Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Sci Rep ; 14(1): 13632, 2024 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871850

RESUMEN

Helicobacter pylori is a prominent gastrointestinal pathogen associated with various gastrointestinal illnesses. It presents substantial health risks due to its antibiotic resistance. Therefore, it is crucial to identify alternative treatments for H. pylori infections. Limosilactobacillus spp exhibit probiotic properties with beneficial effects in humans; however, the mechanisms by which it counteracts H. pylori infection are unknown. This study aimed to evaluate the potential of Limosilactobacillus fermentum T0701 lyophilized cell-free supernatants (LCFS) against H. pylori. The LCFS has varying antimicrobial activities, with inhibition zones of up to 10.67 mm. The minimum inhibitory concentration and minimum bacterial concentration of LCFS are 6.25-25.00 mg/mL and 6.25 mg/mL to > 50.00 mg/mL, respectively, indicating its capability to inhibit H. pylori. There is morphological damage observed in H. pylori treated with LCFS. Additionally, H. pylori adhesion to AGS cells (human gastric adenocarcinoma epithelial cells) reduces by 74.23%, highlighting the LCFS role in preventing bacterial colonization. Moreover, LCFS exhibits no cytotoxicity or morphological changes in AGS cells, and with no detected virulence or antimicrobial resistance genes, further supporting its safety profile. L. fermentum T0701 LCFS shows promise as a safe and effective non-toxic agent against H. pylori, with the potential to prevent gastric colonization.


Asunto(s)
Antibacterianos , Helicobacter pylori , Limosilactobacillus fermentum , Pruebas de Sensibilidad Microbiana , Helicobacter pylori/efectos de los fármacos , Limosilactobacillus fermentum/fisiología , Humanos , Antibacterianos/farmacología , Antibacterianos/química , Liofilización , Probióticos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/tratamiento farmacológico , Línea Celular Tumoral
2.
J Food Sci ; 89(6): 3713-3728, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38638065

RESUMEN

Lactobacillus fermentum can exert antiaging effects, but their roles are strain-specific, and little is known about the molecular mechanisms in some strains. This study investigated the antiaging effects of L. fermentum WC2020 (WC2020) isolated from Chinese fermented pickles and the underlying mechanism of the action in Caenorhabditis elegans. WC2020 enhanced the mean lifespan of L1-stage and L4-stage worms by 22.67% and 12.42%, respectively, compared with Escherichia coli OP50 (OP50), a standard food source for C. elegans. WC2020-induced longevity was accompanied by an increase in body length and mitochondrial transmembrane potential and a reduction in lipid accumulation and the production of reactive oxygen species and malondialdehyde. Moreover, WC2020 increased the production of glutathione, superoxide dismutases, and catalases and altered the transcripts of many phenotype-related genes. Furthermore, WC2020-fed jnk-1 rather than akt-2 or pmk-1 loss-of-function mutants showed similar lifespans to OP50-fed worms. Correspondingly, WC2020 significantly upregulated the expression of jnk-1 rather than genes involved in insulin-like, p38 MAPK, bate-catenin, or TGF-beta pathway. Moreover, the increase in body length, mitochondrial transmembrane potential, and antioxidant capability and the decrease in lipid accumulation induced by WC2020 were not observed in jnk-1 mutants. Additionally, WC2020 increased the expression of daf-16 and the proportion of daf-16::GFP in the nucleus, and increased lifespan disappeared in WC2020-fed daf-16 loss-of-function mutants. In conclusion, WC2020 activated the JNK/DAF-16 pathway to improve mitochondria function, reduce oxidative stress, and then extend the longevity of nematodes, suggesting WC2020 could be a potential probiotic targeting JNK-mediated antioxidant pathway for antiaging in food supplements and bioprocessing. PRACTICAL APPLICATION: Aging has a profound impact on the global economy and human health and could be delayed by specific diets and nutrient resources. This study demonstrated that Lactobacillus fermentum WC2020 could be a potential probiotic strain used in food to promote longevity and health via the JNK-mediated antioxidant pathway.


Asunto(s)
Antioxidantes , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Limosilactobacillus fermentum , Longevidad , Animales , Caenorhabditis elegans/fisiología , Limosilactobacillus fermentum/fisiología , Antioxidantes/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sistema de Señalización de MAP Quinasas , Potencial de la Membrana Mitocondrial , Proteínas Quinasas Activadas por Mitógenos
3.
Food Funct ; 12(17): 7938-7953, 2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34251007

RESUMEN

With the increasing incidence of type 2 diabetes, it is imperative to identify how to effectively prevent or treat this disease. Studies have shown that some lactic acid bacteria can improve type 2 diabetes with almost no side effects. Therefore, in this experimental study, we explored the preventive and therapeutic effects of Lactobacillus fermentum TKSN041 (L. fermentum TKSN041) on streptozotocin-induced type 2 diabetes in rats. The results showed that L. fermentum TKSN041 could reduce the amount of water intake, reduce weight loss, and control the increase in the fasting blood glucose level of diabetic rats. The organ index and tissue section results showed that L. fermentum TKSN041 could reduce the damage caused by diabetes to the liver, kidney, spleen, pancreatic, and brain tissue. Furthermore, L. fermentum TKSN041 decreased the levels of triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL), aminotransferase (AST), alanine aminotransferase (ALT), glycated serum proteins (GSP), malondialdehyde (MDA), interleukin 1 beta (IL-1ß), interleukin 6 (IL-6), and endothelin 1 (ET-1) in serum and increased the serum levels of high-density lipoprotein cholesterol (HDL) and interleukin 10 (IL-10). Finally, L. fermentum TKSN041 up-regulated the mRNA and protein expressions of NF-kappa-B inhibitor-α (IκB-α), AMP-activated protein kinase (AMPK), insulin receptor substrate-1 (IRS-1), liver kinase B1 (LKB1), and glucose transporter 4 (GLUT4) and down-regulated those of nuclear factor-κBp65 (NFκB-p65) and tumor necrosis factor alpha (TNF-α). Furthermore, LF-TKSN041 up-regulated the mRNA expressions of peroxisome proliferator-activated receptor γ (PPAR-γ) and down-regulated neuropeptide Y (NPY), sterol regulatory element-binding protein-1 (SREBF-1), and vascular endothelial growth factor (VEGF). These results suggest that L. fermentum TKSN041 may be a useful intervention factor for the prevention or treatment of type 2 diabetes induced by STZ. Clinical trials are needed to further demonstrate its effectiveness.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Limosilactobacillus fermentum/fisiología , Probióticos/administración & dosificación , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Alanina Transaminasa/genética , Alanina Transaminasa/metabolismo , Animales , Colesterol/metabolismo , Diabetes Mellitus Tipo 2/inducido químicamente , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , PPAR gamma/genética , PPAR gamma/metabolismo , Ratas , Ratas Sprague-Dawley , Estreptozocina , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Food Funct ; 12(13): 6029-6044, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34037025

RESUMEN

In this experiment, Lactobacillus fermentum CQPC08 (LF-CQPC08) isolated from traditionally fermented pickles was used to study its mitigation effect on lead acetate-induced oxidative stress and lead ion adsorption capacity in rats. In vitro experiments showed that the survival rate in artificial gastric juice and the growth efficiency in artificial bile salt of LF-CQPC08 was 93.6% ± 2.2% and 77.2% ± 0.8%, and the surface hydrophobicity rate was 45.5% ± 0.3%. The scavenging rates of hydroxyl radical, superoxide anion, and 1,1-diphenyl-2-picrylhydrazyl (DPPH) were 47.8% ± 0.9%, 63.9% ± 1.2%, and 83.6% ± 1.5%, respectively, and the reduction power was 107.3 ± 2.8 µmol L-1. LF-CQPC08 could not only adsorb 76.9% ± 1.0% lead ions in aqueous solution but also reduce the lead content in serum, liver, kidneys, and brain tissue of Sprague-Dawley (SD) rats, as well as maintain the cell structure and tissue state of the liver and kidneys. In addition, by examining the indicators of inflammation and oxidation in the serum, liver, and kidneys of SD rats, we found that LF-CQPC08 can reduce the proinflammatory factors interleukin (IL)-1 beta (1ß), IL-6, tumor necrosis factor alpha, and interferon gamma in the body, increase the level of anti-inflammatory factor IL-10, enhance the activity of antioxidant enzymes such as superoxide dismutase and catalase and glutathione levels in serum and organ tissues, and reduce the production of reactive oxygen species and accumulation of lipid peroxide malondialdehyde. LF-CQPC08 can also activate the Keap1/Nrf2/ARE signaling pathway to promote high-level expression of the downstream antioxidants heme oxygenase 1 (HO-1), NAD(P)H : quinone oxidoreductase 1 (NQO1), and γ-glutamylcysteine synthetase (γ-GCS). As food-grade lactic acid bacteria, LF-CQPC08 has great potential and research value in removing heavy metals from food and alleviating the toxicity of heavy metals in the future.


Asunto(s)
Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Plomo/efectos adversos , Limosilactobacillus fermentum/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Antioxidantes/farmacología , Sangre , Encéfalo , Citocinas/sangre , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Hemo-Oxigenasa 1/metabolismo , Inflamación , Riñón , Hígado , Masculino , Malondialdehído/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
5.
J Appl Microbiol ; 130(4): 1307-1322, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32638482

RESUMEN

AIM: The aim of this study was to evaluate the molecular mechanisms of Lactobacillus strains in improving ageing of the musculoskeletal system. METHODS AND RESULTS: The anti-ageing mechanism of three probiotics strains Lactobacillus fermentum DR9, Lactobacillus paracasei OFS 0291 and L. helveticus OFS 1515 were evaluated on gastrocnemius muscle and tibia of d-galactose-induced ageing rats. Upon senescence induction, aged rats demonstrated reduced antioxidative genes CAT and SOD expression in both bone and muscle compared to the young rats (P < 0·05). Strain L. fermentum DR9 demonstrated improved expression of SOD in bone and muscle compared to the aged rats (P < 0·05). In the evaluation of myogenesis-related genes, L. paracasei OFS 0291 and L. fermentum DR9 increased the mRNA expression of IGF-1; L. helveticus OFS 1515 and L. fermentum DR9 reduced the expression of MyoD, in contrast to the aged controls (P < 0·05). Protective effects of L. fermentum DR9 on ageing muscle were believed to be contributed by increased AMPK-α2 expression. Among the osteoclastogenesis genes studied, TNF-α expression was highly elevated in tibia of aged rats, while all three probiotics strains ameliorated the expression. Lactobacillus fermentum DR9 also reduced the expression of IL-6 and TRAP in tibia when compared to the aged rats (P < 0·05). All probiotics treatment resulted in declined proinflammatory cytokines IL-1ß in muscle and bone. CONCLUSIONS: Lactobacillus fermentum DR9 appeared to be the strongest strain in modulation of musculoskeletal health during ageing. SIGNIFICANCE AND IMPACT OF THE STUDY: The study demonstrated the protective effects of the bacteria on muscle and bone through antioxidative and anti-inflammatory actions. Therefore, L. fermentum DR9 may serve as a promising targeted anti-ageing therapy.


Asunto(s)
Envejecimiento/efectos de los fármacos , Huesos/efectos de los fármacos , Galactosa/efectos adversos , Lacticaseibacillus paracasei/fisiología , Lactobacillus helveticus/fisiología , Limosilactobacillus fermentum/fisiología , Sistema Musculoesquelético/efectos de los fármacos , Probióticos/administración & dosificación , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Desarrollo Óseo/efectos de los fármacos , Huesos/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Desarrollo Musculoesquelético/efectos de los fármacos , Sistema Musculoesquelético/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
PLoS One ; 15(10): e0239365, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33001998

RESUMEN

The growth of filamentous fungi during the spontaneous cocoa bean fermentation leads to inferior cocoa bean quality and poses a health risk for consumers due to the potential accumulation of mycotoxins. We recently developed anti-fungal cultures with the capacity to inhibit the growth of mycotoxigenic filamentous fungi on cocoa beans. However, it is not clear how these anti-fungal cultures affect the fermentation process and cocoa bean quality. For that, the anti-fungal co-cultures, Lactobacillus fermentum M017-Saccharomyces cerevisiae H290 (A) and Lb. fermentum 223-S. cerevisiae H290 (B), were applied to 180-kg box fermentations in Honduras in three time-independent replications each including a spontaneous control fermentation. The comparison of inoculated and spontaneous fermentation processes revealed that the co-cultures only marginally affected the fermentation process and cocoa bean quality. Microorganisms reached maximal levels of 6.2-7.6 log CFU/g of yeasts and acetic acid bacteria and 7.9-9.5 log CFU/g of lactic acid bacteria during all fermentations and led to maximal metabolite concentrations in bean cotyledons of 4-12 mg/g ethanol, 2-6 mg/g lactic acid and 6-14 mg/g acetic acid. The fermentation and drying processes resulted in 38-90 mg epicatechin equivalents/g in the cotyledons of dried beans. However, the co-cultures led to up to ten times higher mannitol levels in cotyledons of inoculated beans compared to beans during spontaneous fermentation, and caused a slower fermentation process, detectable as up to 8-12 °C lower temperatures in the centre of the fermenting pulp-bean mass and up to 22% lower proportions of well-fermented beans after drying. Co-culture B-with Lb. fermentum 223 -led to improved cocoa bean quality compared to co-culture A-with Lb. fermentum M017 -, i.e. cocoa beans with 0.5-1.9 mg/g less acetic acid, 4-17% higher shares of well-fermented beans and, on a scale from 0 to 10, to 0.2-0.6 units lower astringency, up to 1.1 units lower off-flavours, and 0.2-0.9 units higher cocoa notes. Therefore, the anti-fungal co-culture B is recommended for future applications and its capacity to limit fungal growth and mycotoxin production during industrial-scale cocoa bean fermentation should be investigated in further studies.


Asunto(s)
Cacao/metabolismo , Cacao/microbiología , Técnicas de Cocultivo , Fermentación , Calidad de los Alimentos , Limosilactobacillus fermentum/fisiología , Saccharomyces cerevisiae/fisiología , Alcaloides/análisis , Cacao/química , Concentración de Iones de Hidrógeno , Limosilactobacillus fermentum/crecimiento & desarrollo , Polifenoles/análisis , Saccharomyces cerevisiae/crecimiento & desarrollo , Temperatura , Factores de Tiempo
7.
Nutrients ; 11(10)2019 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-31600934

RESUMEN

The bidirectional communication between the gastrointestinal tract and the central nervous system appears to be functionally linked to the intestinal microbiome, namely the microbiome-gut-brain axis (MGBA). Probiotics with health benefits on psychiatric or neurological illnesses are generally called psychobiotics, and some of them may also be able to improve sleep by targeting the MGBA. This study aimed to investigate the effects of a psychobiotic strain, Lactobacillus fermentum PS150TM (PS150TM), on sleep improvement by using a pentobarbital-induced sleep mouse model. Compared with the vehicle control group, the oral administration of PS150TM, but not the other L. fermentum strains, significantly decreased the sleep latency and increased the sleep duration of mice, suggesting strain-specific sleep-improving effects of PS150TM. Moreover, the ingestion of diphenhydramine, an antihistamine used to treat insomnia, as a drug control group, only increased the sleep duration of mice. We also found that the sleep-improving effects of PS150TM are time- and dose-dependent. Furthermore, the oral administration of PS150TM could attenuate a caffeine-induced sleep disturbance in mice, and PS150TM appeared to increase the expression of the gene encoding the adenosine 1 receptor in the hypothalamus of mice, as assessed by quantitative real-time polymerase chain reaction. Taken together, our results present a potential application of PS150TM as a dietary supplement for sleep improvement.


Asunto(s)
Hipnóticos y Sedantes/farmacología , Limosilactobacillus fermentum/fisiología , Pentobarbital/farmacología , Fármacos Inductores del Sueño , Sueño/efectos de los fármacos , Adenosina/metabolismo , Animales , Cafeína/farmacología , ADN Bacteriano/análisis , ADN Bacteriano/aislamiento & purificación , Expresión Génica , Limosilactobacillus fermentum/genética , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Sueño/fisiología
8.
Microb Pathog ; 131: 246-253, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30974159

RESUMEN

Gastrointestinal (GI) infection is one of the most common types of infectious diseases. Application of probiotic strains in the control of such infections represents a promising approach. In this study, Lactobacillus fermentum strain 4-17 was isolated from kashkineh, an Iranian cereal fermented food, and identified by sequencing its 16S rRNA gene using universal primers. Its probiotic features, including resistance to acid, bile tolerance, antibacterial activity, resistance to intestinal and gastric juices, and hydrophobicity were evaluated. The ability of this strain to adhere to human intestinal cells as well as the anti-adhesive effect of L. fermentum strain 4-17 against E. coli isolated from patients with urinary tract infection was investigated. L. fermentum strain 4-17 was capable of surviving at various conditions such as low pH values, bile salts exposure, and GI tract environment. It showed 43% cell hydrophobicity. The adhesion level of L. fermentum strain 4-17 to human colon adenocarcinoma Caco-2 cells was 8.5% which was also confirmed by scanning electron microscopy (SEM). Furthermore, this strain showed appropriate anti-adhesive properties (including competition, inhibition and replacement properties) against human pathogenic bacteria. These data suggest that L. fermentum strain 4-17 could be examined further for its useful effects and introduced as a novel candidate probiotic to control GI infection disease.


Asunto(s)
Antiinfecciosos/farmacología , Adhesión Bacteriana , Escherichia coli/efectos de los fármacos , Limosilactobacillus fermentum/fisiología , Probióticos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Ácidos y Sales Biliares , Células CACO-2 , Escherichia coli/patogenicidad , Alimentos Fermentados/microbiología , Jugo Gástrico , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Irán , Limosilactobacillus fermentum/genética , Limosilactobacillus fermentum/aislamiento & purificación , Probióticos/uso terapéutico , ARN Ribosómico 16S/genética , Tolerancia a la Sal , Infecciones Urinarias/microbiología
9.
Benef Microbes ; 9(5): 829-841, 2018 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-29798705

RESUMEN

Helicobacter pylori infection is associated with important gastric pathologies. An aggressive proinflammatory immune response is generated in the gastric tissue infected with H. pylori, resulting in gastritis and a series of morphological changes that increase the susceptibility to cancer development. Probiotics could present an alternative solution to prevent or decrease H. pylori infection. Among them, the use of immunomodulatory lactic acid bacteria represents a promising option to reduce the severity of chronic inflammatory-mediated tissue damage and to improve protective immunity against H. pylori. We previously isolated Lactobacillus fermentum UCO-979C from human gastric tissue and demonstrated its capacity to reduce adhesion of H. pylori to human gastric epithelial cells (AGS cells). In this work, the ability of L. fermentum UCO-979C to modulate immune response in AGS cells and PMA phorbol 12-myristate 13-acetate (PMA)-differentiated THP-1 (human monocytic leukaemia) macrophages in response to H. pylori infection was evaluated. We demonstrated that the UCO-979C strain is able to differentially modulate the cytokine response of gastric epithelial cells and macrophages after H. pylori infection. Of note, L. fermentum UCO-979C was able to significantly reduce the production of inflammatory cytokines and chemokines in AGS and THP-1 cells as well as increase the levels of immunoregulatory cytokines, indicating a remarkable anti-inflammatory effect. These findings strongly support the probiotic potential of L. fermentum UCO-979C and provide evidence of its beneficial effects against the inflammatory damage induced by H. pylori infection. Although our findings should be proven in appropriate experiments in vivo, in both H. pylori infection animal models and human trials, the results of the present work provide a scientific rationale for the use of L. fermentum UCO-979C to prevent or reduce H. pylori-induced gastric inflammation in humans.


Asunto(s)
Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/inmunología , Helicobacter pylori/fisiología , Inmunidad Innata/efectos de los fármacos , Limosilactobacillus fermentum/fisiología , Probióticos/farmacología , Animales , Citocinas/genética , Citocinas/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Humanos , Ratones
10.
Microbiology (Reading) ; 164(3): 349-358, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29458690

RESUMEN

Lactobacilli are the dominant bacteria of the vaginal tract of healthy women and they play a major role in the maintenance of mucosal homeostasis, preventing genital infections, such as bacterial vaginosis (BV) and vulvovaginal candidiasis (VVC). It is now known that one mechanism of this protection is the influence that lactobacilli can exert on host immune responses. In this context, we evaluated two Lactobacillus strains (L. plantarum 59 and L. fermentum 137) for their immunomodulatory properties in response to Gardnerella vaginalis (BV) or Candida albicans (VVC) infections in a HeLa cell infection model. G. vaginalis and C. albicans triggered the secretion of pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6 and IL-8) and the activation of NF-κB in HeLa cells, in contrast to L. plantarum 59 and L. fermentum 137. Treatments with the Lactobacillus strains or their cell-free supernatants before (pre-treatment) or after (post-treatment) the challenge with the pathogens resulted in decreased secretion of pro-inflammatory cytokines and decreased activation of NF-κB. The treatments with Lactobacillus strains not only decreased the secretion of IL-8, but also its expression, as confirmed by gene reporter luciferase assay, suggesting transcription-level control by lactobacilli. In conclusion, L. plantarum 59 and L. fermentum 137 were confirmed to have an anti-inflammatory effect against G. vaginalis and C. albicans and they were able to influence signalling in NF-κB pathway, making them interesting candidates as probiotics for the prevention or treatment of BV and VVC.


Asunto(s)
Antiinflamatorios/farmacología , Candida albicans/efectos de los fármacos , Gardnerella vaginalis/efectos de los fármacos , Lactobacillus plantarum/fisiología , Limosilactobacillus fermentum/fisiología , Probióticos/farmacología , Candida albicans/crecimiento & desarrollo , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Citocinas/genética , Citocinas/metabolismo , Femenino , Gardnerella vaginalis/crecimiento & desarrollo , Células HeLa , Humanos , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/efectos de los fármacos
11.
BMC Microbiol ; 17(1): 108, 2017 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-28477627

RESUMEN

BACKGROUND: Genus Clostridium accompanies more than 200 known species and at least 30 among them are associated with human and animal diseases. At the moment, the treatment of clostridial infections is based on use of antibiotics. However, due to the European ban on the use of antibiotics in livestock production, novel therapeutic strategies for treatment of these hardly curable infections have been evaluated. Hence, in this study the antimicrobial effect of newly designed probiotic culture consisted of natural isolates Lactobacillus helveticus BGRA43, Lactobacillus fermentum BGHI14 and Streptococcus thermophilus BGVLJ1-44 against Clostridium difficile and Clostridium perfringens was analyzed. RESULTS: The probiotic culture showed strong in vitro antimicrobial effect on C. difficile (human clinical isolate). In addition, individual strains and the probiotic combination exhibited immunomodulatory activity. The probiotic combination significantly increased the proliferation of GALT lymphocytes. At the other hand, none of the bacterial treatments (individual strains and the combination) induced the production of proinflammatory cytokines IL-6 and IL-1ß by intestinal epithelial cells, Caco-2. Interestingly, Caco-2 cells exposed to the probiotic combination produced significantly elevated amount of TGFß pointing to potential protecting effect of the probiotic. In addition, the results of field trial on spontaneously infected goats revealed reduction of C. perfringens in goats (below the detection threshold) after the probiotic treatment. CONCLUSIONS: The results of this study indicated that the novel probiotic deserves to be further investigated as a promising antimicrobial agent against C. difficile and C. perfringens.


Asunto(s)
Antibiosis , Clostridioides difficile/crecimiento & desarrollo , Clostridium perfringens/crecimiento & desarrollo , Probióticos/uso terapéutico , Animales , Células CACO-2 , Técnicas de Cultivo de Célula , Proliferación Celular , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/veterinaria , Clostridium perfringens/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Femenino , Cabras , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Intestinos/efectos de los fármacos , Intestinos/inmunología , Limosilactobacillus fermentum/fisiología , Lactobacillus helveticus/fisiología , Pruebas de Sensibilidad Microbiana , Fragmentos de Péptidos/metabolismo , Streptococcus thermophilus/fisiología
12.
Electron. j. biotechnol ; Electron. j. biotechnol;25: 75-83, ene. 2017. tab, graf, ilus
Artículo en Inglés | LILACS | ID: biblio-1008594

RESUMEN

Background: Helicobacter pylori is considered as the main risk factor in the development of gastric cancer. In the present study, we performed a detailed characterization of the probiotic properties and the anti-H. pylori activity of a previously isolated lactobacillus strain ­ Lactobacillus fermentum UCO-979C ­ obtained from human gut. Results: The strain tolerated pH 3.0; grew in the presence of 2% bile salts; produced lactic acid and hydrogen peroxide; aggregated in saline solution; showed high hydrophobicity; showed high adherence to glass; Caco-2 and gastric adenocarcinoma human cells (AGS) cells; showed an efficient colonization in Mongolian Gerbils; and potently inhibited the growth and urease activity of H. pylori strains. L. fermentum UCO-979C significantly inhibited H. pylori-induced IL-8 production in AGS cells and reduced the viability of H. pylori. With regard to innocuousness, the strain UCO-979C was susceptible to several antibiotics and did not produce histamine or beta-haemolysis in blood agar containing red blood cells from various origins. Conclusion: The results demonstrated that L. fermentum UCO-979C is a very good candidate as a probiotic for the protection of humans against H. pylori infections.


Asunto(s)
Humanos , Animales , Helicobacter pylori/efectos de los fármacos , Infecciones por Helicobacter/prevención & control , Probióticos/farmacología , Limosilactobacillus fermentum/fisiología , Antibacterianos/farmacología , Neoplasias Gástricas/prevención & control , Ureasa/antagonistas & inhibidores , Interleucina-8/antagonistas & inhibidores , Gerbillinae , Modelos Animales de Enfermedad , Interacciones Hidrofóbicas e Hidrofílicas
13.
Biofouling ; 32(10): 1245-1257, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27834106

RESUMEN

The ability of the human isolate Lactobacillus fermentum UCO-979C to form biofilm and synthesize exopolysaccharide on abiotic and biotic models is described. These properties were compared with the well-known Lactobacillus casei Shirota to better understand their anti-Helicobacter pylori probiotic activities. The two strains of lactobacilli synthesized exopolysaccharide as detected by the Dubois method and formed biofilm on abiotic and biotic surfaces visualized by crystal violet staining and scanning electron microscopy. Concomitantly, these strains inhibited H. pylori urease activity by up to 80.4% (strain UCO-979C) and 66.8% (strain Shirota) in gastric adenocarcinoma (AGS) cells, but the two species showed equal levels of inhibition (~84%) in colorectal adenocarcinoma (Caco-2) cells. The results suggest that L. fermentum UCO-979C has probiotic potential against H. pylori infections. However, further analyses are needed to explain the increased activity observed against the pathogen in AGS cells as compared to L. casei Shirota.


Asunto(s)
Antibiosis , Biopelículas/crecimiento & desarrollo , Helicobacter pylori/crecimiento & desarrollo , Limosilactobacillus fermentum/fisiología , Probióticos , Adhesión Bacteriana , Células CACO-2 , Neoplasias del Colon/microbiología , Helicobacter pylori/efectos de los fármacos , Humanos , Lacticaseibacillus casei/metabolismo , Lacticaseibacillus casei/fisiología , Limosilactobacillus fermentum/metabolismo , Microscopía Electrónica de Rastreo , Polisacáridos Bacterianos/análisis , Polisacáridos Bacterianos/farmacología , Probióticos/farmacología , Neoplasias Gástricas/microbiología
14.
Nutrients ; 8(3): 155, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26978395

RESUMEN

The purpose of the study was to determine the inhibitory effects of Lactobacillus fermentum Suo (LF-Suo) on HCl/ethanol induced gastric injury in ICR (Institute for Cancer Research) mice and explain the mechanism of these effects through the molecular biology activities of LF-Suo. The studied mice were divided into four groups: healthy, injured, LF-Suo-L and LF-Suo-H group. After the LF-Suo intragastric administration, the gastric injury area was reduced compared to the injured group. The serum MOT (motilin), SP (substance P), ET (endothelin) levels of LF-Suo treated mice were lower, and SS (somatostatin), VIP (vasoactive intestinal peptide) levels were higher than the injured group mice. The cytokine IL-6 (interleukin 6), IL-12 (interleukin 12), TNF-α (tumor necrosis factor-α) and IFN-γ (interferon-γ) serum levels were decreased after the LF-Suo treatment. The gastric tissues SOD (superoxide dismutase), GSH-Px (glutathione peroxidase), NO (nitric oxide) and activities of LF-Suo treated mice were increased and MDA (malondialdehyde) activity was decreased compared to the injured group mice. By the RT-PCR assay, LF-Suo raised the occludin, EGF (epidermal growth factor), EGFR (epidermal growth factor receptor), VEGF (vascular endothelial growth factor), Fit-1 (fms-like tyrosine kinase-1), IκB-α (inhibitor kappaB-α), nNOS (neuronal nitric oxide synthase), eNOS (endothelial nitric oxide synthase), Mn-SOD, Cu/Zn-SOD, CAT (catalase) mRNA or protein expressions and reduced the COX-2, NF-κB (nuclear factor kappaB), and iNOS (inducible nitric oxide synthase) expressions in gastric tissues compared to the gastric injured group mice. A high concentration (1.0 × 108 CFU/kg b.w.) of LF-Suo treatment showed stronger anti-gastric injury effects compared to a low concentration of (0.5 × 108 CFU/kg b.w.) of LF-Suo treatment. LF-Suo also showed strong survival in pH 3.0 man-made gastric juice and hydrophobic properties. These results indicate that LF-Suo has potential use as probiotics for its gastric injury treatment effects.


Asunto(s)
Antioxidantes/metabolismo , Etanol , Ácido Clorhídrico , Limosilactobacillus fermentum/fisiología , Probióticos , Úlcera Gástrica/prevención & control , Estómago/microbiología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Jugo Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Hormonas Gastrointestinales/metabolismo , Regulación Enzimológica de la Expresión Génica , Concentración de Iones de Hidrógeno , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos ICR , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/microbiología , Úlcera Gástrica/patología
15.
Sci Rep ; 6: 20318, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26843130

RESUMEN

Lactobacilli are thought to be beneficial for human health, with lactobacilli-associated infections being confined to immune-compromised individuals. However, Lactobacillus fermentum AGR1487 negatively affects barrier integrity in vitro so we hypothesized that it caused a pro-inflammatory response in the host. We compared germ-free rats inoculated with AGR1487 to those inoculated with another L. fermentum strain, AGR1485, which does not affect in vitro barrier integrity. We showed that rats inoculated with AGR1487 had more inflammatory cells in their colon, higher levels of inflammatory biomarkers, and increased colonic gene expression of pro-inflammatory pathways. In addition, our in vitro studies showed that AGR1487 had a greater capacity to activate TLR signaling and induce pro-inflammatory cytokines in immune cells. This study indicates the potential of strains of the same species to differentially elicit inflammatory responses in the host and highlights the importance of strain characterization in probiotic approaches to treat inflammatory disorders.


Asunto(s)
Limosilactobacillus fermentum/fisiología , Boca/microbiología , Probióticos/administración & dosificación , Animales , Biomarcadores/metabolismo , Colitis/etiología , Colitis/metabolismo , Colon/citología , Colon/microbiología , Colon/patología , Citocinas/metabolismo , Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Limosilactobacillus fermentum/aislamiento & purificación , Linfocitos/citología , Linfocitos/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Receptores Toll-Like/metabolismo
16.
Food Microbiol ; 53(Pt A): 51-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26611169

RESUMEN

Few studies have extensively investigated probiotic functions associated with biofilms. Here, we show that strains of Lactobacillus plantarum and Lactobacillus fermentum are able to grow as biofilm on abiotic surfaces, but the biomass density differs between strains. We performed microtiter plate biofilm assays under growth conditions mimicking to the gastrointestinal environment. Osmolarity and low concentrations of bile significantly enhanced Lactobacillus spatial organization. Two L. plantarum strains were able to form biofilms under high concentrations of bile and mucus. We used the agar well-diffusion method to show that supernatants from all Lactobacillus except the NA4 isolate produced food pathogen inhibitory molecules in biofilm. Moreover, TNF-α production by LPS-activated human monocytoid cells was suppressed by supernatants from Lactobacillus cultivated as biofilms but not by planktonic culture supernatants. However, only L. fermentum NA4 showed anti-inflammatory effects in zebrafish embryos fed with probiotic bacteria, as assessed by cytokine transcript level (TNF-α, IL-1ß and IL-10). We conclude that the biofilm mode of life is associated with beneficial probiotic properties of lactobacilli, in a strain dependent manner. Those results suggest that characterization of isolate phenotype in the biofilm state could be additional valuable information for the selection of probiotic strains.


Asunto(s)
Antibiosis , Biopelículas/crecimiento & desarrollo , Lactobacillus plantarum/crecimiento & desarrollo , Lactobacillus plantarum/fisiología , Limosilactobacillus fermentum/crecimiento & desarrollo , Limosilactobacillus fermentum/fisiología , Probióticos , Animales , Bilis/microbiología , Medios de Cultivo/química , Escherichia coli/fisiología , Humanos , Inmunidad Innata , Inmunomodulación , Interleucina-10/biosíntesis , Limosilactobacillus fermentum/inmunología , Lactobacillus plantarum/inmunología , Monocitos/inmunología , Moco/microbiología , Salmonella enterica/fisiología , Factor de Necrosis Tumoral alfa/biosíntesis , Pez Cebra
17.
J Microbiol Biotechnol ; 25(9): 1510-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26095384

RESUMEN

In this work, we wanted to develop a probiotic from famous longevity villages in Korea. We visited eight longevity villages in Korea to collect fecal samples from healthy adults who were aged above 80 years and had regular bowel movements, and isolated lactic-acid-producing bacteria from the samples. Isolated colonies that appeared on MRS agar containing bromophenol blue were identified by means of 16S rRNA sequencing, and 102 of the isolates were identified as lactic-acid-producing bacteria (18 species). Lactobacillus fermentum was the most frequently found species. Eight isolates were selected on the basis of their ability to inhibit the growth of six intestinal pathogens (Escherichia coli O157:H7, Salmonella enterica subsp. enterica Typhimurium, Salmonella enterica subsp. enterica Enteritidis, Enterococcus faecalis, Staphylococcus aureus, and Listeria monocytogenes) and their susceptibility to 15 antimicrobial agents. Among these eight isolates, four Lactobacillus fermentum isolates were found not to produce any harmful enzymes or metabolites. Among them, Lactobacillus fermentum isolate no. 24 showed the strongest binding to intestinal epithelial cells, the highest immune-enhancing activity, anti-inflammation activity, and anti-oxidation activity as well as the highest survival rates in the presence of artificial gastric juice and bile solution. This isolate, designated Lactobacillus fermentum PL9988, has all the characteristics for a good probiotic.


Asunto(s)
Antibacterianos/farmacología , Antibiosis , Heces/microbiología , Limosilactobacillus fermentum/clasificación , Limosilactobacillus fermentum/aislamiento & purificación , Anciano , Animales , Pueblo Asiatico , Adhesión Bacteriana , Toxinas Bacterianas/análisis , Línea Celular , Análisis por Conglomerados , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Ribosómico/química , ADN Ribosómico/genética , Enterococcus faecalis , Enzimas/análisis , Células Epiteliales/microbiología , Escherichia coli O157 , Humanos , Limosilactobacillus fermentum/genética , Limosilactobacillus fermentum/fisiología , Listeria monocytogenes , Longevidad , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Viabilidad Microbiana/efectos de los fármacos , Datos de Secuencia Molecular , Oxidantes/toxicidad , Filogenia , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Staphylococcus aureus
18.
PLoS One ; 10(5): e0126520, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25978374

RESUMEN

Lipopolysaccharide (LPS) has essential role in the pathogenesis of D-galactosamine-sensitized animal models and alcoholic liver diseases of humans, by stimulating release of pro-inflammatory mediators that cause hepatic damage and intestinal barrier impairment. Oral pretreatment of probiotics has been shown to attenuate LPS-induced hepatic injury, but it is unclear whether the effect is direct or due to improvement in the intestinal barrier. The present study tested the hypothesis that pretreatment with probiotics enables the liver to withstand directly LPS-induced hepatic injury and inflammation. In a mouse model of LPS-induced hepatic injury, the levels of hepatic tumor necrosis factor-alpha (TNF-α) and serum alanine aminotransferase (ALT) of mice with depleted intestinal commensal bacteria were not significantly different from that of the control models. Pre-feeding mice for 10 days with Lactobacillus fermentum ZYL0401 (LF41), significantly alleviated LPS-induced hepatic TNF-α expression and liver damage. After LF41 pretreatment, mice had dramatically more L.fermentum-specific DNA in the ileum, significantly higher levels of ileal cyclooxygenase (COX)-2 and interleukin 10 (IL-10) and hepatic prostaglandin E2 (PGE2). However, hepatic COX-1, COX-2, and IL-10 protein levels were not changed after the pretreatment. There were also higher hepatic IL-10 protein levels after LPS challenge in LF41-pretreated mice than in the control mice. Attenuation of hepatic TNF-α was mediated via the PGE2/E prostanoid 4 (EP4) pathway, and serum ALT levels were attenuated in an IL-10-dependent manner. A COX-2 blockade abolished the increase in hepatic PGE2 and IL-10 associated with LF41. In LF41-pretreated mice, a blockade of IL-10 caused COX-2-dependent promotion of hepatic PGE2, without affecting hepatic COX-2 levels. In LF41-pretreated mice, COX2 prevented enhancing TNF-α expression in both hepatic mononuclear cells and the ileum, and averted TNF-α-mediated increase in intestinal permeability. Together, we demonstrated that LF41 pre-feeding enabled the liver to alleviate LPS-induced hepatic TNF-α expression and injury via a PGE2-EP4- and IL-10-dependent mechanism.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Dinoprostona/metabolismo , Limosilactobacillus fermentum/fisiología , Lipopolisacáridos/toxicidad , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Animales , Western Blotting , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Interleucina-10/metabolismo , Masculino , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
19.
Int J Mol Sci ; 15(12): 21875-95, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25464378

RESUMEN

The aim of this study was to investigate the effects of Lactobacillus fermentum Suo (LF-Suo) on activated carbon-induced constipation in ICR (Institute of Cancer Research) mice. ICR mice were orally administered with lactic acid bacteria for 9 days. Body weight, diet intake, drinking amount, defecation status, gastrointestinal transit and defecation time, and the serum levels of MTL (motilin), Gas (gastrin), ET (endothelin), SS (somatostatin), AChE (acetylcholinesterase), SP (substance P), VIP (vasoactive intestinal peptide) were used to evaluate the preventive effects of LF-Suo on constipation. Bisacodyl, a laxative drug, was used as a positive control. The normal, control, 100 mg/kg bisacodyl treatment, LB (Lactobacillus bulgaricus)-, LF-Suo (L)- and LF-Suo (H)-treated mice showed the time to the first black stool defecation at 90, 218, 117, 180, 155 and 137 min, respectively. By the oral administration of LB-, LF-Suo (L), LF-Suo (H) or bisacodyl (100 mg/kg), the gastrointestinal transit was reduced to 55.2%, 72.3%, 85.5% and 94.6%, respectively, of the transit in normal mice, respectively. In contrast to the control mice, the serum levels of MTL, Gas, ET, AChE, SP and VIP were significantly increased and the serum levels of SS were reduced in the mice treated with LF-Suo (p < 0.05). By the RT-PCR (reverse transcription-polymerase chain reaction) and western blot assays, LF-Suo increased the c-Kit, SCF (stem cell factor), GDNF (glial cell line-derived neurotrophic factor) and decreased TRPV1 (transient receptor potential vanilloid 1), NOS (nitric oxide synthase) expressions of small intestine tissue in mice. These results demonstrate that lactic acid bacteria has preventive effects on mouse constipation and LF-Suo demonstrated the best functional activity.


Asunto(s)
Estreñimiento/terapia , Limosilactobacillus fermentum/fisiología , Animales , Peso Corporal , Carbón Orgánico , Estreñimiento/genética , Estreñimiento/fisiopatología , Defecación , Dieta , Modelos Animales de Enfermedad , Conducta de Ingestión de Líquido , Femenino , Tránsito Gastrointestinal , Regulación de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Intestino Delgado/patología , Intestino Delgado/fisiopatología , Ratones , Ratones Endogámicos ICR , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Factores de Tiempo
20.
PLoS One ; 9(11): e112214, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25386948

RESUMEN

Helicobacter pylori (H. pylori) is a major gastric pathogen that has been associated with humans for more than 60,000 years. H. pylori causes different gastric diseases including dyspepsia, ulcers and gastric cancers. Disease development depends on several factors including the infecting H. pylori strain, environmental and host factors. Another factor that might influence H. pylori colonization and diseases is the gastric microbiota that was overlooked for long because of the belief that human stomach was a hostile environment that cannot support microbial life. Once established, H. pylori mainly resides in the gastric mucosa and interacts with the resident bacteria. How these interactions impact on H. pylori-caused diseases has been poorly studied in human. In this study, we analyzed the interactions between H. pylori and two bacteria, Streptococcus mitis and Lactobacillus fermentum that are present in the stomach of both healthy and gastric disease human patients. We have found that S. mitis produced and released one or more diffusible factors that induce growth inhibition and coccoid conversion of H. pylori cells. In contrast, both H. pylori and L. fermentum secreted factors that promote survival of S. mitis during the stationary phase of growth. Using a metabolomics approach, we identified compounds that might be responsible for the conversion of H. pylori from spiral to coccoid cells. This study provide evidences that gastric bacteria influences H. pylori physiology and therefore possibly the diseases this bacterium causes.


Asunto(s)
Mucosa Gástrica/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Limosilactobacillus fermentum/fisiología , Streptococcus mitis/fisiología , Técnicas de Cocultivo , Microbioma Gastrointestinal , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA