1
|
Pillich H, Puri M, Chakraborty T. ActA of Listeria monocytogenes and Its Manifold Activities as an Important Listerial Virulence Factor. Curr Top Microbiol Immunol 2016; 399:113-132. [PMID: 27726006 DOI: 10.1007/82_2016_30] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Listeria monocytogenes is a ubiquitously occurring gram-positive bacterium in the environment that causes listeriosis, one of the deadliest foodborne infections known today. It is a versatile facultative intracellular pathogen capable of growth within the host's cytosolic compartment. Following entry into the host cell, L. monocytogenes escapes from vacuolar compartments to the cytosol, where the bacterium begins a remarkable journey within the host cytoplasm, culminating in bacterial spread from cell to cell, to deeper tissues and organs. This dissemination process depends on the ability of the bacterium to harness central components of the host cell actin cytoskeleton using the surface bound bacterial factor ActA (actin assembly inducing protein). Hence ActA plays a major role in listerial virulence, and its absence renders bacteria intracellularly immotile and essentially non-infectious. As the bacterium, moving by building a network of filamentous actin behind itself that is often referred to as its actin tail, encounters cell-cell contacts it forms double-vacuolar protrusions that allow it to enter the neighboring cell where the cycle then continues. Recent studies have now implicated ActA in other stages of the life cycle of L. monocytogenes. These include extracellular properties of aggregation and biofilm formation to mediate colonization of the gut lumen, promotion and enhancement of bacterial host cell entry, evasion of autophagy, vacuolar exit, as well as nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB) activation. These novel properties provide a new view of ActA and help explain its role as an essential virulence factor of L. monocytogenes.
Collapse
Affiliation(s)
- Helena Pillich
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Madhu Puri
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany.
| |
Collapse
|
2
|
Ansari MA, Zia Q, Kazmi S, Ahmad E, Azhar A, Johnson KE, Zubair S, Owais M. Efficacy of Cell Wall-Deficient Spheroplasts Against Experimental Murine Listeriosis. Scand J Immunol 2015; 82:10-24. [DOI: 10.1111/sji.12296] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/09/2015] [Indexed: 01/19/2023]
Affiliation(s)
- M. A. Ansari
- Interdisciplinary Biotechnology Unit; Aligarh Muslim University; Aligarh India
- Department of Microbiology and Immunology; Rosalind Franklin University of Medicine and Science; North Chicago IL USA
| | - Q. Zia
- Interdisciplinary Biotechnology Unit; Aligarh Muslim University; Aligarh India
| | - S. Kazmi
- Interdisciplinary Biotechnology Unit; Aligarh Muslim University; Aligarh India
| | - E. Ahmad
- Interdisciplinary Biotechnology Unit; Aligarh Muslim University; Aligarh India
| | - A. Azhar
- Interdisciplinary Biotechnology Unit; Aligarh Muslim University; Aligarh India
| | - K. E. Johnson
- Department of Microbiology and Immunology; Rosalind Franklin University of Medicine and Science; North Chicago IL USA
| | - S. Zubair
- Women's College; Aligarh Muslim University; Aligarh India
| | - M. Owais
- Interdisciplinary Biotechnology Unit; Aligarh Muslim University; Aligarh India
| |
Collapse
|
3
|
Acquired resistance of Listeria monocytogenes in and escaped from liver parenchymal cells to gentamicin is caused by being coated with their plasma membrane. Microbes Infect 2014; 16:237-43. [DOI: 10.1016/j.micinf.2013.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/22/2013] [Accepted: 11/09/2013] [Indexed: 11/24/2022]
|
4
|
Osanai A, Li SJ, Asano K, Sashinami H, Hu DL, Nakane A. Fibronectin-binding protein, FbpA, is the adhesin responsible for pathogenesis ofListeria monocytogenesinfection. Microbiol Immunol 2013; 57:253-62. [DOI: 10.1111/1348-0421.12030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 01/08/2013] [Accepted: 01/10/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Arihiro Osanai
- Department of Microbiology and Immunology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho, Hirosaki; Aomori; 036-8562; Japan
| | | | - Krisana Asano
- Department of Microbiology and Immunology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho, Hirosaki; Aomori; 036-8562; Japan
| | - Hiroshi Sashinami
- Department of Microbiology and Immunology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho, Hirosaki; Aomori; 036-8562; Japan
| | - Dong-Liang Hu
- Department of Microbiology and Immunology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho, Hirosaki; Aomori; 036-8562; Japan
| | - Akio Nakane
- Department of Microbiology and Immunology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho, Hirosaki; Aomori; 036-8562; Japan
| |
Collapse
|
5
|
Ansari MA, Zubair S, Tufail S, Ahmad E, Khan MR, Quadri Z, Owais M. Ether lipid vesicle-based antigens impart protection against experimental listeriosis. Int J Nanomedicine 2012; 7:2433-47. [PMID: 22745536 PMCID: PMC3383290 DOI: 10.2147/ijn.s25875] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Incidence of food-borne infections from Listeria monocytogenes, a parasite that has adapted intracellular residence to avoid antibody onslaught, has increased dramatically in the past few years. The apparent lack of an effective vaccine that is capable of evoking the desired cytotoxic T cell response to obliterate this intracellular pathogen has encouraged the investigation of alternate prophylactic strategies. It should also be noted that Archaebacteria (Archae) lipid-based adjuvants enhance the efficacy of subunit vaccines. In the present study, the adjuvant properties of archaeosomes (liposomes prepared from total polar lipids of archaebacteria, Halobacterium salinarum) combined with immunogenic culture supernatant antigens of L. monocytogenes have been exploited in designing a vaccine candidate against experimental listeriosis in murine model. Methods Archaeosome-entrapped secretory protein antigens (SAgs) of L. monocytogenes were evaluated for their immunological responses and tendency to deplete bacterial burden in BALB/c mice challenged with sublethal listerial infection. Various immunological studies involving cytokine profiling, lymphocyte proliferation assay, detection of various surface markers (by flowcytometric analysis), and antibody isotypes (by enzyme-linked immunosorbent assay) were used for establishing the vaccine potential of archaeosome-entrapped secretory proteins. Results Immunization schedule involving archaeosome-encapsulated SAgs resulted in upregulation of Th1 cytokine production along with boosted memory in BALB/c mice. It also showed protective effect by reducing listerial burden in various vital organs (liver and spleen) of the infected mice. However, the soluble form of the antigens (SAgs) and their physical mixture with sham (empty) archaeosomes, besides showing feeble Th1 response, were unable to protect the animals against virulent listerial infection. Conclusion On the basis of the evidence provided by the current data, it is inferred that archaeosome-entrapped SAgs formulation not only enhances cytotoxic T cell response but also helps in the clearance of pathogens and thereby increases the survival of the immunized animals.
Collapse
Affiliation(s)
- Mairaj Ahmed Ansari
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, UP, India
| | | | | | | | | | | | | |
Collapse
|
6
|
|
7
|
Bast A, Schmidt IHE, Brauner P, Brix B, Breitbach K, Steinmetz I. Defense Mechanisms of Hepatocytes Against Burkholderia pseudomallei. Front Microbiol 2012; 2:277. [PMID: 22291688 PMCID: PMC3263921 DOI: 10.3389/fmicb.2011.00277] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 12/24/2011] [Indexed: 01/18/2023] Open
Abstract
The Gram-negative facultative intracellular rod Burkholderia pseudomallei causes melioidosis, an infectious disease with a wide range of clinical presentations. Among the observed visceral abscesses, the liver is commonly affected. However, neither this organotropism of B. pseudomallei nor local hepatic defense mechanisms have been thoroughly investigated so far. Own previous studies using electron microscopy of the murine liver after systemic infection of mice indicated that hepatocytes might be capable of killing B. pseudomallei. Therefore, the aim of this study was to further elucidate the interaction of B. pseudomallei with these cells and to analyze the role of hepatocytes in anti-B. pseudomallei host defense. In vitro studies using the human hepatocyte cell line HepG2 revealed that B. pseudomallei can invade these cells. Subsequently, B. pseudomallei is able to escape from the vacuole, to replicate within the cytosol of HepG2 cells involving its type 3 and type 6 secretion systems, and to induce actin tail formation. Furthermore, stimulation of HepG2 cells showed that IFNγ can restrict growth of B. pseudomallei in the early and late phase of infection whereas the combination of IFNγ, IL-1β, and TNFα is required for the maximal antibacterial activity. This anti-B. pseudomallei defense of HepG2 cells did not seem to be mediated by inducible nitric oxide synthase-derived nitric oxide or NADPH oxidase-derived superoxide. In summary, this is the first study describing B. pseudomallei intracellular life cycle characteristics in hepatocytes and showing that IFNγ-mediated, but nitric oxide- and reactive oxygen species-independent, effector mechanisms are important in anti-B. pseudomallei host defense of hepatocytes.
Collapse
Affiliation(s)
- Antje Bast
- Friedrich Loeffler Institute of Medical Microbiology, University of Greifswald Greifswald, Germany
| | | | | | | | | | | |
Collapse
|
8
|
dos Santos SA, de Andrade Júnior DR, de Andrade DR. TNF-α production and apoptosis in hepatocytes after Listeria monocytogenes and Salmonella Typhimurium invasion. Rev Inst Med Trop Sao Paulo 2011; 53:107-12. [PMID: 21537759 DOI: 10.1590/s0036-46652011000200009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 01/20/2011] [Indexed: 11/22/2022] Open
Abstract
Invasion of hepatocytes by Listeria monocytogenes (LM) and Salmonella Typhimurium (ST) can stimulate tumor necrosis factor alpha (TNF-α) release and induce apoptosis. In this study, we compared the behavior of hepatocytes invaded by three L. monocytogenes serotypes (LM-4a, LM-4b and LM-1/2a) and by ST to understand which bacterium is more effective in the infectious process. We quantified TNF-α release by ELISA, apoptosis rates by annexin V (early apoptosis) and TUNEL (late apoptosis) techniques. The cell morphology was studied too. TNF-α release rate was highest in ST-invaded hepatocytes. ST and LM-1/2a induced the highest apoptosis production rates evaluated by TUNEL. LM-4b produced the highest apoptosis rate measured by annexin. Invaded hepatocytes presented various morphological alterations. Overall, LM-4b and LM-1/2a proved to be the most efficient at cell invasion, although ST adapted faster to the environment and induced earlier hepatocyte TNF-α release.
Collapse
Affiliation(s)
- Sânia Alves dos Santos
- Laboratory of Bacteriology, Department of Infectious Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil.
| | | | | |
Collapse
|
9
|
Kayal S, Charbit A. Listeriolysin O: a key protein ofListeria monocytogeneswith multiple functions. FEMS Microbiol Rev 2006; 30:514-29. [PMID: 16774585 DOI: 10.1111/j.1574-6976.2006.00021.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are produced by a large number of pathogenic Gram-positive bacteria. Most of these single-chain proteins are secreted in the extracellular medium. Among the species producing CDCs, only two species belonging to the genus Listeria (Listeria monocytogenes and Listeria ivanovii) are able to multiply intracellularly and release their toxins in the phagosomal compartment of the infected host cell. This review provides an updated overview on the importance of listeriolysin O (LLO) in the pathogenicity of L. monocytogenes, focusing mainly on two aspects: (1) the structure-function relationship of LLO and (2) its role in intra- and extracellular signalling. We first examine the specific sequence determinants, or protein domains, that make this cytolysin so well adapted to the intracellular lifestyle of L. monocytogenes. The roles that LLO has in cellular signalling events in the context of relations to pathogenesis are also discussed.
Collapse
Affiliation(s)
- Samer Kayal
- Faculté de Médecine, Université René Descartes-Paris 5, INSERM U-570, Paris, France
| | | |
Collapse
|
10
|
Werbrouck H, Grijspeerdt K, Botteldoorn N, Van Pamel E, Rijpens N, Van Damme J, Uyttendaele M, Herman L, Van Coillie E. Differential inlA and inlB expression and interaction with human intestinal and liver cells by Listeria monocytogenes strains of different origins. Appl Environ Microbiol 2006; 72:3862-71. [PMID: 16751490 PMCID: PMC1489604 DOI: 10.1128/aem.02164-05] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2005] [Accepted: 03/19/2006] [Indexed: 02/08/2023] Open
Abstract
In this study, a number of Listeria monocytogenes strains of different origins were evaluated for in vitro invasion capacity for various human cell types (monocytic THP-1, enterocytic Caco-2, and hepatocytic HepG2 cells) and for expression levels of specific virulence genes. For THP-1 cells, no differences between clinical and nonclinical L. monocytogenes strains in invasion capacity or in production of the proinflammatory cytokine interleukin-8 (IL-8) were observed, whereas for the Caco-2 and HepG2 cells, significant differences in invasion capacity were noticed. On average, the clinical strains showed a significantly lower invasion capacity than the nonclinical L. monocytogenes strains. Furthermore, it was shown that the clinical strains induce lower IL-8 levels in HepG2 cells than do the nonclinical strains. This observation led us to study the mRNA expression levels of inlA, inlB, and ami, important virulence genes mediating adhesion and invasion of eukaryotic cells, by real-time reverse transcription-PCR for 27 clinical and 37 nonclinical L. monocytogenes strains. Significant differences in inlA and inlB expression were observed, with clinical strains showing a lower expression level than nonclinical strains. These observations were in accordance with in vitro invasion of Caco-2 and HepG2 cells, respectively. The results of this study indicate that differential expression levels of inlA and inlB possibly play a role in the virulence capacities of L. monocytogenes strains. The lower capacity of clinical strains to invade HepG2 cells and to induce IL-8 is possibly a mechanism of immune evasion used by specific L. monocytogenes strains.
Collapse
Affiliation(s)
- Hadewig Werbrouck
- Institute for Agricultural and Fisheries Research, Unit Technology and Food-Product Quality and Food Safety, Brusselsesteenweg 370, 9090 Melle, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Santos SAD, Andrade DRD, Andrade Júnior DRD. Rat hepatocyte invasion by Listeria monocytogenes and analysis of TNF-alpha role in apoptosis. Rev Inst Med Trop Sao Paulo 2005; 47:73-80. [PMID: 15880217 DOI: 10.1590/s0036-46652005000200003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Listeria monocytogenes, etiological agent of severe human foodborne infection, uses sophisticated mechanisms of entry into host cytoplasm and manipulation of the cellular cytoskeleton, resulting in cell death. The host cells and bacteria interaction may result in cytokine production as Tumor Necrosis Factor (TNF) alpha. Hepatocytes have potential to produce pro-inflammatory cytokines as TNF-alpha when invaded by bacteria. In the present work we showed the behavior of hepatocytes invaded by L. monocytogenes by microscopic analysis, determination of TNF-alpha production by bioassay and analysis of the apoptosis through TUNEL technique. The presence of bacterium, in ratios that ranged from 5 to 50,000 bacteria per cell, induced the rupture of cellular monolayers. We observed the presence of internalized bacteria in the first hour of incubation by electronic microscopy. The levels of TNF-alpha increased from first hour of incubation to sixth hour, ranging from 0 to 3749 pg/mL. After seven and eight hours of incubation non-significant TNF-alpha levels decrease occurred, indicating possible saturation of cellular receptors. Thus, the quantity of TNF-alpha produced by hepatocytes was dependent of the incubation time, as well as of the proportion between bacteria and cells. The apoptosis rate increased in direct form with the incubation time (1 h to 8 + 24 h), ranging from 0 to 43%, as well as with the bacteria : cells ratio. These results show the ability of hepatocyte invasion by non-hemolytic L. monocytogenes, and the main consequences of this phenomenon were the release of TNF-alpha by hepatocytes and the induction of apoptosis. We speculate that hepatocytes use apoptosis induced by TNF-alpha for release bacteria to extracellular medium. This phenomenon may facilitate the bacteria destruction by the immune system.
Collapse
Affiliation(s)
- Sânia Alves dos Santos
- Laboratory of Cellular Microbiology, LIM 54, Department of Infectious Diseases, School of Medicine, University of São Paulo, 05403-000 São Paulo, SP, Brazil.
| | | | | |
Collapse
|
12
|
Bonnemain C, Raynaud C, Réglier-Poupet H, Dubail I, Frehel C, Lety MA, Berche P, Charbit A. Differential roles of multiple signal peptidases in the virulence of Listeria monocytogenes. Mol Microbiol 2004; 51:1251-66. [PMID: 14982622 DOI: 10.1111/j.1365-2958.2004.03916.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Most bacteria contain one type I signal peptidase (Spase I) for cleavage of signal peptides from exported and secreted proteins. Here, we identified a locus encoding three contiguous Spase I genes in the genome of Listeria monocytogenes. The deduced Sip proteins (denoted SipX, SipY and SipZ) are significantly similar to SipS and SipT, the major SPase I proteins of Bacillus subtilis (38% to 44% peptidic identity). We studied the role of these multiple signal peptidases in bacterial pathogenicity by constructing a series of single- and double-chromosomal knock-out mutants. Inactivation of sipX did not affect intracellular multiplication of L. monocytogenes but significantly reduced bacterial virulence (approximately 100-fold). Inactivation of sipZ impaired the secretion of phospholipase C (PC-PLC) and listeriolysin O (LLO), restricted intracellular multiplication and almost abolished virulence (LD(50) of 10(8.3)), inactivation of sipY had no detectable effects. Most importantly, a mutant expressing only SipX was impaired in intracellular survival and strongly attenuated in the mouse (LD(50) of 10(7.2)), whereas, a mutant expressing only SipZ behaved like wild-type EGD in all the assays performed. The data establish that SipX and SipZ perform distinct functions in bacterial pathogenicity and that SipZ is the major Spase I of L. monocytogenes. This work constitutes the first report on the differential role of multiple Spases I in a pathogenic bacterium and suggests a possible post-translational control mechanism of virulence factors expression.
Collapse
Affiliation(s)
- Claire Bonnemain
- INSERM U-570, CHU Necker-Enfants Malades, 156, rue de Vaugirard, 75730 Paris Cedex 15-France
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Milohanic E, Jonquières R, Glaser P, Dehoux P, Jacquet C, Berche P, Cossart P, Gaillard JL. Sequence and binding activity of the autolysin-adhesin Ami from epidemic Listeria monocytogenes 4b. Infect Immun 2004; 72:4401-9. [PMID: 15271896 PMCID: PMC470693 DOI: 10.1128/iai.72.8.4401-4409.2004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 02/26/2004] [Accepted: 04/09/2004] [Indexed: 11/20/2022] Open
Abstract
Ami is an autolytic amidase from Listeria monocytogenes that is targeted to the bacterial surface via its C-terminal cell wall anchoring (CWA) domain. We recently showed that the CWA domain from Ami of L. monocytogenes EGD (serovar 1/2a) (Ami 1/2a) mediated bacterial binding to mammalian cells. Here we studied the sequence and binding properties of Ami from CHUT 82337 (serovar 4b) (Ami 4b). The Ami 4b polypeptide is predicted to be 770 amino acids long (compared with the 917 amino acids of Ami 1/2a from EGD). Ami 1/2a and Ami 4b are almost identical in the N-terminal enzymatic domain (approximately 98% amino acid identity), but the sequence is poorly conserved in the C-terminal CWA domain, with only approximately 54% amino acid identity and eight GW modules in Ami 1/2a compared with six GW modules in Ami 4b. The purified Ami 4b CWA domain efficiently bound serovar 4b bacterial cells and only poorly bound serovar 1/2a bacterial cells. The Ami 4b CWA domain was also significantly less able to bind Hep-G2 human hepatocytic cells than the Ami 1/2a CWA domain. We sequenced the ami regions encoding CWA domains of reference strains belonging to the 12 L. monocytogenes serovars. The phylogenic tree constructed from the sequences yielded a binary division into group I (serovars 1/2a, 1/2b, 1/2c, 3a, 3b, 3c, and 7) and group II (serovars 4a, 4b, 4c, 4d, and 4e). This is the first direct evidence of divergence between serovars 1/2a and 4b in a gene involved in the adhesion of L. monocytogenes to mammalian cells, as well as the first demonstration of allelic polymorphism correlated with the somatic antigen in this species.
Collapse
Affiliation(s)
- Eliane Milohanic
- Laboratoire de Microbiologie, Institut National de la Santé et de la Recherche Médicale U 411, Faculté de Médecine Necker-Enfants Malades, 75730 Paris Cedex 15, France
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Bierne H, Garandeau C, Pucciarelli MG, Sabet C, Newton S, Garcia-del Portillo F, Cossart P, Charbit A. Sortase B, a new class of sortase in Listeria monocytogenes. J Bacteriol 2004; 186:1972-82. [PMID: 15028680 PMCID: PMC374393 DOI: 10.1128/jb.186.7.1972-1982.2004] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2003] [Accepted: 12/02/2003] [Indexed: 01/30/2023] Open
Abstract
Sortases are transamidases that covalently link proteins to the peptidoglycan of gram-positive bacteria. The genome of the pathogenic bacterium Listeria monocytogenes encodes two sortases genes, srtA and srtB. The srtA gene product anchors internalin and some other LPXTG-containing proteins to the listerial surface. Here, we focus on the role of the second sortase, SrtB. Whereas SrtA acts on most of the proteins in the peptidoglycan fraction, SrtB appears to target minor amounts of surface polypeptides. We identified one of the SrtB-anchored proteins as the virulence factor SvpA, a surface-exposed protein which does not contain the LPXTG motif. Therefore, as in Staphylococcus aureus, the listerial SrtB represents a second class of sortase in L. monocytogenes, involved in the attachment of a subset of proteins to the cell wall, most likely by recognizing an NXZTN sorting motif. The DeltasrtB mutant strain does not have defects in bacterial entry, growth, or motility in tissue-cultured cells and does not show attenuated virulence in mice. SrtB-mediated anchoring could therefore be required to anchor surface proteins involved in the adaptation of this microorganism to different environmental conditions.
Collapse
Affiliation(s)
- Hélène Bierne
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Réglier-Poupet H, Frehel C, Dubail I, Beretti JL, Berche P, Charbit A, Raynaud C. Maturation of lipoproteins by type II signal peptidase is required for phagosomal escape of Listeria monocytogenes. J Biol Chem 2003; 278:49469-77. [PMID: 12975369 DOI: 10.1074/jbc.m307953200] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lipoproteins of Gram-positive bacteria are involved in a broad range of functions such as substrate binding and transport, antibiotic resistance, cell signaling, or protein export and folding. Lipoproteins are also known to initiate both innate and adaptative immune responses. However, their role in the pathogenicity of intracellular microorganisms is yet poorly understood. In Listeria monocytogenes, a Gram-positive facultative intracellular human pathogen, surface proteins have important roles in the interactions of the microorganism with the host cells. Among the putative surface proteins of L. monocytogenes, lipoproteins constitute the largest family. Here, we addressed the role of the signal peptidase (SPase II), responsible for the maturation of lipoproteins in listerial pathogenesis. We identified a gene, lsp, encoding a SPase II in the genome of L. monocytogenes and constructed a deltalsp chromosomal deletion mutant. The mutant strain fails to process several lipoproteins demonstrating that lsp encodes a genuine SPase II. This defect is accompanied by a reduced efficiency of phagosomal escape during infection of eucaryotic cells, and leads to an attenuated virulence. We show that lsp gene expression is strongly induced when bacteria are still entrapped inside phagosomes of infected macrophages. The data presented establish, thus, that maturation of lipoproteins is critical for efficient phagosomal escape of L. monocytogenes, a process temporally controlled by the regulation of Lsp production in infected cells.
Collapse
Affiliation(s)
- Hélène Réglier-Poupet
- INSERM U570, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75730 Paris cedex 15, France
| | | | | | | | | | | | | |
Collapse
|
16
|
Mérino D, Réglier-Poupet H, Berche P, Charbit A. A hypermutator phenotype attenuates the virulence of Listeria monocytogenes in a mouse model. Mol Microbiol 2002; 44:877-87. [PMID: 11994166 DOI: 10.1046/j.1365-2958.2002.02929.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The integrity of the genetic material of bacteria is guaranteed by a set of distinct repair mechanisms. The participation of these repair systems in bacterial pathogenicity has been addressed only recently. Here, we study for the first time the participation in virulence of the MutSL mismatch repair system of Listeria monocytogenes. The mutS and mutL genes, which are contiguous in the L. monocytogenes chromosome, were identified after in silico analysis. The deduced MutS shares 62% identity with MutS of Bacillus subtilis and 50% identity with HexA, its homologue in Streptococcus pneumoniae; MutL shares 59% identity with MutL of B. subtilis and 47% identity with HexB of S. pneumoniae. Functional analysis of the mutSL locus was studied by constructing a double knock-out mutant. We showed that the deletion DeltamutSL induces: (i) a 100- to 1000-fold increase in the spontaneous mutation rate; and (ii) a 10- to 15-fold increase in the frequency of transduction, thus demonstrating the role of mutSL of L. monocytogenes in both mismatch repair and homologous recombination. We found that the deletion DeltamutSL moderately affected bacterial virulence, with a 1-log increase in the lethal dose 50% (LD50) in the mouse. Strikingly, repeated passages of the mutant strain in mice reduced virulence further. Competition assays between wild-type and mutant strains showed that the deletion DeltamutSL reduced the capacity of L. monocytogenes to survive and multiply in mice. These results thus demonstrate that, for the intracellular pathogen L. monocytogenes, a hypermutator phenotype is more deleterious than profitable to its virulence.
Collapse
Affiliation(s)
- Delphine Mérino
- INSERM U-411, CHU Necker-Enfants Malades, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| | | | | | | |
Collapse
|
17
|
Garandeau C, Réglier-Poupet H, Dubail I, Beretti JL, Berche P, Charbit A. The sortase SrtA of Listeria monocytogenes is involved in processing of internalin and in virulence. Infect Immun 2002; 70:1382-90. [PMID: 11854224 PMCID: PMC127754 DOI: 10.1128/iai.70.3.1382-1390.2002] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2001] [Revised: 10/17/2001] [Accepted: 12/03/2001] [Indexed: 01/22/2023] Open
Abstract
Listeria monocytogenes is an intracellular gram-positive human pathogen that invades eucaryotic cells. Among the surface-exposed proteins playing a role in this invasive process, internalin belongs to the family of LPXTG proteins, which are known to be covalently linked to the bacterial cell wall in gram-positive bacteria. Recently, it has been shown in Staphylococcus aureus that the covalent anchoring of protein A, a typical LPXTG protein, is due to a cysteine protease, named sortase, required for bacterial virulence. Here, we identified in silico from the genome of L. monocytogenes a gene, designated srtA, encoding a sortase homologue. The role of this previously unknown sortase was studied by constructing a sortase knockout mutant. Internalin was used as a reporter protein to study the effects of the srtA mutation on cell wall anchoring of this LPXTG protein in L. monocytogenes. We show that the srtA mutant (i) is affected in the display of internalin at the bacterial surface, (ii) is significantly less invasive in vitro, and (iii) is attenuated in its virulence in the mouse. These results demonstrate that srtA of L. monocytogenes acts as a sortase and plays a role in the pathogenicity.
Collapse
|
18
|
Vázquez-Boland JA, Kuhn M, Berche P, Chakraborty T, Domínguez-Bernal G, Goebel W, González-Zorn B, Wehland J, Kreft J. Listeria pathogenesis and molecular virulence determinants. Clin Microbiol Rev 2001; 14:584-640. [PMID: 11432815 PMCID: PMC88991 DOI: 10.1128/cmr.14.3.584-640.2001] [Citation(s) in RCA: 1506] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The gram-positive bacterium Listeria monocytogenes is the causative agent of listeriosis, a highly fatal opportunistic foodborne infection. Pregnant women, neonates, the elderly, and debilitated or immunocompromised patients in general are predominantly affected, although the disease can also develop in normal individuals. Clinical manifestations of invasive listeriosis are usually severe and include abortion, sepsis, and meningoencephalitis. Listeriosis can also manifest as a febrile gastroenteritis syndrome. In addition to humans, L. monocytogenes affects many vertebrate species, including birds. Listeria ivanovii, a second pathogenic species of the genus, is specific for ruminants. Our current view of the pathophysiology of listeriosis derives largely from studies with the mouse infection model. Pathogenic listeriae enter the host primarily through the intestine. The liver is thought to be their first target organ after intestinal translocation. In the liver, listeriae actively multiply until the infection is controlled by a cell-mediated immune response. This initial, subclinical step of listeriosis is thought to be common due to the frequent presence of pathogenic L. monocytogenes in food. In normal individuals, the continual exposure to listerial antigens probably contributes to the maintenance of anti-Listeria memory T cells. However, in debilitated and immunocompromised patients, the unrestricted proliferation of listeriae in the liver may result in prolonged low-level bacteremia, leading to invasion of the preferred secondary target organs (the brain and the gravid uterus) and to overt clinical disease. L. monocytogenes and L. ivanovii are facultative intracellular parasites able to survive in macrophages and to invade a variety of normally nonphagocytic cells, such as epithelial cells, hepatocytes, and endothelial cells. In all these cell types, pathogenic listeriae go through an intracellular life cycle involving early escape from the phagocytic vacuole, rapid intracytoplasmic multiplication, bacterially induced actin-based motility, and direct spread to neighboring cells, in which they reinitiate the cycle. In this way, listeriae disseminate in host tissues sheltered from the humoral arm of the immune system. Over the last 15 years, a number of virulence factors involved in key steps of this intracellular life cycle have been identified. This review describes in detail the molecular determinants of Listeria virulence and their mechanism of action and summarizes the current knowledge on the pathophysiology of listeriosis and the cell biology and host cell responses to Listeria infection. This article provides an updated perspective of the development of our understanding of Listeria pathogenesis from the first molecular genetic analyses of virulence mechanisms reported in 1985 until the start of the genomic era of Listeria research.
Collapse
Affiliation(s)
- J A Vázquez-Boland
- Grupo de Patogénesis Molecular Bacteriana, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Milohanic E, Jonquières R, Cossart P, Berche P, Gaillard JL. The autolysin Ami contributes to the adhesion of Listeria monocytogenes to eukaryotic cells via its cell wall anchor. Mol Microbiol 2001; 39:1212-24. [PMID: 11251838 DOI: 10.1111/j.1365-2958.2001.02208.x] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Adherence of pathogenic microorganisms to the cell surface is a key event during infection. We have previously reported the characterization of Listeria monocytogenes transposon mutants defective in adhesion to eukaryotic cells. One of these mutants had lost the ability to produce Ami, a 102 kDa autolytic amidase with an N-terminal catalytic domain and a C-terminal cell wall-anchoring domain made up of repeated modules containing the dipeptide GW ('GW modules'). We generated ami null mutations by plasmid insertion into L. monocytogenes strains lacking the invasion proteins InlA (EGDDeltainlA), InlB (EGDDeltainlB) or both (EGDDeltainlAB). These mutants were 5-10 times less adherent than their parental strains in various cell types. The adhesion capacity of the mutants was restored by complementation with a DNA fragment encoding the Ami cell wall-anchoring domain fused to the Ami signal peptide. The cell-binding activity of the Ami cell wall-anchoring domain was further demonstrated using the purified polypeptide. Growth of the ami null mutants constructed in EGD and EGDDeltainlAB backgrounds was attenuated in the livers of mice inoculated intravenously, indicating a role for Ami in L. monocytogenes virulence. Adhesive properties have recently been reported in the non-catalytic domain of two other autolysins, Staphylococcus epidermidis AtlE and Staphylococcus saprophyticus Aas. Interestingly, we found that these domains were also composed of repeated GW modules. Thus, certain autolysins appear to promote bacterial attachment by means of their GW repeat domains. These molecules may contribute to the colonization of host tissues by Gram-positive bacteria.
Collapse
Affiliation(s)
- E Milohanic
- Laboratoire de Microbiologie, Institut National de la Santé et de la Recherche Médicale U 411, Faculté de Médecine Necker-Enfants Malades, 75730 Paris Cedex 15, France
| | | | | | | | | |
Collapse
|
20
|
Nair S, Milohanic E, Berche P. ClpC ATPase is required for cell adhesion and invasion of Listeria monocytogenes. Infect Immun 2000; 68:7061-8. [PMID: 11083831 PMCID: PMC97816 DOI: 10.1128/iai.68.12.7061-7068.2000] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2000] [Accepted: 09/18/2000] [Indexed: 11/20/2022] Open
Abstract
We studied the role of two members of the 100-kDa heat shock protein family, the ClpC and ClpE ATPases, in cell adhesion and invasion of the intracellular pathogen Listeria monocytogenes. During the early phase of infection, a clpC mutant failed to disseminate to hepatocytes in the livers of infected mice whereas the invasive capacity of a clpE mutant remained unchanged. This was confirmed by a confocal microscopy study on infected cultured hepatocyte and epithelial cell lines, showing a strong reduction of cell invasion only by the clpC mutant. Western blot analysis with specific antisera showed that the absence of ClpC, but not that of ClpE, reduced expression of the virulence factors InlA, InlB, and ActA. ClpC-dependent modulation of these factors occurs at the transcriptional level with a reduction in the transcription of inlA, inlB, and actA in the clpC mutant, in contrast to the clpE mutant. This work provides the first evidence that, in addition to promoting escape from the phagosomes, ClpC is required for adhesion and invasion and modulates the expression of InlA, InlB, and ActA, further supporting the major role of the Clp chaperones in the virulence of intracellular pathogens.
Collapse
Affiliation(s)
- S Nair
- Unité de Physiopathologie Moléculaire des Infections Microbiennes, INSERM U411, Faculté de Médecine Necker, 75730 Paris Cedex 15, France
| | | | | |
Collapse
|
21
|
Alvarez-Domínguez C, Carrasco-Marín E, López-Mato P, Leyva-Cobián F. The contribution of both oxygen and nitrogen intermediates to the intracellular killing mechanisms of C1q-opsonized Listeria monocytogenes by the macrophage-like IC-21 cell line. Immunology 2000; 101:83-9. [PMID: 11012757 PMCID: PMC2327058 DOI: 10.1046/j.1365-2567.2000.00083.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Listeria monocytogenes is a facultative intracellular pathogen which is internalized by host mammalian cells upon binding to their surface. Further listerial growth occurs in the cytosol after escape from the phagosomal-endosomal compartment. We have previously reported that C1q is able to potentiate L. monocytogenes phagocytosis upon bacterial opsonization by ingestion through C1q-binding structures. In this report, we analysed the post-phagocytic events upon internalization of C1q-opsonized L. monocytogenes and found an induction of macrophage (Mphi)-like IC-21 cell bactericidal mechanisms displayed by the production of oxygen and nitrogen metabolites. Both types of molecules are effective in L. monocytogenes killing. Further analysis of the cellular responses promoted by interaction of C1q with its surface binding structures, leads us to consider C1q as a collaborative molecule involved in Mphi activation. Upon interaction with surface binding structures, C1q was able to trigger and/or amplify the production of reactive oxygen and nitrogen intermediates induced by stimuli such as interferon-gamma and L. monocytogenes phagocytosis.
Collapse
Affiliation(s)
- C Alvarez-Domínguez
- Servicio de Inmunología, Hospital Universitario 'Marqués de Valdecilla', Santander, Spain
| | | | | | | |
Collapse
|
22
|
Milohanic E, Pron B, Berche P, Gaillard JL. Identification of new loci involved in adhesion of Listeria monocytogenes to eukaryotic cells. European Listeria Genome Consortium. MICROBIOLOGY (READING, ENGLAND) 2000; 146 ( Pt 3):731-739. [PMID: 10746777 DOI: 10.1099/00221287-146-3-731] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Insertional mutagenesis was performed with Tn1545 in the genetic background of an inIAB deletion mutant to identify new adhesion determinants in Listeria monocytogenes. Four insertion mutants defective in adhesion to eukaryotic cells were identified. Insertion sites were cloned by inverse-PCR and sequenced. The genetic organization of insertion regions was further analysed by screening and sequencing DNA fragments from a HindIII library and by searching databases. Three adhesion-defective mutants each had one copy of Tn1545 inserted into their chromosome. The insertion sites were different in the three mutants: (i) upstream from two ORFs in tandem, similar to dfp and priA of Bacillus subtilis, respectively; (ii) within an ORF encoding a putative 126 amino-acid-polypeptide with no significant similarity to any known protein; (iii) within an ORF similar to a B. subtilis ORF with no known function, just upstream from an operon similar to an ABC (ATP-binding cassette) transporter operon from B. subtilis. The excisants obtained from these mutants using the excision reporter plasmid pTCR9 recovered full adhesion capacity. A fourth mutant was the most severely defective in adhesion. It had five Tn1545 insertions, one of which was upstream from dfp and priA, and another of which was upstream from ami, a gene encoding a surface-exposed autolysin with a C terminus similar to that of InIB. Ami was clearly involved because an ami null mutant constructed in an EGDdeltainIA-F background was adhesion-defective. Thus new regions involved in the adhesion of L. monocytogenes to eukaryotic cells were identified. Further study is required to define more accurately the roles of these regions in the adhesion process itself.
Collapse
Affiliation(s)
- Eliane Milohanic
- Laboratoire de Microbiologie, Institut National de la Santé et de la Recherche Médicale U 411, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75730 Paris Cedex 15, France1
| | - Bénédicte Pron
- Laboratoire de Microbiologie, Institut National de la Santé et de la Recherche Médicale U 411, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75730 Paris Cedex 15, France1
| | - Patrick Berche
- Laboratoire de Microbiologie, Institut National de la Santé et de la Recherche Médicale U 411, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75730 Paris Cedex 15, France1
| | - Jean-Louis Gaillard
- Laboratoire de Microbiologie, Hôpital Raymond Poincaré, 104 boulevard Raymond Poincaré, 92380 Garches, France2
| |
Collapse
|
23
|
Lajarin F, Rubio G, Lorenzo N, Gámiz P, Hernandez-Caselles T, Garcia-Peñarrubia P. Implication of reactive oxygen species in the antibacterial activity against Salmonella typhimurium of hepatocyte cell lines. Free Radic Biol Med 1999; 27:1008-18. [PMID: 10569633 DOI: 10.1016/s0891-5849(99)00148-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We recently described the antibacterial activity of a murine hepatocyte cell line stimulated with interferon-gamma (IFN-gamma), interleukin-1 (IL-1), and lipopolysaccharide (LPS) against intracellular Salmonella organisms. Here we show for the first time the existence of basal antibacterial activity in cultured hepatocyte cell lines. Thus treatment of resting and stimulated hepatocytes with catalase or superoxide dismutase increased bacterial number recovered per monolayer, which suggests that the mechanism involved with antibacterial activity of hepatocytes is mediated by reactive oxygen species (ROS). Also, the capacity of these cell lines to generate intracellular peroxides under resting and stimulated conditions was investigated. This revealed that IL-1 and LPS did not induce any increase in the amount of intracellular peroxides by themselves, but they primed IFN-gamma for maximal induction of peroxides. The intracellular amount of peroxides was highly increased on stimulation with IFN-gamma, IL-1, and LPS, and it was strongly inhibited by catalase. This explains that the mechanism whereby this enzyme inhibits antibacterial activity takes place by decreasing the intracellular pool of peroxides. In turn, experiments performed in the presence of several inhibitors of metabolic pathways involved in ROS generation suggested that cyclo-oxygenase are a source of these species in hepatocyte cell lines. These results attribute a prominent role to the generation of peroxides as effector molecules of antibacterial activity in hepatocyte cell lines. Thus these cells displayed a moderate basal level, which increased on stimulation with proinflammatory cytokines such as IFN-gamma, IL-1, and bacterial products such as LPS. Finally, it has been also shown for the first time that IFN-gamma stimulation induces production of peroxides in human and murine hepatocyte cell lines.
Collapse
Affiliation(s)
- F Lajarin
- Department of Biochemistry and Molecular Biology B and Immunology, School of Medicine, University of Murcia, Spain
| | | | | | | | | | | |
Collapse
|
24
|
Invasion of mammalian cells by Listeria monocytogenes. ADVANCES IN CELLULAR AND MOLECULAR BIOLOGY OF MEMBRANES AND ORGANELLES 1999. [DOI: 10.1016/s1874-5172(99)80006-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
25
|
Maganti S, Pierce MM, Hoffmaster A, Rodgers FG. The role of sialic acid in opsonin-dependent and opsonin-independent adhesion of Listeria monocytogenes to murine peritoneal macrophages. Infect Immun 1998; 66:620-6. [PMID: 9453618 PMCID: PMC107949 DOI: 10.1128/iai.66.2.620-626.1998] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The adhesion of listeriae to host cells employs mechanisms which are complex and not well understood. Listeria monocytogenes is a facultative intracellular pathogen responsible for meningoencephalitis, septicemia, and abortion in susceptible and immunocompromised individuals. Subsequent to colonization and penetration of the gut epithelium, the organism attaches to resident macrophages and replicates intracellularly, thus evading the humoral immune system of the infected host. The focus of these studies was to investigate the attachment of the organism to murine peritoneal macrophages in an opsonin-dependent and opsonin-independent fashion. Assessment of competitive binding experiments by immunofluorescence and enzyme-linked immunosorbent assays showed that adhesion of the organism to macrophages in the presence or absence of opsonins was inhibited (90%) by N-acetylneuraminic acid (NAcNeu). In addition, the lectin from Maackia amurensis, with affinity for NAcNeu-alpha(2,3)galactose, blocked binding of L. monocytogenes to host cells. Oxidation of the surface carbohydrates on the organism by using sodium metaperiodate resulted in a dose-dependent reduction (up to 98%) in adherence to macrophages. Monoclonal antibody to complement receptor 3 did not prevent listeriae from binding to mouse macrophages or from replicating within the infected cells whether or not normal mouse serum was present. Based on our results, we propose the involvement of NAcNeu, a member of the sialic acid group, in the attachment of L. monocytogenes to permissive murine macrophages.
Collapse
Affiliation(s)
- S Maganti
- Department of Microbiology, University of New Hampshire, Durham 03824-2617, USA
| | | | | | | |
Collapse
|
26
|
Unanue ER. Inter-relationship among macrophages, natural killer cells and neutrophils in early stages of Listeria resistance. Curr Opin Immunol 1997; 9:35-43. [PMID: 9039774 DOI: 10.1016/s0952-7915(97)80156-2] [Citation(s) in RCA: 161] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Reports in the past few years have shown the involvement of different cells and cytokines in controlling the infection with the intracellular facultative pathogen Listeria monocytogenes. A synergistic interaction of T-cell-independent and -dependent processes takes place but the nature of these interactions and of the relevant cells and cytokines depends on both the stage of the infection and the tissue that is involved.
Collapse
Affiliation(s)
- E R Unanue
- Department of Pathology, Center for Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St Louis, MO 63110, USA.
| |
Collapse
|
27
|
Alvarez-Domínguez C, Vázquez-Boland JA, Carrasco-Marín E, López-Mato P, Leyva-Cobián F. Host cell heparan sulfate proteoglycans mediate attachment and entry of Listeria monocytogenes, and the listerial surface protein ActA is involved in heparan sulfate receptor recognition. Infect Immun 1997; 65:78-88. [PMID: 8975895 PMCID: PMC174559 DOI: 10.1128/iai.65.1.78-88.1997] [Citation(s) in RCA: 153] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The mechanisms by which the intracellular pathogen Listeria monocytogenes interacts with the host cell surface remain largely unknown. In this study, we investigated the role of heparan sulfate proteoglycans (HSPG) in listerial infection. Pretreatment of bacteria with heparin or heparan sulfate (HS), but not with other glycosaminoglycans, inhibited attachment and subsequent uptake by IC-21 murine macrophages and CHO epithelial-like cells. Specific removal of HS from target cells with heparinase III significantly impaired listerial adhesion and invasion. Mutant CHO cells deficient in HS synthesis bound and internalized significantly fewer bacteria than wild-type cells did. Pretreatment of target cells with the HS-binding proteins fibronectin and platelet factor 4, or with heparinase III, impaired listerial infectivity only in those cells expressing HS. Moreover, a synthetic peptide corresponding to the HS-binding ligand in Plasmodium falciparum circumsporozoite protein (pepPf1) inhibited listerial attachment to IC-21 and CHO cells. A motif very similar to the HS-binding site of pepPf1 was found in the N-terminal region of ActA, the L. monocytogenes surface protein responsible for actin-based bacterial motility and cell-to-cell spread. In the same region of ActA, several clusters of positively charged amino acids which could function as HS-binding domains were identified. An ActA-deficient mutant was significantly impaired in attachment and entry due to altered HS recognition functions. This work shows that specific interaction with an HSPG receptor present on the surface of both professional and nonprofessional phagocytes is involved in L. monocytogenes cytoadhesion and invasion and strongly suggests that the bacterial surface protein ActA may be a ligand mediating HSPG receptor recognition.
Collapse
Affiliation(s)
- C Alvarez-Domínguez
- Servicio de Immunología, Hospital Universitario Marqués de Valdecilla,Instituto Nacional de la Salud, Santander, Spain
| | | | | | | | | |
Collapse
|
28
|
Abstract
Listeria monocytogenes is one of the leading foodborne pathogens and has been implicated in numerous outbreaks in the last 2 decades. Immunocompromised populations are usually the most susceptible to Listeria infections. Although the pathogenic mechanism is a complex process, significant progress has been made in unravelling the mechanism in recent years. It is now clear that numerous extracellular and cell-associated proteins, such as internalin, listeriolysin, actin polymerization protein, phospholipase, metalloprotease, and possibly p60 proteins, are essential for L. monocytogenes entry into mammalian cells, survival inside the phagosome, escape into the cytoplasm, and cell-to-cell spread. Other proteins may be responsible for growth and physiology or to maintain the structural integrity of the bacteria. Monoclonal and polyclonal antibodies have been developed against many of those antigens or their synthetic derivatives that have helped greatly to determine the structure and function of these antigens. The antibodies were also used for the diagnosis and detection, immunocytochemical staining, and serotyping of Listeria. Humoral immune response to live L. monocytogenes cells was examined in naturally or experimentally infected hosts. Studies revealed that only extracellular antigens induced the humoral response, whereas cell-associated antigens had apparently no response. It is speculated that during the occasional bacteremic phase, L. monocytogenes releases extracellular antigens that are then processed by the immune system for antibody production. As L. monocytogenes is an intracellular pathogen, the cell-associated antigens are not persistent in the blood circulation and thus fail to stimulate the humoral immune response.
Collapse
Affiliation(s)
- A K Bhunia
- Department of Food Science and Animal Industries, Alabama A&M University, Huntsville 35762, USA
| |
Collapse
|
29
|
Leal IS, Appelberg R, Silva MT. Neutrophils are involved in the non-specific resistance to listeriosis induced by mycobacterial infections. Immunol Suppl 1996; 89:442-8. [PMID: 8958060 PMCID: PMC1456549 DOI: 10.1046/j.1365-2567.1996.d01-758.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A major role has been recently ascribed to the neutrophil in the resistance to infection by Listeria monocytogenes (L. monocytogenes). Here we evaluated whether such neutrophils played a role in the non-specific resistance to listeriosis that develops in hosts infected by mycobacteria. We found that the depletion of neutrophils completely abrogated the resistance conferred by the activated macrophages induced during the mycobacterial infection. The lack of killing by activated Kupffer cells and the visualization of bacteria proliferating inside peritoneal macrophages in neutrophil-depleted mice allowed us to postulate a role for the cooperation between neutrophils and macrophages in the killing of L. monocytogenes. We also found listerial proliferation in hepatocytes of neutrophil-depleted, mycobacteria-infected mice showing that the neutrophils may be involved in the control of listeria infection of parenchymal cells.
Collapse
Affiliation(s)
- I S Leal
- Centro de Citologia Experimental, University of Porto, Portugal
| | | | | |
Collapse
|
30
|
Harty JT, Bevan MJ. CD8 T-cell recognition of macrophages and hepatocytes results in immunity to Listeria monocytogenes. Infect Immun 1996; 64:3632-40. [PMID: 8751910 PMCID: PMC174274 DOI: 10.1128/iai.64.9.3632-3640.1996] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
CD8 T cells are effective mediators of specific immunity to infection by Listeria monocytogenes, a bacterial pathogen that initially infects macrophages in the spleen and liver and subsequently spreads to hepatocytes and unidentified parenchymal cells in the spleen. To identify the in vivo target cells of L. monocytogenes-immune CD8 T cells, adoptive transfer assays were performed with bone marrow chimeric or transgenic host mice which had been manipulated to alter the major histocompatibility complex molecules expressed on macrophages or hepatocytes. L. monocytogenes-immune CD8 T cells mediate significant immunity in BDF1-->beta 2 M-/- chimeras, comparable to that seen in unmanipulated BDF1 recipients. L. monocytogenes-immune CD8 T cells also mediate significant antilisterial immunity in parent-->F1 chimeras when the CD8 T cells are syngeneic with the bone marrow donor. These data demonstrate that bone marrow-derived macrophages are major targets for L. monocytogenes-immune CD8 T cells in adoptive transfer assays. Interestingly, significant immunity was observed in parent-->F1 chimeras when the L. monocytogenes-immune CD8 T cells were not syngeneic with the bone marrow donor, suggesting that recognition of Listeria-infected non-bone-marrow-derived cells such as hepatocytes may also occur in vivo. Consistent with this possibility, H-2Kb-restricted CD8 T cells specific for the listeriolysin O molecule mediate significant immunity in the liver, but not the spleen, in transgenic mice expressing H-2Kb only on hepatocytes. In addition, Listeria-specific CD8 T cells lyse Listeria-infected hepatocyte-like cells in vitro. Thus, Listeria-infected hepatocytes can be recognized by CD8 T cells in vivo and in vitro.
Collapse
Affiliation(s)
- J T Harty
- Department of Microbiology, University of Iowa, Iowa City 52242, USA.
| | | |
Collapse
|
31
|
Gaillard JL, Jaubert F, Berche P. The inlAB locus mediates the entry of Listeria monocytogenes into hepatocytes in vivo. J Exp Med 1996; 183:359-69. [PMID: 8627149 PMCID: PMC2192459 DOI: 10.1084/jem.183.2.359] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The intracellular parasite Listeria monocytogenes is able to induce its internalization by cultured mammalian cells that are not normally phagocytic. This process requires the expression of the chromosomal locus inlAB. We studied the virulence of an inlAB mutant and of its parent in murine listeriosis. Irrespective of the route of inoculation, the inlAB mutant was severely attenuated for growth in the liver. The livers of mice inoculated with the inlAB mutant displayed much smaller infectious foci than the parent as early as 24 h after infection. Electron microscopy showed that these foci consisted of a few inflammatory cells, with few bacteria; bacteria were rarely found within hepatocytes. In contrast, foci in livers of mice inoculated with the parent consisted of islets of heavily infected hepatocytes that were infiltrated by numerous neutrophils; bacteria seemed intact within hepatocytes and damaged within neutrophils. A direct role of inlAB for the entry of L. monocytogenes into hepatocytes was confirmed in a cell infection system using the murine embryonic hepatocyte cell line TIB73. The inlAB mutant was approximately 20-fold less invasive in trans. The "invasion locus" inlAB contributes to protect L. monocytogenes from the host's innate defense mechanisms by promoting its entry into hepatocytes.
Collapse
Affiliation(s)
- J L Gaillard
- Laboratoire de Microbiologie, Institut National de la Sante et de la Recherche Medicale, Paris, France
| | | | | |
Collapse
|
32
|
Poyart C, Trieu-Cuot P, Berche P. The inlA gene required for cell invasion is conserved and specific to Listeria monocytogenes. MICROBIOLOGY (READING, ENGLAND) 1996; 142 ( Pt 1):173-180. [PMID: 8581163 DOI: 10.1099/13500872-142-1-173] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The Gram-positive bacterium Listeria monocytogenes can actively induce its own uptake by epithelial cells and fibroblasts through a surface-exposed 80 kDa protein, internalin (InIA), encoded by inIA. We studied the distribution and the DNA polymorphism of inIA sequences in a wide variety of wild strains of L. monocytogenes as compared to other Listeria species. This was done by PCR-amplifying inIA sequences encoding the fifteen repeats A and the three repeats B of InIA. inIA-repeated sequences were only found in L. monocytogenes. The amplified fragment of inIA encoding the repeats A displayed an AIuI DNA polymorphism which arises from point mutations. These results indicate that inIA required for cell invasion is specific to L. monocytogenes and that the intragenic repeats only exhibit a genetic heterogeneity due to point mutations and not to recombinations.
Collapse
Affiliation(s)
- Claire Poyart
- INSERM U.411, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75724 Paris Cedex 15, France
| | - Patrick Trieu-Cuot
- INSERM U.411, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75724 Paris Cedex 15, France
| | - Patrick Berche
- INSERM U.411, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75724 Paris Cedex 15, France
| |
Collapse
|
33
|
Dramsi S, Lebrun M, Cossart P. Molecular and genetic determinants involved in invasion of mammalian cells by Listeria monocytogenes. Curr Top Microbiol Immunol 1996; 209:61-77. [PMID: 8742246 DOI: 10.1007/978-3-642-85216-9_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- S Dramsi
- CNRS URA 1300, Institut Pasteur, Paris, France
| | | | | |
Collapse
|
34
|
Szalay G, Hess J, Kaufmann SH. Restricted replication of Listeria monocytogenes in a gamma interferon-activated murine hepatocyte line. Infect Immun 1995; 63:3187-95. [PMID: 7542640 PMCID: PMC173435 DOI: 10.1128/iai.63.8.3187-3195.1995] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The intracellular pathogen Listeria monocytogenes replicates mainly in resting macrophages and hepatocytes residing in infected tissues. Both innate and acquired resistance strongly depend on activation of listericidal capacities of macrophages by gamma interferon (IFN-gamma) produced by natural killer cells and T lymphocytes. In contrast to macrophages, hepatocytes have been considered to serve purely as a cellular habitat, prolonging survival of the pathogen in the host. By using an immortalized murine hepatocyte line, the relationship between L. monocytogenes and this cell type has been analyzed in more detail. Our data reveal that hepatocytes are able to eradicate listeriolysin-deficient (avirulent) L. monocytogenes but fail to control growth of listeriolysin-expressing (virulent) L. monocytogenes organisms. Following stimulation with IFN-gamma, hepatocytes gained the capacity to restrict growth of virulent L. monocytogenes, although less efficiently than the highly listericidal IFN-gamma-activated macrophages. Hepatocytes costimulated with a combination of IFN-gamma, interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-alpha) expressed the highest antilisterial activities. Although IFN-gamma-stimulated hepatocytes produced demonstrable levels of reactive nitrogen intermediates (RNI), the results of inhibition studies do not support a major role for these molecules in antilisterial hepatocyte activities. In contrast, inhibition of RNI produced by macrophages neutralized their antilisterial effects. IFN-gamma-stimulated, L. monocytogenes-infected hepatocytes expressed TNF-alpha mRNA, suggesting that they are a source of this cytokine during listeriosis. These studies suggest a novel function for hepatocytes in listeriosis: first, IFN-gamma-stimulated hepatocytes could contribute to listerial growth restriction in the liver, and second, through secretion of proinflammatory cytokines, they could promote phagocyte influx to the site of listerial growth.
Collapse
Affiliation(s)
- G Szalay
- Department of Immunology, University of Ulm, Germany
| | | | | |
Collapse
|
35
|
Abstract
Cytokine and cytokine receptor gene knockout mice provide powerful experimental systems to characterize the functions of these molecules in resistance to infectious disease. Such mice may also provide unique models of immune deficiency to learn whether manipulation of the immune response can overcome the specific dysfunction. We demonstrate that resistance of IFN gamma gene knockout (GKO-/-) mice to the intracellular bacterium Listeria monocytogenes is severely impaired compared with wild-type mice. However, immunization of GKO-/- mice with an attenuated L. monocytogenes strain generates antigen-specific CD8 T cell responses that can transfer immunity to naive hosts. Furthermore, vaccinated GKO-/- mice themselves exhibit 20,000-fold increased resistance to challenge with virulent L. monocytogenes and this resistance appears to be CD8 T cell mediated. These studies demonstrate that vaccination-induced immunity can overcome the absence of a cytokine that is critical for resistance to acute infection.
Collapse
Affiliation(s)
- J T Harty
- Department of Microbiology, University of Iowa, Iowa City 52242, USA
| | | |
Collapse
|
36
|
Nishibori T, Cooray K, Xiong H, Kawamura I, Fujita M, Mitsuyama M. Correlation between the presence of virulence-associated genes as determined by PCR and actual virulence to mice in various strains of Listeria spp. Microbiol Immunol 1995; 39:343-9. [PMID: 7565175 DOI: 10.1111/j.1348-0421.1995.tb02211.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Five chromosomal genes, prfA, plcA, hlyA, mpl and plcB, are implicated in the virulence of Listeria monocytogenes and some of these genes have been used for the identification of bacteria by polymerase chain reaction (PCR). Using 6 strains of L. monocytogenes and 3 L. innocua strains, the relationship was examined between the presence of five virulence-associated genes and actual virulence to mice in terms of 50% lethal dose (LD50), bacterial viability in the organ of infected mice and the intracellular growth in cultured macrophages. None of the five genes could be amplified by PCR in all the L. innocua strains and they were actually avirulent to mice. All L. monocytogenes strains were shown to be virulent and to have intact virulence-associated genes except for the strain ATCC15313. This particular strain was revealed to be avirulent and defective in hlyA and plcA in PCR amplification. It was suggested that PCR detection of genes prfA, mpl, or plcB may not be sufficient to detect virulent strains of L. monocytogenes. It appeared that the ability to produce listeriolysin O (LLO), which is encoded by hlyA, was critical for the expression of virulence regardless of the amount of LLO produced.
Collapse
Affiliation(s)
- T Nishibori
- Department of Bacteriology, Niigata University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Hess J, Gentschev I, Szalay G, Ladel C, Bubert A, Goebel W, Kaufmann SH. Listeria monocytogenes p60 supports host cell invasion by and in vivo survival of attenuated Salmonella typhimurium. Infect Immun 1995; 63:2047-53. [PMID: 7729919 PMCID: PMC173263 DOI: 10.1128/iai.63.5.2047-2053.1995] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The extracellular protein p60 is a major virulence factor of the intracellular bacterium Listeria monocytogenes. Its roles in pathogen survival in vivo and host cell invasion in vitro were studied. To this end, Salmonella typhimurium SL7207 was used as carrier for secreted p60-HlyA fusion protein by Escherichia coli HlyB and HlyD transport proteins. C57BL/6 mice infected intravenously with this strain suffered from increased bacterial numbers in livers and spleens compared with the p60-nonexpressing control strain, but only transiently. In vitro experiments showed that p60 promotes invasion of recombinant S. typhimurium SL7207 p60 into hepatocytes and resting macrophages independent from complement. Moreover, the uptake of wild-type L. monocytogenes EGD and L. monocytogenes BUG 8, an internalin-deficient strain, into hepatocytes was partially blocked by anti-p60 antibodies. The impaired invasion of dissociated bacterial chains of L. monocytogenes RIII, a p60 expression mutant, into hepatocytes and macrophages was partially restored by addition of p60- or p60-HlyA-enriched bacterial supernatants. These data suggest that the L. monocytogenes surface-associated proteins, p60 and internalin, act in concert to achieve optimal uptake into nonprofessional phagocytes and macrophages. Together, these experiments reveal a substantial impact of p60 on cell invasion and virulence and thus emphasize the importance of the intracellular habitat for survival of L. monocytogenes in the host.
Collapse
Affiliation(s)
- J Hess
- Department of Immunology, University of Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Boury NM, Czuprynski CJ. Listeria monocytogenes infection increases neutrophil adhesion and damage to a murine hepatocyte cell line in vitro. Immunol Lett 1995; 46:111-6. [PMID: 7590905 DOI: 10.1016/0165-2478(95)00027-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Several studies have reported that Listeria monocytogenes multiples within hepatocytes and that inflammatory neutrophils inhibit this intracellular growth in vivo. In the present study, we used a murine embryonic hepatocyte cell line (ATCC TIB73) as an in vitro model to investigate neutrophil-hepatocyte interactions. Murine peritoneal exudate neutrophils adhered more readily to L. monocytogenes-infected hepatocyte monolayers than to uninfected monolayers or monolayers infected with actA- and hly- mutants of L. monocytogenes. L. monocytogenes-infected TIB73 cells increased their surface expression of ICAM-1 as compared with uninfected TIB73 cells. Neutrophil adherence and oxidative stress to TIB73 cells were reduced by pre-incubating the hepatocyte monolayers with anti-ICAM-1 monoclonal antibody and diminished further by pre-incubating the peritoneal exudate neutrophils with an anti-CR3 monoclonal antibody.
Collapse
Affiliation(s)
- N M Boury
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison 53706, USA
| | | |
Collapse
|
39
|
Dramsi S, Biswas I, Maguin E, Braun L, Mastroeni P, Cossart P. Entry of Listeria monocytogenes into hepatocytes requires expression of inIB, a surface protein of the internalin multigene family. Mol Microbiol 1995; 16:251-61. [PMID: 7565087 DOI: 10.1111/j.1365-2958.1995.tb02297.x] [Citation(s) in RCA: 366] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The intracellular bacterium Listeria monocytogenes can invade several types of normally non-phagocytic cells. Entry into cultured epithelial cells requires the expression of inIA, the first gene of an operon, comprising two genes: inIA, which encodes internalin, an 800-amino-acid protein, and inIB, which encodes a 630-amino-acid protein. Several genes homologous to inIA are detected in the genome of L. monocytogenes; InIB is one of them. We have assessed the role of inIB in invasiveness of L. monocytogenes by constructing isogenic chromosomal deletion mutants in the inIAB locus. Our findings indicate that: i) inIB is required for entry of L. monocytogenes into hepatocytes, but not into intestinal epithelial cells; ii) inIB encodes a surface protein; iii) internalin plays a role for entry into some hepatocyte cell lines. These results provide the first insight into the cell tropism displayed by L. monocytogenes.
Collapse
Affiliation(s)
- S Dramsi
- Unite des Intéractions Bactéries-Cellules, Institut Pasteur, Paris, France
| | | | | | | | | | | |
Collapse
|
40
|
|
41
|
|
42
|
Langermans JAM, van Furth R. Cytokines and the host defense againstListeria monocytogenes andSalmonella typhimurium. ACTA ACUST UNITED AC 1994. [DOI: 10.1007/bf01878483] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
43
|
Langermans IA, van Furth R. Cytokines and the host defense against Listeria monocytogenes and Salmonella typhimurium. BIOTHERAPY (DORDRECHT, NETHERLANDS) 1994; 7:169-78. [PMID: 7865348 DOI: 10.1007/978-94-011-0233-9_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The host defense against intracellular pathogens depends largely on activation of phagocytes and is regulated by a complex network of cytokines. Modulation of this cytokine network might lead to new or additional therapies in the treatment of infections with intracellular pathogens. Therefore, insight in the role of various cytokines in the host defense against these pathogens is required. The present contribution summarizes the results of various studies on the role of different cytokines in the host defense against the intracellular bacteria Listeria monocytogenes and Salmonella typhimurium.
Collapse
Affiliation(s)
- I A Langermans
- Department of Infectious Diseases, University Hospital Leiden, The Netherlands
| | | |
Collapse
|