Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 183: 114314, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33152344

RESUMO

Clostridioides difficile infections (CDI) are the leading cause of nosocomial antibiotic-associated diarrhea. C. difficile produces dormant spores that serve as infectious agents. Bile salts in the gastrointestinal tract signal spores to germinate into toxin-producing cells. As spore germination is required for CDI onset, anti-germination compounds may serve as prophylactics. CamSA, a synthetic bile salt, was previously shown to inhibit C. difficile spore germination in vitro and in vivo. Unexpectedly, a single dose of CamSA was sufficient to offer multi-day protection from CDI in mice without any observable toxicity. To study this intriguing protection pattern, we examined the pharmacokinetic parameters of CamSA. CamSA was stable to the gut of antibiotic-treated mice but was extensively degraded by the microbiota of non-antibiotic-treated animals. Our data also suggest that CamSA's systemic absorption is minimal since it is retained primarily in the intestinal lumen and liver. CamSA shows weak interactions with CYP3A4, a P450 hepatic isozyme involved in drug metabolism and bile salt modification. Like other bile salts, CamSA seems to undergo enterohepatic circulation. We hypothesize that the cycling of CamSA between the liver and intestines serves as a slow-release mechanism that allows CamSA to be retained in the gastrointestinal tract for days. This model explains how a single CamSA dose can prevent murine CDI even though spores are present in the animal's intestine for up to four days post-challenge.


Assuntos
Ácidos e Sais Biliares/administração & dosagem , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/prevenção & controle , Microbioma Gastrointestinal/efeitos dos fármacos , Profilaxia Pré-Exposição/métodos , Animais , Ácidos e Sais Biliares/química , Clostridioides difficile/fisiologia , Infecções por Clostridium/fisiopatologia , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
2.
Artigo em Inglês | MEDLINE | ID: mdl-30012758

RESUMO

Clostridium difficile infection (CDI) is the leading cause of antibiotic-associated diarrhea and has gained worldwide notoriety due to emerging hypervirulent strains and the high incidence of recurrence. We previously reported protection of mice from CDI using the antigerminant bile salt analog CamSA. Here we describe the effects of CamSA in the hamster model of CDI. CamSA treatment of hamsters showed no toxicity and did not affect the richness or diversity of gut microbiota; however, minor changes in community composition were observed. Treatment of C. difficile-challenged hamsters with CamSA doubled the mean time to death, compared to control hamsters. However, CamSA alone was insufficient to prevent CDI in hamsters. CamSA in conjunction with suboptimal concentrations of vancomycin led to complete protection from CDI in 70% of animals. Protected animals remained disease-free at least 30 days postchallenge and showed no signs of colonic tissue damage. In a delayed-treatment model of hamster CDI, CamSA was unable to prevent infection signs and death. These data support a putative model in which CamSA reduces the number of germinating C. difficile spores but does not keep all of the spores from germinating. Vancomycin halts division of any vegetative cells that are able to grow from spores that escape CamSA.


Assuntos
Antibacterianos/uso terapêutico , Ácidos e Sais Biliares/uso terapêutico , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Animais , Clostridioides difficile/patogenicidade , Cricetinae , Feminino , Vancomicina/uso terapêutico
3.
Mol Microbiol ; 108(5): 505-518, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29453862

RESUMO

Transcriptional silencing and anti-silencing mechanisms modulate bacterial physiology and virulence in many human pathogens. In Shigella species, many virulence plasmid genes are silenced by the histone-like nucleoid structuring protein H-NS and anti-silenced by the virulence gene regulator VirB. Despite the key role that these regulatory proteins play in Shigella virulence, their mechanisms of transcriptional control remain poorly understood. Here, we characterize the regulatory elements and their relative spacing requirements needed for the transcriptional silencing and anti-silencing of icsP, a locus that requires remotely located regulatory elements for both types of transcriptional control. Our findings highlight the flexibility of the regulatory elements' positions with respect to each other, and yet, a molecular roadblock docked between the VirB binding site and the upstream H-NS binding region abolishes transcriptional anti-silencing by VirB, providing insight into transcriptional anti-silencing. Our study also raises the need to re-evaluate the currently proposed VirB binding site. Models of transcriptional silencing and anti-silencing at this genetic locus are presented, and the implications for understanding these regulatory mechanisms in bacteria are discussed.


Assuntos
Proteínas de Bactérias/genética , Proteínas Repressoras/metabolismo , Shigella flexneri/genética , Shigella flexneri/patogenicidade , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Loci Gênicos/genética , Humanos , Plasmídeos , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Transcrição Gênica , Virulência/genética
4.
PLoS One ; 8(8): e72620, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023628

RESUMO

Clostridium difficile infection (CDI) is a leading cause of antibiotic-associated diarrhea, a major nosocomial complication. The infective form of C. difficile is the spore, a dormant and resistant structure that forms under stress. Although spore germination is the first committed step in CDI onset, the temporal and spatial distribution of ingested C. difficile spores is not clearly understood. We recently reported that CamSA, a synthetic bile salt analog, inhibits C. difficile spore germination in vitro and in vivo. In this study, we took advantage of the anti-germination activity of bile salts to determine the fate of ingested C. difficile spores. We tested four different bile salts for efficacy in preventing CDI. Since CamSA was the only anti-germinant tested able to prevent signs of CDI, we characterized CamSa's in vitro stability, distribution, and cytotoxicity. We report that CamSA is stable to simulated gastrointestinal (GI) environments, but will be degraded by members of the natural microbiota found in a healthy gut. Our data suggest that CamSA will not be systemically available, but instead will be localized to the GI tract. Since in vitro pharmacological parameters were acceptable, CamSA was used to probe the mouse model of CDI. By varying the timing of CamSA dosage, we estimated that C. difficile spores germinated and established infection less than 10 hours after ingestion. We also showed that ingested C. difficile spores rapidly transited through the GI tract and accumulated in the colon and cecum of CamSA-treated mice. From there, C. difficile spores were slowly shed over a 96-hour period. To our knowledge, this is the first report of using molecular probes to obtain disease progression information for C. difficile infection.


Assuntos
Clostridioides difficile/fisiologia , Infecções por Clostridium/microbiologia , Animais , Toxinas Bacterianas/metabolismo , Células CACO-2 , Ceco/efeitos dos fármacos , Ceco/microbiologia , Ceco/patologia , Morte Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Chlorocebus aethiops , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/crescimento & desenvolvimento , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/prevenção & controle , Citoproteção/efeitos dos fármacos , Fezes/microbiologia , Humanos , Hidrolases/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Intestinos/patologia , Camundongos , Nível de Efeito Adverso não Observado , Esporos Bacterianos/efeitos dos fármacos , Esporos Bacterianos/fisiologia , Ácido Taurocólico/efeitos adversos , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/farmacologia , Ácido Taurocólico/toxicidade , Fatores de Tempo , Células Vero
5.
J Infect Dis ; 207(10): 1498-504, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23420906

RESUMO

BACKGROUND: Clostridium difficile infection (CDI) is a leading cause of antibiotic-associated diarrhea. The infective form of C. difficile is the spore, but the vegetative bacterium causes the disease. Because C. difficile spore germination is required for symptomatic infection, antigermination approaches could lead to the prevention of CDI. We recently reported that CamSA, a bile salt analog, inhibits C. difficile spore germination in vitro. METHODS: Mice infected with massive inocula of C. difficile spores were treated with different concentrations of CamSA and monitored for CDI signs. C. difficile spore and vegetative cells were counted in feces from infected mice. RESULTS: A single 50-mg/kg dose of CamSA prevented CDI in mice without any observable toxicity. Lower CamSA doses resulted in delayed CDI onset and less severe signs of disease. Ingested C. difficile spores were quantitatively recovered from feces of CamSA-protected mice. CONCLUSIONS: Our results support a mechanism whereby the antigermination effect of CamSA is responsible for preventing CDI signs. This approach represents a new paradigm in CDI treatment. Instead of further compromising the microbiota of CDI patients with strong antibiotics, antigermination therapy could serve as a microbiota surrogate to curtail C. difficile colonization of antibiotic-treated patients.


Assuntos
Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/prevenção & controle , Animais , Antibacterianos/farmacologia , Ácidos e Sais Biliares/farmacologia , Clostridioides difficile/patogenicidade , Infecções por Clostridium/complicações , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/microbiologia , Colistina/farmacologia , Diarreia/tratamento farmacológico , Diarreia/etiologia , Diarreia/microbiologia , Relação Dose-Resposta a Droga , Fezes/microbiologia , Feminino , Gentamicinas/farmacologia , Canamicina/farmacologia , Metagenoma , Metronidazol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Esporos Bacterianos/efeitos dos fármacos , Esporos Bacterianos/patogenicidade , Vancomicina/farmacologia
6.
J Bacteriol ; 193(1): 274-82, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20971909

RESUMO

Germination of Clostridium difficile spores is the first required step in establishing C. difficile-associated disease (CDAD). Taurocholate (a bile salt) and glycine (an amino acid) have been shown to be important germinants of C. difficile spores. In the present study, we tested a series of glycine and taurocholate analogs for the ability to induce or inhibit C. difficile spore germination. Testing of glycine analogs revealed that both the carboxy and amino groups are important epitopes for recognition and that the glycine binding site can accommodate compounds with more widely separated termini. The C. difficile germination machinery also recognizes other hydrophobic amino acids. In general, linear alkyl side chains are better activators of spore germination than their branched analogs. However, L-phenylalanine and L-arginine are also good germinants and are probably recognized by distinct binding sites. Testing of taurocholate analogs revealed that the 12-hydroxyl group of taurocholate is necessary, but not sufficient, to activate spore germination. In contrast, the 6- and 7-hydroxyl groups are required for inhibition of C. difficile spore germination. Similarly, C. difficile spores are able to detect taurocholate analogs with shorter, but not longer, alkyl amino sulfonic acid side chains. Furthermore, the sulfonic acid group can be partially substituted with other acidic groups. Finally, a taurocholate analog with an m-aminobenzenesulfonic acid side chain is a strong inhibitor of C. difficile spore germination. In conclusion, C. difficile spores recognize both amino acids and taurocholate through multiple interactions that are required to bind the germinants and/or activate the germination machinery.


Assuntos
Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/fisiologia , Glicina/farmacologia , Esporos Bacterianos/efeitos dos fármacos , Esporos Bacterianos/fisiologia , Ácido Taurocólico/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Glicina/análogos & derivados , Glicina/química , Estrutura Molecular , Relação Estrutura-Atividade , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/química , Fatores de Tempo
7.
PLoS One ; 4(7): e6398, 2009 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-19636427

RESUMO

BACKGROUND: The first step of the bacterial lifecycle is the germination of bacterial spores into their vegetative form, which requires the presence of specific nutrients. In contrast to closely related Bacillus anthracis spores, Bacillus cereus spores germinate in the presence of a single germinant, inosine, yet with a significant lag period. METHODS AND FINDINGS: We found that the initial lag period of inosine-treated germination of B. cereus spores disappeared in the presence of supernatants derived from already germinated spores. The lag period also dissipated when inosine was supplemented with the co-germinator alanine. In fact, HPLC-based analysis revealed the presence of amino acids in the supernatant of germinated B. cereus spores. The released amino acids included alanine in concentrations sufficient to promote rapid germination of inosine-treated spores. The alanine racemase inhibitor D-cycloserine enhanced germination of B. cereus spores, presumably by increasing the L-alanine concentration in the supernatant. Moreover, we found that B. cereus spores lacking the germination receptors gerI and gerQ did not germinate and release amino acids in the presence of inosine. These mutant spores, however, germinated efficiently when inosine was supplemented with alanine. Finally, removal of released amino acids in a washout experiment abrogated inosine-mediated germination of B. cereus spores. CONCLUSIONS: We found that the single germinant inosine is able to trigger a two-tier mechanism for inosine-mediated germination of B. cereus spores: Inosine mediates the release of alanine, an essential step to complete the germination process. Therefore, B. cereus spores appear to have developed a unique quorum-sensing feedback mechanism to monitor spore density and to coordinate germination.


Assuntos
Alanina/metabolismo , Bacillus cereus/fisiologia , Inosina/metabolismo , Esporos Bacterianos/fisiologia , Cromatografia Líquida de Alta Pressão
8.
J Cosmet Dermatol ; 5(1): 30-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17173569

RESUMO

BACKGROUND: CoQ10 (ubiquinone, coenzyme Q10) and carotenoids are popular antioxidants used in many skin care products to protect the skin from free radical damage. AIM: To evaluate the effects of CoQ10 and colorless carotenoids on the production of inflammatory mediators in human dermal fibroblasts treated with UV radiation (UVR) and to investigate the possible synergistic effects of these two antioxidants. METHODS: Normal human dermal fibroblast cell cultures were exposed to either 50 mJ of UVR or to IL-1 and then incubated with various concentrations of either CoQ10, the colorless carotenoids, phytoene and phytofluene, or to combinations of these antioxidants. After 24 h in culture, cells and spent medium were harvested and assayed by enzyme-linked immunosorbent assay for prostaglandin E2 (PGE-2), interleukin 6 (IL-6), and matrix metalloproteinase 1 (MMP-1). In addition, the ability of the carotenoids to protect CoQ10 from oxidation by the reactive oxygen species (ROS), hyperchlorite, was also determined. RESULTS: Human fibroblasts respond to UVR or to IL-1 by increasing the production of various inflammatory mediators including PGE-2, IL-1, and IL-6 and proteases such as collagenase (MMP-1). Treatment of fibroblasts with 10 microm of CoQ10 suppressed the UVR- or IL-1-induced increase in PGE-2, IL-6, and MMP-1. The combination of carotenoids and CoQ10 produced an enhanced inhibition of these three inflammatory mediators. Furthermore, the colorless carotenoids, phytoene and phytofluene, protected CoQ10 from degradation by the ROS, hypochlorite. CONCLUSION: CoQ10 is able to suppress the UVR- or IL-1-induced inflammatory response in dermal fibroblasts. Furthermore, this compound can block the UVR induction of the matrix-eroding enzyme, MMP-1. Finally, the combination of carotenoids plus CoQ10 results in enhanced suppression of inflammation. The results suggest that the combination of carotenoids and CoQ10 in topical skin care products may provide enhanced protection from inflammation and premature aging caused by sun exposure.


Assuntos
Anti-Inflamatórios/farmacologia , Carotenoides/farmacologia , Fibroblastos/citologia , Pele/citologia , Ubiquinona/análogos & derivados , Células Cultivadas , Coenzimas , Dinoprostona/metabolismo , Ensaio de Imunoadsorção Enzimática , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Humanos , Interleucina-6/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Ubiquinona/farmacologia , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...