Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Benef Microbes ; 12(6): 583-599, 2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34550056

ABSTRACT

The serotonin transporter (SERT) readily takes up serotonin (5-HT), thereby regulating the availability of 5-HT within the intestine. In the absence of SERT, 5-HT remains in the interstitial space and has the potential to aberrantly activate the many 5-HT receptors distributed on the epithelium, immune cells and enteric neurons. Perturbation of SERT is common in many gastrointestinal disorders as well as mouse models of colitis. Select commensal microbes regulate intestinal SERT levels, but the mechanism of this regulation is poorly understood. Additionally, ethanol upregulates SERT in the brain and dendritic cells, but its effects in the intestine have never been examined. We report that the intestinal commensal microbe Limosilactobacillus (previously classified as Lactobacillus) reuteri ATCC PTA 6475 secretes 83.4 mM ethanol. Consistent with the activity of L. reuteri alcohol dehydrogenases, we found that L. reuteri tolerated various levels of ethanol. Application of L. reuteri conditioned media or exogenous ethanol to human colonic T84 cells was found to upregulate SERT at the level of mRNA. A 4-(4-(dimethylamino) phenyl)-1-methylpyridinium (APP+) uptake assay confirmed the functional activity of SERT. These findings were mirrored in mouse colonic organoids, where L. reuteri metabolites and ethanol were found to upregulate SERT at the apical membrane. Finally, in a trinitrobenzene sulphonic acid model of acute colitis, we observed that mice treated with L. reuteri maintained SERT at the colon membrane compared with mice receiving phosphate buffered saline vehicle control. These data suggest that L. reuteri metabolites, including ethanol, can upregulate SERT and may be beneficial for maintaining intestinal homeostasis with respect to serotonin signalling.


Subject(s)
Colitis , Intestinal Mucosa/metabolism , Limosilactobacillus reuteri , Serotonin Plasma Membrane Transport Proteins , Animals , Colitis/therapy , Ethanol , Limosilactobacillus reuteri/chemistry , Mice , Serotonin , Serotonin Plasma Membrane Transport Proteins/metabolism
2.
Antonie Van Leeuwenhoek ; 105(3): 579-92, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24488273

ABSTRACT

The gut microbiome may modulate intestinal immunity by luminal conversion of dietary amino acids to biologically active signals. The model probiotic organism Lactobacillus reuteri ATCC PTA 6475 is indigenous to the human microbiome, and converts the amino acid L-histidine to the biogenic amine, histamine. Histamine suppresses tumor necrosis factor (TNF) production by human myeloid cells and is a product of L-histidine decarboxylation, which is a proton-facilitated reaction. A transposon mutagenesis strategy was developed based on a single-plasmid nisin-inducible Himar1 transposase/transposon delivery system for L. reuteri. A highly conserved proton-chloride antiporter gene (eriC), a gene widely present in the gut microbiome was discovered by Himar1 transposon (Tn)-mutagenesis presented in this study. Genetic inactivation of eriC by transposon insertion and genetic recombineering resulted in reduced ability of L. reuteri to inhibit TNF production by activated human myeloid cells, diminished histamine production by the bacteria and downregulated expression of histidine decarboxylase cluster genes compared to those of WT 6475. EriC belongs to a large family of ion transporters that includes chloride channels and proton-chloride antiporters and may facilitate the availability of protons for the decarboxylation reaction, resulting in histamine production by L. reuteri. This report leverages the tools of bacterial genetics for probiotic gene discovery. The findings highlight the widely conserved nature of ion transporters in bacteria and how ion transporters are coupled with amino acid decarboxylation and contribute to microbiome-mediated immunomodulation.


Subject(s)
Antiporters/genetics , Antiporters/metabolism , DNA Transposable Elements , Histamine/biosynthesis , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/metabolism , Mutagenesis , Biological Transport , Chlorides/metabolism , Gene Expression , Gene Expression Regulation, Bacterial , Gene Order , Gene Silencing , Genetic Vectors/genetics , Histidine Decarboxylase/genetics , Limosilactobacillus reuteri/drug effects , Multigene Family , Mutagenesis, Insertional , Probiotics , Promoter Regions, Genetic , Protons , Tumor Necrosis Factors/pharmacology
3.
J Bacteriol ; 195(24): 5567-76, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123819

ABSTRACT

Human microbiome-derived strains of Lactobacillus reuteri potently suppress proinflammatory cytokines like human tumor necrosis factor (TNF) by converting the amino acid l-histidine to the biogenic amine histamine. Histamine suppresses mitogen-activated protein (MAP) kinase activation and cytokine production by signaling via histamine receptor type 2 (H2) on myeloid cells. Investigations of the gene expression profiles of immunomodulatory L. reuteri ATCC PTA 6475 highlighted numerous genes that were highly expressed during the stationary phase of growth, when TNF suppression is most potent. One such gene was found to be a regulator of genes involved in histidine-histamine metabolism by this probiotic species. During the course of these studies, this gene was renamed the Lactobacillus reuteri-specific immunoregulatory (rsiR) gene. The rsiR gene is essential for human TNF suppression by L. reuteri and expression of the histidine decarboxylase (hdc) gene cluster on the L. reuteri chromosome. Inactivation of rsiR resulted in diminished TNF suppression in vitro and reduced anti-inflammatory effects in vivo in a trinitrobenzene sulfonic acid (TNBS)-induced mouse model of acute colitis. A L. reuteri strain lacking an intact rsiR gene was unable to suppress colitis and resulted in greater concentrations of serum amyloid A (SAA) in the bloodstream of affected animals. The PhdcAB promoter region targeted by rsiR was defined by reporter gene experiments. These studies support the presence of a regulatory gene, rsiR, which modulates the expression of a gene cluster known to mediate immunoregulation by probiotics at the transcriptional level. These findings may point the way toward new strategies for controlling gene expression in probiotics by dietary interventions or microbiome manipulation.


Subject(s)
Cytokines/antagonists & inhibitors , Gene Expression Regulation, Bacterial , Histamine/metabolism , Immunomodulation , Limosilactobacillus reuteri/immunology , Transcription Factors/metabolism , Animals , Artificial Gene Fusion , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Disease Models, Animal , Genes, Reporter , Humans , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/isolation & purification , Limosilactobacillus reuteri/metabolism , Mice , Microbiota , Promoter Regions, Genetic , Transcription, Genetic , Trinitrobenzenesulfonic Acid/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...