009). Many strains of HuNoV bind to HBGA (reviewed in Tan and Jiang, 2014) but some HuNoV strains also bind sialic acids (Rydell et al., 2009Maalouf et al., 2010Esseili et al., 2012). MNV binds to terminal sialic acids on gangliosides and N- or O-linked glycoproteins in a strain-dependent manner (Taube et al., 2009 2012), although no strains of MNV are known to date that use HBGA (Taube et al., 2010). Thus, while the MNV model does not allow studies into the function of HBGA during NoV pathogenesis, it does provide a system to understand the role of carbohydrates during NoV infection in general.

MNVs and HuNoVs also share the physical properties of the capsid and route of transmission. Like HuNoV, MNV virions are highly stable in the environment and are relatively resistant to heat, dessication, and low pH (Cannon et al., 2006Arthur and Gibson, 2015Kotwal and Cannon, 2014Tuladhar et al., 2012). In addition, viral particles are also highly infectious. The estimated 50% HuNoV infectious dose is between 18 and 2800 particles depending on the study (Teunis et al., 2008Atmar et al., 2014), and MNV infection can be initiated with as few as 10 particles (Liu et al., 2009). These similarities are consistent with their common fecal–oral route of transmission making MNV a widely used surrogate for food safety studies (reviewed in Kniel, 2014).

Given the shared route of transmission, viral shedding in the stool is characteristic for all NoVs. While the infectious potential of shed HuNoV virions remains unknown, genomes are detected in the stool for weeks to months in immunocompetent individuals, long after symptomatic disease has resolved, and for years in immunocompromised individuals (Kaufman et al., 2014Teunis et al., 2014Green, 2014). Infectious MNV is shed for months as most strains cause a persistent infection in wild-type mice (Thackray et al., 2007Hsu et al., 2006). In addition, the MNV-1 strain, which causes a self-resolving, acute infection in wild-type mice with transient shedding, causes long-term shedding in mice lacking components of the adaptive immune system (ie, Rag-deficient mice) (Karst et al., 2003). Thus, different durations of viral shedding can be modeled depending on the combinations of MNV and mouse strains. Multiple factors influencing shedding have been identified; viral gene products (ie, NS1/2 [N-terminal] and VP1 [viral capsid]) (Nice et al., 2013Strong et al., 2012), host factors (eg, adaptive immune responses, interferon λ) (Karst et al., 2003Nice et al., 2014), and the microbiota (Baldridge et al., 2015). While transmission occurs readily between mice in a cage (Manuel et al., 2008) (Fig. 3.4.4), the parameters that influence transmission remain largely unexplored. Nevertheless, a recent study successfully demonstrated the ability of an antiviral compound to disrupt transmission of MNV (Rocha-Pereira et al., 2015), highlighting the utility of the MNV system as a transmission model.

image

Figure 3.4.4 Transmission of MNV between mice in a cage.
One Balb/c mouse each was orally infected with 1 × 106 pfu of MNV.CR3, the same dose of UV-inactivated MNV.CR3, or mock-infected. Two days later, each infected mouse was cohoused with 2 naïve mice for 28 days. Serum was collected from all animals on days 0, 7, 14, 21, and 28 of cohousing. ELISA was performed on a 1:100 dilution of serum as described (Wobus et al., 2004) and absorbance values were graphed. Naïve mice housed with a MNV.CR3-inoculated, but not mock or UV inactivated virus-inoculated cage mate seroconverted, indicating viral infection.

Despite many similarities, important differences exist between MNVs and HuNoVs. One of the greatest differences is that MNV does not cause overt clinical disease in an immunocompetent host. HuNoV infections frequently result in vomiting and diarrhea but mice are unable to vomit and MNV-infected wild-type mice do not develop overt diarrhea. Nevertheless, MNV-infected mice show histopathologic changes in the intestine (mild inflammation, limited villous atrophy) similar to those observed in HuNoV-infected volunteers (Karst et al., 2015Mumphrey et al., 2007). It is also important to note that MNV causes diarrhea in mouse strains with impaired innate immunity, including strains lacking signal transducer and activator of transcription (Stat) 1 or the type I and II interferon receptors (Karst et al., 2003Mumphrey et al., 2007Rocha-Pereira et al., 2013). While the immunodeficient status of the host limits the use of those strains for studies of disease mechanisms, it does provide a small animal disease model for antiviral efficacy testing to develop compounds that not only reduce viral loads but also disease scores, as one recent study elegantly demonstrated (Rocha-Pereira et al., 2013).

Differences exist also at the level of genetic diversity. HuNoV strains identified to date exhibit a broader genetic diversity (up to 45% divergence at the nucleotide level), spanning 3 genogroups (GI, GII, and GIV) with at least 29 genotypes (Zheng et al., 2006) than known MNV strains. MNVs identified to date separate into two genotypes with ∼23% nucleotide divergence (Smith et al., 2012). However, a recently identified rat NoV that clusters with MNVs in GV diverges at the nucleotide level by 31% (Tsunesumi et al., 2012). Thus, it is likely that the diversity of rodent NoVs will increase in the future as more strains are identified. While the broad genetic diversity of HuNoV requires specific considerations during vaccine development, vaccination studies of HuNoV VLPs in mice demonstrated the development of heterologous, cross-genogroup immunity that reduces MNV viral loads upon challenge (LoBue et al., 2009). Thus, the MNV model can also be used as one tool for the development of vaccines with cross-genogroup protective efficacy.

Many features of HuNoV infection and replication remain poorly understood, but studies with MNV may offer clues to these processes. One open question in the field is the cell tropism of NoV. The identity of HuNoV-infected cell types in vivo remains unknown. However, viral antigen from both MNV and HuNoV has been detected within the intestinal lamina propria of infected mice and humans, respectively (Lay et al., 2010Mumphrey et al., 2007), suggesting a tropism for immune cells (Karst and Wobus, 2015). This is consistent with the cell types that support HuNoV and MNV replication in vitro, specifically B cells for HuNoV (Jones et al., 2014) and macrophages, dendritic cells, microglia, and B cells (Jones et al., 2014Wobus et al., 2004Cox et al., 2009) for MNV. Thus, to date a partial overlap in cellular tropism exists between HuNoV and MNV. Future studies are needed to determine whether human and murine NoVs share the tropism for all these different immune cells. Another open question is how the broad genetic diversity of HuNoV influences viral phenotypes. Genetic analysis of MNV strains has already revealed viral determinants of NoV biology, including virulence and glycan utilization (Karst et al., 2014Taube et al., 2012Nice et al., 2013Strong et al., 2012). Similarly, genetic variability in HuNoV may eventually be linked to specific phenotypes. Furthermore, studies with MNV have illuminated many aspects of NoV biology, for example, identifying roles for microfold (M) cells in intestinal entry into the host (Gonzalez-Hernandez et al., 2014), for dendritic cells in dissemination (Elftman et al., 2013), and for components of the innate immune system in controlling infections (McCartney et al., 2008Thackray et al., 2012Changotra et al., 2009). Future studies will need to determine the translatability of these findings to HuNoV infections.

One of the most promising aspects of the MNV model is the opportunity to study an enteric pathogen in its natural host and elucidate how the host’s genetic background and microbial environment influence an encounter with a pathogen. For example, studies using MNV demonstrate that the interaction between enteric viruses, the microbiome, and the host’s genetic background can influence the development of complex conditions such as inflammatory bowel disease (Cadwell et al., 2010). More recent studies have shown how concurrent helminth infections create a type 2 cytokine environment resulting in poor control of NoV replication (Osborne et al., 2014), and that the microbiome as a whole or specific members promote MNV infection and persistence (Jones et al., 2014Baldridge et al., 2015), at least in the latter case via modulating the host immune response.

Overall, MNV provides a unique opportunity to mechanistically dissect, not only the effects of host factors and environment on a naturally coevolved host–pathogen relationship, but also many aspects of NoV biology. Despite the limitations of MNV as a model for clinical disease (ie, need for an immunodeficient host), MNV infection of wild-type mice represents a genetically tractable infection and transmission model.

3.3. The Recovirus Model


Rhesus enteric caliciviruses represent the most recently established HuNoV model. Recoviruses were first described in 2008, after the detection and isolation of a novel calicivirus from fecal samples of rhesus macaques (Farkas et al., 2008). These viruses are phylogenetically distinct from the genus Norovirus and from the proposed genus Recovirus. However, they share many properties with HuNoV, including their genomic organization, large genetic diversity, the ability to bind to HBGA, transmission by the fecal–oral route, and clinical disease presentations. Recovirus genetic diversity encompasses three genogroups and five genotypes (Farkas, 2015). GI and GII strains are found in rhesus macaques (Farkas et al., 2014), while GIII strains were discovered in samples collected from six humans with gastroenteritis in Bangladesh (Smits et al., 2012). The prototypic Tulane virus (TV), a GI.1 strain, replicates efficiently in cell culture in the rhesus macaque kidney epithelial cell line LLC-MK2 (Farkas et al., 2008), has a reverse genetics system available (Wei et al., 2008), and is associated with diarrheal disease in rhesus macaques after experimental infection (Sestak et al., 2012), providing crucial tools for mechanistic studies.

One of the most important advantages of the recovirus model is that the clinical disease presentation, histopathology, and transient serological response to TV are reminiscent of volunteer studies of HuNoV infection (Sestak et al., 2012). The similarities are multi-fold. First, intragastric infection of juvenile macaques with TV causes fever, a semiliquid diarrhea, and viral shedding in feces, although neither vomiting nor viremia were observed (Sestak et al., 2012). Second, intestinal biopsies from the duodenum of these monkeys revealed slightly blunted villi and mild mononuclear infiltration of the lamina propria. Third, experimentally infected macaques developed a robust but transient neutralizing antibody response against TV. However, future studies are needed to determine the ability of these antibodies to protect from infection or disease. Fourth, TV has a tropism for B cells. TV antigen-positive B cells were observed in the intestinal lamina propria, and infection of peripheral blood mononuclear cells obtained from healthy macaques led to an increase in viral RNA and antigen expression within B cells (Sestak et al., 2012). This tropism is similar to at least one HuNoV GII.4 strain (Jones et al., 2014).

Another important similarity of HuNoV and recoviruses is their ability to bind HBGAs. Diverse HuNoV strains have different patterns of HBGA-utilization and dependency (Huang et al., 2005) (see Chapter 3.3). Three HBGA binding patterns have been identified to date for recoviruses (Farkas, 2015). In case of TV, binding occurs with the type 3 form of the A antigen and all forms of the B antigen (Zhang et al., 2015). HBGAs are a known susceptibility factor for HuNoV infection (reviewed in Tan and Jiang, 2014) but the primate colonies used for recovirus studies exhibit a fairly homogeneous HBGA phenotype with greater than 95% of monkeys producing B antigen (Farkas et al., 2010). Thus, future experiments in a more phenotypically diverse population of macaques are needed to investigate whether specific HBGAs determine susceptibility of macaques to recovirus infection. Multiple roles have been proposed for HBGAs and its different forms (ie, free, bacterially associated, cell associated) during NoV infection (Karst and Wobus, 2015). Thus, the ability of recoviruses to bind HBGA and cause disease in a conventionally housed (ie, bacterially colonized) immunocompetent host provides a unique opportunity to understand the role of HBGA during enteric calicivirus infection and disease in the future.

The relationship between genetic and serologic diversity of HuNoVs remains unknown. Study of recoviruses with a similarly broad genetic diversity and available cell culture system has yielded initial insights; namely that genotypic classifications correlate generally with serotypes, although exceptions exist (Farkas, 2015Farkas et al., 2014). These data suggest that the great HuNoV genetic diversity is likely accompanied by broad serological diversity. While this may present challenges in vaccine design, the recently described HuNoV culture system (Jones et al., 2014) will now enable future serotyping studies.

In summary, the recovirus model combines many beneficial features of an ideal HuNoV model. Most importantly, recoviruses can be genetically manipulated in vitro (Wei et al., 2008), replicate efficiently in tissue culture (Farkas et al., 2008), use the same HBGA attachment receptors as HuNoV (Sestak, 2014), and cause similar disease in vivo (Sestak et al., 2012). While the financial cost of in vivo studies and the inability to genetically manipulate the host are drawbacks of this model, the limitations are outweighed by many benefits, foremost the availability of an immunologically and microbially competent disease model. This model might be particularly useful in elucidating various roles of HBGAs in calicivirus pathogenesis, including how changing HBGA binding affinities and escape from neutralizing immune responses influence individual and herd immunity, an important driver of HuNoV evolution (Debbink et al., 2013).

4. Conclusions


Animal models are an integral part of our studies of human viruses. Large animal models are particularly valuable for translational studies and preclinical efficacy testing of antivirals and vaccines, while small animal models, especially knock-out and transgenic mouse models, are invaluable to gain a greater mechanistic understanding of all aspects of virus—host interactions at the organismal and cellular levels.

Although the past two decades have seen much progress in our understanding of HuNoVs, our understanding of these viruses lags behind that of other human viruses. The study of HuNoV has been particularly difficult due to the long-term lack of a cell culture system (Duizer et al., 2004Herbst-Kralovetz et al., 2013Papafragkou et al., 2013Takanashi et al., 2014) and thus independent validation of the recently described B-cell culture model (Jones et al., 2014) is critical. The field also struggles to identify an animal model that closely reproduces HuNoV disease and is widely accepted. No “perfect” model has been developed to date but multiple different animal models for the study of HuNoV are available either by overcoming the species barrier of HuNoV or using HuNoV surrogates. Each of these models has its strengths and limitations (Tables 3.4.1 and 3.4.2) and overcoming some of these limitations remains an important goal for future research. One potentially fruitful avenue could be the generation of host-adapated HuNoVs, which has not been reported for any HuNoV animal model. Improvements might also come from the study of little explored (eg, newborn pigtail macaques) or new animal hosts, the identification of new surrogate viruses, or discovery of additional strains of HuNoV that more readily adapt or transmit to other species. While data obtained from any of the existing or future models are critical for our improved understanding of HuNoVs, validation of findings in the human host will ultimately determine how appropriate each model is to study a given aspect of HuNoV biology.


Table 3.4.2


Summary of the Discussed HuNoV Surrogates With Their Strengths and Limitations






























Host Virus Strengths Limitations
Wild-type mouse (Wobus et al., 2006; Chang et al., 2005; Farkas et al., 2012) MNV (all strains)

Natural host


Fecal shedding


Orally susceptible


Genetically tractable host


Relatively low cost


Similar histopathology to humans (villous blunting, mild intestinal inflammation)


Available cell culture system


Available reverse genetics system


Asymptomatic infection


Does not bind HBGAs


Relatively limited genetic diversity of MNV strains

STAT1-/-AG129 mouse (Karst et al., 2003; Hsu et al., 2006) MNV (eg, MNV-1, MNV-3, CR3)

Orally susceptible


Symptomatic infection


Fecal shedding


Relatively low cost


Available cell culture system


Available reverse genetics system


Immune deficiency


Systemic infection


Exacerbated histopathology


Does not bind HBGAs


Relatively limited genetic diversity

Rhesus macaques (Sestak et al., 2012; Farkas, 2015)

Recoviruses/


Tulane virus


Natural host


Orally susceptible


Symptomatic infection


Virus binds HBGAs


Large genetic diversity


Genomic organization similar to HuNoVs


Fecal shedding


Similar histopathology to humans (villous blunting, mild intestinal inflammation)


Transient neutralizing antibody response like humans


Available cell culture system


Available reverse genetics system


Not a norovirus


High cost

Gnotobiotic pigs (Rydell et al., 2009; Scheuer et al., 2013) PoSaV

Natural host


Orally susceptible


Symptomatic infection


Fecal shedding


Villous atrophy and blunting


Available cell culture system


Available reverse genetics system


Not a norovirus


High cost


Lack of microbiota


Underdeveloped immune system


Does not bind HBGAs




Acknowledgments


We apologize to all colleagues whose work could not be cited due to lengths restrictions. C.E.W. is a recipient of the Friedrich Wilhelm Bessel award from the Alexander von Humboldt Foundation. Work in her laboratory was supported by NIH grants R21/R33 AI102106, R01 AI080611, R21 AI103961, and DARPA grant HR0011-11-C-0093. J.B.C. was funded by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Brasília, Brazil.


References


Arias A, Emmott E, Vashist S, Goodfellow I. Progress towards the prevention and treatment of norovirus infections. Future Microbiol. 2013;8:14751487.


Arthur SE, Gibson KE. Comparison of methods for evaluating the thermal stability of human enteric viruses. Food Environ. Virol. 2015;7(1):1426.


Atmar RL, Opekun AR, Gilger MA, et al. Norwalk virus shedding after experimental human infection. Emerg. Infect. Dis. 2008;14(10):15531557.


Atmar RL, Bernstein DI, Harro CD, et al. Norovirus vaccine against experimental human Norwalk Virus illness. N. Engl. J. Med. 2011;365(23):21782187.


Atmar RL, Opekun AR, Gilger MA, et al. Determination of the 50% human infectious dose for Norwalk virus. J. Infect. Dis. 2014;209(7):10161022.


Baldridge MT, Nice TJ, McCune BT, et al. Commensal microbes and interferon-lambda determine persistence of enteric murine norovirus infection. Science. 2015;347(6219):266269.


Bereszczak JZ, Barbu IM, Tan M, et al. Structure, stability and dynamics of norovirus P domain derived protein complexes studied by native mass spectrometry. J. Struct. Biol. 2012;177(2):273282.


Bernstein DI, Atmar RL, Lyon GM, et al. Norovirus vaccine against experimental human GII.4 virus illness: a challenge study in healthy adults. J. Infect. Dis. 2015;211(6):870878.


Bitler EJ, Matthews JE, Dickey BW, Eisenberg JN, Leon JS. Norovirus outbreaks: a systematic review of commonly implicated transmission routes and vehicles. Epidemiol. Infect. 2013;141(8):15631571.


Bok K, Parra GI, Mitra T, et al. Chimpanzees as an animal model for human norovirus infection and vaccine development. Proc. Natl. Acad. Sci. USA. 2011;108(1):325330.


Bui T, Kocher J, Li Y, et al. Median infectious dose of human norovirus GII.4 in gnotobiotic pigs is decreased by simvastatin treatment and increased by age. J. Gen. Virol. 2013;94(Pt 9):20052016.


Cadwell K. Expanding the Role of the Virome: Commensalism in the Gut. J. Virol. 2015;89(4):19511953.


Cadwell K, Patel KK, Maloney NS, et al. Virus-plus-susceptibility gene interaction determines Crohn’s disease gene Atg16L1 phenotypes in intestine. Cell. 2010;141(7):11351145.


Cannon JL, Papafragkou E, Park GW, Osborne J, Jaykus LA, Vinje J. Surrogates for the study of norovirus stability and inactivation in the environment: A comparison of murine norovirus and feline calicivirus. J. Food Protect. 2006;69(11):27612765.


Chachu KA, Strong DW, LoBue AD, Wobus CE, Baric RS, Virgin IV HW. Antibody is critical for the clearance of murine norovirus infection. J. Virol. 2008;82(13):66106617.


Chang KO, Sosnovtsev SV, Belliot G, Kim Y, Saif LJ, Green KY. Bile acids are essential for porcine enteric calicivirus replication in association with down-regulation of signal transducer and activator of transcription 1. Proc. Natl. Acad. Sci. USA. 2004;101(23):87338738.


Chang KO, Sosnovtsev SV, Belliot G, Wang Q, Saif LJ, Green KY. Reverse genetics system for porcine enteric calicivirus, a prototype sapovirus in the Caliciviridae. J. Virol. 2005;79(3):14091416.


Changotra H, Jia Y, Moore TN, et al. Type I and type II interferon inhibit the translation of murine norovirus proteins. J. Virol. 2009;83(11):56835692.


Chaudhry Y, Skinner MA, Goodfellow IG. Recovery of genetically defined murine norovirus in tissue culture by using a fowlpox virus expressing T7 RNA polymerase. J. Gen. Virol. 2007;88(Pt 8):20912100.


Cheetham S, Souza M, Meulia T, Grimes S, Han MG, Saif LJ. Pathogenesis of a genogroup II human norovirus in gnotobiotic pigs. J. Virol. 2006;80(21):1037210381.


Cheetham S, Souza M, McGregor R, Meulia T, Wang Q, Saif LJ. Binding patterns of human norovirus-like particles to buccal and intestinal tissues of gnotobiotic pigs in relation to A/H histo-blood group antigen expression. J. Virol. 2007;81(7):35353544.


Cox C, Cao S, Lu Y. Enhanced detection and study of murine norovirus-1 using a more efficient microglial cell line. Virol. J. 2009;6:196.


Debbink K, Lindesmith LC, Donaldson EF, et al. Emergence of new pandemic GII.4 Sydney Norovirus strain correlates with escape from herd immunity. J. Infect. Dis. 2013;208(11):18771887.


Duizer E, Schwab KJ, Neill FH, Atmar RL, Koopmans MP, Estes MK. Laboratory efforts to cultivate noroviruses. J. Gen. Virol. 2004;85(Pt 1):7987.


Elftman MD, Gonzalez-Hernandez MB, Kamada N, et al. Multiple effects of dendritic cell depletion on murine norovirus infection. J. Gen. Virol. 2013;94(Pt 8):17611768.


Esseili MA, Wang Q, Saif LJ. Binding of human GII.4 norovirus virus-like particles to carbohydrates of romaine lettuce leaf cell wall materials. Appl. Environ. Microbiol. 2012;78(3):786794.


Farkas T. Rhesus enteric calicivirus surrogate model for human norovirus gastroenteritis. J. Gen. Virol. 2015;96(7):15041514.


Farkas T, Sestak K, Wei C, Jiang X. Characterization of a rhesus monkey calicivirus representing a new genus of Caliciviridae. J. Virol. 2008;82(11):54085416.


Farkas T, Cross RW, Hargitt 3rd E, Lerche NW, Morrow AL, Sestak K. Genetic diversity and histo-blood group antigen interactions of rhesus enteric caliciviruses. J. Virol. 2010;84(17):86178625.


Farkas T, Fey B, Keller G, Martella V, Egyed L. Molecular detection of murine noroviruses in laboratory and wild mice. Vet. Microbiol. 2012;160(3–4):463467.


Farkas T, Lun CW, Fey B. Relationship between genotypes and serotypes of genogroup 1 recoviruses: a model for human norovirus antigenic diversity. J. Gen. Virol. 2014;95(Pt 7):14691478.


Feld JJ, Hoofnagle JH. Mechanism of action of interferon and ribavirin in treatment of hepatitis C. Nature. 2005;436(7053):967972.


Flynn WT, Saif LJ. Serial propagation of porcine enteric calicivirus-like virus in primary porcine kidney cell cultures. J. Clin. Microbiol. 1988;26(2):206212.


Flynn WT, Saif LJ, Moorhead PD. Pathogenesis of porcine enteric calicivirus-like virus in four-day-old gnotobiotic pigs. Am. J. Vet. Res. 1988;49(6):819825.


Gonzalez-Hernandez MB, Liu T, Blanco LP, Auble H, Payne HC, Wobus CE. Murine norovirus transcytosis across an in vitro polarized murine intestinal epithelial monolayer is mediated by M-like cells. J. Virol. 2013;87(23):1268512693.


Gonzalez-Hernandez MB, Liu T, Payne HC, et al. Efficient Norovirus and Reovirus Replication in the Mouse Intestine Requires Microfold (M) Cells. J. Virol. 2014;88(12):69346943.


Gota C, Calabrese L. Induction of clinical autoimmune disease by therapeutic interferon-alpha. Autoimmunity. 2003;36(8):511518.


Graham DY, Jiang X, Tanaka T, Opekun AR, Madore HP, Estes MK. Norwalk virus infection of volunteers: new insights based on improved assays. J. Infect. Dis. 1994;170(1):3443.


Green KY. Caliciviridae: the noroviruses. In: Knipe DM, Howley PM, Cohen JI, Griffin DI, Lamb RA, Martin MA, Racaniello VR, Roizman B, eds. Fields Virology. Philadelphia: Lippincott Williams & Wilkins, a Wolters Kluwer Business; 2013:582608.


Green KY. Norovirus infection in immunocompromised hosts. Clin. Microbiol. Infect. 2014;20(8):717723.


Guo M, Hayes J, Cho KO, Parwani AV, Lucas LM, Saif LJ. Comparative pathogenesis of tissue culture-adapted and wild-type Cowden porcine enteric calicivirus (PEC) in gnotobiotic pigs and induction of diarrhea by intravenous inoculation of wild-type PEC. J. Virol. 2001;75(19):92399251.


Henderson KS. Murine norovirus, a recently discovered and highly prevalent viral agent of mice. Lab. Animal. 2008;37(7):314320.


Herbst-Kralovetz MM, Radtke AL, Lay MK, et al. Lack of norovirus replication and histo-blood group antigen expression in 3-dimensional intestinal epithelial cells. Emerg. Infect. Dis. 2013;19(3):431438.


Hsu CC, Riley LK, Wills HM, Livingston RS. Persistent infection with and serologic cross-reactivity of three novel murine noroviruses. Comp. Med. 2006;56(4):247251.


Huang P, Farkas T, Zhong W, et al. Norovirus and histo-blood group antigens: demonstration of a wide spectrum of strain specificities and classification of two major binding groups among multiple binding patterns. J. Virol. 2005;79(11):67146722.


Hutson AM, Atmar RL, Graham DY, Estes MK. Norwalk virus infection and disease is associated with ABO histo-blood group type. J. Infect. Dis. 2002;185(9):13351337.


Hutson AM, Airaud F, LePendu J, Estes MK, Atmar RL. Norwalk virus infection associates with secretor status genotyped from sera. J. Med. Virol. 2005;77(1):116120.


Jones MK, Watanabe M, Zhu S, et al. Enteric bacteria promote human and mouse norovirus infection of B cells. Science. 2014;346(6210):755759.


Jung K, Wang Q, Kim Y, et al. The effects of simvastatin or interferon-alpha on infectivity of human norovirus using a gnotobiotic pig model for the study of antivirals. PLoS One. 2012;7(7):e41619.


Kandasamy S, Chattha KS, Vlasova AN, Rajashekara G, Saif LJ. Lactobacilli and Bifidobacteria enhance mucosal B cell responses and differentially modulate systemic antibody responses to an oral human rotavirus vaccine in a neonatal gnotobiotic pig disease model. Gut Microbes. 2014;5(5):639651.


Kane M, Case LK, Kopaskie K, et al. Successful transmission of a retrovirus depends on the commensal microbiota. Science. 2011;334(6053):245249.


Karst SM. Pathogenesis of noroviruses, Emerging RNA viruses. Viruses. 2010;2:748781.


Karst SM, Wobus CE. A working model of how noroviruses infect the intestine. PLoS Pathog. 2015;11(2):e1004626.


Karst SM, Wobus CE, Lay M, Davidson J, Virgin IV HW. STAT1-dependent innate immunity to a Norwalk-like virus. Science. 2003;299(5612):15751578.


Karst SM, Wobus CE, Goodfellow IG, Green KY, Virgin HW. Advances in norovirus biology. Cell Host Microbe. 2014;15(6):668680.


Karst SM, Zhu S, Goodfellow IG. The molecular pathology of noroviruses. J. Pathol. 2015;235(2):206216.


Kaufman SS, Green KY, Korba BE. Treatment of norovirus infections: moving antivirals from the bench to the bedside. Antiviral. Res. 2014;105:8091.


Kernbauer E, Ding Y, Cadwell K. An enteric virus can replace the beneficial function of commensal bacteria. Nature. 2014;516(7529):9498.


Kim DS, Hosmillo M, Alfajaro MM, et al. Both alpha2,3- and alpha2,6-Linked Sialic Acids on O-Linked Glycoproteins Act as Functional Receptors for Porcine Sapovirus. PLoS Pathog. 2014;10(6):e1004172.


Kniel KE. The makings of a good human norovirus surrogate. Curr. Opin. Virol. 2014;4:8590.


Kocher J, Bui T, Giri-Rachman E, et al. Intranasal P particle vaccine provided partial cross-variant protection against human GII.4 norovirus diarrhea in gnotobiotic pigs. J. Virol. 2014;88(17):97289743.


Kotwal G, Cannon JL. Environmental persistence and transfer of enteric viruses. Curr. Opin. Virol. 2014;4:3743.


Kuss SK, Best GT, Etheredge CA, et al. Intestinal microbiota promote enteric virus replication and systemic pathogenesis. Science. 2011;334(6053):249252.


Lay MK, Atmar RL, Guix S, et al. Norwalk virus does not replicate in human macrophages or dendritic cells derived from the peripheral blood of susceptible humans. Virology. 2010;406(1):111.


Lee RM, Lessler J, Lee RA, et al. Incubation periods of viral gastroenteritis: a systematic review. BMC Infect. Dis. 2013;13:446.


Lindesmith L, Moe C, Marionneau S, et al. Human susceptibility and resistance to Norwalk virus infection. Nature Med. 2003;9(5):548553.


Liu G, Kahan SM, Jia Y, Karst SM. Primary high-dose murine norovirus 1 infection fails to protect from secondary challenge with homologous virus. J. Virol. 2009;83(13):69636968.


LoBue AD, Thompson JM, Lindesmith L, Johnston RE, Baric RS. Alphavirus-adjuvanted norovirus-like particle vaccines: heterologous, humoral, and mucosal immune responses protect against murine norovirus challenge. J. Virol. 2009;83(7):32123227.


Lopman BA, Reacher MH, Vipond IB, Sarangi J, Brown DW. Clinical manifestation of norovirus gastroenteritis in health care settings. Clin. Infect. Dis. 2004;39(3):318324.


Maalouf H, Zakhour M, Le Pendu J, Le Saux JC, Atmar RL, Le Guyader FS. Distribution in tissue and seasonal variation of norovirus genogroup I and II ligands in oysters. Appl. Environ. Microbiol. 2010;76(16):56215630.


Manuel CA, Hsu CC, Riley LK, Livingston RS. Soiled-bedding sentinel detection of murine norovirus 4. J. Am. Assoc. Lab. Anim. Sci. 2008;47(3):3136.


McCartney SA, Thackray LB, Gitlin L, Gilfillan S, Virgin IV HW, Colonna M. MDA-5 recognition of a murine norovirus. PLoS Pathog. 2008;4(7):e1000108.


McFadden N, Bailey D, Carrara G, et al. Norovirus regulation of the innate immune response and apoptosis occurs via the product of the alternative open reading frame 4. PLoS Pathog. 2011;7(12):e1002413.


Miura T, Sano D, Suenaga A, et al. Histo-blood group antigen-like substances of human enteric bacteria as specific adsorbents for human noroviruses. J. Virol. 2013;87(17):94419451.


Mumphrey SM, Changotra H, Moore TN, et al. Murine norovirus 1 infection is associated with histopathological changes in immunocompetent hosts, but clinical disease is prevented by STAT1-dependent interferon responses. J. Virol. 2007;81(7):32513263.


Nice TJ, Strong DW, McCune BT, Pohl CS, Virgin HW. A single-amino-acid change in murine norovirus NS1/2 is sufficient for colonic tropism and persistence. J. Virol. 2013;87(1):327334.


Nice TJ, Baldridge MT, McCune BT, et al. Interferon lambda cures persistent murine norovirus infection in the absence of adaptive immunity. Science. 2014;347(6219):269273.


Oka T, Wang Q, Katayama K, Saif LJ. Comprehensive review of human sapoviruses. Clin. Microbiol. Rev. 2015;28(1):3253.


Osborne LC, Monticelli LA, Nice TJ, et al. Coinfection. Virus-helminth coinfection reveals a microbiota-independent mechanism of immunomodulation. Science. 2014;345(6196):578582.


Papafragkou E, Hewitt J, Park GW, Greening G, Vinje J. Challenges of culturing human norovirus in three-dimensional organoid intestinal cell culture models. PLoS One. 2013;8(6):e63485.


Radford AD, Gaskell RM, Hart CA. Human norovirus infection and the lessons from animal caliciviruses. Curr. Opin. Infect. Dis. 2004;17(5):471478.


Radford AD, Coyne KP, Dawson S, Porter CJ, Gaskell RM. Feline calicivirus. Vet. Res. 2007;38(2):319335.


Ramani S, Neill FH, Opekun AR, et al. Mucosal and cellular immune responses to Norwalk virus. J. Infect. Dis. 2015;212(3):397405.


Rocha-Pereira J, Jochmans D, Debing Y, Verbeken E, Nascimento MS, Neyts J. The viral polymerase Inhibitor 2’-C-methylcytidine inhibits Norwalk virus replication and protects against Norovirus-induced diarrhea and mortality in a mouse model. J. Virol. 2013;87(21):1179811805.


Rocha-Pereira J, Neyts J, Jochmans D. Norovirus: targets and tools in antiviral drug discovery. Biochem. Pharmacol. 2014;91(1):111.


Rocha-Pereira J, Jochmans D, Neyts J. Prophylactic treatment with the nucleoside analogue 2’-C-methylcytidine completely prevents transmission of norovirus. J. Antimicrob. Chemother. 2015;70(1):190197.


Rockx B, De Wit M, Vennema H, et al. Natural history of human calicivirus infection: a prospective cohort study. Clin. Infect. Dis. 2002;35(3):246253.


Rockx BH, Bogers WM, Heeney JL, van Amerongen G, Koopmans MP. Experimental norovirus infections in non-human primates. J. Med. Virol. 2005;75(2):313320.


Rondy M, Koopmans M, Rotsaert C, et al. Norovirus disease associated with excess mortality and use of statins: a retrospective cohort study of an outbreak following a pilgrimage to Lourdes. Epidemiol. Infect. 2011;139(3):453463.


Rydell GE, Nilsson J, Rodriguez-Diaz J, et al. Human noroviruses recognize sialyl Lewis x neoglycoprotein. Glycobiology. 2009;19(3):309320.


Saif LJ, Ward LA, Yuan L, Rosen BI, To TL. The gnotobiotic piglet as a model for studies of disease pathogenesis and immunity to human rotaviruses. Arch. Virol. Suppl. 1996;12:153161.


Sandoval-Jaime C, Green KY, Sosnovtsev SV. Recovery of murine norovirus and feline calicivirus from plasmids encoding EMCV IRES in stable cell lines expressing T7 polymerase. J. Virol. Methods. 2015;217:17.


Scheuer KA, Oka T, Hoet AE, et al. Prevalence of porcine noroviruses, molecular characterization of emerging porcine sapoviruses from finisher swine in the United States, and unified classification scheme for sapoviruses. J. Clin. Microbiol. 2013;51(7):23442353.


Sestak K. Role of histo-blood group antigens in primate enteric calicivirus infections. World J. Virol. 2014;3(3):1821.


Sestak K, Feely S, Fey B, et al. Experimental inoculation of juvenile rhesus macaques with primate enteric caliciviruses. PLoS One. 2012;7(5):e37973.


Shirato-Horikoshi H, Ogawa S, Wakita T, Takeda N, Hansman GS. Binding activity of norovirus and sapovirus to histo-blood group antigens. Arch. Virol. 2007;152(3):457461.


Smith DB, McFadden N, Blundell RJ, Meredith A, Simmonds P. Diversity of murine norovirus in wild-rodent populations: species-specific associations suggest an ancient divergence. J. Gen. Virol. 2012;93(Pt 2):259266.


Smits SL, Rahman M, Schapendonk CM, et al. Calicivirus from novel Recovirus genogroup in human diarrhea. Emerg. Infect. Dis. 2012;18(7):11921195.


Sommer F, Backhed F. The gut microbiota–masters of host development and physiology. Nature Rev. 2013;11(4):227238.


Souza M, Cheetham SM, Azevedo MS, Costantini V, Saif LJ. Cytokine and antibody responses in gnotobiotic pigs after infection with human norovirus genogroup II.4 (HS66 strain). J. Virol. 2007;81(17):91839192.


Souza M, Costantini V, Azevedo MS, Saif LJ. A human norovirus-like particle vaccine adjuvanted with ISCOM or mLT induces cytokine and antibody responses and protection to the homologous GII.4 human norovirus in a gnotobiotic pig disease model. Vaccine. 2007;25(50):84488459.


Souza M, Azevedo MS, Jung K, Cheetham S, Saif LJ. Pathogenesis and immune responses in gnotobiotic calves after infection with the genogroup II.4-HS66 strain of human norovirus. J. Virol. 2008;82(4):17771786.


Strong DW, Thackray LB, Smith TJ, Virgin HW. Protruding domain of capsid protein is necessary and sufficient to determine murine norovirus replication and pathogenesis in vivo. J. Virol. 2012;86(6):29502958.


Subekti DS, Tjaniadi P, Lesmana M, et al. Experimental infection of Macaca nemestrina with a Toronto Norwalk-like virus of epidemic viral gastroenteritis. J. Med. Virol. 2002;66(3):400406.


Takanashi S, Wang Q, Chen N, et al. Characterization of emerging GII.g/GII.12 noroviruses from a gastroenteritis outbreak in the United States in 2010. J. Clin. Microbiol. 2011;49(9):32343244.


Takanashi S, Saif LJ, Hughes JH, et al. Failure of propagation of human norovirus in intestinal epithelial cells with microvilli grown in three-dimensional cultures. Arch. Virol. 2014;159(2):257266.


Tan M, Jiang X. Histo-blood group antigens: a common niche for norovirus and rotavirus. Exp. Rev. Mol. Med. 2014;16:e5.


Taube S, Perry JW, Yetming K, et al. Ganglioside-linked terminal sialic acid moieties on murine macrophages function as attachment receptors for murine noroviruses. J. Virol. 2009;83(9):40924101.


Taube S, Jiang M, Wobus CE. Glycosphingolipids as receptors for non-enveloped viruses. Viruses. 2010;2(4):10111049.


Taube S, Perry JW, McGreevy E, et al. Murine noroviruses bind glycolipid and glycoprotein attachment receptors in a strain-dependent manner. J. Virol. 2012;86(10):55845593.


Taube S, Kolawole AO, Hohne M, et al. A mouse model for human norovirus. MBio. 2013;4(4):e00450e00513.


Teunis PF, Moe CL, Liu P, et al. Norwalk virus: how infectious is it? J. Med. Virol. 2008;80(8):14681476.


Teunis PF, Sukhrie FH, Vennema H, Bogerman J, Beersma MF, Koopmans MP. Shedding of norovirus in symptomatic and asymptomatic infections. Epidemiol. Infect. 2014:18.


Thackray LB, Wobus CE, Chachu KA, et al. Murine noroviruses comprising a single genogroup exhibit biological diversity despite limited sequence divergence. J. Virol. 2007;81(19):1046010473.


Thackray LB, Duan E, Lazear HM, et al. Critical role for interferon regulatory factor 3 (IRF-3) and IRF-7 in type I interferon-mediated control of murine norovirus replication. J. Virol. 2012;86(24):1351513523.


Thorne LG, Goodfellow IG. Norovirus gene expression and replication. J. Gen. Virol. 2014;95(Pt 2):278291.


Treanor JJ, Atmar RL, Frey SE, et al. A Novel Intramuscular Bivalent Norovirus Virus-Like Particle Vaccine Candidate-Reactogenicity, Safety, and Immunogenicity in a Phase 1 Trial in Healthy Adults. J. Infect. Dis. 2014;210(11):17631771.


Troeger H, Loddenkemper C, Schneider T, et al. Structural and functional changes of the duodenum in human norovirus infection. Gut. 2009;58(8):10701077.


Tsunesumi N, Sato G, Iwasa M, Kabeya H, Maruyama S, Tohya Y. Novel Murine Norovirus-Like Genes in Wild Rodents in Japan. J. Vet. Med. Sci. 2012;74:12211224.


Tuladhar E, Bouwknegt M, Zwietering MH, Koopmans M, Duizer E. Thermal stability of structurally different viruses with proven or potential relevance to food safety. J. Appl. Microbiol. 2012;112(5):10501057.


Uchiyama R, Chassaing B, Zhang B, Gewirtz AT. Antibiotic treatment suppresses rotavirus infection and enhances specific humoral immunity. J. Infect. Dis. 2014;210(2):171182.


Vashist S, Bailey D, Putics A, Goodfellow I. Model systems for the study of human norovirus Biology. Future Virol. 2009;4(4):353367.


Vlasova AN, Chattha KS, Kandasamy S, et al. Lactobacilli and bifidobacteria promote immune homeostasis by modulating innate immune responses to human rotavirus in neonatal gnotobiotic pigs. PLoS One. 2013;8(10):e76962.


Ward VK, McCormick CJ, Clarke IN, et al. Recovery of infectious murine norovirus using pol II-driven expression of full-length cDNA. Proc. Natl. Acad. Sci. USA. 2007;104(26):1105011055.


Wei C, Farkas T, Sestak K, Jiang X. Recovery of infectious virus by transfection of in vitro-generated RNA from tulane calicivirus cDNA. J. Virol. 2008;82(22):1142911436.


Wobus CE, Karst SM, Thackray LB, et al. Replication of Norovirus in cell culture reveals a tropism for dendritic cells and macrophages. PLoS Biol. 2004;2(12):e432.


Wobus CE, Thackray LB, Virgin IV HW. Murine norovirus: a model system to study norovirus biology and pathogenesis. J. Virol. 2006;80(11):51045112.


Wyatt RG, Greenberg HB, Dalgard DW, et al. Experimental infection of chimpanzees with the Norwalk agent of epidemic viral gastroenteritis. J. Med. Virol. 1978;2(2):8996.


Yunus MA, Chung LM, Chaudhry Y, Bailey D, Goodfellow I. Development of an optimized RNA-based murine norovirus reverse genetics system. J. Virol. Methods. 2010;169(1):112118.


Zhang D, Huang P, Zou L, Lowary TL, Tan M, Jiang X. Tulane virus recognizes the A type 3 and B histo-blood group antigens. J. Virol. 2015;89(2):14191427.


Zheng DP, Ando T, Fankhauser RL, Beard RS, Glass RI, Monroe SS. Norovirus classification and proposed strain nomenclature. Virology. 2006;346(2):312323.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Apr 25, 2018 | Posted by in MICROBIOLOGY | Comments Off on Animal Models of Norovirus Infection

Full access? Get Clinical Tree

Get Clinical Tree app for offline access