Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Chem Biol ; 19(5): 556-564, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36894723

RESUMO

Anaerobic microbial metabolism drives critical functions within global ecosystems, host-microbiota interactions, and industrial applications, yet remains ill-defined. Here we advance a versatile approach to elaborate cellular metabolism in obligate anaerobes using the pathogen Clostridioides difficile, an amino acid and carbohydrate-fermenting Clostridia. High-resolution magic angle spinning nuclear magnetic resonance (NMR) spectroscopy of C. difficile, grown with fermentable 13C substrates, informed dynamic flux balance analysis (dFBA) of the pathogen's genome-scale metabolism. Analyses identified dynamic recruitment of oxidative and supporting reductive pathways, with integration of high-flux amino acid and glycolytic metabolism at alanine's biosynthesis to support efficient energy generation, nitrogen handling and biomass generation. Model predictions informed an approach leveraging the sensitivity of 13C NMR spectroscopy to simultaneously track cellular carbon and nitrogen flow from [U-13C]glucose and [15N]leucine, confirming the formation of [13C,15N]alanine. Findings identify metabolic strategies used by C. difficile to support its rapid colonization and expansion in gut ecosystems.


Assuntos
Clostridioides difficile , Anaerobiose , Ecossistema , Espectroscopia de Ressonância Magnética/métodos , Aminoácidos , Alanina
2.
Clin Infect Dis ; 76(3): e540-e543, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35686436

RESUMO

We enrolled arriving international air travelers in a severe acute respiratory syndrome coronavirus 2 genomic surveillance program. We used molecular testing of pooled nasal swabs and sequenced positive samples for sublineage. Traveler-based surveillance provided early-warning variant detection, reporting the first US Omicron BA.2 and BA.3 in North America.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Aeroportos , COVID-19/diagnóstico , Genômica
3.
Cell Host Microbe ; 29(11): 1709-1723.e5, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34637780

RESUMO

We present predictive models for comprehensive systems analysis of Clostridioides difficile, the etiology of pseudomembranous colitis. By leveraging 151 published transcriptomes, we generated an EGRIN model that organizes 90% of C. difficile genes into a transcriptional regulatory network of 297 co-regulated modules, implicating genes in sporulation, carbohydrate transport, and metabolism. By advancing a metabolic model through addition and curation of metabolic reactions including nutrient uptake, we discovered 14 amino acids, diverse carbohydrates, and 10 metabolic genes as essential for C. difficile growth in the intestinal environment. Finally, we developed a PRIME model to uncover how EGRIN-inferred combinatorial gene regulation by transcription factors, such as CcpA and CodY, modulates essential metabolic processes to enable C. difficile growth relative to commensal colonization. The C. difficile interactive web portal provides access to these model resources to support collaborative systems-level studies of context-specific virulence mechanisms in C. difficile.


Assuntos
Clostridioides difficile , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Clostridioides , Clostridioides difficile/genética , Regulação Bacteriana da Expressão Gênica , Redes e Vias Metabólicas/genética , Análise de Sistemas
4.
Cell Host Microbe ; 29(11): 1693-1708.e7, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34637781

RESUMO

Leveraging systems biology approaches, we illustrate how metabolically distinct species of Clostridia protect against or worsen Clostridioides difficile infection in mice by modulating the pathogen's colonization, growth, and virulence to impact host survival. Gnotobiotic mice colonized with the amino acid fermenter Paraclostridium bifermentans survive infection with reduced disease severity, while mice colonized with the butyrate-producer, Clostridium sardiniense, succumb more rapidly. Systematic in vivo analyses revealed how each commensal alters the gut-nutrient environment to modulate the pathogen's metabolism, gene regulatory networks, and toxin production. Oral administration of P. bifermentans rescues conventional, clindamycin-treated mice from lethal C. difficile infection in a manner similar to that of monocolonized animals, thereby supporting the therapeutic potential of this commensal species. Our findings lay the foundation for mechanistically informed therapies to counter C. difficile disease using systems biology approaches to define host-commensal-pathogen interactions in vivo.


Assuntos
Clostridiales/fisiologia , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Infecções por Clostridium/terapia , Clostridium/fisiologia , Simbiose , Aminoácidos/metabolismo , Animais , Arginina/metabolismo , Butiratos/metabolismo , Ceco/metabolismo , Ceco/microbiologia , Clostridiales/crescimento & desenvolvimento , Clostridioides difficile/genética , Clostridioides difficile/fisiologia , Clostridium/crescimento & desenvolvimento , Fermentação , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Redes Reguladoras de Genes , Vida Livre de Germes , Camundongos , Índice de Gravidade de Doença , Biologia de Sistemas , Virulência
5.
Anaerobe ; 59: 1-7, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31077800

RESUMO

Clostridioides difficile is a Gram-positive, anaerobic bacterium. It is known that C. difficile is one of the major causes of antibiotic associated diarrhea. The enhanced antibiotic resistance observed in C. difficile is the result of highly resistant spores produced by the bacterium. In Bacillus subtilis, the sin operon is involved in sporulation inhibition. Two proteins coded within this operon, SinR and SinI, have an antagonistic relationship; SinR acts as an inhibitor to sporulation whereas SinI represses the activity of SinR, thus allowing the bacterium to sporulate. In a previous study, we examined the sin locus in C. difficile and named the two genes associated with this operon sinR and sinR', analogous to sinR and sinI in B. subtilis, respectively. We have shown that SinR and SinR' have pleiotropic roles in pathogenesis pathways and interact antagonistically with each other. Unlike B. subtilis SinI, SinR' in C. difficile carries two domains: the HTH domain and the Multimerization Domain (MD). In this study, we first performed a GST Pull-down experiment to determine the domain within SinR' that interacts with SinR. Second, the effect of these two domains on three phenotypes; sporulation, motility, and toxin production was examined. The findings of this study confirmed the prediction that the Multimerization Domain (MD) of SinR' is responsible for the interaction between SinR and SinR'. It was also discovered that SinR' regulates sporulation, toxin production and motility primarily by inhibiting SinR activity through the Multimerization Domain (MD).


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/biossíntese , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/metabolismo , Regulação Bacteriana da Expressão Gênica , Locomoção , Esporos Bacterianos/crescimento & desenvolvimento , Ligação Proteica , Mapeamento de Interação de Proteínas
6.
PLoS Pathog ; 14(3): e1006940, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29529083

RESUMO

Clostridium difficile is the primary cause of nosocomial diarrhea and pseudomembranous colitis. It produces dormant spores, which serve as an infectious vehicle responsible for transmission of the disease and persistence of the organism in the environment. In Bacillus subtilis, the sin locus coding SinR (113 aa) and SinI (57 aa) is responsible for sporulation inhibition. In B. subtilis, SinR mainly acts as a repressor of its target genes to control sporulation, biofilm formation, and autolysis. SinI is an inhibitor of SinR, so their interaction determines whether SinR can inhibit its target gene expression. The C. difficile genome carries two sinR homologs in the operon that we named sinR and sinR', coding for SinR (112 aa) and SinR' (105 aa), respectively. In this study, we constructed and characterized sin locus mutants in two different C. difficile strains R20291 and JIR8094, to decipher the locus's role in C. difficile physiology. Transcriptome analysis of the sinRR' mutants revealed their pleiotropic roles in controlling several pathways including sporulation, toxin production, and motility in C. difficile. Through various genetic and biochemical experiments, we have shown that SinR can regulate transcription of key regulators in these pathways, which includes sigD, spo0A, and codY. We have found that SinR' acts as an antagonist to SinR by blocking its repressor activity. Using a hamster model, we have also demonstrated that the sin locus is needed for successful C. difficile infection. This study reveals the sin locus as a central link that connects the gene regulatory networks of sporulation, toxin production, and motility; three key pathways that are important for C. difficile pathogenesis.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Movimento Celular/fisiologia , Clostridioides difficile/metabolismo , Infecções por Clostridium/microbiologia , Óperon , Esporos Bacterianos/fisiologia , Sequência de Aminoácidos , Animais , Bacillus subtilis/genética , Bacillus subtilis/crescimento & desenvolvimento , Bacillus subtilis/metabolismo , Proteínas de Bactérias/genética , Ceco/metabolismo , Ceco/microbiologia , Clostridioides difficile/genética , Clostridioides difficile/crescimento & desenvolvimento , Infecções por Clostridium/genética , Infecções por Clostridium/metabolismo , Regulação Bacteriana da Expressão Gênica , Mesocricetus , Camundongos , Coelhos , Regulon , Homologia de Sequência
7.
mSphere ; 2(1)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28217744

RESUMO

Clostridium difficile is an important nosocomial pathogen and the leading cause of hospital-acquired diarrhea. Antibiotic use is the primary risk factor for the development of C. difficile-associated disease because it disrupts normally protective gut flora and enables C. difficile to colonize the colon. C. difficile damages host tissue by secreting toxins and disseminates by forming spores. The toxin-encoding genes, tcdA and tcdB, are part of a pathogenicity locus, which also includes the tcdR gene that codes for TcdR, an alternate sigma factor that initiates transcription of tcdA and tcdB genes. We created a tcdR mutant in epidemic-type C. difficile strain R20291 in an attempt to identify the global role of tcdR. A site-directed mutation in tcdR affected both toxin production and sporulation in C. difficile R20291. Spores of the tcdR mutant were more heat sensitive than the wild type (WT). Nearly 3-fold more taurocholate was needed to germinate spores from the tcdR mutant than to germinate the spores prepared from the WT strain. Transmission electron microscopic analysis of the spores also revealed a weakly assembled exosporium on the tcdR mutant spores. Accordingly, comparative transcriptome analysis showed many differentially expressed sporulation genes in the tcdR mutant compared to the WT strain. These data suggest that regulatory networks of toxin production and sporulation in C. difficile strain R20291 are linked with each other. IMPORTANCEC. difficile infects thousands of hospitalized patients every year, causing significant morbidity and mortality. C. difficile spores play a pivotal role in the transmission of the pathogen in the hospital environment. During infection, the spores germinate, and the vegetative bacterial cells produce toxins that damage host tissue. Thus, sporulation and toxin production are two important traits of C. difficile. In this study, we showed that a mutation in tcdR, the toxin gene regulator, affects both toxin production and sporulation in epidemic-type C. difficile strain R20291.

8.
PLoS One ; 11(10): e0165579, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27768767

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0160107.].

9.
PLoS One ; 11(7): e0160107, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27467167

RESUMO

Clostridium difficile is the principal cause of antibiotic-associated diarrhea. Major metabolic requirements for colonization and expansion of C. difficile after microbiota disturbance have not been fully determined. In this study, we show that glutamate utilization is important for C. difficile to establish itself in the animal gut. When the gluD gene, which codes for glutamate dehydrogenase (GDH), was disrupted, the mutant C. difficile was unable to colonize and cause disease in a hamster model. Further, from the complementation experiment it appears that extracellular GDH may be playing a role in promoting C. difficile colonization and disease progression. Quantification of free amino acids in the hamster gut during C. difficile infection showed that glutamate is among preferred amino acids utilized by C. difficile during its expansion. This study provides evidence of the importance of glutamate metabolism for C. difficile pathogenesis.


Assuntos
Clostridioides difficile/enzimologia , Glutamato Desidrogenase/metabolismo , Animais , Clostridioides difficile/crescimento & desenvolvimento , Cricetinae
10.
Microbiology (Reading) ; 160(Pt 1): 47-55, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24145018

RESUMO

Clostridium difficile produces an NAD-specific glutamate dehydrogenase (GDH), which converts l-glutamate into α-ketoglutarate through an irreversible reaction. The enzyme GDH is detected in the stool samples of patients with C. difficile-associated disease and serves as one of the diagnostic tools to detect C. difficile infection (CDI). We demonstrate here that supernatant fluids of C. difficile cultures contain GDH. To understand the role of GDH in the physiology of C. difficile, an isogenic insertional mutant of gluD was created in strain JIR8094. The mutant failed to produce and secrete GDH as shown by Western blot analysis. Various phenotypic assays were performed to understand the importance of GDH in C. difficile physiology. In TY (tryptose yeast extract) medium, the gluD mutant grew slower than the parent strain. Complementation of the gluD mutant with the functional gluD gene reversed the growth defect in TY medium. The presence of extracellular GDH may have a functional role in the pathogenesis of CDI. In support of this assumption we found higher sensitivity to H2O2 in the gluD mutant as compared to the parent strain. Complementation of the gluD mutant with the functional gluD gene reversed the H2O2 sensitivity.


Assuntos
Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/enzimologia , Farmacorresistência Bacteriana , Glutamato Desidrogenase/metabolismo , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/toxicidade , Clostridioides difficile/genética , Clostridioides difficile/crescimento & desenvolvimento , Meios de Cultura/química , Técnicas de Inativação de Genes , Teste de Complementação Genética , Glutamato Desidrogenase/genética , Mutagênese Insercional
11.
J Bacteriol ; 195(18): 4246-54, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23873908

RESUMO

Toxigenic Clostridium sordellii causes uncommon but highly lethal infections in humans and animals. Recently, an increased incidence of C. sordellii infections has been reported in women undergoing obstetric interventions. Pathogenic strains of C. sordellii produce numerous virulence factors, including sordellilysin, phospholipase, neuraminidase, and two large clostridial glucosylating toxins, TcsL and TcsH. Recent studies have demonstrated that TcsL toxin is an essential virulence factor for the pathogenicity of C. sordellii. In this study, we identified and characterized TcsR as the toxin gene (tcsL) regulator in C. sordellii. High-throughput sequencing of two C. sordellii strains revealed that tcsR lies within a genomic region that encodes TcsL, TcsH, and TcsE, a putative holin. By using ClosTron technology, we inactivated the tcsR gene in strain ATCC 9714. Toxin production and tcsL transcription were decreased in the tcsR mutant strain. However, the complemented tcsR mutant produced large amounts of toxins, similar to the parental strain. Expression of the Clostridium difficile toxin gene regulator tcdR also restored toxin production to the C. sordellii tcsR mutant, showing that these sigma factors are functionally interchangeable.


Assuntos
Proteínas de Bactérias , Toxinas Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica , Fator sigma , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Infecções por Clostridium/microbiologia , Clostridium sordellii/genética , Clostridium sordellii/metabolismo , Clostridium sordellii/patogenicidade , Feminino , Genes Reguladores , Genoma Bacteriano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Dados de Sequência Molecular , Análise de Sequência de DNA , Fator sigma/química , Fator sigma/genética , Fator sigma/metabolismo , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...