Você está na página 1de 6

Journal of General Virology (2007), 88, 1275–1280 DOI 10.1099/vir.0.

82423-0

Short Differential onset of apoptosis in influenza A virus


Communication H5N1- and H1N1-infected human blood
macrophages
Chris K. P. Mok,1 Davy C. W. Lee,1 Chung-Yan Cheung,2 Malik Peiris2
and Allan S. Y. Lau1
Correspondence 1
Immunology Research Laboratory, Department of Paediatrics and Adolescent Medicine,
Allan S. Y. Lau Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR,
asylau@hkucc.hku.hk China
2
Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong,
Pok Fu Lam, Hong Kong SAR, China

Pathogenesis of the highly pathogenic avian influenza virus A/Hong Kong/483/97 (H5N1/97)
remains to be investigated. It was demonstrated recently that H5N1 dysregulation of
proinflammatory cytokines in human macrophages is a p38-kinase-dependent process. The
results indicated that macrophages may play a role in disease severity. To investigate cellular
responses to H5N1 infection further, apoptosis and its related pathways were studied in primary
blood macrophages. Here, it is shown that the H5N1/97 virus triggered apoptosis, including
caspases and PARP activation, in infected macrophages with a delayed onset compared with
H1N1 counterparts. Similar results were also found in human macrophages infected by
precursors of the H5N1/97 virus. Thus, these results showed that the delay in apoptosis onset
in macrophages infected by H5N1/97 and its related precursor subtypes may be a means for
Received 28 July 2006 the pathogens to have longer survival in the cells; this may contribute to the pathogenesis of
Accepted 14 December 2006 H5N1 disease in humans.

The H5N1/97 ‘bird-flu’ incident was the first documented cytokine dysregulation contributes to pathogenesis and
direct transmission of an avian influenza virus to humans, severity of the disease (Fisman, 2000; Headley et al., 1997;
causing devastating infections with severe viral pneumonia To et al., 2001). In delineating the mechanisms of cytokine
and a mortality rate of .30 % (Claas et al., 1998; Subbarao dysregulation, we recently reported that p38K, a mitogen-
et al., 1998; Yuen et al., 1998). The H5N1 virus continued activated protein kinase (MAPK), plays a significant role
to disseminate among migratory birds and led to a wide- in the hyperinduction of tumour necrosis factor alpha
spread outbreak in domestic poultry around the world. (TNF-a) in H5N1-infected macrophages (Lee et al., 2005).
It had caused over 200 human cases as of May 2006 In addition, a recent study showed that H5N1-infected
(WHO avian influenza information; http://www.who.int/ macrophages enhance TNF-related apoptosis-inducing
csr/disease/avian_influenza/en/). The increasing incidence ligand (TRAIL)-induced apoptosis in Jurkat T cells (Zhou
of avian-to-human transmission provides an opportunity et al., 2006). These recent reports indicate that human
for these highly pathogenic avian influenza viruses to adapt blood macrophages may play a critical role in the patho-
to the host environment, which may result in reassortment genesis of H5N1 infection.
of genetic materials between avian and human influenza Among cellular responses against invading viruses, induc-
viruses. Generation of such newly reassorted influenza tion of apoptosis has been postulated to be one of the most
virus may lead to a potential pandemic threat (WHO avian effective host defence mechanisms. The cell-death process
influenza information; http://www.who.int/csr/disease/ results in inhibition of virus replication, limitation of virus
avian_influenza/en/). dissemination and minimization of uncontrolled inflam-
Previously, we demonstrated that H5N1/97 viruses, in matory responses. It has been shown that influenza virus
contrast to human influenza A virus subtypes including induces apoptosis in vivo and in vitro (Brydon et al., 2005;
H1N1 and H3N2, induce high levels of proinflammatory Fesq et al., 1994; Hinshaw et al., 1994; Takizawa et al., 1993,
cytokines in differentiated primary human blood macro- 1999). However, the functional role of influenza virus-
phages (Cheung et al., 2002). It was suggested that this induced apoptosis is still not well defined. To gain insights
into the virulence of the highly pathogenic avian influenza
Supplementary figures are available in JGV Online. virus, together with an understanding of the cellular

0008-2423 G 2007 SGM Printed in Great Britain 1275


C. K. P. Mok and others

responses of macrophages during microbial infection, we Pharmingen), an essential factor in the initiation of
studied the mechanisms of apoptosis in primary blood apoptosis (Soldani & Scovassi, 2002), at 6, 12 and 18 h
macrophages infected with H5N1 (483/97) or human post-infection by using Western analysis (Fig. 1d). Over the
influenza virus H1N1 (54/98). time course, there was a 4- or 8-fold increase in the levels of
the cleaved PARP fragment in H5N1-infected cells at 12 or
Primary blood macrophages were isolated from mono-
18 h post-infection, respectively, compared with the mock-
nuclear cells of healthy blood donors as described pre-
treated cells (Fig. 1d, lanes 6 and 7). In contrast, there
viously (Lee et al., 2005). The cells were mock-treated or
was an 18- or 22-fold increase in PARP fragment levels
infected with influenza viruses (H5N1 or H1N1) at an
m.o.i. of 2 for 30 min and harvested at indicated time in H1N1-infected cells at the corresponding time point
points for analysis. The infectivity of H5N1 and H1N1 (Fig. 1d, lanes 3 and 4). As PARP is the downstream target
viruses on human macrophages was examined by immuno- of the caspase cascade, we then measured the activity of
fluorescent staining using monoclonal antibodies specific two important apoptotic markers, caspases 3 and 8, by
for the nucleoprotein of influenza viruses (DAKO). Western analysis using antibodies from Cell Signaling
Positive staining for the nucleoprotein in influenza virus- Technology and Upstate Biotech, respectively. Our results
infected macrophages was found at 8 h post-infection showed that the cleaved fragment of caspase 3 was barely
(data not shown). Additionally, cell death, as demonstrated detectable in cells infected with H5N1 (Fig. 2a, lane 9) and
by nuclear condensation or nuclear fragmentation, was was not detected in those infected with H9N2/G1 (data
determined by staining the cells with 4,6-diamidine-2- not shown) at 12 h post-infection. In contrast, levels of
phenylindole dihydrochloride (DAPI) at indicated time activated caspase 3 were increased significantly in H1N1-
points post-infection. As shown, the number of dead cells infected cells at 10 h post-infection and persisted at 12 h
was lower in those cells infected by the H5N1 virus than in (Fig. 2a, lanes 4–5). Consistent with the caspase 3 findings,
cells infected by its H1N1 counterpart (Fig. 1a). H5N1 did not induce the degradation of procaspase 8
strongly at 12 h post-infection compared with H1N1
A previous report showed that replication of influenza (Fig. 2b, lanes 9 and 5).
virus is necessary to trigger apoptosis in infected cells
(Price et al., 1997). We thus measured the viral titres of We further examined the functional role of caspase 8 in
H5N1 or H1N1 in the infected macrophages at 6, 10, 18 influenza virus-induced apoptosis by treating virus-infected
and 24 h post-infection. There was no significant difference cells with a specific inhibitor for caspase 8, Z-IETD-FMK
between the viral titres in the macrophages infected by (Takizawa et al., 1999). Following caspase 8 inhibition, our
H1N1, H5N1 (Fig. 1b) or A/Quail/Hong Kong/G1/97 results showed that the level of the cleaved PARP fragment in
(H9N2/G1), the precursor of H5N1/97 (see Supplementary H5N1- or H1N1-infected macrophages was reduced sig-
Fig. S1, available in JGV Online). Hence, the delayed onset nificantly, by 82 and 86 %, respectively, compared with the
of apoptosis in H5N1-infected macrophages was not due to corresponding samples without inhibitor treatment (Fig. 2c,
replication of the avian virus. Furthermore, we performed lanes 3 and 5). Therefore, the differential onset of cell death
kinetic studies to measure the level of the viral genes for the in H5N1 and H1N1 infection was associated with the delayed
polymerase protein (PA) and nucleoprotein (NP) in activation of the caspase cascade.
H5N1- or H1N1-infected macrophages by using a quanti- To investigate the characteristics of the onset of apoptosis
tative RT-PCR assay. Consistent with the virus titration in avian influenza virus infection further, we measured the
results, we did not find any significant differences in the levels of PARP activation in macrophages infected with
transcription levels of PA and NP in H5N1- or H1N1- precursors of H5N1/97, including H9N2/G1, A/Teal/HK/
infected macrophages at 0, 6 or 10 h post-infection (see W312/97 (H6N1) and A/Goose/Guangdong/1/96 (H5N1/
Supplementary Fig. S2, available in JGV Online). 437.6) (Guan et al., 1999; Hoffmann et al., 2000; Subbarao
We next examined the ultrastructural features of H5N1- & Shaw, 2000; Xu et al., 1999), and human influenza viruses,
and H1N1-infected cells at 12 h post-infection under including H1N1 and H3N2, at 18 h post-infection (Fig. 3a,
a transmission electron microscope (Philips EM208S). b). The level of activated PARP in macrophages infected with
The cellular morphology of H5N1-infected macrophages the H5N1/97 virus or its precursors was lower than that in
(Fig. 1c, middle) was comparable to that of mock-treated H1N1- or H3N2-infected cells. Our findings suggested that
cells at 12 h post-infection (Fig. 1c, right). In contrast, macrophages infected with H5N1/97 or its precursors
H1N1-infected cells showed characteristics of apoptotic undergo slower kinetics of apoptotic responses compared
cells, including nuclear condensation and chromatin with H1N1- or H3N2-infected cells. In addition, H9N2/G1
adherence to the nuclear membrane (Fig. 1c, left). Our induced the cleavage of PARP, but not that of caspase 3,
results demonstrated a differential onset of cell death in suggesting that H9N2/G1 activates PARP through a caspase
macrophages infected with H5N1 compared with those 3-indpendent pathway (Hong et al., 2004).
infected with the H1N1 virus.
The detailed mechanisms of delayed onset of apoptosis
We investigated the apoptotic pathways in virus-infected occurring in avian influenza virus-infected human macro-
macrophages further by measuring the activation of phages remain to be investigated. It is plausible that the
caspase-activated poly(ADP–ribose) polymerase (PARP; infected macrophages cannot recognize the avian influenza

1276 Journal of General Virology 88


Delayed onset of apoptosis in H5N1-infected macrophages

(a) 60 (b) 6

Virus titre [log10 (TCID50 ml 1)]


H1N1

_
50 5 H5N1

Cell death (%)


40 4

30 3

20 2

10 1

H1N1 H5N1 Mock 6 10 18 24


Time (h)

(c)

CC

H1N1 H5N1 Mock

(d) Mock H1N1 H5N1


_
Time post-infection (h) 6 12 18 6 12 18
Pro-PARP
Cleaved PARP

Fold induction (1.00) (1.30) (17.41) (21.71) (0.15) (4.36) (8.37)

Actin
(1) (1) (1) (1) (1) (1) (1)

1 2 3 4 5 6 7

Fig. 1. H5N1/97-infected macrophages show less cell death and apoptosis than H1N1-infected macrophages. (a) Primary
human blood macrophages (0.5¾106) were fixed with 4 % paraformaldehyde and stained with DAPI at 18 h post-infection with
H5N1 (483/97) or H1N1 (54/98) virus at an m.o.i. of 2. Values represent the mean±SD of cells from three different donors
analysed statistically by the two-tailed, paired t-test. *P , 0.1. (b) Replication of the H5N1 (483/97) virus in primary human
blood macrophages. Primary human blood macrophages (1¾106) were infected with human influenza virus H1N1 (54/98) or
avian influenza virus H5N1 (483/97) at an m.o.i. of 2. Culture supernatants were collected at 6, 10, 18 and 24 h post-infection.
Viral titres (TCID50) of the samples were measured by titration in Madin–Darby canine kidney (MDCK) cells. The results shown
are representative of experiments performed on cells from three different donors. (c) Primary human blood macrophages
(2¾106) were infected with influenza virus H5N1 (483/97) or H1N1 (54/98) at an m.o.i. of 2. At 12 h post-infection, the cells
were collected for ultrastructural examination by transmission electron microscopy. H1N1-infected macrophages (left); H5N1-
infected macrophages (middle); mock-treated macrophages (right). Magnification, ¾28 000; CC, chromatin condensation. The
results shown are representative of experiments performed on cells from three different donors. (d) Primary human blood
macrophages (1¾106) were harvested at 6, 12 and 18 h after infection with H5N1 (483/97) or H1N1 (54/98) virus at an m.o.i.
of 2. The activated level of PARP was assayed by Western analysis using a monoclonal anti-PARP antibody. Equal loading of
protein samples was demonstrated with anti-actin antibodies. The results shown are representative of experiments performed
on cells from three different donors. The density of the protein band was determined by using Bio-Rad Quantity One imaging
software. Numbers in parentheses are density values of activated PARP relative to that of actin. Mock, uninfected cells.

virus effectively and trigger apoptosis efficiently to fight H3N2 viruses (Cooper & Subbarao, 2000). Whether the
against the invading avian influenza virus. It has been variation of the viral genome sequences affects the
documented that the sequences of H5N1/97 virus are quite interactions between the host and the virus remains to be
different from those of the human influenza H1N1 or determined.

http://vir.sgmjournals.org 1277
C. K. P. Mok and others

Fig. 2. (a) Delayed activation of caspase 3 in


H5N1-infected human macrophages. Primary
human blood macrophages (1¾106) were
(a) Mock H1N1 H5N1
harvested at 6, 8, 10 and 12 h post-infection
Time post-infection (h) _ 6 8 10 12 6 8 10 12 with H5N1 (483/97) or H1N1 (54/98) virus at
Procaspase 3 an m.o.i. of 2. Activation of caspase 3 was
assayed by Western analysis using polyclonal
anti-caspase 3 antibodies. (b) Activation of
Cleaved caspase 3 caspase 8 in H5N1-infected primary human
Fold induction (1.00)(1.87)(3.10)(13.50)(38.08)(1.53)(2.47)(4.51)(6.60) blood macrophages. Primary human macro-
phages (1¾106) were harvested at 6, 8, 10
Actin and 12 h post-infection with H5N1 (483/97)
(1) (1) (1) (1) (1) (1) (1) (1) (1) or H1N1 (54/98) virus at an m.o.i. of 2. The
1 2 3 4 5 6 7 8 9 degradation level of procaspase 8 was
assayed by Western analysis using a mono-
(b) Mock H1N1 H5N1 clonal anti-procaspase 8 antibody. (c)
Involvement of caspase 8 in the apoptosis of
Time post-infection (h) _ 6 8 10 12 6 8 10 12 H5N1-infected human macrophages. Primary
Procaspase 8 human macrophages (1¾106) were pretreated
Fold induction (1.00)(0.99)(1.02)(0.75) (0.25)(0.99) (0.97) (0.73) (0.70)
or not with 50 mM caspase 8 inhibitor Z-IETD-
Actin FMK for 1 h at 37 6C and infected with H5N1
(483/97) or H1N1 (54/98) virus at an m.o.i. of
(1) (1) (1) (1) (1) (1) (1) (1) (1)
2, or left uninfected. Total proteins were
1 2 3 4 5 6 7 8 9
harvested at 18 h post-infection and assayed
(c) Mock Mock H1N1 H1N1 H5N1 H5N1 by Western analysis using anti-PARP anti-
_ _ _ body. Equal loading of protein samples was
Inhibitor + + + Time post-infection = 18 h
demonstrated by using anti-actin antibodies.
Pro-PARP
Cleaved PARP The results shown are representative of
Fold induction (1.17) (1.00) (2.34)(16.61)(1.30) (7.46) experiments performed on cells from three
different donors. The density of the protein
Actin band was determined by using Bio-Rad
(1) (1) (1) (1) (1) (1) Quantity One imaging software. Numbers in
1 2 3 4 5 6 parentheses are density values of activated
caspase 3, procaspase 8 or activated PARP
relative to that of actin. Mock, uninfected cells.

Induction of anti-apoptotic genes and pathways in H5N1- to examine whether this signalling cascade plays a role in the
infected macrophages is another possible mechanism delayed onset of apoptosis in H5N1-infected cells.
leading to the delayed onset of apoptosis. In our previous
Our results also provide additional information on the
study, we observed that p38 MAPK is activated differen-
role of macrophages in the pathogenesis of H5N1 infec-
tially in primary human macrophages after infection by
tion. H5N1-infected patients present with severe viral
H5N1/97 virus. This activation was shown to be associated
pneumonia and lymphopenia. A recent study reported that
with superinduction of TNF-a by H5N1 (Lee et al., 2005).
H5N1-infected macrophages could enhance TRAIL-induced
Moreover, it has been shown that double-stranded RNA
apoptosis in T cells compared with H1N1 infection (Zhou
activation of p38 MAPK results in inhibition of the activity
et al., 2006). Therefore, our results suggest that the
of caspases 3, 8 and 9 (Tadlock et al., 2003). This may be
delayed onset of apoptosis in H5N1-infected macrophages
analogous to the situation in H5N1 infection of macro-
may prolong the interaction of TRAIL-expressing macro-
phages, in which preferential activation of p38 MAPK may
phages with T cells to enhance the induction of apoptotic
contribute to delayed apoptosis compared with H1N1
cell death in the T cells.
infection. However, apoptosis in H5N1-infected macro-
phages was not decreased by using the p38 MAPK-specific In conclusion, we have provided experimental evidence
inhibitor SB203580 (see Supplementary Fig. S3, available that avian influenza viruses, including H5N1/97 and its
in JGV Online). Recent findings showed that the interac- precursors, trigger an apoptotic response mediated by
tion of Ccl5 and Ccr5 induces anti-apoptotic signals for caspase activation that is similar to, but delayed compared
macrophages during viral infection (Tyner et al., 2005). As with, that induced by human influenza viruses including
Ccl5 was highly induced in H5N1-infected human macro- H1N1 and H3N2. Our findings on the delay of apoptosis
phages compared with those infected by human influenza in H5N1 viruses combined with superinduction of
viruses (Cheung et al., 2002), further investigation is required proinflammatory cytokines may contribute, in part, to

1278 Journal of General Virology 88


Delayed onset of apoptosis in H5N1-infected macrophages

Fig. 3. Differential time of onset of apoptosis


(a) in primary human blood macrophages infected
Time post-infection = 18 h H1N1 H3N2 H6N1 H9N2 by different avian influenza viruses. (a) Primary
/G1 blood macrophages (1¾106) were infected
Pro-PARP with H1N1 (54/98), H3N2 (1174/99), H6N1
Cleaved PARP (W312/97) or H9N2 (G1/97) virus at an m.o.i.
Fold induction (30.48) (25.29) (5.92) (12.02) of 2 or treated with mock reagents. Protein
extracts were collected at 18 h post-infection
Actin
and assayed by Western analysis using a
(1) (1) (1) (1) monoclonal anti-PARP antibody. (b) Protein
1 2 3 4 extracts of H5N1 (483/97), H5N1 (437.6/99)
(b) and H1N1 (54/98)-infected primary human
Time post-infection = 18 h H5N1 H5N1 H1N1 Mock blood macrophages, as well as mock-treated
(437.6/99) cells, were collected and assayed under the
Pro-PARP same conditions as in (a). Equal loading of pro-
Cleaved PARP tein samples was demonstrated with anti-actin
Fold induction (7.46) (5.72) (33.08) (1.00) antibodies. The results shown are representative
of experiments performed on cells from three
Actin different donors. The density of the protein band
(1) (1) (1) (1) was determined by using Bio-Rad Quantity One
1 2 3 4 imaging software. Numbers in parentheses are
density values of activated PARP relative to that
of actin. Mock, uninfected cells.

understanding how these novel viruses cause fatal disease donors of the ‘internal’ genes of H5N1 viruses in Hong Kong? Proc
in humans. Natl Acad Sci U S A 96, 9363–9367.
Headley, A. S., Tolley, E. & Meduri, G. U. (1997). Infections and the
inflammatory response in acute respiratory distress syndrome. Chest
Acknowledgements 111, 1306–1321.
This work was supported by research grants to A. S. Y. L. (HKU 7430/ Hinshaw, V. S., Olsen, C. W., Dybdahl-Sissoko, N. & Evans, D.
03 M) and to M. P. (HKU 7459/03) from the Research Grants Council (1994). Apoptosis: a mechanism of cell killing by influenza A and B
of Hong Kong and the Research Fund for Control of Infectious viruses. J Virol 68, 3667–3673.
Disease to A. S. Y. L. (project no. 05050112), as well as the 2003 Vice Hoffmann, E., Stech, J., Leneva, I., Krauss, S., Scholtissek, C., Chin,
Chancellor’s Development Fund from The University of Hong Kong P. S., Peiris, M., Shortridge, K. F. & Webster, R. G. (2000). Charac-
to M. P. terization of the influenza A virus gene pool in avian species in
southern China: was H6N1 a derivative or a precursor of H5N1? J
Virol 74, 6309–6315.
References
Hong, S. J., Dawson, T. M. & Dawson, V. L. (2004). Nuclear and
Brydon, E. W., Morris, S. J. & Sweet, C. (2005). Role of apoptosis and mitochondrial conversations in cell death: PARP-1 and AIF signaling.
cytokines in influenza virus morbidity. FEMS Microbiol Rev 29, 837–850. Trends Pharmacol Sci 25, 259–264.
Cheung, C. Y., Poon, L. L., Lau, A. S., Luk, W., Lau, Y. L., Shortridge, Lee, D. C., Cheung, C. Y., Law, A. H., Mok, C. K., Peiris, M. & Lau, A. S.
K. F., Gordon, S., Guan, Y. & Peiris, J. S. (2002). Induction of (2005). p38 mitogen-activated protein kinase-dependent hyperinduc-
proinflammatory cytokines in human macrophages by influenza A tion of tumor necrosis factor alpha expression in response to avian
(H5N1) viruses: a mechanism for the unusual severity of human influenza virus H5N1. J Virol 79, 10147–10154.
disease? Lancet 360, 1831–1837. Price, G. E., Smith, H. & Sweet, C. (1997). Differential induction of
Claas, E. C., Osterhaus, A. D., Van Beek, R., De Jong, J. C., cytotoxicity and apoptosis by influenza virus strains of differing
Rimmelzwaan, G. F., Senne, D. A., Krauss, S., Shortridge, K. F. & virulence. J Gen Virol 78, 2821–2829.
Webster, R. G. (1998). Human influenza A H5N1 virus related to a Soldani, C. & Scovassi, A. I. (2002). Poly (ADP-ribose) polymerase-1
highly pathogenic avian influenza virus. Lancet 351, 472–477. cleavage during apoptosis: an update. Apoptosis 7, 321–328.
Cooper, L. A. & Subbarao, K. (2000). A simple restriction fragment Subbarao, K. & Shaw, M. W. (2000). Molecular aspects of avian
length polymorphism-based strategy that can distinguish the internal influenza (H5N1) viruses isolated from humans. Rev Med Virol 10,
genes of human H1N1, H3N2, and H5N1 influenza A viruses. J Clin 337–348.
Microbiol 38, 2579–2583.
Subbarao, K., Klimov, A., Katz, J., Regnery, H., Lim, W., Hall, H.,
Fesq, H., Bacher, M., Nain, M. & Gemsa, D. (1994). Programmed cell Perdue, M., Swayne, D., Bender, C. & other authors (1998).
death (apoptosis) in human monocytes infected by influenza A virus. Characterization of an avian influenza A (H5N1) virus isolated from
Immunobiology 190, 175–182. a child with a fatal respiratory illness. Science 279, 393–396.
Fisman, D. N. (2000). Hemophagocytic syndromes and infection. Tadlock, L., Yamagiwa, Y., Marienfeld, C. & Patel, T. (2003). Double-
Emerg Infect Dis 6, 601–608. stranded RNA activates a p38 MAPK-dependent cell survival pro-
Guan, Y., Shortridge, K. F., Krauss, S. & Webster, R. G. (1999). gram in biliary epithelia. Am J Physiol Gastrointest Liver Physiol 284,
Molecular characterization of H9N2 influenza viruses: were they the G924–G932.

http://vir.sgmjournals.org 1279
C. K. P. Mok and others

Takizawa, T., Matsukawa, S., Higuchi, Y., Nakamura, S., Nakanishi, antiapoptotic signals for macrophage survival during viral infection.
Y. & Fukuda, R. (1993). Induction of programmed cell death Nat Med 11, 1180–1187.
(apoptosis) by influenza virus infection in tissue culture cells. J Gen Xu, X., Subbarao, K., Cox, N. J. & Guo, Y. (1999). Genetic characteri-
Virol 74, 2347–2355. zation of the pathogenic influenza A/Goose/Guangdong/1/96 (H5N1)
Takizawa, T., Tatematsu, C., Ohashi, K. & Nakanishi, Y. (1999). virus: similarity of its hemagglutinin gene to those of H5N1 viruses
Recruitment of apoptotic cysteine proteases (caspases) in influenza from the 1997 outbreaks in Hong Kong. Virology 261, 15–19.
virus-induced cell death. Microbiol Immunol 43, 245–252. Yuen, K. Y., Chan, P. K., Peiris, M., Tsang, D. N., Que, T. L.,
To, K. F., Chan, P. K., Chan, K. F., Lee, W. K., Lam, W. Y., Wong, K. F., Shortridge, K. F., Cheung, P. T., To, W. K., Ho, E. T. & other authors
Tang, N. L., Tsang, D. N., Sung, R. Y. & other authors (2001). (1998). Clinical features and rapid viral diagnosis of human disease
Pathology of fatal human infection associated with avian influenza A associated with avian influenza A H5N1 virus. Lancet 351, 467–471.
H5N1 virus. J Med Virol 63, 242–246. Zhou, J., Law, H. K., Cheung, C. Y., Ng, I. H., Peiris, J. S. & Lau, Y. L.
Tyner, J. W., Uchida, O., Kajiwara, N., Kim, E. Y., Patel, A. C., (2006). Functional tumor necrosis factor-related apoptosis-inducing
O’Sullivan, M. P., Walter, M. J., Schwendener, R. A., Cook, ligand production by avian influenza virus-infected macrophages. J
D. N. & other authors (2005). CCL5–CCR5 interaction provides Infect Dis 193, 945–953.

1280 Journal of General Virology 88

Você também pode gostar