Contributions of mast cells and vasoactive products, leukotrienes and chymase, to dengue virus-induced vascular leakage

Ashley L St John, Abhay P S Rathore, Bhuvanakantham Raghavan, Mah-Lee Ng, Soman N Abraham, Ashley L St John, Abhay P S Rathore, Bhuvanakantham Raghavan, Mah-Lee Ng, Soman N Abraham

Abstract

Dengue Virus (DENV), a flavivirus spread by mosquito vectors, can cause vascular leakage and hemorrhaging. However, the processes that underlie increased vascular permeability and pathological plasma leakage during viral hemorrhagic fevers are largely unknown. Mast cells (MCs) are activated in vivo during DENV infection, and we show that this elevates systemic levels of their vasoactive products, including chymase, and promotes vascular leakage. Treatment of infected animals with MC-stabilizing drugs or a leukotriene receptor antagonist restores vascular integrity during experimental DENV infection. Validation of these findings using human clinical samples revealed a direct correlation between MC activation and DENV disease severity. In humans, the MC-specific product, chymase, is a predictive biomarker distinguishing dengue fever (DF) and dengue hemorrhagic fever (DHF). Additionally, our findings reveal MCs as potential therapeutic targets to prevent DENV-induced vasculopathy, suggesting MC-stabilizing drugs should be evaluated for their effectiveness in improving disease outcomes during viral hemorrhagic fevers. DOI:http://dx.doi.org/10.7554/eLife.00481.001.

Keywords: Human; Mouse; Viruses; chymase; dengue virus; leukotrienes; mast cell; vascular leakage.

Conflict of interest statement

The authors declare that no competing interests exist.

Figures

Figure 1.. DENV-Induced MC activation and microstructural…
Figure 1.. DENV-Induced MC activation and microstructural changes surrounding blood vessels.
TEMs acquired from mouse ear tissue 24 hr after either saline injection (A) or 1 × 105 PFU of DENV (B) and (C). Figure labels for (A)–(C): MC: mast cell; V: vessel; Er: erythrocyte. (A) In control tissues, apparently quiescent, granulated, MCs can be visualized in proximity to a blood vessel. (B) A field containing many MCs that appear activated due to their reduced granularity and cytoplasmic projections that are characteristic of MC degranulation. A granule that is being released is circled. This image also contains portions of two vessels, visible on the left and bottom sides. (C) MCs are closely associated with vessels in tissue that shows signs of fluid pooling or edema. Extracellular granules are visible throughout the tissue and are circled. A blood vessel containing an erythrocyte is labeled on the right side of the image and a second vessel, likely a lymphatic vessel, is visible on the left side. DOI:http://dx.doi.org/10.7554/eLife.00481.003
Figure 2.. Vascular pathology and DENV replication…
Figure 2.. Vascular pathology and DENV replication in the WT mouse model.
Cohorts of mice were monitored for 5 days following the experimental establishment of systemic infection using DENV clinical isolate Eden2, by intra-peritoneally. injection. Blood was obtained at 1 hr after infection and at subsequent 24 hr time points then analyzed to determine the (A) hematocrit values and (B) platelet concentration for individual infected mice (n = 3–4 at each time point). A dashed line represents the average baseline values for uninfected, naïve animals. (C) At the same time points, tissue was harvested from the spleen and liver and real time PCR was performed after cDNA conversion in order to quantitate DENV genome copies (PFU equivalents) in the tissue, which was normalized to the tissue mass. Error bars for panels (AC) represent the SEM and are included for all groups; where they are not visible, variation was small. (D) Images of spleen sections along this time course are presented, where 10-μm-thick sections were stained for DENV replication (dsRNA, red), monocytes and/or macrophages (cd11b, blue), and dendritic cells (cd11c, green). Bottom panels in (D) show the isolated dsRNA panel. (E) Co-localization images were generated using ImageJ software and reveal that dsRNA staining co-localized predominantly with the monocyte/macrophage marker cd11b, and to a lesser extent, the DC marker, cd11c. For panels (D and E), the dashed line denotes the border of the spleen in the tissue section. DOI:http://dx.doi.org/10.7554/eLife.00481.004
Figure 3.. Vascular leakage during DENV infection…
Figure 3.. Vascular leakage during DENV infection is MC-dependent.
(A) Graph depicts the serum concentration of MCPT1, which was quantified using serum obtained from WT or Sash mice, 24 and 48 hr after intra-peritoneally. injection with 1 × 106 PFU of DENV. MCPT1 was not detected (ND) in uninfected WT mice and uninfected or infected Sash mice. Error bars represent the SEM of ELISA replicates using pooled serum samples from n = 4 animals. To compare vascular leakage in infected vs uninfected WT, Sash, and Sash-R mice, (B) hematocrit analysis using heparinized blood and (C) quantitation of Evans blue dye leakage into liver tissue were performed 24 hr after infection with DENV. Representative images of mouse livers after saline perfusion are presented below the respective data bars to support that visually perceivable vascular leakage occurred in DENV-infected animals. For (B and C), error bars represent the SEM where values were obtained from individual infected mice n = 3–6 per group. * indicates a significant increase over uninfected controls; p≤0.05. DOI:http://dx.doi.org/10.7554/eLife.00481.005
Figure 3—figure supplement 1.. Representative images of…
Figure 3—figure supplement 1.. Representative images of livers from uninfected mice, or mice infected with 1 × 106 PFU of DENV.
Livers appeared normal during necropsy at 24 hr following injection with Evans blue dye. Perfusion of mice with saline eliminates blood in the vasculature. In mice infected with DENV, vascular permeability can be visualized due to remaining blood and Evans blue dye within tissues after saline perfusion. DOI:http://dx.doi.org/10.7554/eLife.00481.006
Figure 3—figure supplement 2.. Representative images of…
Figure 3—figure supplement 2.. Representative images of the kidneys of WT mice or Sash mice that were infected with 1 × 106 PFU of DENV.
After injection of Evan's blue dye followed by saline perfusion, vascular leakage was visible in the kidneys of WT but not Sash mice after DENV infection. DOI:http://dx.doi.org/10.7554/eLife.00481.007
Figure 4.. Drugs targeting MCs and their…
Figure 4.. Drugs targeting MCs and their products improve DENV-induced vasculopathy.
(A) Evans blue dye perfusion studies and hematocrit analysis were performed to determine the vascular leakage in mice infected intra-peritoneally with 1 × 106 PFU of DENV. Serum was obtained from uninfected mice, DENV-infected and untreated mice, and mice that received MC-stabilizing or MC-product targeting treatments (see ‘Materials and methods’) 24 hr after infection. Error bars represent the SEM of values obtained from individual animals n = 3–6 per group. Data was analyzed by ANOVA with Bonferroni post-tests to determine significance; * indicates a significant increase over control (uninfected) values and ** indicates a significant decrease from DENV-infected, untreated values; p≤0.05. (B) Serum ELISA for MCPT1 was performed using pooled serum from DENV-infected, untreated mice and DENV-infected, cromolyn-treated mice. Significance was determined by ANOVA; δ indicates a significant decrease compared to untreated controls; p≤0.05. (C) Viral genome copies were quantified in the serum of mice infected with DENV that were either untreated or treated with cromolyn. The moderate increase with cromolyn treatment was not significant with p=0.09 and n = 5. (D) Trans-well assays demonstrate the direct activity of MCs and MC products on permeability of a monolayer of EOMA cells. Significance was determined by ANOVA. δ indicates a significant decrease in TER compared to exposure to supernatants from untreated MC or DENV alone treatment (p<0.05). Groups treated with montelukast or chymase inhibitor cocktail significantly increased TER over untreated EOMA cells exposed to supernatant from DENV activated MCs; *p<0.05. Cromolyn treatment during DENV exposure resulted in increased TER over supernatants from untreated DENV-exposed BMMCs **p<0.01. (E) Trans well assays were also performed using peritoneal and pleural cavity MCs isolated by antibody labeling and magnetic separation. Purified MCs, which have abundant eosinophilic cytoplasmic granules, are imaged in the inset. Purified MCs from WT, 5-LO-KO, or TNF-KO mice were untreated or treated with DENV (MOI = 5) for 1 hr prior to isolation of supernatant for exposure to EOMA cells. Supernatants from both WT and TNF-KO MCs resulted in a significant reduction in the TER of EOMA cells with exposure compared to controls, determined by ANOVA; for δ p<0.05. 5-LO-KO showed a trend towards slightly reduced TER, but this was not significant since p=0.06. DENV activated WT MCs promoted significantly reduced relative TER readings compared to DENV activated 5-LO-KO MCs, determined by t-test *p=0.01. Similar results were obtained in a second independent vascular endothelial cell line, SVEC4-10EHR1 (Figure 4—figure supplement 1). DOI:http://dx.doi.org/10.7554/eLife.00481.008
Figure 4—figure supplement 1.. Supernatants from both…
Figure 4—figure supplement 1.. Supernatants from both WT and TNF-KO MCs resulted in a significant reduction in the TER of monolayers of vascular endothelial cell line SVEC4-10EHR1 with exposure compared to controls, determined by ANOVA; for δ p
Supernatants from 5-LO-KO MCs did not promote detectable decreases in TER compared to controls. DENV activated WT MCs promoted significantly reduced relative TER readings compared to DENV activated 5-LO-KO MCs, determined by T-test *p=0.003, demonstrating that MC-derived leukotrienes mediate permeability of endothelial monolayers after DENV activation. DOI:http://dx.doi.org/10.7554/eLife.00481.009
Figure 5.. Cromolyn is effective in the…
Figure 5.. Cromolyn is effective in the IFN-α,β,γ-deficient mouse model to limit DENV-induced vasculopathy.
(A) Images are presented for control (top) and DENV-infected mesentery tissue (bottom) in channel series showing staining for blood vessels (CD31, blue), MC granules (MC-heparin by probing for Avidin, red), and viral replication (NS3, green), as well as the merged image. Mesentery tissue from the DENV-permissive mouse strain, AG129, was isolated from control or DENV-infected tissue at 24 hr after intra-peritoneally injection of 2 × 105 PFU of DENV strain Eden2, followed by immunostaining in whole mount and viewing at 20× magnification. MCs can be observed lining the blood vessels (branches of the mesenteric artery) in control tissue (left). Discrete avidin-staining particles suggest extensive degranulation in DENV-infected mesentery (right). Note that the endothelial junction marker, CD31, appears reduced and that NS3 staining is only present in the DENV-infected panel (right). L designates the lumen of the blood vessel in both panels. (B) Mice deficient in IFN-α,β,γ (strain AG129) were infected with DENV by intra-peritoneally injection of 2 × 105 PFU of Eden2. After 1 day, treatment was initiated for some infected mice by administering intra-peritoneally injections of cromolyn. On day 3, blood was collected from untreated and cromolyn-treated infected groups and uninfected controls. Hematocrit analysis was performed using blood from individual mice n ≥ 3. Error bars represent the SEM and * indicates a significant increase over uninfected controls and ** indicates a significant decrease compared to DENV infection alone. The p-value for the comparison between uninfected vs DENV + cromolyn was not significant. The graph in the right panel depicts the plaque forming units obtained using pooled serum. Error bars represent the SEM of the assay, which was performed in replicates. Where no error bars are apparent, values obtained were the same for each replicate. * indicates a significant increase for the cromolyn-treated animals compared to infection alone. DOI:http://dx.doi.org/10.7554/eLife.00481.010
Figure 6.. Severity of DENV-induced disease in…
Figure 6.. Severity of DENV-induced disease in humans is linked to the MC product chymase.
(A) Graph depicts the chymase concentration in human serum for healthy controls, DENV-negative febrile patients, and patients that were diagnosed with DF or DHF and positive for DENV by molecular tests (see ‘Materials and methods’). For DENV-Neg, DF and DHF patients, serum was collected during acute infection, 2–4 days after the onset of fever. (B) Graph depicts the serum chymase concentration in DF and DHF patients 4–7 days after fever onset (defervescent phase). For (A and B), each dot represents the average concentration for an individual patient (n = 10–108 patients per group). (C) Data is represented as the relative amount of chymase in patient samples obtained in the acute phase (left) or defervescent phase (right), after normalizing to the average chymase concentration in healthy control human serum. For (AC) ANOVA analysis was used to determine significance of samples with Bonferroni’s post-test to determine significance between groups. * indicates a significant increase over healthy controls and DENV-Neg, febrile controls and ** indicates a significant increase over healthy control, DENV-Neg control, and DF groups. p<0.0001. (D) Graph depicts the concentration of chymase in serum samples grouped based on the serotype of DENV with which the patient was infected. Analysis by two-way ANOVA to compare chymase concentrations amongst DF and DHF samples reveals that serotype significantly influenced the chymase levels in patient sera, p<0.0001, although contributing to only 2.6% of the total variation. The concentrations of chymase in (E) DF or (F) DHF patients with either primary (1°) or secondary (2°) infection are shown. Chymase levels were significantly higher during secondary infection *p=0.0049 for DHF patients, but did not differ for DF patients, determined by Student’s unpaired t-test. (G) The concentrations of chymase are plotted vs the corresponding amounts of virus genome copies amplified from serum samples (represented as the inverse of the cross-over threshold [CT] value determined by real time PCR). For DF samples (green), Pearson’s R = 0.06, indicating no correlation. For DHF samples (purple), Pearson’s R = −0.85, indicating a correlation between higher viral genome copies and lower chymase levels. DOI:http://dx.doi.org/10.7554/eLife.00481.011
Figure 6—figure supplement 1.. Serum chymase levels…
Figure 6—figure supplement 1.. Serum chymase levels are presented to represent the repeated measures for individual patients at the early (acute) and late (defervescent) time points of infection and a line connects each patient’s paired values.
When assessed using a repeated measures ANOVA, DF and DHF groups differ highly significantly with pDOI: http://dx.doi.org/10.7554/eLife.00481.012
Figure 7.. Diagram representing the impact of…
Figure 7.. Diagram representing the impact of DENV-induced activation of MCs on the vasculature.
High DENV viral titers in vivo results in the activation of MCs, which release many vasoactive factors in the vicinity of blood vessels including leukotrienes and proteases, such as chymase. These factors act in concert to promote vascular leakage that, when occurring on a systemic level, has pathological consequences for the host. Drugs that target MC products can limit this leakage and vascular pathology. Similarly, the MC-specific product chymase can also be used to predict the severity of hemorrhagic disease in human patients. DOI:http://dx.doi.org/10.7554/eLife.00481.013

References

    1. Abraham SN, St John AL. 2010. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol 10:440–52 doi: 10.1038/nri2782
    1. Assuncao-Miranda I, Amaral FA, Bozza FA, Fagundes CT, Sousa LP, Souza DG, et al. 2010. Contribution of macrophage migration inhibitory factor to the pathogenesis of dengue virus infection. FASEB J 24:218–28 doi: 10.1096/fj.09-139469
    1. Atrasheuskaya A, Petzelbauer P, Fredeking TM, Ignatyev G. 2003. Anti-TNF antibody treatment reduces mortality in experimental dengue virus infection. FEMS Immunol Med Microbiol 35:33–42 doi: 10.1111/j.1574-695X.2003.tb00646.x
    1. Bente DA, Rico-Hesse R. 2006. Models of dengue virus infection. Drug Discov Today Dis Models 3:97–103 doi: 10.1016/j.ddmod.2006.03.014
    1. Blackley S, Kou Z, Chen H, Quinn M, Rose RC, Schlesinger JJ, et al. 2007. Primary human splenic macrophages, but not T or B cells, are the principal target cells for dengue virus infection in vitro. J Virol 81:13325–34 doi: 10.1128/JVI.01568-07
    1. Bois P. 1964. Mast cells and histamine concentration in muscle and liver of dystrophic mice. Am J Physiol 206:338–40
    1. Boonpucknavig S, Vuttiviroj O, Boonpucknavig V. 1981. Infection of young adult mice with dengue virus type 2. Trans R Soc Trop Med Hyg 75:647–53 doi: 10.1016/0035-9203(81)90142-5
    1. Brett J, Gerlach H, Nawroth P, Steinberg S, Godman G, Stern D. 1989. Tumor necrosis factor/cachectin increases permeability of endothelial cell monolayers by a mechanism involving regulatory G proteins. J Exp Med 169:1977–91 doi: 10.1084/jem.169.6.1977
    1. Busse WW, McGill KA, Horwitz RJ. 1999. Leukotriene pathway inhibitors in asthma and chronic obstructive pulmonary disease. Clin Exp Allergy 29(suppl 2):110–5
    1. Chen HC, Hofman FM, Kung JT, Lin YD, Wu-Hsieh BA. 2007. Both virus and tumor necrosis factor alpha are critical for endothelium damage in a mouse model of dengue virus-induced hemorrhage. J Virol 81:5518–26 doi: 10.1128/JVI.02575-06
    1. Chien LJ, Liao TL, Shu PY, Huang JH, Gubler DJ, Chang GJ. 2006. Development of real-time reverse transcriptase PCR assays to detect and serotype dengue viruses. J Clin Microbiol 44:1295–304 doi: 10.1128/JCM.44.4.1295-1304.2006
    1. Dahlen SE, Bjork J, Hedqvist P, Arfors KE, Hammarstrom S, Lindgren JA, et al. 1981. Leukotrienes promote plasma leakage and leukocyte adhesion in postcapillary venules: in vivo effects with relevance to the acute inflammatory response. Proc Natl Acad Sci USA 78:3887–91 doi: 10.1073/pnas.78.6.3887
    1. Fink J, Gu F, Ling L, Tolfvenstam T, Olfat F, Chin KC, et al. 2007. Host gene expression profiling of dengue virus infection in cell lines and patients. PLoS Negl Trop Dis 1:e86 doi: 10.1371/journal.pntd.0000086
    1. Finkelman FD. 2007. Anaphylaxis: lessons from mouse models. J Allergy Clin Immunol 120:506–15 doi: 10.1016/j.jaci.2007.07.033
    1. Flower RJ, Harvey EA, Kingston WP. 1976. Inflammatory effects of prostaglandin D2 in rat and human skin. Br J Pharmacol 56:229–33 doi: 10.1111/j.1476-5381.1976.tb07446.x
    1. Frangogiannis NG, Lindsey ML, Michael LH, Youker KA, Bressler RB, Mendoza LH, et al. 1998. Resident cardiac mast cells degranulate and release preformed TNF-alpha, initiating the cytokine cascade in experimental canine myocardial ischemia/reperfusion. Circulation 98:699–710 doi: 10.1161/01.CIR.98.7.699
    1. Furuta T, Murao LA, Lan NT, Huy NT, Huong VT, Thuy TT, et al. 2012. Association of mast cell-derived VEGF and proteases in Dengue shock syndrome. PLoS Negl Trop Dis 6:e1505 doi: 10.1371/journal.pntd.0001505
    1. Gowen BB, Julander JG, London NR, Wong MH, Larson D, Morrey JD, et al. 2010. Assessing changes in vascular permeability in a hamster model of viral hemorrhagic fever. Virol J 7:240 doi: 10.1186/1743-422X-7-240
    1. Grimbaldeston MA, Chen CC, Piliponsky AM, Tsai M, Tam SY, Galli SJ. 2005. Mast cell-deficient W-sash c-kit mutant Kit W-sh/W-sh mice as a model for investigating mast cell biology in vivo. Am J Pathol 167:835–48 doi: 10.1016/S0002-9440(10)62055-X
    1. Halstead SB. 2007. Dengue. Lancet 370:1644–52 doi: 10.1016/S0140-6736(07)61687-0
    1. He S, Walls AF. 1998. The induction of a prolonged increase in microvascular permeability by human mast cell chymase. Eur J Pharmacol 352:91–8 doi: 10.1016/S0014-2999(98)00343-4
    1. Holian A, Hamilton R, Scheule RK. 1991. Mechanistic aspects of cromolyn sodium action on the alveolar macrophage: inhibition of stimulation by soluble agonists. Agents Actions 33:318–25 doi: 10.1007/BF01986580
    1. Huang C, Wong GW, Ghildyal N, Gurish MF, Sali A, Matsumoto R, et al. 1997. The tryptase, mouse mast cell protease 7, exhibits anticoagulant activity in vivo and in vitro due to its ability to degrade fibrinogen in the presence of the diverse array of protease inhibitors in plasma. J Biol Chem 272:31885–93 doi: 10.1074/jbc.272.50.31885
    1. Huang KJ, Li SY, Chen SC, Liu HS, Lin YS, Yeh TM, et al. 2000. Manifestation of thrombocytopenia in dengue-2-virus-infected mice. J Gen Virol 81:2177–82
    1. Jessie K, Fong MY, Devi S, Lam SK, Wong KT. 2004. Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis 189:1411–8 doi: 10.1086/383043
    1. Koda W, Harada K, Tsuneyama K, Kono N, Sasaki M, Matsui O, et al. 2000. Evidence of the participation of peribiliary mast cells in regulation of the peribiliary vascular plexus along the intrahepatic biliary tree. Lab Invest 80:1007–17 doi: 10.1038/labinvest.3780106
    1. Koraka P, Murgue B, Deparis X, Setiati TE, Suharti C, van Gorp EC, et al. 2003. Elevated levels of total and dengue virus-specific immunoglobulin E in patients with varying disease severity. J Med Virol 70:91–8 doi: 10.1002/jmv.10358
    1. Kunder CA, St John AL, Abraham SN. 2011. Mast cell modulation of the vascular and lymphatic endothelium. Blood 118:5383–93 doi: 10.1182/blood-2011-07-358432
    1. Leff JA, Busse WW, Pearlman D, Bronsky EA, Kemp J, Hendeles L, et al. 1998. Montelukast, a leukotriene-receptor antagonist, for the treatment of mild asthma and exercise-induced bronchoconstriction. N Engl J Med 339:147–52 doi: 10.1056/NEJM199807163390302
    1. Low JG, Ooi EE, Tolfvenstam T, Leo YS, Hibberd ML, Ng LC, et al. 2006. Early Dengue infection and outcome study (EDEN)—study design and preliminary findings. Ann Acad Med Singapore 35:783–9
    1. Mabalirajan U, Kadhiravan T, Sharma SK, Banga A, Ghosh B. 2005. Th(2) immune response in patients with dengue during defervescence: preliminary evidence. Am J Trop Med Hyg 72:783–5
    1. Malasit P. 1987. Complement and dengue haemorrhagic fever/shock syndrome. Southeast Asian J Trop Med Public Health 18:316–20
    1. McClean SP, Arreaza EE, Lett-Brown MA, Grant JA. 1989. Refractory cholinergic urticaria successfully treated with ketotifen. J Allergy Clin Immunol 83:738–41 doi: 10.1016/0091-6749(89)90008-0
    1. McFadden ER, Jnr, Gilbert IA. 1992. Asthma. N Engl J Med 327:1928–37 doi: 10.1056/NEJM199212313272708
    1. McLachlan JB, Hart JP, Pizzo SV, Shelburne CP, Staats HF, Gunn MD, et al. 2003. Mast cell-derived tumor necrosis factor induces hypertrophy of draining lymph nodes during infection. Nat Immunol 4:1199–205 doi: 10.1038/ni1005
    1. Oschatz C, Maas C, Lecher B, Jansen T, Bjorkqvist J, Tradler T, et al. 2011. Mast cells increase vascular permeability by heparin-initiated bradykinin formation in vivo. Immunity 34:258–68 doi: 10.1016/j.immuni.2011.02.008
    1. Paes MV, Pinhao AT, Barreto DF, Costa SM, Oliveira MP, Nogueira AC, et al. 2005. Liver injury and viremia in mice infected with dengue-2 virus. Virology 338:236–46 doi: 10.1016/j.virol.2005.04.042
    1. Rathore AP, Paradkar PN, Watanabe S, Tan KH, Sung C, Connolly JE, et al. 2011. Celgosivir treatment misfolds dengue virus NS1 protein, induces cellular pro-survival genes and protects against lethal challenge mouse model. Antiviral Res 92:453–60 doi: 10.1016/j.antiviral.2011.10.002
    1. Raut CG, Deolankar RP, Kolhapure RM, Goverdhan MK. 1996. Susceptibility of laboratory-bred rodents to the experimental infection with dengue virus type 2. Acta Virol 40:143–6
    1. Rosen L, Khin MM, U T. 1989. Recovery of virus from the liver of children with fatal dengue: reflections on the pathogenesis of the disease and its possible analogy with that of yellow fever. Res Virol 140:351–60 doi: 10.1016/S0923-2516(89)80115-3
    1. Sanchez LF, Hotta H, Hotta S, Homma M. 1986. Degranulation and histamine release from murine mast cells sensitized with dengue virus-immune sera. Microbiol Immunol 30:753–9
    1. Sendo T, Sumimura T, Itoh Y, Goromaru T, Aki K, Yano T, et al. 2003. Involvement of proteinase-activated receptor-2 in mast cell tryptase-induced barrier dysfunction in bovine aortic endothelial cells. Cell Signal 15:773–81 doi: 10.1016/S0898-6568(03)00014-7
    1. Shresta S, Kyle JL, Snider HM, Basavapatna M, Beatty PR, Harris E. 2004. Interferon-dependent immunity is essential for resistance to primary dengue virus infection in mice, whereas T- and B-cell-dependent immunity are less critical. J Virol 78:2701–10 doi: 10.1128/JVI.78.6.2701-2710.2004
    1. St John AL, Abraham SN, Gubler DJ. 2013. Barriers to pre-clinical investigations of anti-dengue immunity and immune-pathology. Nat Rev Immunol.
    1. St John AL, Rathore AP, Yap H, Ng ML, Metcalfe DD, Vasudevan SG, et al. 2011. Immune surveillance by mast cells during dengue infection promotes natural killer (NK) and NKT-cell recruitment and viral clearance. Proc Natl Acad Sci USA 108:9190–5 doi: 10.1073/pnas.1105079108
    1. Tassaneetrithep B, Burgess TH, Granelli-Piperno A, Trumpfheller C, Finke J, Sun W, et al. 2003. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med 197:823–9 doi: 10.1084/jem.20021840
    1. Theoharides TC, Sieghart W, Greengard P, Douglas WW. 1980. Antiallergic drug cromolyn may inhibit histamine secretion by regulating phosphorylation of a mast cell protein. Science 207:80–2 doi: 10.1126/science.6153130
    1. Tseng CS, Lo HW, Teng HC, Lo WC, Ker CG. 2005. Elevated levels of plasma VEGF in patients with dengue hemorrhagic fever. FEMS Immunol Med Microbiol 43:99–102 doi: 10.1016/j.femsim.2004.10.004
    1. Vaughn DW, Green S, Kalayanarooj S, Innis BL, Nimmannitya S, Suntayakorn S, et al. 2000. Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. J Infect Dis 181:2–9 doi: 10.1086/315215
    1. Vitarana T, de Silva H, Withana N, Gunasekera C. 1991. Elevated tumour necrosis factor in dengue fever and dengue haemorrhagic fever. Ceylon Med J 36:63–5
    1. Wang Z, Lai Y, Bernard JJ, Macleod DT, Cogen AL, Moss B, et al. 2012. Skin mast cells protect mice against vaccinia virus by triggering mast cell receptor S1PR2 and releasing antimicrobial peptides. J Immunol 188:345–57 doi: 10.4049/jimmunol.1101703
    1. Williams CM, Galli SJ. 2000. Mast cells can amplify airway reactivity and features of chronic inflammation in an asthma model in mice. J Exp Med 192:455–62 doi: 10.1084/jem.192.3.455
    1. World Health Organization 1997. Dengue hemorrhagic fever: diagnosis, treatment, prevention, and control. 2nd ed Geneva: World Health Organization;
    1. Wu SJ, Grouard-Vogel G, Sun W, Mascola JR, Brachtel E, Putvatana R, et al. 2000. Human skin Langerhans cells are targets of dengue virus infection. Nat Med 6:816–20 doi: 10.1038/77553
    1. Yauch LE, Shresta S. 2008. Mouse models of dengue virus infection and disease. Antiviral Res 80:87–93 doi: 10.1016/j.antiviral.2008.06.010
    1. Zellweger RM, Prestwood TR, Shresta S. 2010. Enhanced infection of liver sinusoidal endothelial cells in a mouse model of antibody-induced severe dengue disease. Cell Host Microbe 7:128–39 doi: 10.1016/j.chom.2010.01.004

Source: PubMed

3
Suscribir