Sie sind auf Seite 1von 9

MAJOR ARTICLE

A Paradoxical Role for Neutrophils


in the Pathogenesis of West Nile Virus
Fengwei Bai,1,4,a Kok-Fai Kong,2,a,b Jianfeng Dai,1 Feng Qian,2 Lin Zhang,2 Charles R. Brown,5 Erol Fikrig,1,3
and Ruth R. Montgomery2
Sections of 1Infectious Diseases and 2Rheumatology, Department of Internal Medicine, and 3The Howard Hughes Medical Institute,
Yale University School of Medicine, and 4L2 Diagnostics, New Haven, Connecticut; and 5Departments of Molecular Microbiology
and Immunology and Veterinary Pathobiology, University of Missouri, Columbia

Downloaded from jid.oxfordjournals.org by guest on February 27, 2011


Polymorphonuclear leukocytes (PMNs) are key in innate immunity, but their role in viral pathogenesis is
incompletely understood. In infection due to West Nile virus (WNV), we found that expression of 2 PMN-
attracting chemokines, Cxcl1 and Cxcl2, was rapidly and dramatically elevated in macrophages. PMNs are
rapidly recruited to the site of WNV infection in mice and support efficient replication of WNV. Mice depleted
of PMNs after WNV inoculation developed higher viremia and experienced earlier death, compared with the
control group, which suggest a protective role for PMNs. In contrast, when PMNs were depleted prior to
infection with WNV, and in mice deficient in Cxcr2 (a chemokine receptor gene), viremia was reduced and
survival was enhanced. Collectively, these data suggest that PMNs have a biphasic response to WNV infection,
serving as a reservoir for replication and dissemination in early infection and later contributing to viral
clearance.

West Nile virus (WNV), a mosquito-transmitted single- alitis [1]. An approved therapy for use in humans does
stranded RNA (ssRNA) virus, belongs to the family not currently exist, and viral pathogenesis is incom-
Flaviviridae, which includes other mosquito-borne hu- pletely understood.
man pathogenic viruses, such as Yellow fever and Den- The immune response to WNV is multifactorial and
gue viruses [1]. WNV has spread rapidly throughout includes both innate and adaptive immunity. Upon
North America since its first introduction into New WNV infection, host innate immune sensors, including
York in 1999 and has caused worldwide morbidity and Toll-like receptors, double-stranded RNA-activated
mortality. Although infection in humans is typically protein kinase, retinoic-acid-inducible protein I, and
asymptomatic, the elderly and immunosuppressed pop- melanoma-differentiation-associated gene 5 (MDA-5),
ulations are particularly susceptible to life-threatening recognize WNV components and initiate anti-viral sig-
neurologic disease, including meningitis and enceph-
naling and production of type I interferon (IFN) and
cytokines [2–4]. Cellular immunity is critical for host
recovery from WNV infection, including contributions
Received 9 April 2010; accepted 20 July 2010; electronically published 4 of CD4+, CD8+, and gd T cell responses, as well as
November 2010.
a
F.B. and K.-F.K. contributed equally to this article. antibody production by B cells [5–8]. The role of nat-
b
Present affiliation: Division of Cell Biology, La Jolla Institute for Allergy and ural killer cells remains unclear [9, 10]. Macrophages
Immunology, La Jolla, California.
Potential conflicts of interest: none reported. are essential in resistance to WNV infection: macro-
Financial support: National Institutes of Health (AI 50031, AI 070343, AI 079348, phage depleted mice show accelerated development of
and AI 059292) and The Howard Hughes Medical Institute. FB was supported by
a career development grant (U54-AI057158-Lipkin to F.B.). WNV encephalitis and a 50% increase in mortality [11].
Reprints or correspondence: Dr Ruth R. Montgomery, Dept of Internal Medicine, Polymorphonuclear leukocytes (PMNs; neutrophils)
Yale University School of Medicine, 300 Cedar St/TAC S413, New Haven, CT
06520-8031 (ruth.montgomery@yale.edu). are the most abundant type of leukocytes in humans,
The Journal of Infectious Diseases 2010; 202(12):1804–1812 a key component of the innate immune response, and
 2010 by the Infectious Diseases Society of America. All rights reserved.
0022-1899/2010/20212-0007$15.00
the first immune cells to be recruited to inflammatory
DOI: 10.1086/657416 foci [12]. PMNs ingest pathogens, release antimicrobial

1804 • JID 2010:202 (15 December) • Bai et al


peptides, and produce reactive oxygen species and extracellular Investigation Committee of Yale University. PMNs were isolated
traps to combat pathogens [13, 14]. Depletion of PMNs or from blood by 3% dextran sedimentation followed by hypo-
mutations in key PMN antimicrobial pathways increases sus- tonic lysis to remove red blood cells [26]. Peripheral blood
ceptibility to numerous pathogens [15]. PMNs also contribute mononuclear cells (PBMCs) were separated by Ficoll-Hypaque
to inflammation in a number of disease processes, including a density gradient centrifugation and plated at 5 ⫻ 10 6 per 35 mm2
complex program of apoptosis [16] and an abundant zinc- well in Roswell Park Memorial Institute 1640 medium con-
chelating bacteriostatic cytoplasmic protein, calprotectin taining 20% human serum (Cambrex) and antibiotics, as above.
(S100A8/A9) [17, 18]. PMNs predominate in the cerebrospinal PBMCs were washed after 2 h and incubated for 6–8 days to
fluid (CSF) of patients with neuroinvasive WNV meningitis obtain mature primary macrophages with 99% viability [26].
and encephalitis [19, 20] and are recruited into the brain as Fluorescence-activated cell scanning (FACS) analy-
early as day 4 after infection in the rodent model of WNV sis. Peritoneal exudate cells were washed twice in PBS con-
infection [21]. We have undertaken the current study to elu- taining 1% FBS and stained for 30 min at 4C with antibodies
cidate the role of PMNs in WNV pathogenesis. for PMNs (anti–granulocyte receptor 1 [Gr-1]), macrophages
(F4/80), T cells (CD4+ and CD8+) and B cells (CD19+) (BD
MATERIALS AND METHODS Biosciences). Cells were fixed with 4% paraformaldehyde. FACS
data were collected using an LSR II Flow cytometer (BD Bi-
WNV infections and experimental animals. WNV isolate

Downloaded from jid.oxfordjournals.org by guest on February 27, 2011


oscience) and were analyzed using FlowJo software (Tree Star)
2471 [22], used in these studies, was propagated 1 time in Vero
[27].
cells and titered in a Vero cell plaque-formation assay. All ex-
Assessment of Chemokines by quantitative polymerase
periments with WNV were performed in a Bio-safety Level 3
chain reaction (Q-PCR) array and enzyme-linked immuno-
animal facility, according to the regulations of Yale University.
sorbent assay (ELISA). Macrophage cultures were washed
C3H/HeJ mice were purchased from the Charles River (Wil-
with PBS prior to total RNA extraction by Qiagen RNeasy Mini
mington, MA). Cxcr2⫺/⫺ mice were fully backcrossed to C3H/
kit (Valencia) and assayed using RT2Profiler PCR Array kits
HeJ background [23]. All infections were performed on age-
(SuperArray Bioscience) according to the user manual. Total
and sex-matched 7–8-week-old mice and protocols were ap-
RNA from macrophages (1 mg) was transcribed into first strand
proved by the Yale University Institutional Animal Care and
complementary DNA (cDNA) and processed in 96-well PCR
Use Committee. C3H wild-type and Cxcr2⫺/⫺ mice were in-
oculated intraperitoneally with 100 mL of phosphate-buffered array plates with 25 mL of Q-PCR master mix per well. Thresh-
saline (PBS) with 1% gelatin or WNV (100–10,000 plaque old cycle values (Ct) were analyzed to compare gene expression
forming units [pfu], as indicated) in PBS with 1% gelatin [24]. between mock- and WNV-infected macrophages.
Peritoneal exudates were collected by lavage with 10 mL of cold Culture supernatants from macrophages were harvested and
PBS. For footpad inoculation, animals were infected with 10 chemokines were quantified in duplicate by ELISA according
pfu of WNV in 50 mL of PBS containing 1% heat-inactivated to the manufacturer’s instructions (R&D Systems).
fetal bovine serum (FBS). Animals were monitored twice daily WNVE and chemokine gene Q-PCR. Total RNA was ex-
for morbidity and mortality for up to 21 days after infection tracted from macrophages, PMNs, and blood using the RNeasy
[24]. kit (Qiagen) and was used to synthesize cDNA by the
Preparation of PMNs and macrophages. Mouse peritoneal AffinityScript Multi Temperature cDNA synthesis kit (Strata-
PMNs and macrophages were elicited by intraperitoneal injec- gene). WNV-specific RNA was quantified by Q-PCR [24].
tion of 1 mL of thioglycollate medium (BD BBL). PMN were TaqMan gene expression assays for Cxcl1, Cxcl2, and Cxcl10
collected 6 h after injection, and macrophages were collected were purchased from Applied Biosystems. For differential RNA
at 3 days after injection [24, 25]. PMNs were isolated by 3% Q-PCR of the WNVE region, primer for positive-sense cDNA
dextran sedimentation and used for experiments immediately. was 5’-TGGGTCAGCACGTTTGTCATTGTG-3’; primer for
Macrophages were cultured overnight at 1 ⫻ 10 6 cells per 35 negative-sense cDNA was 5’-TTGGAAGGAGTGTCTGGAGC-
mm2 dish in 2 mL Dulbecco’s modified eagle medium (DMEM) AACA-3’. Values for each gene were calculated from the ac-
containing 10% FBS and 1000 U/mL penicillin and 1 mg/mL companying standard curve in each Q-PCR reaction plate, and
streptomycin (Invitrogen). Nonadherent cells were washed copy numbers were expressed as a ratio to cellular b-actin
away prior to infection for 4 h with WNV (multiplicity of cDNA copies measured by Q-PCR. Each duplicate measure-
infection [MOI] p 1). Leukocyte populations were isolated ment was divided by the corresponding measurement for actin
from blood using anti–Gr-1+–conjugated magnetic microbeads and then averaged to determine the relative levels in each
(Miltenyi Biotec). sample.
Heparinized blood from healthy human volunteer donors PMN depletion in vivo. Circulating PMNs in mice were
was collected in accordance with the regulations of Human depleted using mouse monoclonal antibodies Gr-1 (clone RB6–

PMN in WNV Infection • JID 2010:202 (15 December) • 1805


8C5) [28] or Ly-6G [29]. Gr-1 was obtained from the Brown followed by Tukey’s test for assessment of significance in FACS
laboratory and as a kind gift from Robert L. Coffman (DNAX assays. We performed survival curve comparisons using the
Research Institute). Ly-6G (clone 1A8) and isotype-matched Kaplan-Meier survival and log-rank test (GraphPad Prism soft-
control antibodies were purchased from BD Biosciences. RB6– ware, version 4.0). Statistical significance was set at P ! .05 for
8C5 cells were cultured in DMEM with 10% FBS, 10 mM all analyses.
Hepes, 2 mM L-glutamine, and antibiotics. RB6–8C5 antibody
was affinity purified from cell culture supernatants using a pro- RESULTS
tein G column (Pharmacia). Seven-week-old female C3H mice
were injected intraperitoneally either with 250 mg of RB6–8C5 Expression of chemokines Cxcl1 and Cxcl2 is rapidly elevated
or isotype matched IgG2b (Clone MOPC-141; Sigma-Aldrich) in macrophages upon WNV infection. To investigate the role
antibodies or with 100 mg Ly-6G and isotype matched IgG2a of chemokines in WNV pathogenesis, we infected thioglycol-
in 100 mL of PBS 1 day prior or 1–2 days after infection with late-elicited peritoneal macrophages from C3H mice with WNV
WNV. (MOI p 1) and analyzed chemokine and chemokine receptor
Statistical analysis. We calculated standard errors of the gene expression profiles by Q-PCR arrays. Expression of Cxcl1
means (SEMs) and analyzed data by nonpaired Student’s t test and Cxcl2, 2 CXC-type chemokines that induce the migration
for single mean comparisons and one-way analysis of variance of PMNs, was dramatically up-regulated by 38-fold over un-

Downloaded from jid.oxfordjournals.org by guest on February 27, 2011

Figure 1. Cxcl1 and Cxcl2 expression is elevated in vitro at early stage of West Nile virus (WNV) infection (A–G). Murine thioglycollate-elicited
peritoneal macrophages from strain C3H mice were infected with WNV (multiplicity of infection [MOI] of 0, 0.1, 0.5 or 1). A, Macrophages were
infected with WNV (MOI p 1) for 4 h, and total RNA was extracted for chemokine quantitative PCR (Q-PCR) array analysis. The data are normalized
to mouse b-actin messenger RNS (mRNA) and are expressed as the relative fold increase over normalized RNA from mock controls (n p 3 ). B and
C, Cxcl1 mRNA was measured by Q-PCR and normalized to mouse b-actin (Actb; unitless ratio  1 standard error of the mean [SEM]) and enzyme-
linked immunosorbent assay (ELISA) results are shown for Cxcl1 protein (pg/mL in the media. D and E, Q-PCR and ELISA results of Cxcl2 expression.
F and G, Q-PCR and ELISA results of Cxcl10 expression. H, Human monocyte–derived macrophages were infected with WNV (MOI p 1 and n p 6).
Cxcl1, Cxcl2, and Cxcl10 mRNA was measured by Q-PCR. The data are normalized to human b-actin mRNA and are expressed as the relative fold
increase over normalized RNA from time 0.

1806 • JID 2010:202 (15 December) • Bai et al


as early as 3 h after infection and decreasing by 24 h, whereas
up-regulation of Cxcl10 only began at 8 h and peaked at 24 h.
Elevated levels of Cxcl1 and Cxcl2 suggest that PMNs may be
recruited rapidly to sites of infection with WNV.
PMNs are rapidly recruited to the site of WNV infection.
To assess the kinetics of recruitment of PMNs, we infected mice
with WNV and assessed peritoneal exudate cells with cell lin-
eage markers for flow cytometry (FACS). Analysis of the com-
position of the exudates at time points up to 72 h showed that
PMNs (Gr-1+ cells) were the most abundant cell type (1.5 
0.3 ⫻ 10 6 cells/mouse; ∼62%) in the peritoneal cavity as early
as 12 h after WNV inoculation (Figure 2A) with a peak abun-
dance at 24 h that began to decrease at 72 h after infection.
Of the other immune cells detected, only CD8+ T cells followed
similar kinetics, but at only ∼50% (7.0  1.7 ⫻ 10 5 cells/mouse)
of the PMN number (Figure 2A). These data suggest that PMNs

Downloaded from jid.oxfordjournals.org by guest on February 27, 2011


are the predominant immune cells that are initially recruited
to sites of WNV infection.
Figure 2. Neutrophils are rapidly recruited to the peritoneal cavity after The efficient accumulation of PMN depends on recruitment
intraperiotoneal inoculation of West Nile virus (WNV). C3H wild-type or
Cxcr2⫺/⫺ mice were infected intraperitoneally with 10,000 plaque-forming
through the chemokines Cxcl1 and Cxcl2. When we conducted
units of WNV, and mice were euthanized at the selected time points up infections in mice deficient in Cxcr2 (Cxcr2⫺/⫺), a receptor gene
to 72 h to collect total cells infiltrating into the peritoneal cavity. A, for chemokines Cxcl1 and Cxcl2, there was a reduction of up
Lavaged cells were stained with granulocyte receptor 1 (Gr-1) (neutrophil to 85% (2.4 ⫻ 10 5 cells) and 64% (4.5  0.003 ⫻ 10 5 cells) of
[neu] marker), F4/80 (macrophage [mac] marker), CD8+, CD4+, and CD19+ PMNs in peritoneal exudates at 24 h and 48 h, respectively
(B cell marker) and differentiated by fluorescence-activated cell scanning
(FACS) (n p 2–3 per group). *P ! .05 , **P ! .01, and ***P ! .001 when (Figure 2B), and levels of CD8+ T cells were also reduced, albeit
compared with Gr-1+ cell group at each time point. Data were analyzed at a lower level (loss of 35% [4.5  0.6 ⫻ 10 5] and 40% of
with use of one-way analysis of variance followed by Tukey’s test. B, CD8+ T [2.7  0.08 ⫻ 10 5] cells/mouse at 24 h and 48 h, re-
FACS results of Cxcr2⫺/⫺ mice infiltrating cells (n p 2 per group). spectively). Lower CD8+ cell counts may be attributable to re-
duced recruitment through their Cxcr2 [31] or to an unknown
infected macrophages at 4 h after infection, and another CXC- role of PMNs in modulating recruitment of adaptive cells.
type chemokine, Cxcl10, was slightly elevated (2-fold) at the Depletion of PMNs prior to but not after WNV challenge
same time point (Figure 1A). The expression of the remaining increases survival. To investigate the role of PMNs in WNV
81 chemokine and receptor genes in the array, including other pathogenesis, we infected mice depleted of PMNs by treatment
CXC-type and CC-type chemokines, did not show up-regu- with the monoclonal antibodies Gr-1 (clone RB6–8C5), which
lation after 4 h of infection with WNV. This is consistent with selectively binds and depletes PMNs and eosinophils [28], or
reports showing that CXC type chemokine interleukin 8 (IL- Ly-6G (clone 1A8), which specifically binds and depletes PMNs
8) is among the first cytokines produced in response to infection [29]. Gr-1 (250 mg) or Ly-6G (100 mg) in 100 mL of PBS reduced
with another related flavivirus, dengue virus [30]. levels of PMNs to 50% of levels in mice injected with isotype-
The findings from the array were confirmed by both Q-PCR matched control antibodies (Figure 3A;11% vs 22%; P ! .05;
and ELISA when we assessed expression of Cxcl1, Cxcl2, and Ly-6G data not shown). When viral load in the blood was
Cxcl10 messenger RNA (mRNA) over a time course of infection quantified on day 2 or 3 after infection by Q-PCR for WNV
by reverse transcription Q-PCR and secreted protein levels by envelope protein gene (WNVE), the level of WNV was signif-
ELISA from the same cells. Expression of Cxcl1 and Cxcl2 was icantly reduced in animals that had been depleted of PMNs 1
markedly up-regulated after WNV infection in vitro (Figure day prior to inoculation (Figure 3B and 3C; P ! .05). Further-
1B–E) at 4 h. In contrast, up-regulation of Cxcl10 expression more, PMN-depleted mice by Gr-1 showed dramatically in-
was lower and developed relatively later, with elevation ob- creased survival (Figure 3D; P ! .001, by log-rank test). The
served at 48 h after infection (Figure 1F and 1G). Consistent effect of PMN depletion by Gr-1 prior to challenge was also
with murine macrophage results, primary human macrophages noted in an alternate route of infection, when mice were in-
infected with WNV (MOI p 1) showed time-dependent in- fected with WNV by footpad injection (10 pfu). These PMN-
creases in mRNA expression of Cxcl1, Cxcl2, and Cxcl10 (Figure depleted animals had lower viral loads and significantly in-
1H), with expression of mRNA for Cxcl1 and Cxcl2 increasing creased survival (Figure 3E; 40% vs 0%; P ! .01), suggesting

PMN in WNV Infection • JID 2010:202 (15 December) • 1807


Downloaded from jid.oxfordjournals.org by guest on February 27, 2011
Figure 3. Neutrophil depletion prior to but not after West Nile virus (WNV) challenge increases mouse resistance (A–D). C3H mice were injected
with neutrophil-depleting antibodies (intraperitoneal injection with a single 250-mg dose of granulocyte receptor 1 (Gr-1) or 100 mg of Ly-6G in 100
mL of phosphate-buffered saline [PBS]) or isotype-matched control antibodies 1 day prior to WNV infection (intraperitoneal injection of 100 plaque-
forming units [pfu]). A, Polymorphonuclear leukocyte (PMN) percentage in blood smears on day 1 after Gr-1 treatment (isotype control, n p 5 ; Gr-1,
n p 4). B, Mice were treated with Gr-1 and WNVE was measured by quantitative polymerase chain reaction (Q-PCR) in blood at day 3 after infection
(isotype control, n p 10; Gr-1, n p 7). C, Mice were treated with Ly-6G, and WNVE was measured by Q-PCR in blood at day 2 after infection
(n p 10 per group). These data are representative of 2 independent experiments. D, Kaplan-Meier survival analysis of isotype-matched control and
Gr-1 antibody–treated mice after intraperitoneal inoculation of WNV (n p 10 per group). E, Survival curves of C3H mice that were injected with Gr-
1 (intraperitoneal injection with 1 dose of 250 mg in 100 mL PBS) 1 day before WNV infection by footpad inoculation (10 pfu; n p 10 per group). F–
H, C3H mice were injected with 2 doses of neutrophil-depleting antibodies (intraperitoneal injection with 250 mg Gr-1 or 100 mg Ly-6G in 100 mL
PBS) on days 1 and 2 after WNV infection (intraperitoneal injection of 100 pfu). F and G, WNVE was measured by Q-PCR in blood specimens at day
3 after infection (n p 10 per group). H, Survival analysis of isotype-matched control and Gr-1 antibody–treated mice after intraperitoneal inoculation
of WNV (n p 10 per group). Survival data shown are representative of at least 2 independent experiments. *P ! .05 and **P ! .001.

that PMN may play a negative role in the host immune response not shown). Taken together, these data suggest, paradoxically,
against WNV infection. that PMN may enhance WNV infection at the early stages of
To investigate whether PMNs play a detrimental role in the infection, whereas in the later course of infection, PMNs appear
defense against WNV, we further dissected the kinetics of the to contribute to control of infection.
PMN response to infection with WNV. Remarkably, when mice PMNs are an important reservoir for WNV replication. To
were infected with WNV first and then depleted of PMNs on further dissect the antiviral mechanisms, we infected murine
days 1 and 2 after infection, treated animals showed higher PMNs and macrophages with WNV ex vivo at a range of MOIs
levels of viral burden (Figure 3F and 3G; P ! .05 ) and reduced and quantified viral load by Q-PCR at 8 h after infection [24,
survival rate, compared with the mice that were administrated 25, 27] and found that PMNs were infected by WNV to a
control antibody (Figure 3H; 0% vs 30%; P ! .001; Ly-6G data significantly higher level than were macrophages (eg, 55-fold

1808 • JID 2010:202 (15 December) • Bai et al


Downloaded from jid.oxfordjournals.org by guest on February 27, 2011
Figure 4. Neutrophils are an important reservoir for West Nile virus (WNV) replication. A, Thioglycollate elicited peritoneal macrophages and
neutrophils from C3H mice were infected with WNV or mock (phosphate-buffered saline [PBS]) for 8 h. Quantitative polymerase chain reaction (Q-
PCR) was performed for WNVE to measure WNV replication (macrophages, n p 18 ; neutrophils, n p 6 ). B, Q-PCR results of WNV-infected human
monocyte–derived macrophages and neutrophils at 8 h (macrophages, n p 5 ; neutrophils, n p 12 ). C, Differential Q-PCR results of WNV-infected
mouse peritoneal macrophages and neutrophils at 8 h. D, C3H mice were infected via intraperitoneal injection with 10,000 plaque-forming units (pfu)
of WNV, and mice were euthanized at day 3 after infection to collect blood (n p 8 per group). Neutrophils were isolated using anti-Gr-1 Macs beads,
and Q-PCR was performed for WNVE in the neutrophils and the Gr-1⫺ cells. *P ! .05, **P ! .01, *** P ! .001, and ****P ! .0001.

and 69-fold higher when infected with MOIs of 1 and 10, in PMN may be attributed to active replication and not only
respectively) (Figure 4A). A similar enhancement of infection to increased permissiveness (Figure 4C).
and replication of WNV in PMNs was noted in human mac- To determine whether PMN serve as a reservoir for WNV
rophages and PMNs (eg, 17-fold and 13-fold; Figure 4B). These in vivo, we infected mice with 10,000 pfu of WNV intraperi-
data suggest that PMNs may act as a reservoir of virus in early toneally and quantified viral load in blood cells at day 3 after
infection. infection. Q-PCR for WNVE measurement in PMN (Gr-1+
We used a differential Q-PCR method to demonstrate active cells) versus other cell types (Gr-1⫺ cells) showed that PMNs,
replication of WNV by distinguishing the positive- and nega- which accounted for only ∼25% of the leukocytes in mouse
tive-sense RNA in WNV-infected cells [32]. Because viral RNA blood, harbored ∼8-fold more WNVE than the other cell types
polymerase generates negative-sense RNA from the positive- combined (Figure 4D; P ! .05). These results suggest that PMNs
sense stranded genomic RNA that serves as a template for may play a critical role in vivo for WNV replication and dis-
replication, accumulation of negative-sense RNA indicates that semination, especially in humans, where PMNs are the pre-
active replication is occurring. In both human and murine dominant cell type in blood.
PMNs, differential Q-PCR detected both positive- and negative- Cxcr25/5 mice show delayed death after lethal WNV
sensed viral RNA, suggesting that the higher levels of WNVE infection. We assessed the course of WNV infection in

PMN in WNV Infection • JID 2010:202 (15 December) • 1809


phase for WNV replication and dissemination, they later con-
tribute to limiting WNV infectivity.
Effects of the cellular milieu may contribute to the shift in
PMN from the early replication-permissive state to the later
antiviral state. In the first 24 h after WNV infection, macro-
phages initiate a robust antiviral program including abundant
type I IFN [27, 33]. When PMN in vitro were treated with IFN
(2000–10,000 IU) for 1 h prior to infection with WNV
(MOI p 1), WNVE gene levels at 20 h were reduced (0.83-
fold; n p 9; P ! .016). Although not comprehensive, this re-
duction suggests that the antiviral responses of surrounding
immune cells, such as macrophages, may promote the antiviral
state of PMNs in vivo.

DISCUSSION

WNV is a neuroinvasive virus and lethality is caused by viral

Downloaded from jid.oxfordjournals.org by guest on February 27, 2011


invasion of the brain and subsequent encephalitis [7, 24, 25].
Early control of WNV, which relies on innate immune re-
sponses, is crucial to restrict WNV dissemination, contain in-
flammation, and prevent permeabilization of the blood-brain-
barrier leading to neural infection and death [33]. Although
immune responses are generally successful at combating infec-
tion due to WNV in young immunocompetent individuals,
WNV attenuates activation of macrophages [34], and increased
Figure 5. Cxcr2⫺/⫺ mice show delayed death after lethal West Nile
virus (WNV) infection. Cxcr2⫺/⫺ mice and wild-type (WT; C3H) mice were susceptibility in the elderly population may be attributable to
challenged with WNV by intraperitoneal injection (100 plaque-forming dysregulation of innate responses in that population [27, 35].
units). A, quantitative polymerase chain reaction (Q-PCR) was performed Activation of PMNs in infectious or inflammatory foci leads
for WNVE in peripheral blood on days 1, 2, and 3 (n p 10 per group). to the release of powerful antimicrobial products and is crucial
B, Survival analysis of WT and Cxcr2⫺/⫺ mice after intraperitoneal in- for effective innate immunity [12]. Evidence supporting a role
oculation of WNV (WT, n p 43 ; Cxcr2⫺/⫺ mice, n p 34). Data are pooled
for PMNs in response to flaviviral infections includes the pre-
from 4 independent experiments. *P ! .05 and **P ! .01.
dominance of PMN noted in infiltrates of cerebrospinal fluid
from human patients with WNV meningitis and encephalitis
Cxcr2⫺/⫺ mice, which have equivalent numbers of PMN as wild- [19, 20]; the enhanced binding of PMNs to dengue virus–
type mice but whose PMNs have reduced chemotactic re- infected endothelial cells [36], suggesting a preferential recruit-
sponses. Consistent with our previous data, the Q-PCR results ment for PMNs; and the increased number of immature PMNs
on day 1 after infection showed that viremia in blood was in the blood of patients at an early stage (5–6 days after onset)
significantly lower in Cxcr2⫺/⫺ mice than in wild-type mice of dengue hemorrhagic fever [37].
(Figure 5A; P ! .05), again suggesting that PMN may support In our examination of the initial chemokine responses to
WNV replication in early WNV infection. However, as infec- WNV infection, we noted that expression of PMN-recruiting
tion progressed, we detected similar levels of viremia in both chemokines was dramatically elevated and that PMN were
Cxcr2⫺/⫺ and wild-type mice at day 2 and a modest but sta- quickly recruited to sites of WNV infection in vivo. Our results
tistically higher viral burden on day 3 after infection in blood showing up-regulation of Cxcl10 somewhat later are consistent
specimens from Cxcr2⫺/⫺ mice, compared with blood speci- with a role in recruitment of CD8+ T cells, which are critical
mens from wild-type mice, which argued in favor of an antiviral to clear viral infection in the central nervous system [38, 39].
role for PMN at this later time point (Figure 5A; 2.7-fold higher; The predominance and early arrival of PMN supports a crit-
P ! .01). Analysis of survival of infected animals, which encom- ical role in response to WNV infection, but depletion of PMNs
passes both early and later responses, showed delayed death of prior to WNV challenge (or studies in Cxcr2⫺/⫺ mice) para-
Cxcr2⫺/⫺ mice, compared with wild-type mice (median dura- doxically lowered viremia and enhanced survival, suggesting
tion of survival, 11 vs 9 days for Cxcr2⫺/⫺ vs wild-type mice; that PMNs initially have an enhancing role in WNV infection.
P p .01; Figure 5B). Collectively, these data support the hy- Mechanisms underlying PMN facilitation of early WNV infec-
pothesis that, although PMNs may be permissive in the early tivity may include: (1) abundant reservoir, because PMNs are

1810 • JID 2010:202 (15 December) • Bai et al


the predominant cell type recruited to the site of infection; (2) 5. Shrestha B, Diamond MS. Role of CD8+ T cells in control of West
Nile virus infection. J Virol 2004; 78:8312–8321.
efficient replication of WNV in PMN; and (3) higher levels of 6. Wang T, Gao Y, Scully E, et al. Gamma delta T cells facilitate adaptive
viral load in PMNs than in other cells circulating in WNV- immunity against West Nile virus infection in mice. J Immunol
infected mice. In support of our finding, elevated replication 2006; 177:1825–1832.
7. Shrestha B, Samuel MA, Diamond MS. CD8+ T cells require perforin
rates in PMNs may contribute to efficient dissemination of
to clear West Nile virus from infected neurons. J Virol 2006; 80:
virus in other systems, as has been shown previously for cy- 119–129.
tomegalovirus and human immunodeficiency virus [40–42]. 8. Diamond MS, Shrestha B, Marri A, Mahan D, Engle M. B cells and
We noted a biphasic effect of the absence of PMNs on the antibody play critical roles in the immediate defense of disseminated
infection by West Nile encephalitis virus. J Virol 2003; 77:2578–2586.
course of WNV infection. Initially, the absence of PMNs led 9. Getts DR, Terry RL, Getts MT, et al. Ly6c+ “inflammatory monocytes”
to lower viremia and delayed death but a later depletion of are microglial precursors recruited in a pathogenic manner in West
PMN led to increased viral burden. Collectively, these data Nile virus encephalitis. J Exp Med 2008; 205:2319–2337.
10. Hershkovitz O, Rosental B, Rosenberg LA, et al. NKp44 receptor me-
indicate a complex and previously unappreciated role for PMN diates interaction of the envelope glycoproteins from the West Nile
in infection with WNV, facilitating WNV infection initially and dengue viruses with NK cells. J Immunol 2009; 183:2610–2621.
through acting as a reservoir for viral replication and allowing 11. Ben-Nathan D, Huitinga I, Lustig S, van Rooijen N and Kobiler D.
West Nile virus neuroinvasion and encephalitis induced by macrophage
dissemination, and only subsequently serving a protective func-
depletion in mice. Arch Virol 1996; 141:459–469.
tion. Studies on the effects of macrophage-derived type I IFN 12. Nathan C. Neutrophils and immunity: challenges and opportunities.

Downloaded from jid.oxfordjournals.org by guest on February 27, 2011


suggest that the cytokine milieu in vivo may influence the shift Nat Rev Immunol 2006; 6:173–182.
13. Kobayashi SD, Voyich JM, Burlak C, DeLeo FR. Neutrophils in the
in PMN antiviral activity. This finding reflects the well-de-
innate immune response. Arch Immunol Ther Exp (Warsz) 2005; 53:
scribed cooperativity of PMNs and macrophages in immune 505–517.
responses [43] and, in particular, a role for type I IFN, which 14. Papayannopoulos V, Zychlinsky A. NETs: a new strategy for using old
has recently been shown to modulate PMN recruitment and weapons. Trends Immunol 2009; 30:513–521.
15. Lekstrom-Himes JA, Gallin JI. Immunodeficiency diseases caused by
turnover in response to infection with the protozoan parasite defects in phagocytes. N Engl J Med 2000; 343:1703–1714.
Leishmania [44]. 16. Kobayashi SD, Voyich JM, Somerville GA, et al. An apoptosis-differ-
Interestingly, a role for PMNs as a reservoir for pathogens entiation program in human polymorphonuclear leukocytes facilitates
resolution of inflammation. J Leukoc Biol 2003; 73:315–322.
as has been described previously for PMN infection by Sta- 17. Sohnle PG, Hunter MJ, Hahn B, Chazin WJ. Zinc-reversible antimi-
phylocccus aureus [45, 46] and may be relevant also in infection crobial activity of recombinant calprotectin (migration inhibitory fac-
with H5N1 Avian influenza [47]. Imaging studies in the skin tor–related proteins 8 and 14). J Infect Dis 2000; 182:1272–1275.
18. Lusitani DL, Malawista SE, Montgomery RR. Calprotectin, an abun-
of sand fly–mediated infections with the intracellular protozoan dant cytosolic protein from human polymorphonuclear leukocytes,
Leishmania reveal that PMNs initially capture the parasites and inhibits the growth of Borrelia burgdorferi. Infect Immun 2003; 71:
contribute to establishment of infection [48]. In addition, as a 4711–4716.
19. Rawal A, Gavin PJ, Sturgis CD. Cerebrospinal fluid cytology in seasonal
vector-transmitted infection, WNV enters the host with vector
epidemic West Nile virus meningo-encephalitis. Diagn Cytopathol
saliva, which has been shown to have immunosuppressive ac- 2006; 34:127–129.
tivities, including the inhibition of PMN functions [49, 50]. 20. Tyler KL, Pape J, Goody RJ, Corkill M, Kleinschmidt-DeMasters BK.
These reports suggest that there may be a more general role of CSF findings in 250 patients with serologically confirmed West Nile
virus meningitis and encephalitis. Neurology 2006; 66:361–365.
PMNs facilitating initial pathogen infections, which may be an 21. Brehin AC, Mouries J, Frenkiel MP, et al. Dynamics of immune cell
important target for early anti-microbial interventions. recruitment during West Nile encephalitis and identification of a new
CD19+B220-BST-2+ leukocyte population. J Immunol 2008; 180:
6760–6767.
22. Anderson JF, Andreadis TG, Vossbrinck CR, et al. Isolation of West
Acknowledgments Nile virus from mosquitoes, crows, and a Cooper’s hawk in Con-
necticut. Science 1999; 286:2331–2333.
We thank Deborah Beck and Xianzhong Liu.
23. Brown CR, Blaho VA, Loiacono CM. Susceptibility to experimental
Lyme arthritis correlates with KC and monocyte chemoattractant pro-
tein-1 production in joints and requires neutrophil recruitment via
References CXCR2. J Immunol 2003; 171:893–901.
24. Bai F, Town T, Qian F, et al. IL-10 signaling blockade controls murine
1. Gubler DJ. The continuing spread of West Nile virus in the western West Nile virus infection. PLoS Pathogens 2009; 5:e1000610.
hemisphere. Clin Infect Dis 2007; 45:1039–1046. 25. Town T, Bai F, Wang T, et al. Tlr7 mitigates lethal West Nile encephalitis
2. Medzhitov R. Toll-like receptors and innate immunity. Nat Rev Im- by affecting interleukin 23-dependent immune cell infiltration and
munol 2001; 1:135–145. homing. Immunity 2009; 30:242–253.
3. Fredericksen BL, Keller BC, Fornek J, Katze MG, Gale M. Establishment 26. Montgomery RR, Lusitani DL, de Boisfleury Chevance A, Malawista
and maintenance of the Innate antiviral response to West Nile virus SE. Human phagocytic cells in the early innate immune response to
involves both RIG-1 and MDA5 signalig through IPS-1. J Virol 2008;82: Borrelia burgdorferi. J Infect Dis 2002; 185:1773–1779.
609–616. 27. Kong K-F, Delroux K, Wang X, et al. Dysregulation of TLR3 impairs
4. Kawai T, Akira S. Antiviral signaling through pattern recognition re- the innate immune response to West Nile virus in the elderly. J Virol
ceptors. J Biochem 2007; 141:137–145. 2008; 82:7613–7623.

PMN in WNV Infection • JID 2010:202 (15 December) • 1811


28. Brown CR, Blaho VA, Loiacono CM. Treatment of mice with the neu- system during West Nile virus encephalitis. J Immunol 2008; 180:
trophil-depleting antibody RB6–8C5 results in early development of 2641–2649.
experimental Lyme arthritis via the recruitment of Gr-1-polymorpho- 40. Grundy JE, Lawson KM, MacCormac LP, Fletcher JM, Yong KL. Cy-
nuclear leukocyte-like cells. Infect Immun 2004; 72:4956–4965. tomegalovirus-infected endothelial cells recruit neutrophils by the se-
29. Daley JM, Thomay AA, Connolly MD, Reichner JS, Albina JE. Use of cretion of C-X-C chemokines and transmit virus by direct neutrophil-
Ly6G-specific monoclonal antibody to deplete neutrophils in mice. J endothelial cell contact and during neutrophil transendothelial
Leukoc Biol 2008; 83:64–70. migration. J Infect Dis 1998; 177:1465–1474.
30. Chen YC, Wang SY. Activation of terminally differentiated human 41. Pugliese A, Vidotto V, Beltramo T, Torre D. Phagocytic activity in
monocytes/macrophages by dengue virus: productive infection, hier- human immunodeficiency virus type 1 infection. Clin Diagn Lab Im-
archical production of innate cytokines and chemokines, and the syn- munol 2005; 12:889–895.
ergistic effect of lipopolysaccharide. J Virol 2002; 76:9877–9887. 42. Gabali AM, Anzinger JJ, Spear GT, Thomas LL. Activation by inflam-
31. Tani K, Su SB, Utsunomiya I, Oppenheim JJ, Wang JM. Interferon- matory stimuli increases neutrophil binding of human immunodefi-
gamma maintains the binding and functional capacity of receptors for ciency virus type 1 and subsequent infection of lymphocytes. J Virol
IL-8 on cultured human T cells. Eur J Immunol 1998; 28:502–507. 2004; 78:10833–10836.
32. Rios M, Zhang MJ, Grinev A, et al. Monocytes-macrophages are a 43. Silva MT. When two is better than one: macrophages and neutrophils
potential target in human infection with West Nile virus through blood work in concert in innate immunity as complementary and cooperative
transfusion. Transfusion 2006; 46:659–667.
partners of a myeloid phagocyte system. J Leukoc Biol 2010; 87:93–106.
33. Daffis S, Samuel MA, Keller BC, Gale M, Diamond MS. Cell-specific
44. Xin L, Vargas-Inchaustegui DA, Raimer SS, et al. Type I IFN receptor
IRF-3 responses protect against West Nile virus infection by interferon-
regulates neutrophil functions and innate immunity to Leishmania
dependent and -independent mechanisms. PLoS Pathog 2007; 3:e106.
parasites. J Immunol 2010; 184:7047–5706.
34. Kong K-F, Wang X, Anderson JF, Fikrig E, Montgomery RR. West Nile
45. Gresham HD, Lowrance JH, Caver TE, Wilson BS, Cheung AL, Lind-

Downloaded from jid.oxfordjournals.org by guest on February 27, 2011


virus attenuates activation of primary human macrophages. Viral Im-
berg FP. Survival of 7Staphylococcus aureus inside neutrophils contrib-
munol 2008; 21:78–82.
utes to infection. J Immunol 2000; 164:3713–3722.
35. Panda A, Qian F, Mohanty S, et al. Age-associated decrease in TLR
function in primary human dendritic cells predicts influenza vaccine 46. Voyich JM, Braughton KR, Sturdevant DE, et al. Insights into mech-
response. J Immunol 2010; 184:2518–2527. anisms used by Staphylococcus aureus to avoid destruction by human
36. Butthep P, Bunyaratvej A, Bhamarapravati N. Dengue virus and en- neutrophils. J Immunol 2005; 175:3907–3919.
dothelial cell: a related phenomenon to thrombocytopenia and gran- 47. Zhao Y, Lu M, Lau LT, et al. Neutrophils may be a vehicle for viral
ulocytopenia in dengue hemorrhagic fever. Southeast Asian J Trop Med replication and dissemination in human H5N1 avian influenza. Clin
Public Health 1993; 24(Suppl 1):246–249. Infect Dis 2008; 47:1575–1578.
37. Liu CC, Huang KJ, Lin YS, Yeh TM, Liu HS, Lei HY. Transient CD4/ 48. Peters NC, Egen JG, Secundino N, et al. In vivo imaging reveals an
CD8 ratio inversion and aberrant immune activation during dengue essential role for neutrophils in leishmaniasis transmitted by sand flies.
virus infection. J Med Virol 2002; 68:241–252. Science 2008; 321:970–974.
38. Klein RS, Lin E, Zhang B, et al. Neuronal CXCL10 directs CD8+ T- 49. Ribeiro JM, Francischetti IM. Role of arthropod saliva in blood feeding:
cell recruitment and control of West Nile virus encephalitis. J Virol sialome and post-sialome perspectives. Annu Rev Entomol 2003; 48:
2005; 79:11457–1166. 73–88.
39. Zhang B, Chan YK, Lu B, Diamond MS, Klein RS. CXCR3 mediates 50. Guo X, Booth CJ, Paley MA, et al. Inhibition of neutrophil function
region-specific antiviral T cell trafficking within the central nervous by two tick salivary proteins. Infect Immun 2009; 77:2320–2329.

1812 • JID 2010:202 (15 December) • Bai et al

Das könnte Ihnen auch gefallen