Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.172
Filter
Add more filters

Publication year range
1.
Cell ; 186(9): 1846-1862.e26, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37028428

ABSTRACT

The use of probiotics by cancer patients is increasing, including among those undergoing immune checkpoint inhibitor (ICI) treatment. Here, we elucidate a critical microbial-host crosstalk between probiotic-released aryl hydrocarbon receptor (AhR) agonist indole-3-aldehyde (I3A) and CD8 T cells within the tumor microenvironment that potently enhances antitumor immunity and facilitates ICI in preclinical melanoma. Our study reveals that probiotic Lactobacillus reuteri (Lr) translocates to, colonizes, and persists within melanoma, where via its released dietary tryptophan catabolite I3A, it locally promotes interferon-γ-producing CD8 T cells, thereby bolstering ICI. Moreover, Lr-secreted I3A was both necessary and sufficient to drive antitumor immunity, and loss of AhR signaling within CD8 T cells abrogated Lr's antitumor effects. Further, a tryptophan-enriched diet potentiated both Lr- and ICI-induced antitumor immunity, dependent on CD8 T cell AhR signaling. Finally, we provide evidence for a potential role of I3A in promoting ICI efficacy and survival in advanced melanoma patients.


Subject(s)
Limosilactobacillus reuteri , Melanoma , Tumor Microenvironment , Humans , Diet , Immune Checkpoint Inhibitors , Limosilactobacillus reuteri/metabolism , Melanoma/therapy , Tryptophan/metabolism , CD8-Positive T-Lymphocytes/immunology , Receptors, Aryl Hydrocarbon/agonists
2.
Cell ; 184(7): 1740-1756.e16, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33705688

ABSTRACT

The core symptoms of many neurological disorders have traditionally been thought to be caused by genetic variants affecting brain development and function. However, the gut microbiome, another important source of variation, can also influence specific behaviors. Thus, it is critical to unravel the contributions of host genetic variation, the microbiome, and their interactions to complex behaviors. Unexpectedly, we discovered that different maladaptive behaviors are interdependently regulated by the microbiome and host genes in the Cntnap2-/- model for neurodevelopmental disorders. The hyperactivity phenotype of Cntnap2-/- mice is caused by host genetics, whereas the social-behavior phenotype is mediated by the gut microbiome. Interestingly, specific microbial intervention selectively rescued the social deficits in Cntnap2-/- mice through upregulation of metabolites in the tetrahydrobiopterin synthesis pathway. Our findings that behavioral abnormalities could have distinct origins (host genetic versus microbial) may change the way we think about neurological disorders and how to treat them.


Subject(s)
Gastrointestinal Microbiome , Locomotion , Social Behavior , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Biopterins/analogs & derivatives , Biopterins/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials , Fecal Microbiota Transplantation , Feces/microbiology , Limosilactobacillus reuteri/metabolism , Limosilactobacillus reuteri/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/microbiology , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/therapy , Principal Component Analysis , Psychomotor Agitation/pathology , Synaptic Transmission
3.
Cell ; 165(7): 1762-1775, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27315483

ABSTRACT

Maternal obesity during pregnancy has been associated with increased risk of neurodevelopmental disorders, including autism spectrum disorder (ASD), in offspring. Here, we report that maternal high-fat diet (MHFD) induces a shift in microbial ecology that negatively impacts offspring social behavior. Social deficits and gut microbiota dysbiosis in MHFD offspring are prevented by co-housing with offspring of mothers on a regular diet (MRD) and transferable to germ-free mice. In addition, social interaction induces synaptic potentiation (LTP) in the ventral tegmental area (VTA) of MRD, but not MHFD offspring. Moreover, MHFD offspring had fewer oxytocin immunoreactive neurons in the hypothalamus. Using metagenomics and precision microbiota reconstitution, we identified a single commensal strain that corrects oxytocin levels, LTP, and social deficits in MHFD offspring. Our findings causally link maternal diet, gut microbial imbalance, VTA plasticity, and behavior and suggest that probiotic treatment may relieve specific behavioral abnormalities associated with neurodevelopmental disorders. VIDEO ABSTRACT.


Subject(s)
Autism Spectrum Disorder/microbiology , Diet, High-Fat , Gastrointestinal Microbiome , Obesity/complications , Social Behavior , Animals , Dysbiosis/physiopathology , Female , Germ-Free Life , Housing, Animal , Limosilactobacillus reuteri , Male , Mice , Mice, Inbred C57BL , Oxytocin/analysis , Oxytocin/metabolism , Pregnancy , Ventral Tegmental Area
4.
Nature ; 607(7919): 563-570, 2022 07.
Article in English | MEDLINE | ID: mdl-35831502

