Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(3): E367-E375, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28049818

RESUMEN

Directed modulation of the colonic bacteria to metabolize lactose effectively is a potentially useful approach to improve lactose digestion and tolerance. A randomized, double-blind, multisite placebo-controlled trial conducted in human subjects demonstrated that administration of a highly purified (>95%) short-chain galactooligosaccharide (GOS), designated "RP-G28," significantly improved clinical outcomes for lactose digestion and tolerance. In these individuals, stool samples were collected pretreatment (day 0), after GOS treatment (day 36), and 30 d after GOS feeding stopped and consumption of dairy products was encouraged (day 66). In this study, changes in the fecal microbiome were investigated using 16S rRNA amplicon pyrosequencing and high-throughput quantitative PCR. At day 36, bifidobacterial populations were increased in 27 of 30 of GOS subjects (90%), demonstrating a bifidogenic response in vivo. Relative abundance of lactose-fermenting Bifidobacterium, Faecalibacterium, and Lactobacillus were significantly increased in response to GOS. When dairy was introduced into the diet, lactose-fermenting Roseburia species increased from day 36 to day 66. The results indicated a definitive change in the fecal microbiome of lactose-intolerant individuals, increasing the abundance of lactose-metabolizing bacteria that were responsive to dietary adaptation to GOS. This change correlated with clinical outcomes of improved lactose tolerance.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Lactosa/metabolismo , Oligosacáridos/administración & dosificación , Adulto , Bifidobacterium/efectos de los fármacos , Colon/metabolismo , Método Doble Ciego , Faecalibacterium/efectos de los fármacos , Heces/microbiología , Femenino , Humanos , Lactobacillus/efectos de los fármacos , Masculino , ARN Ribosómico 16S/metabolismo
2.
EMBO J ; 34(7): 881-95, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25666591

RESUMEN

Intestinal immune regulatory signals govern gut homeostasis. Breakdown of such regulatory mechanisms may result in inflammatory bowel disease (IBD). Lactobacillus acidophilus contains unique surface layer proteins (Slps), including SlpA, SlpB, SlpX, and lipoteichoic acid (LTA), which interact with pattern recognition receptors to mobilize immune responses. Here, to elucidate the role of SlpA in protective immune regulation, the NCK2187 strain, which solely expresses SlpA, was generated. NCK2187 and its purified SlpA bind to the C-type lectin SIGNR3 to exert regulatory signals that result in mitigation of colitis, maintenance of healthy gastrointestinal microbiota, and protected gut mucosal barrier function. However, such protection was not observed in Signr3(-/-) mice, suggesting that the SlpA/SIGNR3 interaction plays a key regulatory role in colitis. Our work presents critical insights into SlpA/SIGNR3-induced responses that are integral to the potential development of novel biological therapies for autoinflammatory diseases, including IBD.


Asunto(s)
Antígenos CD/inmunología , Proteínas Bacterianas/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Lactobacillus acidophilus/inmunología , Lectinas Tipo C/inmunología , Animales , Antígenos CD/genética , Proteínas Bacterianas/genética , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Lactobacillus acidophilus/genética , Lectinas Tipo C/genética , Lipopolisacáridos/genética , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Unión Proteica/genética , Unión Proteica/inmunología , Ácidos Teicoicos/genética , Ácidos Teicoicos/inmunología
3.
Microbiology (Reading) ; 164(9): 1098-1111, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30024364

RESUMEN

Lactobacillus gasseri is a human commensal which carries CRISPR-Cas, an adaptive immune system that protects the cell from invasive mobile genetic elements (MGEs). However, MGEs occasionally escape CRISPR targeting due to DNA mutations that occur in sequences involved in CRISPR interference. To better understand CRISPR escape processes, a plasmid interference assay was used to screen for mutants that escape CRISPR-Cas targeting. Plasmids containing a target sequence and a protospacer adjacent motif (PAM) were transformed for targeting by the native CRISPR-Cas system. Although the primary outcome of the assay was efficient interference, a small proportion of the transformed population overcame targeting. Mutants containing plasmids that had escaped were recovered to investigate the genetic routes of escape and their relative frequencies. Deletion of the targeting spacer in the native CRISPR array was the dominant pattern of escape, accounting for 52-70 % of the mutants from two L. gasseri strains. We repeatedly observed internal deletions in the chromosomal CRISPR array, characterized by polarized excisions from the leader end that spanned 1-15 spacers, and systematically included the leader-proximal targeting spacer. This study shows that deletions of spacers within CRISPR arrays constitute a key escape mechanism to evade CRISPR targeting, while preserving the functionality of the CRISPR-Cas system. This mechanism enables cells to maintain an active immune system, but allows the uptake of potentially beneficial plasmids. Our study revealed the co-occurrence of other genomic mutations associated with various phenotypes, showing how this selection process uncovers population diversification.


Asunto(s)
Secuencias Repetitivas Esparcidas , Lactobacillus gasseri/genética , Mutación , Eliminación de Secuencia , Proteína 9 Asociada a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Plásmidos , Recombinación Genética , Transformación Bacteriana
4.
Proc Natl Acad Sci U S A ; 112(26): 8076-81, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26080436

RESUMEN

Genomic analysis of Streptococcus thermophilus revealed that mobile genetic elements (MGEs) likely contributed to gene acquisition and loss during evolutionary adaptation to milk. Clustered regularly interspaced short palindromic repeats-CRISPR-associated genes (CRISPR-Cas), the adaptive immune system in bacteria, limits genetic diversity by targeting MGEs including bacteriophages, transposons, and plasmids. CRISPR-Cas systems are widespread in streptococci, suggesting that the interplay between CRISPR-Cas systems and MGEs is one of the driving forces governing genome homeostasis in this genus. To investigate the genetic outcomes resulting from CRISPR-Cas targeting of integrated MGEs, in silico prediction revealed four genomic islands without essential genes in lengths from 8 to 102 kbp, totaling 7% of the genome. In this study, the endogenous CRISPR3 type II system was programmed to target the four islands independently through plasmid-based expression of engineered CRISPR arrays. Targeting lacZ within the largest 102-kbp genomic island was lethal to wild-type cells and resulted in a reduction of up to 2.5-log in the surviving population. Genotyping of Lac(-) survivors revealed variable deletion events between the flanking insertion-sequence elements, all resulting in elimination of the Lac-encoding island. Chimeric insertion sequence footprints were observed at the deletion junctions after targeting all of the four genomic islands, suggesting a common mechanism of deletion via recombination between flanking insertion sequences. These results established that self-targeting CRISPR-Cas systems may direct significant evolution of bacterial genomes on a population level, influencing genome homeostasis and remodeling.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Genoma Bacteriano , Streptococcus thermophilus/genética , Eliminación de Gen , Homeostasis , Recombinación Homóloga , Streptococcus thermophilus/crecimiento & desarrollo
5.
Appl Environ Microbiol ; 83(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28411221

RESUMEN

Of the few predicted extracellular glycan-active enzymes, glycoside hydrolase family 13 subfamily 14 (GH13_14) pullulanases are the most common in human gut lactobacilli. These enzymes share a unique modular organization, not observed in other bacteria, featuring a catalytic module, two starch binding modules, a domain of unknown function, and a C-terminal surface layer association protein (SLAP) domain. Here, we explore the specificity of a representative of this group of pullulanases, Lactobacillus acidophilus Pul13_14 (LaPul13_14), and its role in branched α-glucan metabolism in the well-characterized Lactobacillus acidophilus NCFM, which is widely used as a probiotic. Growth experiments with L. acidophilus NCFM on starch-derived branched substrates revealed a preference for α-glucans with short branches of about two to three glucosyl moieties over amylopectin with longer branches. Cell-attached debranching activity was measurable in the presence of α-glucans but was repressed by glucose. The debranching activity is conferred exclusively by LaPul13_14 and is abolished in a mutant strain lacking a functional LaPul13_14 gene. Hydrolysis kinetics of recombinant LaPul13_14 confirmed the preference for short-branched α-glucan oligomers consistent with the growth data. Curiously, this enzyme displayed the highest catalytic efficiency and the lowest Km reported for a pullulanase. Inhibition kinetics revealed mixed inhibition by ß-cyclodextrin, suggesting the presence of additional glucan binding sites besides the active site of the enzyme, which may contribute to the unprecedented substrate affinity. The enzyme also displays high thermostability and higher activity in the acidic pH range, reflecting adaptation to the physiologically challenging conditions in the human gut.IMPORTANCE Starch is one of the most abundant glycans in the human diet. Branched α-1,6-glucans in dietary starch and glycogen are nondegradable by human enzymes and constitute a metabolic resource for the gut microbiota. The role of health-beneficial lactobacilli prevalent in the human small intestine in starch metabolism remains unexplored in contrast to colonic bacterial residents. This study highlights the pivotal role of debranching enzymes in the breakdown of starchy branched α-glucan oligomers (α-limit dextrins) by human gut lactobacilli exemplified by Lactobacillus acidophilus NCFM, which is one of the best-characterized strains used as probiotics. Our data bring novel insight into the metabolic preference of L. acidophilus for α-glucans with short α-1,6-branches. The unprecedented affinity of the debranching enzyme that confers growth on these substrates reflects its adaptation to the nutrient-competitive gut ecological niche and constitutes a potential advantage in cross-feeding from human and bacterial dietary starch metabolism.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glucanos/metabolismo , Glicósido Hidrolasas/metabolismo , Lactobacillus acidophilus/enzimología , Amilopectina/química , Amilopectina/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Estabilidad de Enzimas , Tracto Gastrointestinal/microbiología , Glucanos/química , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Humanos , Hidrólisis , Cinética , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/metabolismo , Especificidad por Sustrato
6.
Proteomics ; 16(9): 1361-75, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26959526

RESUMEN

Whole cell and surface proteomes were analyzed together with adhesive properties of the probiotic bacterium Lactobacillus acidophilus NCFM (NCFM) grown on the emerging prebiotic raffinose, exemplifying a synbiotic. Adhesion of NCFM to mucin and intestinal HT-29 cells increased three-fold after culture with raffinose versus glucose, as also visualized by scanning electron microscopy. Comparative proteomics using 2D-DIGE showed 43 unique proteins to change in relative abundance in whole cell lysates from NCFM grown on raffinose compared to glucose. Furthermore, 14 unique proteins in 18 spots of the surface subproteome underwent changes identified by differential 2DE, including elongation factor G, thermostable pullulanase, and phosphate starvation inducible stress-related protein increasing in a range of +2.1 - +4.7 fold. By contrast five known moonlighting proteins decreased in relative abundance by up to -2.4 fold. Enzymes involved in raffinose catabolism were elevated in the whole cell proteome; α-galactosidase (+13.9 fold); sucrose phosphorylase (+5.4 fold) together with metabolic enzymes from the Leloir pathway for galactose utilization and the glycolysis; ß-galactosidase (+5.7 fold); galactose (+2.9/+3.1 fold) and fructose (+2.8 fold) kinases. The insights at the molecular and cellular levels contributed to the understanding of the interplay of a synbiotic composed of NCFM and raffinose with the host.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Lactobacillus acidophilus/efectos de los fármacos , Probióticos/metabolismo , Proteoma/genética , Rafinosa/farmacología , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Galactosa/metabolismo , Ontología de Genes , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Células HT29 , Humanos , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/crecimiento & desarrollo , Lactobacillus acidophilus/metabolismo , Anotación de Secuencia Molecular , Factor G de Elongación Peptídica/genética , Factor G de Elongación Peptídica/metabolismo , Prebióticos , Proteoma/metabolismo , Coloración y Etiquetado , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo
7.
Appl Environ Microbiol ; 82(18): 5687-97, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27422832

RESUMEN

UNLABELLED: Autolysins, also known as peptidoglycan hydrolases, are enzymes that hydrolyze specific bonds within bacterial cell wall peptidoglycan during cell division and daughter cell separation. Within the genome of Lactobacillus acidophilus NCFM, there are 11 genes encoding proteins with peptidoglycan hydrolase catalytic domains, 9 of which are predicted to be functional. Notably, 5 of the 9 putative autolysins in L. acidophilus NCFM are S-layer-associated proteins (SLAPs) noncovalently colocalized along with the surface (S)-layer at the cell surface. One of these SLAPs, AcmB, a ß-N-acetylglucosaminidase encoded by the gene lba0176 (acmB), was selected for functional analysis. In silico analysis revealed that acmB orthologs are found exclusively in S-layer- forming species of Lactobacillus Chromosomal deletion of acmB resulted in aberrant cell division, autolysis, and autoaggregation. Complementation of acmB in the ΔacmB mutant restored the wild-type phenotype, confirming the role of this SLAP in cell division. The absence of AcmB within the exoproteome had a pleiotropic effect on the extracellular proteins covalently and noncovalently bound to the peptidoglycan, which likely led to the observed decrease in the binding capacity of the ΔacmB strain for mucin and extracellular matrices fibronectin, laminin, and collagen in vitro These data suggest a functional association between the S-layer and the multiple autolysins noncovalently colocalized at the cell surface of L. acidophilus NCFM and other S-layer-producing Lactobacillus species. IMPORTANCE: Lactobacillus acidophilus is one of the most widely used probiotic microbes incorporated in many dairy foods and dietary supplements. This organism produces a surface (S)-layer, which is a self-assembling crystalline array found as the outermost layer of the cell wall. The S-layer, along with colocalized associated proteins, is an important mediator of probiotic activity through intestinal adhesion and modulation of the mucosal immune system. However, there is still a dearth of information regarding the basic cellular and evolutionary function of S-layers. Here, we demonstrate that multiple autolysins, responsible for breaking down the cell wall during cell division, are associated with the S-layer. Deletion of the gene encoding one of these S-layer-associated autolysins confirmed its autolytic role and resulted in reduced binding capacity to mucin and intestinal extracellular matrices. These data suggest a functional association between the S-layer and autolytic activity through the extracellular presentation of autolysins.


Asunto(s)
Acetilglucosaminidasa/metabolismo , Lactobacillus acidophilus/enzimología , Glicoproteínas de Membrana/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Acetilglucosaminidasa/genética , Adhesión Bacteriana , Bacteriólisis , División Celular , Pared Celular/química , Biología Computacional , Eliminación de Gen , Prueba de Complementación Genética , Lactobacillus acidophilus/genética , Glicoproteínas de Membrana/genética , N-Acetil Muramoil-L-Alanina Amidasa/genética , Peptidoglicano/análisis
8.
Appl Environ Microbiol ; 82(20): 6091-6101, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27496774

RESUMEN

Clostridium botulinum and Bacillus anthracis produce potent toxins that cause severe disease in humans. New and improved vaccines are needed for both of these pathogens. For mucosal vaccine delivery using lactic acid bacteria, chromosomal expression of antigens is preferred over plasmid-based expression systems, as chromosomal expression circumvents plasmid instability and the need for antibiotic pressure. In this study, we constructed three strains of Lactobacillus acidophilus NCFM expressing from the chromosome (i) the nontoxic host receptor-binding domain of the heavy chain of Clostridium botulinum serotype A neurotoxin (BoNT/A-Hc), (ii) the anthrax protective antigen (PA), and (iii) both the BoNT/A-Hc and the PA. The BoNT/A-Hc vaccine cassette was engineered to contain the signal peptide from the S-layer protein A from L. acidophilus and a dendritic-cell-targeting peptide. A chromosomal region downstream of lba0889 carrying a highly expressed enolase gene was selected for insertion of the vaccine cassettes. Western blot analysis confirmed the heterologous expression of the two antigens from plasmid and chromosome locations. Stability assays demonstrated loss of the vaccine cassettes from expression plasmids without antibiotic maintenance. RNA sequencing showed high expression of each antigen and that insertion of the vaccine cassettes had little to no effect on the transcription of other genes in the chromosome. This study demonstrated that chromosomal integrative recombinant strains are promising vaccine delivery vehicles when targeted into high-expression chromosomal regions. Levels of expression match high-copy-number plasmids and eliminate the requirement for antibiotic selective maintenance of recombinant plasmids. IMPORTANCE: Clostridium botulinum and Bacillus anthracis produce potent neurotoxins that pose a biochemical warfare concern; therefore, effective vaccines against these bacteria are required. Chromosomal expression of antigens is preferred over plasmid-based expression systems since expressing antigens from a chromosomal location confers an advantage to the vaccine strains by eliminating the antibiotic maintenance required for plasmids and negates issues with plasmid instability that would result in loss of the antigen. Lactic acid bacteria, including Lactobacillus acidophilus, have shown potential for mucosal vaccine delivery, as L. acidophilus is bile and acid tolerant, allowing transit through the gastrointestinal tract where cells interact with host epithelial and immune cells, including dendritic cells. In this study, we successfully expressed C. botulinum and B. anthracis antigens in the probiotic L. acidophilus strain NCFM. Both antigens were highly expressed individually or in tandem from the chromosome of L. acidophilus.


Asunto(s)
Carbunco/microbiología , Bacillus anthracis/genética , Toxinas Botulínicas Tipo A/genética , Botulismo/microbiología , Clostridium botulinum/genética , Expresión Génica , Lactobacillus acidophilus/genética , Carbunco/prevención & control , Bacillus anthracis/metabolismo , Vacunas Bacterianas/genética , Vacunas Bacterianas/metabolismo , Toxinas Botulínicas Tipo A/metabolismo , Botulismo/prevención & control , Cromosomas Bacterianos/genética , Cromosomas Bacterianos/metabolismo , Clostridium botulinum/metabolismo , Lactobacillus acidophilus/metabolismo
9.
Appl Environ Microbiol ; 82(4): 1286-94, 2016 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-26682852

RESUMEN

The Bacillus thuringiensis crystal (Cry) protein Cry5B (140 kDa) and a truncated version of the protein, tCry5B (79 kDa), are lethal to nematodes. Genes encoding the two proteins were separately cloned into a high-copy-number vector with a strong constitutive promoter (pTRK593) in Lactococcus lactis for potential oral delivery against parasitic nematode infections. Western blots using a Cry5B-specific antibody revealed that constitutively expressed Cry5B and tCry5B were present in both cells and supernatants. To increase production, cry5B was cloned into the high-copy-number plasmid pMSP3535H3, carrying a nisin-inducible promoter. Immunoblotting revealed that 3 h after nisin induction, intracellular Cry5B was strongly induced at 200 ng/ml nisin, without adversely affecting cell viability or cell membrane integrity. Both Cry5B genes were also cloned into plasmid pTRK1061, carrying a promoter and encoding a transcriptional activator that invoke low-level expression of prophage holin and lysin genes in Lactococcus lysogens, resulting in a leaky phenotype. Cry5B and tCry5B were actively expressed in the lysogenic strain L. lactis KP1 and released into cell supernatants without affecting culture growth. Lactate dehydrogenase (LDH) assays indicated that Cry5B, but not LDH, leaked from the bacteria. Lastly, using intracellular lysates from L. lactis cultures expressing both Cry5B and tCry5B, in vivo challenges of Caenorhabditis elegans worms demonstrated that the Cry proteins were biologically active. Taken together, these results indicate that active Cry5B proteins can be expressed intracellularly in and released extracellularly from L. lactis, showing potential for future use as an anthelminthic that could be delivered orally in a food-grade microbe.


Asunto(s)
Antihelmínticos/metabolismo , Proteínas Bacterianas/biosíntesis , Caenorhabditis elegans/efectos de los fármacos , Endotoxinas/biosíntesis , Expresión Génica , Proteínas Hemolisinas/biosíntesis , Lactococcus lactis/metabolismo , Proteínas Recombinantes/biosíntesis , Animales , Antihelmínticos/farmacología , Toxinas de Bacillus thuringiensis , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Western Blotting , Clonación Molecular , Endotoxinas/genética , Endotoxinas/farmacología , Dosificación de Gen , Vectores Genéticos , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/farmacología , Lactococcus lactis/genética , Viabilidad Microbiana , Nisina/metabolismo , Plásmidos , Regiones Promotoras Genéticas , Transporte de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Análisis de Supervivencia , Activación Transcripcional/efectos de los fármacos
10.
Appl Environ Microbiol ; 82(9): 2676-2685, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26921419

RESUMEN

Bacterial surface layers (S-layers) are crystalline arrays of self-assembling proteinaceous subunits called S-layer proteins (Slps) that comprise the outermost layer of the cell envelope. Many additional proteins that are associated with or embedded within the S-layer have been identified in Lactobacillus acidophilus NCFM, an S-layer-forming bacterium that is widely used in fermented dairy products and probiotic supplements. One putative S-layer-associated protein (SLAP), LBA0191, was predicted to mediate adhesion to fibronectin based on the in silico detection of a fibronectin-binding domain. Fibronectin is a major component of the extracellular matrix (ECM) of intestinal epithelial cells. Adhesion to intestinal epithelial cells is considered an important trait for probiotic microorganisms during transit and potential association with the intestinal mucosa. To investigate the functional role of LBA0191 (designated FbpB) in L. acidophilus NCFM, an fbpB-deficient strain was constructed. The L. acidophilus mutant with a deletion off bpB lost the ability to adhere to mucin and fibronectin in vitro Homologues off bpB were identified in five additional putative S-layer-forming species, but no homologues were detected in species outside theL. acidophilus homology group.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fibronectinas/metabolismo , Lactobacillus acidophilus/metabolismo , Glicoproteínas de Membrana/metabolismo , Adhesión Bacteriana/fisiología , Proteínas Bacterianas/química , Intestinos/microbiología , Lactobacillus acidophilus/química , Lactobacillus acidophilus/genética , Mutación , Filogenia , Unión Proteica
11.
Appl Environ Microbiol ; 82(1): 134-45, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26475115

RESUMEN

The Lactobacillus acidophilus homology group comprises Gram-positive species that include L. acidophilus, L. helveticus, L. crispatus, L. amylovorus, L. gallinarum, L. delbrueckii subsp. bulgaricus, L. gasseri, and L. johnsonii. While these bacteria are closely related, they have varied ecological lifestyles as dairy and food fermenters, allochthonous probiotics, or autochthonous commensals of the host gastrointestinal tract. Bacterial cell surface components play a critical role in the molecular dialogue between bacteria and interaction signaling with the intestinal mucosa. Notably, the L. acidophilus complex is distinguished in two clades by the presence or absence of S-layers, which are semiporous crystalline arrays of self-assembling proteinaceous subunits found as the outermost layer of the bacterial cell wall. In this study, S-layer-associated proteins (SLAPs) in the exoproteomes of various S-layer-forming Lactobacillus species were proteomically identified, genomically compared, and transcriptionally analyzed. Four gene regions encoding six putative SLAPs were conserved in the S-layer-forming Lactobacillus species but not identified in the extracts of the closely related progenitor, L. delbrueckii subsp. bulgaricus, which does not produce an S-layer. Therefore, the presence or absence of an S-layer has a clear impact on the exoproteomic composition of Lactobacillus species. This proteomic complexity and differences in the cell surface properties between S-layer- and non-S-layer-forming lactobacilli reveal the potential for SLAPs to mediate intimate probiotic interactions and signaling with the host intestinal mucosa.


Asunto(s)
Proteínas Bacterianas/química , Lactobacillus/genética , Glicoproteínas de Membrana/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Lactobacillus/química , Lactobacillus/clasificación , Lactobacillus/metabolismo , Lactobacillus acidophilus/química , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Datos de Secuencia Molecular , Filogenia , Proteómica
12.
Microbiology (Reading) ; 161(Pt 2): 311-321, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25500495

RESUMEN

Surface proteins of probiotic microbes, including Lactobacillus acidophilus and Lactobacillus gasseri, are believed to promote retention in the gut and mediate host-bacterial communications. Sortase, an enzyme that covalently couples a subset of extracellular proteins containing an LPXTG motif to the cell surface, is of particular interest in characterizing bacterial adherence and communication with the mucosal immune system. A sortase gene, srtA, was identified in L. acidophilus NCFM (LBA1244) and L. gasseri ATCC 33323 (LGAS_0825). Additionally, eight and six intact sortase-dependent proteins were predicted in L. acidophilus and L. gasseri, respectively. Due to the role of sortase in coupling these proteins to the cell wall, ΔsrtA deletion mutants of L. acidophilus and L. gasseri were created using the upp-based counterselective gene replacement system. Inactivation of sortase did not cause significant alteration in growth or survival in simulated gastrointestinal juices. Meanwhile, both ΔsrtA mutants showed decreased adhesion to porcine mucin in vitro. Murine dendritic cells exposed to the ΔsrtA mutant of L. acidophilus or L. gasseri induced lower levels of pro-inflammatory cytokines TNF-α and IL-12, respectively, compared with the parent strains. In vivo co-colonization of the L. acidophilus ΔsrtA mutant and its parent strain in germ-free 129S6/SvEv mice resulted in a significant one-log reduction of the ΔsrtA mutant population. Additionally, a similar reduction of the ΔsrtA mutant was observed in the caecum. This study shows for the first time that sortase-dependent proteins contribute to gut retention of probiotic microbes in the gastrointestinal tract.


Asunto(s)
Aminoaciltransferasas/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Tracto Gastrointestinal/microbiología , Lactobacillus acidophilus/enzimología , Lactobacillus acidophilus/fisiología , Lactobacillus/enzimología , Lactobacillus/fisiología , Aminoaciltransferasas/genética , Aminoaciltransferasas/inmunología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Células CACO-2 , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Tracto Gastrointestinal/inmunología , Humanos , Inmunomodulación , Lactobacillus/genética , Lactobacillus/crecimiento & desarrollo , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/crecimiento & desarrollo , Ratones , Porcinos , Factor de Necrosis Tumoral alfa/inmunología
14.
Proc Natl Acad Sci U S A ; 109(26): 10462-7, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22689992

RESUMEN

An imbalance of commensal bacteria and their gene products underlies mucosal and, in particular, gastrointestinal inflammation and a predisposition to cancer. Lactobacillus species have received considerable attention as examples of beneficial microbiota. We have reported previously that deletion of the phosphoglycerol transferase gene that is responsible for lipoteichoic acid (LTA) biosynthesis in Lactobacillus acidophilus (NCK2025) rendered this bacterium able to significantly protect mice against induced colitis when delivered orally. Here we report that oral treatment with LTA-deficient NCK2025 normalizes innate and adaptive pathogenic immune responses and causes regression of established colonic polyps. This study reveals the proinflammatory role of LTA and the ability of LTA-deficient L. acidophilus to regulate inflammation and protect against colonic polyposis in a unique mouse model.


Asunto(s)
Poliposis Adenomatosa del Colon/inmunología , Lactobacillus acidophilus/genética , Lipopolisacáridos/genética , Ácidos Teicoicos/genética , Poliposis Adenomatosa del Colon/patología , Animales , Ratones , Linfocitos T Reguladores/inmunología
15.
Mol Microbiol ; 89(6): 1187-200, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23879596

RESUMEN

Glycogen metabolism contributes to energy storage and various physiological functions in some prokaryotes, including colonization persistence. A role for glycogen metabolism is proposed on the survival and fitness of Lactobacillus acidophilus, a probiotic microbe, in the human gastrointestinal environment. L. acidophilus NCFM possesses a glycogen metabolism (glg) operon consisting of glgBCDAP-amy-pgm genes. Expression of the glg operon and glycogen accumulation were carbon source- and growth phase-dependent, and were repressed by glucose. The highest intracellular glycogen content was observed in early log-phase cells grown on trehalose, which was followed by a drastic decrease of glycogen content prior to entering stationary phase. In raffinose-grown cells, however, glycogen accumulation gradually declined following early log phase and was maintained at stable levels throughout stationary phase. Raffinose also induced an overall higher temporal glg expression throughout growth compared with trehalose. Isogenic ΔglgA (glycogen synthase) and ΔglgB (glycogen-branching enzyme) mutants are glycogen-deficient and exhibited growth defects on raffinose. The latter observation suggests a reciprocal relationship between glycogen synthesis and raffinose metabolism. Deletion of glgB or glgP (glycogen phosphorylase) resulted in defective growth and increased bile sensitivity. The data indicate that glycogen metabolism is involved in growth maintenance, bile tolerance and complex carbohydrate utilization in L. acidophilus.


Asunto(s)
Vías Biosintéticas/genética , Regulación Bacteriana de la Expresión Génica , Glucógeno/biosíntesis , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/metabolismo , Operón , Enzima Ramificadora de 1,4-alfa-Glucano/genética , Enzima Ramificadora de 1,4-alfa-Glucano/metabolismo , Bilis/metabolismo , Carbono/metabolismo , Metabolismo Energético , Eliminación de Gen , Glucosa/metabolismo , Glucógeno Fosforilasa/genética , Glucógeno Fosforilasa/metabolismo , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Humanos , Lactobacillus acidophilus/crecimiento & desarrollo , Rafinosa/metabolismo , Trehalosa/metabolismo
16.
Microb Cell Fact ; 13: 94, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25410006

RESUMEN

In prokaryotic species equipped with glycogen metabolism machinery, the co-regulation of glycogen biosynthesis and degradation has been associated with the synthesis of energy storage compounds and various crucial physiological functions, including global cellular processes such as carbon and nitrogen metabolism, energy sensing and production, stress response and cell-cell communication. In addition, the glycogen metabolic pathway was proposed to serve as a carbon capacitor that regulates downstream carbon fluxes, and in some microorganisms the ability to synthesize intracellular glycogen has been implicated in host persistence. Among lactobacilli, complete glycogen metabolic pathway genes are present only in select species predominantly associated with mammalian hosts or natural environments. This observation highlights the potential involvement of glycogen biosynthesis in probiotic activities and persistence of intestinal lactobacilli in the human gastrointestinal tract. In this review, we summarize recent findings on (i) the presence and potential ecological distribution of glycogen metabolic pathways among lactobacilli, (ii) influence of carbon substrates and growth phases on glycogen metabolic gene expression and glycogen accumulation in L. acidophilus, and (iii) the involvement of glycogen metabolism on growth, sugar utilization and bile tolerance. Our present in vivo studies established the significance of glycogen biosynthesis on the competitive retention of L. acidophilus in the mouse intestinal tract, demonstrating for the first time that the ability to synthesize intracellular glycogen contributes to gut fitness and retention among probiotic microorganisms.


Asunto(s)
Adaptación Fisiológica , Metabolismo de los Hidratos de Carbono , Tracto Gastrointestinal/microbiología , Glucógeno/metabolismo , Lactobacillus acidophilus/metabolismo , Estrés Fisiológico , Adaptación Fisiológica/efectos de los fármacos , Animales , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Carbono/farmacología , Recuento de Colonia Microbiana , Tracto Gastrointestinal/efectos de los fármacos , Glucógeno/biosíntesis , Humanos , Lactobacillus acidophilus/efectos de los fármacos , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/crecimiento & desarrollo , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Ratones , Modelos Biológicos , Mutación/genética , Operón/genética , Células Procariotas/efectos de los fármacos , Células Procariotas/metabolismo , Rafinosa/farmacología , Especificidad de la Especie , Estrés Fisiológico/efectos de los fármacos , Trehalosa/farmacología
17.
Antonie Van Leeuwenhoek ; 106(1): 141-56, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24748373

RESUMEN

For thousands of years, humans have safely consumed microorganisms through fermented foods. Many of these bacteria are considered probiotics, which act through diverse mechanisms to confer a health benefit to the host. However, it was not until the availability of whole-genome sequencing and the era of genomics that mechanisms of probiotic efficacy could be discovered. In this review, we explore the history of the probiotic concept and the current standard of integrated genomic techniques to discern the complex, beneficial relationships between probiotic microbes and their hosts.


Asunto(s)
Bacterias/genética , Investigación Biomédica/historia , Probióticos/historia , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Cantaxantina/historia , Genómica/historia , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Probióticos/química
18.
Proc Natl Acad Sci U S A ; 108(43): 17785-90, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22006318

RESUMEN

Probiotic microbes rely on their ability to survive in the gastrointestinal tract, adhere to mucosal surfaces, and metabolize available energy sources from dietary compounds, including prebiotics. Genome sequencing projects have proposed models for understanding prebiotic catabolism, but mechanisms remain to be elucidated for many prebiotic substrates. Although ß-galactooligosaccharides (GOS) are documented prebiotic compounds, little is known about their utilization by lactobacilli. This study aimed to identify genetic loci in Lactobacillus acidophilus NCFM responsible for the transport and catabolism of GOS. Whole-genome oligonucleotide microarrays were used to survey the differential global transcriptome during logarithmic growth of L. acidophilus NCFM using GOS or glucose as a sole source of carbohydrate. Within the 16.6-kbp gal-lac gene cluster, lacS, a galactoside-pentose-hexuronide permease-encoding gene, was up-regulated 5.1-fold in the presence of GOS. In addition, two ß-galactosidases, LacA and LacLM, and enzymes in the Leloir pathway were also encoded by genes within this locus and up-regulated by GOS stimulation. Generation of a lacS-deficient mutant enabled phenotypic confirmation of the functional LacS permease not only for the utilization of lactose and GOS but also lactitol, suggesting a prominent role of LacS in the metabolism of a broad range of prebiotic ß-galactosides, known to selectively modulate the beneficial gut microbiota.


Asunto(s)
Regulación Bacteriana de la Expresión Génica/fisiología , Lactobacillus acidophilus/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Oligosacáridos/farmacocinética , Prebióticos , beta-Galactosidasa/metabolismo , ADN Complementario/genética , Lactobacillus acidophilus/genética , Análisis por Micromatrices , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , beta-Galactosidasa/genética
19.
Proc Natl Acad Sci U S A ; 108 Suppl 1: 4623-30, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282652

RESUMEN

Imbalance in the regulatory immune mechanisms that control intestinal cellular and bacterial homeostasis may lead to induction of the detrimental inflammatory signals characterized in humans as inflammatory bowel disease. Induction of proinflammatory cytokines (i.e., IL-12) induced by dendritic cells (DCs) expressing pattern recognition receptors may skew naive T cells to T helper 1 polarization, which is strongly implicated in mucosal autoimmunity. Recent studies show the ability of probiotic microbes to treat and prevent numerous intestinal disorders, including Clostridium difficile-induced colitis. To study the molecular mechanisms involved in the induction and repression of intestinal inflammation, the phosphoglycerol transferase gene that plays a key role in lipoteichoic acid (LTA) biosynthesis in Lactobacillus acidophilus NCFM (NCK56) was deleted. The data show that the L. acidophilus LTA-negative in LTA (NCK2025) not only down-regulated IL-12 and TNFα but also significantly enhanced IL-10 in DCs and controlled the regulation of costimulatory DC functions, resulting in their inability to induce CD4(+) T-cell activation. Moreover, treatment of mice with NCK2025 compared with NCK56 significantly mitigated dextran sulfate sodium and CD4(+)CD45RB(high)T cell-induced colitis and effectively ameliorated dextran sulfate sodium-established colitis through a mechanism that involves IL-10 and CD4(+)FoxP3(+) T regulatory cells to dampen exaggerated mucosal inflammation. Directed alteration of cell surface components of L. acidophilus NCFM establishes a potential strategy for the treatment of inflammatory intestinal disorders.


Asunto(s)
Autoinmunidad/inmunología , Colitis/inmunología , Colitis/microbiología , Regulación de la Expresión Génica/inmunología , Lactobacillus acidophilus/metabolismo , Lipopolisacáridos/deficiencia , Animales , Linfocitos T CD4-Positivos/inmunología , Colitis/inducido químicamente , Cartilla de ADN/genética , Sulfato de Dextran/toxicidad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Proteínas de Homeodominio/genética , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Reacción en Cadena de la Polimerasa , Ácidos Teicoicos , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética
20.
BMC Genomics ; 14: 312, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23663691

RESUMEN

BACKGROUND: Probiotic bifidobacteria in combination with prebiotic carbohydrates have documented positive effects on human health regarding gastrointestinal disorders and improved immunity, however the selective routes of uptake remain unknown for most candidate prebiotics. The differential transcriptomes of Bifidobacterium animalis subsp. lactis Bl-04, induced by 11 potential prebiotic oligosaccharides were analyzed to identify the genetic loci involved in the uptake and catabolism of α- and ß-linked hexoses, and ß-xylosides. RESULTS: The overall transcriptome was modulated dependent on the type of glycoside (galactosides, glucosides or xylosides) utilized. Carbohydrate transporters of the major facilitator superfamily (induced by gentiobiose and ß-galacto-oligosaccharides (GOS)) and ATP-binding cassette (ABC) transporters (upregulated by cellobiose, GOS, isomaltose, maltotriose, melibiose, panose, raffinose, stachyose, xylobiose and ß-xylo-oligosaccharides) were differentially upregulated, together with glycoside hydrolases from families 1, 2, 13, 36, 42, 43 and 77. Sequence analysis of the identified solute-binding proteins that determine the specificity of ABC transporters revealed similarities in the breadth and selectivity of prebiotic utilization by bifidobacteria. CONCLUSION: This study identified the differential gene expression for utilization of potential prebiotics highlighting the extensive capabilities of Bifidobacterium lactis Bl-04 to utilize oligosaccharides. Results provide insights into the ability of this probiotic microbe to utilize indigestible carbohydrates in the human gastrointestinal tract.


Asunto(s)
Bifidobacterium/efectos de los fármacos , Bifidobacterium/genética , Perfilación de la Expresión Génica , Oligosacáridos/farmacología , Prebióticos , Transcripción Genética/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Minería de Datos , Variación Genética/genética , Genómica , Familia de Multigenes/genética , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA