Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Med Microbiol ; 310(2): 151398, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31987726

RESUMEN

Clostridium perfringens is the causative agent of human clostridial myonecrosis; the major toxins involved in this disease are α-toxin and perfringolysin O. The RevSR two-component regulatory system has been shown to be involved in regulating virulence in a mouse myonecrosis model. Previous microarray and RNAseq analysis of a revR mutant implied that factors other than the major toxins may play a role in virulence. The RNAseq data showed that the expression of the gene encoding the EngCP endo α-N-acetylgalactosaminidase (CPE0693) was significantly down-regulated in a revR mutant. Enzymes from this family have been identified in several Gram-positive pathogens and have been postulated to contribute to their virulence. In this study, we constructed an engCP mutant of C. perfringens and showed that it was significantly less virulent than its wild-type parent strain. Virulence was restored by complementation in trans with the wild-type engCP gene. We also demonstrated that purified EngCP was able to hydrolyse α-dystroglycan derived from C2C12 mouse myotubes. However, EngCP had little effect on membrane permeability in mice, suggesting that EngCP may play a role other than the disruption of the structural integrity of myofibres. Glycan array analysis indicated that EngCP could recognise structures containing the monosaccharide N-acetlygalactosamine at 4C, but could recognise structures terminating in galactose, glucose and N-acetylglucosamine under conditions where EngCP was enzymatically active. In conclusion, we have obtained evidence that EngCP is required for virulence in C. perfringens and, although classical exotoxins are important for disease, we have now shown that an O-glycosidase also plays an important role in the disease process.


Asunto(s)
Clostridium perfringens/enzimología , Clostridium perfringens/patogenicidad , Gangrena Gaseosa/microbiología , Factores de Virulencia/genética , alfa-N-Acetilgalactosaminidasa/genética , Animales , Permeabilidad de la Membrana Celular , Clostridium perfringens/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Ratones , Ratones Endogámicos BALB C , Análisis de Secuencia de ARN , alfa-N-Acetilgalactosaminidasa/metabolismo
2.
mBio ; 9(2)2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29588405

RESUMEN

To obtain an insight into host-pathogen interactions in clostridial myonecrosis, we carried out comparative transcriptome analysis of both the bacterium and the host in a murine Clostridium perfringens infection model, which is the first time that such an investigation has been conducted. Analysis of the host transcriptome from infected muscle tissues indicated that many genes were upregulated compared to the results seen with mock-infected mice. These genes were enriched for host defense pathways, including Toll-like receptor (TLR) and Nod-like receptor (NLR) signaling components. Real-time PCR confirmed that host TLR2 and NLRP3 inflammasome genes were induced in response to C. perfringens infection. Comparison of the transcriptome of C. perfringens cells from the infected tissues with that from broth cultures showed that host selective pressure induced a global change in C. perfringens gene expression. A total of 33% (923) of C. perfringens genes were differentially regulated, including 10 potential virulence genes that were upregulated relative to their expression in vitro These genes encoded putative proteins that may be involved in the synthesis of cell wall-associated macromolecules, in adhesion to host cells, or in protection from host cationic antimicrobial peptides. This report presents the first successful expression profiling of coregulated transcriptomes of bacterial and host genes during a clostridial myonecrosis infection and provides new insights into disease pathogenesis and host-pathogen interactions.IMPORTANCEClostridium perfringens is the causative agent of traumatic clostridial myonecrosis, or gas gangrene. In this study, we carried out transcriptional analysis of both the host and the bacterial pathogen in a mouse myonecrosis infection. The results showed that in comparison to mock-infected control tissues, muscle tissues from C. perfringens-infected mice had a significantly altered gene expression profile. In particular, the expression of many genes involved in the innate immune system was upregulated. Comparison of the expression profiles of C. perfringens cells isolated from the infected tissues with those from equivalent broth cultures identified many potential virulence genes that were significantly upregulated in vivo These studies have provided a new understanding of the range of factors involved in host-pathogen interactions in a myonecrosis infection.


Asunto(s)
Clostridium perfringens/patogenicidad , Gangrena Gaseosa/genética , Gangrena Gaseosa/inmunología , Animales , Femenino , Gangrena Gaseosa/microbiología , Interacciones Huésped-Patógeno , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos BALB C , Transcriptoma/genética , Virulencia/genética
3.
PLoS One ; 11(9): e0162981, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27637108

RESUMEN

The ability of a pathogenic bacterium to scavenge iron from its host is important for its growth and survival during an infection. Our studies on C. perfringens gas gangrene strain JIR325, a derivative of strain 13, showed that it is capable of utilizing both human hemoglobin and ferric chloride, but not human holo-transferrin, as an iron source for in vitro growth. Analysis of the C. perfringens strain 13 genome sequence identified a putative heme acquisition system encoded by an iron-regulated surface gene region that we have named the Cht (Clostridium perfringens heme transport) locus. This locus comprises eight genes that are co-transcribed and includes genes that encode NEAT domain-containing proteins (ChtD and ChtE) and a putative sortase (Srt). The ChtD, ChtE and Srt proteins were shown to be expressed in JIR325 cells grown under iron-limited conditions and were localized to the cell envelope. Moreover, the NEAT proteins, ChtD and ChtE, were found to bind heme. Both chtDE and srt mutants were constructed, but these mutants were not defective in hemoglobin or ferric chloride utilization. They were, however, attenuated for virulence when tested in a mouse myonecrosis model, although the virulence phenotype could not be restored via complementation and, as is common with such systems, secondary mutations were identified in these strains. In summary, this study provides evidence for the functional redundancies that occur in the heme transport pathways of this life threatening pathogen.


Asunto(s)
Clostridium perfringens/metabolismo , Hemo/metabolismo , Western Blotting , Electroforesis en Gel de Poliacrilamida , Unión Proteica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transcripción Genética
4.
PLoS Pathog ; 12(7): e1005758, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27414650

RESUMEN

Clostridium difficile is a global health burden and the leading cause of antibiotic-associated diarrhoea worldwide, causing severe gastrointestinal disease and death. Three well characterised toxins are encoded by this bacterium in two genetic loci, specifically, TcdB (toxin B) and TcdA (toxin A) in the Pathogenicity Locus (PaLoc) and binary toxin (CDT) in the genomically distinct CDT locus (CdtLoc). Toxin production is controlled by regulators specific to each locus. The orphan response regulator, CdtR, encoded within the CdtLoc, up-regulates CDT production. Until now there has been no suggestion that CdtR influences TcdA and TcdB production since it is not carried by all PaLoc-containing strains and CdtLoc is not linked genetically to PaLoc. Here we show that, in addition to CDT, CdtR regulates TcdA and TcdB production but that this effect is strain dependent. Of clinical relevance, CdtR increased the production of TcdA, TcdB and CDT in two epidemic ribotype 027 human strains, modulating their virulence in a mouse infection model. Strains traditionally from animal lineages, notably ribotype 078 strains, are increasingly being isolated from humans and their genetic and phenotypic analysis is critical for future studies on this important pathogen. Here we show that CdtR-mediated toxin regulation did not occur in other strain backgrounds, including a ribotype 078 animal strain. The finding that toxin gene regulation is strain dependent highlights the regulatory diversity between C. difficile isolates and the importance of studying virulence regulation in diverse lineages and clinically relevant strains. Our work provides the first evidence that TcdA, TcdB and CDT production is linked by a common regulatory mechanism and that CdtR may act as a global regulator of virulence in epidemic 027 strains.


Asunto(s)
Clostridioides difficile/metabolismo , Enterocolitis Seudomembranosa/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Factores de Virulencia/biosíntesis , Virulencia/fisiología , ADP Ribosa Transferasas/biosíntesis , Animales , Proteínas Bacterianas/biosíntesis , Toxinas Bacterianas/biosíntesis , Western Blotting , Modelos Animales de Enfermedad , Enterotoxinas/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
5.
Int J Med Microbiol ; 306(6): 429-42, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27267179

RESUMEN

Clostridium perfringens is a Gram-positive, anaerobic, spore-forming bacterium that causes human gas gangrene (clostridial myonecrosis) and food poisoning. Early studies showed that virulence was regulated by the VirSR two-component signal transduction system. However, our identification of the RevR orphan response regulator indicated that more than one system was involved in controlling virulence. To further characterize this virulence-associated regulator, gel mobility shift experiments, coupled with DNase I footprinting, were used to identify the RevR DNA binding sequence. Bioinformatics analysis suggested that an orphan sensor histidine kinase, CPE1757 (renamed RevS), was the cognate sensor of RevR. Interaction between RevS and RevR was demonstrated by use of a bacterial two-hybrid system and validated by protein-protein interaction studies using biolayer interferometry. To assess the involvement of RevS in virulence regulation, the revS gene was inactivated by Targetron insertion. When isogenic wild-type, revS and complemented revS strains were tested in a mouse myonecrosis model, the revS mutant was found to be attenuated in virulence, which was similar to the attenuation observed previously with the revR mutant. However, transcriptional analysis of selected RevR-regulated genes in the revS mutant revealed a different pattern of expression to a revR mutant, suggesting that the RevSR system is more complex than originally thought. Taken together, the results have led to the identification and characterization of the two essential parts of a new regulatory network that is involved in the regulation of virulence in C. perfringens.


Asunto(s)
Clostridium perfringens/fisiología , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Clostridium perfringens/genética , Huella de ADN , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Histidina Quinasa/genética , Ratones Endogámicos BALB C , Mutagénesis Insercional , Unión Proteica , Mapeo de Interacción de Proteínas , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos , Virulencia
6.
Anaerobe ; 41: 10-17, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27178230

RESUMEN

Bacterial pathogens have adopted numerous mechanisms for acquiring iron from host proteins during an infection, including the direct acquisition of ferric iron from heme-associated proteins or from iron-scavenging siderophores. Ferric iron then is transported into the cytosol, where it can be utilized by the bacterial pathogen. Under anaerobic conditions bacteria can also transport ferrous iron using the transmembrane complex FeoAB, but little is known about iron transport systems in anaerobic bacteria such as the pathogenic clostridia. In this study we sought to characterize the iron acquisition process in Clostridium perfringens. Bioinformatic analysis of the Clostridium perfringens strain 13 genome sequence revealed that it has seven potential iron acquisition systems: three siderophore-mediated systems, one ferric citrate uptake system, two heme-associated acquisition systems and one ferrous iron uptake system (FeoAB). The relative level of expression of these systems was determined using quantitative real-time RT-PCR assays that were specific for one gene from each system. Each of these genes was expressed, with the feoAB genes generating the most abundant iron-uptake related transcripts. To further examine the role of this system in the growth of C. perfringens, insertional inactivation was used to isolate a chromosomal feoB mutant. Growth of this mutant in the presence and absence of iron revealed that it had altered growth properties and a markedly reduced total iron and manganese content compared to the wild type; effects that were reversed upon complementation with the wild-type feoB gene. These studies suggest that under anaerobic conditions FeoB is the major protein required for the uptake of iron into the cell and that it may play an important role in the pathogenesis of C. perfringens infections.


Asunto(s)
Proteínas Bacterianas/genética , Clostridium perfringens/genética , Proteínas de Transporte de Membrana/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/biosíntesis , Clostridium perfringens/metabolismo , Hierro/metabolismo , Manganeso/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Mutación , Transcripción Genética
7.
BMC Genomics ; 17: 391, 2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27216822

RESUMEN

BACKGROUND: Clostridium perfringens causes toxin-mediated diseases, including gas gangrene (clostridial myonecrosis) and food poisoning in humans. The production of the toxins implicated in gas gangrene, α-toxin and perfringolysin O, is regulated by the VirSR two-component regulatory system. In addition, RevR, an orphan response regulator, has been shown to affect virulence in the mouse myonecrosis model. RevR positively regulates the expression of genes that encode hydrolytic enzymes, including hyaluronidases and sialidases. RESULTS: To further characterize the VirSR and RevR regulatory networks, comparative transcriptomic analysis was carried out with strand-specific RNA-seq on C. perfringens strain JIR325 and its isogenic virR and revR regulatory mutants. Using the edgeR analysis package, 206 genes in the virR mutant and 67 genes in the revR mutant were found to be differentially expressed. Comparative analysis revealed that VirR acts as a global negative regulator, whilst RevR acts as a global positive regulator. Therefore, about 95 % of the differentially expressed genes were up-regulated in the virR mutant, whereas 81 % of the differentially expressed genes were down-regulated in the revR mutant. Importantly, we identified 23 genes that were regulated by both VirR and RevR, 18 of these genes, which included the sporulation-specific spoIVA, sigG and sigF genes, were regulated positively and negatively by RevR and VirR, respectively. Furthermore, analysis of the mapped RNA-seq reads visualized as depth of coverage plots showed that there were 93 previously unannotated transcripts in intergenic regions. These transcripts potentially encode small RNA molecules. CONCLUSION: In conclusion, using strand-specific RNA-seq analysis, this study has identified differentially expressed chromosomal and pCP13 native plasmid-encoded genes, antisense transcripts, and transcripts within intergenic regions that are controlled by the VirSR or RevR regulatory systems.


Asunto(s)
Proteínas Bacterianas/genética , Clostridium perfringens/genética , Mutación , Análisis de Secuencia de ARN/métodos , Perfilación de la Expresión Génica/métodos , Regulación Bacteriana de la Expresión Génica , Redes Reguladoras de Genes , Anotación de Secuencia Molecular
8.
PLoS One ; 10(7): e0133217, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26197388

RESUMEN

Among many other virulence factors, Clostridium perfringens produces three sialidases NanH, NanI and NanJ. NanH lacks a secretion signal peptide and is predicted to be an intracellular enzyme, while NanI and NanJ are secreted. Previously, we had identified part of an operon encoding NanE (epimerase) and NanA (sialic acid lyase) enzymes. Further analysis of the entire operon suggests that it encodes a complete pathway for the transport and metabolism of sialic acid along with a putative transcriptional regulator, NanR. The addition of 30 mM N-acetyl neuraminic acid (Neu5Ac) to a semi-defined medium significantly enhanced the growth yield of strain 13, suggesting that Neu5Ac can be used as a nutrient. C. perfringens strain 13 lacks a nanH gene, but has NanI- and NanJ-encoding genes. Analysis of nanI, nanJ, and nanInanJ mutants constructed by homologous recombination revealed that the expression of the major sialidase, NanI, was induced by the addition of Neu5Ac to the medium, and that in separate experiments, the same was true of a nanI-gusA transcriptional fusion. For the nanI and nanJ genes, primer extension identified three and two putative transcription start sites, respectively. Gel mobility shift assays using purified NanR and DNA from the promoter regions of the nanI and nanE genes showed high affinity, specific binding by NanR. We propose that NanR is a global regulator of sialic acid-associated genes and that it responds, in a positive feedback loop, to the concentration of sialic acid in the cell.


Asunto(s)
Proteínas Bacterianas/genética , Infecciones por Clostridium/microbiología , Clostridium perfringens/enzimología , Clostridium perfringens/genética , Regulación Bacteriana de la Expresión Génica , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Clostridium perfringens/crecimiento & desarrollo , Clostridium perfringens/metabolismo , Genes Bacterianos , Humanos , Datos de Secuencia Molecular , Neuraminidasa/metabolismo , Operón , Regiones Promotoras Genéticas , Sitio de Iniciación de la Transcripción , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
9.
Vet Microbiol ; 179(1-2): 23-33, 2015 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-25770894

RESUMEN

The most common animal models used to study Clostridium perfringens infections in humans and animals are reviewed here. The classical C. perfringens-mediated histotoxic disease of humans is clostridial myonecrosis or gas gangrene and the use of a mouse myonecrosis model coupled with genetic studies has contributed greatly to our understanding of disease pathogenesis. Similarly, the use of a chicken model has enhanced our understanding of type A-mediated necrotic enteritis in poultry and has led to the identification of NetB as the primary toxin involved in disease. C. perfringens type A food poisoning is a highly prevalent bacterial illness in the USA and elsewhere. Rabbits and mice are the species most commonly used to study the action of enterotoxin, the causative toxin. Other animal models used to study the effect of this toxin are rats, non-human primates, sheep and cattle. In rabbits and mice, CPE produces severe necrosis of the small intestinal epithelium along with fluid accumulation. C. perfringens type D infection has been studied by inoculating epsilon toxin (ETX) intravenously into mice, rats, sheep, goats and cattle, and by intraduodenal inoculation of whole cultures of this microorganism in mice, sheep, goats and cattle. Molecular Koch's postulates have been fulfilled for enterotoxigenic C. perfringens type A in rabbits and mice, for C. perfringens type A necrotic enteritis and gas gangrene in chickens and mice, respectively, for C. perfringens type C in mice, rabbits and goats, and for C. perfringens type D in mice, sheep and goats.


Asunto(s)
Infecciones por Clostridium/microbiología , Clostridium perfringens/patogenicidad , Gangrena Gaseosa/microbiología , Animales , Bovinos , Pollos , Modelos Animales de Enfermedad , Enterotoxinas/metabolismo , Cabras , Humanos , Ratones , Primates , Conejos , Ratas , Ovinos
10.
Toxins (Basel) ; 7(2): 516-34, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25675415

RESUMEN

Clostridium septicum is the causative agent of atraumatic gas gangrene, with α-toxin, an extracellular pore-forming toxin, essential for disease. How C. septicum modulates the host's innate immune response is poorly defined, although α-toxin-intoxicated muscle cells undergo cellular oncosis, characterised by mitochondrial dysfunction and release of reactive oxygen species. Nonetheless, the signalling events that occur prior to the initiation of oncosis are poorly characterised. Our aims were to characterise the ability of α-toxin to activate the host mitogen activated protein kinase (MAPK) signalling pathway both in vitro and in vivo. Treatment of Vero cells with purified α-toxin activated the extracellular-signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 arms of the MAPK pathway and stimulated the release of TNF-α in a dose-dependent manner. Studies using inhibitors of all three MAPK components suggested that activation of ERK occurred in a Ras-c-Raf dependent manner, whereas activation of JNK and p38 occurred by a Ras-independent mechanism. Toxin-mediated activation was dependent on efficient receptor binding and pore formation and on an influx of extracellular calcium ions. In the mouse myonecrosis model we showed that the MAPK pathway was activated in tissues of infected mice, implying that it has an important role in the disease process.


Asunto(s)
Toxinas Bacterianas/toxicidad , Clostridium septicum/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Citotóxicas Formadoras de Poros/toxicidad , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Toxinas Bacterianas/genética , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Ratones , Músculo Esquelético/enzimología , Proteínas Citotóxicas Formadoras de Poros/genética , Sepsis/enzimología , Sepsis/microbiología , Bazo/enzimología , Células Vero
11.
Anaerobe ; 30: 199-204, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25152227

RESUMEN

Clostridium perfringens is a Gram-positive rod that is widely distributed in nature and is the etiological agent of several human and animal diseases. The complete genome sequence of C. perfringens strain 13 has been determined and multiple two-component signal transduction systems identified. One of these systems, designated here as the MalNO system, was analyzed in this study. Microarray analysis was used to carry out functional analysis of a malO mutant. The results, which were confirmed by quantitative reverse-transcriptase PCR, indicated that genes putatively involved in the uptake and metabolism of maltose were up-regulated in the malO mutant. These effects were reversed by complementation with the wild-type malO gene. Growth of these isogenic strains in medium with and without maltose showed that the malO mutant recovered more quickly from maltose deprivation when compared to the wild-type and complemented strains, leading to the conclusion that the MalNO system regulates maltose utilization in C. perfringens. It is postulated that this regulatory network may allow this soil bacterium and opportunistic pathogen to respond to environmental conditions where there are higher concentrations of maltose or maltodextrins, such as in the presence of decaying plant material in rich soil.


Asunto(s)
Clostridium perfringens/genética , Clostridium perfringens/metabolismo , Regulación Bacteriana de la Expresión Génica , Maltosa/metabolismo , Transducción de Señal , Eliminación de Gen , Perfilación de la Expresión Génica , Prueba de Complementación Genética , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa , Microbiología del Suelo
12.
Appl Environ Microbiol ; 80(12): 3597-3603, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24682304

RESUMEN

TnpX is a site-specific recombinase responsible for the excision and insertion of the transposons Tn4451 and Tn4453 in Clostridium perfringens and Clostridium difficile, respectively. Here, we exploit phenotypic features of TnpX to facilitate genetic mutagenesis and complementation studies. Genetic manipulation of bacteria often relies on the use of antibiotic resistance genes; however, a limited number are available for use in the clostridia. The ability of TnpX to recognize and excise specific DNA fragments was exploited here as the basis of an antibiotic resistance marker recycling system, specifically to remove antibiotic resistance genes from plasmids in Escherichia coli and from marked chromosomal C. perfringens mutants. This methodology enabled the construction of a C. perfringens plc virR double mutant by allowing the removal and subsequent reuse of the same resistance gene to construct a second mutation. Genetic complementation can be challenging when the gene of interest encodes a product toxic to E. coli. We show that TnpX represses expression from its own promoter, PattCI, which can be exploited to facilitate the cloning of recalcitrant genes in E. coli for subsequent expression in the heterologous host C. perfringens. Importantly, this technology expands the repertoire of tools available for the genetic manipulation of the clostridia.


Asunto(s)
Proteínas Bacterianas/metabolismo , Clonación Molecular/métodos , Clostridium perfringens/genética , ADN Bacteriano/genética , Escherichia coli/genética , Genoma Bacteriano , Recombinasas/metabolismo , Proteínas Bacterianas/genética , Clostridium perfringens/enzimología , ADN Nucleotidiltransferasas , Escherichia coli/metabolismo , Prueba de Complementación Genética , Recombinasas/genética , Recombinación Genética
13.
Mol Microbiol ; 91(2): 221-31, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24563915

RESUMEN

The genus Clostridium comprises a large, heterogeneous group of obligate anaerobic, Gram-positive spore forming bacilli. Members of this genus are ubiquitous in the environment and although most species are considered saprophytic, several are pathogenic to both humans and animals. These bacteria cause a variety of diseases including neuroparalysis, gas gangrene, necrotic enteritis, food poisoning, toxic shock syndrome and pseudomembraneous colitis, which in most cases arise as a consequence of the production of potent exotoxins. Treatment options are often limited, underscoring the need for new treatment strategies and novel therapeutics. Understanding the fundamental mechanisms and signals that control toxin production in the pathogenic clostridia may lead to the identification of novel therapeutic targets that can be exploited in the development of new antimicrobial agents.


Asunto(s)
Toxinas Bacterianas/biosíntesis , Clostridium/metabolismo , Genes Bacterianos , Secuencia de Aminoácidos , Regulación Bacteriana de la Expresión Génica , Filogenia , Alineación de Secuencia , Transducción de Señal
14.
J Infect Dis ; 210(3): 483-92, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24550443

RESUMEN

Gas gangrene is a potentially fatal disease that is primarily caused by the ubiquitous, anaerobic bacteria Clostridium perfringens and Clostridium septicum. Treatment is limited to antibiotic therapy, debridement of the infected tissue, and, in severe cases, amputation. The need for new treatment approaches is compelling. Opioid-based analgesics such as buprenorphine and morphine also have immunomodulatory properties, usually leading to faster disease progression. However, here we show that mice pretreated with buprenorphine and morphine do not die from clostridial myonecrosis. Treatment with buprenorphine after the onset of infection also arrested disease development. Protection against myonecrotic disease was specific to C. perfringens-mediated myonecrosis; buprenorphine did not protect against disease caused by C. septicum infection even though infections due to both species are very similar. These data provide the first evidence of a protective role for opioids during infection and suggest that new therapeutic strategies may be possible for the treatment of C. perfringens-mediated myonecrosis.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Buprenorfina/uso terapéutico , Clostridium perfringens , Gangrena Gaseosa/tratamiento farmacológico , Morfina/uso terapéutico , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Naltrexona/uso terapéutico
15.
PLoS One ; 8(9): e73525, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023881

RESUMEN

Clostridium perfringens is ubiquitous in nature and is often found as a commensal of the human and animal gastrointestinal tract. It is the primary etiological agent of clostridial myonecrosis, or gas gangrene, a serious infection that results in extensive tissue necrosis due to the action of one or more potent extracellular toxins. α-toxin and perfringolysin O are the major extracellular toxins involved in the pathogenesis of gas gangrene, but histotoxic strains of C. perfringens, such as strain 13, also produce many degradative enzymes such as collagenases, hyaluronidases, sialidases and the cysteine protease, α-clostripain. The production of many of these toxins is regulated either directly or indirectly by the global VirSR two-component signal transduction system. By isolating a chromosomal mutant and carrying out microarray analysis we have identified an orphan sensor histidine kinase, which we have named ReeS (regulator of extracellular enzymes sensor). Expression of the sialidase genes nanI and nanJ was down-regulated in a reeS mutant. Since complementation with the wild-type reeS gene restored nanI and nanJ expression to wild-type levels, as shown by quantitative reverse transcription-PCR and sialidase assays we concluded that ReeS positively regulates the expression of these sialidase genes. However, mutation of the reeS gene had no significant effect on virulence in the mouse myonecrosis model. Sialidase production in C. perfringens has been previously shown to be regulated by both the VirSR system and RevR. In this report, we have analyzed a previously unknown sensor histidine kinase, ReeS, and have shown that it also is involved in controlling the expression of sialidase genes, adding further complexity to the regulatory network that controls sialidase production in C. perfringens.


Asunto(s)
Clostridium perfringens/metabolismo , Neuraminidasa/biosíntesis , Proteínas Quinasas/metabolismo , Animales , Clostridium perfringens/citología , Clostridium perfringens/enzimología , Clostridium perfringens/genética , Espacio Extracelular/metabolismo , Femenino , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa , Ratones , Ratones Endogámicos BALB C , Mutación , Neuraminidasa/genética , Proteínas Quinasas/genética
16.
mBio ; 4(1): e00019-13, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23386432

RESUMEN

Clostridium perfringens is an anaerobic bacterium that causes numerous important human and animal diseases, primarily as a result of its ability to produce many different protein toxins. In chickens, C. perfringens causes necrotic enteritis, a disease of economic importance to the worldwide poultry industry. The secreted pore-forming toxin NetB is a key virulence factor in the pathogenesis of avian necrotic enteritis and is similar to alpha-hemolysin, a ß-barrel pore-forming toxin from Staphylococcus aureus. To address the molecular mechanisms underlying NetB-mediated tissue damage, we determined the crystal structure of the monomeric form of NetB to 1.8 Å. Structural comparisons with other members of the alpha-hemolysin family revealed significant differences in the conformation of the membrane binding domain. These data suggested that NetB may recognize different membrane receptors or use a different mechanism for membrane-protein interactions. Consistent with this idea, electrophysiological experiments with planar lipid bilayers revealed that NetB formed pores with much larger single-channel conductance than alpha-hemolysin. Channel conductance varied with phospholipid net charge. Furthermore, NetB differed in its ion selectivity, preferring cations over anions. Using hemolysis as a screen, we carried out a random-mutagenesis study that identified several residues that are critical for NetB-induced cell lysis. Mapping of these residues onto the crystal structure revealed that they were clustered in regions predicted to be required for oligomerization or membrane binding. Together these data provide an insight into the mechanism of NetB-mediated pore formation and will contribute to our understanding of the mode of action of this important toxin. IMPORTANCE Necrotic enteritis is an economically important disease of the worldwide poultry industry and is mediated by Clostridium perfringens strains that produce NetB, a ß-pore-forming toxin. We carried out structural and functional studies of NetB to provide a mechanistic insight into its mode of action and to assist in the development of a necrotic enteritis vaccine. We determined the structure of the monomeric form of NetB to 1.8 Å, used both site-directed and random mutagenesis to identify key residues that are required for its biological activity, and analyzed pore formation by NetB and its substitution-containing derivatives in planar lipid bilayers.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Clostridium perfringens/química , Clostridium perfringens/patogenicidad , Enterotoxinas/química , Enterotoxinas/metabolismo , Animales , Toxinas Bacterianas/genética , Transporte Biológico , Cationes/metabolismo , Pollos , Clostridium perfringens/genética , Cristalografía por Rayos X , Análisis Mutacional de ADN , Enterotoxinas/genética , Eritrocitos/efectos de los fármacos , Hemólisis , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína
17.
mBio ; 2(5)2011.
Artículo en Inglés | MEDLINE | ID: mdl-21954306

RESUMEN

UNLABELLED: The pathogenesis of avian necrotic enteritis involves NetB, a pore-forming toxin produced by virulent avian isolates of Clostridium perfringens type A. To determine the location and mobility of the netB structural gene, we examined a derivative of the tetracycline-resistant necrotic enteritis strain EHE-NE18, in which netB was insertionally inactivated by the chloramphenicol and thiamphenicol resistance gene catP. Both tetracycline and thiamphenicol resistance could be transferred either together or separately to a recipient strain in plate matings. The separate transconjugants could act as donors in subsequent matings, which demonstrated that the tetracycline resistance determinant and the netB gene were present on different conjugative elements. Large plasmids were isolated from the transconjugants and analyzed by high-throughput sequencing. Analysis of the resultant data indicated that there were actually three large conjugative plasmids present in the original strain, each with its own toxin or antibiotic resistance locus. Each plasmid contained a highly conserved 40-kb region that included plasmid replication and transfer regions that were closely related to the 47-kb conjugative tetracycline resistance plasmid pCW3 from C. perfringens. The plasmids were as follows: (i) a conjugative 49-kb tetracycline resistance plasmid that was very similar to pCW3, (ii) a conjugative 82-kb plasmid that contained the netB gene and other potential virulence genes, and (iii) a 70-kb plasmid that carried the cpb2 gene, which encodes a different pore-forming toxin, beta2 toxin. IMPORTANCE: The anaerobic bacterium Clostridium perfringens can cause an avian gastrointestinal disease known as necrotic enteritis. Disease pathogenesis is not well understood, although the plasmid-encoded pore-forming toxin NetB, is an important virulence factor. In this work, we have shown that the plasmid that carries the netB gene is conjugative and has a 40-kb region that is very similar to replication and transfer regions found within each of the sequenced conjugative plasmids from C. perfringens. We also showed that this strain contained two additional large plasmids that were also conjugative and carried a similar 40-kb region. One of these plasmids encoded beta2 toxin, and the other encoded tetracycline resistance. To our knowledge, this is the first report of a bacterial strain that carries three closely related but different independently conjugative plasmids. These results have significant implications for our understanding of the transmission of virulence and antibiotic resistance genes in pathogenic bacteria.


Asunto(s)
Toxinas Bacterianas/genética , Clostridium perfringens/genética , Clostridium perfringens/aislamiento & purificación , Farmacorresistencia Bacteriana , Enterocolitis Necrotizante/microbiología , Enterotoxinas/genética , Plásmidos , Antibacterianos/farmacología , Clostridium perfringens/efectos de los fármacos , Clostridium perfringens/patogenicidad , Conjugación Genética , ADN Bacteriano/química , ADN Bacteriano/genética , Técnicas de Inactivación de Genes , Transferencia de Gen Horizontal , Humanos , Datos de Secuencia Molecular , Mutagénesis Insercional , Análisis de Secuencia de ADN
18.
PLoS One ; 6(7): e22762, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829506

RESUMEN

Clostridium perfringens is the causative agent of clostridial myonecrosis or gas gangrene and produces many different extracellular toxins and enzymes, including the cysteine protease α-clostripain. Mutation of the α-clostripain structural gene, ccp, alters the turnover of secreted extracellular proteins in C. perfringens, but the role of α-clostripain in disease pathogenesis is not known. We insertionally inactivated the ccp gene C. perfringens strain 13 using TargeTron technology, constructing a strain that was no longer proteolytic on skim milk agar. Quantitative protease assays confirmed the absence of extracellular protease activity, which was restored by complementation with the wild-type ccp gene. The role of α-clostripain in virulence was assessed by analysing the isogenic wild-type, mutant and complemented strains in a mouse myonecrosis model. The results showed that although α-clostripain was the major extracellular protease, mutation of the ccp gene did not alter either the progression or the development of disease. These results do not rule out the possibility that this extracellular enzyme may still have a role in the early stages of the disease process.


Asunto(s)
Toxinas Bacterianas/metabolismo , Infecciones por Clostridium/enzimología , Clostridium perfringens/patogenicidad , Cisteína Endopeptidasas/metabolismo , Necrosis , Virulencia/genética , Animales , Supervivencia Celular , Células Cultivadas , Infecciones por Clostridium/genética , Infecciones por Clostridium/microbiología , Clostridium perfringens/enzimología , Clostridium perfringens/genética , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Femenino , Hemoglobinas/metabolismo , Proteínas Hemolisinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mutagénesis Insercional , Mutación/genética , Tasa de Supervivencia
19.
Infect Immun ; 79(6): 2145-53, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21402758

RESUMEN

Clostridium perfringens causes clostridial myonecrosis or gas gangrene and produces several extracellular hydrolytic enzymes and toxins, many of which are regulated by the VirSR signal transduction system. The revR gene encodes a putative orphan response regulator that has similarity to the YycF (WalR), VicR, PhoB, and PhoP proteins from other Gram-positive bacteria. RevR appears to be a classical response regulator, with an N-terminal receiver domain and a C-terminal domain with a putative winged helix-turn-helix DNA binding region. To determine its functional role, a revR mutant was constructed by allelic exchange and compared to the wild type by microarray analysis. The results showed that more than 100 genes were differentially expressed in the mutant, including several genes involved in cell wall metabolism. The revR mutant had an altered cellular morphology; unlike the short rods observed with the wild type, the mutant cells formed long filaments. These changes were reversed upon complementation with a plasmid that carried the wild-type revR gene. Several genes encoding extracellular hydrolytic enzymes (sialidase, hyaluronidase, and α-clostripain) were differentially expressed in the revR mutant. Quantitative enzyme assays confirmed that these changes led to altered enzyme activity and that complementation restored the wild-type phenotype. Most importantly, the revR mutant was attenuated for virulence in the mouse myonecrosis model compared to the wild type and the complemented strains. These results provide evidence that RevR regulates virulence in C. perfringens; it is the first response regulator other than VirR to be shown to regulate virulence in this important pathogen.


Asunto(s)
Clostridium perfringens/patogenicidad , Genes Bacterianos/fisiología , Factores de Virulencia/genética , Animales , Infecciones por Clostridium/microbiología , Clostridium perfringens/genética , Cisteína Endopeptidasas/metabolismo , Femenino , Genes Bacterianos/genética , Hialuronoglucosaminidasa/metabolismo , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Virulencia/fisiología
20.
Infect Immun ; 78(7): 3064-72, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20457789

RESUMEN

Clostridium perfringens causes several diseases in domestic livestock, including necrotic enteritis in chickens, which is of concern to the poultry industry due to its health implications and associated economic cost. The novel pore-forming toxin NetB is a critical virulence factor in the pathogenesis of this disease. In this study, we have examined the regulation of NetB toxin production. In C. perfringens, the quorum sensing-dependent VirSR two-component signal transduction system regulates genes encoding several toxins and extracellular enzymes. Analysis of the sequence upstream of the netB gene revealed the presence of potential DNA binding sites, or VirR boxes, that are recognized by the VirR response regulator. In vitro binding experiments showed that purified VirR was able to recognize and bind to these netB-associated VirR boxes. Furthermore, using a reporter gene assay, the netB VirR boxes were shown to be functional. Mutation of the virR gene in two avian C. perfringens strains was shown to significantly reduce the production of the NetB toxin; culture supernatants derived from these strains were no longer cytotoxic to Leghorn male hepatoma cells. Complementation with the virRS operon restored the toxin phenotypes to wild type. The results also showed that the VirSR two-component system regulates the expression of netB at the level of transcription. We postulate that in the gastrointestinal tract of infected birds, NetB production is upregulated when the population of C. perfringens cells reaches a threshold level that leads to activation of the VirSR system.


Asunto(s)
Toxinas Bacterianas/biosíntesis , Infecciones por Clostridium/microbiología , Clostridium perfringens/fisiología , Enterotoxinas/biosíntesis , Percepción de Quorum/fisiología , Factores de Virulencia/fisiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Pollos/microbiología , Clostridium perfringens/genética , Clostridium perfringens/metabolismo , Enteritis/microbiología , Enteritis/veterinaria , Enterotoxinas/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , Genes Bacterianos/fisiología , Masculino , Datos de Secuencia Molecular , Enfermedades de las Aves de Corral/microbiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...