Differential host immune responses in BALB c and C57BL 6 mice to burkholderia pseudomallei infection

166 298 0
Differential host immune responses in BALB c and C57BL 6 mice to burkholderia pseudomallei infection

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

DIFFERENTIAL HOST IMMUNE RESPONSES IN BALB/C AND C57BL/6 MICE TO BURKHOLDERIA PSEUDOMALLEI INFECTION KOO GHEE CHONG (B SC (HONS.), NUS) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF BIOCHEMISTRY NATIONAL UNIVERSITY OF SINGAPORE 2006 i Acknowledgements I really would not have finished this project if not for the many people who have been always there to inspire, love, guide, teach, support and motivate me during the past few years of challenging journey that I have gone through I would like to express my heartfelt gratitude to my supervisor Dr Gan Yunn Hwen, for her constant guidance, advice, patience and understanding I would also like to thanks Ms Lim Soh Chan for her constant help, technical support, advice, prayers, comments, comforting and encouraging words I am grateful to all my friends and labmates, especially Sun Guang Wen, Lee Tien Huat, Liu Boping, Xie Chao, Ong Yong Mei, Chen Kang, Chan Ying Ying, Cheryl Lee, Justin Lee, Ng Kian Hong, Hu Huan, Wong Kok Lun, Ng Hui Ling, Hii Chung Shii, Chen Yahua, Tang Soong Yew, Lim Kok Siong, Lu Guodong, Dawn Koh, Chan Mann Yin, Bian Hao Sheng, Fei Wei Hua, Low Choon Pei, Clarence Kho, Jowett Wong, Lim Chih Gang, Joshua Lau and many others for their assistance, encouragement and friendship To my parents and family members for their love, support and understanding all these years To brothers and sisters from my church for constantly keeping me in prayers Most of all, I am eternally thankful to God for sustaining me and bringing me through all the difficulties, for constantly staying by my side and being the source of my strength, inspiration and hope ii Table of Contents Contents Page Title Page…………………………………………………………………………… .i Acknowledgements……………………………………………………………………….ii Table of Contents……………………………………………………………………… iii Summary………………………………………………………………………………….ix List of Tables…………………………………………………………………………… x List of Figures……………………………………………………………………………vii List of Abbreviations……………………………………………………………………viii Chapter Introduction………………………………………………………………….1 1.1 Melioidosis………………………………………………………………………… 1.2 Prevalence and epidemiology……………………………………………………… 1.3 Melioidosis in Singapore…………………………………………………………… 1.4 Modes of transmission…………………………………………………………… 1.5 Clinical manisfestations…………………………………………………………… 1.6 Diagnosis…………………………………………………………………………… 1.6.1 Identification of Burkholderia pseudomallei……………………………………7 1.6.2 Serological tests…………………………………………………………………8 1.6.3 Molecular identification techniques…………………………………………… 1.7 Management and treatment … …………………………………………………… 1.8 Bacterial pathogensis……………………………………………………………… 11 1.8.1 Gene and genome………………………………………………………………11 1.8.2 Virulence factors……………………………………………………………….11 iii 1.9 Animals model for melioidosis…………………………………………………… 13 1.10 Role of cytokines in immunity………………………………………………… ….15 1.11 Objectives of present study…………………………………………………………18 Chapter Characterization of B pseudomallei mucosal infection model…… … 20 2.1 Introduction…………………………………………………………………………21 2.2 Materials and methods………………………………………………………… 23 2.2.1 Animals……………………………………………………………………… 23 2.2.2 Bacteria ………………………………………………………………………23 2.2.3 LD50 determination… ……………………………………………………… 24 2.2.4 Infection of mice and preparation of organs………………………………… 24 2.2.5 RNA isolation and reverse transcription-polymerase chain reaction (RT-PCR)………………………………………………………………………25 2.2.6 Determination of cytokines concentration by ELISA…………………………26 2.2.7 Flow cytometric analysis………………………………………………………26 2.2.8 Tissue pathology……………………………………………………………….27 2.2.9 Statistical analysis…………………………………………………………… 27 2.3 Results… ………………………………………………………………………….28 2.3.1 LD50 of Burkholderia pseudomallei in BALB/c and C57BL/6 mice………….28 2.3.2 Bacterial loads in the infected organs……………………………… ……… 28 2.3.3 Cytokine Responses……………………………………………………………31 2.3.4 Kinetics of IFN-γ response upon low dose infection………………………… 32 2.3.5 IFN-γ response and bacterial loads in the blood of C57BL/6 mice upon high dose infection………………………………………………………………… 34 iv 2.3.6 Bacterial loads in organs of high dose and low dose challenged C57BL/6 mice…………………………………………………………………………….34 2.3.7 Pathology of infected organs………………………………………………… 36 2.3.8 Infiltration of immune cells during infection with B peudomallei……………39 2.4 Discussion ……………………………………………………………………… 41 Chapter Humoral immune response to Burkholderia pseudomallei………………47 3.1 Introduction………………………………………………………………………… 48 3.2 Materials and methods……………………………………………………………….53 3.2.1 Mice……………………………………………………………………………… 53 3.2.2 Bacteria…………………………………………………………………………….53 3.2.3 Expression of recombinant flagellin (r-FliC)……………… …………………….53 3.2.4 Purification of recombinant protein……………………………………………… 54 3.2.4.1 Preparation of cleared lysate under denaturing condition………………… 54 3.2.4.2 Purification of protein under denaturing condition………………………….55 3.2.4.3 Analysis of purified flagellin by SDS-PAGE……………………………….56 3.2.4.4 Determination of protein concentration by Bradford method …………… 56 3.2.4.5 Dialysis and concentration of purified flagellin…………………………… 56 3.2.5 Immunization of BALB/c mice …………………………………………… 57 3.2.6 Infection with live bacteria and protection study…………………………….57 3.2.7 Collection of serum………………………………………………………… 57 3.2.8 Detection of antigen specific antibodies by ELISA … …………………… 58 3.2.8.1 Flagellin specific antibodies.…………………………………………58 3.2.8.2 Burkholderia pseudomallei specific antibodies …………………… 58 v 3.2.9 Western blot………………………………………………………………… 59 3.3 Results……………………………………………………………………………… 60 3.3.1 Purication of flagellin protein (r-FliC)……………… …………………… 60 3.3.2 Immunization and protection study………………………………………… 61 3.3.3 Flagellin specific antibody responses (total IgG and IgG subclasses)……… 64 3.3.4 Western blot analysis of anti-sera ………………………………………… 66 3.3.5 Specificity of antibodies against whole bacteria…………………………… 66 3.3.6 Low dose-high dose infection in C57BL/6 mice …………………………….68 3.3.6.1 Kinetics of the antibody responses (total IgG, IgG1 and IgG2a)……… 68 3.3.6.2 Flagellin specific antibodies (total IgG, IgG1 and IgG2a)……………….70 3.4 Discussion……………………………………………………………………………71 Chapter Characterization of IFN-γ response in vitro………………………………77 4.1 Introduction… …………………………………………………………………….78 4.2 Materials and methods………………………………………………………………81 4.2.1 Mice……………………………………………………………………………81 4.2.2 Bacteria……………………………………………………………………… 81 4.2.3 Infection with Burkholderia pseudomallei…………………………………….81 4.2.4 Preparation and stimulation of splenocytes in vitro………………………… 82 4.2.5 Cell viability determination……………………………………………………82 4.2.6 Bacterial load determination…………………………… ……………….……83 4.2.7 Magnetic cell separation for cell-type purification………………………… 83 4.2.8 Cytokine determination by ELISA…………………………………………….84 4.2.9 Flow Cytometric analysis…………………………………………………… 84 vi 4.2.10 Intracellular cytokine staining ……………… ………………………………85 4.2.11 Cytokine capturing assay for IFN-γ………………………………………… 85 4.2.12 Isolation of human neutrophils……………………………………………….86 4.2.13 Statistical analyses……………………………………………………………86 4.3 Results……………………………………………………………………………….87 4.3.1 IFN-γ response in the splenocytes stimulated with B pseudomallei in vitro….87 4.3.2 Bacterial loads of the infected splenocytes……………………… ………… 88 4.3.3 Cell viability of infected splenocytes from BALB/c and C57BL/6 mice…… 90 4.3.4 Cytokine profiles in BALB/c and C57BL/6 mice after B pseudomallei infection……………………………………………………………………… 94 4.3.5 The effects of IL-12, IL-18 and IL-10 neutralizing antibodies on IFN-γ response…………………………………………………………………97 4.3.6 Cell types produce IFN-γ in response to bacteria in BALB/c and C57BL/6 mice……………………………………………………… ……… 97 4.3.6.1 Intracellular cytokine staining…………………………………………… 97 4.3.6.2 Cytokine capturing assay………… …………………………………… 97 4.3.6.3 Effect of cell depletion on the production of IFN-γ……………………… 99 4.3.7 Gr-1 expression populations in BALB/c and C57BL/6 splenocytes…………103 4.3.8 Role of T cells in IFN-γ response ……………………………………………107 4.3.9 Role of LPS in IFN-γ response to B pseudomallei………………………… 108 4.3.9.1 The effect of polymyxin B treatment on IFN-γ response to B pseudomallei………………………………………………………………107 4.3.9.2 TLR-4 signaling pathway is required for IFN-γ response to heat-killed Bacteria but dispensable for the IFN-γ response to live bacteria……… 107 3.10 IFN-γ response in human neutrophils……………………………………….110 vii 4.4 Discussion……………………………………………………………………….…110 Chapter Conclusion and future studies……………………………………………116 References……………… … ……………………………………………………… 122 Appendix I: Recipes …………………………………… ………………………… 148 Appendix II: Publications…………………………….…………………………… 151 viii Summary Burkholderia pseudomallei is the causative agent of melioidosis—an endemic disease in the Southeast Asia and Northern Australia Infections can result in clinical manifestations ranging from asymptomatic, chronic suppurative infection to potentially fatal septicemia The aim of this project is to study the host responses and factors which contribute to resistance or susceptibility in two strains of mice showing differential responses to B pseudomallei infection We found that BALB/c mice were highly susceptible to low dose intranasal B pseudomallei infection They developed acute disease and died within weeks, whereas C57BL/6 mice were relatively resistant Susceptibility of BALB/c mice correlates with high bacterial loads in the lung and spleen, infiltration of leukocytes (especially neutrophils), tissue pathology (in the lung and spleen), and hyper-inflammation A transient hyperproduction of IFN-γ was found at 48h post-infection in the serum of BALB/c, but not in the relatively resistant C57BL/6 mice C57BL/6 did not show a complete resistance to infection, as high dose B pseudomallei infected C57BL/6 mice died of septicemia resembling the characteristics of low dose infected BALB/c mice C57BL/6 mice which survived after an initial infection were found to be resistant to subsequent low dose or high dose challenge Resistance of low dose immunized C57BL/6 mice to high dose infection correlated to high serum IgG2a, which was indirect evidence of IFN-γ induced cell-mediated immunity, and with a low IgG1 response In contrast, BALB/c mice immunized with recombinant flagellin (r-FliC) induced high serum IgG1, which did not confer protection against B pseudomallei infecton However, some low dose infected and all high dose infected C57BL/6 mice eventually developed splenic abscesses and died at much later time points ix In vitro study showed that naïve BALB/c splenocytes produced higher IFN-γ than naïve C57BL/6 splenocytes in response to B pseudomallei infection, mirroring what was seen in vivo Splenocytes from BALB/c mice contained higher number of intracellular bacteria than C57BL/6, which could explain why they made more IFN-γ The IFN-γ response was IL-12 dependent and IL-18 could have a synergistic effect, while IL-10 ameliorated the IFN-γ response There was no obvious difference in the cell types making IFN-γ in BALB/c and C57BL/6 splenocytes In both strains of mice, Gr-1 positive cells (particularly the CD8+ T lymphocytes and DX5+ NK cells), and CD4+ cells were the major producers of the IFN-γ in response to B pseudomallei BALB/c splenocytes also contained higher numbers of Gr-1intermediate expressing NK and CD8+ cells Using splenocytes from nude mice, the redundancy of T cells in IFN-γ response of splenocytes to B pseudomallei was observed TLR-4 signaling was found to be essential for IFN-γ response of splenocytes to the heat-killed bacteria, but not for the response to live bacteria Besides hyperproduction of IFN-γ, BALB/c mice produced more TNF-α, IL-1β, IL-6, higher basal IL-18, and more anti-inflammatory cytokine (IL-10) compared to the splenocytes of C57BL/6 mice This study suggests that resistance to infection lies in the innate ability to control the bacterial growth that allows subsequent development of adaptive immunity Despite the importance of IFN-γ for host resistance, uncontrolled production of it could cause immunopathology leading to the fatal outcome x Lagasse, E and I L Weissman 1996 Flow cytometric identification of murine neutrophils and monocytes J Immunol Methods 197(1-2): 139-150 Lauw, F N., A J Simpson, J M Prins, S J van Deventer, W Chaowagul, N J White, and T van der Poll 2000 The CXC chemokines gamma interferon (IFN-gamma)-inducible protein 10 and monokine induced by IFN-gamma are released during severe melioidosis Infect Immun 68:3888-3893 Leakey, A K., G C Ulett, and R G Hirst 1998 BALB/c and C57Bl/6 mice infected with virulent Burkholderia pseudomallei provide contrasting animal models for the acute and chronic forms of human melioidosis Microb Pathog 24(5): 269-75 Lee, M A and Y Liu 2000 Sequencing and characterization of a novel serine metalloprotease from Burkholderia pseudomallei FEMS Microbiol Lett 192(1): 67-72 Leelarasamee, A., and S Bovornkitti 1989 Melioidosis: review and update Rev Infect Dis 11(3): 413-25 Leelarasamee, A 2004 Recent development in melioidosis Curr Opin Infect Dis 17(2): 131-6 Leelarasamee, A., C Chupaprawan, M Chenchittikul, and S Udompanthurat 2004 Etiologies of acute undifferentiated febrile illness in Thailand J Med Assoc Thai 87(5): 46472 Lertmemongkolchai, G., G Cai, C A Hunter, and G J Bancroft 2001 Bystander activation of CD8+ T cells contributes to the rapid production of IFN-γ in response to bacterial pathogens J Immunol 166:1097-1105 137 Liu, B., G C Koo, E H Yap, K L Chua and Y H Gan 2002 Model of differential susceptibility to mucosal Burkholderia pseudomallei infection Infect Immun 70(2): 504-11 Lowe, P., Engler C., and R Norton 2002 Comparison of automated and nonautomated systems for identification of Burkholderia pseudomallei J Clin Microbiol 40:4625-7 Marchant, A., C Bruyns, P Vandenabeele, M Ducarme, C Gerard, A Delvaux, D De Groote, D Abramowicz, T Velu, and M Goldman 1994 Interleukin-10 controls interferon-gamma and tumor necrosis factor production during experimental endotoxemia Eur J Immunol 24:1167-1171 Martin, R M., J L Brady, and A M Lew 1998 The need for IgG2c specific antiserum when isotyping antibodies from C57BL/6 and NOD mice J Immunol Meth 212(2): 187-192 Matsuzaki, J., T Tsuji, K Chamoto, T Takeshima, F Sendo, and T Nishimura 2003 Successful elimination of memory-type CD8+ T cell subsets by the administration of anti-Gr1 monoclonal antibody in vivo Cell Immunol 224:98-105 Mays, E E., and E A Ricketts 1975 Melioidosis: recrudescence associated with bronchogenic carcinoma twenty-six years following initial geographic exposure Chest 68(2): 261-3 McKenzie, S J., and J F Halsey 1984 Cholera toxin B subunit as a carrier protein to stimulate a mucosal immune response J Immunol 133(4): 1818-24 Miralles, I S., C Maciel Mdo, M R Angelo, M M Gondini, L H Frota, C M dos Reis, and E Hofer 2004 Burkholderia pseudomallei: a case report of a human infection in Ceara, Brazil Rev Inst Med Trop Sao Paulo 46(1): 51-4 138 Moore, R A., D DeShazer, S Reckseidler, A Weissman, and D E Woods 1999 Effluxmediated aminoglycoside and macrolide resistance in Burkholderia pseudomallei Antimicrob Agents Chemother 43(3): 465-70 Munckhof, W J., M J Mayo, I Scott, and B J Currie 2001 Fatal human melioidosis acquired in a subtropical Australian city Am J Trop Med Hyg 65(4): 325-8 Nakamura, S., T Otani, Y Ijiri, R Motoda, M Kurimoto, and K Orita 2000 IFNgamma-dependent and -independent mechanisms in adverse effects caused by concomitant administration of IL-18 and IL-12 J Immunol 164:3330-3336 Nakano, H., M Yanagita, and M D Gunn 2001 CD11c(+)B220(+)Gr-1(+) cells in mouse lymph nodes and spleen display characteristics of plasmacytoid dendritic cells J Exp Med 194:1171-1178 Nelson, M., J L Prior, M S Lever, H E Jones, T P Atkins, and R W Titball 2004 Evaluation of lipopolysaccharide and capsular polysaccharide as subunit vaccines against experimental melioidosis J Med Microbiol 53 (12): 1177-82 Ngauy, V., Y Lemeshev, L Sadkowski, and G Crawford 2005 Cutaneous melioidosis in a man who was taken as a prisoner of war by the Japanese during World War II J Clin Microbiol 43(2): 970-2 Olive C., Clair T., Yarwood P., and Good M F 2002 Protection of mice from group A streptococcal infection by intranasal immunisation with a peptide vaccine that contains a conserved M protein B cell epitope and lacks a T cell autoepitope Vaccine 20(21-22): 281625 139 Orellana, C 2004 Melioidosis strikes Singapore Lancet Infect Dis 4(11): 655 Pickup, J C 2004 Inflammation and activated innate immunity in the pathogenesis of type diabetes Diabetes Care 27:813-23 Piggott, J A., and L Hochholzer 1970 Human melioidosis A histopathologic study of acute and chronic melioidosis Arch Pathol 90(2): 101-11 Pruksachartvuthi, S., N Aswapokee, and K Thankerngpol 1990 Survival of Pseudomonas pseudomallei in human phagocytes J Med Microbiol 31(2): 109-14 Puthucheary, S D., editor 2002 Human melioidosis Singapore: University of Singapore Press 7-46 Raja, N S., M Z Ahmed, and N N Singh 2005 Melioidosis: An emerging infectious disease J Postgrad Med 51(2): 140-5 Reckseidler-Zenteno, S L., R DeVinney and D E Woods 2005 The capsular polysaccharide of Burkholderia pseudomallei contributes to survival in serum by reducing complement factor C3b deposition Infect Immun 73(2): 1106-15 Reed, L.J., and H Muench 1938 A simple method for estimating fifty per cent end point Am J Hyg 27:493-497 Richard, A G., T J Kindt, B A Osborne Kuby Immunology 2000 4th edition New York: W.H Freeman 140 Salkowski, C A., K E Thomas, M J Cody, and S N Vogel 2000 Impaired IFN-gamma production in IFN regulatory factor-1 knockout mice during endotoxemia is secondary to a loss of both IL-12 and IL-12 receptor expression J Immunol 165:3970-3977 Santanirand, P., V S Harley, D A Dance, B S Drasar, and G J Bancroft 1999 Obligatory role of gamma interferon for host survival in a murine model of infection with Burkholderia pseudomallei Infect Immun 67(7): 3593-600 Scharton, T M., and P Scott 1993 Natural killer cells are a source of interferon that drives differentiation of CD4+ T cell subsets and induces early resistance to Leishmania major in mice J Exp Med 178:567-577 Schleicher, S., A Hesse, and C Bogdan 2005 Minute numbers of contaminant CD8+ T cells or CD11b+CD11c+NK cells are the source of IFN-γ in IL-12/IL-18-stimulated mouse macrophage populations Blood 105:1319-1328 Schroder, M., C Meisel, K Buhl, N Profanter, N Sievert, H D Volk, and G Grutz 2003 Different Modes of IL-10 and TGF-ß to inhibit cytokine-dependent IFN- production: consequences for reversal of lipopolysaccharide desensitization J Immunol 170:5260-5267 Scott, P 1991 IFN- modulates the early development of Th1 and Th2 responses in a murine model of cutaneous leishmaniasis J Immunol 147:3149-3155 Sexton, M M., A L Jones, W Chaowagul, and D E Woods 1994 Purification and characterization of a protease from Pseudomonas pseudomallei Can J Microbiol 40(11): 90310 Simpson, A J H., M D Smith, G J Weverling, Y Suputtamongkol, B J Angus, W Chaowagul, N J White, and J M Prins 2000 Prognostic value of cytokine 141 concentrations (tumor necrosis factor-alpha, interleukin-6, and interleukin-10) and clinical parameters in severe melioidosis J Infect Dis 181:621-625 Shtrichman, R., and C Samuel 2001 The role of gamma interferon in antimicrobial immunity Curr Opin Microbiol 4:251-259 Smith, C J., J C Allen, M Noor Embi, O Othman, N Razak, and G Ismail 1987 Human melioidosis: an emerging medical problem MIRCEN J Appl Microbiol Biotechnol 3:343-366 So, S.Y 1986 Melioidosis-an endemic problem in Southeast Asia Medicine Digest Asia 4:19-23 Song, Y., C Xie, Y M Ong, Y H Gan, and K L Chua 2005 The BpsIR quorumsensing system of Burkholderia pseudomallei J Bacteriol 187(2): 785-90 Spellberg, B., and Edwards J E., Jr 2001 Type 1/Type immunity in infectious diseases Clin Infect Dis 32(1):76-102 Sprague, L D., and Neubauer H 2004 Melioidosis in animals: a review on epizootiology, diagnosis and clinical presentations J Vet Med 51:305-20 Stanton, A T., and W Fletcher 1921 Melioidosis, a new disease of the tropics Trans Fourth Congr Far East Assoc Trop Med 2:196-198 Stephens-Romero, S D., A J Mednick, and M Feldmesser 2005 The pathogenesis of fatal outcome in murine pulmonary aspergillosis depends on the neutrophil depletion strategy Infect Immun 73:114-125 142 Stevens, M P., M W Wood, L A Taylor, P Monaghan, P Hawes, P W Jones, T S Wallis, and E E Galyov 2002 An Inv/Mxi-Spa-like type III protein secretion system in Burkholderia pseudomallei modulates intracellular behaviour of the pathogen Mol Microbiol 46(3): 649-59 Stevens, M P., and E E Galyov 2004 Exploitation of host cells by Burkholderia pseudomallei Int J Med Microbiol 293(7-8): 549-55 Strauss, J M., D W Ellison, E Gan, S Jason, J L Marcarelli, and G Rapmund 1969 Melioidosis in Malaysia IV Intensive ecological study of Carey Island, Selangor, for Pseudomonas pseudomallei Med J Malaya 24(2): 94-100 Strindelius, L., Filler M., and Sjoholm I 2004 Mucosal immunization with purified flagellin from Salmonella induces systemic and mucosal immune responses in C3H/HeJ mice Vaccine 9;22(27-28):3797-808 Sultan, F., L L Jin, M G Jobling, R K Holmes, and S L Stanley 1998 Mucosal immunogenicity of a holotoxin-like molecule containing the serine-rich Entamoeba histolytica protein (SREHP) fused to the A2 domain of cholera toxin Infect Immun 66(2):462-8 Sun, G W., J Lu, S Pervaiz, W P Cao, and Y H Gan 2005 Caspase-1 dependent macrophage death induced by Burkholderia pseudomallei Cell Microbiol 7(10): 1447-58 Suparak, S., W Kespichayawattana, A Haque, A Easton, S Damnin, G Lertmemongkolchai, G J Bancroft, and S Korbsrisate 2005 Multinucleated giant cell formation and apoptosis in infected host cells is mediated by Burkholderia pseudomallei type III secretion protein BipB J Bacteriol 187(18): 6556-60 143 Suputtamongkol, Y., D A Dance, W Chaowagul, Y Wattanagoon, V Wuthiekanun, and N J White 1991 Amoxycillin-clavulanic acid treatment of melioidosis Trans R Soc Trop Med Hyg 85(5): 672-5 Suputtamongkol, Y., D Kwiatkowski, D A Dance, W Chaowagul, and N J White 1992 Tumor necrosis factor in septicemic melioidosis J Infect Dis 165(3): 561-4 Thomas, A D., J Forbes-Faulkner, and M Parker 1979 Isolation of Pseudomonas pseudomallei from clay layers at defined depths Am J Epidemiol 110(4): 515-21 Thummakul, T., H Wilde, and T Tantawichien 1999 Melioidosis, an environmental and occupational hazard in Thailand Mil Med 164(9): 658-62 Tong, S., S Yang, Z Lu, and W He 1996 Laboratory investigation of ecological factors influencing the environmental presence of Burkholderia pseudomallei Microbiol Immunol 40(6): 451-3 Towbin, H., T Staehelin, and J Gordon 1979 Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications Proc Natl Acad Sci U S A 76(9):4350-4 Tribuddharat, C., R A Moore, P Baker, and D E Woods 2003 Burkholderia pseudomallei class a beta-lactamase mutations that confer selective resistance against ceftazidime or clavulanic acid inhibition Antimicrob Agents Chemother 47(7): 2082-7 Trinchieri, G 2003 Interleukin-12 and the regulation of innate resistance and adaptive immunity Nat Rev Immunol 3:133-146 144 Ulett, G C., N Ketheesan, and R G Hirst 1998 Macrophage-lymphocyte interactions mediate anti-Burkholderia pseudomallei activity FEMS Immunol Med Microbiol 21(4): 2836 Ulett, G C., N Ketheesan, and R G Hirst 2000 Cytokine gene expression in innately susceptible BALB/c mice and relatively resistant C57BL/6 mice during infection with virulent Burkholderia pseudomallei Infect Immun 68(4): 2034-42 Ulett, G C., J T Labrooy, B J Currie, J L Barnes, and N Ketheesan 2005 A model of immunity to Burkholderia pseudomallei: unique responses following immunization and acute lethal infection Microbes Infect 7(11-12): 1263-75 Utaisincharoen, P., N Anuntagool, K Limposuwan, P Chaisuriya, and S Sirisinha 2003 Involvement of beta interferon in enhancing inducible nitric oxide synthase production and antimicrobial activity of Burkholderia pseudomallei-infected macrophages Infect Immun 71(6): 3053-7 Utaisincharoen, P., N Anuntagool, S Arjcharoen, K Limposuwan, P Chaisuriya, and S Sirisinha 2004 Induction of iNOS expression and antimicrobial activity by interferon (IFN)-beta is distinct from IFN-gamma in Burkholderia pseudomallei-infected mouse macrophages Clin Exp Immunol 136(2): 277-83 Vadivellu, J., S D Puthucheary, G.S Gendeh, and N Parasakthi 1995 Serodiagnosis of melioidosis in Malaysia Singpore Med J 36:299-302 Varma, T., T Toliver-Kinsky, C Lin, A Koutrouvelis, J Nichols, and E R Sherwood 2001 Cellular mechanisms causing suppressed gamma interferon secretion in endotoxintolerant mice Infect Immun 69:5249-5263 145 Venuprasad, K., P P Banerjee, S Chattopadhyay, S Sharma, S Pal, P B Parab, D Mitra, and B Saha 2002 Human neutrophil-expressed CD28 interacts with macrophage B7 to induce phosphatidylinositol 3-kinase-dependent IFN-γ secretion and restriction of Leishmania growth J Immunol 169:920-928 Warawa, J and D E Woods 2002 Melioidosis vaccines Expert Rev Vaccines 1(4): 47782 Wellen, K E., G S Hotamisligil 2005 Inflammation, stress, and diabetes J Clin Invest 115(5): 1111-1119 White, N J 2003 Melioidosis Lancet 361:1715-1722 Whitmore, A., Krishnaswami C S 1912 An account of the discovery of a hitherto underscribed infective disease occurring among the population of Rangoon Indian Medical Gazette 47:262-267 Whitmore, A 1913 An account of a glanders-like disease occurring in Rangoon J Hyg 13:1-34 Wong, K T., S D Puthucheary, and J Vadivelu 1995 The histopathology of human melioidosis Histopathology 26(1): 51-5 Wong, K T., S D Puthucheary, and J Vadivelu 1995 The histopathology of human melioidosis Histopathology 26(1): 51-5 Wongratanacheewin, S., W Kespichayawattana, P Intachote, S Pichyangkul, R W Sermswan, A M Krieg, and S Sirisinha 2004 Immunostimulatory CpG oligodeoxynucleotide confers protection in a murine model of infection with Burkholderia pseudomallei Infect Immun 72(8): 4494-502 146 Woods, D E., A L Jones, and P J Hill 1993 Interaction of insulin with Pseudomonas pseudomallei Infect Immun 61(10): 4045-50 Wuthiekanun, V., M D Smith, and N J White 1995 Survival of Burkholderia pseudomallei in the absence of nutrients Trans R Soc Trop Med Hyg 89(5): 491 Wysocka, M., S Robertson, H Riemann, J Caamano, C Hunter, A Mackiewicz, L J Montaner, G Trinchieri, and C L Karp 2001 IL-12 suppression during experimental endotoxin tolerance: dendritic cell loss and macrophage hyporesponsiveness J Immunol 166:7504-7513 Yabuuchi, E., Y Kosako, H Oyaizu, I Yano, H Hotta, Y Hashimoto, T Ezaki, and M Arakawa 1992 Proposal of Burkholderia gen nov and transfer of seven species of the genus Pseudomonas homology group II to the new genus, with the type species Burkholderia cepacia (Palleroni and Holmes 1981) comb nov Microbiol Immunol 36(12): 1251-75 Yahata, S N., K Santa, A Ohta, Y Ohmi, S Habu, and T Nishimura 1996 Functional characterization of NK1.1+ Ly-6C+ cells Immunol Lett 54:5-9 Yang, H., C D Kooi, and P A Sokol 1993 Ability of Pseudomonas pseudomallei malleobactin to acquire transferrin-bound, lactoferrin-bound, and cell-derived iron Infect Immun 61(2): 656-62 Yeaman, G R., J E Collins, J K Currie, P M Guyre, C R Wira, and M W Fanger 1998 IFN-γ is produced by polymorphonuclear neutrophils in human uterine endometrium and by cultured peripheral blood polymorphonuclear neutrophils J Immunol 160:5145-5153 147 Appendix I: Recipes 1.1 Media for bacteria culture Tryptic Soy Agar (TSA): 15.0 g/L bacto-tryptone, g/L soya peptone, 5.0 g/L NaCl, 15.0 g/L agar, pH 7.3 Ashdown slective medium: 10 g/L TSA, % glycerol (w/v), mg/L crystal violet, 50 mg/L neutral red and mg/L gentamicin LB broth: 10 g/L bacto-tryptone, g/L bacto yeast extract, g/L NaCl, pH 7.0 LB agar: LB medium containing 15 g/L bactor agar • All the media used for bacteria culture were sterilized by autoclaving at 121 °C, 15 PSI for 15 1.2 Supplements Kanamycin stock solution: 25 mg/ml in H2O, sterile filter, store in aliquots at – 20 °C Ampicillin stock solution: 50 mg/ml in H2O, sterile filter, store in aliquots at – 20 °C Gentamicin stock solution: 100 mg / ml in H2O, sterile filter, store in aliquots at – 20 °C IPTG (1M): 238 mg/ml in H2O, sterile filter through 0.22 μm filter, store in aliquots at – 20 °C 1.3 Buffers for purification under denature conditions: Buffer B: 100mM NaH2PO4 , 10 mM Tris HCL, M Urea, pH 8.0 Buffer C: 100mM NaH2PO4 , 10 mM Tris HCL, 0.5 % Sodium deoxycholate, M Urea, pH 6.3 Buffer D: 100mM NaH2PO4 , 10 mM Tris HCL, M Urea, pH 5.9 Buffer E: 100mM NaH2PO4 , 10 mM Tris HCL, M Urea, pH 4.5 148 1.4 Reagents for SDS-PAGE Resolving Gel (8%) Gels Gel Solution D 10.5 ml ml ddH2O 4.2 ml ml Solution A 4.2 ml ml % SDD 2.1 ml ml 10% APS* 105 μl 50 μl TEMED* 10.5 μl μl Gels Gel Solution C 2.5 ml 1.25 ml ddH2O 1.5 ml 750 μl Solution A 0.5 ml 250 μl % SDD 0.5 ml 250 μl 10% APS* 12.5 μl 6.25 μl TEMED* 10 μl μl Stacking Gel (4%) Solution A (Bis-acrylamide): acrylamid 45 g, bis-acryalmide 1.2 g, H2O 100 ml Solution B (5x running buffer): 15.14 g Tris (125 mM), 72 g glycine (0.96 M), g SDS (0.5 %), top up with H2O to liter, pH 8.3 Solution C (Stacking buffer): 30.28 g Tris (0.25 M), top up with H2O to liter, pH 6.8 Solution D (Resolving buffer): 90.9 g Tris (0.75M), top up with top up with H2O to liter, pH 8.8 Solution F (Ammonium persulfate): 0.03 g APS in 300 μl H2O (10%) 149 SDS-PAGE running buffer: 25 mM Tris-base, 0.19 M glycine, 0.1 % (w/v) SDS, pH 8.3 Coomassie Blue Staining Fixing solution for SDS-PAGE: Glacial acetic acid: methanol: H2O = 10: 50: 40 Staining solution: 0.05 % Coomassie blue Glacial acetic acid: methanol: H2O = 10: 50: 40 Destaining solution: Glacial acetic acid: methanol: H2O = 7: 5: 88 1.5 Buffers for ELISA 10X PBS: % (w/v) NaCl, g/L 0.2 % (w/v) KCl, 0.2 g/L 1.44 % Na2HPO4, 1.44 g/L 0.24 % KH2PO4, 0.24 g/L Adjust to pH 7.4 Bicarbonate coating buffer: 1.59 g Na2CO3, 2.93 g NaHCO3, top up to L with H2O pH 9.5 Wash buffer PBST: 500 μl Tween 20 to liter 1×PBS Assay buffer (0.5 % BSA-PBST): 0.5 g of BSA was dissolved in 100 ml PBST 1.6 Buffer for flowcytometry staining Fixing buffer: % parafolmadehyde in x PBS Permeabilization/wash buffer: 0.1 % saponin- % FBS – x PBS 150 Appendix II: Publications Journal Papers Boping Liu, Ghee Chong Koo, Eu Hian Yap, Kim Lee Chua and Yunn-Hwen Gan A Model of differential Susceptibility to Mucosal Infection of Burkholderia pseudomallei Infect Immun 2002, 70(2): 504-511 Ghee Chong Koo, Yunn-Hwen Gan The innate interferon gamma response of BALB/c and C57BL/6 mice to in vitro to Burkholderia pseduomallei infection BMC Immunology 2006, 7:19 Poster presentations Ghee Chong Koo, Yunn-Hwen Gan 2004 The Roles of IFN-γ During Burkholderia pseudomallei Infection Poster 31 The 5th Combined Scientific Meeting, NUS (Merit Award) Ghee Chong Koo, Yunn-Hwen Gan 2004 Innate IFN-γ response to Burkholderia pseudomallei Poster 110 The 4th World Melioidosis Congress, Singapore Ghee Chong Koo, Yunn-Hwen Gan 2005 Innate IFN-γ response in mice showing differential susceptibility to Burkholderia pseudomallei infection Poster 209 The Keystone Symposia on Innate Immunity to Pathogens, Colorado, USA (Keystone Symposia Scholarship Winner) 151 ... resistant C5 7BL /6 mice C5 7BL /6 did not show a complete resistance to infection, as high dose B pseudomallei infected C5 7BL /6 mice died of septicemia resembling the characteristics of low dose infected... bacterial counts in C5 7BL /6 mice were decreased 12 h after infection in comparison to BALB/ c mice which suggests an innate immune mechanism against B pseudomallei in the early phase of infection contributing... Bacterial loads of the infected splenocytes……………………… ………… 88 4.3.3 Cell viability of infected splenocytes from BALB/ c and C5 7BL /6 mice? ??… 90 4.3.4 Cytokine profiles in BALB/ c and C5 7BL /6 mice

Ngày đăng: 14/09/2015, 10:39

Từ khóa liên quan

Mục lục

  • Content_9June2007a

  • Correction_Final

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan