ArticlePDF AvailableLiterature Review

Abstract and Figures

Parvoviruses are small non‐enveloped single‐stranded DNA viruses, which depend on host cell nuclear transcriptional and replication machinery. After endosomal exposure of nuclear localization sequence and a phospholipase A2 domain on the capsid surface, and escape into the cytosol, parvovirus capsids enter the nucleus. Due to the small capsid diameter of 18–26 nm, intact capsids can potentially pass into the nucleus through nuclear pore complexes (NPCs). This might be facilitated by active nuclear import, but capsids may also follow an alternative entry pathway that includes activation of mitotic factors and local transient disruption of the nuclear envelope. The nuclear entry is followed by currently undefined events of viral genome uncoating. After genome release, viral replication compartments are initiated and infection proceeds. Parvoviral genomes replicate during cellular S phase followed by nuclear capsid assembly during virus‐induced S/G2 cell cycle arrest. Nuclear egress of capsids occurs upon nuclear envelope degradation during apoptosis and cell lysis. An alternative pathway for nuclear export has been described using active transport through the NPC mediated by the chromosome region maintenance 1 protein, CRM1, which is enhanced by phosphorylation of the N‐terminal domain of VP2. However, other alternative but not yet uncharacterized nuclear export pathways cannot be excluded. Parvovirus capsids enter the nucleus by active import or by an alternative entry pathway that includes local transient disruption of the nuclear envelope. Parvoviral genomes replicate during cellular S phase followed by nuclear capsid assembly. The egress of progeny capsids occurs upon nuclear envelope degradation during cell lysis or by using active transport through the nuclear pore complexes.
This content is subject to copyright. Terms and conditions apply.
Molecular Microbiology. 2022;118:295–308.
|
295wileyonlinelibrary.com/journal/mmi
1 |INTRODUCTION
1.1  | Parvoviruses
Parvovirinae subfamily infects vertebrates including humans. Most
viruses of this subfamily, including minute virus of mice (MVM),
canine parvovirus (CPV), and rat parvovirus (H- 1PV), are autono-
mous, but replication of adeno- associated viruses (AAV) requires the
presence of helper viruses such as adenoviruses or herpesviruses
(dependoparvoviruses) (Cotmore et al., 2019; Pénzes et al., 2020).
While Parvovirinae have an important potential in oncolytic therapy,
AAVs are a major platform in gene therapy. H- 1PV and MVM are
Received: 23 May 2022 
|
Revised: 10 August 2022 
|
Accepted: 13 August 2022
DOI : 10.1111/m mi.14974
MICRO REVIEW
Nuclear entry and egress of parvoviruses
Salla Mattola1| Vesa Aho1| Luisa F. Bustamante- Jaramillo2| Edoardo Pizzioli2|
Michael Kann2,3,4| Maija Vihinen- Ranta 1
This is an op en access ar ticle under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provide d the original work is properly cited.
© 2022 The Authors. Molecular Microbiology published by John Wiley & Sons Ltd.
Michael Kann co- last author.
1Department of Biological and
Environmental Science, University of
Jyvaskyla, Jyvaskyla, Finland
2Department of Infectious Diseases,
Institute of Biomedicine, University of
Gothenburg, Gothenburg, Sweden
3Sahlgrenska Academy, Gothenburg,
Sweden
4Department of Clinical Microbiology,
Region Väs tra Götaland, Sahlgrenska
University Hospital, Gothenburg, Sweden
Correspondence
Michael Kann, Department of Infectious
Diseases, Institute of Biomedicine,
University of Gothenburg, Gothenburg ,
Sweden.
Email: michael.kann@gu.se
Maija Vihinen- Ranta, Department of
Biological and Environmental Science,
University of Jyvaskyla, P O Box 35, 40014
Jyvaskyla, Finland.
Email: maija.vihinen-ranta@jyu.fi
Funding information
Academy of Finland, G rant/Award
Number : 330896; Gr aduate School of
the Unive rsity of Jy vaskyla, Grant/Award
Number : 2022; Jane ja A atos Erkon Säätiö,
Grant/Award Number: 2019
Abstract
Parvoviruses are small non- enveloped single- stranded DNA viruses, which depend
on host cell nuclear transcriptional and replication machinery. After endosomal expo-
sure of nuclear localization sequence and a phospholipase A2 domain on the capsid
surface, and escape into the cytosol, parvovirus capsids enter the nucleus. Due to
the small capsid diameter of 18– 26 nm, intact capsids can potentially pass into the
nucleus through nuclear pore complexes (NPCs). This might be facilitated by active
nuclear import, but capsids may also follow an alternative entry pathway that includes
activation of mitotic factors and local transient disruption of the nuclear envelope.
The nuclear entry is followed by currently undefined events of viral genome uncoat-
ing. After genome release, viral replication compartments are initiated and infection
proceeds. Parvoviral genomes replicate during cellular S phase followed by nuclear
capsid assembly during virus- induced S/G2 cell cycle arrest. Nuclear egress of capsids
occurs upon nuclear envelope degradation during apoptosis and cell lysis. An alterna-
tive pathway for nuclear export has been described using active transport through
the NPC mediated by the chromosome region maintenance 1 protein, CRM1, which
is enhanced by phosphorylation of the N- terminal domain of VP2. However, other
alternative but not yet uncharacterized nuclear export pathways cannot be excluded.
KEYWORDS
import and export, nuclear envelope, nuclear pore complexes, nucleus, parvoviruses
296 
|
    M ATT OL A et al .
known to induce lysis of transformed cells and to activate anticancer
immune responses (Abschuetz et al., 2006; Geletneky et al., 2017;
Gil- Ranedo et al., 2021; Grekova et al., 2012; Hartley et al., 2020;
Marchini et al., 2015). The role of CPV in inducing an antitumor im-
mune response in different tumor models has been discussed (Arora
et al., 2021). The potential of recombinant AAV vectors in gene ther-
apies has been shown by the approval of two AAV therapeutic appli-
cations for the treatment of Leber's congenital amaurosis (Luxturna)
and spinal muscular atrophy (Zolgensma) by the US Food and Drug
Administration (FDA) (Kuzmin et al., 2021; Large et al., 2021). In fact,
the first gene therapy, Glybera medicine, approved in 2012 cor-
rected hereditary lipoprotein lipase deficiency (LPLD). This treat-
ment was stopped in 2018 due to the high cost of c. one million US$
per patient, and only 31 people were treated (Mendell et al., 2021).
Parvoviruses comprise a linear single- stranded DNA of ~4 to
6 kb and an icosahedral capsid of 18– 26 nm in diameter (Cotmore
et al., 198 3; Kaufmann et al., 2004; Mietzsch et al., 2019; Tsao
et al., 1991; Xie et al., 20 02). The viral proteome differs between
members of parvovirus (Cotmore & Tattersall, 2014). Many of the
autonomous parvovirus genome encodes two structural proteins
(VP1 and VP2) and two non- structural proteins (NS1 and NS2)
(Cotmore et al., 1983; Cotmore & Tattersall, 2014), whereas AAV en-
codes at least for three capsid proteins ( VP1, VP2, and VP3) and four
non- structural proteins (Rep40, Rep52, Rep68, and Rep78) (Im &
Muzyczka, 1990; X ie et al., 2002). Capsid p roteins and non- s tructural
proteins are translated from alternatively spliced mRNAs, following
transcription controlled by the early P4 and the late P38 promoter.
While the former guides the expression of NS1 and NS2, the latter
controls the expression of capsid proteins (Christensen et al., 1995;
Cotmore & Tattersall, 1995; Li & Rhode 3rd, 1990). Nonetheless, the
family shows different transcriptional strategies and viruses within
the type species of each genus express a small number of genus-
specific ancillary proteins (Cotmore & Tattersall, 2014).
Parvoviruses use a variety of cell sur face receptors for attach-
ment to their host cells, determining host range and tissue tro-
pism (Govindasamy et al., 2003; Hueffer et al., 2003; Llamas- Saiz
et al., 1996; Michelfelder & Trepel, 2009; Palermo et al., 2006).
CPV uses sialic acid and transferrin receptor (Parker et al., 2001;
Parrish, 199 0), whereas human par vovirus B19V attaches to
erythrocyte P antigen (Brown et al., 1993) and its cellular entry is
facilitated by low pH- mediated interaction with globoside (Bieri
et al., 2021; Bieri & Ros, 2019). The dependoparvovirus AAV2 rec-
ognizes several receptors of target cells including heparan sulfate
proteoglycan, αVβ5 integrin, and basic fibroblast growth factor re-
ceptor 1 (Qing et al., 1999; Summerford et al., 1999; Summerford
& Samulski, 1998). Recently, the AAV receptor (AAVR; KIAA0319L)
was identified as an essential receptor for cell internalization and
trafficking of different AAVs (Meyer & Chapman, 2022; Pillay
et al., 2016). After receptor binding, many parvoviruses enter cells
via clathrin- mediated endocytosis (Bartlett et al., 2000; Cureton
et al., 2012; Parker & Parrish, 2000). The low endosomal pH induces
conformational changes in parvovirus capsid structure, which leads
to exposure of the VP1 N- terminal unique region (VP1u). VP1u of
B19, MVM, and CPV comprises a phospholipase A2 (PLA2) motif,
a nuclear localization sequence (NLS), and three PDZ domains,
which are highly conserved. The PLA2 domain is required for cap-
sid escape from endocytic vesicles (Farr et al., 2005; Popa- Wagner
et al., 2012; Qu et al., 2008; Suikkanen, Antila, et al., 2003b; Zádori
et al., 2001;Ros et al., 2020) presumably by forming holes in the en-
dosomal membrane, while NLS and PDZ domains are implicated in
nuclear import of the capsid.
This review focuses on what has been learned in the past years
about cy toplasmic trafficking, nuclear entry, and exit of parvovirus
capsids.
2 |NUCLEAR ENTRY OF PARVOVIRUS
CAPSIDS
2.1  | Traveling to the nucleus
Subsequent to endosomal escape, the capsids have to reach the
nuclear envelope (NE). Likely, parvoviruses make use of the cellular
microtubule network, as depolymerization of microtubules blocks
CPV infection (Suikkanen, Aaltonen, et al., 2003a), which is also
consistent with their observed velocity toward the nucleus (Mäntylä
et al., 2018). These findings are supported by obser vations on A AV,
showing that their perinuclear accumulation is enhanced by dynein-
and microtubule- mediated transport (Kelkar et al., 2004, 2006; Xiao
& Samulski, 2012). As with CPV, tracking of single AAV particles in
the cytoplasm has demonstrated directed motion of viral capsid to-
ward the nucleus, which is a characteristic of dynein- microtubule
mediated transport (Seisenberger et al., 2001).
Direct transport of the released capsids along microtubules is
likely but it s requirement is not unequivocally proven as microtubule
depolymerization does not affect CPV distribution after microinjec-
tion (Lyi et al., 2 014), which was also obser ved for cells transduced
with recombinant AAV2 vectors (rAAV) (Hirosue et al., 2007). Even
less understood is the observation that the intermediate filament
protein vimentin enhances infection after endosomal escape as
shown for MVM (Fay & Panté, 2013) as intermediate filaments are
not polarized and thus hardly contribute to directed cytoplasmic
transport. The observation showing that vimentin filaments become
disrupted at 14 to 24 h post- MVM infection, well after nuclear entry
should have been completed, indicates an independent phenome-
non that is unrelated to early infection events (Nüesch et al., 2005).
Further, some parvoviruses such as MVM and CPV exploit
ubiquitin- proteasome machinery to enhance their nuclear trans-
location. The presence of a proteosomal inhibitor (MG132) leads
to cytoplasmic perinuclear retainment of capsids. However, the
viral entry, the natural proteolytic cleavage of VP2 to VP3 and
the externalization of the N terminal of VP1 are not affected (Ros
& Kempf, 2004). In contrast, ubiquitination of AAV capsids leads
to their degradation, and treatment with MG132 increases A AV- 2
and AAV- 5 transduction (Ding et al., 2003; Douar et al., 20 01; Yan
et al., 2002; Zhong et al., 2008). It remains an open question if
   
|
297
MAT TOLA et al.
the involvement of proteasomes affects cytosolic transpor t or a
subsequent step.
2.2  | Overview of NPC and nuclear import
Many viruses have adapted to replicate in the host's nuclei, allow-
ing exploitation of cellular machiner y like DNA or RNA polymer-
ases. This requires access to the nucleoplasm, which has led to the
evolution of specific mechanisms for reaching this compartment.
Amongst the best- described approaches to enter the nucleus are
interactions with nuclear pore complexes (NPC) (Fay & Panté, 2015;
Guedán et al., 2021).
NPCs are macromolecular structures crossing the NE, allowing
passive dif fusion only of metabolites and proteins smaller than 30
60 kDa, dependent upon their charge. However, a slow diffusion of
larger molecules up to 230 kDa through the NPC has been observed
(Popken et al., 2015; Timney et al., 2016; Wang & Brat tain, 20 07).
The NPC is composed by approximately 30 proteins termed nucle-
oporins (Nups). The shape of the NPC opening is determined by the
Y complexes (Nup107- Nup160 complex) (Stuwe et al., 2015). These
are crucial for their interactions with the gel- like mesh of highly dis-
ordered nucleoporins present in the NPC channel, which are charac-
terized by high abundance of short stretches of hydrophobic amino
acids comprising phenylalanine (F) and glycine (G) residues. FG-
Nups, which include Nup62, regulate which molecules may traverse
the NE and fix cytoplasmic and nuclear fibers ex truding from the
central part of the NPC (Lyngdoh et al., 2021). Other nucleoporins
like Nup153 and Tpr form a basket- like structure on the nucleoplas-
mic side, which is necessary for impor t and expor t. Many Nups such
as Nup153 and Nup62 are also involved in other, non- transport-
related functions, such as chromosome alignment and binding (Chien
et al., 2020; Hashizume et al., 2013). Nup153 and Nup358 have been
reported to possess conserved zinc finger domains, which are re-
quired for recruitment of coat protein I complex (COPI) coatomers
in the early process of nuclear envelope breakdown (NEBD) during
mitosis (Liu et al., 2003; Prunuske et al., 2006).
Transport of large proteins or nucleoprotein complexes through
the NPC is energy dependent and requires exposure of specific
signaling motifs on cargo surface. Classical nuclear localization
signals (NL Ss) are characterized by shor t stretches of positively
charged amino acids (Arginine and Lysine) exemplified by that of
SV40 (PKKKRKV) (Kalderon et al., 1984). Other signals are proline-
tyrosine NLSs, previously termed M9 domains, which comprise
highly disordered sequences of 20– 30 amino acids interspaced by
hydrophobic or basic residues, as, for example, found in hnRNP A1
(Bradley et al., 2007; Görlich, 1997). Not all NLSs are permanently
exposed. The so- called cryptic NL Ss become exposed only upon
post- translational modifications or protein– protein interactions
(Fagerlund et al., 2002; Gu et al., 2003).
The different signals for nuclear transport through the NPC
allow binding of specialized transport receptors, named importins,
which are divided into importin α and β (also known as KPNA, KPNB)
(Cautain et al., 2015). Of the former, seven members, all involved
in nuclear import, are known to serve as adaptor proteins between
the nuclear import signal on the cargo and importin β to which it
binds via an importin- binding domain. Depending on the species,
between 14 and 20 importin βs have been described. Eleven mem-
bers of human importins βs facilitate nuclear import. These include
transportin (TNPO, also called importin β2), six nuclear export, and
three nuclear import and export (Kimura & Imamoto, 2014; Oldrini
et al., 2017). Importin β not only binds to cargos via importin α but
may also directly interact with cargo- exposed importin- binding do-
mains (Lee et al., 2006; Mitrousis et al., 2008). There is, however,
growing evidence that nuclear transport receptor- independent
pathways exist as it was described for, for example, IκBα (Sachdev
et al., 2000).
The import is initiated by binding of importin to its correspond-
ing nuclear import motif. Via multiple interactions, these complexes
pass the hydrophobic mesh in the central pore channel (Yoshimura
et al., 2014). Upon interacting with Nup153, dissociation of the
cargo from the importin occurs through binding of the Ras- like small
GTPase Ran in its GTP- bound form (Walther et al., 2001).
Nuclear export follows a similar principle in which a nuclear ex-
port signal (NES) containing cargo traverses the NPC toward the cy-
toplasm. NESs are characterized by a hydrophobic profile, as is found
on the HIV Rev protein (LPPLERLT) (Fischer et al., 1995). NESs allow
binding of ex portins, such as Chromosomal Maintenance 1 (CRM1) in
complex with RanGTP (Kehlenbach et al., 1999; Petosa et al., 2004).
After translocation through the NPC, export complexes reach cyto-
plasmic filaments where Nup214 binds to CRM1 allowing the closely
localized Nup358- bound RanGAP to trigger the GTPase function of
Ran, catalyzing the hydrolysis of RanGTP to RanGDP. This results in
a conformational change and the dissociation of the transport- cargo
complex (Hutten et al., 2008; Mahadevan et al., 2013; Ritterhoff
et al., 2016; Wälde et al., 2012).
2.3  | Nuclear entry of parvoviruses
through the NPC
Although the molecular details of parvoviral nuclear import remain
controversial, it has been suggested that intact capsids enter the
nucleus followed by genome release at some distance from the NE
(Bernau d et al., 2018; Mänt ylä et al., 2018). Prev ious studies on MV M
demonstrated that the nuclear release of parvoviral genomes occurs
without complete disassembly of the capsids (Cotmore et al., 1999;
Ros et al., 2006; Ros & Kempf, 2004). However, fast diffusion of
intranuclear CPV capsid fragment s demonstrates the presence of
disassembled capsids (Mäntylä et al., 2018). Irrespectively to the
intranuclear fate of capsids, which is linked to the unknown mecha-
nism of genome release, they may have to be primed for genome re-
lease prior to nuclear import as B19V capsid uncoating is enhanced
by cytoplasmic depletion of divalent cations (Caliaro et al., 2019).
Single particle imaging demonstrated that the first nuclear AAV- 2
capsids are detected already 15 min after adding the viral particles to cell
298 
|
    M ATT OL A et al .
culture (Seisenberger et al., 2001) although others reported that more
than 2 hours are needed for nuclear capsid arrival (Bartlett et al., 2000;
Sonntag et al., 2006; Zhong et al., 2008). The entry of intact capsids
was also observed for CPV either after infection or af ter cytoplasmic
microinjection of viral particles; the latter observation indicating that
acidification and subsequent structural changes are not essential for
nuclear entry (Harbison et al., 2009; Suikkanen, Antila, et al., 2003b;
Vihinen- Ranta et al., 2002). However, these microinjections were per-
formed using parvovirus- susceptible cells and the technically caused
leakage of capsids from the needle prior to injection leads to exposure
of capsids to the cell exterior thus initiating parallel infections.
Two possible pathways of how parvoviral capsids enter the nu-
cleus have been proposed: a “classical” entr y passing the NPCs using
the NLS on VP1u, which binds to nuclear import factors of the im-
portin family (Table 1). This would allow the capsids to pass the NPC
due to their small diameter which is below the 40 nm size limit of the
NPC (Panté & Kann, 2002) (Figure 1). Alternatively, parvoviral cap-
sids may enter the nucleus through transient holes in the NE, which
are induced by their interaction with Nups (Porwal et al., 2013)
(Figure 2). Due to the low efficiency of all parvoviruses, it remains
not fully evident which pathway leads to progeny infection, and a
combination of both pathways appears possible.
As mentioned before, acidification leads to exposure of VP1u,
which comprises a NLS with basic residues as shown for CPV
(Cotmore et al., 1999, 2010 ; Vihinen- Ranta et al., 2002). Similarly, the
externalized N- terminus of AAV2 VP1 and VP2 proteins comprise
three NLS- like motifs which are essential for the progression of in-
fection (Grieger et al., 2006; Hoque et al., 1999; Johnson et al., 2010;
Sonntag et al., 2006), as well as three PDZ- motifs crucial for nuclear
entry and infection (Popa- Wagner et al., 2012). These NLSs may not
only contribute to the nuclear transport of the capsids but also to
the transport of capsid proteins, required for nuclear assembly of
TABLE 1 Key facts on nuclear entry of parvovirus capsids
Nuclear entry
requirements Active transpor t NPC
Interac tion with
Nups NEBD
Autonomous
parvovirus
B19V: Potential depletion
of capsid- associated
divalent cations for
uncoating (Caliaro
et al., 2019)
CPV and H - 1 P V : Ca2+
release for NE
disruption (Porwal
et al., 2013)
CPV: Capsids recruit
importin β to form
capsid- importin β
complex
(Mäntylä et al., 2020)
capsid- importin
β complex is
transported
into the nucleus
(Mänty
et al., 2018)
H - 1 P V :
Coprecipitation
with Nup358,
Nup153 and
Nup62 (Porwal
et al., 2013)
Interaction with
Nups may
trigger PLA2
exposure
to induce
initial Ca2+
release for NE
disintegration
(Porwal
et al., 2013)
MVM: NE invagination and redistribution of
lamin A/C (Cohen et al., 2006)
H - 1 P V : Ca2+ release triggers activation of
mitotic factors (PKC, cdk2/cdk1 and
caspase 3) for NE disintegration by local
lamin B depolymerization. No soluble
cytosolic factors needed in permeabilized
cells (Por wal et al., 2013)
CPV failed to infect cells preloaded with
hepatitis B capsids by microinjection
(Porwal et al., 2013)
Dependo-
parvovirus
AAV 2: Capsid
acidification (pH 5. 2)
and Ca2+ release
was required for NE
disruption (Porwal
et al., 2013)
AAV 2: Three NLS- like
motifs in VP1 and
VP2 essential for
infection (Johnson
et al., 2010).
Three PDZ- motifs on
VP1u essential
for nuclear entry
and infec tion
(Popa- Wagner
et al., 2012).
Labeled capsids pass-
through NPC, no
evidence of NE
disintegration:
(Kelich et al., 2015)
rAAV2: Interaction
with importin β
with or without
interaction
with importin
α (Nicolson &
Samulski, 2014)
AAV 2
coprecipitates
with Nup358,
Nup153 and
Nup62 (Porwal
et al., 2013)
NEBD limited to microinjected AAV2 capsid
exposed to pH 5.2 (Porwal et al., 2013)
NE invagination showed by EM (Cohen
et al., 2006; Cohen & Panté, 20 05)
   
|
299
MAT TOLA et al.
progeny par voviruses. In fact, VP1/VP2 trimers of MVM are actively
imported although using importin β - independent pathway (Riolobos
et al., 2006). Of note, the nuclear import of these trimers requires
VP1/VP2 phosphorylation by Raf- 1 kinase during entr y of the cell
into S phase, which may contribute to MVM specificity for trans-
formed cells (Lombardo et al., 2000; Riolobos et al., 2006).
More direct evidence for importin β- recruitment on assembled
capsids was found in time- lapse microscopy of CPV after infection,
showing that the formation of importin β- CPV- complexes slows the
diffusion of cytoplasmic CPV capsids (Mäntylä et al., 2018, 2020).
Further, importin β- CPV capsid complexes are transported simulta-
neously through the NE (Mäntylä et al., 2018), which is consistent
with data on rAAV- 2 nuclear import, which depends on interac-
tion with importin β alone or in complex with importin α (Nicolson
& Samulski, 2014). However, at least some nuclear capsids remain
decorated with importin β (Mäntylä et al., 2018) arguing against a
classical nuclear import of at least a fraction of capsids, as importin
β becomes removed from the cargo within the nuclear basket during
classical NLS- dependent nuclear import.
However, the number of detected intranuclear capsids is very
low but in agreement with the small number of nuclear replica-
tion compartment foci detected in early stages of MVM and CPV
infection (Ihalainen et al., 2007; Ruiz et al., 2006). It can be thus not
excluded that only a minor fraction of parvoviral capsids initiates
infection.
2.4  | Entry through the NE by increased nuclear
envelope permeability
Various parvoviruses exhibit a unique feature in that they permea-
bilize transiently the NE, as it was shown for H- 1PV, CPV, and AAV2
(Cohen et al., 2006; Cohen & Panté, 2005; Popa- Wagner et al., 2012;
Porwal et al., 2013) (Table 1). This nuclear envelope break- down
(NEBD) occurs within minutes after capsid exposure to nuclei, being
in agreement with a rapid passage of the capsid into the nucleus ob-
served in infection. This led to the hypothesis that these holes in
the nuclear envelope allow nuclear entr y of intact capsids (Figure 2).
Mechanistically, parvoviral NEBD shows similarities to mitosis in
that Ca++, released from the lumen between inner and outer nuclear
membrane, initiates activation of PKCα, which activates Cdk2 and/
or Cdk1, followed by activation of caspase 3 (Figure 2b). The acti-
vation of the kinases allows the hyper- phosphorylation of lamin B,
which was described to cause local lamin (Cohen et al., 2006). Such
depolymerization is required for open holes of up to 190 nm (Porwal
et al., 2013), which are large enough to allow entry of the capsids
or even larger complexes as capsid- importin complexes. The role of
caspase 3 is in the proteolytic cleavage of lamin B and not in the di-
rect disruption of the nuclear membranes (Cohen et al., 2006, 2011;
Cohen & Panté, 2005) (Figure 2c). Caspase 3 is upregulated and ac-
tivated just prior to mitosis (Hsu et al., 2006), being in concordance
with its function during parvoviral- mediated NEBD.
Pore formation depends on interaction with the NPC in par-
ticular by capsid binding to at least three Nups (Nup358, Nup153,
and Nup62). Blocking AAV2 or H- 1PV interaction with Nup153
by hepatitis B virus capsids, which specifically interacts with
Nup153 (Schmitz et al., 2010) inhibited NEBD. The relevance of
this finding for infection was later confirmed by CPV, which failed
to infect cells preloaded with hepatitis B virus capsids by micro-
injection (Mäntylä et al., 2020). As Nup153 is localized in the
nuclear basket close to the inner ring of the NPC, these obser-
vations indicate that the parvoviruses should be associated with
importins in order to reach the nuclear side of the NPC (Figure 2a).
Furthermore, NEBD was accelerated when the capsids were pre-
acidified and neutralized implying the need of VP1u exposure.
In fact, PLA2 exposure could also be achieved by direct interac-
tion of parvoviral capsids (AAV2 and H- 1PV) with Nups leading
to the hypothesis that the accessible PLA2 domain triggers the
initial Ca++ efflux. However, PLA2 activity on MVM capsids has
not been reported to be involved in causing NE disruption (Cohen
et al., 2011) and other mechanisms causing permeabilization can-
not be excluded. This includes amphipathic helices identified on
VP1u (Leisi et al., 2016) as they permeabilize membranes, which
was as shown for endosomal escape of adenoviruses (Wiethoff &
Nemerow, 2015), or the PDZ domains, which exhibit membrane
FIGURE 1 Nuclear entry of parvoviruses through the NPC.
Cytoplasmic parvoviruses (PVs) that have undergone structural
changes within the endosome bind to importin α (KPNA) /
importin β (KPNB). This allows transport through cellular nuclear
pore complexes (NPCs). Upon reaching the nuclear basket, the
PV- importin complex dissociates, releasing the capsid into the
nucleoplasm. Figure created with BioRender.
300 
|
    M ATT OL A et al .
affinity (Fanning & Anderson, 1999) and induce membrane curva-
ture (Herlo et al., 2018).
In summar y, there are two seemingly contradictory models of nu-
clear import of parvoviral capsids but both rely on interaction with
the NPC, which was also demonstrated by single particle tracking of
rAAV (Junod et al., 2021; Kelich et al., 2015). Numerous data support
that this interaction is mediated by importin β, however, the interac-
tion between importin α and the NLS exposed on VP1u on capsid sur-
face is not well understood. Nuclear import of microinjected capsids
(Harbison et al., 2009; Suikkanen, Aaltonen, et al., 20 03a) suggests that
a sub- fraction of capsids might expose their VP1us without acidifica-
tion. Fur ther, it cannot be totally excluded that the nuclear c apsids after
microinjection are derived from infection occurring in parallel.
Differences between the models comprise later events once the
capsids arrive on the nuclear side of the NPC. While the classical im-
port model favors dissociation of the importins from the capsids and
diffusion of the latter deeper into the nucleus, the NEBD model sup-
ports interaction with Nup153 possibly after importin β dissociation,
FIGURE 2 Entry through the NE by increased nuclear envelope permeability. (a) Parvovirus capsids bound to importins (KPNA: Importin
α/KPNB: Importin β) bind to Nups. The binding triggers exposure of PLA2 on VP1u inducing calcium efflux. (b) the release of calcium
activates PKCα, which activates cdk2/cdk1. Caspase 3 is also activated. (c) Hyper- phosphorylation of Lamin B by kinases as well as L amin
B- cleavage by caspase 3 leads to its local degradation. (d) The formation of transient holes allows entry of NPC- bound or cytosolic capsids or
capsid- importin complexes. Figures created with BioRender.
   
|
301
MAT TOLA et al.
disintegration of the NE and entry of the capsid. These could be either
cytosolic capsids (eventually importin β- bound) or the capsids that have
initiated the NEBD, likely after NPC- dissociation which is mediated
by cdk- 1 (Kutay et al., 2021). However, as long as it remains unknown
which capsids initiate infection, none of the models can be excluded.
3 |NUCLEAR EGRESS
Once parvovirus genome has entered the nucleus, successful replica-
tion depends on cell entry into the S phase. The S phase- dependent
activation of DNA replication machinery is needed to provide the re-
sources necessary for viral replication. These cellular factors include
DNA polymerase δ required for conversion of ssDNA to dsDNA tem-
plate for viral gene transcription (Cotmore & Tattersall, 2013). The
progression to the S phase is accompanied by virus- induced cellular
DNA damage, ataxia telangiectasia mutated (ATM)- dependent DNA
damage response (DDR) and pre- mitotic cell cycle arrest in MVM
infection (Adeyemi et al., 2010; Cotmore & Tattersall, 2013; Ruiz
et al., 2011). In AAV infection, cytotoxic viral Rep proteins induce
S- phase arrest (Berthet et al., 2005; Saudan et al., 2000), and UV-
treated A AV particles evoke ATM- and Rad3- related kinase (ATR)-
dependent DDR charac terized by accumulation of cells in the late S
and/or G2 phases (Jurvansuu et al., 2005; Raj et al., 2001; Schwartz
et al., 2009; Winocour et al., 1988). Preventing cell entry from G2
phase to mitosis maintains nuclear structure thereby allowing the
prolonged assembly of new virions (Adeyemi & Pintel, 2014; Chen
et al., 2010; Morita et al., 2003). The empty capsids are formed in the
nucleus, and they mature into DNA- filled capsids at the late S/G2
phase (Gil- Ranedo et al., 2015). Af ter AAV capsid assembly, involv-
ing capsid accumulation in nucleoli (Sonntag et al., 2010; Wistuba
et al., 1997), targeting of viral ssDNA to viral capsid is mediated by
Rep proteins (Bleker et al., 2006; Dubielzig et al., 1999).
Viral infection elicits various responses in the host cell which
can lead to plasma membrane ruptures, formation of membrane
vesicles, nuclear fragmentation, and finally to cell lysis (Labbé &
Saleh, 2008). The cellular egress of many non- enveloped viruses is a
passive process which relies on cell lysis to release viral progeny into
the extracellular space (Daeffler et al., 2003; Georgi & Greber, 2020;
Tollefson et al., 1996). The major form of cell death described for
parvoviruses is apoptosis, however, also necrosis has been detected
(Chen & Qiu, 2010; Nykky et al., 2010).
In apoptotic cells, the NE permeability is regulated by caspase-
dependent and - independent alterations of NPCs and caspase- dependent
cleavage of la mins and other NE prote ins (Ferrando- May, 2005; Kihlmark
et al., 2004; Roehrig et al., 2003; Strasser et al., 2012). As described ear-
lier, nuclear entry of parvovirus capsids is accompanied by the NE dis-
integration and activation of the key enzymes of mitosis (Porwal et al.,
2013). However, nuclear microinjection of H- 1PV capsids does not in-
duce NEBD making it unlikely that this entry- related mechanism is re-
quired for capsid egress from the nucleus.
During parvovirus infection, the disintegration of host DNA
is followed by DNA damage response and activation of apoptosis
TABLE 2 Key facts on nuclear egress of parvovirus capsids
NS2- CRM1 interaction Active tr ansport NPC Phosphorylation Apoptosis
Autonomous
parvoviruses
MVM: CRM1 interacts with the NES in
NS2 (Bodendorf et al., 1999; Eichwald
et al., 2002; Engelsma et al., 20 08; Fornerod
et al., 1997; Maroto et al., 2004; Miller &
Pintel, 2002)
MVM: NS2 NES is required
for active nuclear
export of progeny
viruses (Engelsma
et al., 2008)
MVM: Phosphorylation of serine residues
in the exposed N - terminal end of
VP2 functions as a NES contributing
to active export (Maroto et al., 2004).
Phosphorylation of the capsid surface
enhances nuclear export capacity. VP2
N- terminal phosphorylation is involved in
passive release, but not required for active
transport (Wolfisberg et al., 2016)
CPV: Infection activates caspases 9,8, 3/7.
(Nykky et al., 2010)
Minute virus of canines (MVC): Infection-
induced DNA damage leads to p53-
dependent cell death (Chen & Qiu, 2010)
Dependo-
parvovirus
AAV: Activities of helper virus leads to cell lysis
and viral exit
(Meier et al., 2020, Smith & Enquist, 2002)
302 
|
    M ATT OL A et al .
(Adeyemi et al., 2010; Chen & Qiu, 2010). The cell death events are
mediated by apoptotic caspases (Roos & Kaina, 2006). CPV- infected
cells have a relatively long lifespan even thou gh the initiator cas pases
8 and 9, and effector caspases 3 and 7 are activated early in infection
and remain active until very late in infection, until 48– 72 hpi (Nykky
et al., 2010). Analysis of infected cells has indicated that capsids are
released from host cells already at 12 hpi (Zhao et al., 2016). These
observations support the model that viral capsids egress the nucleus
and the host cell prior to apoptosis- induced cell lysis. After nuclear
exit cytoplasmic MVM progeny capsids are transported through
COPII- vesicles of ER and cisternae of Golgi and continue toward
the cellular periphery in lysosomal/late endosomal vesicles. The ve-
sicular capsid transport and cellular exocytosis depend on gelsolin-
induced degradation of actin (Bär et al., 2008).
In contrast to these cell- destruction- based exit mechanisms,
which were previously thought to be the main pathway for progeny
parvoviral egress, more recent evidence supports that parvoviruses
are also able to actively egress the nucleus into cytosol before pas-
sive release through cell lysis at the final stage of the infection occur s
(Table 2). Active translocation has been previously shown for MVM,
which utilizes the CRM1- mediated active nuclear export pathway
for nuclear exit of capsids through the NPC (Eichwald et al., 2002;
FIGURE 3 Nuclear egress of progeny capsids. Packaging of viral genomes inside capsids causes a conformational change exposing the
VP2 N- terminal on the capsid surface. (a) MVM capsids are actively exported out of the nucleus through NPCs mediated by the interaction
between NS2 NES with CRM1 (Bodendorf et al., 1999; Eichwald et al., 2002; Engelsma et al., 2008; Fornerod et al., 1997; Maroto
et al., 2004; Miller & Pintel, 2002). (b) The phosphorylation of the exposed VP2 N- terminal end on the capsid surface acts as a nuclear
export signal enhancing capsid export out of the nucleus (Maroto et al., 2004). (c) Phosphorylation of the capsid surface enhances capsid
export (Wolfisberg et al., 2016). (d) Activation of apoptosis and necrosis affect the structure of the nuclear lamina, and capsids are released
to the cytoplasm in late infection (Chen & Qiu, 2010; Nykky et al., 2010, (Wolfisberg et al., 2016). Figures created with BioRender.
   
|
303
MAT TOLA et al.
Engelsma et al., 2008) (Figure 3a). CRM1, also called exportin 1, is
a versatile nuclear export receptor which shuttles between the nu-
cleus and cytoplasm (Fornerod et al., 1997) and translocates mul-
tiple cargoes including ribosomal subunits (Ho et al., 2000; Moy &
Silve r, 1999; Thomas & Kutay, 2003). The binding of CRM1 to the
cargo is promoted by RanGTP (Koyama & Matsuura, 2010) and me-
diated by NES. For nuclear export of MVM capsids, CRM1 interacts
with the NES in NS2 in a RanGTP- independent manner (Bodendorf
et al., 1999; Eichwald et al., 2002; Engelsma et al., 2008; Fornerod
et al., 1997; Maroto et al., 2004; Miller & Pintel, 2002).
Several findings, however, support CRM1- independent nuclear
capsid export, being thus most likely NS2- independent. Similar to
nuclear import of MVM VP1/VP2 trimers, nuclear egress of MVM
capsids is enhanced by Raf- 1 kinase- mediated phosphorylation of
three serine residues in the N- terminus of VP2 on capsid surface
(Maroto et al., 2004) (Figure 3b). This pathway relies on exposure of
the N- terminal domain of VP2, which is exposed in DNA- containing
parvovirus capsids during their maturation (Agbandje- McKenna
et al., 1998; Kaufmann et al., 2008; Kontou et al., 2005; Sánchez-
Martínez et al., 2012; Tsao et al., 19 91). Moreover, the phosphoryla-
tion of the capsid surface residues has been linked to nuclear export
capacity prior to the passive release by cell lysis. Although confor-
mational change of the VP2 N- terminus on the capsid surface was
required for phosphorylation, the VP2 N- terminus was dispensable
for nuclear capsid egress (Wolfisberg et al., 2016) (Figure 3c.) Non-
phosphorylated capsids exit the nucleus passively upon NE damage
during apoptosis (Figure 3d). The cellular and nuclear egress of AAV
was earlier thought to rely on cell lysis caused by overexpression of
helper virus, adenovirus or herpesvirus, proteins (Meier et al., 2020;
Smith & Enquist, 2002). Recently, the presence of viral membrane-
associated accessory protein (MAAP) was observed for AAV at the
late stages of infection. MAAP is located in the plasma membrane
and in the n uclear periphe ry (Galiber t et al., 2021; Ogden et al., 2019).
This protein is a viral egress factor, which also promotes AAV capsid
association with extracellular vesicles (Elmore et al., 2021).
4 |CONCLUDING REMARKS
The versatile therapeutic potential of parvovirus has researchers
focused on understanding the full mechanism of infection. In gene
therapy, for which an efficient delivery of modified parvoviral vec-
tors (mostly AAV) is crucial, nuclear entry seems to be a bottleneck
and detailed knowledge may help improving their clinical administra-
tion. Similar to nuclear entry, the studies of viral egress have shown
also controversial result s. Improving the knowledge on export may
assist oncolytic therapy using autonomous parvoviruses, as their po-
tential depends upon efficient spread.
ACKNOWLEDGMENTS
This work was financed by the Jane and Aatos Erkko Foundation
(MVR), Academy of Finland under the award numbers 330896 (MVR),
and the Graduate School of the University of Jyvaskyla (SM).
DATA AVAIL ABILI TY STATEMENT
Data sharing not applicable - no new data generated.
ETHICS STATEMENT
The work presented here did not include human or animal subjects
nor human or animal material or data. Thus, no formal consent or
approval was necessary.
ORCID
Maija Vihinen- Ranta https://orcid.org/0000-0003-0959-1153
REFERENCES
Abschuetz, A., Kehl, T., Geibig, R., Leuchs, B., Rommelaere, J. & Régnier-
Vigouroux, A. (20 06) Oncoly tic murine autonomous parvovirus, a
candidate vector for glioma gene therapy, is innocuous to normal
and immunocompetent mouse glial cells. Cell and Tissue Research,
325, 423– 436. https://doi.org/10.1007/S0044 1- 006- 0199- Z/
FIGUR ES/8
Adeyemi, R .O. & Pintel, D.J. (2014) Parvovirus- induced depletion of cy-
clin B1 prevents mitotic entry of infected cells. PLoS Pathogens, 10,
e1003891. https://doi.org/10.1371/journ al.ppat.1003891
Adeyemi, R .O., Landr y, S., Davis, M.E., Weitzman, M.D. & Pintel, D.J.
(2010) Parvovirus minute virus of mice induces a DNA damage
response that facilitates viral replication. PLoS Pathogens, 6, 1– 11.
https://doi.org/10.1371/journ al.ppat.1001141
Agbandje- McKenna, M., Llamas- Saiz, A.L., Wang, F., Tattersall, P. &
Rossmann, M.G. (1998) Functional implications of the structure
of the murine parvovirus, minute virus of mice. Structure, 6, 1369–
1381. https://doi.org/10.1016/S0969 - 2126(98)00137 - 3
Arora, R., Malla, W.A., Tyagi, A., Mahajan, S., Sajjanar, B. & Tiwari, A.K.
(2021) Canine parvovirus and its non- structural gene 1 as oncolytic
agents: mechanism of ac tion and induction of anti- tumor immune
response. Frontiers in Oncology, 11, 1290. https://doi.org/10.3389/
FONC.2021.64887 3/BIBTEX
Bär, S., Daeffler, L., Rommelaere, J. & Nüesch, J.P.F. (2008) Vesicular
egress of non- enveloped lytic parvoviruses depends on gelsolin
functioning. PLoS Pathogens, 4, e1000 126. https://doi.org/10.1371/
JOURN AL.PPAT.1000126
Bartlett, J.S., Wilcher, R. & Samulski, R .J. (200 0) Infectious entry path-
way of adeno- associated virus and adeno- associated virus vec-
tors. Journal of Virology, 74, 2777– 2785. https://doi.org/10.1128/
JVI.74.6.2777- 2785.2000/ASSET/ 4B651 4B3- 3A9B- 4BFA- A0E2-
849E4 34F8C 24/ASSET S/GRAPH IC/JV060 14300 08.JPEG
Bernau d, J., Rossi, A. , Fis, A., Gard ette, L., Ai llot, L., Bün ing, H. et al. (2018)
Charac terization of AAV vector particle stability at the single-
capsid level. Journal of Biological Physics, 44, 181– 194. h t tp s://d o i.
org/10.1007/S1086 7- 018- 948 8- 5/FIG UR ES/6
Berthet, C., Raj, K., Saudan, P. & Beard, P. (2005) How adeno-
associat ed virus Rep78 protein arr ests cells comp letely in S phase.
Proceedings of the National Academy of Sciences of the United
States of America, 102, 13634– 13639. https://doi.org/10.1073/
PNAS.05045 83102
Bieri, J. & Ros, C. (2019) Globoside is dispensable for par vovirus B19
entry but essential at a Postentry step for productive infection.
Journal of Virology, 93, 1– 15. https://doi.org/10.1128/jvi.00972
- 19
Bieri, J., Leisi, R., Bircher, C. & Ros, C. (2021) Human par vovirus B19
interac ts with globoside under acidic conditions as an essential
step in endocytic trafficking. PLoS Pathogens, 17, 1– 22. h t tps ://do i.
org/10.1371/journ al.ppat.1009434
Bleker, S., Pawlita, M. & Kleinschmidt , J.A. (2006) Impac t of capsid con-
formation and rep- capsid interactions on adeno- associated virus
304 
|
    M ATT OL A et al .
type 2 genome packaging. Journal of Virology, 80, 810– 820. ht t ps: //
doi.org/10.1128/JVI.80.2.810- 820.2006
Bodendorf, U., Cziepluch, C., Jauniaux, J.- C ., Rommelaere, J. & Salomé,
N. (1999) Nuclear export factor CRM1 interacts with non-
structural proteins NS2 from parvovirus minute virus of mice.
Journal of Virology, 73, 7769– 7779. https://doi.org/10.1128/
jvi.73.9.7769- 7779.1999
Bradley, K.J., Bowl, M.R., Williams, S.E., Ahmad, B.N., Partridge, C.J.,
Patmanidi, A.L. et al. (2007) Parafibromin is a nuclear protein with
a functional monopartite nuclear localization signal. Oncogene, 26,
1213– 1221. ht tps://doi.org/10.1038/SJ.ONC.1209893
Brown, K .E., Anderson, S.M. & Young, N.S. (1993) Ery throcyte P antigen:
cellular receptor for B19 par vovirus. Science, 262, 114– 117. htt p s: //
doi .org/10.1126/SCI EN CE.8211117
Caliaro, O., Marti, A ., Ruprecht, N., Leisi, R., Subramanian, S., Hafenstein,
S. et al. (2019) Parvovirus B19 Uncoating occurs in the cytoplasm
without capsid disassembly and it is facilitated by depletion of
capsid- associated divalent cations. Viruses, 11, 430. h ttp s://d oi.
org/10.3390/V1105 0430
Cautain, B., Hill, R., de Pedro, N. & Link, W. (2015) Components and reg-
ulation of nuclear transport processes. The FEBS Journal, 282, 4 45–
462. https://doi.org/10.1111/FEBS.13163
Chen, A .Y. & Qiu, J. (2010) Parvovirus infection- induced cell death
and cell cycle arrest. Future Virology, 5, 731– 743. ht tp s ://do i.
org/10.2217/fvl.10.56
Chen, A .Y., Luo, Y., Cheng, F., Sun, Y. & Qiu, J. (2010) Bocavirus infec-
tion induces mitochondrion- mediated apoptosis and cell cycle ar-
rest at G2/M phase. Journal of Virolog y, 84, 5615– 5626. h t tp s ://do i.
org/10.1128/JVI.02094 - 09
Chien, M.L., Lai, J.H., Lin, T.F., Yang, W.S. & Juang, Y.L. (2020) NUP62 is
required for the maintenance of the spindle assembly checkpoint and
chromosomal stability. International Journal of Biochemistry and Cell
Biology, 128, 105843. https://doi.org/10.1016/j.biocel.2020.105843
Christensen, J., Cotmore, S.F. & Tattersall, P. (1995) Minute virus of
mice transcriptional activator protein NS1 binds directly to the
transactivation region of the viral P38 promoter in a strictly ATP-
dependent manner. Journal of Virology, 69, 5422– 5430. h t tp s://d o i.
org/10.1128/JVI.69.9.5422- 5430.1995
Cohen, S. & Panté, N. (20 05) Pushing the envelope: microinjection of
minute virus of mice into Xenopus oocytes causes damage to
the nuclear envelope. Journal of General Virology, 86, 3243– 3252.
https://doi.org/10.1099/VIR.0.80967 - 0/CITE/REFWORKS
Cohen, S., Behzad, A.R., C arroll, J.B. & Panté, N. (2006) Parvoviral nu-
clear import: bypassing the host nuclear- transport machinery. The
Journal o f General Viro logy, 87, 3209– 3213. https://doi.org/10.1099/
VIR.0.82232 - 0
Cohen, S ., Marr, A.K. , Garcin, P. & Panté, N. (2011) Nucl ear envelope disr up-
tion involving host caspases plays a role in the par vovirus replication
cycle. Journal of Virology, 85, 4863– 4874. https://doi.org/10.1128/
JVI.01999 - 10/ASSET/ 2BF10 4E6- C8F0- 43D8- 9E83- 5C314 ECAF8
FD/ASSET S/GRAPH IC/ZJV99 90944 960008.JPEG
Cotmore, S.F. & Tattersall, P. (1995) DNA replication in the autono-
mous parvoviruses. Seminars in Virology, 6, 271– 281. ht tps ://do i.
org/10.1006/SMVY.1995.0033
Cotmore, S.F. & Tattersall, P. (2013) Parvovirus diversit y and DNA
damage responses. Cold Spring Harbor Perspectives in Biology, 5,
a0 12989. https://doi.org/10.1101/CSHPE RSPECT.A012989
Cotmore, S.F. & Tattersall, P. (2014) Parvoviruses: small does not
mean simple. Annual Review of Virology, 1, 517– 537. htt p s: //d oi.
org/10.1146/annur ev- virol ogy- 03141 3- 085444
Cotmore, S.F., Sturzenbecker, L.J. & Tattersall, P. (1983) The autonomous
parvovirus MVM encodes two nonstructural proteins in addition
to its capsid polypeptides. Virology, 129, 333– 343. h t tps ://do i.
org/10.1016/0042- 6822(83)90172 - 1
Cotmore, S.F., D'Abramo, A .M., Ticknor, C.M. & Tattersall, P. (1999)
Controlled conformational transitions in the MVM Virion expose
the VP1 N- terminus and viral genome without par ticle disassembly.
Virology, 254, 169– 181. ht tps://doi.o rg/10.10 06/VIRO.1998.9520
Cotmore, S.F., Hafenstein, S. & Tattersall, P. (2010) Depletion of virion-
associated divalent cations induces parvovirus minute virus of mice
to eject its genome in a 3′- to- 5′ direction from an otherwise in-
tact viral particle. Journal of Virology, 84, 1945– 1956. htt p s: //d oi .
org/10.1128/JVI.01563 - 09
Cotmore, S.F., Agbandje- McKenna, M., Canuti, M., Chiorini, J.A., Eis-
Hubinger, A.M., Hughes, J. et al. (2019) ICT V virus taxonomy pro-
file: parvoviridae. Journal of Gen eral Virology, 100, 367– 368. h t tp s://
doi.org/10.1099/jgv.0.001212
Cureton, D.K., Harbison, C.E., Cocucci, E., Parrish, C.R. & Kirchhausen, T.
(2012) Limited transferrin receptor clustering allows rapid diffusion
of canine parvovirus into clathrin endocytic structures. Journal of
Virology, 86, 5330– 5340. https://doi.org/10.1128/JVI.07194 - 11
Daeffler, L., Hörlein, R., Rommelaere, J. & Nüesch, J.P.F. (2003)
Modulation of minute virus of mice cytotoxic activities through
site- directed mutagenesis within the NS coding region.
Journal of Virology, 77, 12466– 12478. https://doi.org/10.1128/
jvi.77.23.12466 - 12478.2003
Ding, W., Yan, Z., Zak, R., Saavedra, M., Rodman, D.M. & Engelhardt, J.F.
(2003) Second- strand genome conversion of adeno- associated
virus type 2 (AAV- 2) and A AV- 5 is not rate limiting following apical
infect ion of polarized hu man airway epit helia. Journa l of Virology, 77,
7361– 7366. https://doi.org/10.1128/JVI.77.13.7361- 7366.20 03
Douar, A.- M., Poulard, K., Stockholm, D. & Danos, O. (2001) Intracellular
trafficking of adeno- associated virus vectors: routing to the
late endosomal compartment and proteasome degradation.
Journal of Virology, 75, 1824– 1833. https://doi.org/10.1128/
JVI.75.4.1824- 1833.2001
Dubielzig, R., King, J.A., Weger, S., Kern, A. & Kleinschmidt, J.A. (1999)
Adeno- associated virus type 2 protein interactions: formation of
pre- encapsidation complexes. Journal of Virology, 73, 8989– 8998.
htt ps://doi.org/10.1128/JVI.73.11.8989- 8998.1999
Eichwald, V., Daeffler, L., Klein, M., Rommelaere, J. & Salomé, N. (2002)
The NS2 proteins of parvovirus minute virus of mice are required
for efficient nuclear egress of progeny Virions in mouse cells.
Journal of Virology, 76 , 10307– 10319. https://doi.org/10.1128/
jvi.76.20.10307 - 10319.2002
Elmore, Z .C., Patri ck Havlik, L ., Oh, D.K., A nderson , L., Daabo ul, G., Devl in,
G.W. et al. (2021) The membrane associated accessory protein is an
adeno- associated viral egress factor. Nature Communications, 12, 1–
13. https://doi.org/10.1038/S4146 7- 021- 26485 - 4
Engelsma, D., Valle, N., Fish, A., Salomé, N., Almendral, J.M. & Fornerod,
M. (200 8) A supraphysiological nuclear export signal is required for
parvovirus nuclear export. Molecular Biology of the Cell, 19, 2544–
2552. https://doi.org/10.1091/MBC.E08- 01- 0009
Fagerlund, R., Mélen, K., Kinnunen, L. & Julkunen, I. (2002) Arginine/
lysine- rich nuclear localization signals mediate interactions between
dimeric STATs and importin α5 *. Journal of Biological Chemistry, 277,
30072– 30078. https://doi.org/10.1074/JBC.M2029 43200
Fanning, A .S. & Anderson, J.M. (1999) PDZ domains: fundamental build-
ing blocks in the organization of protein complexes at the plasma
membrane. The Journal of Clinical Investigation, 103, 767– 772.
htt ps://doi .org /10.1172/JCI650 9
Farr, G.A ., Zhang, L .G. & Tattersall, P. (2005) Parvoviral virions deploy a
capsid- tethered lipolytic enzyme to breach the endosomal mem-
brane during cell entry. Proceedings of the National Academy of
Sciences of the United States of America, 102, 17148– 17153. htt ps: //
doi.org/10 .1073/PNA S.05084 7 7102
Fay, N. & Panté, N. (2013) The intermediate filament network protein, vi-
mentin, is required for parvoviral infection. Virology, 444, 181– 190.
https://doi.org/10.1016/J.VIROL.2013.06.009
Fay, N. & Panté, N. (2015) Old foes, new underst andings: nuclear entry of
small non- enveloped DNA viruses. Current Opinion in Virology, 12,
59– 65. https://doi.org/10.1016/J.COVIRO.2015.03.017
   
|
305
MAT TOLA et al.
Ferrando- May, E. (20 05) Nucleocytoplasmic transport in apoptosis. Cell
Death and Differentiation, 12, 1263– 1276. https://doi.org/10.1038/
sj.cdd.4401626
Fischer, U., Huber, J., Boelens, W.C., Mattajt, L.W. & Lührmann, R. (1995)
The HIV- 1 rev activation domain is a nuclear export signal that ac-
cesses an export pathway used by specific cellular RNAs. Cell, 82,
475– 483. https://doi.org/10.1016/0092- 8674(95)90436 - 0
Fornerod, M., Ohno, M ., Yoshida, M. & Mat taj, I.W. (1997) CRM1 is an ex-
port receptor for leucine- rich nuclear export signals. Cell, 90, 1051
1060. https://doi.org/10.1016/S0092 - 8674(00)80371 - 2
Galibert, L., Hyvönen, A ., Eriksson, R.A.E., Mattola, S., Aho, V., Salminen,
S. et al. (2021) Functional roles of the membrane- associated AAV
protein MAAP. Scientific Reports, 11, 1– 19. ht tps://doi.org/10.1038/
S4159 8- 021- 01220 - 7
Geletneky, K., Hajda, J., Angelova, A.L., Leuchs, B., Capper, D., Bar tsch,
A.J. et al. (2017) Oncolytic H- 1 parvovirus shows safety and signs
of immunogenic activity in a first phase I/IIa glioblastoma trial.
Molecular Therapy, 25, 2620– 2634. https://doi.org/10.1016/J.
YMTHE.2017.08.016
Georgi, F. & Greber, U.F. (2020) The adenovirus death protein - a small
membrane protein controls cell lysis and disease. FEBS Letters, 594,
1861– 1878 . https://doi.o rg/10 .1002/1873- 3468.138 48
Gil- Ranedo, J., Hernando, E., Riolobos, L., Domínguez, C., Kann, M. &
Almendral, J. (2015) The mammalian cell cycle regulates parvovirus
nuclear capsid assembly. PLoS Pathogens, 11, e1004920. h t tp s://
doi.org/10.1371/JOURN AL.PPAT.1004920
Gil- Ranedo, J., Gallego- García, C. & Almendral, J.M. (2021) Viral tar-
geting of glioblastoma stem cells with patient- specific genetic and
post- translational p53 deregulations. Cell Reports, 36, 109673.
https://doi.org/10.1016/j.celrep.2021.109673
Görlich, D. (1997) Nuclear protein import. Current Opinion in Cell Biology,
9, 412– 419. https://doi.org/10.1016/S0955 - 0674(97)80015 - 4
Govind asamy, L., Huef fer, K., Parrish , C.R. & A gbandje- McKenna , M. (2003)
Structures of host range- controlling regions of the capsids of canine
and feline parvoviruses and mutants. Journal of Virology, 77, 12 211–
12221. https://doi.org/10.1128/JVI.77.22.12211 - 12221.2003
Grekova, S.P., Raykov, Z., Zawatzk y, R., Rommelaere, J. & Koch, U. (2012)
Activation of a glioma- specific immune response by oncolytic par-
vovirus minute virus of mice infection. Cancer Gene Therapy, 19(7),
468– 475. ht tp s://doi. org/10.10 38/c gt .2 012.20
Grieger, J.C., Snowdy, S. & Samulski, R.J. (2006) Separate basic region
motifs within the adeno- associated virus capsid proteins are essen-
tial for infectivity and assembly. Journal of Virology, 80, 5199– 5210.
https://doi.org/10.1128/JVI.02723 - 05
Gu, Y., Hinner wisch, J., Fredricks, R ., Kalepu, S., Mishra, R.S. & Singh, N.
(2003) Identification of cryptic nuclear localization signals in the
prion protein. Neurobiology of Disease, 12, 133– 149. htt p s: //d oi .
org/10.1016/S0969 - 9961(02)0 0014 - 1
Guedán, A., Caroe, E.R., Barr, G.C.R. & Bishop, K.N. (2021) The role
of capsid in HIV- 1 nuclear entry. Viruses, 13, 1425. ht tps ://do i .
org/10.3390/V1308 1425
Harbison, C.E., Lyi, S.M., Weichert, W.S. & Parrish, C.R. (2009) Early
steps in cell infection by parvoviruses: host- specific dif ferences in
cell receptor binding but similar endosomal trafficking. Journal of
Virology, 83, 1050 4– 10514. https://doi.org/10.1128/JVI.00295 - 09
Hartley, A., Kavishwar, G., Salvato, I. & Marchini, A . (2020) A roadmap
for the success of oncolytic parvovirus- based anticancer therapies.
Annual Review of Virology, 7, 537– 557. https://doi.org/10.1146/
ANNUR EV- VIROL OGY- 01222 0- 023606
Hashizume, C., Moyori, A., Kobayashi, A., Yamakoshi, N., Endo, A . &
Wong, R.W. (2013) Nucleoporin Nup62 maintains centrosome
homeostasis. Cell Cycle, 12, 3804– 3816. https://doi.org/10.4161/
CC. 26671
Herlo, R ., Lund, V.K., Lycas, M .D., Jansen, A .M., Khelashv ili, G., Ander sen,
R.C. et al. (2018) An amphipathic helix directs cellular mem-
brane curvature sensing and function of the BAR domain protein
PICK1. Cell Reports, 23, 2 056– 2069. https://doi.org/10.1016/j .
celrep.2018.04.074
Hirosue, S., Senn, K., Clément, N., Nonnenmacher, M., Gigout, L., Linden,
R.M. et al. (2007) Effect of inhibition of dynein function and
microtubule- altering drugs on AAV2 transduction. Virology, 367,
10 – 18 . https://doi.org/10.1016/J.VIROL.2007.05.009
Ho, J.H.N., Kallstrom, G. & Johnson, A.W. (2000) Nmd3p is a Crm1p-
dependent adapter protein for nuclear export of the large ribo-
somal subunit. The Journal of Cell Biolog y, 151, 1057– 10 66. h t tps ://
doi.org/10.1083/JCB.151.5.1057
Hoque, M ., Ishizu, K., Mat sumoto, A., Han, S.- I ., Arisaka, F., Takayama, M.
et al. (1999) Nuclear transpor t of the major capsid protein is essen-
tial for adeno- associated virus capsid formation. Journal of Virology,
73, 7912– 7915. https://doi.org/10.1128/JVI.73.9.7912- 7915.1999
Hsu, S.L., Yu, C.T.R., Yin , S.C., Tang, M.J., Tien, A.C., Wu, Y.M. et al. (2006)
Caspas e 3, periodica lly expressed a nd activated at G2 /M transition,
is required for nocodazole- induced mitotic checkpoint. Apoptosis,
11(5), 765– 771. https://doi.org/10.10 07/S1049 5- 006- 5880- X
Hueffer, K., Parker, J.S.L., Weicher t, W.S., Geisel, R.E., Sgro, J.- Y. &
Parrish, C.R. (2003) The natural host range shift and subsequent
evolution of canine parvovirus resulted from virus- specific binding
to the canine transferrin receptor. Journal of Virology, 77, 1718–
1726. htt ps://doi.org /10.1128/JVI.7 7.3.1718- 1726 .20 03
Hutten, S., Flotho, A ., Melchior, F. & Kehlenbach, R.H. (2008) The
Nup358- RanGAP complex is required for efficient importin alpha/
beta- dependent nuclear import. Molecular Biology of the Cell, 19,
2300– 2310. https://doi.org/10.1091/MBC.E07- 12- 1279
Ihalainen, T.O., Niskanen, E.A., Jylhävä, J., Turpeinen, T., Rinne, J.,
Timonen, J. et al. (2007) Dynamic s and interactions of par voviral
NS1 protein in the nucleus. Cellular Microbiology, 9, 1946– 1959.
https://doi.org /10.1111/j.1462- 5822.20 07.00 926. x
Im, D.S. & Muzyczka, N. (1990) The AAV origin binding pro-
tein Rep68 is an ATP- dependent site- specific endonucle-
ase with DNA helicase activity. Cell, 61, 447– 457. h ttp s: //d oi.
org/10.1016/0092- 8674(90)90526 - K
Johnson, J.S., Li, C ., DiPrimio, N., Weinberg, M.S., McCown, T.J. &
Samulski, R.J. (2010) Mutagenesis of adeno- associated virus t ype
2 capsid protein VP1 uncovers new roles for basic amino acids in
trafficking and cell- specific transduction. Journal of V irology, 84,
8888– 8902. https://doi.org/10.1128/JVI.00687 - 10
Junod, S.L., Saredy, J. & Yang, W. (2021) Nuclear import of adeno-
associated viruses imaged by high- speed single- molecule micros-
cop y. Viruses, 13, 167. https://doi.org/10.3390/V1302 0167
Jurv ansuu, J., Raj, K ., Stasiak, A . & Beard, P. (2005) Vira l transport of D NA
damage that mimics a st alled replication fork. Jou rnal of Virolog y, 79,
569– 580. https://doi.org/10.1128/JVI.79.1.569- 580.2005
Kalderon, D., Roberts, B.L ., Richardson, W.D. & Smith, A.E. (1984) A
short amino acid sequence able to specify nuclear location. Cell, 39,
499– 509. https://doi.org/10.1016/0092- 8674(84)90457 - 4
Kaufmann, B., Simpson, A.A. & Rossmann, M.G. (20 04) The structure
of human parvovirus B19. Proceedings of the National Academy of
Sciences of the United States of America, 101, 11628– 11633. htt p s: //
doi.org/10 .1073/PNA S.0 4029 92101
Kaufmann, B., Chipman, P.R., Kostyuchenko, V.A., Modrow, S. &
Rossmann, M.G. (2008) Visualization of the ex ternalized VP2 N
termini of infectious human parvovirus B19. Journal of Virology, 82,
7306– 7312. https://doi.org/10.1128/JVI.00512 - 08
Kehlenbach, R.H., Dickmanns, A., Kehlenbach, A., Guan, T. & Gerace, L.
(1999) A role for RanBP1 in the release of CRM1 from the nuclear
pore comp lex in a terminal step of nuclear ex port. The Jou rnal of Cell
Biology, 145, 645– 657.
Kelich, J.M., Ma, J., Dong, B., Wang, Q., Chin, M., Magura, C.M. et al.
(2015) Super- resolution imaging of nuclear import of adeno-
associated virus in live cells. Molecular Therapy - Methods
and Clinical Development, 2, 15047. https://doi.o rg/10.1038/
mtm.2015.47
306 
|
    M ATT OL A et al .
Kelkar, S.A., Pfister, K.K., Crystal, R.G. & Leopold, P.L. (200 4)
Cytoplasmic dynein mediates adenovirus binding to microtubules.
Journal of Virology, 78, 10122– 10132. https://doi.org/10.1128/
JVI.78.18.10122 - 10132.2004
Kelkar, S., De, B.P., Gao, G., Wilson, J.M., Cr ystal, R .G. & Leopold, P.L.
(2006) A common mechanism for cytoplasmic dynein- dependent
microtubule binding shared among adeno- associated virus and ad-
enovirus serotypes. Journal of Virology, 80, 7781– 7785. h t tp s ://do i.
org/10.1128/JVI.00481 - 06
Kihlmark, M., Rustum, C., Eriksson, C., Beckman, M., Iverfeldt, K. &
Hallberg, E. (200 4) Correlation between nucleocytoplasmic trans-
port and caspase- 3- dependent dismantling of nuclear pores during
apoptosis. Experimental Cell Research, 293, 346– 356. ht t ps ://do i .
org/10.1016/j.yexcr.2003.10 .019
Kimura, M. & Imamoto, N. (2014) Biological significance of the impor-
tin- β family- dependent nucleocytoplasmic transport pathways.
Traffic, 15, 727– 748. h ttp s://doi.org /10.1111/ TR A.12174
Kontou, M., Govindasamy, L., Nam, H.- J., Bryant, N., Llamas- Saiz,
A.L ., Foces- Foces, C. et al. (2005) Structur al determinants of tis-
sue tropism and in vivo pathogenicity for the parvovirus minute
virus of mice. Journal of Virology, 79, 10931 10943. h ttp s://d o i.
org /10.1128/JVI .79.17.10931 - 10943.2005
Koyama, M . & Matsuura, Y. (2010) An a llosteric me chanism to disp lace nu-
clear export cargo from CRM1 and RanGTP by RanBP1. The EMBO
Journal, 29, 2002– 2013. ht tps://doi.o rg/10.10 38/EMBOJ.2010.89
Kutay, U., Jühlen, R. & Antonin, W. (2021) Mitotic disassembly and reas-
sembly of nuclear pore complexes. Trends in Cell Biology, 31, 1019–
1033. https://doi.org/10.1016/J.TCB.2021.06.011
Kuzmin, D. A., Shutova, M.v., Johnston, N.R., Smith, O.P., Fedorin, V.v.,
Kukushkin, Y.S. et al. (2021) The clinical landscape for AAV gene
therapies. Nature Reviews. Drug Discovery, 20, 173– 174. h ttp s: //d oi.
org /10.103 8/D4157 3- 021- 00017 - 7
Labbé, K. & Saleh, M. (2008) Cell death in the host response to infec-
tion. Cell Death and Differentiation, 15, 1339– 1349. ht t ps: //d oi .
org /10.103 8/CDD.2008.91
Large, E.E., Silveria, M.A ., Zane, G .M., Weerakoon, O. & Chapman, M.S.
(2021) Adeno- associated virus (AAV) gene delivery: dissecting mo-
lecular interactions upon cell entry. Viruses, 13, 1336. h ttp s://d oi.
org/10.3390/V1307 1336
Lee, B.J., Cansizoglu, A.E., Süel, K.E., Louis, T.H., Zhang, Z. & Chook,
Y.M. (2006) Rules for nuclear localization sequence recognition by
karyopherin beta 2. Cell, 126, 543– 558. https://doi.org/10.1016/J.
CELL.2006.05.049
Leisi, R., di Tommaso, C., Kempf, C. & Ros, C. (2016) The receptor- binding
domain in the VP1u region of parvovirus B19. Viruses, 8, 61. ht t ps : //
doi.org/10.3390/V8030061
Li, X. & Rhode, S.L ., 3rd. (1990) Mutation of lysine 4 05 to serine in
the par vovirus H- 1 NS1 abolishes its functions for viral DNA
replication, late promoter trans activation, and cytotoxicity.
Journal of Virology, 64, 4654– 4660. https://doi.org/10.1128/
JVI.64.10.4654- 4660.1990
Liu, J., Prunuske, A. J., Fager, A.M. & Ullman, K. S. (2003) The COPI com-
plex functions in nuclear envelope breakdown and is recruited by
the nucleoporin Nup153. Developmental Cell, 5, 487– 498. h t tps ://
doi.org/10 .1016/S1534 - 5807( 03)00262 - 4
Llamas- Saiz, A.L., Agbandje- McKenna, M., Parker, J.S.L., Wahid, A.T.M.,
Parris h, C.R. & Rossm ann, M.G. (1996) Structural analysis of a m uta-
tion in ca nine parvovi rus which contro ls antigenicit y and host ran ge.
Virology, 225, 65– 71. https://doi.org/10.10 06/VIRO.1996 .0575
Lombardo, E., Ramírez, J.C., Agbandje- McKenna, M. & Almendral, J.M.
(200 0) A beta- str anded motif drives capsid protein oligomer s of the
parvovirus minute virus of mice into the nucleus for viral assem-
bly. Journal of Virology, 74, 3804– 3814. https://doi.org/10.1128/
JVI.74.8.3804- 3814.2000
Lyi, S.M., Tan, M.J.A. & Parrish, C.R. (2014) Parvovirus parti-
cles and movement in the cellular cytoplasm and ef fects of
the cytoskeleton. Virology, 456– 457, 342– 352. ht t ps: //d oi .
org/10.1016/J.VIROL.2014.04.003
Lyngdoh, D.L ., Nag, N., Uversky, V.N. & Tripathi, T. (2021) Prevalence
and func tionalit y of intrinsic disorder in human FG- nucleoporins.
International Journal of Biological Macromolecules, 175, 156– 170.
https://doi.org/10.1016/J.IJBIO MAC.2021.01.218
Mahadevan, K., Zhang, H., Akef, A., Cui, X.A., Gueroussov, S., Cenik ,
C. et al. (2013) RanBP2/Nup358 potentiates the translation of a
subset of mRNAs encoding secretor y proteins. PLoS Biology, 11,
e1001545. https://doi.org/10.1371/JOURN AL.PBIO.1001545
Mäntylä, E., Chacko, J.v., Aho, V., Parrish, C.R ., Shahin, V., Kann, M. et al.
(2018) Viral highway to nucleus exposed by image correlation anal-
yses. Scientific Reports, 8, 1– 11. https://doi.org/10.1038/s4159 8-
018- 19582 - w
Mäntylä, E., Aho, V., Kann, M. & Vihinen- Ranta, M. (2020) Cytoplasmic
parvovirus capsids recruit importin Beta for nuclear delivery. Journal
of Virology, 94, e 01532- 19. https://doi.org/10.1128/JVI.01532 - 19
Marchini, A., Bonifati, S., Scott, E.M., Angelova, A .L. & Rommelaere, J.
(2015) Oncolytic par voviruses: from basic virology to clinical appli-
cations. Virology Journal, 12, 1– 16. https://doi.org/10.1186/S1298
5- 014- 0223- Y/FIGUR ES/3
Maroto, B., Valle, N., Saffrich, R. & Almendral, J.M. (2004) Nuclear export
of the nonenveloped parvovirus virion is directed by an unordered
protein signal exposed on the capsid surface. Journal of Virology, 78,
10685– 10694. https://doi.org/10.1128/jvi.78.19.10685 - 10694.2004
Meier, A.F., Fraefel, C. & Seyffert, M. (2020) The interplay between
adeno- associated virus and its helper viruses. Viruses, 12, 662.
Mendell, J.R., Al- Zaidy, S.A., Rodino- Klapac, L.R., Goodspeed, K., Gray,
S.J., Kay, C.N. et al. (2021) Current clinical applic ations of in vivo
gene therapy with AAVs. Molecular Therapy, 29, 464– 488. h ttp s://
doi.org/10.1016/J.YMTHE.2020.12.007
Meyer, N.L. & Chapman, M.S. (2022) Adeno- associated virus (AAV)
cell entr y: struc tural insights. Trends in Microbiology, 30, 432– 451.
https://doi.org/10.1016/j.tim.2021.09.005
Michelfelder, S. & Trepel, M. (2009) Adeno- associated viral vectors and
their re directio n to cell- t ype specif ic receptor s. Advances in G enetics,
67, 29– 60. https://doi.org/10.1016/S0065 - 2660(09)67002 - 4
Mietzsch, M., Pénzes, J.J. & Agbandje- Mckenna, M. (2019) Twenty- five
years of structural parvovirology. Viruses, 11, 362. h t tps ://do i.
org/10.3390/V1104 0362
Miller, C.L. & Pintel, D. J. (2002) Interaction bet ween parvovirus NS2 pro-
tein and nuclear export factor Crm1 is important for viral egress
from the nucleus of murine cells. Jour nal of Virology, 76, 3257– 3266.
https://doi.org/10.1128/jvi.76.7.3257- 3266.2002
Mitrousis, G., Olia, A.S., Walker- Kopp, N. & Cingolani, G. (2008)
Molecular basis for the recognition of snurportin 1 by importin
beta. The Journal of Biological Chemistry, 283, 7877– 7884. h ttp s: //
doi.org/10.1074/JBC.M7090 93200
Morita, E., Nakashima, A., Asao, H., Sato, H. & Sugamura, K. (2003)
Human parvovirus B19 nonstructural protein (NS1) induces cell
cycle arrest at G 1 phase. Journal of Virology, 77, 2915– 2921.
https://doi.org/10.1128/JVI.77.5.2915- 2921.2003
Moy, T.I. & Silver, P.A. (1999) Nuclear export of the small ribosomal
subunit requires the ran- GTPase cycle and certain nucleoporins.
Genes & Development, 13, 2118– 2133. https://doi.org/10.1101/
GAD.13.16.2118
Nicolson, S.C. & Samulski, R.J. (2014) Recombinant adeno- associated
virus utilizes host cell nuclear import machinery to enter the nu-
cleus. Journal of Virology, 88, 4132– 414 4. https://doi.org/10.1128/
JVI.02660 - 13
Nüesch, J.P.F., Lachmann, S. & Rommelaere, J. (20 05) Selective al-
terations of the host cell architecture upon infection with par-
vovirus minute virus of mice. Virology, 331, 159– 174. ht tps :// do i.
org/10.1016/J.VIROL.2004.10.019
Nykky, J., Tuusa, J.E., Kirjavainen, S., Vuento, M. & Gilber t, L. (2010)
Mechanisms of cell death in canine par vovirus- infected cells
   
|
307
MAT TOLA et al.
provide intuitive insights to developing nanotools for medicine.
International Journal of Nanomedicine, 5, 417– 428. h ttp s://d o i.
org /10.2147/ijn.s10579
Ogden, P.J., Kelsic, E.D., Sinai, S. & Church, G.M. (2019) Comprehensive
AAV capsid fitness landscape reveals a viral gene and enables
machine- guided design. Science, 366, 1139– 1143. http s://d o i.
org /10.1126/SC IEN CE.A AW290 0
Oldrini, B., Hsieh, W.Y., Erdjument- Bromage, H., Codega, P., Carro, M.S.,
Curiel- García, A. et al. (2017) EGFR feedback- inhibition by ran-
binding protein 6 is disrupted in cancer. Nature Communications, 8,
1– 12. https://doi.org/10.1038/s4146 7- 017- 02185 - w
Palermo, L.M., Hafenstein, S.L. & Parrish, C.R. (2006) Purified feline
and canine transferrin receptors reveal complex interac tions with
the capsids of canine and feline parvoviruses that correspond to
their host ranges. Journal of Virology, 80, 8482– 8492. ht t ps: //d oi .
org/10.1128/JVI.00683 - 06
Panté, N. & K ann, M. (20 02) Nuclear pore complex is able to transport
macromolecules with diameters of 39 nm. Molecular Biology of the
Cell, 13, 425– 434. https://doi.org/10.1091/mbc.01- 06- 0308
Parker, J.S.L. & Parrish, C.R. (2000) Cellular uptake and infection by
canine parvovirus involves rapid dynamin- regulated clathrin-
mediated endocytosis, followed by slower intracellular traffick-
ing. Journal of Virology, 74, 1919– 1930 . https://doi.org/10.1128/
JVI.74.4 .1919- 1930 .20 00
Parker, J.S.L., Murphy, W.J., Wang, D., O'Brien, S.J. & Parrish,
C.R. (2001) Canine and feline parvoviruses can use human
or feline transferrin receptors to bind, enter, and infect cells.
Journal of Virology, 75, 3896– 3902. https://doi.org/10.1128/
JVI.75.8.3896- 3902.2001
Parrish, C.R. (1990) Emergence, natural histor y, and variation of canine,
mink, and feline parvoviruses. Advances in Virus Research, 38, 4 03–
450. ht tps://doi.o rg/10.1016/S0065 - 3527(08)6 0867 - 2
Pénzes, J. J., Söderlund- Venermo, M., Canuti, M., Eis- Hübinger, A.M.,
Hughes, J., Cotmore, S.F. et al. (2020) Reorganizing the family par-
voviridae: a revised taxonomy independent of the canonical ap-
proach based on host association. A rchives of Virology, 165, 2133–
2146 . https://doi.org/10.1007/S0070 5- 020- 04632 - 4
Petosa, C ., Schoehn, G., Askjaer, P., Bauer, U., Moulin, M., Steuerwald, U.
et al. (2004) Architecture of CRM1/exportin1 suggests how coop-
erativity is achieved during formation of a nuclear export complex.
Molecular Cell, 16, 761– 7 75 .
Pillay, S., Meyer, N.L., Puschnik, A.S., Davulcu, O., Diep, J., Ishikawa, Y.
et al. (2016) An essential receptor for adeno- associated virus in-
fection. Nature, 530, 108– 112. ht tps://doi.o rg/10.10 38/NATUR
E16465
Popa- Wagner, R., Porwal, M., Kann, M., Reuss, M., Weimer, M., Florin,
L. et al. (2012) Impact of VP1- specific protein sequence motifs on
adeno- associated virus type 2 intracellular trafficking and nuclear
ent ry. Journal of Virology, 86, 9163– 9174. https://doi.org/10.1128/
JVI.0 0282 - 12/ASSET/ 92553 E70- AD11- 4AE8- 9082- 8A4B2 37012
37/ASSET S/GRAPH IC/ZJV99 90964 010006.JPEG
Popken, P., Ghavami, A., Onck, P.R., Poolman, B. & Veenhoff, L.M. (2015)
Size- dependent lea k of soluble and membrane proteins through the
yeast nuclear pore complex. Molecular Biology of the Cell, 26, 1386–
1394. ht tps://doi.o rg/10.10 91/mbc .E14- 07- 1175
Porwal, M., Cohen, S., Snoussi , K., Popa- Wagner, R., Anderson, F., Dugot-
Senant, N. et al. (2013) Parvoviruses cause nuclear envelope break-
down by activating key enzymes of mitosis. PLoS Pathogens, 9,
e1003671. https://doi.org/10.1371/JOURN AL.PPAT.1003671
Prunus ke, A.J., Li u, J., Elgort , S., Joseph, J. , Dasso, M. & Ullma n, K.S. (2 006)
Nuclear envelope breakdown is coordinated by both Nup358/
RanBP2 and Nup153, two nucleoporins with zinc finger modules.
Molecular Biology of the Cell, 17, 760. https://doi.org/10 .1091/MBC.
E05- 06- 0485
Qing, K., Mah, C., Hansen, J., Zhou, S., Dwarki, V. & Srivastava, A. (1999)
Human fibroblast growth factor receptor 1 is a co- receptor for
infection by adeno- associated virus 2. Nature Medicine, 5, 71– 77.
https://doi.o rg/10.10 38/4758
Qu, X.W., Liu, W.P., Qi, Z.Y., Duan, Z .J., Zheng , L.S., Ku ang, Z.Z . et al. (200 8)
Phospholipase A2- like activity of human bocavirus VP1 unique re-
gion. Biochemical and Biophysical Research Communications, 365,
158– 163. ht tps: //doi.org/10 .1016/J.BBRC.2007.10.164
Raj, K., Ogston, P. & Beard, P. (2001) Virus- mediated killing of
cells that lack p53 activity. Nature, 412, 914– 917. h t tp s ://do i.
org/10.1038/35091082
Riolobos, L., Reguera, J., Mateu, M.G. & Almendral, J.M. (2006) Nuclear
transport of trimeric assembly intermediates exerts a morpho-
genetic control on the icosahedral parvovirus capsid. Journal of
Molecular Biology, 357, 1026– 1038. https://doi.org/10.1016/J.
JMB.20 06.01.019
Ritterhoff, T., Das, H., Hofhaus, G., Schröder, R.R ., Flotho, A . & Melchior,
F. (2016) The RanBP2/RanGAP1*SUMO1/Ubc9 SUMO E3 ligase is
a disassembly machine for Crm1- dependent nuclear export com-
plexes. Nature Communications, 7, 11482. htt ps://doi.org/10.1038/
NCOMM S11482
Roehrig, S., Tabbert, A. & Ferrando- May, E. (2003) In vitro measure-
ment of nuclear permeability changes in apoptosis. Analytical
Biochemistry, 318, 244– 253. https://doi.org/10 .1016/S0 003
- 2697(03)00242 - 2
Roos, W.P. & Kaina, B. (2006) DNA damage- induced cell death by
apoptosis. Trends in Molecular Medicine, 12, 440– 450. ht t ps: //doi .
org/10.1016/j.molmed.2006.07.007
Ros, C. & Kempf, C. (2004) The ubiquitin- proteasome machinery is
essential for nuclear translocation of incoming minute virus
of mice. Virology, 324, 350– 360. https://doi.org/10.1016/J.
VIROL.2004.04.016
Ros, C., Baltzer, C., Mani, B. & Kempf, C. (2006) Parvovirus uncoating in
vitro reve als a mechanism of DNA release without capsid disassem-
bly and striking dif ferences in encapsidated DNA stability. Virology,
345, 137– 147. https://doi.org/10.1016/J.VIROL.2005.09.030
Ros, C., Bieri, J. & Leisi, R. (2020) The vp1u of human parvovirus b19: a
multifunctional capsid protein with biotechnological applications.
Viruses, 12, 1– 21. https://doi.org/10.3390/v1212 1463
Ruiz, Z., D'Abramo, A. & Tattersall, P. (2006) Dif ferential roles for the
C- terminal hexapeptide domains of NS2 splice variants during
MVM infec tion of murine cells. Virology, 349, 382– 395. h t tps ://do i.
org/10.1016/J.VIROL.2006.01.039
Ruiz, Z., Mihaylov, I.S., Cotmore, S.F. & Tattersall, P. (2011) Recruitment
of DNA replication and damage response proteins to viral replica-
tion centers during infection with NS2 mutants of minute virus of
mice (MVM). Virology, 410, 375– 384. https://doi.org/10.1016/J.
VIROL.2010.12.009
Sachdev, S., Bagchi, S., Zhang, D.D., Mings, A.C. & Hannink, M. (2000)
Nuclear import of IκBα is accomplished by a ran- independent
transport pathway. Molecular and Cellular Biology, 20, 1571– 1582.
https://doi.org/10.1128/mcb.20.5.1571- 1582.2000
Sánchez- Martínez, C., Grueso, E., Carroll, M., Rommelaere, J. &
Almendral, J.M. (2012) Essential role of the unordered VP2 n-
terminal domain of the parvovirus MVM capsid in nuclear assem-
bly and endosomal enlargement of the virion fivefold channel
for cell entry. Virology, 432, 45– 56. https://doi.org/10.1016/J.
VIROL.2012.05.025
Saudan, P., Vlach, J. & Beard, P. (2000) Inhibition of S- phase progression
by adeno- associated virus Rep78 protein is mediated by hypophos-
phorylated pRb. The EMBO Journal, 19, 4351– 4361. h t tp s ://do i.
org /10.1093/EMBOJ/ 19.16.4351
Schmitz, A., Schwarz, A., Foss, M., Zhou, L., Rabe, B., Hoellenriegel, J.
et al. (2010) Nu cleoporin 153 ar rests the nucl ear import of hepatitis
B virus capsids in the nuclear basket. PLoS Pathogens, 6, e100 0741.
ht tps://doi. org/10 .1371/JO UR N AL. PPAT.100 0741
Schwartz, R. A., Carson, C.T., Schuberth, C. & Weit zman, M.D. (2009)
Adeno- associated virus replication induces a DNA damage
308 
|
    M ATT OL A et al .
response coordinated by DNA- dependent protein kinase. Journal
of Virology, 83, 6269– 6278. https://doi.org/10.1128/jvi.00318 - 09
Seisen berger, G., Ried , M.U., Endreß, T., Büni ng, H., Hallek , M. & Bräuchl e,
C. (2001) Real- time single- molecule imaging of the infection path-
way of an aden o- asso ciated virus. Science, 294, 1929– 1932. h t tps ://
doi.org/10 .1126/SCIEN CE.1064103
Smith, G .A. & Enquist, L.W. (2002) Break ins and break outs: viral inter-
actions with the cy toskeleton of mammalian cells. Annual Review of
Cell and Developmental Biology, 18, 135– 161.
Sonntag, F., Bleker, S., Leuchs, B., Fischer, R. & Kleinschmidt, J.A . (2006)
Adeno- associated virus type 2 capsids with externalized VP1/ VP2
trafficking domains are generated prior to passage through the cy-
toplasm and are maintained until uncoating occurs in the nucleus.
Journal of Virology, 80, 110 40– 1105 4. https://doi.org/10.1128/
JVI .01056 - 06
Sonntag, F., Schmidt, K. & Kleinschmidt, J.A . (2010) A viral assembly fac-
tor promotes AAV2 capsid formation in the nucleolus. Proceedings
of the National Academy of Sciences of the United States of America,
107, 10220– 10225. https ://doi.org/10.1073/PNAS.10 016 73107/ - /
DCSUP PLEME NTAL/PNAS.20100 1673SI.PDF
Strasser, C., Grote, P., Schäuble, K., Ganz, M. & Ferrando- May, E. (2012)
Regulation of nuclear envelope permeability in cell death and sur-
vival. Nucleus (United States), 3, 540– 551. https://doi.org/10.4161/
nucl.21982
Stuwe, T., Bley, C. J., Thierbach, K., Petrovic, S., Schilbach, S., Mayo, D.J.
et al. (2015) Architecture of the fungal nuclear pore inner ring
complex. Science, 350, 56– 64. https://doi.org/10.1126/SCIEN
CE .A AC9176
Suikkanen, S., Aaltonen, T., Nevalainen, M., Välilehto, O., Lindholm, L.,
Vuento, M. et al. (2003a) Exploitation of microtubule cytoskel-
eton and dynein during parvoviral traffic toward the nucleus.
Journal of Virology, 77, 10270– 10279. https://doi.org/10.1128/
JVI.77.19.10270 - 10279.2003
Suikkanen, S., Antila, M., Jaatinen, A., V ihinen- Ranta, M. & Vuento,
M. (2003b) Release of canine parvovirus from endocytic ves-
icles. Virology, 316, 267– 280. https://doi.org/10.1016/J.
VIROL.2003.08.031
Summerford, C. & Samulski, R.J. (1998) Membrane- associated heparan
sulfate proteoglycan is a receptor for adeno- associated virus t ype 2
virions. Journal of Vir ology, 72, 1438– 1445. https://doi.org/10.1128/
JVI.72.2.14 38- 1445.1998
Summer ford, C., Bartlet t, J.S. & Samulski, R.J. (1999) AlphaVbeta5 in-
tegrin: a co- receptor for adeno- associated virus type 2 infection.
Nature Medicine, 5, 78– 82. https://doi.org/10.1038/4768
Thomas, F. & Kutay, U. (2003) Biogenesis and nuclear export of ribo-
somal subunits in higher eukaryotes depend on the CRM1 ex-
port pathway. Journal of Cell Science, 116, 240 9– 2419. ht t ps ://do i .
org/10.1242/JCS.00464
Timney, B.L., Raveh, B., Mironska, R., Trivedi, J.M., Kim, S.J., Russel, D.
et al. (2016) Simple rules for passive diffusion through the nuclear
pore complex. The Journal of Cell Biology, 215, 57– 76. ht t ps: //d oi .
org/10.1083/ jcb.20160 10 04
Tollefson, A .E., Scaria, A., Hermiston, T.W., Ryerse, J. S., Wold, L. J. &
Wold, W.S. (1996) The adenovirus death protein (E3- 11.6K) is re-
quired at very late stages of infection for efficient cell lysis and
release of adenovirus from infected cells. Journal of Virolog y, 70,
22 96– 2306. https://doi.org/10.1128/jvi.70.4.2296- 2306.1996
Tsao, J., Chapman, M.S., A gbandje, M., Keller, W., Smith, K ., Wu, H. et al.
(1991) The three- dimensional structure of canine parvovirus and
its functional implications. Science, 251, 1456– 146 4. h t tps ://do i.
org/10.1126/SCIEN CE.2006420
Vihinen- Ranta, M., Wang, D., Weichert, W.S. & Parrish, C.R. (2002)
The VP1 N- terminal sequence of canine par vovirus affects nu-
clear transport of c apsids and ef ficient cell infection. Journal of
Virology, 76 , 1884 1891. ht tps://d oi.org/10.1128/JVI.76.4.1884-
1891. 2002
Wälde, S., Thakar, K., Hutten, S., Spillner, C., Nath, A., Rothbauer, U. et al.
(2012) The nucleoporin Nup358/RanBP2 promotes nuclear import
in a cargo- and transport receptor- specific manner. Traffic, 13, 218–
233. ht tps://doi.o rg/10 .1111/J.1600- 085 4.2011.01302.X
Walther, T.C., Fornerod, M., Pickersgill, H., Goldberg, M., Allen, T.D. &
Mattaj, I.W. (2001) The nucleoporin Nup153 is required for nuclear
pore basket formation, nuclear pore complex anchoring and import
of a subset of nuclear protei ns. The EMBO Journal, 20, 5703. ht t ps : //
doi.org/10.1093/EMBOJ/ 20.20.5703
Wang, R. & Brattain, M.G. (2007) The maximal size of protein to diffuse
through the nuclear pore is larger than 60 kDa. FEBS Letters, 581,
3164– 3170. https://doi.org/10.1016/j.febsl et.2007.05.082
Wiethoff, C.M. & Nemerow, G.R. (2015) Adenovirus membrane penetra-
tion: tickling the tail of a sleeping dragon. Virology, 479– 48 0, 591–
599. https://doi.org/10.1016/J.VIROL.2015.03.006
Winocour, E., Callaham, M.F. & Huberman, E. (1988) Perturbation of the
cell cycle by adeno- associated virus. Virology, 167, 393– 399.
Wistuba, A., Kern, A., Weger, S., Grimm, D. & Kleinschmidt, J. A. (1997)
Subcellular compartmentalization of adeno- associated virus
type 2 assembly. Journal of Virology, 71, 1341– 1352. h ttp s://d o i.
org /10.1128/JVI .71.2 .1341- 1352.1997
Wolfisberg, R., Kempf, C. & Ros, C. (2016) Late maturation steps pre-
ceding selective nuclear expor t and egress of progeny parvovi-
rus. Journal of Virology, 90, 5462– 5474. https://doi.org/10.1128/
jvi.02967 - 15
Xiao, P.- J. & Samulski, R.J. (2012) Cytoplasmic trafficking, endosomal
escape, and perinuclear accumulation of adeno- associated virus
type 2 particles are facilitated by microtubule network. Journal of
Virology, 86, 10462– 10473. https://doi.org/10.1128/JVI.00935 - 12
Xie, Q., Bu , W., Bhatia , S., Hare, J., Soma sundaram, T., Azz i, A. et al. (20 02)
The atomic structure of adeno- associated virus (AAV- 2), a vector
for human gene therapy. Proceedings of the National Academy of
Sciences of the United States of America, 99, 10 405– 10410. ht t ps: //
doi.org/10.1073/PNAS.16225 0899
Yan, Z., Zak, R., Luxton, G.W.G., Ritchie, T.C., Bantel- Schaal, U. &
Engelhardt, J.F. (2002) Ubiquitination of both adeno- associated
virus type 2 and 5 capsid proteins affects the transduc tion effi-
ciency of recombinant vectors. Journal of Virology, 76, 2043– 2053.
https://doi.org/10.1128/JVI.76.5.2043- 2053.2002
Yoshimura, S .H., Kumeta, M . & Takeyasu, K. (2014) Stru ctural mechanism
of nuclea r transport m ediated by impor tin β and flexible amphiphilic
proteins. Structure, 22, 1699– 1710. https://doi.org/10.1016/J.
STR.2014.10.009
Zádori, Z., Szelei, J., Lacoste, M.C., Li, Y., Gariépy, S., Raymond, P. et al.
(2001) A viral phospholipase A2 is required for parvovirus infectiv-
it y. Developmental Cell, 1, 291– 302. https://doi.org/10.1016/S1534
- 5807(01)00031 - 4
Zhao, H., Cheng, Y., Wang, J., Lin, P., Yi, L., Sun, Y. et al. (2016) Profiling of
host cell response to successive canine parvovirus infection based
on kinetic proteomic change identification. Scientific Reports, 6, 1–
11. https://doi.org/10.1038/srep2 9560
Zhong, L ., Li, B., Jayandharan, G., Mah, C.S., Govindasamy, L., Agbandje-
McKenna, M. et al. (200 8) Tyrosine- phosphorylation of AAV2 vec-
tors and i ts consequen ces on viral intr acellular tra fficking an d trans-
gene expr ession. Virology, 381, 194– 202. ht tps://doi.org/10.1016/J.
VIROL.2008.08.027
How to cite this article: Mattola, S., Aho, V., Bustamante-
Jaramillo, L. F., Pizzioli, E., Kann, M. & Vihinen- Ranta, M. (2022)
Nuclear entry and egress of parvoviruses. Molecular
Microbiology, 118, 295–308. ht tps://doi.org /10.1111/
mmi.14974
... NP1 processes viral pre-mRNA. NP1 is needed to define the big 3′ exon, the VP-encoding exon (Shao et al., 2021;Mattola et al., 2022). ...
... The efficient assembly of these VLPs is essential for the virus's ability to establish and maintain an infection. Understanding the role of HBoV-1's capsid proteins and their interactions with host cells is crucial for developing effective antiviral therapies and vaccines (Kailasan et al., 2016;Liu et al., 2017;Ros et al., 2017;Malta et al., 2020;Fakhiri and Grimm, 2021;Shao et al., 2021;Mattola et al., 2022;Xie et al., 2022). ...
... Understanding the structural and functional differences between different strains of HBoV is critical for developing effective strategies for diagnosing and treating viral infections. By examining how these viruses interact with host cells and evade the immune system, researchers can gain insight into the underlying mechanisms of viral infection and identify new targets for drug and vaccine development (Babkin et al., 2013;Abramczuk et al., 2015;Liu et al., 2017;Qiu et al., 2017;Ros et al., 2017;Christensen et al., 2019;Malta et al., 2020;Bhat and Almajhdi, 2021;Shao et al., 2021;Mattola et al., 2022;Xie et al., 2022). ...
Article
Full-text available
The single-stranded DNA virus known as human bocavirus 1 (HBoV-1) is an icosahedral, linear member of the Parvoviridae family. In 2005, it was discovered in nasopharyngeal samples taken from kids who had respiratory tract illnesses. The HBoV genome is 4.7–5.7 kb in total length. The HBoV genome comprises three open-reading frames (ORF1, ORF2, and ORF3) that express structural proteins (VP1, VP2, and VP3), viral non-coding RNA, and non-structural proteins (NS1, NS1-70, NS2, NS3, and NP1) (BocaSR). The NS1 and NP1 are crucial for viral DNA replication and are substantially conserved proteins. Replication of the HBoV-1 genome in non-dividing, polarized airway epithelial cells. In vitro, HBoV-1 infects human airway epithelial cells that are strongly differentiated or polarized. Young children who have HBoV-1 are at risk for developing a wide range of respiratory illnesses, such as the common cold, acute otitis media, pneumonia, and bronchiolitis. The most common clinical symptoms are wheezing, coughing, dyspnea, and rhinorrhea. After infection, HBoV-1 DNA can continue to be present in airway secretions for months. The prevalence of coinfections is considerable, and the clinical symptoms can be more severe than those linked to mono-infections. HBoV-1 is frequently detected in combination with other pathogens in various reports. The fecal-oral and respiratory pathways are more likely to be used for HBoV-1 transmission. HBoV-1 is endemic; it tends to peak in the winter and spring. This Review summarizes the knowledge on HBoV-1.
... Their upregulation provides additional evidence supporting the facilitation of nuclear import of AAV particles or viral components, potentially impacting viral replication or gene expression. [20][21][22][23][24] Additionally, several other biosynthetic pathways were upregulated, such as oligosaccharide-lipid intermediate processes (e.g., ALG12, MPDU1), cellular lipid biosynthesis, and glycerol ether biosynthesis (e.g., FAR1, ERLIN1) at 24 hpt. In parallel, downregulated GO biological processes included DNA-templated transcription (e.g., MAZ, SSU72), autophagy (e.g., MTMR9, FOXK1, ATP6V1C1), cellular localization, and ubiquitin protein-dependent catabolic processes (e.g., UBE2C, RNF25, FBXO7). ...
... Protein-protein interaction network reveals role of HEK293T proteins in AAV replication, assembly, and protein synthesis Our investigation showed dynamic regulation of the host proteome, which plays a pivotal role in governing the processes of AAV2 replication, assembly, and protein expression. From an extensive review of existing literature [20][21][22][23]26 and the VirHostNet 2.0 database, 24,27 we identified a subset of 124 differentially expressed host proteins associated with virus-host interactions in optimal transfectants compared with standard transfectants in at least one post-transfection time point assessed (Table S4). ...
Article
Full-text available
The gene therapy field seeks cost-effective, large-scale production of recombinant adeno-associated virus (rAAV) vectors for high-dosage therapeutic applications. Although strategies like suspension cell culture and transfection optimization have shown moderate success, challenges persist for large-scale applications. To unravel molecular and cellular mechanisms influencing rAAV production, we conducted an SWATH-MS proteomic analysis of HEK293T cells transfected using standard, sub-optimal, and optimal conditions. Gene Ontology and pathway analysis revealed significant protein expression variations, particularly in processes related to cellular homeostasis, metabolic regulation, vesicular transport, ribosomal biogenesis, and cellular proliferation under optimal transfection conditions. This resulted in a 50% increase in rAAV titer compared with the standard protocol. Additionally, we identified modifications in host cell proteins crucial for AAV mRNA stability and gene translation, particularly regarding AAV capsid transcripts under optimal transfection conditions. Our study identified 124 host proteins associated with AAV replication and assembly, each exhibiting distinct expression pattern throughout rAAV production stages in optimal transfection condition. This investigation sheds light on the cellular mechanisms involved in rAAV production in HEK293T cells and proposes promising avenues for further enhancing rAAV titer during production.
... According to the current model of AAV transduction, capsids escape into the cytosol after endosomal uptake and become transported into the nucleus as intact capsids. [6][7][8] It is within the nucleus that the ssDNA becomes accessible for further processing. 9 An important step in the uncoating of AAVs is that the extended N-termini of VP1 and VP2 that normally reside within the capsid, emerge outward just before endosomal escape, in a likely pH-triggered event. ...
... [10][11][12][13] This initial step then uncovers different nuclear localization sequences which are vital for nuclear uptake of AAVs either by the nuclear pore complex or via pore formation in the nuclear envelope. 6,8 Once inside the nucleus, however, the process of AAV uncoating and ssDNA release remains elusive. ...
Preprint
Full-text available
Adeno-associated viruses (AAVs) are gaining traction as delivery vehicles for gene therapy although the molecular understanding of AAV-transgene release is still limited. Typically, the process of viral uncoating is investigated ( in vitro ) through thermal stress, revealing capsid disintegration at elevated temperatures. Here, we used single-molecule interferometric scattering microscopy to assess the (in)stability of different empty and filled AAV preparations. By introducing a heat-stable DNA plasmid as an internal standard, we quantitatively probed the impact of heat on AAVs. Generally, empty AAVs exhibited greater heat resistance than genome-filled particles. Our data also indicate that upon DNA release, the capsids do not transform into empty AAVs, but seem to aggregate or disintegrate. Strikingly, some AAVs exhibited an intermediate state with disrupted capsids but preserved bound genome, a feature that experimentally only emerged following incubation with a nuclease. Our data demonstrate that the thermal uncoating process is highly AAV specific ( i . e ., can be influenced by serotype, genome, host system). We argue that nuclease treatment in combination with mass photometry can be used as an additional analytical tool for assessing structural integrity of recombinant and/or clinical AAV vectors.
... La proteína NS2 complementa la función apoptótica de NS1. Este efecto se desarrolla a través de distintas vías en función del virus y de la célula hospedadora (Mattola et al. 2022). ...
Article
Full-text available
Los parvovirus pertenecen a un grupo de virus no envueltos, capaces de infectar en una amplia variedad de hospedadores domésticos y silvestres, pudiendo causar diversos cuadros clínicos como fallas reproductivas en cerdos y bovinos, enteritis en caninos y aves de corral, panleucopenia en felinos, hepatitis en equinos, enfermedad respiratoria y cutánea en humanos. Estos virus son mayormente especie-específicos, aunque hay evidencia de transmisión interespecie, especialmente en animales silvestres. Su genoma está compuesto por una cadena lineal de ADN, de aproximadamente 5 kb, cuyas secuencias terminales son complejos palindrómicos en forma de horquilla, compuestos por 120 a 200 bases. Se encuentran ampliamente distribuidos y son muy estables en diversas condiciones ambientales, capaces de permanecer infectivos durante largos períodos. Los hospedadores susceptibles se infectan por contacto directo con individuos infectados o fómites y la infección puede ocasionar cuadros clínicos con signos diversos según la especie afectada. Además, algunos hospedadores desarrollan cuadros subclínicos que pueden eliminar el virus en secreciones y excreciones. Para su diagnóstico se emplean técnicas serológicas y moleculares, siendo la PCR la de mayor sensibilidad y especificidad. El tratamiento para los cuadros causados en animales de compañía se basa en la reversión de los signos mediante fluidoterapia, el uso de antimicrobianos de amplio espectro, antieméticos, antiácidos y protectores de la mucosa gástrica. Además, se han comenzado a utilizar antivirales, inmunomoduladores y probióticos para revertir el cuadro clínico. Por otra parte, no existe tratamiento para los cuadros clínicos en animales de producción. En producción porcina el manejo se basa en la prevención a través de la utilización de vacunas inactivadas y medidas de bioseguridad. En la presente revisión se describirán los diversos cuadros clínicos asociados a parvovirus en especies hospedadoras de interés en medicina veterinaria, y aspectos referentes a su clasificación taxonómica, epidemiología, patogenia, diagnóstico tratamiento y prevención.
... Parvoviruses enter cells by receptor-mediated endocytosis, which includes the binding of virions to specific receptors on the cell surface, followed by internalization of the virions into the host cell (9,13). However, the HBoV1 receptors remain unknown. ...
Article
Full-text available
Human bocavirus 1 (HBoV1) has appeared as an emerging pathogen, causing mild to life-threatening respiratory tract infections, acute otitis media, and encephalitis in young children and immunocompromised individuals. The lack of cell lines suitable for culturing replicative viruses hinders research on HBoV1. Here, we characterized the susceptibility to HBoV1 of 29 human and 7 animal cell lines, and identified a permissive cell line, MA104. The complete HBoV1 life cycle was achieved in MA104 cells, including viral entry, complete replication, and infectious progeny virion production. Additionally, the suppression of the interferon pathway facilitated the viral genome replication in MA104 cells. RNA-sequencing showed that innate immunity, inflammation, the PI3K-Akt and MAPK signaling pathways, and the cellular membrane system were mobilized in response to HBoV1 infection. Overall, our study is the first to identify a cell line, MA104, that supports the complete HBoV1 life cycle, which will promote research on HBoV1 virology and pathogenesis and benefit drug and vaccine development. IMPORTANCE HBoV1 is an emerging pathogen that mainly causes respiratory tract infections, while the lack of cell lines suitable for culture replicative viruses hindered research on HBoV1. Here, we identify a permissive cell line for HBoV1 infection, MA104, and reveal that the complete life cycle of HBoV1 was supported in MA104 cells. Our findings provide a suitable cell model for the study of HBoV1 and explore its application for antiviral drug evaluation, which is vital for research on HBoV1 virology and pathogenesis, as well as for drug and vaccine development.
... A long-standing unanswered topic is how and where the capsid uncoating for gene expression before entering the nucleus (42,43). Earlier studies have shown that in the infectious parvovirus particles, the capsid surface exposed the 5' end 20-30 nucleotides (44)(45)(46)(47). ...
Article
Full-text available
Parvoviruses are a group of non-enveloped DNA viruses that have a broad spectrum of natural infections, making them important in public health. NS1 is the largest and most complex non-structural protein in the parvovirus genome, which is indispensable in the life cycle of parvovirus and is closely related to viral replication, induction of host cell apoptosis, cycle arrest, DNA damage response (DDR), and other processes. Parvovirus activates and utilizes the DDR pathway to promote viral replication through NS1, thereby increasing pathogenicity to the host cells. Here, we review the latest progress of parvovirus in regulating host cell DDR during the parvovirus lifecycle and discuss the potential of cellular consequences of regulating the DDR pathway, targeting to provide the theoretical basis for further elucidation of the pathogenesis of parvovirus and development of new antiviral drugs.
Article
Full-text available
Recombinant adeno-associated virus (rAAV) is comprised of non-enveloped capsids that can package a therapeutic transgene and are currently being developed and utilized as gene therapy vectors. The therapeutic efficiency of rAAV is dependent on successful cytoplasmic trafficking and transgene delivery to the nucleus. It is hypothesized that an increased understanding of the effects of the cellular environment and biophysical properties of the capsid as it traffics to the nucleus could provide insight to improve vector efficiency. The AAV capsid is exposed to increasing [H⁺] during endo-lysosomal trafficking. Exposure to low pH facilitates the externalization of the viral protein 1 unique region (VP1u). This VP1u contains a phospholipase A2 domain required for endosomal escape and nuclear localization signals that facilitate nuclear targeting and entry. The viral genome is released either after total capsid disassembly or via a concerted DNA ejection mechanism in the nucleus. This study presents the characterization of genome ejection (GE) for two diverse serotypes, AAV2 and AAV5, using temperature. The temperature required to disassemble the virus capsid (TM) is significantly higher than the temperature required to expose the transgene (TE) for both serotypes. This was verified by quantitative PCR (qPCR) and transmission electron microscopy. Additionally, the absence of VP1/VP2 in the capsids and a decrease in pH increase the temperature of GE. Furthermore, cryo-electron microscopy structures of the AAV5 capsid pre- and post-GE reveal dynamics at the twofold, threefold, and fivefold regions of the capsid interior consistent with a concerted egress of the viral genome. IMPORTANCE The development of recombinant adeno-associated virus (rAAV) capsids has grown rapidly in recent years, with five of the eight established therapeutics gaining approval in the past 2 years alone. Clinical progression with AAV2 and AAV5 represents a growing need to further characterize the molecular biology of these viruses. The goal of AAV-based gene therapy is to treat monogenic disorders with a vector-delivered transgene to provide wild-type protein function. A better understanding of the dynamics and conditions enabling transgene release may improve therapeutic efficiency. In addition to their clinical importance, AAV2 and 5 were chosen in this study for their diverse antigenic and biophysical properties compared to more closely related serotypes. Characterization of a shared genome ejection process may imply a conserved mechanism for all rAAV therapies.
Article
Adeno-associated viruses (AAVs) are gaining traction as delivery vehicles for gene therapy although the molecular understanding of AAV-transgene release is still limited. Typically, the process of viral uncoating is investigated (in vitro) through thermal stress, revealing capsid disintegration at elevated temperatures. To assess the (in)stability of different empty and filled AAV preparations, we used the light-scattering-based interferometric microscopy technique of mass photometry that, on a single-particle basis, determines the molecular weight of AAVs. By introducing a heat-stable DNA plasmid as an internal standard, we quantitatively probed the impact of heat on AAVs. Generally, empty AAVs exhibited greater heat resistance than genome-filled particles. Our data also indicate that upon DNA release, the capsids do not transform into empty AAVs, but seem to aggregate or disintegrate. Strikingly, some AAVs exhibited an intermediate state with disrupted capsids but preserved bound genome, a feature that experimentally only emerged following incubation with a nuclease. Our data demonstrate that the thermal uncoating process is highly AAV specific (i.e., can be influenced by serotype, genome, host system). We argue that nuclease treatment in combination with MP can be used as an additional analytical tool for assessing structural integrity of recombinant and/or clinical AAV vectors.
Article
Full-text available
With a limited coding capacity of 4.7 kb, adeno-associated virus (AAV) genome has evolved over-lapping genes to maximise the usage of its genome. An example is the recently found ORF in the cap gene, encoding membrane-associated accessory protein (MAAP), located in the same genomic region as the VP1/2 unique domain, but in a different reading frame. This 13 KDa protein, unique to the dependovirus genus, is not homologous to any known protein. Our studies confirm that MAAP translation initiates from the first CTG codon found in the VP1 ORF2. We have further observed MAAP localised in the plasma membrane, in the membranous structures in close proximity to the nucleus and to the nuclear envelope by co-transfecting with plasmids encoding the wild-type AAV (wt-AAV) genome and adenovirus (Ad) helper genes. While keeping VP1/2 protein sequence identical, both inactivation and truncation of MAAP translation affected the emergence and intracellular distribution of the AAV capsid proteins. We have demonstrated that MAAP facilitates AAV replication and has a role in controlling Ad infection. Additionally, we were able to improve virus production and capsid integrity through a C-terminal truncation of MAAP while other modifications led to increased packaging of contaminating, non-viral DNA. Our results show that MAAP plays a significant role in AAV infection, with profound implications for the production of therapeutic AAV vectors.
Article
Full-text available
Adeno-associated viruses (AAV) rely on helper viruses to transition from latency to lytic infection. Some AAV serotypes are secreted in a pre-lytic manner as free or extracellular vesicle (EV)-associated particles, although mechanisms underlying such are unknown. Here, we discover that the membrane-associated accessory protein (MAAP), expressed from a frameshifted open reading frame in the AAV cap gene, is a novel viral egress factor. MAAP contains a highly conserved, cationic amphipathic domain critical for AAV secretion. Wild type or recombinant AAV with a mutated MAAP start site (MAAPΔ) show markedly attenuated secretion and correspondingly, increased intracellular retention. Trans-complementation with MAAP restored secretion of multiple AAV/MAAPΔ serotypes. Further, multiple processing and analytical methods corroborate that one plausible mechanism by which MAAP promotes viral egress is through AAV/EV association. In addition to characterizing a novel viral egress factor, we highlight a prospective engineering platform to modulate secretion of AAV vectors or other EV-associated cargo.
Article
Full-text available
Adeno-associated virus (AAV) is the leading vector in emerging treatments of inherited diseases. Higher transduction efficiencies and cellular specificity are required for broader clinical application, motivating investigations of virus–host molecular interactions during cell entry. High-throughput methods are identifying host proteins more comprehensively, with subsequent molecular studies revealing unanticipated complexity and serotype specificity. Cryogenic electron microscopy (cryo-EM) provides a path towards structural details of these sometimes heterogeneous virus–host complexes, and is poised to illuminate more fully the steps in entry. Here presented, is progress in understanding the distinct steps of glycan attachment, and receptor-mediated entry/trafficking. Comparison with structures of antibody complexes provides new insights on immune neutralization with implications for the design of improved gene therapy vectors.
Article
Full-text available
Cancer therapy urges targeting of malignant subsets within self-renewing heterogeneous stem cell populations. We dissect the genetic and functional heterogeneity of human glioblastoma stem cells (GSCs) within patients by their innate responses to non-pathogenic mouse parvoviruses that are tightly restrained by cellular physiology. GSC neurospheres accumulate assembled capsids but restrict viral NS1 cytotoxic protein expression by an innate PKR/eIF2α-P response counteractable by electric pulses. NS1 triggers a comprehensive DNA damage response involving cell-cycle arrest, neurosphere disorganization, and bystander disruption of GSC-derived brain tumor architecture in rodent models. GSCs and cancer cell lines permissive to parvovirus genome replication require p53-Ser15 phosphorylation (Pp53S15). NS1 expression is enhanced by exogeneous Pp53S15 induction but repressed by wtp53. Consistently, patient-specific GSC subpopulations harboring p53 gain-of-function mutants and/or Pp53S15 are selective viral targets. This study provides a molecular foundation for personalized biosafe viral therapies against devastating glioblastoma and other cancers with deregulated p53 signaling.
Article
Full-text available
HIV-1 can infect non-dividing cells. The nuclear envelope therefore represents a barrier that HIV-1 must traverse in order to gain access to the host cell chromatin for integration. Hence, nuclear entry is a critical step in the early stages of HIV-1 replication. Following membrane fusion, the viral capsid (CA) lattice, which forms the outer face of the retroviral core, makes numerous interactions with cellular proteins that orchestrate the progress of HIV-1 through the replication cycle. The ability of CA to interact with nuclear pore proteins and other host factors around the nuclear pore determines whether nuclear entry occurs. Uncoating, the process by which the CA lattice opens and/or disassembles, is another critical step that must occur prior to integration. Both early and delayed uncoating have detrimental effects on viral infectivity. How uncoating relates to nuclear entry is currently hotly debated. Recent technological advances have led to intense discussions about the timing, location, and requirements for uncoating and have prompted the field to consider alternative uncoating scenarios that presently focus on uncoating at the nuclear pore and within the nuclear compartment. This review describes recent advances in the study of HIV-1 nuclear entry, outlines the interactions of the retroviral CA protein, and discusses the challenges of investigating HIV-1 uncoating.
Article
Full-text available
Nuclear pore complexes (NPCs) are huge protein assemblies within the nuclear envelope (NE) that serve as selective gates for macromolecular transport between nucleus and cytoplasm. When higher eukaryotic cells prepare for division, they rapidly disintegrate NPCs during NE breakdown such that nuclear and cytoplasmic components mix to enable the formation of a cytoplasmic mitotic spindle. At the end of mitosis, reassembly of NPCs is coordinated with the establishment of the NE around decondensing chromatin. We review recent progress on mitotic NPC disassembly and reassembly, focusing on vertebrate cells. We highlight novel mechanistic insights into how NPCs are rapidly disintegrated into conveniently reusable building blocks, and put divergent models of (post-)mitotic NPC assembly into a spatial and temporal context.
Article
Full-text available
Human gene therapy has advanced from twentieth-century conception to twenty-first-century reality. The recombinant Adeno-Associated Virus (rAAV) is a major gene therapy vector. Research continues to improve rAAV safety and efficacy using a variety of AAV capsid modification strategies. Significant factors influencing rAAV transduction efficiency include neutralizing antibodies, attachment factor interactions and receptor binding. Advances in understanding the molecular interactions during rAAV cell entry combined with improved capsid modulation strategies will help guide the design and engineering of safer and more efficient rAAV gene therapy vectors.
Article
Full-text available
The exploration into the strategies for the prevention and treatment of cancer is far from complete. Apart from humans, cancer has gained considerable importance in animals because of increased awareness towards animal health and welfare. Current cancer treatment regimens are less specific towards tumor cells and end up harming normal healthy cells. Thus, a highly specific therapeutic strategy with minimal side effects is the need of the hour. Oncolytic viral gene therapy is one such specific approach to target cancer cells without affecting the normal cells of the body. Canine parvovirus (CPV) is an oncolytic virus that specifically targets and kills cancer cells by causing DNA damage, caspase activation, and mitochondrial damage. Non-structural gene 1 (NS1) of CPV, involved in viral DNA replication is a key mediator of cytotoxicity of CPV and can selectively cause tumor cell lysis. In this review, we discuss the oncolytic properties of Canine Parvovirus (CPV or CPV2), the structure of the NS1 protein, the mechanism of oncolytic action as well as role in inducing an antitumor immune response in different tumor models.
Article
Full-text available
The glycosphingolipid (GSL) globoside (Gb4) is essential for parvovirus B19 (B19V) infection. Historically considered the cellular receptor of B19V, the role of Gb4 and its interaction with B19V are controversial. In this study, we applied artificial viral particles, genetically modified cells, and specific competitors to address the interplay between the virus and the GSL. Our findings demonstrate that Gb4 is not involved in the binding or internalization process of the virus into permissive erythroid cells, a function that corresponds to the VP1u cognate receptor. However, Gb4 is essential at a post-internalization step before the delivery of the single-stranded viral DNA into the nucleus. In susceptible erythroid Gb4 knockout cells, incoming viruses were arrested in the endosomal compartment, showing no cytoplasmic spreading of capsids as observed in Gb4-expressing cells. Hemagglutination and binding assays revealed that pH acts as a switch to modulate the affinity between the virus and the GSL. Capsids interact with Gb4 exclusively under acidic conditions and dissociate at neutral pH. Inducing a specific Gb4-mediated attachment to permissive erythroid cells by acidification of the extracellular environment led to a non-infectious uptake of the virus, indicating that low pH-mediated binding to the GSL initiates active membrane processes resulting in vesicle formation. In summary, this study provides mechanistic insight into the interaction of B19V with Gb4. The strict pH-dependent binding to the ubiquitously expressed GSL prevents the redirection of the virus to nonpermissive tissues while promoting the interaction in acidic intracellular compartments as an essential step in infectious endocytic trafficking.
Article
The nuclear-cytoplasmic transport of biomolecules is assisted by the nuclear pores composed of evolutionarily conserved proteins termed nucleoporins (Nups). The central Nups, characterized by multiple FG-repeats, are highly dynamic and contain a high level of intrinsically disordered regions (IDPRs). FG-Nups bind several protein partners and play critical roles in molecular interactions and the regulation of cellular functions through their IDPRs. In the present study, we performed a multiparametric bioinformatics analysis to characterize the prevalence and functionality of IDPRs in human FG-Nups. These analyses revealed that the sequence of all FG-Nups contained >50% IDPRs (except Nup54 and Nup358). Nup98, Nup153, and POM121 were extremely disordered with ~80% IDPRs. The functional disorder-based binding regions in the FG-Nups were identified. The phase separation behavior of FG-Nups indicated that all FG-Nups have the potential to undergo liquid-to-liquid phase separation that could stabilize their liquid state. The inherent structural flexibility in FG-Nups is mechanistically and functionally advantageous. Since certain FG-Nups interact with disease-relevant protein aggregates, their complexes can be exploited for drug design. Furthermore, consideration of the FG-Nups from the intrinsic disorder perspective provides critical information that can guide future experimental studies to uncover novel pathways associated with diseases linked with protein misfolding and aggregation.