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 15 毫秒
1.
By producing toxins, Clostridium perfringens causes devastating diseases of both humans and animals. C. perfringens beta toxin (CPB) is the major virulence determinant for type C infections and is also implicated in type B infections, but little is known about the CPB structure-function relationship. Amino acid sequence comparisons of the CPBs made by 8 randomly selected isolates identified two natural variant toxins with four conserved amino acid changes, including a switch of E to K at position 168 (E168K) that introduces a potential trypsin cleavage site into the CPB protein of strain JGS1076. To investigate whether this potential trypsin cleavage site affects sensitivity to trypsin, a primary host defense against this toxin, the two CPB variants were assayed for their trypsin sensitivity. The results demonstrated a significant difference in trypsin sensitivity, which was linked to the E168K switch by using site-directed recombinant CPB (rCPB) mutants. The natural CPB variants also displayed significant differences in their cytotoxicity to human endothelial cells. This cytotoxicity difference was mainly attributable to increased host cell binding rather than the ability to oligomerize or form functional pores. Using rCPB site-directed mutants, differences in cytotoxicity and host cell binding were linked to an A300V amino acid substitution in the strain JGS1076 CPB variant that possessed more cytotoxic activity. Mapping of sequence variations on a CPB structure modeled using related toxins suggests that the E168K substitution is surface localized and so can interact with trypsin and that the A300V substitution is located in a putative binding domain of the CPB toxin.  相似文献   

2.
Growth and toxin production of stable L-forms of Clostridium perfringens grown in a mini-fermentor were monitored. A gradual but steady increment in viable count occurred over a 7-h period, followed by death. The peak of viability preceded the optical density peak by 3 h. Theta, alpha, kappa, and lambda toxins were measured, with theta toxin appearing first in the culture supernate. Growth of the parent bacillus form of C. perfringens was compared under similar conditions. Toxin levels achieved by the bacillus culture exceeded those of the L-form culture four- to eightfold; however, based upon viable count, the L-form organism produced 8 to 16 times as much toxin as did the bacillus. The amounts of extracellular toxin produced by both forms were similar when related to cell protein rather than cell number. Guinea pig inoculation showed that the L-form of C. perfringens did not produce gas gangrene, although it was not entirely without effect. Both guinea pig and human sera were inhibitory to these L-forms, a fact attributable to a heat-liable component in the sera, most likely complement.  相似文献   

3.
Clostridium perfringens TpeL belongs to a family of large clostridial cytotoxins that encompasses Clostridium difficile toxin A (TcdA) and B (TcdB) and Clostridium sordellii lethal toxin (TcsL). We report here the identification of the TpeL-catalyzed modification of small GTPases. A recombinant protein (TpeL1-525) derived from the TpeL N-terminal catalytic domain in the presence of streptolysin O (SLO) induced the rounding of Vero cells and the glycosylation of cellular Rac1. Among several hexoses tested, UDP-N-acetyl-glucosamine (UDP-GlcNAc) and UDP-glucose (UDP-Glc) served as cosubstrates for TpeL1-525-catalyzed modifications. TpeL1-525 catalyzed the incorporation of UDP-Glc into Ha-Ras, Rap1B, and RalA and of UDP-GlcNAc into Rac1, Ha-Ras, Rap1B, and RalA. In Rac1, TpeL and TcdB share the same acceptor amino acid for glycosylation, Thr-35. In Vero cells treated with TpeL1-525 in the presence of SLO, glycosylation leads to a translocation of the majority of Rac1 and Ha-Ras to the membrane. We demonstrate for first time that TpeL uses both UDP-GlcNAc and UDP-Glc as donor cosubstrates and modifies the Rac1 and Ras subfamily by glycosylation to mediate its cytotoxic effects.  相似文献   

4.
Clostridium perfringens type C isolates cause enteritis necroticans in humans or necrotizing enteritis and enterotoxemia in domestic animals. Type C isolates always produce alpha toxin and beta toxin but often produce additional toxins, e.g., beta2 toxin or enterotoxin. Since plasmid carriage of toxin-encoding genes has not been systematically investigated for type C isolates, the current study used Southern blot hybridization of pulsed-field gels to test whether several toxin genes are plasmid borne among a collection of type C isolates. Those analyses revealed that the surveyed type C isolates carry their beta toxin-encoding gene (cpb) on plasmids ranging in size from ∼65 to ∼110 kb. When present in these type C isolates, the beta2 toxin gene localized to plasmids distinct from the cpb plasmid. However, some enterotoxin-positive type C isolates appeared to carry their enterotoxin-encoding cpe gene on a cpb plasmid. The tpeL gene encoding the large clostridial cytotoxin was localized to the cpb plasmids of some cpe-negative type C isolates. The cpb plasmids in most surveyed isolates were found to carry both IS1151 sequences and the tcp genes, which can mediate conjugative C. perfringens plasmid transfer. A dcm gene, which is often present near C. perfringens plasmid-borne toxin genes, was identified upstream of the cpb gene in many type C isolates. Overlapping PCR analyses suggested that the toxin-encoding plasmids of the surveyed type C isolates differ from the cpe plasmids of type A isolates. These findings provide new insight into plasmids of proven or potential importance for type C virulence.Clostridium perfringens isolates are classified into five toxinotypes (A to E) based upon the production of four (α, β, ɛ, and ι) typing toxins (29). Each toxinotype is associated with different diseases affecting humans or animals (25). In livestock species, C. perfringens type C isolates cause fatal necrotizing enteritis and enterotoxemia, where toxins produced in the intestines absorb into the circulation to damage internal organs. Type C-mediated animal diseases result in serious economic losses for agriculture (25). In humans, type C isolates cause enteritis necroticans, which is also known as pigbel or Darmbrand (15, 17), an often fatal disease that involves vomiting, diarrhea, severe abdominal pain, intestinal necrosis, and bloody stools. Acute cases of pigbel, resulting in rapid death, may also involve enterotoxemia (15).By definition, type C isolates must produce alpha and beta toxins (24, 29). Alpha toxin, a 43-kDa protein encoded by the chromosomal plc gene, has phospholipase C, sphingomyelinase, and lethal properties (36). Beta toxin, a 35-kDa polypeptide, forms pores that lyse susceptible cells (28, 35). Recent studies demonstrated that beta toxin is necessary for both the necrotizing enteritis and lethal enterotoxemia caused by type C isolates (33, 37). Besides alpha and beta toxins, type C isolates also commonly express beta2 toxin, perfringolysin O, or enterotoxin (11).There is growing appreciation that naturally occurring plasmids contribute to both C. perfringens virulence and antibiotic resistance. For example, all typing toxins, except alpha toxin, can be encoded by genes carried on large plasmids (9, 19, 26, 30-32). Other C. perfringens toxins, such as the enterotoxin or beta2 toxin, can also be plasmid encoded (6, 8, 12, 34). Furthermore, conjugative transfer of several C. perfringens antibiotic resistance plasmids or toxin plasmids has been demonstrated, supporting a key role for plasmids in the dissemination of virulence or antibiotic resistance traits in this bacterium (2).Despite their pathogenic importance, the toxin-encoding plasmids of C. perfringens only recently came under intensive study (19, 26, 27, 31, 32). The first carefully analyzed C. perfringens toxin plasmids were two plasmid families carrying the enterotoxin gene (cpe) in type A isolates (6, 8, 12, 26). One of those cpe plasmid families, represented by the ∼75-kb prototype pCPF5603, has an IS1151 sequence present downstream of the cpe gene and also carries the cpb2 gene, encoding beta2 toxin. A second cpe plasmid family of type A isolates, represented by the ∼70-kb prototype pCPF4969, lacks the cpb2 gene and carries an IS1470-like sequence, rather than an IS1151 sequence, downstream of the cpe gene. The pCPF5603 and pCPF4969 plasmid families share an ∼35-kb region that includes transfer of a clostridial plasmid (tcp) locus (26). The presence of this tcp locus likely explains the demonstrated conjugative transfer of some cpe plasmids (5) since a similar tcp locus was shown to mediate conjugative transfer of the C. perfringens tetracycline resistance plasmid pCW3 (2).The iota toxin-encoding plasmids of type E isolates are typically larger (up to ∼135 kb) than cpe plasmids of type A isolates (19). Plasmids carrying iota toxin genes often encode other potential virulence factors, such as lambda toxin and urease, as well as a tcp locus (19). Many iota toxin plasmids of type E isolates share, sometimes extensively, sequences with cpe plasmids of type A isolates (19). It has been suggested that many iota toxin plasmids evolved from the insertion of a mobile genetic element carrying the iota toxin genes near the plasmid-borne cpe gene in a type A isolate, an effect that silenced the cpe gene in many type E isolates (3, 19).Plasmids carrying the epsilon toxin gene (etx) vary from ∼48 kb to ∼110 kb among type D isolates (32). In part, these etx plasmid size variations in type D isolates reflect differences in toxin gene carriage. For example, the small ∼48-kb etx plasmids present in some type D isolates lack both the cpe gene and the cpb2 gene. In contrast, larger etx plasmids present in other type D isolates often carry the cpe gene, the cpb2 gene, or both the cpe and cpb2 genes. Thus, the virulence plasmid diversity of type D isolates spans from carriage of a single toxin plasmid, possessing from one to three distinct toxin genes, to carriage of three different toxin plasmids.In contrast to the variety of etx plasmids found among type D isolates, type B isolates often or always share the same ∼65-kb etx plasmid, which is related to pCPF5603 but lacks the cpe gene (27). This common etx plasmid of type B isolates, which carries a cpb2 gene and the tcp locus, is also present in a few type D isolates. Most type B isolates surveyed to date carry their cpb gene, encoding beta toxin, on an ∼90-kb plasmid, although a few of those type B isolates possess an ∼65-kb cpb plasmid distinct from their ∼65-kb etx plasmid (31).To our knowledge, the cpb gene has been mapped to a plasmid (uncharacterized) in only a single type C strain (16). Furthermore, except for the recent localization of the cpe gene to plasmids in type C strains (20), plasmid carriage of other potential toxin genes in type C isolates has not been investigated. Considering the limited information available regarding the toxin plasmids of type C isolates, our study sought to systematically characterize the size, diversity, and toxin gene carriage of toxin plasmids in a collection of type C isolates. Also, to gain insight into possible mobilization of the cpb gene by insertion sequences or conjugative transfer, the presence of IS1151 sequences or the tcp locus on type C toxin plasmids was investigated.  相似文献   

