ArticlePDF Available

Induction of DNA double-strand breaks and cellular senescence by human respiratory syncytial virus

Taylor & Francis
Virulence
Authors:

Abstract and Figures

Human respiratory syncytial virus (HRSV) accounts for the majority of lower respiratory tract infections during infancy and childhood and is associated with significant morbidity and mortality. HRSV provokes a proliferation arrest and characteristic syncytia in cellular systems such as immortalized epithelial cells. We show here that HRSV induces the expression of DNA damage markers and proliferation arrest such as P-TP53, P-ATM, CDKN1A and γH2AFX in cultured cells secondary to the production of mitochondrial reactive oxygen species (ROS). The DNA damage foci contained γH2AFX and TP53BP1, indicative of double-strand breaks (DSBs) and could be reversed by antioxidant treatments such as N-Acetylcysteine (NAC) or reduced glutathione ethyl ester (GSHee). The damage observed is associated with the accumulation of senescent cells, displaying a canonical senescent phenotype in both mononuclear cells and syncytia. In addition, we show signs of DNA damage and aging such as γH2AFX and CDKN2A expression in the respiratory epithelia of infected mice long after viral clearance. Altogether, these results show that HRSV triggers a DNA damage-mediated cellular senescence program probably mediated by oxidative stress. The results also suggest that this program might contribute to the physiopathology of the infection, tissue remodeling and aging, and might be associated to long-term consequences of HRSV infections.
Occurrence of cellular senescence in cells infected with HRSV. (A) Occurrence of cellular senescence during HRSV lytic infection of A549 cells seeded at 5200 cells/cm 2. The cells were processed for SA-bgal assay at 48 h.p.i. (MOI D 3). Magnification: 200X, scale bar: 50 mm. (B) Quantification of SA-bgal expression in A549 cells (in total population and syncytia). (C) and (D) Similar assays in HEp-2 cells. (E) Immuno-SA-bgal, an assay that allows the detection of SA-bgal expression in massively infected HEp-2 cells stained with a-HRSV antibodies (red staining). Magnification: 200X; scale bar: 50 mm. (F) Occurrence and quantification of cellular senescence in a culture of HEp-2 cells persistently infected by HRSV. Magnification: 200X; scale bar: 50 mm. (G) Quantification of IL-6 and TNF-a in supernatants of cultured cells. Bars in the graphs represent the mean § SD of 2-3 experiments. n D 3 replicates. (H) Analysis of apoptosis/necrosis. Eight thousand cells/well were plated in 6-well plates and were infected with HRSV the following day at MOI D 3. At 60 h.p.i., the cells were processed for flow cytometry. After treatment with the fluorescent probes (AnnexinV-AlexaFluor488 TM and Sytox TM ) apoptotic cells show green fluorescence, dead cells show brighter green fluorescence, and live cells show little. These populations were distinguished in the FL1 channel of a FACSCalibur flow cytometer. Treatment of cells with 20 mM camptothecin (CPT) for 12 h was used as positive control for apoptosis. (I) SA-bgal assay using the fluorogenic bgalactosidase substrate C12FDG and flow cytometry (upper panel) or conventional SA-bgal (bottom panel). Eight thousand cells/well were plated in 6-well plates and were infected following day with HRSV at MOI D 3. At 60 h.p.i., the cells were incubated with Bafilomycin A1 and C12FDG and processed for flow cytometry or conventional SAbgal assay (bottom panel) as described in materials and methods. Magnification: 200X; scale bar: 50 mm.
… 
HRSV-induced DNA damage is reduced with antioxidant treatments (A) Quantification of DNA damage foci containing gH2AFX and TP53BP1 in mock and infected A549 cells (48 h.p.i. MOI D 3), treated or not with N-Acetylcysteine (NAC). Cells were treated with 5 mM NAC 90 min before the infection and then after virus adsorption until the end of the experiment. (B) Virus titers in supernatants of A549 infected cells (48 h.p.i. MOI D 3) treated or not with 5 mM NAC. (C) Representative images of DNA damage foci of mock and infected A549 cells (48 h.p.i. MOI D 3), treated or not with N-Acetylcysteine (NAC). Magnification: 600X; scale bar: 10 mm. Arrowheads: DD foci. (D) Increase of intracellular reduced glutathione (GSH) in mock and infected A549 cells (24 h.p.i. MOI D 3) upon treatment with 2,5 mM GSH ethyl ester (GSHee). (E) Quantification of DNA damage foci containing gH2AFX and TP53BP1 in mock and infected A549 cells (48 h.p.i. MOI D 3) treated or not with GSHee. Cells were treated with GSHee 90 min before the infection and then after virus adsorption until the end of the experiment. Virus titers in supernatants of A549 infected cells (48 h.p.i. MOI D 3) treated or not with 2,5 mM GSHee (right panel). (F) Representative images of DNA damage foci in mock and infected A549 cells (48 h.p.i. MOI D 3), treated or not with GSHee. Magnification: 600X; scale bar: 10 mm. (G) Left panel: Expression of transfected catalase (flagged-catalase, in green) and DD foci (TP53BP1 in red) in A549 cells infected with HRSV (48 h.p.i. MOI D 3) and transfected with the plasmid pCMV3-CAT-N-FLAG. Cells labeled as "b" and "c" show catalase overexpression and the absence of DD foci; syncytium labeled as "a" shows the absence of significant catalase overexpression and the presence of nuclei harboring numerous DD foci. Right panel: expression of DD foci (TP53BP1) and HRSV-F glycoprotein (green) in A549 infected cells (48 h.p.i. MOI D 3) Magnification 600X, Scale bar: 10 mM. (h) Left: panel: Quantification of DD foci in A549 cells infected with HRSV (48 h.p.i. MOI D 3) and transfected with the plasmid pCMV3-CT-N-FLAG. CAT ¡ : cells without significant catalase overexpression, CAT C : cells overexpressing catalase. Right panel: virus titers of culture supernatants of A549 cells infected with HRSV (48 h.p.i. MOI D 3) and transfected with either empty vector as a control or catalase (CAT) plasmid. Bars represent the mean § SD of two experimenst; n D 3 replicates. DNA damage foci were counted from >150 cells for each experimental condition.
… 
Content may be subject to copyright.
RESEARCH PAPER
Induction of DNA double-strand breaks and cellular senescence by human
respiratory syncytial virus
Isidoro Mart
ınez
a
,
y
, Ver
onica Garc
ıa-Carpizo
b
,
y
, Trinidad Guijarro
a
, Ana Garc
ıa-Gomez
a
, Diego Navarro
b
,
Ana Aranda
b
, and Alberto Zambrano
a
a
Department of Molecular Pathology, Spanish National Center for Microbiology, Institute of Health Carlos III, Madrid, Spain;
b
Department of
Endocrine Physiopathology and Nervous System, Institute for Biomedical Research (IIBM), CSIC-UAM, Madrid, Spain
ARTICLE HISTORY
Received 12 August 2015
Revised 11 January 2016
Accepted 14 January 2016
ABSTRACT
Human respiratory syncytial virus (HRSV) accounts for the majority of lower respiratory tract
infections during infancy and childhood and is associated with signicant morbidity and mortality.
HRSV provokes a proliferation arrest and characteristic syncytia in cellular systems such as
immortalized epithelial cells. We show here that HRSV induces the expression of DNA damage
markers and proliferation arrest such as P-TP53, P-ATM, CDKN1A and gH2AFX in cultured cells
secondary to the production of mitochondrial reactive oxygen species (ROS). The DNA damage foci
contained gH2AFX and TP53BP1, indicative of double-strand breaks (DSBs) and could be reversed
by antioxidant treatments such as N-Acetylcysteine (NAC) or reduced glutathione ethyl ester
(GSHee). The damage observed is associated with the accumulation of senescent cells, displaying a
canonical senescent phenotype in both mononuclear cells and syncytia. In addition, we show signs
of DNA damage and aging such as gH2AFX and CDKN2A expression in the respiratory epithelia of
infected mice long after viral clearance. Altogether, these results show that HRSV triggers a DNA
damage-mediated cellular senescence program probably mediated by oxidative stress. The results
also suggest that this program might contribute to the physiopathology of the infection, tissue
remodeling and aging, and might be associated to long-term consequences of HRSV infections.
KEYWORDS
cellular senescence; DNA
damage; human respiratory;
ROS; syncytial virus
Introduction
HRSV is an enveloped virus that belongs to the Pneumo-
virus genus of the Paramixoviridae family.
5,14
HRSV rep-
licates in airway epithelial cells activating a variety of
mediators involved in lung immune/inammatory
responses such as proinammatory cytokines, chemo-
kines, interferons and adhesion molecules.
24,26
The gene
expression of these mediators is regulated by key tran-
scription factors including nuclear factor-kB (NF-kB),
the signal transducers and activators of transcription
(STATs) and interferon regulatory factors (IRFs).
Through its action on the signaling pathways implicated
in the activation of these transcription factors, reactive
oxygen species (ROS) formation can regulate the expres-
sion of the immune/inammatory responses.
1,24
Exces-
sive ROS production, however, can lead to oxidative
stress and cause severe damage to cells. Thus, oxidative
stress has been increasingly recognized as a contributing
factor in aging and in multiple pathologies including
lung inammatory diseases.
38,51
HRSV infection is
accompanied by the induction of ROS whose production
by the pulmonary epithelial and endothelial cells is
involved in the activation of transcription factors, oxida-
tive stress and lung damage in infected cells in both ani-
mals and children.
9,10,24,30,31,32,37
Compelling evidence
indicate that antioxidants treatments inhibit both viral
HRSV replication and the activation of NF-kB, STATs
and IRFs.
9,31,37,43
Moreover, antioxidants reduce HRSV-
induced oxidative stress and clinical disease in a mouse
model of infection suggesting a causal relationship
between increased ROS production and lung disease.
10
DNA double-strand breaks (DSBs) may arise from a
variety of exogenous and endogenous sources and in
general by cell stress coming from any deviation from
normal physiological conditions.
4,47,55
One of the most
CONTACT Alberto Zambrano azambra@isciii.es; Department of Molecular Pathology, Spanish National Center for Microbiology, Institute of Health
Carlos III, Ctra. Majadahonda-Pozuelo Km 2, Majadahonda, Madrid 28220, Spain; Alberto Zambrano aaranda@iib.uam.es Instituto de Investigaciones
Biomédicas Alberto SolsArturo Duperier, 4, Madrid-28029, Spain
Color versions of one or more of the gures in this article can be found online at www.tandfonline.com/kvir.
y
These authors contributed equally to this work.
Supplemental data for this article can be accessed on the publishers website.
© 2016 Taylor & Francis
VIRULENCE
2016, VOL. 7, NO. 4, 427442
http://dx.doi.org/10.1080/21505594.2016.1144001
important sources of DSBs is the generation of ROS in
cells. ROS are produced primarily by the mitochondria
and can induce base oxidation, abasic sites, and even
both single and double-DNA strand breaks.
7,16,20,66
A
signicant number of the DNA single-stranded lesions
induced by ROS are converted in DSBs, either by a direct
mechanism or by the repair process itself.
63,72
A compel-
ling amount of evidence has strengthened the notion
that oxidative stress can trigger cellular senescence, a cel-
lular state characterized primarily by an irreversible
proliferative arrest. Besides the absence of proliferation,
senescent cells display a number of morphological
changes and markers that in aggregate dene the senes-
cent phenotype. These include the expression of a senes-
cence-associated b-galactosidase activity (SA-bgal),
expression of tumor suppressors and cell cycle inhibitors,
secretion of diverse cytokines and often also the presence
of DNA damage lesions phenotype.
8,15,18,61
There are a
number of stimuli that induce senescence that ultimately
converge in the activation of TP53 and of key cyclin-
dependent kinase (CDK) inhibitors such as CDKN2A
(p16), CDKN2B (p15), CDKN1A (p21) and CDKN1B
(p27). These mediators trigger a proliferation arrest and
the hypo-phosphorylation of the tumor suppressor RB1
that implements the cellular senescence program.
53
Senescence as well as apoptosis has emerged as physio-
logical mechanisms to prevent proliferation of damaged
cells. However, recent evidence has expanded its role
beyond DNA damage or stress. For instance, senescence
may occur during development
52,53,70
(developmental
senescence) and as a physiological program in adult cells,
such as normal megakaryocytes and placenta syncytio-
trophoblasts,
73
in association with DNA damage
markers, TP53, CDKN2A and CDKN1A.
HRSV induces a proliferation arrest by activating
transforming growth factor band increasing the abun-
dance of cell-cycle regulatory molecules involved in
G
0
/G
1
phase control.
3,25,74
In addition, the infection
courses with delayed low levels of apoptosis due to differ-
ent antiapoptotic (resistance) mechanisms.
12,19,42,49,50
This evidence together with the fact that HRSV is able to
activate the DNA damage response (DDR)
21
led us to
investigate the role of HRSV in DNA damage and cellu-
lar senescence.
We show here the occurrence of cellular senes-
cence during the infection of HRSV. HRSV induces
the expression of DNA damage markers and prolifer-
ation arrest such as P-TP53, P-ATM, CDKN1A and
gH2AFX (H2A histone family member X, phosphory-
lated on Ser 139) in cultured cells secondary to the
production of mitochondrial ROS. The DNA damage
foci contained gH2AFX and TP53BP1, indicative of
DSBs and could be reversed by antioxidants
treatments such as N-Acetylcysteine (NAC) or
reduced glutathione ethyl ester (GSHee). The damage
observed is associated with the accumulation of senes-
cent cells, displaying all the hallmarks of the senes-
cence phenotype in both mononuclear cells and
syncytia. In addition, we show signs of DNA damage
and aging such as gH2AFX and CDKN2A expression
in the lungs of infected mice at different times post-
infection. Altogether, these results show that oxidative
stress induced by HRSV triggers a DNA damage-
mediated cellular senescence program. The results
also suggest that the DNA damage inicted and the
senescence program might contribute to the physio-
pathology of the infection, tissue remodeling and
aging, and might be associated to long-term conse-
quences of HRSV infections.
Results
HRSV induces cellular senescence in cultured cells
We observed that the infection of A549 and HEp-2 cells
by HRSV induces the accumulation of senescent cells
positive for SA-bgal activity. This activity could also be
detected in spontaneous syncytia of both immortalized
cell lines (»2040%), but was greatly magnied in both
mononuclear cells and syncytia upon HRSV infection
(Fig. 1a-d and Fig. S1a-d). The absence of signicant
levels of SA-bgal in the majority of the spontaneous
syncytia suggests that besides the fusion process there is
aspecic mechanism responsible for the SA-bgal activ-
ity associated to the infection of these immortalized
cells. We performed the infections 24 h after seeding
cells at two densities, 5200 cells/cm
2
(Fig. 1a-d)and
31200 cells/cm
2
(Fig. S1a-b) These cellular densities
correspond approximately to 18% and 33% of the typi-
cal density used in conventional infections (93750 cells/
cm
2
) and prevent the typical SA-bgal background due
to cell over conuence. Under these conditions, expo-
sure of A549 cells to HRSV produces a drastic increase
in the levels of senescence, being higher at cellular den-
sities that favor virus dissemination (31200 cells/cm
2
).
This effect was magnied when HEp-2 cells were used
(Fig. 1c,d and Fig. S1b,c). This cell line of laryngeal ori-
gin, the cell line of choice for virus production, shows
high levels of chromosomal instability and is very per-
missive for HRSV replication. Due to the high levels of
senescence found upon HRSV infection, we used an
assay consisting of a combination of SA-bgal activity
and immunocytochemical staining. This technique
termed here Immuno-SA-bgal allows the detection of
senescent cells expressing high levels of the antigen of
choice, in this case, HRSV antigens. As shown in
428 I. MART
INEZ ET AL.
Figure 1. Occurrence of cellular senescence in cells infected with HRSV. (A) Occurrence of cellular senescence during HRSV lytic infection
of A549 cells seeded at 5200 cells/cm
2
. The cells were processed for SA-bgal assay at 48 h.p.i. (MOI D3). Magnication: 200X, scale bar:
50 mm. (B) Quantication of SA-bgal expression in A549 cells (in total population and syncytia). (C) and (D) Similar assays in HEp-2 cells.
(E) Immuno-SA-bgal, an assay that allows the detection of SA-bgal expression in massively infected HEp-2 cells stained with a-HRSV
antibodies (red staining). Magnication: 200X; scale bar: 50 mm. (F) Occurrence and quantication of cellular senescence in a culture of
HEp-2 cells persistently infected by HRSV. Magnication: 200X; scale bar: 50 mm. (G) Quantication of IL-6 and TNF-ain supernatants of
cultured cells. Bars in the graphs represent the mean §SD of 23 experiments. n D3 replicates. (H) Analysis of apoptosis/necrosis. Eight
thousand cells/well were plated in 6-well plates and were infected with HRSV the following day at MOI D3. At 60 h.p.i., the cells were
processed for ow cytometry. After treatment with the uorescent probes (AnnexinV-AlexaFluor488TM and SytoxTM) apoptotic cells
show green uorescence, dead cells show brighter green uorescence, and live cells show little. These populations were distinguished
in the FL1 channel of a FACSCalibur ow cytometer. Treatment of cells with 20 mM camptothecin (CPT) for 12 h was used as positive
control for apoptosis. (I) SA-bgal assay using the uorogenic bgalactosidase substrate C12FDG and ow cytometry (upper panel) or con-
ventional SA-bgal (bottom panel). Eight thousand cells/well were plated in 6-well plates and were infected following day with HRSV at
MOI D3. At 60 h.p.i., the cells were incubated with Balomycin A1 and C12FDG and processed for ow cytometry or conventional SA-
bgal assay (bottom panel) as described in materials and methods. Magnication: 200X; scale bar: 50 mm.
VIRULENCE 429
Fig. 1e, nearly 100% of the cells were infected (red
staining) and a signicant proportion of them (approxi-
mately 70%) were also positive for SA-bgal. Although
to a lesser extent than in lytic infections of HEp-2 cells,
signicant levels of senescence could be detected in
HEp-2 cells persistently infected with HRSV, i.e., cells
that survived a lytic infection and were grown to obtain
a culture that can be propagated indenitely (Fig. 1f).
These cells exhibited a constant virus production for
more than twenty passages and a high heterogeneity
regarding the expression of viral antigens, ranging from
high to undetectable levels.
42
Thus, the coexistence of
uninfected and infected cells in these complex cultures
may account for the high levels of senescence observed
compared to uninfected HEp-2 cells.
In addition to being at and bigger than their normal
counterparts, senescent cells display a senescence-associ-
ated secretory phenotype (SASP), characterized by
expression of proinammatory cytokines, growth factors
and proteases. HRSV induces a rapid activation of sev-
eral mediators involved in the immune/inammatory
responses. Strikingly, the broad array of proinamma-
tory cytokines induced by HRSV resembles the expressed
SASP by canonical senescent cells.
5,14,60
As reported by
others, we detected signicantly higher levels of IL-6 and
TNF-ain supernatants of infected cells when compared
with mock-infected cells (Fig. 1g). In order to know
whether cellular senescence can be associated to cell sur-
vival, we assessed the levels of apoptosis and senescence
after 60 h.p.i. The levels of apoptosis were determined
with the Annexin V/Sytox assay. This assay allows the
detection of live cells (low green uorescence), apoptosis
(moderate green uorescence due to Annexin V binding)
and necrosis (intense green uorescence due to the Sytox
green dye which is impermeable to live cells). The levels
of apoptosis were similar for mock-infected and infected
cells (4% approximately) and relatively low compared to
the levels found when cells were treated with camptothe-
cin, a known inducer of apoptosis (Fig. 1h). On the other
hand, the majority of cells that survived a lytic infection
showed higher intensities of C12FDG uorescence
(senescence) as determined by ow cytometry analysis
using this uorogenic b-galactosidase substrate (Fig. 1i,
upper panel). Cells showing FL1-H intensities over value
10 represented less than 4% of the control cultures, how-
ever, this proportion increased to 30 in the case of
infected cultures. Finally, approximately 60% of cells that
survived a lytic infection were senescent as determined
by the conventional SA-bgal assay (Fig. 1i, bottom
panel). Together, these ndings suggest that cellular
senescence is a biological program associated to HRSV
infections and might be associated to cell survival.
HRSV-induced cellular senescence is associated
to DNA damage
As senescence is primarily a mechanism to prevent pro-
liferation of damaged cells, we reasoned that the senes-
cence signs observed in virus-infected cells could be
associated to DNA damage. Of the different types of
DNA lesions, DSBs are the most deleterious, being
potent inducers of chromosomal rearrangements such as
deletions, translocations or amplications.
67,75
The phos-
phorylated histone H2AFX (gH2AFX) and TP53BP1 are
accurate markers of DNA damage.
2,56,62,64,65
When DSBs
are detected, a signaling cascade is initiated by the phos-
phorylation of H2AFX by Ataxia-Telangiectasia Mutated
kinase (ATM) near the break site, followed by the rapid
recruitment of TP53BP1 on the chromatin surrounding
the DNA lesion. TP53BP1, among other functions, acts
as a molecular scaffold in damaged chromatin during
non-homologous end-joining (NHEJ) repair mecha-
nism.
6,56
We analyzed the expression of DNA damage
(DD) foci containing gH2AFX and TP53BP1 (Fig. 2a,b).
First, we examined their confocal colocalization in A549
cells. As extensively reported before in other cellular sys-
tems, we corroborated their signicant colocalization
and observed an increase of DD foci in infected-A549
cells, in both mononuclear cells and syncytia (Fig. 2a).
Next, we performed conventional microscopy to analyze
and quantify the levels of DD in the same cells and in
HEp-2 cells (Fig. 2b, Fig. S1e, f). We observed a signi-
cant increase of cells containing DD foci and cells con-
taining numerous foci (>5/nucleus) after virus infection.
This effect was exacerbated in the HEp-2 cell line
(Fig. S1e, f). We further examined various markers of the
DNA damage response (DDR), proliferation arrest and
cellular senescence in A549 cells. As shown in Figure. 2c,
the infection of A549 cells induces the phosphorylation
and activation of ATM, TP53 and H2AFX, an elevation
in CDKN1A and CDKN2A expression and RB1 hypo-
phosphorylation. Altogether these ndings indicate that
cellular senescence is a common phenomenon during
HRSV infection and is associated with the presence of
DNA damage.
Reactive oxygen species (ROS) generated during
HRSV infection induce DNA damage
Induction of ROS by HRSV has been extensively docu-
mented.
9,10,24,30,31,32,37
As previously reported, we
observed an elevation in the total ROS levels during the
infection of A549 cells by using the widely used probe
DCFH-DA
9,34
(Fig. 3a). As expected, we also observed a
signicant increase in the intracellular amount of
430 I. MART
INEZ ET AL.
oxidized glutahione (GSSG) concomitantly with a
decrease in reduced glutathione (GSH), upon HRSV
infection as previously reported
32,48
(Fig. 3b). As ROS
are produced primarily by the mitochondria we further
examined whether HRSV could trigger the formation of
superoxide (O
2
.
¡
), the main mitochondrial ROS. HRSV
infection induced a drastic increase in superoxide
generation, detected with the specic mitochondrial
probe MitoSOX by immunouorescence and by ow
cytometry (Fig. 3c,d, Fig. S2 a). Therefore, ROS triggered
by HRSV have at least in part a mitochondrial origin. As
ROS are able to induce DSBs and HRSV behaves as a
potent stressor inducing mitochondrial superoxide, we
reasoned that the DD observed could be alleviated by
Figure 2. HRSV induces DNA damage. (A) Analysis of confocal colocalization of the DNA damage markers gH2AFX and TP53BP1 in
HRSV-infected A549 cells (48 h.p.i. MOI D3). Magnication: 600X; scale bar: 10 mm (B) Conventional indirect immunouorescence show-
ing the presence of gH2AFX and TP53BP1 in mock and infected A549 cells (48 h.p.i. MOI D3). Magnication: 600X; scale bar: 10 mm.
Quantication of the DNA damage foci containing gH2AFX and TP53BP1 is shown at the right panel. (C) Detection by western-blotting
of various DNA damage and proliferation arrest markers in mock and infected A549 cells (30 h.p.i. MOI D3: panels P-TP53, CDKN2A, P-
RB1, P-ATM); 48 h.p.i. MOI D3: panels CDKN1A and gH2FAX. KDa: kilodaltons. Bars in the graphs represent the mean §SD of 2 experi-
ments, n D3 replicates. DNA damage foci were counted from >150 cells for each experimental condition. Data from panel c are of a
representative experiment.
VIRULENCE 431
treatments with antioxidants as previously reported.
39,77
We treated cells with N-Acetylcysteine (NAC) and found
a drastic reduction in the number of damaged nuclei and
a concomitant moderate reduction on virus titers
(Fig. 4A-C). This is consistent with previous reports indi-
cating that antioxidant treatment inhibits HRSV infec-
tion
9,31,37,43
and ameliorates clinical disease and
pulmonary inammation in mice.
10,31
Similar results
were obtained when cells were treated with reduced glu-
tathione ethyl ester (GSHee), a permeable source of GSH
(Fig. 4D-F). Treatment of cells with GSHee increased the
intracellular pool of GSH and induced a reduction in
DD foci and virus titers. When GSHee or NAC were also
included during virus adsorption, virus titers were very
strongly reduced (data not shown), indicating that these
antioxidants might have also an effect on virus integrity,
attachment or entry into the cells. Remarkably, neither
of the two treatments altered substantially the low levels
of constitutive DD foci, suggesting that the antioxidants
are only effective against the de novo DD foci induced by
HRSV. Antioxidants treatment also reduced the occur-
rence of senescence in infected cultures (Fig. S2 b), and
the expression or activation of DD markers such as P-
TP53, gH2AFX and CDKN1A (Fig. S2 c). In addition,
the partial depletion of TP53BP1 mediated by small-
interfering RNAs (siRNAs) increased signicatively the
levels of senescence in infected cultures (Fig. S2 d,e)
probably due to an additional increase of unrepaired
DNA damage. In order to determine whether the oxida-
tive stress itself was responsible of the DD observed, we
transfected a plasmid encoding human catalase to HRSV
infected cells and evaluated the presence of DD foci.
Figure 3. HRSV induces mitochondrial reactive oxygen species (ROS). (A) Measurement of total ROS levels of mock and infected A549
cells (48 h.p.i. MOI D3) using the DCFH-DA probe and uorometry. (B) Measurement of reduced (GSH) and oxidized (GSSG) glutathione
of mock and infected A549 cells (24 h.p.i. MOI D3). (C) Evaluation of mitochondrial ROS in mock and infected A549 cells (24 h.p.i. MOI
D3) with MitoSOX and uorescence microscopy. Positive control: cells treated with 100 mM Paraquat. Images of cells treated with Para-
quat, acquired at the same exposure to that of the rest of the samples (equivalent exposure) and at a reduced exposure, are shown.
Magnication: 400X; scale bar: 20 mm. (D) Assessment of mitochondrial ROS levels of mock and infected A549 cells (24 h.p.i. MOI D3)
with MitoSOX and ow cytometry. The left panel shows representative histograms. The mean intensity of MitoSOX uorescence and the
percentage of positive cells are shown at the right panel. Bars represent the mean §SD of two experiments, ANOVA of data D:
P<0,0001; n D3 replicates.
432 I. MART
INEZ ET AL.
Figure 4. HRSV-induced DNA damage is reduced with antioxidant treatments (A) Quantication of DNA damage foci containing gH2AFX
and TP53BP1 in mock and infected A549 cells (48 h.p.i. MOI D3), treated or not with N-Acetylcysteine (NAC). Cells were treated with
5 mM NAC 90 min before the infection and then after virus adsorption until the end of the experiment. (B) Virus titers in supernatants
of A549 infected cells (48 h.p.i. MOI D3) treated or not with 5 mM NAC. (C) Representative images of DNA damage foci of mock and
infected A549 cells (48 h.p.i. MOI D3), treated or not with N-Acetylcysteine (NAC). Magnication: 600X; scale bar: 10 mm. Arrowheads:
DD foci. (D) Increase of intracellular reduced glutathione (GSH) in mock and infected A549 cells (24 h.p.i. MOI D3) upon treatment with
2,5 mM GSH ethyl ester (GSHee). (E) Quantication of DNA damage foci containing gH2AFX and TP53BP1 in mock and infected A549
cells (48 h.p.i. MOI D3) treated or not with GSHee. Cells were treated with GSHee 90 min before the infection and then after virus
adsorption until the end of the experiment. Virus titers in supernatants of A549 infected cells (48 h.p.i. MOI D3) treated or not with
2,5 mM GSHee (right panel). (F) Representative images of DNA damage foci in mock and infected A549 cells (48 h.p.i. MOI D3), treated
or not with GSHee. Magnication: 600X; scale bar: 10 mm. (G) Left panel: Expression of transfected catalase (agged-catalase, in green)
and DD foci (TP53BP1 in red) in A549 cells infected with HRSV (48 h.p.i. MOI D3) and transfected with the plasmid pCMV3-CAT-N-FLAG.
Cells labeled as band cshow catalase overexpression and the absence of DD foci; syncytium labeled as ashows the absence of sig-
nicant catalase overexpression and the presence of nuclei harboring numerous DD foci. Right panel: expression of DD foci (TP53BP1)
and HRSV-F glycoprotein (green) in A549 infected cells (48 h.p.i. MOI D3) Magnication 600X, Scale bar: 10 mM. (h) Left: panel: Quanti-
cation of DD foci in A549 cells infected with HRSV (48 h.p.i. MOI D3) and transfected with the plasmid pCMV3-CT-N-FLAG. CAT
¡
: cells
without signicant catalase overexpression, CAT
C
: cells overexpressing catalase. Right panel: virus titers of culture supernatants of A549
cells infected with HRSV (48 h.p.i. MOI D3) and transfected with either empty vector as a control or catalase (CAT) plasmid. Bars repre-
sent the mean §SD of two experimenst; n D3 replicates. DNA damage foci were counted from >150 cells for each experimental
condition.
VIRULENCE 433
Catalase has a preeminent role in protecting cells from
oxidative damage caused by ROS.
36,76
Catalase calalyzes
the decomposition of H
2
O
2
to H
2
O and O
2
.H
2
O
2
is a
harmful byproduct of many normal metabolic processes
including the reaction of superoxide dismutase (SOD)
enzyme that converts superoxide radical into O
2
and
H
2
O. As shown in Figure 4g (cells labeled as band c
in the picture on the left), the overexpression of catalase
reduced the occurrence of DD foci while cells with
apparent signs of infection (syncytium labeled as ain
Fig. 4g) showed numerous DD foci similar to those typi-
cal syncytia stained with HRSV-F glycoprotein (Fig. 4g,
picture on the right). The analysis of the DD foci and the
expression of catalase in the infected/transfected popula-
tion indicated that the expression of DD foci was signi-
catively reduced in cells overexpressing catalase (Fig. 4h,
CAT
C
cells, left panel). In contrast to antioxidant treat-
ments, transfection of the plasmid encoding catalase did
not alter signicantly virus titers compared to cells
infected and transfected with the empty vector (control
culture) (Fig. 4h, right panel). These results suggest that
oxidative stress is directly responsible of the damage
observed and link the occurrence of DD damage with
cellular senescence. The physical presence of DNA
breaks in infected cells was also directly detected by
using the terminal deoxynucleotidyl transferase (TdT)
enzyme,
77
(Fig. S2f). We also assessed the repair capabil-
ity of infected cells after irradiation with g-rays (Fig. S3
a-d). Due to the progression of the infection and the sub-
sequent accumulation of de novo foci, the DD load was
still very high compared to control cells. Analysis of the
DDR at a single time suggested that HRSV does not
affect signicantly the early DDR signaling (Fig. S3e, f).
Thus, the DD inicted by HRSV is most likely due to
oxidative stress and does not appear to be caused by a
gross defect in cellular DSBs repair capacity.
Signs of DNA damage and aging in lungs of young
mice infected with HRSV
Mice were either mock infected (n D5) or infected with
HRSV (strains Long and A2, n D6) and the lungs were
analyzed by a highly sensitive multiple immunouores-
cence assay to detect both gH2AFX, a marker of DD and
aging,
44
and HRSV antigens. We found no signicant
differences in DD expression and viral antigens between
the two strains used. As shown in Figure 5b and Fig. S3g,
we observed signs of severely damaged DNA in 23%§2
of HRSV-infected cells, in »30% of the columnar epithe-
lia of bronchioles (CSCE: ciliated simple columnar epi-
thelium). Moderate DNA damage, 1 or 2 foci/nucleus
approximately, could be also detected in »30% of the
alveolar epithelium of infected mice (26%§5 of alveolar
cells with signicant reactivity to HRSV antigens showed
DD foci)(data not shown). In contrast, no signicant
gH2AFX reactivity was detected in the lungs of mock-
infected cells (Fig. 5A,C). We further evaluated the
expression of both gH2AFX and CDKN2A by conven-
tional immunohistochemistry at different times post
infection (4, 11 and 30 days). HRSV-infected mice nor-
mally show a viral replication that peaks at days 45 and
becomes undetectable by day 7.
33
As expected, we did
not observe reactivity to HRSV antigens at days 11 and
30 post infection in the lungs of infected mice (data not
shown), however, we could still detect signs of DNA
damage and senescence in the respiratory epitelium of
some bronchioles (»18%) at those times post infection
(Fig. 5E, F). These results suggest the occurrence of DNA
damage and senescence long time after a primary
infection.
Discussion
We show for the rst time the occurrence of cellular
senescence in cultured immortalized epithelial cells
infected with HRSV. These infected cells show the
canonical cellular senescence phenotype, including the
SA-bgal activity, components of the SASP phenotype
and the presence of accurate markers of DNA damage
and proliferation arrest such as the phosphorylation and
activation of ATM, TP53 and H2AFX, an elevation in
CDKN1A and CDKN2A expression and RB1 hypo-
phosphorylation. SA-bgal activity is evident at 24 h.p.i.
(data not shown) but increased progressively with time
and under conditions that favor virus dissemination
(31200 cells/cm
2
), although it could be also detected in
severely damaged mononuclear infected cells. Besides
HRSV, fusion of cells may be also triggered by patho-
genic viruses such as HIV, ERVWE1 virus, Measles, and
Inuenza.
11,28,35,46,69
This fusogenic property seems to be
a mechanism to support virus dissemination and repro-
duction and has been associated to senescence in some
cases.
11
In the case of HRSV and the immortalized con-
text of the cellular systems used, it seems that cellular
senescence is driven by the DNA damage induced by
HRSV rather than the cell fusion process itself as signs of
cellular senescence can be detected in spontaneous syncy-
tia and in mononuclear infected cells.
As recently reported by others,
21
HRSV is able to acti-
vate the DDR. Here we show evidence indicating that
the DNA damage inicted by HRSV are DSBs. DSBs
observed are primarily of oxidative nature, probably due
to ROS generation. Several reports have previously
reported that HRSV induces ROS expression by different
mechanisms including nicotinamide adenine dinucleo-
tide phosphate (NADPH)-oxidase dependent
434 I. MART
INEZ ET AL.
mechanisms and inhibition of antioxidant enzymes.
22,24
According to the results obtained from the use of the
mitochondrial superoxide indicator MitoSOX, ROS for-
mation by HRSV has at least in part a mitochondrial ori-
gin. ROS are involved in transcription factor activation
and chemokine gene expression, oxidative stress and
lung damage in infected cells in both animals and chil-
dren.
9,10,30,32,37
Antioxidants treatments inhibit viral
HRSV replication and block the activation of
transcription factors implicated in the regulation of
immune/inammatory responses.
9,31,37,43
In addition,
antioxidants reduce HRSV-induced oxidative stress and
clinical symptoms in mice suggesting a link between
increased ROS production and lung disease.
10
We show
that damage inicted by HRSV can be alleviated to a
great extent by antioxidants that also inhibited moder-
ately virus production and reversed the reduced glutathi-
one (GSH)/oxidized GSSG ratio as reported before.
32,48
Antioxidant treatments concomitantly reduced the levels
of cellular senescence induced by HRSV and the activa-
tion of DD markers in cell cultures suggesting that the
cellular senescence observed is mainly triggered by DNA
damage. According to this, the ectopic expression of the
enzyme catalase, that protect cells from oxidative damage
from ROS, reduced the expression of DD foci in infected
cells without altering virus production, in contrast to
antioxidant treatments, suggesting a direct role of the
oxidative stress in the damage observed. The DD
inicted does not seems to be a consequence of a gross
deciency in the cellular DD repair capabilities and the
downregulation of one of the major components of DSB
repair, TP53BP1, slightly increased the levels of senes-
cence induced by HRSV probably due to increased unre-
paired damage. These results suggested a direct link
between DNA damage and the occurrence of senescence.
We have also shown the presence of two accurate bio-
markers of DNA damage and aging in HRSV-infected
lungs of young mice, gH2AFX and CDKN2A. HRSV
infection was evident in some distal and terminal airways
as well as in the respiratory epithelium. A signicant
number of gH2AFX foci were found in some infected
cells lining the terminal airways. However only 12foci/
Figure 5. Expression of markers of DNA damage and aging in
lungs of mice infected by HRSV. (A) Epithelial lung tissue from
mock-infected mice stained with antibodies: aHRSV (pool of anti-
bodies against HRSV) and gH2AFX. (B) Representative image of
multiple immunouorescence labeling with aHRSV and gH2AFX
antibodies of lung tissue from HRSV-infected mice stained. (C),
(D) Representative images of negative controls (no primary) of
the multiple immunouorescence labeling of epithelial lung tis-
sue. Magnications: 600X; scale bar: 10 mm. Arrowheads signal
cells showing reactivity to gH2AFX. AE: alveolar epithelium, Br:
bronchiole, CSCE: ciliated simple columnar epithelium. (E) Repre-
sentative images of conventional immunohistochemistry of
g-H2AFX and CDKN2A on mice lung tissue (mock-infected or
infected with HRSV) at 4 days post-infection. (F) Representative
images of conventional immunohistochemistry of gH2AFX and
CDKN2A on mice lung tissue at 11 and 30 days post-infection.
Arrowheads signal cells showing reactivity to gH2AFX and
CDKN2A. Two experiments, n D5 (mock-infected) or 6 (infected)
replicates (mice) per condition.
VIRULENCE 435
nucleus were observed in infected cells of the alveolar
elds (data not shown). Distal airways of the mouse are
lined with a simple columnar epithelium containing cili-
ated and secretory cells and are an important site for
HRSV replication. The staining of HRSV antigens found
was similar to that observed by others in the human and
mouse airway columnar epithelium.
13,24
gH2AFX and
CDKN2A expression was still evident at 11 and 30 days
post infection indicating a persistence of the primary
damage induced by HRSV during the acute infection.
We found that CDKN2A expression pattern was diffuse
nuclear and cytoplasmic. Although nuclear expression of
CDKN2A is usually associated to cell-cycle control, cyto-
plasmic accumulation has also been associated to senes-
cent phenotype induced by stress.
68
Histological studies suggest that non-ciliated secre-
tory cells give rise to ciliated cells and function as a
long-term self-renewing stem cell population in the
intrapulmonary airways.
59
In addition, it has been
reported that the adult mouse lung contains a minor
population of multipotent epithelial stem cells with the
capacity of self-renewal and to give rise to all epithelial
progenitors that reside in discrete microenviromental
niches along the proximal-distal axis.
45
It is tempting
to speculate that such stem cells could be targets of
HRSV. Indeed, it has been recently reported that HRSV
can infect proliferating airway basal cells and alter cel-
lular composition of the epithelium contributing to air-
way epithelial remodeling.
58
Thus, the DNA damage
inicted and the occurrence of senescence might inu-
ence events long time after the virus has cleared from
thelungbymechanismsbasedonakindofhit-and-
runphenomenon that has been proposed to explain
how transient infections can cause long term airway
disease.
29
Our ndings might have substantial implications for
the pathophysiology of HRSV, the leading cause of bron-
chiolitis and pneumonia in infants and young chil-
dren.
27,54
During the acute infection, the SASP might
contribute to alert the immune system about the damage
inicted. The SASP components would mediate this pro-
cess, facilitating the attraction of immune cells to the
infection sites and the further clearance of infected cells.
Finally, the SASP and cellular senescence may serve as
therapeutic targets.
53,57,71
Although it is too premature to
determine whether cellular senescence would be positive
or detrimental for the pathological consequences of
HRSV infection, current evidence indicates that both
prosenescent and antisenescent therapeutic approaches
can be useful depending on the context. To conrm this
hypothesis further investigations in human primary cells
and in more permissive animal models supporting
HRSV infections are needed.
Methods
Cell culture, viruses, mice and transfections
HEp-2 and A549 cells (ATCC) were maintained in DMEM
medium supplemented with 10% FBS (Biowest), 2 mM
glutamine and 100 U/ml of penicillin and streptomycin
(Lonza). The Long and A2 strains of HRSV were propa-
gated in HEp-2 cells in medium supplemented with 2%
FBS, glutamine and antibiotics. Viruses were puried and
titrated in HEp-2 cells as previously described.
41
All experi-
ments were performed with the Long strain except for the
in vivo experiments in which the A2 strain was also used.
Two-month-old female mice (BALB/c strain) were intra-
nasally infected with 3,3£10
7
pfus of puried virus. At 4,
11 and 30 days post-infection, mice were sacried by cervi-
cal dislocation and the lungs were processed for immuno-
histochemistry. All experiments were performed following
the regulations of the Instituto Carlos III for animal care
and handling. HEp-2 persistently infected by HRSV (L39)
were described elsewhere.
42
N-Acetylcysteine (NAC,
Sigma) and reduced glutathione reduced ethyl ester
(GSHee, Sigma) were prepared as indicated by the manu-
facturers and used at 5 mM and 2,5 mM, respectively.
Tansfections of siRNAs were performed with Lipofect-
amine RNAimax (cat.#13778, Invitrogen) following manu-
facturers instructions. Briey, cells were transfected with
siRNAs (control or TP53BP1 duplexes, sc-37007 and sc-
37455, respectively, Santa Cruz Biotechnology) and the fol-
lowing day infected at MOI 3 with HRSV. After 48 h.p.i.,
the cells were processed for western-blotting and SA-bgal
assays. Transitory transfections of the plasmid pCMV3-
CT-N-FLAG (HG12084-NF, Sino Biological Inc.) express-
ing human catalase or empty vector (pCMV) were per-
formed with Lipofectamine 2000 (Invitrogen, cat. 11668)
in 8-well chamber slides following manufacturers instruc-
tions. Cells were transfected and the following day infected
at MOI 3 with HRSV. After 48 h.p.i., culture supernatants
were collected for virus titration and the cells were proc-
essed for immunouorescence.
Immunouorescence
For immunouorescence, cells were seeded in 8-well
chambers (Thermo Fisher Scientic) at a density of
50000 cells/well. The following day the cells were
infected or treated as indicated in the corresponding
experiments. Immunouorescence was performed as
previously described.
77
Basically, cells were xed in
2% PFA in PBS for 10 min at RT and permeabilized
with 0.1% Triton X-100 and 0.1% sodium citrate
(5 min at RT). Preparations were then washed with
PBS and washing solution (PBS/0.25% BSA/0.1%
Tween 20), blocked for 30 min with blocking
436 I. MART
INEZ ET AL.
solution (washing solution, 2.5% BSA, and 5% nor-
mal serum), and incubated overnight with antibodies
against gH2AFX (1:1000, cat.# 05636 Millipore),
TP53BP1 (1:1000, cat.# NB-100304, Novus Biologi-
cals), antibody 47F (1:50) directed against the F gly-
coprotein of HRSV and anti-Flag (1:500, cat.#
F1804, Sigma-Aldrich). Preparations were then
washed with washing solution and incubated with
secondary antibodies (AF488, AF546 or AF647, Life
Technologies) for 1 h at RT. Nuclei were counter-
stainedwithDAPI,andsamplesweremountedwith
ProLong Diamond (Life Technologies). Cell images
were captured with a uorescence microscopy (Zeiss
Axio) equipped with a camera (Axiocam MRm) and
AxioVision software. DNA damage foci were counted
from >150 cells for each experimental condition.
Analysis of colocalization was performed with a con-
focal microscope (LEICA DMI 600; 63X/1.4 len),
equipped with the following laser lines: Argon (ve
spectral lines 488 to 514 nm), HeNe (633 nm), diode
violet(405nm)anddiodesolidstate(561nm).
Images were obtained with software LAS AF (Leica).
Protein analysis
Cell monolayers were washed with ice-cold PBS and
lysed in triple-detergent lysis buffer [50 mM Tris-HCl,
pH 8.0, 150 mM NaCl, 0.02% sodium azide, 0.1% SDS,
1% NP-40, 0.5% sodium deoxycholate, 100 mg/ml
PMSF, 2 mg/ml pepstatin, 2 mg/ml aprotinin, 2 mg/ml
leupeptin, and phosphatase inhibitors cocktail 2 and 3
(Sigma-Aldrich)]. SDS-PAGE and immunoblotting were
performed under standard conditions. Basically, samples
in Laemmli buffer (30 mg/lane) were separated through
8% or 12% gels and transferred to nitrocellulose mem-
branes for 90 min at RT in the presence of 20% methanol
and 0,1% SDS. Membranes were blocked with 3% BSA in
PBS-Tween 0,05% (PBST-BSA) and incubated ON at
4C with specic antibodies diluted 1:1000 in PBST-
BSA. Antibodies used were: gH2AFX (05636, Milli-
pore), P-CHEK2 (AF1626, R&D Systems), P-RB1 (sc-
16670-R, Santa Cruz Biotech.), P-TP53 (9286S, Cell Sig-
naling), P-ATM (05740, Millipore), CDKN1A (ab7960,
Abcam), CDKN2A (04239, Millipore), TP53BP1 (cat.#
NB-100304, Novus Biologicals) and TUBA1A (used at
1:2000, clone DM1A, Sigma-Aldrich).
Senescence-associated b-galactosidase assays
(SA-bgal) and Immuno-SA-bgal
Conventional SA-bgal assays were performed as previ-
ously described by Dimri et al.
18
SA-bgal detection by
ow cytometry using Balomycin A1 (cat.#B1793,
Sigma-Aldrich) and the uorogenic bgalactosidase sub-
strate C12FDG (cat.# 11590276, Fisher Scientic) was
performed as described by Debacq-Chainiaux et al.
17
Immuno-SA-bgal is an assay consisting of a combina-
tion of immunocytochemistry and SA-bgal assays. Cells
growing in 6-well plates (MW6) were washed twice
with PBS and xed for 3 minutes with 2% formalde-
hyde and 0,2% glutaraldehyde. Cells were permeabi-
lized for 5 min at RT with 0.1% Triton X-100 and 0.1%
sodium citrate, washed with PBS and blocked with BSA
1% in PBS (30 min at RT). Cells were then incubated
for 45 min at RT with a pool of antibodies directed
against HRSV [021/1G, 021/2G, 47F, 67P].
23,40
The
plates were washed twice in a bath of tap water and
incubated for 45 min at RT with a-mouse-HRP second-
ary (ab97040, Abcam) 1:1000 in PBS-BSA 1%. The
plates were washed with tap water before adding the
peroxidase substrate in ELISA buffer [Citric acid
25 mM/Na
2
HPO
4
50 mM pH 5, 0,033 mg/ml AEC sub-
strate (A6926-100TAB, Sigma), 2 ml/ml (30% H
2
O
2
)].
The plates were maintained in the dark until the
appearance of signal (510min).Afterthat,thesub-
strate was removed, the plates were washed twice with
PBS and the SA-b-gal reaction mixture was added. The
plates were incubated in the dark at 37for 24 hours.
Micrographs were taken in a microscope TS100F
(Nikon) equipped with a digital camera DS-L1 (Nikon).
ROS analyses and quantication of GSSG (oxidized
glutathione) and GSH (reduced glutathione)
Cellular ROS levels were assessed with 20 mM
H
2
DCFDA (20,70dichlorodihydrouorescein diacetate,
Sigma-Aldrich) and a microplate uorometer Synergy
(Biotek) as previously described.
77
For the determination
of mitochondrial ROS (superoxide) we used MitoSOX
Red (Life Technologies), uorescence microscopy and
ow citometry. Treatments of cells were carried-out in
triplicates in 8-well chambers (for microscopy) and in
MW6 plates (for ow cytometry). Positive controls con-
sisted of cells treated with 5 mM MitoSOX for 15 min,
then with 100 mM Paraquat for 30 min followed by an
additional 30 min treatment with 5 mM MitoSOX. The
rest of the samples were treated with 5 mM MitoSOX for
30 min. Blank cells consisted of cells treated with vehicle.
The cells growing in 8-well chambers were xed with
PFA 2% in PBS for 10 min, washed and incubated with
DAPI for 15 minutes. Then, the cells were washed with
PBS, mounted with ProLong Diamond (Life Technolo-
gies) and examined under uorescence microscopy. For
ow cytometry, the cells were trypsinized and counted.
The same number of cells for each condition was xed
with PFA, washed and transferred to FACS tubes. The
VIRULENCE 437
measurements were carried out using FASCanto (BD
Bioscience) cytometer. MitoSOX was excited by a laser at
488 nm and the data collected at the FSC, SSC, 585/
42 nm bandpass lter. Cell debris, represented by distinct
low forward and side scatter, were gated out for analysis.
50000 gated events for each condition were
analyzed with the FACSCanto and Flow Jo software. The
mean intensity of MitoSOX uorescence and the per-
centage of positive cells were calculated. For the determi-
nation of GSSG and GSH we used the EnzyChromTM
GSH/GSSG Assay kit (Bioassay Systems) and followed
manufacturers instructions.
Apoptosis/necrosis by ow cytometry
This assay was performed by using the single channel
dead cells apoptosis kit with Annexin
V-Alexa-uor488TM and SytoxTM green dyes from Fisher
Scientic (cat.# V13240). Eight thousand cells per well
were plated in MW6 and were infected with HRSV at
MOI 3 the following day. Sixty h.p.i., attached cells were
processed for ow cytometry as indicated by the manu-
facturers. Treatment of cells with 20 mM camptothecin
(CPT) for 12 h (cat.#C9911, Sigma-Aldrich) was used as
positive control for apoptosis. After treatment with both
probes, apoptotic cells show green uorescence, dead
cells show brighter green uorescence, and live cells
show little. These populations were distinguished in the
FL1 channel of a FACSCalibur ow cytometer (Becton
Dickinson). Twenty thousand events for each condition
were analyzed with the FACSCalibur and Flow Jo
software.
Immunohistochemistry
Mice were sacriced at 4, 11 and 30 days post-infection
by cervical dislocation and lung pieces (5 £5 mm,
approximately) were dissected, xed in 4% buffered for-
malin and embedded in parafn wax. Multiple antigen
labeling was performed following the double-immuno-
uorescent labeling protocol and reagents provided by
Vector Laboratories (cat. # A-2011, A-2016, SP-2001,
PK-6102). Sequential incubations of the antibodies were
made overnight for the rst antibody (a-HRSV: pool of
Abs 021/1G, 021/2G, 47F, 67P, 1:100) and 3 h at RT for
the second antibody (gH2AFX, 1:400). Antigen retrieval
was performed with citrate buffer (pH 6) using a Micro-
wave Tender Cooker (Nordic Ware) and a 700 W micro-
wave (15 min 700W/15 min 350W). Endogenous
peroxidase activity was inhibited with 0.3% H
2
O
2
in
methanol (25 min). Nuclei staining, mounting, and
microscopy acquisition were performed as for the indi-
rect immunouorescence. Images of Figure 5A-D were
processed by deconvolution in the green channel with
Huygens Essential (Scientic Volume Imaging, Hilver-
sum, the Netherlands) based on the classicmaximum
likelihood estimation method. Conventional immuno-
histochemistry was performed following the labeling
protocol and reagents provided by Vector Laboratories
(PK-6101, PK-6102, SP-2001). Antibodies used were
gH2AFX (1:200 cat.# 05636 Millipore) and CDKN2A
(1:200, cat.# 04239, Millipore). Counterstaining with
nuclear fast, mounting and images acquisition were per-
formed as previously described.
77
Immunoassay
The determination of the concentration of human IL-6
and TNF-afrom cell culture supernatatants was assessed
by the ProcartaPlexTM multiplex Immunoassay kit -Mag-
netic beads (eBioscence) based on the Luminex
R
technol-
ogy. Procarta PlexTM Analyst 1.0 software was used for
analysis.
Irradiation of cells with g-ray
Cells growing in 8-well chambers and MW6 plates were
irradiated with 3 Gy (1.2 Gy/min) in a shelf-shielded J. L.
Shepherd Mark I irradiator with a Cesium-137 source.
After irradiation cells were processed for immunouo-
rescence or immunoblotting at the corresponding times.
DNA breaks detection
To detect DNA breaks, we performed an assay based in
the incorporation of dUTP by Terminal deoxynucleoi-
tidyl Transferase enzyme (TdT) and posterior detection
by immunocytochemistry as previously described.
77
Statistical analysis
Statistical signicance of data was determined by apply-
ing a two-tailed Students t test or analysis of variance
followed by the NewmanKeuls or Bonferroni post-tests
for experiments with more than two experimental
groups. P <0.05 is considered signicant. Signicance of
analysis of variance post-test or the Students t test is
indicated in the gures as
(P <0.05),

(P <0.01) and

(P <0.001). Statistics were calculated with the Prism
5 software (GraphPad Software). The results presented
in the gures are means §SD. Experiments were
repeated at least two times.
438 I. MART
INEZ ET AL.
Ethics statement
All experiments with animals were approved by the
Commitee of Bioethics and Animal Welfare of the Insti-
tuto de Salud Carlos III (le reference: PA 31). Protocols
used followed the guidelines for animal protection
reported by the Spanish national law RD 53/2013.
Abbreviations
CPT Camptothecin
DD DNA damage
DDR DNA damage response
DSB double-strand breaks
GSH reduced glutathione
GSHee reduced glutathione ethyl ester
GSSG oxidized glutathione
h.p.i hours post-infection
MOI multiplicity of infection
MW6 6-well plates
NAC N-Acetylcysteine
ROS reactive oxygen species
siRNA small interfering RNAs
Disclosure of potential conicts of interest
No potential conicts of interest were disclosed.
Acknowledgments
We thank the core facilities of the Instituto de Investigaciones
Biom
edicas de Madrid and of the Centro Nacional de Micro-
biolog
ıa for technical help.
Funding
This work was supported by Grants MPY-1038/14 to Alberto
Zambrano, PI 11/00590 to Isidoro Mart
ınez and RD12/0036/
0030 to Ana Aranda from FIS (Instituto de Salud Carlos III)
and Grant BFU2011-28958, from Ministerio de Econom
ıay
Competitividad to Ana Aranda. The authors have no conict-
ing nancial interests.
Author contributions
IM performed and designed experiments. V G-C performed
and designed experiments. TG, A G-G and DG performed
experiments, AA designed experiments and wrote paper. AZ
performed experiments, designed experiments, wrote paper
and conceived the project.
References
[1] Allen RG, Tresini M. Oxidative stress and gene regula-
tion. Free Radical Biol Med 2000; 28:463-99; http://dx.
doi.org/10.1016/S0891-5849(99)00242-7
[2] Anderson L, Henderson C, Adachi Y. Phosphoryla-
tion and rapid relocalization of 53BP1 to nuclear
foci upon DNA damage. Mol Cell Biol 2001;
21:1719-29; PMID:11238909; http://dx.doi.org/
10.1128/MCB.21.5.1719-1729.2001
[3] Bian T, Gibbs JD, Orvell C, Imani F. Respiratory syncy-
tial virus matrix protein induces lung epithelial cell cycle
arrest through a p53 dependent pathway. PloS One 2012;
7:e38052; PMID:22662266; http://dx.doi.org/10.1371/
journal.pone.0038052
[4] Bonner WM, Redon CE, Dickey JS, Nakamura AJ, Sedel-
nikova OA, Solier S, Pommier Y. GammaH2AX and can-
cer. Nat Rev Cancer 2008; 8:957-67; PMID:19005492;
http://dx.doi.org/10.1038/nrc2523
[5] Borchers AT, Chang C, Gershwin ME, Gershwin LJ.
Respiratory syncytial virusa comprehensive review. Clin
Rev Allergy Immunol 2013; 45:331-79; PMID:23575961;
http://dx.doi.org/10.1007/s12016-013-8368-9
[6] Bunting SF, Callen E, Wong N, Chen HT, Polato F, Gunn
A, Bothmer A, Feldhahn N, Fernandez-Capetillo O, Cao
L, et al. 53BP1 inhibits homologous recombination in
Brca1-decient cells by blocking resection of DNA
breaks. Cell 2010; 141:243-54; PMID:20362325; http://
dx.doi.org/10.1016/j.cell.2010.03.012
[7] Cadet J, Douki T, Ravanat JL. Oxidatively generated
base damage to cellular DNA. Free Radical Biol
Med 2010; 49:9-21; http://dx.doi.org/10.1016/j.
freeradbiomed.2010.03.025
[8] Campisi J, dAdda di Fagagna F. Cellular senescence: when
bad things happen to good cells. Nature Rev Mol Cell Biol
2007; 8:729-40; http://dx.doi.org/10.1038/nrm2233
[9] Casola A, Burger N, Liu T, Jamaluddin M, Brasier
AR, Garofalo RP. Oxidant tone regulates RANTES
gene expression in airway epithelial cells infected with
respiratory syncytial virus. Role in viral-induced inter-
feron regulatory factor activation. J Biol Chem 2001;
276:19715-22; PMID:11259439; http://dx.doi.org/
10.1074/jbc.M101526200
[10] Castro SM, Guerrero-Plata A, Suarez-Real G, Adeg-
boyega PA, Colasurdo GN, Khan AM, Garofalo RP,
Casola A. Antioxidant treatment ameliorates respiratory
syncytial virus-induced disease and lung inammation.
Am J Respiratory Critical Care Med 2006; 174:1361-9;
http://dx.doi.org/10.1164/rccm.200603-319OC
[11] Chuprin A, Gal H, Biron-Shental T, Biran A, Amiel A,
Rozenblatt S, Krizhanovsky V. Cell fusion induced by
ERVWE1 or measles virus causes cellular senescence.
Genes Dev 2013; 27:2356-66; PMID:24186980; http://dx.
doi.org/10.1101/gad.227512.113
[12] Coleman CM, Plant K, Newton S, Hobson L, Whyte MK,
Everard ML. The anti-apoptotic effect of respiratory syn-
cytial virus on human peripheral blood neutrophils is
mediated by a monocyte derived soluble factor. Open
Virol J 2011; 5:114-23; PMID:22046209; http://dx.doi.
org/10.2174/1874357901105010114
[13] Collins PL, Graham BS. Viral and host factors in human
respiratory syncytial virus pathogenesis. J Virol 2008;
82:2040-55; PMID:17928346; http://dx.doi.org/10.1128/
JVI.01625-07
[14] Collins PL, Karron RA. Respiratory Syncytial Virus and
Metapneumovirus. Philadelphia: Lippincott Williams &
Wilkins. 2013
VIRULENCE 439
[15] dAdda di Fagagna F. Living on a break: cellular senescence
as a DNA-damage response. Nat Rev Cancer 2008; 8:512-
22; PMID:18574463; http://dx.doi.org/10.1038/nrc2440
[16] De Zio D, Bordi M, Cecconi F. Oxidative DNA
damage in neurons: implication of ku in neuronal
homeostasis and survival. Int J Cell Biol 2012;
2012:752420; PMID:22737170; http://dx.doi.org/
10.1155/2012/752420
[17] Debacq-Chainix F, Erusalimsky JD, Campisi J, Tous-
saint O. Protocols to detect senescence-associated
b-galactosidase (SA-betagal) activity, a biomarker of
senescent cells in culture and in vivo. Nat Protoc
2009; 4:1798-806; PMID:20010931; http://dx.doi.org/
10.1038/nprot.2009.191
[18] Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley
C, Medrano EE, Linskens M, Rubelj I, Pereira-Smith O,
et al. A biomarker that identies senescent human cells
in culture and in aging skin in vivo. Proc Natl Acad Sci
USA 1995; 92:9363-7; PMID:7568133; http://dx.doi.org/
10.1073/pnas.92.20.9363
[19] Domachowske JB, Bonville CA, Mortelliti AJ, Colella CB,
Kim U, Rosenberg HF. Respiratory syncytial virus infec-
tion induces expression of the anti-apoptosis gene IEX-
1L in human respiratory epithelial cells. J Infect Dis 2000;
181:824-30; PMID:10720500; http://dx.doi.org/10.1086/
315319
[20] Evans MD, Dizdaroglu M, Cooke MS. Oxidative DNA
damage and disease: induction, repair and signicance.
Mutation Res 2004; 567:1-61; PMID:15341901; http://dx.
doi.org/10.1016/j.mrrev.2003.11.001
[21] Fang L, Choudhary S, Tian B, Boldogh I, Yang C, Ivan-
ciuc T, Ma Y, Garofalo RP, Brasier AR. Ataxia telangiec-
tasia mutated kinase mediates NF-kappaB serine 276
phosphorylation and interferon expression via the IRF7-
RIG-I amplication loop in paramyxovirus infection. J
Virol 2015; 89:2628-42; PMID:25520509; http://dx.doi.
org/10.1128/JVI.02458-14
[22] Fink K, Duval A, Martel A, Soucy-Flkner A, Grandvx N.
Dual role of NOX2 in respiratory syncytial virus- and sendai
virus-induced activation of NF-kappaB in airway epithelial
cells. J Immunol 2008; 180:6911-22; PMID:18453612; http://
dx.doi.org/10.4049/jimmunol.180.10.6911
[23] Garcia-Barreno B, Palomo C, Penas C, Delgado T, Perez-
Brena P, Melero JA. Marked differences in the antigenic
structure of human respiratory syncytial virus F and G
glycoproteins. J Virol 1989; 63:925-32; PMID:2463385
[24] Garofalo RP, Kolli D, Casola A. Respiratory syncytial
virus infection: mechanisms of redox control and novel
therapeutic opportunities. Antioxidants Redox Signal
2013; 18:186-217; http://dx.doi.org/10.1089/ars.2011.
4307
[25] Gibbs JD, Ornoff DM, Igo HA, Zeng JY, Imani F. Cell
cycle arrest by transforming growth factor beta1 enhan-
ces replication of respiratory syncytial virus in lung epi-
thelial cells. J Virol 2009; 83:12424-31; PMID:19759128;
http://dx.doi.org/10.1128/JVI.00806-09
[26] Haeberle HA, Takizawa R, Casola A, Brasier AR, Dieter-
ich HJ, Van Rooijen N, Gatalica Z, Garofalo RP. Respira-
tory syncytial virus-induced activation of nuclear factor-
kappaB in the lung involves alveolar macrophages and
toll-like receptor 4-dependent pathways. J Infect Dis
2002; 186:1199-206; PMID:12402188; http://dx.doi.org/
10.1086/344644
[27] Hall CB, Weinberg GA, Iwane MK, Blumkin AK,
Edwards KM, Staat MA, Auinger P, Grifn MR, Poehling
KA, Erdman D, et al. The burden of respiratory syncytial
virus infection in young children. N Eng J Med 2009;
360:588-98; http://dx.doi.org/10.1056/NEJMoa0804877
[28] Harrison SC. Viral membrane fusion. Nat Struct Mol Biol
2008; 15:690-8; PMID:18596815; http://dx.doi.org/
10.1038/nsmb.1456
[29] Holtzman MJ, Shornick LP, Grayson MH, Kim EY,
Tyner JW, Patel AC, Agapov E, Zhang Y. Hit-and-
runeffects of paramyxoviruses as a basis for
chronic respiratory disease. Pediatric Infect Dis J
2004; 23:S235-245; http://dx.doi.org/10.1097/01.
inf.0000144674.24802.c1
[30] Hosakote YM, Jantzi PD, Esham DL, Spratt H, Kurosky
A, Casola A, Garofalo RP. Viral-mediated inhibition of
antioxidant enzymes contributes to the pathogenesis of
severe respiratory syncytial virus bronchiolitis. Am J
Respiratory Critical Care Med 2011; 183:1550-60; http://
dx.doi.org/10.1164/rccm.201010-1755OC
[31] Hosakote YM, Komaravelli N, Mautemps N, Liu T,
Garofalo RP, Casola A. Antioxidant mimetics modu-
late oxidative stress and cellular signaling in airway
epithelial cells infected with respiratory syncytial
virus. Am J Physiol Lung Cell Mol Physiol 2012;
303:L991-1000; PMID:23023968; http://dx.doi.org/
10.1152/ajplung.00192.2012
[32] Hosakote YM, Liu T, Castro SM, Garofalo RP, Casola A.
Respiratory syncytial virus induces oxidative stress by
modulating antioxidant enzymes. Am J Resp Cell Mol Biol
2009; 41:348-57; http://dx.doi.org/10.1165/rcmb.2008-
0330OC
[33] Jafri HS, Chavez-Bueno S, Mejias A, Gomez AM, Rios
AM,NassiSS,YusufM,KapurP,HardyRD,Hateld
J, et al. Respiratory syncytial virus induces pneumo-
nia, cytokine response, airway obstruction, and
chronic inammatory inltrates associated with long-
term airway hyperresponsiveness in mice. J Infect Dis
2004; 189:1856-65; PMID:15122522; http://dx.doi.org/
10.1086/386372
[34] Jamaluddin M, Tian B, Boldogh I, Garofalo RP, Brasier
AR. Respiratory syncytial virus infection induces a reac-
tive oxygen species-MSK1-phospho-Ser-276 RelA path-
way required for cytokine expression. J Virol 2009;
83:10605-15; PMID:19706715; http://dx.doi.org/10.1128/
JVI.01090-09
[35] Jardetzky TS, Lamb RA. Activation of paramyxovirus
membrane fusion and virus entry. Curr Opin Virol 2014;
5:24-33; PMID:24530984; http://dx.doi.org/10.1016/j.
coviro.2014.01.005
[36] Lindau-Shepard BA, Shaffer JB. Expression of human cata-
lase in acatalasemic murine SV-B2 cells confers protection
from oxidative damage. Free Radical Biol Med 1993; 15:581-
8; http://dx.doi.org/10.1016/0891-5849(93)90160-V
[37] Liu T, Castro S, Brasier AR, Jamaluddin M, Garofalo RP,
Casola A. Reactive oxygen species mediate virus-induced
STAT activation: role of tyrosine phosphatases. J Biol
Chem 2004; 279:2461-9; PMID:14578356; http://dx.doi.
org/10.1074/jbc.M307251200
440 I. MART
INEZ ET AL.
[38] MacNee W. Oxidative stress and lung inammation in
airways disease. Eur J Pharmacol 2001; 429:195-207;
PMID:11698041; http://dx.doi.org/10.1016/S0014-2999
(01)01320-6
[39] Mansour HH, Hafez HF, Fahmy NM, HanaN. Protec-
tive effect of N-acetylcysteine against radiation induced
DNA damage and hepatic toxicity in rats. Biochem Phar-
macol 2008; 75:773-80; PMID:18028880; http://dx.doi.
org/10.1016/j.bcp.2007.09.018
[40] Martinez I, Dopazo J, Melero JA. Antigenic structure
of the human respiratory syncytial virus G glycopro-
tein and relevance of hypermutation events for the
generation of antigenic variants. J General Virol 1997;
78 (Pt 10):2419-29; http://dx.doi.org/10.1099/0022-
1317-78-10-2419
[41] Martinez I, Lombardia L, Garcia-Barreno B, Dominguez
O, Melero JA. Distinct gene subsets are induced at differ-
ent time points after human respiratory syncytial virus
infection of A549 cells. J General Virol 2007; 88:570-81;
http://dx.doi.org/10.1099/vir.0.82187-0
[42] Martinez I, Lombardia L, Herranz C, Garcia-Barreno B,
Dominguez O, Melero JA. Cultures of HEp-2 cells persis-
tently infected by human respiratory syncytial virus differ
in chemokine expression and resistance to apoptosis as
compared to lytic infections of the same cell type. Virol-
ogy 2009; 388:31-41; PMID:19345972; http://dx.doi.org/
10.1016/j.virol.2009.03.008
[43] Mata M, Martinez I, Melero JA, Tenor H, Cortijo J.
Roumilast inhibits respiratory syncytial virus infection
in human differentiated bronchial epithelial cells. PloS
One 2013; 8:e69670; PMID:23936072; http://dx.doi.org/
10.1371/journal.pone.0069670
[44] Matheu A, Maraver A, Klatt P, Flores I, Garcia-Cao I,
Borras C, Flores JM, Vina J, Blasco MA, Serrano M.
Delayed ageing through damage protection by the Arf/
p53 pathway. Nature 2007; 448:375-9; PMID:17637672;
http://dx.doi.org/10.1038/nature05949
[45] McQualter JL, Yuen K, Williams B, Bertoncello I. Evi-
dence of an epithelial stem/progenitor cell hierarchy
in the adult mouse lung. Proc Natl Acad Sci U S A
2010; 107:1414-9; PMID:20080639; http://dx.doi.org/
10.1073/pnas.0909207107
[46] Melikyan GB. HIV entry: a game of hide-and-fuse? Curr
Opin Virol 2014; 4:1-7; PMID:24525288; http://dx.doi.
org/10.1016/j.coviro.2013.09.004
[47] Mladenov E, Iliakis G. Induction and repair of DNA
double strand breaks: the increasing spectrum of non-
homologous end joining pathways. Mutation Res
2011; 711:61-72; PMID:21329706; http://dx.doi.org/
10.1016/j.mrfmmm.2011.02.005
[48] Mochizuki H, Todokoro M, Arakawa H. RS virus-
induced inammation and the intracellular glutathione
redox state in cultured human airway epithelial cells.
Inammation 2009; 32:252-64; PMID:19548075; http://
dx.doi.org/10.1007/s10753-009-9128-0
[49] Monick MM, Cameron K, Powers LS, Butler NS, McCoy
D, Mallampalli RK, Hunninghake GW. Sphingosine
kinase mediates activation of extracellular signal-related
kinase and Akt by respiratory syncytial virus. Am J Resp
Cell Mol Biol 2004; 30:844-52; http://dx.doi.org/10.1165/
rcmb.2003-0424OC
[50] Monick MM, Cameron K, Staber J, Powers LS, Yarovin-
sky TO, Koland JG, Hunninghake GW. Activation of the
epidermal growth factor receptor by respiratory syncytial
virus results in increased inammation and delayed apo-
ptosis. J Biol Chem 2005; 280:2147-58; PMID:15542601;
http://dx.doi.org/10.1074/jbc.M408745200
[51] Morcillo EJ, Estrela J, Cortijo J. Oxidative stress and
pulmonary inammation: pharmacological interven-
tion with antioxidants. Pharmacol Res 1999; 40:393-
404; PMID:10527653; http://dx.doi.org/10.1006/
phrs.1999.0549
[52] Munoz-Espin D, Canamero M, Maraver A, Gomez-
Lopez G, Contreras J, Murillo-Cuesta S, Rodriguez-Baeza
A, Varela-Nieto I, Ruberte J, Collado M, et al. Pro-
grammed cell senescence during mammalian embryonic
development. Cell 2013; 155:1104-18; PMID:24238962;
http://dx.doi.org/10.1016/j.cell.2013.10.019
[53] Munoz-Espin D, Serrano M. Cellular senescence: from
physiology to pathology. Nat Rev Mol Cell Biol 2014;
15:482-96; PMID:24954210; http://dx.doi.org/10.1038/
nrm3823
[54] Nair H, Nokes DJ, Gessner BD, Dherani M, Madhi SA,
Singleton RJ, OBrien KL, Roca A, Wright PF, Bruce N,
et al. Global burden of acute lower respiratory infections
due to respiratory syncytial virus in young children: a
systematic review and meta-analysis. Lancet 2010;
375:1545-55; PMID:20399493; http://dx.doi.org/10.1016/
S0140-6736(10)60206-1
[55] ODriscoll M, Jeggo PA. The role of double-strand break
repair - insights from human genetics. Nat Rev Genetics
2006; 7:45-54; PMID:16369571; http://dx.doi.org/
10.1038/nrg1746
[56] Panier S, Boulton SJ. Double-strand break repair: 53BP1
comes into focus. Nat Rev Mol Cell Biol 2014; 15:7-18;
PMID:24326623; http://dx.doi.org/10.1038/nrm3719
[57] Perez-Mancera PA, Young AR, Narita M. Inside and out:
the activities of senescence in cancer. Nat Rev Cancer
2014; 14:547-58; PMID:25030953; http://dx.doi.org/
10.1038/nrc3773
[58] Persson BD, Jaffe AB, Fearns R, Danahay H. Respiratory
syncytial virus can infect basal cells and alter human air-
way epithelial differentiation. PloS One 2014; 9:e102368;
PMID:25033192; http://dx.doi.org/10.1371/journal.pone.
0102368
[59] Rawlins EL, Okubo T, Xue Y, Brass DM, Auten RL, Hase-
gawa H, Wang F, Hogan BL. The role of Scgb1a1CClara
cells in the long-term maintenance and repair of lung air-
way, but not alveolar, epithelium. Cell Stem Cell 2009;
4:525-34; PMID:19497281; http://dx.doi.org/10.1016/j.
stem.2009.04.002
[60] Rodier F, Campisi J. Four faces of cellular senescence. J
Cell Biol 2011; 192:547-56; PMID:21321098; http://dx.
doi.org/10.1083/jcb.201009094
[61] Rodier F, Campisi J, Bhaumik D. Two faces of p53: aging and
tumor suppression. Nucleic Acids Res 2007; 35:7475-84;
PMID:17942417; http://dx.doi.org/10.1093/nar/gkm744
[62] Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner
WM. DNA double-stranded breaks induce histone
H2AX phosphorylation on serine 139. J Biol Chem 1998;
273:5858-68; PMID:9488723; http://dx.doi.org/10.1074/
jbc.273.10.5858
VIRULENCE 441
[63] Roth DB, Wilson JH. Illegitimate recombination in mamma-
lian cells. In: Kucherlapati R, Smith G. (eds) Genetic Recom-
bination. ASM Press, Washington, DC 1988, pp. 621-53
[64] Schultz LB, Chehab NH, Malikzay A, Halazonetis TD.
p53 binding protein 1 (53BP1) is an early participant in
the cellular response to DNA double-strand breaks. J Cell
Biol 2000; 151:1381-90; PMID:11134068; http://dx.doi.
org/10.1083/jcb.151.7.1381
[65] Sedelnikova OA, Horikawa I, Zimonjic DB, Popescu NC,
Bonner WM, Barrett JC. Senescing human cells and age-
ing mice accumulate DNA lesions with unrepairable dou-
ble-strand breaks. Nat Cell Biol 2004; 6:168-70;
PMID:14755273; http://dx.doi.org/10.1038/ncb1095
[66] Sedelnikova OA, Redon CE, Dickey JS, Nakamura AJ,
Georgakilas AG, Bonner WM. Role of oxidatively
induced DNA lesions in human pathogenesis. Mutation
Res 2010; 704:152-9; PMID:20060490; http://dx.doi.org/
10.1016/j.mrrev.2009.12.005
[67] Shrivastav M, De Haro LP, Nickoloff JA. Regulation of
DNA double-strand break repair pathway choice. Cell
Res 2008; 18:134-47; PMID:18157161; http://dx.doi.org/
10.1038/cr.2007.111
[68] Spallarossa P, Altieri P, Barisione C, Passalacqua M, Aloi
C, Fugazza G, Frassoni F, Podesta M, Canepa M, Ghi-
gliotti G, et al. p38 MAPK and JNK antagonistically con-
trol senescence and cytoplasmic p16INK4A expression in
doxorubicin-treated endothelial progenitor cells. PloS
One 2010; 5:e15583; PMID:21187925; http://dx.doi.org/
10.1371/journal.pone.0015583
[69] Stegmann T. Membrane fusion mechanisms: the inuenza
hemagglutinin paradigm and its implications for intracel-
lular fusion. Trafc 2000; 1:598-604; PMID:11208147;
http://dx.doi.org/10.1034/j.1600-0854.2000.010803.x
[70] Storer M, Mas A, Robert-Moreno A, Pecoraro M, Ortells
MC, Di Giacomo V, Yosef R, Pilpel N, Krizhanovsky V,
Sharpe J, et al. Senescence is a developmental mechanism
that contributes to embryonic growth and patterning.
Cell 2013; 155:1119-30; PMID:24238961; http://dx.doi.
org/10.1016/j.cell.2013.10.041
[71] Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland
JL. Cellular senescence and the senescent secretory phe-
notype: therapeutic opportunities. J Clin Invest 2013;
123:966-72; PMID:23454759; http://dx.doi.org/10.1172/
JCI64098
[72] Thompson L, Limoli C. Origin, Recognition, signaling
and repair of DNA double-strand breaks in mammalian
cells. In: Caldecott KW, editor. Eukaryotic DNA Damage
Surveillance and Repair. Georgetown, Texas: Landes Bio-
science; 2004; 2004: p 107-45.
[73] Ullah Z, Lee CY, Lilly MA, DePamphilis ML. Develop-
mentally programmed endoreduplication in animals.
Cell Cycle 2009; 8:1501-9; PMID:19372757; http://dx.doi.
org/10.4161/cc.8.10.8325
[74] Wu W, Munday DC, Howell G, Platt G, Barr JN, Hiscox
JA. Characterization of the interaction between human
respiratory syncytial virus and the cell cycle in continu-
ous cell culture and primary human airway epithelial
cells. J Virol 2011; 85:10300-9; PMID:21795354; http://
dx.doi.org/10.1128/JVI.05164-11
[75] Wyman C, Kanaar R. DNA double-strand break repair:
alls well that ends well. Annual Rev Genetics 2006;
40:363-83; http://dx.doi.org/10.1146/annurev.genet.40.110405.
090451
[76] Ye G, Metreveli NS, Donthi RV, Xia S, Xu M, Carlson
EC, Epstein PN. Catalase protects cardiomyocyte func-
tion in models of type 1 and type 2 diabetes. Diabetes
2004; 53:1336-43; PMID:15111504; http://dx.doi.org/
10.2337/diabetes.53.5.1336
[77] Zambrano A, Garcia-Carpizo V, Gallardo ME, Villa-
muera R, Gomez-Ferreria MA, Pascual A, Buisine N,
Sachs LM, Garesse R, et al. The thyroid hormone recep-
tor binduces DNA damage and premature senescence. J
Cell Biol 2014; 204:129-46; PMID:24395638; http://dx.
doi.org/10.1083/jcb.201305084
442 I. MART
INEZ ET AL.
... Oxidative stress induces the damage of DNA and may trigger cellular senescence (with proliferative arrest), which has also been shown in the case of RSV infection in HEp-2 and A549 cells [33]. The observed increase in the expression of the markers of DNA damage and proliferation arrest were the result of the increased production of ROS, at least partially derived from mitochondria [33]. ...
... Oxidative stress induces the damage of DNA and may trigger cellular senescence (with proliferative arrest), which has also been shown in the case of RSV infection in HEp-2 and A549 cells [33]. The observed increase in the expression of the markers of DNA damage and proliferation arrest were the result of the increased production of ROS, at least partially derived from mitochondria [33]. ROS cause (directly or as an effect of repair) DNA doublestrand breaks (DSBs), which may be reversed by the use of antioxidants; interestingly, NAC substantially decreased the number of damaged nuclei, but its protective effect was only observed in the case of de novo DNA damage foci, with virtually no effect on the previously existing ones [33]. ...
... The observed increase in the expression of the markers of DNA damage and proliferation arrest were the result of the increased production of ROS, at least partially derived from mitochondria [33]. ROS cause (directly or as an effect of repair) DNA doublestrand breaks (DSBs), which may be reversed by the use of antioxidants; interestingly, NAC substantially decreased the number of damaged nuclei, but its protective effect was only observed in the case of de novo DNA damage foci, with virtually no effect on the previously existing ones [33]. While a moderate reduction in the RSV titers was achieved with NAC treatment, a much stronger effect was observed when NAC was used during the RSV adsorption, suggesting a possible role for the antioxidants in RSV cell entry, surface attachment, or viral integrity [33]. ...
Article
Full-text available
N-acetylcysteine (NAC) is a mucolytic agent with antioxidant and anti-inflammatory properties. The respiratory syncytial virus (RSV) is one of the most important etiological factors of lower respiratory tract infections, and exposure to air pollution appears to be additionally associated with higher RSV incidence and disease severity. We aimed to systematically review the existing literature to determine which molecular mechanisms mediate the effects of NAC in an RSV infection and air pollution, and to identify the knowledge gaps in this field. A search for original studies was carried out in three databases and a calibrated extraction grid was used to extract data on the NAC treatment (dose, timing), the air pollutant type, and the most significant mechanisms. We identified only 28 studies conducted in human cellular models (n = 18), animal models (n = 7), and mixed models (n = 3). NAC treatment improves the barrier function of the epithelium damaged by RSV and air pollution, and reduces the epithelial permeability, protecting against viral entry. NAC may also block RSV-activated phosphorylation of the epidermal growth factor receptor (EGFR), which promotes endocytosis and facilitates cell entry. EGFR also enhances the release of a mucin gene, MUC5AC, which increases mucus viscosity and causes goblet cell metaplasia; the effects are abrogated by NAC. NAC blocks virus release from the infected cells, attenuates the cigarette smoke-induced shift from necrosis to apoptosis, and reverses the block in IFN-γ-induced antiviral gene expression caused by the inhibited Stat1 phosphorylation. Increased synthesis of pro-inflammatory cytokines and chemokines is induced by both RSV and air pollutants and is mediated by the nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways that are activated in response to oxidative stress. MCP-1 (monocyte chemoattractant protein-1) and RANTES (regulated upon activation, expressed and secreted by normal T cells) partially mediate airway hyperresponsiveness (AHR), and therapeutic (but not preventive) NAC administration reduces the inflammatory response and has been shown to reduce ozone-induced AHR. Oxidative stress-induced DNA damage and cellular senescence, observed during RSV infection and exposure to air pollution, can be partially reversed by NAC administration, while data on the emphysema formation are disputed. The review identified potential common molecular mechanisms of interest that are affected by NAC and may alleviate both the RSV infection and the effects of air pollution. Data are limited and gaps in knowledge include the optimal timing or dosage of NAC administration, therefore future studies should clarify these uncertainties and verify its practical use.
... However, the precise regulatory mechanisms directly linking HCMV to cellular senescence remain unknown. As DDR signaling pathways are critical for the replication of HCMV [21][22][23], it would be interesting to investigate if HCMV can cause or worsen cellular senescence through DDR. In this review, we first provide a detailed explanation of how DNA damage response (DDR) begins and develops as well as how DDR contributes to the establishment of cellular senescence. ...
... senescence, known as virus-induced senescence (VIS) [23,113,114]. Measles virus (MV) infection has been proven to induce p53 and p16-pRb pathway-dependent cellular senescence via cell [115]. Epstein-Barr virus (EBV), Kaposi sarcoma herpesvirus (KSHV) and human respiratory syncytial virus (RSV) infections can trigger DNA damage-mediated cellular senescence through replicative stress or induction of mitochondrial ROS [23,116,117]. ...
... Measles virus (MV) infection has been proven to induce p53 and p16-pRb pathway-dependent cellular senescence via cell [115]. Epstein-Barr virus (EBV), Kaposi sarcoma herpesvirus (KSHV) and human respiratory syncytial virus (RSV) infections can trigger DNA damage-mediated cellular senescence through replicative stress or induction of mitochondrial ROS [23,116,117]. Senescence markers and SASP factors have been found in tissue samples of the nasopharyngeal cavity and lungs of patients suffering from coronavirus disease 2019 (COVID-19) with severe disease progression [20]. A basic research study, assessing the occurrence of VIS, found that human diploid fibroblast models exposed to high-titer retrovirus exhibited typical senescence and the activated cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway after the fifth day of infection [118]. ...
Article
Full-text available
The DNA damage response (DDR) is a signaling cascade that is triggered by DNA damage, involving the halting of cell cycle progression and repair. It is a key event leading to senescence, which is characterized by irreversible cell cycle arrest and the senescence-associated secretory phenotype (SASP) that includes the expression of inflammatory cytokines. Human cytomegalovirus (HCMV) is a ubiquitous pathogen that plays an important role in the senescence process. It has been established that DDR is necessary for HCMV to replicate effectively. This paper reviews the relationship between DDR, cellular senescence, and HCMV, providing new sights for virus-induced senescence (VIS).
... Generally, various viruses enhance the generation of mitochondrial ROS (mtROS), which activates certain host cellular pathways that facilitate viral replication [38][39][40]. This can also be confirmed in cells infected by RSV [41,42]. RSV can induce microtubule/ dynein-dependent mitochondrial perinuclear clustering and translocation toward the microtubule-organizing center, which is concomitant with impaired mitochondrial respiration, loss of mitochondrial membrane potential, and increased production of mtROS. ...
... Persistent DNA damage signaling triggers senescence-associated inflammatory cytokine secretion [141]. A study has shown that the production of RSVinduced mtROS results in the expression of DNA damage markers, such as phosphorylated tumor suppressor p53 (TP53), ATM, cyclin-dependent kinase inhibitor 1A (CDKN1A) and γH2AFX (H2A histone family member X, phosphorylated on Ser 139), and proliferation arrest in cultured cells (Fig. 5) [42]. ATM, as a nuclear ROS and DNA damage sensor, is activated by RSV replication and translocated from the nucleus into the cytosol. ...
... Moreover, NAC is able to reverse DSBs via the inhibition of ROS production. ATM, as a nuclear ROS and DNA damage sensor, is translocated from the nucleus into the cytosol and activates the MSK1-phospho-Ser276 RelA-IRF-RIG-I amplification loop in RSV infection phenotype in both mononuclear cells and syncytia [42]. RSV-induced senescence may be seen as a host defense mechanism, instructing "damaged" cells to cease proliferation. ...
Article
Full-text available
Respiratory syncytial virus (RSV), a member of the Pneumoviridae family, can cause severe acute lower respiratory tract infection in infants, young children, immunocompromised individuals and elderly people. RSV is associated with an augmented innate immune response, enhanced secretion of inflammatory cytokines, and necrosis of infected cells. Oxidative stress, which is mainly characterized as an imbalance in the production of reactive oxygen species (ROS) and antioxidant responses, interacts with all the pathophysiologic processes above and is receiving increasing attention in RSV infection. A gradual accumulation of evidence indicates that ROS overproduction plays an important role in the pathogenesis of severe RSV infection and serves as a major factor in pulmonary inflammation and tissue damage. Thus, antioxidants seem to be an effective treatment for severe RSV infection. This article mainly reviews the information on oxidative stress and ROS-mediated cellular events during RSV infection for the first time.
... This includes impairment of local innate immune mechanisms, an hyperinflammatory phenotype related to age called "inflammaging", and a decline in humoral and cellular responses, termed "immunosenescence" (8)(9)(10). Infection with a number of viruses, such as measles virus, human respiratory syncytial virus, and coronaviruses, can induce premature cellular senescence (11). Interestingly, infection of lung tissue by IAV induced premature aging as indicated by the appearance of senescent cells and subsequently, infection of senescent cells resulted in increased viral replication (12). ...
Preprint
Full-text available
The recent coronavirus disease 2019 (COVID-19) outbreak revealed the susceptibility of elderly patients to respiratory virus infections, showing cell senescence or subclinical persistent inflammatory profiles and favouring the development of severe pneumonia. In our study, we evaluated the potential influence of lung aging on the efficiency of replication of influenza A virus (IAV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV 2), as well as determined the pro-inflammatory and antiviral responses of the distal lung tissue. Using precision-cut lung slices (PCLS) from donors of different ages, we found that pandemic H1N1 and avian H5N1 IAV replicated in the lung parenchyma with high efficacy. In contrast to these IAV strains, SARS-CoV-2 early isolate and Delta variant of concern (VOC) replicated less efficiently in PCLS. Interestingly, both viruses showed reduced replication in PCLS from older compared to younger donors, suggesting that aged lung tissue represents a sub optimal environment for viral replication. Regardless of the age-dependent viral loads, PCLS responded to infection with both viruses by an induction of IL-6 and IP-10/CXCL10 mRNAs, being highest for H5N1. Finally, while SARS-CoV-2 infection was not causing detectable cell death, IAV infection caused significant cytotoxicity and induced significant early interferon responses. In summary, our findings suggest that aged lung tissue might not favour viral dissemination, pointing to a determinant role of dysregulated immune mechanisms in the development of severe disease.
... Leveraging on these results, we speculated that HCMV-infected RPTECs may exhibit features of VIS 25,26 . It is in fact well-established that both DNA and RNA viruses can induce classic senescence hallmarks, such as SA-β-gal activity, increased expression of p16 INK4a , and p21 Waf1 , as well as the secretion of pro-inflammatory SASP molecules, and the generation of reactive oxygen species (ROS) [53][54][55][56] . Furthermore, previous in vitro studies on fibroblasts have shown that viral infection affects the host cell cycle and results in cytoplasm enlargement, induction of replication stress, and release of pro-inflammatory cytokines 15,30,57 . ...
Article
Full-text available
Human cytomegalovirus (HCMV) is an opportunistic pathogen causing severe diseases in immunosuppressed individuals. To replicate its double-stranded DNA genome, HCMV induces profound changes in cellular homeostasis that may resemble senescence. However, it remains to be determined whether HCMV-induced senescence contributes to organ-specific pathogenesis. Here, we show a direct cytopathic effect of HCMV on primary renal proximal tubular epithelial cells (RPTECs), a natural setting of HCMV disease. We find that RPTECs are fully permissive for HCMV replication, which endows them with an inflammatory gene signature resembling the senescence-associated secretory phenotype (SASP), as confirmed by the presence of the recently established SenMayo gene set, which is not observed in retina-derived epithelial (ARPE-19) cells. Although HCMV-induced senescence is not cell-type specific, as it can be observed in both RPTECs and human fibroblasts (HFFs), only infected RPTECs show downregulation of LAMINB1 and KI67 mRNAs, and enhanced secretion of IL-6 and IL-8, which are well-established hallmarks of senescence. Finally, HCMV-infected RPTECs have the ability to trigger a senescence/inflammatory loop in an IL-6-dependent manner, leading to the development of a similar senescence/inflammatory phenotype in neighboring uninfected cells. Overall, our findings raise the intriguing possibility that this unique inflammatory loop contributes to HCMV-related pathogenesis in the kidney.
... This activation leads to p53-related outcomes such as the transcriptional regulation of A3 and other immune genes and apoptosis induction. The findings of Martinez et al. [53] support our results which indicate that RSV infection in A549 cells leads to DNA damage, causing p53 induction. Consistent with these observations, other studies have also shown that RSV infection activates the DNA damage response via ATM signaling [54] and induces oxidative stress through the increased production of reactive oxygen species (ROS) [55,56]; both of these are known activators of p53 [7,8]. ...
Article
Full-text available
Identifying and understanding genetic factors that influence the propagation of the human respiratory syncytial virus (RSV) can lead to health benefits and possibly augment recent vaccine approaches. We previously identified a p53/immune axis in which the tumor suppressor p53 directly regulates the expression of immune system genes, including the seven members of the APOBEC3 family of DNA cytidine deaminases (A3), which are innate immune sentinels against viral infections. Here, we examined the potential p53 and A3 influence in RSV infection, as well as the overall p53-dependent cellular and p53/immune axis responses to infection. Using a paired p53 model system of p53+ and p53- human lung tumor cells, we found that RSV infection activates p53, leading to the altered p53-dependent expression of A3D, A3F, and A3G, along with p53 site-specific binding. Focusing on A3G because of its 10-fold-greater p53 responsiveness to RSV, the overexpression of A3G can reduce RSV viral replication and syncytial formation. We also observed that RSV-infected cells undergo p53-dependent apoptosis. The study was expanded to globally address at the transcriptional level the p53/immune axis response to RSV. Nearly 100 genes can be directly targeted by the p53/immune axis during RSV infection based on our p53BAER analysis (Binding And Expression Resource). Overall, we identify A3G as a potential p53-responsive restriction factor in RSV infection. These findings have significant implications for RSV clinical and therapeutic studies and other p53-influenced viral infections, including using p53 adjuvants to boost the response of A3 genes.
... DNA-PKI/ATMI and CV-B5/F cooperate to trigger irreversible ER stress RNA viruses can cause a DDR by inducing reactive oxygen species (ROS). 26,27 We found that CV-B5/F infection can induce ROS release, which can be exacerbated by NU7441 (Fig. 4a). In addition, CV-B5/F infection increased p-DNA-PK, p-ATM, and p-H2AX levels in NCI-H460 cells (Fig. 4b). ...
Article
Full-text available
With the continuous in-depth study of the interaction mechanism between viruses and hosts, the virus has become a promising tool in cancer treatment. In fact, many oncolytic viruses with selectivity and effectiveness have been used in cancer therapy. Human enterovirus is one of the most convenient sources to generate oncolytic viruses, however, the high seroprevalence of some enteroviruses limits its application which urges to exploit more oncolytic enteroviruses. In this study, coxsackievirus B5/Faulkner (CV-B5/F) was screened for its potential oncolytic effect against non-small cell lung cancers (NSCLCs) through inducing apoptosis and autophagy. For refractory NSCLCs, DNA-dependent protein kinase (DNA-PK) or ataxia telangiectasia mutated protein (ATM) inhibitors can synergize with CV-B5/F to promote refractory cell death. Here, we showed that viral infection triggered endoplasmic reticulum (ER) stress-related pro-apoptosis and autophagy signals, whereas repair for double-stranded DNA breaks (DSBs) contributed to cell survival which can be antagonized by inhibitor-induced cell death, manifesting exacerbated DSBs, apoptosis, and autophagy. Mechanistically, PERK pathway was activated by the combination of CV-B5/F and inhibitor, and the irreversible ER stress-induced exacerbated cell death. Furthermore, the degradation of activated STING by ERphagy promoted viral replication. Meanwhile, no treatment-related deaths due to CV-B5/F and/or inhibitors occurred. Conclusively, our study identifies an oncolytic CV-B5/F and the synergistic effects of inhibitors of DNA-PK or ATM, which is a potential therapy for NSCLCs.
Article
Patients with pulmonary fibrosis (PF) often experience exacerbations of their disease, characterised by a rapid, severe deterioration in lung function that is associated with high mortality. Whilst the pathobiology of such exacerbations is poorly understood, virus infection is a trigger. This study investigated virus-induced injury responses of alveolar and bronchial epithelial cells (AECs and BECs respectively) from patients with PF and age-matched controls (Ctrls). Air liquid interface (ALI) cultures of AECs, comprising type I and II pneumocytes or BECs were inoculated with influenza A virus (H1N1) at 0.1 multiplicity of infection (MOI). Levels of interleukin-6 (IL-6), IL-36γ and IL-1β were elevated in cultures of AECs from PF patients (PF-AECs, n=8-11), being markedly higher than Ctrl-AECs (n=5-6), 48 h post inoculation (pi) (P<0.05); despite no difference in H1N1 RNA copy numbers 24 h pi. Furthermore, the virus-induced inflammatory responses of PF-AECs were greater than BECs (from either PF patients or controls), even though viral loads in the BECs were overall 2 to 3-fold higher than AECs. Baseline levels of the senescence and DNA damage markers, nuclear p21, p16 and H2AXγ were also significantly higher in PF-AECs than Ctrl-AECs and further elevated post-infection. Senescence induction using etoposide augmented virus-induced injuries in AECs (but not viral load), whereas selected senotherapeutics (rapamycin and mitoTEMPO) were protective. This study provides evidence that senescence increases the susceptibility of AECs from PF patients to severe virus-induced injury and suggests targeting senescence may provide an alternative option to prevent or treat the exacerbations that worsen the underlying disease.
Article
Severe viral lower respiratory tract infection (LRTI), resulting in both acute and long-term pulmonary disease, constitutes a substantial burden among young children. Viral LRTI triggers local oxidative stress pathways by infection and inflammation, and supportive care in the pediatric intensive care unit may further aggravate oxidative injury. The main goal of this exploratory study was to identify and monitor breath markers linked to oxidative stress in children over the disease course of severe viral LRTI. Exhaled breath was sampled during invasive ventilation and volatile organic compounds (VOCs) were analyzed using gas-chromatography and mass-spectrometry. VOCs were selected in an untargeted principal component analysis and assessed for change over time. Additionally, identified VOCs were correlated with clinical parameters. Seventy breath samples from 21 patients were analyzed. A total of 15 VOCs were identified that contributed the most to the explained variance of breath markers. Of these 15 VOCs, 10 were previously linked to pathways of oxidative stress. Eight VOCs, including seven alkanes and methyl alkanes, significantly decreased from the initial phase of ventilation to the day of extubation. No correlation was observed with the administered oxygen dose, while 6 VOCs showed a poor-to-strong positive correlation with driving pressure. In this prospective study of children with severe viral LRTI, the majority of VOCs that were most important for the explained variance mirrored clinical improvement. These breath markers could potentially help monitor the pulmonary oxidative status in these patients, but further research with other objective measures of pulmonary injury is required.
Article
Full-text available
We have previously estimated that respiratory syncytial virus (RSV) was associated with 22% of all episodes of (severe) acute lower respiratory infection (ALRI) resulting in 55 000 to 199 000 deaths in children younger than 5 years in 2005. In the past 5 years, major research activity on RSV has yielded substantial new data from developing countries. With a considerably expanded dataset from a large international collaboration, we aimed to estimate the global incidence, hospital admission rate, and mortality from RSV-ALRI episodes in young children in 2015. We estimated the incidence and hospital admission rate of RSV-associated ALRI (RSV-ALRI) in children younger than 5 years stratified by age and World Bank income regions from a systematic review of studies published between Jan 1, 1995, and Dec 31, 2016, and unpublished data from 76 high quality population-based studies. We estimated the RSV-ALRI incidence for 132 developing countries using a risk factor-based model and 2015 population estimates. We estimated the in-hospital RSV-ALRI mortality by combining in-hospital case fatality ratios with hospital admission estimates from hospital-based (published and unpublished) studies. We also estimated overall RSV-ALRI mortality by identifying studies reporting monthly data for ALRI mortality in the community and RSV activity. We estimated that globally in 2015, 33·1 million (uncertainty range [UR] 21·6-50·3) episodes of RSV-ALRI, resulted in about 3·2 million (2·7-3·8) hospital admissions, and 59 600 (48 000-74 500) in-hospital deaths in children younger than 5 years. In children younger than 6 months, 1·4 million (UR 1·2-1·7) hospital admissions, and 27 300 (UR 20 700-36 200) in-hospital deaths were due to RSV-ALRI. We also estimated that the overall RSV-ALRI mortality could be as high as 118 200 (UR 94 600-149 400). Incidence and mortality varied substantially from year to year in any given population. Globally, RSV is a common cause of childhood ALRI and a major cause of hospital admissions in young children, resulting in a substantial burden on health-care services. About 45% of hospital admissions and in-hospital deaths due to RSV-ALRI occur in children younger than 6 months. An effective maternal RSV vaccine or monoclonal antibody could have a substantial effect on disease burden in this age group. The Bill & Melinda Gates Foundation.
Article
Full-text available
Unlabelled: Respiratory syncytial virus (RSV) is a primary etiological agent of childhood lower respiratory tract disease. Molecular patterns induced by active infection trigger a coordinated retinoic acid-inducible gene I (RIG-I)-Toll-like receptor (TLR) signaling response to induce inflammatory cytokines and antiviral mucosal interferons. Recently, we discovered a nuclear oxidative stress-sensitive pathway mediated by the DNA damage response protein, ataxia telangiectasia mutated (ATM), in cytokine-induced NF-κB/RelA Ser 276 phosphorylation. Here we observe that ATM silencing results in enhanced single-strand RNA (ssRNA) replication of RSVand Sendai virus, due to decreased expression and secretion of type I and III interferons (IFNs), despite maintenance of IFN regulatory factor 3 (IRF3)-dependent IFN-stimulated genes (ISGs). In addition to enhanced oxidative stress, RSV replication enhances foci of phosphorylated histone 2AX variant (γH2AX), Ser 1981 phosphorylation of ATM, and IKKγ/NEMO-dependent ATM nuclear export, indicating activation of the DNA damage response. ATM-deficient cells show defective RSV-induced mitogen and stress-activated kinase 1 (MSK-1) Ser 376 phosphorylation and reduced RelA Ser 276 phosphorylation, whose formation is required for IRF7 expression. We observe that RelA inducibly binds the native IFN regulatory factor 7 (IRF7) promoter in an ATM-dependent manner, and IRF7 inducibly binds to the endogenous retinoic acid-inducible gene I (RIG-I) promoter. Ectopic IRF7 expression restores RIG-I expression and type I/III IFN expression in ATM-silenced cells. We conclude that paramyxoviruses trigger the DNA damage response, a pathway required for MSK1 activation of phospho Ser 276 RelA formation to trigger the IRF7-RIG-I amplification loop necessary for mucosal IFN production. These data provide the molecular pathogenesis for defects in the cellular innate immunity of patients with homozygous ATM mutations. Importance: RNA virus infections trigger cellular response pathways to limit spread to adjacent tissues. This "innate immune response" is mediated by germ line-encoded pattern recognition receptors that trigger activation of two, largely independent, intracellular NF-κB and IRF3 transcription factors. Downstream, expression of protective antiviral interferons is amplified by positive-feedback loops mediated by inducible interferon regulatory factors (IRFs) and retinoic acid inducible gene (RIG-I). Our results indicate that a nuclear oxidative stress- and DNA damage-sensing factor, ATM, is required to mediate a cross talk pathway between NF-κB and IRF7 through mediating phosphorylation of NF-κB. Our studies provide further information about the defects in cellular and innate immunity in patients with inherited ATM mutations.
Article
Full-text available
Respiratory syncytial virus (RSV) is a major cause of morbidity and mortality worldwide, causing severe respiratory illness in infants and immune compromised patients. The ciliated cells of the human airway epithelium have been considered to be the exclusive target of RSV, although recent data have suggested that basal cells, the progenitors for the conducting airway epithelium, may also become infected in vivo. Using either mechanical or chemical injury models, we have demonstrated a robust RSV infection of p63+ basal cells in air-liquid interface (ALI) cultures of human bronchial epithelial cells. In addition, proliferating basal cells in 2D culture were also susceptible to RSV infection. We therefore tested the hypothesis that RSV infection of this progenitor cell would influence the differentiation status of the airway epithelium. RSV infection of basal cells on the day of seeding (MOI≤0.0001), resulted in the formation of an epithelium that showed a profound loss of ciliated cells and gain of secretory cells as assessed by acetylated α-tubulin and MUC5AC/MUC5B immunostaining, respectively. The mechanism driving the switch in epithelial phenotype is in part driven by the induced type I and type III interferon response that we demonstrate is triggered early following RSV infection. Neutralization of this response attenuates the RSV-induced loss of ciliated cells. Together, these data show that through infection of proliferating airway basal cells, RSV has the potential to influence the cellular composition of the airway epithelium. The resulting phenotype might be expected to contribute towards both the severity of acute infection, as well as to the longer-term consequences of viral exacerbations in patients with pre-existing respiratory diseases.
Article
Full-text available
There is increasing evidence that the thyroid hormone (TH) receptors (THRs) can play a role in aging, cancer and degenerative diseases. In this paper, we demonstrate that binding of TH T3 (triiodothyronine) to THRB induces senescence and deoxyribonucleic acid (DNA) damage in cultured cells and in tissues of young hyperthyroid mice. T3 induces a rapid activation of ATM (ataxia telangiectasia mutated)/PRKAA (adenosine monophosphate-activated protein kinase) signal transduction and recruitment of the NRF1 (nuclear respiratory factor 1) and THRB to the promoters of genes with a key role on mitochondrial respiration. Increased respiration leads to production of mitochondrial reactive oxygen species, which in turn causes oxidative stress and DNA double-strand breaks and triggers a DNA damage response that ultimately leads to premature senescence of susceptible cells. Our findings provide a mechanism for integrating metabolic effects of THs with the tumor suppressor activity of THRB, the effect of thyroidal status on longevity, and the occurrence of tissue damage in hyperthyroidism.
Article
Full-text available
DNA double-strand break (DSB) signalling and repair is crucial to preserve genomic integrity and maintain cellular homeostasis. p53-binding protein 1 (53BP1) is an important regulator of the cellular response to DSBs that promotes the end-joining of distal DNA ends, which is induced during V(D)J and class switch recombination as well as during the fusion of deprotected telomeres. New insights have been gained into the mechanisms underlying the recruitment of 53BP1 to damaged chromatin and how 53BP1 promotes non-homologous end-joining-mediated DSB repair while preventing homologous recombination. From these studies, a model is emerging in which 53BP1 recruitment requires the direct recognition of a DSB-specific histone code and its influence on pathway choice is mediated by mutual antagonism with breast cancer 1 (BRCA1).
Article
The core aspect of the senescent phenotype is a stable state of cell cycle arrest. However, this is a disguise that conceals a highly active metabolic cell state with diverse functionality. Both the cell-autonomous and the non-cell-autonomous activities of senescent cells create spatiotemporally dynamic and context-dependent tissue reactions. For example, the senescence-associated secretory phenotype (SASP) provokes not only tumour-suppressive but also tumour-promoting responses. Senescence is now increasingly considered to be an integrated and widespread component that is potentially important for tumour development, tumour suppression and the response to therapy.
Article
Recent discoveries are redefining our view of cellular senescence as a trigger of tissue remodelling that acts during normal embryonic development and upon tissue damage. To achieve this, senescent cells arrest their own proliferation, recruit phagocytic immune cells and promote tissue renewal. This sequence of events - senescence, followed by clearance and then regeneration - may not be efficiently completed in aged tissues or in pathological contexts, thereby resulting in the accumulation of senescent cells. Increasing evidence indicates that both pro-senescent therapies and antisenescent therapies can be beneficial. In cancer and during active tissue repair, pro-senescent therapies contribute to minimize the damage by limiting proliferation and fibrosis, respectively. Conversely, antisenescent therapies may help to eliminate accumulated senescent cells and to recover tissue function.
Article
The paramyxoviruses represent a diverse virus family responsible for a wide range of human and animal diseases. In contrast to other viruses, such as HIV and influenza virus, which use a single glycoprotein to mediate host receptor binding and virus entry, the paramyxoviruses require two distinct proteins. One of these is an attachment glycoprotein that binds receptor, while the second is a fusion glycoprotein, which undergoes conformational changes that drive virus-cell membrane fusion and virus entry. The details of how receptor binding by one protein activates the second to undergo conformational changes have been poorly understood until recently. Over the past couple of years, structural and functional data have accumulated on representative members of this family, including parainfluenza virus 5, Newcastle disease virus, measles virus, Nipah virus and others, which suggest a mechanistic convergence of activation models. Here we review the data indicating that paramyxovirus attachment glycoproteins shield activating residues within their N-terminal stalk domains, which are then exposed upon receptor binding, leading to the activation of the fusion protein by a 'provocateur' mechanism.
Article
Human Immunodeficiency Virus (HIV) initiates infection by fusing its envelope membrane with the cell membrane through a process which is triggered through interactions with the cellular receptor and coreceptor. Although the mechanism of HIV fusion has been extensively studied, the point of its entry into cells remains controversial. HIV has long been thought to fuse directly with the cell plasma membrane. However, several lines of evidence suggest that endocytic entry of HIV can lead to infection and, moreover, that endocytosis could be the predominant HIV entry pathway into different cell types. This review discusses recent findings pertinent to HIV entry routes and novel approaches to pinpoint the sites of virus entry.