• 沒有找到結果。

The patients’ sera data were expressed as the median± interquartile range and tested if the val-ues come from a Gaussian distribution by using D’Agostino and Pearson omnibus normality test. If the data meet Gaussian distribution, the significance of differences between each groups was analyzied using One-way ANOVA with Tukey’s method. If the data do not meet the assumptions of normality, they were analyzed with a non-parametric test by Kruskal-Wallis test. Thein vitro and in vivo data are expressed as the mean ± standard deviation (SD) from more than three independent experiments. Student’s t-test was used to analyze the significance

of differences between the test and control groups. One-way ANOVA with Kruskal-Wallis comparison test was used to analyze the significance of differences between multiple groups.

All data were analyzed by GraphPad Prism 5 software. P values <0.05 were considered statisti-cally significant.

Supporting information

S1 Table. Characteristics of dengue patients.

(DOCX)

S1 Fig. The correlations of serum NS1, HPA-1, MMP-9, CD138, MIF levels and viral load in severe dengue patients. The correlations of the concentrations of (A) NS1, (B) HPA-1, (C) MMP-9, (D) CD138 and (E) MIF and viral load in the same group of severe dengue patients were plotted. Linear regressions were analyzed using nonparametric correlation test (panel A, B, C, D and E).

(DOCX)

S2 Fig. DENV NS1 induces HPA-1 activation and vascular leakage. (A) HUVECs were treated with PBS, NS1 or NS1 mixed with anti-NS1 antibodies (2E8) or control mouse IgG (cmIgG) for 24 h, and the HPA-1 level was determined by western blot. The relative HPA-1 protein level (including the proform and active form) was normalized toβ-actin, and the fold change is noted under each band. (B) BALB/c mice were intravenously injected with Evans Blue dye, followed by subcutaneous injections of PBS or different doses of HPA-1, heat-dena-tured HPA-1 or thrombin. After the mice were sacrificed, skin samples were collected and pro-cessed 3 h postinjection.

(DOCX)

S3 Fig. DENV NS1-induced MIF secretion causes glycocalyx degradation and hyperperme-ability in HUVECs. (A) HUVECs were treated with different concentrations of NS1 for the indicated times, followed supernatant collection for the detection of MIF by ELISA. (B) HUVEC monolayers were incubated with the supernatant from control or NS1-treated HUVEC cultures for the indicated times, and the endothelial permeability was then deter-mined by Transwell permeability assay. (C) HUVEC monolayers were incubated with control or NS1-treated HUVEC-conditioned medium for 24 h. The concentration of CD138 in the supernatant after the incubation was determined by ELISA. (D) (E) Control or NS1-treated HUVEC culture supernatant with or without anti-MIF polyclonal antibodies or HPA-1 inhibi-tor (OGT 2115) or anti-NS1 antibodies (2E8) and incubated with HUVECs for 24 h. (D) The endothelial permeability was determined by Transwell permeability assay, and (E) the concen-tration of CD138 in the supernatant was determined by ELISA. (F) HUVEC monolayers were treated with PBS, NS1 or NS1 mixed with anti-NS1 antibodies (2E8) or HPA-1 inhibitor (OGT 2115) for the indicated times, and the concentration of MIF in the supernatant was determined by ELISA; S/N, supernatant;P<0.05,P<0.005,P<0.001; unpaired t-test (panel B and C), Kruskal-Wallis ANOVA (panel D and E).

(DOCX)

S4 Fig. MIF induces HPA-1 activation and glycocalyx shedding in HUVECs. (A) HUVECs were treated with or without MIF recombinant protein (1μg/ml) for the indicated times, and the concentration of CD138 in the supernatant was determined by ELISA. (B) HUVECs were treated with or without MIF recombinant protein (1μg/ml) for 18 h, and the HPA-1 level was determined by western blot. The relative HPA-1 protein level (including the proform and active form) was normalized toβ-actin, and the fold change is noted under each band. (C)

HUVECs were treated as indicated for 18 h and then stained for HPA-1 (red), CD138 (green), and nuclei (blue).P<0.05,P<0.005; unpaired t-test (panel A).

(DOCX)

S5 Fig. DENV NS1 does not induce MIF and MMP-9 secretion in PBMCs. (A) (B) Isolated human PBMCs were treated with or without NS1 (10μg/ml) for the indicated times, and the concentration of (A) MIF and (B) MMP-9 in the supernatant was determined by ELISA.

(DOCX)

S6 Fig. DENV NS1-induced MMP-9 secretion from WBCs causes endothelial hyperperme-ability. Isolated human WBCs were treated with or without NS1 for 24 h, and the supernatants were collected. HUVEC monolayers were incubated with the supernatant from control or NS1-treated WBCs for 6 h; then, endothelial permeability was determined by Transwell per-meability assay. S/N, supernatant;P<0.05; Kruskal-Wallis ANOVA.

(DOCX)

S7 Fig. Cytokine secretion profile of WBCs and THP-1 cells after DENV NS1 stimulation.

(A) (B) (C) Isolated human WBCs and (D) (E) (F) THP-1 cells were treated with or without NS1 for the indicated times, and the concentration of MIF, IL-6 and IL-8 in the supernatant was determined by ELISA;P<0.05,P<0.005,P<0.001; unpaired t-test (panel A, B, C and D).

(DOCX)

S8 Fig. DENV NS1-induced endothelial hyperpermeability is mediated by MIF. (A) HUVECs were transfected with MIF shRNA (shMIF) or scrambled shRNA (shLuc). The cell lysates were collected, and the relative protein level of MIF was measured by western blot. (B) The permeability of shMIF HUVECs and shLuc HUVECs after 24 h of NS1 treatment was detected by Transwell permeability assay. The results are presented as the mean± SD of tripli-cate measurements.

(DOCX)

S9 Fig. Inhibition of MIF and MMP-9 attenuate NS1-induced vascular leakage in mice.

BALB/c mice were intravenously injected with Evans Blue dye, followed by the subcutaneous injection of PBS or different doses of NS1, NS1 with MMP-9 inhibitor I or NS1 with ISO-1 for 6 h. After 5 h, the mice were subcutaneously injected with thrombin as a positive control. After another hour, the mice were sacrificed, and skin samples were collected and processed.

(DOCX)

S10 Fig. The correlations of serum levels of HPA-1 with NS1 and MIF in severe dengue patients. (A) The correlations of the concentrations of (A) NS1, (B) MIF, and HPA-1 in the severe dengue patients were plotted. Linear regressions were analyzed using nonparametric correlation test (panel A and B).

(DOCX)

Acknowledgments

We would like to thank the members of the Center of Infectious Disease and Signaling Research of NCKU for their invaluable input and insight throughout the course of this study. We also thank the technical services provided by the “Bioimaging Core Facility of the National Core Facility for Biopharmaceuticals, Ministry of Science and Technology, Taiwan”.

Author Contributions

Conceptualization: Hong-Ru Chen, Trai-Ming Yeh.

Investigation: Hong-Ru Chen, Chiao-Hsuan Chao.

Methodology: Hong-Ru Chen.

Resources: Ching-Chuan Liu, Tzong-Shiann Ho, Huey-Pin Tsai, Guey-Chuen Perng, Jen-Ren Wang.

Supervision: Trai-Ming Yeh.

Writing – original draft: Hong-Ru Chen.

Writing – review & editing: Guey-Chuen Perng, Yee-Shin Lin, Trai-Ming Yeh.

References

1. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, et al. The global distribution and burden of dengue. Nature. 2013; 496(7446):504–7.https://doi.org/10.1038/nature12060PMID:

23563266

2. Radeva MY, Waschke J. Mind the gap: mechanisms regulating the endothelial barrier. Acta physiol.

2017. PMID:28231640

3. Mehta D, Ravindran K, Kuebler WM. Novel regulators of endothelial barrier function. Am J Physiol Lung Cell Mol Physiol. 2014; 307(12):L924–35.https://doi.org/10.1152/ajplung.00318.2014PMID:25381026 4. Lipowsky HH. The endothelial glycocalyx as a barrier to leukocyte adhesion and its mediation by

extra-cellular proteases. Annals Biomed Eng. 2012; 40(4):840–8. PMID:22065204

5. Schmidt EP, Yang Y, Janssen WJ, Gandjeva A, Perez MJ, Barthel L, et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat Med. 2012; 18 (8):1217–23.https://doi.org/10.1038/nm.2843PMID:22820644

6. Becker BF, Jacob M, Leipert S, Salmon AH, Chappell D. Degradation of the endothelial glycocalyx in clinical settings: searching for the sheddases. Br J Clin Pharmacol. 2015; 80(3):389–402.https://doi.

org/10.1111/bcp.12629PMID:25778676

7. Chelazzi C, Villa G, Mancinelli P, De Gaudio AR, Adembri C. Glycocalyx and sepsis-induced alterations in vascular permeability. Crit Care. 2015; 19:26.https://doi.org/10.1186/s13054-015-0741-zPMID:

25887223

8. Chappell D, Jacob M, Rehm M, Stoeckelhuber M, Welsch U, Conzen P, et al. Heparinase selectively sheds heparan sulphate from the endothelial glycocalyx. Biol Chem. 2008; 389(1):79–82.https://doi.

org/10.1515/BC.2008.005PMID:18095872

9. Purushothaman A, Uyama T, Kobayashi F, Yamada S, Sugahara K, Rapraeger AC, et al. Heparanase-enhanced shedding of syndecan-1 by myeloma cells promotes endothelial invasion and angiogenesis.

Blood. 2010; 115(12):2449–57.https://doi.org/10.1182/blood-2009-07-234757PMID:20097882 10. Yang Y, Macleod V, Miao HQ, Theus A, Zhan F, Shaughnessy JD Jr., et al. Heparanase enhances

syn-decan-1 shedding: a novel mechanism for stimulation of tumor growth and metastasis. J Biol Chem.

2007; 282(18):13326–33.https://doi.org/10.1074/jbc.M611259200PMID:17347152

11. Mulivor AW, Lipowsky HH. Inhibition of glycan shedding and leukocyte-endothelial adhesion in postca-pillary venules by suppression of matrixmetalloprotease activity with doxycycline. Microcirculation.

2009; 16(8):657–66.https://doi.org/10.3109/10739680903133714PMID:19905966

12. Lipowsky HH. Protease Activity and the Role of the Endothelial Glycocalyx in Inflammation. Drug Discov Today Dis Models. 2011; 8(1):57–62.https://doi.org/10.1016/j.ddmod.2011.05.004PMID:22059089 13. Nguyen-Pouplin J, Pouplin T, Van TP, The TD, Thi DN, Farrar J, et al. Dextran fractional clearance

stud-ies in acute dengue infection. PLoS Negl Trop Dis. 2011; 5(8):e1282.https://doi.org/10.1371/journal.

pntd.0001282PMID:21886850

14. Suwarto S, Sasmono RT, Sinto R, Ibrahim E, Suryamin M. Association of Endothelial Glycocalyx and Tight and Adherens Junctions With Severity of Plasma Leakage in Dengue Infection. J Infect Dis. 2017;

215(6):992–9.https://doi.org/10.1093/infdis/jix041PMID:28453844

15. Thomas SJ. NS1: A corner piece in the dengue pathogenesis puzzle? Sci Transl Med. 2015; 7 (304):304fs37.https://doi.org/10.1126/scitranslmed.aad1255PMID:26355028

16. Modhiran N, Watterson D, Muller DA, Panetta AK, Sester DP, Liu L, et al. Dengue virus NS1 protein activates cells via Toll-like receptor 4 and disrupts endothelial cell monolayer integrity. Sci Transl Med.

2015; 7(304):304ra142.https://doi.org/10.1126/scitranslmed.aaa3863PMID:26355031

17. Beatty PR, Puerta-Guardo H, Killingbeck SS, Glasner DR, Hopkins K, Harris E. Dengue virus NS1 trig-gers endothelial permeability and vascular leak that is prevented by NS1 vaccination. Sci Transl Med.

2015; 7(304):304ra141.https://doi.org/10.1126/scitranslmed.aaa3787PMID:26355030

18. Puerta-Guardo H, Glasner DR, Harris E. Dengue Virus NS1 Disrupts the Endothelial Glycocalyx, Lead-ing to Hyperpermeability. PLoS Pathog. 2016; 12(7):e1005738.https://doi.org/10.1371/journal.ppat.

1005738PMID:27416066

19. Chen HR, Chuang YC, Lin YS, Liu HS, Liu CC, Perng GC, et al. Dengue Virus Nonstructural Protein 1 Induces Vascular Leakage through Macrophage Migration Inhibitory Factor and Autophagy. PLoS Negl Trop Dis. 2016; 10(7):e0004828.https://doi.org/10.1371/journal.pntd.0004828PMID:27409803 20. Lue H, Dewor M, Leng L, Bucala R, Bernhagen J. Activation of the JNK signalling pathway by

macro-phage migration inhibitory factor (MIF) and dependence on CXCR4 and CD74. Cell Signal. 2011; 23 (1):135–44.https://doi.org/10.1016/j.cellsig.2010.08.013PMID:20807568

21. Calandra T, Roger T. Macrophage migration inhibitory factor: a regulator of innate immunity. Nat Rev Immunol. 2003; 3(10):791–800.https://doi.org/10.1038/nri1200PMID:14502271

22. Bernhagen J, Calandra T, Bucala R. Regulation of the immune response by macrophage migration inhibitory factor: biological and structural features. J Mol Med. 1998; 76(3–4):151–61. PMID:9535548 23. Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, et al. MIF is a noncognate ligand

of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 2007; 13 (5):587–96.https://doi.org/10.1038/nm1567PMID:17435771

24. Weber C, Kraemer S, Drechsler M, Lue H, Koenen RR, Kapurniotu A, et al. Structural determinants of MIF functions in CXCR2-mediated inflammatory and atherogenic leukocyte recruitment. Proc Natl Acad Sci U S A 2008; 105(42):16278–83. PMID:18852457

25. Alampour-Rajabi S, El Bounkari O, Rot A, Muller-Newen G, Bachelerie F, Gawaz M, et al. MIF interacts with CXCR7 to promote receptor internalization, ERK1/2 and ZAP-70 signaling, and lymphocyte che-motaxis. FASEB J. 2015; 29(11):4497–511.https://doi.org/10.1096/fj.15-273904PMID:26139098 26. Chuang YC, Lei HY, Liu HS, Lin YS, Fu TF, Yeh TM. Macrophage migration inhibitory factor induced by

dengue virus infection increases vascular permeability. Cytokine. 2011; 54(2):222–31.https://doi.org/

10.1016/j.cyto.2011.01.013PMID:21320786

27. Chen LC, Shyu HW, Lei HY, Chen SH, Liu HS, Lin YS, Yeh TM. Dengue Virus Infection Induced NF-κB-dependent Macrophage Migration Inhibitory Factor Production. Am J Infect Dis. 2008; 4(1):22–31.

28. Chen LC, Lei HY, Liu CC, Shiesh SC, Chen SH, Liu HS, et al. Correlation of serum levels of macro-phage migration inhibitory factor with disease severity and clinical outcome in dengue patients. Am J Trop Med Hyg. 2006; 74(1):142–7. PMID:16407359

29. Assuncao-Miranda I, Amaral FA, Bozza FA, Fagundes CT, Sousa LP, Souza DG, et al. Contribution of macrophage migration inhibitory factor to the pathogenesis of dengue virus infection. FASEB J. 2010;

24(1):218–28.https://doi.org/10.1096/fj.09-139469PMID:19776337

30. Her Z, Kam YW, Gan VC, Lee B, Thein TL, Tan JJ, et al. Severity of Plasma Leakage Is Associated With High Levels of Interferon gamma-Inducible Protein 10, Hepatocyte Growth Factor, Matrix Metallo-proteinase 2 (MMP-2), and MMP-9 During Dengue Virus Infection. J Infect Dis. 2017; 215(1):42–51.

https://doi.org/10.1093/infdis/jiw494PMID:28077582

31. Luplertlop N, Misse D, Bray D, Deleuze V, Gonzalez JP, Leardkamolkarn V, et al. Dengue-virus-infected dendritic cells trigger vascular leakage through metalloproteinase overproduction. EMBO Rep.

2006; 7(11):1176–81.https://doi.org/10.1038/sj.embor.7400814PMID:17028575

32. Kubelka CF, Azeredo EL, Gandini M, Oliveira-Pinto LM, Barbosa LS, Damasco PV, et al. Metalloprotei-nases are produced during dengue fever and MMP9 is associated with severity. J Infect. 2010; 61 (6):501–5.https://doi.org/10.1016/j.jinf.2010.09.020PMID:20863849

33. Turner RJ, Sharp FR. Implications of MMP9 for Blood Brain Barrier Disruption and Hemorrhagic Trans-formation Following Ischemic Stroke. Front Cell Neurosci. 2016; 10:56.https://doi.org/10.3389/fncel.

2016.00056PMID:26973468

34. Kong YZ, Yu X, Tang JJ, Ouyang X, Huang XR, Fingerle-Rowson G, et al. Macrophage migration inhibi-tory factor induces MMP-9 expression: implications for destabilization of human atherosclerotic pla-ques. Atherosclerosis. 2005; 178(1):207–15.https://doi.org/10.1016/j.atherosclerosis.2004.08.030 PMID:15585220

35. Yu X, Lin SG, Huang XR, Bacher M, Leng L, Bucala R, et al. Macrophage migration inhibitory factor induces MMP-9 expression in macrophages via the MEK-ERK MAP kinase pathway. J Interferon Cyto-kine Res. 2007; 27(2):103–9.https://doi.org/10.1089/jir.2006.0054PMID:17316137

36. Alcon S, Talarmin A, Debruyne M, Falconar A, Deubel V, Flamand M. Enzyme-linked immunosorbent assay specific to Dengue virus type 1 nonstructural protein NS1 reveals circulation of the antigen in the blood during the acute phase of disease in patients experiencing primary or secondary infections. J Clin Microbiol. 2002; 40(2):376–81.https://doi.org/10.1128/JCM.40.2.376-381.2002PMID:11825945 37. Schonrich G, Raftery MJ. Neutrophil Extracellular Traps Go Viral. Front Immunol. 2016; 7:366.https://

doi.org/10.3389/fimmu.2016.00366PMID:27698656

38. Podolska MJ, Biermann MH, Maueroder C, Hahn J, Herrmann M. Inflammatory etiopathogenesis of systemic lupus erythematosus: an update. J Inflamm Res. 2015; 8:161–71.https://doi.org/10.2147/JIR.

S70325PMID:26316795

39. Dumitru CA, Gholaman H, Trellakis S, Bruderek K, Dominas N, Gu X, et al. Tumor-derived macrophage migration inhibitory factor modulates the biology of head and neck cancer cells via neutrophil activation.

Int J Cancer. 2011; 129(4):859–69.https://doi.org/10.1002/ijc.25991PMID:21328346

40. Clyde K, Kyle JL, Harris E. Recent advances in deciphering viral and host determinants of dengue virus replication and pathogenesis. J Virol. 2006; 80(23):11418–31.https://doi.org/10.1128/JVI.01257-06 PMID:16928749

41. Anderson R. Manipulation of cell surface macromolecules by flaviviruses. Adv Virus Res. 2003;

59:229–74. PMID:14696331

42. Luplertlop N, Misse D. MMP cellular responses to dengue virus infection-induced vascular leakage. Jpn J Infect Dis. 2008; 61(4):298–301. PMID:18653973

43. Voraphani N, Khongphatthanayothin A, Srikaew K, Tontulawat P, Poovorawan Y. Matrix metalloprotei-nase-9 (mmp-9) in children with dengue virus infection. Jpn J Infect Dis. 2010; 63(5):346–8. PMID:

20859002

44. Bonnema DD, Webb CS, Pennington WR, Stroud RE, Leonardi AE, Clark LL, et al. Effects of age on plasma matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs). J Card Fail. 2007; 13(7):530–40.https://doi.org/10.1016/j.cardfail.2007.04.010PMID:17826643

45. Goldberg GI, Strongin A, Collier IE, Genrich LT, Marmer BL. Interaction of 92-kDa type IV collagenase with the tissue inhibitor of metalloproteinases prevents dimerization, complex formation with interstitial collagenase, and activation of the proenzyme with stromelysin. J Biol Chem. 1992; 267(7):4583–91.

PMID:1311314

46. Itoh Y, Nagase H. Preferential inactivation of tissue inhibitor of metalloproteinases-1 that is bound to the precursor of matrix metalloproteinase 9 (progelatinase B) by human neutrophil elastase. J Biol Chem.

1995; 270(28):16518–21. PMID:7622455

47. Wang Y, Rosen H, Madtes DK, Shao B, Martin TR, Heinecke JW, et al. Myeloperoxidase inactivates TIMP-1 by oxidizing its N-terminal cysteine residue: an oxidative mechanism for regulating proteolysis during inflammation. J Biol Chem. 2007; 282(44):31826–34.https://doi.org/10.1074/jbc.M704894200 PMID:17726014

48. Glasner DR, Ratnasiri K, Puerta-Guardo H, Espinosa DA, Beatty PR, Harris E. Dengue virus NS1 cyto-kine-independent vascular leak is dependent on endothelial glycocalyx components. PLoS Pathog.

2017; 13(11):e1006673.https://doi.org/10.1371/journal.ppat.1006673PMID:29121099

49. Gore Y, Starlets D, Maharshak N, Becker-Herman S, Kaneyuki U, Leng L, et al. Macrophage migration inhibitory factor induces B cell survival by activation of a CD74-CD44 receptor complex. J Biol Chem.

2008; 283(5):2784–92.https://doi.org/10.1074/jbc.M703265200PMID:18056708

50. Wu W, Pan C, Meng K, Zhao L, Du L, Liu Q, et al. Hypoxia activates heparanase expression in an NF-kappaB dependent manner. Oncol Rep. 2010; 23(1):255–61. PMID:19956890

51. Hertz T, Beatty PR, MacMillen Z, Killingbeck SS, Wang C, Harris E. Antibody Epitopes Identified in Criti-cal Regions of Dengue Virus Nonstructural 1 Protein in Mouse Vaccination and Natural Human Infec-tions. J Immunol. 2017; 198(10):4025–35.https://doi.org/10.4049/jimmunol.1700029PMID:28381638 52. Lai YC, Chuang YC, Liu CC, Ho TS, Lin YS, Anderson R, et al. Antibodies Against Modified NS1 Wing

Domain Peptide Protect Against Dengue Virus Infection. Sci Rep. 2017; 7(1):6975.https://doi.org/10.

1038/s41598-017-07308-3PMID:28765561

53. Kerschbaumer RJ, Rieger M, Volkel D, Le Roy D, Roger T, Garbaraviciene J, et al. Neutralization of macrophage migration inhibitory factor (MIF) by fully human antibodies correlates with their specificity for the beta-sheet structure of MIF. J Biol Chem. 2012; 287(10):7446–55.https://doi.org/10.1074/jbc.

M111.329664PMID:22238348

54. Tsai HP, Tsai YY, Lin IT, Kuo PH, Chang KC, Chen JC, et al. Validation and Application of a Commer-cial Quantitative Real-Time Reverse Transcriptase-PCR Assay in Investigation of a Large Dengue Virus Outbreak in Southern Taiwan. PLoS Negl Trop Dis. 2016; 10(10):e0005036.https://doi.org/10.

1371/journal.pntd.0005036PMID:27732593

55. Benelli U, Bocci G, Danesi R, Lepri A, Bernardini N, Bianchi F, et al. The heparan sulfate suleparoide inhibits rat corneal angiogenesis and in vitro neovascularization. Exp Eye Res. 1998; 67(2):133–42.

https://doi.org/10.1006/exer.1998.0512PMID:9733580

56. van den Biggelaar M, Hernandez-Fernaud JR, van den Eshof BL, Neilson LJ, Meijer AB, Mertens K, et al. Quantitative phosphoproteomics unveils temporal dynamics of thrombin signaling in human endo-thelial cells. Blood. 2014; 123(12):e22–36.https://doi.org/10.1182/blood-2013-12-546036PMID:

24501219

57. Lee JS, Hmama Z, Mui A, Reiner NE. Stable gene silencing in human monocytic cell lines using lenti-viral-delivered small interference RNA. Silencing of the p110alpha isoform of phosphoinositide 3-kinase reveals differential regulation of adherence induced by 1alpha,25-dihydroxycholecalciferol and bacterial lipopolysaccharide. J Biol Chem. 2004; 279(10):9379–88.https://doi.org/10.1074/jbc.M310638200 PMID:14672955

58. Hong-Ru Chen T-MY. In vitro Assays for Measuring Endothelial Permeability by Transwells and Electri-cal Impedance Systems. Bio Protoc. 2017; 7(9):e2273.

59. Sato H, Takino T, Okada Y, Cao J, Shinagawa A, Yamamoto E, et al. A matrix metalloproteinase expressed on the surface of invasive tumour cells. Nature. 1994; 370(6484):61–5.https://doi.org/10.

1038/370061a0PMID:8015608

日期:107 年 4 月 2 日

一、 參加會議經過

2018 年亞太生命科學及生物工程會議於 3/27-3/29 在日本京都舉行,我於 3/26 早上搭長榮 航空飛機由高雄出發,中午抵達大阪關西機場,入關後轉日本 JR 火車下午抵達京都,3/27 到會場報到,3/28-29 兩天早上聆聽大會安排的特別演講,下午海報展示。這次大會因為在 日本地理位置和台灣較近,而且同一場地還有資訊工程方面的會議在舉行,所以有有許多 台灣各個學校來自不同領域的學者和學生參加,大家也很難得的可以在這幾天跨領域的交 流互動。

計畫編號 A1071-0500 (編號: 107-2321-B-006 -001 -)

計畫名稱 發展新的登革病毒小鼠模式來探討致病機制與治療策略-登革病毒非

相關文件