Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(6): 104781, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37146969

RESUMEN

Intestinal mucous layers mediate symbiosis and dysbiosis of host-microbe interactions. These interactions are influenced by the mucin O-glycan degrading ability of several gut microbes. The identities and prevalence of many glycoside hydrolases (GHs) involved in microbial mucin O-glycan breakdown have been previously reported; however, the exact mechanisms and extent to which these GHs are dedicated to mucin O-glycan degradation pathways warrant further research. Here, using Bifidobacterium bifidum as a model mucinolytic bacterium, we revealed that two ß-N-acetylglucosaminidases belonging to the GH20 (BbhI) and GH84 (BbhIV) families play important roles in mucin O-glycan degradation. Using substrate specificity analysis of natural oligosaccharides and O-glycomic analysis of porcine gastric mucin (PGM) incubated with purified enzymes or B. bifidum carrying bbhI and/or bbhIV mutations, we showed that BbhI and BbhIV are highly specific for ß-(1→3)- and ß-(1→6)-GlcNAc linkages of mucin core structures, respectively. Interestingly, we found that efficient hydrolysis of the ß-(1→3)-linkage by BbhI of the mucin core 4 structure [GlcNAcß1-3(GlcNAcß1-6)GalNAcα-O-Thr] required prior removal of the ß-(1→6)-GlcNAc linkage by BbhIV. Consistent with this, inactivation of bbhIV markedly decreased the ability of B. bifidum to release GlcNAc from PGM. When combined with a bbhI mutation, we observed that the growth of the strain on PGM was reduced. Finally, phylogenetic analysis suggests that GH84 members may have gained diversified functions through microbe-microbe and host-microbe horizontal gene transfer events. Taken together, these data strongly suggest the involvement of GH84 family members in host glycan breakdown.


Asunto(s)
Acetilglucosaminidasa , Proteínas Bacterianas , Bifidobacterium bifidum , Mucinas , Animales , Acetilglucosaminidasa/química , Acetilglucosaminidasa/metabolismo , Proteínas Bacterianas/metabolismo , Bifidobacterium bifidum/clasificación , Bifidobacterium bifidum/enzimología , Bifidobacterium bifidum/genética , Mucinas/metabolismo , Filogenia , Porcinos
2.
BMC Infect Dis ; 24(1): 905, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39223499

RESUMEN

BACKGROUND: Cholera is a diarrheal disease recognized for being caused by toxin-producing Vibrio (V.) cholerae. This study aims to assess the vibriocidal and immunomodulatory properties of derived cell-free supernatants (CFSs) of Bifidobacterium (B.) bifidum and Lactobacillus (L.) acidophilus encapsulated in chitosan nanoparticles (CFSb-CsNPs and CFSa-CsNPs) against clinical multi-drug resistance (MDR) isolates of V. cholerae O1 El Tor. METHODS: We synthesized CFSb-CsNPs and CFSa-CsNPs using the ionic gelation technique. The newly nanostructures were characterized for size, surface zeta potential, morphology, encapsulation efficacy (EE), stability in different pH values and temperatures, release profile, and in vitro cytotoxicity. The antimicrobial and antibiofilm effects of the obtained nanocomposites on clinical MDR isolates (N = 5) of V. cholerae E1 Tor O1 were investigated by microbroth dilution assay and crystal violet staining, respectively. We conducted quantitative real-time PCR (qRT-PCR) to analyze the relative gene expressions of Bap, Rbmc, CTXAB, and TCP in response to CFSb-CsNPs and CFSa-CsNPs. Additionally, the immunomodulatory effects of formulated structures on the expression of TLR2 and TLR4 genes in human colorectal adenocarcinoma cells (Caco-2) were studied. RESULTS: Nano-characterization analyses indicated that CFSb-CsNPs and CFSa-CsNPs exhibit spherical shapes under scanning electron microscopy (SEM) imaging, with mean diameters of 98.16 ± 0.763 nm and 83.90 ± 0.854 nm, respectively. Both types of nanoparticles possess positive surface charges. The EE% of CFSb-CsNPs was 77 ± 4.28%, whereas that of CFSa-CsNPs was 62.5 ± 7.33%. Chitosan (Cs) encapsulation leads to increased stability of CFSs in simulated pH conditions of the gastrointestinal tract in which the release rates for CFSb-CsNPs and CFSa-CsNPs were reached at 58.00 ± 1.24% and 55.01 ± 1.73%, respectively at pH = 7.4. The synergistic vibriocidal effects observed from the co-administration of both CFSb-CsNPs and CFSa-CsNPs, as evidenced by a fractional inhibitory concentration (FIC) index of 0.57, resulting in a significantly lower MIC of 2.5 ± 0.05 mg/mL (p < 0.0001) compare to individual administration. The combined antibacterial effect of CFSb-CsNPs and CFSa-CsNPs on Bap (0.14 ± 0.05), Rbmc (0.24 ± 0.01), CTXAB (0.30 ± 0.09), and TCP (0.38 ± 0.01) gene expression was significant (p < 0.001). Furthermore, co-administration of CFSb-CsNPs and CFSa-CsNPs also demonstrated the potency of suppressing TLR 2/4 (0.20 ± 0.01 and 0.12 ± 0.02, respectively) gene expression (p = 0.0019) and reduced Caco-2 cells attached bacteria to 526,000 ± 51,46 colony-forming units/mL (11.19%) (p < 0.0001). CONCLUSION: Our study revealed that encapsulating CFSs within CsNPs enhances their vibriocidal activity by improving stability and enabling a controlled release mechanism at the site of interaction between the host and bacteria. Additionally, the simultaneous use of CFSb-CsNPs and CFSa-CsNPs exhibited superior vibriocidal potency against MDR V. cholerae O1 El Tor strains, indicating these combinations as a potential new approach against MDR bacteria.


Asunto(s)
Antibacterianos , Bifidobacterium bifidum , Quitosano , Lactobacillus acidophilus , Nanopartículas , Vibrio cholerae O1 , Quitosano/química , Quitosano/farmacología , Lactobacillus acidophilus/efectos de los fármacos , Vibrio cholerae O1/efectos de los fármacos , Humanos , Nanopartículas/química , Antibacterianos/farmacología , Antibacterianos/química , Bifidobacterium bifidum/fisiología , Farmacorresistencia Bacteriana Múltiple , Probióticos/farmacología , Probióticos/administración & dosificación , Biopelículas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Células CACO-2
3.
J Dairy Sci ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38969003

RESUMEN

Human milk oligosaccharides (HMOs) promote the growth and adhesion of bifidobacteria, thus exerting multiple biological functions on intestinal epithelial cells. Bacterial surface proteins play an important role in bacterial-host intestinal epithelial interactions. In this study, we aim to investigate the effects of surface proteins extracted from Bifidobacterium bifidum DNG6 (B. bifidum DNG6) consuming 2'-fucosyllactose (2'-FL) on Caco-2 cells monolayer barrier injury induced by lipopolysaccharide, compared with lactose (Lac) and galacto-oligosaccharides (GOS). Our results indicated that 2'-FL may promote the surface proteins of B. bifidum DNG6 to improve intestinal barrier injury by positively regulating the NF-κB signaling pathway, reducing inflammation(TNF-α reduced to 50.34%, IL-6 reduced to 22.83%, IL-1ß reduced to 37.91%, and IL-10 increased to 63.47%)and strengthening tight junction (ZO-1 2.39 times, Claudin-1 2.79 times, and Occludin 4.70 times). The findings of this study indicate that 2'-FL can further regulate intestinal barrier damage by promoting the alteration of B. bifidum DNG6 surface protein. The findings of this research will also provide theoretical support for the development of synbiotic formulations.

4.
AAPS PharmSciTech ; 25(3): 61, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38485901

RESUMEN

Mesalamine is a first-line drug for the treatment of inflammatory bowel diseases. However, its premature release associated with marketed formulations leads to adverse effects like gastric trouble, vomiting, and diarrhoea. To minimize these side effects, colon-targeted drug delivery is essential. Besides conventional pharmacotherapy, bifidogenic probiotics with anti-inflammatory activity has been reported to elicit a significant impact on the remission of ulcerative colitis. Bifidogenic probiotics being acid-labile necessitate developing a gastro-resistant formulation for enhancing the delivery of viable cells to the colon. The present study was aimed at developing a fixed-dose unit dosage form of mucoadhesive hydrogel beads loaded with mesalamine and Bifidobacterium bifidum further encapsulated in Eudragit® capsules for the targeted drug delivery at colonic pH. The hydrogel beads were prepared by ionotropic gelation, with the effect of single and dual-crosslinking approaches on various formulation characteristics studied. Standard size 00 Eudragit® gastro-resistant capsules were prepared and the dried beads were filled inside the capsule shells. The formulation was then evaluated for various parameters, including physicochemical characterization, in vitro biocompatibility and anti-inflammatory activity. No interaction was observed between the drug and the polymers, as confirmed through FTIR, XRD, and DSC analysis. The mean particle size of the beads was ~ 457-485 µm. The optimized formulation showed a drug entrapment efficiency of 95.4 ± 2.58%. The Eudragit® capsule shells disintegrated in approximately 13 min at pH 7.4. The mucoadhesive hydrogel beads sustained the drug release above 18 h, with 50% of the drug released by the end of 12 h. The optimized formulation demonstrated significant (p < 0.05) gastro-resistance, biocompatibility, sustained drug release, cell viability, and anti-inflammatory activity.


Asunto(s)
Bifidobacterium bifidum , Mesalamina , Ácidos Polimetacrílicos , Hidrogeles/farmacología , Colon , Antiinflamatorios/farmacología
5.
Immun Ageing ; 20(1): 56, 2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37872562

RESUMEN

Sarcopenia is closely associated with gut dysbiosis. Probiotics alleviate gut dysbiosis. Therefore, we selected probiotics Lactobacillus paracasei P62 (Lp) and Bifidobacterium bifidum P61 (Bb), which suppressed muscle RING-finger protein-1 (MuRF1) expression and NF-κB activation in C2C12 cells, and examined their effects on muscle mass loss and dysfunction in aged mice. Oral administration of Lp, Bb, or their mix (LB) increased grip strength and treadmill running distance and time. They significantly increased muscle weight in aged mice. They also increased AKT activation, PGC1α, SIRT1, and myosin heavy chain (MyHC) expression, MyHC-positive cell population, and cell size in the gastrocnemius (GA) muscle, while FOXO3a and NF-κB activation, MuRF1, muscle atrophy F-box, and p16 expression, and NF-κB+CD11c+ cell population decreased. Furthermore, they reduced cognitive impairment-like behavior, IL-6 expression, FOXO3a activation, and NF-κB-positive cell population in the hippocampus, GA, and colon, while hippocampal brain-derived neurotropic factor expression increased. They shifted gut microbiota composition in aged mice: they increased Akkermansiaceae and Bacteroidaceae populations, which were positively correlated with total muscle weight and MyHC expression, and decreased Odoribacteraceae and Deferribacteriaceae populations, which were positively correlated with MuRF1 and IL-6 expression. LB alleviated sarcopenia- and cognitive impairment-like symptoms more potently than Lp or Bb alone. Based on these findings, probiotics, particularly Lp, Bb, and LB, can alleviate aging-dependent sarcopenia and cognitive impairment by regulating gut microbiota-mediated AKT, NF-κB, and/or FOXO3a signaling pathways.

6.
Int J Mol Sci ; 24(9)2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37175825

RESUMEN

Diabetes mellitus (DM) is a metabolic disorder with an alarming incidence rate and a considerable burden on the patient's life and health care providers. An increase in blood glucose level and insulin resistance characterizes it. Internal and external factors such as urbanization, obesity, and genetic mutations could increase the risk of DM. Microbes in the gut influence overall health through immunity and nutrition. Recently, more studies have been conducted to evaluate and estimate the role of the gut microbiome in diabetes development, progression, and management. This review summarizes the current knowledge addressing three main bacterial species: Bifidobacterium adolescentis, Bifidobacterium bifidum, and Lactobacillus rhamnosus and their influence on diabetes and its underlying molecular mechanisms. Most studies illustrate that using those bacterial species positively reduces blood glucose levels and activates inflammatory markers. Additionally, we reported the relationship between those bacterial species and metformin, one of the commonly used antidiabetic drugs. Overall, more research is needed to understand the influence of the gut microbiome on the development of diabetes. Furthermore, more efforts are required to standardize the model used, concentration ranges, and interpretation tools to advance the field further.


Asunto(s)
Diabetes Mellitus , Microbioma Gastrointestinal , Metformina , Humanos , Glucemia , Microbioma Gastrointestinal/fisiología , Hipoglucemiantes
7.
J Food Sci Technol ; 60(12): 2968-2977, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37786598

RESUMEN

Microencapsulation of B. bifidum F-35 was carried out through emulsification technique in order to increase the microbial load while maintaining the rheological functions of set-yogurt. To produce single-layer (SL) microcapsules of whey protein, the pH was adjusted to 6.4 within Transglutaminase-induced gelation. Sodium alginate was processed as the external layer using calcium-induced gelation (pH 5.5) to produce the double-layer (DL) microcapsule. Scanning electron microscopy revealed that SL and DL microcapsules had sizes of 10 and 280 µm, respectively. The highest microbial load was clearly visible in the DL sample. According to texture profile analysis, the DL sample had the highest levels of gumminess, chewiness, and adhesiveness. The free sample outperformed the encapsulated samples in terms of springiness and cohesiveness. Although the SL sample had the highest viscosity, it produced a deformed gel when firmness was measured. In terms of firmness, the DL sample performed quite well. The viability of encapsulated B. bifidum F-35 in DL was higher than SL microcapsules during storage. Microencapsulation of B. bifidum F-35 with whey protein and sodium alginate is a promising technique that could improve the rheological properties of set-yogurt as a popular vehicle for bioactive ingredients.

8.
Microb Cell Fact ; 21(1): 113, 2022 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672695

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is a gastrointestinal disease characterized by diarrhea, rectal bleeding, abdominal pain, and weight loss. Recombinant probiotics producing specific proteins with IBD therapeutic potential are currently considered novel drug substitutes. In this study, a Bifidobacterium bifidum BGN4-SK strain was designed to produce the antioxidant enzymes streptococcal superoxide dismutase (SOD) and lactobacillus catalase (CAT), and a B. bifidum BGN4-pBESIL10 strain was proposed to generate an anti-inflammatory cytokine, human interleukin (IL)-10. In vitro and in vivo efficacy of these genetically modified Bifidobacterium strains were evaluated for colitis amelioration. RESULTS: In a lipopolysaccharide (LPS)-stimulated HT-29 cell model, tumor necrosis factor (TNF)-α and IL-8 production was significantly suppressed in the B. bifidum BGN4-SK treatment, followed by B. bifidum BGN4-pBESIL10 treatment, when compared to the LPS-treated control. Synergistic effects on TNF-α suppression were also observed. In a dextran sodium sulphate (DSS)-induced colitis mouse model, B. bifidum BGN4-SK treatment significantly enhanced levels of antioxidant enzymes SOD, glutathione peroxidase (GSH-Px) and CAT, compared to the DSS-only group. B. bifidum BGN4-SK significantly ameliorated the symptoms of DSS-induced colitis, increased the expression of tight junction genes (claudin and ZO-1), and decreased pro-inflammatory cytokines IL-6, IL-1ß and TNF-α. CONCLUSIONS: These findings suggest that B. bifidum BGN4-SK ameliorated DSS-induced colitis by generating antioxidant enzymes, maintaining the epithelial barrier, and decreasing the production of pro-inflammatory cytokines. Although B. bifidum BGN4-pBESIL10 exerted anti-inflammatory effects in vitro, the enhancement of IL-10 production and alleviation of colitis were very limited.


Asunto(s)
Bifidobacterium bifidum , Colitis , Enfermedades Inflamatorias del Intestino , Probióticos , Animales , Antiinflamatorios/efectos adversos , Antioxidantes/metabolismo , Bifidobacterium bifidum/genética , Colitis/tratamiento farmacológico , Colitis/terapia , Citocinas/metabolismo , Sulfato de Dextran/efectos adversos , Sulfato de Dextran/metabolismo , Modelos Animales de Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Interleucina-10/metabolismo , Lipopolisacáridos , Ratones , Probióticos/uso terapéutico , Superóxido Dismutasa/efectos adversos , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
J Appl Microbiol ; 133(2): 908-921, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35488863

RESUMEN

AIMS: Bronchopulmonary dysplasia (BPD) is a common respiratory disease in newborns; however, there is no effective treatment. We aimed to investigate the effects of the potential probiotics Limosilactobacillus reuteri and Bifidobacterium bifidum on BPD using 16S rDNA sequencing and metabolomics methods. METHODS AND RESULTS: Faecal samples were collected from 10 BPD patients and 10 healthy subjects. 16S rDNA sequencing results showed that microbial diversity was decreased and compositions were affected in BPD. Escherichia-Shigella and Clostridium_sensu_stricto_1 were increased in the BPD group, and Enterobacteriaceae, Megamonas, Blautia, Lactobacillus (Limosilactobacillus), [Eubacterium]_coprostanoligenes_group, Phascolarctobacterium and Bifidobacterium were reduced. Metabolomics analysis identified 129 differentiated metabolites that were changed in BPD patients, and they were associated with a preference for carbohydrate metabolism in translation and metabolism during genetic information processing. Correlation analysis revealed a remarkable relationship between gut microbiota and metabolites. Subsequently, a BPD cell model was constructed to test the effect of the potential probiotics. Cell function experiments verified that treatment with the potential probiotics L. reuteri and B. bifidum promoted proliferation and inhibited apoptosis of hyperoxia-induced MLE-12 cells. In addition, treatment with the potential probiotics L. reuteri and B. bifidum reduced inflammation and oxidative stress damage. CONCLUSIONS: Treatment with the potential probiotics L. reuteri and B. bifidum could alleviate BPD and reduce inflammation and oxidative stress damage. SIGNIFICANCE AND IMPACT: This study was the first to report positive roles for the potential probiotics L. reuteri and B. bifidum in BPD. The potential probiotics L. reuteri and B. bifidum were shown to reduce inflammation and oxidative stress damage in BPD. This study provided new insights on the pathogenesis and treatment of BPD.


Asunto(s)
Bifidobacterium bifidum , Displasia Broncopulmonar , Probióticos , Bifidobacterium bifidum/fisiología , ADN Ribosómico , Humanos , Recién Nacido , Inflamación , Metaboloma , Probióticos/farmacología
10.
Biochem Biophys Res Commun ; 571: 104-109, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34314995

RESUMEN

The acoustic propagation characteristic of ultrasound determines that the energy of ultrasound beam will decrease with the increase of its propagation depth in the body. Similarly, the energy of High Intensity Focused Ultrasound (HIFU) will be attenuated with the increase of HIFU propagation depth in the body. Ensuring sufficient ultrasound energy deposition in the HIFU ablation region for tumor ablation is usually achieved by increasing the ultrasound irradiation power or prolonging the ultrasound ablation time. However, these two methods may damage the normal tissue adjacent to the HIFU ablation region. Herein, we constructed the nanoparticles conjugated with tumor-homing bacteria as the biological tumor-homing synergist to facilitate HIFU-mediated tumor ablation avoiding the potential safety risk. In our strategy, Bifidobacterium bifidum (B.bifidum) was selectively colonized in the hypoxic region of solid tumors after been injected into 4T1 breast cancer bearing-BALB/c mice via the tail vein due to its anaerobic growth characteristic. The amount of B. bifidum with negative surface potential in the hypoxic region of solid tumors was increased by its anaerobic proliferation. Polyethylenimine (PEI) -modified Poly (lactic-co-glycolic) acid nanoparticles loaded sodium bicarbonate (PEI-PLGA-NaHCO3-NPs) with positive surface potential injected into 4T1 breast cancer bearing-BALB/c mice via the tail vein displayed the tumor-homing ability by the electrostatic adsorption with B. bifidum colonized solid tumors. PEI-PLGA-NaHCO3-NPs could release NaHCO3 to produce carbon dioxide (CO2) as cavitation nuclei inside the acidic microenvironment of solid tumors. When HIFU irradiated solid tumors contained with more cavitation nuclei, the ultrasound energy deposition at the tumor region was increased to destroy the tumors more effectively. Meanwhile, the improved efficiency of HIFU-mediated tumor ablation reduced the dependence of the tumor ablation on the ultrasound energy dose, which improved the safety of HIFU-mediated tumor ablation to the non-targeted ablation tissue. This tumor-homing synergist shows the potential application value on the HIFU-mediated tumor ablation in the clinical.


Asunto(s)
Antineoplásicos/farmacología , Bifidobacterium bifidum/aislamiento & purificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/microbiología , Nanopartículas/química , Polietileneimina/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Animales , Antineoplásicos/química , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Ratones , Ratones Endogámicos BALB C , Polietileneimina/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ondas Ultrasónicas
11.
Biochem Biophys Res Commun ; 547: 155-161, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33610915

RESUMEN

Bifidobacterium bifidum is one of the most abundant members of the gut microbiota at the early stage of life. The established association of the bacterium with the human gut confers health benefits. Such successful persistence of B. bifidum necessitates metabolic adaptation to the host-derived carbohydrates, a process which is poorly understood. The current study focuses on revealing the genomic-based phylogeny (phylogenomics) of B. bifidum and utilizing comparative genomics to decipher the glycolytic abilities of bifidobacterial strains isolated from different human body niches (feces, human gut, vagina, and breast milk). When the phylogenomic analysis was performed on 95 B. bifidum strains, currently available on the RefSeq database, the bacterium was clearly distinguished from other members of the Bifidobacterium genus. Furthermore, a pairwise genomic comparison indicated that a large proportion of orthologous gene families were shared among the B. bifidum strains. These findings highlight the notion that the B. bifidum species is genetically similar and may perform similar functions in their host. When 15 B. bifidum genomes representing strains from different human body niches were annotated, the resulting functional profile showed the presence of enriched proteins involved in carbohydrate utilization. Moreover, mining the 15 B. bifidum genomes for the presence of Carbohydrate-Active Enzyme (CAZY) systems, the analysis found the existence of diverse protein families which include glycosyl hydrolases, glycosyl transferases, carbohydrate-binding modules, and carbohydrate esterases. Collectively, these CAZY systems enables B. bifidum to utilize host-derived glycans (e.g., mucin) and diet-derived carbohydrates (e.g., starch). In contrast, a correlation analysis revealed that B. bifidum strains isolated from the different body niches were indistinguishable in the context of presence-absence of CAZY systems. These findings emphasize the valuable use of comparative genomics in deciphering the glycolytic abilities of B. bifidum and consequently its adaptation to carbohydrate utilization in the human gut environment.


Asunto(s)
Bifidobacterium bifidum/genética , Carbohidratos de la Dieta/metabolismo , Microbioma Gastrointestinal , Adaptación Fisiológica/genética , Bifidobacterium bifidum/metabolismo , Biología Computacional/métodos , Genoma Bacteriano , Genómica , Humanos , Filogenia
12.
Microb Cell Fact ; 20(1): 16, 2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33468130

RESUMEN

BACKGROUND: Bifidobacterium spp. are representative probiotics that play an important role in the health of their hosts. Among various Bifidobacterium spp., B. bifidum BGN4 exhibits relatively high cell adhesion to colonic cells and has been reported to have various in vivo and in vitro bio functionalities (e.g., anti-allergic effect, anti-cancer effect, and modulatory effects on immune cells). Interleukin-10 (IL-10) has emerged as a major suppressor of immune response in macrophages and other antigen presenting cells and plays an essential role in the regulation and resolution of inflammation. In this study, recombinant B. bifidum BGN4 [pBESIL10] was developed to deliver human IL-10 effectively to the intestines. RESULTS: The vector pBESIL10 was constructed by cloning the human IL-10 gene under a gap promoter and signal peptide from Bifidobacterium spp. into the E. coli-Bifidobacterium shuttle vector pBES2. The secreted human IL-10 from B. bifidum BGN4 [pBESIL10] was analyzed by sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE), Western Blotting, and enzyme-linked immunosorbent assay (ELISA). More than 1,473 ± 300 ng/mL (n = 4) of human IL-10 was obtained in the cell free culture supernatant of B. bifidum BGN4 [pBESIL10]. This productivity is significantly higher than other previously reported human IL-10 level from food grade bacteria. In vitro functional evaluation of the cell free culture supernatant of B. bifidum BGN4 [pBESIL10] revealed significantly inhibited interleukin-6 (IL-6) production in lipopolysaccharide (LPS)-induced Raw 264.7 cells (n = 6, p < 0.0001) and interleukin-8 (IL-8) production in LPS-induced HT-29 cells (n = 6, p < 0.01) or TNFα-induced HT-29 cells (n = 6, p < 0.001). CONCLUSION: B. bifidum BGN4 [pBESIL10] efficiently produces and secretes significant amounts of biologically active human IL-10. The human IL-10 production level in this study is the highest of all human IL-10 production reported to date. Further research should be pursued to evaluate B. bifidum BGN4 [pBESIL10] producing IL-10 as a treatment for various inflammation-related diseases, including inflammatory bowel disease, rheumatoid arthritis, allergic asthma, and cancer immunotherapy.


Asunto(s)
Bifidobacterium bifidum/metabolismo , Escherichia coli/metabolismo , Interleucina-10/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Secuencia de Bases , Bifidobacterium bifidum/genética , Western Blotting , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Células HT29 , Humanos , Interleucina-10/genética , Ratones , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Células RAW 264.7 , Homología de Secuencia de Ácido Nucleico
13.
Int J Mol Sci ; 22(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34299216

RESUMEN

Bifidobacterium bifidum strains, an important component of probiotic foods, can form biofilms on abiotic surfaces, leading to increased self-resistance. However, little is known about the molecular mechanism of B. bifidum biofilm formation. A time series transcriptome sequencing and untargeted metabolomics analysis of both B. bifidum biofilm and planktonic cells was performed to identify key genes and metabolites involved in biofilm formation. Two hundred thirty-five nonredundant differentially expressed genes (DEGs) (including vanY, pstS, degP, groS, infC, groL, yajC, tadB and sigA) and 219 nonredundant differentially expressed metabolites (including L-threonine, L-cystine, L-tyrosine, ascorbic acid, niacinamide, butyric acid and sphinganine) were identified. Thirteen pathways were identified during the integration of both transcriptomics and metabolomics data, including ABC transporters; quorum sensing; two-component system; oxidative phosphorylation; cysteine and methionine metabolism; glutathione metabolism; glycine, serine and threonine metabolism; and valine, leucine and isoleucine biosynthesis. The DEGs that relate to the integration pathways included asd, atpB, degP, folC, ilvE, metC, pheA, pstS, pyrE, serB, ulaE, yajC and zwf. The differentially accumulated metabolites included L-cystine, L-serine, L-threonine, L-tyrosine, methylmalonate, monodehydroascorbate, nicotinamide, orthophosphate, spermine and tocopherol. These results indicate that quorum sensing, two-component system and amino acid metabolism are essential during B. bifidum biofilm formation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bifidobacterium bifidum/fisiología , Biopelículas/crecimiento & desarrollo , Proteínas Bacterianas/genética , Bifidobacterium bifidum/genética , Bifidobacterium bifidum/metabolismo , Perfilación de la Expresión Génica , Metaboloma , Percepción de Quorum , Transcriptoma , Triticum/microbiología
14.
Int J Mol Sci ; 22(9)2021 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-34063173

RESUMEN

Recent studies have suggested that flavonoids such as quercetin and probiotics such as Bifidobacterium bifidum (Bf) and Lactobacillus gasseri (Lg) could play a relevant role in inhibiting colon cancer cell growth. Our study investigated the role of dietary supplementation with microencapsulated probiotics (Bf and Lg) along with quercetin in the development of mouse colorectal cancer (CRC). Methods: Adenomatous polyposis coli/multiple intestinal neoplasia (ApcMin/+) mice were fed a standard diet or the same diet supplemented with microencapsulated probiotics (Bf and Lg strains, 107 CFU/100 g food) or both probiotics strains plus microencapsulated quercetin (15 mg/100 g food) for 73 days. Changes in body and organ weights, energy metabolism, intestinal microbiota, and colon tissue were determined. The expression of genes related to the Wnt pathway was also analyzed in colon samples. Results: Dietary supplementation with microencapsulated probiotics or microencapsulated probiotics plus quercetin reduced body weight loss and intestinal bleeding in ApcMin/+ mice. An improvement in energy expenditure was observed after 8 weeks but not after 10 weeks of treatment. A supplemented diet with microencapsulated Bf and Lg reduced the number of aberrant crypt foci (ACF) and adenomas by 45% and 60%, respectively, whereas the supplementation with Bf, Lg and quercetin decreased the number of ACF and adenomas by 57% and 80%, respectively. Microencapsulated Bf and Lg in combination with quercetin could exert inhibition of the canonical Wnt/ß-catenin signaling pathway in the colon of ApcMin/+ mice Conclusions: The administration of microencapsulated Bf and Lg, individually or in combination with quercetin, inhibits the CRC development in ApcMin/+ mice.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Bifidobacterium bifidum/citología , Carcinogénesis/patología , Células Inmovilizadas/citología , Neoplasias Colorrectales/patología , Lactobacillus gasseri/citología , Quercetina/farmacología , Animales , Peso Corporal/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Colon/patología , Recuento de Colonia Microbiana , Neoplasias Colorrectales/genética , Metabolismo Energético/efectos de los fármacos , Heces/microbiología , Conducta Alimentaria , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones Endogámicos C57BL , Sangre Oculta , Tamaño de los Órganos/efectos de los fármacos , Probióticos/farmacología , Vía de Señalización Wnt/efectos de los fármacos
15.
Ter Arkh ; 93(4): 456-464, 2021 Apr 15.
Artículo en Ruso | MEDLINE | ID: mdl-36286781

RESUMEN

AIM: To determine the clinical efficacy and safety of the sorbed probiotics Bifidobacterium bifidum 1 (5108 KОЕ) and B. bifidum 1 (5107 KОЕ) in combination with Lactobacillus plantarum 8P-А3 in the complex therapy of pneumonia caused by SARS-CoV-2 in adult patients without severe risk factors. MATERIALS AND METHODS: An open, randomized prospective study included 100 patients (45 men, 55 women), aged 18 to 60 years without risk factors for severe COVID-19 with pneumonia confirmed by computed tomography, and an area of lung lesion no more than 75% (moderate forms). SARS-CoV-2 RNA in nasal and oropharyngeal swabs (RT-PCR) was detected in 72% of the participants, in the rest it was highly probable in terms of the aggregate parameters. Diagnostics of COVID-19 and its severity, the appointment of a standard examination and treatment were carried out in accordance with the Temporary Methodological Recommendations of the Ministry of Health of Russia, version 8 of 09.03.2020. This publication presents the results of using B. bifidum 1 (3 capsules twice a day for 10 days) during the peak of clinical manifestations (in a hospital). RESULTS: In those who received sorbed B. bifidum 1, by the 10th day of treatment, the frequency of weakness was 32% lower (RR 0.55 [95% CI 0.240.73], OR 0.25 [0.110.59]); hypoosmia/dysgeusia by 22% (RR 0.42 [0.050.65], OR 0.40 [0.170.90]) and cough by 24% (RR 0.39 [0.070.60], OR 0.38 [0.170.84]). B. bifidum 1 reduced the average duration of weakness by 3 days [1.14.9], hypoosmia/dysgeusia by 3.2 days [1.35.1], cough by 1.9 days [0.43,4], dyspnea by 1.8 days [0.72.7], diarrhea by 1.7 days [0.13.5]; reduced the risk of antibiotic-associated diarrhea by 20% (RR 0.77 [0.240.93], OR 0.18 [0.050.68]). Due to the deterioration of the condition and the increase in the symptoms of respiratory failure, additional treatment was required less often by 24% (p=0.005). After the end of the intervention, the frequency of virologic debridement, levels of CRP, leukocytes, lymphocytes, platelets and the degree of lung damage on computed tomography did not statistically differ in the compared groups. No side effects of B. bifidum 1 (5108 KОЕ) have been identified. CONCLUSION: The use of sorbed B. bifidum 1 (5108 KОЕ) improved the well-being of patients without risk factors with moderate viral (SARS-CoV-2) pneumonia and reduced the duration of diarrheal syndrome in a short time. The safety profile of their use was high. More research is needed to clarify the anti-inflammatory effects of the sorbed probiotic.

16.
Microb Pathog ; 145: 104207, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32325236

RESUMEN

As of present, a number of studies have shown anti-cancer effects of different strains of probiotics, but the precise host immunological mechanisms of these antitumor effects remain unclear. Thus, the aim of current study was to investigate the preventive-therapeutic effects of oral versus intravenous administration of probiotic Bifidobacterium bifidum on immune response and tumor growth of C57BL/6 mice bearing transplanted TC-1 cell of human papillomavirus (HPV)-related tumor, expressing HPV-16 E6/E7 oncogenes. Our major findings are that the intravenous or oral administration of Bifidobacterium bifidum effectively induces antitumor immune responses and inhibits tumor growth in mice. Compared to oral route only, intravenous administration of probiotic Bifidobacterium bifidum into tumor-bearing mice leads to the activation of tumor-specific IL-12 and IFN-γ, lymphocyte proliferation, CD8+ cytolytic responses that control and eradicate tumor growth. These observations meant intravenous administration of probiotics is an effective anticancer approach through modulation of the immune system. The potential of probiotic Bifidobacterium bifidum as an immunomodulator in the treatment of cervical cancer could be further explored.


Asunto(s)
Alphapapillomavirus , Bifidobacterium bifidum , Probióticos , Neoplasias del Cuello Uterino , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Papillomaviridae , Neoplasias del Cuello Uterino/terapia
17.
Eur J Nutr ; 59(5): 2119-2130, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31342226

RESUMEN

PURPOSE: Many studies have investigated the association between intestinal barrier impairment and the onset of atopic dermatitis (AD). The gut microbiota is essential to maintain physiological homeostasis and immune regulation of host. Therefore, the objectives were to determine the effects of probiotics on the clinical symptoms, immune responses, and gut microbiota in AD patients. METHODS: 109 patients were randomly divided into 4 groups, including placebo group, oligosaccharides group, Bifidobacterium bifidum CCFM16 group, and Lactobacillus plantarum CCFM8610 group. At the end of the experiment, serological indicators, SCORAD, and DLQI indices were assessed. V3-V4 region of the 16S ribosomal RNA gene was sequenced to evaluate changes in the gut microbiota. Linear discriminant analysis (LDA) effect size was used to uncover microbial biomarkers and PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States) was used to predict gene family abundances based on 16S information. RESULTS: The results demonstrated that CCFM8610 significantly decreased the SCORAD index, and increased the serum IL-10 levels. Supplement with CCFM8610 and CCFM16 significantly influenced the alpha diversity, increased the proportion of Bacteroidetes, and reduced the F/B ratio. CCFM8610 treatment downregulated the functional genes of gut microbiota involving Staphylococcus aureus infection and upregulated the steroid hormone biosynthesis. CONCLUSION: The results indicated a positive correlation between decreased SCORAD index and CCFM8610 treatment, and that CCFM8610 regulated the immune responses in AD patients. CCFM8610 treatment influences the gut microbiota composition and functional changes. In conclusion, L. plantarum CCFM8610 exerts the strain-specific amelioration effects on patients with AD. TRIAL REGISTRATION: ChiCTR1800015330 (Clinicaltrials.gov Identifier).


Asunto(s)
Dermatitis Atópica , Microbioma Gastrointestinal , Probióticos , Humanos , Inmunidad , Filogenia , Proyectos Piloto
18.
Cell Mol Biol (Noisy-le-grand) ; 66(7): 111-117, 2020 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-33287930

RESUMEN

Bifidobacterium selectively colonizes the infants' intestinal tract, and the relevant coliform bacteria in adults are particularly beneficial because of their enhanced capability to prevent pathogens of gastro intestine by direct antimicrobial action and relieve infection, which led to their intensification, the antibacterial activities of titanium nanoparticles producing by some bacteria, makes them attractive as a new agent against pathogenic bacteria. In our present study, we used a probiotic bacteria Bifidobacterium bifidum which was isolated from the commercial market capsule to produce TiO2 nanoparticles and study the biologically characterized nanoparticle using various techniques like Scanning Electron Microscopic (SEM), atomic force microscopy (AFM), and study its antimicrobial activity against a bacteria isolated from the stool of patients suffering from acute diarrhea. The results showed that the morphological characteristics of nanoparticles were found to have a spherical shape and mean size of 81 nm by AFM while scanning electron microscope viewed as an oval shape with anatase form synthesized by B. bifidum. TiO2-NP synthesized by B. bifidum had an inhibitory effect against P. aeruginosa, A. baumanii, K. pneumonia at a concentration 16 mg/ml and 32 mg/ml towards E. coli and S. typhi, the minimum inhibitory concentration (MIC) against pathogenic bacteria isolated from acute diarrhea included Pseudomonas aeruginosa, Acinetobacter baumanii, Klebsiella pneumonia, E.coli and salmonella typhi was utilized to determine the antibacterial impact of the synthesized TIO2 nanoparticles. Our biologically synthesized titanium nanoparticles were effective against all the tested pathogenic bacteria at various degrees and had a probable role in significantly greater antimicrobial efficacy against all isolates under study. This trial may have considerable significance for the prevention of antibiotic resistance associated diarrhea in hospitals.


Asunto(s)
Antiinfecciosos/farmacología , Bifidobacterium bifidum/fisiología , Nanopartículas/química , Probióticos/farmacología , Titanio/farmacología , Bifidobacterium bifidum/ultraestructura , Pruebas de Sensibilidad Microbiana , Nanopartículas/ultraestructura
19.
Appl Microbiol Biotechnol ; 104(13): 5915-5928, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32367312

RESUMEN

The gut microbiota plays an important role in colorectal cancer (CRC), and the use of probiotics might be a promising intervention method. The aim of our study was to investigate the beneficial effect of Bifidobacterium bifidum CGMCC 15068 on an azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced colitis-associated CRC (CAC) mouse model. CAC was induced by an intra-peritoneal injection of AOM (10 mg/kg) and three 7-day cycles of 2% DSS in drinking water with a 14-day recovery period between two consecutive DSS administrations. B. bifidum CGMCC 15068 (3 × 109 CFU/mL) was gavaged once daily during the recovery period. Then, the faecal microbial composition and metabolome were profiled using the 16S rRNA sequencing technology and gas chromatography-mass spectrometry (GC-MS), respectively. The administration of B. bifidum CGMCC 15068 attenuated tumourigenesis in the CAC mouse model. In addition, B. bifidum CGMCC 15068 pre-treatment increased the relative abundance of Akkermansia, Desulfovibrionaceae, Romboutsia, Turicibacter, Verrucomicrobiaceae, Ruminococcaceae_UCG_013, Lachnospiraceae_UCG_004, and Lactobacillus. Meanwhile, B. bifidum CGMCC 15068 altered metabolites involved in the citrate cycle (TCA cycle), glycolysis, butyrate metabolism, fatty acid biosynthesis, and galactose metabolism. Several significant correlations were identified between the differentially abundant microbes and metabolites. These findings supported the beneficial role of B. bifidum CGMCC 15068 in intestinal health by modulating dysbiosis and the gut metabolic profile. The manipulation of the gut microbial composition using probiotics might be a promising prevention strategy for CRC. Long-term and large-scale clinical trials are warranted for the potential clinical applications of this strategy in the future.


Asunto(s)
Bifidobacterium bifidum/fisiología , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Metaboloma/efectos de los fármacos , Probióticos/administración & dosificación , Animales , Azoximetano/toxicidad , Carcinogénesis/efectos de los fármacos , Neoplasias Asociadas a Colitis/inducido químicamente , Neoplasias Asociadas a Colitis/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Heces/química , Heces/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Probióticos/farmacología
20.
J Dairy Sci ; 103(11): 9825-9834, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32896399

RESUMEN

Adhesion to the intestinal mucosa is the prerequisite for bifidobacteria to colonize and exert biological functions, whereas the choice of carbon source affects the ability of bifidobacteria to adhere to and interact with intestinal epithelial cells. However, knowledge about the relationship between human milk oligosaccharide consumption by bifidobacteria and its adhesion is still limited. In this study, we aim to investigate the effect of 2'-fucosyllactose (2'-FL) as the carbon source on the growth and adhesion properties of Bifidobacterium bifidum DNG6, and make comparisons with galactooligosaccharides and glucose. We found that the growth and adhesion properties of B. bifidum DNG6 grown in different carbon sources were varied. The 2'-FL as a carbon source improves the adhesion ability of B. bifidum DNG6. The expression of adhesion-associated genes was significantly higher in B. bifidum DNG6 grown in 2'-FL after incubation with Caco-2 cells compared with that in galactooligosaccharides and glucose. Our results indicated that 2'-FL may promote B. bifidum DNG6 adhesion to Caco-2 cells through high expression of genes encoding adhesion proteins. The findings of this study contribute to a better understanding of the involvement of human milk oligosaccharides in the adhesion of bifidobacteria and further support the potential application of 2'-FL as a prebiotic in infant nutritional supplements.


Asunto(s)
Bifidobacterium bifidum/metabolismo , Trisacáridos/metabolismo , Animales , Adhesión Bacteriana , Bifidobacterium bifidum/efectos de los fármacos , Células CACO-2 , Carbono/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestinos/microbiología , Leche Humana/fisiología , Oligosacáridos/metabolismo , Trisacáridos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA