首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Bacillus anthracis spores are enclosed by an exosporium comprised of a basal layer and an external hair-like nap. The filaments of the nap are composed of trimers of the collagen-like glycoprotein BclA. The attachment of essentially all BclA trimers to the exosporium requires the basal layer protein BxpB, and both proteins are included in stable high-molecular-mass exosporium complexes. BclA contains a proteolytically processed 38-residue amino-terminal domain (NTD) that is essential for basal-layer attachment. In this report, we identify three NTD submotifs (SM1a, SM1b, and SM2, located within residues 21 to 33) that are important for BclA attachment and demonstrate that residue A20, the amino-terminal residue of processed BclA, is not required for attachment. We show that the shortest NTD of BclA—or of a recombinant protein—sufficient for high-level basal-layer attachment is a 10-residue motif consisting of an initiating methionine, an apparently arbitrary second residue, SM1a or SM1b, and SM2. We also demonstrate that cleavage of the BclA NTD is necessary for efficient attachment to the basal layer and that the site of cleavage is somewhat flexible, at least in certain mutant NTDs. Finally, we propose a mechanism for BclA attachment and discuss the possibility that analogous mechanisms are involved in the attachment of many different collagen-like proteins of B. anthracis and closely related Bacillus species.Bacillus anthracis, a Gram-positive, rod-shaped, aerobic bacterium, is the causative agent of anthrax (17). When vegetative cells of B. anthracis are starved for certain essential nutrients, they form dormant spores that can survive in harsh soil environments for many years (12, 19). Spore formation starts with asymmetric septation that divides the starved vegetative cell into two genome-containing compartments, a mother cell compartment and a smaller forespore compartment. The mother cell then engulfs the forespore and surrounds it with three protective layers: a cortex composed of peptidoglycan, a closely apposed proteinaceous coat, and a loosely fitting exosporium (11). After a spore maturation stage, the mother cell lyses and releases the mature spore. When spores encounter an aqueous environment containing nutrients, they can germinate and grow as vegetative cells (18). Anthrax is typically caused by contact with spores (17).The outermost layer of B. anthracis spores, the exosporium, has been studied intensively in recent years because it is both the first point of contact with the immune system of an infected host and the target of new detectors for agents of bioterrorism (21, 28, 32). The exosporium of B. anthracis and closely related pathogenic species, such as Bacillus cereus and Bacillus thuringiensis, is a prominent structure consisting of a paracrystalline basal layer and an external hair-like nap (1, 9). The filaments of the nap are formed by trimers of the collagen-like glycoprotein BclA (2, 29). Recent studies suggest that BclA plays a major role in pathogenesis by directing spores to professional phagocytic cells, a critical step in disease progression (4, 21). The basal layer is composed of approximately 20 different proteins (23, 25, 26), several of which have been shown to play key roles in exosporium assembly (3, 13, 27). One of these proteins is BxpB (also called ExsFA) (25, 30, 34), which is required for the attachment of approximately 98% of spore-bound BclA to the basal layer (26, 30). Residual BclA attachment requires the basal layer protein ExsFB, a paralog of BxpB (30).BclA contains three distinct domains: a 38-residue amino-terminal domain (NTD), a central collagen-like region containing a strain-specific number of XXG (mostly PTG) repeats, and a 134-residue carboxyl-terminal domain (CTD) (25, 29, 31). The CTD apparently functions as the major nucleation site for trimerization of BclA (24), and CTD trimers form the globular distal ends of the filaments in the nap (2). The highly extended collagen-like region is extensively glycosylated (5), and its length determines the depth of the nap (2, 31). The NTD is the site of attachment of BclA to the basal layer, and deletion of the NTD prevents this attachment (2). The NTD is normally proteolytically processed to remove the first 19 amino acids, and it is this mature form of BclA that is attached to the basal layer (25, 29). In an earlier report, we suggested that NTD processing of BclA is required for basal-layer attachment, perhaps through a direct covalent linkage to BxpB (26).Recently, Thompson and Stewart identified conserved 11-residue sequences in the NTDs of BclA and the minor B. anthracis collagen-like glycoprotein BclB and showed that these sequences are involved in the incorporation of BclA and BclB into the exosporium. These investigators used a truncated BclA NTD that lacked residues 2 through 19 but included the conserved 11-amino-acid sequence to target enhanced green fluorescent protein (EGFP) to the surface of the developing forespore (33). Thompson and Stewart also reported that cleavage of the BclA NTD occurred after its association with the forespore and suggested that this cleavage was involved indirectly in the attachment process. Actual cleavage sites were not determined in these studies, however. We have performed related studies of the attachment of BclA to the exosporium that provide a more detailed and somewhat different view of this process. In our studies, which are reported here, we identified short segments, or submotifs, of the BclA NTD that can be arranged in different combinations to produce 10-amino-acid motifs sufficient for tight attachment of BclA, and probably most proteins, to the exosporium basal layer. Additionally, we present direct evidence showing that BclA NTD cleavage is required for efficient attachment to the basal layer and that selection of the cleavage site can be somewhat flexible. Finally, we discuss a possible mechanism for BclA attachment and the likelihood that similar mechanisms are used for attachment of many different collagen-like proteins of B. anthracis and closely related Bacillus species.  相似文献   

2.
The sequestration of iron by mammalian hosts represents a significant obstacle to the establishment of a bacterial infection. In response, pathogenic bacteria have evolved mechanisms to acquire iron from host heme. Bacillus anthracis, the causative agent of anthrax, utilizes secreted hemophores to scavenge heme from host hemoglobin, thereby facilitating iron acquisition from extracellular heme pools and delivery to iron-regulated surface determinant (Isd) proteins covalently attached to the cell wall. However, several Gram-positive pathogens, including B. anthracis, contain genes that encode near iron transporter (NEAT) proteins that are genomically distant from the genetically linked Isd locus. NEAT domains are protein modules that partake in several functions related to heme transport, including binding heme and hemoglobin. This finding raises interesting questions concerning the relative role of these NEAT proteins, relative to hemophores and the Isd system, in iron uptake. Here, we present evidence that a B. anthracis S-layer homology (SLH) protein harboring a NEAT domain binds and directionally transfers heme to the Isd system via the cell wall protein IsdC. This finding suggests that the Isd system can receive heme from multiple inputs and may reflect an adaptation of B. anthracis to changing iron reservoirs during an infection. Understanding the mechanism of heme uptake in pathogenic bacteria is important for the development of novel therapeutics to prevent and treat bacterial infections.Pathogenic bacteria need to acquire iron to survive in mammalian hosts (12). However, the host sequesters most iron in the porphyrin heme, and heme itself is often bound to proteins such as hemoglobin (14, 28, 85). Circulating hemoglobin can serve as a source of heme-iron for replicating bacteria in infected hosts, but the precise mechanisms of heme extraction, transport, and assimilation remain unclear (25, 46, 79, 86). An understanding of how bacterial pathogens import heme will lead to the development of new anti-infectives that inhibit heme uptake, thereby preventing or treating infections caused by these bacteria (47, 68).The mechanisms of transport of biological molecules into a bacterial cell are influenced by the compositional, structural, and topological makeup of the cell envelope. Gram-negative bacteria utilize specific proteins to transport heme through the outer membrane, periplasm, and inner membrane (83, 84). Instead of an outer membrane and periplasm, Gram-positive bacteria contain a thick cell wall (59, 60). Proteins covalently anchored to the cell wall provide a functional link between extracellular heme reservoirs and intracellular iron utilization pathways (46). In addition, several Gram-positive and Gram-negative bacterial genera also contain an outermost structure termed the S (surface)-layer (75). The S-layer is a crystalline array of protein that surrounds the bacterial cell and may serve a multitude of functions, including maintenance of cell architecture and protection from host immune components (6, 7, 18, 19, 56). In bacterial pathogens that manifest an S-layer, the “force field” function of this structure raises questions concerning how small molecules such as heme can be successfully passed from the extracellular milieu to cell wall proteins for delivery into the cell cytoplasm.Bacillus anthracis is a Gram-positive, spore-forming bacterium that is the etiological agent of anthrax disease (30, 33). The life cycle of B. anthracis begins after a phagocytosed spore germinates into a vegetative cell inside a mammalian host (2, 40, 69, 78). Virulence determinants produced by the vegetative cells facilitate bacterial growth, dissemination to major organ systems, and eventually host death (76-78). The release of aerosolized spores into areas with large concentrations of people is a serious public health concern (30).Heme acquisition in B. anthracis is mediated by the action of IsdX1 and IsdX2, two extracellular hemophores that extract heme from host hemoglobin and deliver the iron-porphyrin to cell wall-localized IsdC (21, 45). Both IsdX1 and IsdX2 harbor near iron transporter domains (NEATs), a conserved protein module found in Gram-positive bacteria that mediates heme uptake from hemoglobin and contributes to bacterial pathogenesis upon infection (3, 8, 21, 31, 44, 46, 49, 50, 67, 81, 86). Hypothesizing that B. anthracis may contain additional mechanisms for heme transport, we provide evidence that B. anthracis S-layer protein K (BslK), an S-layer homology (SLH) and NEAT protein (32, 43), is surface localized and binds and transfers heme to IsdC in a rapid, contact-dependent manner. These results suggest that the Isd system is not a self-contained conduit for heme trafficking and imply that there is functional cross talk between differentially localized NEAT proteins to promote heme uptake during infection.  相似文献   

3.
The Bacillus anthracis spore is the causative agent of the disease anthrax. The outermost structure of the B. anthracis spore, the exosporium, is a shell composed of approximately 20 proteins. The function of the exosporium remains poorly understood and is an area of active investigation. In this study, we analyzed the previously identified but uncharacterized exosporium protein ExsK. We found that, in contrast to other exosporium proteins, ExsK is present in at least two distinct locations, i.e., the spore surface as well as a more interior location underneath the exosporium. In spores that lack the exosporium basal layer protein ExsFA/BxpB, ExsK fails to encircle the spore and instead is present at only one spore pole, indicating that ExsK assembly to the spore is partially dependent on ExsFA/BxpB. In spores lacking the exosporium surface protein BclA, ExsK fails to mature into high-molecular-mass species observed in wild-type spores. These data suggest that the assembly and maturation of ExsK within the exosporium are dependent on ExsFA/BxpB and BclA. We also found that ExsK is not required for virulence in murine and guinea pig models but that it does inhibit germination. Based on these data, we propose a revised model of exosporium maturation and assembly and suggest a novel role for the exosporium in germination.During starvation, bacteria of the genus Bacillus differentiate into dormant, highly robust cell types called spores, thereby preserving their genomes during stressful and nutrient-poor conditions (10). Spores can withstand extremely harsh environmental insults, including toxic chemicals, UV radiation, and heat (31). When conditions again become favorable for cell survival, spores can return to vegetative cell growth through a process called germination (17, 18, 31, 49). Spores are formed in an approximately 8-h process during which the developing spore first forms as a compartment (the forespore) contained within the surrounding cell (the mother cell) (34). Ultimately, the mother cell envelope lyses, releasing the mature spore into the environment.Spores from all Bacillus species have similar architectures. At the spore interior is the core, which houses the spore chromosome. Surrounding the core is an inner membrane encased in a specialized peptidoglycan called the cortex and finally a series of outer layers that vary significantly among species (10). In some species, including Bacillus subtilis, the outermost structure is a protective layer called the coat, which guards the spore against reactive small molecules, degradative enzymes, and predation by other microbes (11, 17, 20, 38). Spores of other species, including the pathogens Bacillus anthracis, Bacillus cereus, and Bacillus thuringiensis and the nonpathogenic bacteria Bacillus megaterium and Bacillus odysseyi, have an additional structure called the exosporium, which surrounds the coat (24, 32, 47). The exosporium is composed of two structural units: the basal layer, which is a shell of proteins forming a hexagonal array, and a nap of hairlike protrusions extending outward from the basal layer (2, 32). A major component of the nap (and of the spore surface) is the collagen-like protein BclA (40, 43). The proteins that comprise the outer structures (the coat and exosporium) are synthesized in the mother cell cytoplasm, from which location they assemble onto the spore surface to form their respective structures (11).The function of the exosporium is poorly understood. Previous studies have implicated its contribution to germination, resistance to host cells and other stresses, adhesion to inert surfaces, and interactions with epithelial cells and macrophages (1, 6, 7, 13, 33, 41, 48; G. Chen, A. Driks, K. Tawfiq, M. Mallozzi, and S. Patil, submitted for publication). In most cases, however, the roles of individual exosporium proteins in each of these functions remain unclear, in part because the location of each protein within the exosporium is largely unknown.Interestingly, it appears that the exosporium is not essential for virulence of B. anthracis in several animal models (5, 7, 12, 13). Nonetheless, it is possible that in natural infections the exosporium plays a significant role. Because it is involved in attachment, the exosporium is also likely to have a significant impact on the persistence of B. anthracis spores in the environment.To gain insight into the molecular basis of exosporium assembly and function, we studied a previously identified but otherwise uncharacterized exosporium protein, ExsK. Using immunofluorescence microscopy (IFM), we found that ExsK is asymmetrically distributed on the surfaces of mature spores and is also present beneath the exosporium. In the absence of ExsFA/BxpB, ExsK was restricted to one spore pole, suggesting that the encirclement of the spore by ExsK depends on ExsFA/BxpB. Western blot analysis indicated that in mature spores ExsK is present in high-molecular-mass complexes, the formation of which is BclA dependent. Although ExsK is not required for several spore resistance properties or virulence, we found that it is required for normal germination. Our results provide a deeper understanding of the composition, function, and assembly of the B. anthracis exosporium and show that proteins comprising outer-spore structures can have multiple locations.  相似文献   

4.
Bacillus cereus spores are assembled with a series of concentric layers that protect them from a wide range of environmental stresses. The outermost layer, or exosporium, is a bag-like structure that interacts with the environment and is composed of more than 20 proteins and glycoproteins. Here, we identified a new spore protein, ExsM, from a β-mercaptoethanol extract of B. cereus ATCC 4342 spores. Subcellular localization of an ExsM-green fluorescent protein (GFP) protein revealed a dynamic pattern of fluorescence that follows the site of formation of the exosporium around the forespore. Under scanning electron microscopy, exsM null mutant spores were smaller and rounder than wild-type spores, which had an extended exosporium (spore length for the wt, 2.40 ± 0.56 μm, versus that for the exsM mutant, 1.66 ± 0.38 μm [P < 0.001]). Thin-section electron microscopy revealed that exsM mutant spores were encased by a double-layer exosporium, both layers of which were composed of a basal layer and a hair-like nap. Mutant exsM spores were more resistant to lysozyme treatment and germinated with higher efficiency than wild-type spores, and they had a delay in outgrowth. Insertional mutagenesis of exsM in Bacillus anthracis ΔSterne resulted in a partial second exosporium and in smaller spores. In all, these findings suggest that ExsM plays a critical role in the formation of the exosporium.Bacillus cereus and Bacillus anthracis are closely related members of the Bacillus cereus group (47). Although B. cereus is mainly an apathogenic organism, certain isolates can cause two different types of food poisoning, emetic syndrome and diarrheal disease (18). The emetic syndrome is caused by ingestion of cereulide, a heat-resistant toxin produced by vegetative cells contaminating the food (30), while the diarrheal disease occurs when spores germinate in the intestinal tract. Spores are also the infective agent in anthrax, a disease caused by B. anthracis (64).B. cereus and B. anthracis differentiate into spores when faced with nutrient deprivation. The spore is a dormant cell type that can remain viable for decades until favorable conditions induce germination and the resumption of vegetative growth. The remarkable resistance properties of the spore result from its unique architecture, consisting of a series of concentric protective layers (51). The spore core contains the genetic material and is surrounded by the cortex, a thick layer of modified peptidoglycan that promotes a highly dehydrated state. Encasing the core and the cortex, the coat is a multilayer protein shell that provides mechanical and chemical resistance. In addition, both the cortex and coat contribute to spore germination (17). Separated from the coat by an interspace, the exosporium encloses the rest of the spore, and it is composed of an inner basal layer and an outer hair-like nap (25).Being the most external layer of the spore, the exosporium interacts directly with the environment and as such provides a semipermeable barrier that may exclude large molecules, like antibodies and hydrolytic enzymes (3, 23, 24, 54). However, the exosporium does not appear to contribute to the typical resistance properties of the spore (6, 35, 60). Also, the exosporium is not necessary in anthrax pathogenesis when tested under laboratory conditions (7, 27, 59), although it is able to down-modulate the innate immune response to spores and mediate adhesion to host tissues (4, 8, 43, 44). The exosporium may also help the spore avoid premature germination in unsustainable environments, since it contains two enzymes, alanine racemase (Alr) and inosine hydrolase (Iunh), that can inactivate low quantities of the germinants l-alanine and inosine, respectively (6, 48, 55, 61). However, regulation of germination by the exosporium is poorly understood. Mutation of exosporial proteins has resulted in only negligible and inconsistent germination phenotypes (2, 5, 27, 28, 52, 54).The exosporium is composed of at least 20 proteins and glycoproteins in tight or loose association (48, 53, 57, 61, 65). These proteins are synthesized in the mother cell and always start self-assembly at the forespore pole near the middle of the mother cell, concurrently with the cortex and coat formation (42). Exosporium assembly is discontinuous and starts with a synthesis of a substructure known as the cap, which likely contains only a subset of the proteins present in the exosporium (55). After cap formation, construction of the rest of the exosporium requires the expression of ExsY (6). BclA is the main component of the hair-like nap on the external side of the exosporium, and it is linked to the basal layer through interaction with ExsFA/BxpB (54, 58). In addition, CotE participates in the correct attachment of the exosporium to the spore (27).Despite these findings, exosporium assembly continues to be a poorly understood process, and many questions remain regarding its composition and the regulation of its synthesis. In this study, we characterized a new spore protein, ExsM, which plays a key role in assembly of the exosporium. In B. cereus, inactivation of exsM resulted in spores with an unusual double-layer exosporium, and a similar phenotype was also observed in B. anthracis exsM null mutant spores. Finally, double-layer exosporium spores allowed us to study the role of the exosporium in germination and outgrowth.  相似文献   

5.
Bacillus anthracis spores, the etiological agents of anthrax, possess a loosely fitting outer layer called the exosporium that is composed of a basal layer and an external hairlike nap. The filaments of the nap are formed by trimers of the collagenlike glycoprotein BclA. Multiple pentasaccharide and trisaccharide side chains are O linked to BclA. The nonreducing terminal residue of the pentasaccharide side chain is the unusual sugar anthrose. A plausible biosynthetic pathway for anthrose biosynthesis has been proposed, and an antABCD operon encoding four putative anthrose biosynthetic enzymes has been identified. In this study, we genetically and biochemically characterized the activities of these enzymes. We also used mutant B. anthracis strains to determine the effects on BclA glycosylation of individually inactivating the genes of the anthrose operon. The inactivation of antA resulted in the appearance of BclA pentasaccharides containing anthrose analogs possessing shorter side chains linked to the amino group of the sugar. The inactivation of antB resulted in BclA being replaced with only trisaccharides, suggesting that the enzyme encoded by the gene is a dTDP-β-l-rhamnose α-1,3-l-rhamnosyl transferase that attaches the fourth residue of the pentasaccharide side chain. The inactivation of antC and antD resulted in the disappearance of BclA pentasaccharides and the appearance of a tetrasaccharide lacking anthrose. These phenotypes are entirely consistent with the proposed roles for the antABCD-encoded enzymes in anthrose biosynthesis. Purified AntA was then shown to exhibit β-methylcrotonyl-coenzyme A (CoA) hydratase activity, as we predicted. Similarly, we confirmed that purified AntC had aminotransferase activity and that purified AntD displayed N-acyltransferase activity.Bacillus anthracis, the causative agent of anthrax, is a Gram-positive, rod-shaped soil bacterium that forms spores when deprived of essential nutrients (15). Spore formation begins with an asymmetric septation that divides the developing cell into a forespore compartment and a larger mother cell compartment, each of which contains a copy of the genome. The mother cell then engulfs the forespore and surrounds it with three protective layers: a cortex composed of peptidoglycan, a closely apposed proteinaceous coat, and a loosely fitting exosporium (10). Mother cell lysis releases the mature spore, which is dormant and capable of surviving in harsh environments for many years (17). When spores encounter an aqueous environment containing nutrients, they can germinate and grow as vegetative cells (21).Recently, interest in B. anthracis spores has intensified in response to their use as agents of bioterrorism. Of particular interest has been the outermost layer of the spore, the exosporium, which serves as a semipermeable barrier to potentially harmful macromolecules (8, 25) and as the vital first point of contact with the immune system of an infected host (11, 18, 30). The exosporium of B. anthracis and of closely related species, such as Bacillus cereus and Bacillus thuringiensis, is comprised of a paracrystalline basal layer and an external hairlike nap (1). The basal layer contains approximately 20 different proteins (20, 23), while the filaments of the nap are formed by trimers of a single collagenlike glycoprotein called BclA (2, 26). The central region of BclA contains a large number of GXX repeats, and the region varies in length in naturally occurring strains of B. anthracis, resulting in hairlike naps of differing lengths (22, 27). Most of the GXX repeats are GPT, and many of the threonine residues are glycosylated. Two major oligosaccharide side chains are present, a pentasaccharide and a trisaccharide, and both are linked to the protein through reducing terminal N-acetylgalactosamine (GalNAc) residues (3). Several studies have demonstrated that the oligosaccharides are antigenic and are exposed on the surface of Bacillus anthracis spores (14, 29). This makes them prime targets for both detection devices and immunoprophylaxis.We previously reported our use of hydrazinolysis to release BclA oligosaccharides from exosporium preparations (3). The primary product was a tetrasaccharide that formed as a result of the undesirable loss of the reducing terminal GalNAc residue of the pentasaccharide, a process called “peeling.” We determined that the oligosaccharide consisted of a linear chain of three rhamnose residues with a novel deoxyamino sugar at its nonreducing terminus. This unusual sugar, 2-O-methyl-4-(3-hydroxy-3-methylbutamido)-4,6-dideoxy-d-glucose, was given the trivial name anthrose.Rhamnose is the major sugar present in both the trisaccharide and the pentasaccharide, and a four-gene rhamnose biosynthetic operon was previously identified (22). Previously, we proposed a pathway for anthrose biosynthesis (Fig. (Fig.1)1) and identified a four-gene operon (Fig. (Fig.2)2) that is essential for its biosynthesis (5). An in-frame deletion of the first gene of the operon reduced the amount of anthrose by approximately 50%, whereas the deletion of any one of the other three genes totally abolished anthrose synthesis. Here, we describe the characterization of the altered oligosaccharide side chains of the four deletion mutants. We also cloned several genes that we predicted are involved in anthrose biosynthesis and demonstrated that the gene products possessed the expected biochemical activities.Open in a separate windowFIG. 1.Proposed biosynthetic pathway of anthrose. The pathway utilizes dTDP-4-keto-6-deoxy-α-d-glucose, an intermediate in rhamnose biosynthesis, and methylcrotonyl-CoA, derived from leucine catabolism. (Modified from reference 5.)Open in a separate windowFIG. 2.Anthrose operon and flanking genes. The four genes of the anthrose operon are antA (BAS3322), antB (BAS3321), antC (BAS3320), and antD (BAS3319). The operon is flanked by genes that encode a putative collagenase (BAS3323) and a putative methyltransferase (BAS3318). (Modified from reference 5.)  相似文献   

6.
Adhesive pili on the surface of the serotype M1 Streptococcus pyogenes strain SF370 are composed of a major backbone subunit (Spy0128) and two minor subunits (Spy0125 and Spy0130), joined covalently by a pilin polymerase (Spy0129). Previous studies using recombinant proteins showed that both minor subunits bind to human pharyngeal (Detroit) cells (A. G. Manetti et al., Mol. Microbiol. 64:968-983, 2007), suggesting both may act as pilus-presented adhesins. While confirming these binding properties, studies described here indicate that Spy0125 is the pilus-presented adhesin and that Spy0130 has a distinct role as a wall linker. Pili were localized predominantly to cell wall fractions of the wild-type S. pyogenes parent strain and a spy0125 deletion mutant. In contrast, they were found almost exclusively in culture supernatants in both spy0130 and srtA deletion mutants, indicating that the housekeeping sortase (SrtA) attaches pili to the cell wall by using Spy0130 as a linker protein. Adhesion assays with antisera specific for individual subunits showed that only anti-rSpy0125 serum inhibited adhesion of wild-type S. pyogenes to human keratinocytes and tonsil epithelium to a significant extent. Spy0125 was localized to the tip of pili, based on a combination of mutant analysis and liquid chromatography-tandem mass spectrometry analysis of purified pili. Assays comparing parent and mutant strains confirmed its role as the adhesin. Unexpectedly, apparent spontaneous cleavage of a labile, proline-rich (8 of 14 residues) sequence separating the N-terminal ∼1/3 and C-terminal ∼2/3 of Spy0125 leads to loss of the N-terminal region, but analysis of internal spy0125 deletion mutants confirmed that this has no significant effect on adhesion.The group A Streptococcus (S. pyogenes) is an exclusively human pathogen that commonly colonizes either the pharynx or skin, where local spread can give rise to various inflammatory conditions such as pharyngitis, tonsillitis, sinusitis, or erysipelas. Although often mild and self-limiting, GAS infections are occasionally very severe and sometimes lead to life-threatening diseases, such as necrotizing fasciitis or streptococcal toxic shock syndrome. A wide variety of cell surface components and extracellular products have been shown or suggested to play important roles in S. pyogenes virulence, including cell surface pili (1, 6, 32). Pili expressed by the serotype M1 S. pyogenes strain SF370 mediate specific adhesion to intact human tonsil epithelia and to primary human keratinocytes, as well as cultured keratinocyte-derived HaCaT cells, but not to Hep-2 or A549 cells (1). They also contribute to adhesion to a human pharyngeal cell line (Detroit cells) and to biofilm formation (29).Over the past 5 years, pili have been discovered on an increasing number of important Gram-positive bacterial pathogens, including Bacillus cereus (4), Bacillus anthracis (4, 5), Corynebacterium diphtheriae (13, 14, 19, 26, 27, 44, 46, 47), Streptococcus agalactiae (7, 23, 38), and Streptococcus pneumoniae (2, 3, 24, 25, 34), as well as S. pyogenes (1, 29, 32). All these species produce pili that are composed of a single major subunit plus either one or two minor subunits. During assembly, the individual subunits are covalently linked to each other via intermolecular isopeptide bonds, catalyzed by specialized membrane-associated transpeptidases that may be described as pilin polymerases (4, 7, 25, 41, 44, 46). These are related to the classical housekeeping sortase (usually, but not always, designated SrtA) that is responsible for anchoring many proteins to Gram-positive bacterial cell walls (30, 31, 33). The C-terminal ends of sortase target proteins include a cell wall sorting (CWS) motif consisting, in most cases, of Leu-Pro-X-Thr-Gly (LPXTG, where X can be any amino acid) (11, 40). Sortases cleave this substrate between the Thr and Gly residues and produce an intermolecular isopeptide bond linking the Thr to a free amino group provided by a specific target. In attaching proteins to the cell wall, the target amino group is provided by the lipid II peptidoglycan precursor (30, 36, 40). In joining pilus subunits, the target is the ɛ-amino group in the side chain of a specific Lys residue in the second subunit (14, 18, 19). Current models of pilus biogenesis envisage repeated transpeptidation reactions adding additional subunits to the base of the growing pilus, until the terminal subunit is eventually linked covalently via an intermolecular isopeptide bond to the cell wall (28, 41, 45).The major subunit (sometimes called the backbone or shaft subunit) extends along the length of the pilus and appears to play a structural role, while minor subunits have been detected either at the tip, the base, and/or at occasional intervals along the shaft, depending on the species (4, 23, 24, 32, 47). In S. pneumoniae and S. agalactiae one of the minor subunits acts as an adhesin, while the second appears to act as a linker between the base of the assembled pilus and the cell wall (7, 15, 22, 34, 35). It was originally suggested that both minor subunits of C. diphtheriae pili could act as adhesins (27). However, recent data showed one of these has a wall linker role (26, 44) and may therefore not function as an adhesin.S. pyogenes strain SF370 pili are composed of a major (backbone) subunit, termed Spy0128, plus two minor subunits, called Spy0125 and Spy0130 (1, 32). All three are required for efficient adhesion to target cells (1). Studies employing purified recombinant proteins have shown that both of the minor subunits, but not the major subunit, bind to Detroit cells (29), suggesting both might act as pilus-presented adhesins. Here we report studies employing a combination of recombinant proteins, specific antisera, and allelic replacement mutants which show that only Spy0125 is the pilus-presented adhesin and that Spy0130 has a distinct role in linking pili to the cell wall.  相似文献   

7.
8.
Soil substrate membrane systems allow for microcultivation of fastidious soil bacteria as mixed microbial communities. We isolated established microcolonies from these membranes by using fluorescence viability staining and micromanipulation. This approach facilitated the recovery of diverse, novel isolates, including the recalcitrant bacterium Leifsonia xyli, a plant pathogen that has never been isolated outside the host.The majority of bacterial species have never been recovered in the laboratory (1, 14, 19, 24). In the last decade, novel cultivation approaches have successfully been used to recover “unculturables” from a diverse range of divisions (23, 25, 29). Most strategies have targeted marine environments (4, 23, 25, 32), but soil offers the potential for the investigation of vast numbers of undescribed species (20, 29). Rapid advances have been made toward culturing soil bacteria by reformulating and diluting traditional media, extending incubation times, and using alternative gelling agents (8, 21, 29).The soil substrate membrane system (SSMS) is a diffusion chamber approach that uses extracts from the soil of interest as the growth substrate, thereby mimicking the environment under investigation (12). The SSMS enriches for slow-growing oligophiles, a proportion of which are subsequently capable of growing on complex media (23, 25, 27, 30, 32). However, the SSMS results in mixed microbial communities, with the consequent difficulty in isolation of individual microcolonies for further characterization (10).Micromanipulation has been widely used for the isolation of specific cell morphotypes for downstream applications in molecular diagnostics or proteomics (5, 15). This simple technology offers the opportunity to select established microcolonies of a specific morphotype from the SSMS when combined with fluorescence visualization (3, 11). Here, we have combined the SSMS, fluorescence viability staining, and advanced micromanipulation for targeted isolation of viable, microcolony-forming soil bacteria.  相似文献   

9.
Anthrax is a zoonotic disease caused by the gram-positive spore-forming bacterium Bacillus anthracis. Human infection occurs after the ingestion, inhalation, or cutaneous inoculation of B. anthracis spores. The subsequent progression of the disease is largely mediated by two native virulence plasmids, pXO1 and pXO2, and is characterized by septicemia, toxemia, and meningitis. In order to produce meningitis, blood-borne bacteria must interact with and breach the blood-brain barrier (BBB) that is composed of a specialized layer of brain microvascular endothelial cells (BMEC). We have recently shown that B. anthracis Sterne is capable of penetrating the BBB in vitro and in vivo, establishing the classic signs of meningitis; however, the molecular mechanisms underlying the central nervous system (CNS) tropism are not known. Here, we show that attachment to and invasion of human BMEC by B. anthracis Sterne is mediated by the pXO1 plasmid and an encoded envelope factor, BslA. The results of studies using complementation analysis, recombinant BslA protein, and heterologous expression demonstrate that BslA is both necessary and sufficient to promote adherence to brain endothelium. Furthermore, mice injected with the BslA-deficient strain exhibited a significant decrease in the frequency of brain infection compared to mice injected with the parental strain. In addition, BslA contributed to BBB breakdown by disrupting tight junction protein ZO-1. Our results identify the pXO1-encoded BslA adhesin as a critical mediator of CNS entry and offer new insights into the pathogenesis of anthrax meningitis.Bacillus anthracis, the etiologic agent of anthrax, is a gram-positive spore-forming bacterium that is commonly found in soil (29). The bacterium can infect animals and humans by ingestion, inhalation, or cutaneous inoculation of B. anthracis spores (8). Spores are taken up by resident macrophages that migrate to the lymph nodes (15). Here, the spores germinate into vegetative bacteria, multiply, and then disseminate throughout the host, causing septicemia and toxemia (8). Systemic disease can be complicated by the onset of a fulminant and rapidly fatal hemorrhagic meningitis and meningoencephalitis (27). Anthrax meningitis is associated with a high mortality rate despite intensive antibiotic therapy (24). Biopsy studies after an outbreak of inhalational anthrax and experimental studies of inhalational infection in rhesus monkeys demonstrated the presence of bacilli in the central nervous system (CNS) and pathologies consistent with suppurative and hemorrhagic meningitis in the majority of cases (1, 12). The intentional release of B. anthracis spores (19) during the 2001 bioterrorism event resulted in a case of meningitis (19), necessitating a need for a better understanding of the pathogenesis of anthrax meningitis and CNS infection.To cause meningitis, blood-borne bacteria must interact with and breach the blood-brain barrier (BBB). The majority of the BBB is anatomically represented by the cerebral microvascular endothelium; brain microvascular endothelial cells (BMEC) are joined by tight junctions and display a paucity of pinocytosis, thereby effectively limiting the passage of substances and maintaining the CNS microenvironment (4, 5). Despite its highly restrictive nature, certain bacterial pathogens are still able to penetrate the BBB and gain entry into the CNS. The presence of bacilli in the brains of patients (1, 24) and in experimental models of anthrax infection (42, 44) suggests that vegetative B. anthracis cells are able to cross the BBB to initiate meningeal inflammation and the classic pathology associated with meningitis.B. anthracis harbors two large virulence plasmids, pXO1 and pXO2 (8), which are required for full virulence, as strains lacking these plasmids are attenuated in animal models of infection (29). B. anthracis Sterne (pXO1+ pXO2) has been utilized as a vaccine strain (41) but is still widely used in both in vitro and in vivo studies of anthrax infection since it causes lethal disease in mouse models of infection (46). Despite the crucial roles of pXO1 and pXO2 in anthrax disease pathogenesis, very few plasmid-encoded factors have been characterized. The best described are the antiphagocytic polyglutamyl capsule, encoded by biosynthetic enzymes on pXO2, and the anthrax toxin complex comprised of protective antigen, lethal factor (LF), and edema factor (EF), encoded by pXO1 (8, 29). Sequence analysis of the pXO1 plasmid revealed that the majority of plasmid-encoded factors, ∼70%, were of unknown function (31). More recently, in silico analysis identified novel pXO1-encoded proteins with immunogenic potential and relevance for pathogenesis. These included factors with putative adherent and invasive properties (2). Interestingly, two of the immunoreactive proteins were predicted surface layer (S-layer) proteins (2), one of which, B. anthracis S-layer protein A (BslA, pXO1-90), has recently been described and shown to mediate adherence of the vegetative form to host cells (20).Using in vitro and in vivo model systems, we have recently shown that B. anthracis Sterne adheres to and invades brain endothelium (44). This interaction was partially dependent on the pXO1-encoded anthrax toxins; however, the molecular mechanisms that contribute to B. anthracis penetration of the BBB are currently unknown. In this study, we investigate the role of pXO1 in B. anthracis Sterne''s interaction with brain endothelium and identify the encoded BslA adhesin as a critical mediator for BBB attachment and penetration during the pathogenesis of anthrax meningitis.  相似文献   

10.
11.
The Bacillus cereus group includes three closely related species, B. anthracis, B. cereus, and B. thuringiensis, which form a highly homogeneous subdivision of the genus Bacillus. One of these species, B. anthracis, has been identified as one of the most probable bacterial biowarfare agents. Here, we evaluate the sequence and length polymorphisms of the Bacillus collagen-like protein bcl genes as a basis for B. anthracis detection and fingerprinting. Five genes, designated bclA to bclE, are present in B. anthracis strains. Examination of bclABCDE sequences identified polymorphisms in bclB alleles of the B. cereus group organisms. These sequence polymorphisms allowed specific detection of B. anthracis strains by PCR using both genomic DNA and purified Bacillus spores in reactions. By exploiting the length variation of the bcl alleles it was demonstrated that the combined bclABCDE PCR products generate markedly different fingerprints for the B. anthracis Ames and Sterne strains. Moreover, we predict that bclABCDE length polymorphism creates unique signatures for B. anthracis strains, which facilitates identification of strains with specificity and confidence. Thus, we present a new diagnostic concept for B. anthracis detection and fingerprinting, which can be used alone or in combination with previously established typing platforms.The Bacillus cereus group includes three closely related species, B. anthracis, B. cereus, and B. thuringiensis, as well as the more distantly related species B. mycoides and B. weihenstephanensis. These gram-positive, spore-forming bacteria form a highly homogeneous subdivision of the genus Bacillus, which also contains several other organisms belonging to the B. subtilis group. The importance and public awareness of B. cereus group organisms are associated with their distinct phenotypes and pathological effects. B. anthracis is the causative agent of anthrax, a disease that affects humans and animals worldwide and has also been developed as a biological warfare agent (17, 25). B. cereus is an opportunistic human pathogen which is responsible mainly for gastrointestinal illnesses resulting from food contamination (9), whereas B. thuringiensis is an insect pathogen whose toxin is a biological pesticide widely used in global agriculture (38). The systematics of the members of the B. cereus group poses significant challenges due to very high level of chromosomal synteny and protein identity (33). Intense efforts have focused on overcoming these challenges, and there has been a particular focus on developing methods for specific detection of B. anthracis and for differentiating among strains of these closely related organisms.Biodefense and forensic needs prompted large-scale sequencing of multiple bacillus genomes in a search for polymorphic sites for use in typing procedures (33). One type of polymorphism involves variation in the number of repeating nucleotide units that are referred to as variable-number tandem repeats (VNTRs). The resulting variation in the length and mass of the PCR products of these units can be demonstrated by gel and capillary electrophoresis (20), mass spectrometry (29), or microchannel fluidics (30). To date, several different VNTRs have been identified and tested. For example, Keim et al. studied the genetic relationship among a large collection of B. anthracis isolates based on the VNTRs found in the vrr genes (19, 20). Using a similar approach, Valjevac et al. used VNTRs of Bcms loci as markers to assess the phylogeny of members of the B. cereus group (46). Finally, length variation of the collagen-like (CL) region of the bclA gene was employed to differentiate among B. anthracis strains (6, 42).The CL sequences, which are composed of Gly-Xaa-Yaa (i.e., a glycine followed by two additional residues; GXY) repeats, have been identified in silico in more than 100 prokaryotic proteins (34). Recent studies demonstrated that some bacterial CL proteins (CLPs), such as streptococcal protein Scl and BclA, can form the collagen triple helix (4, 14, 48). Bacterial CLPs are typically surface exposed and are found in microorganisms pathogenic to humans and animals. BclA (Bacillus CLP of B. anthracis) is a major spore surface protein (41) and is found in all members of the B. cereus group (6; this study). A second CLP, designated BclB (47), was identified as a component of the B. anthracis exosporium; however, its distribution and structural properties have not been well characterized. Likewise, two closely related proteins, ExsH and ExsJ, contain GXY CL repeats and are presumably located in the exosporium of Bacillus strains (45).In this work we investigated in silico the occurrence and distribution of the bcl genes, presumably encoding CLPs, in all members of the B. cereus group. A new classification of the resulting Bcl protein variants is proposed based on the domain composition and folding of these proteins. As many as 10 bcl genes were found in a single B. cereus strain. Five genes were consistently observed in B. anthracis strains and designated bclA to bclE. We further analyzed sequence polymorphisms among these bcl genes and assessed use of them for B. anthracis detection and strain fingerprinting. Representative members of the B. cereus group and less closely related control bacilli were used to demonstrate specific bclB gene-based detection of B. anthracis spores. Finally, a combination of experiments and mathematical modeling was used to demonstrate how combined use of the bclABCDE sequence polymorphisms can be a powerful tool for strain fingerprinting in biodefense and forensic applications.  相似文献   

12.
A search for bacterium-specific biomarkers in peripheral blood following infection with Bacillus anthracis was carried out with rabbits, using a battery of specific antibodies generated by DNA vaccination against 10 preselected highly immunogenic bacterial antigens which were identified previously by a genomic/proteomic/serologic screen of the B. anthracis secretome. Detection of infection biomarkers in the circulation of infected rabbits could be achieved only after removal of highly abundant serum proteins by chromatography using a random-ligand affinity column. Besides the toxin component protective antigen, the following three secreted proteins were detected in the circulation of infected animals: the chaperone and protease HtrA (BA3660), an NlpC/P60 endopeptidase (BA1952), and a protein of unknown function harboring two SH3 (Src homology 3) domains (BA0796). The three proteins could be detected in plasma samples from infected animals exhibiting 103 to 105 CFU/ml blood and also in standard blood cultures at 3 to 6 h post-bacterial inoculation at a bacteremic level as low as 103 CFU/ml. Furthermore, the three biomarkers appear to be present only in the secretome of B. anthracis, not in those of the related pathogens B. thuringiensis and B. cereus. To the best of our knowledge, this is the first report of direct detection of B. anthracis-specific proteins, other than the toxin components, in the circulation of infected animals.The gram-positive spore-forming bacterium Bacillus anthracis is the causative agent of anthrax, a rare fatal disease which is initiated, in its most severe form, by inhalation of spores. Due to the severity of the disease, the ease of respiratory infection, and the extreme resistance of the spores to unfavorable environmental conditions, B. anthracis is considered a potential biological warfare agent (for a review, see references 8, 10, 35, 56, and 62), and in recent years, the need for novel reliable diagnostic approaches, improved vaccination strategies, novel therapeutic targets, and a better understanding of the pathogenesis of anthrax has been widely acknowledged.Inhaled B. anthracis spores are taken up by alveolar macrophages and germinate into vegetative bacilli which eventually invade the bloodstream, where they multiply massively and secrete toxins and virulence factors. Anthrax is toxinogenic in the sense that the bacterial binary exotoxin is necessary for the onset of the disease (54), yet other factors may be required for the colonization and expansion of bacteria in the host (15, 18, 31, 32, 37, 46, 65, 66, 70, 83). The toxin is composed of the following three proteins: protective antigen (PA), which mediates binding to the receptor on target cells and internalization of the toxin components (14, 74); lethal factor, a zinc protease targeting several mitogen-activated protein kinases (52); and edema factor (EF), a calmodulin-dependent adenylate cyclase (55, 57). The genes encoding the three exotoxin components are located on the native virulence plasmid pXO1. Genes encoding proteins with functions involved in the synthesis of the second major B. anthracis virulence determinant, an immunologically inert polyglutamyl capsule that protects bacteria from phagocytosis, are located on a second native virulence plasmid, pXO2 (56).In humans, the initial symptoms of inhalation anthrax are nonspecific and reminiscent of influenza-like upper respiratory tract infections. The second stage is characterized by high fever, respiratory failure, dyspnea, and shock. Unless patients are treated promptly, death occurs within 24 h of the onset of the second stage, preceded by massive bacteremia (27, 34, 73, 76). The mandatory treatment for anthrax is based on administration of antibiotics (17, 76), yet study of the disease in animal models has clearly established that the time of antibiotic administration postinfection is crucial for the effectiveness of the treatment, strongly supporting the importance of rapid diagnosis (2, 47, 48). At present, due to the severity of the disease and its rapid progression, treatment is administered to each person with confirmed contact with contaminated areas (76).Early accurate diagnosis of anthrax is complicated by the rarity of the disease and the nonspecific nature of the symptoms. Microbiologic identification of anthrax is based on the nonhemolytic nature of the typically white-gray colonies and the rod morphology of the gram-positive nonmotile bacilli detected in clinical samples or blood cultures (16, 19, 30, 73, 78). Immunofluorescence and immunohistochemistry targeted to bacterial proteins are not routinely conducted. Later in the course of the disease, seroconversion in response to the various components of the toxin may serve only as a retrospective confirmation of initial exposure. With the advent of genetic methodologies, B. anthracis in cultures inoculated with clinical and forensic samples can be detected specifically and accurately by PCR, usually designed to amplify genes located on the native virulence plasmids (30). Recently, the use of PA as a disease biomarker was suggested, owing to the presence of this protein in detectable amounts in the circulation of infected animals (53, 71). EF and lethal factor can be detected in the circulation only at later stages of infection (30).In recent years, the search for novel biomarkers of disease, including bacterial infections, has exploited the approach of global biological inspection based on functional genomic or proteomic studies (64, 85). We previously documented such global surveys, combined with a serological study of B. anthracis (5, 6, 20, 21, 22, 38, 39), for identification of vaccine and diagnostic marker candidates among extracellular (secreted or membranal) proteins. These studies indeed revealed a list of proteins that can serve as potential biomarkers, based on their immunogenicity (which probes their in vivo expression), abundance under various culture conditions, and functional relatedness to infection. In the present study, the search was extended by directly addressing the presence of bacterial secreted proteins in the circulation of B. anthracis-infected rabbits, using specific antibodies generated by DNA vaccination against the previously selected immunogenic proteins. Visualization of bacterial proteins in the circulation of infected animals was achieved only following depletion of highly abundant serum proteins by an affinity chromatography protocol. The search enabled the successful detection, in addition to PA, of three secreted proteins uniquely expressed by B. anthracis, i.e., HtrA (BA3660), the BA1952 endopeptidase, and a protein of unknown function (BA0796). All of these proteins are potential virulence-related factors. This is the first communication of the presence of B. anthracis secreted proteins other than the bacterial toxin in the circulation of infected animals, and their identification strongly supports the validity of the reductional screening approach for selection of disease biomarkers.  相似文献   

13.
Analysis of Lyme borreliosis (LB) spirochetes, using a novel multilocus sequence analysis scheme, revealed that OspA serotype 4 strains (a rodent-associated ecotype) of Borrelia garinii were sufficiently genetically distinct from bird-associated B. garinii strains to deserve species status. We suggest that OspA serotype 4 strains be raised to species status and named Borrelia bavariensis sp. nov. The rooted phylogenetic trees provide novel insights into the evolutionary history of LB spirochetes.Multilocus sequence typing (MLST) and multilocus sequence analysis (MLSA) have been shown to be powerful and pragmatic molecular methods for typing large numbers of microbial strains for population genetics studies, delineation of species, and assignment of strains to defined bacterial species (4, 13, 27, 40, 44). To date, MLST/MLSA schemes have been applied only to a few vector-borne microbial populations (1, 6, 30, 37, 40, 41, 47).Lyme borreliosis (LB) spirochetes comprise a diverse group of zoonotic bacteria which are transmitted among vertebrate hosts by ixodid (hard) ticks. The most common agents of human LB are Borrelia burgdorferi (sensu stricto), Borrelia afzelii, Borrelia garinii, Borrelia lusitaniae, and Borrelia spielmanii (7, 8, 12, 35). To date, 15 species have been named within the group of LB spirochetes (6, 31, 32, 37, 38, 41). While several of these LB species have been delineated using whole DNA-DNA hybridization (3, 20, 33), most ecological or epidemiological studies have been using single loci (5, 9-11, 29, 34, 36, 38, 42, 51, 53). Although some of these loci have been convenient for species assignment of strains or to address particular epidemiological questions, they may be unsuitable to resolve evolutionary relationships among LB species, because it is not possible to define any outgroup. For example, both the 5S-23S intergenic spacer (5S-23S IGS) and the gene encoding the outer surface protein A (ospA) are present only in LB spirochete genomes (36, 43). The advantage of using appropriate housekeeping genes of LB group spirochetes is that phylogenetic trees can be rooted with sequences of relapsing fever spirochetes. This renders the data amenable to detailed evolutionary studies of LB spirochetes.LB group spirochetes differ remarkably in their patterns and levels of host association, which are likely to affect their population structures (22, 24, 46, 48). Of the three main Eurasian Borrelia species, B. afzelii is adapted to rodents, whereas B. valaisiana and most strains of B. garinii are maintained by birds (12, 15, 16, 23, 26, 45). However, B. garinii OspA serotype 4 strains in Europe have been shown to be transmitted by rodents (17, 18) and, therefore, constitute a distinct ecotype within B. garinii. These strains have also been associated with high pathogenicity in humans, and their finer-scale geographical distribution seems highly focal (10, 34, 52, 53).In this study, we analyzed the intra- and interspecific phylogenetic relationships of B. burgdorferi, B. afzelii, B. garinii, B. valaisiana, B. lusitaniae, B. bissettii, and B. spielmanii by means of a novel MLSA scheme based on chromosomal housekeeping genes (30, 48).  相似文献   

14.
In this report we provide evidence that the antimicrobial action of stannous salts and a gold drug, auranofin, against Treponema denticola is mediated through inhibition of the metabolism of selenium for synthesis of selenoproteins.The biological use of selenium as a catalyst, incorporated into proteins as selenocysteine, is broad. It plays an essential role in energy metabolism, redox balance, and reproduction in a variety of organisms, from bacterial pathogens to eukaryotic parasites to humans. The results of several epidemiological studies indicate that higher levels of selenium in the mammalian diet can have a negative effect on dental health (2, 17-19, 39). Although the impact of selenium is attributed to its influence on the physical properties of the enamel surface (10), the role of selenium in supporting the oral microbial community has not been studied.The oral cavity is a highly complex microbiome, with a large proportion of its residents uncharacterized due to their fastidious nature and resistance to traditional culture methods (11). Analysis of whole saliva indicates that bacterial metabolism influences the amino acid composition and indicates a role for amino acid fermentation (38). Curtis et al. demonstrated the occurrence of Stickland reactions in dental plaque (9). These reactions were first described in clostridia (35-37). They involve the coupled fermentation of amino acids in which one amino acid is oxidized (Stickland donor) and another (Stickland acceptor) is reduced (29). Treponema denticola, an established resident of the oral cavity, performs Stickland reactions via the selenoprotein glycine reductase (32). Glycine reductase is composed of a multiprotein complex that contains two separate selenoproteins, termed selenoprotein A and selenoprotein B (1, 7, 8, 15, 16). This complex of proteins converts glycine to acetyl phosphate by using inorganic phosphate and the reducing potential from thioredoxin. For the organisms that use this complex, this is a vital source of ATP. Thus far, the requirement for selenocysteine at the active site of this enzyme complex is universally conserved, even though all other selenoproteins that have been identified using computational techniques have a putative cysteine homologue (24).Treponema denticola is considered one of the primary pathogens responsible for periodontitis, a chronic inflammatory disease that is the major cause of adult tooth loss (11, 27, 33). It is the best-studied oral spirochete, commonly found with other spirochetes within the periodontal pocket. It expresses a variety of virulence factors and is capable of adhering to and penetrating endothelial cell monolayers (31). Its health impact may reach beyond the oral cavity. A recent study linked periodontitis with peripheral arterial disease and detected T. denticola, along with other periodontal pathogens, in atherosclerotic plaque (3). Sequence analysis indicates the presence of several selenoproteins in addition to glycine reductase within the genome of T. denticola (24). This organism exhibits a strict growth requirement for selenium (32).A significant literature exists that clearly demonstrates the antimicrobial activity of fluoride compounds against microorganisms associated with dental decay and periodontitis. Both sodium fluoride and stannous fluoride, as well as stannous ions alone, inhibit the growth of T. denticola (21). The inhibitory effect of stannous salts on T. denticola''s growth is unexplained. It should be noted that toothpastes containing stannous fluoride are more effective in reducing gingivitis and plaque (28, 30).Tin, as well as several other trace elements, modulates the effects of acute selenium toxicity (20). Conversely, selenium affects the activity of tin in animal models (4-6). In this study, we examine the possibility that stannous ions interfere with selenium metabolism in T. denticola.  相似文献   

15.
Here, we report a fluorescence in situ hybridization (FISH) method for rapid detection of Cronobacter strains in powdered infant formula (PIF) using a novel peptide nucleic acid (PNA) probe. Laboratory tests with several Enterobacteriaceae species showed that the specificity and sensitivity of the method were 100%. FISH using PNA could detect as few as 1 CFU per 10 g of Cronobacter in PIF after an 8-h enrichment step, even in a mixed population containing bacterial contaminants.Cronobacter strains were originally described as Enterobacter sakazakii (12), but they are now known to comprise a novel genus consisting of six separate genomospecies (20, 21). These opportunistic pathogens are ubiquitous in the environment and various types of food and are occasionally found in the normal human flora (11, 12, 16, 32, 47). Based on case reports, Cronobacter infections in adults are generally less severe than Cronobacter infections in newborn infants, with which a high fatality rate is associated (24).The ability to detect Cronobacter and trace possible sources of infection is essential as a means of limiting the impact of these organisms on neonatal health and maintaining consumer confidence in powdered infant formula (PIF). Conventional methods, involving isolation of individual colonies followed by biochemical identification, are more time-consuming than molecular methods, and the reliability of some currently proposed culture-based methods has been questioned (28). Recently, several PCR-based techniques have been described (23, 26, 28-31, 38). These techniques are reported to be efficient even when low levels of Cronobacter cells are found in a sample (0.36 to 66 CFU/100 g). However, PCR requires DNA extraction and does not allow direct, in situ visualization of the bacterium in a sample.Fluorescence in situ hybridization (FISH) is a method that is commonly used for bacterial identification and localization in samples. This method is based on specific binding of nucleic acid probes to particular DNA or RNA target regions (1, 2). rRNA has been regarded as the most suitable target for bacterial FISH, allowing differentiation of potentially viable cells. Traditionally, FISH methods are based on the use of conventional DNA oligonucleotide probes, and a commercial system, VIT-E sakazakii (Vermicon A.G., Munich, Germany), has been developed based on this technology (25). However, a recently developed synthetic DNA analogue, peptide nucleic acid (PNA), has been shown to provide improved hybridization performance compared to DNA probes, making FISH procedures easier and more efficient (41). Taking advantage of the PNA properties, FISH using PNA has been successfully used for detection of several clinically relevant microorganisms (5, 15, 17, 27, 34-36).  相似文献   

16.
Factors potentially contributing to the lower incidence of Lyme borreliosis (LB) in the far-western than in the northeastern United States include tick host-seeking behavior resulting in fewer human tick encounters, lower densities of Borrelia burgdorferi-infected vector ticks in peridomestic environments, and genetic variation among B. burgdorferi spirochetes to which humans are exposed. We determined the population structure of B. burgdorferi in over 200 infected nymphs of the primary bridging vector to humans, Ixodes pacificus, collected in Mendocino County, CA. This was accomplished by sequence typing the spirochete lipoprotein ospC and the 16S-23S rRNA intergenic spacer (IGS). Thirteen ospC alleles belonging to 12 genotypes were found in California, and the two most abundant, ospC genotypes H3 and E3, have not been detected in ticks in the Northeast. The most prevalent ospC and IGS biallelic profile in the population, found in about 22% of ticks, was a new B. burgdorferi strain defined by ospC genotype H3. Eight of the most common ospC genotypes in the northeastern United States, including genotypes I and K that are associated with disseminated human infections, were absent in Mendocino County nymphs. ospC H3 was associated with hardwood-dominated habitats where western gray squirrels, the reservoir host, are commonly infected with LB spirochetes. The differences in B. burgdorferi population structure in California ticks compared to the Northeast emphasize the need for a greater understanding of the genetic diversity of spirochetes infecting California LB patients.In the United States, Lyme borreliosis (LB) is the most commonly reported vector-borne illness and is caused by infection with the spirochete Borrelia burgdorferi (3, 9, 52). The signs and symptoms of LB can include a rash, erythema migrans, fever, fatigue, arthritis, carditis, and neurological manifestations (50, 51). The black-legged tick, Ixodes scapularis, and the western black-legged tick, Ixodes pacificus, are the primary vectors of B. burgdorferi to humans in the United States, with the former in the northeastern and north-central parts of the country and the latter in the Far West (9, 10). These ticks perpetuate enzootic transmission cycles together with a vertebrate reservoir host such as the white-footed mouse, Peromyscus leucopus, in the Northeast and Midwest (24, 35), or the western gray squirrel, Sciurus griseus, in California (31, 46).B. burgdorferi is a spirochete species with a largely clonal population structure (14, 16) comprising several different strains or lineages (8). The polymorphic ospC gene of B. burgdorferi encodes a surface lipoprotein that increases expression within the tick during blood feeding (47) and is required for initial infection of mammalian hosts (25, 55). To date, approximately 20 North American ospC genotypes have been described (40, 45, 49, 56). At least four, and possibly up to nine, of these genotypes are associated with B. burgdorferi invasiveness in humans (1, 15, 17, 49, 57). Restriction fragment length polymorphism (RFLP) and, subsequently, sequence analysis of the 16S-23S rRNA intergenic spacer (IGS) are used as molecular typing tools to investigate genotypic variation in B. burgdorferi (2, 36, 38, 44, 44, 57). The locus maintains a high level of variation between related species, and this variation reflects the heterogeneity found at the genomic level of the organism (37). The IGS and ospC loci appear to be linked (2, 8, 26, 45, 57), but the studies to date have not been representative of the full range of diversity of B. burgdorferi in North America.Previous studies in the northeastern and midwestern United States have utilized IGS and ospC genotyping to elucidate B. burgdorferi evolution, host strain specificity, vector-reservoir associations, and disease risk to humans. In California, only six ospC and five IGS genotypes have been described heretofore in samples from LB patients or I. pacificus ticks (40, 49, 56) compared to approximately 20 ospC and IGS genotypes identified in ticks, vertebrate hosts, or humans from the Northeast and Midwest (8, 40, 45, 49, 56). Here, we employ sequence analysis of both the ospC gene and IGS region to describe the population structure of B. burgdorferi in more than 200 infected I. pacificus nymphs from Mendocino County, CA, where the incidence of LB is among the highest in the state (11). Further, we compare the Mendocino County spirochete population to populations found in the Northeast.  相似文献   

17.
18.
19.
20.
In the current study, we examined the regulatory interactions of a serine/threonine phosphatase (BA-Stp1), serine/threonine kinase (BA-Stk1) pair in Bacillus anthracis. B. anthracis STPK101, a null mutant lacking BA-Stp1 and BA-Stk1, was impaired in its ability to survive within macrophages, and this correlated with an observed reduction in virulence in a mouse model of pulmonary anthrax. Biochemical analyses confirmed that BA-Stp1 is a PP2C phosphatase and dephosphorylates phosphoserine and phosphothreonine residues. Treatment of BA-Stk1 with BA-Stp1 altered BA-Stk1 kinase activity, indicating that the enzymatic function of BA-Stk1 can be influenced by BA-Stp1 dephosphorylation. Using a combination of mass spectrometry and mutagenesis approaches, three phosphorylated residues, T165, S173, and S214, in BA-Stk1 were identified as putative regulatory targets of BA-Stp1. Further analysis found that T165 and S173 were necessary for optimal substrate phosphorylation, while S214 was necessary for complete ATP hydrolysis, autophosphorylation, and substrate phosphorylation. These findings provide insight into a previously undescribed Stp/Stk pair in B. anthracis.A profile of the intracellular signaling proteins that regulate transition of Bacillus anthracis from dormancy to expression of virulence factors is emerging. Like many prokaryotes, B. anthracis utilizes two-component histidine kinase systems to regulate physiological changes and the expression of virulence factors. These systems include the Spo0 histidine kinase-based phosphorelay pathway (32, 37) and the Bacillus respiratory response A and B system involved in regulating toxin expression (36). Unlike for histidine kinase systems, little is known about reversible serine/threonine phosphorylation events in B. anthracis. These systems are common to eukaryotic cells (3, 14, 25, 40) but were only recently found in prokaryotes to modulate a variety of metabolic and physiological processes (1, 2, 7, 11, 12, 15, 17, 24, 28, 35, 38). Whether reversible serine/threonine phosphorylation contributes to similar events in B. anthracis is not known.The current paradigm for prokaryotic serine/threonine kinases (STK) is based in part on the structure of PknB, a serine/threonine kinase from Mycobacterium tuberculosis that is structurally related to eukaryotic Hanks-type kinases (39). PknB autophosphorylates and is dephosphorylated by an M. tuberculosis phosphatase, PstP, in order to alter kinase activity (4). Similar to the findings for PnkB, Madec et al. identified critical autophosphorylated residues and autophosphorylated domains of PrkC, an STK from Bacillus subtilis (22), which suggested that the phosphorylation state of these residues impacts the activation of PrkC (22). These studies suggested that prokaryotic STKs exhibited activities similar to those of their eukaryotic homologs and were regulated by cognate phosphatases. Hence, studies of serine/threonine phosphatase (STP)/STK pairs may help define a core regulatory module in bacterial physiology and virulence, wherein the kinase autophosphorylates following interaction with stimuli and is subsequently downregulated by a cognate phosphatase when stimulus levels decline.Analysis of the B. anthracis genome indicates that this organism has a single phosphatase-kinase pair encoded within a putative operon. This operon, between nucleotides 3588319 and 3678099 in the genome of B. anthracis Sterne, contains eight candidate open reading frames (ORFs). Six of the potential ORFs encode proteins involved in translation and DNA metabolism, while the phosphatase-encoding ORF (stp1) and the kinase-encoding ORF (stk1) are paired at the 3′ end of this operon. Examination of the genome sequences of several other Gram-positive bacteria indicates that this putative operon and the general orientation of stp1 and stk1 are conserved among members of the Firmicutes group of bacteria. B. anthracis Stp1 (BA-Stp1) and BA-Stk1 homologs influence a variety of bacterial processes. For example, homologs of BA-Stp1 and BA-Stk1 regulate growth in Bacillus subtilis (12), cell viability and segregation in Streptococcus agalactiae (28), competence in Streptococcus pneumoniae (26), and virulence in both Streptococcus pyogenes (17) and Staphylococcus aureus (9). Although kinases homologous to BA-Stk1 influence several bacterial processes in different species, the tandem association of this kinase with a phosphatase does not vary. This observation led us to hypothesize that the phosphatase (BA-Stp1) influences Ba-Stk1 activity by dephosphorylation.In the current study, we analyzed the importance of BA-Stp1 and BA-Stk1 in the virulence of B. anthracis and assessed the biochemical interactions between these two proteins. Results from these studies indicate that this phosphatase-kinase pair contributes to the virulence of B. anthracis, as mutants lacking BA-Stp1 and BA-Stk1 exhibit decreased lethality in a mouse model of pulmonary anthrax. Furthermore, a series of biochemical analyses reveal an interaction between BA-Stk1 and BA-Stp1 where BA-Stk1 autophosphorylates in order to enhance kinase activity and is dephosphorylated by BA-Stp1 as a putative step in downregulating kinase activity as the levels of stimuli subside. Moreover, we have identified candidate serine and threonine residues that appear to modulate kinase activity. These findings provide insight into a previously undescribed serine/threonine phosphatase-kinase system in B. anthracis.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号