Dengue vascular leakage is augmented by mast cell degranulation mediated by immunoglobulin Fcγ receptors

Ayesa Syenina, Cyril J Jagaraj, Siti A B Aman, Aishwarya Sridharan, Ashley L St John, Ayesa Syenina, Cyril J Jagaraj, Siti A B Aman, Aishwarya Sridharan, Ashley L St John

Abstract

Dengue virus (DENV) is the most significant human arboviral pathogen and causes ∼400 million infections in humans each year. In previous work, we observed that mast cells (MC) mediate vascular leakage during DENV infection in mice and that levels of MC activation are correlated with disease severity in human DENV patients (St John et al., 2013b). A major risk factor for developing severe dengue is secondary infection with a heterologous serotype. The dominant theory explaining increased severity during secondary DENV infection is that cross-reactive but non-neutralizing antibodies promote uptake of virus and allow enhanced replication. Here, we define another mechanism, dependent on FcγR-mediated enhanced degranulation responses by MCs. Antibody-dependent mast cell activation constitutes a novel mechanism to explain enhanced vascular leakage during secondary DENV infection.

Keywords: Fc receptor; antibodies; dengue virus; immunology; infectious disease; mast cell; microbiology; mouse; vascular leakage.

Conflict of interest statement

The authors declare that no competing interests exist.

Figures

Figure 1.. DENV-specific IgG enhances MC degranulation…
Figure 1.. DENV-specific IgG enhances MC degranulation and vascular leakage.
(A) MC-degranulation (cell line RBL-2H3) in response to DENV2 is enhanced by dose-dependent increases in the DENV E-specific antibody 4G2. Significance was determined by 2-way ANOVA, where * indicates a significant increase over control, while ** indicates a significant increase over DENV alone without 4G2 antibody. (B) In vivo degranulation of peritoneal MCs was enhanced by DENV compared to isotype controls and 4G2+DENV compared to both controls and DENV infection alone. MC degranulation was determined by flow cytometry to quantitate the heparin-containing granules as an assessment of granularity. Heparin staining of MCs (ckit+) was partitioned into high, mid, low, and negative populations (St John et al., 2011). DENV infection resulted in a significant loss of granulated MCs in the mid partition, and a significant increase in the percentage of cells in the heparin-negative partition (presumably degranulated). This was further accentuated by 4G2 treatment so that approximately 70% of ckit+ cells lacked heparin-staining granules in that group, while both mid and low heparin-staining groups were significantly reduced compared to DENV infection alone. Data were analyzed by 2-way ANOVA with Bonferroni's multiple comparison test to obtain p-values for between group comparisons: *p < 0.05, **p < 0.001, ***p < 0.0001. For each group, n = 3 animals and consistent data were obtained in a second independent trial. (C) Images of MCs were taken by confocal microscopy after cytospinning peritoneal lavage. Slides were stained for MC-heparin to reveal granules and DAPI for nuclei. Control MCs were densely granulated, but degranulation could be observed in samples from DENV-infected animals with and without 4G2 pre-sensitization. More granules appeared extracellular on slides in the 4G2+DENV group. (D) Vascular leakage was measured by obtaining hematocrit values 24 hr and 48 hr after infection with DENV, with and without pre-treatment with 4G2. Significance was determined by 1-way ANOVA with Bonferroni's post-test, where * indicates a significant increase over control, while ** indicates a significant increase over both control and DENV alone. (E) PFU equivalents were measured in mice by real time RT-PCR using RNA isolated from spleens, 24 hr after infection with DENV. Means do not differ significantly by Student's un-paired t-test (p = 0.2073, n = 5). DOI:http://dx.doi.org/10.7554/eLife.05291.002
Figure 2.. Antibody specificity governs IgG-enhanced MC…
Figure 2.. Antibody specificity governs IgG-enhanced MC degranulation and vascular leakage.
(A) MC degranulation in response to DENV1–4 is enhanced by cross-reactive antibody 4G2. (B) DENV2-specific antibody 3H5 enhances MC degranulation in response to DENV2 but not DENV1, 3 or 4. For A and B, data were analyzed by 2-way ANOVA with Bonferroni's post-test to determine p-values for group comparisons; * indicates a significant enhancement over control (p < 0.05), † indicates a significant increase over DENV treatment with isotype control (IC) for a given serotype (p < 0.05), and ns designates ‘not significant’. (C) Hematocrit values and (D) Evan's blue dye leakage into tissues were measured 24 hr after DENV1 infection of mice that had been pretreated with IC, 4G2, or 3H5. Hematocrit values and EBD detection values were elevated over baseline for all DENV1-infected mice; however, only the cross-reactive antibody 4G2 increased hematocrit values and Evans blue dye leakage over DENV1 infection with IC antibody. For C and D, significance was determined by 1-way ANOVA with Bonferroni's post-test; p < 0.05. (E) Images show the appearance of mouse livers after saline perfusion. For mice given control injections of saline, the perfusion results in complete flushing of the blood and EBD from the liver. In contrast, bruising consistent with vascular leakage could be observed with DENV1 alone infection, which appears enhanced in mice pre-treated with DENV1 cross-reactive antibody 4G2, but similar in animals pre-treated with non-binding antibody 3H5. DOI:http://dx.doi.org/10.7554/eLife.05291.003
Figure 3.. Antibody-enhanced vascular leakage is MC…
Figure 3.. Antibody-enhanced vascular leakage is MC dependent.
(A) WT and Sash mice were given IC antibody or 3H5 injections 24 hr prior to infection with DENV2. At 24 hr post-infection, blood was obtained for hematocrit analysis. WT mice given DENV2 after either IC or 3H5 treatment had significantly elevated hematocrit values compared to uninfected controls while hematocrit values of Sash mice were not significantly (ns) elevated over controls. EBD in the liver tissue was measured at 24 hr after infection and 30 min after injection with EBD. (B) Images showing the organs in the peritoneal cavity, taken prior to saline perfusion, from necropsy of representative WT and Sash mice with 3H5-antibody-enhanced DENV2 infection showing the organs in the peritoneal cavity. EBD can be visually observed in the gut of WT mice but not in Sash mice. (C) WT mice with and without 3H5 pretreatment before DENV2 infection were treated with the MC stabilizer, Cromolyn. Vascular leakage was measured by EBD perfusion 24 hr post-infection, followed by quantitation in liver tissue. For panels (AC), significance was determined by 1-way ANOVA with Bonferroni's post-test; * indicates a significant increase over control and ** indicates a significant increase over control as well as the DENV2-infected group that was not pre-treated with 3H5. For (C), δ indicates a significant reduction in vascular leakage after Cromolyn treatment, compared to the untreated group. DOI:http://dx.doi.org/10.7554/eLife.05291.004
Figure 4.. MC FcγRIII-induced degranulation promotes antibody-enhanced…
Figure 4.. MC FcγRIII-induced degranulation promotes antibody-enhanced vascular leakage.
WT and FcγRIII-KO BMMCs were pre-treated with antibodies 4G2 or 3H5, followed by exposure to (A) DENV2 or (B) DENV1 at an MOI of 1. Both 3H5 and 4G2 enhanced WT BMMC degranulation over DENV2 exposure alone in panel (A), but only 4G2 enhanced BMMC degranulation to DENV1 in panel (B). Both the 3H5- and 4G2-enhanced degranulation responses of BMMCs to DENV2, and the 4G2-enhanced degranulation responses to DENV1 were abrogated in FcγRIII-KO BMMCs. For (AB), 3H5 or 4G2 pretreatment did not change degranulation levels of FcγRIII-KO BMMCs over DENV exposure alone. All statistical comparisons in (AB) were performed by 1-way ANOVA with Bonferroni's post-test, *p < 0.05. (C) Degranulation is significantly induced by DENV and UV-DENV in mouse BMMCs. (D) Degranulation responses of WT BMMCs to both DENV and UV-DENV were increased significantly in the presence of antibody 4G2. (E) Antibody 4G2 did not enhance degranulation of FcγRIII-KO BMMCs in response to DENV or UV-DENV. In (DE), baseline control levels are denoted by gray lines. (F) Both WT and FcγRIII-KO BMMCs showed increased levels of DENV2 replication in the presence of 4G2, compared to control BMMCs but the levels were reduced in 4G2-treated FcγRIII-KO BMMCs compared to WT BMMCs treated with 4G2. All comparisons designated by * were significantly different (p < 0.05), as determined by ANOVA with Tukey's multiple comparison test. (G) This panel depicts the experimental design for BMMC adoptive transfer studies. MC-deficient Sash mice were reconstituted with BMMCs produced from either WT or FcγRIII-KO congenic controls. After maturation, all mice were pre-sensitized with 4G2 antibody prior to challenge with DENV2 (1 × 106 pfu) by i.p. injection (or saline control, uninfected). At 24 hr after injection with saline or DENV2, vascular leakage was measured by (H) hematocrit and (I) EBD detection in the liver after saline perfusion, shown as relative detection, normalized to uninfected controls. (GI) Mice reconstituted with FcγRIII-KO BMMCs showed reduced vascular leakage compared to mice reconstituted with WT BMMCs, demonstrating the role of MC-expressed FcγRIII in antibody-enhanced vascular leakage. Significance was determined in (H) by 1-way ANOVA with Bonferroni's post-test, and in (I) by un-paired Student's t-test; *p < 0.05. DOI:http://dx.doi.org/10.7554/eLife.05291.005
Figure 5.. Vascular leakage in response to…
Figure 5.. Vascular leakage in response to live and inactivated DENV after passive immunization.
(A) A schematic depicting the experimental design of serum-adoptive transfer experiments where post-immune serum from DENV2-infected or naïve mice was adoptively transferred into naïve hosts that were then challenged with 1 × 106 pfu of either DENV1 or DENV2. (B) No significant (ns) differences were detected in the levels of DENV1 infection at 24 hr between groups infected after passive transfer of either control serum or serum from DENV2 post immune (p.i.) mice. Significance was determined by Student's un-paired t-test (p = 0.2905, n = 5). DENV1 RNA levels are displayed normalized to actin and the levels in the control group of mice. (C) Hematocrit analysis showed that vascular leakage was enhanced by either a heterologous challenge with DENV1 (p = 0.0001) or a homologous challenge with DENV2 (p = 0.0005), compared to mice provided control serum from naïve animals. For reference, the dashed line represents the baseline hematocrit value for naïve WT mice. (D) Similarly, mice challenged with UV-inactivated DENV (UV-DENV) showed elevated hematocrit values that were similar to mice given live virus. (E) Serum from control (naïve) mice or DENV2 p.i. mice was adoptively transferred to recipient mice. Levels of serum MCPT1 were measured by ELISA 24 hr after challenge of recipient mice with either DENV2 or UV-DENV2. Passive immunization with DENV2 p.i. serum resulted in elevated concentration of the MC activation biomarker, MCPT1, after challenge. For each panel, p ≤ 0.05; n = 5. (F) Mice adoptively transferred serum from mice immunized against the antigen OVA or control serum were challenged with particulate OVA (pOVA). Antigen alone-treated mice did not have elevated hematocrit levels, but mice given OVA p.i. serum had significantly increased hematocrit levels at 24 hr. (G) The experimental design for adoptive transfer experiments where serum from DENV2 post immune animals was transferred into MC-deficient Sash mice, and Sash mice that had been reconstituted (Sash-R) with either FcγRIII-KO BMMCs or WT BMMCs, followed by a heterologous DENV1 challenge. (H) Mice reconstituted with WT MCs had significantly increased vascular leakage based on hematocrit analysis compared to Sash mice alone (p = 0.002), as did mice reconstituted with FcγRIII-KO BMMCs (p = 0.047); however, mice reconstituted with WT MCs showed a further increase over Sash-R mice repleted with FcγRIII-KO BMMCs (p = 0.003). The dashed line represents the baseline average hematocrit value for naive Sash mice. Statistical comparisons in data-containing panels were performed using Student's unpaired t-test. DOI:http://dx.doi.org/10.7554/eLife.05291.006
Author response image 1.
Author response image 1.
DENV burden in the spleen of MC-deficient and -repleted mice. Spleens were isolated 24h after infection with DENV and real time RTPCR was performed to quantify the viral burden in this DENV-target organ. In contrast to the significant differences in vascular leakage, viral burden did not differ significantly for the same number of mice (n=5; p=0.13).

References

    1. Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens. Nature Reviews Immunology. 2010;10:440–452. doi: 10.1038/nri2782.
    1. Anderson KB, Gibbons RV, Cummings DA, Nisalak A, Green S, Libraty DH, Jarman RG, Srikiatkhachorn A, Mammen MP, Darunee B, Yoon IK, Endy TP. A shorter time interval between first and second dengue infections is associated with protection from clinical illness in a school-based cohort in Thailand. The Journal of Infectious Diseases. 2014;209:360–368. doi: 10.1093/infdis/jit436.
    1. Chakravarti A, Kumaria R. Circulating levels of tumour necrosis factor-alpha & interferon-gamma in patients with dengue & dengue haemorrhagic fever during an outbreak. The Indian Journal of Medical Research. 2006;123:25–30.
    1. Daëron M, Bonnerot C, Latour S, Fridman WH. Murine recombinant Fc gamma RIII, but not Fc gamma RII, trigger serotonin release in rat basophilic leukemia cells. Journal of Immunology. 1992;149:1365–1373.
    1. Dejnirattisai W, Jumnainsong A, Onsirisakul N, Fitton P, Vasanawathana S, Limpitikul W, Puttikhunt C, Edwards C, Duangchinda T, Supasa S, Chawansuntati K, Malasit P, Mongkolsapaya J, Screaton G. Cross-reacting antibodies enhance dengue virus infection in humans. Science. 2010;328:745–748. doi: 10.1126/science.1185181.
    1. Goncalvez AP, Engle RE, St Claire M, Purcell RH, Lai CJ. Monoclonal antibody-mediated enhancement of dengue virus infection in vitro and in vivo and strategies for prevention. Proceedings of the National Academy of Sciences of USA. 2007;104:9422–9427. doi: 10.1073/pnas.0703498104.
    1. Guilarde AO, Turchi MD, Siqueira JB, Jr, Feres VC, Rocha B, Levi JE, Souza VA, Boas LS, Pannuti CS, Martelli CM. Dengue and dengue hemorrhagic fever among adults: clinical outcomes related to viremia, serotypes, and antibody response. The Journal of Infectious Diseases. 2008;197:817–824. doi: 10.1086/528805.
    1. Halstead SB. Pathogenesis of dengue: challenges to molecular biology. Science. 1988;239:476–481. doi: 10.1126/science.3277268.
    1. Halstead SB. Dengue. Lancet. 2007;370:1644–1652. doi: 10.1016/S0140-6736(07)61687-0.
    1. Halstead SB, Chow JS, Marchette NJ. Immunological enhancement of dengue virus replication. Nature. 1973;243:24–26.
    1. Halstead SB, Nimmannitya S, Cohen SN. Observations related to pathogenesis of dengue hemorrhagic fever. IV. Relation of disease severity to antibody response and virus recovered. The Yale Journal of Biology and Medicine. 1970;42:311–328.
    1. Halstead SB, O'Rourke EJ. Antibody-enhanced dengue virus infection in primate leukocytes. Nature. 1977a;265:739–741. doi: 10.1038/265739a0.
    1. Halstead SB, O'Rourke EJ. Dengue viruses and mononuclear phagocytes. I. Infection enhancement by non-neutralizing antibody. The Journal of Experimental Medicine. 1977b;146:201–217. doi: 10.1084/jem.146.1.201.
    1. Hazenbos WL, Gessner JE, Hofhuis FM, Kuipers H, Meyer D, Heijnen IA, Schmidt RE, Sandor M, Capel PJ, Daëron M, van de Winkel JG, Verbeek JS. Impaired IgG-dependent anaphylaxis and Arthus reaction in Fc gamma RIII (CD16) deficient mice. Immunity. 1996;5:181–188. doi: 10.1016/S1074-7613(00)80494-X.
    1. Henchal EA, Gentry MK, McCown JM, Brandt WE. Dengue virus-specific and flavivirus group determinants identified with monoclonal antibodies by indirect immunofluorescence. The American Journal of Tropical Medicine and Hygiene. 1982;31:830–836.
    1. Hober D, Poli L, Roblin B, Gestas P, Chungue E, Granic G, Imbert P, Pecarere JL, Vergez-Pascal R, Wattre P, Maniez-Montreuil M. Serum levels of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and interleukin-1 beta (IL-1 beta) in dengue-infected patients. The American Journal of Tropical Medicine and Hygiene. 1993;48:324–331.
    1. Jin C, Shelburne CP, Li G, Potts EN, Riebe KJ, Sempowski GD, Foster WM, Abraham SN. Particulate allergens potentiate allergic asthma in mice through sustained IgE-mediated mast cell activation. The Journal of Clinical Investigation. 2011;121:941–955. doi: 10.1172/JCI43584.
    1. King CA, Marshall JS, Alshurafa H, Anderson R. Release of vasoactive cytokines by antibody-enhanced dengue virus infection of a human mast cell/basophil line. Journal of Virology. 2000;74:7146–7150. doi: 10.1128/JVI.74.15.7146-7150.2000.
    1. Kliks SC, Nimmanitya S, Nisalak A, Burke DS. Evidence that maternal dengue antibodies are important in the development of dengue hemorrhagic fever in infants. The American Journal of Tropical Medicine and Hygiene. 1988;38:411–419.
    1. Koraka P, Murgue B, Deparis X, Setiati TE, Suharti C, van Gorp EC, Hack CE, Osterhaus AD, Groen J. Elevated levels of total and dengue virus-specific immunoglobulin E in patients with varying disease severity. Journal of Medical Virology. 2003;70:91–98. doi: 10.1002/jmv.10358.
    1. Kunder CA, St John AL, Abraham SN. Mast cell modulation of the vascular and lymphatic endothelium. Blood. 2011;118:5383–5393. doi: 10.1182/blood-2011-07-358432.
    1. Kunder CA, St John AL, Li G, Leong KW, Berwin B, Staats HF, Abraham SN. Mast cell-derived particles deliver peripheral signals to remote lymph nodes. The Journal of Experimental Medicine. 2009;206:2455–2467. doi: 10.1084/jem.20090805.
    1. Low JG, Ooi EE, Tolfvenstam T, Leo YS, Hibberd ML, Ng LC, Lai YL, Yap GS, Li CS, Vasudevan SG, Ong A. Early Dengue infection and outcome study (EDEN) - study design and preliminary findings. Annals of the Academy of Medicine, Singapore. 2006;35:783–789.
    1. Malbec O, Daëron M. The mast cell IgG receptors and their roles in tissue inflammation. Immunological Reviews. 2007;217:206–221. doi: 10.1111/j.1600-065X.2007.00510.x.
    1. Okayama Y, Kirshenbaum AS, Metcalfe DD. Expression of a functional high-affinity IgG receptor, Fc gamma RI, on human mast cells: up-regulation by IFN-gamma. Journal of immunology. 2000;164:4332–4339. doi: 10.4049/jimmunol.164.8.4332.
    1. Oshiba A, Hamelmann E, Takeda K, Bradley KL, Loader JE, Larsen GL, Gelfand EW. Passive transfer of immediate hypersensitivity and airway hyperresponsiveness by allergen-specific immunoglobulin (Ig) E and IgG1 in mice. The Journal of Clinical Investigation. 1996;97:1398–1408. doi: 10.1172/JCI118560.
    1. Rothman AL. Immunity to dengue virus: a tale of original antigenic sin and tropical cytokine storms. Nature Reviews Immunology. 2011;11:532–543. doi: 10.1038/nri3014.
    1. Sabin AB. Research on dengue during World War II. The American Journal of Tropical Medicine and Hygiene. 1952;1:30–50.
    1. Sanchez LF, Hotta H, Hotta S, Homma M. Degranulation and histamine release from murine mast cells sensitized with dengue virus-immune sera. Microbiology and Immunology. 1986;30:753–759. doi: 10.1111/j.1348-0421.1986.tb03002.x.
    1. Schulze IT, Schlesinger RW. Plaque assay of dengue and other group B arthropod-borne viruses under methyl cellulose overlay media. Virology. 1963;19:40–48. doi: 10.1016/0042-6822(63)90022-9.
    1. Shresta S, Sharar KL, Prigozhin DM, Beatty PR, Harris E. Murine model for dengue virus-induced lethal disease with increased vascular permeability. Journal of Virology. 2006;80:10208–10217. doi: 10.1128/JVI.00062-06.
    1. Sithiprasasna R, Strickman D, Innis BL, Linthicum KJ. ELISA for detecting dengue and Japanese encephalitis viral antigen in mosquitoes. Annals of Tropical Medicine and Parasitology. 1994;88:397–404.
    1. St John AL. Influence of mast cells on dengue protective immunity and immune pathology. PLOS Pathogens. 2013;9:e1003783. doi: 10.1371/journal.ppat.1003783.
    1. St John AL, Abraham SN, Gubler DJ. Barriers to preclinical investigations of anti-dengue immunity and dengue pathogenesis. Nature Reviews Microbiology. 2013a;11:420–426. doi: 10.1038/nrmicro3030.
    1. St John AL, Rathore AP, Raghavan B, Ng ML, Abraham SN. Contributions of mast cells and vasoactive products, leukotrienes and chymase, to dengue virus-induced vascular leakage. eLife. 2013b;2:e00481. doi: 10.7554/eLife.00481.
    1. St John AL, Rathore AP, Yap H, Ng ML, Metcalfe DD, Vasudevan SG, Abraham SN. Immune surveillance by mast cells during dengue infection promotes natural killer (NK) and NKT-cell recruitment and viral clearance. Proceedings of the National Academy of Sciences of USA. 2011;108:9190–9195. doi: 10.1073/pnas.1105079108.
    1. Sylvestre DL, Ravetch JV. A dominant role for mast cell Fc receptors in the Arthus reaction. Immunity. 1996;5:387–390. doi: 10.1016/S1074-7613(00)80264-2.
    1. Takizawa F, Adamczewski M, Kinet JP. Identification of the low affinity receptor for immunoglobulin E on mouse mast cells and macrophages as Fc gamma RII and Fc gamma RIII. The Journal of Experimental Medicine. 1992;176:469–475. doi: 10.1084/jem.176.2.469.
    1. Vaz NM, Ovary Z. Passive anaphylaxis in mice with gamma-G antibodies. 3. Release of histamine from mast cells by homologous antibodies. Journal of Immunology. 1968;100:1014–1019.
    1. Vaz NM, Prouvost-Danon A. Behaviour of mouse mast cells during anaphylaxis in vitro. Progress in Allergy. 1969;13:111–173.
    1. Wahala WM, Huang C, Butrapet S, White LJ, de Silva AM. Recombinant dengue type 2 viruses with altered e protein domain III epitopes are efficiently neutralized by human immune sera. Journal of Virology. 2012;86:4019–4023. doi: 10.1128/JVI.06871-11.
    1. World Health Organization . Dengue: guidelines for diagnosis, treatment, prevention and control - new edition. Geneva: World Health Organization; 2009.
    1. Yamanaka A, Kosugi S, Konishi E. Infection-enhancing and -neutralizing activities of mouse monoclonal antibodies against dengue type 2 and 4 viruses are controlled by complement levels. Journal of Virology. 2008;82:927–937. doi: 10.1128/JVI.00992-07.

Source: PubMed

3
구독하다