Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 181(7): 1452-1454, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32589955

RESUMO

In this issue of Cell, Alavi et al. report that infection by Vibrio cholerae is blocked by gut microbiome-mediated hydrolysis of bile acids. Cholera therefore joins amebic dysentery and Clostridioides difficile colitis as enteric infections profoundly influenced by the microbiome's impact on bile acid metabolism.


Assuntos
Cólera , Microbioma Gastrointestinal , Microbiota , Vibrio cholerae , Ácidos e Sais Biliares , Humanos
2.
Proc Natl Acad Sci U S A ; 120(19): e2301252120, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37126691

RESUMO

Intestinal bile acids play an essential role in the Clostridioides difficile lifecycle having been shown in vitro to modulate various aspects of pathogenesis, including spore germination, vegetative growth, and more recently the action of the primary virulence determinant, TcdB. Here, we investigated whether physiological levels of the total pool of intestinal bile acids in mice and humans protect against TcdB action. Small molecules extracted from the lumenal contents of the small intestine, cecum, colon, and feces were found to inhibit TcdB in accordance with the differential amounts of total bile acids in each compartment. Extracts from antibiotic-treated and germ-free mice, despite harboring dramatically altered bile acid profiles, unexpectedly also prevented TcdB-induced cell rounding to similar extents. We show that protection, however, is surmountable and can be overcome at higher doses of TcdB-typical to those seen during severe C. difficile infection-suggesting that the protective properties of intestinal bile acids are operant primarily under low to moderate toxin levels. Taken together, these findings demonstrate a role for intestinal bile acids in attenuating virulence, provide insights into asymptomatic carriage of toxigenic C. difficile, and inform strategies to manipulate bile acid levels for therapeutic benefit.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Infecções por Clostridium , Humanos , Camundongos , Animais , Ácidos e Sais Biliares , Infecções por Clostridium/patologia , Intestinos/patologia , Proteínas de Bactérias
3.
J Proteome Res ; 23(6): 2000-2012, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38752739

RESUMO

Biological interpretation of untargeted LC-MS-based metabolomics data depends on accurate compound identification, but current techniques fall short of identifying most features that can be detected. The human fecal metabolome is complex, variable, incompletely annotated, and serves as an ideal matrix to evaluate novel compound identification methods. We devised an experimental strategy for compound annotation using multidimensional chromatography and semiautomated feature alignment and applied these methods to study the fecal metabolome in the context of fecal microbiota transplantation (FMT) for recurrent C. difficile infection. Pooled fecal samples were fractionated using semipreparative liquid chromatography and analyzed by an orthogonal LC-MS/MS method. The resulting spectra were searched against commercial, public, and local spectral libraries, and annotations were vetted using retention time alignment and prediction. Multidimensional chromatography yielded more than a 2-fold improvement in identified compounds compared to conventional LC-MS/MS and successfully identified several rare and previously unreported compounds, including novel fatty-acid conjugated bile acid species. Using an automated software-based feature alignment strategy, most metabolites identified by the new approach could be matched to features that were detected but not identified in single-dimensional LC-MS/MS data. Overall, our approach represents a powerful strategy to enhance compound identification and biological insight from untargeted metabolomics data.


Assuntos
Transplante de Microbiota Fecal , Fezes , Metaboloma , Metabolômica , Espectrometria de Massas em Tandem , Humanos , Fezes/microbiologia , Fezes/química , Cromatografia Líquida/métodos , Metabolômica/métodos , Espectrometria de Massas em Tandem/métodos , Infecções por Clostridium/microbiologia , Infecções por Clostridium/metabolismo , Clostridioides difficile/metabolismo , Ácidos e Sais Biliares/metabolismo , Ácidos e Sais Biliares/análise , Espectrometria de Massa com Cromatografia Líquida
4.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33526676

RESUMO

Primary bile acids (BAs) are a collection of host-synthesized metabolites that shape physiology and metabolism. BAs transit the gastrointestinal tract and are subjected to a variety of chemical transformations encoded by indigenous bacteria. The resulting microbiota-derived BA pool is a mediator of host-microbiota interactions. Bacterial bile salt hydrolases (BSHs) cleave the conjugated glycine or taurine from BAs, an essential upstream step for the production of deconjugated and secondary BAs. Probiotic lactobacilli harbor a considerable number and diversity of BSHs; however, their contribution to Lactobacillus fitness and colonization remains poorly understood. Here, we define and compare the functions of multiple BSHs encoded by Lactobacillus acidophilus and Lactobacillus gasseri Our genetic and biochemical characterization of lactobacilli BSHs lend to a model of Lactobacillus adaptation to the gut. These findings deviate from previous notions that BSHs generally promote colonization and detoxify bile. Rather, we show that BSH enzymatic preferences and the intrinsic chemical features of various BAs determine the toxicity of these molecules during Lactobacillus growth. BSHs were able to alter the Lactobacillus transcriptome in a BA-dependent manner. Finally, BSHs were able to dictate differences in bacterial competition in vitro and in vivo, defining their impact on BSH-encoding bacteria within the greater gastrointestinal tract ecosystem. This work emphasizes the importance of considering the enzymatic preferences of BSHs alongside the conjugated/deconjugated BA-bacterial interaction. These results deepen our understanding of the BA-microbiome axis and provide a framework to engineer lactobacilli with improved bile resistance and use probiotics as BA-altering therapeutics.


Assuntos
Amidoidrolases/genética , Microbioma Gastrointestinal/genética , Interações Hospedeiro-Patógeno/genética , Lactobacillus/enzimologia , Amidoidrolases/metabolismo , Ecossistema , Microbioma Gastrointestinal/fisiologia , Aptidão Genética/genética , Humanos , Lactobacillus/genética , Probióticos/farmacologia , Especificidade por Substrato/genética
5.
J Bacteriol ; 205(9): e0018023, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37695856

RESUMO

Clostridioides difficile is a Gram-positive, spore-forming anaerobe that causes clinical diseases ranging from diarrhea and pseudomembranous colitis to toxic megacolon and death. C. difficile infection (CDI) is associated with antibiotic usage, which disrupts the indigenous gut microbiota and causes the loss of microbial-derived secondary bile acids that normally provide protection against C. difficile colonization. Previous work has shown that the secondary bile acid lithocholate (LCA) and its epimer isolithocholate (iLCA) have potent inhibitory activity against clinically relevant C. difficile strains. To further characterize the mechanisms by which LCA and its epimers iLCA and isoallolithocholate (iaLCA) inhibit C. difficile, we tested their minimum inhibitory concentration against C. difficile R20291 and a commensal gut microbiota panel. We also performed a series of experiments to determine the mechanism of action by which LCA and its epimers inhibit C. difficile through bacterial killing and effects on toxin expression and activity. Additionally, we tested the cytotoxicity of these bile acids through Caco-2 cell apoptosis and viability assays to gauge their effects on the host. Here, we show that the epimers iLCA and iaLCA strongly inhibit C. difficile growth in vitro while sparing most commensal Gram-negative gut microbes. We also show that iLCA and iaLCA have bactericidal activity against C. difficile, and these epimers cause significant bacterial membrane damage at subinhibitory concentrations. Finally, we observe that iLCA and iaLCA decrease the expression of the large cytotoxin tcdA, while LCA significantly reduces toxin activity. Although iLCA and iaLCA are both epimers of LCA, they have distinct mechanisms for inhibiting C. difficile. LCA epimers, iLCA and iaLCA, represent promising compounds that target C. difficile with minimal effects on members of the gut microbiota that are important for colonization resistance. IMPORTANCE In the search for a novel therapeutic that targets Clostridioides difficile, bile acids have become a viable solution. Epimers of bile acids are particularly attractive as they may provide protection against C. difficile while leaving the indigenous gut microbiota largely unaltered. This study shows that LCA epimers isolithocholate (iLCA) and LCA epimers isoallolithocholate (iaLCA) specifically are potent inhibitors of C. difficile, affecting key virulence factors including growth, toxin expression, and activity. As we move toward the use of bile acids as therapeutics, further work will be required to determine how best to deliver these bile acids to a target site within the host intestinal tract.


Assuntos
Clostridioides difficile , Microbioma Gastrointestinal , Humanos , Virulência , Células CACO-2 , Ácidos e Sais Biliares/farmacologia , Ácido Litocólico
6.
Anal Chem ; 95(41): 15357-15366, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37796494

RESUMO

Bile acids play key roles in nutrient uptake, inflammation, signaling, and microbiome composition. While previous bile acid analyses have primarily focused on profiling 5 canonical primary and secondary bile acids and their glycine and taurine amino acid-bile acid (AA-BA) conjugates, recent studies suggest that many other microbial conjugated bile acids (or MCBAs) exist. MCBAs are produced by the gut microbiota and serve as biomarkers, providing information about early disease onset and gut health. Here we analyzed 8 core bile acids synthetically conjugated with 22 proteinogenic and nonproteogenic amino acids totaling 176 MCBAs. Since many of the conjugates were isomeric and only 42 different m/z values resulted from the 176 MCBAs, a platform coupling liquid chromatography, ion mobility spectrometry, and mass spectrometry (LC-IMS-MS) was used for their separation. Their molecular characteristics were then used to create an in-house extended bile acid library for a combined total of 182 unique compounds. Additionally, ∼250 rare bile acid extracts were also assessed to provide additional resources for bile acid profiling and identification. This library was then applied to healthy mice dosed with antibiotics and humans having fecal microbiota transplantation (FMT) to assess the MCBA presence and changes in the gut before and after each perturbation.


Assuntos
Aminoácidos , Ácidos e Sais Biliares , Humanos , Camundongos , Animais , Isomerismo , Espectrometria de Massas , Esteroides
7.
Proc Natl Acad Sci U S A ; 117(12): 6792-6800, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32152097

RESUMO

Intestinal bile acids are known to modulate the germination and growth of Clostridioides difficile Here we describe a role for intestinal bile acids in directly binding and neutralizing TcdB toxin, the primary determinant of C. difficile disease. We show that individual primary and secondary bile acids reversibly bind and inhibit TcdB to varying degrees through a mechanism that requires the combined oligopeptide repeats region to which no function has previously been ascribed. We find that bile acids induce TcdB into a compact "balled up" conformation that is no longer able to bind cell surface receptors. Lastly, through a high-throughput screen designed to identify bile acid mimetics we uncovered nonsteroidal small molecule scaffolds that bind and inhibit TcdB through a bile acid-like mechanism. In addition to suggesting a role for bile acids in C. difficile pathogenesis, these findings provide a framework for development of a mechanistic class of C. difficile antitoxins.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Ácidos e Sais Biliares/metabolismo , Clostridioides difficile/metabolismo , Intestinos/fisiologia , Receptores de Superfície Celular/metabolismo , Células CACO-2 , Clostridioides difficile/crescimento & desenvolvimento , Infecções por Clostridium/microbiologia , Células HCT116 , Humanos
8.
Infect Immun ; 90(8): e0015322, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35862710

RESUMO

C. difficile infection (CDI) is a highly inflammatory disease mediated by the production of two large toxins that weaken the intestinal epithelium and cause extensive colonic tissue damage. Antibiotic alternative therapies for CDI are urgently needed as current antibiotic regimens prolong the perturbation of the microbiota and lead to high disease recurrence rates. Inflammation is more closely correlated with CDI severity than bacterial burden, thus therapies that target the host response represent a promising yet unexplored strategy for treating CDI. Intestinal bile acids are key regulators of gut physiology that exert cytoprotective roles in cellular stress, inflammation, and barrier integrity, yet the dynamics between bile acids and host cellular processes during CDI have not been investigated. Here we show that several bile acids are protective against apoptosis caused by C. difficile toxins in Caco-2 cells and that protection is dependent on conjugation of bile acids. Out of 20 tested bile acids, taurine conjugated ursodeoxycholic acid (TUDCA) was the most potent inhibitor, yet unconjugated UDCA did not alter toxin-induced apoptosis. TUDCA treatment decreased expression of genes in lysosome associated and cytokine signaling pathways. TUDCA did not affect C. difficile growth or toxin activity in vitro whereas UDCA significantly reduced toxin activity in a Vero cell cytotoxicity assay and decreased tcdA gene expression. These results demonstrate that bile acid conjugation can have profound effects on C. difficile as well as the host and that conjugated and unconjugated bile acids may exert different therapeutic mechanisms against CDI.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Antibacterianos/farmacologia , Anticorpos Antibacterianos/farmacologia , Apoptose , Ácidos e Sais Biliares/farmacologia , Células CACO-2 , Infecções por Clostridium/microbiologia , Humanos , Inflamação , Ácido Tauroquenodesoxicólico , Ácido Ursodesoxicólico/farmacologia
9.
J Infect Dis ; 223(12 Suppl 2): S194-S200, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33326565
10.
Nucleic Acids Res ; 47(18): e103, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31269198

RESUMO

Targeted PCR amplification and high-throughput sequencing (amplicon sequencing) of 16S rRNA gene fragments is widely used to profile microbial communities. New long-read sequencing technologies can sequence the entire 16S rRNA gene, but higher error rates have limited their attractiveness when accuracy is important. Here we present a high-throughput amplicon sequencing methodology based on PacBio circular consensus sequencing and the DADA2 sample inference method that measures the full-length 16S rRNA gene with single-nucleotide resolution and a near-zero error rate. In two artificial communities of known composition, our method recovered the full complement of full-length 16S sequence variants from expected community members without residual errors. The measured abundances of intra-genomic sequence variants were in the integral ratios expected from the genuine allelic variants within a genome. The full-length 16S gene sequences recovered by our approach allowed Escherichia coli strains to be correctly classified to the O157:H7 and K12 sub-species clades. In human fecal samples, our method showed strong technical replication and was able to recover the full complement of 16S rRNA alleles in several E. coli strains. There are likely many applications beyond microbial profiling for which high-throughput amplicon sequencing of complete genes with single-nucleotide resolution will be of use.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Microbiota/genética , Filogenia , RNA Ribossômico 16S/genética , Escherichia coli K12/classificação , Escherichia coli K12/genética , Escherichia coli O157/classificação , Escherichia coli O157/genética , Fezes/microbiologia , Humanos , Reação em Cadeia da Polimerase/métodos , Polimorfismo de Nucleotídeo Único/genética , Análise de Sequência de DNA
11.
Infect Immun ; 88(6)2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32205405

RESUMO

Clostridioides difficile infection (CDI) is associated with increasing morbidity and mortality posing an urgent threat to public health. Recurrence of CDI after successful treatment with antibiotics is high, thus necessitating discovery of novel therapeutics against this enteric pathogen. Administration of the secondary bile acid ursodeoxycholic acid (UDCA; ursodiol) inhibits the life cycles of various strains of C. difficilein vitro, suggesting that the FDA-approved formulation of UDCA, known as ursodiol, may be able to restore colonization resistance against C. difficilein vivo However, the mechanism(s) by which ursodiol is able to restore colonization resistance against C. difficile remains unknown. Here, we confirmed that ursodiol inhibits C. difficile R20291 spore germination and outgrowth, growth, and toxin activity in a dose-dependent manner in vitro In a murine model of CDI, exogenous administration of ursodiol resulted in significant alterations in the bile acid metabolome with little to no changes in gut microbial community structure. Ursodiol pretreatment resulted in attenuation of CDI pathogenesis early in the course of disease, which coincided with alterations in the cecal and colonic inflammatory transcriptome, bile acid-activated receptors nuclear farnesoid X receptor (FXR) and transmembrane G-protein-coupled membrane receptor 5 (TGR5), which are able to modulate the innate immune response through signaling pathways such as NF-κB. Although ursodiol pretreatment did not result in a consistent decrease in the C. difficile life cycle in vivo, it was able to attenuate an overly robust inflammatory response that is detrimental to the host during CDI. Ursodiol remains a viable nonantibiotic treatment and/or prevention strategy against CDI. Likewise, modulation of the host innate immune response via bile acid-activated receptors FXR and TGR5 represents a new potential treatment strategy for patients with CDI.


Assuntos
Ácidos e Sais Biliares/metabolismo , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/metabolismo , Infecções por Clostridium/microbiologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Ácido Ursodesoxicólico/farmacologia , Animais , Biomarcadores , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/genética , Biologia Computacional/métodos , Relação Dose-Resposta a Droga , Fatores de Crescimento de Fibroblastos/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Estágios do Ciclo de Vida , Camundongos , Transdução de Sinais , Transcriptoma , Ácido Ursodesoxicólico/fisiologia
12.
Annu Rev Microbiol ; 69: 445-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26488281

RESUMO

Antibiotics have significant and long-lasting effects on the intestinal microbiota and consequently reduce colonization resistance against pathogens, including Clostridium difficile. By altering the community structure of the gut microbiome, antibiotics alter the intestinal metabolome, which includes both host- and microbe-derived metabolites. The mechanisms by which antibiotics reduce colonization resistance against C. difficile are unknown yet important for development of preventative and therapeutic approaches against this pathogen. This review focuses on how antibiotics alter the structure of the gut microbiota and how this alters microbial metabolism in the intestine. Interactions between gut microbial products and C. difficile spore germination, growth, and toxin production are discussed. New bacterial therapies to restore changes in bacteria-driven intestinal metabolism following antibiotics will have important applications for treatment and prevention of C. difficile infection.


Assuntos
Clostridioides difficile/metabolismo , Infecções por Clostridium/microbiologia , Microbioma Gastrointestinal , Animais , Antibacterianos/administração & dosagem , Fermentação , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Evasão da Resposta Imune , Interações Microbianas , Microbiota/efeitos dos fármacos
13.
Anaerobe ; 53: 64-73, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29654837

RESUMO

A significant proportion of individuals develop recurrent Clostridium difficile infection (CDI) following initial disease. Fecal microbiota transplantation (FMT), a highly effective treatment method for recurrent CDI, has been demonstrated to induce microbiota recovery. One of the proposed functions associated with restoration of colonization resistance against C. difficile has been recovery of bile acid metabolism. In this study, we aimed to assess recovery of short chain fatty acids (SCFAs) in addition to bile acids alongside microbial community structure in six patients with recurrent CDI following treatment with FMT over time. Using 16S rRNA gene-based sequencing, we observed marked similarity of the microbiota between recipients following FMT (n = 6, sampling up to 6 months post-FMT) and their respective donors. Sustained increases in the levels of the SCFAs butyrate, acetate, and propionate were observed post-FMT, and variable recovery over time was observed in the secondary bile acids deoxycholate and lithocholate. To correlate these changes with specific microbial taxa at an individual level, we applied a generalized estimating equation approach to model metabolite concentrations with the presence of specific members of the microbiota. Metabolites that increased following FMT were associated with bacteria classified within the Lachnospiraceae, Ruminococcaceae, and unclassified Clostridiales families. In contrast, members of these taxa were inversely associated with primary bile acids. The longitudinal aspect of this study allowed us to characterize individualized patterns of recovery, revealing variability between and within patients following FMT.


Assuntos
Ácidos e Sais Biliares/metabolismo , Infecções por Clostridium/terapia , Ácidos Graxos Voláteis/metabolismo , Transplante de Microbiota Fecal , Prevenção Secundária/métodos , Adulto , Idoso , Feminino , Microbioma Gastrointestinal , Humanos , Estudos Longitudinais , Masculino , Metabolômica , Pessoa de Meia-Idade , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Adulto Jovem
14.
Anaerobe ; 45: 86-100, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28279860

RESUMO

The changing epidemiology of Clostridium difficile infection over the past decades presents a significant challenge in the management of C. difficile associated diseases. The gastrointestinal tract microbiota provides colonization resistance against C. difficile, and growing evidence suggests that gut microbial derived secondary bile acids (SBAs) play a role. We hypothesized that the C. difficile life cycle; spore germination and outgrowth, growth, and toxin production, of strains that vary by age and ribotype will differ in their sensitivity to SBAs. C. difficile strains R20291 and CD196 (ribotype 027), M68 and CF5 (017), 630 (012), BI9 (001) and M120 (078) were used to define taurocholate (TCA) mediated spore germination and outgrowth, growth, and toxin activity in the absence and presence of gut microbial derived SBAs (deoxycholate, isodeoxycholate, lithocholate, isolithocholate, ursodeoxycholate, ω-muricholate, and hyodeoxycholate) found in the human and mouse large intestine. C. difficile strains varied in their rates of germination, growth kinetics, and toxin activity without the addition of SBAs. C. difficile M120, a highly divergent strain, had robust germination, growth, but significantly lower toxin activity compared to other strains. Many SBAs were able to inhibit TCA mediated spore germination and outgrowth, growth, and toxin activity in a dose dependent manner, but the level of inhibition and resistance varied across all strains and ribotypes. This study illustrates how clinically relevant C. difficile strains can have different responses when exposed to SBAs present in the gastrointestinal tract.


Assuntos
Antibacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Ácidos e Sais Biliares/metabolismo , Clostridioides difficile/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Esporos Bacterianos/efeitos dos fármacos , Animais , Antibiose , Clostridioides difficile/crescimento & desenvolvimento , Microbioma Gastrointestinal , Humanos , Camundongos , Esporos Bacterianos/crescimento & desenvolvimento
16.
Anaerobe ; 41: 44-50, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27163871

RESUMO

Clostridium difficile is an anaerobic, Gram positive, spore-forming bacillus that is the leading cause of nosocomial gastroenteritis. Clostridium difficile infection (CDI) is associated with increasing morbidity and mortality, consequently posing an urgent threat to public health. Recurrence of CDI after successful treatment with antibiotics is high, thus necessitating discovery of novel therapeutics against this pathogen. Susceptibility to CDI is associated with alterations in the gut microbiota composition and bile acid metabolome, specifically a loss of microbial derived secondary bile acids. This review aims to summarize in vitro, ex vivo, and in vivo studies done by our group and others that demonstrate how secondary bile acids affect the different stages of the C. difficile life cycle. Understanding the dynamic interplay of C. difficile and microbial derived secondary bile acids within the gastrointestinal tract will shed light on how bile acids play a role in colonization resistance against C. difficile. Rational manipulation of secondary bile acids may prove beneficial as a treatment for patients with CDI.


Assuntos
Ácidos e Sais Biliares/fisiologia , Clostridioides difficile/fisiologia , Enterocolite Pseudomembranosa/microbiologia , Trato Gastrointestinal/microbiologia , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Ácidos e Sais Biliares/farmacologia , Clostridioides difficile/efeitos dos fármacos , Suscetibilidade a Doenças , Resistencia a Medicamentos Antineoplásicos , Enterocolite Pseudomembranosa/tratamento farmacológico , Microbioma Gastrointestinal , Humanos
17.
Infect Immun ; 83(10): 3838-46, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169276

RESUMO

Recurrent Clostridium difficile infection (CDI) is of particular concern among health care-associated infections. The role of the microbiota in disease recovery is apparent given the success of fecal microbiota transplantation (FMT) for recurrent CDI. Here, we present a murine model of CDI relapse to further define the microbiota recovery following FMT. Cefoperazone-treated mice were infected with C. difficile 630 spores and treated with vancomycin after development of clinical disease. Vancomycin treatment suppressed both C. difficile colonization and cytotoxin titers. However, C. difficile counts increased within 7 days of completing treatment, accompanied by relapse of clinical signs. The administration of FMT immediately after vancomycin cleared C. difficile and decreased cytotoxicity within 1 week. The effects of FMT on the gut microbiota community were detectable in recipients 1-day posttransplant. Conversely, mice not treated with FMT remained persistently colonized with high levels of C. difficile, and the gut microbiota in these mice persisted at low diversity. These results suggest that full recovery of colonization resistance against C. difficile requires the restoration of a specific community structure.


Assuntos
Doença Crônica/terapia , Clostridioides difficile/fisiologia , Infecções por Clostridium/terapia , Transplante de Microbiota Fecal , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Infecções por Clostridium/microbiologia , Fezes/microbiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota , Recidiva
18.
Infect Immun ; 83(3): 934-41, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25534943

RESUMO

Clostridium difficile infection (CDI) following antibiotic therapy is a major public health threat. While antibiotic disruption of the indigenous microbiota underlies the majority of cases of CDI, the early dynamics of infection in the disturbed intestinal ecosystem are poorly characterized. This study defines the dynamics of infection with C. difficile strain VPI 10463 throughout the gastrointestinal (GI) tract using a murine model of infection. After inducing susceptibility to C. difficile colonization via antibiotic administration, we followed the dynamics of spore germination, colonization, sporulation, toxin activity, and disease progression throughout the GI tract. C. difficile spores were able to germinate within 6 h postchallenge, resulting in the establishment of vegetative bacteria in the distal GI tract. Spores and cytotoxin activity were detected by 24 h postchallenge, and histopathologic colitis developed by 30 h. Within 36 h, all infected mice succumbed to infection. We correlated the establishment of infection with changes in the microbiota and bile acid profile of the small and large intestines. Antibiotic administration resulted in significant changes to the microbiota in the small and large intestines, as well as a significant shift in the abundance of primary and secondary bile acids. Ex vivo analysis suggested the small intestine as the site of spore germination. This study provides an integrated understanding of the timing and location of the events surrounding C. difficile colonization and identifies potential targets for the development of new therapeutic strategies.


Assuntos
Clostridioides difficile/patogenicidade , Infecções por Clostridium/patologia , Colite/patologia , Trato Gastrointestinal/patologia , Animais , Antibacterianos/efeitos adversos , Ácidos e Sais Biliares/química , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/metabolismo , Infecções por Clostridium/etiologia , Infecções por Clostridium/microbiologia , Infecções por Clostridium/mortalidade , Colite/etiologia , Colite/microbiologia , Colite/mortalidade , Progressão da Doença , Enterotoxinas/biossíntese , Enterotoxinas/metabolismo , Fezes/microbiologia , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/metabolismo , Esporos Bacterianos/patogenicidade , Análise de Sobrevida , Fatores de Tempo
19.
Immunology ; 144(4): 587-97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25327211

RESUMO

Our previous work has shown the significant up-regulation of Il22 and increased phosphorylation of signal transducer and activator of transcription 3 (STAT3) as part of the mucosal inflammatory response to Clostridium difficile infection in mice. Others have shown that phosphorylation of STAT3 at mucosal surfaces includes interleukin-22 (IL-22) and CD160-mediated components. The current study sought to determine the potential role(s) of IL-22 and/or CD160 in the mucosal response to C. difficile infection. Clostridium difficile-infected mice treated with anti-IL-22, anti-CD160 or a combination of the two showed significantly reduced STAT3 phosphorylation in comparison to C. difficile-infected mice that had not received either antibody. In addition, C. difficile-infected mice treated with anti-IL-22/CD160 induced a smaller set of genes, and at significantly lower levels than the untreated C. difficile-infected mice. The affected genes included pro-inflammatory chemokines and cytokines, and anti-microbial peptides. Furthermore, histopathological and flow cytometric assessments both showed a significantly reduced influx of neutrophils in C. difficile-infected mice treated with anti-IL-22/CD160. These data demonstrate that IL-22 and CD160 are together responsible for a significant fraction of the colonic STAT3 phosphorylation in C. difficile infection. They also underscore the additive effects of IL-22 and CD160 in mediating both the pro-inflammatory and pro-survival aspects of the host mucosal response in this infection.


Assuntos
Antígenos CD/imunologia , Clostridioides difficile/patogenicidade , Enterocolite Pseudomembranosa/imunologia , Imunidade nas Mucosas , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Receptores Imunológicos/imunologia , Animais , Antibacterianos , Anticorpos/farmacologia , Antígenos CD/genética , Antígenos CD/metabolismo , Clostridioides difficile/imunologia , Modelos Animais de Doenças , Enterocolite Pseudomembranosa/genética , Enterocolite Pseudomembranosa/metabolismo , Enterocolite Pseudomembranosa/microbiologia , Enterocolite Pseudomembranosa/prevenção & controle , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica , Imunidade nas Mucosas/efeitos dos fármacos , Interleucinas/antagonistas & inibidores , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Fosforilação , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Fator de Transcrição STAT3/imunologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fatores de Tempo , Interleucina 22
20.
Antimicrob Agents Chemother ; 59(3): 1596-604, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25547352

RESUMO

The glycylcycline antibiotic tigecycline was approved in 2005 for the treatment of complicated skin and soft tissue infections and complicated intra-abdominal infections. Tigecycline is broadly active against both Gram-negative and Gram-positive microorganisms, including Clostridium difficile. Tigecycline has a low MIC against C. difficile in vitro and thus may represent an alternate treatment for C. difficile infection (CDI). To assess the use of tigecycline for treatment of established CDI, 5- to 8-week-old C57BL/6 mice were colonized with C. difficile strain 630. After C. difficile colonization was established, mice (n = 10 per group) were treated with either a 5-day course of tigecycline (6.25 mg/kg every 12 h subcutaneously) or a 5-day course of vancomycin (0.4 mg/ml in drinking water) and compared to infected, untreated control mice. Mice were evaluated for clinical signs of CDI throughout treatment and at 1 week posttreatment to assess potential for disease development. Immediately following a treatment course, C. difficile was not detectable in the feces of vancomycin-treated mice but remained detectable in feces from tigecycline-treated and untreated control mice. Toxin activity and histopathological inflammation and edema were observed in the ceca and colons of untreated mice; tigecycline- and vancomycin-treated mice did not show such changes directly after treatment. One week after the conclusion of either antibiotic treatment, C. difficile load, toxin activity, and histopathology scores increased in the cecum and colon, indicating that C. difficile-associated disease occurred. In vitro growth studies confirmed that subinhibitory concentrations of tigecycline were able to suppress toxin activity and spore formation of C. difficile, whereas vancomycin did not. Taken together, these data show how tigecycline is able to alter C. difficile pathogenesis in a mouse model of CDI.


Assuntos
Antibacterianos/farmacologia , Clostridioides difficile/efeitos dos fármacos , Enterocolite Pseudomembranosa/tratamento farmacológico , Minociclina/análogos & derivados , Vancomicina/farmacologia , Animais , Ceco/microbiologia , Colo/microbiologia , Enterocolite Pseudomembranosa/microbiologia , Fezes/microbiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Minociclina/farmacologia , Tigeciclina
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa