Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Microbiol ; 111(1): 287-301, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30352131

RESUMEN

Development of the human gut microbiota commences at birth, with certain bifidobacterial species representing dominant and early colonisers of the newborn gastrointestinal tract. The molecular basis of Bifidobacterium colonisation, persistence and presumed communication with the host has remained obscure. We previously identified tight adherence (Tad) pili from Bifidobacterium breve UCC2003 as an essential colonisation factor. Here, we demonstrate that bifidobacterial Tad pili also promote in vivo colonic epithelial proliferation. A significant increase in cell proliferation was detectable 5 days postadministration of B. breve UCC2003. Using advanced functional genomic approaches, bacterial strains either (a) producing the Tad2003 pili or (b) lacking the TadE or TadF pseudopilins were created. Analysis of the ability of these mutant strains to promote epithelial cell proliferation in vivo demonstrated that the pilin subunit, TadE, is the bifidobacterial molecule responsible for this proliferation response. These findings were confirmed in vitro using purified TadE protein. Our data imply that bifidobacterial Tad pili may contribute to the maturation of the naïve gut in early life through the production of a specific scaffold of extracellular protein structures, which stimulate growth of the neonatal mucosa.


Asunto(s)
Bifidobacterium breve/fisiología , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/fisiología , Mucosa Intestinal/microbiología , Bifidobacterium breve/genética , Línea Celular , Proteínas Fimbrias/genética , Eliminación de Gen , Humanos
2.
BMC Genomics ; 19(1): 33, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29310579

RESUMEN

BACKGROUND: Bifidobacterium longum is a common member of the human gut microbiota and is frequently present at high numbers in the gut microbiota of humans throughout life, thus indicative of a close symbiotic host-microbe relationship. Different mechanisms may be responsible for the high competitiveness of this taxon in its human host to allow stable establishment in the complex and dynamic intestinal microbiota environment. The objective of this study was to assess the genetic and metabolic diversity in a set of 20 B. longum strains, most of which had previously been isolated from infants, by performing whole genome sequencing and comparative analysis, and to analyse their carbohydrate utilization abilities using a gene-trait matching approach. RESULTS: We analysed their pan-genome and their phylogenetic relatedness. All strains clustered in the B. longum ssp. longum phylogenetic subgroup, except for one individual strain which was found to cluster in the B. longum ssp. suis phylogenetic group. The examined strains exhibit genomic diversity, while they also varied in their sugar utilization profiles. This allowed us to perform a gene-trait matching exercise enabling the identification of five gene clusters involved in the utilization of xylo-oligosaccharides, arabinan, arabinoxylan, galactan and fucosyllactose, the latter of which is an abundant human milk oligosaccharide (HMO). CONCLUSIONS: The results showed high diversity in terms of genes and predicted glycosyl-hydrolases, as well as the ability to metabolize a large range of sugars. Moreover, we corroborate the capability of B. longum ssp. longum to metabolise HMOs. Ultimately, their intraspecific genomic diversity and the ability to consume a wide assortment of carbohydrates, ranging from plant-derived carbohydrates to HMOs, may provide an explanation for the competitive advantage and persistence of B. longum in the human gut microbiome.


Asunto(s)
Bifidobacterium longum/genética , Bifidobacterium longum/metabolismo , Metabolismo de los Hidratos de Carbono , Genes Bacterianos , Genoma Bacteriano , Carácter Cuantitativo Heredable , Biodiversidad , Bases de Datos Genéticas , Microbioma Gastrointestinal , Humanos , Lactante , Recién Nacido , Filogenia , Probióticos , Sitios de Carácter Cuantitativo
3.
Appl Environ Microbiol ; 84(9)2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29500268

RESUMEN

Bifidobacterial carbohydrate metabolism has been studied in considerable detail for a variety of both plant- and human-derived glycans, particularly involving the bifidobacterial prototype strain Bifidobacterium breve UCC2003. We recently elucidated the metabolic pathways by which the human milk oligosaccharide (HMO) constituents lacto-N-tetraose (LNT), lacto-N-neotetraose (LNnT) and lacto-N-biose (LNB) are utilized by B. breve UCC2003. However, to date, no work has been carried out on the regulatory mechanisms that control the expression of the genetic loci involved in these HMO metabolic pathways. In this study, we describe the characterization of three transcriptional regulators and the corresponding operator and associated (inducible) promoter sequences, with the latter governing the transcription of the genetic elements involved in LN(n)T/LNB metabolism. The activity of these regulators is dependent on the release of specific monosaccharides, which are believed to act as allosteric effectors and which are derived from the corresponding HMOs targeted by the particular locus.IMPORTANCE Human milk oligosaccharides (HMOs) are a key factor in the development of the breastfed-infant microbiota. They function as prebiotics, selecting for a specific range of microbes, including a number of infant-associated species of bifidobacteria, which are thought to provide a range of health benefits to the infant host. While much research has been carried out on elucidating the mechanisms of HMO metabolism in infant-associated bifidobacteria, to date there is very little understanding of the transcriptional regulation of these pathways. This study reveals a multicomponent transcriptional regulation system that controls the recently identified pathways of HMO metabolism in the infant-associated Bifidobacterium breve prototype strain UCC2003. This not only provides insight into the regulatory mechanisms present in other infant-associated bifidobacteria but also provides an example of a network of sequential steps regulating microbial carbohydrate metabolism.


Asunto(s)
Bifidobacterium breve/genética , Regulación Bacteriana de la Expresión Génica , Leche Humana/microbiología , Oligosacáridos/metabolismo , Elementos Reguladores de la Transcripción/genética , Lactancia Materna , Humanos , Lactante , Recién Nacido , Redes y Vías Metabólicas , Microbiota
4.
Appl Microbiol Biotechnol ; 102(24): 10645-10663, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30306201

RESUMEN

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.


Asunto(s)
Bifidobacterium animalis/crecimiento & desarrollo , Bifidobacterium animalis/genética , Microbiología de Alimentos/métodos , Leche/microbiología , Animales , Bifidobacterium animalis/efectos de los fármacos , Metabolismo de los Hidratos de Carbono , Farmacorresistencia Microbiana , Femenino , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Ratones Endogámicos BALB C , Probióticos
5.
Proc Natl Acad Sci U S A ; 112(16): E1984-93, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25848054

RESUMEN

Escherichia coli and yeast DNA-dependent RNA polymerases are shown to mediate efficient nascent transcript stem loop formation-dependent RNA-DNA hybrid realignment. The realignment was discovered on the heteropolymeric sequence T5C5 and yields transcripts lacking a C residue within a corresponding U5C4. The sequence studied is derived from a Roseiflexus insertion sequence (IS) element where the resulting transcriptional slippage is required for transposase synthesis. The stability of the RNA structure, the proximity of the stem loop to the slippage site, the length and composition of the slippage site motif, and the identity of its 3' adjacent nucleotides (nt) are crucial for transcripts lacking a single C. In many respects, the RNA structure requirements for this slippage resemble those for hairpin-dependent transcription termination. In a purified in vitro system, the slippage efficiency ranges from 5% to 75% depending on the concentration ratios of the nucleotides specified by the slippage sequence and the 3' nt context. The only previous proposal of stem loop mediated slippage, which was in Ebola virus expression, was based on incorrect data interpretation. We propose a mechanical slippage model involving the RNAP translocation state as the main motor in slippage directionality and efficiency. It is distinct from previously described models, including the one proposed for paramyxovirus, where following random movement efficiency is mainly dependent on the stability of the new realigned hybrid. In broadening the scope for utilization of transcription slippage for gene expression, the stimulatory structure provides parallels with programmed ribosomal frameshifting at the translation level.


Asunto(s)
Conformación de Ácido Nucleico , ARN Mensajero/química , Regiones Terminadoras Genéticas , Transcripción Genética , Secuencia de Aminoácidos , Secuencia de Bases , Chloroflexi/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Datos de Secuencia Molecular , Motivos de Nucleótidos/genética , ARN Mensajero/genética , Saccharomyces cerevisiae/genética , Inversión de Secuencia
6.
Appl Environ Microbiol ; 82(22): 6611-6623, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27590817

RESUMEN

Bifidobacteria constitute a specific group of commensal bacteria typically found in the gastrointestinal tract (GIT) of humans and other mammals. Bifidobacterium breve strains are numerically prevalent among the gut microbiota of many healthy breastfed infants. In the present study, we investigated glycosulfatase activity in a bacterial isolate from a nursling stool sample, B. breve UCC2003. Two putative sulfatases were identified on the genome of B. breve UCC2003. The sulfated monosaccharide N-acetylglucosamine-6-sulfate (GlcNAc-6-S) was shown to support the growth of B. breve UCC2003, while N-acetylglucosamine-3-sulfate, N-acetylgalactosamine-3-sulfate, and N-acetylgalactosamine-6-sulfate did not support appreciable growth. By using a combination of transcriptomic and functional genomic approaches, a gene cluster designated ats2 was shown to be specifically required for GlcNAc-6-S metabolism. Transcription of the ats2 cluster is regulated by a repressor open reading frame kinase (ROK) family transcriptional repressor. This study represents the first description of glycosulfatase activity within the Bifidobacterium genus. IMPORTANCE: Bifidobacteria are saccharolytic organisms naturally found in the digestive tract of mammals and insects. Bifidobacterium breve strains utilize a variety of plant- and host-derived carbohydrates that allow them to be present as prominent members of the infant gut microbiota as well as being present in the gastrointestinal tract of adults. In this study, we introduce a previously unexplored area of carbohydrate metabolism in bifidobacteria, namely, the metabolism of sulfated carbohydrates. B. breve UCC2003 was shown to metabolize N-acetylglucosamine-6-sulfate (GlcNAc-6-S) through one of two sulfatase-encoding gene clusters identified on its genome. GlcNAc-6-S can be found in terminal or branched positions of mucin oligosaccharides, the glycoprotein component of the mucous layer that covers the digestive tract. The results of this study provide further evidence of the ability of this species to utilize mucin-derived sugars, a trait which may provide a competitive advantage in both the infant gut and adult gut.


Asunto(s)
Bifidobacterium breve/genética , Heces/microbiología , Genes Bacterianos , Familia de Multigenes , Sulfatasas/genética , Acetilglucosamina/análogos & derivados , Acetilglucosamina/metabolismo , Bifidobacterium breve/enzimología , Bifidobacterium breve/crecimiento & desarrollo , Bifidobacterium breve/metabolismo , Lactancia Materna , ADN Bacteriano/genética , Tracto Gastrointestinal/microbiología , Perfilación de la Expresión Génica , Genoma Bacteriano , Genómica/métodos , Humanos , Lactante , Oligosacáridos/metabolismo , Sulfatasas/clasificación , Sulfatasas/aislamiento & purificación
7.
Appl Environ Microbiol ; 82(24): 7185-7196, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27736791

RESUMEN

The immune-modulating properties of certain bifidobacterial strains, such as Bifidobacterium longum subsp. longum 35624 (B. longum 35624), have been well described, although the strain-specific molecular characteristics associated with such immune-regulatory activity are not well defined. It has previously been demonstrated that B. longum 35624 produces a cell surface exopolysaccharide (sEPS), and in this study, we investigated the role played by this exopolysaccharide in influencing the host immune response. B. longum 35624 induced relatively low levels of cytokine secretion from human dendritic cells, whereas an isogenic exopolysaccharide-negative mutant derivative (termed sEPSneg) induced vastly more cytokines, including interleukin-17 (IL-17), and this response was reversed when exopolysaccharide production was restored in sEPSneg by genetic complementation. Administration of B. longum 35624 to mice of the T cell transfer colitis model prevented disease symptoms, whereas sEPSneg did not protect against the development of colitis, with associated enhanced recruitment of IL-17+ lymphocytes to the gut. Moreover, intranasal administration of sEPSneg also resulted in enhanced recruitment of IL-17+ lymphocytes to the murine lung. These data demonstrate that the particular exopolysaccharide produced by B. longum 35624 plays an essential role in dampening proinflammatory host responses to the strain and that loss of exopolysaccharide production results in the induction of local TH17 responses. IMPORTANCE: Particular gut commensals, such as B. longum 35624, are known to contribute positively to the development of mucosal immune cells, resulting in protection from inflammatory diseases. However, the molecular basis and mechanisms for these commensal-host interactions are poorly described. In this report, an exopolysaccharide was shown to be decisive in influencing the immune response to the bacterium. We generated an isogenic mutant unable to produce exopolysaccharide and observed that this mutation caused a dramatic change in the response of human immune cells in vitro In addition, the use of mouse models confirmed that lack of exopolysaccharide production induces inflammatory responses to the bacterium. These results implicate the surface-associated exopolysaccharide of the B. longum 35624 cell envelope in the prevention of aberrant inflammatory responses.


Asunto(s)
Infecciones por Bifidobacteriales/inmunología , Bifidobacterium longum/inmunología , Polisacáridos Bacterianos/inmunología , Células Th17/inmunología , Animales , Infecciones por Bifidobacteriales/microbiología , Bifidobacterium longum/genética , Citocinas/inmunología , Femenino , Humanos , Interleucina-17/inmunología , Ratones , Ratones Endogámicos BALB C
8.
Microb Cell Fact ; 15: 72, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27142164

RESUMEN

BACKGROUND: Among the oligosaccharides that may positively affect the gut microbiota, xylo-oligosaccharides (XOS) and arabinoxylan oligosaccharides (AXOS) possess promising functional properties. Ingestion of XOS has been reported to contribute to anti-oxidant, anti-bacterial, immune-modulatory and anti-diabetic activities. Because of the structural complexity and chemical heterogeneity, complete degradation of xylan-containing plant polymers requires the synergistic activity of several enzymes. Endo-xylanases and ß-D-xylosidases, collectively termed xylanases, represent the two key enzymes responsible for the sequential hydrolysis of xylan. Xylanase cocktails are used on an industrial scale for biotechnological purposes. Lactobacillus rossiae DSM 15814(T) can utilize an extensive set of carbon sources, an ability that is likely to contribute to its adaptive ability. In this study, the capacity of this strain to utilize XOS, xylan, D-xylose and L-arabinose was investigated. RESULTS: Genomic and transcriptomic analyses revealed the presence of two gene clusters, designated xyl and ara, encoding proteins predicted to be responsible for XOS uptake and hydrolysis and D-xylose utilization, and L-arabinose metabolism, respectively. The deduced amino acid sequence of one of the genes of the xyl gene cluster, LROS_1108 (designated here as xylA), shows high similarity to (predicted) ß-D-xylosidases encoded by various lactic acid bacteria, and belongs to glycosyl hydrolase family 43. Heterologously expressed XylA was shown to completely hydrolyse XOS to xylose and showed optimal activity at pH 6.0 and 40 °C. Furthermore, ß-D-xylosidase activity of L. rossiae DSM 15814(T) was also measured under sourdough conditions. CONCLUSIONS: This study highlights the ability of L. rossiae DSM 15814(T) to utilize XOS, which is a very useful trait when selecting starters with specific metabolic performances for sourdough fermentation or as probiotics.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Lactobacillus/enzimología , Lactobacillus/genética , Xilosidasas/genética , Xilosidasas/metabolismo , Arabinosa/metabolismo , Clonación Molecular , Concentración de Iones de Hidrógeno , Hidrólisis , Lactobacillus/clasificación , Familia de Multigenes , Oligosacáridos/metabolismo , Filogenia , Plásmidos/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Temperatura , Xilosa/metabolismo , Xilosidasas/química
9.
Proc Natl Acad Sci U S A ; 110(27): 11151-6, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23776216

RESUMEN

Bifidobacteria represent one of the dominant groups of microorganisms colonizing the human infant intestine. Commensal bacteria that interact with a eukaryotic host are believed to express adhesive molecules on their cell surface that bind to specific host cell receptors or soluble macromolecules. Whole-genome transcription profiling of Bifidobacterium bifidum PRL2010, a strain isolated from infant stool, revealed a small number of commonly expressed extracellular proteins, among which were genes that specify sortase-dependent pili. Expression of the coding sequences of these B. bifidum PRL2010 appendages in nonpiliated Lactococcus lactis enhanced adherence to human enterocytes through extracellular matrix protein and bacterial aggregation. Furthermore, such piliated L. lactis cells evoked a higher TNF-α response during murine colonization compared with their nonpiliated parent, suggesting that bifidobacterial sortase-dependent pili not only contribute to adherence but also display immunomodulatory activity.


Asunto(s)
Bifidobacterium/fisiología , Fimbrias Bacterianas/fisiología , Aminoaciltransferasas/genética , Aminoaciltransferasas/metabolismo , Animales , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Bifidobacterium/genética , Bifidobacterium/inmunología , Línea Celular , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Citocinas/biosíntesis , Femenino , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/inmunología , Genes Bacterianos , Humanos , Lactante , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Lactococcus lactis/genética , Lactococcus lactis/fisiología , Ratones , Ratones Endogámicos BALB C , Probióticos , Transcriptoma/inmunología
10.
Appl Environ Microbiol ; 81(1): 166-76, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25326305

RESUMEN

Bifidobacterium breve is a common and sometimes very abundant inhabitant of the human gut. Genome sequencing of B. breve JCM 7017 revealed the presence of an extrachromosomal element, designated pMP7017 consisting of >190 kb, thus representing the first reported bifidobacterial megaplasmid. In silico characterization of this element revealed several genomic features supporting a stable establishment of the megaplasmid in its host, illustrated by predicted CRISPR-Cas functions that are known to protect the host against intrusion of foreign DNA. Interestingly, pMP7017 is also predicted to encode a conjugative DNA transfer apparatus and, consistent with this notion, we demonstrate here the conjugal transfer of pMP7017 to representative strains of B. breve and B. longum subsp. longum. We also demonstrate the presence of a megaplasmid with homology to pMP7017 in three B. longum subsp. longum strains.


Asunto(s)
Bifidobacterium/genética , Plásmidos , Sistemas CRISPR-Cas , Conjugación Genética , ADN Bacteriano/química , ADN Bacteriano/genética , Genes Bacterianos , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
11.
BMC Genomics ; 15: 831, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25269955

RESUMEN

BACKGROUND: So-called 936-type phages are among the most frequently isolated phages in dairy facilities utilising Lactococcus lactis starter cultures. Despite extensive efforts to control phage proliferation and decades of research, these phages continue to negatively impact cheese production in terms of the final product quality and consequently, monetary return. RESULTS: Whole genome sequencing and in silico analysis of three 936-type phage genomes identified several putative (orphan) methyltransferase (MTase)-encoding genes located within the packaging and replication regions of the genome. Utilising SMRT sequencing, methylome analysis was performed on all three phages, allowing the identification of adenine modifications consistent with N-6 methyladenine sequence methylation, which in some cases could be attributed to these phage-encoded MTases. Heterologous gene expression revealed that M.Phi145I/M.Phi93I and M.Phi93DAM, encoded by genes located within the packaging module, provide protection against the restriction enzymes HphI and DpnII, respectively, representing the first functional MTases identified in members of 936-type phages. CONCLUSIONS: SMRT sequencing technology enabled the identification of the target motifs of MTases encoded by the genomes of three lytic 936-type phages and these MTases represent the first functional MTases identified in this species of phage. The presence of these MTase-encoding genes on 936-type phage genomes is assumed to represent an adaptive response to circumvent host encoded restriction-modification systems thereby increasing the fitness of the phages in a dynamic dairy environment.


Asunto(s)
Bacteriófagos/enzimología , Bacteriófagos/fisiología , Enzimas de Restricción del ADN/metabolismo , Genómica , Lactococcus lactis/virología , Metiltransferasas/metabolismo , Secuencia de Aminoácidos , Bacteriófagos/genética , Queso/microbiología , Metilación de ADN , Genoma Viral/genética , Metiltransferasas/química , Metiltransferasas/genética , Datos de Secuencia Molecular
12.
BMC Genomics ; 15: 170, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24581150

RESUMEN

BACKGROUND: Bifidobacteria are commonly found as part of the microbiota of the gastrointestinal tract (GIT) of a broad range of hosts, where their presence is positively correlated with the host's health status. In this study, we assessed the genomes of thirteen representatives of Bifidobacterium breve, which is not only a frequently encountered component of the (adult and infant) human gut microbiota, but can also be isolated from human milk and vagina. RESULTS: In silico analysis of genome sequences from thirteen B. breve strains isolated from different environments (infant and adult faeces, human milk, human vagina) shows that the genetic variability of this species principally consists of hypothetical genes and mobile elements, but, interestingly, also genes correlated with the adaptation to host environment and gut colonization. These latter genes specify the biosynthetic machinery for sortase-dependent pili and exopolysaccharide production, as well as genes that provide protection against invasion of foreign DNA (i.e. CRISPR loci and restriction/modification systems), and genes that encode enzymes responsible for carbohydrate fermentation. Gene-trait matching analysis showed clear correlations between known metabolic capabilities and characterized genes, and it also allowed the identification of a gene cluster involved in the utilization of the alcohol-sugar sorbitol. CONCLUSIONS: Genome analysis of thirteen representatives of the B. breve species revealed that the deduced pan-genome exhibits an essentially close trend. For this reason our analyses suggest that this number of B. breve representatives is sufficient to fully describe the pan-genome of this species. Comparative genomics also facilitated the genetic explanation for differential carbon source utilization phenotypes previously observed in different strains of B. breve.


Asunto(s)
Bifidobacterium/genética , Genoma Bacteriano , Genómica , Bifidobacterium/clasificación , Bifidobacterium/metabolismo , Metabolismo de los Hidratos de Carbono , Análisis por Conglomerados , Biología Computacional , Elementos Transponibles de ADN , Orden Génico , Estudios de Asociación Genética , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Metaboloma , Metabolómica , Datos de Secuencia Molecular , Familia de Multigenes , Filogenia
13.
Appl Environ Microbiol ; 80(14): 4414-26, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24814790

RESUMEN

Bifidobacteria constitute a specific group of commensal bacteria that inhabit the gastrointestinal tracts of humans and other mammals. Bifidobacterium breve UCC2003 has previously been shown to utilize several plant-derived carbohydrates that include cellodextrins, starch, and galactan. In the present study, we investigated the ability of this strain to utilize the mucin- and human milk oligosaccharide (HMO)-derived carbohydrate sialic acid. Using a combination of transcriptomic and functional genomic approaches, we identified a gene cluster dedicated to the uptake and metabolism of sialic acid. Furthermore, we demonstrate that B. breve UCC2003 can cross feed on sialic acid derived from the metabolism of 3'-sialyllactose, an abundant HMO, by another infant gut bifidobacterial strain, Bifidobacterium bifidum PRL2010.


Asunto(s)
Bifidobacterium/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Bifidobacterium/genética , Medios de Cultivo , Fragmentación del ADN , ADN Bacteriano/genética , Familia de Multigenes , Análisis de Secuencia por Matrices de Oligonucleótidos , Plásmidos/genética , Análisis de Secuencia de ADN
14.
Appl Environ Microbiol ; 80(18): 5623-35, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25002431

RESUMEN

In this study, three phages infecting Lactobacillus delbrueckii subsp. bulgaricus, named Ld3, Ld17, and Ld25A, were isolated from whey samples obtained from various industrial fermentations. These phages were further characterized in a multifaceted approach: (i) biological and physical characterization through host range analysis and electron microscopy; (ii) genetic assessment through genome analysis; (iii) mass spectrometry analysis of the structural components of the phages; and (iv), for one phage, transcriptional analysis by Northern hybridization, reverse transcription-PCR, and primer extension. The three obtained phage genomes display high levels of sequence identity to each other and to genomes of the so-called group b L. delbrueckii phages c5, LL-Ku, and phiLdb, where some of the observed differences are believed to be responsible for host range variations.


Asunto(s)
Bacteriófagos/aislamiento & purificación , ADN Viral/genética , Especificidad del Huésped , Lactobacillus delbrueckii/virología , Proteínas Virales/análisis , Virión/ultraestructura , Bacteriófagos/química , Bacteriófagos/genética , Bacteriófagos/fisiología , Microbiología de Alimentos , Perfilación de la Expresión Génica , Orden Génico , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Sintenía
15.
Microb Cell Fact ; 13 Suppl 1: S4, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25186128

RESUMEN

The human gastrointestinal tract represents an environment which is a densely populated home for a microbiota that has evolved to positively contribute to host health. At birth the essentially sterile gastrointestinal tract (GIT) is rapidly colonized by microorganisms that originate from the mother and the surrounding environment. Within a short timeframe a microbiota establishes within the (breastfed) infant's GIT where bifidobacteria are among the dominant members, although their numerical dominance disappears following weaning. The numerous health benefits associated with bifidobacteria, and the consequent commercial relevance resulting from their incorporation into functional foods, has led to intensified research aimed at the molecular understanding of claimed probiotic attributes of this genus. In this review we provide the current status on the diversity and ecology of bifidobacteria. In addition, we will discuss the molecular mechanisms that allow this intriguing group of bacteria to colonize and persist in the GIT, so as to facilitate interaction with its host.


Asunto(s)
Bifidobacterium/metabolismo , Bifidobacterium/clasificación , Bifidobacterium/genética , Biodiversidad , Metabolismo de los Hidratos de Carbono , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Hibridación Genómica Comparativa , Tracto Gastrointestinal/microbiología , Genoma Bacteriano , Humanos , Polisacáridos Bacterianos/metabolismo , ARN Ribosómico 16S/genética
16.
Proc Natl Acad Sci U S A ; 108(27): 11217-22, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21690406

RESUMEN

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.


Asunto(s)
Bifidobacterium/genética , Bifidobacterium/fisiología , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/fisiología , Genoma Bacteriano , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Secuencia de Bases , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/ultraestructura , Hibridación Genómica Comparativa , ADN Bacteriano/genética , Femenino , Fimbrias Bacterianas/ultraestructura , Tracto Gastrointestinal/microbiología , Regulación Bacteriana de la Expresión Génica , Vida Libre de Gérmenes , Humanos , Masculino , Metagenoma , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Familia de Multigenes , Mutación , Homología de Secuencia de Aminoácido
17.
Microb Biotechnol ; 17(1): e14405, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38206097

RESUMEN

The 190 kb megaplasmid pMP7017 of Bifidobacterium breve JCM7017 represents the first conjugative and largest plasmid characterised within this genus to date. In the current study, we adopted an integrated approach combining transcriptomics, whole genome comparative analysis and metagenomic data mining to understand the biology of pMP7017 and related megaplasmids, and to assess the impact of plasmid-carriage on the host strain. The data generated revealed variations within basic features of promoter elements which correlate with a high level of transcription on the plasmid and highlight the transcriptional activity of genes encoding both offensive and defensive adaptations, including a Type IIL restriction-modification system, an anti-restriction system and four Type II toxin-antitoxin systems. Furthermore, a highly transcribed tmRNA, which likely provides translational support to the host strain, was identified, making pMP7017 the first plasmid of the Bifidobacterium genus and the smallest plasmid known to express a tmRNA. Analyses of synteny and variability among pMP7017 and related plasmids indicate substantial diversity in gene organisation and accessory gene cargo highlighting diverse (co-)evolution and potential host-specific rearrangements and adaptations. Systematic analysis of the codon usage profile of transcriptionally active pMP7017-encoded genes suggests that pMP7017 originated from (sub)species of Bifidobacterium longum. Furthermore, mining of metagenomic data suggests the presence of pMP7017-homologues in ~10% of microbiome samples, mostly infants and/or mothers from various geographical locations. Comparative transcriptome analysis of the B. breve UCC2003 chromosome in the presence or absence of pMP7017 revealed differential expression of genes representing 8% of the total gene pool. Genes involved in genetic information processing were exclusively upregulated, while altered expression of genes involved in biofilm production and polysaccharide biosynthesis was also observed.


Asunto(s)
Bifidobacterium breve , Humanos , Bifidobacterium breve/genética , Bifidobacterium breve/metabolismo , Transcriptoma , Bifidobacterium/genética , Plásmidos/genética , Perfilación de la Expresión Génica
18.
Appl Environ Microbiol ; 79(20): 6280-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23913435

RESUMEN

Members of the genus Bifidobacterium are common inhabitants of the gastrointestinal tracts of humans and other mammals, where they ferment many diet-derived carbohydrates that cannot be digested by their hosts. To extend our understanding of bifidobacterial carbohydrate utilization, we investigated the molecular mechanisms by which 11 strains of Bifidobacterium breve metabolize four distinct α-glucose- and/or α-galactose-containing oligosaccharides, namely, raffinose, stachyose, melibiose, and melezitose. Here we demonstrate that all B. breve strains examined possess the ability to utilize raffinose, stachyose, and melibiose. However, the ability to metabolize melezitose was not common to all B. breve strains tested. Transcriptomic and functional genomic approaches identified a gene cluster dedicated to the metabolism of α-galactose-containing carbohydrates, while an adjacent gene cluster, dedicated to the metabolism of α-glucose-containing melezitose, was identified in strains that are able to use this carbohydrate.


Asunto(s)
Bifidobacterium/metabolismo , Melibiosa/metabolismo , Oligosacáridos/metabolismo , Rafinosa/metabolismo , Bifidobacterium/aislamiento & purificación , Perfilación de la Expresión Génica , Humanos , Redes y Vías Metabólicas/genética , Familia de Multigenes
19.
Methods Mol Biol ; 2642: 403-427, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36944891

RESUMEN

Research to date on abiotic stress responses in plants has been largely focused on the plant itself, but current knowledge indicates that microorganisms can interact with and help plants during periods of abiotic stress. In our research, we aim to investigate the interkingdom communication between the plant root and the rhizo-microbiota. Our investigation showed that miRNA plays a pivotal role in this interkingdom communication. Here, we describe a protocol for the analysis of miRNA secreted by the plant root, which includes all of the steps from the isolation of the miRNA to the bioinformatics analysis. Because of their short nucleotide length, Next Generation Sequencing (NGS) library preparation from miRNAs can be challenging due to the presence of dimer adapter contaminants. Therefore, we highlight some strategies we adopt to inhibit the generation of dimer adapters during library preparation. Current screens of miRNA targets mostly focus on the identification of targets present in the same organism expressing the miRNA. Our bioinformatics analysis challenges the barrier of evolutionary divergent organisms to identify candidate sequences of the microbiota targeted by the miRNA of plant roots. This protocol should be of interest to researchers investigating interkingdom RNA-based communication between plants and their associated microorganisms, particularly in the context of holobiont responses to abiotic stresses.


Asunto(s)
MicroARNs , MicroARNs/genética , Biblioteca de Genes , Plantas/genética , Programas Informáticos , Estrés Fisiológico/genética , Regulación de la Expresión Génica de las Plantas , ARN de Planta/genética
20.
Appl Environ Microbiol ; 78(4): 1123-31, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22156415

RESUMEN

Tolerance of gut commensals to bile salt exposure is an important feature for their survival in and colonization of the intestinal environment. A transcriptomic approach was employed to study the response of Bifidobacterium breve UCC2003 to bile, allowing the identification of a number of bile-induced genes with a range of predicted functions. The potential roles of a selection of these bile-inducible genes in bile protection were analyzed following heterologous expression in Lactococcus lactis. Genes encoding three transport systems belonging to the major facilitator superfamily (MFS), Bbr_0838, Bbr_0832, and Bbr_1756, and three ABC-type transporters, Bbr_0406-0407, Bbr_1804-1805, and Bbr_1826-1827, were thus investigated and shown to provide enhanced resistance and survival to bile exposure. This work significantly improves our understanding as to how bifidobacteria respond to and survive bile exposure.


Asunto(s)
Antibacterianos/metabolismo , Bifidobacterium/genética , Bifidobacterium/metabolismo , Ácidos y Sales Biliares/metabolismo , Clonación Molecular , Expresión Génica , Genómica/métodos , Lactococcus lactis/genética , Viabilidad Microbiana/efectos de los fármacos , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA