Download Viral and cellular subnuclear structures in human cytomegalovirus

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

SR protein wikipedia , lookup

Protein moonlighting wikipedia , lookup

Endomembrane system wikipedia , lookup

Signal transduction wikipedia , lookup

Amitosis wikipedia , lookup

Interferon wikipedia , lookup

List of types of proteins wikipedia , lookup

Cell nucleus wikipedia , lookup

JADE1 wikipedia , lookup

Transcript
Journal of General Virology (2015), 96, 239–253
Review
DOI 10.1099/vir.0.071084-0
Viral and cellular subnuclear structures in human
cytomegalovirus-infected cells
Blair L. Strang
Correspondence
Institute for Infection & Immunity, St George’s, University of London, London, UK
Blair L. Strang
[email protected]
Received 15 August 2014
Accepted 28 October 2014
In human cytomegalovirus (HCMV)-infected cells, a dramatic remodelling of the nuclear
architecture is linked to the creation, utilization and manipulation of subnuclear structures. This
review outlines the involvement of several viral and cellular subnuclear structures in areas of
HCMV replication and virus–host interaction that include viral transcription, viral DNA synthesis
and the production of DNA-filled viral capsids. The structures discussed include those that
promote or impede HCMV replication (such as viral replication compartments and promyelocytic
leukaemia nuclear bodies, respectively) and those whose role in the infected cell is unclear (for
example, nucleoli and nuclear speckles). Viral and cellular proteins associated with subnuclear
structures are also discussed. The data reviewed here highlight advances in our understanding of
HCMV biology and emphasize the complexity of HCMV replication and virus–host interactions in
the nucleus.
Introduction
The betaherpesvirus human cytomegalovirus (HCMV) is a
widespread opportunistic pathogen that severely affects
immunocompromised and immunodeficient populations
(Mocarski et al., 2007). Like all herpesviruses, HCMV
undergoes either productive or latent infection. During
productive infection, infectious virus is produced, while in
latent infection the virus becomes largely transcriptionally
quiescent (Mocarski et al., 2007). Like other herpesviruses, many events that are critical for productive
HCMV replication take place within the nucleus. These
include essential steps in viral replication, such as: the
transcriptional cascade of immediate-early (IE), early and
late viral RNA transcripts, synthesis of viral DNA and the
production of DNA-containing capsids (Mocarski et al.,
2007). Compared with the prototype human herpesvirus,
the alphaherpesvirus herpes simplex virus (HSV), certain
features of productive HCMV infection are not well
characterized. However, it is clear that several subnuclear
structures are involved in HCMV genome replication and
virus–host interactions. Uncovering the roles of these
structures has led to significant advances in our understanding of HCMV biology. This review summarizes the
involvement of several viral and cellular subnuclear
structures in productive HCMV infection. The participation of each structure in HCMV replication and virus–host
interactions is described from entry of the viral genome
into the nucleus to the egress of viral capsids through the
nuclear membrane. The data discussed highlight recent
advances in our understanding of subnuclear structure
function, introduce viral and cellular factors associated
with subnuclear structures and indicate what questions
have yet to be answered.
071084 G 2015 The Author
Entry of the HCMV genome into the nucleus: the
nuclear pore complex, nuclear speckles,
promyelocytic leukaemia protein nuclear bodies
and pp150 bodies
HCMV genome entry into the nucleus is poorly defined
but may be similar to movement of the HSV DNA
genome through the nuclear pore complex (NPC)
(Kobiler et al., 2012) (Fig. 1a). HCMV capsid and
tegument proteins (potentially HCMV UL48 and UL77,
homologues of HSV UL36 and UL25, respectively)
probably interact with proteins on the cytoplasmic face
of the NPC (potentially including Nup214, Nup358 and
hCG1) (Table 1). This leads to opening of the capsid. The
pressure of viral DNA on the capsid wall leads to release of
a linear DNA form of the HCMV genome. Viral DNA
travels out of the capsid via a portal on a unique vertex of
the capsid comprised of UL104 (HSV UL6 homologue)
(Kobiler et al., 2012). The HCMV genome moves through
the central channel of the NPC into the nucleus (Kobiler
et al., 2012) (Fig. 1a).
The linear HCMV DNA genome circularizes upon entry
into the nucleus via fusion of the genomic termini (McVoy
& Adler, 1994) (Fig. 1b). This could be mediated by either
homologous or non-homologous end-joining DNA repair
mechanisms. The circular form of the genome is utilized as
a template for viral RNA transcription and DNA synthesis.
Very early in infection, viral genomes can be found at the
periphery of the nucleus where transcription from the viral
genome takes place (Ishov et al., 1997). At the periphery of
the nucleus, the major viral IE transcriptional transactivator IE2 co-localizes with cellular RNA polymerase II
transcription factors TATA-binding protein (TBP) and
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Printed in Great Britain
239
B. L. Strang
Capsid
(a)
Portal
Nuclear
membrane
Nuclear pore complex
Nuclear
lamina
(b)
PML
IE2
SC35
?
TFIIB
pp150
TBP
Fig. 1. Entry of the HCMV genome through the nuclear pore
complex and association with viral and cellular factors at the
periphery of the nucleus. (a) The viral capsid (shown in yellow)
docks at the nuclear pore complex. The linear form of the viral DNA
genome exits the capsid through the portal on a unique vertex of
the capsid (shown in grey). (b) The linear form of the genome
circularizes and associates with the viral transcriptional transactivator IE2 (shown in pink), promyelocytic leukaemia protein (PML)containing nuclear bodies (shown in purple) and SC35-containing
bodies thought to be nuclear speckles (shown in yellow) or,
potentially, pp150-containing bodies (shown in orange). The
cellular transcription factors transcription factor IIB (TFIIB) (shown
in blue) and TATA-binding protein (TBP) (shown in green) also
associate with the viral genome.
transcription factor IIB (TFIIB) (Ishov et al., 1997)
(Fig. 1b). It is possible that, like HSV RNA transcription,
HCMV RNA transcription involves the nuclear lamina.
The nuclear lamina is a dense fibrillar protein network at
the nuclear membrane composed of lamin proteins and
lamin-associated proteins (Fig. 1a). In HSV-infected cells,
the lamina component lamin A/C is important for genome
targeting to the lamina, and at the lamina hetero- or
euchromatin on viral promoters modulates HSV RNA
transcription (Silva et al., 2008).
The majority of HCMV genomes entering the nucleus
localize with promyelocytic leukaemia protein nuclear
bodies (PML-NBs, also referred to as ND10 bodies)
and SC35-containing structures within intrachromosomal space. IE2 bridges PML-NBs and SC35-containing
structures (Ishov et al., 1997) (Fig. 1b). As transcription
progresses, viral IE RNA transcripts co-localize with SC35containing bodies but not PML-NBs (Ishov et al., 1997).
SC35 is a major protein component of nuclear speckles,
which are dynamic nuclear structures composed of RNA
and protein and enriched in pre-mRNA splicing factors
(Lamond & Spector, 2003). A role for nuclear speckles in
240
HCMV replication, or that of any other human virus, is
not often commented on. It is likely that nuclear speckles
are involved in HCMV IE gene transcription, but it is
unknown what transcriptional events they might facilitate.
In HSV-infected cells, it has been reported that nuclear
speckles are involved in export of viral RNA transcripts
(Chang et al., 2011). Thus, nuclear speckles may serve in
the processing and export of HCMV RNA transcripts early
in infection. These RNAs probably include the viral RNAs
transcribed from the UL122–123 locus of the HCMV
genome that are multiply spliced to produce mRNA
transcripts encoding the IE proteins IE1 and IE2.
The role of PML-NBs in HCMV replication is one of
the most studied and debated facets of HCMV biology.
The complex relationship between PML-NBs and HCMV
cannot be fully summarized here but is discussed in detail
elsewhere (Everett et al., 2013; Tavalai & Stamminger, 2009,
2011). PML-NBs are dynamic structures that comprise
at least 70 constitutively or transiently present proteins,
including PML protein (which acts a scaffold for PML-NB
formation) (Ishov et al., 1999). The majority of, if not all,
PML-NB proteins are post-translationally modified with the
addition of small ubiquitin-related modifier (SUMO)
moieties. PML-NBs contain several proteins required for
protein SUMOylation and the relationship between protein
function and post-translational modification by SUMO is a
rapidly developing topic in HCMV biology and elsewhere
(Everett et al., 2013).
PML-NBs are found throughout the nucleus. Upon entry
of the viral genome into the nucleus, PML-NBs form on
SUMO-modified PML proteins juxtaposed to incoming
viral genomes. PML-NBs act to repress viral transcription
via the co-operative function of the PML-NB proteins
PML, Sp100 and hDaxx (Glass & Everett, 2013). PML
protein is one of the best characterized PML-NB proteins
involved in HCMV transcriptional repression and acts to
facilitate assembly of PML-NBs containing transcriptionally repressive proteins at HCMV genomes (Tavalai &
Stamminger, 2009, 2011). Sp100 promotes transcriptional
repression through its interaction with the transcriptional
repressor heterochromatin protein (HP1) (Seeler et al.,
1998). SUMO modification of Sp100 promotes interaction
with HP1 (Seeler et al., 2001) but is not required for
localization to PML-NBs (Sternsdorf et al., 1999). hDaxx
is recruited to PML-NBs via an interaction with
SUMOylated PML (Everett et al., 2013) and acts as a
transcriptional co-repressor via its interaction with the
chromatin remodeller ATRX (Ishov et al., 2004; Xue et al.,
2003). PML-NBs contain other transcriptionally repressive
cellular proteins (Tavalai & Stamminger, 2011), and it is
possible other PML-NB proteins may antagonize HCMV
replication.
Crucially, HCMV encodes antagonists of PML-NB
function (Table 1) whose actions lead to removal of
proteins from PML-NBs and the dispersal of PML-NBs
(Everett et al., 2013; Tavalai & Stamminger, 2009, 2011).
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Journal of General Virology 96
http://vir.sgmjournals.org
Table 1. HCMV proteins associated with subnuclear structures
Nuclear
structure
HCMV
associated
proteins
Kinetic class of
HCMV protein*
HCMV protein
functionD
Requirement for Known or proposed
HCMV replicationd cellular partner(s)
References
Nuclear pore
UL48
UL77
L
E
Capsid transport
Portal capping protein
E
E
Nup214, Nup358 hCG1 Kobiler et al. (2012)
Nup214, Nup358 hCG1 Kobiler et al. (2012)
PML-NB
TLR9
UL29
UL30
UL35
UL69
L
L
Unknown
E
E–L
Unknown
A
A
NE
A
Unknown
Unknown
Unknown
Unknown
SUMO
Salsman
Salsman
Salsman
Salsman
Salsman
UL82 (pp71)
L
A
hDaxx, ATRX
Kalejta & Shenk (2003), Lukashchuk et al. (2008)
UL76
UL80a
UL97
UL98
UL122–123 (IE1)
Unknown
L
L
E–L
IE, L
A
E
A
E
E
Unknown
Unknown
Rb
Unknown
PML Sp100
Salsman et al. (2008)
Salsman et al. (2008)
Prichard et al. (2008)
Salsman et al. (2008)
Tavalai et al. (2011)
UL122–123 (IE2)
IE, L
Unknown
Temperance factor
Unknown
Unknown
Multiple regulatory
functions
Transcriptional
transactivator
Unknown
Capsid protein
Viral kinase
Alkaline nuclease
Transcriptional
transactivator
Transcriptional
transactivator
E
US25
US32
E–L
L
Unknown
Unknown
NE
NE
Sp100
SC35
PML
SUMO
Unknown
Tavalai et al. (2011)
Ishov et al. (1997)
Ishov et al. (1997)
Salsman et al. (2008)
Salsman et al. (2008)
Nuclear speckles UL122–123 (IE2)
IE, L
Transcriptional
transactivator
E
SC35
Ishov et al. (1997)
pp150 bodies
L
Cell-cycle regulator
E
A2-CDK
Bogdanow et al. (2013)
IE, L
Transcriptional
transactivator
Transcriptional
transactivator
DNA polymerase
subunit
ssDNA-binding protein
E
Unknown
Ahn et al. (1999), Penfold & Mocarski (1997)
A
Unknown
Ahn et al. (1999)
E
Nucleolin
Ahn et al. (1999), Penfold & Mocarski (1997), Strang et al. (2012b)
E
Unknown
UL32 (pp150)
Viral replication UL122–123 (IE2)
compartments
UL112–
UL44
E–L
UL57
E
al.
al.
al.
al.
al.
(2008)
(2008)
(2008)
(2008)
(2008)
UL84
E–L
DNA replication factor
E
hnRNP K
Penfold & Mocarski (1997)
Strang et al. (2012b)
Kagele et al. (2012), Gao et al. (2008)
TLR5
TRL9
UL29
E
L
L
Unknown
Unknown
Temperance factor
Unknown
Unknown
A
Unknown
Unknown
Unknown
Salsman et al. (2008)
Salsman et al. (2008)
Salsman et al. (2008)
241
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
HCMV and subnuclear structures
Nucleoli
E, E–L
et
et
et
et
et
242
Sharma et al. (2014)
Sharma et al. (2014)
Unknown
Lamin A/C
*From Chambers et al. (1999). E, Early; IE, immediate early; L, late; E–L, early to late.
DProtein functions described in references indicated in the table and/or Fields Virology (Mocarski et al., 2007).
dFrom Yu et al. (2003). E, Essential; NE, non-essential; A, augments replication.
Sharma et al. (2014)
Unknown
E
A
Nuclear egress complex
Viral kinase
Nuclear egress complex
UL53
UL97
Lamina
Unknown
UL50
Nuclear
L
E–L
E
Salsman et al. (2008)
Unknown
UL30
Unknown
Unknown
A
Salsman et al. (2008)
Salsman et al. (2008)
Arcangeletti et al. (2009)
Bender et al. (2014)
Unknown
Unknown
Unknown
Unknown
NE
A
NE
E
Unknown
Unknown
Interferon antagonist
Replication factor
L
Unknown
L
E
UL31
UL76
UL83 (pp65)
UL84
Cajal bodies
Nuclear
structure
Table 1. cont.
HCMV
associated
proteins
Kinetic class of
HCMV protein*
HCMV protein
functionD
Requirement for Known or proposed
HCMV replicationd cellular partner(s)
References
B. L. Strang
In HCMV-infected cells, two major antagonists of PMLNB function are pp71 and IE1. pp71 degrades hDaxx by a
proteasome-dependent, ubiquitin-independent mechanism (Kalejta & Shenk, 2003) and displaces ATRX from
PML-NBs (Lukashchuk et al., 2008). The presence of IE1
promotes the disruption of PML-NBs, induces the
proteasome-independent loss of SUMOylated PML and
Sp100 proteins (Ahn & Hayward, 1997; Kang et al., 2006;
Kim et al., 2011; Korioth et al., 1996; Lee et al., 2004;
Tavalai et al., 2011; Wilkinson et al., 1998; Xu et al., 2001)
and potentially induces the proteasome-dependent
degradation of unSUMOylated Sp100 late in infection
(Tavalai et al., 2011). Importantly, IE1 does not act like
its well-studied counterpart expressed by HSV, ICP0.
ICP0 possesses an E3 ubiquitin ligase activity directly
involved in the proteasome-dependent degradation of
PML protein (Boutell & Everett, 2013). IE1 has no known
ubiquitin ligase or SUMOylase activity (Kang et al.,
2006), but is able to exert an effect that leads to PML-NB
disruption, loss of SUMOylated PML and an increase in
the abundance of unmodified PML. This would suggest
that IE1 can recruit a cellular protein to deSUMOylate
PML within PML-NBs or SUMO is removed from PML
by a cellular deSUMOylase after PML-NBs have been
dispersed in the presence of IE1. The role of IE1 in Sp100
degradation is unclear. IE1 binding to Sp100 (Kim et al.,
2011) may result in Sp100 degradation by an as-yetunknown mechanism. Alternatively, Sp100 degradation may be an indirect result of PML-NB modification
by IE1.
Other HCMV proteins may be involved in PML-NB
destruction or modification (Table 1). Expression of
epitope-tagged versions of US25, UL29 UL30, UL69,
UL76, UL98 or TLR9 is sufficient to modestly reduce the
number of PML-NBs in uninfected cells (Salsman et al.,
2008). These proteins are expressed in different kinetic
classes of viral transcription and have different effects on
HCMV replication, plus the function of several of these
proteins is unknown (Table 1). In transfection experiments epitope-tagged HCMV proteins US32, UL80a and
UL35 co-localize with, but do not obviously degrade,
PML-NBs in uninfected cells (Salsman et al., 2008).
Epitope-tagged US32 and UL35 alter the shape and size of
PML-NBs in uninfected cells. The localization and
function of these proteins in relation to PML-NBs in
HCMV-infected cells has yet to be elucidated. UL80a is a
capsid protein. UL80a association with PML-NBs is
reminiscent of PML cages trapping alphaherpesvirus
varicella-zoster virus (VZV) capsids in infected cells via
physical interaction between PML-NB proteins and VZV
capsids (Reichelt et al., 2011). This mechanism of cellular
defence to infection has not yet been observed in HCMVinfected cells. An epitope-tagged version of the putative
HCMV protein UL3 has been reported to alter PML-NBs
in uninfected cells (Salsman et al., 2008). However, it is
now thought that the HCMV genome does not contain an
ORF encoding UL3 (Gatherer et al., 2011). A final protein
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Journal of General Virology 96
HCMV and subnuclear structures
to consider in the alteration of PML-NBs in HCMVinfected cells is the HCMV viral kinase UL97. UL97
phosphorylates and inactivates the PML-NB protein
retinoblastoma (Rb) (Hume et al., 2008) and expression
of epitope-tagged UL97 in uninfected simian cells
(Prichard et al., 2008), but not uninfected human cells
(Kuny et al., 2010), notably reduces the number of PMLNBs in the cell. Clarification of the roles, if any, of the
UL97 and Rb proteins in PML-NB function in HCMVinfected cells is required.
Future consideration of PML-NB function in HCMVinfected cells will benefit from further exploration of the
interaction of PML-NBs with other viruses, including other
herpesviruses. Indeed, most detailed information concerning the interaction of PML-NBs with viral DNA genomes
comes from the study of HSV infection and analysis of
PML-NB antagonism by the HSV protein ICP0 (Boutell
& Everett, 2013). Interaction of PML-NBs with adenoand papillomaviruses has been reported but is less well
understood. Prominent findings include the ability of the
adenovirus proteins E1B and E4orf3 to antagonize PML
and hDaxx functions (Schreiner et al., 2010; Ullman &
Hearing, 2008) and the repression of papillomavirus RNA
transcription by Sp100 (Stepp et al., 2013). Future studies
of adeno- and papillomavirus replication may illuminate important features of herpesvirus interactions with
PML-NBs.
Subnuclear structures other than PML-NBs could also be
involved in repression of viral transcription. These include
bodies such as the HCMV tegument protein pp150 (UL32).
pp150 is a substrate of cyclin A2-CDK (Table 1) and is a
sensor for cell-cycle progression during HCMV infection.
By an unknown mechanism, pp150 is involved in blocking
viral transcription when levels of cyclin A2-CDK are high
(Bogdanow et al., 2013). It has been suggested that pp150
nuclear bodies are sites of pp150-mediated transcriptional
repression and are the destination of HCMV genomes not
associated with PML-NBs (Bogdanow et al., 2013; Ishov
et al., 1997) (Fig. 1b).
Therefore, upon engagement of the viral capsid with
proteins on the cytoplasmic face of the NPC, the HCMV
genome is delivered into the nucleus through the nuclear
pore. Once in the nucleus, transcription from the viral
genome involves cellular transcription factors and may be
facilitated by interaction of the genome with the nuclear
lamina at the periphery of the nucleus. At the periphery of
the nucleus, the genome encounters nuclear speckles,
which probably facilitate the splicing and export to the
cytoplasm of viral RNA transcripts, and PML-NBs or
pp150 bodies that restrict viral transcription. Restriction of
viral transcription by PML-NBs involves the recruitment of
transcriptional repressors to the viral genome and is
antagonized by virally encoded proteins. Restriction of
viral transcription by pp150 bodies is less well defined and
is associated with progression of the infected cell through
the cell cycle.
http://vir.sgmjournals.org
Replication of the viral genome: HCMV replication
compartments, chromatin partitioning, protein
quality-control compartments and nuclear
speckles
Once transcription of IE and E RNA transcripts is under
way, viral proteins that replicate HCMV genomic DNA are
produced. Viral DNA synthesis and several early and late
transcriptional events occur within non-membrane-bound
compartments created by the virus composed of protein
and nucleic acids. The formation of these ‘viral replication compartments’ is poorly understood but allows the
recruitment and concentration of factors required for viral
replication to specific areas of the infected cell nucleus.
Analysis of alphaherpesvirus infection implies that a
herpesvirus replication compartment can develop from a
single incoming viral genome (Kobiler et al., 2011). In
HCMV-infected cells, replication compartments form in
the vicinity of PML-NBs (Ahn et al., 1999). As compartments develop, they contain viral transcriptional transactivators (including IE2 and UL112–113 proteins), viral
proteins involved in viral DNA synthesis (including UL44
and UL57) and viral DNA (Ahn et al., 1999; Penfold &
Mocarski, 1997; Strang et al., 2012b) (Table 1). Early in the
formation of replication compartments, factors involved in
the DNA damage response, including cH2AX and ATM,
localize to replication compartments (E et al., 2011) and
may recognize viral genomes within compartments. It is
thought that viral transcriptional transactivators drive
E2F1-mediated activation of ATM signalling, which
activates DNA damage response factors such as cH2AX
and p53 (E et al., 2011). Activation of this pathway may be
required for modulation of cell-cycle-related functions that
promote viral replication. ATM signalling promotes the
development of replication compartments (E et al., 2011),
but it is unclear if this is an effect that directly relates to
replication compartment development or an indirect effect
related to events that promote viral RNA transcription or
viral DNA synthesis.
During HCMV replication, several compartments can form
and then coalesce to form a single compartment that
occupies nearly all the nucleus but does not encroach
into nucleoli (Penfold & Mocarski, 1997; Strang et al.,
2012a, b) (Fig. 2a, b). The mechanisms driving coalescence
of HCMV replication compartments have yet to be
defined. Replication compartment coalescence in HSVinfected cells occurs via non-random movement and is
dependent upon nuclear actin, myosin and viral transcription (Chang et al., 2011).
To provide areas in the nucleus for replication compartments to develop, the compaction and movement of
chromatin is observed (O’Dowd et al., 2012; Strang et al.,
2012b). This process is termed ‘chromatin partitioning’. It
is unknown what drives this process, but UL98 and UL76
may be involved (Table 1). UL98 is an alkaline nuclease
capable of digesting DNA (Sheaffer et al., 1997). UL76 is
member of the UL24 gene family, all of which contain a
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
243
B. L. Strang
conserved putative PD-(D/E)XK endonuclease motif
(Knizewski et al., 2006). Although endonuclease activity
of UL76 has yet to be demonstrated, indirect evidence
points to a role for UL76 in the chromosome breakage
required for chromatin partitioning. For example, the
presence of UL76 in uninfected cells is sufficient to
stimulate chromosomal aberrations (Siew et al., 2009),
and in HCMV-infected cells, UL76 is involved in
stimulating a DNA damage response (Costa et al., 2013).
Several facets of HCMV replication compartment organization and function late in infection have recently been
described, each involving the localization of HCMV
protein UL44. UL44 is the putative processivity subunit
of the HCMV DNA polymerase (Ertl & Powell, 1992), and
several different phosphorylated forms of UL44 are present
in the infected cell (Silva et al., 2011). At least one form of
UL44 accumulates at the periphery of replication compartments throughout HCMV replication, and UL44 is also
found at considerably lower levels within certain domains
inside replication compartments, plus in the nucleus
outside replication compartments and also in the cytoplasm (Hamirally et al., 2009; Penfold & Mocarski, 1997;
Strang et al., 2012b). Viral DNA synthesis occurs at foci
within the layer of UL44 at the periphery of replication
compartments (Fig. 2b), and newly synthesized viral DNA
moves into the interior of replication compartments
(Strang et al., 2012b).
The viral ssDNA-binding protein UL57 co-localizes with
UL44 at the periphery of replication compartments and is
found throughout the interior of compartments (Penfold
& Mocarski, 1997; Strang et al., 2012b). UL57 at the
periphery of replication compartments is likely to be
directly involved in viral DNA synthesis. UL57 protein in
the interior of replication compartments is associated with
viral DNA (Penfold & Mocarski, 1997) and is possibly
involved in UL57-mediated DNA strand exchange events,
including recombination and repair of newly synthesized
HCMV DNA.
As well as UL44 and UL57, other factors required for viral
DNA synthesis have been reported. These include the viral
DNA polymerase subunit UL54, replication factor UL84
and the helicase DNA complex (UL70, UL102 and UL105)
(Pari & Anders, 1993; Pari et al., 1993). However, the full
repertoire of viral and cellular proteins acting in concert
during DNA synthesis has yet to be described. These
proteins probably include those viral and cellular proteins
that associate with UL44 and/or UL84 (Gao et al., 2008;
Strang et al., 2009, 2010a, b), several proteins that have illdefined roles in viral DNA synthesis (for example, UL36,
IRS1 and IE2; Pari & Anders, 1993; Pari et al., 1993; Sarisky
& Hayward, 1996), plus viral and cellular proteins involved
in DNA damage responses and viral DNA repair (E et al.,
2011; O’Dowd et al., 2012; Strang & Coen, 2010). How
these and other proteins are organized within HCMV
replication compartments is unclear but may involve
UL44. UL44, a homodimer, is a structural homologue of
244
(a)
Coalescence
DNA replication
(c)
Cajal body
(b)
Replication
compartment
Nucleoli
Perinucleolar
compartment
UPS compartment
SC35
Fig. 2. Viral replication compartments, nuclear speckles, nucleoli
and nucleoli-associated subnuclear structures. (a) Viral replication
compartments form and coalesce to form a single viral replication
compartment that dominates nearly the entire nuclear space.
Throughout these processes, the viral protein UL44 (shown in red)
accumulates at the periphery of replication compartments. (b) Viral
DNA replication occurs at foci (shown in yellow) distributed
throughout the layer of UL44 at the periphery of replication
compartments. Ubiquitin–proteasome system (UPS) compartments (shown in blue) are found within replication compartments.
SC35-containing structures thought to be nuclear speckles
(shown in yellow) are found on the exterior of replication
compartments. (c) Viral replication compartments do not encroach
into nucleoli. In HCMV-infected cells, nucleolin (shown in green)
accumulates at the periphery of nucleoli. Cajal bodies (shown in
pink) and perinucleolar compartments (shown in orange) may be
involved in HCMV replication and virus–host interactions.
the cellular proliferating cell nuclear antigen (PCNA),
which exists as a trimer (Appleton et al., 2004, 2006;
Loregian et al., 2004a, b). PCNA binds multiple proteins of
multiple functions at the DNA replication fork (Moldovan
et al., 2007). UL44 could play a similar role and bind
multiple proteins in replication compartments. Proteomic
studies have indicated that UL44 associates with viral and
cellular proteins found in replication compartments,
including UL84 (Strang et al., 2010a). The binding of
proteins to UL44 is increasingly well characterized. Studies
investigating the binding of UL54 to UL44 have revealed
that, like PCNA, UL44 is able to bind proteins within
‘hydrophobic pockets’; crevices located in the N termini of
UL44 containing several hydrophobic amino acids into
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Journal of General Virology 96
HCMV and subnuclear structures
which proteins binding UL44 insert their binding domains
(Appleton et al., 2004, 2006; Loregian et al., 2004a, b).
Unexpectedly, UL44 can also associates with proteins
outside the hydrophobic pockets, as it has been demonstrated that the association of UL84 with UL44 is not
dependent upon binding in the hydrophobic pocket
(Strang et al., 2009). Rather, association of UL84 with
UL44 takes place elsewhere in the N-terminal domain
of UL44 and is not dependent upon homodimerization of
UL44 (Strang et al., 2009). It is unclear what drives
selection of proteins for binding to UL44, but there are
likely determinants of protein binding in both UL44 and its
interacting proteins. Indeed, it has been demonstrated that
the HCMV proteins IRS1 and TRS1, which have identical
N termini that are required for association with UL44,
compete for UL44 binding (Strang et al., 2010b).
UL44 is also indirectly involved in the recruitment of viral
proteins to replication compartments. The viral uracil
DNA glycosylase UL114 is required for efficient viral DNA
synthesis and base excision repair (Prichard et al., 1996).
Base excision repair maintains genome integrity by
removing uracil from the HCMV genome, which is the
result of either misincorporation of uracil during viral
DNA synthesis or the spontaneous deamination of cytosine
in the viral genome. Although UL114 localizes with UL44
within replication compartments (Prichard et al., 2005), it
does not bind UL44, as first thought (Ranneberg-Nilsen
et al., 2008). Rather, UL114 associates with the catalytic
subunit of the viral DNA polymerase UL54, and UL114,
UL54 and UL44 form a protein complex (Strang & Coen,
2010). Thus, UL44 indirectly recruits UL114 to replication
compartments via the interaction of UL114 with UL54.
There are few reports of cellular proteins localizing to
HCMV replication compartments, although it is known
that both p53 and replication protein A are recruited to the
interior of replication compartments (Fortunato & Spector,
1998). Their roles within replication compartments are
unknown but may involve modulation of cellular responses
to infection and promoting efficient viral DNA synthesis.
Several factors influence the architecture of HCMV
replication compartments. While the N terminus of UL44
is required for binding of UL44-associated proteins, the C
terminus of UL44 is required for the development of
replication compartments and correct localization of UL44
in the nucleus (Silva et al., 2010). Although the C terminus
of UL44 can be extensively phosphorylated by both the
viral kinase UL97 and the cellular kinase cdk1, it is unlikely
that these post-translational modifications affect replication compartment function, as mutational analysis of
phosphorylated residues in the UL44 C terminus has little
effect on viral DNA synthesis and viral replication (Silva
et al., 2011). Efficient viral DNA synthesis is also required
to maintain UL44 localization and compartment structure
(E et al., 2011; Penfold & Mocarski, 1997; Strang et al.,
2012a, b). Unexpectedly, the multifunctional cellular
nucleolar protein nucleolin (Ginisty et al., 1999) is involved
http://vir.sgmjournals.org
in maintaining replication compartment architecture.
UL44 and nucleolin associate via the N terminus of UL44
(Strang et al., 2010a) and in infected cells nucleolin colocalizes with UL44, but not viral DNA, on the exterior
of replication compartments (Strang et al., 2010a, 2012a).
The absence of nucleolin in the infected cell causes
mislocalization of UL44 at the periphery of replication
compartments and compromises efficient viral DNA
synthesis (Strang et al., 2012a). Nucleolin is also required
for UL84 localization with UL44 at the periphery of
replication compartments, although in the absence of
nucleolin, UL84 does not redistribute throughout the
nucleus but relocates to the cytoplasm (Bender et al.,
2014). Thus, nucleolin is required for both proper nuclear
and subnuclear localization of HCMV replication compartment proteins. Furthermore, it is possible that, in
infected cells, nucleolin, UL44, UL84 and either IRS1 or
TRS1 interact within a large multiprotein complex (Strang
et al., 2010a, b). Therefore, nucleolin may have an effect on
the localization of both IRS1 and TRS1 in the infected cell.
Although the organization of factors required for viral DNA
synthesis in replication compartments is increasingly well
understood, perhaps the most important unanswered
questions involving the architecture of replication compartments involve the localization of factors required for RNA
transcription from the viral genome. The location of
transcriptionally active genomes within replication compartments is unknown. There is little information on the
localization of viral and cellular proteins required for viral
RNA transcription. In infected cells, both phosphorylated
(transcriptionally active) and unphosphorylated (transcriptionally inactive) forms of RNA polymerase II concentrate
within nuclear structures thought to be replication compartments (Feichtinger et al., 2011). Within these structures, unphosphorylated and phosphorylated forms of
RNA polymerase II localize with the viral protein UL69
(Feichtinger et al., 2011), which is known to be required for
viral RNA processing and export to the cytoplasm. The
localization of RNA polymerase II in relation to UL44 has
yet to be described; however, viral proteins involved in
transcription and RNA metabolism, for example IE1, IE2
and UL84 (Bender et al., 2014; Silva et al., 2010), are found
throughout the nucleus and co-localize with UL44 in viral
replication compartments. Therefore, it is likely that viral
transcription occurs throughout the interior of replication
compartments and, unlike viral DNA synthesis, is not
restricted to specific areas of replication compartments such
as the layer of UL44 that accumulates at the periphery of
replication compartments.
The presence of subcompartments within HCMV replication compartments has been reported. Components of
the ubiquitin–proteasome system (UPS) assemble into
domains at the periphery of replication compartments
(Fig. 2b), where they co-localize with UL57 (Tran et al.,
2010). The accumulation of UPS proteins at the periphery
of HCMV replication compartments is analogous to the
formation of VICE (virus-induced chaperone enriched)
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
245
B. L. Strang
domains at the periphery of HSV replication compartments. VICE domains degrade misfolded proteins in a
proteasome-dependent fashion to promote efficient virus
replication (Livingston et al., 2009). Thus, compartments
containing UPS components at the periphery of HCMV
replication compartments may act as quality-control areas
for HCMV proteins. It is unclear which HCMV proteins
other than UL57 localize to UPS compartments. However,
HCMV UL76 co-localizes with the UPS component Rpn10/
S5a in discrete areas of infected cell nuclei termed aggressomes
(Lin et al., 2013). Rpn10/S5a is found in UPS compartments
(Tran et al., 2010). Thus, some aggressomes may be UPS
compartments associated with replication compartments that
contain both UL76 and Rpn10/S5a. It is unknown what role
UL76 might have in UPS compartments. Subcompartments
within the interior of replication compartments that do not
contain UL44 or viral DNA have been observed (Strang et al.,
2012b). It is possible that these subcompartments are the
aforementioned UPS compartments.
The presence of SC35-containing structures on the exterior
of HCMV replication compartments has been reported
(Rechter et al., 2009) (Fig. 2b). It is likely that, as in HSVinfected cells (Chang et al., 2011), these are nuclear
speckles that remain associated with HCMV replication
compartments throughout virus replication. The directed
movement of nuclear speckles with HSV replication
compartments, or vice versa, is associated with efficient
viral RNA transcription in HSV-infected cells, in particular
promoting the export of late HSV RNA transcripts to the
cytoplasm (Chang et al., 2011). The function of nuclear
speckles associated with replication compartments in
HCMV-infected cells has yet to be described, although it
is likely that they are involved in viral RNA transcription.
In summary, HCMV replication compartments are necessary for the concentration and organization of factors
required for viral replication. The organization of factors
required for viral DNA synthesis in replication compartments is increasingly well defined and involves UL44. Viral
replication compartments are dynamic structures whose
architecture is influenced by UL44 localization, viral DNA
synthesis and the cellular nucleolar protein nucleolin.
Nucleolin is not only required to maintain replication
compartment architecture but is also involved in the
subcellular localization of at least one viral protein found
in replication compartments. The organization of factors
required for RNA transcription from the HCMV genome
in replication compartments is not well defined. However,
it can be suggested that viral transcription takes place
throughout the interior of replication compartments, in
contrast to viral DNA synthesis, which takes place within a
layer of UL44 at the periphery of replication compartments. Within replication compartments, there are subcompartments, some of which are associated with protein
quality control. Nuclear speckles are associated with viral
replication compartments where they probably enable
viral transcription, possibly by facilitating the export of
viral transcripts to the cytoplasm.
246
Structures associated with HCMV replication:
nucleoli, Cajal bodies and perinucleolar
compartments
To date, little attention has been paid to the role of nucleoli
and associated subnuclear structures such as Cajal bodies
and perinucleolar compartments in HCMV-infected cells.
It is increasing clear, however, that these subnuclear
structures may have important roles in HCMV genome
replication and virus–host interactions.
Nucleoli are dynamic non-membrane-bound protein and
RNA structures that form on rRNA genes. A nucleolus has
a tripartite structure in which the fibrillar centre is
surrounded by a dense fibrillar component (containing
the majority of nucleolin found in nucleoli) that is
surrounded by the granular component. In HCMVinfected cells, nucleoli remain intact (Strang et al.,
2012c), and visualization of nucleolin in HCMV-infected
cells suggests changes in the size of nucleoli upon infection
(Strang et al., 2012a). Also, striking changes to nucleolar
composition and architecture occur, as, following infection, nucleolin levels in nucleoli increase and nucleolin
accumulates at the periphery of nucleoli (Strang et al.,
2012a) (Fig. 2c). These changes in nucleolar composition
and architecture may relate to the recruitment of nucleolin
to replication compartments, a nucleolar stress response to
infection or the production of ribosomes in the HCMVinfected cell. The recruitment of nucleolin to replication compartments may be related to the interaction of
nucleolin with viral proteins found in replication compartments. It has been observed that the HCMV protein UL84
co-localizes with nucleolin in nucleoli and at the periphery of replication compartments (Bender et al., 2014).
Therefore, it is possible that UL84 recruits nucleolin to
replication compartments from nucleoli, which results
in changes to nucleolar composition and architecture.
Nucleoli can act as sensors of environmental, genotoxic or
osmotic stress, and pathogen invasion (Boisvert et al., 2007;
Boulon et al., 2010; Hiscox, 2002; Hiscox et al., 2010).
These stresses result in a variety of outcomes including
changes in nucleolar morphology and the nucleolar
proteome, plus pleiotropic effects of p53 activation
(Boulon et al., 2010). The changes in the nucleolar
proteome and architecture observed in HCMV-infected
cells could relate to a nucleolar stress response. The
nucleolar localization of nucleolin in HCMV-infected cells
is akin to that seen in coronavirus-infected cells. It has been
suggested that localization of nucleolin in coronavirusinfected cells is related to ‘nucleolar capping’ (Dove et al.,
2006). Capping is associated with a nucleolar stress
response caused by factors such as transcriptional inhibition or DNA damage. These stresses cause condensation
and segregation of the fibrillar centre and the granular
component. This is accompanied by the formation of ‘caps’
of nucleoplasmic RNA-binding proteins on the body of
nucleoli (Boulon et al., 2010; Shav-Tal et al., 2005). It has
been reported that nucleolar capping is observed in
HCMV-infected cells (Gaddy et al., 2010). However, it is
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Journal of General Virology 96
HCMV and subnuclear structures
unclear if the reported nucleolar caps are actually
associated with nucleoli. Furthermore, analysis of nucleoli
in HCMV-infected cells by electron microscopy does not
suggest the presence of nucleolar caps (Strang et al., 2012c).
It is unknown whether HCMV infection stimulates
p53-mediated nucleolar stress responses, which include
specific changes to the protein translational profile of the
cell (Boulon et al., 2010). Finally, the principal role of
nucleoli in uninfected cells is to produce ribosomes. Levels
of certain rRNAs increase in infected cells (Tanaka et al.,
1975). Therefore, ribosome production during HCMV
replication may not be compromised. However, the increase
in rRNA production in infected cells could be related to
changes in nucleolar composition and architecture.
An integral part of any of the possibilities discussed could be
the action of HCMV proteins in nucleoli. Several viral
proteins can localize to nucleoli. It has been reported that
HCMV pp65 and UL84 proteins are a nucleolar antigens
(Arcangeletti et al., 2009; Bender et al., 2014). Also, transfection experiments have indicated that several epitopetagged versions of HCMV proteins (TLR5, TLR7, US33,
UL29, UL31 and UL76) localize to the nucleolus in
uninfected cells (Salsman et al., 2008). These proteins are
expressed in different kinetic classes of viral RNA transcription and have different effects on HCMV replication, plus
the function of several of these proteins is unknown (Table
1). Also, it has been reported that epitope-tagged versions of
the putative HCMV proteins TLR7 and UL108 localize to
nucleoli in uninfected cells (Salsman et al., 2008). However,
it is now thought that the HCMV genome does not possess
ORFs encoding TLR7 or UL108 (Gatherer et al., 2011).
Cajal bodies and perinucleolar compartments are subnuclear structures associated with nucleoli (Fig. 2c) and both
may be involved in viral RNA production. Cajal bodies are
non-membrane-bound protein and RNA structures
involved in transcription and maturation of ribonuclear
particles (Ogg & Lamond, 2002). A role for Cajal bodies in
HCMV replication has yet to be described. However,
transfection of epitope-tagged HCMV UL30 leads to
disruption and/or loss of Cajal bodies in uninfected cells
(Table 1). Thus, it has been suggested (Salsman et al., 2008)
that UL30 may allow HCMV to inhibit transcription and
ribonuclear particle maturation. Cajal bodies are thought to
play a role in adenovirus replication, wherein adenovirus
infection results in fragmentation of Cajal bodies (James
et al., 2010). Also, Cajal bodies are required for efficient
production of several late adenoviral RNA transcripts
(James et al., 2010). Thus, it is possible, as in adenovirusinfected cells, that Cajal bodies are disrupted upon HCMV
infection and have a role in HCMV RNA transcription.
However, Cajal bodies are generally absent in slowly
dividing or non-dividing cells (Ogg & Lamond, 2002) and
may not be present in the fibroblast, vascular and neuronal
cell types within which HCMV replicates during productive
infection. It has been suggested that the putative HCMV
protein UL3 is also involved in modification of Cajal bodies
(Salsman et al., 2008). However, as mentioned above, it is
http://vir.sgmjournals.org
now thought that an ORF encoding UL3 is not present in the
HCMV genome (Gatherer et al., 2011).
Perinucleolar compartments are thought to be involved
in RNA polymerase II and III mediated RNA transcription.
In HCMV-infected cells, at least one perinucleolar
compartment protein, polyryrimidine tract-binding protein (PTB), is relocalized from the nucleus to the cytoplasm
or degraded in the nucleus (Gaddy et al., 2010). It has been
suggested (Gaddy et al., 2010) that loss of PTB from
perinucleolar compartments is related to negative regulation of viral splicing by PTB (Cosme et al., 2009).
In summary, upon HCMV infection of the cell, nucleoli
remain intact but changes to nucleolar composition and
architecture are observed. These changes may be related
to a cellular nucleolar stress response, the recruitment of
nucleolin to HCMV replication compartments or the
presence of viral proteins within nucleoli. Subnuclear
structures associated with nucleoli, including Cajal bodies
and perinucleolar compartments, may be involved in viral
RNA production in HCMV-infected cells.
Production of DNA-filled capsids: a viral structure
putatively involved in capsid assembly and
genome packaging
Late viral transcription produces proteins required to form
viral capsids. The assembly of HCMV capsids and packaging
of genomes into them is increasingly well understood
(Gibson, 2008; McVoy et al., 2000; Scheffczik et al., 2002;
Wang et al., 2012). However, it is unclear where within
viral replication compartments capsid assembly and genome
packaging take place. These questions have not been extensively investigated.
There are few data regarding the localization of HCMV
capsid proteins within the infected cell nucleus. However,
it has been reported that a ‘sponge-like’ web of UL80
proteins forms throughout the infected cell nucleus,
dependent upon UL80 self-interaction (Loveland et al.,
2007; Nguyen et al., 2008). UL80 proteins include the
capsid assembly protein precursor (UL80.5) and the
maturational protease precursor (UL80a), which interact
with the major HCMV capsid protein UL86 (Wood et al.,
1997). It is possible that the web of UL80 proteins could act
as a scaffold for capsid assembly and genome packaging
within viral replication compartments.
Packaging of viral DNA into nascent viral capsids probably
takes place within replication compartments. The viral
protein responsible for DNA binding during the packaging
process, UL56, is found within replication compartments.
UL56 localization to replication compartments is dependent on viral DNA synthesis, and UL56 may interact with
UL44 within replication compartments (Giesen et al.,
2000). Furthermore, it has been speculated that packaging
of viral DNA into capsids is responsible for the movement
of newly synthesized viral DNA from the periphery of
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
247
B. L. Strang
replication compartments to the interior of replication
compartments (Strang et al., 2012b).
Therefore, further study is required to understand the
relationship between the production of DNA-containing
viral capsids and viral replication compartments. It can be
proposed, however, that viral capsid proteins play a role in
organizing capsid production by creating a subnuclear
structure on which capsid assembly and DNA packaging
might take place.
Exit of capsids from the nucleus: the nuclear
lamina
To escape the nucleus, DNA-containing capsids must first
travel from within viral replication compartments to the
nuclear periphery. It is unknown what facilitates this
process in HCMV-infected cells. In HSV-infected cells,
directed capsid movement in the nucleus is dependent
upon myosin and actin (Forest et al., 2005). It is possible
that HCMV also utilizes these cellular cargo transporters to
facilitate capsid movement. Recently, it has been reported
that mutation of HCMV UL84 leads to a defect in virus
replication associated with a defect in nuclear egress of
capsids and the clustering of capsids around nucleoli
(Strang et al., 2012c). This would suggest that UL84 is,
directly or indirectly, involved in the efficient movement
of HCMV capsids within the nucleus. It has yet to be
determined what relevance the mislocalization of capsids to
nucleoli has on the movement of capsids in the nucleus.
At the periphery of the nucleus, capsids must traverse the
physical barrier of the nuclear lamina and pass though
the nuclear membrane. There have been several recent
advances in our understanding of these processes. HCMV
proteins mediate events at the nuclear lamina required for
nuclear egress. The viral nuclear egress complex (UL50 and
UL53) recruits the viral kinase UL97 to the nuclear lamina
in HCMV-infected cells (Sharma et al., 2014) (Table 1).
Phosphorylation of the lamina component lamin A/C by
UL97 promotes lamina disruption, akin to that observed
during cell division, resulting in thinning and the
appearance of gaps in the lamina (Hamirally et al., 2009).
This should facilitate capsid movement through the lamina
to the nuclear membrane. The requirement for other viral
(including c-ORF29 and UL45) and cellular (including
emerin, p32 and protein kinase C) proteins implicated in
nuclear egress in this model remains unclear (Milbradt
et al., 2014; Miller et al., 2010). It is unknown what
mechanism selects only DNA-containing capsids for
nuclear egress in HCMV-infected cells. In HSV-infected
cells, this process is governed by the interaction of the
nuclear egress complex and capsid proteins (Yang &
Baines, 2011). A similar mechanism may operate in
HCMV-infected cells.
Once through the lamina, capsids must traverse the nuclear
membrane in a process now thought to be similar to the
passage of large ribonucleoprotein particles from the nucleus
248
to the cytoplasm (Jokhi et al., 2013; Speese et al., 2012).
Briefly, capsids must bud through the inner leaflet of the
nuclear membrane into the intramembrane space. A fusion
event with the outer leaflet of the membrane grants capsids
access to the cytoplasm. Capsids then engage the cytoplasmic
membrane envelopment processes via a cytoplasmic viral
assembly compartment (Alwine, 2012), which leads to virion
exit from the cell. It is thought that the development of the
cytoplasmic assembly compartment next to the nucleus
deforms the nuclear membrane. This creates a concave
surface on the nucleus, giving it a ‘kidney bean’ shape (Azzeh
et al., 2006). To gain direct access to the cytoplasmic
assembly compartment, capsids probably exit the nucleus
through the concave face of the nuclear membrane.
In summary, the process that facilitates the movement of
DNA-containing capsids to the nuclear periphery is ill
defined but probably involves cellular cargo transporters.
Once at the nuclear membrane, a virally encoded nuclear
egress complex possibly selects DNA-containing capsids for
nuclear egress. This nuclear egress complex then facilitates
capsid movement through the nuclear lamina by promoting modification of lamina components which results in
thinning and the appearance of gaps in the lamina. The
capsid moves through the inner and outer leaflets of the
nuclear membrane via the intramembrane space. Once in
the cytoplasm, capsids participate in a complex membrane
envelopment process via a viral cytoplasmic assembly
compartment that leads to virion exit from the cell. There
may be a relationship between the construction of the viral
cytoplasmic assembly compartment, deformation of the
nucleus and exit of capsids through the nuclear membrane
proximal to assembly compartments.
Further questions and future perspectives
As discussed here, there have been notable advances in
our understanding of HCMV replication and virus–
host interactions by examining subnuclear structures.
However, a full examination of the questions that remain
is limited by several factors. These include our inability to
fully dissect viral gene expression or protein function and
explore the composition of viral and cellular subnuclear
structures. To address these issues, technological development is required. Of particular importance to interrogating the role of virally encoded factors will be the
continued development of robust systems to produce
recombinant HCMV viruses, including viruses with lethal
mutations in their genomes. The study of viral genomes
containing lethal mutations can currently be achieved by
studying cells transfected with bacmids containing mutated
HCMV genomes, for example Silva et al. (2010), although
this methodology has its limitations. Examination of viral
cellular subnuclear structure composition requires the
development of well-characterized antibodies recognizing
unmodified and post-translationally modified forms of
viral and cellular proteins. Recombinant viruses and antibodies can then be coupled with increasingly sophisticated
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Journal of General Virology 96
HCMV and subnuclear structures
molecular and cellular techniques. These include live-cell
imaging, super-resolution microscopy, correlative light
electron microscopic analysis (Sharma et al., 2014), chemical
biology techniques (for example, ‘click chemistry’; Strang et
al., 2012b) and proteomic analysis of subnuclear structures
(Andersen et al., 2002; Hiscox et al., 2010).
A number of the observations discussed above rely on the
study of transfection experiments using epitope-tagged
versions of viral proteins in uninfected cells (Kuny et al.,
2010; Prichard et al., 2008; Salsman et al., 2008). It is
unclear what effect overexpression during transfection or
epitope tagging might have on protein function. Thus, data
based on transfection experiments should be interpreted
cautiously. Furthermore, certain aspects of this review rely
on the study of HSV. However, several features of HSV
replication differ from HCMV replication, probably due to
evolutionary divergence among herpesviruses. These
include antagonism of the PML response (Everett et al.,
2013), nucleolar organization (Lymberopoulos & Pearson,
2007; Strang et al., 2012a), the localization of nucleolin
(Lymberopoulos & Pearson, 2007; Strang et al., 2012a) and
replication compartment organization (Liptak et al., 1996;
Strang et al., 2012b). There are areas where examination of
HSV, or other herpesviruses, may not benefit HCMV
research. These points underscore the need to generate
reagents and methodologies to examine events that occur
in HCMV-infected cells.
The use of laboratory viral strains, the cell types studied and
virus latency must be considered in future experiments.
Most of the data outlined in this review were derived from
the study of HCMV laboratory strains. The genomic
complexity of HCMV is still being addressed. Primary
strains of HCMV contain several more ORFs than laboratory
strains of HCMV (Dolan et al., 2004). Also, recent highresolution transcriptome mapping (Gatherer et al., 2011)
and ribosome profiling (Stern-Ginossar et al., 2012) have
revealed the presence of many previously unrecognized viral
RNA transcripts and non-canonical ORFs. Further investigation is required to discover how recently identified
HCMV factors might influence viral or cellular subnuclear
structures. These factors include newly identified transcripts
from the US33 and TRL9 loci (Gatherer et al., 2011), which
may localize to the nucleolus of HCMV-infected cells
(Salsman et al., 2008) (Table 1). Also, the HCMV
transcriptome contains a number of long (Gatherer et al.,
2011) and short (Grey et al., 2005) non-coding RNAs. As yet,
there are no data indicating that HCMV non-coding RNAs
are involved in the function of viral or cellular subnuclear
structures.
The HCMV transcriptome varies between cell types
(Towler et al., 2012), suggesting cell-type-specific requirements for viral factors. Transcripts differentially expressed
between cell lines tested include those encoding UL44
(involved in viral replication compartment architecture),
UL98 (suggested to be involved in chromatin partitioning),
TRL5 (thought to be localized to the nucleolus), UL80
http://vir.sgmjournals.org
proteins (thought to be involved in capsid assembly within
replication compartments) plus UL97 and UL50 (involved
in capsid egress through the nuclear lamina) (Towler et al.,
2012) (Table 1). Thus, the function of viral and cellular
subnuclear structures could vary among cell types.
Virus latency is a hallmark of herpesvirus infection. However,
our understanding of the establishment, maintenance and
reactivation from latency of HCMV is incomplete. Recent in
vitro models of virus infection in haematopoietic cells (for
example, CD14+ and CD34+ cells) have illuminated
important aspects of HCMV latency. The loss of hDaxx in
CD34+ cells is required for transcription of IE viral genes
upon infection by laboratory, but not primary, strains of
HCMV (Saffert et al., 2010). This would argue that PMLNBs repress HCMV transcription from laboratory HCMV
strains and that primary HCMV strains possess as-yetunidentified functions that control viral transcription
during latency. Co-infection of CD34+ cells with both
laboratory and primary strains of HCMV blocks the ability
of histone deacetylases to activate IE gene transcription from
the laboratory strain of HCMV (Saffert et al., 2010). Thus,
transcriptional repression of primary HCMV strains may
relate to chromatin modification by histone deacetylases.
Also, it is unclear if hDaxx has a major role in repression of
HCMV gene transcription during latency. In the embryonic
carcinoma cell line NT2D1, which can recapitulate the
differentiation-dependent regulation of IE gene expression
observed during latency, repression of IE gene expression
has been reported in the presence (Saffert & Kalejta, 2007)
and absence (Groves & Sinclair, 2007) of hDaxx in
undifferentiated NT2D1 cells. This may be due to differences
in the virus strains used in each study. Further experiments
are required to ascertain the relationship between virus
strain, cell type, cell differentiation and gene expression
during HCMV latency.
There are few other data regarding the involvement of other
subnuclear structures in HCMV latency. Furthermore, the
latent HCMV transcriptome in either CD14+ or CD34+
cells (Rossetto et al., 2013) does not produce the high levels
of transcripts encoding HCMV proteins thought to localized
to cellular subnuclear structures during productive HCMV
replication. Thus, if cellular subnuclear structures are
involved in HCMV latency, different repertoires of viral
factors interact with subnuclear structures during productive and latent HCMV infections.
Acknowledgements
Thanks to Andrew Davison, Wade Gibson, David Pasdeloup and
Frazer Rixon for insightful discussions, plus Chris Boutell and Mayuri
Sharma for comments on the manuscript. I would also like to thank
the anonymous reviewers for their constructive comments, which
have greatly improved this manuscript. I sincerely apologize to
colleagues whose important work was not discussed or cited due to
space limitations. This work was supported by St George’s, University
of London, London, UK.
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
249
B. L. Strang
References
expression and viral replication of human cytomegalovirus. J Virol 83,
2839–2850.
Ahn, J. H. & Hayward, G. S. (1997). The major immediate-early
Costa, H., Nascimento, R., Sinclair, J. & Parkhouse, R. M. (2013).
proteins IE1 and IE2 of human cytomegalovirus colocalize with and
disrupt PML-associated nuclear bodies at very early times in infected
permissive cells. J Virol 71, 4599–4613.
Human cytomegalovirus gene UL76 induces IL-8 expression through
activation of the DNA damage response. PLoS Pathog 9, e1003609.
Ahn, J. H., Jang, W. J. & Hayward, G. S. (1999). The human
cytomegalovirus IE2 and UL112-113 proteins accumulate in viral
DNA replication compartments that initiate from the periphery of
promyelocytic leukemia protein-associated nuclear bodies (PODs or
ND10). J Virol 73, 10458–10471.
Alwine, J. C. (2012). The human cytomegalovirus assembly compartment: a masterpiece of viral manipulation of cellular processes that
facilitates assembly and egress. PLoS Pathog 8, e1002878.
Andersen, J. S., Lyon, C. E., Fox, A. H., Leung, A. K., Lam, Y. W.,
Steen, H., Mann, M. & Lamond, A. I. (2002). Directed proteomic
analysis of the human nucleolus. Curr Biol 12, 1–11.
Appleton, B. A., Loregian, A., Filman, D. J., Coen, D. M. & Hogle, J. M.
(2004). The cytomegalovirus DNA polymerase subunit UL44 forms a
C clamp-shaped dimer. Mol Cell 15, 233–244.
Appleton, B. A., Brooks, J., Loregian, A., Filman, D. J., Coen, D. M. &
Hogle, J. M. (2006). Crystal structure of the cytomegalovirus DNA
polymerase subunit UL44 in complex with the C terminus from the
catalytic subunit. Differences in structure and function relative to
unliganded UL44. J Biol Chem 281, 5224–5232.
Dolan, A., Cunningham, C., Hector, R. D., Hassan-Walker, A. F., Lee,
L., Addison, C., Dargan, D. J., McGeoch, D. J., Gatherer, D. & other
authors (2004). Genetic content of wild-type human cytomegalo-
virus. J Gen Virol 85, 1301–1312.
Dove, B. K., You, J. H., Reed, M. L., Emmett, S. R., Brooks, G. &
Hiscox, J. A. (2006). Changes in nucleolar morphology and proteins
during infection with the coronavirus infectious bronchitis virus. Cell
Microbiol 8, 1147–1157.
E, X., Pickering, M. T., Debatis, M., Castillo, J., Lagadinos, A., Wang,
S., Lu, S. & Kowalik, T. F. (2011). An E2F1-mediated DNA damage
response contributes to the replication of human cytomegalovirus.
PLoS Pathog 7, e1001342.
Ertl, P. F. & Powell, K. L. (1992). Physical and functional interaction of
human cytomegalovirus DNA polymerase and its accessory protein
(ICP36) expressed in insect cells. J Virol 66, 4126–4133.
Everett, R. D., Boutell, C. & Hale, B. G. (2013). Interplay between
viruses and host sumoylation pathways. Nat Rev Microbiol 11, 400–
411.
Feichtinger, S., Stamminger, T., Müller, R., Graf, L., Klebl, B.,
Eickhoff, J. & Marschall, M. (2011). Recruitment of cyclin-dependent
Arcangeletti, M. C., Rodighiero, I., De Conto, F., Gatti, R., Orlandini,
G., Ferraglia, F., Motta, F., Covan, S., Razin, S. V. & other authors
(2009). Modulatory effect of rRNA synthesis and ppUL83 nucleolar
kinase 9 to nuclear compartments during cytomegalovirus late
replication: importance of an interaction between viral pUL69 and
cyclin T1. J Gen Virol 92, 1519–1531.
compartmentalization on human cytomegalovirus gene expression in
vitro. J Cell Biochem 108, 415–423.
Forest, T., Barnard, S. & Baines, J. D. (2005). Active intranuclear
Azzeh, M., Honigman, A., Taraboulos, A., Rouvinski, A. & Wolf, D. G.
(2006). Structural changes in human cytomegalovirus cytoplasmic
Fortunato, E. A. & Spector, D. H. (1998). p53 and RPA are sequestered
assembly sites in the absence of UL97 kinase activity. Virology 354,
69–79.
Bender, B. J., Coen, D. M. & Strang, B. L. (2014). Dynamic and
nucleolin-dependent localization of human cytomegalovirus UL84 to
the periphery of viral replication compartments and nucleoli. J Virol
88, 11738–11747.
movement of herpesvirus capsids. Nat Cell Biol 7, 429–431.
in viral replication centers in the nuclei of cells infected with human
cytomegalovirus. J Virol 72, 2033–2039.
Gaddy, C. E., Wong, D. S., Markowitz-Shulman, A. & Colberg-Poley,
A. M. (2010). Regulation of the subcellular distribution of key cellular
RNA-processing factors during permissive human cytomegalovirus
infection. J Gen Virol 91, 1547–1559.
Bogdanow, B., Weisbach, H., von Einem, J., Straschewski, S., Voigt,
S., Winkler, M., Hagemeier, C. & Wiebusch, L. (2013). Human
Gao, Y., Colletti, K. & Pari, G. S. (2008). Identification of human
cytomegalovirus UL84 virus- and cell-encoded binding partners by
using proteomics analysis. J Virol 82, 96–104.
cytomegalovirus tegument protein pp150 acts as a cyclin A2-CDKdependent sensor of the host cell cycle and differentiation state. Proc
Natl Acad Sci U S A 110, 17510–17515.
Gatherer, D., Seirafian, S., Cunningham, C., Holton, M., Dargan, D. J.,
Baluchova, K., Hector, R. D., Galbraith, J., Herzyk, P. & other authors
(2011). High-resolution human cytomegalovirus transcriptome. Proc
Boisvert, F. M., van Koningsbruggen, S., Navascués, J. & Lamond,
A. I. (2007). The multifunctional nucleolus. Nat Rev Mol Cell Biol 8,
Gibson, W. (2008). Structure and formation of the cytomegalovirus
574–585.
Natl Acad Sci U S A 108, 19755–19760.
virion. Curr Top Microbiol Immunol 325, 187–204.
Boulon, S., Westman, B. J., Hutten, S., Boisvert, F. M. & Lamond, A. I.
(2010). The nucleolus under stress. Mol Cell 40, 216–227.
Giesen, K., Radsak, K. & Bogner, E. (2000). Targeting of the gene
Boutell, C. & Everett, R. D. (2013). Regulation of alphaherpesvirus
product encoded by ORF UL56 of human cytomegalovirus into viral
replication centers. FEBS Lett 471, 215–218.
infections by the ICP0 family of proteins. J Gen Virol 94, 465–481.
Ginisty, H., Sicard, H., Roger, B. & Bouvet, P. (1999). Structure and
Chambers, J., Angulo, A., Amaratunga, D., Guo, H., Jiang, Y., Wan,
J. S., Bittner, A., Frueh, K., Jackson, M. R. & other authors (1999).
Glass, M. & Everett, R. D. (2013). Components of promyelocytic
DNA microarrays of the complex human cytomegalovirus genome:
profiling kinetic class with drug sensitivity of viral gene expression.
J Virol 73, 5757–5766.
Chang, L., Godinez, W. J., Kim, I. H., Tektonidis, M., de Lanerolle, P.,
Eils, R., Rohr, K. & Knipe, D. M. (2011). Herpesviral replication
compartments move and coalesce at nuclear speckles to enhance
export of viral late mRNA. Proc Natl Acad Sci U S A 108, E136–E144.
Cosme, R. S., Yamamura, Y. & Tang, Q. (2009). Roles of
polypyrimidine tract binding proteins in major immediate-early gene
250
functions of nucleolin. J Cell Sci 112, 761–772.
leukemia nuclear bodies (ND10) act cooperatively to repress
herpesvirus infection. J Virol 87, 2174–2185.
Grey, F., Antoniewicz, A., Allen, E., Saugstad, J., McShea, A.,
Carrington, J. C. & Nelson, J. (2005). Identification and character-
ization of human cytomegalovirus-encoded microRNAs. J Virol 79,
12095–12099.
Groves, I. J. & Sinclair, J. H. (2007). Knockdown of hDaxx in normally
non-permissive undifferentiated cells does not permit human cytomegalovirus immediate-early gene expression. J Gen Virol 88, 2935–2940.
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Journal of General Virology 96
HCMV and subnuclear structures
Hamirally, S., Kamil, J. P., Ndassa-Colday, Y. M., Lin, A. J., Jahng, W. J.,
Baek, M. C., Noton, S., Silva, L. A., Simpson-Holley, M. & other
authors (2009). Viral mimicry of Cdc2/cyclin-dependent kinase 1
mediates disruption of nuclear lamina during human cytomegalovirus nuclear egress. PLoS Pathog 5, e1000275.
Kuny, C. V., Chinchilla, K., Culbertson, M. R. & Kalejta, R. F. (2010).
Cyclin-dependent kinase-like function is shared by the beta- and
gamma-subset of the conserved herpesvirus protein kinases. PLoS
Pathog 6, e1001092.
Lamond, A. I. & Spector, D. L. (2003). Nuclear speckles: a model for
Hiscox, J. A. (2002). The nucleolus – a gateway to viral infection? Arch
nuclear organelles. Nat Rev Mol Cell Biol 4, 605–612.
Virol 147, 1077–1089.
Lee, H. R., Kim, D. J., Lee, J. M., Choi, C. Y., Ahn, B. Y., Hayward, G. S.
& Ahn, J. H. (2004). Ability of the human cytomegalovirus IE1 protein
Hiscox, J. A., Whitehouse, A. & Matthews, D. A. (2010). Nucleolar
Hume, A. J., Finkel, J. S., Kamil, J. P., Coen, D. M., Culbertson, M. R. &
Kalejta, R. F. (2008). Phosphorylation of retinoblastoma protein by
proteomics and viral infection. Proteomics 10, 4077–4086.
to modulate sumoylation of PML correlates with its functional
activities in transcriptional regulation and infectivity in cultured
fibroblast cells. J Virol 78, 6527–6542.
viral protein with cyclin-dependent kinase function. Science 320, 797–
799.
Lin, S. R., Jiang, M. J., Wang, H. H., Hu, C. H., Hsu, M. S., Hsi, E., Duh,
C. Y. & Wang, S. K. (2013). Human cytomegalovirus UL76 elicits
Ishov, A. M., Stenberg, R. M. & Maul, G. G. (1997). Human
novel aggresome formation via interaction with S5a of the ubiquitin
proteasome system. J Virol 87, 11562–11578.
cytomegalovirus immediate early interaction with host nuclear
structures: definition of an immediate transcript environment. J Cell
Biol 138, 5–16.
Liptak, L. M., Uprichard, S. L. & Knipe, D. M. (1996). Functional order
of assembly of herpes simplex virus DNA replication proteins into
prereplicative site structures. J Virol 70, 1759–1767.
Ishov, A. M., Sotnikov, A. G., Negorev, D., Vladimirova, O. V., Neff, N.,
Kamitani, T., Yeh, E. T., Strauss, J. F., III & Maul, G. G. (1999). PML is
Livingston, C. M., Ifrim, M. F., Cowan, A. E. & Weller, S. K. (2009).
critical for ND10 formation and recruits the PML-interacting protein
daxx to this nuclear structure when modified by SUMO-1. J Cell Biol
147, 221–234.
Virus-Induced Chaperone-Enriched (VICE) domains function as
nuclear protein quality control centers during HSV-1 infection. PLoS
Pathog 5, e1000619.
Ishov, A. M., Vladimirova, O. V. & Maul, G. G. (2004).
Loregian, A., Appleton, B. A., Hogle, J. M. & Coen, D. M. (2004a).
Heterochromatin and ND10 are cell-cycle regulated and phosphorylation-dependent alternate nuclear sites of the transcription
repressor Daxx and SWI/SNF protein ATRX. J Cell Sci 117, 3807–
3820.
Specific residues in the connector loop of the human cytomegalovirus
DNA polymerase accessory protein UL44 are crucial for interaction
with the UL54 catalytic subunit. J Virol 78, 9084–9092.
James, N. J., Howell, G. J., Walker, J. H. & Blair, G. E. (2010). The role
Residues of human cytomegalovirus DNA polymerase catalytic
subunit UL54 that are necessary and sufficient for interaction with
the accessory protein UL44. J Virol 78, 158–167.
of Cajal bodies in the expression of late phase adenovirus proteins.
Virology 399, 299–311.
Jokhi, V., Ashley, J., Nunnari, J., Noma, A., Ito, N., Wakabayashi-Ito,
N., Moore, M. J. & Budnik, V. (2013). Torsin mediates primary
Loregian, A., Appleton, B. A., Hogle, J. M. & Coen, D. M. (2004b).
Loveland, A. N., Nguyen, N. L., Brignole, E. J. & Gibson, W. (2007).
envelopment of large ribonucleoprotein granules at the nuclear
envelope. Cell Rep 3, 988–995.
The amino-conserved domain of human cytomegalovirus UL80a
proteins is required for key interactions during early stages of capsid
formation and virus production. J Virol 81, 620–628.
Kagele, D., Rossetto, C. C., Elorza, M. & Pari, G. S. (2013). Analysis of
Lukashchuk, V., McFarlane, S., Everett, R. D. & Preston, C. M. (2008).
the interactions of viral and cellular factors with human cytomegalovirus lytic origin of replication, oriLyt. Virology 424, 106–114.
Kalejta, R. F. & Shenk, T. (2003). Proteasome-dependent, ubiquitin-
Human cytomegalovirus protein pp71 displaces the chromatinassociated factor ATRX from nuclear domain 10 at early stages of
infection. J Virol 82, 12543–12554.
independent degradation of the Rb family of tumor suppressors by
the human cytomegalovirus pp71 protein. Proc Natl Acad Sci U S A
100, 3263–3268.
Lymberopoulos, M. H. & Pearson, A. (2007). Involvement of UL24 in
herpes-simplex-virus-1-induced dispersal of nucleolin. Virology 363,
397–409.
Kang, H., Kim, E. T., Lee, H. R., Park, J. J., Go, Y. Y., Choi, C. Y. & Ahn,
J. H. (2006). Inhibition of SUMO-independent PML oligomerization
McVoy, M. A. & Adler, S. P. (1994). Human cytomegalovirus DNA
by the human cytomegalovirus IE1 protein. J Gen Virol 87, 2181–
2190.
Kim, Y. E., Lee, J. H., Kim, E. T., Shin, H. J., Gu, S. Y., Seol, H. S., Ling,
P. D., Lee, C. H. & Ahn, J. H. (2011). Human cytomegalovirus infection
causes degradation of Sp100 proteins that suppress viral gene
expression. J Virol 85, 11928–11937.
Knizewski, L., Kinch, L., Grishin, N. V., Rychlewski, L. & Ginalski, K.
(2006). Human herpesvirus 1 UL24 gene encodes a potential PD-(D/
replicates after early circularization by concatemer formation, and
inversion occurs within the concatemer. J Virol 68, 1040–1051.
McVoy, M. A., Nixon, D. E., Hur, J. K. & Adler, S. P. (2000). The ends
on herpesvirus DNA replicative concatemers contain pac2 cis
cleavage/packaging elements and their formation is controlled by
terminal cis sequences. J Virol 74, 1587–1592.
Milbradt, J., Kraut, A., Hutterer, C., Sonntag, E., Schmeiser, C., Ferro,
M., Wagner, S., Lenac, T., Claus, C. & other authors (2014).
E)XK endonuclease. J Virol 80, 2575–2577.
Proteomic analysis of the multimeric nuclear egress complex of
human cytomegalovirus. Mol Cell Proteomics 13, 2132–2146.
Kobiler, O., Brodersen, P., Taylor, M. P., Ludmir, E. B. & Enquist, L. W.
(2011). Herpesvirus replication compartments originate with single
Miller, M. S., Furlong, W. E., Pennell, L., Geadah, M. & Hertel, L.
(2010). RASCAL is a new human cytomegalovirus-encoded protein
incoming viral genomes. MBio 2, e00278-11.
that localizes to the nuclear lamina and in cytoplasmic vesicles at late
times postinfection. J Virol 84, 6483–6496.
Kobiler, O., Drayman, N., Butin-Israeli, V. & Oppenheim, A. (2012).
Virus strategies for passing the nuclear envelope barrier. Nucleus 3,
526–539.
Mocarski, E. S., Shenk, T. & Pass, R. F. (2007). Cytomegaloviruses. In
Fields Virology, 5th edn, vol. 2, pp. 2701–2772. Edited by D. M. Knipe
& P. M. Howley. New York, NY: Lippincott, Williams & Wilkins.
Korioth, F., Maul, G. G., Plachter, B., Stamminger, T. & Frey, J.
(1996). The nuclear domain 10 (ND10) is disrupted by the human
Moldovan, G. L., Pfander, B. & Jentsch, S. (2007). PCNA, the maestro
cytomegalovirus gene product IE1. Exp Cell Res 229, 155–158.
of the replication fork. Cell 129, 665–679.
http://vir.sgmjournals.org
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
251
B. L. Strang
Nguyen, N. L., Loveland, A. N. & Gibson, W. (2008). Nuclear
localization sequences in cytomegalovirus capsid assembly proteins
(UL80 proteins) are required for virus production: inactivating NLS1,
NLS2, or both affects replication to strikingly different extents. J Virol
82, 5381–5389.
O’Dowd, J. M., Zavala, A. G., Brown, C. J., Mori, T. & Fortunato, E. A.
(2012). HCMV-infected cells maintain efficient nucleotide excision
repair of the viral genome while abrogating repair of the host genome.
PLoS Pathog 8, e1003038.
Ogg, S. C. & Lamond, A. I. (2002). Cajal bodies and coilin – moving
towards function. J Cell Biol 159, 17–21.
Pari, G. S. & Anders, D. G. (1993). Eleven loci encoding trans-acting
factors are required for transient complementation of human
cytomegalovirus oriLyt-dependent DNA replication. J Virol 67,
6979–6988.
subcellular localization and alteration of PML nuclear bodies. PLoS
Pathog 4, e1000100.
Sarisky, R. T. & Hayward, G. S. (1996). Evidence that the UL84 gene
product of human cytomegalovirus is essential for promoting oriLytdependent DNA replication and formation of replication compartments in cotransfection assays. J Virol 70, 7398–7413.
Scheffczik, H., Savva, C. G., Holzenburg, A., Kolesnikova, L. &
Bogner, E. (2002). The terminase subunits pUL56 and pUL89 of
human cytomegalovirus are DNA-metabolizing proteins with toroidal
structure. Nucleic Acids Res 30, 1695–1703.
Schreiner, S., Wimmer, P., Sirma, H., Everett, R. D., Blanchette, P.,
Groitl, P. & Dobner, T. (2010). Proteasome-dependent degradation of
Daxx by the viral E1B-55K protein in human adenovirus-infected
cells. J Virol 84, 7029–7038.
Pari, G. S., Kacica, M. A. & Anders, D. G. (1993). Open reading frames
Seeler, J. S., Marchio, A., Sitterlin, D., Transy, C. & Dejean, A.
(1998). Interaction of SP100 with HP1 proteins: a link between
UL44, IRS1/TRS1, and UL36-38 are required for transient complementation of human cytomegalovirus oriLyt-dependent DNA
synthesis. J Virol 67, 2575–2582.
the promyelocytic leukemia-associated nuclear bodies and the
chromatin compartment. Proc Natl Acad Sci U S A 95, 7316–
7321.
Penfold, M. E. & Mocarski, E. S. (1997). Formation of cytomegalo-
Seeler, J. S., Marchio, A., Losson, R., Desterro, J. M., Hay, R. T.,
Chambon, P. & Dejean, A. (2001). Common properties of nuclear
virus DNA replication compartments defined by localization of viral
proteins and DNA synthesis. Virology 239, 46–61.
Prichard, M. N., Duke, G. M. & Mocarski, E. S. (1996). Human
cytomegalovirus uracil DNA glycosylase is required for the normal
temporal regulation of both DNA synthesis and viral replication.
J Virol 70, 3018–3025.
Prichard, M. N., Lawlor, H., Duke, G. M., Mo, C., Wang, Z., Dixon, M.,
Kemble, G. & Kern, E. R. (2005). Human cytomegalovirus uracil DNA
glycosylase associates with ppUL44 and accelerates the accumulation
of viral DNA. Virol J 2, 55.
Prichard, M. N., Sztul, E., Daily, S. L., Perry, A. L., Frederick, S. L., Gill,
R. B., Hartline, C. B., Streblow, D. N., Varnum, S. M. & other authors
(2008). Human cytomegalovirus UL97 kinase activity is required for
body protein SP100 and TIF1alpha chromatin factor: role of SUMO
modification. Mol Cell Biol 21, 3314–3324.
Sharma, M., Kamil, J. P., Coughlin, M., Reim, N. I. & Coen, D. M.
(2014). Human cytomegalovirus UL50 and UL53 recruit viral protein
kinase UL97, not protein kinase C, for disruption of nuclear lamina
and nuclear egress in infected cells. J Virol 88, 249–262.
Shav-Tal, Y., Blechman, J., Darzacq, X., Montagna, C., Dye, B. T.,
Patton, J. G., Singer, R. H. & Zipori, D. (2005). Dynamic sorting of
nuclear components into distinct nucleolar caps during transcriptional inhibition. Mol Biol Cell 16, 2395–2413.
Sheaffer, A. K., Weinheimer, S. P. & Tenney, D. J. (1997). The human
cytomegalovirus UL98 gene encodes the conserved herpesvirus
alkaline nuclease. J Gen Virol 78, 2953–2961.
the hyperphosphorylation of retinoblastoma protein and inhibits the
formation of nuclear aggresomes. J Virol 82, 5054–5067.
Siew, V. K., Duh, C. Y. & Wang, S. K. (2009). Human cytomegalovirus
Ranneberg-Nilsen, T., Dale, H. A., Luna, L., Slettebakk, R.,
Sundheim, O., Rollag, H. & Bjørås, M. (2008). Characterization of
Silva, L., Cliffe, A., Chang, L. & Knipe, D. M. (2008). Role for A-type
human cytomegalovirus uracil DNA glycosylase (UL114) and its
interaction with polymerase processivity factor (UL44). J Mol Biol
381, 276–288.
Rechter, S., Scott, G. M., Eickhoff, J., Zielke, K., Auerochs, S., Müller,
R., Stamminger, T., Rawlinson, W. D. & Marschall, M. (2009). Cyclin-
dependent kinases phosphorylate the cytomegalovirus RNA export
protein pUL69 and modulate its nuclear localization and activity.
J Biol Chem 284, 8605–8613.
Reichelt, M., Wang, L., Sommer, M., Perrino, J., Nour, A. M., Sen, N.,
Baiker, A., Zerboni, L. & Arvin, A. M. (2011). Entrapment of viral
capsids in nuclear PML cages is an intrinsic antiviral host defense
against varicella-zoster virus. PLoS Pathog 7, e1001266.
Rossetto, C. C., Tarrant-Elorza, M. & Pari, G. S. (2013). Cis and trans
acting factors involved in human cytomegalovirus experimental and
natural latent infection of CD14 + monocytes and CD34 + cells. PLoS
Pathog 9, e1003366.
Saffert, R. T. & Kalejta, R. F. (2007). Human cytomegalovirus gene
UL76 induces chromosome aberrations. J Biomed Sci 16, 107.
lamins in herpesviral DNA targeting and heterochromatin modulation. PLoS Pathog 4, e1000071.
Silva, L. A., Loregian, A., Pari, G. S., Strang, B. L. & Coen, D. M.
(2010). The carboxy-terminal segment of the human cytomegalovirus
DNA polymerase accessory subunit UL44 is crucial for viral
replication. J Virol 84, 11563–11568.
Silva, L. A., Strang, B. L., Lin, E. W., Kamil, J. P. & Coen, D. M. (2011).
Sites and roles of phosphorylation of the human cytomegalovirus
DNA polymerase subunit UL44. Virology 417, 268–280.
Speese, S. D., Ashley, J., Jokhi, V., Nunnari, J., Barria, R., Li, Y.,
Ataman, B., Koon, A., Chang, Y. T. & other authors (2012). Nuclear
envelope budding enables large ribonucleoprotein particle export
during synaptic Wnt signaling. Cell 149, 832–846.
Stepp, W. H., Meyers, J. M. & McBride, A. A. (2013). Sp100 provides
intrinsic immunity against human papillomavirus infection. MBio 4,
e00845-e13.
expression is silenced by Daxx-mediated intrinsic immune defense in
model latent infections established in vitro. J Virol 81, 9109–9120.
Stern-Ginossar, N., Weisburd, B., Michalski, A., Le, V. T., Hein,
M. Y., Huang, S. X., Ma, M., Shen, B., Qian, S. B. & other authors
(2012). Decoding human cytomegalovirus. Science 338, 1088–
Saffert, R. T., Penkert, R. R. & Kalejta, R. F. (2010). Cellular and viral
1093.
control over the initial events of human cytomegalovirus experimental latency in CD34+ cells. J Virol 84, 5594–5604.
Sternsdorf, T., Jensen, K., Reich, B. & Will, H. (1999). The nuclear dot
Salsman, J., Zimmerman, N., Chen, T., Domagala, M. & Frappier, L.
(2008). Genome-wide screen of three herpesviruses for protein
252
protein sp100, characterization of domains necessary for dimerization, subcellular localization, and modification by small ubiquitin-like
modifiers. J Biol Chem 274, 12555–12566.
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
Journal of General Virology 96
HCMV and subnuclear structures
Strang, B. L. & Coen, D. M. (2010). Interaction of the human
cytomegalovirus uracil DNA glycosylase UL114 with the viral DNA
polymerase catalytic subunit UL54. J Gen Virol 91, 2029–2033.
Strang, B. L., Sinigalia, E., Silva, L. A., Coen, D. M. & Loregian, A.
(2009). Analysis of the association of the human cytomegalovirus
DNA polymerase subunit UL44 with the viral DNA replication factor
UL84. J Virol 83, 7581–7589.
Strang, B. L., Boulant, S. & Coen, D. M. (2010a). Nucleolin associates
Towler, J. C., Ebrahimi, B., Lane, B., Davison, A. J. & Dargan, D. J.
(2012). Human cytomegalovirus transcriptome activity differs during
replication in human fibroblast, epithelial and astrocyte cell lines.
J Gen Virol 93, 1046–1058.
Tran, K., Mahr, J. A. & Spector, D. H. (2010). Proteasome subunits
relocalize during human cytomegalovirus infection, and proteasome
activity is necessary for efficient viral gene transcription. J Virol 84,
3079–3093.
with the human cytomegalovirus DNA polymerase accessory subunit
UL44 and is necessary for efficient viral replication. J Virol 84, 1771–
1784.
Ullman, A. J. & Hearing, P. (2008). Cellular proteins PML and Daxx
mediate an innate antiviral defense antagonized by the adenovirus E4
ORF3 protein. J Virol 82, 7325–7335.
Strang, B. L., Geballe, A. P. & Coen, D. M. (2010b). Association of
Wang, J. B., Zhu, Y., McVoy, M. A. & Parris, D. S. (2012). Changes in
human cytomegalovirus proteins IRS1 and TRS1 with the viral DNA
polymerase accessory subunit UL44. J Gen Virol 91, 2167–2175.
subcellular localization reveal interactions between human cytomegalovirus terminase subunits. Virol J 9, 315.
Strang, B. L., Boulant, S., Kirchhausen, T. & Coen, D. M. (2012a).
Wilkinson, G. W., Kelly, C., Sinclair, J. H. & Rickards, C. (1998).
Host cell nucleolin is required to maintain the architecture of human
cytomegalovirus replication compartments. MBio 3, 301–311.
Strang, B. L., Boulant, S., Chang, L., Knipe, D. M., Kirchhausen, T. &
Coen, D. M. (2012b). Human cytomegalovirus UL44 concentrates at
the periphery of replication compartments, the site of viral DNA
synthesis. J Virol 86, 2089–2095.
Strang, B. L., Bender, B. J., Sharma, M., Pesola, J. M., Sanders, R. L.,
Spector, D. H. & Coen, D. M. (2012c). A mutation deleting sequences
encoding the amino terminus of human cytomegalovirus UL84
impairs interaction with UL44 and capsid localization. J Virol 86,
11066–11077.
Tanaka, S., Furukawa, T. & Plotkin, S. A. (1975). Human
cytomegalovirus stimulates host cell RNA synthesis. J Virol 15, 297–
304.
Disruption of PML-associated nuclear bodies mediated by the human
cytomegalovirus major immediate early gene product. J Gen Virol 79,
1233–1245.
Wood, L. J., Baxter, M. K., Plafker, S. M. & Gibson, W. (1997). Human
cytomegalovirus capsid assembly protein precursor (pUL80.5) interacts with itself and with the major capsid protein (pUL86) through
two different domains. J Virol 71, 179–190.
Xu, Y., Ahn, J. H., Cheng, M., apRhys, C. M., Chiou, C. J., Zong, J.,
Matunis, M. J. & Hayward, G. S. (2001). Proteasome-independent
disruption of PML oncogenic domains (PODs), but not covalent
modification by SUMO-1, is required for human cytomegalovirus
immediate-early protein IE1 to inhibit PML-mediated transcriptional
repression. J Virol 75, 10683–10695.
Xue, Y., Gibbons, R., Yan, Z., Yang, D., McDowell, T. L., Sechi, S., Qin,
J., Zhou, S., Higgs, D. & Wang, W. (2003). The ATRX syndrome
Tavalai, N. & Stamminger, T. (2009). Interplay between herpesvirus
infection and host defense by PML nuclear bodies. Viruses 1, 1240–
1264.
protein forms a chromatin-remodeling complex with Daxx and
localizes in promyelocytic leukemia nuclear bodies. Proc Natl Acad Sci
U S A 100, 10635–10640.
Tavalai, N. & Stamminger, T. (2011). Intrinsic cellular defense
Yang, K. & Baines, J. D. (2011). Selection of HSV capsids for
mechanisms targeting human cytomegalovirus. Virus Res 157, 128–133.
envelopment involves interaction between capsid surface components
pUL31, pUL17, and pUL25. Proc Natl Acad Sci U S A 108, 14276–14281.
Tavalai, N., Adler, M., Scherer, M., Riedl, Y. & Stamminger, T. (2011).
Evidence for a dual antiviral role of the major nuclear domain 10
component Sp100 during the immediate-early and late phases of the
human cytomegalovirus replication cycle. J Virol 85, 9447–9458.
http://vir.sgmjournals.org
Yu, D., Silva, M. C. & Shenk, T. (2003). Functional map of human
cytomegalovirus AD169 defined by global mutational analysis. Proc
Natl Acad Sci U S A 100, 12396–12401.
Downloaded from www.microbiologyresearch.org by
IP: 88.99.165.207
On: Fri, 11 Aug 2017 15:49:36
253