ABSTRACT

Gut commensal bacteria with the ability to translocate across the intestinal barrier can drive the development of diverse immune-mediated diseases1-4. However, the key factors that dictate bacterial translocation remain unclear. Recent studies have revealed that gut microbiota strains can adapt and evolve throughout the lifetime of the host5-9, raising the possibility that changes in individual commensal bacteria themselves over time may affect their propensity to elicit inflammatory disease. Here we show that within-host evolution of the model gut pathobiont Enterococcus gallinarum facilitates bacterial translocation and initiation of inflammation. Using a combination of in vivo experimental evolution and comparative genomics, we found that E. gallinarum diverges into independent lineages adapted to colonize either luminal or mucosal niches in the gut. Compared with ancestral and luminal E. gallinarum, mucosally adapted strains evade detection and clearance by the immune system, exhibit increased translocation to and survival within the mesenteric lymph nodes and liver, and induce increased intestinal and hepatic inflammation. Mechanistically, these changes in bacterial behaviour are associated with non-synonymous mutations or insertion-deletions in defined regulatory genes in E. gallinarum, altered microbial gene expression programs and remodelled cell wall structures. Lactobacillus reuteri also exhibited broadly similar patterns of divergent evolution and enhanced immune evasion in a monocolonization-based model of within-host evolution. Overall, these studies define within-host evolution as a critical regulator of commensal pathogenicity that provides a unique source of stochasticity in the development and progression of microbiota-driven disease.


Subject(s)
Bacteria , Bacterial Translocation , Biological Evolution , Gastrointestinal Microbiome , Liver , Bacteria/genetics , Bacteria/immunology , Bacteria/pathogenicity , Bacterial Translocation/genetics , Cell Wall/genetics , Enterococcus/genetics , Enterococcus/immunology , Gastrointestinal Microbiome/genetics , Genomics , Host-Pathogen Interactions/immunology , Humans , Inflammation/microbiology , Inflammation/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/immunology , Liver/microbiology , Liver/pathology , Lymph Nodes/microbiology , Mutation , Stochastic Processes , Symbiosis/genetics , Symbiosis/immunology
5.
Nature ; 585(7823): 102-106, 2020 09.
Article in English | MEDLINE | ID: mdl-32848245

ABSTRACT

Accumulating evidence indicates that gut microorganisms have a pathogenic role in autoimmune diseases, including in multiple sclerosis1. Studies of experimental autoimmune encephalomyelitis (an animal model of multiple sclerosis)2,3, as well as human studies4-6, have implicated gut microorganisms in the development or severity of multiple sclerosis. However, it remains unclear how gut microorganisms act on the inflammation of extra-intestinal tissues such as the spinal cord. Here we show that two distinct signals from gut microorganisms coordinately activate autoreactive T cells in the small intestine that respond specifically to myelin oligodendrocyte glycoprotein (MOG). After induction of experimental autoimmune encephalomyelitis in mice, MOG-specific CD4+ T cells are observed in the small intestine. Experiments using germ-free mice that were monocolonized with microorganisms from the small intestine demonstrated that a newly isolated strain in the family Erysipelotrichaceae acts similarly to an adjuvant to enhance the responses of T helper 17 cells. Shotgun sequencing of the contents of the small intestine revealed a strain of Lactobacillus reuteri that possesses peptides that potentially mimic MOG. Mice that were co-colonized with these two strains showed experimental autoimmune encephalomyelitis symptoms that were more severe than those of germ-free or monocolonized mice. These data suggest that the synergistic effects that result from the presence of these microorganisms should be considered in the pathogenicity of multiple sclerosis, and that further study of these microorganisms may lead to preventive strategies for this disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/microbiology , Gastrointestinal Microbiome/immunology , Inflammation/pathology , Spinal Cord/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Germ-Free Life , Inflammation/immunology , Intestine, Small/immunology , Intestine, Small/microbiology , Intestine, Small/pathology , Limosilactobacillus reuteri/chemistry , Limosilactobacillus reuteri/immunology , Limosilactobacillus reuteri/pathogenicity , Male , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/microbiology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/chemistry , Myelin-Oligodendrocyte Glycoprotein/immunology , Spinal Cord/immunology , Th17 Cells/immunology , Th17 Cells/pathology
6.
FASEB J ; 38(2): e23383, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38197892

ABSTRACT

Mastitis is the most frequent disease of cows and has well-recognized detrimental effects on animal wellbeing and dairy farm profitability. With the advent of the postantibiotic era, alternative antibiotic agents, especially probiotics, have received increasing attention in the treatment of mastitis. Based on research showing that Lactobacillus reuteri (L. reuteri) has anti-inflammatory effects, this study explored the protective effects and mechanisms of L. reuteri against mastitis induced by Staphylococcus aureus (S. aureus) in mice. First, mice with S. aureus-induced mastitis were orally administered L. reuteri, and the inflammatory response in the mammary gland was observed. The results showed that L. reuteri significantly inhibited S. aureus-induced mastitis. Moreover, the concentration of oxytocin (OT) and protein expression of oxytocin receptor (OTR) were measured, and inhibition of OTR or vagotomy reversed the protective effect of L. reuteri or its culture supernatant (LCS) on S. aureus-induced mastitis. In addition, in mouse mammary epithelial cells (MMECs), OT inhibited the inflammation induced by S. aureus by inhibiting the protein expression of OTR. It was suggested that L. reuteri protected against S. aureus-induced mastitis by releasing OT. Furthermore, microbiological analysis showed that the composition of the microbiota was altered, and the relative abundance of Lactobacillus was significantly increased in gut and mammary gland after treatment with L. reuteri or LCS. In conclusion, our study found the L. reuteri inhibited the mastitis-induced by S. aureus via promoting the release of OT, and treatment with L. reuteri increased the abundance of Lactobacillus in both gut and mammary gland.


Subject(s)
Gastrointestinal Microbiome , Limosilactobacillus reuteri , Mastitis , Staphylococcal Infections , Female , Humans , Animals , Cattle , Mice , Oxytocin/pharmacology , Oxytocin/therapeutic use , Staphylococcus aureus , Mastitis/therapy , Receptors, Oxytocin , Lactobacillus
7.
Am J Physiol Gastrointest Liver Physiol ; 327(2): G140-G153, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38780469

ABSTRACT

Treatments of colitis, inflammation of the intestine, rely on induction of immune suppression associated with systemic adverse events, including recurrent infections. This treatment strategy is specifically problematic in the increasing population of patients with cancer with immune checkpoint inhibitor (ICI)-induced colitis, as immune suppression also interferes with the ICI-treatment response. Thus, there is a need for local-acting treatments that reduce inflammation and enhance intestinal healing. Here, we investigated the effect and safety of bacterial delivery of short-lived immunomodulating chemokines to the inflamed intestine in mice with colitis. Colitis was induced by dextran sulfate sodium (DSS) alone or in combination with ICI (anti-PD1 and anti-CTLA-4), and Limosilactobacillus reuteri R2LC (L. reuteri R2LC) genetically modified to express the chemokine CXCL12-1α (R2LC_CXCL12, emilimogene sigulactibac) was given perorally. In addition, the pharmacology and safety of the formulated drug candidate, ILP100-Oral, were evaluated in rabbits. Peroral CXCL12-producing L. reuteri R2LC significantly improved colitis symptoms already after 2 days in mice with overt DSS and ICI-induced colitis, which in benchmarking experiments was demonstrated to be superior to treatments with anti-TNF-α, anti-α4ß7, and corticosteroids. The mechanism of action involved chemokine delivery to Peyer's patches (PPs), confirmed by local CXCR4 signaling, and increased numbers of colonic, regulatory immune cells expressing IL-10 and TGF-ß1. No systemic exposure or engraftment could be detected in mice, and product feasibility, pharmacology, and safety were confirmed in rabbits. In conclusion, peroral CXCL12-producing L. reuteri R2LC efficiently ameliorates colitis, enhances mucosal healing, and has a favorable safety profile.NEW & NOTEWORTHY Colitis symptoms are efficiently reduced by peroral administration of probiotic bacteria genetically modified to deliver CXCL12 locally to the inflamed intestine in several mouse models.


Subject(s)
Chemokine CXCL12 , Colitis , Dextran Sulfate , Disease Models, Animal , Limosilactobacillus reuteri , Animals , Colitis/immunology , Colitis/chemically induced , Colitis/drug therapy , Colitis/therapy , Colitis/metabolism , Mice , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Administration, Oral , Rabbits , Probiotics/administration & dosage , Mice, Inbred C57BL , Female , Colon/metabolism , Colon/microbiology , Colon/immunology , Male
8.
Appl Environ Microbiol ; 90(7): e0024724, 2024 07 24.
Article in English | MEDLINE | ID: mdl-38888338

ABSTRACT

The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE: By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.


Subject(s)
Bifidobacterium , Extracellular Vesicles , Limosilactobacillus reuteri , Probiotics , Limosilactobacillus reuteri/metabolism , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/growth & development , Extracellular Vesicles/metabolism , Humans , Bifidobacterium/metabolism , Bifidobacterium/genetics , Bifidobacterium/growth & development
9.
Appl Environ Microbiol ; 90(7): e0081424, 2024 07 24.
Article in English | MEDLINE | ID: mdl-38888337

ABSTRACT

Tetrahydrofolate is a cofactor involved in C1 metabolism including biosynthesis pathways for adenine and serine. In the classical tetrahydrofolate biosynthesis pathway, the steps removing three phosphate groups from the precursor 7,8-dihydroneopterin triphosphate (DHNTP) remain unclear in many bacteria. DHNTP pyrophosphohydrolase hydrolyzes pyrophosphate from DHNTP and produces 7,8-dihydroneopterin monophosphate. Although two structurally distinct DHNTP pyrophosphohydrolases have been identified in the intestinal bacteria Lactococcus lactis and Escherichia coli, the distribution of their homologs is limited. Here, we aimed to identify a third DHNTP pyrophosphohydrolase gene in the intestinal lactic acid bacterium Limosilactobacillus reuteri. In a gene operon including genes involved in dihydrofolate biosynthesis, we focused on the lreu_1276 gene, annotated as Ham1 family protein or XTP/dITP diphosphohydrolase, as a candidate encoding DHNTP pyrophosphohydrolase. The Lreu_1276 recombinant protein was prepared using E. coli and purified. Biochemical analyses of the reaction product revealed that the Lreu_1276 protein displays significant pyrophosphohydrolase activity toward DHNTP. The optimal reaction temperature and pH were 35°C and around 7, respectively. Substrate specificity was relatively strict among 17 tested compounds. Although previously characterized DHNTP pyrophosphohydrolases prefer Mg2+, the Lreu_1276 protein exhibited maximum activity in the presence of Mn2+, with a specific activity of 28.2 ± 2.0 µmol min-1 mg-1 in the presence of 1 mM Mn2+. The three DHNTP pyrophosphohydrolases do not share structural similarity to one another, and the distribution of their homologs does not overlap, implying that the Lreu_1276 protein represents a third structurally novel DHNTP pyrophosphohydrolase in bacteria. IMPORTANCE: The identification of a structurally novel DHNTP pyrophosphohydrolase in L. reuteri provides valuable information in understanding tetrahydrofolate biosynthesis in bacteria that possess lreu_1276 homologs. Interestingly, however, even with the identification of a third family of DHNTP pyrophosphohydrolases, there are still a number of bacteria that do not harbor homologs for any of the three genes while possessing other genes involved in the biosynthesis of the pterin ring structure. This suggests the presence of an unrecognized DHNTP pyrophosphohydrolase gene in bacteria. As humans do not harbor DHNTP pyrophosphohydrolase, the high structural diversity of enzymes responsible for a reaction in tetrahydrofolate biosynthesis may provide an advantage in designing inhibitors targeting a specific group of bacteria in the intestinal microbiota.


Subject(s)
Bacterial Proteins , Limosilactobacillus reuteri , Pyrophosphatases , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/enzymology , Limosilactobacillus reuteri/metabolism , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Pterins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Neopterin/analogs & derivatives
10.
Microb Pathog ; 188: 106541, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38224920

ABSTRACT

Diarrhea is a prevalent health issue in farm animals and poses a significant challenge to the progress of animal husbandry. Recent evidence suggested that probiotics can alleviate diarrhea by maintaining gut microbial balance and enhancing the integrity of the intestinal barrier. However, there is a scarcity of studies investigating the efficacy of equine Lactobacillus reuteri in relieving E. coli-induced diarrhea. Hence, this study aimed to examine the potential of equine-derived Lactobacillus reuteri in alleviating E. coli diarrhea from the perspective of gut microbiota. Results demonstrated that supplementation of Lactobacillus reuteri had the potential to alleviate diarrhea induced by E. coli infection and restore the decline of tight junction genes, such as Claudin-1 and ZO-1. Additionally, Lactobacillus reuteri supplementation can restore the expression of inflammatory factors (IL-6, IL-10, TNF-α, and IFN-γ) and reduce colon inflammatory damage. Diversity analysis, based on amplicon sequencing, revealed a significant reduction in the diversity of gut microbiota during E. coli-induced diarrhea. Moreover, there were notable statistical differences in the composition and structure of gut microbiota among the different treatment groups. E. coli could induce gut microbial dysbiosis by decreasing the abundance of beneficial bacteria, including Lactobacillus, Bifidobacterium, Ligilactobacillus, Enterorhabdus, and Lachnospiraceae_UCG_001, in comparison to the control group. Conversely, supplementation with Lactobacillus reuteri could restore the abundance of beneficial bacteria and increase the diversity of the gut microbiota, thereby reshaping gut microbiota. Additionally, we also observed that supplementation with Lactobacillus reuteri alone improved the gut microbial composition and structure. In summary, the findings suggest that Lactobacillus reuteri can alleviate E. coli-induced diarrhea by preserving the integrity of the intestinal barrier and modulating the composition of the gut microbiota. These results not only contribute to understanding of the mechanism underlying the beneficial effects of Lactobacillus reuteri in relieving diarrhea, but also provide valuable insights for the development of probiotic products aimed at alleviating diarrheal diseases.


Subject(s)
Escherichia coli Infections , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Horses , Animals , Escherichia coli , Diarrhea/therapy , Lactobacillus , Escherichia coli Infections/therapy , Escherichia coli Infections/veterinary , Probiotics/therapeutic use , Probiotics/pharmacology
11.
FASEB J ; 37(5): e22895, 2023 05.
Article in English | MEDLINE | ID: mdl-37000564

ABSTRACT

Lactobacillus species is one of the most commonly used probiotics with a wide range of health-promoting effects, and beneficial effects of the surface protein of the lactobacillus could potentially be involved in the action of probiotics in the gastrointestinal tract. In this study, the anti-inflammatory effect of LPxTG-motif surface protein (LMP) derived from Limosilactobacillus reuteri SH 23 was assessed using a mouse model of colitis induced by dextran sodium sulfate (DSS). The results showed that LMP has the inhibition properties upon the DSS-induced ulcerative colitis of mice via the MAPK-dependent NF-κB pathway. The inflammatory factors TNF-α and IL-6 were inhibited, and the IL-10 secretion was enhanced in the LMP-treated DSS mice model. Furthermore, the diversity of the intestinal microbiota bacteria in this treated group was also influenced, including the increase in the abundance of Lactobacillus and Akkermansia genus in the LMP-treated mice groups, and there is a positive correlation between the IL-10 cytokines with the changes in the intestinal microbiota Lactobacillus and Akkermansia. Therefore, LMP derived from the Limosilactobacillus reuteri SH 23 has the potential to alleviate inflammatory diseases through the balance of the intestinal flora with the inhibition of the inflammatory factors in the NF-κB pathway.


Subject(s)
Colitis, Ulcerative , Colitis , Limosilactobacillus reuteri , Animals , Mice , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Interleukin-10/metabolism , NF-kappa B/metabolism , Membrane Proteins/metabolism , Colitis/metabolism , Disease Models, Animal , Dextran Sulfate/toxicity , Mice, Inbred C57BL , Colon/metabolism
12.
Biotechnol Bioeng ; 121(1): 317-328, 2024 01.
Article in English | MEDLINE | ID: mdl-37747698

ABSTRACT

The lactic acid bacterium Limosilactobacillus reuteri (formerly Lactobacillus reuteri) is a desirable host for the production of 1,3-propanediol (1,3-PDO) from glycerol when 1,3-PDO is used in the food or cosmetic industry. However, the production is hindered by strain instability, causing cell lysis, and difficult gene manipulation. This study reveals that the stability of L. reuteri DSM 20016 and its 1,3-PDO production, especially in the alcohol dehydrogenases (ADHs)-deletion mutants, are greatly enhanced after the deletion of two prophages (Φ3 and Φ4) present in the L. reuteri's chromosome. The resulting phage-free and ADHs-deletion mutant could produce >825 mM 1,3-PDO in 48 h without cell lysis at the theoretical maximum yield on glucose of ~2 mol/mol. Compared to the wild-type strain, the mutant exhibited a 45.2% increase in 1,3-PDO production titer and a 2.1-fold increase in yield. In addition, this study reports that the transformation efficiency of L. reuteri Δadh2Δadh6 mutant strains were greatly enhanced by >300-fold after the deletion of prophage Φ3, probably due to the removal of a restriction-modification (RM) system which resides in the phage genome. With improved stability and higher transformation efficiency, recombinant L. reuteri DSM 20016 Δadh2Δadh6ΔΦ3ΔΦ4 can be a more reliable and amenable host for industrial applications.


Subject(s)
Bacteriophages , Limosilactobacillus reuteri , Prophages/genetics , Limosilactobacillus reuteri/genetics , Propylene Glycols , Propylene Glycol , Glycerol , Alcohol Dehydrogenase/genetics
13.
Arch Microbiol ; 206(4): 140, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441642

ABSTRACT

Limosilactobacillus reuteri is an indigenous inhabitant of the animal gut known for its probiotic effects on the host. In our previous study, a large number of L. reuteri strains were isolated from the gastrointestinal tract of mice recovering from ulcerative colitis, from which we randomly selected L. reuteri RE225 for whole genome sequencing to explore its probiotic properties. The results of next-generation sequencing and third-generation single molecule sequencing showed that L. reuteri RE225 contained many genes encoding functional proteins associated with adhesion, anti-inflammatory and pathogen inhibition. And compared to other L. reuteri strains in NCBI, L. reuteri RE225 has unique gene families with probiotic functions. In order to further explore the probiotic effect of the L. reuteri RE225, the derived peptides were identified by LC-MS/MS, and the peptides with tumor necrosis factor-α binding ability were screened by reverse molecular docking and microscale thermophoresis. Finally, cell experiments demonstrated the anti-inflammatory ability of the peptides. Western blotting and qPCR analyses confirmed that the selected peptides might alleviate LPS-induced inflammation in NCM460 cells by inhibiting JAK2/STAT3 pathway activation.


Subject(s)
Colitis, Ulcerative , Limosilactobacillus reuteri , Animals , Mice , Limosilactobacillus reuteri/genetics , Colitis, Ulcerative/drug therapy , Chromatography, Liquid , Molecular Docking Simulation , Tandem Mass Spectrometry , Peptides/genetics , Peptides/pharmacology , Whole Genome Sequencing
14.
Arch Microbiol ; 206(7): 322, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38907754

ABSTRACT

Limosilactobacillus reuteri DSM17938 is one of the most pivotal probiotics, whose general beneficial effects on the intestinal microbiota are well recognized. Enhancing their growth and metabolic activity can effectively regulate the equilibrium of intestinal microbiota, leading to improved physical health. A common method to promote the growth of Lactobacillus is the addition of prebiotics. Current research suggests that proteins and their hydrolysates from different sources with potential prebiotic activity can also promote the growth of probiotics. In this study, soybean proteins and peptides were effective in promoting the growth, organic acid secretion, and adhesive properties of Limosilactobacillus reuteri DSM17938 to Caco-2 cells. These results illustrate the feasibility of soybean proteins and peptides as prebiotics, providing theoretical and practical advantages for their application.


Subject(s)
Bacterial Adhesion , Limosilactobacillus reuteri , Peptides , Probiotics , Soybean Proteins , Limosilactobacillus reuteri/growth & development , Limosilactobacillus reuteri/metabolism , Soybean Proteins/pharmacology , Soybean Proteins/metabolism , Soybean Proteins/chemistry , Humans , Caco-2 Cells , Bacterial Adhesion/drug effects , Peptides/pharmacology , Prebiotics , Gastrointestinal Microbiome/drug effects , Glycine max/microbiology
15.
Microbiol Immunol ; 68(7): 213-223, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38747013

ABSTRACT

Acute kidney injury (AKI) has considerably high morbidity and mortality but we do not have proper treatment for it. There is an urgent need to develop new prevention or treatment methods. Gut microbiota has a close connection with renal diseases and has become the new therapy target for AKI. In this study, we found the oral administration of the probiotic Limosilactobacillus reuteri had a prevention effect on the AKI induced by lipopolysaccharide (LPS). It reduced serum concentration of creatinine and urea nitrogen and protected the renal cells from necrosis and apoptosis. Meanwhile, L. reuteri improved the gut barrier function, which is destroyed in AKI, and modulated the gut microbiota and relevant metabolites. Compared with the LPS group, L. reuteri increased the proportion of Proteobacteria and reduced the proportion of Firmicutes, changing the overall structure of the gut microbiota. It also influenced the fecal metabolites and changed the metabolite pathways, such as tyrosine metabolism, pentose and glucuronate interconversions, galactose metabolism, purine metabolism, and insulin resistance. These results showed that L. reuteri is a potential therapy for AKI as it helps in sustaining the gut barrier integrity and modulating gut microbiota and related metabolites.


Subject(s)
Acute Kidney Injury , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Gastrointestinal Microbiome/drug effects , Limosilactobacillus reuteri/physiology , Limosilactobacillus reuteri/metabolism , Animals , Acute Kidney Injury/prevention & control , Acute Kidney Injury/metabolism , Mice , Lipopolysaccharides/metabolism , Male , Kidney/microbiology , Kidney/metabolism , Feces/microbiology , Disease Models, Animal , Creatinine/blood , Mice, Inbred C57BL , Apoptosis/drug effects
16.
J Periodontal Res ; 59(3): 552-564, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38193526

ABSTRACT

OBJECTIVE: The present study aimed to evaluate the effects of reuterin, a bioactive isolated from the probiotic Lactobacillus reuteri (L. reuteri) on periodontal tissue regeneration, and provide a new strategy for periodontitis treatment in the future. BACKGROUND: Data discussing the present state of the field: Probiotics are essential for maintaining oral microecological balance. Our previous study confirmed that probiotic L. reuteri extracts could rescue the function of mesenchymal stem cells (MSCs) and promote soft tissue wound healing by neutralizing inflammatory Porphyromonas gingivalis-LPS. Periodontitis is a chronic inflammatory disease caused by bacteria seriously leading to tooth loss. In this study, we isolated and purified reuterin from an extract of L. reuteri to characterize from the extracts of L. reuteri to characterize its role in promoting periodontal tissue regeneration and controlling inflammation in periodontitis. METHODS: Chromatographic analysis was used to isolate and purify reuterin from an extract of L. reuteri, and HNMR was used to characterize its structure. The inflammatory cytokine TNFα was used to simulate the inflammatory environment. Periodontal ligament stem cells (PDLSCs) were treated with TNFα and reuterin after which their effects were characterized using scratch wound cell migration assays to determine the concentration of reuterin, an experimental periodontitis model in rats was used to investigate the function of reuterin in periodontal regeneration and inflammation control in vivo. Real-time PCR, dye transfer experiments, image analysis, alkaline phosphatase activity, Alizarin red staining, cell proliferation, RNA-sequencing and Western Blot assays were used to detect the function of PDLSCs. RESULTS: In vivo, local injection of reuterin promoted periodontal tissue regeneration of experimental periodontitis in rats and reduced local inflammatory response. Moreover, we found that TNFα stimulation caused endoplasmic reticulum (ER) stress in PDLSCs, which resulted in decreased osteogenic differentiation. Treatment with reuterin inhibited the ER stress state of PDLSCs caused by the inflammatory environment and restored the osteogenic differentiation and cell proliferation functions of inflammatory PDLSCs. Mechanistically, we found that reuterin restored the functions of inflammatory PDLSCs by inhibiting the intercellular transmission of ER stress mediated by Cx43 in inflammatory PDLSCs and regulated osteogenic differentiation capacity. CONCLUSION: Our findings identified reuterin isolated from extracts of the probiotic L. reuteri, which improves tissue regeneration and controls inflammation, thus providing a new therapeutic method for treating periodontitis.


Subject(s)
Endoplasmic Reticulum Stress , Glyceraldehyde , Limosilactobacillus reuteri , Probiotics , Propane , Regeneration , Animals , Propane/analogs & derivatives , Propane/pharmacology , Propane/therapeutic use , Probiotics/therapeutic use , Probiotics/pharmacology , Endoplasmic Reticulum Stress/drug effects , Glyceraldehyde/analogs & derivatives , Glyceraldehyde/pharmacology , Rats , Regeneration/drug effects , Periodontitis/microbiology , Periodontal Ligament/drug effects , Humans , Male , Tumor Necrosis Factor-alpha , Rats, Sprague-Dawley , Cell Proliferation/drug effects , Stem Cells/drug effects
17.
Eur J Pediatr ; 183(5): 2311-2324, 2024 May.
Article in English | MEDLINE | ID: mdl-38427038

ABSTRACT

Infantile functional gastrointestinal disorders, such as colic, constipation, diarrhea, and gastroesophageal reflux (regurgitation), often occur in early infancy and, representing one of the causes of significant parental anxiety, lead to a significant strain on the healthcare resources. In this study, we aimed to evaluate the effects of Lactobacillus reuteri drops (L. reuteri NCIMB 30351) on the symptoms of infantile colic, constipation, diarrhea, and gastroesophageal reflux, as well as on the levels of intestinal microbiota in full-term newborns during the first months of life. A randomized, placebo-controlled, single-masked (blinded), post-marketing clinical study was conducted in two clinical units-Children's City Clinical Hospital of Moscow and Medical Center "St. Andrew's Hospitals-NEBOLIT" from March 2020 to May 2022 in 90 infants aged from 1 to 4 months (mean age (± SD) 12.3 ± 5.09 weeks; 53.3% females, 46.7% males). Patients with colic, regurgitation (single symptom or combination of several symptoms), and constipation or diarrhea were randomly allocated in two parallel arms to receive either 5 drops (2 × 108 colony forming unit) of L. reuteri NCIMB 30351 (n = 60) or masked placebo (n = 30) for 25 consecutive days. Two treatment arms had equal numbers of patients with constipation and diarrhea (n = 30 each). Daily crying times and their duration, evacuations, and regurgitations were recorded in a structured diary. The levels of gut microbiota were analyzed by deep sequencing of bacterial 16S rRNA gene. Infants with colic receiving supplementary L. reuteri NCIMB 30351 for 25 days had significant reduction in the numbers of colic (change from baseline - 6.3 (7.34) vs - 3.0 (7.29) in placebo, P < 0.05) and numbers of crying cases and mean duration of crying (decrease from baseline - 144 (70.7) minutes, lower in the diarrhea subgroup than in constipation infants, compared with - 80 (58.9) in placebo, P < 0.0001), as well as regurgitation numbers (decreased by - 4.8 (2.49) with L. reuteri vs - 3 (7.74) with placebo). We also observed increased numbers of evacuations in infants with constipation (L. reuteri 2.2 (2.4) vs 0.9 (1.06) in placebo, P < 0.05). There was a remarkable reduction of evacuations in infants with diarrhea, while not statistically significant. The analysis of bacterial 16S rRNA gene in the collected samples showed that L. reuteri positively influences the proportions of prevalent species, while it negatively affects both conditionally pathogenic and commensal microbes. Additional in vitro test for formation of Clostridium colonies in the presence of the probiotic demonstrated that L. reuteri effectively inhibits the growth of pathogenic Clostridium species. No adverse events were reported in this study.   Conclusion: The uptake of L. reuteri NCIMB 30351 leads to a significant reduction in the number of regurgitations, feeding-induced constipations, and diarrhea as well as mean daily numbers of crying and crying duration in infants during the first months of life. Our results suggest that L. reuteri NCIMB 30351 represents a safe and effective treatment for colic in newborns.  Trial registration: ClinicalTrials.gov : NCT04262648. What is Known: • Infantile functional gastrointestinal disorders, such as colic, constipation, diarrhea, and gastroesophageal reflux (regurgitation), often occur in early infancy and, represent one of the causes of significant parental anxiety. • A number of studies have shown that both the composition and diversity of the intestinal microbiota play important roles in the development and function of the gastrointestinal tract. What is New: • The uptake of L. reuteri NCIMB 30351 leads to a significant reduction in the number of regurgitations, feeding-induced constipations, and diarrhea as well as mean daily numbers of crying and crying duration in infants during the first months of life. • L. reuteri positively influences the proportions of prevalent species, while it negatively affects both conditionally pathogenic and commensal microbes in gut microbiota.


Subject(s)
Gastrointestinal Diseases , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Female , Humans , Infant , Infant, Newborn , Male , Colic/therapy , Colic/microbiology , Constipation/therapy , Constipation/microbiology , Diarrhea/microbiology , Diarrhea/therapy , Gastroesophageal Reflux/microbiology , Gastroesophageal Reflux/therapy , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/therapy , Probiotics/therapeutic use , Probiotics/administration & dosage , Single-Blind Method , Treatment Outcome , Prospective Studies
18.
J Nanobiotechnology ; 22(1): 241, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735933

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) incidence is increasing in recent years due to intestinal flora imbalance, making oral probiotics a hotspot for research. However, numerous studies related to intestinal flora regulation ignore its internal mechanisms without in-depth research. RESULTS: Here, we developed a probiotic microgel delivery system (L.r@(SA-CS)2) through the layer-by-layer encapsulation technology of alginate (SA) and chitosan (CS) to improve gut microbiota dysbiosis and enhance anti-tumor therapeutic effect. Short chain fatty acids (SCFAs) produced by L.r have direct anti-tumor effects. Additionally, it reduces harmful bacteria such as Proteobacteria and Fusobacteriota, and through bacteria mutualophy increases beneficial bacteria such as Bacteroidota and Firmicutes which produce butyric acid. By binding to the G protein-coupled receptor 109A (GPR109A) on the surface of colonic epithelial cells, butyric acid can induce apoptosis in abnormal cells. Due to the low expression of GPR109A in colon cancer cells, MK-6892 (MK) can be used to stimulate GPR109A. With increased production of butyrate, activated GPR109A is able to bind more butyrate, which further promotes apoptosis of cancer cells and triggers an antitumor response. CONCLUSION: It appears that the oral administration of L.r@(SA-CS)2 microgels may provide a treatment option for CRC by modifying the gut microbiota.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Gastrointestinal Microbiome/drug effects , Probiotics/pharmacology , Humans , Fatty Acids, Volatile/metabolism , Animals , Limosilactobacillus reuteri/metabolism , Mice , Chitosan/chemistry , Alginates/chemistry , Alginates/pharmacology , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Administration, Oral , Colorectal Neoplasms/drug therapy , Cell Line, Tumor , Receptors, G-Protein-Coupled/metabolism , Microgels/chemistry , Mice, Inbred BALB C , Butyric Acid/pharmacology , Butyric Acid/metabolism
19.
Biochemistry (Mosc) ; 89(Suppl 1): S71-S89, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38621745

ABSTRACT

The problem of antibiotic resistance is currently very acute. Numerous research and development of new antibacterial drugs are being carried out that could help cope with various infectious agents. One of the promising directions for the search for new antibacterial drugs is the search among the probiotic strains present in the human gastrointestinal tract. This review is devoted to characteristics of one of these probiotic strains that have been studied to date: Limosilactobacillus reuteri. The review discusses its properties, synthesis of various compounds, as well as role of this strain in modulating various systems of the human body. The review also examines key characteristics of one of the most harmful among the currently known pathogenic organisms, Klebsiella, which is significantly resistant to antibiotics existing in medical practice, and also poses a great threat of nosocomial infections. Discussion of characteristics of the two strains, which have opposite effects on human health, may help in creation of new effective antibacterial drugs without significant side effects.


Subject(s)
Lactobacillus , Limosilactobacillus reuteri , Humans , Klebsiella , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
20.
J Infect Chemother ; 30(9): 838-846, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38423298

ABSTRACT

INTRODUCTION: The current study aimed to investigate the role of probiotic Lactobacillus reuteri for the treatment and prevention of breast cancer. MATERIALS AND METHODS: Breast cancer was induced by using Cadmium Chloride (Cd) (2 mg/kg) in group II. Tamoxifen was administered to group III. Group IV was treated with Lactobacillus reuteri. Group V was treated with Cd for one month and divided into three subgroups including VA, VB, and VC which were treated with tamoxifen, Lactobacillus reuteri, and tamoxifen + Lactobacillus reuteri, respectively. RESULTS: Significantly higher levels of TNF-α (40.9 ± 4.2 pg/mL), IL-6 (28.0 ± 1.5 pg/mL), IL-10 (60.2 ± 2.0 pg/mL), IFN-γ (60.2 ± 2.0 pg/mL), ALAT (167.2 ± 6.2 U/l), ASAT (451.6 ± 13.9 U/l), and MDA (553.8 ± 19.6 U/l) was observed in Cd group. In comparison, significantly lower levels of TNF-α (18.0 ± 1.1 pg/mL), IL-6 (9.4 ± 0.4 pg/mL), IL-10 (20.8 ± 1.1 pg/mL), IFN-γ (20.8 ± 1.1 pg/mL), ALAT (85.2 ± 3.6 U/l), ASAT (185 ± 6.9 U/l), and MDA (246.0 ± 7.5 U/l) were observed in group Cd + Tam + LR. Liver histopathology of the Cd group showed hemorrhage and ductal aberrations. However, mild inflammation and healthier branched ducts were observed in treatment groups. Furthermore, the renal control group showed normal glomerular tufts, chronic inflammation from the Cd group, and relatively healthier glomerulus with mild inflammation in treatment groups. CONCLUSION: Hence, the preventive and anticancerous role of probiotic Lactobacillus reuteri is endorsed by the findings of the current study.


Subject(s)
Cadmium Chloride , Limosilactobacillus reuteri , Probiotics , Animals , Female , Probiotics/therapeutic use , Probiotics/pharmacology , Probiotics/administration & dosage , Mice , Breast Neoplasms/drug therapy , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Cytokines/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mammary Neoplasms, Experimental/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL