Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Mol Microbiol ; 53(2): 639-49, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15228540

ABSTRACT

Listeria monocytogenes is a Gram-positive intracellular bacterium responsible for severe opportunistic infections in humans and animals. Signature-tagged mutagenesis (STM) was used to identify a gene named fbpA, required for efficient liver colonization of mice inoculated intravenously. FbpA was also shown to be required for intestinal and liver colonization after oral infection of transgenic mice expressing human E-cadherin. fbpA encodes a 570-amino-acid polypeptide that has strong homologies to atypical fibronectin-binding proteins. FbpA binds to immobilized human fibronectin in a dose-dependent and saturable manner and increases adherence of wild-type L. monocytogenes to HEp-2 cells in the presence of exogenous fibronectin. Despite the lack of conventional secretion/anchoring signals, FbpA is detected using an antibody generated against the recombinant FbpA protein on the bacterial surface by immunofluorescence, and in the membrane compartment by Western blot analysis of cell extracts. Strikingly, FbpA expression affects the protein levels of two virulence factors, listeriolysin O (LLO) and InlB, but not that of InlA or ActA. FbpA co-immunoprecipitates with LLO and InlB, but not with InlA or ActA. Thus, FbpA, in addition to being a fibronectin-binding protein, behaves as a chaperone or an escort protein for two important virulence factors and appears as a novel multifunctional virulence factor of L. monocytogenes.


Subject(s)
Adhesins, Bacterial/genetics , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Virulence Factors/genetics , Virulence Factors/isolation & purification , Adhesins, Bacterial/physiology , Animals , Bacterial Adhesion , Bacterial Toxins/analysis , Cell Line , Cell Membrane/metabolism , Colony Count, Microbial , Disease Models, Animal , Fibronectins/metabolism , Genes, Bacterial , Heat-Shock Proteins/analysis , Hemolysin Proteins , Humans , Intestines/microbiology , Listeriosis/microbiology , Liver/microbiology , Lymph Nodes/microbiology , Membrane Proteins , Mice , Mice, Transgenic , Molecular Chaperones/genetics , Molecular Chaperones/physiology , Mutagenesis, Insertional , Open Reading Frames , Protein Binding , Sequence Homology, Amino Acid , Spleen/microbiology , Virulence/genetics
2.
Cell Microbiol ; 2(6): 465-76, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11207601

ABSTRACT

Entry of the bacterial pathogen Listeria monocytogenes into non-phagocytic mammalian cells is mainly mediated by the InlB protein. Here we show that in the human epithelial cell line HEp-2, the invasion protein InlB activates sequentially a p85beta-p110 class I(A) PI 3-kinase and the phospholipase C-gamma1 (PLC-gamma1) without detectable tyrosine phosphorylation of PLC-gamma1. Purified InlB stimulates association of PLC-gamma1 with one or more tyrosine-phosphorylated proteins, followed by a transient increase in intracellular inositol 1,4,5-trisphosphate (IP3) levels and a release of intracellular Ca2+ in a PI 3-kinase-dependent manner. Infection of HEp-2 cells with wild-type L. monocytogenes bacteria also induces association of PLC-gamma1 with phosphotyrosyl proteins. This interaction is undetectable upon infection with a deltainlB mutant revealing an InlB specific signal. Interestingly, pharmacological or genetic inactivation of PLC-gamma1 does not significantly affect InlB-mediated bacterial uptake, suggesting that InlB-mediated PLC-gamma1 activation and calcium mobilization are involved in post-internalization steps.


Subject(s)
Isoenzymes/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Membrane Proteins/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Type C Phospholipases/metabolism , Bacterial Proteins , Calcium/metabolism , Enzyme Activation , Epithelial Cells/microbiology , Humans , Listeria monocytogenes/physiology , Membrane Proteins/metabolism , Phospholipase C gamma , Phosphorylation , Tumor Cells, Cultured , Tyrosine/metabolism , Virulence
3.
Mol Microbiol ; 34(3): 552-61, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10564496

ABSTRACT

Most virulence genes of Listeria monocytogenes are activated by the transcriptional regulator PrfA. Previous studies have shown that environmental parameters, such as temperature, pH, stress conditions and medium composition, affect the expression of PrfA and PrfA-dependent proteins. In this report, we demonstrate a threefold increase in PrfA protein synthesis during infection of mammalian cells, which correlates with the increased activity of the plcA promoter, the major prfA promoter. Increased PrfA synthesis begins when L. monocytogenes adheres to host cells. In addition, we show that the observed induction of PrfA during the interaction of L. monocytogenes with mammalian cells can be reproduced in vitro using total cell extracts. Our data suggest a role for host proteinase K-sensitive protein(s) in PrfA upregulation.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Listeria monocytogenes/pathogenicity , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Bacterial Adhesion , Blotting, Western , Cell Extracts , Cell Line , Humans , Listeria/genetics , Listeria/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/growth & development , Mice , Peptide Termination Factors , Promoter Regions, Genetic , Transcription, Genetic , Up-Regulation , Virulence/genetics
4.
EMBO J ; 18(14): 3956-63, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10406800

ABSTRACT

Human E-cadherin promotes entry of the bacterial pathogen Listeria monocytogenes into mammalian cells by interacting with internalin (InlA), a bacterial surface protein. Here we show that mouse E-cadherin, although very similar to human E-cadherin (85% identity), is not a receptor for internalin. By a series of domain-swapping and mutagenesis experiments, we identify Pro16 of E-cadherin as a residue critical for specificity: a Pro-->Glu substitution in human E-cadherin totally abrogates interaction, whereas a Glu-->Pro substitution in mouse E-cadherin results in a complete gain of function. A correlation between cell permissivity and the nature of residue 16 in E-cadherins from several species is established. The location of this key specificity residue in a region of E-cadherin not involved in cell-cell adhesion and the stringency of the interaction demonstrated here have important consequences not only for the understanding of internalin function but also for the choice of the animal model to be used to study human listeriosis: mouse, albeit previously widely used, and rat appear as inappropriate animal models to study all aspects of human listeriosis, as opposed to guinea-pig, which now stands as a small animal of choice for future in vivo studies.


Subject(s)
Bacterial Proteins/metabolism , Cadherins/chemistry , Cadherins/metabolism , Listeria monocytogenes/growth & development , Listeria monocytogenes/pathogenicity , Proline/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Disease Models, Animal , Endocytosis , Female , Guinea Pigs , Humans , Listeriosis/microbiology , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Phylogeny , Proline/genetics , Protein Binding , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Species Specificity
5.
J Bacteriol ; 181(2): 418-25, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9882654

ABSTRACT

We have identified a novel gene, gtcA, involved in the decoration of cell wall teichoic acid of Listeria monocytogenes serotype 4b with galactose and glucose. Insertional inactivation of gtcA brought about loss of reactivity with the serotype 4b-specific monoclonal antibody c74.22 and was accompanied by a complete lack of galactose and a marked reduction in the amounts of glucose on teichoic acid. Interestingly, the composition of membrane-associated lipoteichoic acid was not affected. Complementation of the mutants with the cloned gtcA in trans restored galactose and glucose on teichoic acid to wild-type levels. The complemented strains also recovered reactivity with c74.22. Within L. monocytogenes, sequences homologous to gtcA were found in all serogroup 4 isolates but not in strains of any other serotypes. In serotype 4b, gtcA appears to be the first member of a bicistronic operon which includes a gene with homology to Bacillus subtilis rpmE, encoding ribosomal protein L31. In contrast to gtcA, the latter gene appears conserved among all screened serotypes of L. monocytogenes.


Subject(s)
Bacterial Proteins/genetics , Genes, Bacterial , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Teichoic Acids/metabolism , Amino Acid Sequence , Antibodies, Monoclonal , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carbohydrate Conformation , Carbohydrate Sequence , Cell Wall/metabolism , Cloning, Molecular , Glycosylation , Listeria monocytogenes/classification , Molecular Sequence Data , Open Reading Frames , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Serotyping , Teichoic Acids/biosynthesis , Teichoic Acids/chemistry
7.
Infect Immun ; 66(9): 4461-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9712801

ABSTRACT

Listeria monocytogenes is an intracellular pathogen that causes severe central nervous system infection in humans and animals. The ability of this bacterium to penetrate nerve cells was investigated by using rat spinal cell cultures. Entry into distinct cell types, i. e., glial cells and neurons, was monitored by a differential immunofluorescence technique with antibodies against cell type-specific markers and the bacterial pathogen. L. monocytogenes was detected predominantly within macrophages constituting the microglia. Astrocytes and oligodendrocytes, the major components of macroglia, were infected to a lesser extent. Surprisingly, Listeria innocua, a noninvasive and nonpathogenic species, also has the capacity to enter into these three types of glial cells. Entry into neurons was a very rare event. In contrast, we found that L. monocytogenes could efficiently invade neurons when these latter cells were cocultivated with Listeria-infected mouse macrophages. In this case, infection of neurons occurs by cell-to-cell spread via an actA-dependent mechanism. These data support the notion that infected phagocytes can be vectors by which L. monocytogenes gains access to privileged niches such as the central nervous system.


Subject(s)
Listeria monocytogenes/physiology , Neurons/microbiology , Animals , Cell Communication , Cell Line , Cells, Cultured , Mice , Neuroglia/cytology , Neuroglia/microbiology , Neurons/cytology , Rats , Spinal Cord/cytology
8.
Annu Rev Cell Dev Biol ; 14: 137-66, 1998.
Article in English | MEDLINE | ID: mdl-9891781

ABSTRACT

Many pathogens actively exploit the actin cytoskeleton during infection. This exploitation may take place during entry into mammalian cells after engagement of a receptor and/or as series of signaling events culminating in the engulfment of the microorganism. Although actin rearrangements are a common feature of most internalization events (e.g. entry of Listeria, Salmonella, Shigella, Yersinia, Neisseria, and Bartonella), bacterial and other cellular factors involved in entry are specific to each bacterium. Another step during which pathogens harness the actin cytoskeleton takes place in the cytosol, within which some bacteria (Listeria, Shigella, Rickettsia) or viruses (vaccinia virus) are able to move. Movement is coupled to a polarized actin polymerization process, with the formation of characteristic actin tails. Increasing attention has focused on this phenomenon due to its striking similarity to cellular events occurring at the leading edge of locomoting cells. Thus pathogens are convenient systems in which to study actin cytoskeleton rearrangements in response to stimuli at the plasma membrane or inside cells.


Subject(s)
Actins/physiology , Bacteria/pathogenicity , Bacterial Physiological Phenomena , Cytoskeleton/microbiology , Cytoskeleton/physiology , Animals , Cytoskeleton/ultrastructure , Genes, Bacterial , Humans , Movement , Vaccinia virus/physiology
9.
Mol Microbiol ; 25(2): 285-94, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9282740

ABSTRACT

Listeria monocytogenes is an intracellular bacterial pathogen that expresses several surface proteins critical for the infectious process. Such proteins include InlA (internalin) and InlB, involved in bacterial entry into the host cell, and ActA, required for bacterially induced actin-based motility. Although the molecular mechanisms of attachment of InlA and ActA have been characterized, essentially nothing is known about how InlB is anchored to the bacterial surface. Using a genetic approach, we demonstrate that the last 232 amino acids of InlB are both necessary and sufficient for anchoring this protein to the bacterial surface. An InlB mutant protein deleted for the last 232 amino acids was secreted and not detected at the cell surface. A 'domain-swapping' strategy in which these 232 amino acids were used to replace the normal cell wall-anchoring domain of InlA resulted in a chimeric protein that was anchored to the cell surface and able to confer entry. Interestingly, surface association of InlB also occurred when InlB was added externally to bacteria, suggesting that association may be able to occur after secretion. This association was productive for invasion, as it conferred bacterial entry into host cells. The C-terminal anchoring region in InlB contains 80-amino-acid repeats beginning with the sequence GW that is also present in a newly identified surface-associated bacteriolysin of L. monocytogenes, called Ami. Addition of GW repeats to the C-terminal of InlB improves anchoring of the protein to the cell surface. These and other data suggest that such 'GW' repeats may constitute a novel motif for cell-surface anchoring in Listeria and other Gram-positive bacteria. This motif may have important consequences for the release of surface proteins involved in interactions with eukaryotic cells.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Membrane Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Membrane Proteins/genetics , Molecular Sequence Data , Mutation
10.
Infect Immun ; 65(5): 1615-25, 1997 May.
Article in English | MEDLINE | ID: mdl-9125538

ABSTRACT

Listeria monocytogenes is a bacterial pathogen that is able to invade nonphagocytic cells. Two surface proteins, internalin, the inlA gene product, and InlB, play important roles in the entry into cultured mammalian cells. These proteins also have extensive sequence similarities. Previously, Southern hybridization predicted the existence of an internalin multigene family. Recently, InlC, a secreted protein of 30 kDa homologous to InlA and InlB, was identified. In this work, we identified and characterized four new members of the internalin multigene family, inlC2, inlD, inlE, and inlF which encode proteins of 548, 567, 499, and 821 amino acids respectively. inlC2, inlD, and inlE are contiguous on the chromosome of L. monocytogenes EGD, whereas inlF is located in a different chromosomal region. These four inl gene products display the principal features of internalin, namely, a signal sequence, two regions of repeats (or LRR and B repeats), and a putative cell wall anchor sequence containing the sorting motif LPXTG. The four inl genes were maximally expressed albeit at a low level during early exponential growth in bacterial medium at 37 degrees C. The role of these inl genes in L. monocytogenes invasion was assessed by constructing isogenic chromosomal deletion mutants and testing them for entry into various nonphagocytic cells. Unexpectedly, the inlC2, inlD, inlE, and inlF null mutants were not affected for entry into any of the cell lines tested, raising the possibility that these genes are needed for an aspect of pathogenicity other than invasion. The identity of such an aspect remains to be determined.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/physiology , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/microbiology , Amino Acid Sequence , Animals , Blotting, Southern , Cell Wall/genetics , Chromosome Mapping , Chromosomes/chemistry , Chromosomes/genetics , Cloning, Molecular , Colony Count, Microbial , Female , Gene Expression Regulation, Bacterial , Humans , Liver/microbiology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleic Acid Hybridization , Plasmids , Protein Sorting Signals , Recombination, Genetic , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Sequence Analysis, DNA , Sequence Deletion , Sequence Homology, Amino Acid , Spleen/microbiology , Transcription, Genetic , Virulence
11.
Infect Immun ; 65(4): 1515-8, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9119495

ABSTRACT

All virulence genes of Listeria monocytogenes identified to date are positively regulated by PrfA, a transcriptional activator belonging to the Crp-Fnr family. Low temperature and cellobiose are two environmental signals known to repress expression of virulence genes in L. monocytogenes. In the present work, we analyzed the effect of temperature and cellobiose on the expression of the PrfA protein. At low temperature, PrfA was undetected, although prfA monocistronic transcripts are present. In contrast, PrfA was fully expressed in the presence of cellobiose. These results strongly suggest that virulence gene activation depends on both the presence of PrfA and additional regulatory pathways that either modify PrfA or act synergistically with PrfA.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial , Listeria monocytogenes/genetics , Trans-Activators/genetics , Listeria monocytogenes/pathogenicity , Peptide Termination Factors , Transcriptional Activation , Virulence/genetics
13.
Mol Microbiol ; 18(5): 801-11, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8825084

ABSTRACT

We report the identification of a previously unknown Listeria monocytogenes gene, flaR, which modulates DNA topology. Through the analysis of a Tn917 non-motile mutant, LOSC1, in which production of flagellin was abolished, we have identified a bacterial component involved in gene regulation. The transposon had inserted in flaR, an open reading frame of 531 bp, followed by a second open reading frame of 1252 bp in reverse orientation. On the L. monocytogenes physical map, flaR was located in a different region from that of the flaA gene encoding flagellin. Transcriptional analysis showed that the flaR gene product affects the flaA expression and negatively regulates its own expression. When expressed in Escherichia coli, flaR encodes a protein of 18 kDa (FlaR) whose transcription is osmoregulated. In addition, FlaR also influences the expression of reporter genes containing supercoiling-sensitive promoters such as proU or ompC. The data presented here suggest that FlaR is a histone-like bacterial protein which acts at specific sites to influence DNA topology and, therefore, transcription. flaR is the first gene of this class to be described in Gram-positive pathogenic bacteria.


Subject(s)
Bacterial Proteins , DNA, Bacterial/chemistry , DNA-Binding Proteins/metabolism , Genes, Bacterial , Listeria monocytogenes/genetics , Nucleic Acid Conformation , Transcription Factors/metabolism , Amino Acid Sequence , Ampicillin/pharmacology , Base Sequence , Chromosome Mapping , Cloning, Molecular , DNA Transposable Elements , DNA, Superhelical/chemistry , DNA-Binding Proteins/genetics , Drug Resistance, Microbial/genetics , Flagellin/genetics , Gene Expression Regulation, Bacterial , Molecular Sequence Data , Mutation , Transcription Factors/genetics , Transcription, Genetic
14.
Mol Microbiol ; 16(2): 251-61, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7565087

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.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/pathogenicity , Liver/metabolism , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromosome Deletion , Colony Count, Microbial , Epithelium/metabolism , Female , Genes, Bacterial , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Liver/cytology , Liver/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutagenesis, Site-Directed , Tumor Cells, Cultured , Virulence
18.
Mol Microbiol ; 9(5): 931-41, 1993 Sep.
Article in English | MEDLINE | ID: mdl-7934921

ABSTRACT

Entry of Listeria monocytogenes into epithelial cells requires expression of inIA, the first gene of an operon comprising two genes: inIA, which encodes internalin, a 800-amino-acid protein, and inIB, which encodes a 630-amino-acid protein. We report here that the inI locus is transcribed on two transcripts in constant relative ratio: a 5 kb transcript spanning inIA and inIB, and a 2.9 kb transcript that covers only inIA. The promoter is located 397 bp from the GTG initiator of inIA and displays in its -35 region a palindrome similar to that found in promoters controlled by the pleiotropic activator prfA. Transcription of the inI locus is, as are several other L. monocytogenes virulence genes, activated by prfA and regulated by temperature--with higher expression at 37 degrees C versus 25 degrees C--and bacterial growth state. It is maximal during exponential growth and correlates with maximal invasivity of the bacteria in the human epithelial cell line Caco-2. It also correlates with maximum amounts of internalin present on the bacterial surface. Internalin is also detected in substantial amounts in culture supernatants. Taken together, these data suggest that surface-bound internalin plays an important role in bacterial entry but do not exclude a role for the released form.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/pathogenicity , Operon , Promoter Regions, Genetic , Trans-Activators/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Base Sequence , Cell Line , Colonic Neoplasms , Consensus Sequence , DNA Primers , Epithelium/microbiology , Gene Expression Regulation, Bacterial , Genes, Bacterial , Hot Temperature , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/growth & development , Molecular Sequence Data , Peptide Termination Factors , Polymerase Chain Reaction , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Temperature , Transcription, Genetic , Tumor Cells, Cultured
19.
Infect Immun ; 60(1): 219-30, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1309513

ABSTRACT

The lecithinase gene of the intracellular pathogen Listeria monocytogenes, plcB, was identified in a 5,648-bp DNA fragment which expressed lecithinase activity when cloned into Escherichia coli. This fragment is located immediately downstream of the previously identified gene mpl (prtA). It contains five open reading frames, named actA, plcB, and ORFX, -Y, and -Z, which, together with mpl, form an operon, since a 5.7-kb-long transcript originates from a promoter located upstream of mpl (J. Mengaud, C. Geoffroy, and P. Cossart, Infect. Immun. 59:1043-1049, 1991). A second promoter was detected in front of actA which encodes a putative membrane protein containing a region of internal repeats. plcB encodes the lecithinase, a predicted 289-amino-acid protein homologous to the phosphatidylcholine-specific phospholipases C of Bacillus cereus and Clostridium perfringens (alpha-toxin). plcB mutants produce only small plaques on fibroblast monolayers, and an electron microscopic analysis of infected macrophages suggests that lecithinase is involved in the lysis of the two-membrane vacuoles that surround the bacteria after cell-to-cell spread. On the opposite DNA strand, downstream of the operon, three more open reading frames, ldh, ORFA, and ORFB, were found. The deduced amino acid sequence of the first one is homologous to lactate dehydrogenases. Low-stringency Southern hybridization experiments suggest that these three open reading frames lie outside of the L. monocytogenes virulence region: mpl and actA were specific for L. monocytogenes, sequences hybridizing to plcB were detected in L. ivanovii and L. seeligeri, and sequences hybridizing to ORFX, -Y, and -Z were found in L. innocua. In contrast to this, sequences hybridizing to ldh or ORFB were detected in all Listeria species (including the nonpathogenic ones).


Subject(s)
Listeria monocytogenes/enzymology , Operon/genetics , Phospholipases/genetics , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/genetics , Base Sequence , Blotting, Southern , Cloning, Molecular , DNA Transposable Elements , In Vitro Techniques , Listeria monocytogenes/pathogenicity , Mice , Microscopy, Electron , Molecular Sequence Data , Phospholipases/physiology , Promoter Regions, Genetic/genetics , Restriction Mapping , Sequence Homology, Nucleic Acid , Viral Plaque Assay , Virulence/genetics
20.
Mol Microbiol ; 5(9): 2273-83, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1662763

ABSTRACT

Evidence for pleiotropic activation of virulence genes in Listeria monocytogenes is presented. A complementation study of a spontaneous prfA-deletion mutant and analysis of cassette and transposon insertion mutants showed that the gene prfA activates the transcription of four independent genes which code for a phosphatidyl-inositol-specific phospholipase C (gene plcA), listeriolysin O (gene hlyA), a metallo-protease (gene prtA) and a lecithinase (gene prtC). Transcription of prfA is not constitutive. During the growth phase, two peaks of prfA transcript accumulation were observed: the first was during exponential growth, and the second was at the beginning of the stationary phase. In addition, two prfA-specific transcripts of 2.2 kb and 1 kb are detected. Early in exponential growth, prfA is co-transcribed with plcA which lies upstream prfA, giving rise to the 2.2 kb plcA-prfA transcript. In late-exponential growth and at the beginning of the stationary phase, prfA transcripts of 1 kb are predominantly detected. Our results demonstrate that since prfA controls plcA transcription, it also regulates its own synthesis.


Subject(s)
Bacterial Toxins , Gene Expression Regulation, Bacterial , Listeria monocytogenes/genetics , Virulence/genetics , Amino Acid Sequence , Base Sequence , Chromosome Deletion , DNA Mutational Analysis , DNA Transposable Elements , Genetic Complementation Test , Heat-Shock Proteins/genetics , Hemolysin Proteins , Listeria monocytogenes/pathogenicity , Metalloendopeptidases/genetics , Molecular Sequence Data , Phospholipases/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Type C Phospholipases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...