Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Available online at www.sciencedirect.

com

ScienceDirect

Rotavirus cell entry: not so simple after all


Carlos F Arias and Susana López

Rotaviruses are important agents of severe gastroenteritis in RNA (dsRNA) viruses, composed of a triple-layered
young children, and show a very selective cell and tissue protein capsid that surrounds the viral genome [2,3].
tropism, as well as significant age and host restriction. In the The outermost capsid layer is formed by trimers of
last few years, these properties have been associated with the VP7 that constitute the smooth surface of the virus,
initial interaction of the virus with histo-blood group antigens on and by trimers of the spike protein VP4, which functions
the cell surface, although post-attachment interactions have as the virus attachment protein. VP4 has to be processed
also been found to define the susceptibility to infection of by trypsin-like proteases in the intestinal lumen for the
human enteroids. These initial interactions seem also to virus to become infectious, and this cleavage activation
determine the virus entry pathway, as well as the induction of yields two non-covalently bound subunits, VP8 and VP5
signaling cascades that influence the virus intracellular [2]. Cryo-electron microscopy (CryoEM) studies, includ-
vesicular traffic and escape from endosomes. Here we review ing one at near-atomic resolution, have shown that the C-
the current knowledge of the different stages of the virus entry terminal domains of the three VP5 subunits interact to
journey. form a trimeric foot that is sandwiched between the
intermediate capsid layer formed by VP6, and the VP7
Address outer capsid layer [4–6]. The region of VP4 that protrudes
Instituto de Biotecnologı́a, Universidad Nacional Autónoma de México, out of the VP7 layer does not maintain the local trimeric
Av. Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos, Mexico
symmetry observed in the foot (Figure 1). One of the VP5
Corresponding author: Arias, Carlos F (arias@ibt.unam.mx) monomers forms the spike stalk, while the other two VP5
subunits build the dimeric spike body that extends away
from the particle surface, and is capped by two VP8-lectin
Current Opinion in Virology 2021, 49:42–48
domains. The third VP8-lectin domain is presumed
This review comes from a themed issue on Virus entry to dissociate from the particle after proteolytic activation
Edited by Chelsey Spriggs and Billy Tsai [4–6]. VP8 is responsible for the initial attachment of the
virus to the surface of the target cell, while VP5 is
involved in cell membrane penetration [7]. The
uncleaved rotavirus particles show spikes with a structure
https://doi.org/10.1016/j.coviro.2021.03.011 indistinguishable from that of trypsin-digested particles,
1879-6257/ã 2021 Elsevier B.V. All rights reserved. suggesting that the proteolytical cleavage of VP4 does not
alter the rigid structure of the spike, but allows the
conformational change required for membrane disruption
during virus entry [8].

Receptor binding
Introduction RV cell entry is a complex multistep process that begins
The initial step in a viral infection is the attachment of the with the interaction of the virus particle, through the VP8
virus to specific receptors on the cell surface, an interac- galectin domain of VP4, with glycans on the cell surface
tion that frequently triggers cellular signaling cascades [9–11] (Figure 2, A1). During decades, it was believed
that facilitate either virus entry or replication. In most that sialic acid (SA), as part of glycosphingolipids (gang-
instances, regardless of whether the virus is enveloped or liosides), or as terminal acidic sugars on saccharide
not, virus internalization proceeds through an endocytic branches of glycoproteins, was key for RVs attachment
pathway that allows the incoming viral particle to reach an to the cell surface, since the infectivity of several animal
intracellular compartment where the appropriate cellular RV strains widely used as model viruses was greatly
cues that promote virus uncoating and penetration of the reduced by the removal, before infection, of SAs from
vesicular membrane are present [1]. In this review we will the cell surface using sialidases, and were thus considered
summarize what is currently known about the different to be neuraminidase (NA)-sensitive [7]. In addition,
stages of the rotavirus (RV) entry process to reach its site although it was observed that some animal RV strains
of replication, and the cellular counterparts that influence and most human rotaviruses (HRVs) are NA-resistant
this process. [10], some of these strains do recognize SAs, but at
internal positions in the carbohydrate chain, such that
RVs, the cause of severe dehydrating diarrhea in many they are not trimmed by the activity of NAs [12], reinfor-
mammals and an estimated 200,000 human infant deaths cing the idea of the relevance of SA for RV infection.
annually, are nonenveloped, segmented double-stranded However, in the last ten years evidence has highlighted

Current Opinion in Virology 2021, 48:42–48 www.sciencedirect.com


Rotavirus cell entry Arias and López 43

Figure 1

(a) (b)

Current Opinion in Virology

Atomic structure of the VP4 spike.


(a) Model of the rotavirus particle structure generated from the atomic coordinates of the triple-layered capsid and the viral RNA polymerase VP1 (
purple). (b) Atomic structure of the VP4 spike. Ribbon representation (top), color-coded to represent the spike foot (green), stalk (red), body (red)
and head (purple). As indicated, each of the VP4 subunits (VP4-A, VP4-B and VP4-C) is highlighted in color. The N-terminal and C-terminal regions
of the three molecules, contacting one another beneath the VP7 layer, form the trimeric foot of the structure. The b-barrel domain of the VP5 C
subunit forms the stalk while the VP5 A and -B subunits build the spike body. The head is constructed by the VP8 lectin domains of the A and -B
VP4 subunits. The VP4-C lectin domain is not present in the structure, and is presumed to be released during proteolysis. The VP6 and VP7 layers
are depicted, as their 3D structure, in blue and yellow, respectively. Diagram of the VP4 domain organization (bottom), color-coded as in top.
Residues delimiting domains and trypsin cleavage sites are indicated. Modified from Rodrı́guez et al. doi: https://doi.org/10.1371/journal.ppat.
1004157.g001.

the relevant role of histo-blood group antigens (HBGA), neonates, identified a binding site for H-type I glycan
non-sialylated glycoconjugates present on the surface of formed by one of the b-sheets and the C-terminal a-helix
red blood cells, epithelial cells, and in mucosal secretions of P[6] VP8, similar to the binding site described for the P
[13], as RV cell attachment and susceptibility factors [11]. [19] VP8, offering clues about the age-restricted tropism
The X-ray crystallographic structure of the VP8 subunit of some neonatal RV strains [16]. More recently, the H-
of several NA-sensitive and NA-resistant strains has been type 1 binding site in P[8] VP8 was determined by X-ray
determined, and the crystal structure of some of these crystallography, and variations in affinities in different P
proteins in complex with glycans has been reported [8] lineages were found, possibly reflecting subtle struc-
(references in Refs. [10,14]). It has been found that RV tural differences in the binding pocket, and providing a
VP8 binds different glycans in a genotype-dependent fine-tuning mechanism for glycan binding in P[8] RV
manner, and this interaction can be even strain-specific. strains [17,18]. Also, it has been described that [P4] and P
In general, the common theme is that glycans interact [8] VP8s can recognize Leb HBGA glycans in still an
with an open-ended, shallow groove on the galectin fold additional previously undescribed pocket [19]. Alto-
of VP8 formed by two b-sheets, but variations in its width gether, these findings suggest complex and variable
and the amino acid residues exposed, as well as the attachment factor binding patterns that may influence
specific binding site in the groove, determine the type cell susceptibility to infection, as well as age, tissue and
of glycans that can be accommodated, and thus the host restriction, and possibly host susceptibility to specific
genotype specificity [10,11]. However, it was recently RV strains [10,16]. In contrast with these observations,
shown that the P[19] VP8 binds to type 1 glycans at a both NA-sensitive and NA-resistant animal and human
novel binding site, completely different from the previ- RV strains bind to a wide variety of cell lines, although
ously characterized A-type or precursor binding sites in only a subset of them is efficiently infected [20], suggest-
RVs [15]. Furthermore, the VP8 domain of the globally ing that the initial VP8/glycan interaction of rotavirus with
dominant HRV P[4] and P[8] VP4 genotypes was found to the cell surface is promiscuous, and that relevant inter-
bind to A-HBGA, H-HBGA, and B-HBGA types, and the actions for cell infection specificity may occur at a post-
crystal structure of the VP8 of a P[6] RV isolated from binding step.

www.sciencedirect.com Current Opinion in Virology 2021, 48:42–48


44 Virus entry

Figure 2

(a)
2 VP2 VP7

VP6 VP4

Lipid raft
3
Terminal and HBGAs Co-receptors
(b)
sub-terminal SA Dynamin

Clathrin
Actin
filaments
E-P viruses
4 1
Viral transcripts ACTN4
CDC42
5 EE RhoA

Drebrin

Ca++
ILV ESCRT
EEA1
Rab5
vATPase
2
ME Rab7
M6PR

Cathepsins 3
6
Viral transcripts
LE ILV
M6PR
Rab9

L-P viruses
7 TNG

Current Opinion in Virology

Models for RV cell entry.


(a) Classical endocytic pathway. A1, RV cell entry begins with the interaction of VP8 with glycans on the cell surface. Terminal or subterminal SAs,
as well as HBGAs serve as attachment receptors for the viral particle. A2, after initial attachment, RVs have post-binding interactions with integrins
a2b1, aVb3, and aXb2, and with hsc70 that are embedded in lipid rafts; JAM-A and occludin have also been involved at this step; a novel,
human-specific post-attachment cell molecule has also been proposed. A3, Several RV strains are internalized by clathrin-dependent endocytosis,
in a dynamin-dependent and cholesterol-dependent process. RRV strain enters cells by an endocytic pathway that is clathrin-independent and
caveolin-independent, and depends on RhoA, Cdc42, and actinin-4 (ACTN4), but is independent of endosomal acidification. A4, After
internalization, all RV strains reach early endosomes (EEs), and depend on a functional ESCRT system, including the ESCRT-associated ATPase
VPS4A involved in fission of intralumenal vesicles in maturating endosomes (MEs). A5, Once in the MEs, different RV strains follow different routes
to reach the cytosol. Early penetrating (E-P) viruses exit from ME vesicles, since their infectivity is not dependent on the GTPase Rab7. A6, Other
strains depend on the expression of Rab7 and thus these viruses continue a deeper journey to reach late endosomes (LEs) that provide the
environment for these—Late-penetrating (L-P) strains—to enter the cytosol. A7, Cellular factors (Rab9, M6PR, and cathepsins), transported from
the trans-Golgi network (TGN) to LEs, facilitate RV cell infection. (b) An alternative pathway for virus internalization. B1, RV interacts with
ganglioside receptors and this interaction drives an invagination process that results in virus internalization entry in tight-fitting membrane vesicles.
B2, There is a drop of Ca2+ concentration within the tight-fitting vesicles vesicle, causing the release of the Ca2+-dependent VP7 trimer contacts;
VP5 folds back to a post-cleavage conformation, disrupting the membrane vesicle. B3, The DLP is liberated into the cytosol, where it initiates RNA
transcription. After entering the cells through either of the entry pathways described, the transcribed mRNAs translate into the 12 encoded viral
proteins which, after accumulating to a critical amount, Form the virus factories, known as viroplasms, where the replication of the genomic RNA
and initial assembly of the viral progeny occurs.

Current Opinion in Virology 2021, 48:42–48 www.sciencedirect.com


Rotavirus cell entry Arias and López 45

Post-binding interactions Despite all these observations, identification of a protein-


Several sequential interactions involving VP5 and VP7, aceous receptor or coreceptor essential for the infectivity
relevant for cell infection, have been reported in the of either animal or human RVs has been elusive. These
frequently employed monkey kidney epithelial MA104 findings are puzzling and suggest that either a more
cells, and human colonic Caco-2 cells. RVs interact with relevant entry factor has not been found for RV, the virus
integrins a2b1, aVb3, and aXb2 and with the heat shock has the plasticity to use more than one route of entry, or
cognate protein hsc70, embedded in detergent resistant the entry factors for RV are redundant.
membrane domains, and consequently RV entry has been
found to be sensitive to membrane cholesterol depletion Cell internalization
[7,21] (Figure 2, A2). More recently, the tight-junction Many animal and human RV strains are internalized into
proteins JAM-A and occludin were also found to be non-polarized and polarized cells by clathrin-dependent
involved in cell infection of some RV strains at a post- endocytosis [32,33], in a process that is dependent on
attachment step [22]. Of note, gangliosides can also play a dynamin and on the presence of cholesterol on the cell
role at a post-binding step, in addition to the role they surface [29,33,34,35,36,37]. In contrast, the simian RRV
have as attachment factors for both NA-sensitive and NA- strain, which has been widely used as a model virus to
resistant RV strains [23]. Interestingly, recent X-ray crys- study RV biology, seems to be an outlier, since it enters
tallographic and cryo-EM analyses of RV structural cells by an endocytic pathway that is independent of
proteins and double-layered (DLP) and triple-layered clathrin and caveolin, although it is also dependent on
(TLP) particles have shown that the proposed VP7 sites dynamin and cholesterol [33]. RRV entry also depends on
for interaction with avb3 and aXb2, as well as the the activity of the small GTPases RhoA and Cdc42, and
interaction site for hsc70 on VP5, are not exposed on on actinin-4 [36,38,39] (Figure 2, A3). Of interest, the
TLPs, indicating that they could only be accessible after endocytic route employed, whether clathrin-dependent
or during the uncoating process of the virus [14,24]. or clathrin-independent, is determined by the spike pro-
Furthermore, it was observed that the interaction of VP5 tein VP4 [32], and is not defined by the initial VP8/glycan
with the a2 subunit of integrin a2b1 depends on a interaction since, with exception of RRV, both NA-sen-
conformational change of VP5 [6,14,24]. This confor- sitive and NA-resistant strains, including those that bind
mational change was suggested to be present in the RRV HBGAs, enter cells through clathrin-mediated endocyto-
NA-resistant nar3 mutant VP5 domain, since this virus sis [32]. The VP4/cell factor interaction that defines the
binds directly to integrin a2b1, obviating the need for the endocytic pathway used has yet to be determined,
VP8-glycan interaction [25,26]. although it appears to occur after initial virus binding.

More recently, a collection of RV VP8-specific recombinant The dynamin-dependent endocytic entry of RV strains is
mAbs, generated from human, intestinal antibody-secret- reinforced by the observation that the actin-binding pro-
ing B cells, were found to be non-neutralizing for P[4], P[6] tein drebrin inhibits RV entry in a virus strain-indepen-
and P[8] HRV strains when assayed in MA104 cells [27]. dent manner through suppressing dynamin-mediated
However, unexpectedly, these ‘non-neutralizing’ antibo- endocytosis [35] (Figure 1, A3). Dbn1 KO mice infected
dies did neutralize P[8] human rotaviruses in enteroid with RRV exhibited higher incidence of diarrhea and
cultured cells as well as in the human epithelial colonic more viral antigen shedding compared with the wild-type
cell line HT-29 [28]. The X-ray crystallographic structure littermates, suggesting a negative factor role for Dbn1 in
of a P[4] VP8, in complex with a single-chain fragment of vivo. Furthermore, pharmacological inhibition of Dbn1
one of these MAbs (MAb9), identified an interaction site significantly promoted RV infection in human intestinal
located away from the HBGA binding site [28] recently enteroids [35,40], suggesting that a dynamin-dependent
described for a P[4] VP8/H type 1 antigen complex [16]. endocytic process also occurs in the human gut.
The observation that mAb9 does not block VP8 binding to
HBGA, suggests that it blocks a virus–cell interaction that In an alternative pathway for virus internalization, it was
occurs only in human intestinal cells (not in MA104 cells), reported that RRV interacts with ganglioside receptors on
and points to a specific RV receptor that could be recog- the surface of BSC-1 cells and rapidly starts an invagina-
nized after the initial VP8-glycan interaction [28]. The tion process driven by the virus particle itself that results
existence of a specific post-attachment receptor is addition- in its engulfment in tight-fitting membrane endocytic
ally supported by the observation that rotavirus infection of vesicles (Figure 2, B1). This process was found to be
polarized cell monolayers, including human intestinal independent of dynamin and clathrin, and was completed
enteroids, occurs preferentially through the basolateral in about 10 min. [24]. In this study, the uncoating
membrane [28,29,30]. The access of virions to putative process of TLPs was followed by live-cell fluorescence
coreceptors in the basolateral side could be facilitated by imaging, and was detected as a rapid diffusional motion of
signaling through the RhoA/ROCK/MLC pathway, the DLP away from residual outer-layer proteins; this
induced by virus attachment to cell receptors on the apical event was interpreted as release of the DLP from the
side, that mediates the disruption of tight junctions [31]. endocytic vesicle and productive entry of the virus [24].

www.sciencedirect.com Current Opinion in Virology 2021, 48:42–48


46 Virus entry

In contrast with these findings, when the read out was an accumulation of cholesterol within LE compartments alter-
infectivity assay, it was found that RRV enters MA104 ing their function, and have also been shown to significantly
cells with a half-time of 35 min [33]. The different impair the infection of influenza A virus, vesicular stomatitis
observations regarding the use of dynamin for virus virus, and dengue virus [47–49]. So, it is suggested that the
endocytosis, the kinetics of virus internalization, and substantial accumulation of cholesterol in the LE compart-
the route proposed for productive entry might reflect ment decreases the exit of L-P RVs from these vesicles,
the predominance of different pathways depending on thereby arresting cytoplasmic virus replication [46].
the experimental conditions, the cells, or the virus strains
employed. Alternatively, the biological relevance of the The transport of cellular factors from the trans-Golgi
different proposed models of virus internalization could network (TGN) to LEs also seem to facilitate RV cell
be different. infection. Newly synthesized lysosomal acid hydrolases
are delivered from the TGN to endosomes by mannose-6-
Intracellular vesicular traffic phosphate receptors (M6PRs), and recycling of M6PRs
When infectivity assays have been used as the readout for back to the Golgi network depends on the small GTPase
productive RV infection, endocytosis through a cholesterol Rab9 (18). In this regard, most L-P viruses depend on the
and dynamin dependent process has been identified as the activities of Rab9, the cation-dependent M6PR, and
route of entry. In this pathway, regardless the cell surface cathepsins B, L, and S, suggesting that these acidic
receptor recognized, and the requirement of clathrin for cell hydrolases facilitate the exit of transcriptionally active
internalization [32,33,36,37,41], all RV strains reach early L-P viruses into the cytoplasm (Figure 2, A7). Endoly-
endosomes (EEs) as a first step (Figure 2, A4) [36,41,42]. All sosomal cysteine cathepsins are also required for proces-
RVs tested also require a functional ESCRT system, sing the capsid or the surface proteins of other viruses,
including the activity of the ESCRT-associated ATPase such as reovirus, Ebola virus, severe acute respiratory
VPS4A involved in the formation of endosomal intralum- syndrome (SARS) coronavirus, and Nipah virus (see
inal vesicles in maturating endosomes (MEs) [36]. After references in Ref. [41]).
reaching MEs, different RV strains follow different routes
to reach the cytosol. Early penetrating (E-P) viruses, like Membrane penetration
RRV and SA11-4S, exit from ME vesicles (Figure 2, A5), To reach the cytosol and initiate replication within the
since their infectivity does not depend on the GTPase cell, RVs must traverse a lipid membrane, whether this
Rab7 [36,41]. The infectivity of other RV strains tested being the plasma membrane or the membrane of an
depends, at least partially, on the expression of this endocytic vesicle. Based on a detailed structure of infec-
GTPase, suggesting that these viruses continue a deeper tious RRV, a mechanistic model for RV entry has been
journey into the cell to reach late endosomes (LEs) that proposed [6,24,50]. In this model, after a trypsin-acti-
likely provide the optimal environment for these strains to vated virus particle is endocytosed in tight-fitting vesi-
enter the cytosol [41,43] (Figure 2, A6). As noted for cell cles, there is a drop of Ca2+ concentration within the
internalization, the spike VP4 also determines the differ- vesicle, possibly mediated by the interaction of hydro-
ential trafficking of RVs; a single amino acid change in VP4 phobic loops on VP5 with the vesicular membrane, that
can dictate not only the pathway of endocytosis used but causes the loosening of the Ca2+-dependent VP7 trimer
also whether the virus reaches EEs or LEs during the entry contacts, releasing this protein from the viral particle and
process [41]. allowing VP5 to fold back to its most stable, post-cleavage
conformation (Figure 2, B2), rearrangement that disrupts
Escape from late endosomes the vesicular membrane and liberates the DLPs into the
Several conditions have been reported to be important for the cytosol to begin transcription [24] (Figure 2, B3). Using
efficient escape of RVs from LEs. Late-penetrating (L-P) reconstituted RRV TLPs with VP4 labeled with a fluo-
human DS-1 and bovine NCDV RV strains stimulate the rescent Ca2+ sensor, it was observed that after engulf-
PI3K/Akt and MEK/ERK pathways very early in infection; ment of the virus particle is completed, a slow (1-min
specific inhibitors of these signaling pathways do not prevent duration) loss of Ca2+ precedes the onset of VP7 dissoci-
virus intracellular trafficking, but trap the viral particles in ation by about 2 min and DLP release by about 7 min,
LEs, preventing their exit to the cytosol [44]. As a conse- supporting the model for the molecular mechanism of
quence, the infectious viral progeny is reduced 100–1000- membrane disruption by RV [50]. In a related study, it
fold. RV-induced phosphorylated PI3K, Akt, and ERK was described that viral RNA synthesis can be detected as
directly interact with the subunit E of the V-ATPase proton early as 15 min after virus adsorption [51]. Regardless the
pump [44], required for endosomal acidification and uncoat- factors that facilitate RV membrane penetration previ-
ing of L-P, but not E-P RV strains such as RRV [36,37,44,45]. ously discussed for different rotavirus strains, and the
In addition to an acidic pH, it has been shown that 25- vesicular compartment from which DLPs are liberated
hydroxycholesterol and 27-hydroxycholesterol inhibit into the cytosol, the mechanism of membrane disruption
human rotavirus infection by sequestering viral particles into driven by a conformational change of VP5 explains all
late endosomes [46]. These oxysterols cause the available biochemical, microscopy and structural data.

Current Opinion in Virology 2021, 48:42–48 www.sciencedirect.com


Rotavirus cell entry Arias and López 47

Conclusions 6. Settembre EC, Chen JZ, Dormitzer PR, Grigorieff N, Harrison SC:
Atomic model of an infectious rotavirus particle. EMBO J 2011,
Advances in the complex process of RV cell entry have 30:408-416.
been challenging, and many questions remain. Is there a 7. Arias CF, Silva-Ayala D, Lopez S: Rotavirus entry: a deep journey
critical cellular receptor for virus entry? To what extent into the cell with several exits. J Virol 2015, 89:890-893.
post-attachment interactions are required for RV infec- 8. Rodriguez JM, Chichon FJ, Martin-Forero E, Gonzalez-
tion? What is the VP4/cell interaction that determines the Camacho F, Carrascosa JL, Caston JR, Luque D: New insights
into rotavirus entry machinery: stabilization of rotavirus spike
route of virus entry and intracellular trafficking? What are conformation is independent of trypsin cleavage. PLoS Pathog
the cellular cues required for the escape of E-P and L-P 2014, 10:e1004157.
viruses from endocytic vesicles? Are there more than one 9. Dormitzer PR, Sun ZY, Wagner G, Harrison SC: The rhesus
pathway of virus productive entry? Do RV strains other rotavirus VP4 sialic acid binding domain has a galectin fold with
a novel carbohydrate binding site. EMBO J 2002, 21:885-897.
than RRV are internalized through the tight-fitting vesi-
cle model? To what extent the VP8/glycan interaction 10. Ramani S, Hu L, Venkataram Prasad BV, Estes MK: Diversity in
rotavirus-host glycan interactions: a "sweet" spectrum. Cell
defines the host susceptibility to RV infection? Are other Mol Gastroenterol Hepatol 2016, 2:263-273.
viral or host factors involved? Does genotype-dependent 11. Venkataram Prasad BV, Shanker S, Hu L, Choi JM, Crawford SE,
variation of glycan recognition have implications for tis- Ramani S, Czako R, Atmar RL, Estes MK: Structural basis of
sue and host restriction? Virus tracking by live-cell imag- glycan interaction in gastroenteric viral pathogens. Curr Opin
Virol 2014, 7:119-127.
ing systems coupled with novel superresolution micros-
copy techniques, application of the recently implemented 12. Haselhorst T, Fleming FE, Dyason JC, Hartnell RD, Yu X,
Holloway G, Santegoets K, Kiefel MJ, Blanchard H, Coulson BS
reverse genetic system for manipulating the rotavirus et al.: Sialic acid dependence in rotavirus host cell invasion.
genome, as well as the availability of human intestinal Nat Chem Biol 2009, 5:91-93.
enteroids that represent a system that more faithfully 13. Marionneau S, Cailleau-Thomas A, Rocher J, Le Moullac-
Vaidye B, Ruvoen N, Clement M, Le Pendu J: ABH and Lewis
mimics virus infection of enterocytes in the human gut, histo-blood group antigens, a model for the meaning of
should allow us to gain insight into some of these ques- oligosaccharide diversity in the face of a changing world.
tions. The continuous use of RNAi and CRISPR)/Cas9 Biochimie 2001, 83:565-573.
technology, high resolution structural information of the 14. Rodriguez JM, Luque D: Structural insights into rotavirus entry.
Adv Exp Med Biol 2019, 1215:45-68.
rotavirus capsid proteins in complex with glycans or other
cell surface molecules, cryo-electron microscopy (Cryo- 15. Liu Y, Xu S, Woodruff AL, Xia M, Tan M, Kennedy MA, Jiang X:
 Structural basis of glycan specificity of P[19] VP8*:
EM) and cryo-electron tomography (Cryo-ET) analyses implications for rotavirus zoonosis and evolution. PLoS Pathog
should also contribute to this end. 2017, 13:e1006707.
Description of a novel glycan binding site in P[19] VP8*, distinct from the
previously characterized A-type or precursor binding sites in RVs.
Conflict of interest statement 16. Hu L, Sankaran B, Laucirica DR, Patil K, Salmen W, Ferreon ACM,
Nothing declared.  Tsoi PS, Lasanajak Y, Smith DF, Ramani S et al.: Glycan
recognition in globally dominant human rotaviruses. Nat
Commun 2018, 9:2631.
Describes an H type-1 binding site that is conserved in the globally
Acknowledgements dominant P[4], P[6], and P[8] genotypes of human RVs. Provides a
structural basis for the age-restricted tropism of some P[6] RVs.
The authors would like to acknowledge support from grants A1-S-15356 and
Fordecyt 302965 from the National Council for Science and Technology, 17. Gozalbo-Rovira R, Ciges-Tomas JR, Vila-Vicent S, Buesa J,
CONACyT-Mexico (SL). We thank Daniel Luque and Javier M. Rodrı́guez  Santiso-Bellon C, Monedero V, Yebra MJ, Marina A, Rodriguez-
for kindly providing Figure 1. Diaz J: Unraveling the role of the secretor antigen in human
rotavirus attachment to histo-blood group antigens. PLoS
Pathog 2019, 15:e1007865.
Determination of the H-type 1 binding site in P[8] VP8. Postulate that
References and recommended reading variations in affinities in different P[8] lineages may provide a fine-tuning
Papers of particular interest, published within the period of review, mechanism for glycan binding in P[8] RV strains.
have been highlighted as:
18. Sun X, Li D, Peng R, Guo N, Jin M, Zhou Y, Xie G, Pang L, Zhang Q,
 of special interest Qi J et al.: Functional and structural characterization of P[19]
 of outstanding interest rotavirus VP8* interaction with histo-blood group antigens. J
Virol 2016, 90:9758-9765.
1. Cossart P, Helenius A: Endocytosis of viruses and bacteria. 19. Xu S, Ahmed LU, Stuckert MR, McGinnis KR, Liu Y, Tan M,
Cold Spring Harb Perspect Biol 2014, 6.  Huang P, Zhong W, Zhao D, Jiang X et al.: Molecular basis of P[II]
2. Estes MKG: Rotaviruses.. H.B. In Fields Virology, edn 6, vol 2. major human rotavirus VP8* domain recognition of histo-
Edited by Knipe DMH, Howley PM. Wolters Kluwer Health/ blood group antigens. PLoS Pathog 2020, 16:e1008386.
Lippincott Williams & Willkins; 2013:1347-1401. Reveals a binding site for Leb HBGA glycans in the human P[8] and P[4]
RV VP8 domains, distinct from the site on these proteins that binds H type
3. Tate JE, Burton AH, Boschi-Pinto C, Parashar UD, World Health 1 gycans.
Organization-Coordinated Global Rotavirus Surveillance Network:
Global, regional, and national estimates of rotavirus mortality 20. Ciarlet M, Crawford SE, Cheng E, Blutt SE, Rice DA, Bergelson JM,
in children <5 years of age, 2000-2013. Clin Infect Dis 2016, 62 Estes MK: VLA-2 (alpha2beta1) integrin promotes rotavirus
(Suppl. 2):S96-S105. entry into cells but is not necessary for rotavirus attachment. J
Virol 2002, 76:1109-1123.
4. Li Z, Baker ML, Jiang W, Estes MK, Prasad BV: Rotavirus architecture
at subnanometer resolution. J Virol 2009, 83:1754-1766. 21. Coulson BS, Londrigan SL, Lee DJ: Rotavirus contains integrin
ligand sequences and a disintegrin-like domain that are
5. Prasad BV, Wang GJ, Clerx JP, Chiu W: Three-dimensional implicated in virus entry into cells. Proc Natl Acad Sci U S A
structure of rotavirus. J Mol Biol 1988, 199:269-275. 1997, 94:5389-5394.

www.sciencedirect.com Current Opinion in Virology 2021, 48:42–48


48 Virus entry

22. Torres-Flores JM, Silva-Ayala D, Espinoza MA, Lopez S, Arias CF: 38. Sanchez-San Martin C, C.Sanchez-San MartinLopez T, T.
The tight junction protein JAM-A functions as coreceptor for LopezArias CF, C.F.AriasLopez S: S.LopezCharacterization of
rotavirus entry into MA104 cells. Virology 2015, 475:172-178. rotavirus cell entry. J Virol 2004, 78:2310-2318.
23. Martinez MA, Lopez S, Arias CF, Isa P: Gangliosides have a functional 39. Trejo-Cerro O, O.Trejo-CerroAguilar-Hernandez N, N.Aguilar-
role during rotavirus cell entry. J Virol 2013, 87:1115-1122. HernandezSilva-Ayala D, D.Silva-AyalaLopez S, S.LopezArias CF:
C.F.AriasThe actin cytoskeleton is important for rotavirus
24. Abdelhakim AH, Salgado EN, Fu X, Pasham M, Nicastro D, internalization and RNA genome replication. Virus Res 2019,
 Kirchhausen T, Harrison SC: Structural correlates of rotavirus 263:27-33.
cell entry. PLoS Pathog 2014, 10:e1004355.
Novel molecular and high-resolution structural approaches are applied to 40. Finkbeiner SR, S.R.FinkbeinerZeng XL, X.L.ZengUtama B, B.
provide evidence for a molecular model of membrane disruption during  UtamaAtmar RL, R.L.AtmarShroyer NF, N.F.ShroyerEstes MK: M.
RRV cell entry. K.EstesStem cell-derived human intestinal organoids as an
infection model for rotaviruses. mBio 2012, 3:e00159-00112.
25. Zarate S, Espinosa R, Romero P, Guerrero CA, Arias CF, Lopez S: First description of intestinal organoids for infection with human
Integrin alpha2beta1 mediates the cell attachment of the rotaviruses.
rotavirus neuraminidase-resistant variant nar3. Virology 2000,
278:50-54. 41. Diaz-Salinas MA, M.A.Diaz-SalinasSilva-Ayala D, D.Silva-
AyalaLopez S, S.LopezArias CF: C.F.AriasRotaviruses reach late
26. Méndez E, Arias CF, López S: Binding to sialic acids is not an endosomes and require the cation-dependent mannose-6-
essential step for the entry of animal rotaviruses to epithelial phosphate receptor and the activity of cathepsin proteases to
cells in culture. J Virol 1993, 67:5253-5259. enter the cell. J Virol 2014, 88:4389-4402.
27. Nair N, Feng N, Blum LK, Sanyal M, Ding S, Jiang B, Sen A, 42. Wolf M, M.WolfDeal EM, E.M.DealGreenberg HB: H.B.
Morton JM, He XS, Robinson WH et al.: Sci Transl Med 2017, 9. GreenbergRhesus rotavirus trafficking during entry into MA104
28. Feng N, Hu L, Ding S, Sanyal M, Zhao B, Sankaran B, Ramani S, cells is restricted to the early endosome compartment. J Virol
 McNeal M, Yasukawa LL, Song Y et al.: Human VP8* mAbs 2012, 86:4009-4013.
neutralize rotavirus selectively in human intestinal epithelial 43. Diaz-Salinas MA, M.A.Diaz-SalinasCasorla LA, L.A.
cells. These MAbs neutralize only in human intestinal cells (not in CasorlaLopez T, T.LopezLopez S, S.LopezArias CF: C.F.
MA104 cells), and point to a specific RV receptor that could be AriasMost rotavirus strains require the cation-independent
recognized after the initial VP8-glycan interaction.. mannose-6-phosphate receptor, sortilin-1, and cathepsins to
29. Cevallos Porta D, D.Cevallos PortaLopez S, S.LopezArias CF, C.F. enter cells. Virus Res 2018, 245:44-51.
AriasIsa P: P.IsaPolarized rotavirus entry and release from
44. Soliman M, M.SolimanSeo JY, J.Y.SeoKim DS, D.S.KimKim JY, J.
differentiated small intestinal cells. Virology 2016, 499:65-71.
 Y.KimPark JG, J.G.ParkAlfajaro MM, M.M.AlfajaroBaek YB, Y.B.
30. Realpe M, M.RealpeEspinosa R, R.EspinosaLopez S, S. BaekCho EH, E.H.ChoKwon J, J.KwonChoi JS J.S.Choiet al.:
LopezArias CF: C.F.AriasRotaviruses require basolateral Activation of PI3K, Akt, and ERK during early rotavirus
molecules for efficient infection of polarized MDCKII cells. infection leads to V-ATPase-dependent endosomal
Virus Res 2010, 147:231-241. acidification required for uncoating. PLoS Pathog 2018, 14:
e1006820.
31. Soliman M, M.SolimanCho EH, E.H.ChoPark JG, J.G.ParkKim JY, Identification of the early virus induction of PI3K/Akt and MEK/ERK
J.Y.KimAlfajaro MM, M.M.AlfajaroBaek YB, Y.B.BaekKim DS, D.S. signaling pathways to facilitate the exit of L-P RV strains from late
KimKang MI, M.I.KangPark SI, S.I.ParkCho KO: K.O. endosomes into the cytoplasm.
ChoRotavirus-induced early activation of the RhoA/ROCK/
MLC signaling pathway mediates the disruption of tight 45. Chemello ME, M.E.ChemelloAristimuno OC, O.C.
junctions in polarized MDCK cells. Sci Rep 2018, 8:13931. AristimunoMichelangeli F, F.MichelangeliRuiz MC: M.C.
RuizRequirement for vacuolar H+ -ATPase activity and Ca2+
32. Diaz-Salinas MA, M.A.Diaz-SalinasRomero P, P. gradient during entry of rotavirus into MA104 cells. J Virol
RomeroEspinosa R, R.EspinosaHoshino Y, Y.HoshinoLopez S, S. 2002, 76:13083-13087.
LopezArias CF: C.F.AriasThe spike protein VP4 defines the
endocytic pathway used by rotavirus to enter MA104 cells. J 46. Civra A, A.CivraFrancese R, R.FranceseGamba P, P.
Virol 2013, 87:1658-1663. GambaTesta G, G.TestaCagno V, V.CagnoPoli G, G.PoliLembo D:
D.Lembo25-Hydroxycholesterol and 27-hydroxycholesterol
33. Gutierrez M, M.GutierrezIsa P, P.IsaSanchez-San Martin C, C. inhibit human rotavirus infection by sequestering viral
Sanchez-San MartinPerez-Vargas J, J.Perez-VargasEspinosa R, particles into late endosomes. Redox Biol 2018, 19:318-330.
R.EspinosaArias CF, C.F.AriasLopez S: S.LopezDifferent
rotavirus strains enter MA104 cells through different 47. Amini-Bavil-Olyaee S, S.Amini-Bavil-OlyaeeChoi YJ, Y.J.
endocytic pathways: the role of clathrin-mediated ChoiLee JH, J.H.LeeShi M, M.ShiHuang IC, I.C.HuangFarzan M,
endocytosis. J Virol 2010, 84:9161-9169. M.FarzanJung JU: J.U.JungThe antiviral effector IFITM3
disrupts intracellular cholesterol homeostasis to block viral
34. Green VA, V.A.GreenPelkmans L: L.PelkmansA systems survey entry. Cell Host Microbe 2013, 13:452-464.
of progressive host-cell reorganization during rotavirus
infection. Cell Host Microbe 2016, 20:107-120. 48. Lakadamyali M, M.LakadamyaliRust MJ, M.J.RustZhuang X: X.
ZhuangEndocytosis of influenza viruses. Microbes Infect 2004,
35. Li B, B.LiDing S, S.DingFeng N, N.FengMooney N, N. 6:929-936.
 MooneyOoi YS, Y.S.OoiRen L, L.RenDiep J, J.DiepKelly MR, M.R.
KellyYasukawa LL, L.L.YasukawaPatton JT J.T.Pattonet al.: 49. Pattanakitsakul SN, S.N.PattanakitsakulPoungsawai J, J.
Drebrin restricts rotavirus entry by inhibiting dynamin- PoungsawaiKanlaya R, R.KanlayaSinchaikul S, S.
mediated endocytosis. Proc Natl Acad Sci U S A 2017, 114: SinchaikulChen ST, S.T.ChenThongboonkerd V: V.
E3642-E3651. ThongboonkerdAssociation of Alix with late endosomal
Reports that the actin-binding protein drebrin inhibits RV entry in a virus lysobisphosphatidic acid is important for dengue virus
strain-independent manner through suppressing dynamin-mediated infection in human endothelial cells. J Proteome Res 2010,
endocytosis. It functions in enteroids and in the mouse model. 9:4640-4648.
36. Silva-Ayala D, D.Silva-AyalaLopez T, T.LopezGutierrez M, M. 50. Salgado EN, E.N.SalgadoGarcia Rodriguez B, B.Garcia
GutierrezPerrimon N, N.PerrimonLopez S, S.LopezArias CF: C.F.  RodriguezNarayanaswamy N, N.NarayanaswamyKrishnan Y, Y.
AriasGenome-wide RNAi screen reveals a role for the ESCRT KrishnanHarrison SC: S.C.HarrisonVisualization of calcium ion
complex in rotavirus cell entry. Proc Natl Acad Sci U S A 2013, loss from rotavirus during cell entry. J Virol 2018, 92.
110:10270-10275. Detailed kinetics of Ca2+ escape from tight endocytic vesicles that supports
the model for the molecular mechanism of membrane disruption by RV.
37. Wolf M, M.WolfVo PT, P.T.VoGreenberg HB: H.B.
GreenbergRhesus rotavirus entry into a polarized epithelium is 51. Salgado EN, E.N.SalgadoUpadhyayula S, S.
endocytosis dependent and involves sequential VP4 UpadhyayulaHarrison SC: S.C.HarrisonSingle-particle detection
conformational changes. J Virol 2011, 85:2492-2503. of transcription following rotavirus entry. J Virol 2017, 91.

Current Opinion in Virology 2021, 48:42–48 www.sciencedirect.com

You might also like