Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
PLoS Biol ; 17(6): e3000334, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31206517

RESUMO

Escherichia coli represents a classical intestinal gram-negative commensal. Despite this commensalism, different E. coli strains can mediate disparate immunogenic properties in a given host. Symbiotic E. coli strains such as E. coli Nissle 1917 (EcN) are attributed beneficial properties, e.g., promotion of intestinal homeostasis. Therefore, we aimed to identify molecular features derived from symbiotic bacteria that might help to develop innovative therapeutic alternatives for the treatment of intestinal immune disorders. This study was performed using the dextran sodium sulphate (DSS)-induced colitis mouse model, which is routinely used to evaluate potential therapeutics for the treatment of Inflammatory Bowel Diseases (IBDs). We focused on the analysis of flagellin structures of different E. coli strains. EcN flagellin was found to harbor a substantially longer hypervariable region (HVR) compared to other commensal E. coli strains, and this longer HVR mediated symbiotic properties through stronger activation of Toll-like receptor (TLR)5, thereby resulting in interleukin (IL)-22-mediated protection of mice against DSS-induced colitis. Furthermore, using bone-marrow-chimeric mice (BMCM), CD11c+ cells of the colonic lamina propria (LP) were identified as the main mediators of these flagellin-induced symbiotic effects. We propose flagellin from symbiotic E. coli strains as a potential therapeutic to restore intestinal immune homeostasis, e.g., for the treatment of IBD patients.


Assuntos
Escherichia coli/metabolismo , Flagelina/genética , Simbiose/genética , Animais , Colite/induzido quimicamente , Colite/imunologia , Modelos Animais de Doenças , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Flagelina/metabolismo , Mucosa Intestinal , Intestinos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Simbiose/fisiologia , Receptor 5 Toll-Like/metabolismo
2.
Cell Microbiol ; 20(2)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29156489

RESUMO

The human gastrointestinal tract is a complex ecosystem in which epithelial cells and microorganisms of the intestinal microbiota live in symbiosis. Certain members of the microbiota, in particular Escherichia coli strains of the B2 phylotype, carry the polyketide synthase-island encoding the genotoxin colibactin. Colibactin is a nonribosomal peptide or polyketide-nonribosomal peptide hybrid of still unsolved structure, which induces DNA double strand breaks (DSBs) in eukaryotic cells. However, direct contact between live bacteria and host cell is required in order to elicit these genotoxic effects. In this study, we used a variety of cell culture models, among them, a 3D cell culture approach based on decellularised small intestinal submucosa, to investigate whether the intestinal mucus layer has the potential to interfere with colibactin activity. We demonstrate that the expression of mucins and the formation of an adherent mucus layer significantly increased with increasing complexity of cell culture. Moreover, we show that the presence of an adherent mucus layer on epithelial cells attenuates the genotoxic activity of colibactin, by preventing the induction of DNA-DSBs. Removal of the adherent mucus layer restored the occurrence of DNA-DSBs.


Assuntos
Trato Gastrointestinal/microbiologia , Muco/microbiologia , Mutagênicos/metabolismo , Peptídeos/metabolismo , Policetídeos/metabolismo , Linhagem Celular Tumoral , Dano ao DNA/fisiologia , Escherichia coli/patogenicidade , Microbioma Gastrointestinal/fisiologia , Ilhas Genômicas/fisiologia , Células HT29 , Humanos , Simbiose/fisiologia , Virulência/fisiologia
3.
Infect Immun ; 82(5): 1801-12, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24549324

RESUMO

Enteropathogenic Escherichia coli (EPEC) is recognized as an important intestinal pathogen that frequently causes acute and persistent diarrhea in humans and animals. The use of probiotic bacteria to prevent diarrhea is gaining increasing interest. The probiotic E. coli strain Nissle 1917 (EcN) is known to be effective in the treatment of several gastrointestinal disorders. While both in vitro and in vivo studies have described strong inhibitory effects of EcN on enteropathogenic bacteria, including pathogenic E. coli, the underlying molecular mechanisms remain largely unknown. In this study, we examined the inhibitory effect of EcN on infections of porcine intestinal epithelial cells with atypical enteropathogenic E. coli (aEPEC) with respect to single infection steps, including adhesion, microcolony formation, and the attaching and effacing phenotype. We show that EcN drastically reduced the infection efficiencies of aEPEC by inhibiting bacterial adhesion and growth of microcolonies, but not the attaching and effacing of adherent bacteria. The inhibitory effect correlated with EcN adhesion capacities and was predominantly mediated by F1C fimbriae, but also by H1 flagella, which served as bridges between EcN cells. Furthermore, EcN seemed to interfere with the initial adhesion of aEPEC to host cells by secretion of inhibitory components. These components do not appear to be specific to EcN, but we propose that the strong adhesion capacities enable EcN to secrete sufficient local concentrations of the inhibitory factors. The results of this study are consistent with a mode of action whereby EcN inhibits secretion of virulence-associated proteins of EPEC, but not their expression.


Assuntos
Escherichia coli Enteropatogênica/fisiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli/classificação , Proteínas de Fímbrias/metabolismo , Flagelos/fisiologia , Probióticos/farmacologia , Animais , Aderência Bacteriana , Linhagem Celular , Escherichia coli Enteropatogênica/patogenicidade , Escherichia coli Enteropatogênica/ultraestrutura , Células Epiteliais/microbiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Fímbrias/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Mucosa Intestinal/citologia , Suínos , Virulência
4.
Int J Med Microbiol ; 303(1): 1-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23312798

RESUMO

The largest EHEC outbreak up to now in Germany occurred in 2011. It was caused by the non-O157:H7 Shiga-toxinogenic enterohemorrhagic E. coli strain O104:H4. This strain encodes in addition to the Shiga toxin 2 (Stx2), responsible for the hemolytic uremic syndrome (HUS), several adhesins such as aggregative adherence fimbriae. Currently, there is no effective prophylaxis and treatment available for EHEC infections in humans. Especially antibiotics are not indicated for treatment as they may induce Stx production, thus worsening the symptoms. Alternative therapeutics are therefore desperately needed. We tested the probiotic Escherichia coli strain Nissle 1917 (EcN) for antagonistic effects on two O104:H4 EHEC isolates from the 2011 outbreak and on the classical O157:H7 EHEC strain EDL933. These tests included effects on adherence, growth, and Stx production in monoculture and co-culture together with EcN. The inoculum of each co-culture contained EcN and the respective EHEC strain either at a ratio of 1:1 or 10:1 (EcN:EHEC). Adhesion of EHEC strains to Caco-2 cells and to the mucin-producing LS-174T cells was reduced significantly in co-culture with EcN at the 1:1 ratio and very dramatically at the 10:1 ratio. This inhibitory effect of EcN on EHEC adherence was most likely not due to occupation of adhesion sites on the epithelial cells, because in monocultures EcN adhered with much lower bacterial numbers than the EHEC strains. Both EHEC strains of serotype O104:H4 showed reduced growth in the presence of EcN (10:1 ratio). EHEC strain EDL933 grew in co-culture with EcN only during the first 2h of incubation. Thereafter, EHEC counts declined. At 24h, the numbers of viable EDL933 was at or slightly below the numbers at the time of inoculation. The amount of Stx2 after 24h co-incubation with EcN (EcN:EHEC ratio 10:1) was for all 3 EHEC strains tested significantly reduced in comparison to EHEC monocultures. Obviously, EcN shows very efficient antagonistic activity on the EHEC strains of serotype O104:H4 and O157:H7 tested here regarding adherence to human gut epithelial cells, bacterial growth, and Stx2 production in vitro.


Assuntos
Escherichia coli Êntero-Hemorrágica/fisiologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/prevenção & controle , Escherichia coli/fisiologia , Probióticos , Aderência Bacteriana/fisiologia , Células CACO-2 , Linhagem Celular Tumoral , Técnicas de Cocultura , Surtos de Doenças/prevenção & controle , Escherichia coli Êntero-Hemorrágica/crescimento & desenvolvimento , Células Epiteliais/microbiologia , Escherichia coli/crescimento & desenvolvimento , Infecções por Escherichia coli/epidemiologia , Escherichia coli O157/crescimento & desenvolvimento , Escherichia coli O157/fisiologia , Alemanha/epidemiologia , Humanos , Toxina Shiga II/metabolismo
5.
Int J Med Microbiol ; 302(6): 276-87, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22748509

RESUMO

The probiotic Escherichia coli strain Nissle 1917 (EcN) is one of the few probiotics licensed as a medication in several countries. Best documented is its effectiveness in keeping patients suffering from ulcerative colitis (UC) in remission. This might be due to its ability to induce the production of human ß-defensin 2 (HBD2) in a flagellin-dependent way in intestinal epithelial cells. In contrast to ulcerative colitis, for Crohn's disease (CD) convincing evidence is lacking that EcN might be clinically effective, most likely due to the genetically based inability of sufficient defensin production in CD patients. As a first step in the development of an alternative approach for the treatment of CD patients, EcN strains were constructed which were able to produce human α-defensin 5 (HD5) or ß-defensin 2 (HBD2). For that purpose, codon-optimized defensin genes encoding either the proform with the signal sequence of human α-defensin 5 (HD5) or the gene encoding HBD2 with or without the signal sequence were cloned in an expression vector plasmid under the control of the T7 promoter. Synthesis of the encoded defensins was shown by Western blots after induction of expression and lysis of the recombinant EcN strains. Recombinant mature HBD2 with an N-terminal His-tag could be purified by Ni-column chromatography and showed antimicrobial activity against E. coli, Salmonella enterica serovar Typhimurium and Listeria monocytogenes. In a second approach, that part of the HBD2 gene which encodes mature HBD2 was fused with the yebF gene. The resulting fusion protein YebFMHBD2 was secreted from the encoding EcN mutant strain after induction of expression. Presence of YebFMHBD2 in the medium was not the result of leakage from the bacterial cells, as demonstrated in the spent culture supernatant by Western blots specific for ß-galactosidase and maltose-binding protein. The dialyzed and concentrated culture supernatant inhibited the growth of E. coli, S. enterica serovar Typhimurium and L. monocytogenes in radial diffusion assays as well as in liquid culture. This demonstrates EcN to be a suitable probiotic E. coli strain for the production of certain defensins.


Assuntos
Escherichia coli/genética , Engenharia Genética/métodos , alfa-Defensinas/biossíntese , beta-Defensinas/biossíntese , Antibacterianos/isolamento & purificação , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Western Blotting , Cromatografia de Afinidade , Clonagem Molecular , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Vetores Genéticos/genética , Humanos , Listeria monocytogenes/efeitos dos fármacos , Plasmídeos/genética , Probióticos/metabolismo , Regiões Promotoras Genéticas , Sinais Direcionadores de Proteínas , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Salmonella typhimurium/efeitos dos fármacos , Transformação Genética , alfa-Defensinas/genética , beta-Defensinas/genética
6.
Int J Med Microbiol ; 302(7-8): 304-14, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23131416

RESUMO

The flagellum of the probiotic Escherichia coli strain Nissle 1917 (EcN) is not just responsible for motility, but also for EcN's ability to induce the production of human ß-defensin 2. Here, we report a third function of this EcN organell. In this study we investigated the role of the EcN flagellum in adhesion to different host tissues by ex vivo and in vitro studies. Ex vivo studies with cryosections of human gut biopsies revealed that the flagellum of EcN is most likely important for efficient adhesion to the human intestinal tract. These results and in vitro studies with different epithelial cells indicated that the presence of mucus is important for efficient mediation of adhesion by the flagellum of EcN. We observed direct interaction between isolated flagella from EcN wild type and porcine mucin 2 as well as human mucus. However, we could not observe any interaction of the flagella with murine mucus. For the first time, we identified the mucus component gluconate as one receptor for the binding of flagella from EcN and were able to exclude the flagellin domain D3 as a responsible interaction partner. We propose that the flagellum of EcN is its major adhesin in vivo, which enables this probiotic strain to compete efficiently for binding sites on host tissue with several bacterial pathogens.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Escherichia coli/fisiologia , Flagelos/fisiologia , Muco/microbiologia , Animais , Feminino , Gluconatos/metabolismo , Humanos , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Muco/química , Suínos
7.
Int J Med Microbiol ; 300(1): 57-62, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19783474

RESUMO

Probiotics are gaining more and more interest as alternatives for antibiotics or anti-inflammatory drugs. However, their mode of action is poorly understood. This review will present examples of probiotic actions from three general modes of actions into which probiotic effects can be classified. Probiotics might modulate the host's immune system, affect other microorganisms directly or act on microbial products, host products or food components. What kind of effect(s) a certain probiotic executes depends on its metabolic properties, the molecules presented at its surface or on the components secreted. Even integral parts of the bacterial cell such as its DNA or peptidoglycan might be of importance for its probiotic effectiveness. The individual combination of such properties in a certain probiotic strain determines its specific probiotic action and as a consequence its effective application for the prevention and/or treatment of a certain disease.


Assuntos
Anti-Infecciosos/farmacologia , Antimetabólitos/farmacologia , Bactérias/imunologia , Bactérias/metabolismo , Fatores Imunológicos/farmacologia , Probióticos/farmacologia , Humanos
8.
Int J Med Microbiol ; 300(7): 449-56, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20547100

RESUMO

Despite promising results and increasing attention in bacterial cancer therapy, surprisingly little is known about initial tumor colonization and the interaction between bacteria and surrounding tumor tissue. Here, we analyzed the role of chemotaxis, motility, and metabolism both in Escherichia coli and Salmonella enterica serovar Typhimurium strains upon intravenous injection into tumor-bearing mice. In contrast to previous models, we found that chemotaxis and motility do not play a significant role in tumor colonization and bacterial distribution within the tumor. Rather, the whole colonization and intratumoral migration process seems to be a passive mechanism that is influenced by the reticuloendothelial system of the host, by the tumor microenvironment and by the bacterial metabolism. These conclusions were supported by experimental data demonstrating that disruption of the basic branch of the aromatic amino acid biosynthetic pathway and depletion of macrophages, in contrast to flagellar mutations, led to significant changes in bacterial accumulation in tumors of live mice.


Assuntos
Quimiotaxia , Escherichia coli/fisiologia , Locomoção , Macrófagos/microbiologia , Neoplasias/microbiologia , Salmonella typhimurium/fisiologia , Aminoácidos Aromáticos/metabolismo , Animais , Carga Bacteriana , Vias Biossintéticas/genética , Escherichia coli/imunologia , Escherichia coli/metabolismo , Feminino , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Neoplasias/imunologia , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo
9.
Infect Immun ; 77(11): 4696-703, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19720753

RESUMO

A genomic island encoding the biosynthesis and secretion pathway of putative hybrid nonribosomal peptide-polyketide colibactin has been recently described in Escherichia coli. Colibactin acts as a cyclomodulin and blocks the eukaryotic cell cycle. The origin and prevalence of the colibactin island among enterobacteria are unknown. We therefore screened 1,565 isolates of different genera and species related to the Enterobacteriaceae by PCR for the presence of this DNA element. The island was detected not only in E. coli but also in Klebsiella pneumoniae, Enterobacter aerogenes, and Citrobacter koseri isolates. It was highly conserved among these species and was always associated with the yersiniabactin determinant. Structural variations between individual strains were only observed in an intergenic region containing variable numbers of tandem repeats. In E. coli, the colibactin island was usually restricted to isolates of phylogenetic group B2 and inserted at the asnW tRNA locus. Interestingly, in K. pneumoniae, E. aerogenes, C. koseri, and three E. coli strains of phylogenetic group B1, the functional colibactin determinant was associated with a genetic element similar to the integrative and conjugative elements ICEEc1 and ICEKp1 and to several enterobacterial plasmids. Different asn tRNA genes served as chromosomal insertion sites of the ICE-associated colibactin determinant: asnU in the three E. coli strains of ECOR group B1, and different asn tRNA loci in K. pneumoniae. The detection of the colibactin genes associated with an ICE-like element in several enterobacteria provides new insights into the spread of this gene cluster and its putative mode of transfer. Our results shed light on the mechanisms of genetic exchange between members of the family Enterobacteriaceae.


Assuntos
DNA Bacteriano/genética , Enterobacteriaceae/genética , Proteínas de Escherichia coli/genética , Genes Bacterianos/genética , Ilhas Genômicas/genética , Sequência de Bases , Citometria de Fluxo , Imunofluorescência , Transferência Genética Horizontal , Células HeLa , Humanos , Microscopia Confocal , Dados de Sequência Molecular , Fenótipo , Reação em Cadeia da Polimerase
10.
Int J Med Microbiol ; 299(7): 467-78, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19467927

RESUMO

Enterotoxigenic Escherichia coli (ETEC) are a leading cause of diarrhoea in piglets and newborn calves. Massive efforts have therefore been made to develop a vaccine for the induction of protective mucosal immunity against ETEC. Since it has been shown that the probiotic strain E. coli Nissle 1917 (EcN) can serve as a safe carrier for targeted delivery of recombinant molecules to the intestinal mucosa, we constructed the recombinant strain EcN pMut2-kanK88 (EcN-K88) stably expressing the determinant for the K88 fimbrial adhesin of ETEC on the bacterial surface. After oral application of EcN-K88 to mice for one week, EcN-K88 as well as wild-type EcN and EcN mock-transformed with the plasmid vector only could be detected in faecal samples for a minimum of 7 days after the last feeding, indicating that EcN can transiently colonise the murine intestine. Oral application of EcN-K88 resulted in significant IgG serum titres against K88 as early as 7 days after the initial feeding with EcN-K88, but no significant IgA titres. In contrast, we failed to detect any specific T cell responses towards the K88 antigen both in spleen and mesenteric lymph nodes. Although dendritic cells readily upregulated maturation and activation markers in response to K88 stimulation, accompanied by secretion of interleukin (IL)-12, IL-6, IL-10, and tumour necrosis factor, restimulation of T cells from mice having received EcN-K88 with K88-loaded dendritic cells did not result in detectable T cell proliferation and IL-2 secretion, but rather induced an IL-10 bias. While the serum antibody responses clearly demonstrate that K88 is recognized by the humoral immune system, our findings indicate that oral application of probiotic EcN expressing the K88 fimbrial adhesin does not induce a selective T cell response towards the antigen.


Assuntos
Antígenos de Bactérias/biossíntese , Antígenos de Bactérias/imunologia , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/imunologia , Vacinas contra Escherichia coli/imunologia , Escherichia coli/imunologia , Proteínas de Fímbrias/biossíntese , Proteínas de Fímbrias/imunologia , Administração Oral , Animais , Anticorpos Antibacterianos/sangue , Contagem de Colônia Microbiana , Citocinas/metabolismo , Vacinas contra Escherichia coli/administração & dosagem , Fezes/microbiologia , Feminino , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/imunologia , Linfócitos T/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
11.
Int Arch Allergy Immunol ; 149(3): 219-30, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19218814

RESUMO

BACKGROUND: Recent clinical trials, epidemiological studies and animal experiments have suggested that probiotics may help suppress the development of allergic responses. OBJECTIVE: To investigate whether the application of the probiotic Escherichia coli strain Nissle 1917 (EcN) protects mice from developing ovalbumin (OVA)-specific T helper-2 responses in the airways. METHODS: OVA-specific Th2 responses were induced by 2 intraperitoneal (i.p.) injections with OVA/alum followed by 1 intranasal (i.n.) challenge with OVA. EcN was given orally during the entire sensitization and challenge period, together with OVA/alum during the i.p. sensitizations, or i.n. before or during the airway challenge with OVA. RESULTS: We found that when the bacteria were given together with OVA/alum airway eosinophilia, airway hyper-reactivity, goblet cell metaplasia and IL-5 levels in the bronchoalveolar lavage and mediastinal lymph node cell cultures were reduced. This effect was associated with increased numbers of IFN-gamma producing T helper-1 cells and IFN-gamma levels in the airways and strongly increased OVA-specific IgG(2a) titers in the serum. The suppressive effect on airway eosinophilia was dependent on IFN-gamma but not TLR-4. Applying EcN i.n. or orally did not reduce the development of allergen-specific Th2 responses. CONCLUSIONS: Our results suggest that EcN can inhibit the development of allergic responses when the bacteria are present at the site of Th2 cell priming and that this immunomodulatory effect is due to a shift from Th2 to Th1 response. The data support the hypothesis that probiotics may help reduce allergic responses and that EcN may also be used as adjuvant therapy to induce allergen-specific Th1 responses.


Assuntos
Hiper-Reatividade Brônquica/prevenção & controle , Células Dendríticas/imunologia , Escherichia coli/imunologia , Hipersensibilidade/imunologia , Probióticos/uso terapêutico , Células Th2/imunologia , Adjuvantes Imunológicos/farmacologia , Administração Intranasal , Administração Oral , Alérgenos/imunologia , Compostos de Alúmen/farmacologia , Animais , Hiper-Reatividade Brônquica/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/microbiologia , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Eosinofilia/imunologia , Eosinofilia/metabolismo , Eosinofilia/microbiologia , Feminino , Células Caliciformes/imunologia , Células Caliciformes/patologia , Hipersensibilidade/metabolismo , Hipersensibilidade/microbiologia , Hipersensibilidade/prevenção & controle , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-5/biossíntese , Interleucina-5/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/microbiologia , Células Th2/metabolismo , Células Th2/microbiologia
12.
Front Microbiol ; 10: 2783, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31849915

RESUMO

Rapidly growing antibiotic resistance among gastrointestinal pathogens, and the ability of antibiotics to induce the virulence of these pathogens makes it increasingly difficult to rely on antibiotics to treat gastrointestinal infections. The probiotic Escherichia coli strain Nissle 1917 (EcN) is the active component of the pharmaceutical preparation Mutaflor® and has been successfully used in the treatment of gastrointestinal disorders. Gut bacteriophages are dominant players in maintaining the microbial homeostasis in the gut, however, their interaction with incoming probiotic bacteria remains to be at conception. The presence of bacteriophages in the gut makes it inevitable for any probiotic bacteria to be phage resistant, in order to survive and successfully colonize the gut. This study addresses the phage resistance of EcN, specifically against lytic T4 phage infection. From various experiments we could show that (i) EcN is resistant toward T4 phage infection, (ii) EcN's K5 polysaccharide capsule plays a crucial role in T4 phage resistance and (iii) EcN's lipopolysaccharide (LPS) inactivates T4 phages and notably, treatment with the antibiotic polymyxin B which neutralizes the LPS destroyed the phage inactivation ability of isolated LPS from EcN. Combination of these identified properties in EcN was not found in other tested commensal E. coli strains. Our results further indicated that N-acetylglucosamine at the distal end of O6 antigen in EcN's LPS could be the interacting partner with T4 phages. From our findings, we have reported for the first time, the role of EcN's K5 capsule and LPS in its defense against T4 phages. In addition, by inactivating the T4 phages, EcN also protects E. coli K-12 strains from phage infection in tri-culture experiments. Our research highlights phage resistance as an additional safety feature of EcN, a clinically successful probiotic E. coli strain.

13.
Lancet Infect Dis ; 19(7): e237-e245, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31031171

RESUMO

Natural products have been a rich source of compounds with structural and chemical diversity for drug discovery. However, antibiotic resistance in bacteria has been reported for nearly every antibiotic once it is used in clinical practice. In the past decade, pharmaceutical companies have reduced their natural product discovery projects because of challenges, such as high costs, low return rates, and high rediscovery rates. The largely unexplored marine environment harbours substantial diversity and is a large resource to discover novel compounds with novel modes of action, which is essential for the treatment of drug-resistant bacterial infections. In this Review, we report compounds derived from marine sources that have shown in-vivo and in-vitro efficacy against drug-resistant bacteria. Analysis of the physicochemical properties of these marine natural products with activity against drug-resistant bacteria showed that 60% of the compounds have oral bioavailability potential. Their overall distribution pattern of drug characteristics agrees with the observation that marketed antibacterial drugs have a polar distribution, with a lower median calculated logP. The aim of this Review is to summarise the diversity of these marine natural products, with a special focus on analysis of drug bioavailability. Such biologically active compounds, with high degrees of bioavailability, have the potential to be developed as effective drugs against infectious diseases.


Assuntos
Antibacterianos , Organismos Aquáticos/efeitos dos fármacos , Infecções Bacterianas/tratamento farmacológico , Produtos Biológicos/uso terapêutico , Descoberta de Drogas , Farmacorresistência Bacteriana/efeitos dos fármacos , Antibacterianos/química , Antibacterianos/farmacologia , Organismos Aquáticos/química , Produtos Biológicos/química , Humanos
14.
Bio Protoc ; 8(12): e2879, 2018 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-34285991

RESUMO

The mucosal surfaces of the gastrointestinal, respiratory, reproductive, and urinary tracts, and the surface of the eye harbor a resident microflora that lives in symbiosis with their host and forms a complex ecosystem. The protection of the vulnerable epithelium is primarily achieved by mucins that form a gel-like structure adherent to the apical cell surface. This mucus layer constitutes a physical and chemical barrier between the microbial flora and the underlying epithelium. Mucus is critical to the maintenance of a homeostatic relationship between the microbiota and its host. Subtle deviations from this dynamic interaction may result in major implications for health. The protocol in this article describes the procedures to grow low mucus-producing HT29 and high mucus-producing HT29-MTX-E12 cells, maintain cells and use them for mucus quantification by ELISA. Additionally, it is described how to assess the amount of secreted adherent mucus. This system can be used to study the protective effect of mucus, e.g., against bacterial toxins, to test the effect of different culture conditions on mucus production or to analyze diffusion of molecules through the mucus layer. Since the ELISA used in this protocol is available for different species and mucus proteins, also other cell types can be used.

15.
Fitoterapia ; 125: 191-198, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29108932

RESUMO

In previous studies the aerial parts of Achillea fragrantissima were found to have substantial antileishmanial and antitrypanosomal activity. A bioassay-guided fractionation of a dichloromethane extract yielded the isolation of the essential anti-trypanosomal compounds of the plant. Seven sesquiterpene lactones (including Achillolide-A), two flavonoids, chrysosplenol-D and chrysosplenetine, and four alkamides (including pellitorine) were identified. This is the first report for the isolation of the sesquiterpene lactones 3 and 4, chrysosplenetine and the group of alkamides from this plant. Bioevaluation against Trypanosoma brucei brucei TC221 (T.b brucei) using the Alamar-Blue assay revealed the novel alkamide 13 to have an IC50 value of 40.37µM. A compound library, derived from the alkamide pellitorine (10), was synthesized and bioevaluated in order to find even more active substances. The most active compounds 26 and 27 showed activities in submicromolar concentrations and selectivity indices of 20.1 and 45.6, respectively, towards macrophage cell line J774.1. Toxicity of 26 and 27 was assessed using the greater wax moth Galleria mellonella larvae as an in vivo model. No significant toxicity was observed for the concentration range of 1.25-20mM.


Assuntos
Achillea/química , Ácidos Graxos Insaturados/farmacologia , Flavonas/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Animais , Linhagem Celular , Ácidos Graxos Insaturados/isolamento & purificação , Flavonas/isolamento & purificação , Camundongos , Estrutura Molecular , Mariposas , Componentes Aéreos da Planta/química , Alcamidas Poli-Insaturadas/isolamento & purificação , Testes de Toxicidade , Tripanossomicidas/isolamento & purificação
16.
Front Microbiol ; 9: 929, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29896160

RESUMO

Shiga toxin (Stx) producing E. coli (STEC) such as Enterohemorrhagic E. coli (EHEC) are the major cause of foodborne illness in humans. In vitro studies showed the probiotic Escherichia coli strain Nissle 1917 (EcN) to efficiently inhibit the production of Stx. Life threatening EHEC strains as for example the serotype O104:H4, responsible for the great outbreak in 2011 in Germany, evolutionary developed from certain E. coli strains which got infected by stx2-encoding lambdoid phages turning the E. coli into lysogenic and subsequently Stx producing strains. Since antibiotics induce stx genes and Stx production, EHEC infected persons are not recommended to be treated with antibiotics. Therefore, EcN might be an alternative medication. However, because even commensal E. coli strains might be converted into Stx-producers after becoming host to a stx encoding prophage, we tested EcN for stx-phage genome integration. Our experiments revealed the resistance of EcN toward not only stx-phages but also against lambda-phages. This resistance was not based on the lack of or by mutated phage receptors. Rather it involved the expression of a phage repressor (pr) gene of a defective prophage in EcN which was able to partially protect E. coli K-12 strain MG1655 against stx and lambda phage infection. Furthermore, we observed EcN to inactivate phages and thereby to protect E. coli K-12 strains against infection by stx- as well as lambda-phages. Inactivation of lambda-phages was due to binding of lambda-phages to LamB of EcN whereas inactivation of stx-phages was caused by a thermostable protein of EcN. These properties together with its ability to inhibit Stx production make EcN a good candidate for the prevention of illness caused by EHEC and probably for the treatment of already infected people.

17.
Front Microbiol ; 9: 1473, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30050506

RESUMO

Staphylococcus epidermidis, the common inhabitant of human skin and mucosal surfaces has emerged as an important pathogen in patients carrying surgical implants and medical devices. Entering the body via surgical sites and colonizing the medical devices through formation of multi-layered biofilms leads to refractory and persistent device-related infections (DRIs). Staphylococci organized in biofilms are more tolerant to antibiotics and immune responses, and thus are difficult-to-treat. The consequent morbidity and mortality, and economic losses in health care systems has strongly necessitated the need for development of new anti-bacterial and anti-biofilm-based therapeutics. In this study, we describe the biological activity of a marine sponge-derived Streptomyces sp. SBT348 extract in restraining staphylococcal growth and biofilm formation on polystyrene, glass, medically relevant titan metal, and silicone surfaces. A bioassay-guided fractionation was performed to isolate the active compound (SKC3) from the crude SBT348 extract. Our results demonstrated that SKC3 effectively inhibits the growth (MIC: 31.25 µg/ml) and biofilm formation (sub-MIC range: 1.95-<31.25 µg/ml) of S. epidermidis RP62A in vitro. Chemical characterization of SKC3 by heat and enzyme treatments, and mass spectrometry (HRMS) revealed its heat-stable and non-proteinaceous nature, and high molecular weight (1258.3 Da). Cytotoxicity profiling of SKC3 in vitro on mouse fibroblast (NIH/3T3) and macrophage (J774.1) cell lines, and in vivo on the greater wax moth larvae Galleria mellonella revealed its non-toxic nature at the effective dose. Transcriptome analysis of SKC3 treated S. epidermidis RP62A has further unmasked its negative effect on central metabolism such as carbon flux as well as, amino acid, lipid, and energy metabolism. Taken together, these findings suggest a potential of SKC3 as a putative drug to prevent staphylococcal DRIs.

18.
Lancet Infect Dis ; 17(2): e30-e41, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27979695

RESUMO

Antibiotics have revolutionised medicine in many aspects, and their discovery is considered a turning point in human history. However, the most serious consequence of the use of antibiotics is the concomitant development of resistance against them. The marine environment has proven to be a very rich source of diverse natural products with significant antibacterial, antifungal, antiviral, antiparasitic, antitumour, anti-inflammatory, antioxidant, and immunomodulatory activities. Many marine natural products (MNPs)-for example, neoechinulin B-have been found to be promising drug candidates to alleviate the mortality and morbidity rates caused by drug-resistant infections, and several MNP-based anti-infectives have already entered phase 1, 2, and 3 clinical trials, with six approved for usage by the US Food and Drug Administration and one by the EU. In this Review, we discuss the diversity of marine natural products that have shown in-vivo efficacy or in-vitro potential against drug-resistant infections of fungal, viral, and parasitic origin, and describe their mechanism of action. We highlight the drug-like physicochemical properties of the reported natural products that have bioactivity against drug-resistant pathogens in order to assess their drug potential. Difficulty in isolation and purification procedures, toxicity associated with the active compound, ecological impacts on natural environment, and insufficient investments by pharmaceutical companies are some of the clear reasons behind market failures and a poor pipeline of MNPs available to date. However, the diverse abundance of natural products in the marine environment could serve as a ray of light for the therapy of drug-resistant infections. Development of resistance-resistant antibiotics could be achieved via the coordinated networking of clinicians, microbiologists, natural product chemists, and pharmacologists together with pharmaceutical venture capitalist companies.


Assuntos
Anti-Infecciosos/administração & dosagem , Produtos Biológicos/uso terapêutico , Drogas em Investigação , Biologia Marinha , Animais , Farmacorresistência Bacteriana , Farmacorresistência Fúngica , Drogas em Investigação/uso terapêutico , Humanos , Micoses , Doenças Parasitárias , Estados Unidos , Viroses
19.
Front Microbiol ; 8: 236, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28261188

RESUMO

Staphylococcus epidermidis and Staphylococcus aureus are opportunistic pathogens that cause nosocomial and chronic biofilm-associated infections. Indwelling medical devices and contact lenses are ideal ecological niches for formation of staphylococcal biofilms. Bacteria within biofilms are known to display reduced susceptibilities to antimicrobials and are protected from the host immune system. High rates of acquired antibiotic resistances in staphylococci and other biofilm-forming bacteria further hamper treatment options and highlight the need for new anti-biofilm strategies. Here, we aimed to evaluate the potential of marine sponge-derived actinomycetes in inhibiting biofilm formation of several strains of S. epidermidis, S. aureus, and Pseudomonas aeruginosa. Results from in vitro biofilm-formation assays, as well as scanning electron and confocal microscopy, revealed that an organic extract derived from the marine sponge-associated bacterium Streptomyces sp. SBT343 significantly inhibited staphylococcal biofilm formation on polystyrene, glass and contact lens surfaces, without affecting bacterial growth. The extract also displayed similar antagonistic effects towards the biofilm formation of other S. epidermidis and S. aureus strains tested but had no inhibitory effects towards Pseudomonas biofilms. Interestingly the extract, at lower effective concentrations, did not exhibit cytotoxic effects on mouse fibroblast, macrophage and human corneal epithelial cell lines. Chemical analysis by High Resolution Fourier Transform Mass Spectrometry (HRMS) of the Streptomyces sp. SBT343 extract proportion revealed its chemical richness and complexity. Preliminary physico-chemical characterization of the extract highlighted the heat-stable and non-proteinaceous nature of the active component(s). The combined data suggest that the Streptomyces sp. SBT343 extract selectively inhibits staphylococcal biofilm formation without interfering with bacterial cell viability. Due to absence of cell toxicity, the extract might represent a good starting material to develop a future remedy to block staphylococcal biofilm formation on contact lenses and thereby to prevent intractable contact lens-mediated ocular infections.

20.
Nat Prod Res ; 31(23): 2818-2823, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28287277

RESUMO

The production of shiga toxin (Stx) is a critical step in the establishment and progress of enterohemorrhagic Escherichia coli (EHEC) infections. The possible release of Stx from dead and dying bacteria, and the risk of resistance development have restricted the usage of antibiotics against EHEC. The chlorinated quaternary ammonium compound, strepthonium A, was isolated from the culture of Streptomyces sp. SBT345 that was cultivated from the Mediterranean sponge Agelas oroides. The structure was elucidated and confirmed by spectroscopic analyses including 1D and 2D NMR, ESI-HRMS, as well as ESI-HRMS2. Strepthonium A follows Lipinski's rule of five with respect to its molecular weight, CLogP values and the number of hydrogen acceptors and donors. Verotoxin ELISA assay demonstrated that Strepthonium A reduced the Stx production in EHEC strain EDL933 at 80 µM concentration without growth inhibition. This study demonstrates the potential of strepthonium A in restraining the production of Stx in EHEC infections.


Assuntos
Antibacterianos/farmacologia , Escherichia coli Êntero-Hemorrágica/efeitos dos fármacos , Toxina Shiga/metabolismo , Streptomyces/metabolismo , Antibacterianos/química , Escherichia coli Êntero-Hemorrágica/crescimento & desenvolvimento , Escherichia coli Êntero-Hemorrágica/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Espectroscopia de Ressonância Magnética , Estrutura Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA