Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Gut Microbes ; 16(1): 2334967, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38630006

RESUMEN

Human milk oligosaccharides (HMOs) are vital milk carbohydrates that help promote the microbiota-dependent growth and immunity of infants. Sialic acid (SA) is a crucial component of sialylated milk oligosaccharides (S-MOs); however, the effects of SA supplementation in lactating mothers on S-MO biosynthesis and their breastfed infants are unknown. Probiotic intervention during pregnancy or lactation demonstrates promise for modulating the milk glycobiome. Here, we evaluated whether SA and a probiotic (Pro) mixture could increase S-MO synthesis in lactating mothers and promote the microbiota development of their breastfed neonates. The results showed that SA+Pro intervention modulated the gut microbiota and 6'-SL contents in milk of maternal rats more than the SA intervention, which promoted Lactobacillus reuteri colonization in neonates and immune development. Deficient 6'-SL in the maternal rat milk of St6gal1 knockouts (St6gal1-/-) disturbed intestinal microbial structures in their offspring, thereby impeding immune tolerance development. SA+Pro intervention in lactating St6gal1± rats compromised the allergic responses of neonates by promoting 6'-SL synthesis and the neonatal gut microbiota. Our findings from human mammary epithelial cells (MCF-10A) indicated that the GPR41-PI3K-Akt-PPAR pathway helped regulate 6'-SL synthesis in mammary glands after SA+Pro intervention through the gut - breast axis. We further validated our findings using a human-cohort study, confirming that providing SA+Pro to lactating Chinese mothers increased S-MO contents in their breast milk and promoted gut Bifidobacterium spp. and Lactobacillus spp. colonization in infants, which may help enhance immune responses. Collectively, our findings may help alter the routine supplementation practices of lactating mothers to modulate milk HMOs and promote the development of early-life gut microbiota and immunity.


Asunto(s)
Microbioma Gastrointestinal , Ácido N-Acetilneuramínico , Femenino , Lactante , Embarazo , Humanos , Animales , Ratas , Lactancia , Estudios de Cohortes , Fosfatidilinositol 3-Quinasas , Leche Humana , Inmunidad
2.
Microorganisms ; 12(4)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38674582

RESUMEN

Hyperuricemia is a prevalent metabolic disorder that arises from abnormal purine metabolism and reduced excretion of uric acid (UA). The gut microbiota plays a significant role in the biosynthesis and excretion of UA. Probiotics capable of purine degradation possess the potential to prevent hyperuricemia. Our study aimed to screen probiotics in areas with abundant dairy products and longevity populations in China, which could attenuate the level of UA and explore the underlying mechanism. In this study, twenty-three lactic acid bacteria isolated from healthy Chinese infant feces and traditional fermented foods such as hurood and lump milk were evaluated for the ability to tolerance acid, bile, artificial gastric juice, and artificial intestinal juice to determine the potential of the candidate strains as probiotics. Eight strains were identified as possessing superior tolerance to simulated intestinal conditions and were further analyzed by high-performance liquid chromatography (HPLC), revealing that Limosilactobacillus reuteri HCS02-001 (Lact-1) and Lacticaseibacillus paracasei HCS17-040 (Lact-2) possess the most potent ability to degrade purine nucleosides. The effect of Lact-1 and Lact-2 on hyperuricemia was evaluated by intervening with them in the potassium oxonate and adenine-induced hyperuricemia Balb/c mice model in vivo. Our results showed that the level of serum UA in hyperuricemic mice can be efficiently reduced via the oral administration of Lact-1 (p < 0.05). It significantly inhibited the levels of liver inflammatory cytokines and hepatic xanthine oxidase through a TLR4/MyD88/NF-κB pathway across the gut-liver axis. Furthermore, UA transporters ABCG2 and SLC2A9 were substantially upregulated by the intervention of this probiotic. Fecal ATP levels were significantly induced, while fecal xanthine dehydrogenase and allantoinase levels were increased following probiotics. RNA sequencing of HT-29 cells line treated with Lact-1 and its metabolites demonstrated significant regulation of pathways related to hyperuricemia. In summary, these findings demonstrate that Limosilactobacillus reuteri HCS02-001 possesses a capacity to ameliorate hyperuricemia by inhibiting UA biosynthesis via enhancing gastrointestinal barrier functions and promoting UA removal through the upregulation of urate transporters, thereby providing a basis for the probiotic formulation by targeting the gut microbiota.

3.
Gut Microbes ; 15(2): 2256749, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37741825

RESUMEN

Gestational diabetes mellitus (GDM) is an increasing public health concern that significantly increases the risk of early childhood allergic diseases. Altered maternal milk glycobiome may strongly affect gut microbiota and enteric-specific Treg cell-mediated development of immune tolerance in GDM infants. In this study, we found that, compared with healthy Chinese mothers, mothers with GDM had significantly lower levels of total and specific human milk oligosaccharides (HMOs) in their colostrum that subsequently increased with extension of lactation. This alteration in HMO profiles significantly delayed colonization of Lactobacillus and Bifidobacterium spp. in their breast-fed infants, resulting in a distinct gut microbial structure and metabolome. Further experiments in GDM mouse models indicated that decreased contents of milk oligosaccharides, mainly 3'-sialyllactose (3'-SL), in GDM maternal mice reduced colonization of bacteria, such as L. reuteri and L. johnsonii, in the neonatal gut, which impeded development of RORγt+ regulatory T (Treg) cell-mediated immune tolerance. Treatment of GDM neonates with 3'-SL, Lactobacillus reuteri (L. reuteri) and L. johnsonii promoted the proliferation of enteric Treg cells and expression of transcription factor RORγt, which may have contributed to compromising ovalbumin (OVA)-induced allergic responses. In vitro experiments showed that 3'-SL, metabolites of L. johnsonii, and lysates of L. reuteri stimulated differentiation of mouse RORγt+ Treg cells through multiple regulatory effects on Toll-like receptor, MAPK, p53, and NOD-like receptor signaling pathways. This study provides new ideas for the development of gut microbiota and immune tolerance in GDM newborns.


Asunto(s)
Diabetes Gestacional , Microbioma Gastrointestinal , Preescolar , Recién Nacido , Lactante , Femenino , Embarazo , Humanos , Animales , Ratones , Linfocitos T Reguladores , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Madres , Leche Humana , Bacterias
4.
Phytomedicine ; 121: 155082, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37722243

RESUMEN

BACKGROUND: Probiotic fermentation is a promising strategy for improving the nutritional and functional properties of traditional Chinese medicines (TCMs). Ganoderma lucidum and Raphani Semen are famous TCMs that have been shown to help alleviate immune system disorders. However, few studies have experimentally investigated the effects of probiotic-fermented G.lucidum and Raphani Semen on the immune system. PURPOSE: We established the in vitro fermentation of G. lucidum and Raphani Semen with a probiotic mixture (Bifidobacterium longum, Lactobacillus acidophilus, and l. fermentum) (GRFB), investigated its ameliorating effect against cyclophosphamide (CTX)-induced immunosuppression, and explored its possible mechanisms. METHODS: First, the different components in GRFB were identified by high-performance liquid chromatography. Second, its immune-stimulatory activities were evaluated in CTX-treated mice. Lastly, its possible in vitro and in vivo mechanisms were studied. RESULTS: Probiotic fermentation of G. lucidum and Raphani Semen altered some of its chemical constituents, potentially helping improve the ability of GRFB to alleviate immunosuppression. As expected, GRFB effectively ameliorated CTX-induced immunosuppression by increasing the number of splenic lymphocytes and regulating the secretion of serum and ileum cytokines. GRFB supplementation also effectively improved intestinal integrity in CTX-treated mice by upregulating tight junction proteins. It also protects against CTX-induced intestinal dysbiosis by increasing the abundance of beneficial bacteria and reducing the abundance of harmful bacteria. GRFB could directly promote intestinal immunity but not systemic immunity in vitro, suggesting a microbiota-dependent regulation of GRFB. Interestingly, cohousing CTX-induced immunosuppressed mice with GRFB-treated mice promoted their symptoms recovery. Enhanced CTX-induced immunosuppression by GRFB in vitro depended on the gut microbiota. Remarkably, a Kyoto Encyclopedia of Genes and Genomes analysis showed that the GRFB-reprogrammed microbiota was significantly enriched in DNA damage repair pathways, which contribute to repairing the intestinal mucosal barrier. CONCLUSION: This is the first study to suggest that compare with unfermented G. lucidum and Raphani Semen, GRFB can more effectively promote intestinal immunity and manipulate the gut microbiota to promote immunostimulatory activity and repair immunosuppression-induced intestinal barrier damage by biotransforming G.lucidum and Raphani Semen components.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Reishi , Animales , Ratones , Fermentación , Probióticos/farmacología , Probióticos/uso terapéutico , Ciclofosfamida/efectos adversos , Inmunidad , Terapia de Inmunosupresión , Semillas
5.
Sci Rep ; 13(1): 10105, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37344615

RESUMEN

The aim of this study was to evaluate the effect of Lactobacillus delbrueckii subsp. lactis (L.del) on vaginal microbiota (VM) dysbiosis and vaginal radiation injury in gynecologic cancer patients. The inhibitory effects of L.del on cervical cancer cells were also studied in vitro. Gynecologic cancer patients receiving radiotherapy were randomized into control and L.del intervention groups. The control group received radiotherapy, while the intervention group received radiotherapy and L.del intervention (1 capsule/day placed into the deep vagina from the first day of radiotherapy until the end of treatment). Vaginal swab samples were collected on the first day pre-treatment and the last day post-treatment. DNA from 54 patients was extracted and assessed by the 16S rRNA sequencing method. Radiotherapy resulted in vaginal microbiome dysbiosis characterized by increased phylogenetic diversity and increased abundance of Brevundimonas, Streptococcus and Prevotella, but a decreased abundance of Lactobacillus. Level 2 vaginal radiation injury was positively associated with the abundance of Brevundimonas and gram-negative non-fermenting bacteria. Administration of L.del attenuated the reduction of Lactobacillus while also inhibiting the abundance of Streptococcus and Prevotella, thereby ameliorating radiotherapy-related vaginal microbiota dysbiosis. CLD inhibited the in vitro proliferation of SiHa cells by altering the expression of BCL2, HPV16-E6, HPV16-E7, IL6, MAP7, BAX, Caspase-3, Caspase-9 and LTF. In conclusion, L. del application can alleviate radiation-induced vaginal dysbiosis and restore Lactobacillus dominance of the vaginal microbiome. Moreover, CLD was found to inhibit cell growth and promote the apoptosis of SiHa cells in vitro. The registration number for this clinical trial is ChiCTR1900021784.


Asunto(s)
Disbiosis , Neoplasias , Femenino , Humanos , Disbiosis/microbiología , ARN Ribosómico 16S/genética , Filogenia , Vagina/microbiología , Lactobacillus/genética , Streptococcus/genética
6.
Front Nutr ; 8: 766606, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34859034

RESUMEN

Breast milk is rich in sialic acids (SA), which are commonly combined with milk oligosaccharides and glycoconjugates. As a functional nutrient component, SA-containing milk components have received increasing attention in recent years. Sialylated human milk oligosaccharides (HMOs) have been demonstrated to promote the growth and metabolism of beneficial gut microbiota in infants, bringing positive outcomes to intestinal health and immune function. They also exhibit antiviral and bacteriostatic activities in the intestinal mucosa of new-borns, thereby inhibiting the adhesion of pathogens to host cells. These properties play a pivotal role in regulating the intestinal microbial ecosystem and preventing the occurrence of neonatal inflammatory diseases. In addition, some recent studies also support the promoting effects of sialylated HMOs on neonatal bone and brain development. In addition to HMOs, sialylated glycoproteins and glycolipids are abundant in milk, and are also critical to neonatal health. This article reviews the current research progress in the regulation of sialylated milk oligosaccharides and glycoconjugates on neonatal gut microbiota and health.

7.
Cell Death Dis ; 12(12): 1094, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34799549

RESUMEN

Vaginal dysbiosis often occurs in patients with cervical cancer. The fucosylation of mucosal epithelial cells is closely related to microbial colonization, and play an important role in protecting the vaginal mucosal epithelial cells. However, no reports on the relationship between vaginal dysbiosis and abnormal mucosal epithelial cell fucosylation, and their roles in the occurrence and development of cervical cancer are unavailable. Here we report that core fucosylation levels were significantly lower in the serum, exfoliated cervical cells and tumor tissue of cervical cancer patients. Core fucosyltransferase gene (Fut8) knockout promoted the proliferation and migration of cervical cancer cells. In patients with cervical cancer, the vaginal dysbiosis, and the abundance of Lactobacillus, especially L. iners, was significantly reduced. Meanwhile, the abundance of L.iners was positively correlated with core fucosylation levels. The L. iners metabolite lactate can activate the Wnt pathway through the lactate-Gpr81 complex, which increases the level of core fucosylation in epidermal cells, inhibiting the proliferation and migration of cervical cancer cells, and have application prospects in regulating the vaginal microecology and preventing cervical cancer.


Asunto(s)
Células Epiteliales/metabolismo , Fucosiltransferasas/metabolismo , Lactobacillus/patogenicidad , Neoplasias del Cuello Uterino/microbiología , Estudios de Casos y Controles , Femenino , Humanos , Persona de Mediana Edad , Microambiente Tumoral
8.
Can J Infect Dis Med Microbiol ; 2021: 3044534, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603565

RESUMEN

Gestational diabetes mellitus (GDM) is defined as "diagnosed as impaired glucose tolerance for the first time during pregnancy," which can lead to adverse pregnancy outcomes and produces divergent effects on mothers and newborns. In recent years, with the continuous expansion of obese people, GDM shows an upward trend. The abundant and diverse members of the human gut microbiota exert critical roles in the maintenance of human health. Studies have shown that GDM may be associated with disordered gut microbiota in both mothers and newborns. Taking into account the potential effects on maternal and consequently neonatal health, in this review, we analyzed the available data and discussed the current knowledge about the potential relationship between GDM and intestinal dysbiosis in mothers and newborns. In addition, we also discussed the influencing factors derived from GDM mothers on the gut microbiome of their newborns, including the vertical transmission of microbiota from mothers, the alteration of milk components of GDM mothers, and using of probiotics. Hoping that new insights into the role of the gut microbiota in GDM could lead to the development of integrated strategies to prevent and treat these metabolic disorders.

9.
Food Nutr Res ; 652021.
Artículo en Inglés | MEDLINE | ID: mdl-34349613

RESUMEN

BACKGROUND: Omega-3 polyunsaturated fatty acids (PUFAs) play beneficial roles in metabolism and health. Little is known about the effects of different doses of omega-3 PUFAs on gut microbiota. OBJECTIVE: In this study, we focus on the effects of different doses of omega-3 PUFAs on gut microbiota and immunity. DESIGN: BALB/c mice was first treated with ceftriaxone sodium for 7 days, and then they received saline or different doses of omega-3 PUFAs (30, 60 and 90 mg omega-3 PUFAs) via daily gavage for 21 days. Alterations of cecum microbiota; the tight junction proteins, zonula occludens 3 (ZO3) and occludin, in the ileal wall; serum lipopolysaccharide (LPS); Interleukin-10 (IL-10), interleukin-1ß (IL-1ß), and Tumour Necrosis Factor α (TNF-α) ; mucus SIgA levels were measured. RESULTS: Compared with the ceftriaxone sodium administration group, significant increases in bacterial richness and diversity were observed in the 60- and 90-mg omega-3 PUFA groups, while only a slight increase was observed in the 30-mg omega-3 PUFA group. A higher percentage of several genera, including Lactobacillus, Helicobacter, and Ruminococcus, and a lower percentage of Bacteroides, Clostridium, and Prevotella were observed in the 60- and 90-mg omega-3 PUFA groups when compared with those in the 30-mg group. The expression of ZO3 and occludin proteins increased in 60- and 90-mg omega-3 PUFA groups compared with the natural recovery group. The mucus SIgA and serum IL-10 levels were increased, and serum levels of LPS, IL-1ß, and TNF-α were decreased in the 60- and 90-mg omega-3 PUFA groups when compared with those in the ceftriaxone sodium-treated group. CONCLUSION: Different doses of omega-3 PUFAs have different therapeutic effects on the intestinal microbiota. The 60- and 90-mg omega-3 PUFA supplementation had better recovery effects on the gut microbiota and immunity than those of the 30 mg omega-3 PUFAs supplementation.

10.
Biomed Pharmacother ; 141: 111909, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34328088

RESUMEN

Ganoderma lucidum is a legendary traditional Chinese medicine with various bioactivities. This study was conducted (a) to explore the in vitro fermentation of the water extracts of G. lucidum fruiting body with Lactobacillus acidophilus and Bifidobacterium breve and (b) to investigate the effect of fermentation broth (GLFB) on dexamethasone (DEX)-induced immunosuppressed mice. Our results demonstrated that probiotic fermentation of G. lucidum fruiting body extracts underwent structural changing of major ganoderic acid components, such as ganoderic acid A (GA) into GC2, and this fermentation process involves changing of several metabolic pathways in the probiotic strains. GLFB could significantly improve the immunity, intestinal integrity, and gut microbiota dysbiosis in DEX-treated mice, and the immunostimulatory activity of GLFB was found closely related to its direct regulation on the expansion of CD4+ T cells in Peyer's patches of mice. These data implied that probiotic fermentation of G. lucidum fruiting body extracts promoted its immunostimulatory activity via biotransformation of components such as GA. This research provides a theoretical support for the development and application of G. lucidum fermentation by probiotics.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Dexametasona/farmacología , Cuerpos Fructíferos de los Hongos/química , Inmunosupresores/farmacología , Probióticos/metabolismo , Reishi/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Fermentación , Microbioma Gastrointestinal/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Intestinos/efectos de los fármacos , Lanosterol/análogos & derivados , Lanosterol/farmacología , Recuento de Linfocitos , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos BALB C , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/efectos de los fármacos , Reishi/química
11.
Front Cell Infect Microbiol ; 11: 647048, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33842393

RESUMEN

This study aimed at determining the beneficial effect of Clostridium butyricum (CB) RH2 on ceftriaxone-induced dysbacteriosis. To this purpose, BALB/c mice were exposed to ceftriaxone (400 mg/ml) or not (control) for 7 days, and administered a daily oral gavage of low-, and high-dose CB RH2 (108 and 1010 CFU/ml, respectively) for 2 weeks. CB RH2 altered the diversity of gut microbiota, changed the composition of gut microbiota in phylum and genus level, decreased the F/B ratio, and decreased the pro-inflammatory bacteria (Deferribacteres, Oscillibacter, Desulfovibrio, Mucispirillum and Parabacteroides) in ceftriaxone-treated mice. Additionally, CB RH2 improved colonic architecture and intestinal integrity by improving the mucous layer and the tight junction barrier. Furthermore, CB RH2 also mitigated intestinal inflammation through decreasing proinflammatory factors (TNF-α and COX-2) and increasing anti-inflammatory factors (IL-10). CB RH2 had direct effects on the expansion of CD4+ T cells in Peyer's patches (PPs) in vitro, which in turn affected their immune response upon challenge with ceftriaxone. All these data suggested that CB RH2 possessed the ability to modulate the intestinal mucosal and systemic immune system in limiting intestinal alterations to relieve ceftriaxone-induced dysbacteriosis.


Asunto(s)
Clostridium butyricum , Disbiosis , Animales , Ceftriaxona , Mucosa Intestinal , Intestinos , Ratones , Ratones Endogámicos BALB C
12.
Can J Infect Dis Med Microbiol ; 2020: 8837156, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33312314

RESUMEN

OBJECTIVE: Diarrhea in infants is a serious gastrointestinal dysfunction characterized by vomiting and watery bowel movements. Without proper treatment, infants will develop a dangerous electrolyte imbalance. Diarrhea is accompanied by intestinal dysbiosis. This study compared the gut microbiota between healthy infants and diarrheic infants. It also investigated the effects of age and pathogen type on the gut microbiota of infants with diarrhea, providing data for the proper treatment for diarrhea in infants. MATERIALS AND METHODS: DNA was collected from the fecal samples of 42 Chinese infants with diarrhea and 37 healthy infants. The healthy infants and infants with diarrhea were divided into four age groups: 0-120, 120-180, 180-270, and 270-365 days. Using PCR and 16S rRNA high-throughput sequencing, the diarrhea-causing pathogens in these infants were identified and then categorized into four groups: Salmonella infection, Staphylococcus aureus infection, combined Salmonella and Staphylococcus aureus infection, and others (neither Salmonella nor Staphylococcus aureus). RESULTS: The species diversity of gut microbiota in diarrheic infants was significantly reduced compared with that in healthy infants. Infants with diarrhea had a lower abundance of Lactobacillus spp. and Bacillus spp. (P < 0.001) and a significant richness of Klebsiella spp. and Enterobacter spp. (P < 0.001). Similar gut microbiota patterns were found in diarrheic infants in all four age groups. However, different pathogenic infections have significant effects on the gut microbiota of diarrheic infants. For instance, the relative abundance of Klebsiella spp. and Streptococcus spp. was significantly increased (P < 0.001) in infants infected with Staphylococcus aureus; meanwhile, the richness of bacteria such as Enterobacter spp. was significantly increased in the Salmonella infection group (P < 0.001). CONCLUSION: The microbiota in infants with diarrhea has changed significantly, characterized by decreased species diversity and abundance of beneficial bacteria and significant increase in the proportion of conditional pathogens. Meanwhile, the gut microbiota of infants with diarrhea at different ages was similar, but different pathogenic infections affect the gut microbiota characteristics. Therefore, early identification of changes in gut microbiota in infants with diarrhea and the adoption of appropriate pathogen type-specific interventions may effectively alleviate the disease and reduce adverse reactions.

13.
Food Sci Nutr ; 8(8): 4388-4398, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32884719

RESUMEN

Constipation is one of the most common functional gastrointestinal disorders accompanied with intestinal dysbiosis. Laxatives for constipation usually have side effects. Bee honey is a natural food with unique composition, antimicrobial properties, and bifidogenic effect. In order to assess whether honey can ameliorate loperamide-induced constipation in BALB/c mice through the alteration of the gut microbiota, the present study was undertaken. Mice were given Jarrah honey (7.5 g/kg body weight) by gavage once per day for 5 days. Fecal water content, intestinal transit rate together with the colon concentrations of substance P (SP), vasoactive intestinal peptide (VIP), and serotonin (5-hydroxytryptamine; 5-HT) were evaluated. Furthermore, we determined the effect of honey treatment on gut microbiota in mice using stool genomic 16S rRNA sequencing. As a result, honey showed an obvious improvement in fecal water content and alleviated constipation by modulating the microbial composition of the microbiota, and this was highly associated with a proportional decrease in gut Desulfovibrio. In addition, we found that the colon level of neurotransmitters SP and VIP was significantly related to microbial variations. Our results indicate that gut microbiota is involved in the alleviation of loperamide-induced constipation by honey supplementation in mice, and it could be considered as an evaluating parameter in constipation therapy strategies.

14.
Biomed Res Int ; 2020: 9067821, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32509874

RESUMEN

Gut dysbiosis induced by high-fat diet (HFD) may result in low-grade inflammation leading to diverse inflammatory diseases. The beneficial effects of probiotics and prebiotics on obesity have been reported previously. However, their benefits in promoting human health and the underlying mechanisms still need to be further characterized. This study is aimed at understanding how probiotic Bacillus licheniformis Zhengchangsheng® (BL) and prebiotic xylooligosaccharides (XOS) influence the health of a rat model with HF (60 kcal %) diet-induced obesity. Five groups of male Sprague Dawley (SD) rats were fed a normal fat diet (CON) or an HFD with or without BL and XOS supplementation for 3 weeks. Lipid profiles, inflammatory biomarkers, and microbiota composition were analyzed at the end of the experiment. Rats fed an HFD exhibited increased body weight and disordered lipid metabolism. In contrast, combined BL and XOS supplementation inhibited body weight gain and returned lipid metabolism to normal. Furthermore, BL and XOS administration changed the gut microbiota composition and modulated specific bacteria such as Prevotellaceae, Desulfovibrionaceae, and Ruminococcaceae. In addition, supplements of combined BL and XOS obviously reduced the serum LPS level, which was significantly related to microbial variations. Our findings suggest that modulation of the gut microbiota as a result of probiotic BL and prebiotic XOS supplementation has a positive effect on HFD-induced obesity in rats.


Asunto(s)
Bacillus licheniformis , Microbioma Gastrointestinal/efectos de los fármacos , Glucuronatos , Obesidad , Oligosacáridos , Probióticos , Administración Oral , Animales , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Medicamentos Herbarios Chinos , Glucuronatos/administración & dosificación , Glucuronatos/farmacología , Masculino , Obesidad/metabolismo , Obesidad/microbiología , Oligosacáridos/administración & dosificación , Oligosacáridos/farmacología , Prebióticos/administración & dosificación , Probióticos/administración & dosificación , Probióticos/farmacología , Ratas , Ratas Sprague-Dawley , Aumento de Peso/efectos de los fármacos
15.
mSystems ; 4(2)2019.
Artículo en Inglés | MEDLINE | ID: mdl-30944880

RESUMEN

Intestinal bacterial dysbiosis has been increasingly linked to ankylosing spondylitis (AS), which is a prototypic and best studied subtype of spondyloarthritis (SpA). Fungi and bacteria coexist in the human gut and interact with each other. Although they have been shown to contribute actively to health or disease, no studies have investigated whether the fungal microbiota in AS patients is perturbed. In this study, fecal samples from 22 AS patients, with clinical and radiographic assessments, and 16 healthy controls (HCs) were collected to systematically characterize the gut microbiota and mycobiota in AS patients by 16S rRNA gene- and ITS2-based DNA sequencing. Our results showed that the microbiota of AS patients was characterized by increased abundance of Proteobacteria and decreased Bacteroidetes, which was contributed by enrichment of Escherichia-Shigella, Veillonella, Lachnospiraceae NK4A136 group, and reduction of Prevotella strain 9, Megamona, and Fusobacterium. The gut mycobiota of AS patients was characterized by higher levels of Ascomycota, especially the class of Dothideomycetes, and decreased abundance of Basidiomycota, which was mainly contributed by the decease of Agaricales. Compared to HCs, decreased ITS2/16S biodiversity ratios and altered bacterial-fungal interkingdom networks were observed in AS patients. Compared with nonsteroidal anti-inflammatory drugs (NSAIDs), treating AS patients with biological agents induced obvious changes in the gut mycobiota, and this result was highly associated with disease activity indexes, including AS disease activity index (ASDAS) C-reactive protein (asCRP), erythrocyte sedimentation rate (ESR), and Bath AS disease activity index (BASDAI). In addition, altered mycobiota in AS patients was also found associated with the degree of radiographic damage. IMPORTANCE The human gut is colonized by diverse fungi (mycobiota), and fungi have long been suspected in the pathogenesis of SpA. Our study unraveled a disease-specific interkingdom network alteration in AS, suggesting that fungi, or the interkingdom interactions between bacteria and fungi, may play an essential role in AS development. However, our study is limited by sample size, and in-depth mechanism studies and additional large-scale investigations characterizing the gut mycobiome in AS patients are needed to form a foundation for research into the relationship between mycobiota dysbiosis and AS development.

16.
mBio ; 10(2)2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940702

RESUMEN

The maternal milk glycobiome is crucial for shaping the gut microbiota of infants. Although high core fucosylation catalyzed by fucosyltransferase 8 (Fut8) is a general feature of human milk glycoproteins, its role in the formation of a healthy microbiota has not been evaluated. In this study, we found that the core-fucosylated N-glycans in milk of Chinese mothers selectively promoted the colonization of specific gut microbial groups, such as Bifidobacterium spp. and Lactobacillus spp. in their breast-fed infants during lactation. Compared with Fut8+/+ (WT) mouse-fed neonates, the offspring fed by Fut8+/- maternal mice had a distinct gut microbial profile, which was featured by a significant reduction of Lactobacillus spp., Bacteroides spp., and Bifidobacterium spp. and increased abundance of members of the Lachnospiraceae NK4A136 group and Akkermansia spp. Moreover, these offspring mice showed a lower proportion of splenic CD19+ CD69+ B lymphocytes and attenuated humoral immune responses upon ovalbumin (OVA) immunization. In vitro studies demonstrated that the chemically synthesized core-fucosylated oligosaccharides possessed the ability to promote the growth of tested Bifidobacterium and Lactobacillus strains in minimal medium. The resulting L-fucose metabolites, lactate and 1,2-propanediol, could promote the activation of B cells via the B cell receptor (BCR)-mediated signaling pathway.IMPORTANCE This study provides novel evidence for the critical role of maternal milk protein glycosylation in shaping early-life gut microbiota and promoting B cell activation of neonates. The special core-fucosylated oligosaccharides might be promising prebiotics for the personalized nutrition of infants.


Asunto(s)
Linfocitos B/inmunología , Bifidobacterium/metabolismo , Fucosa/metabolismo , Tracto Gastrointestinal/inmunología , Lactobacillus/metabolismo , Leche Humana/química , Polisacáridos/metabolismo , Animales , China , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Lactante , Activación de Linfocitos , Ratones , Polisacáridos/química
17.
Nutrition ; 62: 63-73, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30852460

RESUMEN

OBJECTIVE: High fructose consumption exacerbates purine degradation and intestinal dysbiosis, which are closely related to the development of hyperuricemia. Probiotics are powerful weapons to combat metabolic disturbance and intestinal dysbiosis. Previously we isolated a Lactobacillus strain named DM9218 that could reduce the serum uric acid (UA) level by assimilating purine nucleosides. The present study aimed to evaluate the effects of DM9218 on high-fructose-induced hyperuricemia and to elucidate the underlying mechanisms. METHODS: Mice were fed a normal diet, a high-fructose diet, or high-fructose diet with DM9218. Metabolic parameters, fructose- and UA-related metabolites, and fecal microbiota were investigated. Whole-genome sequencing of strain DM9218 was also conducted. In addition, an inosine hydrolase from DM9218 was heterologously expressed in Escherichia coli, and its inosine-degrading activity was detected. RESULTS: Our results indicated that DM9218 could decrease serum UA level and hepatic xanthine oxidase activity in fructose-fed mice. It could protect against high-fructose-induced liver damage and retard UA accumulation by degrading inosine. The modulation effect of DM9218 on high-fructose-induced intestinal dysbiosis resulted in enhancement of intestinal barrier function and reduction of liver lipopolysaccharide, which was closely correlated with the down-regulation of inflammatory cytokine-stimulated xanthine oxidase expression and activity. CONCLUSIONS: Lactobacillus brevis DM9218 is a probiotic strain with the potential to ameliorate fructose-induced hyperuricemia.


Asunto(s)
Disbiosis/tratamiento farmacológico , Fructosa/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Hiperuricemia/tratamiento farmacológico , Inosina/metabolismo , Levilactobacillus brevis , Animales , Dieta/efectos adversos , Dieta/métodos , Modelos Animales de Enfermedad , Disbiosis/metabolismo , Disbiosis/orina , Hiperuricemia/etiología , Hiperuricemia/orina , Inosina/orina , Intestinos/efectos de los fármacos , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Probióticos
18.
Food Funct ; 9(8): 4317-4327, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-30040095

RESUMEN

Accumulating evidence suggests that diet could shape the host gut microbiome composition. Herein, we investigated the effects of maternal high fat diet (HFD) consumption on the gut microbiota and serum profile of mice offspring, and attempted to explore the beneficial roles of maternal probiotics intervention. Female C57BL/6J mice were fed with normal diet, HFD or HFD with daily probiotics (B. breve DM8310, L. acidophilus DM8302, L. casei DM8121 and S. thermophilus DM8309) by gavage starting 6 weeks prior to breeding and continued throughout gestation and lactation. Pups of HFD dams had higher levels of total cholesterol (TC), low-density lipoprotein (LDL), glucose, insulin and leptin compared to those of chow-fed dams. Maternal probiotics intervention resulted in a decrease in the lipid levels in all the pups, while the glucose, insulin and leptin levels were decreased only in adult female pups compared to those from HFD-fed dams; the decreased levels were similar to those in the pups of chow-fed dams. In line with these plasma changes, maternal HFD persistently altered the composition of the offspring gut microbiota in a sex specific way. Maternal probiotics intervention could ameliorate gut microbiota dysbiosis in the offspring. Such intervention showed better effects particularly for the female pups at adulthood. In conclusion, maternal HFD-induced gut microbiota dysbiosis and metabolic disorder could persist through the adulthood of the offspring. Maternal probiotics intervention can negate the detrimental effects of maternal HFD on the gut microbiota and metabolism in the offspring in a sex specific way.


Asunto(s)
Microbioma Gastrointestinal , Obesidad/metabolismo , Obesidad/microbiología , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/microbiología , Animales , Colesterol/sangre , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Leptina/sangre , Lipoproteínas LDL/sangre , Masculino , Herencia Materna , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Probióticos/administración & dosificación
19.
mSystems ; 3(6)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30637338

RESUMEN

The milk glycobiome has a significant impact on the gut microbiota of infants, which plays a pivotal role in health and development. Fucosylated human milk oligosaccharides (HMOs) and N-glycans on milk proteins are beneficial for the development of healthy gut microbiota, and the fucosylation levels of these glycans can be affected by the maternal fucosyltransferase 2 gene (FUT2). Here, we present results of longitudinal research on paired milk and stool samples from 56 Chinese mothers (CMs) and their breast-fed children. Changes of HMOs and fucosylated N-glycans in milk of CMs at different lactation stages were detected, which allowed characterization of the major differences in milk glycans and consequential effects on the gut microbiome of infants according to maternal FUT2 status. Significant differences in the abundance of total and fucosylated HMOs between secretor and nonsecretor CMs were noted, especially during early lactation. Despite a tendency toward decreasing milk protein concentrations, the fucosylation levels of milk N-glycans increased during late lactation. The changes in the levels of fucosylated HMOs and milk N-glycans were highly correlated with the growth of Bifidobacterium spp. and Lactobacillus spp. in the gut of infants during early and later lactation, respectively. Enriched expression of genes encoding glycoside hydrolases, glycosyl transferases, ATP-binding cassette (ABC) transporters, and permeases in infants fed by secretor CMs contributed to the promotion of these bacteria in infants. Our data highlight the important role of fucosylated milk glycans in shaping the gut microbiome of infants and provide a solid foundation for development of "personalized" nutrition for Chinese infants. IMPORTANCE Human milk glycans provide a broad range of carbon sources for gut microbes in infants. Levels of protein glycosylation in human milk vary during lactation and may also be affected by the stages of gestation and lactation and by the secretor status of the mother. This was the first study to evaluate systematically dynamic changes in human milk oligosaccharides and fucosylated N-glycans in the milk of Chinese mothers with different secretor statuses during 6 months of lactation. Given the unique single nucleotide polymorphism site (rs1047781, A385T) on the fucosyltransferase 2 gene among Chinese populations, our report provides a specific insight into the milk glycobiome of Chinese mothers, which may exert effects on the gut microbiota of infants that differ from findings from other study cohorts.

20.
Front Immunol ; 8: 824, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28747917

RESUMEN

Bacterial consortium transplantation (BCT) is a promising alternative to fecal microbiota transplantation in treating inflammatory bowel disease (IBD). Here, we showed that a defined bacterial consortium derived from healthy mice was able to enhance the intestinal barrier function of mice with dextran sulfate sodium (DSS)-induced colitis. Interestingly, we found that the bacterial consortium significantly promoted the expansion of IL-17A-producing γδT (γδT17) cells in colonic lamina propria, which was closely associated with changing of intestinal microbial composition. The increased IL-17A secretion upon treatment with microbial products derived from the bacterial consortium was accompanied with upregulation of TLR2 expression by γδT cells, and it might be responsible for the upregulation of mucosal barrier function through IL-17R-ACT1-mediated recovery of the disrupted occludin subcellular location. Changing of some specific microbial groups such as Bifidobacterium and Bacillus spp. was closely correlated with the promotion of TLR2+ γδT cells. Our results support that BCT can restore the alliance between commensal microbiota and intestinal γδT cells, which contributes to the improvement of intestinal barrier function. This study provides new insight into the development of bacteria transplantation therapy for the treatment of IBD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA