Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Nature ; 607(7919): 563-570, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35831502

RESUMO

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.


Assuntos
Bactérias , Translocação Bacteriana , Evolução Biológica , Microbioma Gastrointestinal , Fígado , Bactérias/genética , Bactérias/imunologia , Bactérias/patogenicidade , Translocação Bacteriana/genética , Parede Celular/genética , Enterococcus/genética , Enterococcus/imunologia , Microbioma Gastrointestinal/genética , Genômica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamação/microbiologia , Inflamação/patologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/imunologia , Fígado/microbiologia , Fígado/patologia , Linfonodos/microbiologia , Mutação , Processos Estocásticos , Simbiose/genética , Simbiose/imunologia
2.
Appl Environ Microbiol ; 90(7): e0024724, 2024 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-38888338

RESUMO

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.


Assuntos
Bifidobacterium , Vesículas Extracelulares , Limosilactobacillus reuteri , Probióticos , Limosilactobacillus reuteri/metabolismo , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/crescimento & desenvolvimento , Vesículas Extracelulares/metabolismo , Humanos , Bifidobacterium/metabolismo , Bifidobacterium/genética , Bifidobacterium/crescimento & desenvolvimento
3.
Appl Environ Microbiol ; 90(7): e0081424, 2024 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-38888337

RESUMO

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.


Assuntos
Proteínas de Bactérias , Limosilactobacillus reuteri , Pirofosfatases , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/enzimologia , Limosilactobacillus reuteri/metabolismo , Pirofosfatases/genética , Pirofosfatases/metabolismo , Pterinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Neopterina/análogos & derivados
4.
Biotechnol Bioeng ; 121(1): 317-328, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37747698

RESUMO

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.


Assuntos
Bacteriófagos , Limosilactobacillus reuteri , Prófagos/genética , Limosilactobacillus reuteri/genética , Propilenoglicóis , Propilenoglicol , Glicerol , Álcool Desidrogenase/genética
5.
Arch Microbiol ; 206(4): 140, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38441642

RESUMO

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.


Assuntos
Colite Ulcerativa , Limosilactobacillus reuteri , Animais , Camundongos , Limosilactobacillus reuteri/genética , Colite Ulcerativa/tratamento farmacológico , Cromatografia Líquida , Simulação de Acoplamento Molecular , Espectrometria de Massas em Tandem , Peptídeos/genética , Peptídeos/farmacologia , Sequenciamento Completo do Genoma
6.
Microb Cell Fact ; 22(1): 86, 2023 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-37120528

RESUMO

BACKGROUND: Expression systems for lactic acid bacteria have been developed for metabolic engineering applications as well as for food-grade recombinant protein production. But the industrial applications of lactic acid bacteria as cell factories have been limited due to low biomass formation resulted in low efficiency of biomanufacturing process. Limosilactobacillus reuteri KUB-AC5 is a safe probiotic lactic acid bacterium that has been proven as a gut health enhancer, which could be developed as a mucosal delivery vehicle for vaccines or therapeutic proteins, or as expression host for cell factory applications. Similar to many lactic acid bacteria, its oxygen sensitivity is a key factor that limits cell growth and causes low biomass production. The aim of this study is to overcome the oxidative stress in L. reuteri KUB-AC5. Several genes involved in oxidative and anti-oxidative stress were investigated, and strain improvement for higher cell densities despite oxidative stress was performed using genetic engineering. RESULTS: An in-silico study showed that L. reuteri KUB-AC5 genome possesses an incomplete respiratory chain lacking four menaquinone biosynthesis genes as well as a complete biosynthesis pathway for the production of the precursor. The presence of an oxygen consuming enzyme, NADH oxidase (Nox), leads to high ROS formation in aerobic cultivation, resulting in strong growth reduction to approximately 25% compared to anaerobic cultivation. Recombinant strains expressing the ROS scavenging enzymes Mn-catalase and Mn-superoxide dismutase were successfully constructed using the pSIP expression system. The Mn-catalase and Mn-SOD-expressing strains produced activities of 873 U/ml and 1213 U/ml and could minimize the ROS formation in the cell, resulting in fourfold and sevenfold higher biomass formation, respectively. CONCLUSIONS: Expression of Mn-catalase and Mn-SOD in L. reuteri KUB-AC5 successfully reduced oxidative stress and enhanced growth. This finding could be applied for other lactic acid bacteria that are subject to oxidative stress and will be beneficial for applications of lactic acid bacteria for cell factory applications.


Assuntos
Limosilactobacillus reuteri , Probióticos , Limosilactobacillus reuteri/genética , Catalase/metabolismo , Espécies Reativas de Oxigênio , Estresse Oxidativo , Oxigênio , Superóxido Dismutase/metabolismo , Probióticos/metabolismo
7.
J Appl Microbiol ; 134(1)2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36724218

RESUMO

AIMS: At conception, the infant gut barrier is immature, gradually developing with regular intake of maternal milk. This study addressed whether the barrier-strengthening effect of breast feeding might be attributable, at least in part, to autochthonous beneficial human milk bacteria. METHODS AND RESULTS: Twelve bacterial strains from the breast milk of Pakistani mothers who underwent cesarean delivery (NPL-88, NPL-157, NPL-179, NPL-181, NPL-388 (Limosilactobacillus reuteri), NPL-76, NPL-495, NPL-504 (Limosilactobacillus fermentum), NPL-415 (Lactobacillus pentosus), NPL-412, NPL-416 (Lactiplantibacilllus plantarum) and NPL-374 (Bifidobacterium longum) were shortlisted based on their tolerance to acidic pH (2.8-4.2) and bile (0.1-0.3%). The effect of these bacteria on gut barrier function in the presence and absence of pathogens was assessed as changes in transepithelial electrical resistance (TEER) in the human T84 colonic epithelial cell line and in murine enteroid-derived monolayers (EDMs). The TEER of T84 cells monolayers rose in the presence of most of the human milk strains, being most pronounced in case of L. reuteri NPL-88 (34% within five h), exceeding the effect of the well-known probiotic L. acidophilus (20%). qRT-PCR, western blot and immunofluorescent staining associated the increase in TEER with enhanced expression of tight junction proteins. Pretreatment of murine EDMs with NPL-88 also largely prevented the ability of the pathogen, Salmonella, to decrease TEER (87 ± 1.50%; P < 0.0001, n = 4). CONCLUSIONS: Human milk lactic acid bacteria are potential probiotics that can strengthen gut barrier function and protect breastfed neonates against enteric infections.


Assuntos
Limosilactobacillus fermentum , Limosilactobacillus reuteri , Probióticos , Lactente , Feminino , Recém-Nascido , Camundongos , Humanos , Animais , Leite Humano , Limosilactobacillus reuteri/genética , Bactérias , Probióticos/metabolismo
8.
Nature ; 552(7684): 244-247, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29211710

RESUMO

Microbiome-wide association studies have established that numerous diseases are associated with changes in the microbiota. These studies typically generate a long list of commensals implicated as biomarkers of disease, with no clear relevance to disease pathogenesis. If the field is to move beyond correlations and begin to address causation, an effective system is needed for refining this catalogue of differentially abundant microbes and to allow subsequent mechanistic studies. Here we demonstrate that triangulation of microbe-phenotype relationships is an effective method for reducing the noise inherent in microbiota studies and enabling identification of causal microbes. We found that gnotobiotic mice harbouring different microbial communities exhibited differential survival in a colitis model. Co-housing of these mice generated animals that had hybrid microbiotas and displayed intermediate susceptibility to colitis. Mapping of microbe-phenotype relationships in parental mouse strains and in mice with hybrid microbiotas identified the bacterial family Lachnospiraceae as a correlate for protection from disease. Using directed microbial culture techniques, we discovered Clostridium immunis, a previously unknown bacterial species from this family, that-when administered to colitis-prone mice-protected them against colitis-associated death. To demonstrate the generalizability of our approach, we used it to identify several commensal organisms that induce intestinal expression of an antimicrobial peptide. Thus, we have used microbe-phenotype triangulation to move beyond the standard correlative microbiome study and identify causal microbes for two completely distinct phenotypes. Identification of disease-modulating commensals by microbe-phenotype triangulation may be more broadly applicable to human microbiome studies.


Assuntos
Clostridium/isolamento & purificação , Clostridium/fisiologia , Colite/microbiologia , Colite/prevenção & controle , Microbioma Gastrointestinal , Fenótipo , Animais , Peso Corporal , Sobrevivência Celular , Clostridium/genética , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Vida Livre de Germes , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/fisiologia , Camundongos , Proteínas Associadas a Pancreatite/metabolismo , Ruminococcus/genética , Ruminococcus/fisiologia , Simbiose
9.
J Ind Microbiol Biotechnol ; 50(1)2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-37974056

RESUMO

Evaluating the safety of probiotic microorganisms is an important part of the development of probiotic products. In this study, we have performed a systematic safety assessment of Limosilactobacillus reuteri American Type Culture Collection (ATCC) PTA 4659 based on genome analysis, antibiotic susceptibility testing, phenotypic characterization, and a human clinical safety study. Genome sequence analysis showed that the strain is free from virulence and antibiotic resistance genes. Connected to this, phenotypic characterization showed that the strain is susceptible to the main classes of antibiotics. Limosilactobacillus reuteri ATCC PTA 4659 was shown to produce histamine, which has previously been described as an anti-inflammatory mediator produced by certain L. reuteri strains. However, the amount of histamine, a biogenic amine, poses no safety concern of a potential product. The strain was investigated in a human clinical safety study and was shown to survive passage through the gastrointestinal tract, both when administered at high [1 × 1011 colony-forming units (CFU)/day] and low doses (1 × 109 CFU/day). The clinical safety evaluation showed that the doses administered are safe for human consumption. Furthermore, carbohydrate utilization, mucus adhesion, and tolerance to acid and bile were studied. It was shown that L. reuteri ATCC PTA 4659 has a very high adhesion to mucus and tolerance to both gastric pH and bile, all potentially important properties for a probiotic strain. Altogether, this study has demonstrated that Limosilactobacillus reuteri ATCC PTA 4659 is safe for human consumption and along with its phenotypic characteristics and previously described anti-inflammatory effects, makes it a promising strain for future probiotic development. NCT01033539.


Assuntos
Limosilactobacillus reuteri , Probióticos , Humanos , Antibacterianos/farmacologia , Anti-Inflamatórios , Genômica , Histamina , Limosilactobacillus reuteri/genética
10.
BMC Microbiol ; 22(1): 298, 2022 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-36510154

RESUMO

BACKGROUND: pTE15 is a ~ 15-kb narrow-host-range indigenous plasmid from Lactobacillus reuteri N16 that does not replicate in selected Bacillus spp., Staphylococcus spp., and other Lactobacillus spp. METHODS: Combined deletion analysis the minireplicon essential of pTE15 with replicon-probe vector pUE80 (-) to confirmed sufficient for replication and from the ssDNA intermediate detection, plasmid amplification tested by chloramphenicol treatment, and replication origin sequence analysis to delineated the novel theta-type replication of pTE15. RESULTS: Single-stranded intermediate of pTE15 DNA was not detected in L. reuteri, indicating that this plasmid does not replicate via a rolling circle mechanism. The replicon of pTE15 did not display the structural organization typical of rolling-circle plasmids, nor were they similar to known rolling-circle plasmids. We further provided evidence that this plasmid applied a new mode of theta-type replication mechanism: (1) the size of this plasmid was > 10-kb; (2) the minireplicon consisted of AT-rich (directed repeat, iteron) and DnaA sequences; (3) the minireplicon did not contain double-strand origin (DSO) and essential rep genes, and it also showed no single-strand origin (SSO) structure; (4) the intermediate single-stranded DNA products were not observed for pTE15 replication; (5) the minireplicon did not contain a typical essential replication protein, Rep, (6) its copy number was decreased by chloramphenicol treatment, and (7) genes in pTE15 replication region encoded truncated RepA (TRepA), RepB and RepC, which were replication-associated proteins, but they were not essential for pTE15 replication. CONCLUSIONS: Collectively, our results strongly suggested that the indigenous plasmid pTE15 of L. reuteri N16 belongs to a new class of theta replicons.


Assuntos
Limosilactobacillus reuteri , Limosilactobacillus reuteri/genética , Sequência de Bases , Replicon , Plasmídeos/genética , Origem de Replicação , Replicação do DNA , DNA de Cadeia Simples
11.
Amino Acids ; 54(4): 663-673, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34657206

RESUMO

This study was designed to gain information about the underlying mechanisms of the effects of a food-occurring free oxidized amino acid, α-aminoadipic acid (AAA), on the probiotic Lactobacillus reuteri PL503. This bacterium was incubated in colonic-simulated conditions (37 °C for 24 h in microaerophilic conditions) and exposed to three food-compatible AAA concentrations, namely, 1 mM, 5 mM, and 10 mM. A control group with no AAA exposure was also considered. Each of the four experimental conditions was replicated three times and samplings were collected at 12, 16, 20, and 24 h. The downregulation of the uspA gene by AAA (0.5-fold decrease as compared to control) suggests that AAA is identified as a potential chemical threat. The dhaT gene, implicated in the antioxidant defense, was found to be upregulated in bacteria treated with 1 and 5 mM AAA (up to twofold increase, as compared to control), which suggest the ability of the oxidized amino acid to impair the redox status of the bacterium. In fact, AAA caused an increased production of reactive oxygen species (ROS) and the accretion of post-translational changes (protein carbonylation) in L. reuteri (up to 13 nmol allysine/mg protein vs 1.8 nmol allysine/mg protein in control). These results suggest that probiotic bacteria identify oxidized amino acids as harmful species and activate mechanisms that may protect themselves and the host against their noxious effects.


Assuntos
Limosilactobacillus reuteri , Probióticos , Ácido 2-Aminoadípico/metabolismo , Aminoácidos/metabolismo , Expressão Gênica , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/metabolismo , Lisina/metabolismo , Oxirredução , Probióticos/farmacologia
12.
Lett Appl Microbiol ; 74(4): 593-603, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35014712

RESUMO

Lactic acid bacteria are micro-organisms used for probiotic purposes and form major parts of human and mammalian intestinal microbiota, exerting important health-promoting effects on the host. Here, we evaluated Lactobacillus reuteri strain S5 isolated from the intestines of healthy white feather broilers. Lactobacillus reuteri S5 grew best after 20 h of incubation in MRS medium. Lactic acid production was 1·42 mmol l-1 at 24 h, which was well tolerated. Activities of T-AOC, GSH-Px and T-SOD in the cell-free fermentation supernatant of L. reuteri S5 were higher than those in the bacteria, and the strain showed good hydrophobicity in vitro. The dominant carbon and nitrogen sources of L. reuteri S5 were glucose and soybean meal. A high-quality complete genome map of L. reuteri S5 was obtained using a Pacbio nanopore third-generation sequencing platform. The results showed that L. reuteri S5 possesses a complete primary metabolic pathway, encoding the main functional enzymes of the glycolysis pathway and pentose phosphate pathway. The genome contains genes encoding antioxidants and conferring tolerance to inorganic salt ions, acids and bile salts. This study shows that L. reuteri S5 is a probiotic strain with excellent probiotic characteristics and has great potential for the development of feed additives to promote animal health.


Assuntos
Lactobacillales , Limosilactobacillus reuteri , Probióticos , Animais , Galinhas , Intestinos/microbiologia , Limosilactobacillus reuteri/genética , Mamíferos , Probióticos/metabolismo
13.
J Proteome Res ; 20(5): 2447-2457, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33705137

RESUMO

The ability to survive in the harsh gastrointestinal tract (GIT) environment is essential for Lactobacillus reuteri (L. reuteri) exhibiting beneficial effects. In this study, we found that the hydrophobicity and auto-aggregation of L. reuteri SH23 were significantly decreased and biofilm production was also significantly decreased when L. reuteri SH23 passes through the simulated GIT. Furthermore, according to the comparative transcriptome analysis, gene expression involved in the cell envelope, metabolic processes, common stress response, regulatory systems, and transporters were also affected. Meanwhile, label-free quantitative proteomics was used to identify the differential expression of surface proteins of L. reuteri in response to simulated gastrointestinal fluid. Proteins related to the ABC transporters (Lreu_0517, Lreu_0098, and Lreu_0296) and LPxTG anchor domain proteins were upregulated in the cell surface after gastrointestinal fluid treatment, which is useful for adherence and colonization of L. reuteri in the GIT. Additionally, the recombinant Mub protein could also enhance the survival ability of L. reuteri SH23 in GIT stress environment. This study provides a comprehensive understanding of the adaptation and adhesion mechanisms of L. reuteri SH23 under the gastrointestinal tract by the transcriptomics and proteomics analysis, and mucus-binding proteins were involved in the adhesion and GIT tolerance process.


Assuntos
Limosilactobacillus reuteri , Probióticos , Aderência Bacteriana , Limosilactobacillus reuteri/genética , Muco , Proteômica , Transcriptoma
14.
BMC Biotechnol ; 21(1): 46, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34330235

RESUMO

BACKGROUND: Limosilactobacillus reuteri (earlier known as Lactobacillus reuteri) is a well-studied lactic acid bacterium, with some specific strains used as probiotics, that exists in different hosts such as human, pig, goat, mouse and rat, with multiple body sites such as the gastrointestinal tract, breast milk and mouth. Numerous studies have confirmed the beneficial effects of orally administered specific L. reuteri strains, such as preventing bone loss and promoting regulatory immune system development. L. reuteri ATCC PTA 6475 is a widely used strain that has been applied in the market as a probiotic due to its positive effects on the human host. Its health benefits may be due, in part, to the production of beneficial metabolites. Considering the strain-specific effects and genetic diversity of L. reuteri strains, we were interested to study the metabolic versatility of these strains. RESULTS: In this study, we aimed to systematically investigate the metabolic features and diversities of L. reuteri strains by using genome-scale metabolic models (GEMs). The GEM of L. reuteri ATCC PTA 6475 was reconstructed with a template-based method and curated manually. The final GEM iHL622 of L. reuteri ATCC PTA 6475 contains 894 reactions and 726 metabolites linked to 622 metabolic genes, which can be used to simulate growth and amino acids utilization. Furthermore, we built GEMs for the other 35 L. reuteri strains from three types of hosts. The comparison of the L. reuteri GEMs identified potential metabolic products linked to the adaptation to the host. CONCLUSIONS: The GEM of L. reuteri ATCC PTA 6475 can be used to simulate metabolic capabilities and growth. The core and pan model of 35 L. reuteri strains shows metabolic capacity differences both between and within the host groups. The GEMs provide a reliable basis to investigate the metabolism of L. reuteri in detail and their potential benefits on the host.


Assuntos
Genoma Bacteriano , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/metabolismo , Animais , Cabras , Especificidade de Hospedeiro , Humanos , Limosilactobacillus reuteri/crescimento & desenvolvimento , Camundongos , Ratos , Suínos
15.
Proc Natl Acad Sci U S A ; 115(8): 1895-1900, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29432190

RESUMO

Impaired wound closure is a growing medical problem associated with metabolic diseases and aging. Immune cells play important roles in wound healing by following instructions from the microenvironment. Here, we developed a technology to bioengineer the wound microenvironment and enhance healing abilities of the immune cells. This resulted in strongly accelerated wound healing and was achieved by transforming Lactobacilli with a plasmid encoding CXCL12. CXCL12-delivering bacteria administrated topically to wounds in mice efficiently enhanced wound closure by increasing proliferation of dermal cells and macrophages, and led to increased TGF-ß expression in macrophages. Bacteria-produced lactic acid reduced the local pH, which inhibited the peptidase CD26 and consequently enhanced the availability of bioactive CXCL12. Importantly, treatment with CXCL12-delivering Lactobacilli also improved wound closure in mice with hyperglycemia or peripheral ischemia, conditions associated with chronic wounds, and in a human skin wound model. Further, initial safety studies demonstrated that the topically applied transformed bacteria exerted effects restricted to the wound, as neither bacteria nor the chemokine produced could be detected in systemic circulation. Development of drugs accelerating wound healing is limited by the proteolytic nature of wounds. Our technology overcomes this by on-site chemokine production and reduced degradation, which together ensure prolonged chemokine bioavailability that instructed local immune cells and enhanced wound healing.


Assuntos
Quimiocina CXCL12/administração & dosagem , Quimiocina CXCL12/farmacologia , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/metabolismo , Cicatrização , Animais , Proliferação de Células , Regulação da Expressão Gênica , Terapia Genética , Humanos , Macrófagos/metabolismo , Camundongos , Plasmídeos , Pele , Técnicas de Cultura de Tecidos , Fator de Crescimento Transformador beta/metabolismo , Ferimentos e Lesões/terapia
16.
Antonie Van Leeuwenhoek ; 113(8): 1191-1200, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32410086

RESUMO

Adhesion of probiotic bacteria to the mucus layer lining the gastrointestinal tract is necessary for its effective colonisation and specific therapeutic effects. Enrichment of growth medium with mucin might stimulate bacterial adhesion, probably by increasing the expression of surface structures responsible for bacteria-gut epithelia and/or mucus interactions. The aim of this study was to determine if pre-cultivation of potentially probiotic strain Lactobacillus reuteri E (LRE) with mucin stimulates its adherence to colon cell line HT-29 and if the increased adhesion modulates mucin expression in these cells. The mucin-producing HT-29 cell line was co-cultivated for 2 h with LRE grown in MRS broth or MRS broth enriched with pig gastric mucin (LRE + M). The adherence ability of LRE was evaluated microscopically and by plate counting. The relative gene expression was measured by qPCR. Pre-cultivation of LRE in mucin enriched medium significantly increased its adhesion to 14 days HT-29 in comparison with LRE by both methods (28.64% vs. 23.83%, evaluated microscopically, and 14.31 ± 3.95 × 106 CFU ml-1 vs. 8.54 ± 0.43 × 106 CFU ml-1, evaluated by plate counting). MUC2, MUC5AC, and IL-10 were significantly upregulated after co-cultivation with LRE + M in comparison to LRE and control group (lactobacilli-free HT-29). Obtained results suggest that pre-cultivation of lactobacilli with mucin may not only stimulate their adhesion abilities but also promote their effectiveness to modulate the pathways involved in the pathophysiology of some diseases, e.g., with defective mucin synthesis in ulcerative colitis or colorectal cancer.


Assuntos
Adesão Celular , Células HT29/metabolismo , Limosilactobacillus reuteri/metabolismo , Mucinas/metabolismo , Animais , Colo , Células Epiteliais/microbiologia , Humanos , Mucosa Intestinal/metabolismo , Lactobacillus , Limosilactobacillus reuteri/genética , Probióticos , Suínos
17.
Food Microbiol ; 86: 103343, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31703887

RESUMO

The bacterial conversion of glutamine to glutamate is catalyzed by glutamine-amidotransferases or glutaminases. Glutamine deamination contributes to the formation of the bioactive metabolites glutamate, γ-aminobutyrate (GABA) and γ-glutamyl peptides, and to acid resistance. This study aimed to investigate the distribution of glutaminase(s) in lactobacilli, and to evaluate their contribution in L. reuteri to amino acid metabolism and acid resistance. Phylogenetic analysis of the glutaminases gls1, gls2 and gls3 in the genus Lactobacillus demonstrated that glutaminase is exclusively present in host-adapted species of lactobacilli. The disruption gls1, gls2 and gls3 in L. reuteri 100-23 had only a limited effect on the conversion of glutamine to glutamate, GABA, or γ-glutamyl peptides in sourdough. The disruption of all glutaminases in L. reuteri 100-23Δgls1Δgls2Δgls3 but not disruption of gls2 and gls3 eliminated the protective effect of glutamine on the survival of the strain at pH 2.5. Glutamine also enhanced acid resistance of L. reuteri 100-23ΔgadB and L. taiwanensis 107q, strains without glutamate decarboxylase activity. Taken together, the study demonstrates that glutaminases of lactobacilli do not contribute substantially to glutamine metabolism but enhance acid resistance. Their exclusive presence in host-adapted lactobacilli provides an additional link between the adaptation of lactobacilli to specific habitats and their functionality when used as probiotics and starter cultures.


Assuntos
Ácidos/metabolismo , Glutaminase/metabolismo , Limosilactobacillus reuteri/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fermentação , Ácido Glutâmico/metabolismo , Glutaminase/genética , Glutamina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Lactobacillus/enzimologia , Lactobacillus/genética , Lactobacillus/metabolismo , Limosilactobacillus reuteri/enzimologia , Limosilactobacillus reuteri/genética , Filogenia , Roedores/microbiologia
18.
Food Microbiol ; 91: 103507, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32539950

RESUMO

Bacillus spp. cause ropy bread spoilage of bread, which is characterized by a rotten fruity odor, followed by discoloration and degradation of the crumb. Bacillus spp. are wheat grain endophytes and form heat resistant endospores, therefore, process hygiene and heating during baking do not prevent ropy spoilage. This study used 8 strains of Bacillus subtilis and Bacillus amyloliquefaciens to determine whether the presence and the copy number of spoVA2mob operon influences survival after baking; in addition, the spoilage phenotype was correlated with the presence of amylolytic enzymes in genomes of Bacillus spp.. The presence and copy number of the spoVA2mob operon had only a minor effect on survival of Bacillus endospores. Strains of B. amyloliquefaciens caused ropy spoilage faster than strains of B. subtilis, this difference correlated to the number and type of extracellular amylases encoded in the genomes of the strains of B. amyloliquefaciens and B. subtilis. The inhibitory effect of sourdough on ropy spoilage was determined by addition of 3-24% sourdough fermented with L. reuteri TMW1.656. Addition of 12% and 24% sourdough, corresponding to a bread pH of 5.93 ± 0.041 and 5.53 ± 0.040, respectively, delayed ropy spoilage for 2 and more than 5 d, respectively. The comparison of addition of 12% sourdough fermented with the reutericyclin producing L. reuteri TMW1.656 and the isogenic reutericyclin-negative strain L. reuteri TMW1.656ΔgtfAΔrtcN demonstrated that reutericyclin produced in sourdough inhibits growth of Bacillus in bread. In conclusion, sourdough inhibits germination of Bacillus spores in bread and the effect of sourdough is enhanced by reutericyclin.


Assuntos
Bacillus/metabolismo , Proteínas de Bactérias/genética , Pão/microbiologia , Ácido Tenuazônico/análogos & derivados , Amilases/genética , Amilases/metabolismo , Bacillus/classificação , Bacillus/genética , Bacillus/crescimento & desenvolvimento , Proteínas de Bactérias/metabolismo , Variações do Número de Cópias de DNA , Fermentação , Microbiologia de Alimentos , Concentração de Íons de Hidrogênio , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/metabolismo , Viabilidade Microbiana , Óperon , Esporos Bacterianos/classificação , Esporos Bacterianos/genética , Esporos Bacterianos/crescimento & desenvolvimento , Temperatura , Ácido Tenuazônico/metabolismo
19.
Prep Biochem Biotechnol ; 50(8): 820-826, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32282271

RESUMO

Glucansucrases (GTFs) catalyzes the synthesis of α-glucans from sucrose and oligosaccharides in the presence of an acceptor sugar by transferring glucosyl units to the acceptor molecule with different linkages. The acceptor reactions can be affected by several parameters and this study aimed to determine the optimal reaction parameters for the production of glucansucrase-based oligosaccharides using sucrose and maltose as the donor and acceptor sugars, respectively via a hybrid technique of Response Surface Method (RSM) and Particle Swarm Optimization (PSO). The experimental design was performed using Central Composite Design and the tested parameters were enzyme concentration, acceptor:donor ratio and the reaction period. The optimization studies showed that enzyme concentration was the most effective parameter for the final oligosaccharides yields. The optimal values of the significant parameters determined for enzyme concentration and acceptor:donor ratio were 3.45 U and 0.62, respectively. Even the response surface plots for input parameters verified the PSO results, an experimental validation study was performed for the reverification. The experimental verification results obtained were also consistent with the PSO results. These findings will help our understanding in the role of different parameters for the production of oligosaccharides in the acceptor reactions of GTFs.


Assuntos
Glicosiltransferases/metabolismo , Limosilactobacillus reuteri/enzimologia , Oligossacarídeos/metabolismo , Biocatálise , Escherichia coli/genética , Escherichia coli/metabolismo , Glicosiltransferases/genética , Microbiologia Industrial , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/metabolismo , Maltose/metabolismo , Modelos Biológicos , Sacarose/metabolismo
20.
Glycobiology ; 29(1): 45-58, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30371779

RESUMO

Lactobacillus reuteri is a gut symbiont inhabiting the gastrointestinal tract of numerous vertebrates. The surface-exposed serine-rich repeat protein (SRRP) is a major adhesin in Gram-positive bacteria. Using lectin and sugar nucleotide profiling of wild-type or L. reuteri isogenic mutants, MALDI-ToF-MS, LC-MS and GC-MS analyses of SRRPs, we showed that L. reuteri strains 100-23C (from rodent) and ATCC 53608 (from pig) can perform protein O-glycosylation and modify SRRP100-23 and SRRP53608 with Hex-Glc-GlcNAc and di-GlcNAc moieties, respectively. Furthermore, in vivo glycoengineering in E. coli led to glycosylation of SRRP53608 variants with α-GlcNAc and GlcNAcß(1→6)GlcNAcα moieties. The glycosyltransferases involved in the modification of these adhesins were identified within the SecA2/Y2 accessory secretion system and their sugar nucleotide preference determined by saturation transfer difference NMR spectroscopy and differential scanning fluorimetry. Together, these findings provide novel insights into the cellular O-protein glycosylation pathways of gut commensal bacteria and potential routes for glycoengineering applications.


Assuntos
Adesinas Bacterianas/química , Limosilactobacillus reuteri/química , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Glicosilação , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/metabolismo , Mutação , Ressonância Magnética Nuclear Biomolecular , Sequências Repetitivas de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA