Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Appl Microbiol Biotechnol ; 102(24): 10645-10663, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30306201

RESUMO

Members of the Bifidobacterium genus are widely used as probiotics in fermented milk products. Bifidobacterium animalis subsp. animalis CNCM I-4602 grows and survives poorly in reconstituted skimmed milk (RSM). Availing of genome and transcriptome information, this poor growth and survival phenotype in milk was substantially improved by the addition of certain compounds, such as yeast extract, uric acid, glutathione, cysteine, ferrous sulfate, and a combination of magnesium sulfate and manganese sulfate. Carbohydrate utilization of CNCM I-4602 was also investigated, allowing the identification of several carbohydrate utilization gene clusters, and highlighting this strain's inability to utilize lactose, unlike the type strain of this subspecies, B. animalis subsp. animalis ATCC25527 and the B. animalis subsp. lactis subspecies. In addition, the ability of B. animalis subsp. animalis CNCM I-4602 to colonize a murine model was investigated, which showed that this strain persists in the murine gut for a period of at least 4 weeks. Associated in vivo transcriptome analysis revealed that, among other genes, a gene cluster encoding a predicted type IVb tight adherence (Tad) pilus was upregulated, indicating that this extracellular structure plays a role in the colonization/adaptation of the murine gastrointestinal tract by this strain.


Assuntos
Bifidobacterium animalis/crescimento & desenvolvimento , Bifidobacterium animalis/genética , Microbiologia de Alimentos/métodos , Leite/microbiologia , Animais , Bifidobacterium animalis/efeitos dos fármacos , Metabolismo dos Carboidratos , Resistência Microbiana a Medicamentos , Feminino , Microbioma Gastrointestinal , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Camundongos Endogâmicos BALB C , Probióticos
2.
Nature ; 473(7346): 174-80, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21508958

RESUMO

Our knowledge of species and functional composition of the human gut microbiome is rapidly increasing, but it is still based on very few cohorts and little is known about variation across the world. By combining 22 newly sequenced faecal metagenomes of individuals from four countries with previously published data sets, here we identify three robust clusters (referred to as enterotypes hereafter) that are not nation or continent specific. We also confirmed the enterotypes in two published, larger cohorts, indicating that intestinal microbiota variation is generally stratified, not continuous. This indicates further the existence of a limited number of well-balanced host-microbial symbiotic states that might respond differently to diet and drug intake. The enterotypes are mostly driven by species composition, but abundant molecular functions are not necessarily provided by abundant species, highlighting the importance of a functional analysis to understand microbial communities. Although individual host properties such as body mass index, age, or gender cannot explain the observed enterotypes, data-driven marker genes or functional modules can be identified for each of these host properties. For example, twelve genes significantly correlate with age and three functional modules with the body mass index, hinting at a diagnostic potential of microbial markers.


Assuntos
Bactérias/classificação , Intestinos/microbiologia , Metagenoma , Bactérias/genética , Técnicas de Tipagem Bacteriana , Biodiversidade , Biomarcadores/análise , Europa (Continente) , Fezes/microbiologia , Feminino , Humanos , Masculino , Metagenômica , Filogenia
3.
J Infect Dis ; 210(7): 1029-41, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24706936

RESUMO

We evaluated the protective effects of fermented dairy products (FDPs) in an infection model, using the mouse pathogen Citrobacter rodentium (CR). Treatment of mice with FDP formulas A, B, and C or a control product did not affect CR colonization, organ specificity, or attaching and effacing lesion formation. Fermented dairy product A (FDP-A), but neither the supernatant from FDP-A nor ß-irradiated (IR) FDP-A, caused a significant reduction in colonic crypt hyperplasia and CR-associated pathology. Profiling the gut microbiota revealed that IR-FDP-A promoted higher levels of phylotypes belonging to Alcaligenaceae and a decrease in Lachnospiraceae (Ruminococcus) during CR infection. Conversely, FDP-A prevented a decrease in Ruminococcus and increased Turicibacteraceae (Turicibacter). Importantly, loss of Ruminococcus and Turicibacter has been associated with susceptibility to dextran sodium sulfate-induced colitis. Our results demonstrate that viable bacteria in FDP-A reduced CR-induced colonic crypt hyperplasia and prevented the loss of key bacterial genera that may contribute to disease pathology.


Assuntos
Biota , Citrobacter rodentium/crescimento & desenvolvimento , Colo/patologia , Laticínios , Dieta/métodos , Infecções por Enterobacteriaceae/prevenção & controle , Hiperplasia/prevenção & controle , Animais , Colo/microbiologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Feminino , Hiperplasia/patologia , Camundongos Endogâmicos C57BL
4.
FEMS Microbiol Rev ; 47(2)2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36725208

RESUMO

Molecular technologies, including high-throughput sequencing, have expanded our perception of the microbial world. Unprecedented insights into the composition and function of microbial communities have generated large interest, with numerous landmark studies published in recent years relating the important roles of microbiomes and the environment-especially diet and nutrition-in human, animal, and global health. As such, food microbiomes represent an important cross-over between the environment and host. This is especially true of fermented food microbiomes, which actively introduce microbial metabolites and, to a lesser extent, live microbes into the human gut. Here, we discuss the history of fermented foods, and examine how molecular approaches have advanced research of these fermented foods over the past decade. We highlight how various molecular approaches have helped us to understand the ways in which microbes shape the qualities of these products, and we summarize the impacts of consuming fermented foods on the gut. Finally, we explore how advances in bioinformatics could be leveraged to enhance our understanding of fermented foods. This review highlights how integrated molecular approaches are changing our understanding of the microbial communities associated with food fermentation, the creation of unique food products, and their influences on the human microbiome and health.


Assuntos
Alimentos Fermentados , Microbiota , Animais , Humanos , Dieta , Fermentação , Sequenciamento de Nucleotídeos em Larga Escala
5.
Gut Microbes ; 15(1): 2244720, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37589280

RESUMO

Fermented foods and beverages are a significant source of dietary bacteria that enter the gastrointestinal (GI) tract. However, little is known about how these microbes survive and adapt to the small intestinal environment. Colony-forming units (CFU) enumeration and viability qPCR of Lacticaseibacillus rhamnosus CNCM I-3690 in the ileal effluent of 10 ileostomy subjects during 12-h post consumption of a dairy product fermented with this strain demonstrated the high level of survival of this strain during human small intestine passage. Metatranscriptome analyses revealed the in situ transcriptome of L. rhamnosus in the small intestine, which was contrasted with transcriptome data obtained from in vitro cultivation. These comparative analyses revealed substantial metabolic adaptations of L. rhamnosus during small intestine transit, including adjustments of carbohydrate metabolism, surface-protein expression, and translation machinery. The prominent presence of L. rhamnosus in the effluent samples did not elicit an appreciable effect on the composition of the endogenous small intestine microbiome, but significantly altered the ecosystem's overall activity profile, particularly of pathways associated with carbohydrate metabolism. Strikingly, two of the previously recognized gut-brain metabolic modules expressed in situ by L. rhamnosus (inositol degradation and glutamate synthesis II) are among the most dominantly enriched activities in the ecosystem's activity profile. This study establishes the survival capacity of L. rhamnosus in the human small intestine and highlights its functional adjustment in situ, which we postulate to play a role in the probiotic effects associated with this strain.


Assuntos
Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Microbiota , Probióticos , Humanos , Íleo
6.
J Bacteriol ; 193(19): 5560-1, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21914878

RESUMO

Bifidobacterium animalis subsp. lactis CNCM I-2494 is part of a commercialized fermented dairy product with documented health benefits revealed by multiple randomized placebo-controlled clinical trials. Here we report the complete genome sequence of this strain, which has a circular genome of 1,943,113 bp with 1,660 open reading frames and 4 ribosomal operons.


Assuntos
Bifidobacterium/genética , Genoma Bacteriano/genética , Probióticos , Dados de Sequência Molecular
7.
Microbiology (Reading) ; 156(Pt 8): 2495-2505, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20430809

RESUMO

Escherichia coli haemolysin A (HlyA), an RTX toxin, is secreted probably as an unfolded intermediate, by the type I (ABC transporter-dependent) pathway, utilizing a C-terminal secretion signal. However, the mechanism of translocation and post-translocation folding is not understood. We identified a mutation (hlyA99) at the extreme C terminus, which is dominant in competition experiments, blocking secretion of the wild-type toxin co-expressed in the same cell. This suggests that unlike recessive mutations which affect recognition of the translocation machinery, the hlyA99 mutation interferes with some later step in secretion. Indeed, the mutation reduced haemolytic activity of the toxin and the activity of beta-lactamase when the latter was fused to a C-terminal 23 kDa fragment of HlyA carrying the hlyA99 mutation. A second mutant (hlyAdel6), lacking the six C-terminal residues of HlyA, also showed reduced haemolytic activity and neither mutant protein regained normal haemolytic activity in in vitro unfolding/refolding experiments. Tryptophan fluorescence spectroscopy indicated differences in structure between the secreted forms of wild-type HlyA and the HlyA Del6 mutant. These results suggested that the mutations affected the correct folding of both HlyA and the beta-lactamase fusion. Thus, we propose a dual function for the HlyA C terminus involving an important role in post-translocation folding as well as targeting HlyA for secretion.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Proteínas Hemolisinas/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas Hemolisinas/genética , Hemólise , Dados de Sequência Molecular , Mutação , Dobramento de Proteína , beta-Lactamases/metabolismo
8.
Sci Rep ; 9(1): 5398, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30931953

RESUMO

The gut barrier plays an important role in human health. When barrier function is impaired, altered permeability and barrier dysfunction can occur, leading to inflammatory bowel diseases, irritable bowel syndrome or obesity. Several bacteria, including pathogens and commensals, have been found to directly or indirectly modulate intestinal barrier function. The use of probiotic strains could be an important landmark in the management of gut dysfunction with a clear impact on the general population. Previously, we found that Lactobacillus rhamnosus CNCM I-3690 can protect intestinal barrier functions in mice inflammation model. Here, we investigated its mechanism of action. Our results show that CNCM I-3690 can (i) physically maintain modulated goblet cells and the mucus layer and (ii) counteract changes in local and systemic lymphocytes. Furthermore, mice colonic transcriptome analysis revealed that CNCM I-3690 enhances the expression of genes related to healthy gut permeability: motility and absorption, cell proliferation; and protective functions by inhibiting endogenous proteases. Finally, SpaFED pili are clearly important effectors since an L. rhamnosus ΔspaF mutant failed to provide the same benefits as the wild type strain. Taken together, our data suggest that CNCM I-3690 restores impaired intestinal barrier functions via anti-inflammatory and cytoprotective responses.


Assuntos
Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Lacticaseibacillus rhamnosus/fisiologia , Muco/metabolismo , Probióticos/farmacologia , Animais , Células CACO-2 , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Citoproteção/efeitos dos fármacos , Dinitrofluorbenzeno/análogos & derivados , Perfilação da Expressão Gênica/métodos , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/metabolismo , Células HEK293 , Células HT29 , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Intestinos/microbiologia , Intestinos/fisiologia , Lacticaseibacillus rhamnosus/genética , Camundongos , Mutação , Permeabilidade/efeitos dos fármacos , Substâncias Protetoras/farmacologia
9.
Immun Inflamm Dis ; 4(3): 289-99, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27621812

RESUMO

INTRODUCTION: The microbiota controls a variety of biological functions, including immunity, and alterations of the microbiota in early life are associated with a higher risk of developing allergies later in life. Several probiotic bacteria, and particularly lactic acid bacteria, were described to reduce both the induction of allergic responses and allergic manifestations. Although specific probiotic strains were used in these studies, their protective effects on allergic responses also might be common for all lactobacilli. METHODS: To determine whether allergic effector cells inhibition is a common feature of lactobacilli or whether it varies among lactobacilli strains, we compared the ability of 40 strains of the same Lactobacillus paracasei species to inhibit IgE-dependent mouse mast cell and human basophil activation. RESULTS: We uncovered a marked heterogeneity in the inhibitory properties of the 40 Lactobacillus strains tested. These segregated into three to four clusters depending on the intensity of inhibition. Some strains inhibited both mouse mast cell and human basophil activation, others strains inhibited only one cell type and another group induced no inhibition of activation for either cell type. CONCLUSIONS: Individual Lactobacillus strains of the same species differentially inhibit IgE-dependent activation of mouse mast cells and human basophils, two cell types that are critical in the onset of allergic manifestations. Although we failed to identify specific bacterial genes associated with inhibition by gene-trait matching analysis, our findings demonstrate the complexity of the interactions between the microbiota and the host. These results suggest that some L. paracasei strains might be more beneficial in allergies than others strains and provide the bases for a rational screening of lactic acid bacteria strains as next-generation probiotics in the field of allergy.

10.
PLoS One ; 9(4): e94875, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24743599

RESUMO

Bifidobacterium animalis subsp. lactis CNCM I-2494 is a component of a commercialized fermented dairy product for which beneficial effects on health has been studied by clinical and preclinical trials. To date little is known about the molecular mechanisms that could explain the beneficial effects that bifidobacteria impart to the host. Restriction-modification (R-M) systems have been identified as key obstacles in the genetic accessibility of bifidobacteria, and circumventing these is a prerequisite to attaining a fundamental understanding of bifidobacterial attributes, including the genes that are responsible for health-promoting properties of this clinically and industrially important group of bacteria. The complete genome sequence of B. animalis subsp. lactis CNCM I-2494 is predicted to harbour the genetic determinants for two type II R-M systems, designated BanLI and BanLII. In order to investigate the functionality and specificity of these two putative R-M systems in B. animalis subsp. lactis CNCM I-2494, we employed PacBio SMRT sequencing with associated methylome analysis. In addition, the contribution of the identified R-M systems to the genetic accessibility of this strain was assessed.


Assuntos
Bifidobacterium/genética , Metilação de DNA , Genômica/métodos , Mapeamento por Restrição/métodos , Análise de Sequência/métodos , Sequência de Bases , Escherichia coli/genética , Genoma Bacteriano/genética , Plasmídeos/genética
11.
PLoS One ; 8(7): e68731, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894338

RESUMO

Lactobacillus paracasei is a member of the normal human and animal gut microbiota and is used extensively in the food industry in starter cultures for dairy products or as probiotics. With the development of low-cost, high-throughput sequencing techniques it has become feasible to sequence many different strains of one species and to determine its "pan-genome". We have sequenced the genomes of 34 different L. paracasei strains, and performed a comparative genomics analysis. We analysed genome synteny and content, focussing on the pan-genome, core genome and variable genome. Each genome was shown to contain around 2800-3100 protein-coding genes, and comparative analysis identified over 4200 ortholog groups that comprise the pan-genome of this species, of which about 1800 ortholog groups make up the conserved core. Several factors previously associated with host-microbe interactions such as pili, cell-envelope proteinase, hydrolases p40 and p75 or the capacity to produce short branched-chain fatty acids (bkd operon) are part of the L. paracasei core genome present in all analysed strains. The variome consists mainly of hypothetical proteins, phages, plasmids, transposon/conjugative elements, and known functions such as sugar metabolism, cell-surface proteins, transporters, CRISPR-associated proteins, and EPS biosynthesis proteins. An enormous variety and variability of sugar utilization gene cassettes were identified, with each strain harbouring between 25-53 cassettes, reflecting the high adaptability of L. paracasei to different niches. A phylogenomic tree was constructed based on total genome contents, and together with an analysis of horizontal gene transfer events we conclude that evolution of these L. paracasei strains is complex and not always related to niche adaptation. The results of this genome content comparison was used, together with high-throughput growth experiments on various carbohydrates, to perform gene-trait matching analysis, in order to link the distribution pattern of a specific phenotype to the presence/absence of specific sets of genes.


Assuntos
Variação Genética , Genoma Bacteriano , Genômica , Lactobacillus/genética , Metabolismo dos Carboidratos/genética , Análise por Conglomerados , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Ácidos Graxos/metabolismo , Ordem dos Genes , Transferência Genética Horizontal , Lactobacillus/classificação , Lactobacillus/metabolismo , Anotação de Sequência Molecular , Filogenia , Plasmídeos/genética
12.
mBio ; 3(3)2012.
Artigo em Inglês | MEDLINE | ID: mdl-22617141

RESUMO

UNLABELLED: In an attempt to explore the microbial content of functionally critical niches of the mouse gastrointestinal tract, we targeted molecular microbial diagnostics of the crypts that contain the intestinal stem cells, which account for epithelial regeneration. As current evidence indicates, the gut microbiota affects epithelial regeneration; bacteria that are likely to primarily participate in this essential step of the gut, microbiota cross talk, have been identified. We show in this article that only the cecal and colonic crypts harbor resident microbiota in the mouse and that regardless of the line and breeding origin of these mice, this bacterial population is unexpectedly dominated by aerobic genera. Interestingly, this microbiota resembles the restricted microbiota found in the midgut of invertebrates; thus, the presence of our so-called "crypt-specific core microbiota" (CSCM) in the mouse colon potentially reflects a coevolutionary process under selective conditions that can now be addressed. We suggest that CSCM could play both a protective and a homeostatic role within the colon. This article is setting the bases for such studies, particularly by providing a bona fide--and essentially cultivable--crypt microbiota of reference. IMPORTANCE: Metagenomic typing of the whole-gut luminal microbiome was recently provided, revealing great opportunities for physiological and physiopathological analysis of the host-microbiota interface. On this basis, it appears increasingly important to analyze which niches of the gut exposed to a particular microbiota are of major functional importance, specifically focusing on the crypt, which accounts for permanent epithelial renewal, and to analyze how this microbiota compares to its luminal counterpart in composition and quantity. Crypt-specific core microbiotas may show themselves as important elements regarding crypt protection and homeostasis of its functions.


Assuntos
Bactérias/isolamento & purificação , Colo/microbiologia , Metagenoma , Camundongos/microbiologia , Animais , Bactérias/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL
13.
Sci Transl Med ; 3(106): 106ra106, 2011 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22030749

RESUMO

Understanding how the human gut microbiota and host are affected by probiotic bacterial strains requires carefully controlled studies in humans and in mouse models of the gut ecosystem where potentially confounding variables that are difficult to control in humans can be constrained. Therefore, we characterized the fecal microbiomes and metatranscriptomes of adult female monozygotic twin pairs through repeated sampling 4 weeks before, 7 weeks during, and 4 weeks after consumption of a commercially available fermented milk product (FMP) containing a consortium of Bifidobacterium animalis subsp. lactis, two strains of Lactobacillus delbrueckii subsp. bulgaricus, Lactococcus lactis subsp. cremoris, and Streptococcus thermophilus. In addition, gnotobiotic mice harboring a 15-species model human gut microbiota whose genomes contain 58,399 known or predicted protein-coding genes were studied before and after gavage with all five sequenced FMP strains. No significant changes in bacterial species composition or in the proportional representation of genes encoding known enzymes were observed in the feces of humans consuming the FMP. Only minimal changes in microbiota configuration were noted in mice after single or repeated gavage with the FMP consortium. However, RNA-Seq analysis of fecal samples and follow-up mass spectrometry of urinary metabolites disclosed that introducing the FMP strains into mice results in significant changes in expression of microbiome-encoded enzymes involved in numerous metabolic pathways, most prominently those related to carbohydrate metabolism. B. animalis subsp. lactis, the dominant persistent member of the FMP consortium in gnotobiotic mice, up-regulates a locus in vivo that is involved in the catabolism of xylooligosaccharides, a class of glycans widely distributed in fruits, vegetables, and other foods, underscoring the importance of these sugars to this bacterial species. The human fecal metatranscriptome exhibited significant changes, confined to the period of FMP consumption, that mirror changes in gnotobiotic mice, including those related to plant polysaccharide metabolism. These experiments illustrate a translational research pipeline for characterizing the effects of FMPs on the human gut microbiome.


Assuntos
Produtos Fermentados do Leite/microbiologia , Trato Gastrointestinal/microbiologia , Metagenoma/fisiologia , Animais , Bifidobacterium , Feminino , Vida Livre de Germes , Humanos , Lactobacillus , Masculino , Metagenoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Probióticos/administração & dosagem , Gêmeos Monozigóticos
14.
Int J Food Microbiol ; 132(1): 54-8, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19361877

RESUMO

We applied two methods of broth microdilution and Etest for measuring minimal inhibition concentration (MIC) of lactic acid bacteria and bifidobacteria for 15 antimicrobial agents to compare the feasibility, reproducibility, and equivalence of the two methods. Both methods were originally described by the European projects PROSAFE and ACE-ART. In 84% combinations of strains and antimicrobial agents MIC differences between the two methods were within one Log(2) dilution. In the case of rifampicin the difference between the two methods was more than ten-fold. We further determined MICs of 70 strains (14 strains of Lactobacillus delbrueckii ssp. bulgaricus, 16 strains of Lactococcus lactis, 30 strains of Streptococcus thermophilus, and 10 strains of Bifidobacterium longum) by the broth microdilution method. In most cases, MIC distributions were uni-modal and within 5 Log(2) dilutions except for the MIC distribution of L. lactis to the aminoglycoside group which was broader. These data are a good basis for improving knowledge of antimicrobial susceptibility of lactic acid bacteria and bifidobacteria, and can be used to revise tentative epidemiological cut-off values.


Assuntos
Antibacterianos/farmacologia , Bifidobacterium/efeitos dos fármacos , Lactobacillaceae/efeitos dos fármacos , Testes de Sensibilidade Microbiana/métodos , Bifidobacterium/crescimento & desenvolvimento , Lactobacillaceae/crescimento & desenvolvimento
15.
Appl Environ Microbiol ; 73(20): 6601-11, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17704267

RESUMO

Robust genotyping methods for Lactobacillus casei are needed for strain tracking and collection management, as well as for population biology research. A collection of 52 strains initially labeled L. casei or Lactobacillus paracasei was first subjected to rplB gene sequencing together with reference strains of Lactobacillus zeae, Lactobacillus rhamnosus, and other species. Phylogenetic analysis showed that all 52 strains belonged to a single compact L. casei-L. paracasei sequence cluster, together with strain CIP107868 (= ATCC 334) but clearly distinct from L. rhamnosus and from a cluster with L. zeae and CIP103137(T) (= ATCC 393(T)). The strains were genotyped using amplified fragment length polymorphism, multilocus sequence typing based on internal portions of the seven housekeeping genes fusA, ileS, lepA, leuS, pyrG, recA, and recG, and tandem repeat variation (multilocus variable-number tandem repeats analysis [MLVA] using nine loci). Very high concordance was found between the three methods. Although amounts of nucleotide variation were low for the seven genes (pi ranging from 0.0038 to 0.0109), 3 to 12 alleles were distinguished, resulting in 31 sequence types. One sequence type (ST1) was frequent (17 strains), but most others were represented by a single strain. Attempts to subtype ST1 strains by MLVA, ribotyping, clustered regularly interspaced short palindromic repeat characterization, and single nucleotide repeat variation were unsuccessful. We found clear evidence for homologous recombination during the diversification of L. casei clones, including a putative intragenic import of DNA into one strain. Nucleotides were estimated to change four times more frequently by recombination than by mutation. However, statistical congruence between individual gene trees was retained, indicating that recombination is not frequent enough to disrupt the phylogenetic signal. The developed multilocus sequence typing scheme should be useful for future studies of L. casei strain diversity and evolution.


Assuntos
Proteínas de Bactérias/genética , Lacticaseibacillus casei/classificação , Lacticaseibacillus casei/genética , Recombinação Genética , Proteínas Ribossômicas/genética , Análise de Sequência de DNA , Proteínas de Bactérias/metabolismo , Técnicas de Tipagem Bacteriana , Sequência de Bases , DNA Bacteriano/análise , Dados de Sequência Molecular , Filogenia , Proteínas Ribossômicas/metabolismo
16.
Antonie Van Leeuwenhoek ; 82(1-4): 187-216, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12369188

RESUMO

Lactic acid bacteria (LAB) constitute a heterogeneous group of bacteria that are traditionally used to produce fermented foods. The industrialization of food bio-transformations increased the economical importance of LAB, as they play a crucial role in the development of the organoleptique and hygienic quality of fermented products. Therefore, the reliability of starter strains in terms of quality and functional properties (important for the development of aroma and texture), but also in terms of growth performance and robustness has become essential. These strains should resist to adverse conditions encountered in industrial processes, for example during starter handling and storage (freeze-drying, freezing or spray-drying). The development of new applications such as life vaccines and probiotic foods reinforces the need for robust LAB since they may have to survive in the digestive tract, resist the intestinal flora, maybe colonize the digestive or uro-genital mucosa and express specific functions under conditions that are unfavorable to growth (for example, during stationary phase or storage). Also in nature, the ability to quickly respond to stress is essential for survival and it is now well established that LAB, like other bacteria, evolved defense mechanisms against stress that allow them to withstand harsh conditions and sudden environmental changes. While genes implicated in stress responses are numerous, in LAB the levels of characterization of their actual role and regulation differ widely between species. The functional conservation of several stress proteins (for example, HS proteins, Csp, etc) and of some of their regulators (for example, HrcA, CtsR) renders even more striking the differences that exist between LAB and the classical model micro-organisms. Among the differences observed between LAB species and B. subtilis, one of the most striking is the absence of a sigma B orthologue in L. lactis ssp. lactis as well as in at least two streptococci and probably E. faecalis. The overview of LAB stress responses also reveals common aspects of stress responses. As in other bacteria, adaptive responses appear to be a usual mode of stress protection in LAB. However, the cross-protection to other stress often induced by the expression of a given adaptive response, appears to vary between species. This observation suggests that the molecular bases of adaptive responses are, at least in part, species (or even subspecies) specific. A better understanding of the mechanisms of stress resistance should allow to understand the bases of the adaptive responses and cross protection, and to rationalize their exploitation to prepare LAB to industrial processes. Moreover, the identification of crucial stress related genes will reveal targets i) for specific manipulation (to promote or limit growth), ii) to develop tools to screen for tolerant or sensitive strains and iii) to evaluate the fitness and level of adaptation of a culture. In this context, future genome and transcriptome analyses will undoubtedly complement the proteome and genetic information available today, and shed a new light on the perception of, and the response to, stress by lactic acid bacteria.


Assuntos
Lactobacillus/fisiologia , Lactococcus/fisiologia , Aclimatação , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/biossíntese , Temperatura Baixa , Lactobacillus/crescimento & desenvolvimento , Lactococcus/crescimento & desenvolvimento , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA