Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 16(9): 918-26, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26237551

RESUMEN

Mucosal immunity protects a host from intestinal inflammation and infection and is profoundly influenced by symbiotic bacteria. Here we report that in mice symbiotic bacteria directed selective cargo sorting in Paneth cells to promote symbiosis through Nod2, a cytosolic bacterial sensor, and the multifunctional protein kinase LRRK2, both encoded by inflammatory bowel disease (IBD)-associated genes. Commensals recruited Nod2 onto lysozyme-containing dense core vesicles (DCVs), which was required for DCV localization of LRRK2 and a small GTPase, Rab2a. Deficiency of Nod2, LRRK2 or Rab2a or depletion of commensals resulted in lysosomal degradation of lysozyme. Thus, commensal bacteria and host factors orchestrate the lysozyme-sorting process to protect the host from enteric infection, implicating Paneth cell dysfunction in IBD pathogenesis.


Asunto(s)
Enterocolitis/inmunología , Inmunidad Mucosa/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Intestinos/inmunología , Listeriosis/inmunología , Células de Paneth/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Simbiosis/inmunología , Animales , Enterocolitis/genética , Inmunidad Mucosa/genética , Enfermedades Inflamatorias del Intestino/genética , Intestinos/microbiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Listeriosis/genética , Lisosomas , Ratones , Ratones Noqueados , Muramidasa , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/inmunología , Proteínas Serina-Treonina Quinasas/genética , Vesículas Secretoras/inmunología , Simbiosis/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/inmunología
2.
J Transl Med ; 19(1): 510, 2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34922582

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) remains a life-threatening disease in neonates. Numerous studies have shown a correlation between the intestinal microbiota and NEC, but the causal link remains unclear. This study aimed to demonstrate the causal role of gut microbiota in NEC and explore potential mechanisms involved. METHODS: Eighty-one fecal samples from patients with NEC and eighty-one matched controls (matched to the NEC infants by gestational age, birth weight, date of birth, mode of delivery and feeding patterns) were collected. To explore if altered gut microbiota contributes to the pathogenesis of NEC, fecal microbiota transplantation (FMT) was carried out in germ-free (GF) mice prior to a NEC-induction protocol that included exposure to hypoxia and cold stress. Butyric acid was also administered to demonstrate its role in NEC. The fecal microbiota from patients and mice were analyzed by 16S rRNA gene sequencing analysis. Short chain fatty acid (SCFA) levels were measured by gas chromatography-mass spectrometry (GC-MS). The ontogeny of T cells and regulatory T cells (Tregs) in lamina propria mononuclear cells (LPMC) from the ileum of patients and mice were isolated and analyzed by flow cytometry.The transcription of inflammatory cytokines was quantified by qRT-PCR. RESULTS: NEC patients had increased Proteobacteria and decreased Firmicutes and Bacteroidetes compared to fecal control samples, and the level of butyric acid in the NEC group was lower than the control group. FMT in GF mice with samples from NEC patients achieved a higher histological injury scores when compared to mice that received FMT with control samples. Alterations in microbiota and butyrate levels were maintained in mice following FMT. The ratio of Treg/CD4+T (Thelper) cells was reduced in both NEC patients and mice modeling NEC following FMT. CONCLUSIONS: The microbiota was found to have NEC and the microbial butyrate-Treg axis was identified as a potential mechanism for the observed effects.


Asunto(s)
Enterocolitis Necrotizante , Microbiota , Animales , Butiratos , Enterocolitis Necrotizante/microbiología , Humanos , Recién Nacido , Ratones , ARN Ribosómico 16S/genética , Linfocitos T Reguladores
3.
Funct Integr Genomics ; 20(3): 355-365, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31677064

RESUMEN

Gut microbiota can affect multiple brain functions and cause behavioral alterations through the microbiota-gut-brain axis. In our previous study, we found that the absence of gut microbiota can influence the expression of microRNAs and mRNAs in the hippocampal region of the germ-free (GF) mice. Long non-coding RNAs (lncRNAs) are increasingly being recognized as an important functional transcriptional regulator in the brain. In the present study, we aim to identify possible biological pathways and functional networks for lncRNA-associated transcript of the gut microbiota in relation to the brain function. The profiles of lncRNA and mRNA from specific pathogen-free (SPF), colonized GF (CGF), and GF mice were generated using the Agilent Mouse LncRNA Array v2.0. Differentially expressed (DE) lncRNAs and mRNAs were identified, and lncRNA target genes were also predicted. Ingenuity pathway analysis (IPA) was performed to analyze related signaling pathways and biological functions associated with these dysregulated mRNAs and target genes. Validation with quantitative real-time PCR was performed on several key genes. Compared with SPF mice a total of 2230 DE lncRNAs were found in GF mice. Among these, 1355 were upregulated and 875 were downregulated. After comparing the target genes of DE lncRNAs with mRNA datasets, 669 overlapping genes were identified. IPA core analyses revealed that most of these genes were highly associated with cardiac hypertrophy, nuclear factors of activated T cells (NFAT) gonadotropin-releasing hormone (GnRH), calcium, and cAMP-response element-binding protein (CREB) signaling pathways. Additionally, mRNA expression levels of APP, CASP9, IGFBP2, PTGDS, and TGFBR2 genes that are involved in central nervous system functions were significantly changed in the GF mouse hippocampus. Through this study, for the first time, we describe the effect of gut microbiota on the hippocampal lncRNA regulation. This will help in enhancing the overall knowledge about microbiota-gut-brain axis.


Asunto(s)
Microbioma Gastrointestinal , Hipocampo/metabolismo , ARN Largo no Codificante/genética , ARN Mensajero/genética , Animales , Redes Reguladoras de Genes , Masculino , Ratones , Ratones Endogámicos BALB C , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Organismos Libres de Patógenos Específicos , Transcriptoma
4.
Ann Rheum Dis ; 79(5): 646-656, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32205337

RESUMEN

OBJECTIVES: Emerging evidence suggests that the microbiome plays an important role in the pathogenesis of osteoarthritis (OA). We aimed to test the two-hit model of OA pathogenesis and potentiation in which one 'hit' is provided by an adverse gut microbiome that activates innate immunity; the other 'hit' is underlying joint damage. METHODS: Medical history, faecal and blood samples were collected from human healthy controls (OA-METS-, n=4), knee OA without metabolic syndrome (OA+METS-, n=7) and knee OA with metabolic syndrome (OA+METS+, n=9). Each group of human faecal samples, whose microbial composition was identified by 16S rRNA sequencing, was pooled and transplanted into germ-free mice 2 weeks prior to meniscal/ligamentous injury (MLI) (n≥6 per group). Eight weeks after MLI, mice were evaluated for histological OA severity and synovitis, systemic inflammation and gut permeability. RESULTS: Histological OA severity following MLI was minimal in germ-free mice. Compared with the other groups, transplantation with the OA+METS+ microbiome was associated with higher mean systemic concentrations of inflammatory biomarkers (interleukin-1ß, interleukin-6 and macrophage inflammatory protein-1α), higher gut permeability and worse OA severity. A greater abundance of Fusobacterium and Faecalibaterium and lesser abundance of Ruminococcaceae in transplanted mice were consistently correlated with OA severity and systemic biomarkers concentrations. CONCLUSION: The study clearly establishes a direct gut microbiome-OA connection that sets the stage for a new means of exploring OA pathogenesis and potentially new OA therapeutics. Alterations of Fusobacterium, Faecalibaterium and Ruminococcaceae suggest a role of these particular microbes in exacerbating OA.


Asunto(s)
Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal , Síndrome Metabólico/complicaciones , Osteoartritis de la Rodilla/terapia , Animales , Biomarcadores/análisis , Biopsia con Aguja , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Meniscos Tibiales/patología , Meniscos Tibiales/cirugía , Síndrome Metabólico/patología , Ratones Endogámicos C57BL , Análisis Multivariante , Osteoartritis de la Rodilla/patología , Distribución Aleatoria , Valores de Referencia , Análisis de Regresión , Medición de Riesgo
5.
J Immunol ; 198(9): 3729-3736, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28330897

RESUMEN

Paneth cells play an important role in maintaining intestinal homeostasis by secreting a large number of antimicrobial peptides into the intestinal lumen. In this study, we found that Rip2 is required for lysozyme sorting in Paneth cells in a manner that is dependent on Nod2, LRRK2, and Rab2a. Rip2 deficiency in mouse led to lysosomal degradation of lysozyme in Paneth cells and prevented the recruitment of Rab2a onto dense core vesicles (DCVs). Like Nod2 and LRRK2, Rip2 localizes to DCVs in Paneth cells, and its DCV localization depends on Nod2 and LRRK2. Thus, we delineated a genetic pathway, consisting of Nod2-LRRK2-Rip2-Rab2a, which is required for lysozyme sorting. Taken together, our results indicate that the lysozyme-sorting process in Paneth cells is orchestrated by a number of host factors and highlight the importance of Paneth cell function in intestinal homeostasis.


Asunto(s)
Enfermedad de Crohn/metabolismo , Intestinos/patología , Muramidasa/metabolismo , Células de Paneth/metabolismo , Vesículas Secretoras/metabolismo , Animales , Células Cultivadas , Homeostasis , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Células de Paneth/patología , Transporte de Proteínas , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
6.
Gastroenterology ; 153(6): 1621-1633.e6, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28823860

RESUMEN

BACKGROUND & AIMS: Altered gut microbiota is implicated in development of colorectal cancer (CRC). Some intestinal bacteria have been reported to potentiate intestinal carcinogenesis by producing genotoxins, altering the immune response and intestinal microenvironment, and activating oncogenic signaling pathways. We investigated whether stool from patients with CRC could directly induce colorectal carcinogenesis in mice. METHODS: We obtained stored stool samples from participants in a metagenome study performed in Hong Kong. Conventional (male C57BL/6) mice were given azoxymethane to induce colon neoplasia after receiving a course of antibiotics in drinking water. Mice were gavaged twice weekly with stool from 5 patients with CRC or 5 healthy individuals (controls) for 5 weeks. Germ-free C57BL/6 mice were gavaged once with stool from 5 patients with CRC or 5 controls. We collected intestinal tissues from mice and performed histology, immunohistochemistry, expression microarray, quantitative polymerase chain reaction, immunoblot, and flow cytometry analyses. We performed 16S ribosomal RNA gene sequencing analysis of feces from mice. RESULTS: Significantly higher proportions of conventional mice fed with stool from individuals with CRC than control stool developed high-grade dysplasia (P < .05) and macroscopic polyps (P < .01). We observed a higher proportion of proliferating (Ki-67-positive) cells in colons of germ-free mice fed with stool from patients with CRC vs those fed with stool from controls (P < .05). Feces from germ-free and conventional mice fed with stool from patients with CRC vs controls contained different microbial compositions, with lower richness in mice fed with stool from patients with CRC. Intestines collected from conventional and germ-free mice fed with stool from patients with CRC had increased expression of cytokines that modulate inflammation, including C-X-C motif chemokine receptor 1, C-X-C motif chemokine receptor 2, interleukin 17A (IL17A), IL22, and IL23A. Intestines from conventional and germ-free mice fed with stool from patients with CRC contained higher proportions of T-helper 1 (Th1) cells (2.25% vs 0.44%) and Th17 cells (2.08% vs 0.31%) (P < .05 for each) than mice fed with stool from controls. Real-time polymerase chain reaction arrays revealed up-regulation of genes involved in cell proliferation, stemness, apoptosis, angiogenesis, invasiveness, and metastasis in mice fed with stool from patients with CRC. CONCLUSIONS: We fed stool samples from patients with CRC and heathy individuals to germ-free mice and conventional mice with azoxymethane. We found stool from patients with CRC to increase the numbers of polyps, levels of intestinal dysplasia and proliferation, markers of inflammation, and proportions of Th1 and Th17 cells in colon, compared with stool from individuals without CRC. This study provides evidence that the fecal microbiota from patients with CRC can promote tumorigenesis in germ-free mice and mice given a carcinogen.


Asunto(s)
Transformación Celular Neoplásica , Colon/microbiología , Pólipos del Colon/microbiología , Neoplasias Colorrectales/microbiología , Heces/microbiología , Microbioma Gastrointestinal , Animales , Azoximetano , Estudios de Casos y Controles , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Colon/metabolismo , Colon/patología , Pólipos del Colon/inducido químicamente , Pólipos del Colon/metabolismo , Pólipos del Colon/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Vida Libre de Gérmenes , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Antígeno Ki-67/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/microbiología , Masculino , Ratones Endogámicos C57BL , Células TH1/metabolismo , Células TH1/microbiología , Células Th17/metabolismo , Células Th17/microbiología
7.
J Pineal Res ; 64(2)2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28875556

RESUMEN

Melatonin influences intestinal microbiota and the pathogenesis of various diseases. This study was conducted to explore whether melatonin alleviates weanling stress through intestinal microbiota in a weanling mouse model. Melatonin supplementation in weanling mice (provided in the drinking water at a dosage of 0.2 mg/mL for 2 weeks) significantly improved body weight gain (1.4 ± 0.03 g/day in melatonin group vs 1.2 ± 0.06 g/day in control group) and intestinal morphology (ie, villus length, crypt depth, and villus to crypt ratio), but had little effect on the proliferation or apoptosis of intestinal cells, the numbers of Paneth cells and goblet cells, as well as the expression of makers related to enterocytes (sucrase) and endocrine cells (chromogranin A and peptide YY) in the ileum. Melatonin supplementation had little effect on serum levels of amino acids or stress-related parameters (eg, SOD, TNF-α, and angiotensin I). 16S rRNA sequencing suggested that melatonin supplementation increased the richness indices of intestinal microbiota (observed species, Chao 1, and ACE) and shaped the composition of intestinal microbiota (eg, increase in the abundance of Lactobacillus [19 ± 3% in melatonin group vs 6 ± 2% in control group]), which was demonstrated using an ex vivo proliferation assay and colonic loop proliferation assay. Melatonin supplementation also significantly influenced the metabolism of intestinal microbiota, such as amino acid metabolism and drug metabolism. More importantly, in antibiotic-treated weanling mice and germ-free weanling mice, melatonin failed to affect body weight gain or intestinal morphology. Melatonin significantly reduced (by about 60%) the bacterial load in enterotoxigenic Escherichia coli (ETEC)-infected weanling mice, but had little effect on ETEC load in antibiotic-pretreated animals. In conclusion, melatonin affects body weight gain, intestinal morphology, and intestinal ETEC infection through intestinal microbiota in weanling mice. The findings highlight the importance of intestinal microbiota in mediating the various physiological functions of melatonin in the host.


Asunto(s)
Antioxidantes/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Melatonina/farmacología , Estrés Fisiológico/efectos de los fármacos , Animales , Mucosa Intestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR
8.
Curr Microbiol ; 69(3): 365-73, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24807625

RESUMEN

The Kunming (KM) mouse is a closed colony mouse strain widely used in Chinese pharmacology, toxicology, and microbiology research laboratories. However, few studies have examined human flora-associated (HFA) microbial communities in KM mice. In this study, HFA models were built from germ-free KM and C57BL/6J mouse strains, and gut microbial diversity was analyzed by denaturing gradient gel electrophoresis (DGGE) and DNA sequencing. We found that the two strains of HFA mice were significantly different based on the UPGMA dendrogram and the Richness index, but dice similarity coefficients of mouse replicates were not significantly different between HFA-KM and HFA-C57BL/6J. Most of the dominant phyla of human gut microflora could be transferred into the guts of the two mouse strains. However, the predominant genus that formed in HFA-KM was Clostridium sp. and that in HFA-C57BL/6J was Blautia sp. These results imply that genotypes difference between the two mice strains is a critical factor in shaping the intestinal microflora. However, genetic differences of individuals within KM mouse populations failed to lead to individual difference in microflora. Successful generation of HFA-KM mice will facilitate studies examining how diet affects gut microbial structure, and will enable comparative studies for uncovering genetic factors that shape gut microbial communities.


Asunto(s)
Biota , Tracto Gastrointestinal/microbiología , Microbiota , Animales , ADN Bacteriano/química , ADN Bacteriano/genética , Electroforesis en Gel de Gradiente Desnaturalizante , Humanos , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
9.
Vascular ; 22(5): 328-35, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23929425

RESUMEN

PURPOSE: To determine the effects of hypercholesterolemia in pregnant mice on the susceptibility to atherosclerosis in adult life through a new animal modeling approach. METHODS: Male offspring from apoE-/- mice fed with regular (R) or high (H) cholesterol chow during pregnancy were randomly subjected to regular (Groups R-R and H-R, n = 10) or high cholesterol diet (Groups R-H and H-H, n = 10) for 14 weeks. Plasma lipid profiles were determined in all rats. The abdominal aorta was examined for the severity of atherosclerotic lesions in offspring. RESULTS: Lipids significantly increased while high-density lipoprotein-cholesterol/low-density lipoprotein-cholesterol decreased in mothers fed high cholesterol chow after delivery compared with before pregnancy (p < 0.01). Groups R-H and H-R indicated dyslipidemia and significant atherosclerotic lesions. Group H-H demonstrated the highest lipids, lowest high-density lipoprotein-cholesterol/low-density lipoprotein-cholesterol, highest incidence (90%), plaque area to luminal area ratio (0.78 ± 0.02) and intima to media ratio (1.57 ± 0.05). CONCLUSION: Hypercholesterolemia in pregnant mice may increase susceptibility to atherosclerosis in their adult offspring.


Asunto(s)
Aorta Abdominal/patología , Aterosclerosis/etiología , Aterosclerosis/patología , Susceptibilidad a Enfermedades , Hipercolesterolemia/complicaciones , Hipercolesterolemia/patología , Lípidos/sangre , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas para Inmunoenzimas , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones , Ratones Noqueados , Embarazo , Distribución Aleatoria , Factores de Riesgo
10.
Curr Microbiol ; 67(3): 313-21, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23604540

RESUMEN

The establishment of human flora-associated animal models allows the in vivo manipulation of host, microbial, and environmental parameters to influence the gut microbial community. However, it is difficult to simulate infant gut microbiota in germ-free animals because of the variation and dynamic state of infant microbial communities. In this study, the effects of age and strain on intestinal microbiota were observed in an infant human flora-associated (IHFA) mouse model. To establish an IHFA model, postnatal day (PND) 1 germ-free mice (Kunming, n = 10; BALB/c, n = 10) were infected with feces from a breast-fed infant. Microbiota in the feces of BALB/c mice (at PND 7, 14, and 21), and Kunming mice (at PND 14) were analyzed by PCR-denaturing gradient gel electrophoresis. Bifidobacteria and lactobacilli levels in the feces of BALB/c and Kunming mice (PND 7/14/21) were detected by quantitative real-time PCR. The Dice similarity coefficient (Cs) for the fecal microbiota of IHFA mice in comparison with the HD donor sample was higher for BALB/c mice than for Kunming mice (P < 0.05). In addition, the DCs at PND 7 were lower than those at PND 14 and PND 21 in both mouse strains (P < 0.05). The Bifidobacteria and Lactobacillus species colonizing the BALB/c mice were similar to those in the Kunming mice (at PND 7/14/21). The bifidobacteria counts increased with age in both mouse strains, whereas the lactobacilli counts decreased with age in both strains. These results suggest that both age and strain influence microbiota patterns in the IHFA mouse model.


Asunto(s)
Microbiota , Animales , Electroforesis en Gel de Gradiente Desnaturalizante , Vida Libre de Gérmenes , Humanos , Lactante , Metagenoma , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Curr Microbiol ; 67(2): 170-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23483307

RESUMEN

The objective of this study was to analyze human fecal Lactobacillus community and its relationship with rheumatoid arthritis. Samples taken from rheumatoid arthritis (RA) patients and healthy individuals were analyzed by quantitative real-time PCR. Bacterial DNA was extracted from feces, and amplicons of the Lactobacillus-specific regions of 16S rRNA were analyzed by denaturing gradient gel electrophoresis. The richness, Shannon-Wiener index, and evenness of gut microbiota of both groups were analyzed to compare fecal Lactobacillus community structures. Results of this study demonstrated that fecal microbiota of RA patients contained significantly more Lactobacillus (10.62 ± 1.72 copies/g) than the control group (8.93 ± 1.60 copies/g). Significant increases were observed in RA patients in terms of the richness, Shannon-Wiener, and evenness measures, indicating more bacterial species, and increased bacterial diversity and abundance. These results suggest a potential relationship between Lactobacillus communities and the development and progression of rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/microbiología , Heces/microbiología , Lactobacillus/aislamiento & purificación , Adulto , Biodiversidad , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Lactobacillus/clasificación , Lactobacillus/genética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa
12.
Wei Sheng Yan Jiu ; 42(1): 103-6, 2013 Jan.
Artículo en Zh | MEDLINE | ID: mdl-23596718

RESUMEN

OBJECTIVE: To study the role of the intestinal floras in the effect of soy isflavones (SI) on lipid metabolism by germ free (GF) mice. METHODS: GF mice were fed by high fat diet and different doses of SI. After 4 weeks, the indexes of blood lipid and oxidation and weigh livers, intestines and hearts were tested. RESULTS: The weight of livers and intestines were increased, while the weight of hearts was decreased by feeding high fat diet to GF mice. But, SI can reduced the increase of intestine and decrease of heart. SI can also raise the level of HDL-c, while it can't improve antioxidant level. CONCLUSION: SI can protect hearts and intestines from GF mice which were fed by fat lipid diets: SI also increases the level of HDL-c which is benefit of prevention of atherosclerosis. So the beneficial effects of SI not completely dependent on the equol which is produced by intestinal flora.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Glycine max/química , Isoflavonas/uso terapéutico , Metabolismo de los Lípidos/efectos de los fármacos , Animales , Sistema Digestivo/microbiología , Vida Libre de Gérmenes , Isoflavonas/farmacología , Masculino , Ratones
13.
Front Microbiol ; 13: 1086058, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36605506

RESUMEN

To contribute to the conservation of endangered animals, the utilization of model systems is critical to analyze the function of their gut microbiota. In this study, the results of a fecal microbial transplantation (FMT) experiment with germ-free (GF) mice receiving giant panda or horse fecal microbiota showed a clear clustering by donor microbial communities in GF mice, which was consistent with the results of blood metabolites from these mice. At the genus level, FMT re-established approximately 9% of the giant panda donor microbiota in GF mice compared to about 32% for the horse donor microbiota. In line with this, the difference between the panda donor microbiota and panda-mice microbiota on whole-community level was significantly larger than that between the horse donor microbiota and the horse-mice microbiota. These results were consistent with source tracking analysis that found a significantly higher retention rate of the horse donor microbiota (30.9%) than the giant panda donor microbiota (4.0%) in GF mice where the microbiota remained stable after FMT. Further analyzes indicated that the possible reason for the low retention rate of the panda donor microbiota in GF mice was a low relative abundance of Clostridiaceae in the panda donor microbiota. Our results indicate that the donor microbiota has a large effect on GF mice microbiota after FMT.

14.
Curr Microbiol ; 62(4): 1107-12, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21132498

RESUMEN

Human flora-associated (HFA) mice are frequently applied in studying the ecology and metabolism of human gut microbiota. However, the development and stability of the genus Bacteriodes, a prominent bacteria group of human gut microbiota, in HFA mice have not yet fully been examined. In this study, PCR-denaturing gradient gel electrophoresis (DGGE) analysis was employed to monitor the Bacteriodes community in the fecal microbiota of six HFA Kunming mice during a period of 3 weeks. Based on the DGGE banding patterns, the majority of prominent bands in the HFA mice DGGE profile were also typical bands in the human DGGE profile, despite the absence of three bands (corresponding to two different B. thetaiotaomicron strains and one B. intestinalis strain) from the human DGGE profile. The Dice coefficient of similarity for the fecal microbiota of HFA mice in comparison to the human donor sample ranged between 74 ± 6% and 81 ± 7%. The phylogeny of bands in the DGGE profile showed that the dominant Bacteriodes species in the fecal microbiota of HFA mice were B. thetaiotaomicron, representing 66.7% of all bands. Our results indicate that the genus Bacteriodes in the fecal microbiota of HFA mice was selected from the human donor and could remain relatively stable over time.


Asunto(s)
Bacteroides/aislamiento & purificación , Tracto Gastrointestinal/microbiología , Metagenoma , Animales , Bacteroides/clasificación , Bacteroides/genética , ADN Bacteriano/genética , ADN Ribosómico/genética , Electroforesis en Gel de Poliacrilamida , Vida Libre de Gérmenes , Humanos , Ratones , Modelos Animales , Datos de Secuencia Molecular , Filogenia
15.
Nutrition ; 90: 111274, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34004414

RESUMEN

OBJECTIVE: This study explores the effects of fecal microbiota from children with vitamin A (VA) deficiency on colonic mucosal barrier function. METHODS: The composition of gut microbes was identified in children with different VA levels, then feces from children with normal VA or VA deficiency was collected separately and transplanted into germ-free (GF) mice, respectively. Three weeks after transplantation, the colon morphology, colonic tight junction proteins, gut microbes, and metabolites were evaluated. RESULTS: In children, Bifidobacterium and Bacteroides were positively correlated with VA levels. Colonization of VA deficiency fecal microbiota markedly impaired colonic development in GF mice, down-regulated colonic tight junction-related proteins occludin and claudin-1, and reduced immunoglobulin A secretion. Furthermore, fecal microbiota transplantation with different VA levels altered composition of gut microbes and bile acid metabolism pathways in GF mice. CONCLUSION: These data suggest that fecal microbiota from children with VA deficiency attenuates colonic barrier function in GF mice, which may be achieved by changing the bile acid metabolic pathways.


Asunto(s)
Microbioma Gastrointestinal , Deficiencia de Vitamina A , Animales , Ácidos y Sales Biliares , Colon , Heces , Ratones
17.
Front Nutr ; 8: 633738, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708788

RESUMEN

Human flora-associated (HFA) mouse models allow us to design interventions for human disease research to test specific hypotheses and explore the complex commensal microbiome while avoiding the ethical limitations of using humans as models to directly study intestinal flora diseases. However, few studies have investigated the effect of a humanized diet profile (coarse-feed diet; CFD) on colonization efficiency and gut microbial diversity in HFA mice. We tested the colonization efficiency and gut microbial diversity in germ-free Kunming (KM) mice fed a CFD or a purified feed diet (PFD) at 1, 2, and 4 weeks. Although the colonization efficiencies differed significantly (67.50-70.00% vs. 72.69-85.96%) in the HFA mice, the colonization efficiency of the PFD-fed HFA mice (85.96%) was significantly higher than that of the CFD-fed mice (69.61%) at 2 weeks. At 4 weeks, the colonization efficiency of the PFD-fed mice (72.69%) was comparable to that of the CFD-fed mice (70.00%). Additionally, the gut microbial diversity of the CFD-fed HFA mice was similar to that of a human fecal donor. Regarding the Kyoto Encyclopedia of Genes and Genomes colonic microbiota metabolic pathways, the CFD-fed HFA mice showed more similarities to the human donor than to the PFD-fed mice in amino sugar and nucleotide sugar metabolism, biosynthesis of amino acids, carbon metabolism, purine metabolism, and phosphotransferase systems. In conclusion, the humanized diet profiles of the CFD and PFD could help establish human microbiotas in mice. Constructing HFA mouse models fed a CFD for 4 weeks may be useful in researching human-derived intestinal diseases.

18.
J Genet Genomics ; 48(9): 815-824, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34400364

RESUMEN

Carrageenans (CGNs) are widely used in foods and pharmaceuticals although their safety remains controversial. To investigate the effects of CGNs and CGN-degrading bacteria in the human colon, we screened for CGN degradation by human fecal microbiota, and for inflammatory response to CGNs and/or CGN-degrading bacteria in germ free mice. Thin-layer chromatography indicated that high molecular weight (MW) CGNs (≥100 kDa) remained undegraded in the presence of human fecal microbiota, whereas low MW CGNs, i.e., κ-carrageenan oligosaccharides (KCO, ~4.5 kDa) were degraded when exposed to seven of eight human fecal samples, although sulfate groups were not removed during degradation. Bacteroides xylanisolvens and Escherichia coli isolates from fecal samples apparently degraded KCO synergistically, with B. xylanisolvens serving as the primary degrader. Combined treatment of KCO with KCO-degrading bacteria led to greater pro-inflammatory effects in the colon and rectum of germ-free mice than either KCO or bacteria alone. Similarly, p-p38-, CD3-, and CD79a-positive immune cells were more abundant in combined treatment group mice than in either single treatment group. Our study shows that KCO-degrading bacteria and the low MW products of KCO can promote proinflammatory effects in mice, and represent two key markers for evaluating CGN safety in foods or medicines.


Asunto(s)
Carragenina
19.
Transl Psychiatry ; 11(1): 568, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34744165

RESUMEN

Major depressive disorder (MDD) is a serious mental illness. Increasing evidence from both animal and human studies suggested that the gut microbiota might be involved in the onset of depression via the gut-brain axis. However, the mechanism in depression remains unclear. To explore the protein changes of the gut-brain axis modulated by gut microbiota, germ-free mice were transplanted with gut microbiota from MDD patients to induce depression-like behaviors. Behavioral tests were performed following fecal microbiota transplantation. A quantitative proteomics approach was used to examine changes in protein expression in the prefrontal cortex (PFC), liver, cecum, and serum. Then differential protein analysis and weighted gene coexpression network analysis were used to identify microbiota-related protein modules. Our results suggested that gut microbiota induced the alteration of protein expression levels in multiple tissues of the gut-brain axis in mice with depression-like phenotype, and these changes of the PFC and liver were model specific compared to chronic stress models. Gene ontology enrichment analysis revealed that the protein changes of the gut-brain axis were involved in a variety of biological functions, including metabolic process and inflammatory response, in which energy metabolism is the core change of the protein network. Our data provide clues for future studies in the gut-brain axis on protein level and deepen the understanding of how gut microbiota cause depression-like behaviors.


Asunto(s)
Trastorno Depresivo Mayor , Microbioma Gastrointestinal , Animales , Conducta Animal , Eje Cerebro-Intestino , Depresión , Disbiosis , Humanos , Ratones , Proteómica
20.
Curr Microbiol ; 60(1): 6-11, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19768504

RESUMEN

In this study, we investigated the impact of neonatal amoxicillin treatment on the development of the murine intestinal Lactobacillus community. Suckling BALB/c mice received a daily intragastric gavage of amoxicillin or saline from postnatal day 7 (PND 7) to PND 20. Just after the treatment (PND 21) and 5 weeks later (PND 56), the colon digesta samples were analyzed by Lactobacillus-specific quantitative real-time PCR analysis and PCR-denaturing gradient gel electrophoresis (DGGE) technique. Real-time PCR results showed that the levels of lactobacilli in the treatment group were similar to those in the control at PND 56. However, in DGGE analysis the number of DGGE bands and Shannon index were decreased significantly in comparison with control (P < 0.05). The dominant Lactobacillus strain in the murine colon changed from L. johnsonii to L. murinus. These results demonstrated that neonatal amoxicillin treatment led to a significant impact on the biodiversity of the murine intestinal Lactobacillus community within a long-time period.


Asunto(s)
Amoxicilina/farmacología , Antibacterianos/farmacología , Intestinos/microbiología , Lactobacillus/efectos de los fármacos , Animales , Animales Recién Nacidos , Biodiversidad , Intestinos/efectos de los fármacos , Lactobacillus/fisiología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA