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.
Production of verocytotoxin or Shiga-like toxin (Stx), particularly Stx2, is the basis of hemolytic uremic syndrome, a frequently lethal outcome for subjects infected with Stx2-producing enterohemorrhagic Escherichia coli (EHEC) strains. The toxin is formed by a single A subunit, which promotes protein synthesis inhibition in eukaryotic cells, and five B subunits, which bind to globotriaosylceramide at the surface of host cells. Host enzymes cleave the A subunit into the A1 peptide, endowed with N-glycosidase activity to the 28S rRNA, and the A2 peptide, which confers stability to the B pentamer. We report the construction of a DNA vaccine (pStx2ΔAB) that expresses a nontoxic Stx2 mutated form consisting of the last 32 amino acids of the A2 sequence and the complete B subunit as two nonfused polypeptides. Immunization trials carried out with the DNA vaccine in BALB/c mice, alone or in combination with another DNA vaccine encoding granulocyte-macrophage colony-stimulating factor, resulted in systemic Stx-specific antibody responses targeting both A and B subunits of the native Stx2. Moreover, anti-Stx2 antibodies raised in mice immunized with pStx2ΔAB showed toxin neutralization activity in vitro and, more importantly, conferred partial protection to Stx2 challenge in vivo. The present vector represents the second DNA vaccine so far reported to induce protective immunity to Stx2 and may contribute, either alone or in combination with other procedures, to the development of prophylactic or therapeutic interventions aiming to ameliorate EHEC infection-associated sequelae.Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC) strains are important food-borne pathogens representing the major etiological agents of hemorrhagic colitis and hemolytic uremic syndrome (HUS), a life-threatening disease characterized by hemolytic anemia, thrombocytopenia, and renal failure (19). The infection correlates with ingestion of contaminated meat or vegetables but is also transmitted by water or even person-to-person contact (8, 14, 44). Sporadic or massive outbreaks have been reported in several developed countries but, in Argentina, HUS is endemic and represents a serious public health problem with high morbidity and mortality rates (29, 40). Production of verocytotoxin or Shiga-like toxin (Stx) is the basis of EHEC pathogenesis (18, 20). The toxin is formed by a single A subunit, which possesses N-glycosidase activity to the 28S rRNA and promotes protein synthesis inhibition in eukaryotic cells, and five B subunits, which bind to globotriaosylceramide at the surface of host cells (9, 28). Although two major types (Stx1 and Stx2) and several subtypes have been described, Stx2 and Stx2c are the most frequently found toxins in severe HUS cases among EHEC-infected subjects (12, 41). The degree of antigenic cross-reactivity between Stx2 and Stx1 is low, and several authors have reported that the two toxins do not provide heterologous protection, particularly concerning the B subunits (45, 47). On the other hand, Stx2c and Stx2d variants are readily neutralized by antibodies against Stx2 (27).Despite the magnitude of the social and economic impacts caused by EHEC infections, no licensed vaccine or effective therapy is presently available for human use. So far, attempts to develop vaccine formulations against EHEC-associated sequelae have relied mainly on induction of serum anti-Stx antibody responses. Several approaches have been pursed to generate immunogenic anti-Stx vaccine formulations and include the use of live attenuated bacterial strains (2, 32), protein-conjugated polysaccharides (21), purified B subunit (33, 48), B-subunit-derived synthetic peptides (15), and mutated Stx1 and Stx2 nontoxic derivatives (5, 6, 13, 16, 37, 39, 42, 45).In a previous report we described anti-Stx2 DNA vaccines encoding either the B subunit or a fusion protein between the B subunit and the first N-terminal amino acid of the A1 subunit (8). The DNA vaccine encoding the hybrid protein elicited Stx-specific immune responses in mice and partial protection to Stx2 challenge (1, 33). Recent data have indicated that epitopes leading to generation of Stx-neutralizing antibodies are present on both the B as well as the A subunit (34, 45, 46). In addition, further evidence indicates that the A2 subunit contains some of the most immunogenic epitopes of the Stx2 toxin (4). Thus, in line with such evidence, we attempted the construction of a new DNA vaccine encoding the last 32 amino acids from the A2 subunit, in addition to the complete B subunit of Stx2, as separated polypeptides which could enhance the immunogenicity and protective effects of the vaccine formulation. In the present report, we describe the generation of a new DNA vaccine encoding both Stx2 A2 and B subunits as an approach to elicit protective antibody responses to Stx2. The results obtained demonstrate that immunization with this vaccine formulation results in systemic antibody responses to Stx2 A and B subunits and toxin neutralization activity both in vitro and in vivo.  相似文献   

2.
Shiga-like toxin 2 (Stx2)-producing enterohemorrhagic Escherichia coli (referred to as EHEC or STEC) strains are the primary etiologic agents of hemolytic-uremic syndrome (HUS), which leads to renal failure and high mortality rates. Expression of Stx2 is the most relevant virulence-associated factor of EHEC strains, and toxin neutralization by antigen-specific serum antibodies represents the main target for both preventive and therapeutic anti-HUS approaches. In the present report, we describe two Salmonella enterica serovar Typhimurium aroA vaccine strains expressing a nontoxic plasmid-encoded derivative of Stx2 (Stx2ΔAB) containing the complete nontoxic A2 subunit and the receptor binding B subunit. The two S. Typhimurium strains differ in the expression of flagellin, the structural subunit of the flagellar shaft, which exerts strong adjuvant effects. The vaccine strains expressed Stx2ΔAB, either cell bound or secreted into the extracellular environment, and showed enhanced mouse gut colonization and high plasmid stability under both in vitro and in vivo conditions. Oral immunization of mice with three doses of the S. Typhimurium vaccine strains elicited serum anti-Stx2B (IgG) antibodies that neutralized the toxic effects of the native toxin under in vitro conditions (Vero cells) and conferred partial protection under in vivo conditions. No significant differences with respect to gut colonization or the induction of antigen-specific antibody responses were detected in mice vaccinated with flagellated versus nonflagellated bacterial strains. The present results indicate that expression of Stx2ΔAB by attenuated S. Typhimurium strains is an alternative vaccine approach for HUS control, but additional improvements in the immunogenicity of Stx2 toxoids are still required.Shiga-like toxins (Stx) play a crucial role in the pathogenesis of enterohemorrhagic Escherichia coli (EHEC) strains, which may lead to hemorrhagic colitis, central nervous system disturbances, and hemolytic-uremic syndrome (HUS) (27, 33). HUS involves acute renal failure, thrombocytopenia, and microangiopathic hemolytic anemia, with mortality rates ranging from 1% to 4% (45, 50). EHEC strains may express different serotypes, including the widely distributed O157:H7 serotype, and infection correlates with the ingestion of contaminated ground beef and cow manure-contaminated water, vegetables, juices, and other products (13, 18). The incidence of EHEC-associated HUS cases is particularly high in developed countries, and high incidence rates have been recorded in Argentina, where cultural and diverse epidemiological factors contribute to the widespread dissemination of the disease among children and teenagers (38).EHEC strains may express two different Stx types. Stx1 is virtually identical to Stx produced by Shigella dysenteriae, while Stx2 shows only 56% homology to Stx1 at the amino acid sequence level (14, 33, 51). Both toxin types are formed by one A subunit and five B subunits, which bind to glycosphingolipid receptors, such as globotriaosyl ceramide (Gb3), on host cell membranes and promote retrograde toxin transport through the Golgi complex and endoplasmic reticulum. In the cell cytoplasm, Stx2 subunit A is processed into two fragments; one of them (A1) is endowed with N-glycosidase activity, which depurinates a specific adenine residue of the eukaryotic 28S rRNA, inhibits protein synthesis, and induces apoptosis of the target cell (18, 51).After ingestion and gut colonization, Stx molecules are released by the bacterial cells and translocate across the gut epithelium to reach, via the bloodstream, capillary endothelial cells at renal glomeruli, where the most relevant tissue damage occurs (33, 45, 50). Epidemiological data indicate that individuals infected with Stx2-producing bacterial strains, and some closely related variants, have a high probability of developing HUS (45, 50). In addition, Stx2 expression has been shown to increase gut colonization by bacterial cells due to induction of increased receptor expression by enterocytes (39).So far, there is no effective prophylactic or therapeutic approach for the prevention of HUS development among EHEC-infected individuals. The treatments available involve platelet transfusion in cases of severe anemia, hemodialysis, and supportive care (7, 50). A more direct anti-Stx treatment under clinical or preclinical evaluation involves the use of synthetic Stx glycolipid receptor analogs and humanized anti-Stx monoclonal antibodies (44, 52).Attempts to develop prophylactic anti-HUS vaccines are focused on the generation of Stx-neutralizing antibodies or the blockade of gut colonization. The vaccine strategies based on Stx2 that have been tested under experimental conditions have included DNA vaccines (5, 12), protein-conjugated polysaccharides (28), purified recombinant B subunits (24, 25, 29, 30, 47, 53, 55, 58), and B-subunit-derived synthetic peptides (19, 20). Anti-EHEC vaccine approaches based on the blockade of gut colonization have employed intimin and type III secreted proteins, such as EspA and EspB (3, 37, 54).Live bivalent anti-Stx vaccines based on genetically modified, attenuated Vibrio cholerae or Salmonella enterica serovar Typhimurium strains have been reported to induce anti-StxB antibody responses following oral administration to mice or rabbits (1, 10, 49). Attenuated Salmonella strains, used as orally administered vaccine vectors for the expression of heterologous antigens, show several advantages over conventional parenterally delivered cellular or acellular vaccine formulations (15, 16). Attenuated Salmonella strains are safe, are easily administered by untrained personnel, and, more relevantly, may induce systemic and secreted antigen-specific antibody and cell-based immune responses against self and heterologous antigens. In addition, whole bacterial cells carry on their surfaces several molecular structures known to activate both innate and adaptive immune responses. These molecules, such as lipopolysaccharide and flagellin, act as strong adjuvants, both systemically and at mucosal surfaces.Flagellins, the structural subunit of flagellar filaments, contribute both to the virulence of bacterial pathogens and to the activation of inflammatory responses in mammalian hosts. Bacterial flagellins have been shown to bind both extracellular and intracellular receptors of antigen-presenting cells, leading to inflammation and increased adaptive immune responses, including the generation of antigen-specific antibodies and T cells (2, 26). The strong adjuvant effects of Salmonella flagellins, either when admixed with purified antigens or when used as hybrid proteins genetically fused to the target antigens, have been demonstrated recently (4, 8, 22, 23, 36). However, there is no clear evidence that the expression of flagellin affects the immunogenicity of heterologous antigens expressed by attenuated Salmonella vaccine strains.In the present study, we generated new experimental anti-HUS vaccine formulations based on two recombinant attenuated S. Typhimurium aroA vaccine strains differing in the expression of flagellin. The two strains were genetically modified in order to express a nontoxic Stx2 derivative consisting of the whole Stx2 B subunit and a partially deleted A subunit encompassing the first amino acid of the A1 subunit genetically fused to the whole A2 subunit (Stx2ΔAB). The Stx2ΔAB protein was previously tested in mice immunized with a DNA vaccine (5). The results of the present study show that the S. Typhimurium vaccine strains express and secrete the recombinant toxin and induce both systemic and mucosal anti-StxB antibodies with anti-Stx2 neutralization activity, conferring partial protection against intravenous (i.v.) challenge with Stx2.  相似文献   

3.
In addition to causing diarrhea, Escherichia coli O157:H7 infection can lead to hemolytic-uremic syndrome (HUS), a severe disease characterized by hemolysis and renal failure. Differences in HUS frequency among E. coli O157:H7 outbreaks have been noted, but our understanding of bacterial factors that promote HUS is incomplete. In 2006, in an outbreak of E. coli O157:H7 caused by consumption of contaminated spinach, there was a notably high frequency of HUS. We sequenced the genome of the strain responsible (TW14359) with the goal of identifying candidate genetic factors that contribute to an enhanced ability to cause HUS. The TW14359 genome contains 70 kb of DNA segments not present in either of the two reference O157:H7 genomes. We identified seven putative virulence determinants, including two putative type III secretion system effector proteins, candidate genes that could result in increased pathogenicity or, alternatively, adaptation to plants, and an intact anaerobic nitric oxide reductase gene, norV. We surveyed 100 O157:H7 isolates for the presence of these putative virulence determinants. A norV deletion was found in over one-half of the strains surveyed and correlated strikingly with the absence of stx1. The other putative virulence factors were found in 8 to 35% of the O157:H7 isolates surveyed, and their presence also correlated with the presence of norV and the absence of stx1, indicating that the presence of norV may serve as a marker of a greater propensity for HUS, similar to the correlation between the absence of stx1 and a propensity for HUS.Escherichia coli O157:H7 is a human pathogen that infects more than 73,000 North Americans per year (39). Although infection by this organism typically causes symptoms such as watery or bloody diarrhea, it may also lead to the development of hemolytic-uremic syndrome (HUS), an infection sequela characterized by hemolysis and renal failure that can result in long-lasting kidney damage. Variables that contribute to the development of HUS include host factors, such as age (51), and the genetic background of the enterohemorrhagic E. coli (EHEC) isolate. Currently, no effective prophylaxis exists for HUS (45). Antibiotic treatment of E. coli O157:H7 infections is contraindicated as it is associated with increased infection sequelae (45, 58).Humans become infected with EHEC by consuming contaminated food. EHEC are noninvasive pathogens that primarily colonize the human colon. Serotype O157:H7 is the predominant EHEC serotype in North America. The other commonly isolated EHEC serotypes include O26:H11, O103:H2, O111:NM, and O113:H21 (34). The systemic absorption of Shiga toxins produced by intestinal EHEC is thought to damage endothelial cells and to cause HUS (31). Shiga toxins are A-B-type toxins that inhibit protein synthesis. The genes encoding these potent toxins are borne on prophages that are related to phage λ. There are two main variants of Shiga toxin, Stx1 and Stx2. Stx2 is more cytotoxic than Stx1 in cell culture and animal models (27, 46, 48), and epidemiologic observations have revealed that the risk of developing HUS following an EHEC infection is heightened if the isolate produces Stx2 (4). Several variants of Stx2 exist, and Stx2c is the variant most commonly found in O157:H7 strains. Stx2 and Stx2c have the same biological function and possess identical A subunits and B subunits that share at least 97% identity (10).Although important for virulence, Stx2 does not appear to be the only EHEC factor that significantly influences whether patients infected with EHEC develop HUS. A comparison of statistics for several outbreaks caused by Stx2-producing O157:H7 strains showed that the rate of HUS can vary from less than 1% to 26% (23), indicating that strain-specific factors of stx2-carrying O157:H7 strains are involved in determining clinical outcomes. To date, the most significant factor identified as a factor contributing to the variability is the presence of the stx1 gene. O157:H7 strains that lack stx1 but carry one or two stx2 alleles are more likely to cause infections resulting in HUS (11, 35, 36).A comparison of the genome sequences of O157:H7 outbreak isolates that have resulted in different HUS rates may provide further insight into genetic factors that contribute to this severe sequela of EHEC infection. The genome sequences of two O157:H7 strains that caused low frequencies of HUS are available. The Sakai strain, the cause of the 1996 outbreak in Japan, caused ∼8,000 infections in people, the majority of whom were children, and the rate of HUS was 1.2% (32). In 1982, EDL933 caused the first diarrhea outbreak linked to the O157:H7 serotype and involved 44 individuals but no recorded HUS cases (41).Sakai shares 4.1 Mb of DNA with the commensal E. coli K-12 strain MG1655 and has 296 novel DNA segments more than 19 bp long, termed S-loops, that account for 1.39 Mb. EDL933 shares 4.1 Mb with E. coli K-12 strain MG1655 and has 177 unique sequence segments more than 50 bp long, termed O-islands, that account for 1.34 Mb (19). For both the Sakai and EDL933 genomes there is significant evidence of horizontal transfer due to the presence of numerous prophage-related elements and the pO157 virulence plasmid. The virulence factors carried on the O157:H7-specific DNA segments, as well as pO157, include stx1, stx2, the locus of enterocyte effacement (LEE), which confers the ability to cause attaching and effacing lesions on enterocytes and, notably, encodes a type III secretion system (TTSS) (22), at least 39 TTSS effectors encoded either on the LEE or at other chromosomal locations (49), numerous fimbrial and nonfimbrial adhesins, and more than one hemolysin (56).No genome sequence is available yet for an O157:H7 outbreak isolate that has caused an outbreak resulting in a significantly higher HUS rate. One O157:H7 isolate, TW14359, caused an outbreak associated with contaminated spinach that sickened 205 individuals in September and October of 2006. A total of 15% of the afflicted individuals developed HUS (5, 28). This rate is significantly higher than the average annual rate of 4.1% for O157:H7 cases that develop HUS (39). The relatively high percentage of adults, ∼8%, who developed HUS in the TW14359 outbreak also likely reflects the greater virulence of this strain (6). Furthermore, Manning et al. performed a phylogenetic analysis of TW14359 utilizing 96 single-nucleotide polymorphisms (SNPs) and demonstrated that this isolate belongs to a more virulent clade of O157:H7 strains (clade 8); the majority of these isolates lack stx1 and carry stx2 (28). A partial genome sequence consisting of 200 contigs of the TW14359 genome was also reported by Manning et al., which was found to contain stx2 and stx2c. While an analysis of these sequence data identified the genes of the two reference isolates that were also present in TW14359 and identified backbone SNPs, it did not provide a list of novel genetic features or provide assembled DNA segments containing repetitive DNA elements, such as phage-like elements. Here we describe the entire genome sequence of this isolate and, focusing on novel genetic material, identify potential genetic features of TW14359 that may promote this strain''s outstanding pathogenicity.  相似文献   

4.
Shiga toxin (Stx)-producing Escherichia coli (STEC) strains of serogroup O91 are the most common human pathogenic eae-negative STEC strains. To facilitate diagnosis and subtyping of these pathogens, we genotypically and phenotypically characterized 100 clinical STEC O91 isolates. Motile strains expressed flagellar antigens H8 (1 strain), H10 (2 strains), H14 (52 strains), and H21 (20 strains) or were H nontypeable (Hnt) (10 strains); 15 strains were nonmotile. All nonmotile and Hnt strains possessed the fliC gene encoding the flagellin subunit of the H14 antigen (fliCH14). Most STEC O91 strains possessed enterohemorrhagic E. coli hlyA and expressed an enterohemolytic phenotype. Among seven stx alleles identified, stx2dact, encoding mucus- and elastase-activatable Stx2d, was present solely in STEC O91:H21, whereas most strains of the other serotypes possessed stx1. Moreover, only STEC O91:H21 possessed the cdt-V cluster, encoding cytolethal distending toxin V; the toxin was regularly expressed and was lethal to human microvascular endothelial cells. Infection with STEC O91:H21 was associated with hemolytic-uremic syndrome (P = 0.0015), whereas strains of the other serotypes originated mostly in patients with nonbloody diarrhea. We conclude that STEC O91 clinical isolates belong to at least four lineages that differ by H antigens/fliC types, stx genotypes, and non-stx putative virulence factors, with accumulation of virulence determinants in the O91:H21 lineage. Isolation of STEC O91 from patients'' stools on enterohemolysin agar and the rapid initial subtyping of these isolates using fliC genotyping facilitate the identification of these emerging pathogens in clinical and epidemiological studies and enable prediction of the risk of a severe clinical outcome.Shiga toxin (Stx)-producing Escherichia coli (STEC) strains cause diarrhea and a life-threatening hemolytic-uremic syndrome (HUS) worldwide (23, 44). STEC strains isolated from patients usually possess, in addition to one or more stx genes, the eae gene, encoding adhesin intimin (7, 11, 16, 25, 26, 41, 49). However, a subset of STEC strains associated with human disease lack eae, and among these, strains of serogroup O91 are the most common (2, 7, 35, 37, 47, 48). In Germany during the last 5 years, serogroup O91 accounted for 6.4% to 11.0% of all STEC strains reported from human infections and was therefore the fourth-most-common STEC serogroup (after O157, O26, and O103) isolated (47, 48; http://www.rki.de). However, in contrast to eae-positive STEC strains of the three leading serogroups, which cause disease mostly in young children (47), STEC O91 is the most common serogroup isolated from adult patients (48).Despite their association with human diseases worldwide (7, 9, 11, 13, 14, 30, 35, 37, 38, 40, 47, 48), the spectrum of serotypes of STEC O91 isolates from patients and the pathogenic traits of such strains are poorly understood. Moreover, characteristics of STEC O91 strains which could assist with their isolation from human stools and further subtyping in clinical microbiological laboratories have not been systematically investigated or reported. To gain insight into the serotype composition and putative virulence factors of STEC O91 strains causing human disease and to identify characteristics which can facilitate laboratory diagnosis of these organisms, we determined the motility and flagellar phenotypes, fliC types, stx genotypes, non-stx putative virulence loci, and diagnostically useful phenotypes of 100 clinical STEC O91 isolates. Moreover, we investigated possible associations between bacterial characteristics and clinical infection phenotypes.  相似文献   

5.
Hemolytic-uremic syndrome (HUS) caused by Shiga toxin-producing Escherichia coli infection is a leading cause of pediatric acute renal failure. Bacterial toxins produced in the gut enter the circulation and cause a systemic toxemia and targeted cell damage. It had been previously shown that injection of Shiga toxin 2 (Stx2) and lipopolysaccharide (LPS) caused signs and symptoms of HUS in mice, but the mechanism leading to renal failure remained uncharacterized. The current study elucidated that murine cells of the glomerular filtration barrier were unresponsive to Stx2 because they lacked the receptor glycosphingolipid globotriaosylceramide (Gb3) in vitro and in vivo. In contrast to the analogous human cells, Stx2 did not alter inflammatory kinase activity, cytokine release, or cell viability of the murine glomerular cells. However, murine renal cortical and medullary tubular cells expressed Gb3 and responded to Stx2 by undergoing apoptosis. Stx2-induced loss of functioning collecting ducts in vivo caused production of increased dilute urine, resulted in dehydration, and contributed to renal failure. Stx2-mediated renal dysfunction was ameliorated by administration of the nonselective caspase inhibitor Q-VD-OPH in vivo. Stx2 therefore targets the murine collecting duct, and this Stx2-induced injury can be blocked by inhibitors of apoptosis in vivo.Shiga toxin-producing Escherichia coli (STEC) is the principal etiologic agent of diarrhea-associated hemolytic-uremic syndrome (HUS) (42, 60, 66). Renal disease is thought to be due to the combined action of Shiga toxins (Shiga toxin 1 [Stx1] and Stx2), the primary virulence factors of STEC, and bacterial lipopolysaccharide (LPS) on the renal glomeruli and tubules (6, 42, 60, 66). Of these, Stx2 is most frequently associated with the development of HUS (45). Shiga toxin enters susceptible cell types after binding to the cell surface receptor glycosphingolipid globotriaosylceramide (Gb3) and specifically depurinates the 28S rRNA, thereby inhibiting protein synthesis (42, 60, 66). The damage initiates a ribotoxic stress response consisting of mitogen-activated protein (MAP) kinase activation, and this response can be associated with cytokine release and cell death (21, 22, 25-27, 61, 69, 73). This cell death is often caspase-dependent apoptosis (18, 61). Gb3 is expressed by human glomerular endothelial cells, podocytes, and multiple tubular epithelial cell types, and damage markers for these cells can be detected in urine samples from HUS patients (10-12, 15, 49, 73). Shiga toxin binds to these cells in renal sections from HUS patients, and along with the typical fibrin-rich glomerular microangiopathy, biopsy sections demonstrate apoptosis of both glomerular and tubular cell types (9, 29, 31).Concomitant development of the most prominent features of HUS: anemia, thrombocytopenia, and renal failure, requires both Shiga toxin and LPS in the murine model (30, 33). Nevertheless, our previous work demonstrated that renal failure is mediated exclusively by Stx2 (33). While it is established that Gb3 is the unique Shiga toxin receptor (46), the current literature regarding the mechanism by which Shiga toxin causes renal dysfunction in mice is inconsistent. Even though Gb3 has been localized to some murine renal tubules and tubular damage has been observed (19, 23, 46, 53, 65, 68, 72, 74), the specific types of tubules affected have been incompletely characterized. Although multiple groups have been unable to locate the Shiga toxin receptor Gb3 in glomeruli in murine renal sections (19, 53), one group has reported that murine glomerular podocytes possess Gb3 and respond to Stx2 in vitro (40), and another group has reported that renal tubular capillaries express the Gb3 receptor (46). Furthermore, murine glomerular abnormalities, including platelet and fibrin deposition, occur in some murine HUS models (28, 30, 33, 46, 59, 63). We demonstrate here that murine glomerular endothelial cells and podocytes are unresponsive to Stx2 because they do not produce the glycosphingolipid receptor Gb3 in vitro or in vivo. Further, murine renal tubules, including collecting ducts, express Gb3 and undergo Stx2-induced apoptosis, resulting in dysfunctional urine production and dehydration.  相似文献   

6.
Shiga toxin 2 (Stx2) is a major virulence factor in infections with Stx-producing Escherichia coli (STEC), which causes gastrointestinal diseases and sometimes fatal systemic complications. Recently, we developed an oral Stx2 inhibitor known as Ac-PPP-tet that exhibits remarkable therapeutic potency in an STEC infection model. However, the precise mechanism underlying the in vivo therapeutic effects of Ac-PPP-tet is unknown. Here, we found that Ac-PPP-tet completely inhibited fluid accumulation in the rabbit ileum caused by the direct injection of Stx2. Interestingly, Ac-PPP-tet accumulated in the ileal epithelial cells only through its formation of a complex with Stx2. The formation of Ac-PPP-tet-Stx2 complexes in cultured epithelial cells blocked the intracellular transport of Stx2 from the Golgi apparatus to the endoplasmic reticulum, a process that is essential for Stx2 cytotoxicity. Thus, Ac-PPP-tet is the first Stx neutralizer that functions in the intestine by altering the intracellular transport of Stx2 in epithelial cells.Infection with Shiga toxin (Stx)-producing Escherichia coli (STEC) in humans causes gastrointestinal diseases that are often followed by potentially fatal systemic complications such as acute encephalopathy and hemolytic-uremic syndrome (12, 22, 25, 26). Stx is produced in the gut, traverses the epithelium, and passes into the circulation. Circulating Stx then causes vascular damage in specific target tissues such as the brain and the kidney, resulting in systemic complications. For this reason, development of a neutralizer that specifically binds to and inhibits Stx in the gut and/or in the circulation would be a promising therapeutic approach.Stx is classified into two subgroups, Stx1 and Stx2. Stx2 is more closely related to the severity of STEC infections than Stx1 (6, 23, 31, 33). Stx consists of a catalytic A subunit and a pentameric B subunit. The former has 28S rRNA N-glycosidase activity and inhibits eukaryotic protein synthesis, while the latter is responsible for binding to the functional cell surface receptor Gb3 [Galα(1-4)-Galβ(1-4)-Glcβ1-ceramide] (11, 17, 25). The crystal structure of Stx reveals the presence of three distinctive binding sites (i.e., sites 1, 2, and 3) on each B subunit monomer for the trisaccharide moiety of Gb3 (7, 16). Highly selective and potent binding of Stx to Gb3 is attributed mainly to the multivalent interaction between the B subunit pentamer and the trisaccharide. This so-called clustering effect has formed the basis for the development of several synthetic Stx neutralizers that contain the trisaccharide in multiple configurations (3, 5, 14, 18, 19, 36). These neutralizers can strongly bind to Stx and inhibit its cytotoxic activity. Some are also effective in STEC infection models (18, 19, 36). However, the clinical application of these neutralizers has been substantially hampered by the synthetic complexity of the trisaccharide moiety.We have recently screened a library of novel tetravalent peptides that exert a clustering effect and have identified four peptide motifs that are superior to trisaccharide in binding Stx (20). Tetravalent forms of these peptides bind with high affinity to one trisaccharide-binding site (site 3) of Stx2 and effectively inhibit Stx2 cytotoxicity. This is particularly true of the neutralizer designated PPP-tet, which contains four Pro-Pro-Pro-Arg-Arg-Arg-Arg motifs. PPP-tet protects mice from a fatal dose of E. coli O157:H7, even when the peptide is orally administered after the establishment of infection (20). Moreover, the addition of acetyl groups to all the amino termini of PPP-tet (yielding Ac-PPP-tet) makes this compound resistant to proteolysis and markedly enhances its protective activity against STEC infection, indicating that Ac-PPP-tet holds promise as a therapeutic agent for STEC infections.After binding to Gb3, Stx is first transported to the Golgi apparatus in a retrograde manner and then transported to the endoplasmic reticulum (ER). On the other hand, the Stx catalytic A subunit is released into the cytoplasm, where it inhibits protein synthesis (27, 29). The retrograde transport of Stx is known to be essential for Stx cytotoxicity (2, 27, 28). In Vero cells, one of the cell types most sensitive to Stx, PPP-tet prevents Stx2 cytotoxicity by inducing the aberrant transport of Stx from the Golgi apparatus to an acidic compartment rather than to the ER, leading to the degradation of Stx (20). An advantage of PPP-tet is its ability to partially permeate cells, which allows it to inhibit the cytotoxicity of Stx2 already incorporated into cells (20). Nevertheless, the precise mechanism by which PPP-tet and Ac-PPP-tet function in vivo, as well as the identities of the organs or cells targeted by these compounds, is unknown.To understand how orally administered Ac-PPP-tet functions in vivo, we investigated the effect of Ac-PPP-tet on fluid accumulation in the rabbit ileum caused by the direct injection of Stx2. The rabbit ileal loop system is highly valid for evaluating the toxicity of Stx2 produced in the intestine after infection. We also examined the localization of the tetravalent peptide and Stx2 in the intact rabbit ileum, cultured ileal specimens, and Caco-2 intestinal epithelial cells. Our results reveal that Ac-PPP-tet functions as a potent Stx2 neutralizer in the intestine by altering the intracellular transport of Stx2 in epithelial cells.  相似文献   

7.
Mice have been extensively employed as an animal model of renal damage caused by Shiga toxins. In this study, we examined the role of the proinflammatory cytokine tumor necrosis factor alpha (TNF-α) in the development of toxin-mediated renal disease in mice. Mice pretreated with TNF-α and challenged with Shiga toxin type 1 (Stx1) showed increased survival compared to that of mice treated with Stx1 alone. Conversely, mice treated with Stx1 before TNF-α administration succumbed more quickly than mice given Stx1 alone. Increased lethality in mice treated with Stx1 followed by TNF-α was associated with evidence of glomerular damage and the loss of renal function. No differences in renal histopathology were noted between animals treated with Stx1 alone and the TNF-α pretreatment group, although we noted a sparing of renal function when TNF-α was administered before toxin. Compared to that of treatment with Stx1 alone, treatment with TNF-α after toxin altered the renal cytokine profile so that the expression of proinflammatory cytokines TNF-α and interleukin-1β (IL-1β) increased, and the expression of the anti-inflammatory cytokine IL-10 decreased. Increased lethality in mice treated with Stx1 followed by TNF-α was associated with higher numbers of dUTP-biotin nick end labeling-positive renal tubule cells, suggesting that increased lethality involved enhanced apoptosis. These data suggest that the early administration of TNF-α is a candidate interventional strategy blocking disease progression, while TNF-α production after intoxication exacerbates disease.Shiga toxins are a family of genetically and functionally related cytotoxic proteins expressed by the enteric pathogens Shigella dysenteriae serotype 1 and certain serotypes of Escherichia coli. Antigenic similarity to Shiga toxin expressed by S. dysenteriae serotype 1 is used to define Shiga toxin type 1 (Stx1) and type 2 (Stx2) expressed by Shiga toxin-producing E. coli (STEC) (44). Shiga toxins consist of a single A subunit in noncovalent association with a pentamer of B subunits. B subunits mediate binding to the neutral glycolipid receptor globotriaosylceramide (Gb3), while the A subunit possesses an N-glycosidase activity (38). Following toxin internalization and routing to the endoplasmic reticulum (ER), a fragment of the toxin A subunit generated by furin or a furin-like protease is translocated across the ER membrane and mediates the cleavage of a single adenine residue (A4256 in the rat) from the 28S rRNA component of ribosomes (39). Stx-induced depurination leads to the disruption of elongation factor-dependent aminoacyl-tRNA binding to nascent polypeptides (30). Thus, Shiga toxins are potent protein synthesis inhibitors, with 50% cytotoxic doses measured in pg/ml amounts for many cell types in vitro. Shiga toxins also activate the ribotoxic and ER stress pathways, which are important in the activation of proinflammatory cytokine/chemokine production and apoptosis (6, 22, 41).The ingestion of small quantities of Stx-producing bacteria may lead to the development of bloody diarrhea with progression to acute renal failure, designated diarrhea-associated hemolytic uremic syndrome (D+HUS) (33). Epidemiologic studies have shown that the ingestion of STEC strains expressing Stx2 alone or Stx1 and Stx2 are more likely to progress to life-threatening extraintestinal complications (3, 17, 31). D+HUS is a leading cause of pediatric acute renal failure. D+HUS is characterized by rapid-onset oligouria or anuria, azotemia, microangiopathic hemolytic anemia with schistocytosis, and thrombocytopenia (33, 47). The histopathological examination of D+HUS renal tissues showed that glomerular microvascular endothelial cells were frequently swollen and detached from the basement membrane, and glomerular capillary lumina may be occluded with fibrin-rich microthrombi (21, 36).Numerous animal models have been employed to characterize the role of Stx1 and Stx2 in pathogenesis. Studies utilizing nonhuman primates showed that Shiga toxins are essential virulence determinants in the development of microangiopathic lesions. Fontaine et al. (9) showed that macaque monkeys fed toxigenic strains of S. dysenteriae developed colonic microvascular lesions, while baboons given purified intravenous Stx1 developed acute renal failure (48). The bolus intravenous administration of Stx1 or Stx2 into baboons revealed that the animals were more sensitive to Stx2, although the mean times to death were prolonged in Stx2-treated animals compared to that with Stx1 treatment. Both toxins mediated hematologic changes such as thrombocytopenia and schistocytosis, and both toxins produced renal pathology, but with different presentations. Renal damage caused by Stx1 was characterized by moderate congestion at the cortico-medullary junction, while Stx2-treated animals showed severe medullary congestion with cortical ischemia (42). Mice fed Stx2-producing E. coli or given a single bolus injection of purified Shiga toxins died without the development of glomerular thrombotic microangiopathy (50, 54). However, the administration of multiple low doses of Stx2 allowed the animals to survive initial toxin challenge and develop glomerular lesions characteristic of HUS in humans (40). In addition to the toxins, host response factors may contribute to D+HUS pathogenesis. Prodromal hemorrhagic colitis may alter normal colonic barrier function, and patients with D+HUS may be endotoxemic or show evidence of elevated antibody titers against lipopolysaccharides (LPS) expressed by Stx-producing E. coli (2, 10, 26). LPS elicit the expression of a broad array of pro- and anti-inflammatory cytokines and chemokines (45). In accordance with this, D+HUS patients frequently have increased serum or urinary proinflammatory cytokine and chemokine levels (15, 23). Studies using small-animal models support the hypothesis that additional bacterial and host response factors facilitate the development of renal disease. Keepers et al. (19) demonstrated that the coadministration of Stx2 and LPS to C57BL/6 mice did not produce major changes in lethality but resulted in pathophysiological changes more consistent with disease in humans: intraglomerular platelet and fibrin deposition, decreased renal function, neutrophilia, and lymphocytopenia. Barrett et al. (1) showed that the timing of toxin and LPS challenges were critical in disease outcome. LPS enhanced the lethal effects of purified Stx2 when administered to rabbits or mice after toxin challenge, whereas LPS protected the animals from Stx2 toxicity when administered before the toxin. Palermo et al. (32) showed that the LPS-induced modulation of Stx2 lethality was cytokine time and dose dependent. Mice given low doses of TNF-α or IL-1β 1 h before Stx2 treatment showed increased lethality when treated with Stx2, while mice given higher doses of IL-1β (sufficient to elicit corticosteroid production) were protected from Stx2 lethality.The proinflammatory cytokines TNF-α and IL-1β sensitize vascular endothelial cells to the cytotoxic action of Shiga toxins in vitro (24, 34, 53) through a mechanism involving the increased expression of genes involved in the biosynthesis of Gb3 (43). Murine and human macrophages or macrophage-like cell lines express proinflammatory cytokines and chemokines when treated with purified Shiga toxins (12, 35, 51). Keepers et al. (18) showed a marked monocytic cell infiltrate into the kidneys of mice given Stx2 and LPS. Collectively, these data suggest that the innate immune response to Shiga toxins, in the presence or absence of LPS, alters the outcome of renal disease. In the present study, we have examined the role of a single proinflammatory cytokine, TNF-α, in pathogenesis using the mouse model of Stx-induced renal damage. Our data suggest that the timing of TNF-α production affects the outcome of disease in that the presence of elevated TNF-α levels prior to toxin challenge protects animals from disease, while high TNF-α levels occurring after toxin administration result in accelerated lethality.  相似文献   

8.
9.
Enterohemorrhagic Escherichia coli (EHEC) strains are responsible for food poisoning in developed countries via consumption of vegetal and animal food sources contaminated by ruminant feces, and some strains (O26, O111, and O118 serogroups) are also responsible for diarrhea in young calves. The prevalence of 27 putative adhesins of EHEC and of bovine necrotoxigenic E. coli (NTEC) was studied with a collection of 43 bovine and 29 human enteropathogenic (EPEC) and EHEC strains and 5 non-EPEC/non-EHEC (1 bovine and 4 human) O26 strains, using specific PCRs. Four “groups” of adhesins exist, including adhesins present in all O26 strains, adhesins present in most O26 strains, adhesins present in a few O26 strains, and adhesins not present in O26 strains. The common profile of EHEC/EPEC strains was characterized by the presence of loc3, loc5, loc7, loc11, loc14, paa, efa1, iha, lpfAO26, and lpfAO113 genes and the absence of loc1, loc2, loc6, loc12, loc13, saa, and eibG genes. Except for the lpfAO26 gene, which was marginally associated with bovine EHEC/EPEC strains in comparison with human strains (P = 0.012), none of the results significantly differentiated bovine strains from human strains. One adhesin gene (ldaE) was statistically (P < 0.01) associated with O26 EHEC/EPEC strains isolated from diarrheic calves in comparison with strains isolated from healthy calves. ldaE-positive strains could therefore represent a subgroup possessing the specific property of producing diarrhea in young calves. This is the first time that the distribution of putative adhesins has been described for such a large collection of EHEC/EPEC O26 strains isolated from both humans and cattle.Enteropathogenic (EPEC) and enterohemorrhagic (EHEC) Escherichia coli strains represent two important classes of enteric pathogens that cause diarrhea in humans and animals. They have in common the ability to produce a histopathological lesion on enterocytes, called an “attaching and effacing” lesion. The intimate attachment of the bacteria to enterocytes and the localized effacement of microvilli are the main characteristics of the attaching and effacing lesion (26).EPEC strains are an important cause of infant diarrhea in developing countries and are often associated with high mortality rates (8). Human EPEC strains are subdivided into classical (type 1) and nonclassical (type 2) strains on the basis of the production of bundle-forming pili or the presence of the encoding genes. Nonclassical EPEC strains are also present in different animal species. In bovines, nonclassical EPEC strains are associated with diarrhea in young calves of up to 3 months of age (9).EHEC strains are considered to have evolved from EPEC strains through the acquisition of bacteriophages encoding Shiga toxins (Stxs) (31, 45). EHEC strains cause several clinical syndromes in humans (mainly in children and elderly people), such as diarrhea, hemorrhagic colitis, hemolytic-uremic syndrome, and thrombotic thrombocytopenic purpura. These have been responsible for large outbreaks in many developed countries, especially Japan, the United States, and the United Kingdom (26). Transmission can occur via consumption of vegetal and animal foodstuffs contaminated by ruminant feces (mainly cattle) (7). Some EHEC strains are also responsible for undifferentiated diarrhea in young calves of up to 3 months of age (24).EPEC and EHEC strains can belong to more than 1,000 O:H serotypes. In EHEC infections, O157:H7 is the main serotype responsible for several outbreaks and sporadic cases of hemorrhagic colitis and hemolytic-uremic syndrome, but non-O157 serogroups (such as O26, O145, O111, and O103) can also be associated frequently with severe illness in humans (5, 35). Though most, if not all, EHEC serogroups are carried by healthy animal ruminants, a few are associated with diarrhea in calves (O5, O26, O111, O118, etc.). Human and animal EPEC strains also belong to a series of O antigenic groups, including O26, O55, O86, O111, O114, O119, O125, O126, O127, O128, O142, and O158 (6). Thus, several serogroups are present in both pathotypes (EHEC and EPEC) and can infect both humans and cattle. Although classical EPEC strains have always been regarded as host specific, EHEC strains have not, and the actual situation regarding nonclassical EPEC strains remains unknown.The first step in EPEC and EHEC infection is the initial adherence of bacteria to intestinal cells. This adherence step could be the basis for any host specificity via the production of colonization factors, such as the bundle-forming pilus adhesin of classical human EPEC strains.Low et al. analyzed 14 putative fimbrial gene clusters revealed by the EHEC O157:H7 Sakai sequence (21). Of these 14 putative fimbriae, several had already been described under other names, including LpfA1 (42), LpfA2 (43), F9 (20), type 1 fimbriae (32) (34), and curli fimbriae (30). Long polar fimbria (Lpf)-encoding genes had also been described previously, including lpfAO26 and lpfAO113, described by Toma et al. (41) and Doughty et al. (12), respectively.In addition, other putative adhesins have been described, as follows: a 67-kDa adherence-conferring protein (Iha) similar to Vibrio cholerae IrgA confers the capacity to adhere to epithelial cells in a diffuse pattern (38); Efa1 (EHEC factor for adherence), described by Nicholls et al. (27), mediates the binding of bacteria to CHO cells in vitro; ToxB, a protein encoded by a gene located on the 93-kb pO157 plasmid, is required for full adherence of the EHEC O157:H7 Sakai strain (39); Saa is an autoagglutinating adhesin identified in locus of enterocyte effacement-negative verotoxigenic E. coli strains (29); EibG is a protein responsible for the chain-like adherence phenotype of Saa-negative verotoxigenic E. coli strains (22); Paa (porcine attaching and effacing-associated) adhesin, described by An et al. (1), is involved in the early steps of the adherence mechanism of porcine EPEC strains (2); and the hemorrhagic coli pilus (HCP), whose inactivation of the main subunit (hcpA gene) reduces adherence to cultured human intestinal and bovine renal epithelial cells and to porcine and bovine gut explants, was observed in EHEC O157:H7 (46).The aim of this study was to establish the prevalence in bovine and human EPEC and EHEC strains belonging to the O26 serogroup of a total of 23 putative adhesins previously described for EHEC strains and of four fimbrial and afimbrial adhesins associated with bovine necrotoxigenic E. coli (NTEC) (36). The presence of these genes was correlated, on the one hand, with the source of isolation, and on the other hand, with EHEC/EPEC virulence factors (eae, stx1, stx2, and EHEC hlyA).  相似文献   

10.
Enterohemorrhagic Escherichia coli O157 (EHEC O157) is a food-borne pathogen that has raised worldwide public health concern. The development of simple and rapid strain-typing methods is crucial for the rapid detection and surveillance of EHEC O157 outbreaks. In the present study, we developed a multiplex PCR-based strain-typing method for EHEC O157, which is based on the variability in genomic location of IS629 among EHEC O157 strains. This method is very simple, in that the procedures are completed within 2 h, the analysis can be performed without the need for special equipment or techniques (requiring only conventional PCR and agarose gel electrophoresis systems), the results can easily be transformed into digital data, and the genes for the major virulence markers of EHEC O157 (the stx1, stx2, and eae genes) can be detected simultaneously. Using this method, 201 EHEC O157 strains showing different XbaI digestion patterns in pulsed-field gel electrophoresis (PFGE) analysis were classified into 127 types, and outbreak-related strains showed identical or highly similar banding patterns. Although this method is less discriminatory than PFGE, it may be useful as a primary screening tool for EHEC O157 outbreaks.Enterohemorrhagic Escherichia coli O157:H7 (EHEC O157) is a food-borne pathogen that causes diarrhea, hemorrhagic colitis, and hemolytic-uremic syndrome in humans (18). Since its initial identification as a food-borne pathogen in 1982, EHEC O157 has been implicated in numerous outbreaks and sporadic cases, mainly in industrialized countries (8). To prevent and control EHEC O157 infections, rapid detection of outbreaks and the identification of contamination sources are crucial. Thus, suitable tools for epidemiologic studies and systematic surveillance, such as rapid and efficient strain-typing systems, are needed.Among the currently available methods for molecular typing of EHEC O157 strains, pulsed-field gel electrophoresis (PFGE) has the highest discrimination power and is widely used for epidemiologic studies and the surveillance of O157:H7 infections (1, 12, 19, 21). However, PFGE requires strong technical skills and 1 or more days to generate the results. It is also difficult to obtain consistently reproducible results among different laboratories, which hinders interlaboratory data comparisons. Other strain-typing methods for EHEC O157, including PCR-restriction fragment length polymorphism (15, 16), polymorphic amplified typing sequences (5, 6), and multiple-locus variable-number tandem repeat analysis (9), have also been developed. Although these methods have their own advantages, they require special techniques and/or equipment and are time-consuming.Previously, we determined the whole-genome sequence of E. coli O157:H7 strain RIMD 0509952 (referred to as EHEC O157 Sakai) and identified a total of 98 copies of insertion sequence (IS) elements in the genome (2). Among these, IS629 and ISEc8 predominated, with 23 copies of IS629 and 11 copies of ISEc8 being identified. Using whole-genome PCR scanning (WGPScanning) and microarray analyses of eight EHEC O157 clinical isolates (10, 11), we identified numerous genomic segments that carried structural polymorphisms (ranging from several hundred base pairs to a few thousand base pairs) among the eight EHEC O157 strains and EHEC O157 Sakai. Recently, we analyzed all of these segments and found insertions/deletions of IS629 and ISEc8 in the generation of these structural polymorphisms. In particular, the genomic locations of IS629 varied significantly between the strains; we identified a total of 77 genomic loci into which IS629 was inserted in some of the nine strains that we examined (13).In the present study, based on the variable genomic location of IS629 among EHEC O157 strains, we have developed a simple and rapid multiplex PCR-based, strain-typing method for EHEC O157 strains, which we term the O157 IS-printing method.  相似文献   

11.
Shiga toxin-producing Escherichia coli is a principal source of regional outbreaks of bloody diarrhea and hemolytic-uremic syndrome in the United States and worldwide. Primary bacterial virulence factors are Shiga toxin types 1 and 2 (Stx1 and Stx2), and we performed parallel analyses of the pathophysiologies elicited by the toxins in nonhuman primate models to identify shared and unique consequences of the toxemias. After a single intravenous challenge with purified Stx1 or Stx2, baboons (Papio) developed thrombocytopenia, anemia, and acute renal failure with loss of glomerular function, in a dose-dependent manner. Differences in the timing and magnitude of physiologic responses were observed between the toxins. The animals were more sensitive to Stx2, with mortality at lower doses, but Stx2-induced renal injury and mortality were delayed 2 to 3 days compared to those after Stx1 challenge. Multiplex analyses of plasma inflammatory cytokines revealed similarities (macrophage chemoattractant protein 1 [MCP-1] and tumor necrosis factor alpha [TNF-α]) and differences (interleukin-6 [IL-6] and granulocyte colony-stimulating factor [G-CSF]) elicited by the toxins with respect to the mediator induced and timing of the responses. Neither toxin induced detectable levels of plasma TNF-α. To our knowledge, this is the first time that the in vivo consequences of the toxins have been compared in a parallel and reproducible manner in nonhuman primates, and the data show similarities to patient observations. The availability of experimental nonhuman primate models for Stx toxemias provides a reproducible platform for testing antitoxin compounds and immunotherapeutics with outcome criteria that have clinical meaning.Infection with Shiga toxin-producing Escherichia coli (STEC) results in intestinal cramps and bloody diarrhea, followed 5 to 12 days later in some patients by the development of hemolytic-uremic syndrome (HUS) (16, 18). HUS is characterized clinically by the triad of thrombocytopenia, hemolytic microangiopathy, and renal injury and is the leading cause of acute renal failure in otherwise healthy children in the United States. An antibiotic regimen is not recommended, and treatment options are limited to critical care support (47). Patients with diarrhea-associated HUS can have long-term renal impairment of varying severity, and approximately one-fourth of patients have neurologic sequelae, including seizures, coma/stupor, cortical blindness, ataxia, and paraplegia (10, 14).The natural infection route is gastrointestinal, via contaminated food or water. The bacteria colonize the intestinal lumen, with most strains forming characteristic attaching-and-effacing lesions, and the organisms may synthesize and release one or more toxins that are primary virulence factors contributing to the clinical manifestations of HUS (19). The toxins are AB5 holotoxins, referred to as Shiga toxins due to their functional and structural similarities to Shiga toxin expressed by Shigella dysenteriae serotype 1 (4). Shiga toxin type 1 (Stx1) is essentially identical to the Shigella toxin (4), differing by one amino acid, but shares only 58% amino acid identity with Shiga toxin type 2 (Stx2). Stx1 and Stx2 have distinct spatial conformations (8) and dissociation rates from receptor-lipid surfaces (24). STEC strains may secrete one or both toxins and several toxin variants, and clinical studies have demonstrated that HUS is most often associated with the expression of Stx2 (3), particularly following infection with E. coli O157:H7 strains (12, 20). All Shiga toxins share a cellular intoxication mechanism in which B subunits oligomerize into pentamers for interaction with a cell surface globotriaosylceramide Gb3 (CD77) receptor. Following binding, holotoxins are internalized via clathrin-dependent or clathrin-independent mechanisms and undergo retrograde transport through the trans-Golgi network and Golgi apparatus to reach the endoplasmic reticulum (33, 46). During transport, the A subunit undergoes limited proteolysis, and once in the endoplasmic reticulum, a fragment of the A subunit translocates into the cytoplasm, where its N-glycosidase activity inactivates the 28S rRNA component of eukaryotic ribosomes to inhibit protein synthesis and cause cell death (25, 43).While Stx1 and Stx2 share many characteristics, they are not identical and there is evidence that toxin-specific activities may be clinically relevant. Both toxins are internalized after binding to Gb3, but the mechanisms of their intracellular trafficking through polarized intestinal epithelial cells to reach the intestinal endothelium are very different (15). Also, endothelial sensitivities to Stx1 and Stx2 differ depending on the vascular bed, with intestinal endothelium being more sensitive to the Shiga toxins than saphenous vein endothelium (12), and glomerular endothelial cells are about 1,000 times more sensitive to Stx2 than human umbilical vein endothelial cells (17). The mechanisms for these differences are not completely understood but may be related to receptor density, toxin effects on endoplasmic reticulum stress responses and apoptosis (22, 41), or local availability of sensitizing cytokines (5, 7, 11).Most animal models show greater sensitivity to Stx2, including murine, rabbit, and gnotobiotic piglet models, although renal and neurologic micropathologies differ from those in humans and between animal species (6, 9, 45). Earlier studies with the baboon (Papio) model showed that a bolus infusion of purified Stx1 induced intestinal injury, kidney glomerular injury, microangiopathic anemia, thrombocytopenia, and neurologic abnormalities similar to those in humans, suggesting that the baboon represents a promising preclinical animal model (44). A systemic inflammatory response was minimal after Stx1 challenge, but urinary tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) levels were consistent with local kidney inflammatory responses. Baboons were also more sensitive to Stx2 (38), but a direct comparison of the pathophysiologies elicited by the two toxins was difficult because of differing experimental designs. We sought to expand these earlier studies of baboons to identify similarities and differences elicited by Stx1 and Stx2 under reproducible experimental conditions. Given the clinical relevance of Stx2 production during STEC infection in patients, we were particularly interested in responses after Stx2 challenge, for which few data are available from the baboon model. We present the metabolic, physiologic, and inflammatory responses in baboons after intravenous challenge with Stx1 or Stx2. The observed differences in pathophysiology elicited by the two toxins may contribute to a better understanding of the differences in clinical manifestations produced by the toxins.  相似文献   

12.
We examined the susceptibilities to fluconazole of 642 bloodstream infection (BSI) isolates of Candida glabrata and grouped the isolates by patient age and geographic location within the United States. Susceptibility of C. glabrata to fluconazole was lowest in the northeast region (46%) and was highest in the west (76%). The frequencies of isolation and of fluconazole resistance among C. glabrata BSI isolates were higher in the present study (years 2001 to 2007) than in a previous study conducted from 1992 to 2001. Whereas the frequency of C. glabrata increased with patient age, the rate of fluconazole resistance declined. The oldest age group (≥80 years) had the highest proportion of BSI isolates that were C. glabrata (32%) and the lowest rate of fluconazole resistance (5%).Candidemia is without question the most important of the invasive mycoses (6, 33, 35, 61, 65, 68, 78, 86, 88). Treatment of candidemia over the past 20 years has been enhanced considerably by the introduction of fluconazole in 1990 (7, 10, 15, 28, 29, 31, 40, 56-58, 61, 86, 90). Because of its widespread usage, concern about the development of fluconazole resistance among Candida spp. abounds (2, 6, 14, 32, 47, 53, 55, 56, 59, 60, 62, 80, 86). Despite these concerns, fluconazole resistance is relatively uncommon among most species of Candida causing bloodstream infections (BSI) (5, 6, 22, 24, 33, 42, 54, 56, 65, 68, 71, 86). The exception to this statement is Candida glabrata, of which more than 10% of BSI isolates may be highly resistant (MIC ≥ 64 μg/ml) to fluconazole (6, 9, 15, 23, 30, 32, 36, 63-65, 71, 87, 91). Suboptimal fluconazole dosing practices (low dose [<400 mg/day] and poor indications) may lead to an increased frequency of isolation of C. glabrata as an etiological agent of candidemia in hospitalized patients (6, 17, 29, 32, 35, 41, 47, 55, 60, 68, 85) and to increased fluconazole (and other azole) resistance secondary to induction of CDR efflux pumps (2, 11, 13, 16, 43, 47, 50, 55, 69, 77, 83, 84) and may adversely affect the survival of treated patients (7, 10, 29, 40, 59, 90). Among the various Candida species, C. glabrata alone has increased as a cause of BSI in U.S. intensive care units since 1993 (89). Within the United States, the proportion of fungemias due to C. glabrata has been shown to vary from 11% to 37% across the different regions (west, midwest, northeast, and south) of the country (63, 65) and from <10% to >30% within single institutions over the course of several years (9, 48). It has been shown that the prevalence of C. glabrata as a cause of BSI is potentially related to many disparate factors in addition to fluconazole exposure, including geographic characteristics (3, 6, 63-65, 71, 88), patient age (5, 6, 25, 35, 41, 42, 48, 63, 82, 92), and other characteristics of the patient population studied (1, 32, 35, 51). Because C. glabrata is relatively resistant to fluconazole, the frequency with which it causes BSI has important implications for therapy (21, 29, 32, 40, 41, 45, 56, 57, 59, 80, 81, 86, 90).Previously, we examined the susceptibilities to fluconazole of 559 BSI isolates of C. glabrata and grouped the isolates by patient age and geographic location within the United States over the time period from 1992 to 2001 (63). In the present study we build upon this experience and report the fluconazole susceptibilities of 642 BSI isolates of C. glabrata collected from sentinel surveillance sites throughout the United States for the time period from 2001 through 2007 and stratify the results by geographic region and patient age. The activities of voriconazole and the echinocandins against this contemporary collection of C. glabrata isolates are also reported.  相似文献   

13.
Escherichia coli O157:H7 and other Shiga toxin (Stx)-producing E. coli (STEC) bacteria are not enteroinvasive but can cause hemorrhagic colitis. In some STEC-infected individuals, a life-threatening sequela of infection called the hemolytic uremic syndrome may develop that can lead to kidney failure. This syndrome is linked to the production of Stx by the infecting organism. For Stx to reach the kidney, the toxin must first penetrate the colonic epithelial barrier. However, the Stx receptor, globotriaosylceramide (Gb3), has been thought to be absent from human intestinal epithelial cells. Thus, the mechanisms by which the toxin associates with and traverses through the intestine en route to the kidneys have been puzzling aspects of STEC pathogenesis. In this study, we initially determined that both types of Stx made by STEC, Stx1 and Stx2, do in fact bind to colonic epithelia in fresh tissue sections and to a colonic epithelial cell line (HCT-8). We also discovered that globotetraosylceramide (Gb4), a lower-affinity toxin receptor derived from Gb3, is readily detectable on the surfaces of human colonic tissue sections and HCT-8 cells. Furthermore, we found that Gb3 is present on a fraction of HCT-8 cells, where it presumably functions to bind and internalize Stx1 and Stx2. In addition, we established by quantitative real-time PCR (qRT-PCR) that both fresh colonic epithelial sections and HCT-8 cells express Gb3 synthase mRNA. Taken together, our data suggest that Gb3 may be present in small quantities in human colonic epithelia, where it may compete for Stx binding with the more abundantly expressed glycosphingolipid Gb4.Shiga toxins (Stxs) are highly potent ribotoxic virulence factors associated with the worst pathological manifestations of infection by Escherichia coli serotype O157:H7 and other Stx-producing E. coli (STEC) bacteria. Two major types of Stxs are produced by STEC, Stx1 and Stx2, and an organism may produce one or both toxins. Stx1 and Stx2 share enzymatic and structural features but are immunologically distinct. More than 110,000 cases of STEC infection are estimated to occur each year in the United States, and about 75,000 of those cases are caused by E. coli O157:H7 infection. Many individuals infected with E. coli O157:H7 present with severe abdominal pain and bloody diarrhea, of which the latter may be caused by the action of Stxs on endothelial cells that line the small blood vessels (microvasculature) in the gastrointestinal tract (4, 26, 42, 44). In some patients, STEC infection leads to a serious sequela called the hemolytic uremic syndrome (HUS). The HUS is characterized by a triad of clinical features that include thrombocytopenia, hemolytic anemia, and acute kidney failure, and it occurs most frequently in children less than 10 years of age (2, 12). Of note, HUS associated with E. coli O157:H7 infection is a major cause of acute kidney failure in children in the United States and worldwide (6, 61). One hypothesis for how the renal injury occurs in HUS is that blood-borne Stx induces apoptosis in endothelial cells in the glomerular microvasculature (19). Thrombi then form in these damaged blood vessels, a characteristic pathological feature of the HUS. Death of renal tubular cells has also been linked to Stx production in humans and in mouse models of E. coli O157:H7 infection (7, 34, 56).How Stx moves from the lumen of the bowel to the blood vessels that lie below the surface of the gastrointestinal tract to reach the kidneys has not been determined. Presumably, the toxin breaches the epithelial barrier of the colon at or near the site of colonization by the noninvasive E. coli O157:H7. However, the colonic epithelium has been reported to lack globotriaosylceramide (Gb3), the established and preferred receptor for Stx1 and Stx2. The consensus in the literature that the Stx receptor, Gb3, is not present in the human colonic epithelial cells was originally based on conclusions that were drawn from analysis of the general glycolipid composition of the human large intestine (17, 52). In those studies, glycolipids from either mucosal scrapes or the entire mucosal layer were examined by thin-layer chromatography (TLC). Although these mucosal specimens were reported to contain small but measurable levels of Gb3, the samples included not only epithelial cells but also Gb3-enriched endothelial cells. Therefore, evidence of the presence of Gb3 on the cell surface of large bowel epithelial cells was inconclusive. In another investigation, trace amounts of Gb3 were found in epithelial cells isolated by sequential washes of colon tissue with buffer that contained EDTA and reducing agent to gently remove cells layer by layer; however, again, nonepithelial cell contamination could not be ruled out (16). Holgersson et al. ultimately concluded that large bowel epithelial cells do not express “glycolipid-borne Galα1-4 Gal sequence” (components of Gb3) based on the failure to detect Gb3 on the cell surface with Gb3-specific antibody (16). Nevertheless, some of these early studies do suggest that Gb3 may be present in trace amounts in colonic epithelia. These findings, however, appear to have been discounted by many in the field, who assert, based on the negative immunodetection by Holgersson et al. described above (16) and findings of subsequent studies (36, 48), that Gb3 is not present on the colonic epithelium (29).Given the presumed lack of colonic epithelial Gb3, a number of theories have been promulgated to explain how Stx is able to penetrate the epithelial barrier to reach the bloodstream (29). Two related hypotheses are as follows: (i) Stx follows a paracellular course and moves between cells during immune cell infiltration that follows infection (55), or (ii) Stx transits to the bloodstream from the bowel lumen through possible “holes” in the mucosa that result from attachment-and-effacement (A&E) lesions and Stx damage (33). A third hypothesis is that Stx moves by a transcellular route from the lumen to the vasculature in the lamina propria, such as through an alternative but undefined trafficking pathway that does not involve Gb3. In support of the latter conjecture, Stx was found to transcytose polarized tissue culture cells that appeared to lack Gb3 in the absence of cellular damage (1). However, in a human organ culture model, Stx-mediated intestinal epithelial damage was observed (48), a finding that seems to support the second hypothesis above. A fourth premise to explain how Stx crosses the mucosal barrier in the absence of Gb3 on enterocytes is that an alternative receptor exists (11). Such a putative receptor could either mediate Stx-induced cytotoxicity and thus cause a breach in the epithelial barrier or allow transcellular movement of the toxin across the epithelial cell. However, evidence in support of an alternative receptor to Gb3 is not strong. Rather, accumulated data suggest that Gb3 is the sole functional receptor for all Stxs (19, 22, 24, 27, 32, 34, 40, 43, 50, 51, 59), and they can be summarized by 4 lines of evidence. First, cellular cytotoxicity is directly proportional to cell surface Gb3 content (21, 46, 47). Second, Gb3 provided to resistant cells via liposomes induces Stx1 sensitivity in those cells, whereas Gb4 does not (58). Third, Gb3 expression appears to correlate with clinical manifestations of STEC disease. The highest Gb3 content is found in the microvascular glomeruli and proximal tubule cells of the kidney, consistent with renal pathology in HUS (41). Other Gb3-rich regions include the colonic microvascular endothelia, with its associated lamina propria (bloody colitis) (21), and endothelial vasculature of the cerebellum (neurological symptoms) (46). This link between high Gb3 content and Stx toxicity is also seen in animal models. In rabbits, intravenous administration of Stx1 or Stx2 produces vascular lesions in those tissues with high concentrations of Gb3, i.e., the intestine and the central nervous system (47). Furthermore, rabbit kidneys lack Gb3, and no tissue injury is observed there (7). Fourth, Gb3 synthase knockout mice are resistant to Stx effects, although in these mice, other glycosphingolipids, including Gb4, are eliminated as well (43).In this study, we asked whether Stxs could bind to sections of human colonic epithelia and if so by what receptor(s). Based on our initial observation that small amounts of Stx1 and Stx2 were capable of binding to the surface of colonic epithelia, we investigated the possibility that small amounts of Gb3 are present on colonic epithelial cells. Here we present the novel finding that Gb3 synthase mRNA can be detected in epithelial cells isolated by laser capture microdissection (LCM) from normal human colonic tissue sections and that Gb4, a glycosphingolipid derived from Gb3 (Fig. (Fig.1),1), is expressed in this tissue. Furthermore, we demonstrate that while both Stx1 and Stx2 can bind to the epithelial surfaces of those normal human colonic tissue sections, Stx1 binds at relatively higher levels. In addition, we demonstrate that the cytotoxicity of Stx1 and Stx2 for the human colonic cell line HCT-8 can be enhanced 20- to 60-fold by genetically manipulating the cells to increase the cell surface Gb3 content. Collectively, these data suggest that small amounts of Gb3 may mediate Stx uptake in normal colonic tissue, an event that leads to cell death, and that abundant Gb4 may compete with Gb3 for Stx binding.Open in a separate windowFIG. 1.Globotetraosylceramide (Gb4) synthesis pathway (38). The synthesis of Gb4 requires globotriaosylceramide (Gb3) as a substrate. Biosyntheses of Gb3 and Gb4 are accompanied by intracellular transport, presumably by a vesicle-bound exocytotic membrane flow, from the endoplasmic reticulum (ER) through the Golgi cisternae to the plasma membrane (57). Abbreviations: LacCer, lactose-ceramide; GlcCer, glucose-ceramide.  相似文献   

14.
15.
Acanthamoeba culbertsoni is an opportunistic pathogen that causes granulomatous amoebic encephalitis (GAE), a chronic and often fatal disease of the central nervous system (CNS). A hallmark of GAE is the formation of granulomas around the amoebae. These cellular aggregates consist of microglia, macrophages, lymphocytes, and neutrophils, which produce a myriad of proinflammatory soluble factors. In the present study, it is demonstrated that A. culbertsoni secretes serine peptidases that degrade chemokines and cytokines produced by a mouse microglial cell line (BV-2 cells). Furthermore, soluble factors present in cocultures of A. culbertsoni and BV-2 cells, as well as in cocultures of A. culbertsoni and primary neonatal rat cerebral cortex microglia, induced apoptosis of these macrophage-like cells. Collectively, the results indicate that A. culbertsoni can apply a multiplicity of cell contact-independent modes to target macrophage-like cells that exert antiamoeba activities in the CNS.Acanthamoeba culbertsoni belongs to a group of free-living amoebae, such as Balamuthia mandrillaris, Naegleria fowleri, and Sappinia pedata, that can cause disease in humans (46, 56). Acanthamoeba spp. are found worldwide and have been isolated from a variety of environmental sources, including air, soil, dust, tap water, freshwater, seawater, swimming pools, air conditioning units, and contaminated contact lenses (30). Trophozoites feed on bacteria and algae and represent the infective form (47, 56). However, under unfavorable environmental conditions, such as extreme changes in temperature or pH, trophozoites transform into a double-walled, round cyst (22, 45).Acanthamoeba spp. cause an infection of the eye known as amoebic keratitis (AK), an infection of the skin referred to as cutaneous acanthamoebiasis, and a chronic and slowly progressing disease of the central nervous system (CNS) known as granulomatous amoebic encephalitis (GAE) (22, 23, 30, 56). GAE is most prevalent in humans who are immunocompromised (30, 33, 40) and has been reported to occur among individuals infected with the human immunodeficiency virus (HIV) (28). It has been proposed that Acanthamoeba trophozoites access the CNS by passage through the olfactory neuroepithelium (32) or by hematogenous spread from a primary nonneuronal site of infection (23, 24, 33, 53).In immune-competent individuals, GAE is characterized by the formation of granulomas. These cellular aggregates consist of microglia, macrophages, polymorphonuclear cells, T lymphocytes, and B lymphocytes (24, 30). The concerted action of these immune cells results in sequestration of amoebae and is instrumental in slowing the progression of GAE. This outcome is consistent with the observation that granulomas are rarely observed in immunocompromised individuals (34) and in mice with experimentally induced immune suppression following treatment with the cannabinoid delta-9-tetrahydrocannabinol (Δ9-THC) (8).Microglia are a resident population of macrophages in the CNS. These cells, along with CNS-invading peripheral macrophages, appear to play a critical early effector role in the control of Acanthamoeba spread during GAE (4, 5, 29, 31). In vitro, microglia have been shown to produce an array of chemokines and cytokines in response to Acanthamoeba (31, 51). However, these factors appear not to have a deleterious effect on these amoebae (29).Acanthamoeba spp. produce serine peptidases, cysteine peptidases, and metallopeptidases (1, 2, 9, 10, 14, 16, 18, 19, 21, 25, 26, 37, 38, 41, 42, 52). In the present study, it is demonstrated that serine peptidases secreted by A. culbertsoni degrade chemokines and cytokines that are produced by immortalized mouse BV-2 microglia-like cells. In addition, soluble factors present in cocultures of A. culbertsoni and BV-2 cells induced apoptosis of the BV-2 cells. Collectively, these results suggest a mode through which A. culbertsoni can evade immune responsiveness in the CNS.  相似文献   

16.
Moraxella catarrhalis is a human pathogen causing otitis media in infants and respiratory infections in adults, particularly patients with chronic obstructive pulmonary disease. The surface protein Hag (also designated MID) has previously been shown to be a key adherence factor for several epithelial cell lines relevant to pathogenesis by M. catarrhalis, including NCIH292 lung cells, middle ear cells, and A549 type II pneumocytes. In this study, we demonstrate that Hag mediates adherence to air-liquid interface cultures of normal human bronchial epithelium (NHBE) exhibiting mucociliary activity. Immunofluorescent staining and laser scanning confocal microscopy experiments demonstrated that the M. catarrhalis wild-type isolates O35E, O12E, TTA37, V1171, and McGHS1 bind principally to ciliated NHBE cells and that their corresponding hag mutant strains no longer associate with cilia. The hag gene product of M. catarrhalis isolate O35E was expressed in the heterologous genetic background of a nonadherent Haemophilus influenzae strain, and quantitative assays revealed that the adherence of these recombinant bacteria to NHBE cultures was increased 27-fold. These experiments conclusively demonstrate that the hag gene product is responsible for the previously unidentified tropism of M. catarrhalis for ciliated NHBE cells.Moraxella catarrhalis is a gram-negative pathogen of the middle ear and lower respiratory tract (29, 40, 51, 52, 69, 78). The organism is responsible for ∼15% of bacterial otitis media cases in children and up to 10% of infectious exacerbations in patients with chronic obstructive pulmonary disease (COPD). The cost of treating these ailments places a large financial burden on the health care system, adding up to well over $10 billion per annum in the United States alone (29, 40, 52, 95, 97). In recent years, M. catarrhalis has also been increasingly associated with infections such as bronchitis, conjunctivitis, sinusitis, bacteremia, pneumonia, meningitis, pericarditis, and endocarditis (3, 12, 13, 17-19, 24, 25, 27, 51, 67, 70, 72, 92, 99, 102-104). Therefore, the organism is emerging as an important health problem.M. catarrhalis infections are a matter of concern due to high carriage rates in children, the lack of a preventative vaccine, and the rapid emergence of antibiotic resistance in clinical isolates. Virtually all M. catarrhalis strains are resistant to β-lactams (34, 47, 48, 50, 53, 65, 81, 84). The genes specifying this resistance appear to be gram positive in origin (14, 15), suggesting that the organism could acquire genes conferring resistance to other antibiotics via horizontal transfer. Carriage rates as high as 81.6% have been reported for children (39, 104). In one study, Faden and colleagues analyzed the nasopharynx of 120 children over a 2-year period and showed that 77.5% of these patients became colonized by M. catarrhalis (35). These investigators also observed a direct relationship between the development of otitis media and the frequency of colonization. This high carriage rate, coupled with the emergence of antibiotic resistance, suggests that M. catarrhalis infections may become more prevalent and difficult to treat. This emphasizes the need to study pathogenesis by this bacterium in order to identify vaccine candidates and new targets for therapeutic approaches.One key aspect of pathogenesis by most infectious agents is adherence to mucosal surfaces, because it leads to colonization of the host (11, 16, 83, 93). Crucial to this process are surface proteins termed adhesins, which mediate the binding of microorganisms to human cells and are potential targets for vaccine development. M. catarrhalis has been shown to express several adhesins, namely UspA1 (20, 21, 59, 60, 77, 98), UspA2H (59, 75), Hag (also designated MID) (22, 23, 37, 42, 66), OMPCD (4, 41), McaP (61, 100), and a type 4 pilus (63, 64), as well as the filamentous hemagglutinin-like proteins MhaB1, MhaB2, MchA1, and MchA2 (7, 79). Each of these adhesins was characterized by demonstrating a decrease in the adherence of mutant strains to a variety of human-derived epithelial cell lines, including A549 type II pneumocytes and Chang conjunctival, NCIH292 lung mucoepidermoid, HEp2 laryngeal, and 16HBE14o-polarized bronchial cells. Although all of these cell types are relevant to the diseases caused by M. catarrhalis, they lack important aspects of the pathogen-targeted mucosa, such as the features of cilia and mucociliary activity. The ciliated cells of the respiratory tract and other mucosal membranes keep secretions moving out of the body so as to assist in preventing colonization by invading microbial pathogens (10, 26, 71, 91). Given this critical role in host defense, it is interesting to note that a few bacterial pathogens target ciliated cells for adherence, including Actinobacillus pleuropneumoniae (32), Pseudomonas aeruginosa (38, 108), Mycoplasma pneumoniae (58), Mycoplasma hyopneumoniae (44, 45), and Bordetella species (5, 62, 85, 101).In the present study, M. catarrhalis is shown to specifically bind to ciliated cells of a normal human bronchial epithelium (NHBE) culture exhibiting mucociliary activity. This tropism was found to be conserved among isolates, and analysis of mutants revealed a direct role for the adhesin Hag in binding to ciliated airway cells.  相似文献   

17.
Monoclonal antibody (MAb) 11E10 recognizes the Shiga toxin type 2 (Stx2) A1 subunit. The binding of 11E10 to Stx2 neutralizes both the cytotoxic and lethal activities of Stx2, but the MAb does not bind to or neutralize Stx1 despite the 61% identity and 75% similarity in the amino acids of the A1 fragments. In this study, we sought to identify the segment or segments on Stx2 that constitute the 11E10 epitope and to determine how recognition of that region by 11E10 leads to inactivation of the toxin. Toward those objectives, we generated a set of chimeric Stx1/Stx2 molecules and then evaluated the capacity of 11E10 to recognize those hybrid toxins by Western blot analyses and to neutralize them in Vero cell cytotoxicity assays. We also compared the amino acid sequences and crystal structures of Stx1 and Stx2 for stretches of dissimilarity that might predict a binding epitope on Stx2 for 11E10. Through these assessments, we concluded that the 11E10 epitope is comprised of three noncontiguous regions surrounding the Stx2 active site. To determine how 11E10 neutralizes Stx2, we examined the capacity of 11E10/Stx2 complexes to target ribosomes. We found that the binding of 11E10 to Stx2 prevented the toxin from inhibiting protein synthesis in an in vitro assay but also altered the overall cellular distribution of Stx2 in Vero cells. We propose that the binding of MAb 11E10 to Stx2 neutralizes the effects of the toxin by preventing the toxin from reaching and/or inactivating the ribosomes.Escherichia coli O157:H7 and other Shiga toxin (Stx)-producing E. coli (STEC) strains cause approximately 110,000 cases of infection and over 90 deaths each year in the United States according to the Centers for Disease Control and Prevention (16). Infections with STEC can lead to diarrhea, hemorrhagic colitis, and hemolytic uremic syndrome (HUS). HUS occurs in about 6 to 15% of individuals after infection with E. coli O157:H7 (15)—but less frequently with other STEC strains (5)—and is characterized by hemolytic anemia, thrombotic thrombocytopenia, and renal failure. The development of this sequela is linked to the expression of Stxs by the bacteria (18).The Stx family comprises two serogroups, Stx/Stx1 and Stx2, and polyclonal antisera raised against either Stx1 or Stx2 do not cross-neutralize the other toxin (29, 30). Stx is produced by Shigella dysenteriae type 1 and differs by only 1 amino acid from the Stx1 made by the prototypic STEC O157:H7 strain, EDL933. A single isolate of STEC can express Stx1 (or one of its variants), Stx2 (or one of its variants), or both toxins. Variants of each toxin type are defined by either a biological or immunological difference from the prototypical toxin (31). Stx1 variants include Stx1c and Stx1d, while the variants of Stx2 are Stx2c, Stx2d, Stx2d-activatable (Stx2dact), Stx2e, and Stx2f (reviewed in reference 18).Stxs are complex holotoxins with a stoichiometry of five identical binding (B) subunits and a single active (A) domain. These AB5 molecules are potent cytotoxins with an N-glycosidase activity that stops protein synthesis by inactivation of the 60S ribosome (6); this activity eventually leads to eukaryotic cell death. The ∼32-kDa A subunit contains the enzymatic activity of the toxin with the active site glutamic acid residue at position 167. The A subunit is asymmetrically cleaved by trypsin or furin into an enzymatically active ∼28-kDa A1 fragment and an ∼4-kDa A2 peptide. The A2 peptide remains linked to the large enzymatic domain through a disulfide bond and is encircled by the five identical B subunits of ∼7.7 kDa. The B subunits of the Stxs typically bind to the eukaryotic glycolipid receptor globotriaosylceramide (Gb3), also known as CD77. The mature A and B subunits of Stx1 and Stx2 are approximately 68 and 73% similar at the amino acid level. The crystal structures of Stx and Stx2 have been resolved, and the two structures are remarkably similar (7, 8). Nevertheless, there are some features of these three-dimensional models that differ (summarized in reference 8).Currently, there are no Food and Drug Administration-approved therapies in the United States to treat STEC infections. However, our research group is one of several that investigate passive immunization strategies to neutralize the Stxs associated with STEC infections (3, 4, 10, 13, 19, 20). Our passive immunization strategy is based on murine monoclonal antibodies (MAbs) developed in this laboratory that specifically bind to and neutralize Stx/Stx1 or Stx2 (21, 28). The MAb 11E10 was generated by immunization of BALB/c mice with Stx2 turned into a toxoid (“toxoided”) by treatment with formaldehyde (21). MAb 11E10 specifically recognizes the A1 fragment of Stx2 and neutralizes Stx2 for Vero cells and mice but does not bind to or neutralize Stx/Stx1 (21). The murine MAb 11E10 was modified to contain a human constant region to reduce the potential for an antibody recipient to generate an antimouse antibody response (4). This human-mouse chimeric antibody, called cαStx2, successfully underwent phase I clinical testing (3). In this report, we define the epitope on the A subunit of Stx2 recognized by the murine MAb 11E10 (and, therefore, also by cαStx2) and present evidence that the MAb blocks the enzymatic action of the toxin in vitro and also alters toxin trafficking in Vero cells.  相似文献   

18.
Pseudallescheria boydii has long been known to cause white grain mycetoma in immunocompetent humans, but it has recently emerged as an opportunistic pathogen of humans, causing potentially fatal invasive infections in immunocompromised individuals and evacuees of natural disasters, such as tsunamis and hurricanes. The diagnosis of P. boydii is problematic since it exhibits morphological characteristics similar to those of other hyaline fungi that cause infectious diseases, such as Aspergillus fumigatus and Scedosporium prolificans. This paper describes the development of immunoglobulin M (IgM) and IgG1 κ-light chain monoclonal antibodies (MAbs) specific to P. boydii and certain closely related fungi. The MAbs bind to an immunodominant carbohydrate epitope on an extracellular 120-kDa antigen present in the spore and hyphal cell walls of P. boydii and Scedosporium apiospermum. The MAbs do not react with S. prolificans, Scedosporium dehoogii, or a large number of clinically relevant fungi, including A. fumigatus, Candida albicans, Cryptococcus neoformans, Fusarium solani, and Rhizopus oryzae. The MAbs were used in immunofluorescence and double-antibody sandwich enzyme-linked immunosorbent assays (DAS-ELISAs) to accurately differentiate P. boydii from other infectious fungi and to track the pathogen in environmental samples. Specificity of the DAS-ELISA was confirmed by sequencing of the internally transcribed spacer 1 (ITS1)-5.8S-ITS2 rRNA-encoding regions of environmental isolates.Pseudallescheria boydii is an infectious fungal pathogen of humans (7, 16, 40, 58, 59). It is the etiologic agent of white grain mycetoma in immunocompetent humans (7) and has emerged over recent years as the cause of fatal disseminated infections in individuals with neutropenia, AIDS, diabetes, renal failure, bone marrow or solid organ transplants, systemic lupus erythematous, and Crohn''s disease; in those undergoing corticosteroid treatment; and in leukemia and lymphoma patients (1, 2, 3, 18, 27, 31, 32, 34, 36, 37, 38, 47, 49, 52). The fungus is the most prevalent species after Aspergillus fumigatus in the lungs of cystic fibrosis patients (8), where it causes allergic bronchopulmonary disease (5) and chronic lung lesions simulating aspergillosis (24). Near-drowning incidents and recent natural disasters, such as the Indonesian tsunami in 2004, have shown P. boydii and the related species Scedosporium apiospermum and Scedosporium aurantiacum to be the causes of fatal central nervous system infections and pneumonia in immunocompetent victims who have aspirated polluted water (4, 11, 12, 21, 22, 25, 30, 33, 57). Its significance as a potential pathogen of disaster evacuees has led to its recent inclusion in the Centers for Disease Control and Prevention list of infectious etiologies in persons with altered mental statuses, central nervous system syndromes, or respiratory illness.P. boydii is thought to be an underdiagnosed fungus (60), and misidentification is one of the reasons that the mortality rate due to invasive pseudallescheriasis is high. Detection of invasive P. boydii infections, based on cytopathology and histopathology, is problematic since it can occur in tissue and bronchoalveolar and bronchial washing specimens with other hyaline septated fungi, such as Aspergillus and Fusarium spp. (7, 23, 53, 60), which exhibit similar morphological characteristics upon microscopic examination (2, 23, 24, 28, 37, 44, 53, 60). Early diagnosis of infection by P. boydii and differentiation from other agents of hyalohyphomycosis is imperative, since it is refractory to antifungal compounds, such as amphotericin B, that are commonly administered for the control of fungal infections (10, 39, 58).The immunological diagnosis of Pseudallescheria infections has focused on the detection of antigens by counterimmunoelectrophoresis, and by immunohistological techniques using polyclonal fluorescent antibodies, but cross-reactions with antigens from other fungi, such as Aspergillus species, occurs (7, 19, 23). Pinto and coworkers (41, 42) isolated a peptidorhamnomannan from hyphae of P. boydii and proposed the antigen as a diagnostic marker for the pathogen. Cross-reactivity with Sporothrix schenckii and with Aspergillus have, however, been noted (23, 41). Furthermore, it is uncertain whether a similar antigen is present in the related pathogenic species S. prolificans, an important consideration in patient groups susceptible to mixed Scedosporium infections (6, 18).Hybridoma technology allows the production of highly specific MAbs that are able to differentiate between closely related species of fungi (54, 55, 56). The purpose of this paper is to report the development of MAbs specific to P. boydii and certain closely related species and their use to accurately discriminate among P. boydii, A. fumigatus, and other human pathogenic fungi by using immunofluorescence and double-antibody sandwich enzyme-linked immunosorbent assays (DAS-ELISAs).Currently, the natural environmental habitat of P. boydii is unknown, but nutrient-rich, brackish waters, such as estuaries, have been suggested (9, 17). In combination with a semiselective isolation procedure, I show how the DAS-ELISA can be used to rapidly and accurately track the pathogen in naturally infested estuarine muds, and in doing so illustrate the potential of the DAS-ELISA as a diagnostic platform for detection of P. boydii and related species within the Pseudallescheria complex.  相似文献   

19.
Campylobacter jejuni is a leading cause of food-borne illness in the United States. Despite significant recent advances, its mechanisms of pathogenesis are poorly understood. A unique feature of this pathogen is that, with some exceptions, it lacks homologs of known virulence factors from other pathogens. Through a genetic screen, we have identified a C. jejuni homolog of the VirK family of virulence factors, which is essential for antimicrobial peptide resistance and mouse virulence.Campylobacter jejuni is a leading cause of infectious diarrhea in industrialized and developing countries (2, 67). Although most often self-limiting, C. jejuni infections can also lead to severe disease and harmful sequelae, such as Guillain-Barré syndrome (4, 55). Despite the significant progress made during the past few years, the mechanisms of C. jejuni pathogenesis remain poorly understood. A number of potential virulence factors have been identified, and in some cases, their role in virulence and/or colonization has been demonstrated in animal models of infection. For example, motility has been shown to be crucial in order for C. jejuni to colonize or cause disease in several animal models of infection (1, 15, 30, 54). A variety of surface structures, such as adhesins (34, 40, 64) and polysaccharides (5, 6), and glycosylation systems (38, 74), which presumably modify some of these surface structures, have also been shown to be important for infection. Additional studies have revealed the importance of specific metabolic pathways in C. jejuni growth both in vitro and within animals (16, 25, 31, 60, 76). The ability of C. jejuni to invade and survive within nonphagocytic cells has also been proposed to be an important virulence determinant (21, 41, 57, 58, 68, 75, 80).The available genome sequences of several C. jejuni strains have provided significant insight into C. jejuni physiology and metabolism (22, 32, 62, 63, 65). Remarkably, however, analysis of these C. jejuni genome sequences has revealed very few homologs of common virulence factors from other pathogens. A notable exception is the toxin CDT (cytolethal distending toxin), which is also encoded by several other important bacterial pathogens (36, 44, 45). In this paper we describe the identification of a transposon insertion mutant in C. jejuni 81-176, which results in increased susceptibility to antimicrobial peptides and a significant defect in the ability of the organism to cause disease in an animal model of infection. The insertion mutant was mapped to the CJJ81176_1087 open reading frame (Cj1069 in the C. jejuni NCT 11168 reference strain), which encodes a protein with very significant amino acid sequence similarity to the VirK (DUF535) family of virulence factors (13, 20, 56).  相似文献   

20.
Intracellular Salmonella enterica serovar Typhimurium (serovar Typhimurium) occupies a Salmonella-containing vacuole (SCV) where bacterial effector proteins are secreted into the host cell using type III secretion systems (T3SS). Cytoskeletal motor proteins and T3SS-delivered effector proteins facilitate SCV positioning to juxtanuclear positions where bacterial replication occurs. Here, we show that this characteristic SCV positioning is not maintained by all SCVs during infection of HeLa cells. Notably, juxtanuclear SCV localization that occurs by 8 to 14 h postinfection is followed by significant centrifugal displacement of a subset of SCVs toward the host cell periphery by 24 h postinfection. This novel phenotype requires bacterial protein synthesis, a functional Salmonella pathogenicity island 2 (SPI-2)-encoded T3SS, intact microtubules, and kinesin-1 motor protein. Bacteria lacking PipB2, a kinesin-recruiting T3SS effector, did not exhibit centrifugal displacement and remained at juxtanuclear positions throughout 24 h of infection. While levels of the SPI-2 effectors PipB2 and SifA increased during 24 h postinfection, a corresponding decrease in levels of the SPI-1 T3SS effectors SipA and SopB, both known to mediate juxtanuclear SCV positioning, was observed. A fluorescence-based assay indicated that wild-type serovar Typhimurium transferred from infected to uninfected epithelial cells while strains deficient in SPI-2 T3SS secretion or PipB2 did not. Our results reveal a novel SCV phenotype implicated in the cell-to-cell spread of serovar Typhimurium during infection.Salmonella enterica serovar Typhimurium (serovar Typhimurium) is a cause of gastroenteritis in humans and typhoid-like disease in certain strains of mice (55). Serovar Typhimurium is a facultative intracellular pathogen that can actively invade nonphagocytic cells through the delivery of bacterial proteins, termed effectors, into the host cell cytosol using the type III secretion system (T3SS) encoded by Salmonella pathogenicity island 1 (SPI-1) (19). Following entry, serovar Typhimurium typically resides in a membrane-bound compartment termed the Salmonella-containing vacuole (SCV) (16, 50, 54). From here, additional effector proteins required for intracellular replication and virulence are delivered into the host cytosol using a second T3SS encoded by another pathogenicity island, SPI-2 (10, 28, 40, 48, 58). These effectors modulate various host cell activities, including endosomal trafficking, actin assembly dynamics, and microtubule-based transport (5, 8, 24, 33, 50, 51, 58).One characteristic trait of SCVs is their localization to a juxtanuclear, Golgi apparatus-associated region of the host cell several hours postinfection (1, 5, 41, 45). In general, with some variation likely due to differing experimental methods, this distinct localization is observed as early as 4 h postinfection (hpi) and is maintained at 8 to 16 hpi (2, 5, 13, 24, 26, 41, 42, 45). A recent study has shown that serovar Typhimurium can redirect host secretory traffic, resulting in an accumulation of post-Golgi vesicles around the SCV (36). It has been proposed that serovar Typhimurium targets the Golgi apparatus to acquire nutrients and/or membrane materials for maintenance of a replicative niche within the SCV (23, 45).Several different effectors secreted by the SPI-2 T3SS appear to mediate this centralized positioning of SCVs, including SseF, SseG, and SifA (2, 5, 13, 41, 42, 45). Deletion of sseF or sseG results in SCVs that are displaced from their usual juxtanuclear, Golgi compartment-associated position (1, 13, 45). SseF and SseG have been shown to interact with each other (13) and appear to promote the recruitment of the minus-end-directed microtubule motor dynein to SCVs to permit their juxtanuclear localization (2). SseG has also been proposed to act by tethering SCVs to the Golgi region (42).Deletion of sifA also results in SCVs that are displaced from the nucleus and located toward the host cell periphery (5). SifA was shown to interact with a host SifA- and kinesin-interacting protein that negatively regulates the recruitment of plus-end-directed kinesin-1 motors to the SCV, thus favoring the inward migration and maintenance of the SCV around the nucleus (5). In apparent opposition to SifA, the SPI-2 effector PipB2 has been shown to recruit kinesin-1 to the SCV (26). However, the characteristic positioning of SCVs to juxtanuclear regions suggests that the kinesin-inhibitory action of SifA may be dominant over the effects of PipB2 (26), at least at 8 to 14 hpi.Interestingly, some effectors secreted by the SPI-1 T3SS that is traditionally associated with Salmonella invasion appear to persist in host cells (6, 14) and are also implicated in modulating intracellular SCV positioning (6, 57). We recently demonstrated a role for the SPI-1 T3SS effector SopB in maintaining the juxtanuclear positioning of SCVs through the action of nonmuscle myosin II actin motors (57). Another SPI-1 effector, SipA, has also been shown to persist in host cells after bacterial entry and appears to act with SifA to ensure perinuclear positioning of SCVs (6). Hence, it appears that stringent control of microtubule and actin motor activity on the SCV by both SPI-1 and SPI-2 T3SS effectors is an important facet of SCV intracellular positioning (26).Overall, much remains to be resolved regarding the mediators and implications of intracellular SCV positioning. By remaining at the juxtanuclear region, the bacteria likely modify their compartments into a replicative niche where nutrient acquisition and SCV maintenance can occur (23, 36, 45). As a result of replication, high numbers of intracellular bacteria would presumably lead to host cell lysis, resulting in bacterial release; however, little is known about any mechanism(s) of Salmonella escape from host cells.The present study was conducted to examine the intracellular positioning of SCVs over the course of a 24-h infection. We show that at later stages of epithelial cell infection, the positioning of a significant proportion of SCVs is not maintained at a juxtanuclear region but is situated closer to the host cell periphery. This outward displacement of SCVs was dependent upon the SPI-2 T3SS, host microtubules and kinesin, and the SPI-2 effector PipB2. Furthermore, the dynamic positioning of SCVs is associated with a decrease in protein levels of SPI-1 effectors previously shown to mediate juxtanuclear positioning. Results from a cell-to-cell infection assay indicate that serovar Typhimurium strains that did not exhibit peripheral displacement at later stages of infection were also impaired in their ability to infect newly introduced host cells. Our results provide new insight into the nature of SCV positioning and demonstrate that intracellular SCV positioning is a dynamic process, with implications for bacterial cell-to-cell transfer.  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