5.
Clostridium perfringens was found to produce alpha toxin in a synthetic medium containing zinc; in medium containing no zinc, a little toxin was detected in the early logarithmic phase of growth and it disappeared rapidly. No intracellular accumulation of alpha toxin protein occurred whether or not zinc was present in the medium. In zinc-deficient medium, the organisms produced and released into the surrounding medium the protein specifically precipitable with alpha antitoxin in an amount comparable to that of alpha toxin produced in the zinc-containing medium. The protein combined rapidly in some unknown way with zinc to form the active and stable alpha toxin.  相似文献   

6.
7.
目的探讨艰难梭菌毒素A/B及其相关毒素基因检测对艰难梭菌相关性腹泻的诊断价值。方法收集2015年1月至2015年10月我院ICU病房腹泻疑似艰难梭菌感染患者的粪便标本133例作为研究对象,分别采用艰难梭菌培养法、CDAB法、PCR毒素基因检测法及艰难梭菌毒素GDH检测法进行检测,以艰难梭菌培养法结果为金标准,计算各方法的诊断指标所包含有的特异度、敏感度、阴性预测值和阳性预测值等。结果本研究收集的133例粪便标本中,经过金标准粪便样本厌氧培养法检出阳性结果 20例,阴性结果 113例。CDAB法具有低敏感度(0.550)和高特异度(0.912),诊断符合率为0.932,BD-PCR毒素基因检测法具有高敏感度(0.950)和高特异度(0.929),诊断符合率为0.932,艰难梭菌毒素GDH检测法的高敏感度(0.900)和低特异度(0.779),诊断符合率为0.797。结论对于疑似艰难梭菌感染,可联合艰难梭菌GDH、艰难梭菌毒素A/B(CDAB)或进行荧光定量PCR毒素基因共同检测,有效降低检测时间,为临床医师及时提供准确的诊断依据,并制定行之有效的治疗措施。  相似文献   

8.
After binding, Clostridium perfringens enterotoxin (CPE) initially localizes in a small (~90-kDa) complex in plasma membranes. This event is followed by formation of a second membrane complex, referred to as large (160-kDa) complex. Contrary to a previous hypothesis proposing that CPE inserts into intestinal brush border membranes (BBMs) when this toxin is localized in the small complex, this study shows that BBMs do not offer CPE localized in the small complex protection from pronase. However, our experiments indicate that BBMs do substantially protect CPE from pronase when this toxin is localized in large complex. Since the onset of CPE-induced permeability alterations closely coincides with large-complex formation, these new results suggest that CPE-induced alterations in permeability may result from pore formation due to the partial membrane insertion of CPE when this toxin is present in large complex.  相似文献   

9.
The ability of Clostridium perfringens type C to cause human enteritis necroticans (EN) is attributed to beta toxin (CPB). However, many EN strains also express C. perfringens enterotoxin (CPE), suggesting that CPE could be another contributor to EN. Supporting this possibility, lysate supernatants from modified Duncan-Strong sporulation (MDS) medium cultures of three CPE-positive type C EN strains caused enteropathogenic effects in rabbit small intestinal loops, which is significant since CPE is produced only during sporulation and since C. perfringens can sporulate in the intestines. Consequently, CPE and CPB contributions to the enteropathogenic effects of MDS lysate supernatants of CPE-positive type C EN strain CN3758 were evaluated using isogenic cpb and cpe null mutants. While supernatants of wild-type CN3758 MDS lysates induced significant hemorrhagic lesions and luminal fluid accumulation, MDS lysate supernatants of the cpb and cpe mutants caused neither significant damage nor fluid accumulation. This attenuation was attributable to inactivating these toxin genes since complementing the cpe mutant or reversing the cpb mutation restored the enteropathogenic effects of MDS lysate supernatants. Confirming that both CPB and CPE are needed for the enteropathogenic effects of CN3758 MDS lysate supernatants, purified CPB and CPE at the same concentrations found in CN3758 MDS lysates also acted together synergistically in rabbit small intestinal loops; however, only higher doses of either purified toxin independently caused enteropathogenic effects. These findings provide the first evidence for potential synergistic toxin interactions during C. perfringens intestinal infections and support a possible role for CPE, as well as CPB, in some EN cases.  相似文献   

10.
11.
Toxins A and B are the main virulence factors of Clostridium difficile and are the targets for molecular diagnostic tests. Here, we describe a new toxin A-negative, toxin B-positive, binary toxin CDT (Clostridium difficile transferase)-negative (A B+ CDT) toxinotype(XXXII) characterized by a variant type of pathogenicity locus (PaLoc) without tcdA and with atypical organization of the PaLoc integration site.  相似文献   

12.
Mono-ADP ribosylation of actin by bacterial toxins, such as Clostridium perfringens iota or Clostridium botulinum C2 toxins, results in rapid depolymerization of actin filaments and cell rounding. Here we report that treatment of African green monkey kidney (Vero) cells with iota toxin resulted in delayed caspase-dependent death. Unmodified actin did not reappear in toxin-treated cells, and enzyme-active toxin was detectable in the cytosol for at least 24 h. C2 toxin showed comparable, long-lived effects in cells, while a C2 toxin control lacking ADP-ribosyltransferase activity did not induce cell death. To address whether the remarkable stability of the iota and C2 toxins in cytosol was crucial for inducing cell death, we treated cells with C/SpvB, the catalytic domain of Salmonella enterica SpvB. Although C/SpvB also mono-ADP ribosylates actin as do the iota and C2 toxins, cells treated with a cell-permeating C/SpvB fusion toxin became rounded but recovered and remained viable. Moreover, unmodified actin reappeared in these cells, and ADP-ribosyltransferase activity due to C/SpvB was not detectable in the cytosol after 24 h, a result most likely due to degradation of C/SpvB. Repeated application of C/SpvB prevented recovery of cells and reappearance of unmodified actin. In conclusion, a complete but transient ADP ribosylation of actin was not sufficient to trigger apoptosis, implying that long-term stability of actin-ADP-ribosylating toxins, such as iota and C2, in the cytosol is crucial for inducing delayed, caspase-dependent cell death.Various bacterial toxins destroy the actin cytoskeleton of eukaryotic cells by mono-ADP ribosylation of G-actin at arginine-177 (1, 3, 23). ADP-ribosylated actin caps the barbed, fast-growing ends of actin filaments (F-actin), thus preventing further assembly of unmodified G-actin into filaments (32). Although ADP-ribosylated G-actin does not affect the pointed, slow-growing ends of F-actin, the critical concentration for actin polymerization does increase and leads to complete depolymerization of actin filaments (33). Therefore, treatment of cells with these toxins disrupts the actin cytoskeleton and causes rounding of adherent cells within hours.Binary ADP-ribosylating toxins that target actin can be divided into family members that include Clostridium botulinum C2 toxin (20), Clostridium perfringens iota toxin (29), CDT from Clostridium difficile (26), Clostridium spiroforme toxin, also known as CST (25), and the vegetative insecticidal proteins from Bacillus cereus (9). The clostridial and bacillus binary toxins are typical exotoxins, produced by extracellular bacteria, that ultimately enter the cytosol of targeted cells without the toxin-producing bacteria. In contrast, SpvB from Salmonella enterica (21, 31) also targets actin but is delivered into the host cell''s cytosol by intracellularly located bacteria.All binary actin-ADP-ribosylating toxins are composed of two nonlinked proteins, a binding/translocation component and a separate enzyme component (3). In recent years, the structures, modes of action, and cellular uptake mechanisms of the C2 and iota toxins have been discovered to various degrees. The binding/translocation components of the C2 (C2IIa) and iota (Ib) toxins, respectively, mediate cell surface docking of the enzyme components C2I and Ia, followed by cellular uptake and translocation of an enzyme component(s) from acidified endosomes into the cytosol (3). Although the C2 and iota toxins share comparable structures and modes of action, there are striking differences regarding modification of actin isoforms and individual steps during toxin internalization (3). Iota toxin, CDT, and CST are very closely related, and their components are interchangeable, unlike the C2 toxin (8, 22, 24, 25). Thus, CDT and CST are referred to as the iota-like toxins, with C. perfringens iota toxin representing the prototype. Iota toxin is an enterotoxin that naturally causes diarrhea in calves and lambs, is lethal for mice, and is dermonecrotic for guinea pigs (28, 29, 30). The iota-like toxins of C. difficile and C. spiroforme are also associated with gastrointestinal diseases of humans and animals (for a review, see reference 3).Although the immediate cytopathic effects induced by iota toxin have been investigated in detail, the long-term effects on mammalian cells following intoxication and in particular the fate of internalized Ia ADP-ribosyltransferase are unknown. In this study, we have shown that iota toxin-mediated ADP ribosylation of actin and subsequent cell rounding are irreversible, resulting in delayed caspase-dependent death. We detected enzyme-active Ia in the cytosol 48 h after application of iota toxin to cells, indicating that the Ia domain harboring ADP-ribosyltransferase activity remained stable in the cytosol. Prompted by our recent observation that C2 toxin delays apoptosis and persists as an active ADP-ribosyltransferase in the cytosol of intoxicated cells (10), we have now addressed whether the long-lived nature of clostridial actin-ADP-ribosylating toxins in the cytosol is crucial for delayed cell death. To this end, we also investigated whether a fusion toxin containing the catalytic domain of S. enterica SpvB (C/SpvB) mono-ADP ribosylates G-actin as do the C2 and iota toxins (11, 27).  相似文献   

13.
Four hybridoma cell lines producing monoclonal antibodies to Clostridium perfringens enterotoxin were established by fusion of mouse myeloma and spleen cells obtained from mice immunized with the enterotoxin and its toxoid. An enzyme-linked immunosorbent assay indicated that the two antibodies, 2-B-4 and 3-G-10, bound to those regions that were located close each other; the others, 3-B-2 and 2-H-2, bound to other independent regions on the enterotoxin. Release of 51Cr from Vero cells with the enterotoxin was inhibited by either 2-B-4 or 3-G-10, both of which inhibited the binding of 125I-labeled enterotoxin to the cells. Neither binding nor cytotoxicity of the enterotoxin was affected by 2-H-2; 3-B-2 only barely inhibited the binding but neutralized the enterotoxin shown by 51Cr release. It seems justified to conclude that 3-B-2 blocks the toxic action after the enterotoxin has bound to Vero cells.  相似文献   

14.
Enterotoxin produced by a sporulating culture of Clostridium perfringens type C, which had been isolated from a case of severe necrotic enteritis, was purified. The molecular weight was estimated to be 36,000 by gel chromatography on Sephadex G-100 and 33,400 by ultracentrifugation. The sedimentation coefficient S20,W was 2.92. The toxin protein exhibited unusual behavior on sodium dodecyl sulfate gels, and toxin aggregates having molecular weights of 68,000, 85,000, 105,000, and 140,000 were obtained. The purified enterotoxin often separated, apparently due to slight charge differences, into two protein bands on 7% polyacrylamide gels. Electrofocusing of enterotoxin on polyacrylamide gels gave an approximate isoelectric point of 4.3, with the enterotoxin being fractionated into four distinct protein bands. The specific toxicity of the enterotoxin was about 1,900 mouse mean lethal doses per mg of calculated nitrogen. The data obtained indicate that the enterotoxin from C. perfringens type C is identical in most respects to that obtained from type A strains. Whether or not this toxin plays a role in the necrotic enteritis caused by type C strains is unknown at present.  相似文献   

15.
Clostridium perfringens type D causes disease in sheep, goats, and other ruminants. Type D isolates produce, at minimum, alpha and epsilon (ETX) toxins, but some express up to five different toxins, raising questions about which toxins are necessary for the virulence of these bacteria. We evaluated the contribution of ETX to C. perfringens type D pathogenicity in an intraduodenal challenge model in sheep, goats, and mice using a virulent C. perfringens type D wild-type strain (WT), an isogenic ETX null mutant (etx mutant), and a strain where the etx mutation has been reversed (etx complemented). All sheep and goats, and most mice, challenged with the WT isolate developed acute clinical disease followed by death in most cases. Sheep developed various gross and/or histological changes that included edema of brain, lungs, and heart as well as hydropericardium. Goats developed various effects, including necrotizing colitis, pulmonary edema, and hydropericardium. No significant gross or histological abnormalities were observed in any mice infected with the WT strain. All sheep, goats, and mice challenged with the isogenic etx mutant remained clinically healthy for ≥24 h, and no gross or histological abnormalities were observed in those animals. Complementation of etx knockout restored virulence; most goats, sheep, and mice receiving this complemented mutant developed clinical and pathological changes similar to those observed in WT-infected animals. These results indicate that ETX is necessary for type D isolates to induce disease, supporting a key role for this toxin in type D disease pathogenesis.  相似文献   

16.
The iota toxin which is produced by Clostridium perfringens type E, is a binary toxin consisting of two independent polypeptides: Ia, which is an ADP-ribosyltransferase, and Ib, which is involved in the binding and internalization of the toxin into the cell. Two degenerate oligonucleotide probes deduced from partial amino acid sequence of each component of C. spiroforme toxin, which is closely related to the iota toxin, were used to clone three overlapping DNA fragments containing the iota-toxin genes from C. perfringens type E plasmid DNA. Two genes, in the same orientation, coding for Ia (387 amino acids) and Ib (875 amino acids) and separated by 243 noncoding nucleotides were identified. A predicted signal peptide was found for each component, and the secreted Ib displays two domains, the propeptide (172 amino acids) and the mature protein (664 amino acids). The Ia gene has been expressed in Escherichia coli and C. perfringens, under the control of its own promoter. The recombinant polypeptide obtained was recognized by Ia antibodies and ADP-ribosylated actin. The expression of the Ib gene was obtained in E. coli harboring a recombinant plasmid encompassing the putative promoter upstream of the Ia gene and the Ia and Ib genes. Two residues which have been found to be involved in the NAD+ binding site of diphtheria and pseudomonas toxins are conserved in the predicted Ia sequence (Glu-14 and Trp-19). The predicted amino acid Ib sequence shows 33.9% identity with and 54.4% similarity to the protective antigen of the anthrax toxin complex. In particular, the central region of Ib, which contains a predicted transmembrane segment (Leu-292 to Ser-308), presents 45% identity with the corresponding protective antigen sequence which is involved in the translocation of the toxin across the cell membrane.  相似文献   

17.
The important veterinary pathogen Clostridium perfringens type B is unique for producing the two most lethal C. perfringens toxins, i.e., epsilon-toxin and beta-toxin. Our recent study (K. Miyamoto, J. Li, S. Sayeed, S. Akimoto, and B. A. McClane, J. Bacteriol. 190:7178-7188, 2008) showed that most, if not all, type B isolates carry a 65-kb epsilon-toxin-encoding plasmid. However, this epsilon-toxin plasmid did not possess the cpb gene encoding beta-toxin, suggesting that type B isolates carry at least one additional virulence plasmid. Therefore, the current study used Southern blotting of pulsed-field gels to localize the cpb gene to ∼90-kb plasmids in most type B isolates, although a few isolates carried a ∼65-kb cpb plasmid distinct from their etx plasmid. Overlapping PCR analysis then showed that the gene encoding the recently discovered TpeL toxin is located ∼3 kb downstream of the plasmid-borne cpb gene. As shown earlier for their epsilon-toxin-encoding plasmids, the beta-toxin-encoding plasmids of type B isolates were found to carry a tcp locus, suggesting that they are conjugative. Additionally, IS1151-like sequences were identified upstream of the cpb gene in type B isolates. These IS1151-like sequences may mobilize the cpb gene based upon detection of possible cpb-containing circular transposition intermediates. Most type B isolates also possessed a third virulence plasmid that carries genes encoding urease and lambda-toxin. Collectively, these findings suggest that type B isolates are among the most plasmid dependent of all C. perfringens isolates for virulence, as they usually carry three potential virulence plasmids.Isolates of the Gram-positive, spore-forming anaerobe Clostridium perfringens are classified (31) into five different types (A to E), depending upon their production of four (alpha, beta, epsilon, and iota) lethal typing toxins. All C. perfringens types produce alpha-toxin; in addition, type B isolates produce both beta- and epsilon-toxins, type C isolates produce beta-toxin, type D isolates produce epsilon-toxin and type E isolates produce iota-toxin. Except for the chromosomal alpha-toxin gene (plc), all C. perfringens typing toxins are encoded by genes resident on large plasmids (11, 22, 23, 32, 33). Large plasmids can also encode other C. perfringens toxins, such as the enterotoxin (CPE) or beta2-toxin (8, 9, 14, 35), as well as other potential virulence factors such as urease (12, 23).The large virulence plasmids of C. perfringens are only now being characterized (23, 28, 29, 33). The first analyzed, and still most studied, C. perfringens toxin plasmids are the CPE-encoding plasmids of type A isolates (14, 28). In type A isolates, most plasmids carrying the enterotoxin gene (cpe) belong to one of two families: (i) a 75.3-kb plasmid with a cpe locus containing an IS1151 element and the cpb2 gene encoding beta2-toxin or (ii) a 70.5-kb plasmid that lacks the cpb2 gene and carries a cpe locus with an IS1470-like sequence instead of an IS1151 element. Sequence comparisons (28) revealed that these two cpe plasmid families of type A isolates share a conserved region of ∼35 kb that includes the transfer of clostridial plasmid (tcp) locus, which is related to the conjugative transposon Tn916. Confirming that cpe plasmids can be conjugative, mixed mating studies have directly demonstrated transfer of the cpe plasmid from type A isolate F4969 to other C. perfringens isolates (5). A similar tcp locus is also shared by the tetracycline resistance plasmid pCW3 and several other toxin plasmids (2, 23, 28, 29, 33), as discussed below. Mutagenesis analyses demonstrated the importance of several genes in the tcp locus for conjugative transfer of pCW3 (2) and, by extension, presumably the tcp-carrying, conjugative toxin plasmids, such as the cpe plasmid of isolate F4969 (5) and some etx plasmids of type D isolates (19).Although the sequence of an iota-toxin-encoding plasmid has not yet been published, pulsed-field gel electrophoresis (PFGE) and PCR analyses determined that these plasmids are typically larger than the cpe plasmids of type A isolates (23). Specifically, iota-toxin plasmids are often ≥100 kb in size, reaching up to a size of ∼135 kb. These plasmids of type E isolates often encode, in addition to the iota-toxin, other potential virulence factors such as lambda-toxin and urease. These plasmids also carry a tcp locus, suggesting that they may be capable of conjugative transfer. Interestingly, many iota-toxin plasmids appear to be related, sometimes extensively, to the cpe plasmids of type A isolates. Consequently, it has been suggested (3, 23) that many iota-toxin plasmids arose from insertion of an iota-toxin gene-carrying mobile genetic element near the cpe gene on a tcp-carrying type A plasmid. This insertional event apparently inactivated the cpe gene, so most or all type E isolates now carry silent cpe genes (3, 23).The epsilon-toxin-encoding plasmids of type D isolates show considerable size variations (33), ranging from ∼48 kb to ∼110 kb. These size variations in type D etx plasmids reflect, in part, differences among their toxin gene carriage. The small 48-kb etx plasmids present in some type D isolates typically lack either the cpe gene or the cpb2 gene (encoding beta2-toxin), while the larger (>75-kb) etx plasmids found in other type D isolates can also carry the cpe gene, the cpb2 gene, or both the cpe and cpb2 genes. Consequently, some type D isolates carry a toxin plasmid encoding only etx, other type D isolates carry a toxin plasmid with up to three different functional toxin genes (etx, cpb2, and cpe), and the remaining type D isolates carry their etx, cpe, and cpb2 genes on up to three distinct plasmids.C. perfringens type B isolates uniquely produce both beta- and epsilon-toxins, the two most lethal C. perfringens toxins (13). These bacteria are important pathogens of sheep but also cause disease in goats, calves, and foals (26). For unknown reasons, diseases caused by C. perfringens type B isolates apparently are restricted to certain geographic regions (24, 25, 26). C. perfringens type B enterotoxemias initiate when these bacteria proliferate in the gut, accompanied by toxin production. Those toxins initially affect the intestines but later are absorbed and act systemically. Studies from our group (13) showed that beta- and epsilon-toxins each contribute to lethality in a mouse model involving intravenous injection of type B culture supernatants.There has been characterization of only one type B virulence plasmid to date. Our recent study (29) showed that most, if not all, type B isolates carry a common etx plasmid of ∼65 kb that also possesses a tcp locus and a cpb2 gene, although not the cpb gene encoding beta-toxin. Interestingly, the type B etx plasmid is highly (80%) related to the ∼75-kb cpe- and cpb2-carrying plasmid found in some type A isolates (28). The ∼65-kb etx plasmid present in most, if not all, type B isolates is also carried by a minority of type D isolates (29).The absence of the cpb gene from their etx plasmids suggested that most type B isolates might carry additional virulence plasmids. Therefore, the current study was performed to better address virulence plasmid carriage and diversity among type B disease isolates.  相似文献   

18.
Toxigenic strains of Clostridium difficile have been reported to produce both toxins A and B nearly always, and nontoxigenic strains have been reported to produce neither of these toxins. Recent studies indicate that it is not always true. We established a PCR assay to differentiate toxin A-negative, toxin B-positive (toxin A−, toxin B+) strains from both toxin-positive (toxin A+, toxin B+) strains and both toxin-negative (toxin A−, toxin B−) strains as an alternative to cell culture assay and enzyme-linked immunosorbent assay (ELISA). By using the PCR primer set NK11 and NK9 derived from the repeating sequences of the toxin A gene, a shorter segment (ca. 700 bp) was amplified from toxin A−, toxin B+ strains compared to the size of the segment amplified from toxin A+, toxin B+ strains (ca. 1,200 bp), and no product was amplified from toxin A−, toxin B− strains. We examined a total of 421 C. difficile isolates by PCR. Of these, 48 strains showed a shorter segment by the PCR, were negative by ELISAs for the detection of toxin A, and were positive by cell culture assay. Although the cytotoxin produced by the toxin A−, toxin B+ strains was neutralized by anti-toxin B serum, the appearance of the cytotoxic effects on Vero cell monolayers was distinguishable from that of toxin A+, toxin B+ strains. By immunoblotting, the 44 toxin A−, toxin B+ strains were typed to serogroup F and the remaining four strains were serogroup X. Pulsed-field gel electrophoresis separated the 48 strains into 19 types. The PCR assay for the detection of the repeating sequences combined with PCR amplification of the nonrepeating sequences of either the toxin A or the toxin B gene is indicated to be useful for differentiating toxin A−, toxin B+ strains from toxin A+, toxin B+ and toxin A−, toxin B− strains and will contribute to elucidation of the precise role of toxin A−, toxin B+ strains in intestinal diseases.  相似文献   

19.
20.
Hybridomas secreting monoclonal antibodies (MABs) specific for Clostridium perfringens type A enterotoxin were produced by fusion of P3X63Ag8.653 myeloma cells with spleen cells from BALB/c mice immunized with purified enterotoxin. Wells containing hybridomas secreting immunoglobulin G (IgG) antibodies against enterotoxin were specifically identified by an indirect enzyme-linked immunosorbent assay (ELISA), and 10 ELISA-positive hybridomas were selected and cloned twice by limiting dilution. All 10 hybridomas produced MABs containing immunoglobulin G1 heavy chains and kappa (kappa) light chains. These hybridomas were then grown as ascitic tumors in mice, and MABs were purified from the ascites fluids with DEAE Affi-gel blue. The specificity of the MABs for enterotoxin was demonstrated by immunoblotting and ELISA. Competitive radioimmunoassay with 125I-MABs suggests that these MABs recognized at least four epitopes on the enterotoxin molecule. The enterotoxin-neutralizing ability of MABs from both hybridoma culture supernatants and ascites fluids was assessed by using a 3H-nucleotide-release Vero (African green monkey kidney) cell assay. Only 2 of the 10 hybridomas produced MABs which completely (greater than 90%) neutralized the biologic activity of enterotoxin. Preincubation of 125I-enterotoxin with MABs demonstrated that MAB neutralizing ability correlated with MAB-specific inhibition of specific binding of enterotoxin to intestinal brush border membranes.  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