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










Publication year range
1.
bioRxiv ; 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39005317

ABSTRACT

Bifidobacteria are among the earliest colonizers of the human gut, conferring numerous health benefits. While multiple Bifidobacterium strains are used as probiotics, accumulating evidence suggests that the individual responses to probiotic supplementation may vary, likely due to a variety of factors, including strain type(s), gut community composition, dietary habits of the consumer, and other health/lifestyle conditions. Given the saccharolytic nature of bifidobacteria, the carbohydrate composition of the diet is one of the primary factors dictating the colonization efficiency of Bifidobacterium strains. Therefore, a comprehensive understanding of bifidobacterial glycan metabolism at the strain level is necessary to rationally design probiotic or synbiotic formulations that combine bacterial strains with glycans that match their nutrient preferences. In this study, we systematically reconstructed 66 pathways involved in the utilization of mono-, di-, oligo-, and polysaccharides by analyzing the representation of 565 curated metabolic functional roles (catabolic enzymes, transporters, transcriptional regulators) in 2973 non-redundant cultured Bifidobacterium isolates and metagenome-assembled genomes (MAGs). Our analysis uncovered substantial heterogeneity in the predicted glycan utilization capabilities at the species and strain level and revealed the presence of a yet undescribed phenotypically distinct subspecies-level clade within the Bifidobacterium longum species. We also identified Bangladeshi isolates harboring unique gene clusters tentatively implicated in the breakdown of xyloglucan and human milk oligosaccharides. Predicted carbohydrate utilization phenotypes were experimentally characterized and validated. Our large-scale genomic analysis considerably expands the knowledge of carbohydrate metabolism in bifidobacteria and provides a foundation for rationally designing single- or multi-strain probiotic formulations of a given bifidobacterial species as well as synbiotic combinations of bifidobacterial strains matched with their preferred carbohydrate substrates.

2.
Proc Natl Acad Sci U S A ; 120(39): e2311422120, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37733741

ABSTRACT

Understanding how members of the human gut microbiota prioritize nutrient resources is one component of a larger effort to decipher the mechanisms defining microbial community robustness and resiliency in health and disease. This knowledge is foundational for development of microbiota-directed therapeutics. To model how bacteria prioritize glycans in the gut, germfree mice were colonized with 13 human gut bacterial strains, including seven saccharolytic Bacteroidaceae species. Animals were fed a Western diet supplemented with pea fiber. After community assembly, an inducible CRISPR-based system was used to selectively and temporarily reduce the absolute abundance of Bacteroides thetaiotaomicron or B. cellulosilyticus by 10- to 60-fold. Each knockdown resulted in specific, reproducible increases in the abundances of other Bacteroidaceae and dynamic alterations in their expression of genes involved in glycan utilization. Emergence of these "alternate consumers" was associated with preservation of community saccharolytic activity. Using an inducible system for CRISPR base editing in vitro, we disrupted translation of transporters critical for utilizing dietary polysaccharides in Phocaeicola vulgatus, a B. cellulosilyticus knockdown-responsive taxon. In vitro and in vivo tests of the resulting P. vulgatus mutants allowed us to further characterize mechanisms associated with its increased fitness after knockdown. In principle, the approach described can be applied to study utilization of a range of nutrients and to preclinical efforts designed to develop therapeutic strategies for precision manipulation of microbial communities.


Subject(s)
Bacteroides thetaiotaomicron , Bacteroides , Humans , Animals , Mice , Bacteroides/genetics , Polysaccharides , Bacteroides thetaiotaomicron/genetics , Biological Assay , Diet, Western
3.
Sci Transl Med ; 14(640): eabk1107, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35417188

ABSTRACT

Disrupted development of the gut microbiota is a contributing cause of childhood malnutrition. Bifidobacterium longum subspecies infantis is a prominent early colonizer of the infant gut that consumes human milk oligosaccharides (HMOs). We found that the absolute abundance of Bifidobacterium infantis is lower in 3- to 24-month-old Bangladeshi infants with severe acute malnutrition (SAM) compared to their healthy age-matched counterparts. A single-blind, placebo-controlled trial (SYNERGIE) was conducted in 2- to 6-month-old Bangladeshi infants with SAM. A commercial U.S. donor-derived B. infantis strain (EVC001) was administered daily with or without the HMO lacto-N-neotetraose for 28 days. This intervention increased fecal B. infantis abundance in infants with SAM, although to levels still 10- to 100-fold lower than in untreated healthy controls. EVC001 treatment promoted weight gain that was associated with reduced intestinal inflammation markers in infants with SAM. We cultured fecal B. infantis strains from Bangladeshi infants and colonized gnotobiotic mice with these cultured strains. The gnotobiotic mice were fed a diet representative of that consumed by 6-month-old Bangladeshi infants, with or without HMO supplementation. One B. infantis strain, Bg_2D9, expressing two gene clusters involved in uptake and utilization of N-glycans and plant-derived polysaccharides, exhibited superior fitness over EVC001. The fitness advantage of Bg_2D9 was confirmed in a gnotobiotic mouse model of mother-to-infant gut microbiota transmission where dams received a pretreatment fecal community from a SAM infant in the SYNERGIE trial. Whether Bg_2D9 is superior to EVC001 for treating malnourished infants who consume a diet with limited breastmilk requires further clinical testing.


Subject(s)
Bifidobacterium longum subspecies infantis , Severe Acute Malnutrition , Animals , Bifidobacterium , Feces/microbiology , Humans , Infant , Mice , Milk, Human , Single-Blind Method , Weight Gain
4.
Cell Host Microbe ; 29(4): 664-673.e5, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33571448

ABSTRACT

Greater understanding of the spatial relationships between members of the human gut microbiota and available nutrients is needed to gain deeper insights about community dynamics and expressed functions. Therefore, we generated a panel of artificial food particles with each type composed of microscopic paramagnetic beads coated with a fluorescent barcode and one of 60 different dietary or host glycan preparations. Analysis of 160 Bacteroides and Parabacteroides strains disclosed diverse strain-specific and glycan-specific binding phenotypes. We identified carbohydrate structures that correlated with binding by specific bacterial strains in vitro and noted strain-specific differences in the catabolism of glycans that mediate adhesion. Mixed in vitro cultures revealed that these adhesion phenotypes are maintained in more complex communities. Additionally, orally administering glycan beads to gnotobiotic mice confirmed specificity in glycan binding. This approach should facilitate analyses of how strains occupying the same physical niche interact, and it should advance the development of synbiotics, more nutritious foods, and microbiota-based diagnostics.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Polysaccharides/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteroides , Food , Gastrointestinal Tract/microbiology , Humans , Male , Mice , Mice, Inbred C57BL , Polysaccharides/administration & dosage
5.
Cell Host Microbe ; 27(6): 899-908.e5, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32348782

ABSTRACT

Undernourished children in low-income countries often exhibit poor responses to oral vaccination. Perturbed microbiota development is linked to undernutrition, but whether and how microbiota changes affect vaccine responsiveness remains unclear. Here, we show that gnotobiotic mice colonized with microbiota from undernourished Bangladeshi children and fed a Bangladeshi diet exhibited microbiota-dependent differences in mucosal IgA responses to oral vaccination with cholera toxin (CT). Supplementation with a nutraceutical consisting of spirulina, amaranth, flaxseed, and micronutrients augmented CT-IgA production. Mice initially colonized with a microbiota associated with poor CT responses exhibited improved immunogenicity upon invasion of bacterial taxa from cagemates colonized with a more "responsive" microbiota. Additionally, a consortium of five cultured bacterial invaders conferred augmented CT-IgA responses in mice fed the supplemented diet and colonized with the "hypo-responsive" community. These results provide preclinical proof-of-concept that diet and microbiota influence mucosal immune responses to CT vaccination and identify a candidate synbiotic formulation.


Subject(s)
Cholera , Gastrointestinal Microbiome/physiology , Malnutrition , Prebiotics , Vaccination , Animals , Bacteria/classification , Child , Cholera Toxin/pharmacology , Diet , Dietary Supplements , Disease Models, Animal , Germ-Free Life , Humans , Immunity, Mucosal , Immunoglobulin A , Male , Mice , Mice, Inbred C57BL , Mucous Membrane/immunology , Probiotics
6.
Proc Natl Acad Sci U S A ; 116(24): 11988-11996, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31138692

ABSTRACT

Undernutrition in children is a pressing global health problem, manifested in part by impaired linear growth (stunting). Current nutritional interventions have been largely ineffective in overcoming stunting, emphasizing the need to obtain better understanding of its underlying causes. Treating Bangladeshi children with severe acute malnutrition with therapeutic foods reduced plasma levels of a biomarker of osteoclastic activity without affecting biomarkers of osteoblastic activity or improving their severe stunting. To characterize interactions among the gut microbiota, human milk oligosaccharides (HMOs), and osteoclast and osteoblast biology, young germ-free mice were colonized with cultured bacterial strains from a 6-mo-old stunted infant and fed a diet mimicking that consumed by the donor population. Adding purified bovine sialylated milk oligosaccharides (S-BMO) with structures similar to those in human milk to this diet increased femoral trabecular bone volume and cortical thickness, reduced osteoclasts and their bone marrow progenitors, and altered regulators of osteoclastogenesis and mediators of Th2 responses. Comparisons of germ-free and colonized mice revealed S-BMO-dependent and microbiota-dependent increases in cecal levels of succinate, increased numbers of small intestinal tuft cells, and evidence for activation of a succinate-induced tuft cell signaling pathway linked to Th2 immune responses. A prominent fucosylated HMO, 2'-fucosyllactose, failed to elicit these changes in bone biology, highlighting the structural specificity of the S-BMO effects. These results underscore the need to further characterize the balance between, and determinants of, osteoclastic and osteoblastic activity in stunted infants/children, and suggest that certain milk oligosaccharides may have therapeutic utility in this setting.


Subject(s)
Bone and Bones/drug effects , Germ-Free Life/drug effects , Malnutrition/drug therapy , Milk, Human/metabolism , Oligosaccharides/administration & dosage , Osteoblasts/drug effects , Osteoclasts/drug effects , Animals , Bacteria/drug effects , Cattle , Diet , Disease Models, Animal , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Humans , Infant , Intestine, Small/microbiology , Male , Malnutrition/microbiology , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
7.
J Biol Chem ; 290(20): 12630-49, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25795776

ABSTRACT

The adaptive immune response to the human gut microbiota consists of a complex repertoire of antibodies interacting with a broad range of taxa. Fusing intestinal lamina propria lymphocytes from mice monocolonized with Bacteroides thetaiotaomicron to a myeloma fusion partner allowed us to recover hybridomas that captured naturally primed, antigen-specific antibody responses representing multiple isotypes, including IgA. One of these hybridomas, 260.8, produced a monoclonal antibody that recognizes an epitope specific for B. thetaiotaomicron isolates in a large panel of hospital- and community-acquired Bacteroides. Whole genome transposon mutagenesis revealed a 19-gene locus, involved in LPS O-antigen polysaccharide synthesis and conserved among multiple B. thetaiotaomicron isolates, that is required for 260.8 epitope expression. Mutants in this locus exhibited marked fitness defects in vitro during growth in rich medium and in gnotobiotic mice colonized with defined communities of human gut symbionts. Expression of the 260.8 epitope was sustained during 10 months of daily passage in vitro and during 14 months of monocolonization of gnotobiotic wild-type, Rag1-/-, or Myd88-/- mice. Comparison of gnotobiotic Rag1-/- mice with and without subcutaneous 260.8 hybridomas disclosed that this IgA did not affect B. thetaiotaomicron population density or suppress 260.8 epitope production but did affect bacterial gene expression in ways emblematic of a diminished host innate immune response. Our study illustrates an approach for (i) generating diagnostic antibodies, (ii) characterizing IgA responses along a continuum of specificity/degeneracy that defines the IgA repertoire to gut symbionts, and (iii) identifying immunogenic epitopes that affect competitiveness and help maintain host-microbe mutualism.


Subject(s)
Antibodies, Bacterial/immunology , Bacteroides/immunology , Epitopes/immunology , Immunoglobulin A/immunology , Intestinal Mucosa/immunology , Animals , Antibodies, Bacterial/genetics , Bacteroides/genetics , DNA Transposable Elements , Epitopes/genetics , Genetic Loci/immunology , Humans , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Mutagenesis , Mutation , O Antigens/genetics , O Antigens/immunology , Species Specificity
8.
Science ; 341(6141): 1237439, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23828941

ABSTRACT

A low-error 16S ribosomal RNA amplicon sequencing method, in combination with whole-genome sequencing of >500 cultured isolates, was used to characterize bacterial strain composition in the fecal microbiota of 37 U.S. adults sampled for up to 5 years. Microbiota stability followed a power-law function, which when extrapolated suggests that most strains in an individual are residents for decades. Shared strains were recovered from family members but not from unrelated individuals. Sampling of individuals who consumed a monotonous liquid diet for up to 32 weeks indicated that changes in strain composition were better predicted by changes in weight than by differences in sampling interval. This combination of stability and responsiveness to physiologic change confirms the potential of the gut microbiota as a diagnostic tool and therapeutic target.


Subject(s)
Gastrointestinal Tract/microbiology , Metagenome , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Body Composition , Caloric Restriction , Family , Feces/microbiology , Female , Genome, Bacterial/genetics , Genomic Instability , Humans , Male , Models, Biological , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Time Factors , Weight Loss , Young Adult
9.
Proc Natl Acad Sci U S A ; 108 Suppl 1: 4599-606, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21317366

ABSTRACT

The human gut microbiota harbors three main groups of H(2)-consuming microbes: methanogens including the dominant archaeon, Methanobrevibacter smithii, a polyphyletic group of acetogens, and sulfate-reducing bacteria. Defining their roles in the gut is important for understanding how hydrogen metabolism affects the efficiency of fermentation of dietary components. We quantified methanogens in fecal samples from 40 healthy adult female monozygotic (MZ) and 28 dizygotic (DZ) twin pairs, analyzed bacterial 16S rRNA datasets generated from their fecal samples to identify taxa that co-occur with methanogens, sequenced the genomes of 20 M. smithii strains isolated from families of MZ and DZ twins, and performed RNA-Seq of a subset of strains to identify their responses to varied formate concentrations. The concordance rate for methanogen carriage was significantly higher for MZ versus DZ twin pairs. Co-occurrence analysis revealed 22 bacterial species-level taxa positively correlated with methanogens: all but two were members of the Clostridiales, with several being, or related to, known hydrogen-producing and -consuming bacteria. The M. smithii pan-genome contains 987 genes conserved in all strains, and 1,860 variably represented genes. Strains from MZ and DZ twin pairs had a similar degree of shared genes and SNPs, and were significantly more similar than strains isolated from mothers or members of other families. The 101 adhesin-like proteins (ALPs) in the pan-genome (45 ± 6 per strain) exhibit strain-specific differences in expression and responsiveness to formate. We hypothesize that M. smithii strains use their different repertoires of ALPs to create diversity in their metabolic niches, by allowing them to establish syntrophic relationships with bacterial partners with differing metabolic capabilities and patterns of co-occurrence.


Subject(s)
Adhesins, Bacterial/genetics , Gastrointestinal Tract/microbiology , Genome, Archaeal , Methanobrevibacter/genetics , Twins , Adult , Base Sequence , Female , Formates/analysis , Humans , Metagenomics , Methanobrevibacter/metabolism , Molecular Sequence Data , Polymorphism, Single Nucleotide/genetics , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Species Specificity
10.
J Exp Med ; 206(13): 3061-72, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-20008521

ABSTRACT

Variation of surface antigen expression is a mechanism used by microbes to adapt to and persist within their host habitats. Helicobacter pylori, a persistent bacterial colonizer of the human stomach, can alter its surface Lewis (Le) antigen expression. We examined H. pylori colonization in mice to test the hypothesis that host phenotype selects for H. pylori (Le) phenotypes. When wild-type and Le(b)-expressing transgenic FVB/N mice were challenged with H. pylori strain HP1, expressing Le(x) and Le(y), we found that bacterial populations recovered after 8 mo from Le(b)-transgenic, but not wild-type, mice expressed Le(b). Changes in Le phenotype were linked to variation of a putative galactosyltransferase gene (beta-(1,3)galT); mutagenesis and complementation revealed its essential role in type I antigen expression. These studies indicate that H. pylori evolves to resemble the host's gastric Le phenotype, and reveal a bacterial genetic locus that is subject to host-driven selection pressure.


Subject(s)
Helicobacter pylori/immunology , Lewis Blood Group Antigens/analysis , Lewis X Antigen/analysis , Oligosaccharides/physiology , Adhesins, Bacterial/analysis , Animals , Antibodies, Bacterial/blood , Flow Cytometry , Fucosyltransferases/genetics , Galactosyltransferases/genetics , Lipopolysaccharides/toxicity , Mice , Mice, Transgenic , Phenotype
11.
J Biol Chem ; 284(44): 30383-94, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19723631

ABSTRACT

Helicobacter pylori infection is associated with gastric adenocarcinoma in some humans, especially those that develop an antecedent condition, chronic atrophic gastritis (ChAG). Gastric epithelial progenitors (GEPs) in transgenic gnotobiotic mice with a ChAG-like phenotype harbor intracellular collections of H. pylori. To characterize H. pylori adaptations to ChAG, we sequenced the genomes of 24 isolates obtained from 6 individuals, each sampled over a 4-year interval, as they did or did not progress from normal gastric histology to ChAG and/or adenocarcinoma. H. pylori populations within study participants were largely clonal and remarkably stable regardless of disease state. GeneChip studies of the responses of a cultured mouse gastric stem cell-like line (mGEPs) to infection with sequenced strains yielded a 695-member dataset of transcripts that are (i) differentially expressed after infection with ChAG-associated isolates, but not with a "normal" or a heat-killed ChAG isolate, and (ii) enriched in genes and gene functions associated with tumorigenesis in general and gastric carcinogenesis in specific cases. Transcriptional profiling of a ChAG strain during mGEP infection disclosed a set of responses, including up-regulation of hopZ, an adhesin belonging to a family of outer membrane proteins. Expression profiles of wild-type and DeltahopZ strains revealed a number of pH-regulated genes modulated by HopZ, including hopP, which binds sialylated glycans produced by GEPs in vivo. Genetic inactivation of hopZ produced a fitness defect in the stomachs of gnotobiotic transgenic mice but not in wild-type littermates. This study illustrates an approach for identifying GEP responses specific to ChAG-associated H. Pylori strains and bacterial genes important for survival in a model of the ChAG gastric ecosystem.


Subject(s)
Gastritis, Atrophic/microbiology , Gene Expression Profiling , Helicobacter pylori/genetics , Stem Cells/microbiology , Animals , Bacterial Proteins/genetics , Cell Line , Chronic Disease , Gastric Mucosa/cytology , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Helicobacter pylori/physiology , Humans , Mice , Stem Cells/metabolism , Sweden , Transcription, Genetic
12.
Cell Host Microbe ; 2(5): 328-39, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18005754

ABSTRACT

Colonization of germ-free mice with a normal gut microbiota elicits bacteria-specific IgA antibody responses. The effects of these responses on microbial and host biology remain poorly defined. Therefore, we developed a gnotobiotic mouse model where the microbiota is reduced to one bacterial species, and the antibody repertoire to a single, monoclonal IgA against the bacterium's capsular polysaccharide. Bacteroides thetaiotaomicron was introduced into germ-free wild-type, immunodeficient Rag1(-/-), or Rag1(-/-) mice harboring IgA-producing hybridoma cells. Without IgA, B. thetaiotaomicron elicits a more robust innate immune response and reacts to this response by inducing genes that metabolize host oxidative products. IgA reduces intestinal proinflammatory signaling and bacterial epitope expression, thereby balancing suppression of the oxidative burst with the antibody's negative impact on bacterial fitness. These results underscore the adaptive immune system's critical role in establishing a sustainable host-microbial relationship. Immunoselection of bacterial epitope expression may contribute to the remarkable strain-level diversity in this ecosystem.


Subject(s)
Antibodies, Bacterial/physiology , Bacteroides Infections/immunology , Bacteroides Infections/microbiology , Bacteroides/physiology , Homeostasis , Immunoglobulin A/physiology , Symbiosis , Animals , Bacterial Capsules/immunology , Cecum/immunology , Cecum/microbiology , Epitopes/immunology , Gene Expression , Genes, Bacterial/genetics , Genes, RAG-1 , Germ-Free Life , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism
13.
Proc Natl Acad Sci U S A ; 100(6): 3467-72, 2003 Mar 18.
Article in English | MEDLINE | ID: mdl-12629225

ABSTRACT

Helicobacter pylori infection of the human stomach is common and typically benign, although a subset of hosts develops severe pathology. Infection occurs in an organ with distinct microenvironments characterized by pronounced differences in the composition of acid-producing parietal cells. In this study, we examine determinants of bacterial tropism to various gastric niches by using germ-free normal and transgenic mice with an engineered parietal cell ablation. Mice were colonized for 8 weeks with a clinical isolate (Hp1) that expresses adhesins recognized by epithelial NeuAcalpha2,3Galbeta1,4 glycan receptors. In normal mice, Hp1 has tropism for a parietal cell-deficient niche where sialylated glycans are expressed by a narrow band of pit cells positioned at the boundary between the squamous epithelium (forestomach) and the proximal glandular epithelium. Lymphoid aggregates that develop in this niche, but not elsewhere in the stomach, were analyzed by GeneChip and quantitative RT-PCR studies of laser capture microdissected mucosa and yielded a series of biomarkers indicative of immune cell activation and maturation. Genetic ablation of parietal cells produced a new source of NeuAcalpha2,3Galbeta1,4 glycans in amplified gastric epithelial lineage progenitors, with accompanying expansion of Hp1 within the glandular epithelium. Lymphoid aggregates that develop in this formerly acid-protected epithelium have molecular features similar to those observed at the forestomach/glandular junction. These findings demonstrate the important roles played by parietal cells and glycan receptors in determining the positioning of H. pylori within the gastric ecosystem, and emphasize the need to consider the evolution of pathology within a given host in a niche-specific context.


Subject(s)
Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Parietal Cells, Gastric/microbiology , Parietal Cells, Gastric/pathology , Animals , Diphtheria Toxin/genetics , Gastritis/etiology , Gene Expression Profiling , Germ-Free Life , Helicobacter Infections/etiology , Helicobacter pylori/isolation & purification , Humans , Lymphoid Tissue/pathology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Peptide Fragments/genetics
14.
J Biol Chem ; 277(37): 34191-7, 2002 Sep 13.
Article in English | MEDLINE | ID: mdl-12105196

ABSTRACT

Helicobacter pylori infects the stomachs of half of all humans. It has a relatively benign relationship with most hosts but produces severe pathology, including gastric cancer, in others. Identifying the relative contributions of host, microbial, and environmental factors to the outcome of infection has been challenging. Here we describe one approach for identifying microbial genes that affect the magnitude of host responses to infection. Single colony purified H. pylori isolates were obtained from 25 cases and 71 controls in a Swedish case-control study of gastric cancer. Strains were first phenotyped based on their ability to produce adhesins that recognize two classes of human gastric epithelial receptors. Thirteen binding strains and two non-binding controls were then subjected to whole genome genotyping using H. pylori DNA microarrays. A cohort of "variable" genes was identified based on a microarray-determined call of "absent" in at least one member of the strain panel. Each strain was subsequently introduced into two types of germ-free transgenic mice, each programmed to express a different host factor postulated to pose increased risk for development of severe pathology. Expression of biomarkers of host defense was quantitated 4 weeks after inoculation, and the magnitude of the response correlated with bacterial genotype. The proportion of genes encoding HsdS homologs (specificity subunit of hetero-oligomeric type I restriction-modification systems) was significantly higher in the pool of 18 variable genes whose presence directly correlated with a robust host response than their proportion in the remaining 352 members of the variable gene pool. This suggests that the functions of these HsdS homologs may include control of expression of microbial determinants that affect the extent of gastric responses to this potentially virulent pathogen.


Subject(s)
Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Stomach Neoplasms/microbiology , Animals , Biomarkers , Case-Control Studies , Genome, Bacterial , Genotype , Germ-Free Life , Helicobacter pylori/classification , Humans , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Species Specificity , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL