You are on page 1of 17

Critical Reviews in Microbiology

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/imby20

Rotavirus-related systemic diseases: clinical


manifestation, evidence and pathogenesis

Ziqin Dian, Yi Sun, Guiqian Zhang, Ya Xu, Xin Fan, Xuemei Yang, Qiuwei Pan,
Maikel Peppelenbosch & Zhijiang Miao

To cite this article: Ziqin Dian, Yi Sun, Guiqian Zhang, Ya Xu, Xin Fan, Xuemei Yang, Qiuwei Pan,
Maikel Peppelenbosch & Zhijiang Miao (2021) Rotavirus-related systemic diseases: clinical
manifestation, evidence and pathogenesis, Critical Reviews in Microbiology, 47:5, 580-595, DOI:
10.1080/1040841X.2021.1907738

To link to this article: https://doi.org/10.1080/1040841X.2021.1907738

© 2021 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group.

Published online: 06 Apr 2021.

Submit your article to this journal

Article views: 5588

View related articles

View Crossmark data

Citing articles: 7 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=imby20
CRITICAL REVIEWS IN MICROBIOLOGY
2021, VOL. 47, NO. 5, 580–595
https://doi.org/10.1080/1040841X.2021.1907738

REVIEW ARTICLE

Rotavirus-related systemic diseases: clinical manifestation, evidence and


pathogenesis
Ziqin Diana, Yi Suna, Guiqian Zhanga, Ya Xua, Xin Fana, Xuemei Yangb, Qiuwei Panc,
Maikel Peppelenboschc and Zhijiang Miaoc
a
Department of Clinical laboratory, The First People’s Hospital of Yunnan province, Kunming, Yunnan, China; bDepartment of Clinical
laboratory, Kunming Children’s Hospital, Kunming, Yunnan, China; cDepartment of Gastroenterology and Hepatology, Erasmus MC-
University Medical Center, Rotterdam, The Netherlands

ABSTRACT ARTICLE HISTORY


Rotaviruses, double-stranded, non-enveloped RNA viruses, are a global health concern, associated Received 7 November 2020
with acute gastroenteritis and secretory-driven watery diarrhoea, especially in infants and young Revised 12 March 2021
children. Conventionally, rotavirus is primarily viewed as a pathogen for intestinal enterocytes. Accepted 16 March 2021
This notion is challenged, however, by data from patients and animal models documenting Published online 30 March
2021
extra-intestinal clinical manifestations and viral replication following rotavirus infection. In add-
ition to acute gastroenteritis, rotavirus infection has been linked to various neurological disor- KEYWORDS
ders, hepatitis and cholestasis, type 1 diabetes, respiratory illness, myocarditis, renal failure and Rotavirus; systemic disease;
thrombocytopenia. Concomitantly, molecular studies have provided insight into potential mecha- complication; clinical
nisms by which rotavirus can enter and replicate in non-enterocyte cell types and evade host manifestation; pathogenesis
immune responses. Nevertheless, it is fair to say that the extra-intestinal aspect of the rotavirus
infectious process is largely being overlooked by biomedical professionals, and there are gaps in
the understanding of mechanisms of pathogenesis. Thus with the aim of increasing public and
professional awareness we here provide a description of our current understanding of rotavirus-
related extra-intestinal clinical manifestations and associated molecular pathogenesis. Further
understanding of the processes involved should prove exceedingly useful for future diagnosis,
treatment and prevention of rotavirus-associated disease.

Introduction vaccines) has raised hopes that the virus can eventually
be constrained (Burnett et al., 2020; Simsek et al. 2021),
Rotaviruses (RVs) are non-enveloped double-stranded
clinical management of rotavirus-associated disease is
RNA (dsRNA) viruses that belong to the Reoviridae fam-
complicated by the absence of specific antiviral therapy
ily (Trask et al. 2012). Their genome comprises 11 seg-
(Crawford et al. 2017; Banyai et al. 2018). The canonical
ments of dsRNA encoding six viral structural proteins
view is that rotavirus infection is restricted to the intes-
(VP1 to VP4, VP6 and VP7) and six non-structural pro-
tine, but this notion has become challenged (Kraft
teins (NSP1 to NSP6) (Trask et al. 2012) (Figure 1). Up to mez-Rial
1958; Blutt and Conner 2007; Ramig 2007; Go
now, ten rotavirus species (A to J) have been identified. et al. 2019). During the acute phase of rotavirus infec-
Four of these (A, B, C, and H) cause human disease, tion, both antigenemia and viremia are readily detected
with the rotavirus A species [featuring strains Wa, DS-1 in both experimental animals and children, demonstrat-
and AU-1 (Matthijnssens et al. 2008)] being the most ing that the virus can reach a multitude of potential
significant in this respect (Crawford et al. 2017). Human host compartments (Blutt et al. 2003; 2007; Ramig 2007;
rotaviruses are important enteric pathogen, infecting Gomez-Rial et al. 2019). In apparent agreement, the
the epithelial compartment of the small intestine, and presence of either rotaviral RNA, protein or infectious
are a major cause of gastroenteritis in infants and particles during human or animal infection, has been
young children (Crawford et al. 2017; Banyai et al. documented in brain, liver, spleen, lung, heart, kidney,
2018). Although the introduction of anti-rotavirus vac- pancreas, thymus, adrenal gland, bladder testis and
cines (Rotateq and Rotarix, two live-attenuated oral immune cells (Kraft 1958; Crawford et al. 2006; Blutt

CONTACT Zhijiang Miao miaozhijiang@yeah.net Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center,
Rotterdam, The Netherlands
ß 2021 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
CRITICAL REVIEWS IN MICROBIOLOGY 581

Figure 1. Rotavirus infection-induced acute gastroenteritis. The rotavirus particle contains 11 segments of dsRNA as its genome, and the
capsid consists of three layers. The intestinal epithelium is characterized by the villi and crypt. Mature enterocytes locate at the top and
middle of villi allow SGLT-1-mediated absorption of water and salt. The ECs locate at the middle of villi are highly specialized cells with
endocrine function. Stem cells and Paneth cells locate in the crypt are functionally responsible for differentiation and secretion, respect-
ively. Thus, under normal conditions, the intact intestinal membrane barrier can keep homeostasis (absorption and secretion). While rota-
virus infection in mature enterocytes causes absorptive enterocyte death and secretory Paneth cell replacement, leading to shortening
and blunting of villi, and increasing infiltration of inflammatory cells in the lamina propria. The NSP4 stimulates the epithelium and ECs
to increase intracellular calcium, which further induces the secretion of 5-HT from ECs through a calcium-dependent manner. The
secreted 5-HT activates EGCs to increase release of GDNF which induces the expression of tight junction-associated protein ZO-1 in
infected and bystander cells. Thus neurotrophic factors and 5-HT can protect the intestinal barrier function during rotavirus insult.
Moreover, the number of enterocytes remaining following infection is functionally sufficient to allow SGLT-1-mediated absorption of
water and salt. However, 5-HT can also activate the intrinsic primary afferent nerves of the myenteric plexus. Such activation activates
the nerves of the submucosal plexus and leads to increase of intestinal motility which can be attenuated by an opioid-receptor-antagon-
ist (Loperamide). The activation of submucosal nerves further induce VIP release from nerve endings adjacent to crypt cells. Then, VIP
induces crypt cells to secret NaCl and water into the intestinal lumen by increasing cellular cAMP levels, which ultimately leading to
secretory diarrhoea. During these processes, the stimulated afferent vagal signalling activates the nausea and vomiting centre of the
brain to generate efferent vagal signalling which then results in vomiting by stimulating a nerve-muscle vomiting reflex in the stomach.
This event can be attenuated by 5-HT receptor antagonists (Ondansetron). Finally, intestinal rotavirus infection results in secretory-driven
watery diarrhoea accompanying by vomiting. dsRNA: double-stranded RNA; VPs: rotavirus structural proteins (VP4 is cleaved into VP5
and VP8); NSP4: rotavirus non-structural protein; ENS: enteric nervous system; NaCl: sodium chloride; Ca2þ: calcium ions; SGLT-1:
sodium/glucose cotransporter 1; 5-HT: 5-hydroxytryptamine (serotonin); VIP: vasoactive intestinal peptide; ECs: enterochromaffin cells;
EGCs: enteric glial cells; GDNF: glial cell-derived neurotrophic factor; ZO-1: zona occludens 1; cAMP: cyclic adenosine monophosphate.

and Conner 2007; Ramig 2007). Accordingly, extra-intes- infection, and the pathological mechanisms driving
tinal complications during rotavirus therapy have been extra-intestinal rotavirus-associated disease.
widely reported (Mossel and Ramig 2003; Ramig 2007;
Gomez-Rial et al. 2019). Nevertheless, it is fair to say
that the extra-intestinal aspect of rotavirus infection is Rotavirus-induced gastrointestinal disease
largely ignored by the biomedical community. This
Acute gastroenteritis
prompted us to assess the evidence for non-intestinal
diseases associated with rotavirus infection, the pos- Before considering potential extra-intestinal rotavirus
sible size of the clinical problem associated with such infection, it is important to review the conventional
582 Z. DIAN ET AL.

Figure 2. Rotavirus-related systemic diseases and proposed mechanisms. The primary infection of rotavirus in the intestine leads
to acute gastroenteritis. However, the tropism and pathogenetic role of rotavirus infection is not only limited to the gastrointes-
tinal tract. The extra-intestinal dissemination and infection of rotavirus in non-intestinal tissues such as the spleen, liver, heart,
lung, kidney, testis, bladder, adrenal gland, pancreas and brain have been confirmed, which may sometimes lead to additional
extra-intestinal complications. Here we present rotavirus-induced acute gastroenteritis and the frequently reported complications
related to rotavirus extra-intestinal spread and propose the potential mechanisms. RV: rotavirus; VP7: viral structural protein;
NSP4: non-structural protein; ENS: enteric nervous system; NO: nitric oxide; IFN-c: interferon gamma.

presentation of the disease, acute gastroenteritis (36 G genotypes and 51 P genotypes have been classi-
(Figures 1 and 2). About 258 million diarrhoea episodes fied in species A rotavirus binary serotype system based
and 129 000–200 000 associated deaths per year are on the two outer capsid proteins VP4 and VP7
attributed to rotavirus infection, making it the leading (Crawford et al. 2017; Simsek et al. 2021), respectively),
cause of gastroenteritis in young children (Tate et al. even if subsequent infections are less severe than first
2016; Crawford et al. 2017; Banyai et al. 2018; Hartman infections (Velazquez et al. 1996; Mormile 2016). By the
et al. 2019). As the vast majority of cases occurs in age of five, nearly every child in the world will have
developing countries, rotavirus infection is seen as a been infected with rotavirus at least once (Tate
global health concern, but also in developing countries et al. 2012).
rotavirus associated gastroenteritis remains a major The primary outcome of rotavirus infection is acute
cause of hospitalization in children (Crawford et al. gastroenteritis. Its clinical features range from subclin-
2017). Following the introduction of anti-rotavirus vac- ical infection and mild watery diarrhoea to severe diar-
cination, hospitalizations and mortality has shown a rhoea with vomiting and fever that can result in
25–55% decline (Crawford et al. 2017; Troeger et al. dehydration with shock, electrolyte imbalance, and
2018; Aliabadi et al. 2019; Burke et al. 2019; Burnett death (Hjelt et al. 1985; Haffejee and Moosa 1990;
et al., 2020). Nonetheless, 8–10% of all children with Leung et al. 2005; Parashar et al. 2013; Esona and
gastroenteritis and 35–40% of hospitalized children are Gautam 2015) (Table 1). The importance of antiviral
positive for rotavirus (Banyai et al. 2018). Compounding immunity in controlling disease is highlighted by the
the clinical problem is that children often suffer from observation that the symptomology is more severe in
multiple episodes of rotavirus infection as multiple children with more immature immune systems
serotypes are present in the population and both vac- (Crawford et al. 2017; Banyai et al. 2018), with disease
cine and prior disease fail to provide full immunity being the worst in the youngest children, where as
Table 1. Clinical manifestation and evidence summary of rotavirus-related systemic diseases.
Evidence
Systemic disease Clinical manifestation Human Animal model
Acute gastroenteritis 1. major AG symptoms including watery diarrhoea, 1. RV infection is the leading cause of active RV infection in animal models induces morphological,
vomiting, low-grade fever, dehydration and electrolyte gastroenteritis among young children (Tate et al. 2016; anatomical and functional changes in the small intestine
imbalance (Hjelt et al. 1985; Haffejee and Moosa 1990; Crawford et al. 2017; Banyai et al. 2018; Troeger (Hagbom et al. 2012; 2016; 2016; Crawford et al. 2017):
Leung et al. 2005; Parashar et al. 2013; Esona and et al. 2018); 1. piglets (thinning of the intestinal wall, completely
Gautam 2015); 2. RV induces more severe symptoms in the youngest eroded intestinal villi, conversion of columnar villus
2. other associated symptoms including: low children (Leung et al. 2005; Parashar et al. 2013; epithelium to cuboidal) (Shepherd et al. 1979);
inflammation, shock, coma, abdominal cramps, nausea, Hartman et al. 2019); 2. calves (blunting of villus, conversion of columnar villus
malaise, lethargy, pharyngeal erythema, rhinitis and 3. RV hospitalizations and deaths decreased by 25–55% epithelium to cuboidal) (Reynolds et al. 1985);
palpable cervical lymph nodes (Hjelt et al. 1985; following vaccination (Crawford et al. 2017; Troeger 3. lambs (less severe histopathological changes and mild
Haffejee and Moosa 1990; Leung et al. 2005; Parashar et al. 2018; Aliabadi et al. 2019; Burke et al. 2019; clinical disease) (Greenberg et al. 1994);
et al. 2013; Esona and Gautam 2015) Burnett et al., 2020); 4. mice (remarkable swollen and vacuolated enterocytes
4. histological and pathological examinations indicate that but limited villus blunting) (Dharakul et al. 1988;
RV infection and replication in the intestinal epithelium Osborne et al. 1988; Ward et al. 1990; Lundgren and
(mature enterocytes and ECs) leading to significant Svensson 2001; Hagbom et al. 2016)
structural and functional alterations (severe mucosal
damage including shortening and blunting of villi and
increased immune infiltration in the lamina propria)
(Hagbom et al. 2012; 2016; Crawford et al. 2017)
Neurological diseases 1. seizures (diarrhoea, vomiting, fever, febrile, convulsions, 1. more CNS diseases (2–8% incidence of seizures 1. isolation of virus particles by cell culture from the brain
generalized bleeding) (Lloyd et al. 2010; Yeom omara et al. 2002; Lloyd et al. 2010; Pardo-
(Iturriza-G of porcine RV-inoculated neonatal pigs (Janke et al.
et al. 2019); Seco et al. 2015 )) are reported in RV-gastroenteritis 1988; Shaw et al. 1989);
2. meningitis (diarrhoea, vomiting, irritable, afebrile, children (Wong et al. 1984; Ushijima et al. 1986; 2. infectious virus is detected in the brain of heterologous
convulsions) (Wong et al. 1984); Nishimura et al. 1993; Lynch et al. 2001; Fukuda et al. RRV (MMU-18006) or homologous murine strain (EDIM-
3. encephalopathy (diarrhoea, fever, dehydration, lethargy, 2009; Takanashi et al. 2010; Karampatsas et al. 2015; 5099) inoculated mice (Kraft 1958; Shaw et al. 1989;
convulsions, coma) (Lynch et al. 2001; Wong 2001; Laizane et al. 2019); higher positivity of RV in children Czech-Schmidt et al. 2001)
Karampatsas et al. 2015); with seizures or convulsions (Herrmann et al. 1993;
4. encephalitis (diarrhoea, vomiting, fever, dehydration, Wong 2001; Yeom et al. 2019);
convulsions, afebrile) (Ushijima et al. 1986) 2. rotavirus vaccination has resulted in reduced incidence
of paediatric seizures in the USA and Spain (Payne
et al. 2014; Pardo-Seco et al. 2015; Burke et al. 2018);
3. RV particles, antigens, antibodies and RNA are found in
CSF (Wong et al. 1984; Keidan et al. 1992; Pager et al.
2000; Lynch et al. 2001; Iturriza-Gomara et al. 2002); RV
infection and replication (viral VP6, NSP1, NSP3, NSP4,
and RNA) in brain tissue (Nakano et al. 2011);
4. infection and replication (structural protein VP7 and
non-structural protein NSP4) of RRV in neurons in vitro
(Weclewicz et al. 1998)
Hepatobiliary diseases 1. AG symptoms (diarrhoea, vomiting, fever, dehydration) 1. abnormal liver function results from RV infection 1. RV infection (viral structural protein VP7) and active
(Gilger et al. 1992); (Kovacs et al. 1986; Teitelbaum and Daghistani 2007); replication (viral RNA, NSP4 and virus particles) in
2. liver dysfunction (elevated serum sodium and liver 2. RV infection (viral antigen) and replication (viral VP6, murine biliary tracts and livers (Uhnoo et al. 1990;
transaminases such as AST, ALT and GGT, hepatitis, NSP1, NSP3, NSP4, and RNA) in liver tissue (Gilger et al. Riepenhoff-Talty et al. 1993; Shivakumar et al. 2004);
liver steatosis, cirrhosis) (Gilger et al. 1992; Teitelbaum 1992; Riepenhoff-Talty et al. 1996; Nakano et al. 2011); 2. hepatobiliary-tropic RVs strain-specifically induce high
and Daghistani 2007); 3. continuous RV infection (viral RNA, VP4, VP6, and incidence of hepatitis, bile duct obstruction and
3. biliary symptoms (extrahepatic biliary atresia, infectious virus) in human liver organoids results in mortality, as well as human BA-like symptoms and
choledochal cyst, cholestasis, fibrosis) (Gilger et al. direct cytopathogenesis (Chen et al. 2020) hispathological changes in murine models (Uhnoo et al.
CRITICAL REVIEWS IN MICROBIOLOGY

1992; Riepenhoff-Talty et al. 1996; Bezerra et al. 2002) 1990; Riepenhoff-Talty et al. 1993; Shivakumar
et al. 2004)
(continued)
583
Table 1. Continued.
584

Evidence
Systemic disease Clinical manifestation Human Animal model
Type 1 diabetes 1. AG symptoms (diarrhoea, vomiting, fever, dehydration) 1. more acute pancreatitis cases with transient abnormal 1. replication of human and monkey RV (infectious virus)
(Parri et al. 2010; Basturk et al. 2017); size and function of pancreas are reported in RV- in monkey islet cells (Coulson et al. 2002);
2. acute pancreatitis (abdominal pain, elevated serum gastroenteritis children (Nigro 1991; Parri et al. 2010; 2. monkey RRV infection increases insulitis development
transaminases such as pancreatic amylase and lipase, Basturk et al. 2017); and accelerates diabetes in NOD mouse (Graham
Z. DIAN ET AL.

transient moderately enlarged oedematous pancreas) 2. the incidence of T1D decreased in RV-vaccinated et al. 2008);
(Parri et al. 2010; Basturk et al. 2017) children younger 4 years in the USA and Australia 3. RRV antigen and infectious virus are found in the
(Perrett et al. 2019; Rogers et al. 2019); pancreas of NOD mice and rats (Crawford et al. 2006;
3. RV infection (viral antigen and RNA) in the pancreas Graham et al. 2007);
(Lynch et al. 2003; Ramig 2007) 4. RRV infection induces transient pancreatic involution
and hyperglycaemia in weanling mice (Honeyman
et al. 2014)
Respiratory illness 1. AG symptoms (diarrhoea, vomiting, fever, dehydration) 1. respiratory illness is the major extra-intestinal symptom 1. HRV (Wa) induces viremia, antigenemia and nasal virus
(Santosham et al. 1983; Fragoso et al. 1986; Zheng that preceded or concurrently occurred with rotavirus- shedding in piglets (Azevedo et al. 2005);
et al. 1991; Grech et al. 2001); induced gastroenteritis in children (Fragoso et al. 1986; 2. respiratory infection (viral antigen in pnueumocytes
2. respiratory symptoms (cough, mild respiratory distress, Sharma et al. 2002; Grimprel et al. 2008); and lymphoid cells of lungs, viral RNA in nasal fluid
rhinitis, rhinorrhea, nasal discharge, congestion, 2. RV infection (viral antigen) in upper respiratory tract secretions and lungs) of GARV (KJ9-1) induces
pneumonia, otitis media (Goldwater et al. 1979; Zhaori (oropharyngeal aspirates (Zheng et al. 1991) and pneumonia and increases infiltration-mediated
et al. 1991; Santosham et al. 1983; Gurwith et al. 1981) nasopharyngeal secretions (Fragoso et al. 1986)) and cellularity in calves (Kim et al. 2011);
lower respiratory tract (tracheal aspirate (Zhaori 3. RRV infection and replication (viral antigen, structural
et al. 1991)); and non-structural protein and infectious virus) in the
3. RV infection (viral RNA) in lung tissue (Lynch lower respiratory tract (macrophages and pneumocytes)
et al. 2003); induce parenchymas inflammation and respiratory
4. RV as a part of virome frequently identified in secretion of virus in rats (Crawford et al. 2006);
respiratory tract samples (Taboada et al. 2014; Madi 4. homologous (murine rotavirus) and heterologous RRV
et al. 2018; Thi Kha Tu et al. 2020) infection and replication (viral RNA, NSP4 and virus
particles) in the lung tissue of newborn mice (Fenaux
et al. 2006); intranasal but not oral inoculation of RRV
induces immune protection against challenge in mice
(Coffin and Clark 2001)
Heart diseases 1. elevated level of CK-MB (Zheng et al. 2017); 1. infection and replication (viral VP6, NSP1, NSP3, NSP4, 1. viral antigen is detected in the heart of RRV-infected
2. myocyte necrosis (Cioc and Nuovo 2002); and RNA) in heart tissue possibly induce hypertrophic rats (Crawford et al. 2006)
3. myocarditis (Grech et al. 2001; Cioc and Nuovo 2002); cardiomyopathy and myocardial damage (Cioc and
4. hypertrophic cardiomyopathy with focal disorganized Nuovo 2002; Lynch et al. 2003; Nakano et al. 2011;
myocardial architecture (Nakano et al. 2011); Zheng et al. 2017)
Renal failure 1. AG symptoms (diarrhoea, vomiting, fever, dehydration) 1. diarrhoea-induced dehydration is associated with pre- 1. infectious RV (EDIM) is found in the kidney of normal
(Fujinaga et al. 2005; Ashida et al. 2012; Kira and post-renal failure (Ashida et al. 2012; Kira et al. mice (Kraft 1958);
et al. 2016); 2016; Tsukida et al. 2018); 2. RV-A induces cellular degeneration and immune
2. renal and urogenital symptoms (acute obstructive 2. RV infection (viral antigen and RNA) and replication infiltration in the kidney of suckling mice (Kashyap
uropathy-ureteral stones (Kira et al. 2016; Ashida et al. (viral non-structural protein) in kidney tissue (Gilger et al. 2018);
2012; Fujinaga et al. 2005), oliguria (Fujinaga et al. et al. 1992; Lynch et al. 2003) 3. homologous (murine rotavirus) and/or heterologous
2005), hypovolemia (Fujinaga et al. 2005; Shirasu et al. RRV infection and replication (viral antigen, NSP4, RNA
2015), aciduria (Tsukida et al. 2018), azotaemia and virus particles) in the kidney of newborn mice and
(Fujinaga et al. 2005), hyperuricaemia and rats (Crawford et al. 2006; Fenaux et al. 2006);
hyponatremia (Ashida et al. 2012; Tsukida et al. 2018)) 4. monkey kidney cells are highly susceptible for RV
infection in vitro (Ward et al. 1984)
AG: acute gastroenteritis; RV-A: species A rotavirus; RRV: rhesus rotavirus; HRV: human rotavirus; GARV: bovine rotavirus; ECs: enterochromaffin cells; NSP1/3/4: rotavirus non-structural proteins; VP6/7: rotavirus
structural viral proteins; ENS: enteric nervous system; CSF: cerebrospinal fluid; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gammaglutamyl transferase; BA: biliary atresia; T1D: type 1 dia-
betes; NOD: Non-Obese Diabetic; CK-MB: Creatine Kinase-MB, an indication for myocardial damage.
CRITICAL REVIEWS IN MICROBIOLOGY 585

severe dehydration, electrolyte disturbances and emesis that diarrhoea is an active process and not the result
are not uncommon following rotavirus infection (Leung from loss of absorptive function is supported by the
et al. 2005; Parashar et al. 2013; Hartman et al. 2019). In success of rehydration in rotavirus-induced diarrhoea
both humans and animal models (piglets, calves, lambs (Rautanen et al. 1997; Hagbom et al. 2012) or the obser-
and mice) rotavirus infection may provoke histological vation that zinc-mediated inhibition of cAMP-depend-
and functional alterations changes in the small intes- ent chloride secretion counteracts rotavirus-induced
tine, which include intestinal wall hypotrophy, loss of diarrhoea (Baqui et al. 2002; Dalgic et al. 2011; Hagbom
epithelial microvilli, villous atrophy and intraepithelial et al. 2012). Altogether, these studies indicate that rota-
lymphocytosis, and loss of disaccharidase activity virus-induced watery diarrhoea is of a secretory nature.
(Dharakul et al. 1988; Haffejee and Moosa 1990; At the molecular level, rotavirus replication in enter-
Crawford et al. 2017) (Figure 1). Examination of intes- ocytes and ECs results in expression and release of the
tinal biopsies from rotavirus-infected children, however, rotaviral non-structural protein 4 (NSP4), which is a
show poor correlation between histological abnormal- major viral enterotoxin due to its virporin activity and
ities and diarrhoea (Ko €hler et al. 1990; Ramig 2004; subsequently provoked increased levels of intracellular
Hagbom et al. 2016). Interestingly, rotavirus-induced Ca2þ (Van Den Brink et al. 1999; Hagbom et al. 2012;
anatomical changes were linked to the slightly Hyser et al. 2012). This provokes release of 5-HT by the
increased intussusception (a serious condition where an EC compartment, in turn activating the primary afferent
intestinal segment folds into an adjacent part of the nerves of the myenteric plexus of the enteric nervous
intestine) observed primarily in application of the first- system (ENS) (Hagbom et al. 2011; 2012; 2016; Crawford
generation vaccine, but more substantial evidence has et al. 2017). The resulting neurotransmission in turn is
demonstrated that neither natural rotavirus infection responsible for increased intestinal motility as well as
nor vaccination are significantly associated with intus- the release of vasoactive intestinal peptide (VIP) from
susception (Robinson et al. 2004; Lu et al. 2019; Burnett synaptic terminals located adjacent to crypt cells
et al., 2020). In general, rotavirus-induced gastrointes- (Hagbom et al. 2012; 2016; Crawford et al. 2017). As a
tinal disease manifests as acute gastroenteritis is associ- hormone, VIP stimulates secretion of sodium, chloride,
ated with watery diarrhoea, low-grade fever, vomiting, potassium and indirectly of water into the small intes-
low inflammation, and intestinal wall damage (Hagbom tine diarrhoea through increased cAMP production
et al. 2020). (Hagbom et al. 2012; 2016; Crawford et al. 2017).
The pathogenesis of rotavirus gastroenteritis is multi- During this process the afferent component of the ner-
factorial (Figures 1 and 2). Conventionally, gastroenter- vus vagus becomes stimulated, and this provokes vom-
itis is viewed as the consequence of rotavirus infecting iting through a reflex relay to the stomach (Hagbom
mature enterocyte and enterochromaffin cells (ECs, one et al. 2011; 2012; 2016; Crawford et al. 2017), again 5-
type of enteroendocrine cell) at the tip and the body of HT being a prominent neurotransmitter in thus process
intestinal villi, causing loss of enterocytes and second- as evident by the attenuation of emesis following the
ary expansion of the Paneth cell compartment. use of receptor antagonist (Ondansetron) (Yamashiro
Consequently, secretion of digestive enzymes and dis- et al. 1989; Kordasti et al. 2004; Hagbom et al. 2012;
accharidase is impaired, in turn increasing osmolarity of Crawford et al. 2017). Apart from provoking 5-HT
the intestinal lumen and thus provoking diarrhoea and release, NSP4 also stimulates production of nitric oxide
malabsorption (Colbere-Garapin et al. 2007; Hagbom (NO) release and this also modulates ENS signalling
et al. 2016). This concept has been challenged by obser- (Hagbom et al. 2012). Thus the notion that rotavirus-
vation in experimental rodents that show that the num- provoked diarrhoea is secretory in nature can be mech-
ber of enterocytes remaining following infection is anistically substantiated.
functionally sufficient to allow sodium/glucose cotrans- Apart from diarrhoea, rotavirus infection provokes
porter (SGLT-1)-mediated absorption of water and salt systemic activation of immunity as for instance evident
(Hagbom et al. 2011; 2012; 2016). Thus it has been pro- from the fever that often accompanies disease
posed that the diarrhoea is an active defense mechan- (Figure 2). Clinical studies show that rotavirus gastro-
ism controlled by neurotrophic factors and the enteritis is associated with increased levels of the pro-
serotonin 5-hydroxytryptamine (5-HT) (Istrate et al. inflammatory cytokines interleukin-1b (IL-1b), IL-6, IL-8
2014; Hagbom et al. 2020), in which the enteric nervous and interferon gamma (IFN-c) (Mormile 2016; Crawford
system is activated as Loperamide (an opioid receptor et al. 2017), which have been linked to fever and mal-
agonist) appears to attenuate rotavirus-induced diar- aise. Much of the tissue damage observed in rotavirus
rhoea (Li et al. 2007; Hagbom et al. 2012). The notion gastroenteritis may be secondary to immune cell
586 Z. DIAN ET AL.

activation by such cytokines and not as much from dir- to 8% (Iturriza-Go mara et al. 2002; Lloyd et al. 2010;
ect viral cytopathic effects (Mormile 2016). Especially Pardo-Seco et al. 2015).
serum IL-8 appears a distinctive biomarker for clinical Supporting a neurological substrate for rotavirus rep-
rotavirus-mediated disease (Chen et al. 2014; Mormile lication is that rotavirus particles, antigens, antibodies
2016). IFN-c production may well be instrumental in and RNA have been detected in cerebrospinal fluid
inducing clearance of virus as it is a canonical mediator (CSF) (Wong et al. 1984; Keidan et al. 1992; Pager et al.
of cell autonomous and T cell-mediated immune 2000; Lynch et al. 2001; Iturriza-Go mara et al. 2002).
responses (Mormile 2016). Indeed, adult life is charac- Indeed, study showed that neurons can sustain infec-
terized by stronger IFN-c production and subsequent T tion with rhesus rotavirus (RRV), NSP4 protein being
cells activation, as compared to infants, which fits well detectable in dendritic processes (Weclewicz et al.
with the decreased susceptibility of adults to rotavirus 1998). In vivo data in animals are more ambiguous but
infection as compared to children (Mormile 2016). brain cell cultures from pigs and mice inoculated with
While we understand the interaction of rotavirus with heterologous RRV (MMU-18006, species A, serotype G3)
mucosal immunity fairly well, potential interactions out- or homologous murine strain (EDIM-5099) respectively,
side the intestine are, however, less well understood. showed productive infection (Kraft 1958; Janke et al.
1988; Czech-Schmidt et al. 2001). Accordingly, in the
USA and Spain, rotavirus vaccination has resulted in
Rotavirus-related extra-intestinal reduced incidence of paediatric seizures (Payne et al.
complications 2014; Pardo-Seco et al. 2015; Burke et al. 2018),
Central nervous system diseases although later studies failed to detect such an effect
(Biggart et al. 2018; Orrico-Sanchez et al. 2018; Go
mez-
Extra-intestinal manifestations reported following
Rial et al. 2019). Hence, the overall contribution of rota-
paediatric rotavirus infection in infants and children
virus neuronal infection as a trigger for seizures
most commonly involve the central nervous system
remains unclear.
(CNS) (Ramig 2007; Go mez-Rial et al. 2019). Following
Four hypotheses have been brought forward for
early reports from East Asia, also Western countries explaining neurological manifestations of rotavirus
reported cases apparently rotavirus-related cases of infection (Figure 2). The presence of rotavirus in the
neurological disorders, such as seizures, meningitis, CSF, which is mostly documented by reverse transcrip-
encephalopathy, encephalitis, and cerebellitis (Wong tion-polymerase chain reaction (RT-PCR) data (presence
et al. 1984; Ushijima et al. 1986; Nishimura et al. 1993; of viral genome), and the infectious capacity of neurons
Lynch et al. 2001; Iturriza-Gomara et al. 2002; Fukuda to sustain rotavirus infection points towards direct rota-
et al. 2009; Takanashi et al. 2010; Karampatsas et al. virus-mediated neuronal cytotoxicity (Kraft 1958; Wong
2015; Laizane et al. 2019). The severity of the disease et al. 1984; Keidan et al. 1992; Czech-Schmidt et al.
varies widely from well-tolerated seizures and mild 2001; Lynch et al. 2001; Iturriza-Go mara et al. 2002;
encephalitis/encephalopathy concomitant with cerebel- Ramig 2007; Nakano et al. 2011), but it also possible
litis to death, corresponding to neurological manifesta- that the rotavirus presence signals the presence of
tions ranging from benign convulsions and reversible other pathogens that mediate the actual neurotoxicity
white matter injury to fatal Reye’s syndrome (Lynch (e.g. zika virus, influenza virus, or adenovirus)
et al. 2001; Karampatsas et al. 2015) (Table 1). Seizures (Karampatsas et al. 2015). A further alternative is that
presenting as benign convulsions are the most com- the neuronal damage is the consequence of circulating
monly reported. Neurological manifestations of rota- NSP4, possibly in conjunction with NO generation
virus may be underestimated as rotavirus stool levels in (DiFazio et al. 2007). NSP4 can act as a virporin and
newborn infants presenting convulsion were consider- thus provoke Ca2þ-mediated excitotoxicity (Hagbom
ably higher than the healthy control group or rotavirus- et al. 2012; 2016; Crawford et al. 2017), whereas NO
negative control groups, whereas co-presentation of production (which is a reactive free radical (Goldwater
convulsion and acute gastroenteritis in children is et al. 2001)) may further provoke neuronal damage.
higher as expected when compared to other aetiologies Interestingly, a significant elevation of NO was observed
(Herrmann et al. 1993; Wong 2001; Yeom et al. 2019). in the spinal fluid of patients suffering from rotavirus-
Rotavirus infection represents an independent risk fac- induced seizures (DiFazio et al. 2007). Finally, rotavirus-
tor for neonatal seizure increasing risk by four times activated host immune responses may provoke
(Yeom et al. 2019). Strikingly, in paediatric rotavirus neurological damage as well. A pilot study showed that
gastroenteritis, the incidence of seizures ranges from 2 the expression of monocyte chemoattractant protein-1
CRITICAL REVIEWS IN MICROBIOLOGY 587

and macrophage inflammatory protein-1b was higher (over 80%) and mortality (over 90%) due to obstruction
in rotavirus-infected newborns with white matter injury of extrahepatic bile ducts was observed in neonatal
than in newborns without such damage or in healthy mice following inoculation of the animals with RRV
controls (Yeom et al. 2019). Another small study (MMU-18006) and with an apparent tropism to the bile
reported elevated levels of IL-6, IL-10 and IFN-c and duct (Shivakumar et al. 2004; Ramig 2007).
immune cells in the CSF of rotavirus-infected patients Furthermore, in neonatal BALB/c mice infected with
with mild encephalopathy (Miyata et al. 2012; two hepatobiliary-tropic RV strains (HCR-3 and RRV, spe-
Karampatsas et al. 2015). Thus the neurological mani- cies A), active virus replication was detected in the bil-
festations of rotavirus infection may well relate to mul- iary tract and liver as early as two days after inoculation
tiple aetiological mechanisms. (Riepenhoff-Talty et al. 1993), and later jaundice, acholic
stool and other hallmarks of BA were present as well
(Riepenhoff-Talty et al. 1993; Petersen et al. 1997;
Hepatobiliary diseases
Ramig 2007). In apparent agreement, transcriptomic
An increasing body of evidence relates rotavirus to analysis of BA infants finds evidence of ongoing inflam-
acute and chronic liver disease and to cholestatic dis- mation and activation of IFN-c-mediated antiviral
ease (Ramig 2007). Rotavirus-induced acute and chronic immunity (Bezerra et al. 2002), whereas in experimental
hepatitis usually results in liver dysfunction, and rota- animals adoptive transfer of B cells into rotavirus-
virus-induced cholestasis often leads to obstructive infected B cell-deficient mice was essential for biliary
jaundice. Biliary atresia (BA), characterized by perinatal damage (Bednarek et al. 2018). Thus a role for rotavirus
progressive obliteration of the extrahepatic bile duct, is infection in the development of BA either through dir-
one of the most frequent causes of neonatal cholestasis ect cytopathy or indirect through immunological dam-
and will progress as biliary cirrhosis and end-stage liver age have been proposed.
disease requiring liver transplantation (Shivakumar An underappreciated aspect with regard to the dis-
et al. 2004; Allen et al. 2007). A diagnosis of acute hepa- cussion on the role of rotavirus in BA development is
titis, prompted by elevated alanine aminotransferase that biliary and hepatic manifestations of the infection
(ALT), aspartate aminotransferase (AST) and gammaglu- appear highly strain and context dependent. Whereas
tamyl transferase (GGT) levels, in children with rota- inoculation of experimental rodents with the bovine
virus-gastroenteritis has been reported (Table 1) and (WC3), human (Wa) strains or the murine strain EDIM-
associated with the presence of hepatic positivity for 5099 provoked no sign of hepatobiliary disease (Uhnoo
rotaviral non-structural proteins in immunostaining et al. 1990; Allen et al. 2007), in the same model the
(Gilger et al. 1992; Teitelbaum and Daghistani 2007; simian rotavirus strain (SA11-SM) produces hepatocyte
Nakano et al. 2011). Especially, in immunocompromised infection but not cholangiocyte infection and the SA11-
or immunodeficient children with rotavirus infection, FM strain (identical to SA11-SM except for the presence
liver involvement is often obvious (Gilger et al. 1992; of the bovine rotavirus VP4 protein gene) targets biliary
Ramig 2007). A significant association between rota- epithelial cells causing bile duct obstruction and high
virus and cholestatic disease was also reported, in mortality (Figure 2). Furthermore, disease outcome in
which species C rotavirus RNA was detected in liver rotavirus-infected mouse models are highly age-
biopsy samples of extrahepatic BA (EHBA) infants dependent: whereas rotaviral infection of neonatal mice
(Riepenhoff-Talty et al. 1996). Recently it was shown (<2 days of age) results in hepatobiliary inflammation
that human liver organoids are highly susceptible to and obstruction, such infection in adult mice is asymp-
simian rotavirus (SA11, species A, serotype G3) infection tomatic (Riepenhoff-Talty et al. 1993; Shivakumar et al.
and sustain the entire life cycle of the virus (Chen et al. 2004; Mack et al. 2006; Hertel and Estes 2012).
2020). In the liver of severe combined immunodefi- Experiments in which effect of rotavirus infection in foe-
ciency (SCID) mice and immunocompetent BALB/c tal and adult liver organoids are contrasted may shed
mice, the heterologous RRV (MMU-18006) provoked light on to which extent the outcome of cholangiocyte
hepatitis, the immunocompetent BALB/c mice more infection are host age mediated (Saxena et al. 2016;
resistant to this effect, in line with human data (Uhnoo Ramani et al. 2018). Notably, immunization against rota-
et al. 1990; Ramig 2007). Thus host immunity appears virus in female mice protects newborn mice from devel-
to be a critical factor with respect to the development oping rotavirus-induced cholestasis and EHBA (Czech-
of rotavirus-induced hepatobiliary diseases (Uhnoo Schmidt et al. 2001). It is thus well possible that rota-
et al. 1990; Shivakumar et al. 2004; Mack et al. 2006) virus vaccination of pregnant women will substantially
(Figure 2). Importantly, high incidence of cholestasis reduce the incidence of neonatal BA.
588 Z. DIAN ET AL.

Type 1 diabetes pain and elevated serum pancreatic enzymes, following


an earlier rotavirus gastroenteritis have been reported
Type 1 diabetes (T1D) is an autoimmune disease char-
in children (Nigro 1991; Parri et al. 2010; Basturk et al.
acterized by the destruction of insulin-producing pan-
2017) (Table 1). The role for rotavirus in the pancreatitis
creatic b cells, and is frequently and also increasingly
is supported by the presence of stool rotavirus particles
diagnosed in children in the USA (Rogers et al. 2017;
and the absence of other pathogens therein. Also, in
Burke et al. 2020). Although the aetiology of T1D is not
two cases of pancreatitis-associated paediatric fatalities,
fully understood yet, links with different pathogens are
rotavirus antigen and RNA was detected in the pan-
well-recognized (Filippi and von Herrath 2008; Rewers
creas (Lynch et al. 2003; Ramig 2007). Hence, paediatri-
and Ludvigsson 2016; Burke et al. 2020). Rotavirus has
been proposed as an etiologic trigger for T1D (Table 1), cians should be aware of the possibility that severe
but also for other autoimmune disease such as coeliac rotavirus-mediated gastroenteritis can become compli-
disease (Smatti et al. 2019; Burke et al. 2020; Go mez- cated by the development of acute pancreatitis.
Rial et al. 2020). Support for rotavirus as a potential
causative agent in the development of T1D comes from Respiratory illness
the observation that incidence of T1D decreased by
Spreading of rotavirus infection is classically attributed
3–14% in rotavirus-vaccinated children aged 0–4 years
solely to fecal-oral transmission (Esona and Gautam
in the USA and Australia (Perrett et al. 2019; Burke et al.
mez-Rial et al. 2020). However, Finnish studies 2015). Nevertheless, the possibility of a contribution of
2020; Go
respiratory rotavirus transmission cannot be discounted
in children up to 10 years and American studies in chil-
dren with follow-up of 5–12 years, failed to detect asso- (Goldwater et al. 1979). Indeed, occurrence of respira-
ciation between rotavirus vaccination and T1D tory symptoms (rhinorrhea, nasal congestion, coughing,
development (Vaarala et al. 2017; Hemming-Harlo et al. vomiting or pneumonia; Table 1) shortly after or con-
2019; Burke et al. 2020; Glanz et al. 2020). Animal stud- comitant with rotavirus gastroenteritis appears com-
ies, however, largely support the idea that rotavirus mon in infants and children (30–65%) (Goldwater et al.
infection can be a trigger of T1D. Both in vitro as well in 1979; Gurwith et al. 1981; Santosham et al. 1983;
neonates in vivo, infection by rotavirus of Non-Obese Fragoso et al. 1986; Zhaori et al. 1991; Brouard et al.
Diabetic (NOD) mouse islet cells has been convincingly 2000; Sharma et al. 2002; Grimprel et al. 2008; Yu et al.
demonstrated (Ramig 2007; Graham et al. 2008; Pane 2012). Furthermore, rotavirus antigen was found in oro-
and Coulson 2015) and results in transient (pre-)diabetic pharyngeal, nasopharyngeal and tracheal aspirates
symptoms like hyperglycaemia, pancreatic cell death (Yolken and Murphy 1982; Fragoso et al. 1986; Zhaori
and reduced insulin production (Honeyman et al. 2014). et al. 1991; Zheng et al. 1991), and RT-PCR detected
Mechanistically, this effect may linked to the spread of rotavirus RNA in the lungs from a patient with fatal
rotavirus in infected NOD mice to the pancreatic lymph diarrhoea-associated disease (Lynch et al. 2003). Also,
nodes (PLN) and mesenteric lymph nodes (MLN) with the advent of metagenomics, the rotavirus gen-
(Webster et al. 2013) where the infection leads to the ome is frequently found in tracheal aspirates or throat
activation of antigen presenting cells and accumulation and nasal swabs from patients with respiratory tract
of autoreactive lymphocytes (Graham et al. 2008; Pane infections, concurrently with bona-fide respiratory
et al. 2014; Pane and Coulson 2015), which may pro- viruses (adenovirus, influenza A virus, respiratory syn-
voke T1D development (Burke et al. 2020) (Figure 2). cytial virus and rhinovirus) and bacteria (Taboada et al.
The induction of anti-islet autoimmunity may be further 2014; Madi et al. 2018; Thi Kha Tu et al. 2020). In appar-
stimulated by similarity of the rotavirus VP7 protein to ent agreement, in Sus scrofa intranasal inoculation with
two T1D-associated autoantigens, glutamic acid decarb- virulent human rotavirus (HRV, Wa) was not inferior to
oxylase (GAD65) and insulinoma antigen 2 (IA-2) pro- oral or gavage inoculation with regard to provoking
tein. Support for this notion comes from the diarrhoea, and viremia relative to oral or gavage-medi-
observation that the presence of anti-GAD65 antibody ated inoculation, and provoked nasal viral shedding
levels in children shows positive correlation with anti- (Azevedo et al. 2005). In Bos bovis, colostrum-deprived
rotavirus IgG levels (Burke et al. 2020). With the more and thus infection-prone calves were orally inoculated
widespread introduction of anti-virus vaccination with the bovine rotaviruses (GARVs, KJ9-1, species A)
around the globe, the overall contribution of rotavirus and developed transient interstitial pneumonia accom-
to the aetiology of T1D should become more evident. panied by the presence of GARV antigen and viral RNA
Although a rare complication, various cases of acute in the pneumocytes and lung lymphocytes (Kim et al.
pancreatitis, accompanied by diarrhoea, abdominal 2011). Analogously, in neonatal rats, replicating RRV
CRITICAL REVIEWS IN MICROBIOLOGY 589

was detected in alveolar macrophages and pneumo- reported, kidney lesions due to cellular degeneration
cytes (Crawford et al. 2006). The latter study described and mild infiltration of mononuclear cells have been
that rotavirus antigen levels in the lower respiratory identified in mice infected with rotavirus (Kashyap et al.
tract and stomach showed strong correlation and thus 2018). The fact that monkey kidney cells (secondary
proposed that rotavirus was released into respiratory and permanent cell lines) are highly susceptible for RV
secretions and swallowed. Also in newborn mice, fol- infection in vitro is compatible with the view that kid-
lowing murine rotavirus or heterologous RRV infection, neys can be productively infected in vivo (Ward et al.
viral RNA, NSP4 protein and viral particles were found 1984; Gilger et al. 1992; Lynch et al. 2003; Crawford
in lung tissues (Fenaux et al. 2006), whereas it has also et al. 2006; Fenaux et al. 2006).
been shown that intranasal immunization of mice with In addition to the kidneys and the heart, also the
inactivated RRV protects against subsequent rotavirus bone marrow may be affected following rotavirus infec-
challenge (Coffin and Clark 2001). Intriguingly, oral tion. Thrombocytopenia and significantly lower mean
immunization was not protective, possibly suggesting platelet volume (MPV) are more common in children
alternative handling of the rotavirus vaccine by the with rotavirus gastroenteritis than in controls (Mete
bronchial and intestinal mucosal immune system. In et al. 2014; Tanju et al. 2014; Ai et al. 2016), but as there
toto, there seems to be a contribution of the respiratory is no significant association between MPV and gastro-
compartment in rotavirus-provoked pathogenesis, even enteritis score (Mete et al. 2014; Tanju et al. 2014), the
while it remains unclear whether respiratory illness pro- relation may be indirect, for instance relating to dis-
vokes susceptibility to rotavirus infection or rotavirus turbed vitamin uptake.
infection can provoke respiratory illness (Taboada et al.
2014; Madi et al. 2018; Thi Kha Tu et al. 2020) (Figure 2). Conclusions
As the leading cause of severe acute gastroenteritis,
Other extra-intestinal complications rotavirus infection has a substantial effect on morbidity
A number of case reports have described additional and mortality in infants and young children. Rotavirus
extra-intestinal complications of rotavirus infection, primarily infects the intestinal enterocytes and induces
including myocarditis, renal failure and thrombocyto- secretory-driven watery diarrhoea. In addition, both
penia (Ramig 2007) (Table 1 and Figure 2). Indeed, both human and animal experimentation demonstrates that
autopsy examinations and animal studies have extra-intestinal tissue can be susceptible to rotavirus
described the presence of rotavirus RNA, antigens and infection in turn provoking further pathology, which
replication in heart and kidney (Cioc and Nuovo 2002; may be the consequence of virus-mediated destruction
Lynch et al. 2003; Crawford et al. 2006; Ramig 2007). of the cells involved, or indirect secondary to anti-viral
Post-mortem histopathological reports showed that car- immune responses. Nevertheless, the involvement of
diac rotavirus infection could contribute to hyper- rotavirus in extra-intestinal tissue damage is much bet-
trophic cardiomyopathy with focal disorganized ter documented in animal models than in humans, and
myocardial architecture, myocyte necrosis and myocar- there are gaps in the understanding of mechanisms of
ditis (Grech et al. 2001; Cioc and Nuovo 2002; Nakano pathogenesis. It is to be expected that together with
et al. 2011). Prominent lymphocytic infiltration was rotavirus-mediated gastroenteritis such extra-intestinal
found in the myocardium in cases of rotavirus myocar- manifestations will become less prevalent as conse-
ditis, suggesting that antiviral immunity may provoke quence from the introduction of anti-rotavirus vaccin-
myocardial damage (Cioc and Nuovo 2002) and indeed ation. For now, however, clinicians, particularly
paediatricians caring for rotavirus-infected children
rotavirus infection has been associated to increased
should be aware of the possible extra-intestinal compli-
Creatine Kinase-MB signalling damage to the heart dur-
cations of rotavirus infection.
ing infection (Zheng et al. 2017).
With respect to renal manifestations, interpretation
is complicated by that the rotavirus diarrhoea-induced Acknowledgments
dehydration can lead to pre- and post-renal failure and
The authors thank freepik and edraw for their assistance
thus renal problems following infection may not reflect with regard to graphic illustrations.
direct infection of the kidneys (Fujinaga et al. 2005;
Ashida et al. 2012; Shirasu et al. 2015; Kira et al. 2016;
Tsukida et al. 2018). Nevertheless, although direct rota- Disclosure statement
virus damage to renal epithelial cells has not been The authors declare that they have no conflict of interest.
590 Z. DIAN ET AL.

Funding promote immune-mediated bile duct injury in murine bil-


iary atresia. Hepatology. 68:1890–1904.
This work was supported by the following: the National Bezerra JA, Tiao G, Ryckman FC, Alonso M, Sabla GE,
Natural Science Foundation of China [81960606 to Z.D.]; Shneider B, Sokol RJ, Aronow BJ. 2002. Genetic induction
Yunnan Fundamental Research Projects [grant no. of proinflammatory immunity in children with biliary atre-
2017FE468(-124) to Z.D.]; the China Scholarship Council for sia. Lancet. 360:1653–1659.
funding PhD fellowships [201708530234 to Z.M.]. Biggart R, Finn A, Marlow R. 2018. Lack of impact of rotavirus
vaccination on childhood seizure hospitalizations in
England – an interrupted time series analysis. Vaccine. 36:
4589–4592.
Author contributions Blutt SE, Conner ME. 2007. Rotavirus: to the gut and beyond!
Q.P. and Z.M. designed and directed the project; Z.D. wrote Curr Opin Gastroenterol. 23:39–43.
the manuscript with support from M.P., Y.S., G.Z. and Y.X.; Blutt SE, Kirkwood CD, Parren ~o V, Warfield KL, Ciarlet M,
X.F. and X.Y. searched and reviewed the literatures, and they Estes MK, Bok K, Bishop RF, Conner ME. 2003. Rotavirus
aided in interpreting the results. Z.D., Y.S., X.F. and X.Y. antigenaemia and viraemia: a common event? Lancet. 362:
assessed the screened literatures; Z.M. designed the figures; 1445–1449.
G.Z. and Y.X. drew the figures; Q.P. and M.P. directed the Blutt SE, Matson DO, Crawford SE, Staat MA, Azimi P,
revision of the manuscript; all authors provided critical feed- Bennett BL, Piedra PA, Conner ME. 2007. Rotavirus antige-
back and contributed to the final version of the manuscript. nemia in children is associated with viremia. PLoS Med. 4:
e121.
Brouard J, Freymuth F, Vabret A, et al. 2000. Viral co-infec-
ORCID tions in immunocompetent infants with bronchiolitis: pro-
spective epidemiologic study] Co-infections virales lors des
Zhijiang Miao http://orcid.org/0000-0001-7778-2010 bronchiolites du nourrisson immunocompetent: etude pro-
spective epidemiologique. Arch Pediatr. 7:531s–535s.
Burke RM, Tate JE, Dahl RM, Aliabadi N, Parashar UD. 2018.
References Rotavirus vaccination is associated with reduced seizure
Ai Q, Yin J, Chen S, Qiao L, Luo N. 2016. Rotavirus-associated hospitalization risk among commercially insured US chil-
immune thrombocytopenic purpura in children: a retro- dren. Clin Infect Dis. 67:1614–1616.
spective study. Exp Ther Med. 12:2187–2190. Burke RM, Tate JE, Dahl RM, Saydah S, Imperatore G, Gregg
Aliabadi N, Antoni S, Mwenda JM, Weldegebriel G, Biey JNM, EW, Parashar UD. 2020. Rotavirus Vaccination and Type 1
Cheikh D, Fahmy K, Teleb N, Ashmony HA, Ahmed H, Diabetes Risk Among US Children With Commercial
et al. 2019. Global impact of rotavirus vaccine introduction Insurance. JAMA Pediatr. 174:383–385.
on rotavirus hospitalisations among children under 5 years Burke RM, Tate JE, Jiang B, et al. 2020. Rotavirus and type 1
of age, 2008-16: findings from the Global Rotavirus diabetes-is there a connection? A synthesis of the evi-
dence. J Infect Dis. 222:1076–1083.
Surveillance Network. Lancet Glob Health. 7:e893–e903.
Burke RM, Tate JE, Kirkwood CD, Steele AD, Parashar UD.
Allen SR, Jafri M, Donnelly B, McNeal M, Witte D, Bezerra J,
2019. Current and new rotavirus vaccines. Curr Opin Infect
Ward R, Tiao GM. 2007. Effect of rotavirus strain on the
Dis. 32:435–444.
murine model of biliary atresia. J Virol. 81:1671–1679.
Burnett E, Parashar UD, Tate JE. 2020. Associations of intus-
Ashida A, Fujieda M, Ohta K, Nakakura H, Matsumura H,
susception with adenovirus, rotavirus, and other patho-
Morita T, Igarashi T, Tamai H. 2012. Clinical characteristics
gens: a review of the literature. Pediatr Infect Dis J. 39:
of obstructive uropathy associated with rotavirus gastro-
1127–1130.
enteritis in Japan. Clin Nephrol. 77:49–54.
Burnett E, Parashar UD, Tate JE. 2020. Real-world effective-
Azevedo MS, Yuan L, Jeong KI, Gonzalez A, Nguyen TV, Pouly
ness of rotavirus vaccines, 2006-19: a literature review and
S, Gochnauer M, Zhang W, Azevedo A, Saif LJ, et al. 2005. meta-analysis. Lancet Glob Health. 8:e1195–e1202.
Viremia and nasal and rectal shedding of rotavirus in Chen S, Li P, Wang Y, Yin Y, de Ruiter PE, Verstegen MMA,
gnotobiotic pigs inoculated with Wa human rotavirus. J Peppelenbosch MP, van der Laan LJW, Pan Q. 2020.
Virol. 79:5428–5436. Rotavirus infection and cytopathogenesis in human biliary
Banyai K, Estes MK, Martella V, Parashar UD. 2018. Viral organoids potentially recapitulate biliary atresia develop-
gastroenteritis. Lancet. 392:175–186. ment. mBio. 11:e01968-20.
Baqui AH, Black RE, El Arifeen S, Yunus M, Chakraborty J, Chen SM, Lin CP, Tsai JD, Chao YH, Sheu JN. 2014. The sig-
Ahmed S, Vaughan JP. 2002. Effect of zinc supplementa- nificance of serum and fecal levels of interleukin-6 and
tion started during diarrhoea on morbidity and mortality interleukin-8 in hospitalized children with acute rotavirus
in Bangladeshi children: community randomised trial. BMJ. and norovirus gastroenteritis. Pediatr Neonatol. 55:
325:1059. 120–126.
Basturk A, Artan R, Yilmaz A. 2017. Rotavirus gastroenteritis Cioc AM, Nuovo GJ. 2002. Histologic and in situ viral findings
and a rare case accompanying acute pancreatitis. Prz in the myocardium in cases of sudden, unexpected death.
Gastroenterol. 12:68–69. Mod Pathol. 15:914–922.
Bednarek J, Traxinger B, Brigham D, Roach J, Orlicky D, Wang Coffin SE, Clark SL. 2001. Induction of intestinal rotavirus-spe-
D, Pelanda R, Mack CL. 2018. Cytokine-producing B cells cific antibodies in respiratory, but not gut, lymphoid
CRITICAL REVIEWS IN MICROBIOLOGY 591

tissues following mucosal immunization of mice with inac- Rotavirus encephalopathy: pathogenesis reviewed. J
tivated rotavirus. Virology. 291:235–240. Paediatr Child Health. 37:206–209.
Colbere-Garapin F, Martin-Latil S, Blondel B, Mousson L, mez-Rial J, Rivero-Calle I, Salas A, Martino
Go n-Torres F. 2020.
Pelletier I, Autret A, François A, Niborski V, Grompone G, Rotavirus and autoimmunity. J Infect. 81:183–189.
Catonnet G, et al. 2007. Prevention and treatment of mez-Rial J, Sanchez-Batan S, Rivero-Calle I, Pardo-Seco J,
Go
enteric viral infections: possible benefits of probiotic bac- Martino n-Martınez JM, Salas A, Martino n-Torres F. 2019.
teria. Microbes Infect. 9:1623–1631. Rotavirus infection beyond the gut. Infect Drug Resist. 12:
Coulson BS, Witterick PD, Tan Y, Hewish MJ, Mountford JN, 55–64.
Harrison LC, Honeyman MC. 2002. Growth of rotaviruses in Graham KL, O’Donnell JA, Tan Y, Sanders N, Carrington EM,
primary pancreatic cells. J Virol. 76:9537–9544. Allison J, Coulson BS. 2007. Rotavirus infection of infant
Crawford SE, Patel DG, Cheng E, Berkova Z, Hyser JM, Ciarlet and young adult nonobese diabetic mice involves extrain-
M, Finegold MJ, Conner ME, Estes MK. 2006. Rotavirus vir- testinal spread and delays diabetes onset. J Virol. 81:
emia and extraintestinal viral infection in the neonatal rat 6446–6458.
model. J Virol. 80:4820–4832. Graham KL, Sanders N, Tan Y, Allison J, Kay TWH, Coulson
Crawford SE, Ramani S, Tate JE, Parashar UD, Svensson L, BS. 2008. Rotavirus infection accelerates type 1 diabetes in
Hagbom M, Franco MA, Greenberg HB, O’Ryan M, Kang G, mice with established insulitis. JVI. 82:6139–6149.
et al. 2017. Rotavirus infection. Nat Rev Dis Primers. 3: Grech V, Calvagna V, Falzon A, Mifsud A. 2001. Fatal, rota-
17083–17083. virus-associated myocarditis and pneumonitis in a 2-year-
Czech-Schmidt G, Verhagen W, Szavay P, Leonhardt J, old boy. Ann Trop Paediatr. 21:147–148.
Petersen C. 2001. Immunological gap in the infectious ani- Greenberg HB, Clark HF, Offit PA. 1994. Rotavirus pathology
mal model for biliary atresia. J Surg Res. 101:62–67. and pathophysiology. Curr Top Microbiol Immunol. 185:
Dalgic N, Sancar M, Bayraktar B, Pullu M, Hasim O. 2011. 255–283.
Probiotic, zinc and lactose-free formula in children with Grimprel E, Rodrigo C, Desselberger U. 2008. Rotavirus dis-
rotavirus diarrhea: are they effective? Pediatr Int. 53: ease: impact of coinfections. Pediatr Infect Dis J. 27:
677–682. S3–S10.
Dharakul T, Riepenhoff-Talty M, Albini B, Ogra PL. 1988. Gurwith M, Wenman W, Hinde D, Feltham S, Greenberg H.
Distribution of rotavirus antigen in intestinal lymphoid tis- 1981. A prospective study of rotavirus infection in infants
sues: potential role in development of the mucosal and young children. J Infect Dis. 144:218–224.
immune response to rotavirus. Clin Exp Immunol. 74: Haffejee IE, Moosa A. 1990. Rotavirus studies in Indian
14–19. (Asian) South African infants with acute gastro-enteritis: II.
DiFazio MP, Braun L, Freedman S, Hickey P. 2007. Rotavirus- Clinical aspects and outcome. Ann Trop Paediatr. 10:
induced seizures in childhood. J Child Neurol. 22: 245–254.
1367–1370. Hagbom M, De Faria FM, Winberg ME, Westerberg S,
Esona MD, Gautam R. 2015. Rotavirus. Clin Lab Med. 35: Nordgren J, Sharma S, Keita ÅV, Loitto V, Magnusson KE,
363–391. Svensson L, et al. 2020. Neurotrophic factors protect the
Fenaux M, Cuadras MA, Feng N, Jaimes M, Greenberg HB. intestinal barrier from rotavirus insult in mice. mBio. 11:
2006. Extraintestinal spread and replication of a homolo- 31964731.
gous EC rotavirus strain and a heterologous rhesus rota- Hagbom M, Istrate C, Engblom D, Karlsson T, Rodriguez-Diaz
virus in BALB/c mice. JVI. 80:5219–5232. J, Buesa J, Taylor JA, Loitto VM, Magnusson KE, Ahlman H,
Filippi CM, von Herrath MG. 2008. Viral trigger for type 1 dia- et al. 2011. Rotavirus stimulates release of serotonin (5-HT)
betes: pros and cons. Diabetes. 57:2863–2871. from human enterochromaffin cells and activates brain
Fragoso M, Kumar A, Murray DL. 1986. Rotavirus in nasopha- structures involved in nausea and vomiting. PLoS Pathog.
ryngeal secretions of children with upper respiratory tract 7:e1002115.
infections. Diagn Microbiol Infect Dis. 4:87–88. Hagbom M, Sharma S, Lundgren O, Svensson L. 2012.
Fujinaga S, Kaneko K, Ohtomo Y, Takada M, Kobayashi K, Towards a human rotavirus disease model. Curr Opin
Tada M, Yamashiro Y. 2005. Acute renal failure due to Virol. 2:408–418.
obstructive uric acid stones associated with rotavirus Hagbom M, Svensson L, et al. 2016. Chapter 2.6 – Rotavirus
gastroenteritis. Pediatr Nephrol. 20:239–240. disease mechanisms. In: Svensson L, Desselberger U,
Fukuda S, Kishi K, Yasuda K, Sejima H, Yamaguchi S. 2009. Greenberg HB, editors. Viral gastroenteritis. Boston (MA):
Rotavirus-associated encephalopathy with a reversible Academic Press; p. 189–218.
splenial lesion. Pediatric Neurology. 40:131–133. Hagbom M, Svensson L. 2016. Rotavirus disease mechanisms.
Gilger MA, Matson DO, Conner ME, Rosenblatt HM, Finegold In: Svensson L, Desselberger U, Greenberg HB, Estes MK,
MJ, Estes MK. 1992. Extraintestinal rotavirus infections in editors. Viral gastroenteritis. London (UK): Academic Press;
children with immunodeficiency. J Pediatr. 120:912–917. p. 189–218.
Glanz JM, Clarke CL, Xu S, Daley MF, Shoup JA, Schroeder EB, Hartman S, Brown E, Loomis E, Russell HA. 2019.
Lewin BJ, McClure DL, Kharbanda E, Klein NP, et al. 2020. Gastroenteritis in children. Am Fam Physician. 99:159–165.
Association between rotavirus vaccination and type 1 dia- Hemming-Harlo M, L€ahdeaho ML, M€aki M, Vesikari T. 2019.
betes in children. JAMA Pediatr. 174:455–462. Rotavirus vaccination does not increase type 1 diabetes
Goldwater PN, Chrystie IL, Banatvala JE. 1979. Rotaviruses and may decrease celiac disease in children and adoles-
and the respiratory tract. Br Med J. 2:1551. cents. Pediatr Infect Dis J. 38:539–541.
Goldwater PN, Rowland K, Thesinger M, Abbott K, Grieve A, Herrmann B, Lawrenz-Wolf B, Seewald C, et al. 1993. [5th day
Palombo EA, Masendycz PJ, Wilkinson I, Bear J. 2001. convulsions of the newborn infant in rotavirus infections]
592 Z. DIAN ET AL.

5-Tages-Kr€ampfe des Neugeborenen bei Kordasti S, Sjo€vall H, Lundgren O, et al. 2004. Serotonin and
Rotavirusinfektionen. Monatsschr Kinderheilkd. 141: vasoactive intestinal peptide antagonists attenuate rota-
120–123. virus diarrhoea. Gut. 53:952–957.
Hertel PM, Estes MK. 2012. Rotavirus and biliary atresia: can Kovacs A, Chan L, Hotrakitya C, Overturf G, Portnoy B. 1986.
causation be proven? Curr Opin Gastroenterol. 28:10–17. Serum transaminase elevations in infants with rotavirus
Hjelt K, Krasilnikoff PA, Grauballe PC, Rasmussen SW. 1985. gastroenteritis. J Pediatr Gastroenterol Nutr. 5:873–877.
Clinical features in hospitalised children with acute gastro- Kraft LM. 1958. Observations on the control and natural his-
enteritis. Does the rotavirus syndrome exist? Acta Paediatr tory of epidemic diarrhea of infant mice (EDIM). Yale J Biol
Scand. 74:96–101. Med. 31:121–137.
Honeyman MC, Laine D, Zhan Y, Londrigan S, Kirkwood C, Laizane G, Smane L, Nokalna I, Gardovska D, Feemster KA.
Harrison LC. 2014. Rotavirus infection induces transient 2019. Rotavirus-associated seizures and reversible corpus
pancreatic involution and hyperglycemia in weanling callosum lesion. Acta Med Litu. 26:113–117.
mice. PLoS One. 9:e106560. Leung AK, Kellner JD, Davies HD. 2005. Rotavirus gastroenter-
Hongou K, Konishi T, Yagi S, Araki K, Miyawaki T. 1998. itis. Adv Ther. 22:476–487.
Rotavirus encephalitis mimicking afebrile benign convul- Li ST, Grossman DC, Cummings P. 2007. Loperamide therapy
sions in infants. Pediatr Neurol. 18:354–357. for acute diarrhea in children: systematic review and
Hyser JM, Collinson-Pautz MR, Utama B, Estes MK. 2010. meta-analysis. PLoS Med. 4:e98.
Rotavirus disrupts calcium homeostasis by NSP4 viroporin Lloyd MB, Lloyd JC, Gesteland PH, Bale JF. 2010. Rotavirus
activity. mBio. 1:e00265-10. gastroenteritis and seizures in young children. Pediatr
Hyser JM, Utama B, Crawford SE, Estes MK. 2012. Genetic Neurol. 42:404–408.
divergence of rotavirus nonstructural protein 4 results in Lu HL, Ding Y, Goyal H, Xu HG. 2019. Association between
distinct serogroup-specific viroporin activity and intracellu- rotavirus vaccination and risk of intussusception among
lar punctate structure morphologies. J Virol. 86:4921–4934. neonates and infants: a systematic review and meta-ana-
Istrate C, Hagbom M, Vikstro €m E, Magnusson KE, Svensson L. lysis. JAMA Netw Open. 2:e1912458.
2014. Rotavirus infection increases intestinal motility but Lundgren O, Svensson L. 2001. Pathogenesis of rotavirus
not permeability at the onset of diarrhea. J Virol. 88: diarrhea. Microbes Infect. 3:1145–1156.
3161–3169. Lynch M, Lee B, Azimi P, Gentsch J, Glaser C, Gilliam S,
Iturriza-Gomara M, Auchterlonie IA, Zaw W, Molyneaux P, Chang H-GH, Ward R, Glass RI. 2001. Rotavirus and central
Desselberger U, Gray J. 2002. Rotavirus gastroenteritis and nervous system symptoms: cause or contaminant? Case
central nervous system (CNS) infection: characterization of reports and review. Clin Infect Dis. 33:932–938.
the VP7 and VP4 genes of rotavirus strains isolated from Lynch M, Shieh WJ, Tatti K, Gentsch JR, Harris TF, Jiang B,
paired fecal and cerebrospinal fluid samples from a child Guarner J, Bresee JS, Greenwald M, Cullen S, et al. 2003.
with CNS disease. J Clin Microbiol. 40:4797–4799. The pathology of rotavirus-associated deaths, using new
Janke BH, Morehouse LG, Solorzano RF. 1988. Single and molecular diagnostics. Clin Infect Dis. 37:1327–1333.
mixed infections of neonatal pigs with rotaviruses and Mack CL, Tucker RM, Lu BR, Sokol RJ, Fontenot AP, Ueno Y,
enteroviruses: virological studies. Can J Vet Res. 52: Gill RG. 2006. Cellular and humoral autoimmunity directed
360–363. at bile duct epithelia in murine biliary atresia. Hepatology.
Karampatsas K, Spyridou C, Morrison IR, Tong CYW, 44:1231–1239.
Prendergast AJ. 2015. Rotavirus-associated mild encephal- Madi N, Al-Nakib W, Mustafa AS, Habibi N. 2018.
opathy with a reversible splenial lesion (MERS)-case report Metagenomic analysis of viral diversity in respiratory sam-
and review of the literature. BMC Infect Dis. 15:446. ples from patients with respiratory tract infections in
Kashyap G, Singh R, Malik YS, Agrawal RK, Singh KP, Kumar Kuwait. J Med Virol. 90:412–420.
P, Sahoo M, Gupta D, Singh R. 2018. Experimental bovine Matthijnssens J, Ciarlet M, Heiman E, Arijs I, Delbeke T,
rotavirus-A (RV-A)infection causes intestinal and extra- McDonald SM, Palombo EA, Iturriza-Go mara M, Maes P,
intestinal pathology in suckling mice. Microb Pathog. 121: Patton JT, et al. 2008. Full genome-based classification of
22–26. rotaviruses reveals a common origin between human Wa-
Keidan I, Shif I, Keren G, Passwell JH. 1992. Rotavirus enceph- Like and porcine rotavirus strains and human DS-1-like
alopathy: evidence of central nervous system involvement and bovine rotavirus strains. J Virol. 82:3204–3219.
during rotavirus infection. Pediatr Infect Dis J. 11:773–775. Mete E, Akelma AZ, Cizmeci MN, Bozkaya D, Kanburoglu MK.
Kim HJ, Park JG, Matthijnssens J, Lee JH, Bae YC, Alfajaro 2014. Decreased mean platelet volume in children with
MM, Park SI, Kang MI, Cho KO. 2011. Intestinal and extra- acute rotavirus gastroenteritis. Platelets. 25:51–54.
intestinal pathogenicity of a bovine reassortant rotavirus Miyata R, Tanuma N, Hayashi M, Imamura T, Takanashi JI,
in calves and piglets. Vet Microbiol. 152:291–303. Nagata R, Okumura A, Kashii H, Tomita S, Kumada S, et al.
Kira S, Mitsui T, Zakoji H, Aoki T, Sawada N, Miyamoto T, 2012. Oxidative stress in patients with clinically mild
Takeda M. 2016. Postrenal failure due to urinary stones encephalitis/encephalopathy with a reversible splenial
associated with acute viral gastroenteritis: three case lesion (MERS). Brain Dev. 34:124–127.
reports. Case Rep Urol. 2016:1375923. Mormile R. 2016. Severe gastroenteritis and acute pancrea-
Ko€hler T, Erben U, Wiedersberg H, et al. 1990. [Histological titis following rotavirus infection in children: the age-
findings of the small intestinal mucosa in rotavirus infec- related failure of IFN-c? Immunol Lett. 175:58–59.
tions in infants and young children] Histologische Befunde Mossel EC, Ramig RF. 2003. A lymphatic mechanism of rota-
der Du €nndarmschleimhaut bei Rotavirusinfektionen im virus extraintestinal spread in the neonatal mouse. J Virol.
S€auglings- und Kleinkindalter. Kinderarztl Prax. 58:323–327. 77:12352–12356.
CRITICAL REVIEWS IN MICROBIOLOGY 593

Nakano I, Taniguchi K, Ishibashi-Ueda H, Maeno Y, Petersen C, Biermanns D, Kuske M, Sch€akel K, Meyer-


Yamamoto N, Yui A, Komoto S, Wakata Y, Matsubara T, Jungh€anel L, Mildenberger H. 1997. New aspects in a mur-
Ozaki N, et al. 2011. Sudden death from systemic rotavirus ine model for extrahepatic biliary atresia. J Pediatr Surg.
infection and detection of nonstructural rotavirus proteins. 32:1190–1195.
J Clin Microbiol. 49:4382–4385. Ramani S, Crawford SE, Blutt SE, Estes MK. 2018. Human
Nigro G. 1991. Pancreatitis with hypoglycemia-associated organoid cultures: transformative new tools for human
convulsions following rotavirus gastroenteritis. J Pediatr virus studies. Curr Opin Virol. 29:79–86.
Gastroenterol Nutr. 12:280–282. Ramig RF. 2004. Pathogenesis of intestinal and systemic rota-
Nishimura S, Ushijima H, Nishimura S, Shiraishi H, Kanazawa virus infection. J Virol. 78:10213–10220.
C, Abe T, Kaneko K, Fukuyama Y. 1993. Detection of rota- Ramig RF. 2007. Systemic rotavirus infection. Expert Rev anti
virus in cerebrospinal fluid and blood of patients with Infect Ther. 5:591–612.
convulsions and gastroenteritis by means of the reverse Rautanen T, Kurki S, Vesikari T. 1997. Randomised double
transcription polymerase chain reaction. Brain Dev. 15: blind study of hypotonic oral rehydration solution in diar-
457–459. rhoea. Arch Dis Child. 76:272–274.
Operario DJ, Platts-Mills JA, Nadan S, Page N, Seheri M, Rewers M, Ludvigsson J. 2016. Environmental risk factors for
Mphahlele J, Praharaj I, Kang G, Araujo IT, Leite JPG, et al. type 1 diabetes. Lancet. 387:2340–2348.
2017. Etiology of severe acute watery diarrhea in children Reynolds DJ, Hall GA, Debney TG, Parsons KR. 1985.
in the Global Rotavirus Surveillance Network using quanti- Pathology of natural rotavirus infection in clinically normal
tative polymerase chain reaction. J Infect Dis. 216:220–227. calves. Res Vet Sci. 38:264–269.
Orrico-Sanchez A, Lo  pez-Lacort M, Mun~oz-Quiles C, Dıez- Riepenhoff-Talty M, Gouvea V, Evans MJ, Svensson L,
Domingo J. 2018. Lack of impact of rotavirus vaccines on Hoffenberg E, Sokol RJ, Uhnoo I, Greenberg SJ, Schakel K,
seizure-related hospitalizations in children under 5 years Zhaori G, et al. 1996. Detection of group C rotavirus in
old in Spain. Hum Vaccin Immunother. 14:1534–1538. infants with extrahepatic biliary atresia. J Infect Dis. 174:
Osborne MP, Haddon SJ, Spencer AJ, Collins J, Starkey WG, 8–15.
Wallis TS, Clarke GJ, Worton KJ, Candy DC, Stephen J, Riepenhoff-Talty M, Schaekel K, Clark HF, Mueller W, Uhnoo I,
et al. 1988. An electron microscopic investigation of time- Rossi T, Fisher J, Ogra PL. 1993. Group A rotaviruses pro-
related changes in the intestine of neonatal mice infected duce extrahepatic biliary obstruction in orally inoculated
with murine rotavirus. J Pediatr Gastroenterol Nutr. 7: newborn mice. Pediatr Res. 33:394–399.
236–248. Robinson CG, Hernanz-Schulman M, Zhu Y, Griffin MR,
Pager C, Steele D, Gwamanda P, Driessen M. 2000. A neo- Gruber W, Edwards KM. 2004. Evaluation of anatomic
natal death associated with rotavirus infection–detection changes in young children with natural rotavirus infection:
of rotavirus dsRNA in the cerebrospinal fluid. S Afr Med J. is intussusception biologically plausible? J Infect Dis. 189:
90:364–365. 1382–1387.
Pane JA, Coulson BS. 2015. Lessons from the mouse: poten- Rodriguez-Calvo T, Sabouri S, Anquetil F, von Herrath MG.
tial contribution of bystander lymphocyte activation by 2016. The viral paradigm in type 1 diabetes: who are the
viruses to human type 1 diabetes. Diabetologia. 58: main suspects? Autoimmun Rev. 15:964–969.
1149–1159. Rogers MAM, Basu T, Kim C. 2019. Lower incidence rate of
Pane JA, Webster NL, Zufferey C, Coulson BS. 2014. Rotavirus type 1 diabetes after receipt of the rotavirus vaccine in
acceleration of murine type 1 diabetes is associated with the United States, 2001-2017. Sci Rep. 9:7727.
increased MHC class I-restricted antigen presentation by B Rogers MAM, Kim C, Banerjee T, Lee JM. 2017. Fluctuations
cells and elevated proinflammatory cytokine expression by in the incidence of type 1 diabetes in the United States
T cells. Virus Res. 179:73–84. from 2001 to 2015: a longitudinal study. BMC Med. 15:
Parashar UD, Nelson EA, Kang G. 2013. Diagnosis, manage- 199.
ment, and prevention of rotavirus gastroenteritis in chil- Santosham M, Yolken RH, Quiroz E, Dillman L, Oro G, Reeves
dren. BMJ. 347:f7204. WC, Sack RB. 1983. Detection of rotavirus in respiratory
Pardo-Seco J, Cebey-Lo pez M, Martino
n-Torres N, Salas A, secretions of children with pneumonia. J Pediatr. 103:
Gomez-Rial J, Rodriguez-Tenreiro C, Martinon-Sanchez JM, 583–585.
Martino n-Torres F. 2015. Impact of rotavirus vaccination Saxena K, Blutt SE, Ettayebi K, Zeng XL, Broughman JR,
on childhood hospitalization for seizures. Pediatr Infect Dis Crawford SE, Karandikar UC, Sastri NP, Conner ME, Opekun
J. 34:769–773. AR, et al. 2016. Human intestinal enteroids: a new model
Parri N, Innocenti L, Collini S, Bechi F, Mannelli F. 2010. to study human rotavirus infection, host restriction, and
Acute pancreatitis due to rotavirus gastroenteritis in a pathophysiology. J Virol. 90:43–56.
child. Pediatr Emerg Care. 26:592–593. Sharma R, Hudak ML, Premachandra BR, Stevens G, Monteiro
Payne DC, Baggs J, Zerr DM, Klein NP, Yih K, Glanz J, Curns CB, Bradshaw JA, Kaunitz AM, Hollister RA. 2002. Clinical
AT, Weintraub E, Parashar UD. 2014. Protective association manifestations of rotavirus infection in the neonatal inten-
between rotavirus vaccination and childhood seizures in sive care unit. Pediatr Infect Dis J. 21:1099–1105.
the year following vaccination in US children. Clin Infect Shaw DP, Morehouse LG, Solorzano RF. 1989. Rotavirus repli-
Dis. 58:173–177. cation in colostrum-fed and colostrum-deprived pigs. Am
Perrett KP, Jachno K, Nolan TM, Harrison LC. 2019. J Vet Res. 50:1966–1970.
Association of rotavirus vaccination with the incidence of Shepherd RW, Butler DG, Cutz E, Gall DG, Hamilton JR. 1979.
type 1 diabetes in children. JAMA Pediatr. 173:280–282. The mucosal lesion in viral enteritis. Extent and dynamics
594 Z. DIAN ET AL.

of the epithelial response to virus invasion in transmissible Uhnoo I, Riepenhoff-Talty M, Dharakul T, Chegas P, Fisher
gastroenteritis of piglets. Gastroenterology. 76:770–777. JE, Greenberg HB, Ogra PL. 1990. Extramucosal spread
Shirasu A, Ashida A, Matsumura H, Nakakura H, Tamai H. and development of hepatitis in immunodeficient and
2015. Clinical characteristics of rotavirus gastroenteritis normal mice infected with rhesus rotavirus. J Virol. 64:
with urinary crystals. Pediatr Int. 57:917–921. 361–368.
Shivakumar P, Campbell KM, Sabla GE, Miethke A, Tiao G, Ushijima H, Bosu K, Abe T, Shinozaki T. 1986. Suspected rota-
McNeal MM, Ward RL, Bezerra JA. 2004. Obstruction of virus encephalitis. Arch Dis Child. 61:692–694.
extrahepatic bile ducts by lymphocytes is regulated by Vaarala O, Jokinen J, Lahdenkari M, Leino T. 2017. Rotavirus
IFN-gamma in experimental biliary atresia. J Clin Invest. vaccination and the risk of celiac disease or type 1 dia-
114:322–329. betes in Finnish children at early life. Pediatr Infect Dis J.
Simsek C, Corman VM, Everling HU, Lukashev AN, Rasche A, 36:674–675.
Maganga GD, Binger T, Jansen D, Beller L, Deboutte W, Van Den Brink GR, Bloemers SM, Van Den Blink B, Tertoolen
et al. 2021. At least seven distinct rotavirus genotype con- LGJ, Van Deventer SJH, Peppelenbosch MP. 1999. Study of
stellations in bats with evidence of reassortment and zoo- calcium signaling in non-excitable cells. Microsc Res Tech.
notic transmissions. mBio. 12:e02755-20. 46:418–433.
Smatti MK, Cyprian FS, Nasrallah GK, Al Thani AA, Almishal Velazquez FR, Matson DO, Calva JJ, Guerrero L, Morrow AL,
RO, Yassine HM. 2019. Viruses and autoimmunity: a review Carter-Campbell S, Glass RI, Estes MK, Pickering LK, Ruiz-
on the potential interaction and molecular mechanisms. Palacios GM, et al. 1996. Rotavirus infection in infants as
Viruses. 11:762. protection against subsequent infections. N Engl J Med.
Taboada B, Espinoza MA, Isa P, Aponte FE, Arias-Ortiz MA, 335:1022–1028.
Monge-Martınez J, Rodrıguez-Vazquez R, Dıaz-Hernandez Ward RL, Knowlton DR, Pierce MJ. 1984. Efficiency of human
F, Zarate-Vidal F, Wong-Chew RM, et al. 2014. Is there still rotavirus propagation in cell culture. J Clin Microbiol. 19:
room for novel viral pathogens in pediatric respiratory 748–753.
tract infections? PLoS One. 9:e113570. Ward RL, McNeal MM, Sheridan JF. 1990. Development of an
Takanashi J, Miyamoto T, Ando N, Kubota T, Oka M, Kato Z, adult mouse model for studies on protection against rota-
Hamano S, Hirabayashi S, Kikuchi M, Barkovich AJ, et al. virus. J Virol. 64:5070–5075.
2010. Clinical and radiological features of rotavirus cere- Webster NL, Zufferey C, Pane JA, Coulson BS. 2013.
bellitis. AJNR Am J Neuroradiol. 31:1591–1595. Alteration of the thymic T cell repertoire by rotavirus
Tanju C, Ekrem G, Berksoy Emel A, et al. 2014. Mean platelet infection is associated with delayed type 1 diabetes
volume as a negative marker of inflammation in children development in non-obese diabetic mice. PLoS One. 8:
with rotavirus gastroenteritis. Iran J Pediatr. 24:617–622. e59182.
Tate JE, Burton AH, Boschi-Pinto C, et al. 2016. Global, Weclewicz K, Svensson L, Kristensson K. 1998. Targeting of
regional, and national estimates of rotavirus mortality in endoplasmic reticulum-associated proteins to axons and
children <5 years of age, 2000-2013. Clin Infect Dis. 62: dendrites in rotavirus-infected neurons. Brain Res Bull. 46:
S96–S105. 353–360.
Tate JE, Burton AH, Boschi-Pinto C, Steele AD, Duque J, Wong CJ, Price Z, Bruckner DA. 1984. Aseptic meningitis in
Parashar UD, WHO-coordinated Global Rotavirus an infant with rotavirus gastroenteritis. Pediatr Infect Dis.
Surveillance Network. 2012. 2008 estimate of worldwide 3:244–246.
rotavirus-associated mortality in children younger than 5 Wong V. 2001. Acute gastroenteritis-related encephalopathy.
years before the introduction of universal rotavirus vaccin- J Child Neurol. 16:906–910.
ation programmes: a systematic review and meta-analysis. Yamashiro Y, Shimizu T, Oguchi S, Sato M. 1989.
Lancet Infect Dis. 12:136–141. Prostaglandins in the plasma and stool of children with
Teitelbaum JE, Daghistani R. 2007. Rotavirus causes hepatic rotavirus gastroenteritis. J Pediatr Gastroenterol Nutr. 9:
transaminase elevation. Dig Dis Sci. 52:3396–3398. 322–327.
Thi Kha Tu N, Thi Thu Hong N, Thi Han Ny N, My Phuc T, Thi Yeom JS, Jo JY, Park JS, Kim YS, Chung JY, Han TH, Seo JH,
Thanh Tam P, Doorn H. R v, Dang Trung Nghia H, Thao Park ES, Lim JY, Woo HO, et al. 2019. Monocyte chemo-
Huong D, An Han D, Thi Thu Ha L, et al. 2020. The virome attractant protein (MCP)-1 in rotavirus-associated white
of acute respiratory diseases in individuals at risk of zoo- matter injury in newborns. Neuropediatrics. 50:228–234.
notic infections. Viruses. 12:960. Yeom JS, Park JS, Kim YS, Kim RB, Choi DS, Chung JY, Han
Trask SD, McDonald SM, Patton JT. 2012. Structural insights TH, Seo JH, Park ES, Lim JY, et al. 2019. Neonatal seizures
into the coupling of virion assembly and rotavirus replica- and white matter injury: role of rotavirus infection and
tion. Nat Rev Microbiol. 10:165–177. probiotics. Brain Dev. 41:19–28.
Troeger C, Khalil IA, Rao PC, Cao S, Blacker BF, Ahmed T, Yolken R, Murphy M. 1982. Sudden infant death syndrome
Armah G, Bines JE, Brewer TG, Colombara DV, et al. 2018. associated with rotavirus infection. J Med Virol. 10:
Rotavirus vaccination and the global burden of rotavirus 291–296.
diarrhea among children younger than 5 years. JAMA Yu TH, Tsai CN, Lai MW, Chen CC, Chao HC, Lin CW, Chiu CH,
Pediatr. 172:958–965. Chen SY. 2012. Antigenemia and cytokine expression in
Tsukida K, Goto M, Yamaguchi N, Imagawa T, Tamura D, rotavirus gastroenteritis in children. J Microbiol Immunol
Yamagata T. 2018. Rotavirus gastroenteritis-associated Infect. 45:265–270.
urinary tract calculus in an infant. Turk J Pediatr. 60: Zhaori GT, Fu LT, Xu YH, Guo YR, Peng ZJ, Shan WS. 1991.
769–770. Detection of rotavirus antigen in tracheal aspirates of
CRITICAL REVIEWS IN MICROBIOLOGY 595

infants and children with pneumonia. Chin Med J. 104: Zheng J, Zheng H, Gupta RK, Li H, Shi H, Pan L, Gong S,
830–833. Liang H. 2017. Interrelationship of rotavirus infection and
Zheng BJ, Chang RX, Ma GZ, Xie JM, Liu Q, Liang XR, Ng MH. Creatine Kinase-MB isoenzyme levels in children hospital-
1991. Rotavirus infection of the oropharynx and respira- ized with acute gastroenteritis in Guangzhou, China, 2012-
tory tract in young children. J Med Virol. 34:29–37. 2015. Sci Rep. 7:7674.

You might also like