Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Blood ; 129(6): 729-739, 2017 02 09.
Article in English | MEDLINE | ID: mdl-27879260

ABSTRACT

Bone marrow suppression is an adverse effect associated with many antibiotics, especially when administered for prolonged treatment courses. Recent advances in our understanding of steady-state hematopoiesis have allowed us to explore the effects of antibiotics on hematopoietic progenitors in detail using a murine model. Antibiotic-treated mice exhibited anemia, thrombocytosis, and leukopenia, with pronounced pan-lymphopenia as demonstrated by flow cytometric analysis of peripheral blood. Bone marrow progenitor analysis revealed depletion of hematopoietic stem cells and multipotent progenitors across all subtypes. Granulocytes and B cells were also diminished in the bone marrow, whereas the number of CD8+ T cells increased. Reductions in progenitor activity were not observed when cells were directly incubated with antibiotics, suggesting that these effects are indirect. Hematopoietic changes were associated with a significant contraction of the fecal microbiome and were partially rescued by fecal microbiota transfer. Further, mice raised in germ-free conditions had hematopoietic abnormalities similar to those seen in antibiotic-treated mice, and antibiotic therapy of germ-free mice caused no additional abnormalities. The effects of antibiotics were phenocopied in Stat1-deficient mice, with no additional suppression by antibiotics in these mice. We conclude that microbiome depletion as a result of broad-spectrum antibiotic treatment disrupts basal Stat1 signaling and alters T-cell homeostasis, leading to impaired progenitor maintenance and granulocyte maturation. Methods to preserve the microbiome may reduce the incidence of antibiotic-associated bone marrow suppression.


Subject(s)
Anemia/chemically induced , Anti-Bacterial Agents/adverse effects , Gastrointestinal Microbiome/drug effects , Hematopoiesis/drug effects , Leukopenia/chemically induced , STAT1 Transcription Factor/genetics , Thrombocytosis/chemically induced , Anemia/microbiology , Anemia/pathology , Anemia/therapy , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Gene Expression , Germ-Free Life/drug effects , Germ-Free Life/genetics , Granulocytes/drug effects , Granulocytes/metabolism , Granulocytes/pathology , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukopenia/microbiology , Leukopenia/pathology , Leukopenia/therapy , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT1 Transcription Factor/deficiency , Signal Transduction , Thrombocytosis/microbiology , Thrombocytosis/pathology , Thrombocytosis/therapy
2.
Proc Natl Acad Sci U S A ; 113(50): E8141-E8150, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911839

ABSTRACT

Th17 cells accrue in the intestine in response to particular microbes. In rodents, segmented filamentous bacteria (SFB) induce intestinal Th17 cells, but analogously functioning microbes in humans remain undefined. Here, we identified human symbiont bacterial species, in particular Bifidobacterium adolescentis, that could, alone, induce Th17 cells in the murine intestine. Similar to SFB, B. adolescentis was closely associated with the gut epithelium and engendered cognate Th17 cells without attendant inflammation. However, B. adolescentis elicited a transcriptional program clearly distinct from that of SFB, suggesting an alternative mechanism of promoting Th17 cell accumulation. Inoculation of mice with B. adolescentis exacerbated autoimmune arthritis in the K/BxN mouse model. Several off-the-shelf probiotic preparations that include Bifidobacterium strains also drove intestinal Th17 cell accumulation.


Subject(s)
Bifidobacterium adolescentis/immunology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Th17 Cells/immunology , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/immunology , Arthritis, Experimental/microbiology , Bifidobacterium adolescentis/isolation & purification , Female , Gene Expression Profiling , Germ-Free Life/genetics , Germ-Free Life/immunology , Humans , Immunity, Mucosal , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Male , Mice , Mice, Inbred C57BL , Probiotics , Symbiosis/genetics , Symbiosis/immunology , Th17 Cells/cytology
3.
Int J Mol Sci ; 20(7)2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30934845

ABSTRACT

Commensal microbiota contribute to gut homeostasis by inducing transcription of mucosal genes. Analysis of the impact of various microbiota on intestinal tissue provides an important insight into the function of this organ. We used cDNA microarrays to determine the gene expression signature of mucosa isolated from the small intestine and colon of germ-free (GF) mice and animals monoassociated with two E. coli strains. The results were compared to the expression data obtained in conventionally reared (CR) mice. In addition, we analyzed gene expression in colon organoids derived from CR, GF, and monoassociated animals. The analysis revealed that the complete absence of intestinal microbiota mainly affected the mucosal immune system, which was not restored upon monoassociation. The most important expression changes observed in the colon mucosa indicated alterations in adipose tissue and lipid metabolism. In the comparison of differentially expressed genes in the mucosa or organoids obtained from GF and CR mice, only six genes were common for both types of samples. The results show that the increased expression of the angiopoietin-like 4 (Angptl4) gene encoding a secreted regulator of lipid metabolism indicates the GF status.


Subject(s)
Gene Expression Profiling , Germ-Free Life/genetics , Intestinal Mucosa/metabolism , Organoids/metabolism , Animals , Biomarkers/metabolism , Colon/metabolism , Escherichia coli/physiology , Gene Expression Regulation , Immune System/metabolism , Immunity, Mucosal , Mice, Inbred BALB C , Microbiota
4.
Drug Metab Dispos ; 43(10): 1572-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25956306

ABSTRACT

Intestinal bacteria have been shown to be important in regulating host intermediary metabolism and contributing to obesity. However, relatively less is known about the effect of intestinal bacteria on the expression of hepatic drug-processing genes in the host. This study characterizes the expression of hepatic drug-processing genes in germ-free (GF) mice using RNA-Seq. Total RNA were isolated from the livers of adult male conventional and GF C57BL/6J mice (n = 3 per group). In the livers of GF mice, the mRNA of the aryl hydrocarbon receptor target gene Cyp1a2 was increased 51%, and the mRNA of the peroxisome proliferator-activated receptor α (PPARα) target gene Cyp4a14 was increased 202%. Conversely, the mRNA of the constitutive androstane receptor (CAR) target gene Cyp2b10 was decreased 57%, and the mRNA of the pregnane X receptor target gene Cyp3a11 was decreased 87% in GF mice. Although other non-Cyp phase-1 enzymes in the livers of GF mice were only moderately affected, there was a marked down-regulation in the phase-2 enzymes glutathione S-transferases p1 and p2, as well as a marked up-regulation in the major bile acid transporters Na(+)-taurocholate cotransporting polypeptide and organic anion-transporting polypeptide 1b2, and the cholesterol transporter ATP-binding cassette transporter Abcg5/Abcg8. This study demonstrates that intestinal bacteria regulate the expression of a large number of drug-processing genes, which suggests that intestinal bacteria are responsible for some individual differences in drug responses.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Germ-Free Life/genetics , Liver , Pharmaceutical Preparations , RNA/genetics , Sequence Analysis, RNA/methods , Animals , Cytochrome P-450 Enzyme System/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism
5.
Proc Natl Acad Sci U S A ; 109(39): E2605-14, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22949679

ABSTRACT

How fish larvae are protected from infection before the maturation of adaptive immunity, a process which may take up to several weeks in most species, has long been a matter of speculation. Using a germ-free model, we show that colonization by commensals in newly hatched zebrafish primes neutrophils and induces several genes encoding proinflammatory and antiviral mediators, increasing the resistance of larvae to viral infection. Commensal microbe recognition was found to be mediated mainly through a TLR/MyD88 signaling pathway, and professional phagocytes were identified as the source of these immune mediators. However, the induction of proinflammatory and antiviral genes, but not of antimicrobial effector genes, also required the covalent modification of histone H3 at gene promoters. Interestingly, chromatin modifications were not altered by commensal microbes or hatching. Taken together, our results demonstrate that gene-specific chromatin modifications are associated with the protection of zebrafish larvae against infectious agents before adaptive immunity has developed and prevent pathologies associated with excessive inflammation during development.


Subject(s)
Bacteria/immunology , Chromatin/immunology , Germ-Free Life/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptors/immunology , Zebrafish Proteins/immunology , Zebrafish/immunology , Animals , Chromatin/genetics , Chromatin/metabolism , Germ-Free Life/genetics , Histones/genetics , Histones/immunology , Histones/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Zebrafish/classification , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
J Immunol ; 186(3): 1531-7, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21178008

ABSTRACT

Retinoic acid-related orphan receptor (ROR)γt(+) TCRαß(+) cells expressing IL-17, termed Th17 cells, are most abundant in the intestinal lamina propria. Symbiotic microbiota are required for the generation of Th17 cells, but the requirement for microbiota-derived Ag is not documented. In this study, we show that normal numbers of Th17 cells develop in the intestine of mice that express a single TCR in the absence of cognate Ag, whereas the microbiota remains essential for their development. However, such mice, or mice monocolonized with the Th17-inducing segmented filamentous bacteria, fail to induce normal numbers of Foxp3(+) RORγt(+) T cells, the regulatory counterpart of IL-17(+)RORγt(+) T cells. These results demonstrate that a complex microbiota and cognate Ag are required to generate a properly regulated set of RORγt(+) T cells and Th17 cells.


Subject(s)
Cell Proliferation , Interleukin-17/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Receptors, Antigen, T-Cell/deficiency , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Amino Acid Sequence , Animals , CD4 Lymphocyte Count , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Germ-Free Life/genetics , Germ-Free Life/immunology , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/pathology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Interleukin-17/genetics , Intestinal Mucosa/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes, Helper-Inducer/pathology
7.
BMC Genomics ; 13: 335, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22823934

ABSTRACT

BACKGROUND: Epidemiological studies have suggested that the encounter with commensal microorganisms during the neonatal period is essential for normal development of the host immune system. Basic research involving gnotobiotic mice has demonstrated that colonization at the age of 5 weeks is too late to reconstitute normal immune function. In this study, we examined the transcriptome profiles of the large intestine (LI), small intestine (SI), liver (LIV), and spleen (SPL) of 3 bacterial colonization models-specific pathogen-free mice (SPF), ex-germ-free mice with bacterial reconstitution at the time of delivery (0WexGF), and ex-germ-free mice with bacterial reconstitution at 5 weeks of age (5WexGF)-and compared them with those of germ-free (GF) mice. RESULTS: Hundreds of genes were affected in all tissues in each of the colonized models; however, a gene set enrichment analysis method, MetaGene Profiler (MGP), demonstrated that the specific changes of Gene Ontology (GO) categories occurred predominantly in 0WexGF LI, SPF SI, and 5WexGF SPL, respectively. MGP analysis on signal pathways revealed prominent changes in toll-like receptor (TLR)- and type 1 interferon (IFN)-signaling in LI of 0WexGF and SPF mice, but not 5WexGF mice, while 5WexGF mice showed specific changes in chemokine signaling. RT-PCR analysis of TLR-related genes showed that the expression of interferon regulatory factor 3 (Irf3), a crucial rate-limiting transcription factor in the induction of type 1 IFN, prominently decreased in 0WexGF and SPF mice but not in 5WexGF and GF mice. CONCLUSION: The present study provides important new information regarding the molecular mechanisms of the so-called "hygiene hypothesis".


Subject(s)
Bacteria/metabolism , Germ-Free Life/genetics , Germ-Free Life/immunology , Immune System/growth & development , Immune System/microbiology , Oligonucleotide Array Sequence Analysis/methods , Animals , Animals, Newborn , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Developmental , Intestine, Small/growth & development , Intestine, Small/metabolism , Liver/growth & development , Liver/metabolism , Mice , Models, Biological , Multigene Family/genetics , Organ Specificity/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Spleen/growth & development , Spleen/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
8.
J Clin Invest ; 118(1): 205-16, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18060042

ABSTRACT

TLRs may contribute to the progression of rheumatoid arthritis through recognition of microbial or host-derived ligands found in arthritic joints. Here, we show that TLR2 and TLR4, but not TLR9, are involved in the pathogenesis of autoimmune arthritis and play distinct roles in the regulation of T cells and cytokines. We investigated the involvement of TLR2, TLR4, and TLR9 in the progression of arthritis using IL-1 receptor antagonist-knockout (IL1rn-/-) mice, which spontaneously develop an autoimmune T cell-mediated arthritis. Spontaneous onset of arthritis was dependent on TLR activation by microbial flora, as germ-free mice did not develop arthritis. Clinical and histopathological evaluation of IL1rn-/-Tlr2-/- mice revealed more severe arthritis, characterized by reduced suppressive function of Tregs and substantially increased IFN-gamma production by T cells. IL1rn-/-Tlr4-/- mice were, in contrast, protected against severe arthritis and had markedly lower numbers of Th17 cells and a reduced capacity to produce IL-17. A lack of Tlr9 did not affect the progression of arthritis. While any therapeutic intervention targeting TLR2 still seems complicated, the strict position of TLR4 upstream of a number of pathogenic cytokines including IL-17 provides an interesting potential therapeutic target for rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/immunology , Germ-Free Life/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/microbiology , Disease Models, Animal , Germ-Free Life/genetics , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-17/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
9.
Genes (Basel) ; 12(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33430247

ABSTRACT

Recently, it has been shown that the community of gut microorganisms plays a crucial role in host performance with respect to parasite tolerance. Knowledge, however, is lacking on the role of the gut microbiome in mediating host tolerance after parasite re-exposure, especially considering multiple parasite infections. We here aimed to fill this knowledge gap by studying the role of the gut microbiome on tolerance in Daphnia magna upon multiple parasite species re-exposure. Additionally, we investigated the role of the host genotype in the interaction between the gut microbiome and the host phenotypic performance. A microbiome transplant experiment was performed in which three germ-free D. magna genotypes were exposed to a gut microbial inoculum and a parasite community treatment. The gut microbiome inocula were pre-exposed to the same parasite communities or a control treatment. Daphnia performance was monitored, and amplicon sequencing was performed to characterize the gut microbial community. Our experimental results showed that the gut microbiome plays no role in Daphnia tolerance upon parasite re-exposure. We did, however, find a main effect of the gut microbiome on Daphnia body size reflecting parasite specific responses. Our results also showed that it is rather the Daphnia genotype, and not the gut microbiome, that affected parasite-induced host mortality. Additionally, we found a role of the genotype in structuring the gut microbial community, both in alpha diversity as in the microbial composition.


Subject(s)
Daphnia/genetics , Gastrointestinal Microbiome/immunology , Genotype , Host-Parasite Interactions/genetics , Parasites/immunology , Animals , Body Size/genetics , Body Size/immunology , Daphnia/immunology , Daphnia/microbiology , Daphnia/parasitology , Germ-Free Life/genetics , Germ-Free Life/immunology , Host-Parasite Interactions/immunology
10.
Biochem J ; 423(1): 41-52, 2009 Sep 14.
Article in English | MEDLINE | ID: mdl-19614564

ABSTRACT

The social amoeba Dictyostelium discoideum has become established as a simple model for the examination of cell-cell interactions, and early studies suggested that shifts in glycosylation profiles take place during its life cycle. In the present study, we have applied HPLC and mass spectrometric methods to show that the major N-glycans in axenic cultures of the AX3 strain are oligomannosidic forms, most of which carry core fucose and/or intersecting and bisecting N-acetylglucosamine residues, including the major structure with the composition Man8GlcNAc4Fuc1. The postulated alpha1,3-linkage of the core fucose correlates with the cross-reactivity of Dictyostelium glycoproteins with a horseradish peroxidase antiserum; a corresponding core alpha1,3-fucosyltransferase activity capable of modifying oligomannosidic N-glycans was detected in axenic Dictyostelium extracts. The presence of fucose on the N-glycans and the reactivity to the antiserum, but not the fucosyltransferase activity, are abolished in the fucose-deficient HL250 strain. In later stages of development, N-glycans at the mound and culmination stages show a reduction in both the size and the degree of modification by intersecting/bisecting residues compared with mid-exponential phase cultures, consistent with the hypothesis that glycosidase and glycosyltransferase expression levels are altered during the slime mould life cycle.


Subject(s)
Dictyostelium/growth & development , Dictyostelium/metabolism , Fucose/metabolism , Polysaccharides/metabolism , Amino Acid Sequence , Animals , Carbohydrate Dehydrogenases/genetics , Carbohydrate Dehydrogenases/metabolism , Carbohydrate Sequence , Cells, Cultured , Dictyostelium/genetics , Dictyostelium/ultrastructure , Fucose/chemistry , Fucosyltransferases/metabolism , Germ-Free Life/genetics , Germ-Free Life/physiology , Glycosylation , Polysaccharides/chemistry
11.
Sci Rep ; 10(1): 7805, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385373

ABSTRACT

Transplantation of germ-free (GF) mice with microbiota from mice or humans stimulates the intestinal immune system in disparate ways. We transplanted a human microbiota into GF C57BL/6 mice and a murine C57BL/6 microbiota into GF C57BL/6 mice and Swiss-Webster (SW) mice. Mice were bred to produce an offspring generation. 56% of the Operational Taxonomic Units (OTUs) present in the human donor microbiota established in the recipient mice, whereas 81% of the C57BL/6 OTUs established in the recipient C57BL/6 and SW mice. Anti-inflammatory bacteria such as Faecalibacterium and Bifidobacterium from humans were not transferred to mice. Expression of immune-related intestinal genes was lower in human microbiota-mice and not different between parent and offspring generation. Expression of intestinal barrier-related genes was slightly higher in human microbiota-mice. Cytokines and chemokines measured in plasma were differentially present in human and mouse microbiota-mice. Minor differences in microbiota and gene expression were found between transplanted mice of different genetics. It is concluded that important immune-regulating bacteria are lost when transplanting microbiota from humans to C57BL/6 mice, and that the established human microbiota is a weak stimulator of the murine immune system. The results are important for study design considerations in microbiota transplantation studies involving immunological parameters.


Subject(s)
Bacteria/immunology , Gastrointestinal Microbiome/immunology , Immune System/microbiology , Transplants/microbiology , Animals , Bifidobacterium , Colon/microbiology , Gastrointestinal Microbiome/genetics , Germ-Free Life/genetics , Humans , Mice , Mice, Inbred C57BL
12.
FEBS Lett ; 593(6): 594-600, 2019 03.
Article in English | MEDLINE | ID: mdl-30779346

ABSTRACT

Intergenerational inheritance of transcriptional responses induced by low temperature rearing has recently been shown in Drosophila. Besides germline inheritance, fecal transfer experiments indirectly suggested that the acquired microbiome may also have contributed to the transcriptional responses in offspring. Here, we analyze expression data on inheritance of the cold-induced effects in conjunction with previously reported transcriptomic differences between flies with a microbiota or axenic flies and provide support for a contribution of the acquired microbiome to the offspring phenotype. Also, based on a similar analysis in conjunction with diet- and metabolism-related fly transcriptome data, we predicted and, then, experimentally confirmed that cold regulates triglyceride levels both inter- as well as trans-generationally.


Subject(s)
Adaptation, Physiological/genetics , Drosophila melanogaster/genetics , Epigenesis, Genetic , Genes, Insect , Inheritance Patterns , Triglycerides/metabolism , Animals , Cold Temperature , Drosophila melanogaster/metabolism , Drosophila melanogaster/microbiology , Feces/microbiology , Female , Germ-Free Life/genetics , Male , Microbiota/physiology , Phenotype , Transcriptome
13.
Sci Rep ; 8(1): 7267, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29740156

ABSTRACT

Oral microbiome is potentially correlated with many diseases, such as dental caries, periodontitis, oral cancer and some systemic diseases. Twin model, as an effective method for studying human microbiota, is widely used in research of relationship between oral microbiota and dental caries. However, there were few researches focusing on caries discordant twins. In this study, in vitro assays were conducted combined with 16S rRNA sequencing analysis on oral microbiota sampled from twins who presented discordant caries experience and mice model was developed as well. Results showed that oral microbiota from caries-active twin possessed higher metabolic activity and produced more lactic production. 16S rRNA sequencing analysis showed that more than 80% of family taxa could be transferred into gnotobiotic-mice. Key caries-associated genera were significantly different between twins and the same difference in genus level could be found in mice as well (p < 0.05). This study suggested that oral microbiota of twins could be distinguished from each other despite the similarities in genetic make-up, living environment, and lifestyle. The difference in microbiota was applied to develop a mice model which may facilitate the investigation of core microbiota of dental caries.


Subject(s)
Dental Caries/microbiology , Microbiota/genetics , Mouth/microbiology , Periodontitis/genetics , Animals , Bacteria/genetics , Bacteria/pathogenicity , Dental Caries/genetics , Dental Caries/pathology , Dental Plaque/genetics , Dental Plaque/microbiology , Germ-Free Life/genetics , Humans , Mice , Periodontitis/microbiology , RNA, Ribosomal, 16S/genetics , Twins, Monozygotic/genetics
14.
Sci Rep ; 8(1): 10745, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30013139

ABSTRACT

Germ-free (GF) pigs have clear microbiological backgrounds, and are extensively used as large animal models in the biomedical sciences. However, investigations of the transcriptomic differences between GF and cesarean-derived conventional (CV) piglets are limited. To improve our understanding of GF pigs, and to increase the utility of pigs as an alternative non-rodent model, we used RNA sequencing to profile gene expression in five tissues (the oral mucosae, jejunum, colon, liver, and spleen) of four male GF piglets and four male CV piglets from the same litter. We identified 14 genes that were differentially expressed in all five tissues. Seven of these common differentially expressed genes (DEGs) were interferon-inducible genes, and all 14 were consistently downregulated in the GF piglets as compared to the CV piglets. Compared to the other tissues tested, the expression of transcription factors (TFs) in the colon was most affected by the absence of a microbiota. The expression patterns of immune-related genes were downregulated in the GF piglets as compared to the CV piglets, indicating that the intestinal microbiota influenced gene expression in other tissues besides the gut. Gene Ontology (GO) analysis indicated that, in pigs, the intestinal microbiota affected the expression of genes related to immune system function and development.


Subject(s)
Gastrointestinal Microbiome/immunology , Germ-Free Life/genetics , Immune System/growth & development , RNA, Messenger/metabolism , Transcriptome/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Animals, Newborn/immunology , Colon/metabolism , Down-Regulation/immunology , Gene Expression Profiling , Jejunum/metabolism , Liver/metabolism , Male , Models, Animal , Mouth Mucosa/metabolism , Sequence Analysis, RNA , Spleen/metabolism , Swine/genetics , Swine/growth & development , Swine/immunology , Transcriptome/immunology
15.
BMC Genomics ; 8: 215, 2007 Jul 05.
Article in English | MEDLINE | ID: mdl-17615075

ABSTRACT

BACKGROUND: To gain insight into host-microbe interactions in a piglet model, a functional genomics approach was used to address the working hypothesis that transcriptionally regulated genes associated with promoting epithelial barrier function are activated as a defensive response to the intestinal microbiota. Cesarean-derived germfree (GF) newborn piglets were colonized with adult swine feces, and villus and crypt epithelial cell transcriptomes from colonized and GF neonatal piglets were compared using laser-capture microdissection and high-density porcine oligonucleotide microarray technology. RESULTS: Consistent with our hypothesis, resident microbiota induced the expression of genes contributing to intestinal epithelial cell turnover, mucus biosynthesis, and priming of the immune system. Furthermore, differential expression of genes associated with antigen presentation (pan SLA class I, B2M, TAP1 and TAPBP) demonstrated that microbiota induced immune responses using a distinct regulatory mechanism common for these genes. Specifically, gene network analysis revealed that microbial colonization activated both type I (IFNAR) and type II (IFNGR) interferon receptor mediated signaling cascades leading to enhanced expression of signal transducer and activator of transcription 1 (STAT1), STAT2 and IFN regulatory factor 7 (IRF7) transcription factors and the induction of IFN-inducible genes as a reflection of intestinal epithelial inflammation. In addition, activated RNA expression of NF-kappa-B inhibitor alpha (NFkappaBIA; a.k.a I-kappa-B-alpha, IKBalpha) and toll interacting protein (TOLLIP), both inhibitors of inflammation, along with downregulated expression of the immunoregulatory transcription factor GATA binding protein-1 (GATA1) is consistent with the maintenance of intestinal homeostasis. CONCLUSION: This study supports the concept that the intestinal epithelium has evolved to maintain a physiological state of inflammation with respect to continuous microbial exposure, which serves to sustain a tight intestinal barrier while preventing overt inflammatory responses that would compromise barrier function.


Subject(s)
Gene Expression Profiling , Germ-Free Life/genetics , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Proliferation , Electron Transport/genetics , Gene Regulatory Networks , Host-Parasite Interactions/genetics , Immunity, Mucosal/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Small/immunology , Intestine, Small/microbiology , Metabolic Networks and Pathways/genetics , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Signal Transduction/genetics , Swine , Transcription, Genetic
16.
Sci Transl Med ; 8(366): 366ra164, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27881825

ABSTRACT

To model how interactions among enteropathogens and gut microbial community members contribute to undernutrition, we colonized gnotobiotic mice fed representative Bangladeshi diets with sequenced bacterial strains cultured from the fecal microbiota of two 24-month-old Bangladeshi children: one healthy and the other underweight. The undernourished donor's bacterial collection contained an enterotoxigenic Bacteroides fragilis strain (ETBF), whereas the healthy donor's bacterial collection contained two nontoxigenic strains of B. fragilis (NTBF). Analyses of mice harboring either the unmanipulated culture collections or systematically manipulated versions revealed that ETBF was causally related to weight loss in the context of its native community but not when introduced into the healthy donor's community. This phenotype was transmissible from the dams to their offspring and was associated with derangements in host energy metabolism manifested by impaired tricarboxylic acid cycle activity and decreased acyl-coenzyme A utilization. NTBF reduced ETBF's expression of its enterotoxin and mitigated the effects of ETBF on the transcriptomes of other healthy donor community members. These results illustrate how intraspecific (ETBF-NTBF) and interspecific interactions influence the effects of harboring B. fragilis.


Subject(s)
Child Nutrition Disorders/microbiology , Gastrointestinal Microbiome , Animals , Bacteroides fragilis/isolation & purification , Bangladesh , Cachexia/microbiology , Child, Preschool , Diet , Disease Models, Animal , Feces/microbiology , Female , Gene Expression Regulation, Bacterial , Germ-Free Life/genetics , Humans , Infant , Male , Mice , Phenotype
17.
Vet Immunol Immunopathol ; 171: 7-16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26964712

ABSTRACT

Toll-like receptors (TLRs), key initiators of innate immune responses, recognize antigens and are essential in linking innate and adaptive immune responses. Misrecognition and over-stimulation/expression of TLRs may contribute to the development of chronic inflammatory diseases and autoimmune diseases. However, appropriate and mature TLR responses are associated with the establishment of resistance against some infectious diseases. In this study, we assessed the mRNA expression profile of TLRs 1-10 in splenic and ileal mononuclear cells (MNCs) and dendritic cells (DCs) of germ-free (GF) and conventional pigs at different ages. We found that the TLR mRNA expression profiles were distinct between GF and conventional pigs. The expression profiles were also significantly different between splenic and ileal MNCs/DCs. Comparison of the TLR expression profiles in GF and conventional newborn and young pigs demonstrated that exposure to commensal microbiota may play a more important role than age in TLR mRNA expression profiles. To our knowledge, this is the first report that systematically assesses porcine TLRs 1-10 mRNA expression profiles in MNCs and DCs from GF and conventional pigs at different ages. These results further highlighted that the commensal microbiota of neonates play a critical role through TLR signaling in the development of systemic and mucosal immune systems.


Subject(s)
Aging/metabolism , Toll-Like Receptors/metabolism , Aging/genetics , Animals , Dendritic Cells/metabolism , Gene Expression Profiling , Germ-Free Life/genetics , Ileum/cytology , Ileum/metabolism , Leukocytes, Mononuclear/metabolism , Microbiota/immunology , RNA, Messenger/metabolism , Spleen/cytology , Spleen/metabolism , Swine , Toll-Like Receptors/genetics
18.
ILAR J ; 56(2): 169-78, 2015.
Article in English | MEDLINE | ID: mdl-26323627

ABSTRACT

The gastrointestinal (GI) microbiota forms a mutualistic relationship with the host through complex and dynamic interactions. Because of the complexity and interindividual variation of the GI microbiota, investigating how members of the microbiota interact with each other, as well as with the host, is daunting. The altered Schaedler flora (ASF) is a model community of eight microorganisms that was developed by R.P. Orcutt and has been in use since the late 1970s. The eight microorganisms composing the ASF were all derived from mice, can be cultured in vitro, and are stably passed through multiple generations (at least 15 years or more by the authors) in gnotobiotic mice continually bred in isolator facilities. With the limitations associated with conventional, mono- or biassociated, and germfree mice, use of mice colonized with a consortium of known bacteria that naturally inhabit the murine gut offers a powerful system to investigate mechanisms governing host-microbiota relationships, and how members of the GI microbiota interact with one another. The ASF community offers significant advantages to study homeostatic as well as disease-related interactions by taking advantage of a well-defined, limited community of microorganisms. For example, quantification and spatial distribution of individual members, microbial genetic manipulation, genomic-scale analysis, and identification of microorganism-specific host immune responses are all achievable using the ASF model. This review compiles highlights associated with the 37-year history of the ASF, including descriptions of its continued use in biomedical research to elucidate the complexities of host-microbiome interactions in health and disease.


Subject(s)
Gastrointestinal Microbiome/physiology , Animals , Bacteria/metabolism , Gastrointestinal Microbiome/genetics , Germ-Free Life/genetics , Germ-Free Life/physiology , Humans , Mice
19.
ILAR J ; 56(2): 179-91, 2015.
Article in English | MEDLINE | ID: mdl-26323628

ABSTRACT

The intestinal microbiota has long been known to play an important role in the maintenance of health. In addition, alterations of the intestinal microbiota have recently been associated with a range of immune-mediated and metabolic disorders. Characterizing the composition and functionality of the intestinal microbiota, unravelling relevant microbe-host interactions, and identifying disease-relevant microbes are therefore currently of major interest in scientific and medical communities. Experimental animal models for the respective diseases of interest are pivotal in order to address functional questions on microbe-host interaction and to clarify the clinical relevance of microbiome alterations associated with disease initiation and development. This review presents an overview of the outcomes of highly sophisticated experimental studies on microbe-host interaction in animal models of inflammatory diseases, with a focus on inflammatory bowel disease (IBD). We will address the advantages and drawbacks of analyzing microbe-host interaction in complex colonized animal models compared with gnotobiotic animal models using monoassociation, simplified microbial consortia (SMC), or microbial humanization.


Subject(s)
Gastrointestinal Microbiome/physiology , Animals , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Germ-Free Life/genetics , Germ-Free Life/physiology , Host-Pathogen Interactions , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology
20.
Acta Trop ; 83(1): 37-42, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12062791

ABSTRACT

For the first time, we have reported the establishment and serial propagation of an axenic culture of Leishmania chagasi amastigote-like forms. Parasites were characterized by microscopic evaluation and by the expression of two stage-specific genes, A2 and Ldccys2 amastigote-specific cysteine protease. The differentiated amastigote-like forms were maintained by serial cultivation.


Subject(s)
Leishmania/growth & development , Animals , Cells, Cultured , Gene Expression Regulation , Germ-Free Life/genetics , Leishmania/genetics
SELECTION OF CITATIONS
SEARCH DETAIL