Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(24): e2401686121, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38838019

RESUMO

S-layers are crystalline arrays found on bacterial and archaeal cells. Lactobacillus is a diverse family of bacteria known especially for potential gut health benefits. This study focuses on the S-layer proteins from Lactobacillus acidophilus and Lactobacillus amylovorus common in the mammalian gut. Atomic resolution structures of Lactobacillus S-layer proteins SlpA and SlpX exhibit domain swapping, and the obtained assembly model of the main S-layer protein SlpA aligns well with prior electron microscopy and mutagenesis data. The S-layer's pore size suggests a protective role, with charged areas aiding adhesion. A highly similar domain organization and interaction network are observed across the Lactobacillus genus. Interaction studies revealed conserved binding areas specific for attachment to teichoic acids. The structure of the SlpA S-layer and the suggested incorporation of SlpX as well as its interaction with teichoic acids lay the foundation for deciphering its role in immune responses and for developing effective treatments for a variety of infectious and bacteria-mediated inflammation processes, opening opportunities for targeted engineering of the S-layer or lactobacilli bacteria in general.


Assuntos
Glicoproteínas de Membrana , Ácidos Teicoicos , Ácidos Teicoicos/metabolismo , Ácidos Teicoicos/química , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/química , Lactobacillus/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Modelos Moleculares , Lactobacillus acidophilus/metabolismo , Lactobacillus acidophilus/genética
2.
Anaerobe ; 62: 102104, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31562947

RESUMO

In our previous studies on irritable bowel syndrome (IBS) -associated microbiota by molecular methods, we demonstrated that a particular 16S rRNA gene amplicon was more abundant in the feces of healthy subjects or mixed type IBS (IBS-M) -sufferers than in the feces of individuals with diarrhea-type IBS (IBS-D). In the current study, we demonstrated that this, so called Ct85-amplicon, consists of a cluster of very heterogeneous 16S rRNA gene sequences, and defined six 16S rRNA gene types, a to f, within this cluster, each representing a novel species-, genus- or family level taxon. We then designed specific PCR primers for these sequence types, mapped the distribution of the Ct85-cluster sequences and that of the newly defined sequence types in several animal species and compared the sequence types present in the feces of healthy individuals and IBS sufferers using two IBS study cohorts, Finnish and Dutch. Various Ct85-cluster sequence types were detected in the fecal samples of several companion and production animal species with remarkably differing prevalences and abundances. The Ct85 sequence type composition of swine closely resembled that of humans. One of the five types (d) shared between humans and swine was not present in any other animals tested, while one sequence type (b) was found only in human samples. In both IBS study cohorts, one type (e) was more prevalent in healthy individuals than in the IBS-M group. By revealing various sequence types in the widespread Ct85-cluster and their distribution, the results improve our understanding of these uncultured bacteria, which is essential for future efforts to cultivate representatives of the Ct85-cluster and reveal their roles in IBS.


Assuntos
Microbioma Gastrointestinal , Metagenoma , Metagenômica , Animais , Análise por Conglomerados , Bases de Dados Genéticas , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Mamíferos , Metagenômica/métodos , Tipagem Molecular , Filogenia , RNA Ribossômico 16S/genética
3.
BMC Microbiol ; 16(1): 226, 2016 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-27688074

RESUMO

BACKGROUND: The role of Lactobacillus cell wall components in the protection against pathogen infection in the gut is still largely unexplored. We have previously shown that L. amylovorus DSM 16698T is able to reduce the enterotoxigenic F4+ Escherichia coli (ETEC) adhesion and prevent the pathogen-induced membrane barrier disruption through the regulation of IL-10 and IL-8 expression in intestinal cells. We have also demonstrated that L. amylovorus DSM 16698T protects host cells through the inhibition of NF-kB signaling. In the present study, we investigated the role of L. amylovorus DSM 16698T cell wall components in the protection against F4+ETEC infection using the intestinal Caco-2 cell line. METHODS: Purified cell wall fragments (CWF) from L. amylovorus DSM 16698T were used either as such (uncoated, U-CWF) or coated with S-layer proteins (S-CWF). Differentiated Caco-2/TC7 cells on Transwell filters were infected with F4+ETEC, treated with S-CWF or U-CWF, co-treated with S-CWF or U-CWF and F4+ETEC for 2.5 h, or pre-treated with S-CWF or U-CWF for 1 h before F4+ETEC addition. Tight junction (TJ) and adherens junction (AJ) proteins were analyzed by immunofluorescence and Western blot. Membrane permeability was determined by phenol red passage. Phosphorylated p65-NF-kB was measured by Western blot. RESULTS: We showed that both the pre-treatment with S-CWF and the co- treatment of S-CWF with the pathogen protected the cells from F4+ETEC induced TJ and AJ injury, increased membrane permeability and activation of NF-kB expression. Moreover, the U-CWF pre-treatment, but not the co-treatment with F4+ETEC, inhibited membrane damage and prevented NF-kB activation. CONCLUSIONS: The results indicate that the various components of L. amylovorus DSM 16698T cell wall may counteract the damage caused by F4+ETEC through different mechanisms. S-layer proteins are essential for maintaining membrane barrier function and for mounting an anti-inflammatory response against F4+ETEC infection. U-CWF are not able to defend the cells when they are infected with F4+ETEC but may activate protective mechanisms before pathogen infection.

4.
Anaerobe ; 39: 60-7, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26946362

RESUMO

In our previous studies on the intestinal microbiota in irritable bowel syndrome (IBS), we identified a bacterial phylotype with higher abundance in patients suffering from diarrhea than in healthy controls. In the present work, we have isolated in pure culture strain RT94, belonging to this phylotype, determined its whole genome sequence and performed an extensive genomic analysis and phenotypical testing. This revealed strain RT94 to be a strict anaerobe apparently belonging to a novel species with only 94% similarity in the 16S rRNA gene sequence to the closest relatives Ruminococcus torques and Ruminococcus lactaris. The G + C content of strain RT94 is 45.2 mol% and the major long-chain cellular fatty acids are C16:0, C18:0 and C14:0. The isolate is metabolically versatile but not a mucus or cellulose utilizer. It produces acetate, ethanol, succinate, lactate and formate, but very little butyrate, as end products of glucose metabolism. The mechanisms underlying the association of strain RT94 with diarrhea-type IBS are discussed.


Assuntos
Bactérias Anaeróbias/isolamento & purificação , Diarreia/diagnóstico , Genoma Bacteriano , Infecções por Bactérias Gram-Positivas/diagnóstico , Síndrome do Intestino Irritável/diagnóstico , RNA Ribossômico 16S/genética , Ruminococcus/isolamento & purificação , Ácido Acético/metabolismo , Bactérias Anaeróbias/classificação , Bactérias Anaeróbias/genética , Composição de Bases , Sequência de Bases , Diarreia/microbiologia , Etanol/metabolismo , Ácidos Graxos/metabolismo , Formiatos/metabolismo , Trato Gastrointestinal/microbiologia , Glucose/metabolismo , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Síndrome do Intestino Irritável/microbiologia , Ácido Láctico/metabolismo , Filogenia , Ruminococcus/classificação , Ruminococcus/genética , Análise de Sequência de DNA , Ácido Succínico/metabolismo
5.
Biochim Biophys Acta ; 1838(8): 2099-104, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24796504

RESUMO

The reassembly of the S-layer protein SlpA of Lactobacillus brevis ATCC 8287 on positively charged liposomes was studied by small angle X-ray scattering (SAXS) and zeta potential measurements. SlpA was reassembled on unilamellar liposomes consisting of 1-palmitoyl-2-oleyl-sn-glycero-3-phosphocholine and 1,2-dioleoyl-3-trimethylammonium-propane, prepared by extrusion through membranes with pore sizes of 50nm and 100nm. Similarly extruded samples without SlpA were used as a reference. The SlpA-containing samples showed clear diffraction peaks in their SAXS intensities. The lattice constants were calculated from the diffraction pattern and compared to those determined for SlpA on native cell wall fragments. Lattice constants for SlpA reassembled on liposomes (a=9.29nm, b=8.03nm, and γ=84.9°) showed a marked change in the lattice constants b and γ when compared to those determined for SlpA on native cell wall fragments (a=9.41nm, b=6.48nm, and γ=77.0°). The latter are in good agreement with values previously determined by electron microscopy. This indicates that the structure formed by SlpA is stable on the bacterial cell wall, but SlpA reassembles into a different structure on cationic liposomes. From the (10) reflection, the lower limit of crystallite size of SlpA on liposomes was determined to be 92nm, corresponding to approximately ten aligned lattice planes.


Assuntos
Proteínas de Bactérias/química , Parede Celular/metabolismo , Levilactobacillus brevis/metabolismo , Lipossomos , Espalhamento a Baixo Ângulo , Propriedades de Superfície , Difração de Raios X
6.
BMC Microbiol ; 14: 199, 2014 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-25070625

RESUMO

BACKGROUND: Adhesiveness to intestinal epithelium, beneficial immunomodulating effects and the production of pathogen-inhibitory compounds are generally considered as beneficial characteristics of probiotic organisms. We showed the potential health-promoting properties and the mechanisms of probiotic action of seven swine intestinal Lactobacillus amylovorus isolates plus the type strain (DSM 20531T) by investigating their adherence to porcine intestinal epithelial cells (IPEC-1) and mucus as well as the capacities of the strains to i) inhibit the adherence of Escherichia coli to IPEC-1 cells, ii) to produce soluble inhibitors against intestinal pathogens and iii) to induce immune signaling in dendritic cells (DCs). Moreover, the role of the L. amylovorus surface (S) -layers - symmetric, porous arrays of identical protein subunits present as the outermost layer of the cell envelope - in adherence to IPEC-1 cells was assessed using a novel approach which utilized purified cell wall fragments of the strains as carriers for the recombinantly produced S-layer proteins. RESULTS: Three of the L. amylovorus strains studied adhered to IPEC-1 cells, while four strains inhibited the adherence of E. coli, indicating additional mechanisms other than competition for binding sites being involved in the inhibition. None of the strains bound to porcine mucus. The culture supernatants of all of the strains exerted inhibitory effects on the growth of E. coli, Salmonella, Listeria and Yersinia, and a variable, strain-dependent induction was observed of both pro- and anti-inflammatory cytokines in human DCs. L. amylovorus DSM 16698 was shown to carry two S-layer-like proteins on its surface in addition to the major S-layer protein SlpA. In contrast to expectations, none of the major S-layer proteins of the IPEC-1 -adhering strains mediated bacterial adherence. CONCLUSIONS: We demonstrated adhesive and significant pathogen inhibitory efficacies among the swine intestinal L. amylovorus strains studied, pointing to their potential use as probiotic feed supplements, but no independent role could be demonstrated for the major S-layer proteins in adherence to epithelial cells. The results indicate that many intestinal bacteria may coexist with and confer benefits to the host by mechanisms not attributable to adhesion to epithelial cells or mucus.


Assuntos
Antibiose , Aderência Bacteriana , Lactobacillus acidophilus/química , Lactobacillus acidophilus/fisiologia , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/farmacologia , Probióticos , Animais , Células Cultivadas , Células Epiteliais/microbiologia , Escherichia coli/fisiologia , Intestinos/microbiologia , Lactobacillus acidophilus/isolamento & purificação , Listeria/crescimento & desenvolvimento , Muco/microbiologia , Salmonella/crescimento & desenvolvimento , Suínos , Yersinia/crescimento & desenvolvimento
7.
Appl Microbiol Biotechnol ; 97(12): 5225-43, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23677442

RESUMO

Bacterial surface (S) layers are the outermost proteinaceous cell envelope structures found on members of nearly all taxonomic groups of bacteria and Archaea. They are composed of numerous identical subunits forming a symmetric, porous, lattice-like layer that completely covers the cell surface. The subunits are held together and attached to cell wall carbohydrates by non-covalent interactions, and they spontaneously reassemble in vitro by an entropy-driven process. Due to the low amino acid sequence similarity among S-layer proteins in general, verification of the presence of an S-layer on the bacterial cell surface usually requires electron microscopy. In lactobacilli, S-layer proteins have been detected on many but not all species. Lactobacillus S-layer proteins differ from those of other bacteria in their smaller size and high predicted pI. The positive charge in Lactobacillus S-layer proteins is concentrated in the more conserved cell wall binding domain, which can be either N- or C-terminal depending on the species. The more variable domain is responsible for the self-assembly of the monomers to a periodic structure. The biological functions of Lactobacillus S-layer proteins are poorly understood, but in some species S-layer proteins mediate bacterial adherence to host cells or extracellular matrix proteins or have protective or enzymatic functions. Lactobacillus S-layer proteins show potential for use as antigen carriers in live oral vaccine design because of their adhesive and immunomodulatory properties and the general non-pathogenicity of the species.


Assuntos
Lactobacillus/metabolismo , Glicoproteínas de Membrana/metabolismo , Aderência Bacteriana , Biotecnologia/métodos , Técnicas de Visualização da Superfície Celular/métodos , Ponto Isoelétrico , Lactobacillus/genética , Glicoproteínas de Membrana/genética , Peso Molecular , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica
8.
Animals (Basel) ; 11(11)2021 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-34828014

RESUMO

Some Enterococcus faecalis and E. faecium strains are used as probiotics or feed additives. Adherence to the intestinal mucosa is considered a crucial step for intestinal bacteria to colonize and further interact with the host epithelium and the immune system. In dogs, there are no studies investigating the adhesion of E. faecalis and E. faecium to paraffin-embedded intestinal mucosa. Therefore, we aimed to investigate the adhesion of E. faecalis and E. faecium to the intestinal mucosa of six healthy beagles using bacteria derived from dogs and chickens. In addition, we aimed to validate a method to test the adhesion of Alexa Fluor-labeled bacteria to paraffin-embedded canine intestinal mucosa. The results of our study show that both canine- and chicken-derived E. faecalis strains adhered significantly better than E. faecium to the duodenal mucosa of healthy beagles (p = 0.002). In addition, canine E. faecalis and E. faecium adhered in higher numbers to canine duodenal mucosa, compared to chicken-derived strains of the same species (p = 0.015 for E. faecalis and p = 0.002 for E. faecium). The determination of the hydrophobicity of bacteria revealed that canine E. faecalis had the highest hydrophobicity level (36.6%), followed by chicken E. faecalis (20.4%), while canine E. faecium (5.7%) and chicken E. faecium (4.5%) had the lowest levels. Our results suggest that both the bacterial species and the host origin of the strain may influence mucosal adhesion.

9.
Appl Microbiol Biotechnol ; 87(2): 657-68, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20229202

RESUMO

Lactobacillus brevis ATCC 8287 possesses a surface (S)-layer protein SlpA, the gene of which is very efficiently expressed. To study the expression signals of the slpA gene, several different reporter plasmids, based on the low-copy-number vector pKTH2121 derived from pGK12, were constructed. In the reporter plasmids, only one of the two consecutive slpA promoters (P1, P2) was placed upstream of the Lactobacillus helveticus proline iminopeptidase (pepI) gene, and defined parts of the sequences upstream of the promoter were deleted. As indicated by reporter enzyme activities, both promoters were efficiently recognized at different growth stages in L. brevis. An upstream region important for the full activity of P1 was identified. The quantification of pepI-specific mRNA in L. brevis and SDS-PAGE indicated that slpA expression is not regulated at the post-transcriptional level and revealed no regulation of slpA promoters under the conditions tested. The high expression levels of both slpA and the reporter gene in L. brevis were found to remain at a high level after the addition of bile or pancreatin in the growth medium or after a change of the carbon source, which is advantageous for the potential use of SlpA as a carrier in live oral vaccines.


Assuntos
Proteínas de Bactérias/genética , Levilactobacillus brevis/genética , Regiões Promotoras Genéticas , Proteínas de Bactérias/metabolismo , Sequência de Bases , Regulação Bacteriana da Expressão Gênica , Levilactobacillus brevis/metabolismo , Dados de Sequência Molecular
10.
J Bacteriol ; 191(10): 3339-49, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19304849

RESUMO

Bacterial surface layer (S-layer) proteins are excellent candidates for in vivo and in vitro nanobiotechnological applications because of their ability to self-assemble into two-dimensional lattices that form the outermost layer of many Eubacteria and most Archaea species. Despite this potential, knowledge about their molecular architecture is limited. In this study, we investigated SlpA, the S-layer protein of the potentially probiotic bacterium Lactobacillus brevis ATCC 8287 by cysteine-scanning mutagenesis and chemical modification. We generated a series of 46 mutant proteins by replacing single amino acids with cysteine, which is not present in the wild-type protein. Most of the replaced amino acids were located in the self-assembly domain (residues 179 to 435) that likely faces the outer surface of the lattice. As revealed by electron microscopy, all the mutant proteins were able to form self-assembly products identical to that of the wild type, proving that this replacement does not dramatically alter the protein conformation. The surface accessibility of the sulfhydryl groups introduced was studied with two maleimide-containing marker molecules, TMM(PEG)(12) (molecular weight [MW], 2,360) and AlexaFluor488-maleimide (MW = 720), using both monomeric proteins in solution and proteins allowed to self-assemble on cell wall fragments. Using the acquired data and available domain information, we assigned the mutated residues into four groups according to their location in the protein monomer and lattice structure: outer surface of the lattice (9 residues), inner surface of the lattice (9), protein interior (12), and protein-protein interface/pore regions (16). This information is essential, e.g., in the development of therapeutic and other health-related applications of Lactobacillus S-layers.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Levilactobacillus brevis/química , Levilactobacillus brevis/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/ultraestrutura , Levilactobacillus brevis/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/ultraestrutura , Microscopia Eletrônica , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Solventes/química , Difração de Raios X
11.
BMC Microbiol ; 8: 165, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18828902

RESUMO

BACKGROUND: Lactobacillus brevis ATCC 8287 is covered by a regular surface (S-) layer consisting of a 435 amino acid protein SlpA. This protein is completely unrelated in sequence to the previously characterized S-layer proteins of Lactobacillus acidophilus group. RESULTS: In this work, the self-assembly and cell wall binding domains of SlpA were characterized. The C-terminal self-assembly domain encompassed residues 179-435 of mature SlpA, as demonstrated by the ability of N-terminally truncated recombinant SlpA to form a periodic structure indistinguishable from that formed by full length SlpA. Furthermore, a trypsin degradation analysis indicated the existence of a protease resistant C-terminal domain of 214 amino acids. By producing a set of C-terminally truncated recombinant SlpA (rSlpA) proteins the cell wall binding region was mapped to the N-terminal part of SlpA, where the first 145 amino acids of mature SlpA alone were sufficient for binding to isolated cell wall fragments of L. brevis ATCC 8287. The binding of full length rSlpA to the cell walls was not affected by the treatment of the walls with 5% trichloroacetic acid (TCA), indicating that cell wall structures other than teichoic acids are involved, a feature not shared by the Lactobacillus acidophilus group S-layer proteins characterized so far. Conserved carbohydrate binding motifs were identified in the positively charged N-terminal regions of six Lactobacillus brevis S-layer proteins. CONCLUSION: This study identifies SlpA as a two-domain protein in which the order of the functional domains is reversed compared to other characterized Lactobacillus S-layer proteins, and emphasizes the diversity of potential cell wall receptors despite similar carbohydrate binding sequence motifs in Lactobacillus S-layer proteins.


Assuntos
Proteínas de Bactérias/química , Parede Celular/metabolismo , Levilactobacillus brevis/genética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/ultraestrutura , Clonagem Molecular , DNA Bacteriano/genética , Escherichia coli/química , Escherichia coli/genética , Vetores Genéticos , Levilactobacillus brevis/química , Dados de Sequência Molecular , Mapeamento de Peptídeos , Plasmídeos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Análise de Sequência de Proteína
12.
Sci Rep ; 8(1): 13792, 2018 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-30206238

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

13.
Sci Rep ; 8(1): 10437, 2018 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-29993024

RESUMO

Recent research suggests that the microbial colonization of the mammalian intestine may begin before birth, but the observations are controversial due to challenges in the reliable sampling and analysis of low-abundance microbiota. We studied the perinatal microbiota of calves by sampling them immediately at birth and during the first postnatal week. The large size of the bovine newborns allows sampling directly from rectum using contamination-shielded swabs. Our 16S rDNA data, purged of potential contaminant sequences shared with negative controls, indicates the existence of a diverse low-abundance microbiota in the newborn rectal meconium and mucosa. The newborn rectal microbiota was composed of Firmicutes, Proteobacteria, Actinobacteria and Bacteroidetes. The microbial profile resembled dam oral rather than fecal or vaginal vestibular microbiota, but included typical intestinal taxa. During the first postnatal day, the rectum was invaded by Escherichia/Shigella and Clostridia, and the diversity collapsed. By 7 days, diversity was again increasing. In terms of relative abundance, Proteobacteria were replaced by Firmicutes, Bacteroidetes and Actinobacteria, including Faecalibacterium, Bacteroides, Lactobacillus, Butyricicoccus and Bifidobacterium. Our observations suggest that mammals are seeded before birth with a diverse microbiota, but the microbiota changes rapidly in the early postnatal life.


Assuntos
Microbioma Gastrointestinal , Animais , Animais Recém-Nascidos , Bactérias/isolamento & purificação , Bifidobacterium/isolamento & purificação , Biodiversidade , Bovinos , Escherichia/isolamento & purificação , Lactobacillus/isolamento & purificação , Proteobactérias/isolamento & purificação , RNA Ribossômico 16S/genética , Reto/microbiologia
14.
Int J Nanomedicine ; 13: 7939-7957, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30568441

RESUMO

INTRODUCTION: Antibiotic resistance is a growing concern in health care. Methicillin-resistant Staphylococcus aureus (MRSA), forming biofilms, is a common cause of resistant orthopedic implant infections. Gentamicin is a crucial antibiotic preventing orthopedic infections. Silica-gentamicin (SiO2-G) delivery systems have attracted significant interest in preventing the formation of biofilms. However, compelling scientific evidence addressing their efficacy against planktonic MRSA and MRSA biofilms is still lacking, and their safety has not extensively been studied. MATERIALS AND METHODS: In this work, we have investigated the effects of SiO2-G nanohybrids against planktonic MRSA as well as MRSA and Escherichia coli biofilms and then evaluated their toxicity in zebrafish embryos, which are an excellent model for assessing the toxicity of nanotherapeutics. RESULTS: SiO2-G nanohybrids inhibited the growth and killed planktonic MRSA at a minimum concentration of 500 µg/mL. SiO2-G nanohybrids entirely eradicated E. coli cells in biofilms at a minimum concentration of 250 µg/mL and utterly deformed their ultrastructure through the deterioration of bacterial shapes and wrinkling of their cell walls. Zebrafish embryos exposed to SiO2-G nanohybrids (500 and 1,000 µg/mL) showed a nonsignificant increase in mortality rates, 13.4±9.4 and 15%±7.1%, respectively, mainly detected 24 hours post fertilization (hpf). Frequencies of malformations were significantly different from the control group only 24 hpf at the higher exposure concentration. CONCLUSION: Collectively, this work provides the first comprehensive in vivo assessment of SiO2-G nanohybrids as a biocompatible drug delivery system and describes the efficacy of SiO2-G nanohybrids in combating planktonic MRSA cells and eradicating E. coli biofilms.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Farmacorresistência Bacteriana/efeitos dos fármacos , Gentamicinas/farmacologia , Nanopartículas/toxicidade , Dióxido de Silício/química , Testes de Toxicidade , Animais , Embrião não Mamífero/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Humanos , Larva/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Nanopartículas/química , Nanopartículas/ultraestrutura , Peixe-Zebra/embriologia
15.
PLoS One ; 13(8): e0202160, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30161141

RESUMO

The development of an early complex gut microbiota may play an important role in the protection against intestinal dysbiosis later in life. The significance of the developed microbiota for gut barrier functionality upon interaction with pathogenic or beneficial bacteria is largely unknown. The transcriptome of differently perfused jejunal loops of 12 caesarian-derived pigs, neonatally associated with microbiota of different complexity, was studied. Piglets received pasteurized sow colostrum at birth (d0), a starter microbiota (Lactobacillus amylovorus (LAM), Clostridium glycolicum, and Parabacteroides) on d1-d3, and a placebo inoculant (simple association, SA) or an inoculant consisting of sow's diluted feces (complex association, CA) on d3-d4. On d 26-37, jejunal loops were perfused for 8 h with either enterotoxigenic Escherichia coli F4 (ETEC), purified F4 fimbriae, LAM or saline control (CTRL). Gene expression of each intestinal loop was analyzed by Affymetrix Porcine Gene 1.1_ST array strips. Gene Set Enrichment Analysis was performed on expression values. Compared to CTRL, 184 and 74; 2 and 139; 2 and 48 gene sets, were up- and down-regulated by ETEC, F4 and LAM, respectively. ETEC up-regulated networks related to inflammatory and immune responses, RNA processing, and mitosis. There was a limited overlap in up-regulated gene sets between ETEC and F4 fimbriae. LAM down-regulated genes related to inflammatory and immune responses, as well as to cellular compound metabolism. In CA pigs, 57 gene sets were up-regulated by CA, while 73 were down-regulated compared to SA. CA up-regulated gene sets related to lymphocyte modulation and to cellular defense in all loop perfusions. In CA pigs, compared to SA pigs, genes for chemokine and cytokine activity and for response to external stimuli were down-regulated in ETEC-perfused loops and up-regulated in CTRL. The results highlight the importance of the nature of neonatal microbial colonization in the response to microbial stimuli later in life.


Assuntos
Escherichia coli Enterotoxigênica , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Microbioma Gastrointestinal , Jejuno/metabolismo , Jejuno/microbiologia , Lactobacillus acidophilus , Animais , Escherichia coli Enterotoxigênica/classificação , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Interações Hospedeiro-Patógeno/genética , Lactobacillus acidophilus/classificação , Lactobacillus acidophilus/genética , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/microbiologia
16.
Nanomaterials (Basel) ; 7(9)2017 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-28878170

RESUMO

Infected superficial wounds were traditionally controlled by topical antibiotics until the emergence of antibiotic-resistant bacteria. Silver (Ag) is a kernel for alternative antibacterial agents to fight this resistance quandary. The present study demonstrates a method for immobilizing small-sized (~5 nm) silver nanoparticles on silica matrix to form a nanosilver-silica (Ag-SiO₂) composite and shows the prolonged antibacterial effects of the composite in vitro. The composite exhibited a rapid initial Ag release after 24 h and a slower leaching after 48 and 72 h and was effective against both methicillin-resistant Staphylococcus aureus (MRSA) and Escherichia coli ( E . coli ). Ultraviolet (UV)-irradiation was superior to filter-sterilization in retaining the antibacterial effects of the composite, through the higher remaining Ag concentration. A gauze, impregnated with the Ag-SiO₂ composite, showed higher antibacterial effects against MRSA and E . coli than a commercial Ag-containing dressing, indicating a potential for the management and infection control of superficial wounds. Transmission and scanning transmission electron microscope analyses of the composite-treated MRSA revealed an interaction of the released silver ions with the bacterial cytoplasmic constituents, causing ultimately the loss of bacterial membranes. The present results indicate that the Ag-SiO₂ composite, with prolonged antibacterial effects, is a promising candidate for wound dressing applications.

17.
Front Microbiol ; 8: 657, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28450859

RESUMO

Lactobacillus ruminis, an autochthonous member of the gastrointestinal microbiota of humans and many animals, is a less characterized but interesting species for many reasons, including its intestinal prevalence and possible positive roles in host-microbe crosstalk. In this study, we isolated a novel L. ruminis strain (GRL 1172) from porcine feces and analyzed its functional characteristics and niche adaptation factors in parallel with those of three other L. ruminis strains (a human isolate, ATCC 25644, and two bovine isolates, ATCC 27780 and ATCC 27781). All the strains adhered to fibronectin, type I collagen, and human colorectal adenocarcinoma cells (HT-29), but poorly to type IV collagen, porcine intestinal epithelial cells (IPEC-1), and human colon adenocarcinoma cells (Caco-2). In competition assays, all the strains were able to inhibit the adhesion of Yersinia enterocolitica and enterotoxigenic Escherichia coli (ETEC, F4+) to fibronectin, type I; collagen, IPEC-1, and Caco-2 cells, and the inhibition rates tended to be higher than in exclusion assays. The culture supernatants of the tested strains inhibited the growth of six selected pathogens to varying extents. The inhibition was solely based on the low pH resulting from acid production during growth. All four L. ruminis strains supported the barrier function maintenance of Caco-2 cells, as shown by the modest increase in trans-epithelial electrical resistance and the prevention of dextran diffusion during co-incubation. However, the strains could not prevent the barrier damage caused by ETEC in the Caco-2 cell model. All the tested strains and their culture supernatants were able to provoke Toll-like receptor (TLR) 2-mediated NF-κB activation and IL-8 production in vitro to varying degrees. The induction of TLR5 signaling revealed that flagella were expressed by all the tested strains, but to different extents. Flagella and pili were observed by electron microscopy on the newly isolated strain GRL 1172.

18.
Genome Announc ; 4(2)2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27056237

RESUMO

The autochthonousLactobacillus brevisstrain D6, isolated from smoked fresh cheese, carries a 45-kDa S-layer protein. Strain D6 has shown adhesion to extracellular matrix proteins and to Caco-2 intestinal epithelial cells, as well as immunomodulatory potential and beneficial milk technological properties. Hence, it could be used as a potential probiotic starter culture for cheese production.

19.
PLoS One ; 11(4): e0153135, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27054581

RESUMO

Weaning triggers an adaptation of the gut function including luminal lactate generation by lactobacilli, depending on gastrointestinal site. We hypothesized that both lactobacilli and lactate influence porcine intestinal epithelial cells. In vivo experiments showed that concentration of lactate was significantly higher in gastric, duodenal and jejunal chyme of suckling piglets compared to their weaned counterparts. In an in vitro study we investigated the impact of physiological lactate concentration as derived from the in vivo study on the porcine intestinal epithelial cells IPEC-1 and IPEC-J2. We detected direct adherence of lactobacilli on the apical epithelial surface and a modulated F-actin structure. Application of lactobacilli culture supernatant alone or lactate (25 mM) at low pH (pH 4) changed the F-actin structure in a similar manner. Treatment of IPEC cultures with lactate at near neutral pH resulted in a significantly reduced superoxide-generation in Antimycin A-challenged cells. This protective effect was nearly completely reversed by inhibition of cellular lactate uptake via monocarboxylate transporter. Lactate treatment enhanced NADH autofluorescence ratio (Fcytosol/Fnucleus) in non-challenged cells, indicating an increased availability of reduced nucleotides, but did not change the overall ATP content of the cells. Lactobacilli-derived physiological lactate concentration in intestine is relevant for alleviation of redox stress in intestinal epithelial cells.


Assuntos
Antimicina A/farmacologia , Células Epiteliais/efeitos dos fármacos , Intestinos/citologia , Ácido Láctico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Actinas/química , Actinas/efeitos dos fármacos , Animais , Aderência Bacteriana , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Lactobacillus/fisiologia , Masculino , Suínos
20.
PLoS One ; 10(12): e0145718, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26709916

RESUMO

Sortase-dependent surface pili (or fimbriae) in Gram-positive bacteria are well documented as a key virulence factor for certain harmful opportunistic pathogens. However, it is only recently known that these multi-subunit protein appendages are also belonging to the "friendly" commensals and now, with this new perspective, they have come to be categorized as a niche-adaptation factor as well. In this regard, it was shown earlier that sortase-assembled piliation is a native fixture of two human intestinal commensalics (i.e., Lactobacillus rhamnosus and Bifidobacterium bifidum), and correspondingly where the pili involved have a significant role in cellular adhesion and immunomodulation processes. We now reveal that intestinal indigenous (or autochthonous) Lactobacillus ruminis is another surface-piliated commensal lactobacillar species. Heeding to in silico expectations, the predicted loci for the LrpCBA-called pili are organized tandemly in the L. ruminis genome as a canonical fimbrial operon, which then encodes for three pilin-proteins and a single C-type sortase enzyme. Through electron microscopic means, we showed that these pilus formations are a surface assemblage of tip, basal, and backbone pilin subunits (respectively named LrpC, LrpB, and LrpA) in L. ruminis, and also when expressed recombinantly in Lactococcus lactis. As well, by using the recombinant-piliated lactococci, we could define certain ecologically relevant phenotypic traits, such as the ability to adhere to extracellular matrix proteins and gut epithelial cells, but also to effectuate an induced dampening on Toll-like receptor 2 signaling and interleukin-8 responsiveness in immune-related cells. Within the context of the intestinal microcosm, by wielding such niche-advantageous cell-surface properties the LrpCBA pilus would undoubtedly have a requisite functional role in the colonization dynamics of L. ruminis indigeneity. Our study provides only the second description of a native-piliated Lactobacillus species, but at the same time also involves the structural and functional characterization of a third type of lactobacillar pilus.


Assuntos
Aminoaciltransferases/genética , Proteínas de Bactérias/genética , Cisteína Endopeptidases/genética , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/fisiologia , Lactobacillus/genética , Lactobacillus/fisiologia , Aderência Bacteriana , Sequência de Bases , Células CACO-2 , Simulação por Computador , DNA Bacteriano/genética , Fímbrias Bacterianas/ultraestrutura , Microbioma Gastrointestinal , Expressão Gênica , Genes Bacterianos , Células HEK293 , Humanos , Lactobacillus/ultraestrutura , Lactococcus lactis/genética , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Óperon , Fenótipo , Proteínas Recombinantes/genética , Homologia de Sequência do Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA