用户名: 密码: 验证码:
QS系统VjbR在布鲁氏菌胞内生存中作用的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
布鲁氏菌病是一种危害严重的人兽共患病,在世界范围内有广泛流行,给人类健康和经济发展带来巨大损失。布鲁氏菌是一种胞内寄生菌,深入探讨其独特的胞内生存机制和毒力因素,对于布鲁氏菌致病机制的理解、新型疫苗的研发以及布鲁氏菌病的临床治疗等具有重要意义。
     布鲁氏菌有很多感应和处理环境信号的调控系统及毒力因子,如QS密度感应系统。当布鲁氏菌侵入宿主细胞时,QS系统可以调控布鲁氏菌各种靶基因以适应胞内各种不利的环境信号,从而增强抵抗巨噬细胞杀伤的能力。布鲁氏菌QS系统有两个调控蛋白:VjbR和BlxR。目前对BlxR的毒力表型以及调控机制已经进行了详细的研究,但是对VjbR的调控机制尚不完全清楚。另外,本实验室前期研究中发现,T4SS的virB操纵子对vjbR有正调控作用,且virB是细菌重要的毒力基因,可增强布鲁氏菌胞内存活能力。同时已有文献报道virB和vjbR关系密切,相互影响。那么vjbR是否与细菌毒力表型相关?是否可以调控virB从而影响布鲁氏菌胞内生存?vjbR的调控机制是怎样的呢?这些问题的回答,有利于解释和深入探讨布鲁氏菌胞内生存机制和致病机制。
     为研究布鲁氏菌QS系统调控蛋白VjbR与布鲁氏菌毒力和胞内生存的关系,阐明其调控机制,本研究首先构建vjbR基因缺失突变株和互补株。通过对突变株、互补株和野生株生长曲线的观察,发现野生株的生长速度较突变株和互补株快,提示vjbR可能通过调控特定基因而正调控布鲁氏菌的生长。胞内存活及小鼠体内生存等表型实验显示,vjbR突变株虽然可以入侵巨噬细胞并在小鼠的脾脏和肝脏细胞内生存,但生存能力减弱,同时,我们在体外模拟了巨噬细胞内的多种胁迫条件,如高盐、高渗、酸、热休克和氧压力等,与野生株和互补株相比,突变株在这些刺激条件下的生存率都有不同程度的降低,表明vjbR对于布鲁氏菌适应胞内恶性环境、抵抗环境压力、建立慢性感染是必需的。与此同时,我们又研制了布鲁氏菌全基因组DNA芯片。利用全基因组芯片,比较分析了vjbR突变株与16M野生株转录谱差异,鉴定vjbR调控的靶基因,并对这些基因的功能进行详细阐述,为QS调控网络的研究提供靶标基因。本研究中,我们对布鲁氏菌QS系统在毒力中发挥的作用及其调控机制进行了详细的探讨,为布鲁氏菌治病机制的研究提供大量有价值的信息。
Brucellosis, which is caused by Brucella, is one of the most important bacterial zoonoses endemic in the world, especially in developing countries. These pathogens can affect a broad range of mammals and cause serious economic losses. The unique pathogenicity is the research focus on this pathogen. Brucella is an intracellular bacterium, and the virulence depends upon its ability to survive and replicate within host cells. Quorum sensing is a cell-density dependent global regulation system, which is used for cell to cell communication mediated by the signal molecule, N-acyl-homoserine lactone (AHL). However, the molecular mechanisms underlying regulation remain unclear.
     In the present study, homologous recombination was used to construct the vjbR deletion mutant, and then, the survival in macrophages and in mice of wild-type and mutant were compared to define related phenotypes affected by vjbR. The transcriptome of the mutant was compared to the wild-type strain to define the differentially transcribed genes, which may be regulated by vjbR. By combining the function information of the differentially transcribed genes and the alterations of the phenotypes in the mutant, the regulation mechanism was putatively explained.
     To define the vjbR related phenotypes and the target genes regulated by vjbR, the vjbR mutant and its complementary strain were to be constructed. Firstly, the kanamycin gene of pBBR1MCS-2 was amplified by PCR method and cloned at the multicloning site of pUC19 to generate a new suicide plasmid pUC19K, which was then used to construct the deletion mutant strain of 16M to generate16MΔvjbR. And then, the ORF of vjbR gene was cloned into pMD18-T vector to construct the complementary plasmid, which was then transformed into 16MΔvjbR to generate the complementary strain 16MΔvjbR-C. Semi-quantitative reverse transcription PCR results showed that transcription of the vjbR genes was detected in the wild-type and complementary strain, but not in the vjbR mutant, indicating that vjbR was successfully inactivated in the mutant and restored in the complementary strain.
     Then, the related intracellular survival phenotypes of wild-type, mutant and complementary strain were compared. Growth curve assays in vitro showed that the wild-type virulent strain 16M displayed higher growth rate than the vjbR mutant and the complementary strain at logarithmic phase. This implied that QS positively regulate growth of Brucella by regulating other related genes. Survival in macrophages and in mice showed that the mutant could invade the macrophages but the disruption of vjbR led to a decreased survival in macrophages and a drastic reduction in spleen or liver colonization in mice, implying that vjbR is essential for intracellular survival and chronic infection of Brucella. Survival under stress conditions showed that the vjbR mutant was sensitive to the stress conditions which simulated intracellular environments, including high salt, high osmosis, low pH, heat shock and oxidative stress. The vjbR mutant was more sensitive to high salt, osmotic stress, polymyxin B and sodium deoxycholate, indicating that vjbR is important for membrane integrity, and resistance to hostile environments. The vjbR mutant was also sensitive to oxidative and acidic stress, which simulated intracellular environment, indicating its role in adaptation to these harsh environments.
     After the elucidation of the intracellular survival phenotypes, a comparative transcriptome analysis was used to define the genes regulated by vjbR, with the aims to explain the possible regulation mechanisms of vjbR at transcription level. As a tool to perform the comparative transcriptome analysis, the non-redundant whole-genome DNA microarrays were developed based on the genomic sequences of B. melitensis strain 16M. A total number of 3968 genes was amplified by PCR and printed in duplicate onto glass slides. Fluorescently labeled probes were prepared by priming of genomic DNAs or cDNAs with random hexamers and extension with Klenow. Labelled DNAs or cDNAs were hybridized to the microarrays by methods of two-fluorescence comparative hybridization. The results were validated with semi-quantitative reverse transcription PCR.
     With the DNA microarray, we compared the transcription profiles of the vjbR mutant to that of 16M. Firstly, quantitative RT-PCR was used to determine the stress condition under which the transcription of vjbR was greatly activated. The results showed that the vjbR was greatly induced under acidified GEM pH4.0 for 2h. Then, comparative transcription analysis was performed under this condition. By a 2 fold change criteria, sixty three genes, including those encoding the type IV secetion system, energey/lipid/amino acid/carbohydrate/ion metabolism, cell envelop and hepothetical protein, were differentially transcribed in vjbR mutant.
     The results above indicated that the vjbR mutant showed several phenotype changes, reduced survival in the macrophage and mice, and further comfirmed its role in intracellular survival by affecting related phenotypes. Results from comparative transcription profiles showed that the vjbR may contribute the adaptation of Brucella to hostile environments and survival in the host cell by regulating the expression of related genes. These results expand our knowledge about roles of vjbR and molecular mechanism of Brucella intracellular survival.
引文
[1] BOSCHIROLI M L, FOULONGNE V, O'CALLAGHAN D. Brucellosis: a worldwide zoonosis [J]. Curr Opin Microbiol, 2001, 4:58-64.
    [2] YOUNG E J. An overview of human brucellosis [J]. Clin Infect Dis, 1995, 21:283-289; quiz 290.
    [3] SCHURIG G G, SRIRANGANATHAN N, CORBEL M J. Brucellosis vaccines: past, present and future [J]. Vet Microbiol, 2002, 90:479-496.
    [4]尚德秋.布氏菌病研究进展[J].中国地方病防治杂志, 2004, 19:204-212.
    [5] DEQIU S, DONGLOU X, JIMING Y, Epidemiology and control of brucellosis in China, in Vet Microbiol. 2002. p. 165-182.
    [6]尚德秋.布鲁氏菌病再度肆虐及其原因[J].中国地方病防治杂志, 2001, 16:29-34.
    [7] CORBEL M J. Brucellosis: an overview [J]. Emerg Infect Dis, 1997, 3:213-221.
    [8] VERGER J M, GRAYON M, TIBOR A, et al. Differentiation of Brucella melitensis, B. ovis and B. suis biovar 2 strains by use of membrane protein or cytoplasmic protein-specific gene probes [J]. Res Microbiol, 1998, 149:509-517.
    [9] FOSTER G, JAHANS K L, REID R J, et al. Isolation of Brucella species from cetaceans, seals and an otter [J]. Vet Rec, 1996, 138:583-586.
    [10] ROSS H M, FOSTER G, REID R J, et al. Brucella species infection in sea-mammals [J]. Vet Rec, 1994, 134:359.
    [11] ROSS H M, JAHANS K L, MACMILLAN A P, et al. Brucella species infection in North Sea seal and cetacean populations [J]. Vet Rec, 1996, 138:647-648.
    [12] SCHOLZ H C, HUBALEK Z, NESVADBOVA J, et al. Isolation of Brucella microti from soil [J]. Emerg Infect Dis, 2008, 14:1316-1317.
    [13]齐景文.布鲁氏菌病概述[J].中国兽医杂志, 2004, 40:50-52.
    [14] KO J, SPLITTER G A. Molecular host-pathogen interaction in brucellosis: current understanding and future approaches to vaccine development for mice and humans [J]. Clin Microbiol Rev, 2003, 16:65-78.
    [15] CHEVALIER P, BONNEFOY E, KIRKORIAN G, et al. Brucella pancarditis with fatal outcome [J]. Presse Med, 1996, 25:628-630.
    [16] MCQUISTON J R, VEMULAPALLI R, INZANA T J, et al. Genetic characterization of a Tn5-disrupted glycosyltransferase gene homolog in Brucella abortus and its effect on lipopolysaccharide composition and virulence [J]. Infect Immun, 1999, 67:3830-3835.
    [17] ARENAS G N, STASKEVICH A S, ABALLAY A, et al. Intracellular trafficking of Brucella abortus in J774 macrophages [J]. Infect Immun, 2000, 68:4255-4263.
    [18] PIZARRO-CERDA J, MERESSE S, PARTON R G, et al. Brucella abortus transits through the autophagic pathway and replicates in the endoplasmicreticulum of nonprofessional phagocytes [J]. Infect Immun, 1998, 66: 5711- 5724.
    [19] GROSS A, TERRAZA A, OUAHRANI-BETTACHE S, et al. In vitro Brucella suis infection prevents the programmed cell death of human monocytic cells [J]. Infect Immun, 2000, 68:342-351.
    [20] PORTE F, LIAUTARD J P, KOHLER S. Early acidification of phagosomes containing Brucella suis is essential for intracellular survival in murine macrophages [J]. Infect Immun, 1999, 67:4041-4047.
    [21] BOSCHIROLI M L, OUAHRANI-BETTACHE S, FOULONGNE V, et al. The Brucella suis virB operon is induced intracellularly in macrophages [J]. Proc Natl Acad Sci U S A, 2002, 99:1544-1549.
    [22] ESCHENBRENNER M, WAGNER M A, HORN T A, et al. Comparative proteome analysis of Brucella melitensis vaccine strain Rev 1 and a virulent strain, 16M [J]. J Bacteriol, 2002, 184:4962-4970.
    [23] AL-MARIRI A, TIBOR A, MERTENS P, et al. Induction of immune response in BALB/c mice with a DNA vaccine encoding bacterioferritin or P39 of Brucella spp [J]. Infect Immun, 2001, 69:6264-6270.
    [24] DELPINO M V, CASSATARO J, FOSSATI C A, et al. Brucella outer membrane protein Omp31 is a haemin-binding protein [J]. Microbes Infect, 2006, 8:1203-1208.
    [25] GUZMAN-VERRI C, MANTEROLA L, SOLA-LANDA A, et al. The two- component system BvrR/BvrS essential for Brucella abortus virulence regulates the expression of outer membrane proteins with counterparts in members of the Rhizobiaceae [J]. Proc Natl Acad Sci U S A, 2002, 99:12375- 12380.
    [26] LIN J, HUANG S, ZHANG Q. Outer membrane proteins: key players for bacterial adaptation in host niches [J]. Microbes Infect, 2002, 4:325-331.
    [27] WEISER J N, GOTSCHLICH E C. Outer membrane protein A (OmpA) contributes to serum resistance and pathogenicity of Escherichia coli K-1 [J]. Infect Immun, 1991, 59:2252-2258.
    [28] BOUNOUS D I, ENRIGHT F M, GOSSETT K A, et al. Phagocytosis, killing, and oxidant production by bovine monocyte-derived macrophages upon exposure to Brucella abortus strain 2308 [J]. Vet Immunol Immunopathol, 1993, 37:243-256.
    [29] CHEVILLE N F, KUNKLE R A, JENSEN A E, et al. Persistence of Brucella abortus in the livers of T cell-deficient nude mice [J]. Lab Invest, 1995, 73:96-102.
    [30] CANNING P C, ROTH J A, DEYOE B L. Release of 5'-guanosine monophosphate and adenine by Brucella abortus and their role in the intracellular survival of the bacteria [J]. J Infect Dis, 1986, 154:464-470.
    [31] RILEY L K, ROBERTSON D C. Ingestion and intracellular survival of Brucellaabortus in human and bovine polymorphonuclear leukocytes [J]. Infect Immun, 1984, 46:224-230.
    [32] PIZARRO-CERDA J, MORENO E, GORVEL J P. Invasion and intracellular trafficking of Brucella abortus in nonphagocytic cells [J]. Microbes Infect, 2000, 2:829-835.
    [33] KOHLER S, LAYSSAC M, NAROENI A, et al. Secretion of listeriolysin by Brucella suis inhibits its intramacrophagic replication [J]. Infect Immun, 2001, 69:2753-2756.
    [34] RAMBOW-LARSEN A A, PETERSEN E M, GOURLEY C R, et al. Brucella regulators: self-control in a hostile environment [J]. Trends Microbiol, 2009, 17:371-377.
    [35] TAMINIAU B, DAYKIN M, SWIFT S, et al. Identification of a quorum- sensing signal molecule in the facultative intracellular pathogen Brucella melitensis [J]. Infect Immun, 2002, 70:3004-3011.
    [36] FUQUA W C, WINANS S C, GREENBERG E P. Quorum sensing in bacteria: the LuxR-LuxI family of cell density-responsive transcriptional regulators [J]. J Bacteriol, 1994, 176:269-275.
    [37] DAVIES D G, M. R. PARSEK, J. P. PEARSON, B. H. IGLEWSKI, J. W. COSTERTON, GREENBERG. A E P. The involvement of cell-to-cell signals in the development of a bacterial biofilm [J]. Science, 1998, 295–298.
    [38] RAMBOW-LARSEN A A, RAJASHEKARA G, PETERSEN E, et al. Putative quorum-sensing regulator BlxR of Brucella melitensis regulates virulence factors including the type IV secretion system and flagella [J]. J Bacteriol, 2008, 190:3274-3282.
    [39] LEONARD S, FEROOZ J, HAINE V, et al. FtcR is a new master regulator of the flagellar system of Brucella melitensis 16M with homologs in Rhizobiaceae [J]. J Bacteriol, 2007, 189:131-141.
    [40] DE JONG M F, SUN Y H, DEN HARTIGH A B, et al. Identification of VceA and VceC, two members of the VjbR regulon that are translocated into macrophages by the Brucella type IV secretion system [J]. Mol Microbiol, 2008.
    [41] DELRUE R M, DESCHAMPS C, LEONARD S, et al. A quorum-sensing regulator controls expression of both the type IV secretion system and the flagellar apparatus of Brucella melitensis [J]. Cell Microbiol, 2005, 7:1151-1161.
    [42] BOSCHIROLI M L, OUAHRANI-BETTACHE S, FOULONGNE V, et al. Type IV secretion and Brucella virulence [J]. Vet Microbiol, 2002, 90:341- 348.
    [43] O'CALLAGHAN D, CAZEVIEILLE C, ALLARDET-SERVENT A, et al. A homologue of the Agrobacterium tumefaciens VirB and Bordetella pertussis Ptl type IV secretion systems is essential for intracellular survival of Brucella suis [J]. Mol Microbiol, 1999, 33:1210-1220.
    [44] ROUOT B, ALVAREZ-MARTINEZ M T, MARIUS C, et al. Production of thetype IV secretion system differs among Brucella species as revealed with VirB5- and VirB8-specific antisera [J]. Infect Immun, 2003, 71:1075-1082.
    [45] CELLI J, DE CHASTELLIER C, FRANCHINI D M, et al. Brucella evades macrophage killing via VirB-dependent sustained interactions with the endoplasmic reticulum [J]. J Exp Med, 2003, 198:545-556.
    [46] SOLA-LANDA A, PIZARRO-CERDA J, GRILLO M J, et al. A two- component regulatory system playing a critical role in plant pathogens and endosymbionts is present in Brucella abortus and controls cell invasion and virulence [J]. Mol Microbiol, 1998, 29:125-138.
    [47] LOPEZ-GONI I, GUZMAN-VERRI C, MANTEROLA L, et al. Regulation of Brucella virulence by the two-component system BvrR/BvrS [J]. Vet Microbiol, 2002, 90:329-339.
    [48] ALTON G G, JONES L M, PIETZ D E. Laboratory techniques in brucellosis [J]. Monogr Ser World Health Organ, 1975, 1-163.
    [49] HALLING S M. On the presence and organization of open reading frames of the nonmotile pathogen Brucella abortus similar to class II, III, and IV flagellar genes and to LcrD virulence superfamily [J]. Microb Comp Genomics, 1998, 3:21-29.
    [50] DELVECCHIO V G, KAPATRAL V, ELZER P, et al. The genome of Brucella melitensis [J]. Vet Microbiol, 2002, 90:587-592.
    [51] PAULSEN I T, SESHADRI R, NELSON K E, et al. The Brucella suis genome reveals fundamental similarities between animal and plant pathogens and symbionts [J]. Proc Natl Acad Sci U S A, 2002, 99:13148-13153.
    [52] MORENO E, MORIYON I. Brucella melitensis: a nasty bug with hidden credentials for virulence [J]. Proc Natl Acad Sci U S A, 2002, 99:1-3.
    [53] LESTRATE P, DELRUE R M, DANESE I, et al. Identification and characterization of in vivo attenuated mutants of Brucella melitensis [J]. Mol Microbiol, 2000, 38:543-551.
    [54] LESTRATE P, DRICOT A, DELRUE R M, et al. Attenuated signature-tagged mutagenesis mutants of Brucella melitensis identified during the acute phase of infection in mice [J]. Infect Immun, 2003, 71:7053-7060.
    [55] FRETIN D, FAUCONNIER A, KOHLER S, et al. The sheathed flagellum of Brucella melitensis is involved in persistence in a murine model of infection [J]. Cell Microbiol, 2005, 7:687-698.
    [56] HANZELKA B L, GREENBERG E P. Evidence that the N-terminal region of the Vibrio fischeri LuxR protein constitutes an autoinducer-binding domain [J]. J Bacteriol, 1995, 177:815-817.
    [57] CHOI S H, GREENBERG E P. The C-terminal region of the Vibrio fischeri LuxR protein contains an inducer-independent lux gene activating domain [J]. Proc Natl Acad Sci U S A, 1991, 88:11115-11119.
    [58] ZHU J, WINANS S C. The quorum-sensing transcriptional regulator TraR requires its cognate signaling ligand for protein folding, protease resistance, and dimerization [J]. Proc Natl Acad Sci U S A, 2001, 98:1507-1512.
    [59] UZUREAU S, GODEFROID M, DESCHAMPS C, et al. Mutations of the quorum sensing-dependent regulator VjbR lead to drastic surface modifications in Brucella melitensis [J]. J Bacteriol, 2007, 189:6035-6047.
    [60] DE IANNINO N I, BRIONES G, IANNINO F, et al. Osmotic regulation of cyclic 1,2-beta-glucan synthesis [J]. Microbiology, 2000, 146 ( Pt 7):1735-1742.
    [61] BRIONES G, INON DE IANNINO N, ROSET M, et al. Brucella abortus cyclic beta-1,2-glucan mutants have reduced virulence in mice and are defective in intracellular replication in HeLa cells [J]. Infect Immun, 2001, 69:4528-4535.
    [62] BOHIN J P. Osmoregulated periplasmic glucans in Proteobacteria [J]. FEMS Microbiol Lett, 2000, 186:11-19.
    [63] ARELLANO-REYNOSO B, LAPAQUE N, SALCEDO S, et al. Cyclic beta-1,2-glucan is a Brucella virulence factor required for intracellular survival [J]. Nat Immunol, 2005, 6:618-625.
    [64] KELETI G, FEINGOLD D S, YOUNGNER J S. Interferon induction in mice by lipopolysaccharide from Brucella abortus [J]. Infect Immun, 1974, 10:282-283.
    [65] TUMURKHUU G, KOIDE N, TAKAHASHI K, et al. Characterization of biological activities of Brucella melitensis lipopolysaccharide [J]. Microbiol Immunol, 2006, 50:421-427.
    [66] MORENO E, BERMAN D T, BOETTCHER L A. Biological activities of Brucella abortus lipopolysaccharides [J]. Infect Immun, 1981, 31:362-370.
    [67] GOLDSTEIN J, HOFFMAN T, FRASCH C, et al. Lipopolysaccharide (LPS) from Brucella abortus is less toxic than that from Escherichia coli, suggesting the possible use of B. abortus or LPS from B. abortus as a carrier in vaccines [J]. Infect Immun, 1992, 60:1385-1389.
    [68] RASOOL O, FREER E, MORENO E, et al. Effect of Brucella abortus lipopolysaccharide on oxidative metabolism and lysozyme release by human neutrophils [J]. Infect Immun, 1992, 60:1699-1702.
    [69] LAPAQUE N, TAKEUCHI O, CORRALES F, et al. Differential inductions of TNF-alpha and IGTP, IIGP by structurally diverse classic and non-classic lipopolysaccharides [J]. Cell Microbiol, 2006, 8:401-413.
    [70] EISENSCHENK F C, HOULE J J, HOFFMANN E M. Mechanism of serum resistance among Brucella abortus isolates [J]. Vet Microbiol, 1999, 68:235-244.
    [71] MARTINEZ DE TEJADA G, PIZARRO-CERDA J, MORENO E, et al. The outer membranes of Brucella spp. are resistant to bactericidal cationic peptides [J]. Infect Immun, 1995, 63:3054-3061.
    [72] FREER E, MORENO E, MORIYON I, et al. Brucella-Salmonella lipopolysaccharide chimeras are less permeable to hydrophobic probes and moresensitive to cationic peptides and EDTA than are their native Brucella spp. counterparts [J]. J Bacteriol, 1996, 178:5867-5876.
    [73] VELASCO J, BENGOECHEA J A, BRANDENBURG K, et al. Brucella abortus and its closest phylogenetic relative, Ochrobactrum spp., differ in outer membrane permeability and cationic peptide resistance [J]. Infect Immun, 2000, 68:3210-3218.
    [74] DETILLEUX P G, DEYOE B L, CHEVILLE N F. Penetration and intracellular growth of Brucella abortus in nonphagocytic cells in vitro [J]. Infect Immun, 1990, 58:2320-2328.
    [75] PORTE F, NAROENI A, OUAHRANI-BETTACHE S, et al. Role of the Brucella suis lipopolysaccharide O antigen in phagosomal genesis and in inhibition of phagosome-lysosome fusion in murine macrophages [J]. Infect Immun, 2003, 71:1481-1490.
    [76] JIMENEZ DE BAGUES M P, TERRAZA A, GROSS A, et al. Different responses of macrophages to smooth and rough Brucella spp.: relationship to virulence [J]. Infect Immun, 2004, 72:2429-2433.
    [77] FORESTIER C, MORENO E, PIZARRO-CERDA J, et al. Lysosomal accumulation and recycling of lipopolysaccharide to the cell surface of murine macrophages, an in vitro and in vivo study [J]. J Immunol, 1999, 162:6784-6791.
    [78] FORESTIER C, MORENO E, MERESSE S, et al. Interaction of Brucella abortus lipopolysaccharide with major histocompatibility complex class II molecules in B lymphocytes [J]. Infect Immun, 1999, 67:4048-4054.
    [79] FORESTIER C, DELEUIL F, LAPAQUE N, et al. Brucella abortus lipopolysaccharide in murine peritoneal macrophages acts as a down-regulator of T cell activation [J]. J Immunol, 2000, 165:5202-5210.
    [80] RAFIE-KOLPIN M, ESSENBERG R C, WYCKOFF J H, 3RD. Identification and comparison of macrophage-induced proteins and proteins induced under various stress conditions in Brucella abortus [J]. Infect Immun, 1996, 64:5274-5283.
    [81] TEIXEIRA-GOMES A P, CLOECKAERT A, ZYGMUNT M S. Characterization of heat, oxidative, and acid stress responses in Brucella melitensis [J]. Infect Immun, 2000, 68:2954-2961.
    [82] DELORY M, HALLEZ R, LETESSON J J, et al. An RpoH-like heat shock sigma factor is involved in stress response and virulence in Brucella melitensis 16M [J]. J Bacteriol, 2006, 188:7707-7710.
    [83] SIEIRA R, COMERCI D J, PIETRASANTA L I, et al. Integration host factor is involved in transcriptional regulation of the Brucella abortus virB operon [J]. Mol Microbiol, 2004, 54:808-822.
    [84] DOZOT M, BOIGEGRAIN R A, DELRUE R M, et al. The stringent response mediator Rsh is required for Brucella melitensis and Brucella suis virulence, andfor expression of the type IV secretion system virB [J]. Cell Microbiol, 2006, 8:1791-1802.
    [85] ALI AZAM T, IWATA A, NISHIMURA A, et al. Growth phase-dependent variation in protein composition of the Escherichia coli nucleoid [J]. J Bacteriol, 1999, 181:6361-6370.
    [86] YU D H, HU X D, CAI H. A combined DNA vaccine encoding BCSP31, SOD, and L7/L12 confers high protection against Brucella abortus 2308 by inducing specific CTL responses [J]. DNA Cell Biol, 2007, 26:435-443.
    [87] MUNOZ-MONTESINO C, ANDREWS E, RIVERS R, et al. Intraspleen delivery of a DNA vaccine coding for superoxide dismutase (SOD) of Brucella abortus induces SOD-specific CD4+and CD8+T cells [J]. Infect Immun, 2004, 72: 2081- 2087.
    [88] CARON E, CELLIER M, LIAUTARD J P, et al. Complementation of a DnaK-deficient Escherichia coli strain with the dnaK/dnaJ operon of Brucella ovis reduces the rate of initial intracellular killing within the monocytic cell line U937 [J]. FEMS Microbiol Lett, 1994, 120:335-340.
    [89] CELLIER M F, TEYSSIER J, NICOLAS M, et al. Cloning and characterization of the Brucella ovis heat shock protein DnaK functionally expressed in Escherichia coli [J]. J Bacteriol, 1992, 174:8036-8042.
    [90] PHILLIPS R W, ROOP R M, 2ND. Brucella abortus HtrA functions as an authentic stress response protease but is not required for wild-type virulence in BALB/c mice [J]. Infect Immun, 2001, 69:5911-5913.
    [91] ELZER P H, PHILLIPS R W, KOVACH M E, et al. Characterization and genetic complementation of a Brucella abortus high-temperature-requirement A (htrA) deletion mutant [J]. Infect Immun, 1994, 62:4135-4139.
    [92] PHILLIPS R W, ELZER P H, ROOP R M, II. A Brucella melitensis high temperature requirement A (htrA) deletion mutant demonstrates a stress response defective phenotype in vitro and transient attenuation in the BALB/c mouse model [J]. Microb Pathog, 1995, 19:227-284.
    [93] PHILLIPS R W, ELZER P H, ROBERTSON G T, et al. A Brucella melitensis high-temperature-requirement A (htrA) deletion mutant is attenuated in goats and protects against abortion [J]. Res Vet Sci, 1997, 63:165-167.
    [94] CONDE-ALVAREZ R, GRILLO M J, SALCEDO S P, et al. Synthesis of phosphatidylcholine, a typical eukaryotic phospholipid, is necessary for full virulence of the intracellular bacterial parasite Brucella abortus [J]. Cell Microbiol, 2006, 8:1322-1335.
    [95] LEVIER K, PHILLIPS R W, GRIPPE V K, et al. Similar requirements of a plant symbiont and a mammalian pathogen for prolonged intracellular survival [J]. Science, 2000, 287:2492-2493.
    [96] EDMONDS M D, CLOECKAERT A, BOOTH N J, et al. Attenuation of aBrucella abortus mutant lacking a major 25 kDa outer membrane protein in cattle [J]. Am J Vet Res, 2001, 62:1461-1466.
    [97] EDMONDS M D, CLOECKAERT A, ELZER P H. Brucella species lacking the major outer membrane protein Omp25 are attenuated in mice and protect against Brucella melitensis and Brucella ovis [J]. Vet Microbiol, 2002, 88:205-221.
    [98] SHA Z, STABEL T J, MAYFIELD J E. Brucella abortus catalase is a periplasmic protein lacking a standard signal sequence [J]. J Bacteriol, 1994, 176:7375-7377.
    [99] EWALT D R, BRICKER B J. Validation of the abbreviated Brucella AMOS PCR as a rapid screening method for differentiation of Brucella abortus field strain isolates and the vaccine strains, 19 and RB51 [J]. J Clin Microbiol, 2000, 38:3085-3086.
    [100] JAYAPAL M, MELENDEZ A J. DNA microarray technology for target identification and validation [J]. Clin Exp Pharmacol Physiol, 2006, 33:496-503.
    [101] KOSTRZYNSKA M, BACHAND A. Application of DNA microarray technology for detection, identification, and characterization of food-borne pathogens [J]. Can J Microbiol, 2006, 52:1-8.
    [102] MANDRUZZATO S. Technological platforms for microarray gene expression profiling [J]. Adv Exp Med Biol, 2007, 593:12-18.
    [103] ROSA G J, DE LEON N, ROSA A J. Review of microarray experimental design strategies for genetical genomics studies [J]. Physiol Genomics, 2006, 28:15-23.
    [104] BRYANT P A, VENTER D, ROBINS-BROWNE R, et al. Chips with everything: DNA microarrays in infectious diseases [J]. Lancet Infect Dis, 2004, 4:100-111.
    [105] SCHOOLNIK G K. Microarray analysis of bacterial pathogenicity [J]. Adv Microb Physiol, 2002, 46:1-45.
    [106] SCHOOLNIK G K. Functional and comparative genomics of pathogenic bacteria [J]. Curr Opin Microbiol, 2002, 5:20-26.
    [107] EGLAND K A, GREENBERG E P. Conversion of the Vibrio fischeri transcriptional activator, LuxR, to a repressor [J]. J Bacteriol, 2000, 182:805-811.
    [108] LEADBETTER J R, GREENBERG E P. Metabolism of acyl-homoserine lactone quorum-sensing signals by Variovorax paradoxus [J]. J Bacteriol, 2000, 182:6921-6926.
    [109] MANN M, HOJRUP P, ROEPSTORFF P. Use of mass spectrometric molecular weight information to identify proteins in sequence databases [J]. Biol Mass Spectrom, 1993, 22:338-345.
    [110] MARCHESINI M I, UGALDE J E, CZIBENER C, et al. N-terminal-capturing screening system for the isolation of Brucella abortus genes encoding surface exposed and secreted proteins [J]. Microb Pathog, 2004, 37:95-105.
    [111] BUMANN D, AKSU S, WENDLAND M, et al. Proteome analysis of secreted proteins of the gastric pathogen Helicobacter pylori [J]. Infect Immun, 2002, 70:3396-3403.
    [112] VANET A, LABIGNE A. Evidence for specific secretion rather than autolysis in the release of some Helicobacter pylori proteins [J]. Infect Immun, 1998, 66:1023-1027.
    [113] WATARAI M, KIM S, ERDENEBAATAR J, et al. Cellular prion protein promotes Brucella infection into macrophages [J]. J Exp Med, 2003, 198:5-17.
    [114] THIEL M, BRUCHHAUS I. Comparative proteome analysis of Leishmania donovani at different stages of transformation from promastigotes to amastigotes [J]. Med Microbiol Immunol, 2001, 190:33-36.
    [115] DELVECCHIO V G, KAPATRAL V, REDKAR R J, et al. The genome sequence of the facultative intracellular pathogen Brucella melitensis [J]. Proc Natl Acad Sci U S A, 2002, 99:443-448.
    [116] ESCHENBRENNER M, HORN T A, WAGNER M A, et al. Comparative proteome analysis of laboratory grown Brucella abortus 2308 and Brucella melitensis 16M [J]. J Proteome Res, 2006, 5:1731-1740.
    [117] MANTEROLA L, GUZMAN-VERRI C, CHAVES-OLARTE E, et al. BvrR/ BvrS-controlled outer membrane proteins Omp3a and Omp3b are not essential for Brucella abortus virulence [J]. Infect Immun, 2007, 75:4867-4874.
    [118] CONNOLLY J P, COMERCI D, ALEFANTIS T G, et al. Proteomic analysis of Brucella abortus cell envelope and identification of immunogenic candidate proteins for vaccine development [J]. Proteomics, 2006, 6:3767-3780.
    [119] RATLEDGE C, DOVER L G. Iron metabolism in pathogenic bacteria [J]. Annu Rev Microbiol, 2000, 54:881-941.
    [120] PERUCHO M, HANAHAN D, LIPSICH L, et al. Isolation of the chicken thymidine kinase gene by plasmid rescue [J]. Nature, 1980, 285:207-210.
    [121] ORTIZ-MARTIN I, MACHO A P, LAMBERSTEN L, et al. Suicide vectors for antibiotic marker exchange and rapid generation of multiple knockout mutants by allelic exchange in Gram-negative bacteria [J]. J Microbiol Methods, 2006, 67:395-407.
    [122] REYRAT J M, PELICIC V, GICQUEL B, et al. Counterselectable markers: untapped tools for bacterial genetics and pathogenesis [J]. Infect Immun, 1998, 66:4011-4017.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700