Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 440
Filtrar
1.
J Sci Food Agric ; 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38982876

RESUMEN

INTRODUCTION: Cystic fibrosis (CF) patients frequently experience gut microbiota dysbiosis. Probiotic supplementation is a potential therapeutic approach to modify gut microbiota and improve CF management through the gut-lung axis. The aim of this study was to investigate the effect of Lactobacillus reuteri supplementation on pulmonary function test, respiratory symptoms and growth in CF patients. METHODS: A randomized, placebo-controlled clinical trial was carried out on 40 children with CF aged from 6 to 20 years. Participants were designated to receive either L. reuteri or placebo daily for 4 months. Pulmonary function tests, weight, height and body mass index (BMI) z-scores were measured pre and post treatment. RESULTS: The median baseline BMI of the patients was 16.28 kg m-2. A significant change in the probiotic group's BMI z-score after the study period was observed (P = 0.034) but not for weight and height z-scores (P > 0.05). After treatment, Pseudomonas aeruginosa grew in sputum cultures of seven in the placebo and one patient in the intervention group (P = 0.03) while at baseline it grew in the sputum of four patients in each group. There was no significant difference in forced expiratory volume in the first second, forced expiratory flow at 25-75% or forced vital capacity change between the two groups after the treatment period (P > 0.05). Additionally, no significant differences were found in pulmonary exacerbations, hospitalization frequencies or COVID-19 infection between the two groups during the study (P > 0.05). CONCLUSION: The results suggest that L. reuteri supplementation may impact the growth of severely malnourished CF patients. Furthermore, it may be concluded that this strain might reduce P. aeruginosa in the sputum culture of CF patients. © 2024 Society of Chemical Industry.

2.
Mol Nutr Food Res ; : e2300927, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937862

RESUMEN

SCOPE: While probiotics-based therapies have exhibited potential in alleviating alcohol-associated liver disease (ALD), the specific role of postbiotics derived from Lactobacillus reuteri (L. reuteri) in ALD remains elusive. This study aims to investigate the impact of postbiotics on ameliorating alcohol-induced hepatic steatosis and the underlying mechanisms. METHODS AND RESULTS: Using network pharmacology, the study elucidates the targets and pathways impacted by postbiotics from L. reuteri, identifying the farnesoid X receptor (FXR) as a promising target for postbiotics against ALD, and lipid metabolism and alcoholism act as crucial pathways associated with postbiotics-targeting ALD. Furthermore, the study conducts histological and biochemical analyses coupled with LC/MS to evaluate the protective effects and mechanisms of postbiotics against ALD. Postbiotics may modulate bile acid metabolism in vivo by regulating FXR signaling, activating the FXR/FGF15 pathway, and influencing the enterohepatic circulation of bile acids (BAs). Subsequently, postbiotics regulate hepatic FXR activated by BAs and modulate the expression of FXR-mediated protein, including short regulatory partner (SHP) and sterol regulatory element binding protein-1c (SREBP-1c), thereby ameliorating hepatic steatosis in mice with ALD. CONCLUSION: Postbiotics effectively alleviate ethanol-induced hepatic steatosis by regulating the FXR/SHP/SREBP-1c axis, as rigorously validated in both in vivo and in vitro.

3.
Therap Adv Gastroenterol ; 17: 17562848241258021, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38846173

RESUMEN

Background: Given the growing problem of antibiotic resistance, it is crucial to improve Helicobacter pylori (H. pylori) treatment interventions or provide adjunctive therapy. The objective of this meta-analysis was to evaluate whether Lactobacillus reuteri (L. reuteri) could improve H. pylori eradication rate, reduce the incidence of adverse events (AEs), and alleviate gastrointestinal symptoms. Design: A meta-analysis of randomized controlled trials (RCTs) comparing L. reuteri supplementation therapy with placebo was conducted. Sources and methods: We retrieved relevant studies from PubMed, Embase, and the Cochrane Library. The primary outcome was H. pylori eradication rate, and the scores on the Gastrointestinal Symptom Rating Scale and AEs were secondary outcomes. Results: Eight RCTs including 1087 patients were included in this analysis. The L. reuteri supplementation group showed significantly higher H. pylori eradication rates in both intention-to-treat (ITT) and per-protocol (PP) analysis [ITT: 80.0% versus 72.6%; p = 0.005, relative risk (RR): 1.10; 95% confidence interval (CI): 1.03-1.17; number needed to treat (NNT) = 14; PP: 81.8% versus 75.0%; p = 0.006, RR: 1.09; 95% CI: 1.03-1.16; NNT = 15]. Patients treated with L. reuteri showed greater improvements in gastrointestinal symptoms (pooled mean difference: -2.43, 95% CI: -4.56 to -0.29, p = 0.03). The incidence of AEs was significantly reduced in the L. reuteri supplementation group based on ITT and PP analysis (ITT: p < 0.00001, RR: 0.72, 95% CI: 0.67-0.78; PP: p < 0.00001, RR: 0.70, 95% CI: 0.65-0.77). Conclusion: The present meta-analysis demonstrated that supplementation with L. reuteri was beneficial for improving the eradication rate of H. pylori, reducing the overall incidence of side effects, and relieving gastrointestinal symptoms in patients during treatment. The findings provide new insights into clinical decision-making. Trial registration PROSPERO: CRD42023424052.


Lactobacillus reuteri compared with placebo as an adjuvant in Helicobacter pylori eradication therapy: a meta-analysis of randomized controlled trials Given the growing problem of antibiotic resistance, it is crucial to improve Helicobacter pylori (H. pylori) treatment interventions or provide adjunctive therapy. Eight randomized controlled trials (RCTs) including 1087 patients were included in this analysis. The present meta-analysis demonstrated that supplementing with L. reuteri tends to increase the eradication rate of H. pylori, reduce the overall incidence of antibiotic-related side effects, and alleviate gastrointestinal symptoms in patients during treatment, providing new insights for clinical decision-making.

4.
Nutrients ; 16(12)2024 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-38931255

RESUMEN

Gut microbiota are the microbial organisms that play a pivotal role in intestinal health and during disease conditions. Keeping in view the characteristic functions of gut microbiota, in this study, Lactobacillus reuteri TPC32 (L. reuteri TPC32) was isolated and identified, and its whole genome was analyzed by the Illumina MiSeq sequencing platform. The results revealed that L. reuteri TPC32 had high resistance against acid and bile salts with fine in vitro antibacterial ability. Accordingly, a genome sequence of L. reuteri TPC32 has a total length of 2,214,495 base pairs with a guanine-cytosine content of 38.81%. Based on metabolic annotation, out of 2,212 protein-encoding genes, 118 and 101 were annotated to carbohydrate metabolism and metabolism of cofactors and vitamins, respectively. Similarly, drug-resistance and virulence genes were annotated using the comprehensive antibiotic research database (CARD) and the virulence factor database (VFDB), in which vatE and tetW drug-resistance genes were annotated in L. reuteri TPC32, while virulence genes are not annotated. The early prevention of L. reuteri TPC32 reduced the Salmonella typhimurium (S. Typhimurium) infection in mice. The results show that L. reuteri TPC32 could improve the serum IgM, decrease the intestinal cytokine secretion to relieve intestinal cytokine storm, reinforce the intestinal biochemical barrier function by elevating the sIgA expression, and strengthen the intestinal physical barrier function. Simultaneously, based on the 16S rRNA analysis, the L. reuteri TPC32 results affect the recovery of intestinal microbiota from disease conditions and promote the multiplication of beneficial bacteria. These results provide new insights into the biological functions and therapeutic potential of L. reuteri TPC32 for treating intestinal inflammation.


Asunto(s)
Microbioma Gastrointestinal , Genoma Bacteriano , Limosilactobacillus reuteri , Probióticos , Secuenciación Completa del Genoma , Animales , Ratones , Porcinos , Salmonella typhimurium/genética , Salmonella typhimurium/efectos de los fármacos , Antibacterianos/farmacología , Factores de Virulencia/genética
5.
Int J Food Microbiol ; 418: 110735, 2024 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-38761714

RESUMEN

This research aimed to develop, optimize, and evaluate a new antifungal nanoemulsion system based on the crude reuterin-synergistic essential oils (EOs) hybrid to overcome the EOs application limits. At first, the antifungal effects of the Lactobacillus plantarum and Lactobacillus reuteri cell-free extracts (CFE) were tested against the Botrytis cinerea, Penicillium expansum, and Alternaria alternata as indicator fungus using broth microdilution method. The L. reuteri CFE with the MIC of 125 µL/mL for B. cinerea and 250 µL/mL for P. expansum and A. alternata showed more inhibitory effects than L. plantarum. Next, reuterin as a significant antibacterial compound in the L. reuteri CFE was induced in glycerol-containing culture media. To reach a nanoemulsion with maximum antifungal activity and stability, the reuterin concentration, Tween 80 %, and ultrasound time were optimized using response surface methodology (RSM) with a volumetric constant ratio of 5 % v/v oil phase including triple synergistic EOs (thyme, cinnamon, and rosemary) at MIC concentrations. Based on the Box-Behnken Design, the maximum antifungal effect was observed in the treatment with 40 mM reuterin, 1 % Tween 80, and 3 min of ultrasound. The growth inhibitory diameter zones of B. cinerea, P. expansum, and A. alternata were estimated 6.15, 4.25, and 4.35 cm in optimum nanoemulsion, respectively. Also, the minimum average particle size diameter (16.3 nm) was observed in nanoemulsion with reuterin 40 mM, Tween 80 5 %, and 3 min of ultrasound treatment. Zeta potential was relatively high within -30 mV range in all designed nanoemulsions which indicates the nanoemulsion's stability. Also, the prepared nanoemulsions, despite initial particle size showed good stability in a 90-d storage period at 25 °C. In vivo assay, showed a significant improvement in the protection of apple fruit treated with reuterin-EOs nanoemulsions against fungal spoilage compared to free reuterin nanoemulsion. Treatment of apples with nanoemulsion containing 40 mM reuterin showed a maximum inhibitory effect on B. cinerea (5.1 mm lesion diameter compared to 29.2 mm for control fruit) within 7 d at 25 °C. In summary, the present study demonstrated that reuterin-synergistic EOs hybrid with boosted antifungal activities can be considered as a biopreservative for food applications.


Asunto(s)
Antifúngicos , Emulsiones , Gliceraldehído , Aceites Volátiles , Propano , Aceites Volátiles/farmacología , Aceites Volátiles/química , Emulsiones/farmacología , Propano/farmacología , Propano/química , Antifúngicos/farmacología , Antifúngicos/química , Gliceraldehído/farmacología , Gliceraldehído/análogos & derivados , Pruebas de Sensibilidad Microbiana , Limosilactobacillus reuteri/efectos de los fármacos , Penicillium/efectos de los fármacos , Penicillium/crecimiento & desarrollo , Botrytis/efectos de los fármacos , Botrytis/crecimiento & desarrollo , Alternaria/efectos de los fármacos , Alternaria/crecimiento & desarrollo
6.
Adv Sci (Weinh) ; 11(24): e2309725, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38647360

RESUMEN

The interplay between bacteria and their host influences the homeostasis of the human immune microenvironment, and this reciprocal interaction also affects the process of tissue damage repair. A variety of immunomodulatory commensal bacteria reside in the body, capable of delivering membrane vesicles (MVs) to host cells to regulate the local immune microenvironment. This research revealed, for the initial time, the significant enhancement of mucosal and cutaneous wound healing by MVs secreted by the human commensal Lactobacillus reuteri (RMVs) through modulation of the inflammatory environment in wound tissue. Local administration of RMVs reduces the proportion of pro-inflammatory macrophages in inflamed tissues and mitigates the level of local inflammation, thereby facilitating the healing of oral mucosa and cutaneous wounds. The elevated oxidative stress levels in activated pro-inflammatory macrophages can be modulated by RMVs, resulting in phenotypic transformation of macrophages. Furthermore, 3-hydroxypropionaldehyde present in RMVs can decrease the mitochondrial permeability of macrophages and stabilize the mitochondrial membrane potential, thereby promoting the conversion of macrophages to an anti-inflammatory phenotype. This study pioneers the significance of commensal bacterial MVs in tissue injury repair and presents a novel concept for the repair of tissue damage.


Asunto(s)
Limosilactobacillus reuteri , Macrófagos , Mitocondrias , Cicatrización de Heridas , Macrófagos/inmunología , Macrófagos/metabolismo , Cicatrización de Heridas/inmunología , Cicatrización de Heridas/fisiología , Animales , Ratones , Mitocondrias/metabolismo , Humanos , Modelos Animales de Enfermedad , Piel/microbiología , Piel/inmunología , Piel/metabolismo
7.
Iran J Microbiol ; 16(1): 90-96, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38682053

RESUMEN

Background and Objectives: Human herpes virus type 1 (HSV-1) is a neurotropic pathogen that is infected more than 70% of the world population. The increasing of viral resistance to antiviral drugs and the emergence of side effects has motivated researchers to study the use of probiotics as new antiviral agents. The aim of the present study was to study for the first time the potential antiviral activity of Lactobacillus reuteri (L. reuteri) supernatant against HSV-1. Materials and Methods: After measuring the cytotoxicity of L. reuteri supernatant by MTT assay, 1:16 dilution of it was added to HeLa cells before and after HSV-1 infection, after 1.5 hours incubation with HSV-1, and simultaneously with HSV-1 infection. After 48 hours of incubation at 37°C, the viral titer and expression levels of UL54, UL52 and UL27 genes were measured by tissue culture infectious dose 50 (TCID50 ) and Real-Time PCR methods, respectively. Results: HSV-1 titer in the treatment conditions before infection, incubation with HSV-1, simultaneously with infection and after infection was reduced by 0.42, 3.42, 1.83, and 0.83 log 10 TCID50/ml, respectively. When the bacterial supernatant was first incubated with the virus and then added to the cell, or when it was added simultaneously with the virus, the expression of the UL27, UL52, and UL54 genes decreased significantly (p<0.05). When the bacterial supernatant is added to the cell before or after virus infection, the expression of UL52 and UL54 genes does not change significantly (P>0.05). Conclusion: The study findings indicated that the supernatant of L. reuteri has a potent anti-HSV-1 effect especially if it is incubated with the virus before inoculation into the cell. Its possible antiviral mechanism is to inhibit the virus by binding to it or changing the surface structure of the virus. Metabolites of L. reuteri can be considered as a novel inhibitor of HSV-1 infection.

8.
Adv Sci (Weinh) ; 11(18): e2307233, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38487926

RESUMEN

The gut microbiome has emerged as a potential target for the treatment of cardiovascular disease. Ischemia/reperfusion (I/R) after myocardial infarction is a serious complication and whether certain gut bacteria can serve as a treatment option remains unclear. Lactobacillus reuteri (L. reuteri) is a well-studied probiotic that can colonize mammals including humans with known cholesterol-lowering properties and anti-inflammatory effects. Here, the prophylactic cardioprotective effects of L. reuteri or its metabolite γ-aminobutyric acid (GABA) against acute ischemic cardiac injury caused by I/R surgery are demonstrated. The prophylactic gavage of L. reuteri or GABA confers cardioprotection mainly by suppressing cardiac inflammation upon I/R. Mechanistically, GABA gavage results in a decreased number of proinflammatory macrophages in I/R hearts and GABA gavage no longer confers any cardioprotection in I/R hearts upon the clearance of macrophages. In vitro studies with LPS-stimulated bone marrow-derived macrophages (BMDM) further reveal that GABA inhibits the polarization of macrophages toward the proinflammatory M1 phenotype by inhibiting lysosomal leakage and NLRP3 inflammasome activation. Together, this study demonstrates that the prophylactic oral administration of L. reuteri or its metabolite GABA attenuates macrophage-mediated cardiac inflammation and therefore alleviates cardiac dysfunction after I/R, thus providing a new prophylactic strategy to mitigate acute ischemic cardiac injury.


Asunto(s)
Modelos Animales de Enfermedad , Limosilactobacillus reuteri , Ratones Endogámicos C57BL , Probióticos , Ácido gamma-Aminobutírico , Animales , Limosilactobacillus reuteri/metabolismo , Ratones , Ácido gamma-Aminobutírico/metabolismo , Probióticos/administración & dosificación , Probióticos/uso terapéutico , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Macrófagos/metabolismo , Microbioma Gastrointestinal , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/prevención & control
9.
Biol Direct ; 19(1): 23, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38500127

RESUMEN

BACKGROUND: This study seeks to investigate the impacts of Lactobacillus reuteri (L. reuteri) on hepatic ischemia-reperfusion (I/R) injury and uncover the mechanisms involved. METHODS: Mice in the I/R groups were orally administered low and high doses of L.reuteri (L.reuteri-low and L. reuteri-hi; 1 × 1010 CFU/d and 1 × 1011 CFU/d), for 4 weeks prior to surgery. Following this, mice in the model group were treated with an Nrf2 inhibitor (ML-385), palmitoylcarnitine, or a combination of both. RESULTS: After treatment with L. reuteri, mice exhibited reduced levels of serum aminotransferase (ALT), aspartate aminotransferase (AST), and myeloperoxidase (MPO) activity, as well as a lower Suzuki score and apoptosis rate. L. reuteri effectively reversed the I/R-induced decrease in Bcl2 expression, and the significant increases in the levels of Bax, cleaved-Caspase3, p-p65/p65, p-IκB/IκB, p-p38/p38, p-JNK/JNK, and p-ERK/ERK. Furthermore, the administration of L. reuteri markedly reduced the inflammatory response and oxidative stress triggered by I/R. This treatment also facilitated the activation of the Nrf2/HO-1 pathway. L. reuteri effectively counteracted the decrease in levels of beneficial gut microbiota species (such as Blautia, Lachnospiraceae NK4A136, and Muribaculum) and metabolites (including palmitoylcarnitine) induced by I/R. Likewise, the introduction of exogenous palmitoylcarnitine demonstrated a beneficial impact in mitigating hepatic injury induced by I/R. However, when ML-385 was administered prior to palmitoylcarnitine treatment, the previously observed effects were reversed. CONCLUSION: L. reuteri exerts protective effects against I/R-induced hepatic injury, and its mechanism may be related to the promotion of probiotic enrichment, differential metabolite homeostasis, and the Nrf2/HO-1 pathway, laying the foundation for future clinical applications.


Asunto(s)
Microbioma Gastrointestinal , Limosilactobacillus reuteri , Daño por Reperfusión , Ratones , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/uso terapéutico , Palmitoilcarnitina/uso terapéutico , Daño por Reperfusión/prevención & control , Daño por Reperfusión/tratamiento farmacológico , Isquemia
10.
Gut Microbes ; 16(1): 2313769, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38353638

RESUMEN

Melatonin has various physiological effects, such as the maintenance of circadian rhythms, anti-inflammatory functions, and regulation of intestinal barriers. The regulatory functions of melatonin in gut microbiota remodeling have also been well clarified; however, the role of gut microbiota in regulating host melatonin production remains poorly understood. To address this, we studied the contribution of gut microbiota to host melatonin production using gut microbiota-perturbed models. We demonstrated that antibiotic-treated and germ-free mice possessed diminished melatonin levels in the serum and elevated melatonin levels in the colon. The influence of the intestinal microbiota on host melatonin production was further confirmed by fecal microbiota transplantation. Notably, Lactobacillus reuteri (L. R) and Escherichia coli (E. coli) recapitulated the effects of gut microbiota on host melatonin production. Mechanistically, L. R and E. coli activated the TLR2/4/MyD88/NF-κB signaling pathway to promote expression of arylalkylamine N-acetyltransferase (AANAT, a rate-limiting enzyme for melatonin production), and MyD88 deficiency in colonic epithelial cells abolished the influence of intestinal microbiota on colonic melatonin production. Collectively, we revealed a specific underlying mechanism of gut microbiota to modulate host melatonin production, which might provide novel therapeutic ideas for melatonin-related diseases.


Asunto(s)
Microbioma Gastrointestinal , Melatonina , Animales , Ratones , Escherichia coli , Factor 88 de Diferenciación Mieloide/genética , Proteínas Adaptadoras Transductoras de Señales , Células Epiteliales
11.
Int J Biol Macromol ; 261(Pt 2): 129733, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38307433

RESUMEN

The function of miRNAs in intestinal inflammatory injury regulation has been studied extensively. However, the targeted delivery of these functional nucleic acid molecules to specific organs through encapsulation carriers and exerting their functional effects remain critical challenges for further research. Here, we constructed miR-146a-5p overexpression plasmid and validated the anti-inflammatory properties in the cell model. Then, the plasmid was encapsulated by the Pickering double emulsion system to investigate the role of Pickering double emulsion system in LPS-induced acute intestinal inflammatory injury. The results showed that the Pickering double emulsion system could effectively protect the integrity of plasmids in the intestinal tract, alleviate intestinal inflammatory injury, and upregulate the relative abundance of Lactobacillus reuteri. Mechanically, in vivo and in vitro experiments have shown that miR-146a-5p inhibits TLR4/NF-κB pathway to alleviate intestinal inflammation. In addition, miR-146a-5p can also regulate intestinal homeostasis by targeting the RNA polymerase sigma factor RpoD and α-galactosidase A, thereby affecting the growth of Lactobacillus reuteri. Above all, this study reveals a potential mechanism for miR-146a-5p to treat intestinal inflammation and provides a new delivery strategy for miRNAs to regulate intestinal homeostasis.


Asunto(s)
Microbioma Gastrointestinal , MicroARNs , Humanos , Emulsiones , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/genética
12.
Int J Biol Macromol ; 262(Pt 2): 130152, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38365143

RESUMEN

Supplementing probiotics or indigestible carbohydrates is a usual strategy to prevent or revert unhealthy states of the gut by reshaping gut microbiota. One criterion that probiotics are efficacious is the capacity to survive in the gastrointestinal tract. Biofilm is the common growth mode of microorganisms with high tolerances toward harsh environments. Suitable scaffolds are crucial for successful biofilm culture and large-scale production of biofilm-phenotype probiotics. However, the role of scaffolds containing indigestible carbohydrates in biofilm formation has not been studied. In this study, porous zein/cellulose composite scaffolds provided nitrogen sources and carbon sources simultaneously at the solid/liquid interfaces, being beneficial to the biofilm formation of Lactobacillus reuteri. The biofilms showed 2.1-17.4 times higher tolerances in different gastrointestinal conditions. In human fecal fermentation, the biofilms combined with the zein/cellulose composite scaffolds act as the "synbiotics" positively modulating the gut microbiota and the short-chain fatty acids (SCFAs), where biofilms provide probiotics and scaffolds provide prebiotics. The "synbiotics" show a more positive regulation ability than planktonic L. reuteri, presenting potential applications in gut health interventions. These results provide an understanding of the synergistic effects of biofilm-phenotype probiotics and indigestible carbohydrates contained in the "synbiotics" in gut microbiota modulation.


Asunto(s)
Microbioma Gastrointestinal , Limosilactobacillus reuteri , Probióticos , Simbióticos , Zeína , Humanos , Celulosa , Porosidad , Prebióticos , Carbohidratos , Biopelículas
13.
Microb Pathog ; 188: 106541, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38224920

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Probióticos , Caballos , Animales , Escherichia coli , Diarrea/terapia , Lactobacillus , Infecciones por Escherichia coli/terapia , Infecciones por Escherichia coli/veterinaria , Probióticos/uso terapéutico , Probióticos/farmacología
14.
Pediatr Neonatol ; 65(2): 145-151, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37684161

RESUMEN

BACKGROUND: Group B Streptococci (GBS) are common vaginal bacteria found in 20-30% of pregnant women and a significant cause of invasive infections in newborns. Recently, attention has been focused on the efficacy of probiotics during the perinatal period. However, the effect of probiotic intake on the mother-to-child transmission (MTCT) of GBS remains unknown. METHODS: Pregnant women with positive GBS results from vaginal and rectal swab cultures at 35-37 weeks of gestation were randomly assigned to the probiotic group or the control group in an open-label manner at the Department of Obstetrics and Gynecology, San-ikukai Hospital, Tokyo, Japan. The probiotic group received Lactobacillus reuteri during antenatal checkups from 35 to 37-week gestation to 1 month after delivery. Rectal swabs were obtained from the newborns at 5 days and at 1 month of age. Whole-genome sequencing was performed to test for GBS strains in the mother, whose newborn carried GBS at the 1-month checkup. Multi-locus sequence typing and single nucleotide polymorphism analyses were performed to identify MTCT. RESULTS: Overall, 67 mother-infant pairs were included, with 31 in the probiotic group and 36 in the control group. The positivity rate of GBS in newborns at 1 month of age was 10% (n = 3) in the probiotic group and 28% (n = 10) in the control group. In newborns carrying GBS at 1 month of age, genetic analysis revealed that the MTCT rate was 6% in the probiotic group and 22% in the control group, although the difference was not statistically significant (p = 0.0927). CONCLUSION: No statistically significant difference was found; however, the consumption of L. reuteri by women with GBS-positive pregnancies may inhibit the MTCT of GBS.


Asunto(s)
Complicaciones Infecciosas del Embarazo , Probióticos , Infecciones Estreptocócicas , Embarazo , Femenino , Recién Nacido , Humanos , Madres , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Estudios Prospectivos , Tipificación de Secuencias Multilocus , Infecciones Estreptocócicas/prevención & control , Streptococcus agalactiae , Probióticos/uso terapéutico
15.
Mol Nutr Food Res ; 68(2): e2300337, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38048544

RESUMEN

SCORE: This study identifies the coding gene (aldB) of acetolactate decarboxylase (ALDC) as an important regulatory gene of the intracellular pH in Lactobacillus reuteri (L. reuteri), uncovering the important role of ALDC in regulating intracellular pH, morphological features, and antagonism properties in the probiotic organism L. reuteri. METHODS AND RESULTS: The aldB mutant (ΔaldB) of L. reuteri is established using the homologous recombination method. Compare to the wild-type (WT) strain, the ΔaldB strain shows a smaller body size, grows more slowly, and contains more acid in the cell cytoplasm. The survival rate of the ΔaldB strain is much lower in low pH and simulated gastric fluid (SGF) than that of the WT strain, but higher in simulated intestinal fluid (SIF). The antagonism test demonstrates the ΔaldB strain can inhibit Listeria monocytogenes (L. monocytogenes) and Salmonella more effectively than the WT strain. Additionally, there is a dramatic decrease in the adhesion rate of Salmonella to Caco-2 and HT-29 cells in the presence of the ΔaldB strain compared to the WT strain. Simultaneously analyze, the auto-aggregation, co-aggregation, cell surface hydrophobicity (CSH), hemolytic, temperature, NaCl, oxidative stress, and antibiotic susceptibility of the ΔaldB strain are consistent with the features of probiotics. CONCLUSION: This study highlights that the aldB gene plays a significant role in the growth and antibacterial properties of L. reuteri.


Asunto(s)
Carboxiliasas , Limosilactobacillus reuteri , Probióticos , Humanos , Células CACO-2 , Probióticos/farmacología , Concentración de Iones de Hidrógeno
16.
Lett Appl Microbiol ; 77(1)2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38126116

RESUMEN

Fecal microbiota transplantation from patients with depression/inflammatory bowel disease (PDI) causes depression with gut inflammation in mice. Here, we investigated the effects of six Lactobacillus reuteri strains on brain-derived neurotropic factor (BDNF), serotonin, and interleukin (IL)-6 expression in neuronal or macrophage cells and PDI fecal microbiota-cultured microbiota (PcM)-induced depression in mice. Of these strains, L6 most potently increased BDNF and serotonin levels in corticosterone-stimulated SH-SY5Y and PC12 cells, followed by L3. L6 most potently decreased IL-6 expression in lipopolysaccharide (LPS)-stimulated macrophages. When L1 (weakest in vitro), L3, and L6 were orally administered in mice with PcM-induced depression, L6 most potently suppressed depression-like behaviors and hippocampal TNF-α and IL-6 expression and increased hippocampal serotonin, BDNF, 5HT7, GABAARα1, and GABABR1b expression, followed by L3 and L1. L6 also suppressed TNF-α and IL-6 expression in the colon. BDNF or serotonin levels in corticosterone-stimulated neuronal cells were negatively correlated with depression-related biomarkers in PcM-transplanted mice, while IL-6 levels in LPS-stimulated macrophage were positively correlated. These findings suggest that IL-6 expression-suppressing and BDNF/serotonin expression-inducing LBPs in vitro, particularly L6, may alleviate gut microbiota-involved depression with colitis in vivo.


Asunto(s)
Microbioma Gastrointestinal , Limosilactobacillus reuteri , Neuroblastoma , Ratas , Humanos , Ratones , Animales , Interleucina-6/genética , Depresión/terapia , Factor de Necrosis Tumoral alfa/genética , Lipopolisacáridos/toxicidad , Corticosterona/farmacología , Serotonina/farmacología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Ansiedad/terapia , Ansiedad/etiología , Ratones Endogámicos C57BL
17.
Biomedicines ; 11(12)2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-38137342

RESUMEN

This study aimed to elucidate the unique chemical compositions of plasma-activated water (PAW) and the potential antibacterial efficacy of PAW as a novel vaginal cleanser. We analyzed the ion compositions (four anions: F-, Cl-, NO3-, SO42-; five cations: Na+, NH4+, K+, Mg2+, Ca2+) of several formulations of PAW generated at different electrical powers (12 and 24 V) at various treatment time points (1, 10, and 20 min), and stay durations (immediate, 30, and 60 min). As treatment duration increased, hypochlorous acid (HOCl), Ca2+, and Mg2+ concentrations increased and Cl- concentration decreased. Higher electrical power and longer treatment duration resulted in increased HOCl levels, which acts to prevent the growth of general microorganisms. Notably, PAW had no antibacterial effects against the probiotic, Lactobacillus reuteri, which produces lactic acid and is important for vaginal health. These findings indicate that PAW contains HOCl and some cations (Ca2+ and Mg2+), which should help protect against pathogens of the vaginal mucosa and have a cleansing effect within the vaginal environment while not harming beneficial bacteria.

18.
J Agric Food Chem ; 71(48): 19101-19110, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37988599

RESUMEN

There is an increasing global demand for probiotics because of their numerous health benefits. However, a significant percentage of commercially available probiotic products have microbial quantities that are not in accordance with their product labels. In quantifying bacteria, the viable plate count is the standard method but is considered laborious and time-consuming. We demonstrate the use of an attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy coupled with chemometrics to quantify a pure culture of Lactobacillus reuteri (L. reuteri) ProTectis grown in deMan, Rogosa, and Sharpe broth. The chemometric partial least-squares model generated was able to accurately quantify viable plate count (VPC) (root-mean-square error of cross-validation (RMSECV) = 0.115 log CFU mL-1, root-mean-square error of prediction (RMSEP) = 0.145 log CFU mL-1, R2 = 0.982). These results provide proof of concept for this quantification technique and can potentially be developed and applied for the quantification of L. reuteri ProTectis in food products.


Asunto(s)
Limosilactobacillus reuteri , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Quimiometría , Análisis de Fourier , Análisis de los Mínimos Cuadrados
19.
Exp Neurobiol ; 32(5): 313-327, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37927130

RESUMEN

Mental health is influenced by the gut-brain axis; for example, gut dysbiosis has been observed in patients with major depressive disorder (MDD). Gut microbial changes by fecal microbiota transplantation or probiotics treatment reportedly modulates depressive symptoms. However, it remains unclear how gut dysbiosis contributes to mental dysfunction, and how correction of the gut microbiota alleviates neuropsychiatric disorders. Our previous study showed that chronic consumption of Lactobacillus reuteri ATG-F4 (F4) induced neurometabolic alterations in healthy mice. Here, we investigated whether F4 exerted therapeutic effects on depressive-like behavior by influencing the central nervous system. Using chronic unpredictable stress (CUS) to induce anhedonia, a key symptom of MDD, we found that chronic F4 consumption alleviated CUS-induced anhedonic behaviors, accompanied by biochemical changes in the gut, serum, and brain. Serum and brain metabolite concentrations involved in tryptophan metabolism were regulated by CUS and F4. F4 consumption reduced the elevated levels of serotonin (5-HT) in the brain observed in the CUS group. Additionally, the increased expression of Htr1a, a subtype of the 5-HT receptor, in the medial prefrontal cortex (mPFC) of stressed mice was restored to levels observed in stress-naïve mice following F4 supplementation. We further demonstrated the role of Htr1a using AAV-shRNA to downregulate Htr1a in the mPFC of CUS mice, effectively reversing CUS-induced anhedonic behavior. Together, our findings suggest F4 as a potential therapeutic approach for relieving some depressive symptoms and highlight the involvement of the tryptophan metabolism in mitigating CUS-induced depressive-like behaviors through the action of this bacterium.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA