Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 13(1): 1-24, 2021.
Article in English | MEDLINE | ID: mdl-33960282

ABSTRACT

Emerging evidence indicates maternal microbiota as one major reservoir for pioneering microbes in infants. However, the global distinct and identical features of mother-infant gut microbiota at various taxonomic resolutions and metabolic functions across cohorts and potential of infant microbial prediction based on their paired mother's gut microbiota remain unclear. Here, we analyzed 376 mother-infant dyads (468 mother and 1024 infant samples) of eight studies from six countries and observed higher diversity at species and strain levels in maternal gut microbiota but not their metabolic functions. A number of 290 species were shared in at least one mother-infant dyad, with 26 species (five at strain level) observed across cohorts. The profile of mother-infant shared species and strains was further influenced by delivery mode and feeding regimen. The mother-sourced species in infants exhibited similar strain heterogeneity but more metabolic functions compared to other-sourced species, suggesting the comparable stability and fitness of shared and non-shared species and the potential role of shared species in the early gut microbial community, respectively. Predictive models showed moderate performance accuracy for shared species and strains occurrences in infants. These generalized mother-infant shared species and strains may be considered as the primary targets for future work toward infant microbiome development and probiotics exploration.


Subject(s)
Bacteria/isolation & purification , Gastrointestinal Microbiome , Adult , Bacteria/classification , Bacteria/genetics , Feces/microbiology , Female , Genome, Bacterial , Humans , Infant , Male , Metagenome , Metagenomics , Mothers , Phylogeny
2.
Gut Microbes ; 13(1): 1-17, 2021.
Article in English | MEDLINE | ID: mdl-33406976

ABSTRACT

The human gut microbiome is a diverse and complex ecosystem that plays a critical role in health and disease. The composition of the gut microbiome has been well studied across all stages of life. In recent years, studies have investigated the production of endospores by specific members of the gut microbiome. An endospore is a tough, dormant structure formed by members of the Firmicutes phylum, which allows for greater resistance to otherwise inhospitable conditions. This innate resistance has consequences for human health and disease, as well as in biotechnology. In particular, the formation of endospores is strongly linked to antibiotic resistance and the spread of antibiotic resistance genes, also known as the resistome. The term sporobiota has been used to define the spore-forming cohort of a microbial community. In this review, we present an overview of the current knowledge of the sporobiota in the human gut. We discuss the development of the sporobiota in the infant gut and the perinatal factors that may have an effect on vertical transmission from mother to infant. Finally, we examine the sporobiota of critically important food sources for the developing infant, breast milk and powdered infant formula.


Subject(s)
Endospore-Forming Bacteria/physiology , Gastrointestinal Microbiome , Adult , Drug Resistance, Bacterial , Endospore-Forming Bacteria/classification , Endospore-Forming Bacteria/isolation & purification , Humans , Infant , Infant Formula/microbiology , Infectious Disease Transmission, Vertical , Milk, Human/microbiology , Spores, Bacterial/classification , Spores, Bacterial/isolation & purification , Spores, Bacterial/physiology
3.
Sci Rep ; 10(1): 17265, 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33037276

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Microorganisms ; 8(9)2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32872165

ABSTRACT

Bifidobacteria are among the first colonisers of the gastrointestinal tract of breast-fed newborns due to, among other things, their ability to metabolise oligosaccharides naturally occurring in human milk. The presence of bifidobacteria in the infant gut has been shown to promote intestinal health and homeostasis as well as to preserve a functional gut barrier, thus positively influencing host health and well-being. Among human-associated gut commensals, Bifidobacterium bifidum has been described as the only species capable of the extracellular degradation of both mucin-type glycans and HMOs, thereby giving this species a special role as a commensal gut forager of both host and diet-derived glycans. In the present study, we assess the possible beneficial properties and probiotic potential of B. bifidum strain CNCM I-4319. In silico genome analysis and growth experiments confirmed the expected ability of this strain to consume HMOs and mucin. By employing various animal models, we were also able to assess the ability of B. bifidum CNCM I-4319 to preserve gut integrity and functionality from stress-induced and inflammatory damage, thereby enforcing its potential as an effective probiotic strain.

5.
FEMS Microbiol Rev ; 44(6): 763-781, 2020 11 24.
Article in English | MEDLINE | ID: mdl-32821932

ABSTRACT

Maternal health status is vital for the development of the offspring of humans, including physiological health and psychological functions. The complex and diverse microbial ecosystem residing within humans contributes critically to these intergenerational impacts. Perinatal factors, including maternal nutrition, antibiotic use and maternal stress, alter the maternal gut microbiota during pregnancy, which can be transmitted to the offspring. In addition, gestational age at birth and mode of delivery are indicated frequently to modulate the acquisition and development of gut microbiota in early life. The early-life gut microbiota engages in a range of host biological processes, particularly immunity, cognitive neurodevelopment and metabolism. The perturbed early-life gut microbiota increases the risk for disease in early and later life, highlighting the importance of understanding relationships of perinatal factors with early-life microbial composition and functions. In this review, we present an overview of the crucial perinatal factors and summarise updated knowledge of early-life microbiota, as well as how the perinatal factors shape gut microbiota in short and long terms. We further discuss the clinical consequences of perturbations of early-life gut microbiota and potential therapeutic interventions with probiotics/live biotherapeutics.


Subject(s)
Gastrointestinal Microbiome/physiology , Delivery, Obstetric/statistics & numerical data , Female , Gestational Age , Humans , Infant, Newborn , Pregnancy
6.
Sci Rep ; 9(1): 15427, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659215

ABSTRACT

A number of bifidobacterial species are found at a particularly high prevalence and abundance in faecal samples of healthy breastfed infants, a phenomenon that is believed to be, at least partially, due to the ability of bifidobacteria to metabolize Human Milk Oligosaccharides (HMOs). In the current study, we isolated a novel strain of Bifidobacterium kashiwanohense, named APCKJ1, from the faeces of a four-week old breastfed infant, based on the ability of the strain to utilise the HMO component fucosyllactose. We then determined the full genome sequence of this strain, and employed the generated data to analyze fucosyllactose metabolism in B. kashiwanohense APCKJ1. Transcriptomic and growth analyses, combined with metabolite analysis, in vitro hydrolysis assays and heterologous expression, allowed us to elucidate the pathway for fucosyllactose metabolism in B. kashiwanohense APCKJ1. Homologs of the key genes for this metabolic pathway were identified in particular in infant-derived members of the Bifdobacterium genus, revealing the apparent niche-specific nature of this pathway, and allowing a broad perspective on bifidobacterial fucosyllactose and L-fucose metabolism.


Subject(s)
Bifidobacterium/metabolism , Breast Feeding , Feces/microbiology , Gastrointestinal Microbiome , Milk, Human/metabolism , Oligosaccharides/metabolism , Female , Humans , Infant, Newborn , Male
7.
Appl Microbiol Biotechnol ; 102(24): 10645-10663, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30306201

ABSTRACT

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.


Subject(s)
Bifidobacterium animalis/growth & development , Bifidobacterium animalis/genetics , Food Microbiology/methods , Milk/microbiology , Animals , Bifidobacterium animalis/drug effects , Carbohydrate Metabolism , Drug Resistance, Microbial , Female , Gastrointestinal Microbiome , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Bacterial , Mice, Inbred BALB C , Probiotics
8.
Sci Rep ; 8(1): 10633, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30006593

ABSTRACT

Bifidobacteria are common members of the gastro-intestinal microbiota of a broad range of animal hosts. Their successful adaptation to this particular niche is linked to their saccharolytic metabolism, which is supported by a wide range of glycosyl hydrolases. In the current study a large-scale gene-trait matching (GTM) effort was performed to explore glycan degradation capabilities in B. breve. By correlating the presence/absence of genes and associated genomic clusters with growth/no-growth patterns across a dataset of 20 Bifidobacterium breve strains and nearly 80 different potential growth substrates, we not only validated the approach for a number of previously characterized carbohydrate utilization clusters, but we were also able to discover novel genetic clusters linked to the metabolism of salicin and sucrose. Using GTM, genetic associations were also established for antibiotic resistance and exopolysaccharide production, thereby identifying (novel) bifidobacterial antibiotic resistance markers and showing that the GTM approach is applicable to a variety of phenotypes. Overall, the GTM findings clearly expand our knowledge on members of the B. breve species, in particular how their variable genetic features can be linked to specific phenotypes.


Subject(s)
Bifidobacterium breve/genetics , Genetic Association Studies , Genomics , Multigene Family , Benzyl Alcohols/metabolism , Bifidobacterium breve/metabolism , Biosynthetic Pathways/genetics , Computational Biology , Disk Diffusion Antimicrobial Tests , Drug Resistance, Bacterial/genetics , Glucosides/metabolism , Mutagenesis , Polysaccharides, Bacterial/biosynthesis , Sucrose/metabolism
9.
Nucleic Acids Res ; 46(4): 1860-1877, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29294107

ABSTRACT

Bifidobacterium breve represents one of the most abundant bifidobacterial species in the gastro-intestinal tract of breast-fed infants, where their presence is believed to exert beneficial effects. In the present study whole genome sequencing, employing the PacBio Single Molecule, Real-Time (SMRT) sequencing platform, combined with comparative genome analysis allowed the most extensive genetic investigation of this taxon. Our findings demonstrate that genes encoding Restriction/Modification (R/M) systems constitute a substantial part of the B. breve variable gene content (or variome). Using the methylome data generated by SMRT sequencing, combined with targeted Illumina bisulfite sequencing (BS-seq) and comparative genome analysis, we were able to detect methylation recognition motifs and assign these to identified B. breve R/M systems, where in several cases such assignments were confirmed by restriction analysis. Furthermore, we show that R/M systems typically impose a very significant barrier to genetic accessibility of B. breve strains, and that cloning of a methyltransferase-encoding gene may overcome such a barrier, thus allowing future functional investigations of members of this species.


Subject(s)
Bifidobacterium breve/genetics , DNA Methylation , DNA Modification Methylases/genetics , Genome, Bacterial , Bifidobacterium breve/classification , Bifidobacterium breve/enzymology , DNA Restriction Enzymes/genetics , Gene Transfer, Horizontal , Genomics , Nucleotide Motifs , Phylogeny
10.
BMC Genomics ; 18(1): 991, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29281966

ABSTRACT

BACKGROUND: Bifidobacterium breve represents a common member of the infant gut microbiota and its presence in the gut has been associated with host well being. For this reason it is relevant to investigate and understand the molecular mechanisms underlying the establishment, persistence and activities of this gut commensal in the host environment. RESULTS: The assessment of vegetative promoters in the bifidobacterial prototype Bifidobacterium breve UCC2003 was performed employing a combination of RNA tiling array analysis and cDNA sequencing. Canonical -10 (TATAAT) and -35 (TTGACA) sequences were identified upstream of transcribed genes or operons, where deviations from this consensus correspond to transcription level variations. A Random Forest analysis assigned the -10 region of B. breve promoters as the element most impacting on the level of transcription, followed by the spacer length and the 5'-UTR length of transcripts. Furthermore, our transcriptome study also identified rho-independent termination as the most common and effective termination signal of highly and moderately transcribed operons in B. breve. CONCLUSION: The present study allowed us to identify genes and operons that are actively transcribed in this organism during logarithmic growth, and link promoter elements with levels of transcription of essential genes in this organism. As homologs of many of our identified genes are present across the whole genus Bifidobacterium, our dataset constitutes a transcriptomic reference to be used for future investigations of gene expression in members of this genus.


Subject(s)
Bifidobacterium breve/genetics , Promoter Regions, Genetic , Transcriptome , Bifidobacterium breve/metabolism , Gene Expression Profiling , Genes, Essential , Oligonucleotide Array Sequence Analysis , RNA, Small Untranslated/biosynthesis , Riboswitch , Sequence Analysis, RNA , Transcription Initiation, Genetic , Transcription Termination, Genetic
11.
Appl Environ Microbiol ; 82(22): 6611-6623, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27590817

ABSTRACT

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.


Subject(s)
Bifidobacterium breve/genetics , Feces/microbiology , Genes, Bacterial , Multigene Family , Sulfatases/genetics , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/metabolism , Bifidobacterium breve/enzymology , Bifidobacterium breve/growth & development , Bifidobacterium breve/metabolism , Breast Feeding , DNA, Bacterial/genetics , Gastrointestinal Tract/microbiology , Gene Expression Profiling , Genome, Bacterial , Genomics/methods , Humans , Infant , Oligosaccharides/metabolism , Sulfatases/classification , Sulfatases/isolation & purification
12.
FEMS Microbiol Lett ; 362(4)2015 Feb.
Article in English | MEDLINE | ID: mdl-25688064

ABSTRACT

Bifidobacterium breve strains are numerically prevalent among the gut microbiota of healthy, breast-fed infants. The metabolism of sialic acid, a ubiquitous monosaccharide in the infant and adult gut, by B. breve UCC2003 is dependent on a large gene cluster, designated the nan/nag cluster. This study describes the transcriptional regulation of the nan/nag cluster and thus sialic acid metabolism in B. breve UCC2003. Insertion mutagenesis and transcriptome analysis revealed that the nan/nag cluster is regulated by a GntR family transcriptional repressor, designated NanR. Crude cell extract of Escherichia coli EC101 in which the nanR gene had been cloned and overexpressed was shown to bind to two promoter regions within this cluster, each of which containing an imperfect inverted repeat that is believed to act as the NanR operator sequence. Formation of the DNA-NanR complex is prevented in the presence of sialic acid, which we had previously shown to induce transcription of this gene cluster.


Subject(s)
Bifidobacterium/genetics , Bifidobacterium/metabolism , N-Acetylneuraminic Acid/metabolism , Regulatory Elements, Transcriptional , Repressor Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA, Bacterial/genetics , Escherichia coli/genetics , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Multigene Family , Mutagenesis, Insertional , Operator Regions, Genetic , Promoter Regions, Genetic , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sequence Alignment , Sequence Analysis
13.
BMC Microbiol ; 14: 282, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25420416

ABSTRACT

BACKGROUND: Bifidobacteria constitute a specific group of commensal bacteria that commonly inhabit the mammalian gastrointestinal tract. Bifidobacterium breve UCC2003 was previously shown to utilize a variety of plant/diet/host-derived carbohydrates, including cellodextrin, starch and galactan, as well as the mucin and HMO-derived monosaccharide, sialic acid. In the current study, we investigated the ability of this strain to utilize parts of a host-derived source of carbohydrate, namely the mucin glycoprotein, when grown in co-culture with the mucin-degrading Bifidobacterium bifidum PRL2010. RESULTS: B. breve UCC2003 was shown to exhibit growth properties in a mucin-based medium, but only when grown in the presence of B. bifidum PRL2010, which is known to metabolize mucin. A combination of HPAEC-PAD and transcriptome analyses identified some of the possible monosaccharides and oligosaccharides which support this enhanced co-cultivation growth/viability phenotype. CONCLUSION: This study describes the potential existence of a gut commensal relationship between two bifidobacterial species. We demonstrate the in vitro ability of B. breve UCC2003 to cross-feed on sugars released by the mucin-degrading activity of B. bifidum PRL2010, thus advancing our knowledge on the metabolic adaptability which allows the former strain to colonize the (infant) gut by its extensive metabolic abilities to (co-)utilize available carbohydrate sources.


Subject(s)
Bifidobacterium/growth & development , Bifidobacterium/metabolism , Culture Media/chemistry , Microbial Interactions , Mucins/metabolism , Bifidobacterium/physiology , Carbohydrate Metabolism , Proteolysis
14.
Appl Environ Microbiol ; 80(14): 4414-26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24814790

ABSTRACT

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.


Subject(s)
Bifidobacterium/metabolism , N-Acetylneuraminic Acid/metabolism , Bifidobacterium/genetics , Culture Media , DNA Fragmentation , DNA, Bacterial/genetics , Multigene Family , Oligonucleotide Array Sequence Analysis , Plasmids/genetics , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...