Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
mBio ; 12(3): e0100821, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34076467

ABSTRACT

The Dot/Icm type IV secretion system (T4SS) of Legionella pneumophila is essential for lysosomal evasion and permissiveness of macrophages for intracellular proliferation of the pathogen. In contrast, we show that polymorphonuclear cells (PMNs) respond to a functional Dot/Icm system through rapid restriction of L. pneumophila. Specifically, we show that the L. pneumophila T4SS-injected amylase (LamA) effector catalyzes rapid glycogen degradation in the PMNs cytosol, leading to cytosolic hyperglucose. Neutrophils respond through immunometabolic reprogramming that includes upregulated aerobic glycolysis. The PMNs become activated with spatial generation of intracellular reactive oxygen species within the Legionella-containing phagosome (LCP) and fusion of specific and azurophilic granules to the LCP, leading to rapid restriction of L. pneumophila. We conclude that in contrast to macrophages, PMNs respond to a functional Dot/Icm system, and specifically to the effect of the injected amylase effector, through rapid engagement of major microbicidal processes and rapid restriction of the pathogen. IMPORTANCE Legionella pneumophila is commonly found in aquatic environments and resides within a wide variety of amoebal hosts. Upon aerosol transmission to humans, L. pneumophila invades and replicates with alveolar macrophages, causing pneumonia designated Legionnaires' disease. In addition to alveolar macrophages, neutrophils infiltrate into the lungs of infected patients. Unlike alveolar macrophages, neutrophils restrict and kill L. pneumophila, but the mechanisms were previously unclear. Here, we show that the pathogen secretes an amylase (LamA) enzyme that rapidly breakdowns glycogen stores within neutrophils, and this triggers increased glycolysis. Subsequently, the two major killing mechanisms of neutrophils, granule fusion and production of reactive oxygen species, are activated, resulting in rapid killing of L. pneumophila.


Subject(s)
Legionella pneumophila/immunology , Neutrophils/microbiology , Type IV Secretion Systems/immunology , Bacterial Proteins/metabolism , Cytosol/microbiology , Glycogen/metabolism , Glycolysis , Humans , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Legionnaires' Disease/microbiology , Phagosomes/immunology , Phagosomes/microbiology , Reactive Oxygen Species/immunology , Type IV Secretion Systems/genetics
2.
Biomolecules ; 11(1)2021 01 15.
Article in English | MEDLINE | ID: mdl-33467718

ABSTRACT

The ubiquitin pathway is highly conserved across the eukaryotic domain of life and plays an essential role in a plethora of cellular processes. It is not surprising that many intracellular bacterial pathogens often target the essential host ubiquitin pathway. The intracellular bacterial pathogen Legionella pneumophila injects into the host cell cytosol multiple classes of classical and novel ubiquitin-modifying enzymes that modulate diverse ubiquitin-related processes in the host cell. Most of these pathogen-injected proteins, designated as effectors, mimic known E3-ubiquitin ligases through harboring F-box or U-box domains. The classical F-box effector, AnkB targets host proteins for K48-linked polyubiquitination, which leads to excessive proteasomal degradation that is required to generate adequate supplies of amino acids for metabolism of the pathogen. In contrast, the SidC and SdcA effectors share no structural similarity to known eukaryotic ligases despite having E3-ubiquitin ligase activity, suggesting that the number of E3-ligases in eukaryotes is under-represented. L. pneumophila also injects into the host many novel ubiquitin-modifying enzymes, which are the SidE family of effectors that catalyze phosphoribosyl-ubiquitination of serine residue of target proteins, independently of the canonical E1-2-3 enzymatic cascade. Interestingly, the environmental bacterium, L. pneumophila, has evolved within a diverse range of amoebal species, which serve as the natural hosts, while accidental transmission through contaminated aerosols can cause pneumonia in humans. Therefore, it is likely that the novel ubiquitin-modifying enzymes of L. pneumophila were acquired by the pathogen through interkingdom gene transfer from the diverse natural amoebal hosts. Furthermore, conservation of the ubiquitin pathway across eukaryotes has enabled these novel ubiquitin-modifying enzymes to function similarly in mammalian cells. Studies on the biological functions of these effectors are likely to reveal further novel ubiquitin biology and shed further lights on the evolution of ubiquitin.


Subject(s)
Adaptation, Physiological , Amoeba/physiology , Biological Evolution , Host-Pathogen Interactions/physiology , Legionella pneumophila/physiology , Ubiquitination
3.
Sci Rep ; 7(1): 5123, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698607

ABSTRACT

Within macrophages and amoeba, the Legionella-containing vacuole (LCV) membrane is derived from the ER. The bona fide F-box AnkB effector protein of L. pneumophila strain AA100/130b is anchored to the cytosolic side of the LCV membrane through host-mediated farnesylation of its C-terminal eukaryotic "CaaX" motif. Here we show that the AnkB homologue of the Paris strain has a frame shift mutation that led to a loss of the CaaX motif and a concurrent generation of a unique C-terminal KNKYAP motif, which resembles the eukaryotic di-lysine ER-retention motif (KxKxx). Our phylogenetic analyses indicate that environmental isolates of L. pneumophila have a potential positive selection for the ER-retention KNKYAP motif. The AnkB-Paris effector is localized to the LCV membrane most likely through the ER-retention motif. Its ectopic expression in HEK293T cells localizes it to the perinuclear ER region and it trans-rescues the ankB mutant of strain AA100/130b in intra-vacuolar replication. The di-lysine ER retention motif of AnkB-Paris is indispensable for function; most likely as an ER retention motif that enables anchoring to the ER-derived LCV membrane. Our findings show divergent evolution of the ankB allele in exploiting either host farnesylation or the ER retention motif to be anchored into the LCV membrane.


Subject(s)
Ankyrins/chemistry , Ankyrins/genetics , Endoplasmic Reticulum/microbiology , Legionella/pathogenicity , Vacuoles/microbiology , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Endoplasmic Reticulum/metabolism , Frameshift Mutation , HEK293 Cells , Humans , Legionella/genetics , Lysine/metabolism , Phylogeny , Prenylation , Vacuoles/metabolism , Virulence Factors/chemistry , Virulence Factors/genetics
4.
Infect Immun ; 83(6): 2487-95, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25847958

ABSTRACT

Legionella pneumophila utilizes the Dot/Icm type IV translocation system to proliferate within a vacuole in a wide variety of natural amoebal hosts and in alveolar macrophages of the human accidental host. Although L. pneumophila utilizes host amino acids as the main sources of carbon and energy, it is not known whether de novo synthesis of amino acids by intravacuolar L. pneumophila contributes to its nutrition. The aroB and aroE genes encode enzymes for the shikimate pathway that generates the aromatic amino acids Phe, Trp, and Tyr. Here we show the aroB and aroE mutants of L. pneumophila to be defective in growth in human monocyte-derived macrophages (hMDMs) but not in Acanthamoeba spp. The aroB and aroE mutants are severely attenuated in intrapulmonary proliferation in the A/J mouse model of Legionnaires' disease, and the defect is fully complemented by the respective wild-type alleles. The two mutants grow normally in rich media but do not grow in defined media lacking aromatic amino acids, and the growth defect is rescued by inclusion of the aromatic amino acids, which are essential for production of the pyomelanin pigment. Interestingly, supplementation of infected hMDMs with the three aromatic amino acids or with Trp alone rescues the intramacrophage defect of the aroE but not the aroB mutant. Therefore, the shikimate pathway of L. pneumophila is differentially required for optimal growth within human macrophages, which are auxotrophic for Trp and Phe, but is dispensable for growth within the Acanthamoeba spp. that synthesize the aromatic amino acids.


Subject(s)
Acanthamoeba/microbiology , Legionella pneumophila/physiology , Macrophages/microbiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Amino Acids, Aromatic , Animals , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Humans , Mice , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , U937 Cells , Virulence
5.
PLoS One ; 9(12): e114914, 2014.
Article in English | MEDLINE | ID: mdl-25485627

ABSTRACT

BACKGROUND: Legionella pneumophila is an intracellular bacterial pathogen that invades and replicates within alveolar macrophages through injection of ∼ 300 effector proteins by its Dot/Icm type IV translocation apparatus. The bona fide F-box protein, AnkB, is a nutritional virulence effector that triggers macrophages to generate a surplus of amino acids, which is essential for intravacuolar proliferation. Therefore, the ankB mutant represents a novel genetic tool to determine the transcriptional response of human monocyte-derived macrophages (hMDMs) to actively replicating L. pneumophila. METHODOLOGY/PRINCIPAL FINDINGS: Here, we utilized total human gene microarrays to determine the global transcriptional response of hMDMs to infection by wild type or the ankB mutant of L. pneumophila. The transcriptomes of hMDMs infected with either actively proliferating wild type or non-replicative ankB mutant bacteria were remarkably similar. The transcriptome of infected hMDMs was predominated by up-regulation of inflammatory pathways (IL-10 anti-inflammatory, interferon signaling and amphoterin signaling), anti-apoptosis, and down-regulation of protein synthesis pathways. In addition, L. pneumophila modulated diverse metabolic pathways, particularly those associated with bio-active lipid metabolism, and SLC amino acid transporters expression. CONCLUSION/SIGNIFICANCE: Taken together, the hMDM transcriptional response to L. pneumophila is independent of intra-vacuolar replication of the bacteria and primarily involves modulation of the immune response and metabolic as well as nutritional pathways.


Subject(s)
Biomarkers/metabolism , Gene Expression Profiling , Legionella pneumophila/genetics , Legionnaires' Disease/genetics , Macrophages/metabolism , Monocytes/metabolism , Ankyrins/genetics , Cell Proliferation , Cells, Cultured , Humans , Legionella pneumophila/pathogenicity , Legionnaires' Disease/microbiology , Macrophages/cytology , Macrophages/microbiology , Monocytes/cytology , Monocytes/microbiology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vacuoles/genetics , Vacuoles/microbiology , Virulence/genetics
7.
Environ Microbiol ; 16(2): 350-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24112119

ABSTRACT

Legionella pneumophila, the causative agent of Legionnaires' disease, invades and proliferates within a diverse range of free-living amoeba in the environment, but upon transmission to humans, the bacteria hijack alveolar macrophages. Intracellular proliferation of L. pneumophila in two evolutionarily distant hosts is facilitated by bacterial exploitation of conserved host processes that are targeted by bacterial protein effectors injected into the host cell. A key aspect of microbe-host interaction is microbial extraction of nutrients from the host, but understanding of this is still limited. AnkB functions as a nutritional virulence factor and promotes host proteasomal degradation of polyubiquitinated proteins generating gratuitous levels of limiting host cellular amino acids. Legionella pneumophila is auxotrophic for several amino acids including cysteine, which is a metabolically preferred source of carbon and energy during intracellular proliferation, but is limiting in both amoebae and humans. We propose that synchronization of bacterial amino acids auxotrophy with the host is a driving force in pathogenic evolution and nutritional adaptation of L. pneumophila and other intracellular bacteria to life within the host cell. Understanding microbial strategies of nutrient generation and acquisition in the host will provide novel antimicrobial strategies to disrupt pathogen access to essential sources of carbon and energy.


Subject(s)
Adaptation, Physiological/genetics , Amino Acids/metabolism , Amoeba/microbiology , Biological Evolution , Host-Pathogen Interactions , Legionella pneumophila/physiology , Amoeba/metabolism , Ankyrins/genetics , Ankyrins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine/metabolism , Humans , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Macrophages/microbiology , Proteasome Endopeptidase Complex/metabolism , Ubiquitinated Proteins/metabolism , Vacuoles/microbiology , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Science ; 334(6062): 1553-7, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22096100

ABSTRACT

Legionella pneumophila proliferates in environmental amoeba and human cells within the Legionella-containing vacuole (LCV). The exported AnkB F-box effector of L. pneumophila is anchored into the LCV membrane by host-mediated farnesylation. Here, we report that host proteasomal degradation of Lys(48)-linked polyubiquitinated proteins, assembled on the LCV by AnkB, generates amino acids required for intracellular bacterial proliferation. The severe defect of the ankB null mutant in proliferation within amoeba and human cells is rescued by supplementation of a mixture of amino acids or cysteine, serine, pyruvate, or citrate, similar to rescue by genetic complementation. Defect of the ankB mutant in intrapulmonary proliferation in mice is rescued upon injection of a mixture of amino acids or cysteine. Therefore, Legionella promotes eukaryotic proteasomal degradation to generate amino acids needed as carbon and energy sources for bacterial proliferation within evolutionarily distant hosts.


Subject(s)
Amino Acids/metabolism , Legionella pneumophila/growth & development , Legionella pneumophila/metabolism , Proteasome Endopeptidase Complex/metabolism , Acanthamoeba/microbiology , Animals , Cell Proliferation , F-Box Proteins/genetics , F-Box Proteins/metabolism , HEK293 Cells , Humans , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , Lysine/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mice , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitinated Proteins/metabolism , Vacuoles/metabolism , Vacuoles/microbiology
10.
Front Microbiol ; 2: 266, 2011.
Article in English | MEDLINE | ID: mdl-22291682

ABSTRACT

To identify factors contributing to the ability of tubercle bacilli to grow in the lung during active infection, we analyzed RNA expression patterns in bacteria present in patient sputum. Prominent among bacterial transcripts identified were those encoding secreted peptides of the Esat-6 subfamily that includes EsxK and EsxL (Rv1197 and Rv1198). H37Rv esxKL and esxJI transcripts were differentially expressed under different growth conditions, and disruption of these genes altered growth phase kinetics in typical laboratory batch broth cultures. These growth defects, including the reduced intracellular growth of an ΔesxKL mutant in primary human macrophages, were reversed by either low multiplicity co-infection or co-culture with wild-type bacteria, demonstrating the ability of the secreted factors to rescue isogenic mutants. Complementing either only esxL or esxI alone (Rv1198 or Rv1037c) also reduced observed growth defects, indicating these genes encode factors capable of contributing to growth. Our studies indicate that the Mycobacterium tuberculosis Mtb9.9 family secreted factors EsxL and EsxI can act in trans to modulate growth of intracellular bacteria, and are highly expressed during active human lung infection.

11.
J Exp Med ; 207(8): 1713-26, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-20660614

ABSTRACT

Farnesylation involves covalent linkage of eukaryotic proteins to a lipid moiety to anchor them into membranes, which is essential for the biological function of Ras and other proteins. A large cadre of bacterial effectors is injected into host cells by intravacuolar pathogens through elaborate type III-VII translocation machineries, and many of these effectors are incorporated into the pathogen-containing vacuolar membrane by unknown mechanisms. The Dot/Icm type IV secretion system of Legionella pneumophila injects into host cells the F-box effector Ankyrin B (AnkB), which functions as platforms for the docking of polyubiquitinated proteins to the Legionella-containing vacuole (LCV) to enable intravacuolar proliferation in macrophages and amoeba. We show that farnesylation of AnkB is indispensable for its anchoring to the cytosolic face of the LCV membrane, for its biological function within macrophages and Dictyostelium discoideum, and for intrapulmonary proliferation in mice. Remarkably, the protein farnesyltransferase, RCE-1 (Ras-converting enzyme-1), and isoprenyl cysteine carboxyl methyltransferase host farnesylation enzymes are recruited to the LCV in a Dot/Icm-dependent manner and are essential for the biological function of AnkB. In conclusion, this study shows novel localized recruitment of the host farnesylation machinery and its anchoring of an F-box effector to the LCV membrane, and this is essential for biological function in vitro and in vivo.


Subject(s)
Ankyrins/metabolism , Eukaryotic Cells/metabolism , Eukaryotic Cells/microbiology , Legionella pneumophila/physiology , Protein Prenylation/physiology , Animals , Ankyrins/chemistry , Ankyrins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Proliferation , Dictyostelium/metabolism , Dictyostelium/microbiology , Endopeptidases/genetics , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Membranes/metabolism , Legionella pneumophila/cytology , Legionnaires' Disease/microbiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred Strains , Protein Interaction Domains and Motifs/genetics , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Protein Prenylation/drug effects , Protein Transport/drug effects , Protein Transport/genetics , RNA Interference , Transfection , U937 Cells , Ubiquitinated Proteins/metabolism , Vacuoles/metabolism , Vacuoles/microbiology
12.
Infect Immun ; 78(5): 2079-88, 2010 May.
Article in English | MEDLINE | ID: mdl-20194593

ABSTRACT

The Dot/Icm-translocated ankyrin B (AnkB) effector of Legionella pneumophila exhibits molecular mimicry of eukaryotic F-box proteins and is essential for intracellular replication in macrophages and protozoa. In addition to two eukaryotic-like ankyrin (ANK) domains, AnkB harbors a conserved eukaryotic F-box domain, which is involved in polyubiquitination of proteins throughout the eukaryotic kingdom. We have recently shown that the F-box domain of the AnkB effector is essential for decoration of the Legionella-containing vacuole (LCV) with polyubiquitinated proteins within macrophages and protozoan hosts. To decipher the role of the two ANK domains in the function of AnkB, we have constructed in-frame deletion of either or both of the ANK domain-encoding regions (ankB Delta A1, ankB Delta A2, and ankB Delta A1A2) to trans-complement the ankB null mutant. Deletion of the ANK domains results in defects in intracellular proliferation and decoration of the LCV with polyubiquitinated proteins. Export of the truncated variants of AnkB was reduced, and this may account for the observed defects. However, while full-length AnkB ectopically expressed in mammalian cells trans-rescues the ankB null mutant for intracellular proliferation, ectopic expression of AnkB Delta A1, AnkB Delta A2, and AnkB Delta A1A2 fails to trans-rescue the ankB null mutant. Importantly, ectopically expressed full-length AnkB is targeted to the host cell plasma membrane, where it recruits polyubiquitinated proteins. In contrast, AnkB Delta A1, AnkB Delta A2, and AnkB Delta A1A2 are diffusely distributed throughout the cytosol and fail to recruit polyubiquitinated proteins. We conclude that the two eukaryotic-like ANK domains of AnkB are essential for intracellular proliferation, for targeting AnkB to the host membranes, and for decoration of the LCV with polyubiquitinated proteins.


Subject(s)
Amoeba/microbiology , Ankyrins/physiology , Bacterial Proteins/physiology , Host-Pathogen Interactions , Legionella pneumophila/pathogenicity , Macrophages/microbiology , Virulence Factors/physiology , Cell Membrane/chemistry , Gene Deletion , Genetic Complementation Test , Humans , Sequence Deletion
13.
Front Microbiol ; 1: 131, 2010.
Article in English | MEDLINE | ID: mdl-21687755

ABSTRACT

The Dot/Icm type IV translocated Ankyrin B (AnkB) effector of Legionella pneumophila is modified by the host prenylation machinery that anchors it into the outer leaflet of the Legionella-containing vacuole (LCV), which is essential for biological function of the effector in vitro and in vivo. Prenylation involves the covalent linkage of an isoprenoid lipid moiety to a C-terminal CaaX motif in eukaryotic proteins enabling their anchoring into membranes. We show here that the LCV harboring an ankB null mutant is decorated with prenylated proteins in a Dot/Icm-dependent manner, indicating that other LCV membrane-anchored proteins are prenylated. In silico analyses of four sequenced L. pneumophila genomes revealed the presence of eleven other genes that encode proteins with a C-terminal eukaryotic CaaX prenylation motif. Of these eleven designated Prenylated effectors of Legionella (Pel), seven are also found in L. pneumophila AA100. We show that six L. pneumophila AA100 Pel proteins exhibit distinct cellular localization when ectopically expressed in mammalian cells and this is dependent on action of the host prenylation machinery and the conserved cysteine residue of the CaaX motif. Although inhibition of the host prenylation machinery completely blocks intra-vacuolar proliferation of L. pneumophila, it only had a modest effect on intracellular trafficking of the LCV. Five of the Pel proteins are injected into human macrophages by the Dot/Icm type IV translocation system of L. pneumophila. Taken together, the Pel proteins are novel Dot/Icm-translocated effectors of L. pneumophila that are post-translationally modified by the host prenylation machinery, which enables their anchoring into cellular membranes, and the prenylated effectors contribute to evasion of lysosomal fusion by the LCV.

14.
Front Microbiol ; 1: 122, 2010.
Article in English | MEDLINE | ID: mdl-21687758

ABSTRACT

Microbial pathogens have evolved exquisite mechanisms to interfere and intercept host biological processes, often through molecular mimicry of specific host proteins. Ubiquitination is a highly conserved eukaryotic post-translational modification essential in determining protein fate, and is often hijacked by pathogenic bacteria. The conserved SKP1/CUL1/F-box (SCF) E3 ubiquitin ligase complex plays a key role in ubiquitination of proteins in eukaryotic cells. The F-box protein component of the SCF complex provides specificity to ubiquitination by binding to specific cellular proteins, targeting them to be ubiquitinated by the SCF complex. The bacterial pathogens. Legionella pneumophila, Agrobacterium tumefaciens, and Ralstonia solanacearum utilize type III or IV translocation systems to inject into the host cell eukaryotic-like F-box effectors that interact with the host SKP1 component of the SCF complex to trigger ubiquitination of specific host cells targets, which is essential to promote proliferation of these pathogens. Our bioinformatic analyses have identified at least 74 genes encoding putative F-box proteins belonging to 22 other bacterial species, including human pathogens, plant pathogens, and amebal endosymbionts. Therefore, subversion of the host ubiquitination machinery by bacterial F-box proteins may be a widespread strategy amongst pathogenic bacteria. The findings that bacterial F-box proteins harbor Ankyrin repeats as protein-protein interaction domains, which are present in F-box proteins of primitive but not higher eukaryotes, suggest acquisition of many bacterial F-box proteins from primitive eukaryotic hosts rather than the mammalian host.

15.
J Bacteriol ; 190(11): 3955-61, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18390665

ABSTRACT

Mycobacterium tuberculosis maintains a large genetic capacity necessary for growth in different environments during infection and survival upon aerosol transmission to new hosts. Screening for bacterial RNAs produced in response to host interactions produced candidate lists where we noted proXVWZ, annotated as encoding a putative glycine betaine or proline transporter. As high surface-to-volume ratios make bacterial cells particularly vulnerable to changes in water availability, we investigated the contributions of this transporter to the ability of M. tuberculosis to colonize macrophages. An H37Rv proXVWZ mutant was impaired for initial survival and intracellular growth and exhibited reduced growth at elevated medium osmolarity. This defect could be complemented by restoring proXVWZ and was attributable to a failure to accumulate the compatible solute glycine betaine. We then demonstrated that ProXVWZ allows M. tuberculosis to obtain betaine from host macrophages and thereby contributes to early steps in colonizing this niche.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Betaine/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Gene Expression Regulation, Bacterial/physiology , Humans , Mice , Mice, Inbred C57BL , Mutation , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Osmolar Concentration , Osmotic Pressure , Phagocytosis/physiology , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Time Factors
16.
Retina ; 27(8): 1125-30, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18040257

ABSTRACT

PURPOSE: The authors examined the effect of blood on susceptibility to experimental endophthalmitis. METHODS: Forty rabbits received an injection of 5-25 colony-forming units of Staphylococcus epidermidis into the vitreous of the right eye. Twenty of these same eyes received a subsequent intravitreal injection of 0.2 mL blood while the remaining 20 received an intravitreal injection of 0.2 mL of a salt solution. All eyes were examined daily for signs of endophthalmitis. Vitreous cultures were obtained on day 2 from 30 of the 40 rabbits. Twenty rabbits were assigned for culture and euthanasia at day 5 and those remaining were cultured and killed at day 7. RESULTS: In rabbits with blood and bacteria, 10 of 15 (67%) were culture positive at 2 days, compared to 2 of 15 (13%) that received salt solution and bacteria (P < 0.01). At days 5 and 7 there was no statistically significant difference in culture results. However, inflammatory scores were significantly higher at days 3-7 in rabbits with blood compared to those with salt solution (P

Subject(s)
Blood , Endophthalmitis/microbiology , Eye Infections, Bacterial/microbiology , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/physiology , Vitreous Body/microbiology , Animals , Bacteriological Techniques , Disease Models, Animal , Disease Susceptibility , Male , Rabbits , Specific Pathogen-Free Organisms
17.
FEMS Microbiol Lett ; 237(2): 297-302, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15321676

ABSTRACT

The sarA locus of Staphylococccus aureus regulates the synthesis of over 100 genes on the S. aureus chromosome. We now report the effects of sarA inactivation on intrinsic multidrug resistance expression by S. aureus. In a strain-dependent fashion, sarA::kan mutants of three unrelated strains of S. aureus demonstrated significantly increased susceptibility to five or more of the following substances: the antibiotics ciprofloxacin, fusidic acid, and vancomycin; the DNA-intercalating agent ethidium; and four common household cleaner formulations. In addition, all three sarA::kan mutants demonstrated significantly increased accumulation of ciprofloxacin and one sarA::kan mutant demonstrated increased ethidium accumulation. Our data therefore indicate that sarA plays a role in the intrinsic multidrug resistance mechanism expressed by S. aureus, in part by regulating drug accumulation.


Subject(s)
Bacterial Proteins/physiology , Drug Resistance, Multiple, Bacterial , Staphylococcus aureus/drug effects , Trans-Activators/physiology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Ciprofloxacin/metabolism , Ciprofloxacin/pharmacology , Ethidium/metabolism , Mutation , Staphylococcus aureus/cytology , Trans-Activators/genetics , Vancomycin/pharmacology
18.
Appl Environ Microbiol ; 68(11): 5417-21, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12406732

ABSTRACT

Mutants of Staphylococcus aureus strain COL resistant to a household pine oil cleaner (POC) were isolated on laboratory media containing POC. S. aureus mutants expressing the POC resistance (POC(r)) phenotype also demonstrate reduced susceptibility to the cell wall-active antibiotics vancomycin and oxacillin. The POC(r) phenotype is reliant on the S. aureus alternative transcription factor SigB, since inactivation of sigB abolished expression of elevated POC resistance and the reductions in vancomycin and oxacillin susceptibilities. The isolation of suppressor mutants of COLsigB::kan, which maintain the sigB::kan allele, indicates that the POC(r) phenotype can also be expressed to a lesser degree via a sigB-independent mechanism. These results bolster a growing body of reports suggesting that common disinfectants can select for bacteria with reduced susceptibilities to antibiotics. A series of in vitro-selected glycopeptide-intermediate S. aureus (GISA) isolates also expressed reductions in POC susceptibility compared to parent strains. Viewed collectively, our evidence suggests that mutations leading to the POC(r) phenotype may also be involved with the mechanism that leads to the GISA phenotype.


Subject(s)
Bacterial Proteins/metabolism , Drug Resistance, Bacterial/physiology , Oxacillin/pharmacology , Pinus/chemistry , Plant Oils/pharmacology , Sigma Factor/metabolism , Staphylococcus aureus/drug effects , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Staphylococcus aureus/isolation & purification
19.
J Med Microbiol ; 50(1): 104-106, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11192496

ABSTRACT

Salicylate was shown to increase the frequency at which a fusidic acid-susceptible strain of Staphylococcus aureus underwent mutation to become fusidic acid-resistant. These fusidic acid-resistant mutants had alterations in spectinomycin and kanamycin resistance levels indicative of mutations in fusA, the gene that encodes elongation factor-G, the target of fusidic acid.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fusidic Acid/pharmacology , Mutation , Staphylococcus aureus/drug effects , Drug Resistance, Microbial/genetics , Microbial Sensitivity Tests , Sodium Salicylate/pharmacology , Staphylococcus aureus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...