Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Bacteriol ; : e0017524, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38953644

RESUMO

Clostridioides difficile causes a serious diarrheal disease and is a common healthcare-associated bacterial pathogen. Although it has a major impact on human health, the mechanistic details of C. difficile intestinal colonization remain undefined. C. difficile is highly sensitive to oxygen and requires anaerobic conditions for in vitro growth. However, the mammalian gut is not devoid of oxygen, and C. difficile tolerates moderate oxidative stress in vivo. The C. difficile genome encodes several antioxidant proteins, including a predicted superoxide reductase (SOR) that is upregulated upon exposure to antimicrobial peptides. The goal of this study was to establish SOR enzymatic activity and assess its role in protecting C. difficile against oxygen exposure. Insertional inactivation of sor rendered C. difficile more sensitive to superoxide, indicating that SOR contributes to antioxidant defense. Heterologous C. difficile sor expression in Escherichia coli conferred protection against superoxide-dependent growth inhibition, and the corresponding cell lysates showed superoxide scavenging activity. Finally, a C. difficile SOR mutant exhibited global proteome changes under oxygen stress when compared to the parent strain. Collectively, our data establish the enzymatic activity of C. difficile SOR, confirm its role in protection against oxidative stress, and demonstrate SOR's broader impacts on the C. difficile vegetative cell proteome.IMPORTANCEClostridioides difficile is an important pathogen strongly associated with healthcare settings and capable of causing severe diarrheal disease. While considered a strict anaerobe in vitro, C. difficile has been shown to tolerate low levels of oxygen in the mammalian host. Among other well-characterized antioxidant proteins, the C. difficile genome encodes a predicted superoxide reductase (SOR), an understudied component of antioxidant defense in pathogens. The significance of the research reported herein is the characterization of SOR's enzymatic activity, including confirmation of its role in protecting C. difficile against oxidative stress. This furthers our understanding of C. difficile pathogenesis and presents a potential new avenue for targeted therapies.

2.
PLoS Pathog ; 12(10): e1005946, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27741317

RESUMO

Clostridium difficile is a diarrheagenic pathogen associated with significant mortality and morbidity. While its glucosylating toxins are primary virulence determinants, there is increasing appreciation of important roles for non-toxin factors in C. difficile pathogenesis. Cell wall glycopolymers (CWGs) influence the virulence of various pathogens. Five C. difficile CWGs, including PSII, have been structurally characterized, but their biosynthesis and significance in C. difficile infection is unknown. We explored the contribution of a conserved CWG locus to C. difficile cell-surface integrity and virulence. Attempts at disrupting multiple genes in the locus, including one encoding a predicted CWG exporter mviN, were unsuccessful, suggesting essentiality of the respective gene products. However, antisense RNA-mediated mviN downregulation resulted in slight morphology defects, retarded growth, and decreased surface PSII deposition. Two other genes, lcpA and lcpB, with putative roles in CWG anchoring, could be disrupted by insertional inactivation. lcpA- and lcpB- mutants had distinct phenotypes, implying non-redundant roles for the respective proteins. The lcpB- mutant was defective in surface PSII deposition and shedding, and exhibited a remodeled cell surface characterized by elongated and helical morphology, aberrantly-localized cell septae, and an altered surface-anchored protein profile. Both lcpA- and lcpB- strains also displayed heightened virulence in a hamster model of C. difficile disease. We propose that gene products of the C. difficile CWG locus are essential, that they direct the production/assembly of key antigenic surface polysaccharides, and thereby have complex roles in virulence.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/ultraestrutura , Clostridioides difficile/patogenicidade , Clostridioides difficile/ultraestrutura , Infecções por Clostridium/virologia , Fatores de Virulência/metabolismo , Animais , Parede Celular/química , Cricetinae , Modelos Animais de Doenças , Imunofluorescência , Immunoblotting , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Mesocricetus , Microscopia Eletrônica , Mutagênese Insercional , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Polissacarídeos/química , Polissacarídeos/metabolismo , Virulência
3.
Infect Immun ; 83(3): 1139-49, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25561713

RESUMO

Attaching and effacing (A/E) pathogens adhere intimately to intestinal enterocytes and efface brush border microvilli. A key virulence strategy of A/E pathogens is the type III secretion system (T3SS)-mediated delivery of effector proteins into host cells. The secreted protein EspZ is postulated to promote enterocyte survival by regulating the T3SS and/or by modulating epithelial signaling pathways. To explore the role of EspZ in A/E pathogen virulence, we generated an isogenic espZ deletion strain (ΔespZ) and corresponding cis-complemented derivatives of rabbit enteropathogenic Escherichia coli and compared their abilities to regulate the T3SS and influence host cell survival in vitro. For virulence studies, rabbits infected with these strains were monitored for bacterial colonization, clinical signs, and intestinal tissue alterations. Consistent with data from previous reports, espZ-transfected epithelial cells were refractory to infection-dependent effector translocation. Also, the ΔespZ strain induced greater host cell death than did the parent and complemented strains. In rabbit infections, fecal ΔespZ strain levels were 10-fold lower than those of the parent strain at 1 day postinfection, while the complemented strain was recovered at intermediate levels. In contrast to the parent and complemented mutants, ΔespZ mutant fecal carriage progressively decreased on subsequent days. ΔespZ mutant-infected animals gained weight steadily over the infection period, failed to show characteristic disease symptoms, and displayed minimal infection-induced histological alterations. Terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) staining of intestinal sections revealed increased epithelial cell apoptosis on day 1 after infection with the ΔespZ strain compared to animals infected with the parent or complemented strains. Thus, EspZ-dependent host cell cytoprotection likely prevents epithelial cell death and sloughing and thereby promotes bacterial colonization.


Assuntos
Enterócitos/microbiologia , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Microvilosidades/microbiologia , Animais , Apoptose , Carga Bacteriana , Sistemas de Secreção Bacterianos/genética , Enterócitos/patologia , Escherichia coli Enteropatogênica/metabolismo , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/metabolismo , Fezes/microbiologia , Deleção de Genes , Expressão Gênica , Teste de Complementação Genética , Interações Hospedeiro-Patógeno , Humanos , Masculino , Microvilosidades/patologia , Coelhos , Virulência
4.
Nature ; 458(7242): 1176-9, 2009 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-19252482

RESUMO

Clostridium difficile is the leading cause of infectious diarrhoea in hospitals worldwide, because of its virulence, spore-forming ability and persistence. C. difficile-associated diseases are induced by antibiotic treatment or disruption of the normal gastrointestinal flora. Recently, morbidity and mortality resulting from C. difficile-associated diseases have increased significantly due to changes in the virulence of the causative strains and antibiotic usage patterns. Since 2002, epidemic toxinotype III NAP1/027 strains, which produce high levels of the major virulence factors, toxin A and toxin B, have emerged. These toxins have 63% amino acid sequence similarity and are members of the large clostridial glucosylating toxin family, which are monoglucosyltransferases that are pro-inflammatory, cytotoxic and enterotoxic in the human colon. Inside host cells, both toxins catalyse the transfer of glucose onto the Rho family of GTPases, leading to cell death. However, the role of these toxins in the context of a C. difficile infection is unknown. Here we describe the construction of isogenic tcdA and tcdB (encoding toxin A and B, respectively) mutants of a virulent C. difficile strain and their use in the hamster disease model to show that toxin B is a key virulence determinant. Previous studies showed that purified toxin A alone can induce most of the pathology observed after infection of hamsters with C. difficile and that toxin B is not toxic in animals unless it is co-administered with toxin A, suggesting that the toxins act synergistically. Our work provides evidence that toxin B, not toxin A, is essential for virulence. Furthermore, it is clear that the importance of these toxins in the context of infection cannot be predicted exclusively from studies using purified toxins, reinforcing the importance of using the natural infection process to dissect the role of toxins in disease.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/patogenicidade , Animais , Aderência Bacteriana , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/biossíntese , Toxinas Bacterianas/genética , Toxinas Bacterianas/farmacologia , Linhagem Celular , Clostridioides difficile/genética , Cricetinae , Modelos Animais de Doenças , Enterotoxinas/genética , Enterotoxinas/metabolismo , Humanos , Mutação , Virulência
5.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G374-80, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24904077

RESUMO

The diarrheagenic pathogen enteropathogenic Escherichia coli (EPEC) dynamically modulates the survival of infected host intestinal epithelial cells. In the initial stages of infection, several prosurvival signaling events are activated in host cells. These include the phosphorylation of epidermal growth factor receptor (EGFR) and the consequent activation of the phosphatidylinositol-3 kinase/Akt pathway. While studying this pathway in infected epithelial cells, we observed EGFR depletion at later stages of infection, followed subsequently by a decrease in phospho-EGFR. EGFR loss was not dependent on receptor phosphorylation, or on canonical proteasome- and lysosome-dependent processes. Although a type III secretion mutant (ΔescN) stimulated EGFR phosphorylation, it failed to induce receptor degradation. To identify the specific EPEC effector molecule(s) that influenced EGFR stability, epithelial cells infected with isogenic mutant EPEC strains were examined. An EPEC ΔespF strain failed to induce EGFR degradation, whereas EPEC ΔespZ accentuated receptor loss in infected cells. Given the known and contrasting effects of EspF and EspZ on caspase activation, and the known role of proteases in cleaving EGFR, we explored the effect of caspase inhibitors on infection-dependent EGFR loss. The pan-caspase inhibitor Q-VD-OPh blocked EPEC-induced EGFR cleavage in a dose-dependent manner. Taken together, our data suggest that EPEC EspF stimulates caspase-dependent EGFR cleavage and loss, whereas EspZ inhibits this process. Whereas EGFR phosphorylation contributes to the survival of host cells early in infection, EspF-driven caspase activation and consequent EGFR loss likely induce a precipitous increase in host cell death later in the infectious process.


Assuntos
Escherichia coli Enteropatogênica/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Receptores ErbB/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Transdução de Sinais , Células CACO-2 , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Inibidores de Caspase/farmacologia , Morte Celular , Relação Dose-Resposta a Droga , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Receptores ErbB/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Intestinos/efeitos dos fármacos , Intestinos/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Mutação , Fosforilação , Estabilidade Proteica , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
6.
Gut Microbes ; 16(1): 2333483, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38532703

RESUMO

Although the role of the intestinal microbiota in the pathogenesis of inflammatory bowel disease (IBD) is beyond debate, attempts to verify the causative role of IBD-associated dysbiosis have been limited to reports of promoting the disease in genetically susceptible mice or in chemically induced colitis. We aimed to further test the host response to fecal microbiome transplantation (FMT) from Crohn's disease patients on mucosal homeostasis in ex-germ-free (xGF) mice. We characterized and transferred fecal microbiota from healthy patients and patients with defined Crohn's ileocolitis (CD_L3) to germ-free mice and analyzed the resulting microbial and mucosal homeostasis by 16S profiling, shotgun metagenomics, histology, immunofluorescence (IF) and RNAseq analysis. We observed a markedly reduced engraftment of CD_L3 microbiome compared to healthy control microbiota. FMT from CD_L3 patients did not lead to ileitis but resulted in colitis with features consistent with CD: a discontinued pattern of colitis, more proximal colonic localization, enlarged isolated lymphoid follicles and/or tertiary lymphoid organ neogenesis, and a transcriptomic pattern consistent with epithelial reprograming and promotion of the Paneth cell-like signature in the proximal colon and immune dysregulation characteristic of CD. The observed inflammatory response was associated with persistently increased abundance of Ruminococcus gnavus, Erysipelatoclostridium ramosum, Faecalimonas umbilicate, Blautia hominis, Clostridium butyricum, and C. paraputrificum and unexpected growth of toxigenic C. difficile, which was below the detection level in the community used for inoculation. Our study provides the first evidence that the transfer of a dysbiotic community from CD patients can lead to spontaneous inflammatory changes in the colon of xGF mice and identifies a signature microbial community capable of promoting colonization of pathogenic and conditionally pathogenic bacteria.


Assuntos
Clostridioides difficile , Colite , Doença de Crohn , Microbioma Gastrointestinal , Microbiota , Humanos , Camundongos , Animais , Doença de Crohn/microbiologia , Transplante de Microbiota Fecal , Disbiose/microbiologia
7.
J Infect Dis ; 205(1): 128-33, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22124129

RESUMO

Toxin A has historically been regarded as the primary virulence determinant in Clostridium difficile infection, but naturally occurring toxin A-negative, toxin B-positive (A-/B+) C. difficile strains are known to be virulent. To determine the role of toxin B in these strains, we immunized hamsters with a toxoid prepared from purified toxin B to determine whether they would be protected from lethal challenge with an A-/B+ strain of C. difficile.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/imunologia , Infecções por Clostridium/imunologia , Enterotoxinas/metabolismo , Toxoides/imunologia , Vacinação , Fatores de Virulência/metabolismo , Animais , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Cricetinae , Toxoides/administração & dosagem
8.
Front Med (Lausanne) ; 10: 1238159, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37928470

RESUMO

Background: Clostridioides difficile Infection (CDI) is a healthcare-associated diarrheal disease prevalent worldwide. A common diagnostic algorithm relies on a two-step protocol that employs stool enzyme immunoassays (EIAs) to detect the pathogen, and its toxins, respectively. Active CDI is deemed less likely when the Toxin EIA result is negative, even if the pathogen-specific EIA is positive for C. difficile. We recently reported, however, that low-toxin-producing C. difficile strains recovered from Toxin-negative ('discrepant') clinical stool specimens can be fully pathogenic, and cause lethality in a rodent CDI model. To document frequency of discrepant CDI specimens, and evaluate C. difficile strain diversity, we performed longitudinal surveillance at a Southern Arizona tertiary-care hospital. Methods: Diarrheic stool specimens from patients with clinical suspicion of CDI were obtained over an eight-year period (2015-2022) from all inpatient and outpatient Units of a > 600-bed Medical Center in Southern Arizona. Clinical laboratory EIA testing identified C. difficile-containing specimens, and classified them as Toxin-positive or Toxin-negative. C. difficile isolates recovered from the stool specimens were DNA fingerprinted using an international phylogenetic lineage assignment system ("ribotyping"). For select isolates, toxin abundance in stationary phase supernatants of pure cultures was quantified via EIA. Results: Of 8,910 diarrheic specimens that underwent diagnostic testing, 1733 (19.4%) harbored C. difficile. Our major findings were that: (1) C. difficile prevalence and phylogenetic diversity was stable over the 8-year period; (2) toxigenic C. difficile was recovered from 69% of clinically Tox-neg ('discrepant') specimens; (3) the six most prevalent USA ribotypes were recovered in significant proportions (>60%) from Tox-neg specimens; and (4) toxin-producing C. difficile recovered from discrepant specimens produced less toxin than strains of the same ribotype isolated from non-discrepant specimens. Conclusion: Our study highlights the dominance of Toxin EIA-negative CDI specimens in a clinical setting and the high frequency of known virulent ribotypes in these specimens. Therefore, a careful reevaluation of the clinical relevance of diagnostically-discrepant specimens particularly in the context of missed CDI diagnoses and C. difficile persistence, is warranted.

9.
Microbiome ; 11(1): 169, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37533066

RESUMO

BACKGROUND: Upper small intestinal dietary lipids activate a gut-brain axis regulating energy homeostasis. The prebiotic, oligofructose (OFS) improves body weight and adiposity during metabolic dysregulation but the exact mechanisms remain unknown. This study examines whether alterations to the small intestinal microbiota following OFS treatment improve small intestinal lipid-sensing to regulate food intake in high fat (HF)-fed rats. RESULTS: In rats fed a HF diet for 4 weeks, OFS supplementation decreased food intake and meal size within 2 days, and reduced body weight and adiposity after 6 weeks. Acute (3 day) OFS treatment restored small intestinal lipid-induced satiation during HF-feeding, and was associated with increased small intestinal CD36 expression, portal GLP-1 levels and hindbrain neuronal activation following a small intestinal lipid infusion. Transplant of the small intestinal microbiota from acute OFS treated donors into HF-fed rats also restored lipid-sensing mechanisms to lower food intake. 16S rRNA gene sequencing revealed that both long and short-term OFS altered the small intestinal microbiota, increasing Bifidobacterium relative abundance. Small intestinal administration of Bifidobacterium pseudolongum to HF-fed rats improved small intestinal lipid-sensing to decrease food intake. CONCLUSION: OFS supplementation rapidly modulates the small intestinal gut microbiota, which mediates improvements in small intestinal lipid sensing mechanisms that control food intake to improve energy homeostasis. Video Abstract.


Assuntos
Microbioma Gastrointestinal , Ratos , Animais , RNA Ribossômico 16S/genética , Obesidade/metabolismo , Peso Corporal , Gorduras na Dieta , Dieta Hiperlipídica/efeitos adversos
10.
Infect Immun ; 80(11): 3850-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22907816

RESUMO

The diarrheagenic pathogen enteropathogenic Escherichia coli (EPEC) limits the death of infected enterocytes early in infection. A number of bacterial molecules and host signaling pathways contribute to the enhanced survival of EPEC-infected host cells. EspZ, a type III secreted effector protein that is unique to EPEC and related "attaching and effacing" (A/E) pathogens, plays a role in limiting host cell death, but the precise host signaling pathways responsible for this phenotype are not known. We hypothesized that EspZ contributes to the survival of infected intestinal epithelial cells by interfering with apoptosis. Consistent with previous studies, scanning electron microscopy analysis of intestinal epithelial cells infected with an EPEC espZ mutant (ΔespZ) showed increased levels of apoptotic and necrotic cells compared to cells infected with the isogenic parent strain. Correspondingly, higher levels of cytosolic cytochrome c and increased activation of caspases 9, 7, and 3 were observed for ΔespZ strain-infected cells compared to wild-type (WT) EPEC-infected cells. Finally, espZ-transfected epithelial cells were significantly protected from staurosporine-induced, but not tumor necrosis factor alpha (TNF-α)/cycloheximide-induced, apoptosis. Thus, EspZ contributes to epithelial cell survival by mechanisms that include the inhibition of the intrinsic apoptotic pathway. The enhanced survival of infected enterocytes by molecules such as EspZ likely plays a key role in optimal colonization by A/E pathogens.


Assuntos
Apoptose/fisiologia , Escherichia coli Enteropatogênica/metabolismo , Células Epiteliais/metabolismo , Proteínas de Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Células Cultivadas , Células Epiteliais/microbiologia , Proteínas de Escherichia coli/genética , Humanos , Intestinos/microbiologia , Transdução de Sinais
11.
Anaerobe ; 18(6): 614-20, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23017940

RESUMO

Clostridium difficile is a leading cause of hospital-acquired bacterial infections in the United States, and the increased incidence of recurrent C. difficile infections is particularly problematic. The molecular mechanisms of C. difficile colonization, including its ability to evade host innate immune responses, is poorly understood. We hypothesized that epidemic-associated C. difficile clinical isolates would exhibit increased resistance to mammalian, gut-associated, cationic antimicrobial peptides such as the cathelicidin LL-37. Standardized susceptibility tests as well as comparative proteomic analyses revealed that C. difficile strains varied in their responses to LL-37, with epidemic-associated 027 ribotype isolates displaying greater resistance. Further, exposure of C. difficile strains to sub-lethal concentrations of LL-37 resulted in increased resistance to subsequent peptide challenge, suggesting the presence of inducible resistance mechanisms. Correspondingly, LL-37 exposure altered the C. difficile proteome, with marked changes in abundance of cell wall biosynthesis proteins, surface layer proteins, ABC transporters and lysine metabolism pathway components. Taken together, these results suggest that innate immune avoidance mechanisms could facilitate robust colonization by C. difficile.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Clostridioides difficile/química , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/microbiologia , Proteoma/análise , Clostridioides difficile/isolamento & purificação , Infecções por Clostridium/imunologia , Farmacorresistência Bacteriana , Humanos , Evasão da Resposta Imune , Testes de Sensibilidade Microbiana , Estados Unidos
12.
Curr Opin Microbiol ; 66: 86-91, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35124522

RESUMO

Clostridioides difficile is a leading cause of the healthcare-associated disease C. difficile infection (CDI), which has an annual US burden of over 200 000 cases. CDI mitigation strategies have been complicated by the emergence, and widespread distribution, of phylogenetically diverse lineages, as well as pathogen recalcitrance to genetic manipulation. In this review, we highlight past and current efforts to elucidate C. difficile surface glycopolymer biology since these molecules are essential for colonization, disease, and immunity elicitation, and may therefore have potential as CDI anti-infective targets.


Assuntos
Clostridioides difficile , Infecção Hospitalar , Clostridioides , Clostridioides difficile/genética , Humanos
13.
Front Microbiol ; 13: 871152, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35633701

RESUMO

The alternative sigma factor SigL (Sigma-54) facilitates bacterial adaptation to the extracellular environment by modulating the expression of defined gene subsets. A homolog of the gene encoding SigL is conserved in the diarrheagenic pathogen Clostridioides difficile. To explore the contribution of SigL to C. difficile biology, we generated sigL-disruption mutants (sigL::erm) in strains belonging to two phylogenetically distinct lineages-the human-relevant Ribotype 027 (strain BI-1) and the veterinary-relevant Ribotype 078 (strain CDC1). Comparative proteomics analyses of mutants and isogenic parental strains revealed lineage-specific SigL regulons. Concomitantly, loss of SigL resulted in pleiotropic and distinct phenotypic alterations in the two strains. Sporulation kinetics, biofilm formation, and cell surface-associated phenotypes were altered in CDC1 sigL::erm relative to the isogenic parent strain but remained unchanged in BI-1 sigL::erm. In contrast, secreted toxin levels were significantly elevated only in the BI-1 sigL::erm mutant relative to its isogenic parent. We also engineered SigL overexpressing strains and observed enhanced biofilm formation in the CDC1 background, and reduced spore titers as well as dampened sporulation kinetics in both strains. Thus, we contend that SigL is a key, pleiotropic regulator that dynamically influences C. difficile's virulence factor landscape, and thereby, its interactions with host tissues and co-resident microbes.

14.
Emerg Microbes Infect ; 11(1): 1982-1993, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35880487

RESUMO

Clostridioides difficile is a leading cause of healthcare-associated infections worldwide. Currently, there is a lack of consensus for an optimal diagnostic method for C. difficile infection (CDI). Multi-step diagnostic algorithms use enzyme immunosorbent analysis (EIA)-based detection of C. difficile toxins TcdA/TcdB in stool, premised on the rationale that EIA toxin-negative (Tox-) patients have less severe disease and shorter diarrhoea duration. The aim of this study was to characterize toxigenic (i.e. tcdA/tcdB-positive) C. difficile strains isolated from diarrheic patient stool with an EIA Tox- (i.e. "discrepant") CDI diagnostic test result. Recovered strains were DNA fingerprinted (ribotyped), subjected to multiple toxin, genome and proteome evaluations, and assessed for virulence. Overall, of 1243 C. difficile-positive patient stool specimens from Southern Arizona hospitals, 31% were discrepant. For RT027 (the most prevalent ribotype)-containing specimens, 34% were discrepant; the corresponding RT027 isolates were cytotoxic to cultured fibroblasts, but their total toxin levels were comparable to, or lower than, the historic low-toxin-producing C. difficile strain CD630. Nevertheless, these low-toxin RT027 strains (LT-027) exhibited similar lethality to a clade-matched high-toxin RT027 strain in Golden Syrian hamsters, and heightened colonization and persistence in mice. Genomics and proteomics analyses of LT-027 strains identified unique genes and altered protein abundances, respectively, relative to high-toxin RT027 strains. Collectively, our data highlight the robust virulence of LT-027 C. difficile, provide a strong argument for reconsidering the clinical significance of a Tox- EIA result, and underscore the potential limitations of current diagnostic protocols.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Infecções por Clostridium , Animais , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Clostridioides , Clostridioides difficile/genética , Camundongos , Virulência
15.
Gut Microbes ; 14(1): 2143224, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36476073

RESUMO

The diarrheagenic pathogen enteropathogenic Escherichia coli is responsible for significant childhood mortality and morbidity. EPEC and related attaching-and-effacing (A/E) pathogens use a type III secretion system to hierarchically deliver effector proteins into host cells and manipulate epithelial structure and function. Subversion of host mitochondrial biology is a key aspect of A/E pathogen virulence strategy, but the mechanisms remain poorly defined. We demonstrate that the early-secreted effector EspZ and the late-secreted effector EspH have contrasting effects on host mitochondrial structure and function. EspZ interacts with FIS1, a protein that induces mitochondrial fragmentation and mitophagy. Infection of epithelial cells with either wildtype EPEC or an isogenic espZ deletion mutant (ΔespZ) robustly upregulated FIS1 abundance, but a marked increase in mitochondrial fragmentation and mitophagy was seen only in ΔespZ-infected cells. FIS1-depleted cells were protected against ΔespZ-induced fission, and EspZ-expressing transfected epithelial cells were protected against pharmacologically induced mitochondrial fission and membrane potential disruption. Thus, EspZ interacts with FIS1 and blocks mitochondrial fragmentation and mitophagy. In contrast to WT EPEC, ΔespH-infected epithelial cells had minimal FIS1 upregulation and exhibited hyperfused mitochondria. Consistent with the contrasting impacts on organelle shape, mitochondrial membrane potential was preserved in ΔespH-infected cells, but profoundly disrupted in ΔespZ-infected cells. Collectively, our studies reveal hitherto unappreciated roles for two essential EPEC virulence factors in the temporal and dynamic regulation of host mitochondrial biology.


Bacterial pathogens strategically manipulate host cell structures and functions during the process of colonization and expansion, and this eventually contributes to disease symptoms. The diarrhea-causing pathogen enteropathogenic Escherichia coli (EPEC) secretes proteins into host cells to alter their behavior. Two secreted proteins, EspZ and EspH, were previously shown to be essential for causing disease in animal models. In this study, we demonstrate that interplay between EspZ/EspH and host factors modulates the structure and function of host cell mitochondria. Among their various roles, mitochondria generate energy, produce important biomolecules, and protect cells from damage. EPEC infection of epithelial cells results in increased abundance of a key mitochondrial outer-membrane protein, FIS1. FIS1 plays a housekeeping role by breaking down unhealthy mitochondria and targeting them for elimination from cells. In the early stages of infection, EspZ interacts with FIS1 and blocks its action, thereby protecting the host mitochondrial network and consequently, enhancing host cell viability. Our studies are consistent with a model wherein EspZ-dependent preservation of mitochondrial integrity early in infection allows for bacterial colonization. Later in infection, however, EspH-dependent increase in FIS1 results in significant mitochondrial fragmentation and host cell death; this likely facilitates pathogen dispersal. Taken together, EspZ and EspH dynamically impact host biology, and consequently, infection outcomes. Overall, an appreciation of the mechanisms by which EspZ and EspH manipulate host cells could eventually lead to host-directed interventions for EPEC diarrhea, which is currently not vaccine-preventable.


Assuntos
Escherichia coli Enteropatogênica , Microbioma Gastrointestinal , Escherichia coli Enteropatogênica/genética
16.
Urolithiasis ; 51(1): 15, 2022 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-36507964

RESUMO

Kidney stone cultures can be beneficial in identifying bacteria not detected in urine, yet how stone cultures are performed among endourologists, under what conditions, and by what laboratory methods remain largely unknown. Stone cultures are not addressed by current clinical guidelines. A comprehensive REDCap electronic survey sought responses from directed (n = 20) and listserv elicited (n = 108) endourologists specializing in kidney stone disease. Questions included which clinical scenarios prompt a stone culture order, how results influence post-operative antibiotics, and what microbiology lab protocols exist at each institution with respect to processing and resulting stone cultures. Logistic regression statistical analysis determined what factors were associated with performing stone cultures. Of 128 unique responses, 11% identified as female and the mean years of practicing was 16 (range 1-46). A specific 'stone culture' order was available to only 50% (64/128) of those surveyed, while 32% (41/128) reported culturing stone by placing a urine culture order. The duration of antibiotics given for a positive stone culture varied, with 4-7 days (46%) and 8-14 days (21%) the most reported. More years in practice was associated with fewer stone cultures ordered, while higher annual volume of percutaneous nephrolithotomy was associated with ordering more stone cultures (p < 0.01). Endourologists have differing practice patterns with respect to ordering stone cultures and utilizing the results to guide post-operative antibiotics. With inconsistent microbiology lab stone culture protocols across multiple institutions, more uniform processing is needed for future studies to assess the clinical benefit of stone cultures and direct future guidelines.


Assuntos
Cálculos Renais , Nefrolitotomia Percutânea , Feminino , Humanos , Nefrolitotomia Percutânea/métodos , Cálculos Renais/urina , Urinálise , Bactérias , Estudos Multicêntricos como Assunto
17.
J Bacteriol ; 192(19): 4904-11, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20675495

RESUMO

Toxigenic Clostridium difficile strains produce two toxins (TcdA and TcdB) during the stationary phase of growth and are the leading cause of antibiotic-associated diarrhea. C. difficile isolates of the molecular type NAP1/027/BI have been associated with severe disease and hospital outbreaks worldwide. It has been suggested that these "hypervirulent" strains produce larger amounts of toxin and that a mutation in a putative negative regulator (TcdC) allows toxin production at all growth phases. To rigorously explore this possibility, we conducted a quantitative examination of the toxin production of multiple hypervirulent and nonhypervirulent C. difficile strains. Toxin gene (tcdA and tcdB) and toxin gene regulator (tcdR and tcdC) expression was also monitored. To obtain additional correlates for the hypervirulence phenotype, sporulation kinetics and efficiency were measured. In the exponential phase, low basal levels of tcdA, tcdB, and tcdR expression were evident in both hypervirulent and nonhypervirulent strains, but contrary to previous assumptions, toxin levels were below the detectable thresholds. While hypervirulent strains displayed robust toxin production during the stationary phase of growth, the amounts were not significantly different from those of the nonhypervirulent strains tested; further, total toxin amounts were directly proportional to tcdA, tcdB, and tcdR gene expression. Interestingly, tcdC expression did not diminish in stationary phase, suggesting that TcdC may have a modulatory rather than a strictly repressive role. Comparative genomic analyses of the closely related nonhypervirulent strains VPI 10463 (the highest toxin producer) and 630 (the lowest toxin producer) revealed polymorphisms in the tcdR ribosome binding site and the tcdR-tcdB intergenic region, suggesting that a mechanistic basis for increased toxin production in VPI 10463 could be increased TcdR translation and read-through transcription of the tcdA and tcdB genes. Hypervirulent isolates produced significantly more spores, and did so earlier, than all other isolates. Increased sporulation, potentially in synergy with robust toxin production, may therefore contribute to the widespread disease now associated with hypervirulent C. difficile strains.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/metabolismo , Enterotoxinas/metabolismo , Proteínas Repressoras/metabolismo , Esporos Bacterianos/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Clostridioides difficile/genética , Enterotoxinas/genética , Ensaio de Imunoadsorção Enzimática , Humanos , Proteínas Repressoras/genética , Esporos Bacterianos/genética , Virulência/genética , Virulência/fisiologia
18.
Anaerobe ; 16(6): 614-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20934524

RESUMO

Poultry necrotic enteritis (NE) is caused by specific strains of Clostridium perfringens, most of which are type A. The role of alpha toxin (CPA) in NE has been called into question by the finding that an engineered cpa mutant retains full virulence in vivo[9]. This is in contrast to the finding that immunization with CPA toxoids protects against NE. We confirmed the earlier findings, in that 14-day-old Cornish × Rock broiler chicks challenged with a cpa mutant developed lesions compatible with NE in >90% of birds inoculated with the mutant. However, CPA was detected in amounts ranging from 10 to >100 ng per g of gut contents and mucosa in birds inoculated with the cpa mutant, the wildtype strain from which the mutant was constructed, and our positive control strain. There was a direct relationship between lesion severity and amount of CPA detected (R = 0.89-0.99). These findings suggest that the role of CPA in pathogenesis of NE requires further investigation.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas de Ligação ao Cálcio/toxicidade , Clostridium perfringens/genética , Clostridium perfringens/patogenicidade , Enterite/veterinária , Intestinos/microbiologia , Proteínas Mutantes/toxicidade , Doenças das Aves Domésticas/microbiologia , Fosfolipases Tipo C/toxicidade , Animais , Toxinas Bacterianas/genética , Aves , Proteínas de Ligação ao Cálcio/genética , Galinhas , Enterite/microbiologia , Histocitoquímica , Intestinos/patologia , Proteínas Mutantes/genética , Fosfolipases Tipo C/genética
19.
Gut Microbes ; 12(1): 1802865, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33092487

RESUMO

Clostridioides difficile infection (CDI) is a common healthcare- and antibiotic-associated diarrheal disease. If mis-diagnosed, or incompletely treated, CDI can have serious, indeed fatal, consequences. The clinical and economic burden imposed by CDI is great, and the US Centers for Disease Control and Prevention has named the causative agent, C. difficile (CD), as an Urgent Threat To US healthcare. CDI is also a significant problem in the agriculture industry. Currently, there are no FDA-approved preventives for this disease, and the only approved treatments for both human and veterinary CDI involve antibiotic use, which, ironically, is associated with disease relapse and the threat of burgeoning antibiotic resistance. Research efforts in multiple laboratories have demonstrated that non-toxin factors also play key roles in CDI, and that these are critical for disease. Specifically, key CD adhesins, as well as other surface-displayed factors have been shown to be major contributors to host cell attachment, and as such, represent attractive targets for anti-CD interventions. However, research on anti-virulence approaches has been more limited, primarily due to the lack of genetic tools, and an as-yet nascent (but increasingly growing) appreciation of immunological impacts on CDI. The focus of this review is the conceptualization and development of specific anti-virulence strategies to combat CDI. Multiple laboratories are focused on this effort, and the field is now at an exciting stage with numerous products in development. Herein, however, we focus only on select technologies (Figure 1) that have advanced near, or beyond, pre-clinical testing (not those that are currently in clinical trial), and discuss roadblocks associated with their development and implementation.


Assuntos
Clostridioides difficile/patogenicidade , Infecções por Clostridium/tratamento farmacológico , Diarreia/tratamento farmacológico , Animais , Antibacterianos/uso terapêutico , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/genética , Clostridioides difficile/fisiologia , Infecções por Clostridium/microbiologia , Diarreia/microbiologia , Humanos , Virulência/efeitos dos fármacos
20.
Sci Rep ; 10(1): 22135, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33335199

RESUMO

Clostridioides difficile infection (CDI) is a major healthcare-associated diarrheal disease. Consistent with trends across the United States, C. difficile RT106 was the second-most prevalent molecular type in our surveillance in Arizona from 2015 to 2018. A representative RT106 strain displayed robust virulence and 100% lethality in the hamster model of acute CDI. We identified a unique 46 KB genomic island (GI1) in all RT106 strains sequenced to date, including those in public databases. GI1 was not found in its entirety in any other C. difficile clade, or indeed, in any other microbial genome; however, smaller segments were detected in Enterococcus faecium strains. Molecular clock analyses suggested that GI1 was horizontally acquired and sequentially assembled over time. GI1 encodes homologs of VanZ and a SrtB-anchored collagen-binding adhesin, and correspondingly, all tested RT106 strains had increased teicoplanin resistance, and a majority displayed collagen-dependent biofilm formation. Two additional genomic islands (GI2 and GI3) were also present in a subset of RT106 strains. All three islands are predicted to encode mobile genetic elements as well as virulence factors. Emergent phenotypes associated with these genetic islands may have contributed to the relatively rapid expansion of RT106 in US healthcare and community settings.


Assuntos
Clostridioides difficile/classificação , Clostridioides difficile/genética , Genoma Bacteriano , Ilhas Genômicas , Genômica , Fenótipo , Filogenia , Ribotipagem , Animais , Antibacterianos/farmacologia , Arizona/epidemiologia , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/isolamento & purificação , Infecções por Clostridium/epidemiologia , Infecções por Clostridium/microbiologia , Cricetinae , Infecção Hospitalar/epidemiologia , Farmacorresistência Bacteriana , Variação Genética , Genômica/métodos , Genótipo , Humanos , Testes de Sensibilidade Microbiana , Prevalência , Vigilância em Saúde Pública , Ribotipagem/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa