Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
Benef Microbes ; 15(5): 527-536, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39147374

RESUMEN

The aim of this animal study was to compare the primary/secondary stability and micro-CT bone and tissue volumes of implants that were immersed in Limosilactobacillus reuteri, cholecalciferol-D3 (vitamin D) and injectable platelet-rich fibrin (i-PRF) suspensions/solutions before placement in bone. 40 implants (10 in each group) were placed in the iliac crest of 5 sheep. The implants were immersed in L. reuteri, vitamin D or i-PRF solutions for five minutes before placement or left unsoaked as controls. Implant stability was determined by ISQ values and bone volume around implants was histomorphometrically analysed by micro-CT evaluation. At 4 weeks, implants in the L. reuteri group showed the highest secondary stability and 2- and 3D BV/TV values. Both L. reuteri and vitamin D immersed implants had higher osseointegration values compared to the implants in the i-PRF group and controls. There were no statistical differences between L. reuteri and vitamin D immersed implants. Within the limits of the study, the results suggest that immersing implants in L. reuteri or vitamin D suspensions/solutions before implant placement in bone may have beneficial effects on osseointegration.


Asunto(s)
Implantes Dentales , Limosilactobacillus reuteri , Oseointegración , Animales , Implantes Dentales/microbiología , Limosilactobacillus reuteri/fisiología , Ovinos , Oseointegración/efectos de los fármacos , Fibrina Rica en Plaquetas , Microtomografía por Rayos X , Colecalciferol/farmacología , Colecalciferol/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos
2.
Dev Comp Immunol ; 160: 105241, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39121939

RESUMEN

Probiotics play an essential role in the largemouth bass (Micropterus salmoides) aquaculture sector. They aid the fish in sickness prevention, intestinal structure improvement, food absorption, and immune system strengthening. In this experiment, Bacillus subtilis (BS, 107 CFU/g) and Lactobacillus reuteri (LR, 107 CFU/g) were added to the feed and then fed to M. salmoides for 35 days. The effects of two probiotics on the growth, immunity, and metabolism of M. salmoides organisms were studied. The results revealed that the BS group significantly increased the growth rate and specific growth rate of M. salmoides, while both the BS and LR groups significantly increase the length of villi M. salmoides intestines. The BS group significantly increased the levels of AKP, T-AOC, and CAT in the blood of M. salmoides, as well as AKP levels in the intestine. Furthermore, the BS group significantly increased the expression of intestinal genes Nrf2, SOD1, GPX, and CAT, while significantly decreasing the expression of the keap1 gene. M. salmoides gut microbial analysis showed that the abundance of Planctomycetota was significantly different in both control and experimental groups. Analyzed at the genus level, the abundance of Citrobacter, Paracoccus, Luedemannella, Sphingomonas, Streptomyces and Xanthomonas in the both control and experimental groups were significantly different. The BS group's differentially expressed genes were predominantly enriched in oxidative phosphorylation pathways in the intestine, indicating that they had a good influence on intestinal metabolism and inflammation suppression. In contrast, differentially expressed genes in the LR group were primarily enriched in the insulin signaling and linoleic acid metabolism pathways, indicating improved intestine metabolic performance. In conclusion, B. subtilis and L. reuteri improve the growth and health of M. salmoides, indicating tremendous potential for enhancing intestinal metabolism and providing significant application value.


Asunto(s)
Alimentación Animal , Bacillus subtilis , Lubina , Suplementos Dietéticos , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Probióticos , Animales , Probióticos/administración & dosificación , Lubina/inmunología , Lubina/crecimiento & desarrollo , Lubina/microbiología , Limosilactobacillus reuteri/inmunología , Limosilactobacillus reuteri/fisiología , Microbioma Gastrointestinal/inmunología , Intestinos/inmunología , Intestinos/microbiología , Acuicultura , Proteínas de Peces/metabolismo , Proteínas de Peces/genética
3.
Front Cell Infect Microbiol ; 14: 1401462, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39091675

RESUMEN

Introduction: Bacterial urinary tract infections (UTI) are among the most common infectious diseases worldwide. The rise of multidrug-resistant (MDR) uropathogenic Escherichia coli (UPEC) UTI cases is a significant threat to healthcare systems. Several probiotic bacteria have been proposed as an alternative to combat MDR UTI. Lactic acid bacteria in the genus Limosilactobacillus are some of the most studied and used probiotics. However, strain-specific effects play a critical role in probiotic properties. L. reuteri KUB-AC5 (AC5), isolated from the chicken gut, confers antimicrobial and immunobiotic effects against some human pathogens. However, the antibacterial and immune modulatory effects of AC5 on UPEC have never been explored. Methods: Here, we investigated both the direct and indirect effects of AC5 against UPEC isolates (UTI89, CFT073, and clinical MDR UPEC AT31) in vitro. Using a spot-on lawn, agar-well diffusion, and competitive growth assays, we found that viable AC5 cells and cell-free components of this probiotic significantly reduced the UPEC growth of all strains tested. The human bladder epithelial cell line UM-UC-3 was used to assess the adhesion and pathogen-attachment inhibition properties of AC5 on UPEC. Results and discussion: Our data showed that AC5 can attach to UM-UC-3 and decrease UPEC attachment in a dose-dependent manner. Pretreatment of UPEC-infected murine macrophage RAW264.7 cells with viable AC5 (multiplicity of infection, MOI = 1) for 24 hours enhanced macrophage-killing activity and increased proinflammatory (Nos2, Il6, and Tnfa) and anti-inflammatory (Il10) gene expression. These findings indicate the gut-derived AC5 probiotic could be a potential urogenital probiotic against MDR UTI.


Asunto(s)
Limosilactobacillus reuteri , Macrófagos , Probióticos , Escherichia coli Uropatógena , Probióticos/farmacología , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/inmunología , Limosilactobacillus reuteri/fisiología , Animales , Ratones , Macrófagos/inmunología , Macrófagos/microbiología , Humanos , Urotelio/microbiología , Infecciones Urinarias/microbiología , Infecciones Urinarias/prevención & control , Línea Celular , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Células RAW 264.7 , Células Epiteliales/microbiología , Pollos , Adhesión Bacteriana/efectos de los fármacos
4.
Poult Sci ; 103(10): 104072, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39068698

RESUMEN

The continuous expansion of intestinal stem cells (ISCs) is crucial for maintaining the renewal of the intestinal epithelium, particularly in inflammatory conditions. It remains largely unknown how the internal microbiota repair damage to the internal mucosal barrier. Hence, investigating potential anti-inflammatory probiotics from the intestinal symbolic microbes of broilers and analyzing their mechanism of action to support the intestinal mucosal barrier function can offer novel regulatory tools to alleviate broiler enteritis. In this research, we utilized in vivo broilers plus ex vivo organoids model to thoroughly examine the effectiveness of Lactobacillus reuteri (LR) in protecting the integrity of the intestinal mucosa during lipopolysaccharide-induced (LPS-induced) enteritis in broilers. The findings indicated that LR feeding maintained intestinal morphological and structural integrity, enhanced proliferation of intestinal epithelial cells, and inhibited cell apoptosis and inflammatory response against the deleterious effects triggered by LPS. Simultaneously, LR enhanced ISCs activity and stimulated intestinal epithelial regeneration to protect the intestinal barrier during LPS-induced injury conditions. The coculture system of LR and ileum organoids revealed that LR increased the growth of organoids and attenuated LPS-stimulated damage to organoids. Furthermore, the LPS-induced decrease in ISC activity was rescued by reactivation of Wnt/ß-catenin signaling by LR ex vivo and in vivo. This research revealed that LR promoted the expansion of ISCs and intestinal epithelial cell renewal by regulating the Wnt/ß-catenin signaling pathway, thereby maintaining the integrity of the intestinal mucosal barrier. This finding provided theoretical support for lactobacillus as a probiotic additive in livestock feed to improve intestinal inflammation and treat intestinal diseases.


Asunto(s)
Pollos , Mucosa Intestinal , Limosilactobacillus reuteri , Lipopolisacáridos , Probióticos , Células Madre , Vía de Señalización Wnt , Animales , Limosilactobacillus reuteri/fisiología , Probióticos/farmacología , Probióticos/administración & dosificación , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Vía de Señalización Wnt/efectos de los fármacos , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/inducido químicamente , Enfermedades de las Aves de Corral/prevención & control , Enteritis/veterinaria , Enteritis/inducido químicamente , Enteritis/prevención & control , Enteritis/microbiología , Dieta/veterinaria , Masculino , Alimentación Animal/análisis
5.
Adv Exp Med Biol ; 1449: 157-174, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39060737

RESUMEN

The most frequent functional gastrointestinal disorders (FGID) in children include infantile colic, constipation, functional abdominal pain (FAP), and irritable bowel syndrome (IBS). Unfortunately, treatment options for FGID in children are limited, therefore many dietary interventions have been evaluated, including probiotics. This chapter summarizes currently available evidence and recommendations for probiotic use in the treatment of frequent FGIDs in children. The strongest evidence exists for the use of Limosilactobacillus (L.) reuteri DSM 17938 and Bifidobacterium animalis subsp. lactis BB-12 for the treatment of infantile colic in breastfed infants. Limited but yet encouraging evidence exists for Lacticaseibacillus rhamnosus GG (LGG) for the treatment of IBS and L. reuteri DSM 17938 for FAP.


Asunto(s)
Enfermedades Gastrointestinales , Probióticos , Probióticos/uso terapéutico , Humanos , Enfermedades Gastrointestinales/terapia , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/dietoterapia , Niño , Lactante , Síndrome del Colon Irritable/terapia , Síndrome del Colon Irritable/dietoterapia , Síndrome del Colon Irritable/microbiología , Limosilactobacillus reuteri/fisiología
6.
Mol Nutr Food Res ; 68(14): e2300927, 2024 Jul.
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.


Asunto(s)
Ácidos y Sales Biliares , Etanol , Limosilactobacillus reuteri , Ratones Endogámicos C57BL , Receptores Citoplasmáticos y Nucleares , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Limosilactobacillus reuteri/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Animales , Receptores Citoplasmáticos y Nucleares/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos , Ácidos y Sales Biliares/metabolismo , Ratones , Hepatopatías Alcohólicas/prevención & control , Probióticos/farmacología , Factores de Crecimiento de Fibroblastos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Humanos
7.
Food Funct ; 15(12): 6629-6641, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38812427

RESUMEN

Gastrointestinal inflammation and intestinal barrier function have important effects on human health. Alcohol, an important foodborne hazard factor, damages the intestinal barrier, increasing the risk of disease. Lactobacillus reuteri strains have been reported to reduce gastrointestinal inflammation and strengthen the intestinal barrier. In this study, we selected three anti-inflammatory L. reuteri strains to evaluate their role in the protection of the intestinal barrier and their immunomodulatory activity in a mouse model of gradient alcohol intake. Among the three strains tested (FSCDJY33M3, FGSZY33L6, and FCQHCL8L6), L. reuteri FSCDJY33M3 was found to protect the intestinal barrier most effectively, possibly due to its ability to reduce the expression of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-alpha (TNF-α) and increase the expression of tight junction proteins (occludin, claudin-3). Genomic analysis suggested that the protective effects of L. reuteri FSCDJY33M3 may be related to functional genes and glycoside hydrolases associated with energy production and conversion, amino acid transport and metabolism, carbohydrate transport and metabolism, and DNA replication, recombination, and repair. These genes include COG2856, COG1804, COG2071, and COG1061, which encode adenine deaminase, acyl-CoA transferases, glutamine amidotransferase, RNA helicase, and glycoside hydrolases, including GH13_20, GH53, and GH70. Our results identified functional genes that may be related to protection against alcohol-induced intestinal barrier damage, which might be useful for screening lactic acid bacterial strains that can protect the intestinal barrier.


Asunto(s)
Etanol , Mucosa Intestinal , Limosilactobacillus reuteri , Probióticos , Limosilactobacillus reuteri/fisiología , Animales , Ratones , Mucosa Intestinal/metabolismo , Probióticos/farmacología , Masculino , Ratones Endogámicos C57BL , Humanos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Intestinos/microbiología
8.
PLoS One ; 19(5): e0304358, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38820403

RESUMEN

Osteoporosis is an important health problem that occurs due to an imbalance between bone formation and resorption. Hormonal deficiency post-menopause is a significant risk factor. The probiotic Limosilactobacillus reuteri has been reported to prevent ovariectomy (Ovx)-induced bone loss in mice and reduce bone loss in postmenopausal women. Despite the numerous health benefits of probiotics, as they are live bacteria, the administration is not risk-free for certain groups (e.g., neonates and immunosuppressed patients). We evaluated the effects of L. reuteri (ATCC PTA 6475) and its heat-killed (postbiotic) form on Ovx-induced bone loss. Adult female mice (BALB/c) were randomly divided into four groups: group C-control (sham); group OVX-C-Ovx; group OVX-POS-Ovx + heat-killed probiotic; group OVX-PRO-Ovx + probiotic. L. reuteri or the postbiotic was administered to the groups (1.3x109 CFU/day) by gavage. Bacterial morphology after heat treatment was accessed by scanning electron microscopy (SEM). The treatment started one week after Ovx and lasted 28 days (4 weeks). The animals were euthanized at the end of the treatment period. Bone microarchitecture and ileum Occludin and pro-inflammatory cytokines gene expression were evaluated by computed microtomography and qPCR techniques, respectively. The Ovx groups had lower percentage of bone volume (BV/TV) and number of bone trabeculae as well as greater total porosity compared to the control group. Treatment with live and heat-killed L. reuteri resulted in higher BV/TV and trabecular thickness than the Ovx group. The heat treatment caused some cell surface disruptions, but its structure resembled that of the live probiotic in SEM analysis. There were no statistical differences in Occludin, Il-6 and Tnf-α gene expression. Both viable and heat-killed L. reuteri prevented bone loss on ovariectomized mice, independently of gut Occludin and intestinal Il-6 and Tnf-α gene expression.


Asunto(s)
Limosilactobacillus reuteri , Osteoporosis , Ovariectomía , Probióticos , Animales , Femenino , Limosilactobacillus reuteri/fisiología , Probióticos/administración & dosificación , Probióticos/farmacología , Ratones , Osteoporosis/prevención & control , Ratones Endogámicos BALB C , Calor
9.
Microbiol Immunol ; 68(7): 213-223, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38747013

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Probióticos , Microbioma Gastrointestinal/efectos de los fármacos , Limosilactobacillus reuteri/fisiología , Limosilactobacillus reuteri/metabolismo , Animales , Lesión Renal Aguda/prevención & control , Lesión Renal Aguda/metabolismo , Ratones , Lipopolisacáridos/metabolismo , Masculino , Riñón/microbiología , Riñón/metabolismo , Heces/microbiología , Modelos Animales de Enfermedad , Creatinina/sangre , Ratones Endogámicos C57BL , Apoptosis/efectos de los fármacos
10.
Vet Res Commun ; 48(4): 2331-2342, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38771449

RESUMEN

Pentatrichomonas hominis is a common intestinal parasitic protozoan that causes abdominal pain and diarrhea, and poses a zoonotic risk. Probiotics, known for enhancing immunity and pathogen resistance, hold promise in combating parasitic infections. This study aimed to evaluate two porcine-derived probiotics, Lactobacillus reuteri LR1 and Lactobacillus plantarum LP1, against P. hominis infections in pigs. Taxonomic identity was confirmed through 16 S rRNA gene sequencing, with L. reuteri LR1 belonging to L. reuteri species and L. plantarum LP1 belonging to L. plantarum species. Both probiotics exhibited robust in vitro growth performance. Co-culturing intestinal porcine epithelial cell line (IPEC-J2) with these probiotics significantly improved cell viability compared with the control group. Pre-incubation probiotics significantly enhanced the mRNA expression of anti-oxidative response genes in IPEC-J2 cells compared with the PHGD group, with L. reuteri LR1 and L. plantarum LP1 significantly up-regulating CuZn-SOD、CAT and Mn-SOD genes expression (p < 0.05). The anti-oxidative stress effect of L. reuteri LR1 was significantly better than that of L. plantarum LP1 (p < 0.05). Furthermore, pre-incubation with the probiotics alleviated the P. hominis-induced inflammatory response. L. reuteri LR1 and L. plantarum LP1 significantly down-regulated IL-6、IL-8 and TNF-α gene expression(p < 0.05) compared with the PHGD group. The probiotics also mitigated P. hominis-induced apoptosis. L. reuteri LR1 and L. plantarum LP1 significantly down-regulated Caspase3 and Bax gene expression (p < 0.05), significantly up-regulated Bcl-2 gene expression (p < 0.05) compared with the PHGD group. Among them, L. plantarum LP1 showed better anti-apoptotic effect. These findings highlight the probiotics for mitigating P. hominis infections in pigs. Their ability to enhance anti-oxidative responses, alleviate inflammation, and inhibit apoptosis holds promise for therapeutic applications. Simultaneously, probiotics can actively contribute to inhibiting trichomonal infections, offering a novel approach for preventing and treating diseases such as P. hominis. Further in vivo studies are required to validate these results and explore their potential in animal and human health.


Asunto(s)
Lactobacillus plantarum , Probióticos , Animales , Probióticos/farmacología , Porcinos , Línea Celular , Lactobacillus plantarum/fisiología , Limosilactobacillus reuteri/fisiología , Trichomonadida/fisiología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/parasitología
11.
J Agric Food Chem ; 70(38): 11935-11943, 2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36111836

RESUMEN

Bacteria colonizing the gastrointestinal tract generally grow well in biofilms. In recent years, probiotic biofilms have been considered the most promising fourth-generation probiotics. However, the research into the functions of probiotic biofilms is just starting. In this study, Lactobacillus reuteri DSM 17938 biofilms formed on electrospun cellulose acetate nanofibrous scaffolds were contrasted with planktonic cells. Pathogen inhibition analysis of Escherichia coli, Staphylococcus aureus, and Listeria monocytogenes suggested a significant distinction between the planktonic and biofilm groups. In human fecal fermentation, L. reuteri remodeled the microbiota by decreasing the relative abundances of Proteobacteria, Escherichia-Shigella, and Desulfovibrio and increasing the relative abundances of Phascolarctobacterium, Bacteroides, and Lactobacillus. Moreover, L. reuteri biofilms played more positive roles in microbiota modulation and short-chain fatty acid production than planktonic L. reuteri. These findings provide an understanding of the beneficial effects of probiotic biofilms, laying a foundation for the application of probiotic biofilms as a health promoter.


Asunto(s)
Limosilactobacillus reuteri , Microbiota , Probióticos , Biopelículas , Ácidos Grasos Volátiles/farmacología , Fermentación , Humanos , Limosilactobacillus reuteri/fisiología , Probióticos/farmacología
12.
Front Immunol ; 13: 897395, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35911699

RESUMEN

Intestinal epithelial barrier injury disrupts immune homeostasis and leads to many intestinal disorders. Lactobacillus reuteri (L. reuteri) strains can influence immune system development and intestinal function. However, the underlying mechanisms of L. reuteri LR1 that regulate inflammatory response and intestinal integrity are still unknown. The present study aimed to determine the effects of LR1 on the ETEC K88-induced intestinal epithelial injury on the inflammatory response, intestinal epithelial barrier function, and the MLCK signal pathway and its underlying mechanism. Here, we showed that the 1 × 109 cfu/ml LR1 treatment for 4 h dramatically decreased interleukin-8 (IL-8) and IL-6 expression. Then, the data indicated that the 1 × 108 cfu/ml ETEC K88 treatment for 4 h dramatically enhanced IL-8, IL-6, and tumor necrosis factor-α (TNF-α) expression. Furthermore, scanning electron microscope (SEM) data indicated that pretreatment with LR1 inhibited the ETEC K88 that adhered on IPEC-J2 and alleviated the scratch injury of IPEC J2 cells. Moreover, LR1 pretreatment significantly reversed the declined transepithelial electrical resistance (TER) and tight junction protein level, and enhanced the induction by ETEC K88 treatment. Additionally, LR1 pretreatment dramatically declined IL-8, IL-17A, IL-6, and TNF-α levels compared with the ETEC K88 group. Then, ETEC K88-treated IPEC-J2 cells had a higher level of myosin light-chain kinase (MLCK), higher MLC levels, and a lower Rho-associated kinase (ROCK) level than the control group, while LR1 pretreatment significantly declined the MLCK and MLC expression and enhanced ROCK level in the ETEC K88-challenged IPEC-J2 cells. Mechanistically, depletion of MLCK significantly declined MLC expression in IPEC-J2 challenged with ETEC K88 compared to the si NC+ETEC K88 group. On the other hand, the TER of the si MLCK+ETEC K88 group was higher and the FD4 flux in the si MLCK+ETEC K88 group was lower compared with the si NC+ETEC K88 group. In addition, depletion of MLCK significantly enhanced Claudin-1 level and declined IL-8 and TNF-α levels in IPEC-J2 pretreated with LR1 followed by challenging with ETEC K88. In conclusion, our work indicated that L. reuteri LR1 can decline inflammatory response and improve intestinal epithelial barrier function through suppressing the MLCK signal pathway in the ETEC K88-challenged IPEC-J2.


Asunto(s)
Escherichia coli Enterotoxigénica , Infecciones por Escherichia coli , Mucosa Intestinal , Limosilactobacillus reuteri , Animales , Línea Celular , Escherichia coli Enterotoxigénica/inmunología , Infecciones por Escherichia coli/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Enfermedades Intestinales/microbiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiología , Limosilactobacillus reuteri/fisiología , Quinasa de Cadena Ligera de Miosina/metabolismo , Transducción de Señal , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
13.
Front Immunol ; 13: 865982, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35320932

RESUMEN

Defects in gut barrier function are implicated in gastrointestinal (GI) disorders like inflammatory bowel disease (IBD), as well as in systemic inflammation. With the increasing incidence of IBD worldwide, more attention should be paid to dietary interventions and therapeutics with the potential to boost the natural defense mechanisms of gut epithelial cells. The current study aimed to investigate the protective effects of Limosilactobacillus reuteri ATCC PTA 4659 in a colitis mouse model and delineate the mechanisms behind it. Wild-type mice were allocated to the control group; or given 3% dextran sulfate sodium (DSS) in drinking water for 7 days to induce colitis; or administered L. reuteri for 7 days as pretreatment; or for 14 days starting 7 days before subjecting to the DSS. Peroral treatment with L. reuteri improved colitis severity clinically and morphologically and reduced the colonic levels of Tumor necrosis factor-α (TNF-α) (Tnf), Interleukin 1-ß (Il1ß), and nterferon-γ (Ifng), the crucial pro-inflammatory cytokines in colitis onset. It also prevented the CD11b+Ly6G+ neutrophil recruitment and the skewed immune responses in mesenteric lymph nodes (MLNs) of CD11b+CD11c+ dendritic cell (DC) expansion and Foxp3+CD4+ T-cell reduction. Using 16S rRNA gene amplicon sequencing and RT-qPCR, we demonstrated a colitis-driven bacterial translocation to MLNs and gut microbiota dysbiosis that were in part counterbalanced by L. reuteri treatment. Moreover, the expression of barrier-preserving tight junction (TJ) proteins and cytoprotective heat shock protein (HSP) 70 and HSP25 was reduced by colitis but boosted by L. reuteri treatment. A shift in expression pattern was also observed with HSP70 in response to the pretreatment and with HSP25 in response to L. reuteri-DSS. In addition, the changes of HSPs were found to be correlated to bacterial load and epithelial cell proliferation. In conclusion, our results demonstrate that the human-derived L. reuteri strain 4659 confers protection in experimental colitis in young mice, while intestinal HSPs may mediate the probiotic effects by providing a supportive protein-protein network for the epithelium in health and colitis.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Limosilactobacillus reuteri , Animales , Colitis/patología , Sulfato de Dextran/toxicidad , Proteínas de Choque Térmico , Limosilactobacillus reuteri/fisiología , Ratones , ARN Ribosómico 16S/genética , Proteínas de Uniones Estrechas , Factor de Necrosis Tumoral alfa
14.
Nutrients ; 14(6)2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35334930

RESUMEN

Cardiovascular disease has one of the highest global incidences and mortality rates. Atherosclerosis is the main cause of cardiovascular disease, and hypercholesterolaemia and hyperlipidaemia are the main risk factors for the development of atherosclerosis. Decreasing serum cholesterol and triglyceride concentrations is considered to be an effective strategy to prevent atherosclerotic cardiovascular disease. Previous studies have shown that many diseases are related to gut microbiota dysbiosis. The positive regulation of the gut microbiota by probiotics may prevent or treat certain diseases. In this study, Lactobacillus reuteri CCFM8631 treatment was shown to decrease plasma total cholesterol (TC), low-density lipoprotein-cholesterol, aspartate transaminase, alanine transaminase and trimethylamine N-oxide concentrations, decrease liver TC and malondialdehyde concentrations and recover liver superoxide dismutase concentrations in mice fed a Paigen atherogenic diet. In addition, L. reuteri increased the faecal short-chain fatty acid content (acetate, propionate and butyrate), which was accompanied by an increase in the relative abundance of faecal Deferribacteres, Lachnospiraceae NK4A136 group, Lactobacillus and Dubosiella; a decrease in the relative abundance of Erysipelatoclostridium and Romboutsia and the activation of butanoate and vitamin B6 metabolism, leading to the alleviation of hypercholesterolaemia.


Asunto(s)
Microbioma Gastrointestinal , Hipercolesterolemia , Hiperlipidemias , Limosilactobacillus reuteri , Animales , Dieta Aterogénica , Hiperlipidemias/etiología , Limosilactobacillus reuteri/fisiología , Ratones
15.
Biochem Biophys Res Commun ; 593: 137-143, 2022 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-35066403

RESUMEN

Depression, characterized by low mood, is a complex mental disorder that is a serious threat to human health. Depression is thought to be caused by a combination of genetic, environmental and psychological factors. However, the pathophysiology of depression remains unclear. In the present study, we found that Dcf1 knockout (KO) mice had depression-like symptoms and disruptive changes in gamma-aminobutyric acid (GABA) concentration and GABA receptor expression were found in the hippocampus of Dcf1 KO and WT mice. Furthermore, the gut microbiota composition of Dcf1 KO mice was significantly different from that of wildtype (WT) mice and Dcf1 KO mice showed lower Firmicutes and Lactobacillus content compared to WT mice. In addition, the depression-like behavior of Dcf1 KO mice was alleviated by the administration of microbiota. More surprisingly, after treatment with Lactobacillus murine and Lactobacillus reuteri, two Lactobacillus species with proportionally greater differences in content between the WT and KO groups, KO mice showed similar GABA content, as well as restored GABA-related receptor expression, as the WT group. Our data elucidated a possible mechanism of depression induction by gut microbiota in Dcf1 KO mice and provide a new avenue to explore the treatment of depression by gut microbiota.


Asunto(s)
Depresión/terapia , Microbioma Gastrointestinal , Intestinos/trasplante , Lactobacillus/fisiología , Limosilactobacillus reuteri/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Probióticos/administración & dosificación , Animales , Depresión/etiología , Depresión/metabolismo , Depresión/patología , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Sci Rep ; 11(1): 23567, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34876641

RESUMEN

Optimisation of cultivation conditions in the industrial production of probiotics is crucial to reach a high-quality product with retained probiotic functionality. Flow cytometry-based descriptors of bacterial morphology may be used as markers to estimate physiological fitness during cultivation, and can be applied for online monitoring to avoid suboptimal growth. In the current study, the effects of temperature, initial pH and oxygen levels on cell growth and cell size distributions of Limosilactobacillus reuteri DSM 17938 were measured using multivariate flow cytometry. A pleomorphic behaviour was evident from the measurements of light scatter and pulse width distributions. A pattern of high growth yielding smaller cells and less heterogeneous populations could be observed. Analysis of pulse width distributions revealed significant morphological heterogeneities within the bacterial cell population under non-optimal growth conditions, and pointed towards low temperature, high initial pH, and high oxygen levels all being triggers for changes in morphology towards cell chain formation. However, cell size did not correlate to survivability after freeze-thaw or freeze-drying stress, indicating that it is not a key determinant for physical stress tolerance. The fact that L. reuteri morphology varies depending on cultivation conditions suggests that it can be used as marker for estimating physiological fitness and responses to its environment.


Asunto(s)
Limosilactobacillus reuteri/citología , Limosilactobacillus reuteri/crecimiento & desarrollo , Probióticos , Técnicas Bacteriológicas , Citometría de Flujo , Liofilización , Humanos , Concentración de Iones de Hidrógeno , Limosilactobacillus reuteri/fisiología , Microscopía Electrónica de Rastreo , Oxígeno , Fenotipo , Probióticos/aislamiento & purificación , Estrés Fisiológico , Temperatura
17.
BMC Microbiol ; 21(1): 271, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34615458

RESUMEN

BACKGROUND: The purpose of the study was to investigate the effect of probiotics on biofilm acidogenicity and on the number of salivary Streptococcus mutans and lactobacilli in orthodontic patients. METHODS: This RCT was conducted on 28 young adults who were undergoing orthodontic treatment. The short-term prospective clinical trial lasted for three weeks. The test group rinsed daily with drops containing two Lactobacillus reuteri strains diluted in water, while the placebo group used drops without probiotics. The subjects were enrolled eight months since the beginning of orthodontic treatment. Plaque-pH, saliva and dental biofilm samples were obtained at baseline, one week and three weeks post intervention. RESULTS: Twenty-seven subjects successfully completed the trial period, only one drop out in the test group. No side effects were reported. A statistically significant increase in plaque pH at three weeks post-intervention was found for the test group (p < 0.05), while insignificant changes in the pH value were found for the placebo group in comparison to baseline (p > 0.05). In addition, the AUC7.0 showed a significant difference at three weeks between the test and placebo (p = 0.00002). The three-week samples of stimulated whole saliva showed a statistically insignificant difference in the number of S. mutans and lactobacilli between the two groups (p > 0.05). The qPCR analysis showed the ability of the two strains to get colonized in the dental biofilm without a significant effect on the microbial counts. CONCLUSION/CLINICAL IMPLICATIONS: A mixture of Lactobacillus reuteri has the ability to reduce the pH fall at the three-week follow-up. However, the short-term use of probiotics does not appear to have an effect on the number of salivary Streptococcus mutans and lactobacilli in saliva and on the dental biofilm. TRIAL REGISTRATION: Clinicaltrial.gov (Identifier: NCT04593017 / (19/10/2020)).


Asunto(s)
Antibiosis , Susceptibilidad a Caries Dentarias , Placa Dental/microbiología , Limosilactobacillus reuteri/fisiología , Ortodoncia/métodos , Saliva/química , Saliva/microbiología , Adulto , Humanos , Concentración de Iones de Hidrógeno , Lactobacillales/fisiología , Streptococcus mutans/fisiología , Adulto Joven
18.
Benef Microbes ; 12(2): 137-145, 2021 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-33789556

RESUMEN

Pharyngitis and tonsillitis are the most common acute respiratory infections (ARIs) in children aged ≤5 years. The analysis of published data showed that some probiotics could decrease the frequency and number of days with ARIs. This study evaluated the safety and efficacy of Limosilactobacillus reuteri ATCC PTA 5289 and DSM 17938 to reduce the duration and severity of ARI symptoms. This randomised controlled trial included children aged from 6 months to 5 years, with pharyngitis or tonsillitis, who were randomised to receive a probiotic product containing L. reuteri ATCC PTA 5289 and L. reuteri DSM 17938 or placebo, as drops, ingested orally for 10 days as adjuvants to the use of non-steroidal anti-inflammatory drugs. The main outcomes were the duration and severity of ARI symptoms. The secondary outcomes were changes in salivary immunoglobulin A and inflammatory biomarkers. There was no fever on day 2 and subsequent days in the L. reuteri group (37.3 ±0.5 °C vs 38.6±0.3 °C, P<0.05). Beginning on day 3, the severity of sore throat (5±0.9 vs 8±1.2, P<0.05) was lower in the L. reuteri group. Significant differences in the days with runny nose, nasal congestion, days of non-programmed visits to the medical office or emergency department, levels in tumoral necrosis factor-alpha (TNF-alpha) and related costs of treatment were observed in the L. reuteri group. The frequency of adverse events was similar between the groups. Therefore, L. reuteri ATCC PTA 5289 combined with L. reuteri DSM 17938 is a safe and effective adjunct to reduce the symptoms of pharyngitis or tonsillitis in children.


Asunto(s)
Limosilactobacillus reuteri/fisiología , Faringitis/tratamiento farmacológico , Probióticos/administración & dosificación , Tonsilitis/tratamiento farmacológico , Preescolar , Femenino , Humanos , Inmunoglobulina A/inmunología , Lactante , Masculino , Faringitis/inmunología , Saliva/inmunología , Tonsilitis/inmunología , Factor de Necrosis Tumoral alfa/inmunología
19.
Cell Rep Med ; 2(3): 100206, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33763652

RESUMEN

Extremely low birth weight (ELBW) infants often develop an altered gut microbiota composition, which is related to clinical complications, such as necrotizing enterocolitis and sepsis. Probiotic supplementation may reduce these complications, and modulation of the gut microbiome is a potential mechanism underlying the probiotic effectiveness. In a randomized, double-blind, placebo-controlled trial, we assessed the effect of Lactobacillus reuteri supplementation, from birth to post-menstrual week (PMW)36, on infant gut microbiota. We performed 16S amplicon sequencing in 558 stool samples from 132 ELBW preterm infants at 1 week, 2 weeks, 3 weeks, 4 weeks, PMW36, and 2 years. Probiotic supplementation results in increased bacterial diversity and increased L. reuteri abundance during the 1st month. At 1 week, probiotic supplementation also results in a lower abundance of Enterobacteriaceae and Staphylococcaceae. No effects were found at 2 years. In conclusion, probiotics may exert benefits by modulating the gut microbiota composition during the 1st month in ELBW infants.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal/genética , Recien Nacido con Peso al Nacer Extremadamente Bajo/crecimiento & desarrollo , Recien Nacido Extremadamente Prematuro/crecimiento & desarrollo , Limosilactobacillus reuteri/fisiología , Probióticos/administración & dosificación , Actinobacteria/clasificación , Actinobacteria/genética , Actinobacteria/aislamiento & purificación , Bacteroidetes/clasificación , Bacteroidetes/genética , Bacteroidetes/aislamiento & purificación , Biodiversidad , Heces/microbiología , Femenino , Firmicutes/clasificación , Firmicutes/genética , Firmicutes/aislamiento & purificación , Fusobacterias/clasificación , Fusobacterias/genética , Fusobacterias/aislamiento & purificación , Humanos , Lactante , Masculino , Proteobacteria/clasificación , Proteobacteria/genética , Proteobacteria/aislamiento & purificación , ARN Ribosómico 16S/genética , Verrucomicrobia/clasificación , Verrucomicrobia/genética , Verrucomicrobia/aislamiento & purificación
20.
Cell ; 184(7): 1740-1756.e16, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33705688

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Locomoción , Conducta Social , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores , Trasplante de Microbiota Fecal , Heces/microbiología , Limosilactobacillus reuteri/metabolismo , Limosilactobacillus reuteri/fisiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/microbiología , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/terapia , Análisis de Componente Principal , Agitación Psicomotora/patología , Transmisión Sináptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA