Sei sulla pagina 1di 11

crossmark

Constitutively Expressed IFITM3 Protein in Human Endothelial Cells


Poses an Early Infection Block to Human Influenza Viruses
Xiangjie Sun, Hui Zeng, Amrita Kumar, Jessica A. Belser, Taronna R. Maines, Terrence M. Tumpey
Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention,
Atlanta, Georgia, USA

ABSTRACT
A role for pulmonary endothelial cells in the orchestration of cytokine production and leukocyte recruitment during influenza
virus infection, leading to severe lung damage, has been recently identified. As the mechanistic pathway for this ability is not
fully known, we extended previous studies on influenza virus tropism in cultured human pulmonary endothelial cells. We found
that a subset of avian influenza viruses, including potentially pandemic H5N1, H7N9, and H9N2 viruses, could infect human
pulmonary endothelial cells (HULEC) with high efficiency compared to human H1N1 or H3N2 viruses. In HULEC, human influ-
enza viruses were capable of binding to host cellular receptors, becoming internalized and initiating hemifusion but failing to
uncoat the viral nucleocapsid and to replicate in host nuclei. Unlike numerous cell types, including epithelial cells, we found that
pulmonary endothelial cells constitutively express a high level of the restriction protein IFITM3 in endosomal compartments.
IFITM3 knockdown by small interfering RNA (siRNA) could partially rescue H1N1 virus infection in HULEC, suggesting
IFITM3 proteins were involved in blocking human influenza virus infection in endothelial cells. In contrast, selected avian influ-
enza viruses were able to escape IFITM3 restriction in endothelial cells, possibly by fusing in early endosomes at higher pH or by
other, unknown mechanisms. Collectively, our study demonstrates that the human pulmonary endothelium possesses intrinsic
immunity to human influenza viruses, in part due to the constitutive expression of IFITM3 proteins. Notably, certain avian in-
fluenza viruses have evolved to escape this restriction, possibly contributing to virus-induced pneumonia and severe lung disease
in humans.

IMPORTANCE
Avian influenza viruses, including H5N1 and H7N9, have been associated with severe respiratory disease and fatal outcomes in
humans. Although acute respiratory distress syndrome (ARDS) and progressive pulmonary endothelial damage are known to be
present during severe human infections, the role of pulmonary endothelial cells in the pathogenesis of avian influenza virus in-
fections is largely unknown. By comparing human seasonal influenza strains to avian influenza viruses, we provide greater in-
sight into the interaction of influenza virus with human pulmonary endothelial cells. We show that human influenza virus infec-
tion is blocked during the early stages of virus entry, which is likely due to the relatively high expression of the host antiviral
factors IFITMs (interferon-induced transmembrane proteins) located in membrane-bound compartments inside cells. Overall,
this study provides a mechanism by which human endothelial cells limit replication of human influenza virus strains, whereas
avian influenza viruses overcome these restriction factors in this cell type.

I nfluenza A viruses are important respiratory pathogens in hu-


mans and are responsible for approximately 250,000 to 500,000
fatal cases of influenza during annual epidemics worldwide (1).
the 1918 and 2009 H1N1 viruses) or recently isolated HPAI H5N1
viruses possess the ability to replicate in human lower respiratory
tract tissues and induce exacerbated innate immune responses
Occasionally, influenza A viruses of novel strains or subtypes (6–9). This is demonstrated by early recruitment of inflammatory
against which the general human population has no preexisting leukocytes to the lung and excessive cytokine production, ulti-
immunity emerge and cause severe pandemics, as was demon- mately leading to acute respiratory distress syndrome (ARDS) and
strated in 1918, 1957, 1968, and, most recently, in 2009 (2). Mean- high mortality rates (10, 11). While the molecular mechanisms of
while, certain influenza A viruses of avian origin are capable of severe illness caused by influenza virus infection have not been
crossing host species barriers, resulting in sporadic infection in completely uncovered, it is believed that aberrant proinflamma-
humans. Among these viruses, highly pathogenic avian influenza
(HPAI) H5N1 viruses cause the highest mortality rate in humans,
approximately 60% based on WHO reports (3). While exhibiting Received 29 June 2016 Accepted 27 September 2016

reduced mortality in humans, low-pathogenicity avian influenza Accepted manuscript posted online 5 October 2016

(LPAI) viruses of the H7N9 subtype have also been associated with Citation Sun X, Zeng H, Kumar A, Belser JA, Maines TR, Tumpey TM. 2016.
Constitutively expressed IFITM3 protein in human endothelial cells poses an early
severe disease, with over 700 reported cases since their initial de- infection block to human influenza viruses. J Virol 90:11157–11167.
tection in humans in 2013 (4, 5). doi:10.1128/JVI.01254-16.
Human influenza A viruses primarily target epithelial cells in Editor: B. Williams, Hudson Institute of Medical Research
the upper respiratory tract due to their abundant expression of Address correspondence to Terrence M. Tumpey, tft9@cdc.gov.
␣-2,6-linked sialic acids, the preferred receptors for human influ- Copyright © 2016, American Society for Microbiology. All Rights Reserved.
enza viruses (1). However, pandemic influenza viruses (including

December 2016 Volume 90 Number 24 Journal of Virology jvi.asm.org 11157


Sun et al.

tory cytokine production and the resulting damage to the epithe- expressing high levels of IFITM proteins before interferon induc-
lial-endothelial barrier of the pulmonary alveolus play an impor- tion.
tant role in the development of severe disease (12). Recently, it has
been revealed that pulmonary endothelial cells are central orches- MATERIALS AND METHODS
trators of cytokine production and leukocyte recruitment in mice Cells and viruses. Primary human lung blood microvascular endothelial
inoculated with the 2009 pandemic H1N1 virus (13). The work cells (HMVEC) (Lonza, Walkersville, MD), immortalized human lung
suggests that despite not representing a primary site for influenza microvascular endothelial cells (HULEC), and human umbilical vein en-
virus replication, pulmonary endothelial cells contribute to the dothelial cells (HUVEC) (obtained from the Scientific Resources Pro-
severity of the infection (13). Moreover, in vitro studies have gram, CDC, Atlanta, GA) were cultured in endothelial cell basal medium
shown that influenza virus infection can upregulate the expression 2 (EBM-2) (Lonza) with supplements as previously described (17). Hu-
of several endothelial adhesion molecules (14, 15), which may man brain vascular endothelial cells (HBVEC), kindly provided by Mo-
facilitate extravasation of neutrophils and macrophages into the nique Stins, Johns Hopkins University, were cultured in RPMI 1640 me-
alveoli. The persistent influx of such inflammatory cells can lead to dium supplemented with 10% fetal bovine serum (FBS), 10% Nu-Serum
(Fisher Scientific), 2 mM L-glutamine, 1 mM sodium pyruvate, 1⫻ min-
damage of the epithelial-endothelial barrier by releasing reactive
imal essential medium (MEM) nonessential amino acids, MEM vitamins,
oxygen species, cytokines, and neutrophil extracellular traps (16). and penicillin-streptomycin (100 U/ml). Human bronchial epithelium
Additionally, pulmonary endothelial cells are susceptible to HPAI (Calu-3) cells were cultured in Eagle’s MEM supplemented with 10% FBS,
H5N1 virus infection in vitro in an envelope-dependent manner 2 mM L-glutamine, 1 mM sodium pyruvate, and nonessential amino ac-
and express high levels of proinflammatory cytokines upon infec- ids. Human lung epithelial A549 cells and Madin-Darby canine kidney
tion, whereas most human influenza viruses display only limited (MDCK) cells were grown in Dulbecco’s modified Eagle medium
infectivity under these conditions (17–19). However, the exact (DMEM) supplemented with 10% FBS. Primary human bronchial epithe-
molecular mechanism governing how selected highly pathogenic lial (NHBE) cells were grown in serum-free and hormone-supplemented
H5N1 viruses, but not human influenza viruses, possess the ability bronchial epithelial growth medium according to the manufacturer’s in-
to replicate and induce excessive cytokine production is still structions (Lonza). The A549 cells stably expressing IFITM3 or the vector
largely unknown. pQCXIP (Clontech) were originally developed and kindly provided by
Gregory B. Melikyan and Mariana Marin, Emory University, and were
IFITMs (interferon-induced transmembrane proteins) were
maintained with 1.5 mg/ml of puromycin.
first identified as type I and type II interferon (IFN)-induced pro- Influenza viruses A/Puerto Rico/8/1934 (PR8) (H1N1), A/Vietnam/
teins in 1984 (20) and comprise a family of small, 10- to 15-kDa 1203/2004 (VN/04) (H5N1), A/Anhui/1/2013 (Anhui/1) (H7N9), A/
proteins. In humans, there are four functional IFITM genes: the chicken/Vietnam/NCVD-1156/2011 (CK/11) (H9N2), A/Netherlands/
IFITM1, IFITM2, IFITM3, and IFITM5 genes, with IFITM4 being 219/2003 (NL/03) (H7N7), A/shoveler/Egypt/00215-NAMRU3/2007
a pseudogene (21). Although previous studies on IFITMs mainly (Shov/07) (H7N9), A/Brisbane/59/2007 (Bris/07) (seasonal H1N1), A/
focused on their roles in embryonic development, their functions Panama/2007/1999 (Pan/99) (seasonal H3N2), and 2009 pandemic
as host antiviral factors were only recently discovered by RNA A/Mexico/4482/2009 (Mex/09) (H1N1) were grown in the allantoic cav-
interference genomic screening for host factors involved in in- ities of 10-day-old embryonated hen’s eggs for 24 to 48 h at 33°C to 37°C.
fluenza virus infection (22). It was subsequently revealed that A/Brisbane/59/2007 (Bris/07) (seasonal H1N1) and 2009 pandemic
A/Mexico/4482/2009 (Mex/09) (H1N1) were propagated in MDCK cells
IFITMs can restrict the early stages of replication of a wide variety
as described previously (28). Allantoic fluid or cell culture supernatant
of viruses, including influenza virus (22), flavivirus (dengue and was clarified by centrifugation, aliquoted, and stored at ⫺70°C; virus titers
West Nile viruses) (23, 24), filovirus (Marburg virus and Ebola were determined by standard plaque assay using MDCK cells. All research
viruses) (23), and coronavirus (23). Among all the IFITM mem- with HPAI H5, H7, and H9 subtype viruses was conducted under bio-
bers present in humans, IFITM3 has shown to be the most potent safety level 3 (BSL3) containment, including enhancements required by
antiviral factor in restricting influenza virus infection, as IFITM3 the U.S. Department of Agriculture and the National Select Agent Pro-
knockout mice displayed enhanced morbidity and mortality asso- gram.
ciated with seasonal or 2009 pandemic H1N1 influenza virus in- The reassortant viruses bearing HA and NA genes from VN/VN/04
fection (25, 26). Furthermore, the single-nucleotide polymor- (H5N1) or Anhui/1 (H7N9) virus and internal genes from the PR8 donor
phism (SNP) (12252-C) in the IFITM3 gene, which results in virus (VN:PR8 and Anhui:PR8) (kindly provided by Li-Mei Chen, Influ-
enza Division, CDC) were propagated in the allantoic cavities of 10-day-
decreased IFITM3 protein expression, has been linked with a
old embryonated hen’s eggs for 48 h at 35°C under BSL2⫹ laboratory
higher risk of hospitalization among individuals infected with sea- conditions.
sonal or 2009 pandemic H1N1 virus, as well as the novel H7N9 Virus purification. PR8 viruses were propagated in the allantoic cav-
virus (26, 27). ities of 10-day-old embryonated hen’s eggs and then concentrated and
In this study, we investigated the mechanism by which human purified by equilibrium density centrifugation through a 30 to 60% linear
endothelial cells limit replication of human influenza viruses and sucrose gradient as previously described (29). The concentrations of pu-
how avian influenza viruses overcome these restriction factors in rified virus were determined using a Bio-Rad protein assay kit (Bio-Rad
this cell type. Our results show that human influenza virus infec- Laboratories, Hercules, CA) and then diluted to 2 mg/ml in phosphate-
tion is blocked during the early stages of virus entry, precisely, buffered saline (PBS) for virus labeling.
after hemifusion, likely due to the relatively high constitutive ex- Virus infection. HULEC or A549 cells were incubated with influenza
virus at multiplicities of infection (MOI) ranging from 1 to 5 in viral
pression of the host antiviral factor IFITM3 located in endosomal
infection medium (base culture medium supplemented with 0.3% bovine
and lysosomal compartments. Conversely, certain avian influenza serum albumin [BSA]) for 1 h, followed by washing with viral infection
viruses may circumvent this restriction by fusing at a higher pH in medium, and cultured with fresh infection medium at 37°C in a 5% CO2
early endosomes or by other, unknown mechanisms. Overall, our atmosphere for 8 h (time of peak expression) before being fixed with 4%
study suggests human hosts are able to restrict influenza virus paraformaldehyde for 20 min. Viral infectivity was quantified based on
infection in pulmonary endothelial cells partially by constitutively the percentage of nucleoprotein (NP)-positive cells by indirect immuno-

11158 jvi.asm.org Journal of Virology December 2016 Volume 90 Number 24


Influenza Virus Infection in Pulmonary Endothelial Cells

fluorescence microscopy (counting at least 200 cells per infection) using a Immunofluorescence microscopy and confocal imaging. Images
mouse monoclonal anti-influenza A virus NP clone A1 and A3 blend were captured with a Zeiss Axioskop 2 fluorescence microscope with a
(Millipore, Billerica, MA). 20⫻ or 40⫻ objective lens or an LSM 710 Zeiss inverted confocal micro-
Acid-induced fusion assay at the cell plasma membrane (acid bypass scope with a 40⫻, 63⫻, or 100⫻ oil objective lens and processed using
assay). Influenza viruses at an MOI of 5 were bound to A549 cells and Zen 2010 (Zeiss) and Adobe Photoshop (Adobe Inc.). The colocalization
HULEC on ice for 1 h, washed twice with prechilled viral infection me- coefficient was analyzed with Zen 2010 and calculated based on the for-
dium to remove any unbound virus particles, and then incubated with mula described by Manders et al. (34) from an average of at least 10
prewarmed fusion buffer (20 mM HEPES, 2 mM CaCl2, 150 mM NaCl, 20 individual cells. For colocalization studies, the following reagents were
mM citric acid monohydrate-sodium citrate tribasic dehydrate, pH 5.0) used: rabbit anti-EEA1 antibody (Cell Signaling), CellLight Rab7-green
for 2 min at 37°C to induce virus fusion at the cell surface. Following acid fluorescent protein (GFP) (ThermoFisher Scientific), rabbit anti-human
treatment, the cells were washed twice with prechilled viral infection me- CD107a (LAMP-1) and anti-human CD63 antibodies (BD Biosciences),
dium and then incubated for 8 h with viral infection medium containing and mouse anti-EEA1 and LAMP-1 from Santa Cruz Biotechnology Inc.
30 mM NH4Cl to prevent viral infection via endocytic pathways. Virus Western blotting. Human endothelial cells and epithelial cells from
infection mediated by viral fusion at the cell surface was quantified based either cell lines or primary culture were grown to confluence in T-75
flasks. The cells were trypsinized and resuspended at 2 ⫻ 106/ml before
on the percentage of NP-positive cells, using immunofluorescence mi-
being spun down and lysed in 2⫻ Laemmli sample buffer (Bio-Rad) con-
croscopy.
taining 5% ␤-mercaptoethanol. The samples were boiled at 95°C for 5
Virus internalization assay. Concentrated and purified influenza PR8
min before being loaded into a 4 to 15% Mini-Protean Tris-glycine pre-
viruses were labeled with biotin and used in a virus internalization assay as
cast gel (Bio-Rad). Rabbit anti-IFITM3 (ThermoFisher Scientific) and
described previously (30). Briefly, 1 ml of purified virus (2 mg/ml) was
anti-␤-actin (Sigma-Aldrich) antibodies were used to detect IFITM3 and
incubated with 10 mM Sulfo-NHS-SS-biotin (Pierce) for 2 h at 4°C, fol- actin expression in cell lysates, respectively.
lowed by ultracentrifugation to remove any unincorporated biotin. The Influenza virus pH optimum of fusion. Analysis of the pH threshold
labeled viruses were resuspended in PBS at a concentration of 1 mg/ml of fusion was evaluated by virus-induced hemolysis assay as described by
and filtered through a 0.22-␮m filter before use. During the virus inter- Shelton et al. (35). Briefly, viruses at 128 HA units (HAU) per 50 ␮l or PBS
nalization assay, 10 ␮l of biotin-labeled virus was incubated with cells in mock control was mixed with 50 ␮l of 1% (vol/vol) turkey red blood cells
viral infection medium for 1 h at 4°C, followed by washing with ice-cold and incubated at 4°C in a 96-well plate for 1 h; then, the mixtures were
viral infection medium to remove any unbound virus particles. To induce pelleted and resuspended in 100 ␮l of fusion buffer (20 mM HEPES, 2 mM
internalization, the virus-bound cells were shifted to 37°C for 1 h in viral CaCl2, 150 mM NaCl, 20 mM citric acid monohydrate-sodium citrate
infection medium. To distinguish cell surface-bound virus particles from tribasic dehydrate) with various pH values (from 4.8 to 7.4) at 37°C for 1
internalized virions, cells were incubated with excess quantities of uncon- h to trigger hemolysis mediated by viral fusion activity. The released he-
jugated streptavidin (2 mg/ml; Thermo Fisher Scientific Inc., Rockford, moglobin was measured as the optical density at 405 nm (OD405). The OD
IL) on ice after internalization to mask biotins from surface-bound vi- value from mock PBS control at each pH value was subtracted to calculate
ruses, after which the cells were fixed and permeabilized with 0.5% Triton hemolysis. The maximal hemolysis at a given pH was normalized to 100%,
X-100 and visualized by immunofluorescence microscopy by staining and the hemolysis at other pH values was expressed as a percentage of the
with Alexa 488-conjugated streptavidin (Thermo Fisher Scientific Inc.). maximal hemolysis.
Virus fusion assay. Concentrated and purified PR8 virus at 100 ␮g/ml
was labeled with R18 and SP-DiOC18 (Thermo Fisher Scientific Inc.) at RESULTS
final concentrations of 0.4 ␮M and 0.2 ␮M, as described previously (31, A group of selected avian influenza viruses exhibited extended
32). Ten microliters of labeled PR8 viruses was bound to cells grown on tropism in human pulmonary endothelial cells. It has been doc-
24-well plates on ice for 1 h, followed by washing three times with ice-cold
umented previously that human influenza viruses of the H1N1
viral infection medium, before the plates were transferred to a 37°C incu-
and H3N2 subtypes possess low infectivity in human pulmonary
bator with 0.5% CO2 for 1.5 h to induce viral internalization and fusion.
The cells were fixed with 4% paraformaldehyde (PFA) and stained with
endothelial cells, whereas HPAI H5N1 viruses are able to infect
DAPI (4=,6-diamidino-2-phenylindole) before being scanned with an and replicate efficiently in these cells (17–19). In order to further
LSM 710 Zeiss inverted confocal microscope at wavelengths of 510 to 525 examine the tropism of influenza viruses in pulmonary endothe-
nm (green) and 575 to 640 nm (red) simultaneously. Viral fusion in en- lial cells, we infected HULEC with human or avian influenza vi-
dosomes is indicated by a fluorescence color shift from the red to the green ruses for 8 h; viral infectivity was measured by NP expression and
channel. examined by immunofluorescence microscopy. Different sub-
Detection of influenza virus nucleocapsid uncoating. PR8 viruses at types were used for HULEC infection, and parallel viral infections
an MOI of 40 were bound to HULEC and A549 cells on ice for 1 h in viral in human lung epithelial A549 cells, which are highly susceptible
infection medium, after which the virus-bound cells were incubated at to human and avian influenza viruses, were included. The amount
37°C for 3.5 h following washing with viral infection medium. The cells of virus corresponding to an MOI of 1 to 5, which produced 80 to
were fixed, permeabilized, and stained with the anti-M1 monoclonal an- 95% infectivity in A549 cells, was chosen to infect HULEC, and
tibody HB64 (ATCC, Manassas, VA) by indirect immunofluorescence virus infectivity in both cell types was determined (Fig. 1A). To
microscopy. directly compare viral infectivity among different viruses, viral
siRNA transfection. HULEC and A549 cells grown in 24-well plates
infection efficiency was expressed as the ratio of the percentage of
were transfected with a mixture of IFITM3 small interfering RNA (siRNA)
NP-positive HULEC versus that of NP-positive permissive A549
(catalog number 284737) or negative-control siRNA (catalog number
AM4611) and Lipofectamine 2000 (Thermo Fisher Scientific Inc.) follow- cells. As shown in Fig. 1B, human influenza viruses of H1 and H3
ing the manufacturer’s recommended transfection procedures as de- subtypes exhibited very limited viral infection rates in HULEC
scribed previously (33); the knockdown effect on the IFITM3 protein with an infection ratio below 0.1 compared to viral infection rates
expression level was confirmed by Western blotting and by immunofluo- in A549 cells. However, unlike human influenza viruses, the avian
rescence microscopy with rabbit anti-human IFITM3 antibody (catalog influenza viruses, VN/04 (HPAI H5N1), Anhui/1 (LPAI H7N9),
number PA5-11274; Thermo Fisher Scientific Inc.) at 72 h posttransfec- and CK/11 (LPAI H9N2), could efficiently infect HULEC at an
tion. infection ratio of 0.65 or greater. Interestingly, the H7 viruses

December 2016 Volume 90 Number 24 Journal of Virology jvi.asm.org 11159


Sun et al.

FIG 1 Influenza virus infectivity in HULEC. Influenza viruses at an MOI of 1 to 5, i.e., seasonal PR8 (H1N1), Bris/07 (H1N1), Pan/99 (H3N2) and 2009
pandemic Mex/09 (H1N1), VN/04 (H5N1), A/Anhui/1 (H7N9), CK/11 (H9N2), NL/03 (H7N7), Shov/07 (H7N9), and Anhui:PR8 and VN:PR8, were used to
infect HULEC and A549 cells for 8 h, and viral infectivity was quantified based on the percentage of NP-positive cells by immunofluorescence microscopy
(counting at least 200 cells per infection). (A) Viral infectivity in HULEC and A549 cells. (B) Ratios of viral infectivity in HULEC versus A549 cells. The error bars
represent the standard deviations (SD) of the mean from three independent experiments. Statistical analysis was performed to compare the ratio of viral
infectivity in HULEC versus A549 cells between human and avian influenza viruses by an unpaired t test with Welch’s correction.

NL/03 (HPAI H7N7) and Shov/07 (LPAI H7N9), the latter of rates of both viruses in HULEC reached up to 95% upon inducing
which shares high sequence homology with the HA and NA genes fusion at the cell surface, comparable to their infectivity in A549
from Anhui/1 virus, showed only intermediate infectivity in cells under the same conditions (Fig. 2). These findings indicate
HULEC, as their infectivity in these cells was approximately 35 to that human influenza viruses can bind to endothelial cells and fuse
40% of that observed in A549 cells (Fig. 1). at the cell surface in low-pH environments at efficiencies compa-
To better understand which viral genes are involved in the rable to those in permissive A549 cells, thus providing the means
extended tropism of avian influenza viruses in HULEC, recombi- for the viral genome to enter the cytoplasm and initiate replica-
nant PR8 viruses harboring the HA and NA genes from Anhui/1 tion. Our results from this acid bypass assay confirmed that the
or VN/04 (lacking the polybasic cleavage site) were tested in the infection block to human influenza virus in HULEC occurs down-
viral infection assay. Both recombinant influenza viruses showed stream of viral binding but upstream of viral replication in the
enhanced viral infectivity in HULEC compared to PR8 virus, al- host nucleus.
beit at slightly lower infection rates than wild-type (wt) Anhui/1 Human influenza viruses can become internalized and initi-
or VN/04 virus (Fig. 1). This suggests that the HA and NA genes ate hemifusion in endosomes but fail to uncoat during entry
represent the main determinants of influenza virus tropism in into endothelial cells. During influenza virus entry into host cells,
HULEC. Taken together, our results show that, unlike human the virus first binds to cellular receptors and then becomes inter-
H1N1 and H3N2 viruses, avian influenza viruses of the H5, H7, nalized by endocytosis into endosomal compartments in which
and H9 subtypes have gained extended cellular tropism in human the viral membrane fuses with the endosomal membrane to re-
pulmonary endothelial cells in an apparently viral envelope pro- lease the viral contents into the cytosol and initiate replication.
tein-dependent manner. The fusion between the influenza virus membrane and host endo-
Human influenza viruses can infect endothelial cells by fus- somal membrane starts with lipid mixing (hemifusion), which is
ing at the cell surface. To elucidate the stage of human influenza then followed by fusion pore formation (36). After we determined
virus infection that is blocked in endothelial cells, we first set out that the block in human influenza virus infection in endothelial
to determine whether virus infectivity could be rescued by forcing cells occurs downstream of viral binding but upstream of viral
human influenza viruses to fuse at the endothelial cell surface. For replication in the nucleus, we next performed stepwise entry as-
this purpose, viruses were incubated with endothelial cells on ice says to determine the exact step at which the infection is inhibited.
for 1 h to allow virus binding, and then the cells were exposed to First, we examined whether human influenza virus can bind and
pH 5.0 buffer to force fusion between surface-bound influenza be internalized by endocytosis in HULEC. For this purpose, we
viruses and the host plasma membrane. In this assay approach, if used biotin-labeled PR8 virus in a virus binding and internaliza-
viruses are able to bind and fuse at the cell surface, the viral nu- tion assay as described previously (30). As shown in Fig. 3A (left
cleocapsid will be directly released into the cytosol instead of being column), PR8 virus was able to bind to both HULEC and A549
transported through endocytic pathways, as occurs during normal cells by staining with Alexa 488-streptavidin following virus incu-
viral entry. At 8 h after inducing fusion at the cell surface, viral bation with cells at 4°C for 1 h. This further confirms that the virus
infectivity in the presence of 30 mM NH4Cl, which was used to has no defect in binding to HULEC compared to A549 cells. As
block viral entry via the endocytic pathway, was quantified based expected, the signal from cell surface-bound virions could be
on NP staining. As a control, viral infection in permissive A549 blocked by preincubation with excessive amounts of unconju-
cells was used to demonstrate that PR8 (H1N1) and Pan/99 gated streptavidin (Fig. 3A, second column from left). Upon in-
(H3N2) viruses could achieve approximately 75% and 95% infec- ducing internalization at 37°C for 1 h, internalized viruses stained
tivity, respectively, by inducing fusion at the cell surface upon with Alexa 488-streptavidin following unconjugated streptavidin
exposure to pH 5.0, but not pH 7.4, conditions (Fig. 2). Interest- preincubation were visible in both HULEC and A549 cells (Fig.
ingly, although PR8 and Pan/99 viruses showed only very low 3A, right column), suggesting that PR8 virus can become internal-
infectivity in HULEC by the normal viral entry route, the infection ized into HULEC with an efficiency similar to that in A549 cells.

11160 jvi.asm.org Journal of Virology December 2016 Volume 90 Number 24


Influenza Virus Infection in Pulmonary Endothelial Cells

FIG 2 Human influenza virus infection in HULEC induced by fusing at the cell surface (acid bypass infection). Cell surface-bound PR8 H1N1 and Panama
H3N2 viruses at an MOI of 5 were exposed to fusion buffer (pH 5.0 or 7.4) at 37°C for 2 min, after which the cells were cultured for 8 h in the presence of 30 mM
NH4Cl to block virus fusion in the endocytic pathway. (Left) Immunofluorescence microscopy (20⫻ objective lens). Cells were stained with anti-NP antibody
(green) and DAPI (blue) for nuclei. (Right) Viral infectivity expressed as the mean percentage of NP-positive cells from three independent experiments. The error
bars represent SD. Two-way analysis of variance (ANOVA) was done to compare viral infectivity at pH 5.0 and at pH 7.4, ***, P ⬍ 0.001.

As PR8 virus demonstrated efficient binding and internaliza- this cell line generally similar to those observed for HULEC (data
tion into both HULEC and A549 cells, we next investigated if PR8 not shown).
virus could fuse in the endosomes of endothelial cells. Following Following confirmation of successful viral lipid mixing in en-
previously established dual-wavelength imaging methods to mon- dosomes of endothelial cells, we next monitored uncoating of the
itor influenza virus fusion (31), we labeled purified PR8 virus with nucleocapsid by M1 protein staining with HB64 antibody in the
dual lipophilic dyes, R18 (red) and SP-DiOC18 (green). In this presence of 1 mM cycloheximide to prevent synthesis of new viral
assay, lipid mixing between the viral membrane and host endo- proteins (37). The HB64 antibody recognizes the epitope in M1
somal membrane correlates with the fluorescence color shift from protein, which becomes more accessible once the M1 protein is
red to green due to the release of self-quenching DiOC18 and dispersed into the cytosol (38). In control A549 cells at 3.5 h
dissolution of fluorescent resonance energy transfer (FRET) from postinternalization, PR8 M1 protein displayed bright dispersed
DiOC18 to R18 in the labeled viruses. As shown in Fig. 3B (top cytoplasmic signals, indicating that viruses had undergone un-
row), upon binding at 4°C, cell-bound viruses could be visualized coating events, releasing M1 proteins into the cytosol (Fig. 3C). In
only in the red channel (R18) in HULEC. Upon inducing inter- contrast, in HULEC, the M1 protein exhibited a punctate staining
nalization for 1.5 h, the green fluorescence signal was increased, pattern, suggesting the M1 protein was confined in vacuole-like
indicating that PR8 virus can initiate hemifusion (lipid mixing) in compartments and failed to be released into the cytosol. In order
HULEC (Fig. 3B, middle row). As a negative fusion control, virus to determine the location of M1 proteins in HULEC, we per-
internalization was induced in the presence of 30 mM NH4Cl to formed colocalization immunostaining with an early endosome
inhibit endosomal acidification; as a result, internalized viruses marker (EEA1) and a late endosome/lysosome marker (LAMP-1).
could no longer fuse in HULEC, as demonstrated by the lack of The overlap coefficient of M1 with EEA1 was 0.34, and it was 0.73
green signal in Fig. 3B (bottom row). PR8 virus fusion in A549 for M1 with LAMP-1 (Fig. 3C), demonstrating that PR8 virus M1
cells was included as a positive control, with results obtained in protein was mainly trapped in late endosome/lysosome compart-

December 2016 Volume 90 Number 24 Journal of Virology jvi.asm.org 11161


Sun et al.

ments during entry into HULEC. Taken together, these results


show that the blocking of human influenza virus infection in
HULEC is at a posthemifusion step; as a result, virus can no longer
uncoat nucleocapsid and release the viral genome into the cytosol.
The virus replication restriction factor IFITM3 is constitu-
tively expressed in human endothelial cells but at a significantly
lower level in epithelial cells. The infection block in HULEC at a
posthemifusion stage prompted us to investigate whether the
IFITM3 protein, which has recently been suggested to block
influenza virus infection at the same stage (39), plays a similar role
in influenza virus infection in endothelial cells. IFITM proteins
are generally expressed in cells at a basal level but can be signifi-
cantly induced by type I and type II IFNs (20). We first examined
by Western blotting whether human endothelial cells constitu-
tively express IFITM3 without interferon induction. As shown
in Fig. 4A, cell lysates from endothelial cells, including HULEC,
HMVEC, and HUVEC, showed high levels of endogenous IFITM3
protein expression; HBVEC expressed a much lower level of IFITM3
protein. Compared to endothelial cells, human lung epithelial
cells, including A549 and Calu-3 cells, failed to display IFITM3
protein expression under the same detection conditions, and
NHBE cells had only marginal IFITM3 protein expression com-
pared to that in HULEC.
Using immunofluorescence microscopy, we next examined
the locations of endogenous IFITM3 proteins in HULEC. In this
cell type, IFITM3 showed a punctate staining pattern close to nu-
clear regions. In a colocalization study, we found that the overlap
coefficient between IFITM3 and the early endosome marker EEA1
was 0.76, and it was 0.67 between IFITM3 and the late endosome/
lysosome marker LAMP-1, indicating endogenous IFITM3 is
mainly located in endosomal compartments (Fig. 4B). Taken to-
gether, our results show that, unlike most respiratory epithelial
cells, human pulmonary endothelial cells constitutively express
high levels of IFITM3 in endosomes, which may potentially re-
strict influenza virus infection.
IFITM3 knockdown by siRNA in HULEC partially enhances
human influenza virus infection. To determine whether consti-
tutively expressed IFITM3 proteins are involved in the restriction
of influenza virus infection in HULEC, we used silencing RNA
(siRNA) to knock down IFITM3 expression in HULEC prior to
virus infection. As shown in Fig. 5A, 100 pmol of IFITM3 siRNA
FIG 3 PR8 virus binding, internalization, fusion, and uncoating in HULEC and could significantly downregulate IFITM3 expression, as demon-
A549 cells. (A) Biotin-labeled PR8 virus binding and internalization. Cell surface- strated by Western blotting (Fig. 5A). Following viral infection
bound PR8 virus following binding at 4°C for 1 h was visualized by Alexa-488
with PR8 virus, IFITM3 siRNA-transfected cells showed signifi-
streptavidin (strep) (left column); the signal from surface-bound PR8 viruses
could be blocked by preincubation with unconjugated streptavidin (1 mg/ml) cantly enhanced viral infectivity (P ⬍ 0.001), with approximately
(second column from left). Upon inducing internalization at 37°C for 1 h, the cells 12-fold (21% versus 1.7%) and 8-fold (47% versus 5.8%) in-
were incubated with unconjugated streptavidin to block the signal from surface- creases compared to cells transfected with negative-control siRNA
bound viruses, and then the cells were fixed and permeabilized before being at MOI of 0.25 and 1, respectively (Fig. 5B). However, knocking
stained with Alexa-488 streptavidin for internalized PR8 virus visualization (right
column). Both cell surface-bound and internalized viruses were shown by Alexa- down IFITM3 expression in HULEC had a less significant effect
488 streptavidin staining without unconjugated preincubatioin (third column on Anhui:PR8, as Anhui:PR8 virus infectivity increased from 28%
from left). The images were taken under a 63⫻ oil objective lens. (B) Virus-medi- to 35% at an MOI of 0.25 and increased from 46% to 56% at an
ated lipid mixing in endosomes. R18 (red) and SP-DiOC18 (green) dual-color- MOI of 1 (Fig. 5B). The different restriction effects of endoge-
labeled PR8 viruses induced internalization at 37°C for 1.5 h; the lipid mixing
nously expressed IFITM3 in HULEC on human and avian influ-
(hemifusion) between the viral membrane and the host endosomal membrane
was indicated by increased intensity of green fluorescence due to the release of enza viruses seem to be cell type specific. Thus, IFITM3 proteins
self-quenching DiOC18 and dissolution of FRET. The images were taken under a
63⫻ oil objective lens. (C) (Top) PR8 virus uncoating. PR8 virus M1 proteins were
visualized by immunofluorescence using anti-M1 antibody (HB64) at 3.5 h
postinternalization and DAPI (blue) for nuclei (40⫻ objective lens). (Bottom) pixels) of the green and red signals were calculated with the program Zen from
The PR8 virus M1 proteins (green) in HULEC were colocalized with the early Zeiss and are marked in the right corners of the images. The images were taken
endosome marker EEA1 (red) and the lysosomal marker LAMP-1 (red), and the with an LSM 710 Zeiss inverted confocal microscope under a 40⫻ objective lens
overlap coefficients (ranging from 0 to 1, with 1 representing perfectly colocalized with digital zoom 2. The scale bars represent 10 ␮m.

11162 jvi.asm.org Journal of Virology December 2016 Volume 90 Number 24


Influenza Virus Infection in Pulmonary Endothelial Cells

FIG 4 Constitutive IFITM3 expression in human endothelial cells. (A) IFITM3 expression in endothelial cell lysates (HULEC, HMVEC, HUVEC, and HBVEC)
and epithelial cell lysates (Calu-3, A549, and NHBE cells) was detected by anti-IFITM3 antibody in a Western blot assay; actin expression monitored by
anti-␤-actin antibody blotting was used as an internal loading control. Actin and IFITM3 protein expression levels in the Western blot were quantified with a
ChemiDoc MP system with Image Lab software. The relative IFITM3 protein expression was plotted as the mean ratio of the intensity of the IFITM3 signal to that
of the actin internal control from three independent experiments. (B) Localization of IFITM3 in HULEC. The endogenous IFITM3 proteins (red) in HULEC
were colocalized with the early endosome marker EEA1 (green) and the lysosomal marker LAMP-1 (green), and the overlap coefficients of the green and red
signals were calculated with the program Zen from Zeiss and marked in the right corners of the images. DAPI (blue) was used to stain nuclei. 63⫻ oil objective
lenses were used in microscopy.

overexpressed in A549 cells exhibited similar degrees of inhi- A higher virus fusion threshold is not associated with escape
bition of all the influenza viruses we tested (Fig. 5C), which was from IFITM3 restriction. Thus far, we have shown that human
consistent with the results of a previous study (22). Our results influenza virus entry into endothelial cells is blocked at a pos-
demonstrate that constitutively expressed IFITM3 proteins are themifusion step, partially due to the constitutive expression of
able to selectively restrict human influenza virus infection in IFITM3. However, certain avian influenza viruses, including
endothelial cells but have a less potent effect on certain avian HPAI H5N1, LPAI H9N2, and novel LPAI H7N9 viruses, show
influenza viruses. significantly higher infection rates than H1N1 and H3N2 human

December 2016 Volume 90 Number 24 Journal of Virology jvi.asm.org 11163


Sun et al.

FIG 6 pH fusion thresholds for influenza viruses. Influenza viruses at 128


HAU were incubated with 1% turkey erythrocytes at 4°C for 1 h before fusion
was induced with 100 ␮l of fusion buffer at pHs ranging from 4.8 to 7.4. The
hemolysis was measured as the OD405 from duplicate samples, and the maxi-
mal hemolysis was normalized to 100%. The hemolysis efficiencies at various
pHs were expressed as percentages of the maximal hemolysis. The graph rep-
resents the average hemolysis efficiencies from three independent experi-
ments. The pH thresholds for fusion (the pH range at which 50% of maximal
hemolysis was achieved) are shown in the table. Viral infectivities in HULEC
were classified as high, low, or intermediate based on the viral infection rates
shown in Fig. 1.

FIG 5 Constitutively expressed IFITM3 proteins restrict PR8 virus infection enza virus entry. To test this possibility, we used a virus-mediated
in HULEC. (A) Different amounts (12.5 to 100 pmol) of IFITM3 siRNA and
negative-control siRNA were transfected in HULEC, and the expression of hemolysis assay to compare viral fusion pHs for selected human
IFITM3 was examined at 72 h posttransfection by Western blotting with anti- and avian influenza viruses. As shown in Fig. 6, human H1N1
IFITM3 antibody; anti-actin antibody was included as an internal loading influenza viruses, including PR8, Bris/09, and Mex/09, preferen-
control. (B) HULEC transfected with 100 pmol of IFITM3 siRNA for 72 h were tially fused at a relatively low pH (⬍5.3). In contrast, the viruses
infected with PR8 or Anhui:PR8 virus at MOI of 0.25 and 1 for 8 h before viral
infectivity was quantified based on NP staining by immunofluorescence mi- that displayed the highest infectivity in HULEC (Anhui/13
croscopy. The graph represents the mean infectivities of three independent [H7N9] and VN/04 [H5N1] viruses) had a fusion threshold of
experiments, with the error bars showing SD. Two-way ANOVA statistical approximately pH 5.8 (Fig. 6). The H7 viruses NL/03 (H7N7) and
analysis was performed to compare viral infectivities between IFITM3 siRNA Shov/07 (H7N9), which showed intermediate infection rates in
and negative-control siRNA groups; ***, P ⬍ 0.001; *, P ⬍ 0.05. (C) A549 cells
HULEC, possessed slightly lower pH fusion thresholds (pH 5.5)
stably expressing IFITM3 or vector pQCXIP were infected with the indicated
influenza viruses at an MOI of approximately 1, and viral infectivity at 8 h than the Anhui/13 (H7N9) and VN/04 (H5N1) viruses. However,
postinfection was quantified by NP-positive cells in immunofluorescence mi- human Pan/99 (H3N2) virus shared a similar pH fusion profile
croscopy. Shown are the means of three independent experiments with SD. with VN/04 virus despite the different infection rates of these two
Two-way ANOVA statistical analysis was performed to compare viral infectiv- viruses in HULEC (Fig. 1 and 5). Similarly, CK/11 (H9N2) virus
ities in IFITM3 and vector-expressing A549 cells.
had a pH fusion range similar to that of human H1N1 viruses but
was able to infect HULEC with much higher efficiency than H1N1
virus. Our results suggest that although having a relatively high
influenza viruses in endothelial cells. One hypothesis for how fusion threshold might prompt some avian influenza viruses, like
avian influenza viruses circumvent this infection block is that Anhui/1 and VN/04, to fuse early before reaching late endosomes
avian influenza viruses can fuse in early endosomes at relatively to avoid IFITM3 restriction, other avian influenza viruses, such as
high pH values before traveling to late endosomes, where IFITM3 CK/11 virus, may explore other, unknown routes to escape this
proteins have been suggested to be located and to restrict influ- restriction in endothelial cells.

11164 jvi.asm.org Journal of Virology December 2016 Volume 90 Number 24


Influenza Virus Infection in Pulmonary Endothelial Cells

DISCUSSION showed only intermediate levels of IFITM3 expression. Although


Human pulmonary cells constitute approximately 30% of the to- comprehensive immunohistochemical staining for IFITM3 ex-
tal cell population in the lung, providing the lining for the network pression in human lung tissues is still lacking, various levels of
of capillaries in the alveolus (40). Recently, pulmonary endothe- endogenous IFITM3 expression in mouse respiratory epithelial
lium dysfunction caused by cytokines released from alveolar epi- cells and pulmonary endothelial cells have been revealed (25).
thelial cells and leukocytes has been suggested to play an impor- This suggests that endogenous expression of IFITM3 may be tis-
tant role in virus-induced lung damage in severe cases of influenza sue or cell type specific; accordingly, higher endogenous expres-
virus infection (41). Additionally, given the close proximity of the sion of IFITM3 might provide a certain degree of advantage in
pulmonary endothelium and alveolar epithelium, endothelial protecting the host from influenza virus infection prior to inter-
cells exposed to influenza virus particles released from infected feron production. Although the antiviral activity of IFITM3 has
neighboring epithelial cells may lead to cell death, increased en- been well established, the exact mechanism by which IFITM3 re-
dothelial permeability, and vascular destruction, further contrib- stricts viral infection is not yet fully understood. Early evidence
uting to epithelial-endothelial barrier damage (41). However, pre- suggests that IFITM3 restricts viral-membrane hemifusion, pos-
vious studies in vitro have shown that pulmonary endothelial cells sibly by affecting the membrane’s molecular order and fluidity
are not susceptible to infection with most human influenza vi- (43) or by modulating cholesterol homeostasis in the endosome
ruses, as only HPAI H5N1 viruses were able to replicate efficiently by interacting with VAPA (vesicle-associated membrane protein-
and induce excessive production of proinflammatory cytokines associated protein A), resulting in inhibition of viral fusion (44).
(17, 19). Here, we extend previous work on influenza virus tro- However, it was subsequently found that IFITM3 restricts viral
infection by blocking the formation of fusion pores following vi-
pism in human pulmonary endothelial cells by identifying cellular
rus-endosome hemifusion, possibly by modulating endosomal-
and viral factors that contribute to viral infection in this cell type.
membrane rigidity (39). Furthermore, IFITM3 subcellular local-
We showed that human pulmonary endothelial HULEC are less
izations, which can be modulated by protein posttranslational
permissive to human influenza virus infection than a subset of
modification, has proven to be important for its antiviral activi-
avian influenza viruses, including highly pathogenic H5N1 (VN/
ties, and depending on the cell types and stimulation status,
04), LPAI H7N9, and H9N2 viruses. Despite limited expression of
IFITM3 may localize differently in cells (45). In unstimulated
␣-2,6-linked sialic acid residues (17), the receptor for human in-
WI-38 human primary fibroblasts, the majority of IFITM3 resides
fluenza viruses, these viruses are still able to bind to endothelial
in the ER and becomes distributed in a vesicular pattern through-
cells with efficiency comparable to that of binding on epithelial
out the cell upon IFN exposure (22). When overexpressed in A549
cells. This was shown not only by the biotin-labeled-virus binding cells, IFITM3 predominantly localizes in enlarged compartments
assay (Fig. 3A), but also by an acid bypass infection assay (Fig. 2). shared with markers of endosomes and lysosomes (23). In our
We further showed that human PR8 virus could be internalized study, we showed that the constitutively expressed IFITM3 in
and successfully initiate hemifusion in endothelial cells but could HULEC mainly localized in endosomal and lysosomal compart-
not uncoat and release the viral genome into the cytosol to initiate ments, as indicated by their colocalization with the early endo-
subsequent replication processes. Although human influenza vi- some and late endosome/lysosome markers EEA1 and LAMP-1,
rus infection is blocked at the early stages of viral entry in endo- respectively, suggesting that endogenous IFITM3 proteins in
thelial cells, this does not necessarily mean that the pulmonary HULEC already reside in the compartments involved in the influ-
endothelium cannot be activated through direct contact with hu- enza virus entry pathway and might be intrinsically primed for
man influenza viruses. It has been previously reported that a UV- blocking influenza virus infection. However, we did not observe
inactivated, replication-deficient human seasonal H3N2 virus was IFITM3-enriched large compartments, as previously revealed in
capable of causing a significant reduction in endothelial permea- overexpressed or IFN-induced A549 cells; whether this reflects a
bility by inducing degradation of the tight-junction protein Clau- difference between cell types or between endogenously expressed
din 5. This was accomplished without causing cytotoxic effects on versus overexpressed IFITM3 requires further investigation (39).
the endothelium, and binding of the virus alone was not sufficient In our study, we demonstrated that PR8 viral infection in HULEC
to induce permeability changes (14). We postulate that host pat- was blocked after a hemifusion step. This suggests a potential role
tern recognition receptors (PRR) residing in host endocytic path- for endosome-residing constitutively expressed IFITM3 proteins
ways, such as Toll-like receptor 7 (TLR7) or TLR8 in endosomes, in this block, based on our findings that knocking down IFITM3
might be involved in recognizing viral components and activating expression with siRNA significantly improved PR8 virus infectiv-
signal transductions in endothelial cells in a viral-replication-in- ity in HULEC. However, the improved infectivity in HULEC upon
dependent manner (42). Despite the fact that avian influenza vi- IFITM3 siRNA treatment is still lower than that in A549 cells,
ruses, such as HPAI H5N1 and LPAI H7N9, were able to replicate suggesting a role for other restriction factors in endothelial cells.
efficiently in endothelial cells and to induce strong cytokine pro- Moreover, a subset of avian influenza viruses from the H5, H7,
duction in vitro, further studies are needed to elucidate their exact and H9 subtypes seemed to be able to partially escape this IFITM3
role in viral pathogenesis in vivo, as postmortem analysis of pa- restriction block in endothelial cells, as IFITM3 siRNA knock-
tients who succumbed to H5N1 infection rarely showed endothe- down had only a moderate effect on improving the infectivity of a
liotropism (6). recombinant PR8 virus bearing the Anhui/1 H7N9 HA and NA.
In this study, we revealed that, unlike human bronchial epithe- Future studies exploring whether interferon-induced IFITM3 in
lial Calu-3 and alveolar basal epithelium-derived A549 cells, hu- pulmonary endothelial cells, which may have different subcellular
man primary (HMVEC) or transformed (HULEC) pulmonary locations, has an effect on avian influenza virus infection similar
endothelial cells, as well as HUVEC, constitutively express high to that of constitutively expressed IFITM3 are warranted.
levels of IFITM3 without IFN induction, whereas NHBE cells Most influenza viruses have a pH fusion range of 5.0 to 5.5,

December 2016 Volume 90 Number 24 Journal of Virology jvi.asm.org 11165


Sun et al.

which matches the pH in the late endosome (46). However, cer- Roizman B, Straus SE (ed), Fields virology, 5th ed. Lippincott Williams &
tain influenza viruses, such as the novel H7N9 virus Anhui/1, have Wilkins, Philadelphia, PA.
2. Kuiken T, Riteau B, Fouchier RA, Rimmelzwaan GF. 2012. Pathogenesis
a higher pH threshold (pH 5.8) for fusion; such viruses may be of influenza virus infections: the good, the bad and the ugly. Curr Opin
able to fuse in early endosomes, which have pH values ranging Virol 2:276 –286. http://dx.doi.org/10.1016/j.coviro.2012.02.013.
from 5.4 to 6.2, depending on the cell type (47, 48). IFITM3 is 3. Poovorawan Y, Pyungporn S, Prachayangprecha S, Makkoch J. 2013.
mostly located in late endosomes or lysosomes when overex- Global alert to avian influenza virus infection: from H5N1 to H7N9. Pat-
pressed or upon stimulation by IFN (23, 49) and has less inhibi- hog Glob Health 107:217–223. http://dx.doi.org/10.1179/2047773213Y
.0000000103.
tory effects on viruses, such as vesicular stomatitis virus (VSV), 4. Liu Q, Liu DY, Yang ZQ. 2013. Characteristics of human infection with
that preferentially fuse at early endosomes or late endosome/mul- avian influenza viruses and development of new antiviral agents. Acta
tivesicular bodies (50); therefore, IFITM3 might be less potent in Pharmacol Sin 34:1257–1269. http://dx.doi.org/10.1038/aps.2013.121.
restricting influenza viruses, which can fuse in early endosomes. 5. WHO. 2016. Monthly risk assessment summary: influenza at the human-
animal interface. http://www.who.int/influenza/human_animal_interface
In our study, we did observe that VN/04 H5N1 and Anhui/13
/HAI_Risk_Assessment/en. Accessed 16 May 2016.
H7N9 viruses, which showed efficient viral replication in HULEC, 6. Gu J, Xie Z, Gao Z, Liu J, Korteweg C, Ye J, Lau LT, Lu J, Gao Z, Zhang
had a higher pH threshold (pH ⬎ 5.5) for fusion, indicating that B, McNutt MA, Lu M, Anderson VM, Gong E, Yu AC, Lipkin WI. 2007.
the viruses could potentially escape IFITM3 restriction by fusing H5N1 infection of the respiratory tract and beyond: a molecular pathology
early, before reaching late endosomes. However, CK/11 H9N2 study. Lancet 370:1137–1145. http://dx.doi.org/10.1016/S0140-6736(07)
61515-3.
virus was able to achieve high infectivity in HULEC despite having 7. Perrone LA, Tumpey TM. 2007. Reconstruction of the 1918 pandemic
a low pH fusion threshold, suggesting that certain influenza vi- influenza virus: how revealing the molecular secrets of the virus responsi-
ruses may have developed different strategies to counteract ble for the worst pandemic in recorded history can guide our response to
IFITM3 restriction in endothelial cells. future influenza pandemics. Infect Disord Drug Targets 7:294 –303. http:
Altogether, we demonstrated that human influenza virus in- //dx.doi.org/10.2174/187152607783018772.
8. Yeh E, Luo RF, Dyner L, Hong DK, Banaei N, Baron EJ, Pinsky BA.
fection in human pulmonary endothelial cells is less efficient than 2010. Preferential lower respiratory tract infection in swine-origin 2009
that of selected strains of H5, H7, and H9 avian influenza viruses, A(H1N1) influenza. Clin Infect Dis 50:391–394. http://dx.doi.org/10
partially due to the high constitutive expression of IFITM3 in late .1086/649875.
endosomes and lysosomes, which may block the procession from 9. Gao R, Bhatnagar J, Blau DM, Greer P, Rollin DC, Denison AM,
Deleon-Carnes M, Shieh WJ, Sambhara S, Tumpey TM, Patel M, Liu L,
hemifusion to fusion pore formation during the viral entry pro- Paddock C, Drew C, Shu Y, Katz JM, Zaki SR. 2013. Cytokine and
cess. In comparison, certain avian influenza viruses were able to chemokine profiles in lung tissues from fatal cases of 2009 pandemic in-
escape the restriction in endothelial cells by fusing early in the fluenza A (H1N1): role of the host immune response in pathogenesis. Am
endocytic pathway at a higher pH value or by other, unknown J Pathol 183:1258 –1268. http://dx.doi.org/10.1016/j.ajpath.2013.06.023.
mechanisms. However, we cannot rule out the possibility that 10. Herold S, Becker C, Ridge KM, Budinger GR. 2015. Influenza virus-
induced lung injury: pathogenesis and implications for treatment. Eur
other, unidentified restriction factors may exist in human pulmo- Respir J 45:1463–1478. http://dx.doi.org/10.1183/09031936.00186214.
nary endothelial cells that confer their resistance to influenza virus 11. Fukuyama S, Kawaoka Y. 2011. The pathogenesis of influenza virus
infection. An additional factor to be mentioned is the binding infections: the contributions of virus and host factors. Curr Opin Immu-
preference of the laboratory-adapted human influenza PR8 virus, nol 23:481– 486. http://dx.doi.org/10.1016/j.coi.2011.07.016.
12. Short KR, Kroeze EJ, Fouchier RA, Kuiken T. 2014. Pathogenesis of
with ␣-2,6- and ␣-2,3-linked sialic acid dual receptor binding influenza-induced acute respiratory distress syndrome. Lancet Infect Dis
specificity. This virus was able to bind endothelial cells (followed 14:57– 69. http://dx.doi.org/10.1016/S1473-3099(13)70286-X.
by internalization of the virus), but whether PR8 viruses are trans- 13. Teijaro JR, Walsh KB, Cahalan S, Fremgen DM, Roberts E, Scott F,
ported into the same endocytic compartments as avian influenza Martinborough E, Peach R, Oldstone MB, Rosen H. 2011. Endothelial
viruses, which preferentially bind to ␣-2,3-linked sialic acids, is cells are central orchestrators of cytokine amplification during influenza
virus infection. Cell 146:980 –991. http://dx.doi.org/10.1016/j.cell.2011
unknown. Previously, it had been suggested that HPAI H5N1 in- .08.015.
fluenza viruses with different receptor binding specificities might 14. Armstrong SM, Wang C, Tigdi J, Si X, Dumpit C, Charles S, Gamage
be sensed or recognized differently in human monocyte-derived A, Moraes TJ, Lee WL. 2012. Influenza infects lung microvascular endo-
macrophages and dendritic cells, subsequently leading to distinct thelium leading to microvascular leak: role of apoptosis and claudin-5.
PLoS One 7:e47323. http://dx.doi.org/10.1371/journal.pone.0047323.
signaling cascades (51). Whether influenza virus receptor speci- 15. Hiyoshi M, Indalao IL, Yano M, Yamane K, Takahashi E, Kido H. 2015.
ficity is involved in human pulmonary endothelial cell restriction Influenza A virus infection of vascular endothelial cells induces GSK-
to human influenza viruses requires further study. 3beta-mediated beta-catenin degradation in adherens junctions, with a
resultant increase in membrane permeability. Arch Virol 160:225–234.
ACKNOWLEDGMENTS http://dx.doi.org/10.1007/s00705-014-2270-5.
16. Narasaraju T, Yang E, Samy RP, Ng HH, Poh WP, Liew AA, Phoon
The findings and conclusions in this report are ours and do not necessarily MC, van Rooijen N, Chow VT. 2011. Excessive neutrophils and neutro-
represent the official position of the Centers for Disease Control and Pre- phil extracellular traps contribute to acute lung injury of influenza pneu-
vention. monitis. Am J Pathol 179:199 –210. http://dx.doi.org/10.1016/j.ajpath
X. Sun was supported by the Oak Ridge Institute for Science and Ed- .2011.03.013.
ucation. 17. Zeng H, Pappas C, Belser JA, Houser KV, Zhong W, Wadford DA,
Stevens T, Balczon R, Katz JM, Tumpey TM. 2012. Human pulmonary
FUNDING INFORMATION microvascular endothelial cells support productive replication of highly
This work, including the efforts of Xiangjie Sun, was funded by HHS | pathogenic avian influenza viruses: possible involvement in the pathogen-
esis of human H5N1 virus infection. J Virol 86:667– 678. http://dx.doi.org
Centers for Disease Control and Prevention (CDC). /10.1128/JVI.06348-11.
18. Chan MC, Chan RW, Yu WC, Ho CC, Chui WH, Lo CK, Yuen KM,
REFERENCES Guan YI, Nicholls JM, Peiris JS. 2009. Influenza H5N1 virus infection of
1. Palese PSM. 2007. Orthomyxoviridae: the viruses and their replication, p polarized human alveolar epithelial cells and lung microvascular endothe-
1647–1689 In Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA, lial cells. Respir Res 10:102. http://dx.doi.org/10.1186/1465-9921-10-102.

11166 jvi.asm.org Journal of Virology December 2016 Volume 90 Number 24


Influenza Virus Infection in Pulmonary Endothelial Cells

19. Ocana-Macchi M, Bel M, Guzylack-Piriou L, Ruggli N, Liniger M, mediated endocytosis during vesicular stomatitis virus entry into host
McCullough KC, Sakoda Y, Isoda N, Matrosovich M, Summerfield A. cells. Virology 338:53– 60. http://dx.doi.org/10.1016/j.virol.2005.05.006.
2009. Hemagglutinin-dependent tropism of H5N1 avian influenza virus 34. Manders EMM, Verbeek FJ, Aten JA. 1993. Measurement of co-
for human endothelial cells. J Virol 83:12947–12955. http://dx.doi.org/10 localization of objects in dual-colour confocal images. J Microsc 169:375–
.1128/JVI.00468-09. 382. http://dx.doi.org/10.1111/j.1365-2818.1993.tb03313.x.
20. Friedman RL, Manly SP, McMahon M, Kerr IM, Stark GR. 1984. 35. Shelton H, Roberts KL, Molesti E, Temperton N, Barclay WS. 2013.
Transcriptional and posttranscriptional regulation of interferon-induced Mutations in haemagglutinin that affect receptor binding and pH stability
gene expression in human cells. Cell 38:745–755. http://dx.doi.org/10 increase replication of a PR8 influenza virus with H5 HA in the upper
.1016/0092-8674(84)90270-8. respiratory tract of ferrets and may contribute to transmissibility. J Gen
21. Hickford D, Frankenberg S, Shaw G, Renfree MB. 2012. Evolution of Virol 94:1220 –1229. http://dx.doi.org/10.1099/vir.0.050526-0.
vertebrate interferon inducible transmembrane proteins. BMC Genomics 36. Razinkov VI, Melikyan GB, Cohen FS. 1999. Hemifusion between cells
13:155. http://dx.doi.org/10.1186/1471-2164-13-155. expressing hemagglutinin of influenza virus and planar membranes can pre-
22. Brass AL, Huang IC, Benita Y, John SP, Krishnan MN, Feeley EM, Ryan cede the formation of fusion pores that subsequently fully enlarge. Biophys J
BJ, Weyer JL, van der Weyden L, Fikrig E, Adams DJ, Xavier RJ, Farzan 77:3144 –3151. http://dx.doi.org/10.1016/S0006-3495(99)77144-4.
M, Elledge SJ. 2009. The IFITM proteins mediate cellular resistance to 37. Banerjee I, Yamauchi Y, Helenius A, Horvath P. 2013. High-content
influenza A H1N1 virus, West Nile virus, and dengue virus. Cell 139:1243– analysis of sequential events during the early phase of influenza A virus
1254. http://dx.doi.org/10.1016/j.cell.2009.12.017. infection. PLoS One 8:e68450. http://dx.doi.org/10.1371/journal.pone
23. Huang IC, Bailey CC, Weyer JL, Radoshitzky SR, Becker MM, Chiang .0068450.
JJ, Brass AL, Ahmed AA, Chi X, Dong L, Longobardi LE, Boltz D, Kuhn 38. Martin K, Helenius A. 1991. Transport of incoming influenza virus nu-
JH, Elledge SJ, Bavari S, Denison MR, Choe H, Farzan M. 2011. Distinct cleocapsids into the nucleus. J Virol 65:232–244.
patterns of IFITM-mediated restriction of filoviruses, SARS coronavirus, 39. Desai TM, Marin M, Chin CR, Savidis G, Brass AL, Melikyan GB. 2014.
and influenza A virus. PLoS Pathog 7:e1001258. http://dx.doi.org/10.1371 IFITM3 restricts influenza A virus entry by blocking the formation of
/journal.ppat.1001258. fusion pores following virus-endosome hemifusion. PLoS Pathog 10:
24. Jiang D, Weidner JM, Qing M, Pan XB, Guo H, Xu C, Zhang X, Birk e1004048. http://dx.doi.org/10.1371/journal.ppat.1004048.
A, Chang J, Shi PY, Block TM, Guo JT. 2010. Identification of five 40. Piantadosi CA, Schwartz DA. 2004. The acute respiratory distress syn-
interferon-induced cellular proteins that inhibit west Nile virus and den- drome. Ann Intern Med 141:460 – 470. http://dx.doi.org/10.7326/0003
gue virus infections. J Virol 84:8332– 8341. http://dx.doi.org/10.1128/JVI -4819-141-6-200409210-00012.
.02199-09. 41. Armstrong SM, Darwish I, Lee WL. 2013. Endothelial activation and
25. Bailey CC, Huang IC, Kam C, Farzan M. 2012. Ifitm3 limits the severity dysfunction in the pathogenesis of influenza A virus infection. Virulence
of acute influenza in mice. PLoS Pathog 8:e1002909. http://dx.doi.org/10 4:537–542. http://dx.doi.org/10.4161/viru.25779.
.1371/journal.ppat.1002909. 42. Ichinohe T, Pang IK, Iwasaki A. 2010. Influenza virus activates inflam-
26. Everitt AR, Clare S, Pertel T, John SP, Wash RS, Smith SE, Chin CR, masomes via its intracellular M2 ion channel. Nat Immunol 11:404 – 410.
Feeley EM, Sims JS, Adams DJ, Wise HM, Kane L, Goulding D, Digard http://dx.doi.org/10.1038/ni.1861.
P, Anttila V, Baillie JK, Walsh TS, Hume DA, Palotie A, Xue Y, 43. Li K, Markosyan RM, Zheng YM, Golfetto O, Bungart B, Li M, Ding S,
Colonna V, Tyler-Smith C, Dunning J, Gordon SB, Gen II, Investiga- He Y, Liang C, Lee JC, Gratton E, Cohen FS, Liu SL. 2013. IFITM
tors M, Smyth RL, Openshaw PJ, Dougan G, Brass AL, Kellam P. 2012. proteins restrict viral membrane hemifusion. PLoS Pathog 9:e1003124.
IFITM3 restricts the morbidity and mortality associated with influenza. http://dx.doi.org/10.1371/journal.ppat.1003124.
Nature 484:519 –523. http://dx.doi.org/10.1038/nature10921. 44. Amini-Bavil-Olyaee S, Choi YJ, Lee JH, Shi M, Huang IC, Farzan M,
27. Wang Z, Zhang A, Wan Y, Liu X, Qiu C, Xi X, Ren Y, Wang J, Dong Jung JU. 2013. The antiviral effector IFITM3 disrupts intracellular cho-
Y, Bao M, Li L, Zhou M, Yuan S, Sun J, Zhu Z, Chen L, Li Q, Zhang lesterol homeostasis to block viral entry. Cell Host Microbe 13:452– 464.
Z, Zhang X, Lu S, Doherty PC, Kedzierska K, Xu J. 2014. Early hyper- http://dx.doi.org/10.1016/j.chom.2013.03.006.
cytokinemia is associated with interferon-induced transmembrane pro- 45. Chesarino NM, McMichael TM, Yount JS. 2014. Regulation of the traffick-
tein-3 dysfunction and predictive of fatal H7N9 infection. Proc Natl Acad ing and antiviral activity of IFITM3 by post-translational modifications. Fu-
Sci U S A 111:769 –774. http://dx.doi.org/10.1073/pnas.1321748111. ture Microbiol 9:1151–1163. http://dx.doi.org/10.2217/fmb.14.65.
28. Zeng H, Pappas C, Katz JM, Tumpey TM. 2011. The 2009 pandemic 46. Sorkin A, Von Zastrow M. 2002. Signal transduction and endocytosis:
H1N1 and triple-reassortant swine H1N1 influenza viruses replicate effi- close encounters of many kinds. Nat Rev Mol Cell Biol 3:600 – 614. http:
ciently but elicit an attenuated inflammatory response in polarized human //dx.doi.org/10.1038/nrm883.
bronchial epithelial cells. J Virol 85:686 – 696. http://dx.doi.org/10.1128 47. Murphy RF, Powers S, Cantor CR. 1984. Endosome pH measured in
/JVI.01568-10. single cells by dual fluorescence flow cytometry: rapid acidification of
29. Sun X, Cao W, Pappas C, Liu F, Katz JM, Tumpey TM. 2014. Effect of insulin to pH 6. J Cell Biol 98:1757–1762. http://dx.doi.org/10.1083/jcb.98
receptor binding specificity on the immunogenicity and protective effi- .5.1757.
cacy of influenza virus A H1 vaccines. Virology 464-465:156 –165. http: 48. Rybak SL, Murphy RF. 1998. Primary cell cultures from murine kidney
//dx.doi.org/10.1016/j.virol.2014.07.004. and heart differ in endosomal pH. J Cell Physiol 176:216 –222.
30. Sun X, Whittaker GR. 2007. Role of the actin cytoskeleton during influ- 49. Feeley EM, Sims JS, John SP, Chin CR, Pertel T, Chen LM, Gaiha GD,
enza virus internalization into polarized epithelial cells. Cell Microbiol Ryan BJ, Donis RO, Elledge SJ, Brass AL. 2011. IFITM3 inhibits influ-
9:1672–1682. http://dx.doi.org/10.1111/j.1462-5822.2007.00900.x. enza A virus infection by preventing cytosolic entry. PLoS Pathog
31. Sakai T, Ohuchi M, Imai M, Mizuno T, Kawasaki K, Kuroda K, 7:e1002337. http://dx.doi.org/10.1371/journal.ppat.1002337.
Yamashina S. 2006. Dual wavelength imaging allows analysis of mem- 50. Weidner JM, Jiang D, Pan XB, Chang J, Block TM, Guo JT. 2010.
brane fusion of influenza virus inside cells. J Virol 80:2013–2018. http://dx Interferon-induced cell membrane proteins, IFITM3 and tetherin, inhibit
.doi.org/10.1128/JVI.80.4.2013-2018.2006. vesicular stomatitis virus infection via distinct mechanisms. J Virol 84:
32. Stauffer S, Feng Y, Nebioglu F, Heilig R, Picotti P, Helenius A. 2014. 12646 –12657. http://dx.doi.org/10.1128/JVI.01328-10.
Stepwise priming by acidic pH and a high K⫹ concentration is required 51. Ramos I, Bernal-Rubio D, Durham N, Belicha-Villanueva A, Lowen
for efficient uncoating of influenza A virus cores after penetration. J Virol AC, Steel J, Fernandez-Sesma A. 2011. Effects of receptor binding spec-
88:13029 –13046. http://dx.doi.org/10.1128/JVI.01430-14. ificity of avian influenza virus on the human innate immune response. J
33. Sun X, Yau VK, Briggs BJ, Whittaker GR. 2005. Role of clathrin- Virol 85:4421– 4431. http://dx.doi.org/10.1128/JVI.02356-10.

December 2016 Volume 90 Number 24 Journal of Virology jvi.asm.org 11167

Potrebbero piacerti anche