Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Proc Natl Acad Sci U S A ; 121(24): e2401686121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838019

ABSTRACT

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.


Subject(s)
Membrane Glycoproteins , Teichoic Acids , Teichoic Acids/metabolism , Teichoic Acids/chemistry , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/chemistry , Lactobacillus/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Models, Molecular , Lactobacillus acidophilus/metabolism , Lactobacillus acidophilus/genetics
2.
Appl Microbiol Biotechnol ; 105(3): 1123-1131, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33417041

ABSTRACT

Many studies have established the functional properties of Lacticaseibacillus rhamnosus GG, previously known as Lactobacillus rhamnosus GG, marketed worldwide as a probiotic. The extraordinary capacity of L. rhamnosus GG to bind to human mucus and influence the immune system especially stand out. Earlier, we have shown the key role of its SpaCBA sortase-dependent pili encoded by the spaCBA-srtC1 gene cluster herein. These heterotrimeric pili consist of a shaft pilin SpaA, a basal pilin SpaB, and tip pilin SpaC that contains a mucus-binding domain. Here, we set out to characterize a food-grade non-GMO mutant of L. rhamnosus GG, strain PA11, which secretes its pilins, rather than coupling them to the cell surface, due to a defect in the housekeeping sortase A. The sortase-negative strain PA11 was extensively characterized using functional genomics and biochemical approaches and found to secrete the SpaCBA pili into the supernatant. Given the functional importance and uniqueness of the mucus-binding pili of L. rhamnosus GG, strain PA11 offers novel opportunities towards the characterization and further therapeutic application of SpaCBA pili and their low-cost, large-scale production. KEY POINTS: •Creation of pilus-secreting mutant (PA11) of the key probiotic LGG. •Strain PA11 is defective in a functional housekeeping sortase SrtA. •Strain PA11 opens novel biotherapeutic application avenues. Graphical abstract.


Subject(s)
Lacticaseibacillus rhamnosus , Probiotics , Bacterial Proteins/genetics , Fimbriae Proteins , Fimbriae, Bacterial/genetics , Humans , Lacticaseibacillus rhamnosus/genetics , Mucus
3.
J Struct Biol ; 211(3): 107571, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32653644

ABSTRACT

Adhesion to cell surfaces is an essential and early prerequisite for successful host colonization by bacteria, and in most instances involves the specificities of various adhesins. Among bacterial Gram-positives, some genera and species mediate attachment to host cells by using long non-flagellar appendages called sortase-dependent pili. A case in point is the beneficial Lactobacillus rhamnosus GG gut-adapted strain that produces the so-called SpaCBA pilus, a structure noted for its promiscuous binding to intestinal mucus and collagen. Structurally, SpaCBA pili are heteropolymers of three different pilin-protein subunits, each with its own location and function in the pilus: backbone SpaA for length, basal SpaB for anchoring, and tip SpaC for adhesion. Previously, we solved the SpaA tertiary structure by X-ray crystallography and also reported on the crystallization of SpaB and SpaC. Here, we reveal the full-length high-resolution (1.9 Å) crystal structure of SpaC, a first for a sortase-dependent pilus-bearing commensal. The SpaC structure, unlike the representative four-domain architecture of other Gram-positive tip pilins, espouses an atypically longer five-domain arrangement that includes N-terminal 'binding' and C-terminal 'stalk' regions of two and three domains, respectively. With the prospect of establishing new mechanistic insights, we provide a structural basis for the multi-substrate binding nature of SpaC, as well as a structural model that reconciles its exclusive localization at the SpaCBA pilus tip.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Fimbriae, Bacterial/chemistry , Lacticaseibacillus rhamnosus/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Bacterial Proteins/genetics , Binding Sites , Collagen/metabolism , Crystallography, X-Ray , Fimbriae, Bacterial/metabolism , Lacticaseibacillus rhamnosus/cytology , Membrane Proteins/genetics , Microscopy, Atomic Force , Microscopy, Immunoelectron , Models, Molecular , Molecular Docking Simulation , Protein Domains
4.
Anaerobe ; 62: 102104, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31562947

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Metagenome , Metagenomics , Animals , Cluster Analysis , Databases, Genetic , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Mammals , Metagenomics/methods , Molecular Typing , Phylogeny , RNA, Ribosomal, 16S/genetics
5.
J Struct Biol ; 207(1): 74-84, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31026587

ABSTRACT

For some Gram-positive genera and species, the long-extended and adhesive sortase-dependent pilus plays an essential role during host colonization, biofilm formation, and immune modulation. Lactobacillus rhamnosus GG is a gut-adapted commensal strain that harbors the operonic genes for the SpaCBA and SpaFED pili, both being comprised of three different protein subunits termed the backbone, tip, and basal pilins. Crystal structures of the backbone pilins (SpaA and SpaD) have recently been solved, and here we describe the high-resolution (1.5 Å) structural determination of the SpaE basal pilin. SpaE consists of two immunoglobulin-like CnaB domains, with each displaying a spontaneously formed internal isopeptide bond, though apparently slow forming in the N-terminal domain. Remarkably, SpaE contains an atypically lengthy unstructured C-terminal tail, along with an YPKN pilin motif peptide, which is normally reserved for backbone subunits. Based on our analysis of the crystal structure data, we provide a molecular model for the basal positioning of the SpaE pilin within the SpaFED pilus.


Subject(s)
Bacterial Proteins/chemistry , Fimbriae, Bacterial/chemistry , Lacticaseibacillus rhamnosus/chemistry , Amino Acid Motifs , Crystallography, X-Ray , Fimbriae Proteins/chemistry , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Models, Molecular
6.
Appl Environ Microbiol ; 82(13): 3783-92, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27084020

ABSTRACT

UNLABELLED: Lactobacillus rhamnosus GG is a lactic acid bacterium widely marketed by the food industry. Its genomic analysis led to the identification of a gene cluster encoding mucus-binding SpaCBA pili, which is located in a genomic island enriched in insertion sequence (IS) elements. In the present study, we analyzed by genome-wide resequencing the genomic integrity of L. rhamnosus GG in four distinct evolutionary experiments conducted for approximately 1,000 generations under conditions of no stress or salt, bile, and repetitive-shearing stress. Under both stress-free and salt-induced stress conditions, the GG population (excluding the mutator lineage in the stress-free series [see below]) accumulated only a few single nucleotide polymorphisms (SNPs) and no frequent chromosomal rearrangements. In contrast, in the presence of bile salts or repetitive shearing stress, some IS elements were found to be activated, resulting in the deletion of large chromosomal segments that include the spaCBA-srtC1 pilus gene cluster. Remarkably, a high number of SNPs were found in three strains obtained after 900 generations of stress-free growth. Detailed analysis showed that these three strains derived from a founder mutant with an altered DNA polymerase subunit that resulted in a mutator phenotype. The present work confirms the stability of the pilus production phenotype in L. rhamnosus GG under stress-free conditions, highlights the possible evolutionary scenarios that may occur when this probiotic strain is extensively cultured, and identifies external factors that affect the chromosomal integrity of GG. The results provide mechanistic insights into the stability of GG in regard to its extensive use in probiotic and other functional food products. IMPORTANCE: Lactobacillus rhamnosus GG is a widely marketed probiotic strain that has been used in numerous clinical studies to assess its health-promoting properties. Hence, the stability of the probiotic functions of L. rhamnosus GG is of importance, and here we studied the impact of external stresses on the genomic integrity of L. rhamnosus GG. We studied three different stresses that are relevant for understanding its robustness and integrity under both ex vivo conditions, i.e., industrial manufacturing conditions, and in vivo conditions, i.e., intestinal tract-associated stress. Overall, our findings contribute to predicting the genomic stability of L. rhamnosus GG and its ecological performance.


Subject(s)
Gene Rearrangement , Genomic Instability , Lacticaseibacillus rhamnosus/growth & development , Lacticaseibacillus rhamnosus/genetics , Mutation , Polymorphism, Genetic , Probiotics , DNA Transposable Elements , Phenotype , Recombination, Genetic
7.
BMC Microbiol ; 16(1): 226, 2016 Sep 29.
Article in English | MEDLINE | ID: mdl-27688074

ABSTRACT

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.

8.
PLoS Genet ; 9(8): e1003683, 2013.
Article in English | MEDLINE | ID: mdl-23966868

ABSTRACT

Lactobacillus rhamnosus is a lactic acid bacterium that is found in a large variety of ecological habitats, including artisanal and industrial dairy products, the oral cavity, intestinal tract or vagina. To gain insights into the genetic complexity and ecological versatility of the species L. rhamnosus, we examined the genomes and phenotypes of 100 L. rhamnosus strains isolated from diverse sources. The genomes of 100 L. rhamnosus strains were mapped onto the L. rhamnosus GG reference genome. These strains were phenotypically characterized for a wide range of metabolic, antagonistic, signalling and functional properties. Phylogenomic analysis showed multiple groupings of the species that could partly be associated with their ecological niches. We identified 17 highly variable regions that encode functions related to lifestyle, i.e. carbohydrate transport and metabolism, production of mucus-binding pili, bile salt resistance, prophages and CRISPR adaptive immunity. Integration of the phenotypic and genomic data revealed that some L. rhamnosus strains possibly resided in multiple niches, illustrating the dynamics of bacterial habitats. The present study showed two distinctive geno-phenotypes in the L. rhamnosus species. The geno-phenotype A suggests an adaptation to stable nutrient-rich niches, i.e. milk-derivative products, reflected by the alteration or loss of biological functions associated with antimicrobial activity spectrum, stress resistance, adaptability and fitness to a distinctive range of habitats. In contrast, the geno-phenotype B displays adequate traits to a variable environment, such as the intestinal tract, in terms of nutrient resources, bacterial population density and host effects.


Subject(s)
Genome, Bacterial , Lacticaseibacillus rhamnosus/genetics , Phylogeny , Animals , Genetic Association Studies , Genomics , Lacticaseibacillus rhamnosus/classification , Milk/microbiology , Phenotype , Population Density
9.
Anaerobe ; 39: 60-7, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26946362

ABSTRACT

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.


Subject(s)
Bacteria, Anaerobic/isolation & purification , Diarrhea/diagnosis , Genome, Bacterial , Gram-Positive Bacterial Infections/diagnosis , Irritable Bowel Syndrome/diagnosis , RNA, Ribosomal, 16S/genetics , Ruminococcus/isolation & purification , Acetic Acid/metabolism , Bacteria, Anaerobic/classification , Bacteria, Anaerobic/genetics , Base Composition , Base Sequence , Diarrhea/microbiology , Ethanol/metabolism , Fatty Acids/metabolism , Formates/metabolism , Gastrointestinal Tract/microbiology , Glucose/metabolism , Gram-Positive Bacterial Infections/microbiology , Humans , Irritable Bowel Syndrome/microbiology , Lactic Acid/metabolism , Phylogeny , Ruminococcus/classification , Ruminococcus/genetics , Sequence Analysis, DNA , Succinic Acid/metabolism
10.
Gut ; 64(10): 1562-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25527456

ABSTRACT

OBJECTIVE: An adequate bowel cleansing is essential for a successful colonoscopy. Although purgative consumption is safe for the patient, there is little consensus on how the intestinal microbiota is affected by the procedure, especially regarding the potential long-term consequences. DESIGN: 23 healthy subjects were randomised into two study groups consuming a bowel preparation (Moviprep), either in two separate doses of 1 L or as a single 2-L dose. Participants donated faecal samples at the baseline, after bowel cleansing, 14 and 28 days after the treatment. The intestinal microbiota composition was determined with phylogenetic microarray as well as quantitative PCR analysis and correlated with the previously quantified faecal serine proteases. RESULTS: The lavage introduced an instant and substantial change to the intestinal microbiota. The total microbial load was decreased by 31-fold and 22% of the participants lost the subject-specificity of their microbiota. While the bacterial levels and community composition were essentially restored within 14 days, the rate of recovery was dose dependent: consumption of the purgative in a single dose had a more severe effect on the microbiota composition than that of a double dose, and notably increased the levels of Proteobacteria, Fusobacteria and bacteria related to Dorea formicigenerans. The abundance of the latter also correlated with the amount of faecal serine proteases that were increased after purging. CONCLUSIONS: Our results suggest that the bowel cleansing using two separate dosages introduces fewer alterations to the intestinal microbiota than a single dose and hence may be preferred in clinical practice.


Subject(s)
Bacteria/drug effects , Colon/microbiology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/microbiology , Polyethylene Glycols/administration & dosage , Bacteria/genetics , Bacteria/isolation & purification , Colonoscopy/methods , Dose-Response Relationship, Drug , Feces/microbiology , Follow-Up Studies , Healthy Volunteers , Humans , RNA, Bacterial/analysis , Therapeutic Irrigation
11.
Proteomics ; 15(20): 3544-52, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26255997

ABSTRACT

Obesity is associated with the intestinal microbiota in humans but the underlying mechanisms are yet to be fully understood. Our previous phylogenetic study showed that the faecal microbiota profiles of nonobese versus obese and morbidly obese individuals differed. Here, we have extended this analysis with a characterization of the faecal metaproteome, in order to detect differences at a functional level. Proteins were extracted from crude faecal samples of 29 subjects, separated by 1D gel electrophoresis and characterized using RP LC-MS/MS. The peptide data were analyzed in database searches with two complementary algorithms, OMSSA and X!Tandem, to increase the number of identifications. Evolutionary genealogy of genes: nonsupervised orthologous groups (EggNOG) database searches resulted in the functional annotation of over 90% of the identified microbial and human proteins. Based on both bacterial and human proteins, a clear clustering of obese and nonobese samples was obtained that exceeded the phylogenetic separation in dimension. Moreover, integration of the metaproteomics and phylogenetic datasets revealed notably that the phylum Bacteroidetes was metabolically more active in the obese than nonobese subjects. Finally, significant correlations between clinical measurements and bacterial gene functions were identified. This study emphasizes the importance of integrating data of the host and microbiota to understand their interactions.


Subject(s)
Gastrointestinal Tract/microbiology , Microbiota/genetics , Obesity, Morbid/genetics , Proteome/genetics , Adult , Bacteroides/genetics , Bacteroides/isolation & purification , Feces/microbiology , Female , Gastrointestinal Tract/pathology , Humans , Male , Obesity, Morbid/microbiology , Obesity, Morbid/pathology , Phylogeny , Prevotella/genetics , Prevotella/isolation & purification , Tandem Mass Spectrometry
12.
J Biol Chem ; 289(22): 15764-75, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24753244

ABSTRACT

In Gram-positive bacteria, sortase-dependent pili mediate the adhesion of bacteria to host epithelial cells and play a pivotal role in colonization, host signaling, and biofilm formation. Lactobacillus rhamnosus strain GG, a well known probiotic bacterium, also displays on its cell surface mucus-binding pilus structures, along with other LPXTG surface proteins, which are processed by sortases upon specific recognition of a highly conserved LPXTG motif. Bioinformatic analysis of all predicted LPXTG proteins encoded by the L. rhamnosus GG genome revealed a remarkable conservation of glycine residues juxtaposed to the canonical LPXTG motif. Here, we investigated and defined the role of this so-called triple glycine (TG) motif in determining sortase specificity during the pilus assembly and anchoring. Mutagenesis of the TG motif resulted in a lack or an alteration of the L. rhamnosus GG pilus structures, indicating that the TG motif is critical in pilus assembly and that they govern the pilin-specific and housekeeping sortase specificity. This allowed us to propose a regulatory model of the L. rhamnosus GG pilus biogenesis. Remarkably, the TG motif was identified in multiple pilus gene clusters of other Gram-positive bacteria, suggesting that similar signaling mechanisms occur in other, mainly pathogenic, species.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/enzymology , Lacticaseibacillus rhamnosus/enzymology , Aminoacyltransferases/genetics , Bacterial Adhesion/physiology , Bacterial Proteins/genetics , Cysteine Endopeptidases/genetics , Enzyme Activation/physiology , Fimbriae Proteins/genetics , Fimbriae, Bacterial/ultrastructure , Glycine/genetics , Lacticaseibacillus rhamnosus/genetics , Lacticaseibacillus rhamnosus/ultrastructure , Microscopy, Electron, Transmission , Mutagenesis, Site-Directed , Probiotics , Signal Transduction/physiology , Substrate Specificity
13.
Biochim Biophys Acta ; 1838(8): 2099-104, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24796504

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Cell Wall/metabolism , Levilactobacillus brevis/metabolism , Liposomes , Scattering, Small Angle , Surface Properties , X-Ray Diffraction
14.
Appl Environ Microbiol ; 81(6): 2050-62, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25576613

ABSTRACT

Recently, spaCBA-encoded pili on the cell surface of Lactobacillus rhamnosus GG were identified to be key molecules for binding to human intestinal mucus and Caco-2 intestinal epithelial cells. Here, we investigated the role of the SpaCBA pilus of L. rhamnosus GG in the interaction with macrophages in vitro by comparing the wild type with surface mutants. Our results show that SpaCBA pili play a significant role in the capacity for adhesion to macrophages and also promote bacterial uptake by these phagocytic cells. Interestingly, our data suggest that SpaCBA pili also mediate anti-inflammatory effects by induction of interleukin-10 (IL-10) mRNA and reduction of interleukin-6 (IL-6) mRNA in a murine RAW 264.7 macrophage cell line. These pili appear to mediate these effects indirectly by promoting close contact with the macrophages, facilitating the exertion of anti-inflammatory effects by other surface molecules via yet unknown mechanisms. Blockage of complement receptor 3 (CR3), previously identified to be a receptor for streptococcal pili, significantly decreased the uptake of pilus-expressing strains in RAW 264.7 cells, while the expression of IL-10 and IL-6 mRNA by these macrophages was not affected by this blocking. On the other hand, blockage of Toll-like receptor 2 (TLR2) significantly reduced the expression of IL-6 mRNA irrespective of the presence of pili.


Subject(s)
Bacterial Adhesion , Cytokines/metabolism , Fimbriae, Bacterial/immunology , Lacticaseibacillus rhamnosus/immunology , Macrophages/immunology , Macrophages/microbiology , Phagocytosis , Animals , Cell Line , Immune Tolerance , Lacticaseibacillus rhamnosus/physiology , Mice
15.
Pediatr Res ; 77(4): 528-35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25580735

ABSTRACT

BACKGROUND: Bacterial contact in utero modulates fetal and neonatal immune responses. Maternal probiotic supplementation reduces the risk of immune-mediated disease in the infant. We investigated the immunomodulatory properties of live Lactobacillus rhamnosus GG and its SpaC pilus adhesin in human fetal intestinal models. METHODS: Tumor necrosis factor (TNF)-α mRNA expression was measured by qPCR in a human fetal intestinal organ culture model exposed to live L. rhamnosus GG and proinflammatory stimuli. Binding of recombinant SpaC pilus protein to intestinal epithelial cells (IECs) was assessed in human fetal intestinal organ culture and the human fetal intestinal epithelial cell line H4 by immunohistochemistry and immunofluorescence, respectively. TLR-related gene expression in fetal ileal organ culture after exposure to recombinant SpaC was assessed by qPCR. RESULTS: Live L. rhamnosus GG significantly attenuates pathogen-induced TNF-α mRNA expression in the human fetal gut. Recombinant SpaC protein was found to adhere to the fetal gut and to modulate varying levels of TLR-related gene expression. CONCLUSION: The human fetal gut is responsive to luminal microbes. L. rhamnosus GG significantly attenuates fetal intestinal inflammatory responses to pathogenic bacteria. The L. rhamnosus GG pilus adhesin SpaC binds to immature human IECs and directly modulates IEC innate immune gene expression.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Developmental , Ileum/embryology , Ileum/microbiology , Lacticaseibacillus rhamnosus/metabolism , Membrane Proteins/metabolism , Toll-Like Receptors/metabolism , Adhesins, Bacterial/metabolism , Bacterial Adhesion , Cytokines/metabolism , Epithelial Cells/cytology , Fimbriae, Bacterial , Humans , Immunohistochemistry , Inflammation , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-10/metabolism , Microscopy, Fluorescence , Probiotics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-1 Type I/metabolism , Recombinant Proteins/metabolism , Salmonella typhimurium , Tumor Necrosis Factor-alpha/metabolism
16.
Gut ; 63(11): 1737-45, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24310267

ABSTRACT

BACKGROUND: About 10% of patients with IBS report the start of the syndrome after infectious enteritis. The clinical features of postinfectious IBS (PI-IBS) resemble those of diarrhoea-predominant IBS (IBS-D). While altered faecal microbiota has been identified in other IBS subtypes, composition of the microbiota in patients with PI-IBS remains uncharacterised. OBJECTIVE: To characterise the microbial composition of patients with PI-IBS, and to examine the associations between the faecal microbiota and a patient's clinical features. DESIGN: Using a phylogenetic microarray and selected qPCR assays, we analysed differences in the faecal microbiota of 57 subjects from five study groups: patients with diagnosed PI-IBS, patients who 6 months after gastroenteritis had either persisting bowel dysfunction or no IBS symptoms, benchmarked against patients with IBS-D and healthy controls. In addition, the associations between the faecal microbiota and health were investigated by correlating the microbial profiles to immunological markers, quality of life indicators and host gene expression in rectal biopsies. RESULTS: Microbiota analysis revealed a bacterial profile of 27 genus-like groups, providing an Index of Microbial Dysbiosis (IMD), which significantly separated patient groups and controls. Within this profile, several members of Bacteroidetes phylum were increased 12-fold in patients, while healthy controls had 35-fold more uncultured Clostridia. We showed correlations between the IMD and expression of several host gene pathways, including amino acid synthesis, cell junction integrity and inflammatory response, suggesting an impaired epithelial barrier function in IBS. CONCLUSIONS: The faecal microbiota of patients with PI-IBS differs from that of healthy controls and resembles that of patients with IBS-D, suggesting a common pathophysiology. Moreover, our analysis suggests a variety of host-microbe associations that may underlie intestinal symptoms, initiated by gastroenteritis.


Subject(s)
Feces/microbiology , Gastroenteritis/physiopathology , Irritable Bowel Syndrome/physiopathology , Microbiota , Receptor Cross-Talk/physiology , Adult , Female , Gene Expression Profiling , Humans , Irritable Bowel Syndrome/diagnosis , Male , Middle Aged
17.
Appl Environ Microbiol ; 80(22): 7001-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25192985

ABSTRACT

Lactobacillus rhamnosus GG is one of the best-characterized lactic acid bacteria and can be considered a probiotic paradigm. Comparative and functional genome analysis showed that L. rhamnosus GG harbors a genomic island including the spaCBA-srtC1 gene cluster, encoding the cell surface-decorating host-interacting pili. Here, induced mutagenesis was used to study pilus biogenesis in L. rhamnosus GG. A combination of two powerful approaches, mutation selection and next-generation sequencing, was applied to L. rhamnosus GG for the selection of pilus-deficient mutants from an enriched population. The isolated mutants were first screened by immuno-dot blot analysis using antiserum against pilin proteins. Relevant mutants were selected, and the lack of pili was confirmed by immunoelectron microscopy. The pilosotype of 10 mutant strains was further characterized by analyzing pilin expression using Western blot, dot blot, and immunofluorescence methods. A mucus binding assay showed that the mutants did not adhere to porcine intestinal mucus. Comparative genome sequence analysis using the Illumina MiSeq platform allowed us to determine the nature of the mutations in the obtained pilus-deficient derivatives. Three major classes of mutants with unique genotypes were observed: class I, with mutations in the srtC1 gene; class II, with a deletion containing the spaCBA-srtC1 gene cluster; and class III, with mutations in the spaA gene. Only a limited number of collateral mutations were observed, and one of the pilus-deficient derivatives with a deficient srtC1 gene contained 24 other mutations. This strain, PB12, can be considered a candidate for human trials addressing the impact of the absence of pili.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/genetics , Fimbriae, Bacterial/metabolism , Intestinal Mucosa/microbiology , Lacticaseibacillus rhamnosus/physiology , Animals , Bacterial Proteins/metabolism , Fimbriae, Bacterial/genetics , Genomics , Genotype , Humans , Lacticaseibacillus rhamnosus/genetics , Swine
18.
BMC Microbiol ; 14: 199, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25070625

ABSTRACT

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.


Subject(s)
Antibiosis , Bacterial Adhesion , Lactobacillus acidophilus/chemistry , Lactobacillus acidophilus/physiology , Membrane Glycoproteins/analysis , Membrane Glycoproteins/pharmacology , Probiotics , Animals , Cells, Cultured , Epithelial Cells/microbiology , Escherichia coli/physiology , Intestines/microbiology , Lactobacillus acidophilus/isolation & purification , Listeria/growth & development , Mucus/microbiology , Salmonella/growth & development , Swine , Yersinia/growth & development
19.
Curr Top Microbiol Immunol ; 358: 323-46, 2013.
Article in English | MEDLINE | ID: mdl-22094893

ABSTRACT

Our intestinal tract is colonized since birth by multiple microbial species that show a characteristic succession in time. Notably the establishment of the microbiota in early life is important as it appears to impact later health. While apparently stable in healthy adults, the intestinal microbiota is changing significantly during aging. After 100 years of symbiosis marked changes have been observed that may relate to an increased level of intestinal inflammation. There is considerable interest in the microbiota in health and disease as it may provide functional biomarkers, the possibility to differentiate subjects, and avenues for interventions. This chapter reviews the present state of the art on the research to investigate the contribution of the intestinal microbiota to human health. Specific attention will be given to the healthy microbiota and aberrations due to disturbances such as celiac disease, irritable bowel syndrome, inflammatory bowel disease, obesity and diabetes, and non-alcoholic fatty liver disease.


Subject(s)
Bacteria/metabolism , Disease/history , Health/history , Intestines/microbiology , Metagenome , History, 20th Century , History, 21st Century , Humans , Intestinal Mucosa/metabolism
20.
Proc Natl Acad Sci U S A ; 108(27): 11217-22, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21690406

ABSTRACT

Development of the human gut microbiota commences at birth, with bifidobacteria being among the first colonizers of the sterile newborn gastrointestinal tract. To date, the genetic basis of Bifidobacterium colonization and persistence remains poorly understood. Transcriptome analysis of the Bifidobacterium breve UCC2003 2.42-Mb genome in a murine colonization model revealed differential expression of a type IVb tight adherence (Tad) pilus-encoding gene cluster designated "tad(2003)." Mutational analysis demonstrated that the tad(2003) gene cluster is essential for efficient in vivo murine gut colonization, and immunogold transmission electron microscopy confirmed the presence of Tad pili at the poles of B. breve UCC2003 cells. Conservation of the Tad pilus-encoding locus among other B. breve strains and among sequenced Bifidobacterium genomes supports the notion of a ubiquitous pili-mediated host colonization and persistence mechanism for bifidobacteria.


Subject(s)
Bifidobacterium/genetics , Bifidobacterium/physiology , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/physiology , Genome, Bacterial , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Base Sequence , Bifidobacterium/growth & development , Bifidobacterium/ultrastructure , Comparative Genomic Hybridization , DNA, Bacterial/genetics , Female , Fimbriae, Bacterial/ultrastructure , Gastrointestinal Tract/microbiology , Gene Expression Regulation, Bacterial , Germ-Free Life , Humans , Male , Metagenome , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Molecular Sequence Data , Multigene Family , Mutation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL