Open access peer-reviewed chapter

Norovirus Structure and Classification

Written By

Manisha Rani, Sushma Rajyalakshmi, Sunitha Pakalapaty and Nagamani Kammilli

Submitted: 14 February 2021 Reviewed: 03 May 2021 Published: 01 September 2021

DOI: 10.5772/intechopen.98216

From the Edited Volume

Norovirus

Edited by Gyula Mózsik

Chapter metrics overview

568 Chapter Downloads

View Full Metrics

Abstract

Norovirus are a major cause of acute gastroenteritis worldwide. Diarrheal disease is now the fourth common cause of mortality children under the age of 5 years but remain the 2nd most cause of morbidity. NoV are associated with 18% diarrheal diseases worldwide where rotavirus vaccinations has been successfully introduced. NoV has become major cause of gastroenteritis in children. NoV belong to family caliciviridae. They are non-enveloped, single stranded positive sense RNA Viruses. The genome consists of 3 Open reading frames, ORF-1 codes for non-structural protein, ORF-2 codes for major capsid protein VP1 and ORF-3 for minor capsid protein VP2. Based on sequence difference of the capsid gene (VP1), NoV have been classified in to seven genogroup GI-GVII with over 30 genotypes. Genogroups I, II, IV are associated with human infection. Despite this extensive diversity a single genotype GII.4 has been alone to be the more prevalent. Basic epidemiological disease burden data are generated from developing countries. NoV are considered fast evolving viruses and present an extensive diversity that is driven by acquisition of point mutations and recombinations. Immunity is strain or genotype specific with little or no protection conferred across genogroups. Majority of outbreaks and sporadic norovirus cases worldwide are associated with a single genotype, GII.4 which was responsible for 62% of reported NoV outbreaks in 5 continents from 2001 to 2007. GII.4 variants have been reported as major cause of global gastroenteritis pandemics starting in 1995 frequent emergence of novel GII.4 variants is known to be due to rapid evolution and antigenic variation in response to herd immunity. Novel GII.4 variants appear almost every 2 years. Recent GII.4 variant reported include Lordsdale 1996, Farmington Hills 2002, Hunter 2004, Yerseke 2006a, Den Haag 2006b, Apeldoon 2007, New Orleans 2009,most recently Sydney 2012. Detailed molecular epidemiologic investigation of NoV is associated for understanding the genetic diversity of NoV strain and emergence of novel NoV variants. However, reports have revealed that not all individuals develop symptoms and a significant proportion remains asymptomatic after NoV infections.

Keywords

  • Acute gastroenteritis
  • ORF
  • Genogroups
  • Immunity

1. Introduction

The acute gastroenteritis is a major health problems, one of the most common infectious diseases among humans [1, 2]. The annual incidence of diarrheal disease is estimated of annual number is over 4.5 billion cases worldwide [3] The global estimated of annual number of mortalities with gastroenteritis vary between 3.5–5 million cases in majority of deaths occurring among people in developing countries [4]. Diarrhoea remains into 10 most common communicable diseases found in India [5] (annual mortality is 2.5 million deaths each year in children less than five years of age in developing countries) [6].

In human enteric viruses account for more than half of all cases of gastroenteritis worldwide [7, 8]. Viral causes of gastroenteritis are follow: norovirus, rotavirus, adenovirus (group F- type 40/41), astrovirus and sapovirus [9, 10, 11].

Advertisement

2. Norovirus

Norwalk virus is the prototype strain of Norovirus and was associated with an outbreak of gastroenteritis at an elementary school in Norwalk, Ohio, in 1968. The discovery of Norovirus as the aetiological agent of the outbreak was made by Albert Kapikian in 1972 [12].

Using immune electron microscopy (IEM), stool samples were examined from a volunteer who had been experimentally inoculated with a faecal filtrate from the original outbreak. From these studies, Kapikian proposed the name “Norwalk virus” as the causative agent of the outbreak [12]. This was the first human virus specifically associated with gastroenteritis.

2.1 Classification

Norovirus, previously known as Norwalk-like viruses, belongs to the family, Caliciviridae [13]. The Caliciviridae family is comprised of four genera, Norovirus, Sapovirus, Lagovirus and Vesivirus [14]. Norovirus and Sapovirus are found in the genera Norovirus and Sapovirus, respectively, whilst other caliciviruses of veterinary importance, rabbit hemorrhagic disease virus and feline calicivirus, are found in Lagovirus and Vesivirus, respectively. Recently, two additional generas have been proposed within the Caliciviridae family, provisionally named Becovirus or Nabovirus, a bovine enteric calicivirus [15, 16, 17]. All six genera infect animals, but only Norovirus and Sapovirus contain strains that infect both humans and animals.

Based on phylogenetic analysis of the full length nucleotide sequence of the capsid gene [VP1], the Norovirus genus is divided into five genogroups (GI, GII, GIII, GIV and GV). Norovirus GI, GII and GIV are associated with human gastroenteritis. Norovirus GII includes porcine, as well as human strains, GIII contains only bovine strains, and GV contains only murine strains [18].

2.2 Structure

Norovirus is a small virion of 27 to 32 nm in diameter and has a buoyant density of 1.33 to 1.41 g/cm3 in caesium chloride [19, 20]. It is a non-enveloped, single-stranded, positive-sense, RNA virus with a genome of 7.4 to 7.7 kb [21, 22]. The RNA is polyadenylated at the 3′ end. All calicivirus genomes begin with a GU [nucleotide sequence] at the 5′ end terminal. A 5′ end sequence, of between 16 and 28 nucleotides depending on the genus is repeated internally in the genome and corresponds to the start of the subgenomic RNA [located at the start of the capsid gene, VP1]. This sequence is thought to be part of an RNA-dependent RNA polymerase [RdRp] promoter [23].

The Norovirus genome contains three ORFs: ORF1, ORF2 and ORF3.

The initial characterisation of the genome was based on the sequence homology of ORF1 in human calicivirus to characterised proteins of picornaviruses [24]. These conserved motifs included a “2C-like” helicase [a nucleoside triphosphatase, NTPase, in Norovirus genome], a “3C-like” protease [3CLpro] and a “3D-like” RdRp (RdRp in Norovirus genome). Proteolytic mapping and enzymatic studies using site directed mutagenesis and recombinant expression systems have revealed the presence of three other non-structural proteins within the Norovirus polyprotein, including a 3A-like protein, a viral protein-genome linked (VpG) and a N-terminal protein of unknown function [25, 26, 27, 28, 29]. All six non-structural proteins proceed N to C terminus in the Norovirus polyprotein (Figure 1).

Figure 1.

Genomic organisation of NoV. The genomic organisation and nucleotide positions are shown with reference to human NoV/Lordsdale virus/1993/UK, GenBank accession number X86557. The NoV genome is organised into three ORFs, with the 3′ end of ORF1 overlapping the 5′ end of ORF2 by 20 bp, and the 3’end of ORF2 overlaps the 5′ end of ORF3 by one bp. ORF1 encodes for six non-structural proteins: N terminal protein, nucleoside triphosphate (NTPase), a 3A-like protein, viral protein-genome linked (VpG), “3C-like” protease (3CLpro) and an RdRp. ORF2 encodes for the major structural protein, VP1, which self assembles into the viral capsid. ORF3 encodes for a minor structural protein, VP2, involved in stabilisation of VP1. The polyadenylated tail at the 3’end of the genome is indicated by [a]. The two putative RdRp promoter sites are shown below the image as black boxes.

ORF1 encodes a 200 kDa polyprotein which undergoes proteolytic cleavage mediated by a virus-encoded 3CLpro, located upstream of the RdRp. Proteolytic processing is rapid, co-translational and results in the production of six non-structural proteins [30, 31]. ORF2 encodes the major structural protein, VP1 [60 kDa], which is responsible for capsid-related functions, including self-assembly and capsid formation, host interactions and immunogenicity of the virus [32, 33, 34].

The ORF3 region encodes a small basic protein of 20 to 30 kDa involved in expression and stability of the VP1 capsid protein [34]. Downstream from ORF3, a 42 to 78 nucleotide non-translated region is present and attached to a polyadenylated tail [35].

2.3 Non-structural proteins

2.3.1 N terminal protein

Expression of the Norovirus N terminal protein demonstrated that the N terminal protein was localised to the golgi apparatus and led to its disassembly into discrete aggregates [36]. In addition, the N terminal protein interacts with the vesicle-associated membrane protein–associated protein A [VAP-A], which plays a role in regulated vesicle transport [37, 38]. Therefore, the N terminal protein is predicated to interact with intracellular membranes and may act as an anchor to membrane-bound replication complexes of Norovirus [39].

2.3.2 NTPase

NTPase protein (alternatively designated p41) of the Norovirus GI strain, Southampton virus, has NTPase activity and a helicase domain. The protein sequence of the Norovirus p41 protein showed regions of high similarity to the 2C protein of enteroviruses. Norovirus may hydrolyse NTPs for a function distinct from nucleic acid unwinding [40]. The specific role of p41 in the viral replicative cycle has not yet been determined.

2.3.3 3A-like protein

A parallel between picornaviruses and caliciviruses have been demonstrated for the 3A and 3A-like protein, respectively [41, 42, 43]. The 3A-like protein [alternatively designated p22 or p20 for Norovirus GI and GII, respectively] in the Norovirus genome occupies a position similar to that of the 3A protein in picornavirus. The specific function of the Norovirus 3A-like protein is unknown, but it has been suggested to be involved in cellular membrane trafficking and replication complexes [44].

2.3.4 VpG

VpG is essential for the production of infectious caliciviruses [45]. Human VpG has been shown to bind to translational initiation factors in vitro and may also be involved in the recruitment of ribosomes to viral RNA. Recently, VpG has been suggested to play a role in RNA replication [46, 47]. VpG was uridylylated in vitro by the RdRp, suggesting it may function as a protein-primer during RNA replication. Another study by Belliot and colleagues demonstrated that Norovirus VpG was nucleotidylylated by the proteinase-polymerase form of the human Norovirus RdRp. This occurred in a template-independent manner in the presence of Mn2+; furthermore, the linkage between RNA to VpG was covalent. Mutational analysis identified tyrosine 27 of the Norovirus VpG as the target amino acid for this linkage, which was susceptible to phosphodiesterase treatment. Thus, the linkage of RNA to VpG via a phosphodiester bond was confirmed. In addition, there was evidence for the presence of an RNA element in the 3′ end of the polyadenylated genome which enhanced nucleotidylylation of the VpG by the RdRp in the presence of Mg2+ [48].

2.3.5 “3C-like” protease

Norovirus 3CLpro (19 kDa) is crucial to the proteolytic processing of ORF1 polyprotein into six non-structural proteins. Characterisation of Norovirus 3CLpro has revealed an active nucleophilic residue in the conserved GDCG motif, common to all chymotrypsin-like 3Cpro. The motif contains amino acid residues essential to formation of an active site. The amino acid residues exists as a catalytic triad in Norovirus, and include cysteine (Cys139), histidine (His 30), and glutamate (Glu 54), which function as a nucleophile, general base, and anion, respectively. All three amino acid residues are important to the enzymatic activity for proteolysis [49, 50].

It has also been suggested that the Norovirus 3CLpro can cleave the host encoded poly [A]-binding protein, and as a result, cellular translation is inhibited. This suggests an important mechanism of host cell modulation during viral replication [51].

2.3.6 RNA-dependent RNA polymerase

The Norovirus RdRp is a non-structural protein involved in the replication of the Norovirus genome. It has been proposed that Norovirus proteinase-polymerase precursor is a bifunctional enzyme with protease and RdRp activity both exhibited during viral replication [52].

2.4 Structural proteins

2.4.1 VP1

ORF2 encodes the major capsid protein, VP1, of the Norovirus genome. The VP1 capsid protein can be divided into three domains, the N terminal domain, shell [S] domain, which is buried inside the capsid, and a protruding [P]domain. A flexible hinge connects the S and P domains. The S domain is highly conserved and is essential for the formation of the icosahedral capsid shell. The P domain comprises of two subdomains: P1, a moderately conserved subdomain and P2, which is hypervariable in its nucleotide sequence. The P2 subdomain of the norovirus genome is the most exposed region of the capsid structure, hence, it contains immune and cellular recognition binding sites [53, 54, 55, 56].

2.4.2 VP2

ORF3 encodes a small minor structural protein, VP2, of the Norovirus genome. VP2 is highly variable in sequence between strains and varies in length from 208 to 268 amino acids. The function of VP2 involves the upregulation of VP1 expression and stabilisation of the VP1 in the virus structure. Furthermore, VP2 protects VP1 from disassembly and protease degradation [57]. The role of VP2 in viral replication is unknown, but it may interact with RNA, due to its highly basic, and therefore be involved in packaging of the viral genome. In addition, the VP2 protein is reported to be involved in the formation of infectious viral particles [58, 59].

2.4.3 Transmission

A highly infectious agent, Norovirus is primarily transmitted through person-to person and commonly via the faecal oral route. Aerosolised vomitus containing Norovirus is another transmission mode by which the virus disseminates in outbreaks of gastroenteritis [60, 61]. A study by Marks et al. reported attack rates of Norovirus infections of up to 60% in individuals in close proximity (who were seated next to and on the adjacent table in a restaurant) to the index person who vomited. The attack rate of infection was directly proportional to the distance from the vomiter. Other sources of transmission include the consumption of contaminated food (oysters, vegetables, fresh and frozen produce) [62, 63, 64, 65] or water (drinking, ice or recreational) [66, 67, 68]. In addition, fomite contamination in an outbreak setting has been demonstrated as an alternative transmission route [69, 70].

Advertisement

3. Clinical features and pathogenesis of norovirus

3.1 Clinical manifestation

Norovirus infection is characterised by an onset of vomiting, diarrhoea, nausea, and may also be accompanied by variable systemic symptoms including, fever, headache, chills or myalgia [71, 72, 73, 74]. Diarrhoeal stool is non-bloody, lacks mucus and may be loose or watery. Following an incubation period of 1 to 2 days, the illness is usually mild and self-limited, which generally persists for a short duration of 1 to 3 days. A 68% sensitivity and 99% specificity was determined when the criteria was used in conjunction with laboratory detection techniques, including ELISA and nucleic acid amplification assays [75]. Norovirus infection affects all age groups and is often more severe in the elderly, the young, and in transplant and immunocompromised patients [76, 77, 78, 79]. Studies have shown that symptoms can persist for up to five days or longer and infection may progress to chronic disease [80]. Prolonged viral shedding can occur in the presence or absence of clinical symptoms and death may occur [81, 82, 83, 84, 85, 86].

3.2 Pathogenesis

The pathogenicity of NoV was studied in human volunteers inoculated with the prototype strain, Norwalk virus [NoV GI]. Acute infection with NoV resulted in a histopathological lesion in the jejunum and correlated with a broadening and blunting of the villi and crypt cell hyperplasia of the small intestinal tract. These observations provided suggestive evidence that NoV replication is restricted to the small intestine.

Additional studies in volunteers who developed an illness or characteristic lesion, showed the levels of the small intestinal brush border enzymatic activities [alkaline phosphatase, sucrase and trehalase] were significantly reduced, resulting in transient carbohydrate malabsorption [87]. Furthermore, there was a marked delay in gastric emptying. It has been suggested that the reduced gastric motility is responsible for symptoms, specifically nausea and vomiting associated with gastroenteritis.

3.3 Immunity

The immunogenicity associated with Norovirus disease is not well defined. Early studies on host immune responses to Norovirus infection were based on human challenge studies by oral immunisation with either infectious virus or recombinant VLPs [88, 89, 90, 91, 92, 93]. Challenge studies have shown that short-term immunity lasts for six to 14 weeks, and is strain specific [94]. Thus, infection is induced following challenge to a serologically distinct strain. Interestingly, individuals with high levels of pre-existing antibodies against Norwalk virus were reportedly more susceptible to infection than individuals who had a non-detectable or had low levels of serum antibodies after challenge with the same strain.

More recently, the structural recognition site of HBGAs by Norovirus has been determined by mutagenesis and crystallographic studies [95]. Based on crystallographic structures, the receptor site involved in host-cell recognition was the P domain, more specifically the outermost P2 surface on the Norovirus capsid gene [96, 97]. Such findings will provide an understanding into the complex interaction between HBGAs and Norovirus, and could lead to intervention strategies to block attachment of virus to host recognition sites. The study of the role of genetic mechanisms in Norovirus infection is a new area in Norovirus immunology, and further studies are required to understand the complex interactions between specific Norovirus genotypes (particularly, newly emergent Norovirus strains) and susceptibility to infection.

3.4 Replication

Little is known about human Norovirus biology, in particular, human Norovirus replication, immunogenicity and pathogenicity due to the lack of an in vitro cell culture and small animal model systems [98]. However, in recent times our understanding of calicivirus replication has come from other studies, including the animal calicivirus, Feline calicivirus [99], and the use of a gnotobiotic pig as an animal model for the study of human Norovirus pathogenesis [100]. However, a significant advancement in the study of Norovirus biology was the development of the first in vitro cell culture system for the cultivation of murine norovirus 1 (MNV-1) [101]. MNV-1 was used to study immunity and pathogenesis of Norovirus in a mouse model. Subsequently, MNV-1 was successfully propagated in the murine macrophage cell line RAW 264.7 and revealed a tropism for cells of the haematopoietic lineage, specifically the macrophages and dendritic cells. It was proposed that macrophages could contribute to the spread of Norovirus through the host. Norovirus infection of dendritic cells in the lumen of the intestine also provides a point of infection for Norovirus; however, it remains unclear if human Norovirus targets such cells. Regardless, MNV share many molecular and biological properties with human Norovirus, and therefore, provides an important animal model to understand the biology and pathogenesis of human Norovirus infection. Other systems which have provided significant information regarding Norovirus replication are the replicon and reverse genetics systems.

Molecular advances have led to the development of a Norovirus replicon and a recombinant T7 vaccinia virus expressed Norovirus [102, 103, 104, 105]. Studies have shown Norovirus RNA is infectious and capable of replication in three cell types: human hepatoma Huh7-cells, hamster BHK21 cells and human embryonic kidney [HEK] 293 T/17 cells. However, the main limitation of these systems was the inability for virions to spread to other neighbouring cells in the culture system. The inability to culture human Norovirus has been suggested to occur at the level of attachment and entry into the cells. Another in vitro cell culture system for human Norovirus was recently reported based on a rotating wall vessel bioreactor technology to engineer a 3D model of the human small intestinal epithelium (Figure 2).

Figure 2.

Intestinal biopsy of jejunal tissue from a human volunteer infected with Norwalk virus. (A-left fig) Normal jejunal biopsy before administration of Norwalk virus. Villi and cellularmorphology appear normal. (B-right fig) Jejunal biopsy after administration with the viral agent. Villi are broadened and flattened; epithelial lining cells appear disorganised. Image taken from [106].

However, the model may not provide direct evidence of in vitro propagation of human Norovirus and needs further investigation [107]. Nevertheless, the system can offer an insight into host-cell interaction in Norovirus infection.

Recently, an infectious reverse genetics system for MNV that generates.

infectious virus from a genomic complementary DNA [cDNA] clone under the control of an RNA polymerase II promoter was described. The principle of the Norovirus reverse genetics system was demonstrated by mutagenesis of the protease polymerase cleavage site to show that the protease-polymerase cleavage was essential for the recovery of infectious MNV [108]. Overall, the development of such systems provides an approach to perform functional analyses of the Norovirus genome, as well as the study of the molecular biology and replication of Norovirus.

3.5 RNA recombination

RNA recombination is an important mechanism in the evolution of RNA viruses. Recombination in viruses can affect phylogenetic groupings, increase the virulence and pathogenicity of the virus, and affect anti-viral drug design. By exchange of genetic material through recombination, a new variant of the virus is produced [109]. In recent years, human Norovirus recombinants have been detected frequently in cases of gastroenteritis worldwide. This increase in prevalence of Norovirus recombinants suggests that infection with at least two virus strains is common. The proposed site of recombination in Norovirus is within the highly conserved ORF1/ORF2 overlap [110, 111, 112].

3.6 Treatment and prevention

Norovirus associated gastroenteritis is mild and self-limiting, and generally resolves without complications. However, death from Norovirus associated gastroenteritis has been previously reported [113, 114, 115]. In severe cases of Norovirus infection, hospitalisation is required and the administration of an oral fluid and electrolyte treatment is often required to replace the loss of fluids. The oral administration of bismuth subsalicylate after the onset of symptoms has been demonstrated to reduce the duration of abdominal cramps and gastrointestinal symptoms during experimentally induced Norovirus illness in adults [116]. The best control measure for the prevention of Norovirus infection is with good hygiene practices. These include, thorough and frequent hand washing, and the disposal or disinfection of contaminated materials. In addition, extra measures should be implemented in healthcare facilities to prevent large-scale outbreaks, such as restriction of staff movement between wards containing infected patients, the isolation of symptomatic patients, the exclusion of affected staff from work until 48 h after the cessation of symptoms, and the closure of affected units to limit the spread of infection. However, the impact of preventive measures in affected institutions is reduced due to the environmental stability of Norovirus outside the host. This is due to the fact that Norovirus has a non-enveloped structure, is acid stable, persists in the environment and resistant to chlorination of up to 300 ng/ml. Furthermore, quaternary ammonium disinfectants are ineffective in the disinfection of Norovirus [117, 118]. Although a combination of detergent and sodium hypochlorite solution has been reported to be effective in the decontamination of surfaces [119]. Therefore, to prevent and control the spread of Norovirus disease, strict hand hygiene and use of effective disinfectants should be enforced during outbreaks. Importantly, for the efficient implementation of precautionary measures in an outbreak setting, a rapid detection system for the diagnosis of a Norovirus infection would be ideal.

Advertisement

4. Laboratory diagnosis

4.1 Detection of norovirus

Detection of the aetiological agent of gastroenteritis is important as only bacterial and parasitic agents are treatable by current therapeutic agents.

Furthermore, for clinical and epidemiological studies the availability of detection methods for viral nucleic acid, viral antigen, or antibody responses is valuable. Various methods have been used for the diagnosis of Norovirus infection, including electron microscopy [EM], IEM, radioimmunoassays, ELISAs and viral RNA based nucleic acid amplification assays. Of the available detection methods, the most commonly used assays for Norovirus diagnosis include ELISAs and RT-PCR [120].

4.1.1 Electron microscopy

The detection of Norovirus has traditionally relied on EM. It enables the identification of Norovirus by their characteristic morphology. However, the sensitivity of EM detection is low, requiring at least 106 viral particles/g of stool for visualisation. Therefore, this technique is useful only for specimens collected immediately upon the onset of illness when substantial quantities of viral shedding occur. Furthermore, EM is a robust tool but time consuming, requires a high level of technical skill, is labour intensive and not available to all clinical laboratories. Thus, EM is not feasible for large epidemiological studies. Modifications of the EM method, such as, or solid phase IEM [121, 122, 123, 124, 125] have also been used to aid in virus identification. Both these methods are based on antigen–antibody reactions. However, like EM, the application of IEM is rarely applied to epidemiological investigations.

4.1.2 Elisa

An ELISA offers an efficient diagnostic method for the identification of Norovirus infection. The rapid turnover and simplicity for screening a large number of samples makes ELISAs an ideal system for use in a diagnostic laboratory. Norovirus are antigenically diverse and therefore assays may be limited in the detection of a broad range of Norovirus strains in circulation. This has probably contributed to the poor performance assessments [sensitivity and specificity] of commercially available ELISAs in different countries when compared to sensitive molecular methods, such as RT-PCR [126, 127, 128]. The potential for ELISAs to give false negatives and false-positives due to poor sensitivity and poor specificity, respectively, has limited their use for diagnosing outbreaks where large numbers of samples are being tested.

4.1.3 Reverse transcription-polymerase chain reaction

RT-PCR has remained the most reliable means of diagnosing Norovirus infection as it is the most sensitive routine method used compared to EM and ELISA [129, 130]. The availability of RT-PCR amplification has greatly facilitated sequencing and genome characterisation of Norovirus strains [131, 132, 133]. The RT-PCR assay employs primers that target conserved regions of the Norovirus genome, such as the RdRp and/or the VP1 gene. Until recently, Norovirus RT-PCR assays have used primers that targeted the RdRp [3′ end of ORF1 of the Norovirus genome], which is highly conserved among Norovirus. By sequence analysis of the capsid gene [VP1] in the Norovirus genome, another conserved region located at the 5′ end of the capsid gene was identified. This region offered better segregation of Norovirus genotypes by phylogenetic analysis. Moreover, analysis of both regions, RdRp and VP1 is necessary for the detection of Norovirus recombinant strains. These technical advances have improved detection and enhanced epidemiologic surveillance by molecular genotyping and sequence analysis. However, conventional RT-PCR assays have progressively been replaced by real-time RT-PCR, which is more sensitive, faster and offers quantification of RNA viruses. This technology is not only quicker but enables quantitation using the Ct of the unknown target RNA sample compared directly to the Ct of a standard curve, which contains a defined number of copies of the target virus. The Ct value is the basis for accurate and reproducible quantitation using real-time RT-PCR. The application of a standard curve in a real-time RT-PCR assay also enables the determination of viral kinetic parameters associated with Norovirus infection, such as the number of viruses excreted [that is, a measure of viral load in a sample], duration of viral excretion and the viral decay rate.

Several real-time nucleic acid amplification assays have been developed for the detection and quantitation of Norovirus RNA in clinical specimens, by the use of SYBR Green dye chemistry, and probes, including taq-man probes and hybridization probes [134, 135].

References

  1. 1. Murray, C. J. L., and A. D. Lopez. 1997. Mortality by cause for eight regions of he world: global burden of disease study. Lancet 349:1269-1276
  2. 2. WHO 2002, posting date. Revised global burden of disease (GBD) 2002estimates- incidence. [Online.]
  3. 3. WHO 2002, posting date. Revised global burden of disease (GBD) 2002 estimates- incidence. [Online.]
  4. 4. Snyder, J. D., and M. H. Merson. 1982. The magnitude of the global problem of acute diarrhoeal disease: a review of active surveillance data. Bull World Health Organ 60:605-631
  5. 5. Top 13 Common Communicable Diseases Found in India. [Online.]
  6. 6. Kosek, M., C. Bern, and R. L. Guerrant. 2003. The global burden of diarrhoeal disease, as estimated from studies published between 1992 and 2000. Bull World Health Organ 81:197-204
  7. 7. Hansman, G. S., L. T. Doan, T. A. Kguyen, S. Okitsu, K. Katayama, S. Ogawa, K. Natori, N. Takeda, Y. Kato, O. Nishio, M. Noda, and H. Ushijima. 2004. Detection of norovirus and sapovirus infection among children with gastroenteritis in Ho Chi Minh City, Vietnam. Arch Virol 149:1673-1688
  8. 8. Chen, S. Y., Y. C. Chang, Y. S. Lee, H. C. Chao, K. C. Tsao, T. Y. Lin, T. Y. Ko, C. N. Tsai, and C. H. Chiu. 2007. Molecular epidemiology and clinical manifestations of viral gastroenteritis in hospitalized pediatric patients in Northern Taiwan. J Clin MicrobioI 45:2054-2057
  9. 9. Harrington, P. R., J. Vinje, C. L. Moe, and R. S. Baric. 2004. Norovirus capture with histo-blood group antigens reveals novel virus-ligand interactions. J Virol 78:3035-3045
  10. 10. Clark, B., and M. McKendrick. 2004. A review of viral gastroenteritis. Curr Opin Infect Dis 17:461-469
  11. 11. Wilhelmi, I., E. Roman, and A. Sanchez-Fauquier. 2003. Review: Viruses causing gastroenteritis. Clin Infect Dis 9:247-262
  12. 12. Kapikian, A. Z., R. G. Wyatt, R. Dolin, T. S. Thornhill, A. R. Kalica, and R.M. Chanock. 1972. Visualization by immune electron microscopy of a27-nm particle associated with acute infectious nonbacterial gastroenteritis. J Virol 10:1075-1081
  13. 13. Mayo, M. A. 2002. A summary of taxonomic changes recently approved by ICTV. Arch Virol 147:1655-1656
  14. 14. Green, K. Y., T. Ando, M. S. Balayan, T. Berke, I. N. Clarke, M. K. Estes, D. O. Matson, S. Nakata, J. D. Neill, M. J. Studdert, and H. J. Thiel. 2000. Taxonomy of the caliciviruses. J Infect Dis 181 Suppl 2:S322-S330
  15. 15. Bridger, J. C., G. A. Hall, and J. F. Brown. 1984. Characterization of a calici-like virus (Newbury agent) found in association with astrovirus in bovine diarrhea. Infect Immun 43:133-138
  16. 16. Oliver, S. L., E. Asobayire, A. M. Dastjerdi, and J. C. Bridger. 2006.Genomic characterization of the unclassified bovine enteric virus Newbury agent-1 (Newbury1) endorses a new genus in the family Caliciviridae. Virology 350:240-250
  17. 17. Smiley, J. R., K. O. Chang, J. Hayes, J. Vinje, and L. J. Saif. 2002.Characterization of an entero pathogenic bovine calicivirus representing a potentially new calicivirus genus. J Virol 76:10089-10098
  18. 18. Zheng, D. P., T. Ando, R. L. Fankhauser, R. S. Beard, R. I. Glass, and S. Monroe. 2006. Norovirus classification and proposed strain nomenclature. Virology 346:312-323
  19. 19. Kapikian, A. Z., M. K. Estes, and R. M. Chanock. 1996. Norwalk group ofviruses, p. 783-810. In B. N. Knipe and P. M. Howley (ed.), Fields. Lippincott, Raven, Philadelphia
  20. 20. Kapikian, A. Z., J. L. Gerin, R. G. Wyatt, T. S. Thornhill, and R. M. Chanock. 1973. Density in cesium chloride of the 27 nm 8FIIa particle associated with acute infectious nonbacterial gastroenteritis: determination by ultracentrifugation and immune electron microscopy Proc Soc ExpBiol Med 142:874-877
  21. 21. Green, K. Y., T. Ando, M. S. Balayan, T. Berke, I. N. Clarke, M. K. Estes, D. O. Matson, S. Nakata, J. D. Neill, M. J. Studdert, and H. J. Thiel. 2000. Taxonomy of the caliciviruses. J Infect Dis 181 Suppl 2:S322-S330
  22. 22. Jiang, X., D. Y. Graham, K. Wang, and M. K. Estes. 1990. Norwalk virus genome cloning and characterisation. Science 250:1580-1583
  23. 23. Meyers, G., C. Wirblich, and H. J. Thiel. 1991. Genomic and subgenomic RNAs of rabbit hemorrhagic disease virus are both protein-linked and packaged into particles. Virology 184: 677-686
  24. 24. Neill, J. D. 1990. Nucleotide sequence of a region of the feline calicivirus genome which encodes picornavirus-like RNA-dependent RNA polymerase, cysteine protease and 2C polypeptides. Virus Res 17:145-160
  25. 25. Dunham, D. M., X. Jiang, T. Berke, A. W. Smith, and D. O. Matson. 1998. Genomic mapping of a calicivirus VPg. Arch Virol 143:2421-2430
  26. 26. Liu, B., I. N. Clarke, and P. R. Lambden. 1996. Polyprotein processing in Southampton virus: identification of 3C-like protease cleavage sites by in vitro mutagenesis. J Virol 70:2605-2610
  27. 27. Liu, B. L., G. J. Viljoen, I. N. Clarke, and P. R. Lambden. 1999.Identification of further proteolytic cleavage sites in the Southampton calicivirus polyprotein by expression of the viral protease in E. coli. J Gen Virol 80 (Pt 2):291-296
  28. 28. Pfister, T., and E. Wimmer. 2001. Polypeptide p41 of a Norwalk-like virus is a nucleic acid-independent nucleoside triphosphatase. J Virol 75:1611-1619
  29. 29. Seah, E. L., J. A. Marshall, and P. J. Wright. 1999. Open reading frame 1of the Norwalk-like virus Camberwell: completion of sequence and expression in mammalian cells. J Virol 73:10531-10535
  30. 30. Liu, B., I. N. Clarke, and P. R. Lambden. 1996. Polyprotein processing in Southampton virus: identification of 3C-like protease cleavage sites by in vitro mutagenesis. J Virol70:2605-2610
  31. 31. Sosnovtseva, S. A., S. V. Sosnovtsev, and K. Y. Green. 1999. Mapping of the feline calicivirus proteinase responsible for autocatalytic processing of the nonstructural polyprotein and identification of a stable proteinase-polymerase precursor protein. J Virol 73:6626-6633
  32. 32. Chen, S. Y., Y. C. Chang, Y. S. Lee, H. C. Chao, K. C. Tsao, T. Y. Lin, T. Y.Ko, C. N. Tsai, and C. H. Chiu. 2007. Molecular epidemiology and clinical manifestations of viral gastroenteritis in hospitalized pediatric patients in Northern Taiwan. J Clin Microbiol 45:2054-2057
  33. 33. Jiang, X., M. Wang, D. Y. Graham, and M. K. Estes. 1992. Expression, self-assembly, and antigenicity of the Norwalk virus capsid protein. J Virol 66:6527-6532
  34. 34. Bertolotti-Ciarlet, A., S. E. Crawford, A. M. Hutson, and M. K. Estes. 2003. The 3’ End of Norwalk virus mRNA contains determinants that regulate the expression and stability of the viral capsid protein VP1: a novel function for the VP2 protein. J Virol 77:11603-11615
  35. 35. Green, K. Y., R. M. Chanock, and A. Z. Kapikan. 2001. Human Calicivirus, p. 841-874. In D. M. Knipe and P. M. Howley (ed.), Fields Virology, 4 ed, vol. 1. Lippincott Williams & Wilkins, Philadelphia
  36. 36. Fernandez-Vega, V., S. V. Sosnovtsev, G. Belliot, A. D. King, T. Mitra, A. Gorbalenya, and K. Y. Green. 2004. Norwalk virus N-terminal nonstructural protein is associated with disassembly of the Golgi complex in transfected cells. J Virol 78:4827-4837
  37. 37. Ettayebi, K., and M. E. Hardy. 2003. Norwalk virus nonstructural proteinp48 forms a complex with the SNARE regulator VAP-A and prevents cell surface expression of vesicular stomatitis virus G protein. J Virol77:11790-11797
  38. 38. Weir, M. L., H. Xie, A. Klip, and W. S. Trimble. 2001. VAP-A binds promiscuously to both v- and tSNAREs. Biochem Biophys Res Commun 286:616-621
  39. 39. Ettayebi, K., and M. E. Hardy. 2003. Norwalk virus nonstructural proteinp48 forms a complex with the SNARE regulator VAP-A and prevents cell surface expression of vesicular stomatitis virus G protein. J Virol 77:11790-11797
  40. 40. Pfister, T., and E. Wimmer. 2001. Polypeptide p41 of a Norwalk-like virus is a nucleic acid-independent nucleoside triphosphatase. J Virol75:1611-1619
  41. 41. Blakeney, S. J., A. Cahill, and P. A. Reilly. 2003. Processing of Norwalk virus nonstructural proteins by a 3C-like cysteine proteinase. Virology 308:216-224
  42. 42. Konig, M., H. J. Thiel, and G. Meyers. 1998. Detection of viral proteins after infection of cultured hepatocytes with rabbit hemorrhagic disease virus. J Virol 72:4492-4497
  43. 43. Sosnovtsev, S. V., M. Garfield, and K. Y. Green. 2002. Processing map and essential cleavage sites of the nonstructural polyprotein encoded by ORF1 of the feline calicivirus genome. J Virol 76:7060-7072
  44. 44. Belliot, G., S. V. Sosnovtsev, T. Mitra, C. Hammer, M. Garfield, and K. Y.Green. 2003. In vitro proteolytic processing of the MD145 norovirus ORF1 nonstructural polyprotein yields stable precursors and products similar to those detected in calicivirus-infected cells. J Virol 77:10957-10974
  45. 45. Burroughs, J. N., and F. Brown. 1978. Presence of a covalently linked protein on calicivirus RNA. J Gen Virol 41:443-446
  46. 46. Belliot, G., S. V. Sosnovtsev, K. O. Chang, P. McPhie, and K. Y. Green. 2008. Nucleotidylylation of the VPg protein of a human norovirus by its proteinase-polymerase precursor protein. Virology 374:33-49
  47. 47. Rohayem, J., I. Robel, K. Jager, U. Scheffler, and W. Rudolph. 2006.Protein-primed and de novo initiation of RNA synthesis by Norovirus 3Dpol. J Virol 80: 7060-7069
  48. 48. Belliot, G., S. V. Sosnovtsev, K. O. Chang, P. McPhie, and K. Y. Green. 2008. Nucleotidylylation of the VPg protein of a human norovirus by its proteinase-polymerase precursor protein. Virology 374:33-49
  49. 49. Someya, Y., N. Takeda, and T. Miyamura. 2002. Identification of active-site amino acid residues in the Chiba virus 3C-like protease. J Virol 76:5949-5958
  50. 50. Zeitler, C. E., M. K. Estes, and B. V. Venkataram Prasad. 2006. X-raycrystallographic structure of the Norwalk virus protease at 1.5-a resolution. J Virol 80:5050-5058
  51. 51. Kuyumcu-Martinez, M., G. Belliot, S. V. Sosnovtsev, K. O. Chang, K. Y. Green, and R. E. Lloyd. 2004. Calicivirus 3C-like proteinase inhibits cellular translation by cleavage of poly(A)-binding protein. J Virol 78:8172-8182
  52. 52. Belliot, G., H. Laveran, and S. S. Monroe. 1997. Detection and genetic differentiation of human astroviruses: phylogenetic grouping varies by coding region. Arch Virol 142:1323-1334
  53. 53. Hale, A. D., T. N. Tanaka, N. Kitamoto, M. Ciarlet, X. Jiang, N. Takeda, D.W. Brown, and M. K. Estes. 2000. Identification of an epitope common to genogroup1 “norwalk-like viruses”. J Clin Microbiol 38:1656-1660
  54. 54. Hardy, M. E., T. N. Tanaka, N. Kitamoto, L. J. White, J. M. Ball, X. Jiang, and M. K. Estes. 1996. Antigenic mapping of the recombinant Norwalk virus capsid protein using monoclonal antibodies. Virology 217:252-261
  55. 55. Lochridge, V. P., K. L. Jutila, J. W. Graff, and M. E. Hardy. 2005.Epitopes in the P2 domain of norovirus VP1 recognized by monoclonal antibodies that block cell interactions. J Gen Virol 86:2799-2806
  56. 56. Tan, M., P. Huang, J. Meller, W. Zhong, T. Farkas, and X. Jiang. 2003.Mutations within the P2 domain of norovirus capsid affect binding to human histo-blood group antigens: evidence for a binding pocket. J Virol 77:12562-12571
  57. 57. Bertolotti-Ciarlet, A., S. E. Crawford, A. M. Hutson, and M. K. Estes. 2003. The 3’ End of Norwalk virus mRNA contains determinants that regulate the expression and stability of the viral capsid protein VP1: a novel function for the VP2 protein. J Virol 77:11603-11615
  58. 58. Glass, P. J., L. J. White, J. M. Ball, I. Leparc-Goffart, M. E. Hardy, and M.K. Estes. 2000. Norwalk virus open reading frame 3 encodes a minor structural protein. J Virol 74:6581-6591
  59. 59. Asanaka, M., R. L. Atmar, V. Ruvolo, S. E. Crawford, F. H. Neill, and M.K. Estes. 2005. Replication and packaging of Norwalk virus RNA incultured mammalian cells. Proc Natl AcadSci U S A 102:10327-10332
  60. 60. Marks, P. J., I. B. Vipond, D. Carlisle, D. Deakin, R. E. Fey, and E. O. Caul. 2000. Evidence for airborne transmission of Norwalk-like virus (NLV) in a hotel restaurant. Epidemiol Infect 124:481-487
  61. 61. Marks, P. J., I. B. Vipond, F. M. Regan, K. Wedgwood, R. E. Fey, and E.O. Caul. 2003. A school outbreak of Norwalk-like virus: evidence for airborne transmission. Epidemiol Infect 131:727-736
  62. 62. Cheng, P. K., D. K. Wong, T. W. Chung, and W. W. Lim. 2005. Norovirus contamination found in oysters worldwide. J Med Virol 76:593-597
  63. 63. Herwaldt, B. L., J. F. Lew, C. L. Moe, D. C. Lewis, C. D. Humphrey, S. Monroe, E. W. Pon, and R. I. Glass. 1994. Characterization of a variant strain of Norwalk virus from a food-borne outbreak of gastroenteritis on a cruise ship in Hawaii. J ClinMicrobiol 32:861-866
  64. 64. Long, S. M., G. K. Adak, S. J. O’Brien, and I. A. Gillespie. 2002. General outbreaks of infectious intestinal disease linked with salad vegetables and fruit, England and Wales, 1992-2000. Commun Dis Public Health 5:101-105
  65. 65. Martinez, A., A. Dominguez, N. Torner, L. Ruiz, N. Camps, I. Barrabeig, Arias, J. Alvarez, P. Godoy, P. Balana, A. Pumares, R. Bartolome, D. Ferrer, U. Perez, R. Pinto, J. Buesa, and C. V. Gastroenteritis Study Group. 2008. Epidemiology of food borne Norovirus outbreaks in Catalonia, Spain. BMC Infect Dis 8:47
  66. 66. Maunula, L., I. T. Miettinen, and C. H. von Bonsdorff. 2005. Norovirus outbreaks from drinking water. Emerg Infect Dis 11:1716-1721
  67. 67. Podewils, L. J., L. Zanardi Blevins, M. Hagenbuch, D. Itani, A. Burns, C. Otto, L. Blanton, S. Adams, S. S. Monroe, M. J. Beach, and M. Widdowson. 2007. Outbreak of norovirus illness associated with a swimming pool. Epidemiol Infect 135:827-833
  68. 68. Webby, R. J., K. S. Carville, M. D. Kirk, G. Greening, R. M. Ratcliff, S. Crerar, K. Dempsey, M. Sarna, R. Stafford, M. Patel, and G. Hall. 2007. Internationally distributed frozen oyster meat causing multiple outbreaks of norovirus infection in Australia. Clin Infect Dis 44:1026-1031
  69. 69. Jones, E. L., A. Kramer, M. Gaither, and C. P. Gerba. 2007. Role of fomite contamination during an outbreak of norovirus on houseboats. Int J Environ Health Res 17:123-131
  70. 70. Wu, H. M., M. Fornek, K. J. Schwab, A. R. Chapin, K. Gibson, E. Schwab, C. Spencer, and K. Henning. 2005. A norovirus outbreak at along-term-care facility: the role of environmental surface contamination. Infect Control Hosp Epidemiol 26:802-810
  71. 71. Agus, S. G., R. Dolin, R. G. Wyatt, A. J. Tousimis, and R. S. Northrup. 1973. Acute infectious nonbacterial gastroenteritis: intestinal histopathology. Histologic and enzymatic alterations during illness produced by the Norwalk agent in man. Ann Intern Med 79:18-25
  72. 72. Dolin, R., N. R. Blacklow, H. DuPont, S. Formal, R. F. Buscho, J. A. Kasel, R. P. Chames, R. Hornick, and R. M. Chanock. 1971. Transmission of acute infectious nonbacterial gastroenteritis to volunteers by oral administration of stool filtrates. J Infect Dis 123:307-312
  73. 73. Turcios, R. M., M. A. Widdowson, A. C. Sulka, P. S. Mead, and R. I. Glass. 2006. Reevaluation of epidemiological criteria for identifying outbreaks of acute gastroenteritis due to norovirus: United States, 1998-2000. Clin Infect Dis 42:964-969
  74. 74. Gallimore, C. I., D. Lewis, C. Taylor, A. Cant, A. Gennery, and J. J. Gray. 2004. Chronic excretion of a norovirus in a child with cartilage hairhypoplasia (CHH). J Clin Virol 30:196-204
  75. 75. Goller, J. L., A. Dimitriadis, A. Tan, H. Kelly, and J. A. Marshall. 2004.Long-term features of norovirus gastroenteritis in the elderly. J Hosp Infect 58:286-291
  76. 76. Murata, T., N. Katsushima, K. Mizuta, Y. Muraki, S. Hongo, and Y. Matsuzaki. 2007. Prolonged norovirus shedding in infants <or=6 months of age with gastroenteritis. Pediatr Infect Dis J 26:46-49
  77. 77. Rockx, B., M. de Wit, H. Vennema, J. Vinje, E. De Bruin, Y. Van Duynhoven, and M. Koopmans. 2002. Natural history of human calicivirus infection: a prospective cohort study. Clin Infect Dis 35:246-253
  78. 78. Lee, B. E., X. L. Pang, J. L. Robinson, D. Bigam, S. S. Monroe, and J. K. Preiksaitis. 2008. Chronic Norovirus and Adenovirus infection in a solid organ transplant recipient. Pediatr Infect Dis J 27:360-362
  79. 79. Parashar, U. D., L. Dow, R. L. Fankhauser, C. D. Humphrey, J. Miller, T. Ando, K. S. Williams, C. R. Eddy, J. S. Noel, T. Ingram, J. S. Bresee, S.S. Monroe, and R. I. Glass. 1998. An outbreak of viral gastroenteritis associated with consumption of sandwiches: implications for the control of transmission by food handlers. Epidemiol Infect 121:615-621
  80. 80. Ozawa, K., T. Oka, N. Takeda, and G. S. Hansman. 2007. Norovirus infections in symptomatic and asymptomatic food handlers in Japan. J ClinMicrobiol 45:3996-4005
  81. 81. Lee, B. E., X. L. Pang, J. L. Robinson, D. Bigam, S. S. Monroe, and J. K. Preiksaitis. 2008. Chronic Norovirus and Adenovirus infection in a solid organ transplant recipient. Pediatr Infect Dis J 27:360-362
  82. 82. Jenkins, P., S. R. Greene, J. R. Archer, D. Hoang-Johnson, M. Quinn, P. Duncan, G. Johnson, B. I. Rosen, P. Smith, V. Reddy, J. Schlegelmilch, J. Pendarvis, M. Donovan, J. E. Gunn, M. A. Barry, M. Davies, J. Vinje, M.-A. Widdowson, Z. Moore, J. K. Schaffin, and J. E. Tate. 2007.Norovirus activity-United States, 2006-2007. MMWR Morb Mortal Wkly Rep 56:842-846
  83. 83. Graham, D. Y., X. Jiang, T. Tanaka, A. R. Opekun, H. P. Madore, and M.K. Estes. 1994. Norwalk virus infection of volunteers: new insights basedon improved assays. J Infect Dis 170:34-43
  84. 84. Gallimore, C. I., D. Cubitt, N. du Plessis, and J. J. Gray. 2004. Asymptomatic and symptomatic excretion of noroviruses during a hospital outbreak of gastroenteritis. J ClinMicrobiol 42:2271-2274
  85. 85. Schreiber, D. S., N. R. Blacklow, and J. S. Trier. 1973. The mucosal lesion of the proximal small intestine in acute infectious nonbacterial gastroenteritis. N Engl J Med 288:1318-1323
  86. 86. Agus, S. G., R. Dolin, R. G. Wyatt, A. J. Tousimis, and R. S. Northrup. 1973. Acute infectious nonbacterial gastroenteritis: intestinal histopathology. Histologic and enzymatic alterations during illness produced by the Norwalk agent in man. Ann Intern Med 79:18-25
  87. 87. Baron, R. C., H. B. Greenberg, G. Cukor, and N. R. Blacklow. 1984.Serological responses among teenagers after natural exposure to Norwalk virus. J Infect Dis 150:531-534
  88. 88. Greenberg, H. B., J. Valdesuso, A. Z. Kapikian, R. M. Chanock, R. G. Wyatt, W. Szmuness, J. Larrick, J. Kaplan, R. H. Gilman, and D. A. Sack. 1979. Prevalence of antibody to the Norwalk virus in various countries. Infect Immun 26:270-273
  89. 89. Johnson, P. C., J. J. Mathewson, H. L. DuPont, and H. B. Greenberg. 1990. Multiple-challenge study of host susceptibility to Norwalk gastroenteritis in US adults. J Infect Dis 161:18-21
  90. 90. Wolf, S., J. Hewitt, M. Rivera-Aban, and G. E. Greening. 2008. Detection and characterisation of F+ RNA bacteriophages in water and shellfish: Application of a multiplex real-time reverse transcription PCR. J Virol Methods 149:123-128
  91. 91. Matsui, S. M., and H. B. Greenberg. 2000. Immunity to calicivirus infection. J Infect Dis 181:S331-S335
  92. 92. Parrino, T. A., D. S. Schreiber, J. S. Trier, A. Z. Kapikian, and N. R. Blacklow. 1977. Clinical immunity in acute gastroenteritis caused by Norwalk agent. N Engl J Med 297:86-89
  93. 93. Bu, W., A. Mamedova, M. Tan, M. Xia, X. Jiang, and R. S. Hegde. 2008.Structural basis for the receptor binding specificity of Norwalk virus. J Virol 82:5340-5347
  94. 94. Cao, S., Z. Lou, M. Tan, Y. Chen, Y. Liu, Z. Zhang, X. C. Zhang, X. Jiang, X. Li, and Z. Rao. 2007. Structural basis for the recognition of blood group trisaccharides by Norovirus. J Virol8:5949-5957
  95. 95. Choi, J.-M., A. M. Huston, M. K. Estes, and B. V. V. Prasad. 2008.Atomic resolution structural characterisation of recognition of histo-blood group antigens by Norwalk virus. Proc Natl AcadSci U S A 105:9175-9180
  96. 96. Duizer, E., K. J. Schwab, F. H. Neill, R. L. Atmar, M. P. Koopmans, and M.K. Estes. 2004. Laboratory efforts to cultivate noroviruses. J Gen Virol 85:79-87
  97. 97. Chang, K. O., D. W. George, J. B. Patton, K. Y. Green, and S. V. Sosnovtsev. 2008. Leader of the capsid protein in feline calicivirus promotes the replication of Norwalk virus in cell culture. J Virol 82:9206-9217
  98. 98. Cheetham, S., M. Souza, T. Meulia, S. Grimes, M. G. Han, and L. J. Saif. 2006. Pathogensis of a genogroup II human norovirus in gnotobiotic pigs. J Virol 80:10372-10381
  99. 99. Karst, S. M., C. E. Wobus, M. Lay, J. Davidson, and H. W. t. Virgin. 2003. STAT1-dependent innate immunity to a Norwalk-like virus. Science 299:1575-1578
  100. 100. Asanaka, M., R. L. Atmar, V. Ruvolo, S. E. Crawford, F. H. Neill, and M.K. Estes. 2005. Replication and packaging of Norwalk virus RNA incultured mammalian cells. Proc Natl AcadSci U S A102:10327-10332
  101. 101. Chang, K. O., S. V. Sosnovtsev, G. Belliot, A. D. King, and K. Y. Green. 2006. Stable expression of a Norwalk virus RNA replicon in a human hepatoma cell line. Virology 353:463-473
  102. 102. Guix, S., M. Asanaka, K. Katayama, S. E. Crawford, F. H. Neill, R. L. Atmar, and M. K. Estes. 2007. Norwalk virus RNA is infectious inmammalian cells. J Virol 81:12238-12248
  103. 103. Katayama, K., G. S. Hansman, T. Oka, S. Ogawa, and N. Takeda. 2006. Investigation of norovirus replication in a human cell line. Arch Virol 151:1291-1308
  104. 104. Chan, M. C., Y. P. Wong, and W. K. Leung. 2007. Cell culture assay for human noroviruses. Emerg Infect Dis 13:1117; author reply 1117-8
  105. 105. Ward, V. K., C. J. McCormick, I. N. Clarke, O. Salim, C. E. Wobus, L. Thackray, H. W. t. Virgin, and P. R. Lambden. 2007. Recovery ofinfectious murine norovirus using pol II-driven expression of full-length cDNA. Proc Natl Acad Sci U S A104:11050-11055
  106. 106. Blacklow, N. R., J. E. Herrmann, and W. D. Cubitt. 1987. Immunobiology of Norwalk virus. Ciba Found Symp 128:144-161
  107. 107. Worobey, M., and E. C. Holmes. 1999. Evolutionary aspects ofrecombination in RNA viruses. J Gen Virol 80:2535-2543
  108. 108. Ambert-Balay, K., F. Bon, F. Le Guyader, P. Pothier, and E. Kohli. 2005. Characterization of new recombinant noroviruses. J ClinMicrobiol 43: 5179-5186
  109. 109. Bull, R. A., G. S. Hansman, L. E. Clancy, M. M. Tanaka, W. D. Rawlinson, and P. A. White. 2005. Norovirus recombination in ORF1/ORF2 overlap. Emerg Infect Dis 11:1079-1085
  110. 110. Phan, T. G., K. Kaneshi, Y. Ueda, S. Nakaya, S. Nishimura, A. Yamamoto, K. Sugita, S. Takanashi, S. Okitsu, and H. Ushijima. 2007.Genetic heterogeneity, evolution, and recombination in noroviruses. J Med Virol 79:1388-1400
  111. 111. Green, K. Y., G. Belliot, J. L. Taylor, J. Valdesuso, J. F. Lew, A. Z. Kapikian, and F. Y. Lin. 2002. A predominant role for Norwalk-like virusesas agents of epidemic gastroenteritis in Maryland nursing homes for the elderly. J Infect Dis 185:133-146
  112. 112. Jenkins, P., S. R. Greene, J. R. Archer, D. Hoang-Johnson, M. Quinn, P. Duncan, G. Johnson, B. I. Rosen, P. Smith, V. Reddy, J. Schlegelmilch, J. Pendarvis, M. Donovan, J. E. Gunn, M. A. Barry, M. Davies, J. Vinje, M.-A. Widdowson, Z. Moore, J. K. Schaffin, and J. E. Tate. 2007.Norovirus activity--United States, 2006-2007. MMWR Morb Mortal Wkly Rep 56:842-846
  113. 113. Verhoef, L., E. Duizer, H. Vennema, J. Siebenga, C. Swaan, L. Isken, M. Koopmans, K. Balay, P. Pothier, P. McKeown, G. van Dijk, P. Capdepon, and G. Delmas. 2008. Import of norovirus infections in the Netherlands and Ireland following pilgrimages to Lourdes, 2008- preliminary report. Euro Surveill 13:pii=19025
  114. 114. Steinhoff, M. C., R. G. Douglas, Jr., H. B. Greenberg, and D. R. Callahan. 1980. Bismuth subsalicylate therapy of viral gastroenteritis. Gastroenterology 78:1495-1499
  115. 115. Doultree, J. C., J. D. Druce, C. J. Birch, D. S. Bowden, and J. A. Marshall. 1999. Inactivation of feline calicivirus, a Norwalk virus surrogate. J Hosp Infect 41:51-57
  116. 116. Duizer, E., P. Bijkerk, B. Rockx, A. De Groot, F. Twisk, and M. Koopmans. 2004. Inactivation of caliciviruses. Appl Environ Microbiol 70:4538-4543
  117. 117. Barker, J., I. B. Vipond, and S. F. Bloomfield. 2004. Effects of cleaning and disinfection in reducing the spread of Norovirus contamination via environmental surfaces. J Hosp Infect 58:42-49
  118. 118. Rabenau, H. F., M. Sturmer, S. Buxbaum, A. Walczok, W. Preiser, and H.W. Doerr. 2003. Laboratory diagnosis of norovirus: which method is thebest? Intervirology 46:232-238
  119. 119. Dolin, R., R. C. Reichman, K. D. Roessner, T. S. Tralka, R. T. Schooley, W. Gary, and D. Morens. 1982. Detection by immune electron microscopy of the Snow Mountain agent of acute viral gastroenteritis. J Infect Dis 146:184-189
  120. 120. Kapikian, A. Z., R. G. Wyatt, R. Dolin, T. S. Thornhill, A. R. Kalica, and R.M. Chanock. 1972. Visualization by immune electron microscopy of a27-nm particle associated with acute infectious nonbacterial gastroenteritis. J Virol 10:1075-1081
  121. 121. Thornhill, T. S., R. G. Wyatt, A. R. Kalica, R. Dolin, R. M. Chanock, and A.Z. Kapikian. 1977. Detection by immune electron microscopy of 26- to 27-nm viruslike particles associated with two family outbreaks of gastroenteritis. J Infect Dis 135:20-27
  122. 122. Lewis, D. C. 1990. Three serotypes of Norwalk-like virus demonstrated by solid-phase immune electron microscopy. J Med Virol 30:77-81
  123. 123. Lewis, D. C., N. F. Lightfoot, and J. V. Pether. 1988. Solid-phase immune electron microscopy with human immunoglobulin M for serotyping of Norwalk-like viruses. J ClinMicrobiol 26:938-942
  124. 124. Burton-MacLeod, J. A., E. M. Kane, R. S. Beard, L. A. Hadley, R. I. Glass, and T. Ando. 2004. Evaluation and comparison of two commercial enzyme-linked immunosorbent assay kits for detection of antigenically diverse human noroviruses in stool samples. J ClinMicrobiol 42:2587-2595
  125. 125. De Bruin, E., E. Duizer, H. Vennema, and M. P. Koopmans. 2006.Diagnosis of Norovirus outbreaks by commercial ELISA or RT-PCR. J Virol Methods
  126. 126. Dimitriadis, A., L. D. Bruggink, and J. A. Marshall. 2006. Evaluation ofthe Dako IDEIA norovirus EIA assay for detection of norovirus using faecal specimens from Australian gastroenteritis outbreaks. Pathology 38:157-165
  127. 127. Rabenau, H. F., M. Sturmer, S. Buxbaum, A. Walczok, W. Preiser, and H.W. Doerr. 2003. Laboratory diagnosis of norovirus: which method is thebest? Intervirology 46:232-238
  128. 128. Richards, A. F., B. Lopman, A. Gunn, A. Curry, D. Ellis, H. Cotterill, S. Ratcliffe, M. Jenkins, H. Appleton, C. I. Gallimore, J. J. Gray, and D. W. Brown. 2003. Evaluation of a commercial ELISA for detecting Norwalk-likevirus antigen in faeces. J Clin Virol 26:109-115
  129. 129. Jiang, X., D. Y. Graham, K. Wang, and M. K. Estes. 1990. Norwalk virus genome cloning and characterisation. Science 250:1580-1583
  130. 130. Katayama, K., H. Shirato-Horikoshi, S. Kojima, T. Kageyama, T. Oka, F. Hoshino, S. Fukushi, M. Shinohara, K. Uchida, Y. Suzuki, T. Gojobori, and N. Takeda. 2002. Phylogenetic analysis of the complete genome of 18 Norwalk-like viruses. Virology 299:225-239
  131. 131. Lambden, P. R., E. O. Caul, C. R. Ashley, and I. N. Clarke. 1993.Sequence and genome organization of a human small round-structured (Norwalk-like) virus. Science 259:516-519
  132. 132. Ferreira, M. S., M. P. Xavier, T. M. Fumian, M. Victoria, S. A. Oliveira, L.H. Pena, J. P. Leite, and M. P. Miagostovich. 2008. Acutegastroenteritis cases associated with noroviruses infection in the state of Rio de Janeiro. J Med Virol 80:338-344
  133. 133. Fukuda, S., S. Takao, M. Kuwayama, Y. Shimazu, and K. Miyazaki. 2006. Rapid detection of norovirus from fecal specimens by real-time reverse transcription-loop-mediated isothermal amplification assay. J ClinMicrobiol 44:1376-1381
  134. 134. Hohne, M., and E. Schreier. 2004. Detection and characterization ofnorovirus outbreaks in Germany: application of a one-tube RT-PCR using a fluorogenic real-time detection system. J Med Virol 72:312-319
  135. 135. Kageyama, T., S. Kojima, M. Shinohara, K. Uchida, S. Fukushi, F. B. Hoshino, N. Takeda, and K. Katayama. 2003. Broadly reactive and highlysensitive assay for Norwalk-like viruses based on real-time quantitative reverse transcription-PCR. J ClinMicrobiol 41:1548-1557

Written By

Manisha Rani, Sushma Rajyalakshmi, Sunitha Pakalapaty and Nagamani Kammilli

Submitted: 14 February 2021 Reviewed: 03 May 2021 Published: 01 September 2021