Download Word

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

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

Document related concepts

Ebola virus disease wikipedia , lookup

Pandemic wikipedia , lookup

Trichinosis wikipedia , lookup

Dirofilaria immitis wikipedia , lookup

Norovirus wikipedia , lookup

Middle East respiratory syndrome wikipedia , lookup

Sarcocystis wikipedia , lookup

Schistosomiasis wikipedia , lookup

West Nile fever wikipedia , lookup

Chickenpox wikipedia , lookup

Marburg virus disease wikipedia , lookup

HIV wikipedia , lookup

Antiviral drug wikipedia , lookup

Oesophagostomum wikipedia , lookup

Hepatitis C wikipedia , lookup

Coccidioidomycosis wikipedia , lookup

Hospital-acquired infection wikipedia , lookup

Herpes simplex virus wikipedia , lookup

Neonatal infection wikipedia , lookup

Henipavirus wikipedia , lookup

Human cytomegalovirus wikipedia , lookup

Hepatitis B wikipedia , lookup

Lymphocytic choriomeningitis wikipedia , lookup

Transcript
1
2
Altered immune response of immature dendritic cells upon dengue virus
infection in the presence of specific antibodies
3
4
Silvia Torres1,2, Jacky Flipse1, Vinit C. Upasani1, Heidi van der Ende- Metselaar1, Silvio
5
Urcuqui-Inchima2, Jolanda M. Smit1, Izabela A. Rodenhuis-Zybert1
6
1Department
7
Center Groningen, Groningen, The Netherlands.
8
2Grupo
9
No. 52-21, Medellín, Colombia.
of Medical Microbiology, University of Groningen and University Medical
Inmunovirologia, Facultad de Medicina, Universidad de Antioquia UdeA, calle 70
10
Email addresses:
11
ST: [email protected]
12
JF: [email protected]
13
VU: [email protected]
14
SUI: [email protected],
15
HEM: [email protected],
16
JMS: [email protected],
17
IRZ: [email protected]
18
Corresponding author (Rodenhuis-Zybert, IA): Email: [email protected]
19
20
Word count: 2563
21
1
22
23
Abstract
24
Dengue virus (DENV) replication is known to prevent maturation of infected DCs thereby
25
impeding the development of adequate immunity. During secondary DENV infection,
26
dengue-specific antibodies can suppress DENV replication in immature DCs (immDCs),
27
however how dengue-antibody complexes (DENV-IC) influence DCs phenotype remains
28
elusive. Here, we evaluated the maturation state and cytokine profile of immDCs exposed
29
to DENV-ICs. Indeed, DENV infection of immDCs in the absence of antibodies was
30
hallmarked by blunted upregulation of CD83, CD86 and the major histocompatibility
31
complex molecule HLA-DR. In contrast, DENV infection in the presence of neutralizing
32
antibodies triggered full DCs maturation and induced a balanced inflammatory cytokine
33
response. Moreover, DENV infection at non-neutralizing conditions prompted upregulation
34
of CD83 and CD86 but not that of HLA-DR and triggered production of pro-inflammatory
35
cytokines. The effect of DENV-IC was found to rely on the engagement of FcγRIIa.
36
Altogether, our data show that the presence of DENV-IC alters the phenotype and cytokine
37
profile of DCs.
38
39
Introduction
40
Dengue disease is the most prevalent mosquito-borne viral disease with approximately 400
41
million infections per year worldwide (Bhatt et al., 2013). The disease is caused by
42
infection with any of the four serotypes of dengue virus (DENV1-4). Primary (1°) infections
43
are often asymptomatic and provide life-long protection against homotypic re-infection yet
44
short-term (1-3 years) protection against heterotypic re-infections (Reich et al., 2013).
45
Strikingly, after the cross-protective period, individuals re-infected with a heterologous
46
virus serotype are at risk of developing severe disease (Halstead, 2007). Severe disease is
47
also seen during 1° infections in infants with low titres of circulating maternal dengue
48
antibodies (Kliks et al., 1988; Simmons et al., 2007; Hause et al., 2015). Clearly, antibodies
49
play an important role in the outcome of DENV infection. Indeed, in vitro and in vivo studies
2
50
demonstrated that DENV infectivity depends on the concentrations and/or avidity of
51
antibodies present during infection. DENV infection is contained in the presence of
52
neutralizing antibody titers (de Alwis et al., 2011; Endy et al., 2004; van der Schaar et al.,
53
2009). At non-neutralizing antibody concentrations however, enhanced infection of
54
immune cells (antibody-dependent enhancement (ADE) is seen due to viral entry via Fcγ
55
receptors (FcγR) (Rodenhuis-Zybert et al., 2010; Modhiran et al., 2010; Rodrigo et al., 2006;
56
Moi et al., 2010).
57
Dendritic cells (DCs) are the front line of defense against invading pathogens (Lutz &
58
Schuler, 2002). Upon antigen recognition, immature DCs (immDCs) acquire a mature
59
phenotype (matDCs) characterized by high surface expression of costimulatory molecules,
60
major histocompatibility complex II molecules (HLA-DR) and secretion of pro- and anti-
61
inflammatory cytokines (Banchereau & Steinman, 1998; Cella et al., 1997). Activation of
62
DCs is crucial for shaping the innate responses and initiation of adaptive immunity (Dudek
63
et al., 2013). However, DCs also represent an important target for DENV replication during
64
1° and 2° infections (Marovich et al., 2001; Wu et al., 2000; Schmid and Harris et al. 2014).
65
Indeed, in the absence of antibodies (direct infection), DENV not only efficiently infects
66
immDCs (Boonnak et al., 2008; Nightingale et al.,2008) but is also able to blunt their
67
maturation (Chang et al., 2012; Munoz-Jordan et al.,2003; Munoz-Jordan, 2010; Rodriguez-
68
Madozet al., 2010). High titers of DENV-specific antibodies are capable of neutralizing
69
DENV infection in immDCs (Boonnak et al., 2008). In the presence of non-neutralizing
70
antibody concentrations however, immDCs may support ADE of DENV infection (Boonnak
71
et al., 2008; Dejnirattisai et al., 2011 ). Remarkably, to date, little is known how DENV-
72
specific antibodies influence the activation of immDCs upon DENV infection.
73
In this study, we evaluated the effect of DENV-specific antibodies on immDC activation.
74
ImmDCs were generated from human monocytes isolated from buffy coats obtained from
75
healthy, anonymous volunteers (Sanquin Bloodbank) and processed in line with the
76
declaration of Helsinki. Briefly, human peripheral blood mononuclear cells (PBMC) were
77
isolated from buffy coats by centrifugation using Ficoll-Paque™ Plus (GE Healthcare).
78
Monocytes were isolated from PBMCs by gelatin adherence as previously described (Miller
79
et al., 2008) and allowed to differentiate into immDCs by culturing at 37oC in complete
3
80
RPMI medium (CM) with 20% fetal bovine serum (FBS), 500U/ml recombinant granulocyte
81
macrophage colony-stimulating factor (rGM-CSF), and 250U/ml recombinant interleukin-4
82
(IL-4; both from Prospec-Tany). Three-quarters of the culture medium was replaced every
83
second day for 6 days to generate immDCs. After 6 days of culture, the phenotype of the
84
cells was characterized as Lin-1neg, HLA-DRpos and CD11cpos (Supplementary Figure 1A),
85
thus confirming the differentiation of monocytes into immDCs (Boonnak et al., 2008). To
86
generate matDCs that would serve as a positive control in our experiments, immDCs were
87
stimulated at day 6 with MCM mimic containing 15 μg/ml recombinant IL-6, 500ng/ml
88
recombinant IL-1β, 500ng/ml TNF-α and 100μg/ml prostaglandin E2 (all from Bio
89
Connect), as described in Boonnak et al., 2008. At 24 hours post-stimulation, DCs showed
90
increased expression of CD40, CD83, CD86, HLA-DR compared to non-stimulated cells,
91
which confirms the inherent capacity of our immDCs to mature (Supplementary Figure 1B).
92
First, we analyzed the permissiveness of immDCs to DENV-2 infection. DENV-2 strain
93
16681 was propagated in Aedes albopictus cell line C6/36, as described before (Rodenhuis-
94
Zybert et al., 2010). The infectivity of DENV was determined by measuring the number of
95
plaque-forming units (PFU) by plaque assay (PA) on BHK-15 cells and the number of
96
genome-equivalent copies (GEc) by quantitative RT-PCR (qRT-PCR), as described
97
previously (van der Schaar et al., 2007; Zybert et al., 2008). The cells were infected with
98
DENV at multiplicity of infection (MOI) 0.1, 1, 10 at 37⁰C. At 2 hpi, the inoculum was
99
removed, cells were washed and cultured in fresh CM for another 22-24 hours. At 24-
100
26hpi, cells and supernatants were harvested and separated by low-speed centrifugation
101
(5’, 250g). The percentage of DENV-positive cells was evaluated by flow cytometry using
102
the DENV-2 envelope (E)-specific antibody 3H5 (Millipore) followed by a secondary anti-
103
mouse Alexa-647-labeled antibody (Figure 1A). As expected, the infection rate was found
104
to be MOI-dependent with the percentage of percentage of DENV E-positive varying
105
between the donors. Progeny virus production was measured in the cell-free supernatants
106
by plaque assay and qRT-PCR to obtain both PFU and GEc titers (Figure 1B). In agreement
107
with previous reports (Boonnak et al., 2008; Boonnak et al., 2011), immDCs are highly
108
permissive to DENV-2 replication.
4
109
To test the effect of antibodies on the permissiveness of immDCs to DENV-2, immDCs were
110
infected at MOI of 1 with DENV-2 alone (Direct infection) or DENV-2 immune-complexes
111
(DENV-IC). To generate DENV-IC, the virus was pre-incubated for 1 hour at 37°C with 10-
112
fold sequential dilutions of DENV-2-immune serum in a 1:1 volume ratio. In all
113
experiments, two distinct convalescent DENV-2 immune sera (kindly provided by G.
114
Comach Biomed-UC, Lardidev, Maracay, Venezuela; and T. Kochel, U.S. Naval Medical
115
Research Center Detachment, Lima, Peru) were used as a source of polyclonal antibodies.
116
The capacity of the sera to neutralize and enhance the DENV infection had been previously
117
confirmed in macrophage cell lines, human PBMCs and matDCs (Rodenhuis-Zybert et al.,
118
2010 and unpublished). The DENV-immune sera used exhibited potent neutralizing activity
119
and inhibited DENV-2 infection at dilutions of 10-2 to 10-6 for three distinct donors
120
(Supplementary Figure 2, data for 10-3 dilution is shown in Figure 1C and 1D,). The PFU
121
and GEc titers were below the detection limit (20PFU/mL and 400 GEc/mL, respectively)
122
following infection with DENV pre-opsonized with 10-3 diluted serum (Figure 1D).
123
Therefore, we used this serum dilution for neutralization conditions (Neutr). As expected,
124
pre-incubation of DENV-2 with higher dilutions of the serum (Supplementary Figure 2; data
125
for 10- 8 dilution is shown in Figure 1) recovered DENV infectivity to the levels of direct
126
infection, as evidenced by flow cytometric analysis (Figure 1C) as well as DENV PFU and
127
GEc titers in the cell supernatants (Figure 1D). Hence, 10-8 serum dilution was used for non-
128
neutralizing conditions (Non-neutr) in subsequent experiments. Importantly, in line with a
129
previous report (Boonnak et al., 2008), none of the sera dilutions was able to enhance
130
DENV infection in immDCs (Supplementary Figure 2).
131
The effect of direct DENV infection on the maturation of immDCs has been studied
132
previously (Boonnak et al., 2008; Boonnak et al., 2001; Libraty et al., 2001; Navarro-
133
Sanchez et al., 2005; Nightingale et al., 2008; Palmer et al., 2005; Sun et al., 2009) yet the
134
effect of DENV-ICs on DCs remains unknown. To assess this, we next evaluated the surface
135
expression of the DCs maturation markers CD83, CD86 and HLA-DR 24hpi following
136
exposure of the cells to direct, UV-inactivated virus, non-neutralizing and neutralizing
137
DENV infection conditions. Twenty four hpi, cells were harvested, placed into cytometry
138
tubes and treated with FcRs blocking buffer (True Stain FcX, Biolegend) for 10 min at room
5
139
temperature. Subsequently, the cells were washed in staining medium [EDTA, saponin
140
(both from Sigma) and 2% FBS. Cell viability was evaluated using LIVE/DEAD® Fixable
141
Dead Cell Stain Kit (Invitrogen). Phenotyping of the cells with a panel of DCs markers and
142
intracellular DENV staining were performed, as described previously (Richter et al., 2014;
143
van der Schaar et al., 2008). Mock-infected (with or without the addition of neutralizing/
144
non-neutralizing DENV-immune serum) and MCM mimic-stimulated cells (matDCs) were
145
added as negative and positive controls, respectively.
146
Figure 2 illustrates the effects of different infection conditions on the levels of CD83, CD86
147
and HLA-DR compared to matched mock infections. Expression of CD40 was upregulated
148
during all infection conditions (data not shown). In line with above-mentioned studies,
149
direct DENV infection did not promote significant upregulation of the maturation markers.
150
Exposure of cell to UV-inactivated DENV-2 (Direct UVi, complete inactivation confirmed by
151
PA) led to a modest increase in expression levels of CD83 and CD86, but had no effect on
152
HLA-DR. Interestingly, exposure of cells to DENV at neutralizing serum concentrations
153
triggered a significant increase in the expression of CD83, CD86 and HLA-DR when
154
compared to direct infection condition. In fact, the expression level of the maturation
155
markers found in DCs infected at neutralizing conditions was either higher (p>0.1 for
156
CD86) or comparable to that of matDCs (Figure 2). The observed upregulation was not
157
solely due to the blockage of DENV replication as expression of CD83 and HLA-DR
158
following exposure to UV-inactivated DENV where significantly lower (Figure 2).
159
Furthermore, DENV infection at non-neutralizing conditions also led to an increase in the
160
expression pattern of the maturation markers (with p<0.1 for CD86) as compared to direct
161
infection. Together, these results imply that the presence of DENV-IC promoted the
162
maturation capacity of DCs.
163
Subsequently, we examined whether differences in the expression level of DC markers
164
translated to a particular cytokine expression profile. To this end, we evaluated the levels
165
of pro- (IL-6, TNF-α), and anti-inflammatory cytokines (IL-10, IL-4) in the supernatants of
166
DCs infected at conditions as described above (Figure 3A). In line with the lack of DCs
167
maturation, direct infection did not significantly alter the levels of the aforementioned
168
cytokines when compared to mock infection. Importantly, when immDCs were exposed to
6
169
neutralizing DENV-IC, the cells secreted significantly higher amounts of IL-6, TNF-α, IL-4
170
and IL-10 when compared to direct infection. In line with the data obtained for surface
171
markers, addition of the neutralizing serum alone (Figure 3B), as well as Direct UVi (data
172
not shown), did not trigger significant changes in the cytokine response. Taken together,
173
the observed increase upon infection in presence of the serum was likely due to the
174
presence of DENV-IC. This notion was further supported by the observation that non-
175
neutralizing DENV-infection conditions resulted in higher, albeit variable between the
176
donors, levels of IL-6, IL-4 and TNF-α than those induced by the direct infection (Figure
177
3A). Interestingly, it has recently been reported that induction of pro-inflammatory
178
cytokines following immune-complex stimulation depends mainly on FcγRIIa (Vogelpoel
179
L.T.C. et al., 2014). Ligation of FcγRIIa is also required for the production of IL-6 and TNF-α
180
following infection of matDCs under ADE conditions (Boonnak et al., 2008). To assess the
181
role of FcγRIIa in the activation of immDCs in the context of DENV-infection, we pre-treated
182
the immDCs with for 30 minutes with 5mg/mL of FcγRIIa-blocking antibody, clone IV.3
183
(Stem Cell). Figure 3B and 3C show that blocking FcγRIIa had no effect on the levels of
184
cytokines in the conditions of mock infections. However, it did prevent the production of
185
cytokines during infection in the presence of neutralizing (Figure 3B) and non-neutralizing
186
(Figure 3C) serum concentrations. Thus, FcγRIIa ligation was responsible for the
187
production of IL-6 and TNF-α following infection with DENV-IC. Of note, blocking of FcγRIIa
188
had no effect on the neutralizing capacity of the antibodies or on the level of infection at
189
non-neutralizing conditions (Supplementary Figure S3).
190
In summary, our data demonstrate that the presence of DENV-IC triggers distinct DC
191
phenotypes and cytokine profiles.
192
The main role of DCs is to sense, process and present antigens of invading pathogens to
193
cells of the adaptive immune system (Banchereau & Steinman, 1998). Viruses as HIV-1,
194
measles virus, vaccinia virus and DENV target DCs for replication (Boonnak et al., 2008; de
195
Witte et al.,2006; Dejnirattisai et al., 2011; Ho et al., 2001; Liu et al., 2008; Marovich et al.,
196
2001; Nightingale et al., 2008; Rinaldo, 2013; Wu et al., 2000). Given the pivotal role of DCs
197
in promoting adaptive immune responses, it is not surprising that many viruses impair the
198
ability of infected DCs to initiate adaptive immunity (Lilley & Ploegh, 2005; Oreshkova et
7
199
al., 2015). Indeed, several studies have shown that DC maturation is blunted upon DENV
200
infection (Chang et al., 2012; Munoz-Jordan et al., 2003; Rodriguez-Madoz et al., 2010;
201
Palmer et al., 2005). In agreement with this, we found that DENV replication impedes DCs
202
maturation. Although not tested here, previous study has shown that DENV infection in DCs
203
impairs their antigen-presenting cell function (Palmer et al., 2005). Indeed, clinical data
204
show that antigen-presenting cells in patients suffering from acute DV infections are unable
205
to stimulate T-cell responses to mitogens and DV antigens (Mathew, A., et al, 1999). Our
206
data suggest that the presence of DENV-specific antibodies may exert distinct
207
immunomodulatory effects in immDCs during 2° infection. At conditions of antibody-
208
mediated virus neutralization, the expression of HLA-DR, CD83 and CD86 is up-regulated to
209
levels similar as mock-infected matDCs. Indeed, binding of ICs to FcRs on DCs is known to
210
trigger phagocytosis, presentation of antigenic peptides on MHC class I and class II
211
molecules and, depending on the FcR, differential cytokine production (den Dunnen et al.,
212
2012; Nimmerjahn & Ravetch, 2008; Vogelpoel L.T.C. et al., 2014). In line with this, we
213
observed increased production of pro-inflammatory (IL-6, and TNF-α) and anti-
214
inflammatory (IL-4 and IL-10) cytokines at conditions of DENV neutralization.
215
Exposure of immDCs to DENV-ICs at non-neutralizing conditions triggered a significant
216
increase of CD83 and CD86 but did not alter the expression patterns of the HLA-DR.
217
Moreover, TNF-α, IL-6, and IL-4 but not IL-10 were released at this condition. The presence
218
of IL-4 is known to inhibit IL-10 production by DCs (Yao et al., 2005). It is possible that the
219
lack of IL-10 production is due to the significantly elevated levels of IL-4 released from DCs
220
exposed to non-neutralizing conditions. Notably, balanced levels of TNF-α and IL-10 have
221
been shown to be important for control the inflammatory responses (van der Poll et al.,
222
1995, Gudmundsson et al., 1998). Thus it is tempting to speculate that this might be yet
223
another mechanism contributing to a cytokine storm observed in course of severe dengue
224
(Costa et al., 2013; Pang et al., 2007; Soundravally et al., 2013). Mechanistically, the overall
225
increase in cytokine production upon infection with non-neutralized DENV-ICs compared
226
to direct infection suggests that FcRs-mediated infection triggers different downstream
227
signaling pathways.
8
228
The presence of DENV-specific antibodies during DENV infection of DCs has consequences
229
for the maturation of immDCs and subsequent cytokine responses. Our results corroborate
230
earlier findings that DENV-2 can blunt the maturation and activation of exposed DCs.
231
Importantly, we show that the presence of high concentrations of DENV-specific antibodies
232
does not only neutralize DENV infection of immDCs but also rescues the ability of immDCs
233
to mature and produce pro- and anti-inflammatory cytokines. On the other hand, infection
234
in the presence of non-neutralizing antibody titers may induce a phenotype of DCs with
235
reduced ability to present antigens while triggering mainly pro-inflammatory responses.
236
Our data also showed that the ability of DCs to acquire these distinct phenotypes relies on
237
FcγRIIa ligation. Further studies should investigate whether this partially impaired DCs
238
phenotype contributes to the aberrant T responses and exacerbation of inflammation seen
239
during severe disease (Green & Rothman, 2006; Mangada & Rothman, 2005; Rothman,
240
2011).
241
242
Acknowledgments
243
JF and JMS were supported by Dutch Scientific Organization (NWO) VIDI-grant to JMS. ST
244
was supported by Colciencias, Colombia (#111551928777) and SUI by Universidad de
245
Antioquia, (Programa de Sostenibilidad 2016-2017) and UMCG. IRZ was supported by
246
NWO VENI-grant to IRZ. The funders had no role in study design, data collection and
247
analysis, decision to publish, or preparation of the manuscript.
248
249
Author Contributions
250
Conceived and designed the experiments: ST, JF, VU, IRZ
251
Performed the experiments: ST, JF, VU, HEM
252
Analyzed the data: ST, JF, VU, IRZ, JMS, SUI
253
Contributed with reagents/materials/analysis tools: SUI and JMS
9
254
Wrote the paper: ST, VU, JMS and IRZ
255
256
References
257
Banchereau, J., & Steinman, R. M. (1998). Dendritic cells and the control of immunity.
258
Nature, 392(6673), 245-252. doi:10.1038/32588
259
Bhatt, S., Gething, P. W., Brady, O. J., Messina, J. P., Farlow, A. W., Moyes, C. L., Hay, S. I.
260
(2013). The global distribution and burden of dengue. Nature, 496(7446), 504-507.
261
doi:10.1038/nature12060; 10.1038/nature12060
262
Boonnak, K., Dambach, K. M., Donofrio, G. C., Tassaneetrithep, B., & Marovich, M. A. (2011).
263
Cell type specificity and host genetic polymorphisms influence antibody-dependent
264
enhancement of dengue virus infection. Journal of Virology, 85(4), 1671-1683.
265
doi:10.1128/JVI.00220-10; 10.1128/JVI.00220-10
266
Boonnak, K., Slike, B. M., Burgess, T. H., Mason, R. M., Wu, S. J., Sun, P., Marovich, M. A.
267
(2008). Role of dendritic cells in antibody-dependent enhancement of dengue virus
268
infection.
269
10.1128/JVI.02484-07
270
Cella, M., Sallusto, F., & Lanzavecchia, A. (1997). Origin, maturation and antigen presenting
271
function of dendritic cells. Current Opinion in Immunology, 9(1), 10-16.
272
Chang, T. H., Chen, S. R., Yu, C. Y., Lin, Y. S., Chen, Y. S., Kubota, T., Lin, Y. L. (2012). Dengue
273
virus serotype 2 blocks extracellular signal-regulated kinase and nuclear factor-kappaB
274
activation
275
doi:10.1371/journal.pone.0041635; 10.1371/journal.pone.0041635
276
Chawla, T., Chan, K. R., Zhang, S. L., Tan, H. C., Lim, A. P., Hanson, B. J., & Ooi, E. E. (2013).
277
Dengue virus neutralization in cells expressing fc gamma receptors. PloS One, 8(5), e65231.
278
doi:10.1371/journal.pone.0065231; 10.1371/journal.pone.0065231
Journal
to
of
Virology,
downregulate
82(8),
cytokine
3939-3951.
production.
10
doi:10.1128/JVI.02484-07;
PloS
One,
7(8),
e41635.
279
Costa, V. V., Fagundes, C. T., Souza, D. G., & Teixeira, M. M. (2013). Inflammatory and innate
280
immune responses in dengue infection: Protection versus disease induction. The American
281
Journal
282
10.1016/j.ajpath.2013.02.027
283
de Alwis, R., Beltramello, M., Messer, W. B., Sukupolvi-Petty, S., Wahala, W. M., Kraus, A., de
284
Silva, A. M. (2011). In-depth analysis of the antibody response of individuals exposed to
285
primary dengue virus infection. PLoS Neglected Tropical Diseases, 5(6), e1188.
286
doi:10.1371/journal.pntd.0001188; 10.1371/journal.pntd.0001188
287
de Witte, L., Abt, M., Schneider-Schaulies, S., van Kooyk, Y., & Geijtenbeek, T. B. (2006).
288
Measles virus targets DC-SIGN to enhance dendritic cell infection. Journal of Virology,
289
80(7), 3477-3486. doi:10.1128/JVI.80.7.3477-3486.2006
290
Dejnirattisai, W., Webb, A. I., Chan, V., Jumnainsong, A., Davidson, A., Mongkolsapaya, J., &
291
Screaton, G. (2011). Lectin switching during dengue virus infection. The Journal of
292
Infectious Diseases, 203(12), 1775-1783. doi:10.1093/infdis/jir173; 10.1093/infdis/jir173
293
den Dunnen, J., Vogelpoel, L. T., Wypych, T., Muller, F. J., de Boer, L., Kuijpers, T. W., de Jong,
294
E. C. (2012). IgG opsonization of bacteria promotes Th17 responses via synergy between
295
TLRs
296
doi:10.1182/blood-2011-12-399931; 10.1182/blood-2011-12-399931
297
Dudek, A. M., Martin, S., Garg, A. D., & Agostinis, P. (2013). Immature, Semi-Mature, and
298
Fully Mature Dendritic Cells: Toward a DC-Cancer Cells Interface That Augments
299
Anticancer
300
http://doi.org/10.3389/fimmu.2013.00438
301
Endy, T. P., Nisalak, A., Chunsuttitwat, S., Vaughn, D. W., Green, S., Ennis, F. A., Libraty, D. H.
302
(2004). Relationship of preexisting dengue virus (DV) neutralizing antibody levels to
303
viremia and severity of disease in a prospective cohort study of DV infection in thailand.
304
The Journal of Infectious Diseases, 189(6), 990-1000. doi:10.1086/382280
of
and
Pathology,
FcgammaRIIa
Immunity.
182(6),
in
1950-1961.
human
dendritic
Frontiers
in
11
doi:10.1016/j.ajpath.2013.02.027;
cells.
Blood,
120(1),
Immunology,
4,
112-121.
438.
305
Green, S., & Rothman, A. (2006). Immunopathological mechanisms in dengue and dengue
306
hemorrhagic
307
doi:10.1097/01.qco.0000244047.31135.fa
308
Halstead, S. B. (2003). Neutralization and antibody-dependent enhancement of dengue
309
viruses. Advances in Virus Research, 60, 421-467.
310
Halstead, S. B. (2007). Dengue. Lancet, 370(9599), 1644-1652. doi:10.1016/S0140-
311
6736(07)61687-0
312
Hause AM, Perez-Padilla J, Horiuchi K, Han GS, Hunsperger E1, Aiwazian J, Margolis HS,
313
Tomashek KM (2015). Epidemiology of Dengue Among Children Aged < 18 Months-Puerto
314
Rico, 1999-2011.doi: 10.4269/ajtmh.15-0382
315
Ho, L. J., Wang, J. J., Shaio, M. F., Kao, C. L., Chang, D. M., Han, S. W., & Lai, J. H. (2001).
316
Infection of human dendritic cells by dengue virus causes cell maturation and cytokine
317
production. Journal of Immunology (Baltimore, Md.: 1950), 166(3), 1499-1506.
318
Kliks S.C., Nimmanitya S, Nisalak A, Burke D.S. (1988)Evidence that maternal dengue
319
antibodies are important in the development of dengue hemorrhagic fever in infants.Am J
320
Trop Med Hyg. 1988 Mar;38(2):411-9.
321
Libraty, D. H., Pichyangkul, S., Ajariyakhajorn, C., Endy, T. P., & Ennis, F. A. (2001). Human
322
dendritic cells are activated by dengue virus infection: Enhancement by gamma interferon
323
and implications for disease pathogenesis. Journal of Virology, 75(8), 3501-3508.
324
doi:10.1128/JVI.75.8.3501-3508.2001
325
Lilley, B. N., & Ploegh, H. L. (2005). Viral modulation of antigen presentation: Manipulation
326
of cellular targets in the ER and beyond. Immunological Reviews, 207, 126-144.
327
doi:10.1111/j.0105-2896.2005.00318.x
328
Liu, L., Chavan, R., & Feinberg, M. B. (2008). Dendritic cells are preferentially targeted
329
among hematolymphocytes by modified vaccinia virus ankara and play a key role in the
330
induction of virus-specific T cell responses in vivo. BMC Immunology, 9, 15-2172-9-15.
331
doi:10.1186/1471-2172-9-15; 10.1186/1471-2172-9-15
fever.
Current
Opinion
in
12
Infectious
Diseases,
19(5),
429-436.
332
Lutz, M. B., & Schuler, G. (2002). Immature, semi-mature and fully mature dendritic cells:
333
Which signals induce tolerance or immunity? Trends in Immunology, 23(9), 445-449.
334
Mangada, M. M., & Rothman, A. L. (2005). Altered cytokine responses of dengue-specific
335
CD4+ T cells to heterologous serotypes. Journal of Immunology (Baltimore, Md.: 1950),
336
175(4), 2676-2683.
337
Marovich, M., Grouard-Vogel, G., Louder, M., Eller, M., Sun, W., Wu, S. J., . . . Mascola, J.
338
(2001). Human dendritic cells as targets of dengue virus infection. The Journal of
339
Investigative Dermatology.Symposium Proceedings / the Society for Investigative
340
Dermatology, Inc.[and] European Society for Dermatological Research, 6(3), 219-224.
341
doi:10.1046/j.0022-202x.2001.00037.x
342
Mathew A, Kurane I, Green S, Vaughn D.W., Kalayanarooj S, Suntayakorn S, Ennis F. A.,
343
Rothman A. L. (1999). Impaired T Cell Proliferation in Acute Dengue Infection. J Immunol
344
1999; 162:5609-5615.
345
Miller, R. L., Meng, T. C., & Tomai, M. A. (2008). The antiviral activity of toll-like receptor 7
346
and 7/8 agonists. Drug News & Perspectives, 21(2), 69-87.
347
Modhiran, N., Kalayanarooj, S., & Ubol, S. (2010). Subversion of Innate Defenses by the
348
Interplay between DENV and Pre-Existing Enhancing Antibodies: TLRs Signaling Collapse.
349
PLoS
350
http://doi.org/10.1371/journal.pntd.0000924
351
Moi M.L., Lim C.K., Takasaki T., Kurane I. (2010). Involvement of the Fc gamma receptor IIA
352
cytoplasmic domain in antibody-dependent enhancement of dengue virus infection. J Gen
353
Virol. 2010 Jan;91(Pt 1):103-11. doi: 10.1099/vir.0.014829-0. Epub 2009 Sep 23.
354
Munoz-Jordan, J. L. (2010). Subversion of interferon by dengue virus. Current Topics in
355
Microbiology
356
10.1007/978-3-642-02215-9_3
357
Munoz-Jordan, J. L., Sanchez-Burgos, G. G., Laurent-Rolle, M., & Garcia-Sastre, A. (2003).
358
Inhibition of interferon signaling by dengue virus. Proceedings of the National Academy of
Neglected
and
Tropical
Immunology,
338,
Diseases,
35-44.
13
4(12),
e924.
doi:10.1007/978-3-642-02215-9_3;
359
Sciences
of
the
360
doi:10.1073/pnas.2335168100
361
Navarro-Sanchez, E., Despres, P., & Cedillo-Barron, L. (2005). Innate immune responses to
362
dengue
363
doi:10.1016/j.arcmed.2005.04.007
364
Nightingale, Z. D., Patkar, C., & Rothman, A. L. (2008). Viral replication and paracrine effects
365
result in distinct, functional responses of dendritic cells following infection with dengue 2
366
virus. Journal of Leukocyte Biology, 84(4), 1028-1038. doi:10.1189/jlb.0208105;
367
10.1189/jlb.0208105
368
Nimmerjahn, F., & Ravetch, J. V. (2008). Fcgamma receptors as regulators of immune
369
responses. Nature Reviews.Immunology, 8(1), 34-47. doi:10.1038/nri2206
370
Oreshkova, N., Wichgers Schreur, P. J., Spel, L., Vloet, R. P. M., Moormann, R. J. M., Boes, M., &
371
Kortekaas, J. (2015). Nonspreading Rift Valley Fever Virus Infection of Human Dendritic
372
Cells Results in Downregulation of CD83 and Full Maturation of Bystander Cells. PLoS ONE,
373
10(11), e0142670. http://doi.org/10.1371/journal.pone.0142670
374
Palmer, D. R., Sun, P., Celluzzi, C., Bisbing, J., Pang, S., Sun, W., . . . Burgess, T. (2005).
375
Differential effects of dengue virus on infected and bystander dendritic cells. Journal of
376
Virology, 79(4), 2432-2439. doi:10.1128/JVI.79.4.2432-2439.2005
377
Pang, T., Cardosa, M. J., & Guzman, M. G. (2007). Of cascades and perfect storms: The
378
immunopathogenesis of dengue haemorrhagic fever-dengue shock syndrome (DHF/DSS).
379
Immunology and Cell Biology, 85(1), 43-45. doi:10.1038/sj.icb.7100008
380
Regnault, A., Lankar, D., Lacabanne, V., Rodriguez, A., Thery, C., Rescigno, M., Amigorena, S.
381
(1999). Fcgamma receptor-mediated induction of dendritic cell maturation and major
382
histocompatibility complex class I-restricted antigen presentation after immune complex
383
internalization. The Journal of Experimental Medicine, 189(2), 371-380.
384
Reich, N. G., Shrestha, S., King, A. A., Rohani, P., Lessler, J., Kalayanarooj, S., Cummings, D. A.
385
(2013). Interactions between serotypes of dengue highlight epidemiological impact of
virus.
United
Archives
States
of
of
Medical
14
America,
100(24),
Research,
14333-14338.
36(5),
425-435.
386
cross-immunity. Journal of the Royal Society, Interface / the Royal Society, 10(86),
387
20130414. doi:10.1098/rsif.2013.0414; 10.1098/rsif.2013.0414
388
Richter, M. K. S., da Silva Voorham, J. M., Torres Pedraza, S., Hoornweg, T. E., van de Pol, D. P.
389
I., Rodenhuis-Zybert, I. A., Wilschut J., Smit, J. M. (2014). Immature Dengue Virus Is
390
Infectious in Human Immature Dendritic Cells via Interaction with the Receptor Molecule
391
DC-SIGN. PLoS ONE, 9(6), e98785. http://doi.org/10.1371/journal.pone.0098785
392
Rinaldo, C. R. (2013). HIV-1 trans infection of CD4(+) T cells by professional antigen
393
presenting
394
10.1155/2013/164203
395
Rodenhuis-Zybert, I. A., van der Schaar, H. M., da Silva Voorham, J. M., van der Ende-
396
Metselaar, H., Lei, H. Y., Wilschut, J., & Smit, J. M. (2010). Immature dengue virus: A veiled
397
pathogen?
398
10.1371/journal.ppat.1000718
399
Rodrigo, W. W. S. I., Jin, X., Blackley, S. D., Rose, R. C., & Schlesinger, J. J. (2006). Differential
400
Enhancement of Dengue Virus Immune Complex Infectivity Mediated by Signaling-
401
Competent and Signaling-Incompetent Human FcγRIA (CD64) or FcγRIIA (CD32). Journal
402
of Virology, 80(20), 10128–10138. http://doi.org/10.1128/JVI.00792-06
403
Rodriguez-Madoz, J. R., Belicha-Villanueva, A., Bernal-Rubio, D., Ashour, J., Ayllon, J., &
404
Fernandez-Sesma, A. (2010). Inhibition of the type I interferon response in human
405
dendritic cells by dengue virus infection requires a catalytically active NS2B3 complex.
406
Journal of Virology, 84(19), 9760-9774. doi:10.1128/JVI.01051-10; 10.1128/JVI.01051-10
407
Rodriguez-Madoz, J. R., Bernal-Rubio, D., Kaminski, D., Boyd, K., & Fernandez-Sesma, A.
408
(2010). Dengue virus inhibits the production of type I interferon in primary human
409
dendritic cells. Journal of Virology, 84(9), 4845-4850. doi:10.1128/JVI.02514-09;
410
10.1128/JVI.02514-09
cells.
PLoS
Scientifica,
Pathogens,
6(1),
2013,
164203.
e1000718.
15
doi:10.1155/2013/164203;
doi:10.1371/journal.ppat.1000718;
411
Rothman, A. L. (2011). Immunity to dengue virus: A tale of original antigenic sin and
412
tropical
413
doi:10.1038/nri3014; 10.1038/nri3014
414
Sanchez, V., Hessler, C., DeMonfort, A., Lang, J., & Guy, B. (2006). Comparison by flow
415
cytometry of immune changes induced in human monocyte-derived dendritic cells upon
416
infection with dengue 2 live-attenuated vaccine or 16681 parental strain. FEMS
417
Immunology
418
695X.2005.00008.x
419
Schmid, M. A., & Harris, E. (2014). Monocyte Recruitment to the Dermis and Differentiation
420
to Dendritic Cells Increases the Targets for Dengue Virus Replication. PLoS Pathogens,
421
10(12), e1004541. http://doi.org/10.1371/journal.ppat.1004541
422
Schuurhuis, D. H., van Montfoort, N., Ioan-Facsinay, A., Jiawan, R., Camps, M., Nouta, J.,
423
Ossendorp, F. (2006). Immune complex-loaded dendritic cells are superior to soluble
424
immune complexes as antitumor vaccine. Journal of Immunology (Baltimore, Md.: 1950),
425
176(8), 4573-4580. ]
426
Simmons, C. P., Chau, T. N., Thuy, T. T., Tuan, N. M., Hoang, D. M., Thien, N. T., Farrar, J.
427
(2007). Maternal antibody and viral factors in the pathogenesis of dengue virus in infants.
428
The Journal of Infectious Diseases, 196(3), 416-424. doi:10.1086/519170
429
Soundravally, R., Hoti, S. L., Patil, S. A., Cleetus, C. C., Zachariah, B., Kadhiravan, T., Kumar, B.
430
A. (2013). Association between proinflammatory cytokines and lipid peroxidation in
431
patients with severe dengue disease around defervescence. International Journal of
432
Infectious Diseases : IJID : Official Publication of the International Society for Infectious
433
Diseases, doi:10.1016/j.ijid.2013.09.022; 10.1016/j.ijid.2013.09.022
434
Sun, P., Fernandez, S., Marovich, M. A., Palmer, D. R., Celluzzi, C. M., Boonnak, K., Burgess, T.
435
H. (2009). Functional characterization of ex vivo blood myeloid and plasmacytoid dendritic
436
cells
437
doi:10.1016/j.virol.2008.10.022; 10.1016/j.virol.2008.10.022
cytokine
after
and
storms.
Medical
infection
Nature
Reviews
Microbiology,
with
dengue
16
46(1),
virus.
Immunology,
113-123.
Virology,
11(8),
532-543.
doi:10.1111/j.1574-
383(2),
207-215.
438
Van der Schaar, H. M., Rust, M. J., Waarts, B.-L., van der Ende-Metselaar, H., Kuhn, R. J.,
439
Wilschut, J., Zhuang X., & Smit, J. M. (2007). Characterization of the Early Events in Dengue
440
Virus Cell Entry by Biochemical Assays and Single-Virus Tracking . Journal of Virology,
441
81(21), 12019–12028. http://doi.org/10.1128/JVI.00300-07
442
Van der Schaar, H. M., Rust, M. J., Chen, C., van der Ende-Metselaar, H., Wilschut, J., Zhuang,
443
X., & Smit, J. M. (2008). Dissecting the Cell Entry Pathway of Dengue Virus by Single-Particle
444
Tracking
445
http://doi.org/10.1371/journal.ppat.1000244
446
Van der Schaar, H. M., Wilschut, J. C., & Smit, J. M. (2009). Role of antibodies in controlling
447
dengue
448
doi:10.1016/j.imbio.2008.11.008; 10.1016/j.imbio.2008.11.008
449
Vogelpoel, L.T.C. et al. (2014). Fc gamma receptor-TLR-cross-talk elicits pro-inflammatory
450
cytokine production by human M2 macrophages.
451
10.1038/ncomms6444
452
WHO. (1997). Dengue hemorrhagic fever: Diagnosis, treatment, prevention andcontrol.
453
world health organization
454
Wu, S. J., Grouard-Vogel, G., Sun, W., Mascola, J. R., Brachtel, E., Putvatana, R., Frankel, S. S.
455
(2000). Human skin langerhans cells are targets of dengue virus infection. Nature Medicine,
456
6(7), 816-820. doi:10.1038/77553
457
Yao, Y., Li, W., Kaplan, M. H., & Chang, C. H. (2005). Interleukin (IL)-4 inhibits IL-10 to
458
promote IL-12 production by dendritic cells. The Journal of Experimental Medicine,
459
201(12), 1899-1903. doi:10.1084/jem.20050324
460
Zybert, I. A., van der Ende-Metselaar, H., Wilschut, J., & Smit, J. M. (2008). Functional
461
importance of dengue virus maturation: Infectious properties of immature virions. The
462
Journal of General Virology, 89(Pt 12), 3047-3051. doi:10.1099/vir.0.2008/002535-0;
463
10.1099/vir.0.2008/002535-0
in
Living
virus
Cells.
infection.
PLoS
Pathogens,
Immunobiology,
464
17
4(12),
214(7),
e1000244.
613-629.
Nat. Commun. 5:5444 doi:
465
Figure legends
466
Figure 1. DENV infection of immDCs in the absence or presence of DENV-immune
467
serum. ImmDCs were infected with DENV-2 at MOI 0.1, 1 or 10. Cells and cell supernatants
468
were harvested at 24 hours post-infection (hpi). (A) The percentage of DENV-positive cells
469
was determined using mAb 3H5. (B) Quantitative analysis of the infectious properties of
470
DENV in immDCs. White bars: number of infectious particles (Log10 PFU/ml); black bars:
471
number of genome-equivalent copies (log10 GEc/mL). Results are representative of 10
472
independent experiments with 3 donors. (C) ImmDCs were infected either in the absence of
473
DENV immune serum, under neutralizing (here represented by 10-3) or non-neutralizing
474
conditions (here represented by 10-8). (D) Quantitative analysis of the infectious properties
475
of DENV in immDCs under various infection conditions. White bars: number of infectious
476
particles (Log10 PFU/ml); black bars: number of genome-equivalent copies (log10 GEc/mL).
477
Results are representative of ≥10 experiments with three donors. Bars represent the
478
standard error of the mean (SEM), n.d. denotes not detected.
479
480
Figure 2. Differences in DC phenotype following direct DENV infection and infection
481
in the presence of antibodies. ImmDCs were infected with C6/36-derived virus or UV-
482
inactivated (UVi) DENV at MOI 1 in the absence or presence of DENV-immune serum. The
483
mean fluorescence intensity (MFI) of the expression of co-stimulatory markers CD83 and
484
CD86 and the major histocompatibility complex molecule HLA-DR was assessed by flow
485
cytometry at 24 hpi. The bars represent fold-changes between different infection
486
conditions and their matched mocks obtained from at least 3 independent experiments ±
487
SEM. Statistical analysis was done by use of Mann-Whitney U-test (*P< 0.05 ** P < 0.01).
488
Stars above the bars indicate differences when compared to mock-infected cells.
489
490
Figure 3. DENV-immune complexes stimulate cytokine secretion by DCs in a FcγRIIa-
491
dependent manner. DENV infection (MOI 1) was performed as described in the legend to
492
Figure 2. (A) IL-6, TNF-α, IL-10 and IL-4 production was measured by a Cytokine Bead
493
Assay (CBA, BD Biosciences). (B & C) The effect of anti-FcγRIIa antibody pre-treatment on
494
the levels of pro-inflammatory cytokines produced by immDCs following (B) neutralizing
18
495
and (C) non-neutralizing infection conditions. The data shown are representative of at least
496
3 independent experiments ±SD. Statistical analysis was done by use of Mann-Whitney U-
497
test (* P< 0.05; ** P < 0.01, *** P <0.001). Stars above the bars indicate differences when
498
compared to mock-infected cells.
499
500
Suplementary data
501
Supplementary Figure 1. Phenotypic analysis of immDCs and matDCs. (A) Phenotypic
502
analysis of monocyte-derived immDCs. A total of 1.5x105 cells were counted. Histograms
503
show the fluorescence intensity of typical dendritic cell markers: Lin-1, HLA-DR and CD11c.
504
Dashed line: isotype control; continuous line: specific antibody. (B) MFI of the co-
505
stimulatory markers CD40, CD83, and CD86 of immDCs and matDCs.
506
507
Supplementary Figure 2. Effect of the dengue- immune sera dilution on DENV-2
508
infection of immDCs. Infection was performed as described in the legend to Figure 1. Cell-
509
free-supernatants of 24hpi were analyzed by plaque assay. The data shown are
510
representative of two independent experiments with three donors.
511
512
513
Supplementary Figure 3. Effect of FcγRIIa blockage on DENV infection in immDCs in
514
the presence of neutralizing and non-neutralizing sera DENV infection was performed
515
as described in the legend to Figure 3. Cell-free-supernatants of 24hpi were analyzed by
516
plaque assay. The data shown are representative of three independent experiments.
517
19