You are on page 1of 9

The Journal of Immunology

Expression of Cytokine, Chemokine, and Adhesion Molecules


during Endothelial Cell Activation Induced by Antibodies
against Dengue Virus Nonstructural Protein 11

Chiou-Feng Lin,* Shu-Chen Chiu,* Yu-Ling Hsiao,* Shu-Wen Wan,* Huan-Yao Lei,*
Ai-Li Shiau,* Hsiao-Sheng Liu,* Trai-Ming Yeh,† Shun-Hua Chen,* Ching-Chuan Liu,‡ and
Yee-Shin Lin2*
Vascular dysfunction is a hallmark associated with disease onset in dengue hemorrhagic fever and dengue shock syndrome. In
addition to direct viral damage, immune responses to dengue virus (DV) infection may also underlie the pathogenesis of disease.
We have proposed a mechanism of molecular mimicry in which Abs directed against DV nonstructural protein 1 (NS1) cross-react
with endothelial cells and induce damage. In this study, we demonstrated the inflammatory endothelial cell activation induced by
anti-DV NS1 via the transcription factor NF-␬B-regulated pathway. Protein phosphorylation and NF-␬B activation were observed
after anti-DV NS1 stimulation in a human microvascular endothelial cell line-1. The cytokine and chemokine production, including
IL-6, IL-8, and MCP-1, but not RANTES, in endothelial cells increased after treatment with anti-DV NS1 Abs. The expression of
IL-6, IL-8, and MCP-1 was blocked by the preabsorption of anti-DV NS1 with DV NS1 or by the inhibition of NF-␬B activation.
Furthermore, the increases in both ICAM-1 expression and the ability of human PBMC to adhere to endothelial cells were also
observed, and these effects were inhibited by pretreatment with anti-ICAM-1 or anti-MCP-1 Abs. Therefore, in addition to
endothelial cell apoptosis, as previously reported, inflammatory activation occurs in endothelial cells after stimulation by anti-DV
NS1 Abs. These results suggest the involvement of anti-DV NS1 Abs in the vasculopathy of DV infection. The Journal of
Immunology, 2005, 174: 395– 403.

P atients with dengue virus (DV)3 infection present a wide and vasculopathy (7, 35–37). Plasma leakage, also called the hem-
range of diseases from mild dengue fever to life-threaten- orrhagic syndrome, occurs once vessel endothelium is disrupted
ing dengue hemorrhagic fever and dengue shock syn- and is followed by the loss of its barrier function. The pathogenesis
drome (DHF/DSS) (1, 2). There is no vaccine against DHF/DSS of endothelial dysfunction resulting in vascular leakage remains
because its pathogenic mechanisms are still not fully understood unclear, however. In addition to direct viral damage, such as the
(3– 6). In dengue pathogenesis, the involvement of viral and im- induction of apoptosis in endothelial cells as observed in vitro (27),
mune responses has been hypothesized (3–9). Virus variation and complement activation and cytokine and chemokine production
virus load involving Ab-dependent enhancement of infection have induced by DV infection appear to be involved in the induction of
been suggested to be responsible for the progression and severity endothelial cell damage (26, 27, 38, 39). DV can also indirectly
of dengue disease (10 –17). Cell activation, impaired proliferation, modulate endothelial cell function through Ab-enhanced infection
cytokine and chemokine production, and apoptosis are caused after of DV in peripheral blood monocytes (33). Furthermore, the in-
DV infection by a direct (18 –32) or indirect route (33, 34). It is volvement of anti-endothelial cell autoantibodies in DV pathogen-
likely that different mechanisms are involved in the pathogenesis esis has been proposed (40 – 42). Endothelial cell apoptosis caused
of DV infection. by autoantibodies may be related to the transient leakage syndrome
DHF/DSS represents a pathological complication caused by in dengue vasculopathy.
multiple symptoms, including thrombocytopenia, coagulopathy, Inflammatory immune responses facilitate the progression of
disease severity in DHF/DSS (7–9). Lymphocytes, monocytes and
Departments of *Microbiology and Immunology, †Medical Technology, and ‡Pedi- macrophages, mast cells, endothelial cells, liver cells, and den-
atrics, National Cheng Kung University Medical College, Tainan, Taiwan dritic cells are the targets of DV, and these cells could produce
Received for publication March 22, 2004. Accepted for publication October 19, 2004. cytokines or chemokines, or both, after DV infection (20 –22, 24 –
The costs of publication of this article were defrayed in part by the payment of page 27, 33, 38, 39, 43, 44). Increased levels of cytokines, including
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
IL-2, IL-6, IL-8, IL-10, IL-13, IL-18, IFN-␥, TNF-␣, and MCP-1,
1 have been observed in patients with DV infections (8, 27, 45–52).
This work was supported by Grant NSC 91-3112-B006-002 from the National Sci-
ence Council, Taiwan. The roles of various cytokines and chemokines in the pathogenesis
2
Address correspondence and reprint requests to Dr. Yee-Shin Lin, Department of of DHF/DSS need to be clarified, especially on endothelial cell
Microbiology and Immunology, National Cheng Kung University Medical College, 1 activation, which is related to vasculopathy.
University Road, Tainan 701, Taiwan. E-mail address: yslin1@mail.ncku.edu.tw
Previous studies in our laboratory showed the pathogenic role of
3
Abbreviations used in this paper: DV, dengue virus; AEC, 3-amino-9-ethylcarba- Abs against nonstructural protein 1 (NS1) present in dengue pa-
zole; AECA, anti-endothelial cell Ab; DHF, dengue hemorrhagic fever; DSS, dengue
shock syndrome; EGM, endothelial cell growth medium; HMEC-1, human micro- tients or generated in mice (40, 41). Anti-DV NS1 Abs cross-
vascular endothelial cell line-1; IKK, I-K␤ kinase; JEV, Japanese encephalitis virus; reacted with endothelial cells and induced apoptosis via an NO-

L-NAME, N -nitro-L-arginine methyl ester; NS1, nonstructural protein 1; PDTC, pyr-
rolidine dithiocarbamate; zVAD-fmk, benzyloxycarbonylvalylalanylaspartic acid flu- mediated pathway (40, 42). Pathogenic anti-DV NS1 may
oromethyl ketone. contribute to the progression of DHF/DSS (41). In the present

Copyright © 2005 by The American Association of Immunologists, Inc. 0022-1767/05/$02.00


396 ANTI-DENGUE NS1 Ab-INDUCED ENDOTHELIAL CELL ACTIVATION

study, we demonstrated protein tyrosine phosphorylation and (pH 7.5), 10 mM EDTA, 0.02% NaN3, and a protease inhibitor mixture
NF-␬B activation after endothelial cells had been bound by anti- (Boehringer Mannheim). After being freeze thawed once, cell lysates were
centrifuged at 12,000 rpm for 20 min at 4°C. The supernatants were col-
DV NS1 Abs. We also demonstrated that the increased levels of
lected and boiled in sample buffer for 5 min. Following SDS-PAGE, pro-
cytokine and chemokine expression were regulated by NF-␬B, and teins were transferred to polyvinylidene difluoride membrane (Millipore),
that anti-DV NS1 stimulation caused adhesion molecule expres- blocked overnight at 4°C in PBS-T (PBS plus 0.1% Tween 20) containing
sion and PBMC adhesion to endothelial cells. 5% skim milk, and probed with Abs against phosphotyrosine at 4°C over-
night. After being washed with PBS-T, blots were incubated with a 1/5000
dilution of HRP-conjugated goat anti-rabbit IgG for 1 h at 4°C. The protein
Materials and Methods bands were developed with a 3-amino-9-ethylcarbazole (AEC) substrate kit
Mice (Zymed Laboratories).
For immunostaining followed by flow cytometric analysis, cells were
BALB/cByJ breeder mice were obtained from The Jackson Laboratory and washed in PBS, fixed with 1% paraformaldehyde in PBS at room temper-
maintained on standard laboratory food and water ad libitum in our medical ature for 10 min, and permeabilized with 70% ethanol. After being washed
college laboratory animal center. Their 8-wk-old progeny were used for the with PBS three times, cells were incubated with FITC-conjugated mAbs
generation of Abs in the present study. against phosphotyrosine for 1 h at 4°C and analyzed by flow cytometry
(FACSCalibur; BD Biosciences) with excitation set at 488 nm.
Abs and reagents
Both purified and FITC-conjugated mAbs against phosphotyrosine (clone Western blotting and EMSA for detection of NF-␬B activation
PY20) were purchased from Oncogene Research Products. Rabbit Abs Nuclear translocation of NF-␬B was detected by Western blotting. Har-
specific for NF-␬B p65 were purchased from Chemicon International. vested cells were lysed with buffer A (10 mM HEPES (pH 7.8), 5 mM
Anti-human IL-6, IL-8, MCP-1, RANTES, and ICAM-1 mAbs were from MgCl2, 10 mM KCl, 1 mM ZnCl2, 0.2 mM EDTA, 1 mM Na3VO4, 10 mM
BD Pharmingen. Neutralizing anti-human MCP-1 and ICAM-1 mAbs were NaF, 0.5 mM DTT, and 0.5 mM PMSF), incubated on ice for 10 min, and
also from BD Pharmingen, and mouse mAb to ␤-actin was from Sigma- centrifuged at 12,000 rpm for 20 min at 4°C. The supernatants were col-
Aldrich. FITC- or HRP-conjugated goat anti-mouse or anti-rabbit IgG, lected as the cytosolic lysates, and the nuclear extracts were obtained from
NF-␬B inhibitor pyrrolidine dithiocarbamate (PDTC), protein synthesis in- pellets lysed in buffer B (20 mM HEPES (pH 7.8), 5 mM MgCl2, 300 mM
hibitor cycloheximide, caspase inhibitor benzyloxycarbonylvalylalany- NaCl, 1 mM ZnCl2, 0.2 mM EDTA, 25% glycerol, 1 mM Na3VO4, 10 mM
laspartic acid fluoromethyl ketone (zVAD-fmk), and NO synthase inhibitor NaF, 0.5 mM DTT, and 0.5 mM PMSF) and incubated for 15 min on ice
N␻-nitro-L-arginine methyl ester (L-NAME) were obtained from with occasional mixing. Following SDS-PAGE, proteins were transferred
Sigma-Aldrich. to polyvinylidene difluoride membrane, blocked overnight at 4°C in PBS-T
Polyclonal Abs against DV or Japanese encephalitis virus (JEV) NS1 containing 5% skim milk, and probed with Abs against NF-␬B p65 subunit
were obtained from BALB/c mice immunized i.p. with purified recombi- at 4°C overnight. After being washed with PBS-T, blots were incubated
nant DV-2 (New Guinea C strain) or JEV (NT109 strain) NS1 proteins, as with a 1/5000 dilution of HRP-conjugated goat anti-rabbit IgG for 1 h at
described previously (40). The IgG fractions from hyperimmunized mouse 4°C. The protein bands were developed with an AEC substrate kit.
sera were purified with a protein G-Sepharose affinity chromatography col- For EMSA, nuclear extracts were collected, as described above, and
umn (Amersham Biosciences) and recovered with HCl-glycine. The reac- separated by 6% acrylamide gel. The DIG Gel Shift kit (Roche Diagnos-
tivity of purified IgG against NS1 was confirmed by SDS-PAGE and West- tics) was used according to the manufacturer’s instructions. The detection
ern blot. The control IgG was eluted from a protein G column loaded with probe specific for NF-␬B consisted of a digoxigenin-labeled double-strand
normal mouse sera. The endotoxin concentration of each of these prepa- oligonucleotide (5⬘-AGTTGAGGGGACTTTCCCAGGC-3⬘). Specificity
rations was ⬍0.03 EU/␮g, as determined by a Limulus amebocyte lysate of protein-DNA complexes was detected by immunoreactivity with sheep
assay (Associates of Cape Cod). anti-digoxigenin conjugated with alkaline phosphatase and developed us-
ing chemiluminescent substrate disodium 3-(4-methoxyspiro{1,2-dioxet-
Patient sera ane-3-,2⬘-(5⬘-chloro)tricyclo[3.3.1.13.7]decan]-4-yl) phenyl phosphate
Dengue patient sera were obtained from N. Hung (Department of Dengue (Roche). The generated chemiluminescent signals were recorded on x-ray
Hemorrhagic Fever, Children’s Hospital No. 1. Seventeen serum samples film by autoradiography.
were collected from patients with DHF disease severity grades I-III. Di-
agnosis of DHF was based on the clinical criteria established by the World Immunohistochemistry and flow cytometry for detection of
Health Organization. Sera from five healthy volunteers were used as the cytokine, chemokine, and adhesion molecule
normal controls. Monolayers of HMEC-1 cells were cultured on sterile glass slides, fol-
lowed by treatment with mouse anti-DV NS1, anti-JEV NS1, or control
Cell and virus culture
IgG for various time intervals. For flow cytometric analysis, cells were
Human microvascular endothelial cell line-1 (HMEC-1) was passed in cul- detached using 1000 U/ml trypsin and 0.5 mM EDTA. Cells were fixed and
ture plates containing endothelial cell growth medium (EGM; Cambrex) permeabilized with Cytofix/Cytoperm kit (BD Pharmingen). For immuno-
composed of 2% FBS, 1 ␮g/ml hydrocortisone, 10 ng/ml epidermal growth histochemical staining, fixed cells were incubated with 0.3% H2O2 in PBS
factor, and antibiotics (40). Cells were detached using 1000 U/ml trypsin for 5 min to quench endogenous peroxidase activity and then washed again
and 0.5 mM EDTA. Only those cultures from three to five passages with with PBS. Abs specific for IL-6, IL-8, RANTES, MCP-1, and ICAM-1
a viability of ⬎95% by eosin-Y staining were used for experiments. Baby were added to cells and incubated for 1 h at 4°C. After being washed with
hamster kidney cell line and C6/36 cells were cultured in DMEM medium PBS, cells were incubated with HRP- or FITC-conjugated anti-primary Ab
containing 10% FBS and antibiotics. PBMC were isolated from normal for 1 h at 4°C. After being washed with PBS, cells were developed using
volunteer blood using Ficoll-Paque isolation (Amersham Biosciences), ac- the AEC substrate kit (Zymed Laboratories), counterstained with hema-
cording to the standard procedures, and were used for adhesion assay. toxylin (Sigma-Aldrich), and viewed with light microscopy or analyzed by
Dengue-2 virus (PL046, Taiwan isolated) was maintained in the C6/36 flow cytometry with excitation set at 488 nm.
cells. Briefly, monolayers of C6/36 were incubated with DV at a multi-
plicity of infection of 0.01 and incubated at 26°C in 5% CO2 for 5 days. ELISA
The cultured medium was harvested, and cell debris was removed by cen- The concentrations of cytokines and chemokines, including IL-6, IL-8, and
trifugation at 900 ⫻ g for 10 min. After further centrifugation at 16,000 ⫻ MCP-1, in culture supernatants and DHF patient sera were determined
g for 10 min, the virus supernatant was collected and stored at ⫺70°C until using ELISA kits (R&D Systems). The manufacturer’s instructions were
use. Virus titer was determined by plaque assay using the BHK-21 cell line, followed, and the concentrations were determined by spectrophotometry at
as described previously (26). 450 nm (Molecular Devices).
Western blotting and flow cytometry for detection of protein RT-PCR
tyrosine phosphorylation
The expression of MCP-1 and RANTES mRNA was determined using
HMEC-1 cells underwent starvation in serum-free EGM for 1 h before the RT-PCR analysis. Total cellular RNA from endothelial cells was extracted
experiment. Next, the cells were treated with mouse anti-DV NS1, anti- using TRIzol reagent (Invitrogen Life Technologies), according to the
JEV NS1, or control IgG for various time intervals. Cells were then har- manufacturer’s instructions. The concentration of RNA was quantified by
vested and lysed with a buffer containing 1% Triton X-100, 50 mM Tris spectrophotometry at 260 nm (U-2000; Hitachi). The cDNA was prepared
The Journal of Immunology 397

by reverse transcription, as previously described (40), and PCR was per-


formed using a PCR controller (GeneAmp PCR System 2400;
PerkinElmer). PCR was conducted in 50 ␮l of the reaction mixture (1.5
mM MgCl2 and 0.2 mM each of dATP, dGTP, dCTP, and dTTP) contain-
ing primers at 1.5 ␮M each, 0.2 ␮g/ml RNase A (Sigma-Aldrich), and 1 U
of TaqDNA polymerase (Promega), as follows: 94°C for 1 min; 30 thermal
cycles at 95°C for 1 min, 55°C for 1 min, and 72°C for 2 min; and a final
cycle at 72°C for 5 min. The oligonucleotide primers for human MCP-1
(sense, 5⬘-CAAACTGAAGCTCGCACTCTCGCC-3⬘ and antisense, 5⬘-AT
TCTTGGGTTGTGGAGTGAGTGTTCA-3⬘), RANTES (sense, 5⬘-TGCC
TCCCCATATTCCTCGG-3⬘ and antisense, 5⬘-TCATGTTTGCCAGTAA
GC-3⬘), and ␤-actin (sense, 5⬘-AGCGGGAAATCGTGCGTG-3⬘ and anti-
sense, 5⬘-CAGGGTACATGGTGGTGGTGCC-3⬘) were used according to
previously published sequences (27, 40). DV primer set (sense, 5⬘-GATATG
GGTTATTGGATAGA-3⬘ and antisense, 5⬘-CTGATTTCCATCCCGTA-3⬘)
was used as an infected control (26). The PCR products were analyzed by
1.5% agarose gel electrophoresis, stained with ethidium bromide, and viewed
with UV light.
Transient transfection
In experiments using dominant-negative mutant, HMEC-1 cells were co-
transfected with the dominant-negative IKK-␤ (the kind gift of C. Chen,
Department of Pharmacology, National Taiwan University) or the empty
vector pcDNA3.1 (the kind gift of M. Lai, Department of Biochemistry,
National Cheng Kung University). Briefly, HMEC-1 cells were grown to FIGURE 1. Tyrosine phosphorylation of cellular proteins after treat-
50% confluent in six-well plate, then transfected with the dominant-nega- ment with anti-DV NS1 Abs in endothelial cells. After serum-free starva-
tive IKK-␤ (0.5 ␮g) or the empty vector (1.0 ␮g) using Lipofectamine tion for 1 h, HMEC-1 cells were treated with 5 ␮g of anti-DV NS1, anti-
2000 reagent (Invitrogen Life Technologies). After 24 h of transfection, the
JEV NS1, or control IgG, or replaced with EGM (used as positive controls
cells were used for experiments.
for cell signaling) composed of 2% FBS for 30 min. The tyrosine-phos-
Adhesion assay phorylated protein levels were analyzed by Western blotting (A, upper
panel) and quantified by measuring OD (A, lower panel). The cells were
HMEC-1 cells (5 ⫻ 104 cells/well) were plated into eight-well glass cham-
stained with FITC-conjugated phosphorylated tyrosine-specific IgG and
ber slides (Nalge Nunc International). When monolayers were confluent,
the cells were stimulated with anti-DV NS1, anti-JEV NS1, or control IgG analyzed using flow cytometry. A histogram and the percentage of positive
in serum-free culture medium. After 24 h of incubation, the cells were cells are shown (B, upper panel). Time-kinetic changes of protein tyrosine
washed once with medium and incubated for 2 h at 37°C with isolated phosphorylation induced by control IgG (F), anti-DV NS1 (Œ), and EGM
PBMC (1 ⫻ 105 cells/well) in a total volume of 250 ␮l/well. At the end of (f) are shown, respectively. The untreated cells (E) were used as negative
the incubation period, the nonadherent cells were removed by washing controls (B, lower panel).
twice with 0.1% BSA in PBS. Adherent cells were stained with Liu’s stain
(TONYAR Biotech) and viewed with light microscopy. The adherent cells cells after they had been treated with DHF patient sera (data not
were counted on three consecutive microscopic fields (53, 54).
shown).
Statistical analysis We next investigated the involvement of NF-␬B in the anti-DV
Comparisons between various treatments were performed by Student’s t NS1-triggered signaling pathways. After Ab treatment for 3 or
test with SigmaPlot version 4.0 for Windows (Cytel Software). Values 24 h, endothelial cell extracts were separated for cytoplasmic and
were considered statistically significant at p ⬍ 0.05. nuclear fractions, and then followed by immunoblotting for NF-
␬B. The expression of NF-␬B decreased in the cytoplasmic frac-
Results tion and increased in the nuclear fraction in cells with anti-DV
Protein tyrosine phosphorylation and NF-␬B activation induced NS1 treatment compared with those with anti-JEV NS1 or control
by anti-DV NS1 Abs in human endothelial cells IgG treatment (Fig. 2A). Activation of NF-␬B as indicated by its
In a previous study (40), we showed that anti-DV NS1 cross-re- translocation from cytoplasm to nucleus was also observed by im-
acted with endothelial cells. To assess the signal transduction in- munohistochemical staining (data not shown). The DNA-binding
duced by anti-DV NS1 in endothelial cells, we investigated protein ability of NF-␬B was determined by EMSA, which showed pos-
phosphorylation and NF-␬B activation in the present study. We itive signals in the anti-DV NS1-treated cells compared with the
used Western blotting to assay the levels of protein tyrosine phos- untreated, anti-JEV NS1, or control IgG group (Fig. 2B). The
phorylation from Ab-treated HMEC-1 cells (Fig. 1A, upper panel). DNA-binding ability of NF-␬B was inhibited when anti-DV NS1
We also determined the relative ODs of bands (Fig. 1A, lower was preabsorbed with rDV NS1 proteins. The specificity of the
panel). Results showed tyrosine phosphorylation of proteins in cross-reactivity of anti-NS1 Abs was thus confirmed. To further
HMEC-1 after binding with anti-DV NS1, but not with anti-JEV demonstrate whether NF-␬B activation is requisite for its DNA-
NS1 or control IgG. EGM-treated cells were used as the positive binding ability mediated by anti-DV NS1, the effect of NF-␬B
control for protein phosphorylation induced by growth factors. inhibitor PDTC was examined. Results showed that the addition of
Protein tyrosine phosphorylation was further analyzed by flow cy- PDTC to cell cultures caused an inhibition of the DNA-binding
tometry as presented by the percentages of fluorescence-positive activity of NF-␬B in anti-DV NS1-treated cells (data not shown).
cells (Fig. 1B, upper panel). The time-kinetic changes of phos-
phorylated cells after Ab stimulation are shown in Fig. 1B (lower Production of cytokines and chemokines in endothelial cells
panel). Results indicated that protein tyrosine phosphorylation after anti-DV NS1 stimulation
reached its highest level 30 min after anti-DV NS1 treatment. The Immune activation of endothelial cells along with the induction of
specificity of Ab stimulation was demonstrated by the blockage of proinflammatory responses underlie dengue vascular disorders (7,
protein tyrosine phosphorylation when anti-DV NS1 Abs were 9). We therefore examined whether anti-DV NS1 Abs could in-
pretreated with rDV NS1 proteins (data not shown). Tyrosine duce inflammatory factors in endothelial cells. Results from im-
phosphorylation of cellular proteins was also observed in HMEC-1 munohistochemical staining revealed the expression of IL-6, IL-8,
398 ANTI-DENGUE NS1 Ab-INDUCED ENDOTHELIAL CELL ACTIVATION

FIGURE 2. NF-␬B activation induced by anti-DV NS1 Abs in endo-


thelial cells. A, HMEC-1 cells were untreated or treated with 5 ␮g of
anti-DV NS1, anti-JEV NS1, or control IgG for 3 or 24 h, and the expres- FIGURE 3. Production of IL-6, IL-8, and MCP-1, but not RANTES, by
sion of NF-␬B in cytoplasm and nucleus was determined using Western endothelial cells after treatment with anti-DV NS1 Abs. HMEC-1 cells
blotting. ␤-actin was used as an internal control of total cell lysate before were treated with 5 ␮g of anti-DV NS1, anti-JEV NS1, or control IgG for
cytoplasmic and nuclear extraction. B, The DNA-binding ability of NF-␬B 24 h. The expression of IL-6, IL-8, MCP-1, and RANTES was detected
in HMEC-1 cells after treatment with anti-DV NS1 (5 or 25 ␮g), anti-JEV with immunohistochemical staining using specific Abs, as described in
NS1 (25 ␮g), or control IgG (25 ␮g) for 3 h was detected by EMSA using Materials and Methods (A, the positive cells are indicated by arrowheads),
NF-␬B-specific probes. Cells treated with anti-DV NS1 preabsorbed with and with flow cytometry analysis (B). For flow cytometric analysis, three
25 ␮g of rDV NS1 protein were tested for the specificity of Ab stimulation. individual cultures were performed, and the averages of the percentages of
NF-␬B p65 fragment was used as a positive control. Free probe was used positive cells are shown (mean ⫾ SD). Cells treated with anti-DV NS1
as a loading control. preabsorbed with 5 ␮g of recombinant DV or JEV NS1 protein were tested
for the specificity of Ab stimulation.

Requirement for NF-␬B in anti-DV NS1-induced cytokine and


and MCP-1 by HMEC-1 cells after they had been treated with chemokine production
anti-DV NS1 for 24 h (Fig. 3A). The expression of RANTES, Anti-DV NS1 induced NF-␬B activation (Fig. 2). We further ex-
however, could not be detected in anti-DV NS1-treated cells. Ex- amined the involvement of NF-␬B modulation on the expression
pression of IL-6, IL-8, and MCP-1 was further quantified using of IL-6, IL-8, and MCP-1. After treatment with anti-DV NS1 in
both flow cytometric analysis (Fig. 3B) and ELISA (Table I). HMEC-1 cultures for 24 h, the protein levels of IL-6, IL-8, and
Treatment with anti-JEV NS1 Abs showed only basal levels of MCP-1 increased. NF-␬B inhibitor PDTC, but not NO synthase
IL-6, IL-8, MCP-1, and RANTES, similar to those with control inhibitor L-NAME or caspase inhibitor zVAD-fmk, was able to
IgG. The ELISA (Table I) and immunohistochemical staining inhibit the anti-DV NS1-mediated elevations of IL-6, IL-8, and
(data not shown) showed IL-6, IL-8, and MCP-1 production after MCP-1 expression (Fig. 5, left). Further confirming the require-
anti-DV NS1 treatment for as short as 3– 6 h. The specificity of the ment for NF-␬B activation, HMEC-1 cells transfected with dom-
cross-reactivity of anti-NS1 Abs was further confirmed by the in- inant-negative IKK-␤ showed a blockage in IL-6, IL-8, and
hibitory effect of preabsorption of anti-DV NS1 with recombinant MCP-1 production compared with the vector control (Fig. 5,
DV NS1, but not JEV NS1 proteins (Fig. 3B). Further studies right). These results revealed that NF-␬B in anti-DV NS1-induced
showed that the mRNA expression of MCP-1, but not RANTES, endothelial cell activation up-regulates cytokine and chemokine
was elevated after anti-DV NS1 stimulation in HMEC-1 cells, as expression.
detected via RT-PCR (Fig. 4). By contrast, cells infected with DV
showed increased mRNA expression in RANTES, but not in MCP-1 facilitates the expression of ICAM-1 induced by anti-DV
MCP-1. The ELISA quantification demonstrated high levels of NS1 in endothelial cells
MCP-1 in DHF patient sera at different disease grades compared The activation of endothelial cells involves not only the induction
with healthy controls (Table II), similar to results in a study of DSS of cytokines and chemokines; our data showed that the expression
patients (27). of adhesion molecules was also augmented. After anti-DV NS1
The Journal of Immunology 399

Table I. Production of IL-6, IL-8, and MCP-1 by endothelial cells after


treatment with anti-DV NS1 Abs for various time periodsa

Ab
Time Dose
(h) (␮g) Control IgG Anti-JEV NS1 Anti-DV NS1

IL-6 (pg/cell)
0 0.12 ⫾ 0.02
3 5 0.30 ⫾ 0.11 0.14 ⫾ 0.10 0.22 ⫾ 0.10
6 5 0.31 ⫾ 0.10 0.22 ⫾ 0.08 1.05 ⫾ 0.04ⴱ
12 5 0.32 ⫾ 0.15 0.35 ⫾ 0.15 4.03 ⫾ 0.55ⴱⴱ
24 5 0.42 ⫾ 0.14 0.39 ⫾ 0.11 5.75 ⫾ 1.20ⴱⴱ
24 25 0.45 ⫾ 0.11 0.41 ⫾ 0.12 4.15 ⫾ 1.07ⴱⴱ
IL-8 (pg/cell)
0 0.07 ⫾ 0.01
3 5 0.10 ⫾ 0.01 0.12 ⫾ 0.05 0.52 ⫾ 0.12ⴱ FIGURE 4. The expression of MCP-1, but not RANTES mRNA by
6 5 0.12 ⫾ 0.03 0.20 ⫾ 0.01 2.75 ⫾ 1.07ⴱ endothelial cells after anti-DV NS1 Ab treatment. Total cellular mRNA
12 5 0.23 ⫾ 0.09 0.22 ⫾ 0.11 5.33 ⫾ 1.34ⴱⴱ was extracted at 24 h from HMEC-1 cells after treatment with 5 ␮g of
24 5 0.21 ⫾ 0.11 0.31 ⫾ 0.15 7.65 ⫾ 2.52ⴱⴱ anti-DV NS1, anti-JEV NS1, or control IgG or with 100 multiplicity of
24 25 0.29 ⫾ 0.08 0.61 ⫾ 0.20 5.97 ⫾ 1.30ⴱⴱ infection of dengue-2 virus, and detected using RT-PCR. ␤-actin was used
MCP-1 (fg/cell) as an internal control.
0 0.29 ⫾ 0.09
3 5 0.30 ⫾ 0.01 0.29 ⫾ 0.11 0.52 ⫾ 0.01
6 5 0.43 ⫾ 0.01 0.41 ⫾ 0.08 1.75 ⫾ 0.09ⴱ
12 5 0.46 ⫾ 0.07 0.58 ⫾ 0.05 4.33 ⫾ 0.50ⴱⴱ
24 5 0.45 ⫾ 0.03 0.71 ⫾ 0.01 4.65 ⫾ 0.23ⴱⴱ expressed on endothelial cells was involved in this binding effect.
24 25 0.54 ⫾ 0.14 0.87 ⫾ 0.14 4.03 ⫾ 1.07ⴱⴱ We also tested the potential regulatory role of MCP-1 on ICAM-
a
ELISA kits were used for the assay. Three individual cultures were performed in 1-mediated PBMC adherence to endothelial cells. A reduction in
each group, and the averages of values are expressed as mean ⫾ SD; ⴱ, p ⬍ 0.05 vs
control IgG; ⴱⴱ, p ⬍ 0.01 vs control IgG.
PBMC adherence was demonstrated when endothelial cells were
pretreated with anti-MCP-1 Abs (Fig. 7). These results thus sug-
gested that the ICAM-1-mediated ability of PBMC to adhere to
endothelial cells was at least partially regulated by MCP-1. To
stimulation, an increase in the expression of ICAM-1 was detected
further investigate whether the up-regulation of cell adherence was
with immunohistochemical staining (Fig. 6A). The increase in
dependent on new protein synthesis, HMEC-1 cells were pre-
ICAM-1 expression was confirmed using flow cytometric analysis
treated with protein synthesis inhibitor cycloheximide. Results
(see Fig. 6B for time kinetics and dose-response data). To further
showed that cycloheximide treatment inhibited PBMC adhesion,
investigate the regulation of ICAM-1 expression by NF-␬B in anti-
suggesting a requirement for protein synthesis of ICAM-1 or
DV NS1-treated cells, PDTC was used. Results showed a reduc-
MCP-1 to increase PBMC adherence to HMEC-1 cells (Fig. 7B).
tion of ICAM-1 expression once cells were pretreated with PDTC
(data not shown). MCP-1 elevates ICAM-1 expression (55–57).
We therefore investigated the relationship between MCP-1 and Discussion
ICAM-1 expression after anti-DV NS1 stimulation. Pretreatment Dengue disease is an emerging public health problem, and an in-
with MCP-1-neutralizing Abs reduced the expression of ICAM-1, creasing number of cases are being reported (5, 6, 58). Vascular
suggesting that the ICAM-1 expressed was regulated, at least in leakage, liver abnormality, and hemorrhagic diathesis are the life-
part, by MCP-1 in anti-DV NS1-treated endothelial cells. Similar threatening complications that occur in dengue patients with DHF/
results were observed using both immunohistochemical staining DSS. Their pathogenic mechanisms, however, are not well under-
(Fig. 6A) and flow cytometric analysis (Fig. 6B). Further studies stood. We have proposed a mechanism of molecular mimicry in
using anti-DV NS1 F(ab⬘)2 confirmed that the activation of endo- which Abs directed against DV NS1 would cross-react with en-
thelial cells, as detected by ICAM-1 expression, was not via an dothelial cells and cause damage. Our studies showed anti-
Fc-mediated signaling pathway (data not shown). endothelial cell Abs (AECA) in dengue patient sera (41). More-
over, endothelial cell damage induced by anti-DV NS1 by the
Regulation of ICAM-1 and MCP-1 on the adherence of PBMC production of NO may play a role in the disruption of vessel en-
to anti-DV NS1-treated endothelial cells dothelium and contribute to the AECA-induced pathogenesis of
vasculopathy (40, 42). When taken together with previous findings
The ability of immune cells to adhere to DV-infected endothelial
that anti-DV NS1 Abs also cross-reacted with platelets (59) and
cells was investigated next. Based on the findings shown in Fig. 6,
anti-DV NS1 Ab-induced expression of ICAM-1 was down-regu-
lated by anti-MCP-1. To explore the effects of ICAM-1 up-regu-
lation on endothelial cell and immune cell interaction, the adhesion
Table II. MCP-1 levels in DHF patient seraa
of PBMC to anti-DV NS1-treated HMEC-1 cells was assessed.
Higher levels of PBMC adhering to endothelial cells could be ob-
Group MCP-1 (pg/ml)
served in cells treated with anti-DV NS1 for 24 h, compared with
those with anti-JEV NS1 or control IgG (Fig. 7A). The increase in Controls (n ⫽ 5) 72.9 ⫾ 18.9
PBMC adherence was quantified by counting the numbers of ad- DHF patients (n ⫽ 17) 2695.6 ⫾ 444.0ⴱⴱⴱ
Grade I (n ⫽ 1) 3009.5 ⫾ 351.4ⴱⴱⴱ
herent cells under a microscope (see Fig. 7B for time kinetics and
Grade II (n ⫽ 13) 2610.8 ⫾ 383.2ⴱⴱⴱ
dose-response data). We further investigated whether the increase Grade III (n ⫽ 3) 2958.5 ⫾ 618.6ⴱⴱⴱ
in ICAM-1 expression was related to the ability of PBMC to ad- a
ELISA kits were used for the assay. Three individual cultures were performed
here to endothelial cells. Pretreatment with ICAM-1-neutralizing for each person, and the averages of values are expressed as mean ⫾ SD; ⴱⴱⴱ, p ⬍
Abs inhibited the adherence of PBMC, suggesting that ICAM-1 0.001 vs controls.
400 ANTI-DENGUE NS1 Ab-INDUCED ENDOTHELIAL CELL ACTIVATION

FIGURE 5. The involvement of NF-␬B in the expression of IL-6, IL-8,


and MCP-1 in endothelial cells after anti-DV NS1 stimulation. In the pres- FIGURE 6. MCP-1 regulates the expression of ICAM-1 in endothelial
ence or absence of PDTC (100 ␮M), L-NAME (10 ␮M), or zVAD-fmk (10 cells after anti-DV NS1 Ab stimulation. A, HMEC-1 cells were treated with
␮M) for 1-h pretreatment or transfected with dominant-negative IKK-␤ 5 ␮g of anti-DV NS1, anti-JEV NS1, or control IgG for 24 h. Cells were
(⌬IKK-␤) or empty vector, HMEC-1 cells were treated with 5 ␮g of anti- labeled with mouse anti-human ICAM-1 Abs, followed by HRP- or FITC-
DV NS1, anti-JEV NS1, or control IgG for 24 h. The expression of IL-6, conjugated goat anti-mouse IgG. Cells treated with anti-DV NS1 in the
IL-8, and MCP-1 was detected using ELISA, as described in Materials and presence of 5 ␮g of neutralizing anti-MCP-1 were also tested. The ICAM-1
Methods. Three individual cultures were performed in ELISA, and the expression was detected using immunohistochemical staining. B, HMEC-1
averages are shown (mean ⫾ SD). cells were treated with 1, 5, or 25 ␮g of anti-DV NS1, anti-JEV NS1, or
control IgG for 6, 12, or 24 h. The ICAM-1 expression was detected using
flow cytometry analysis. Three individual cultures were performed, and the
percentages of positive cells are shown (mean ⫾ SD).
that transient thrombocytopenia in DV-infected mice was associ-
ated with the generation of anti-platelet Abs (60), our results sug-
gest that the onset of autoimmune responses in DV infection may related to dengue pathogenesis remain to be investigated. Re-
have implications in DHF immunopathogenesis (9). Several other cently, DV NS1 has been reported to express in a GPI-linked form
reports have also shown the autoimmune responses in DV infec- that results in signal transduction, as evidenced by tyrosine phos-
tion (61, 62). phorylation of cellular proteins (64). In this study, we showed
In addition to cell apoptosis (40), we showed in this study that tyrosine phosphorylation and NF-␬B activation after anti-DV NS1
immune activation in endothelial cells also occurred after anti-DV stimulation in endothelial cells without DV infection. The role of
NS1 stimulation. Protein tyrosine phosphorylation and NF-␬B ac- protein tyrosine phosphorylation in the NF-␬B-mediated immune
tivation in HMEC-1 cells were observed. There was also an in- activation requires further clarification. To explore the signaling
crease in the expression of cytokines and chemokines, including pathways mediated by anti-DV NS1, the surface molecules of en-
IL-6, IL-8, and MCP-1, but not RANTES. The involvement of dothelial cells, GPI linked or not, recognized by these autoanti-
NF-␬B in the expression of IL-6, IL-8, and MCP-1 was also dem- bodies need to be identified. Studies on potential autoantigens us-
onstrated. Up-regulation of ICAM-1 expression and ICAM-1-me- ing anti-DV NS1 Abs and DHF/DSS patient sera are in progress.
diated adherence of PBMC to endothelial cells were observed after Various autoimmune diseases associated with vascular dysfunc-
anti-DV NS1 stimulation. The expression of ICAM-1 was modu- tion may be related to the generation of AECA (42). Vasculopathy
lated, at least in part, by MCP-1 and by NF-␬B. Taken together, in autoimmune diseases involves both cytotoxicity and inflamma-
these facts indicate that endothelial cell activation that involves tion facilitated by AECA via a direct or an indirect route. The
NF-␬B-regulated cytokine and chemokine production and adhe- elevated expression of inflammatory mediators, including cyto-
sion molecule expression, as well as adherence of immune cells to kines (IL-1 and IL-6), chemokines (IL-8 and MCP-1), and adhe-
endothelial cells, may represent the inflammatory mechanisms me- sion molecules (ICAM-1, VCAM-1, and E-selectin), has been
diated by anti-DV NS1 Abs. The signaling pathways induced by shown in AECA-stimulated endothelial cells (65– 68). However,
anti-DV NS1 in endothelial cells leading to immune activation and the molecular mechanisms of AECA-mediated endothelial cell ac-
inflammation are shown in Fig. 8. Although our previous study tivation remain poorly defined. Studies on the mechanisms of en-
showed an NO-mediated mechanism of apoptosis induced by anti- dothelial activation have shown that degradation of I-␬B followed
DV NS1, immune activation is NF-␬B dependent, but NO and by NF-␬B activation might contribute to the induction of proin-
apoptosis independent. flammatory responses (67). Activation of NF-␬B was also detected
Several in vitro studies have shown that endothelial cells are the when anti-DV NS1 cross-reacted with uninfected endothelial cells.
targets of DV and that induction of proinflammatory responses and A recent study provided a possible molecular mechanism of AE-
apoptotic cell death are directly caused by DV (26, 27, 63). We CA’s regulation of ICAM-1 expression through ERK1/2 activation
have shown somewhat similar effects mediated by anti-DV NS1 in Behcet’s disease (69).
Abs, specifically that endothelial cells underwent apoptosis (40 – In dengue pathogenesis, elevated levels of inflammatory IL-6
42) and, in the present study, induced proinflammatory effects. The and IL-8 were correlated with clinical presentation of DHF (46 –
combined effects of DV and anti-NS1 Abs on endothelial cells 48, 52). Also, endothelial cells infected with DV induced IL-6 and
The Journal of Immunology 401

FIGURE 8. The signaling pathways mediated by anti-DV NS1 Abs that


lead to endothelial cell activation. Signal transduction in endothelial cells
after anti-NS1 stimulation involves protein tyrosine phosphorylation and
NF-␬B activation. NF-␬B is required for the expression of cytokines (e.g.,
IL-6) and chemokines (e.g., IL-8 and MCP-1). Expression of ICAM-1
induced by anti-NS1 is, at least in part, regulated by NF-␬B and by MCP-1.
Furthermore, the ability of PBMC to adhere to endothelial cells is mediated
by ICAM-1. The endothelial cell activation induced by anti-DV NS1 Abs
leading to inflammatory responses may be related to DHF pathogenesis.
FIGURE 7. The ability of PBMC to adhere to anti-DV NS1-treated en-
dothelial cells is blocked by pretreatment with anti-ICAM-1 and anti-
MCP-1. A, HMEC-1 cells were cultured in eight-well glass chamber slides
and treated with 5 ␮g of anti-DV NS1, anti-JEV NS1, or control IgG for indirect damage in endothelial cells. Activation of endothelial cells
24 h. In anti-DV NS1-treated groups, cells cultured with 5 ␮g of neutral- via DV-infected peripheral blood monocytes has been reported
izing anti-ICAM-1 or anti-MCP-1 were also tested. Cells were washed with (33). Endothelial cell activation and apoptosis might play a role in
PBS, and then cocultured with fresh human PBMC for 2 h. Cell adhesion the fulminant, but short-lived vascular leakage of DHF pathogen-
was observed using Liu’s stain. B, Time kinetics and dose response of esis (27). The clinical features indicate that vascular endothelia in
anti-DV NS1 on the PBMC adhesion to endothelial cells in the presence or serosal tissues are preferentially affected by dengue pathogenetic
absence of 100 ␮M cycloheximide or 5 ␮g of anti-ICAM-1 or anti-MCP-1.
mechanisms, resulting in ascites and pleural effusion for most of
The numbers of adherent cells were quantified in each tested culture.
the plasma leakage in dengue shock syndrome (70). Tissue culture
fluid from DV-infected monocytes has activated endothelial cells
IL-8 production (26, 27). In the present study, we showed that (33). Serum inflammatory cytokines may relate to the development
anti-DV NS1 promoted proinflammatory endothelial cell activa- of plasma leakage and disease severity (8, 52). Given all of these
tion causing increases in IL-6 and IL-8 protein expression and data, endothelial vascular dysfunction in dengue pathogenesis in-
secretion. Furthermore, anti-DV NS1 induces the generation of volves multiple factors, including the dengue virus itself and anti-
MCP-1 in endothelial cells. A previous study showed MCP-1 in DV NS1, direct and indirect effects on endothelial cells, and im-
DSS patient plasma and pleural fluid, but not in DV-infected en- mune activation and apoptosis.
dothelial cells (27). We found, in the present study, MCP-1 pro-
duction by endothelial cells after anti-DV NS1 stimulation; MCP-1 Acknowledgment
in DHF patient sera was also confirmed. In addition to the two We acknowledge the editorial assistance of Bill Franke.
chemokines IL-8 and MCP-1, RANTES was also checked in this
study and was found not induced by anti-DV NS1 stimulation, References
although it was produced in endothelial cells after DV infection 1. Henchal, E. A., and J. R. Putnak. 1990. The dengue viruses. Clin. Microbiol. Rev.
3:376.
(27). MCP-1-mediated elevation of ICAM-1 expression and facil- 2. Gubler, D. J. 1998. Dengue and dengue hemorrhagic fever. Clin. Microbiol. Rev.
itation of leukocyte transmigration have been suggested (53–55). 11:480.
Results in this study showed that MCP-1 up-regulated ICAM-1 3. Bhakdi, S., and M. D. Kazatchkine. 1990. Pathogenesis of dengue: an alternative
hypothesis. Southeast Asian J. Trop. Med. Pub. Hlth. 21:652.
expression in anti-DV NS1-treated HMEC-1 cells. Endothelial cell 4. Bielefeldt-Ohmann, H. 1997. Pathogenesis of dengue virus diseases: missing
activation followed by elevated expression of ICAM-1 may con- pieces in the jigsaw. Trends Microbiol. 5:409.
tribute to the adherence of immune cells to endothelial cells in the 5. Pancharoen, C., U. Thisyakorn, and C. Thisyakorn. 2001. Dengue infection. J. In-
fect. Dis. Antimicrob. Agents 18:115.
inflammatory responses associated with dengue disease pathogen- 6. Halstead, S. B. 2002. Dengue. Curr. Opin. Infect. Dis. 15:471.
esis. The involvement of other cytokines, chemokines, and adhe- 7. Rothman, A. L., and F. A. Ennis. 1999. Immunopathogenesis of dengue hemor-
rhagic fever. Virology 257:1.
sion molecules is currently under investigation. 8. Green, S., D. W. Vaughn, S. Kalayanarooj, S. Nimmannitya, S. Suntayakorn,
In the present study, we showed the inflammatory initiator role A. Nisalak, R. Lew, B. L. Innis, I. Kurane, A. L. Rothman, and F. A. Ennis. 1999.
of anti-DV NS1 resulting in endothelial cell activation. In addition Early immune activation in acute dengue illness is related to development of
plasma leakage and disease severity. J. Infect. Dis. 179:755.
to the increased expression of cytokines and chemokines, a pos- 9. Lei, H. Y., T. M. Yeh, H. S. Liu, Y. S. Lin, S. H. Chen, and C. C. Liu. 2001.
sible chemotactic effect on PBMC adherence may contribute to the Immunopathogenesis of dengue virus infection. J. Biomed. Sci. 8:377.
402 ANTI-DENGUE NS1 Ab-INDUCED ENDOTHELIAL CELL ACTIVATION

10. Halstead, S. B., C. N. Venkateshan, M. K. Gentry, and L. K. Larsen. 1984. 38. Bosch, I., K. Xhaja, L. Estevez, G. Raines, H. Melichar, R. V. Warke,
Heterogeneity of infection enhancement of dengue 2 strains by monoclonal an- M. V. Fournier, F. A. Ennis, and A. L. Rothman. 2002. Increased production of
tibodies. J. Immunol. 132:1529. interleukin-8 in primary human monocytes and in human epithelial and endothe-
11. Littaua, R., I. Kurane, and F. A. Ennis. 1990. Human IgG Fc receptor II mediates lial cell lines after dengue virus challenge. J. Virol. 76:5588.
antibody-dependent enhancement of dengue virus infection. J. Immunol. 39. Warke, R. V., K. Xhaja, K. J. Martin, M. F. Fournier, S. K. Shaw, N. Brizuela,
144:3183. N. De Bosch, D. Lapointe, F. A. Ennis, A. L. Rothman, and I. Bosch. 2003.
12. Mady, B. J., D. V. Erbe, I. Kurane, M. W. Fanger, and F. A. Ennis. 1991. Dengue virus induces novel changes in gene expression of human umbilical vein
Antibody-dependent enhancement of dengue virus infection mediated by bispe- endothelial cells. J. Virol. 77:11822.
cific antibodies against cell surface molecules other than Fc␥ receptors. J. Im- 40. Lin, C. F., H. Y. Lei, A. L. Shiau, H. S. Liu, T. M. Yeh, S. H. Chen, C. C. Liu,
munol. 147:3139. S. C. Chiu, and Y. S. Lin. 2002. Endothelial cell apoptosis induced by antibodies
13. Morens, D. M. 1994. Antibody-dependent enhancement of infection and the against dengue virus nonstructural protein 1 via production of nitric oxide. J. Im-
pathogenesis of viral disease. Clin. Infect. Dis. 19:500. munol. 169:657.
14. Rico-Hesse, R., L. M. Harrison, R. A. Salas, D. Tovar, A. Nisalak, C. Ramos, 41. Lin, C. F., H. Y. Lei, A. L. Shiau, C. C. Liu, H. S. Liu, T. M. Yeh, S. H. Chen,
J. Boshell, M. T. R. de Mesa, R. M. R. Nogueira, and A. Travassos da Rosa. 1997. and Y. S. Lin. 2003. Antibodies from dengue patient sera cross-react with en-
Origins of dengue type 2 viruses associated with increased pathogenicity in the dothelial cells and induce damage. J. Med. Virol. 69:82.
Americas. Virology 230:244. 42. Lin, Y. S., C. F. Lin, C. C. Liu, H. S. Liu, T. M. Yeh, S. H. Chen, and H. Y. Lei.
15. Leitmeyer, K. C., D. W. Vaughn, D. M. Watts, R. Salas, I. Villalobos de Chacon, 2004. Antibody-mediated endothelial cell damage via nitric oxide. Curr. Pharm.
C. Ramos, and R. Rico-Hesse. 1999. Dengue virus structural differences that Design 10:213.
correlate with pathogenesis. J. Virol. 73:4738. 43. Lin, Y. L., C. C. Liu, J. I. Chuang, H. Y. Lei, T. M. Yeh, Y. S. Lin, Y. H. Huang,
16. Diamond, M. S., D. Edgil, T. G. Roberts, B. Lu, and E. Harris. 2000. Infection and H. S. Liu. 2000. Involvement of oxidative stress, NF-IL-6, and RANTES
of human cells by dengue virus is modulated by different cell types and viral expression in dengue-2-virus-infected human liver cells. Virology 276:114.
strains. J. Virol. 74:7814.
44. King, C. A., R. Anderson, and J. S. Marshall. 2002. Dengue virus selectively
17. Vaughn, D. W., S. Green, S. Kalayanarooj, B. L. Innis, S. Nimmannitya,
induces human mast cell chemokine production. J. Virol. 76:8408.
S. Suntayakorn, T. P. Endy, B. Raengsakulrach, A. L. Rothman, F. A. Ennis, and
A. Nisalak. 2000. Dengue viremia titer, antibody response pattern, and virus 45. Kurane, I., B. L. Innis, S. Nimmannitya, A. Nisalak, A. Meager, J. Janus, and
serotype correlate with disease severity. J. Infect. Dis. 181:2. F. A. Ennis. 1991. Activation of T lymphocytes in dengue virus infections: high
18. Kurane, I., B. L. Innis, A. Nisalak, C. Hoke, S. Nimmannitya, A. Meager, and levels of soluble interleukin 2 receptor, soluble CD4, soluble CD8, interleukin 2,
F. A. Ennis. 1989. Human T cell responses to dengue virus antigens: proliferative and interferon-␥ in sera of children with dengue. J. Clin. Invest. 88:1473.
responses and interferon ␥ production. J. Clin. Invest. 83:506. 46. Hober, D., L. Poli, B. Roblin, P. Gestas, E. Chungue, G. Granic, P. Imbert,
19. Mathew, A., I. Kurane, S. Green, D. W. Vaughn, S. Kalayanarooj, J. Pecarere, R. Vergez-Pascal, P. Wattre, and M. Maniez-Montreuil. 1993. Serum
S. Suntayakorn, F. A. Ennis, and A. L. Rothman. 1999. Impaired T cell prolif- levels of tumor necrosis factor-␣ (TNF-␣), interleukin-6 (IL-6), and interleu-
eration in acute dengue infection. J. Immunol. 162:5609. kin-1␤ (IL-1␤) in dengue-infected patients. Am. J. Trop. Med. Hyg. 48:324.
20. Lin, Y. W., K. J. Wang, H. Y. Lei, Y. S. Lin, T. M. Yeh, H. S. Liu, C. C. Liu, 47. Raghupathy, R., U. C. Chaturvedi, H. Al-Sayer, E. A. Elbishbishi, R. Agarwal,
and S. H. Chen. 2002. Virus replication and cytokine production in dengue virus- R. Nagar, S. Kapoor, A. Misra, A. Mathur, H. Nusrat, et al. 1998. Elevated levels
infected human B lymphocytes. J. Virol. 76:12242. of IL-8 in dengue hemorrhagic fever. J. Med. Virol. 56:280.
21. King, C. A., J. S. Marshall, H. Alshurafa, and R. Anderson. 2000. Release of 48. Juffrie, M., G. M. van Der Meer, C. E. Hack, K. Haasnoot, K. Sutaryo,
vasoactive cytokines by antibody-enhanced dengue virus infection of a human A. J. Veerman, and L. G. Thijs. 2000. Inflammatory mediators in dengue virus
mast cell/basophil line. J. Virol. 74:7146. infection in children: interleukin-8 and its relationship to neutrophil degranula-
22. Marianneau, P., A. M. Steffan, C. Royer, M. T. Drouet, D. Jaeck, A. Kirn, and tion. Infect. Immun. 68:702.
V. Deubel. 1999. Infection of primary cultures of human Kupffer cells by Dengue 49. Mustafa, A. S., E. A. Elibishbishi, R. Agarwal, and U. C. Chaturvedi. 2001.
virus: no viral progeny synthesis, but cytokine production is evident. J. Virol. Elevated levels of interleukin-13 and IL-18 in patients with dengue hemorrhagic
73:5201. fever. FEMS Immunol. Med. Microbiol. 30:229.
23. Wu, S. J., G. Grouard-Vogel, W. Sun, J. R. Mascola, E. Brachtel, R. Putvatana, 50. Gagnon, S. J., M. Mori, I. Kurane, S. Green, D. W. Vaughn, S. Kalayanarooj,
M. K. Louder, L. Filgueira, M. A. Marovich, H. K. Wong, et al. 2000. Human S. Suntayakorn, F. A. Ennis, and A. L. Rothman. 2002. Cytokine gene expression
skin Langerhans cells are targets of dengue virus infection. Nat. Med. 6:816. and protein production in peripheral blood mononuclear cells of children with
24. Ho, L. J., J. J. Wang, M. F. Shaio, C. L. Kao, D. M. Chang, S. W. Han, and acute dengue virus infections. J. Med. Virol. 67:41.
J. H. Lai. 2001. Infection of human dendritic cells by dengue virus causes cell 51. Suharti, C., E. C. van Gorp, W. M. Dolmans, T. E. Setiati, C. E. Hack,
maturation and cytokine production. J. Immunol. 166:1499. R. Djokomoeljanto, and J. W. van der Meer. 2003. Cytokine patterns during
25. Libraty, D. H., S. Pichyangkul, C. Ajariyakhajorn, T. P. Endy, and F. A. Ennis. dengue shock syndrome. Eur. Cytokine Network 14:172.
2001. Human dendritic cells are activated by dengue virus infection: enhance- 52. Hung, N. T., H. Y. Lei, N. T. Lan, Y. S. Lin, K. J. Huang, B. Lien le, C. F. Lin,
ment by ␥ interferon and implications for disease pathogenesis. J. Virol. 75:3501. T. M. Yeh, Q. Ha do, V. T. Huong, et al. 2004. Dengue hemorrhagic fever in
26. Huang, Y. H., H. Y. Lei, H. S. Liu, Y. S. Lin, C. C. Liu, and T. M. Yeh. 2000. infants: a study of clinical and cytokine profiles. J. Infect. Dis. 189:221.
Dengue virus infects human endothelial cells and induces IL-6 and IL-8 produc- 53. Frostegard, J., J. Nilsson, A. Haegerstrand, A. Hamsten, H. Wigzell, and
tion. Am. J. Trop. Med. Hyg. 63:71. M. Gidlund. 1990. Oxidized low density lipoprotein induces differentiation and
27. Avirutnan, P., P. Malasit, B. Seliger, S. Bhakdi, and M. Husmann. 1998. Dengue adhesion of human monocytes and the monocytic cell line U937. Proc. Natl.
virus infection of human endothelial cells leads to chemokine production, com- Acad. Sci. USA 87:904.
plement activation, and apoptosis. J. Immunol. 161:6338. 54. Adams, M. R., W. Jessup, D. Hailstones, and D. S. Celermajer. 1997. L-Arginine
28. Despres, P., M. Flamand, P.-E. Ceccaldi, and V. Deubel. 1996. Human isolates reduces human monocyte adhesion to vascular endothelium and endothelial ex-
of dengue type 1 virus induce apoptosis in mouse neuroblastoma cells. J. Virol. pression of cell adhesion molecules. Circulation 95:662.
70:4090.
55. Weiss, J. M., S. A. Downie, W. D. Lyman, and J. W. Berman. 1998. Astrocyte-
29. Marianneau, P., A. Cardona, L. Edelman, V. Deubel, and P. Despres. 1997. derived monocyte-chemoattractant protein-1 directs the transmigration of leuko-
Dengue virus replication in human hepatoma cells activates NF-␬B which in turn cytes across a model of the human blood-brain barrier. J. Immunol. 161:6896.
induces apoptotic cell death. J. Virol. 71:3244.
56. Tekstra, J., H. Beekhuizen, J. S. Van De Gevel, I. J. Van Benten, C. W. Tuk, and
30. Despres, P., M. P. Frenkiel, P. E. Ceccaldi, C. Duarte dos Santos, and V. Deubel.
R. H. Beelen. 1999. Infection of human endothelial cells with Staphylococcus
1998. Apoptosis in the mouse central nervous system in response to infection
aureus induces the production of monocyte chemotactic protein-1 (MCP-1) and
with mouse-neurovirulent dengue viruses. J. Virol. 72:823.
monocyte chemotaxis. Clin. Exp. Immunol. 117:489.
31. Marianneau, P., M. Flamand, V. Deubel, and P. Despres. 1998. Apoptotic cell
death in response to dengue virus infection: the pathogenesis of dengue haem- 57. Viedt, C., R. Dechend, J. Fei, G. M. Hansch, J. Kreuzer, and S. R. Orth. 2002.
orrhagic fever revisited. Clin. Diagn. Virol. 10:113. MCP-1 induces inflammatory activation of human tubular epithelial cells: in-
32. Jan, J. T., B. H. Chen, S. H. Ma, C. I. Liu, H. P. Tsai, H. C. Wu, S. Y. Jiang, volvement of the transcription factors, nuclear factor-␬B and activating protein-1.
K. D. Yang, and M. F. Shaio. 2000. Potential dengue virus-triggered apoptotic J. Am. Soc. Nephrol. 13:1534.
pathway in human neuroblastoma cells: arachidonic acid, superoxide anion, and 58. Monath, T. P. 1994. Dengue: the risk to developed and developing countries.
NF-␬B are sequentially involved. J. Virol. 74:8680. Proc. Natl. Acad. Sci. USA 91:2395.
33. Anderson, R., S. Wang, C. Osiowy, and A. C. Issekutz. 1997. Activation of 59. Lin, C. F., H. Y. Lei, C. C. Liu, H. S. Liu, T. M. Yeh, S. T. Wang, T. I. Yang,
endothelial cells via antibody-enhanced dengue virus infection of peripheral F. C. Sheu, C. F. Kuo, and Y. S. Lin. 2001. Generation of IgM anti-platelet
blood monocytes. J. Virol. 71:4226. autoantibody in dengue patients. J. Med. Virol. 63:143.
34. Gagnon, S. J., F. A. Ennis, and A. L. Rothman. 1999. Bystander target cell lysis 60. Huang, K. J., S. Y. J. Li, S. C. Chen, H. S. Liu, Y. S. Lin, T. M. Yeh, C. C. Liu,
and cytokine production by dengue virus-specific human CD4⫹ cytotoxic T- and H. Y. Lei. 2000. Manifestation of thrombocytopenia in dengue-2-virus-in-
lymphocyte clones. J. Virol. 73:3623. fected mice. J. Gen. Virol. 81:217.
35. Nimmannitya, S. 1987. Clinical spectrum and management of dengue haemor- 61. Falconar, A. K. I. 1997. The dengue virus nonstructural-1 protein (NS1) gener-
rhagic fever. Southeast Asian J. Trop. Med. Pub. Hlth. 18:392. ates antibodies to common epitopes on human blood clotting, integrin/adhesin
36. Bhamarapravati, N. 1989. Hemostatic defects in dengue hemorrhagic fever. Rev. proteins and binds to human endothelial cells: potential implications in haemor-
Infect. Dis. 11:S826. rhagic fever pathogenesis. Arch. Virol. 142:897.
37. Kalayanarooj, S., D. W. Vaughn, S. Nimmannitya, S. Green, S. Suntayakorn, 62. Oishi, K., S. Inoue, M. T. Cinco, E. M. Dimaano, M. T. Alera, J. A. Alfon,
N. Kunentrasai, W. Viramitrachai, S. Ratanachueke, S. Kiatpolpoj, B. L. Innis, et F. Abanes, D. J. Cruz, R. R. Matias, H. Matsuura, et al. 2003. Correlation be-
al. 1997. Early clinical and laboratory indictors of acute dengue illness. J. Infect. tween increased platelet-associated IgG and thrombocytopenia in secondary den-
Dis. 176:313. gue virus infections. J. Med. Virol. 71:259.
The Journal of Immunology 403

63. Andraws, B. S., A. N. Theofilopoulos, C. J. Peters, D. J. Loskutoff, W. E. Brandt, 67. Yazici, Z. A., E. Raschi, A. Patel, C. Testoni, M. O. Borghi, A. M. Graham,
and F. J. Dixon. 1978. Replication of dengue and junin viruses in cultured rabbit P. L. Meroni, and N. Lindsey. 2001. Human monoclonal anti-endothelial cell IgG
and human endothelial cells. Infect. Immun. 20:776. derived from a systemic lupus erythematosus patient binds and activates human
64. Jacobs, M. G., P. J. Robinson, C. Bletchly, J. M. Mackenzie, and P. R. Young. endothelium in vitro. Int. Immunol. 13:349.
2000. Dengue virus nonstructural protein 1 is expressed in a glycosyl-phospha- 68. Praprotnik, S., M. Blank, Y. Levy, S. Tavor, M. C. Boffa, B. Weksler, A. Eldor,
tidylinositol-linked form that is capable of signal transduction. FASEB J. and Y. Shoenfeld. 2001. Anti-endothelial cell antibodies from patients with
14:1603. thrombotic thrombocytopenic purpura specifically activate small vessel endothe-
65. Carvalho, D., C. O. S. Savage, C. M. Black, and J. D. Pearson. 1996. IgG anti- lial cells. Int. Immunol. 13:203.
endothelial cell autoantibodies from scleroderma patients induce leukocyte ad-
hesion to human vascular endothelial cells in vitro: induction of adhesion mol- 69. Lee, K. H., H. J. Cho, H. S. Kim, W. J. Lee, S. Lee, and D. Bang. 2002. Acti-
ecule expression and involvement of endothelial-derived cytokines. J. Clin. vation of extracellular signal regulated kinase 1/2 in human dermal microvascular
Invest. 97:111. endothelial cells stimulated by anti-endothelial cell antibodies in sera of patients
66. Del Papa, N., E. Raschi, G. Moroni, P. Panzeri, M. O. Borghi, C. Ponticelli, with Behçet’s disease. J. Dermatol. Sci. 30:63.
A. Tincani, G. Balestrieri, and P. L. Meroni. 1999. Anti-endothelial cell IgG fractions 70. Innis, B. L. 1995. Dengue and dengue hemorrhagic fever. In Kass Handbook of
from systemic lupus erythematosus patients bind to human endothelial cells and Infectious Diseases: Exotic Viral Diseases. J. S. Porterfield, ed. Chapman and
induce a pro-adhesive and a pro-inflammatory phenotype in vitro. Lupus 8:423. Hall, London, p. 103.

You might also like