Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.680
Filtrar
1.
Adv Appl Microbiol ; 127: 223-252, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38763528

RESUMEN

The intestine tract is a vital site for the body to acquire nutrients, serving as the largest immune organ. Intestinal health is crucial for maintaining a normal physiological state. Abundant microorganisms reside in the intestine, colonized in a symbiotic manner. These microorganisms can generate various metabolites that influence host physiological activities. Microbial metabolites serve as signaling molecules or metabolic substrates in the intestine, and some intestinal microorganisms act as probiotics and promote intestinal health. Researches on host, probiotics, microbial metabolites and their interactions are ongoing. This study reviews the effects of gut bacteria and their metabolites on intestinal health to provide useful references for animal husbandry.


Asunto(s)
Bacterias , Microbioma Gastrointestinal , Probióticos , Animales , Probióticos/metabolismo , Bacterias/metabolismo , Bacterias/genética , Intestinos/microbiología
2.
Nutrients ; 16(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38732540

RESUMEN

Zinc deficiency affects the physical and intellectual development of school-age children, while studies on the effects on intestinal microbes and metabolites in school-age children have not been reported. School-age children were enrolled to conduct anthropometric measurements and serum zinc and serum inflammatory factors detection, and children were divided into a zinc deficiency group (ZD) and control group (CK) based on the results of serum zinc. Stool samples were collected to conduct metagenome, metabolome, and diversity analysis, and species composition analysis, functional annotation, and correlation analysis were conducted to further explore the function and composition of the gut flora and metabolites of children with zinc deficiency. Beta-diversity analysis revealed a significantly different gut microbial community composition between ZD and CK groups. For instance, the relative abundances of Phocaeicola vulgatus, Alistipes putredinis, Bacteroides uniformis, Phocaeicola sp000434735, and Coprococcus eutactus were more enriched in the ZD group, while probiotic bacteria Bifidobacterium kashiwanohense showed the reverse trend. The functional profile of intestinal flora was also under the influence of zinc deficiency, as reflected by higher levels of various glycoside hydrolases in the ZD group. In addition, saccharin, the pro-inflammatory metabolites, and taurocholic acid, the potential factor inducing intestinal leakage, were higher in the ZD group. In conclusion, zinc deficiency may disturb the gut microbiome community and metabolic function profile of school-age children, potentially affecting human health.


Asunto(s)
Heces , Microbioma Gastrointestinal , Zinc , Humanos , Microbioma Gastrointestinal/fisiología , Zinc/deficiencia , Zinc/sangre , Niño , Masculino , Femenino , Heces/microbiología , Bacterias/clasificación , Bacterias/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Metaboloma , Intestinos/microbiología
3.
Gut Microbes ; 16(1): 2347722, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38706205

RESUMEN

The intestine is prone to radiation damage in patients undergoing radiotherapy for pelvic tumors. However, there are currently no effective drugs available for the prevention or treatment of radiation-induced enteropathy (RIE). In this study, we aimed at investigating the impact of indole-3-carboxaldehyde (I3A) derived from the intestinal microbiota on RIE. Intestinal organoids were isolated and cultivated for screening radioprotective tryptophan metabolites. A RIE model was established using 13 Gy whole-abdominal irradiation in male C57BL/6J mice. After oral administration of I3A, its radioprotective ability was assessed through the observation of survival rates, clinical scores, and pathological analysis. Intestinal stem cell survival and changes in the intestinal barrier were observed through immunofluorescence and immunohistochemistry. Subsequently, the radioprotective mechanisms of I3A was investigated through 16S rRNA and transcriptome sequencing, respectively. Finally, human colon cancer cells and organoids were cultured to assess the influence of I3A on tumor radiotherapy. I3A exhibited the most potent radioprotective effect on intestinal organoids. Oral administration of I3A treatment significantly increased the survival rate in irradiated mice, improved clinical and histological scores, mitigated mucosal damage, enhanced the proliferation and differentiation of Lgr5+ intestinal stem cells, and maintained intestinal barrier integrity. Furthermore, I3A enhanced the abundance of probiotics, and activated the AhR/IL-10/Wnt signaling pathway to promote intestinal epithelial proliferation. As a crucial tryptophan metabolite, I3A promotes intestinal epithelial cell proliferation through the AhR/IL-10/Wnt signaling pathway and upregulates the abundance of probiotics to treat RIE. Microbiota-derived I3A demonstrates potential clinical application value for the treatment of RIE.


Asunto(s)
Microbioma Gastrointestinal , Indoles , Ratones Endogámicos C57BL , Probióticos , Receptores de Hidrocarburo de Aril , Vía de Señalización Wnt , Animales , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Humanos , Probióticos/administración & dosificación , Probióticos/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Indoles/metabolismo , Indoles/farmacología , Protectores contra Radiación/farmacología , Organoides/metabolismo , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/prevención & control , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/efectos de la radiación , Intestinos/microbiología , Intestinos/efectos de la radiación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética
4.
BMC Microbiol ; 24(1): 156, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724913

RESUMEN

BACKGROUND: To establish a method to induce Campylobacter jejuni colonization in the intestines of C57BL/6 mice through antibiotic-induced microbiome depletion. RESULTS: Fifty-four female C57BL/6 mice were divided into the normal, control, and experimental groups. The experimental group was administered intragastric cefoperazone sodium and sulbactam sodium (50 mg/mL) for 2 days; then, the experimental and control mice were intragastrically administered 200 µL C. jejuni, which was repeated once more after 2 days. Animal feces were collected, and the HipO gene of C. jejuni was detected using TaqMan qPCR from day 1 to day 14 after modeling completion. Immunofluorescence was used to detect intestinal C. jejuni colonization on day 14, and pathological changes were observed using hematoxylin and eosin staining. Additionally, 16S rDNA analyses of the intestinal contents were conducted on day 14. In the experimental group, C. jejuni was detected in the feces from days 1 to 14 on TaqMan qPCR, and immunofluorescence-labeled C. jejuni were visibly discernable in the intestinal lumen. The intestinal mucosa was generally intact and showed no significant inflammatory-cell infiltration. Diversity analysis of the colonic microbiota showed significant inter-group differences. In the experimental group, the composition of the colonic microbiota differed from that in the other 2 groups at the phylum level, and was characterized by a higher proportion of Bacteroidetes and a lower proportion of Firmicutes. CONCLUSIONS: Microbiome depletion induced by cefoperazone sodium and sulbactam sodium could promote long-term colonization of C. jejuni in the intestines of mice.


Asunto(s)
Antibacterianos , Infecciones por Campylobacter , Campylobacter jejuni , Cefoperazona , Heces , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , ARN Ribosómico 16S , Sulbactam , Animales , Campylobacter jejuni/efectos de los fármacos , Campylobacter jejuni/crecimiento & desarrollo , Femenino , Antibacterianos/farmacología , Cefoperazona/farmacología , Heces/microbiología , Infecciones por Campylobacter/microbiología , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Sulbactam/farmacología , ARN Ribosómico 16S/genética , Intestinos/microbiología , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Mucosa Intestinal/microbiología , Mucosa Intestinal/efectos de los fármacos , ADN Bacteriano/genética , ADN Ribosómico/genética
5.
World J Microbiol Biotechnol ; 40(6): 194, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38713319

RESUMEN

The development and utilization of probiotics have many environmental benefits when they are used to replace antibiotics in animal production. In this study, intestinal lactic acid bacteria were isolated from the intestines of Cherry Valley ducks. Probiotic lactic acid bacterial strains were screened for antibacterial activity and tolerance to produce a Lactobacillus spp. mixture. The effects of the compound on the growth performance and intestinal flora of Cherry Valley ducks were studied. Based on the results of the antibacterial activity and tolerance tests, the highly active strains Lactobacillus casei 1.2435, L. salivarius L621, and L. salivarius L4 from the intestines of Cherry Valley ducks were selected. The optimum ratio of L. casei 1.2435, L. salivarius L621, and L. salivarius L4 was 1:1:2, the amount of inoculum used was 1%, and the fermentation time was 14 h. In vivo experiments showed that compared with the control group, the relative abundances of intestinal Lactobacillus and Blautia were significantly increased in the experimental group fed the lactobacilli compound (P < 0.05); the relative abundances of Parabacteroides, [Ruminococcus]_torques_group, and Enterococcus were significantly reduced (P < 0.05), and the growth and development of the dominant intestinal flora were promoted in the Cherry Valley ducks. This study will provide more opportunities for Cherry Valley ducks to choose microecological agents for green and healthy breeding.


Asunto(s)
Patos , Microbioma Gastrointestinal , Intestinos , Lactobacillus , Probióticos , Animales , Probióticos/farmacología , Patos/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Lactobacillus/aislamiento & purificación , Intestinos/microbiología , Fermentación , Alimentación Animal , ARN Ribosómico 16S/genética , Antibacterianos/farmacología
6.
Microbiome ; 12(1): 89, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38745230

RESUMEN

BACKGROUND: Non-toxic approaches to enhance radiotherapy outcomes are beneficial, particularly in ageing populations. Based on preclinical findings showing that high-fibre diets sensitised bladder tumours to irradiation by modifying the gut microbiota, along with clinical evidence of prebiotics enhancing anti-cancer immunity, we hypothesised that dietary fibre and its gut microbiota modification can radiosensitise tumours via secretion of metabolites and/or immunomodulation. We investigated the efficacy of high-fibre diets combined with irradiation in immunoproficient C57BL/6 mice bearing bladder cancer flank allografts. RESULT: Psyllium plus inulin significantly decreased tumour size and delayed tumour growth following irradiation compared to 0.2% cellulose and raised intratumoural CD8+ cells. Post-irradiation, tumour control positively correlated with Lachnospiraceae family abundance. Psyllium plus resistant starch radiosensitised the tumours, positively correlating with Bacteroides genus abundance and increased caecal isoferulic acid levels, associated with a favourable response in terms of tumour control. Psyllium plus inulin mitigated the acute radiation injury caused by 14 Gy. Psyllium plus inulin increased caecal acetate, butyrate and propionate levels, and psyllium alone and psyllium plus resistant starch increased acetate levels. Human gut microbiota profiles at the phylum level were generally more like mouse 0.2% cellulose profiles than high fibre profiles. CONCLUSION: These supplements may be useful in combination with radiotherapy in patients with pelvic malignancy. Video Abstract.


Asunto(s)
Fibras de la Dieta , Suplementos Dietéticos , Microbioma Gastrointestinal , Inulina , Ratones Endogámicos C57BL , Psyllium , Neoplasias de la Vejiga Urinaria , Animales , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Inulina/administración & dosificación , Neoplasias de la Vejiga Urinaria/radioterapia , Neoplasias de la Vejiga Urinaria/patología , Humanos , Femenino , Traumatismos por Radiación/prevención & control , Intestinos/microbiología , Intestinos/efectos de la radiación , Linfocitos T CD8-positivos
7.
Fish Shellfish Immunol ; 149: 109593, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697374

RESUMEN

A type of fermented bile acids (FBAs) has been produced through a biological method, and its effects on growth performance, metabolism, and intestinal microbiota in largemouth bass were investigated. The results demonstrated that incorporating 0.03 %-0.05 % FBAs diet could improve the final weight, weight gain and specific growth rate, and decrease the feed conversion ratio. Dietary FBAs did not significantly affect the levels of high-density lipoprotein, low-density lipoprotein, and triglycerides, but decreased the activities of α-amylase in most groups. Adding FBAs to the diet significantly increased the integrity of the microscopic structure of the intestine, thickened the muscular layer of the intestine, and notably enhanced its intestinal barrier function. The addition of FBAs to the diet increased the diversity of the gut microbiota in largemouth bass. At the phylum level, there was an increase in the abundance of Proteobacteria, Firmicutes, Tenericutes and Cyanobacteria and a significant decrease in Actinobacteria and Bacteroidetes. At the genus level, the relative abundance of beneficial bacteria Mycoplasma in the GN6 group and Coprococcus in the GN4 group significantly increased, while the pathogenic Enhydrobacter was inhibited. Meanwhile, the highest levels of AKP and ACP were observed in the groups treated with 0.03 % FBAs, while the highest levels of TNF-α and IL-10 were detected in the group treated with 0.04 % FBAs. Additionally, the highest levels of IL-1ß, IL-8T, GF-ß, IGF-1, and IFN-γ were noted in the group treated with 0.06 % FBAs. These results suggested that dietary FBAs improved growth performance and intestinal wall health by altering lipid metabolic profiles and intestinal microbiota in largemouth bass.


Asunto(s)
Alimentación Animal , Lubina , Ácidos y Sales Biliares , Dieta , Microbioma Gastrointestinal , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ácidos y Sales Biliares/metabolismo , Alimentación Animal/análisis , Lubina/crecimiento & desarrollo , Lubina/inmunología , Dieta/veterinaria , Intestinos/microbiología , Fermentación , Metaboloma , Suplementos Dietéticos/análisis , Distribución Aleatoria
8.
Gut Microbes ; 16(1): 2353399, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38757687

RESUMEN

Intestinal stem cells (ISCs) play a pivotal role in gut physiology by governing intestinal epithelium renewal through the precise regulation of proliferation and differentiation. The gut microbiota interacts closely with the epithelium through myriad of actions, including immune and metabolic interactions, which translate into tight connections between microbial activity and ISC function. Given the diverse functions of the gut microbiota in affecting the metabolism of macronutrients and micronutrients, dietary nutrients exert pronounced effects on host-microbiota interactions and, consequently, the ISC fate. Therefore, understanding the intricate host-microbiota interaction in regulating ISC homeostasis is imperative for improving gut health. Here, we review recent advances in understanding host-microbiota immune and metabolic interactions that shape ISC function, such as the role of pattern-recognition receptors and microbial metabolites, including lactate and indole metabolites. Additionally, the diverse regulatory effects of the microbiota on dietary nutrients, including proteins, carbohydrates, vitamins, and minerals (e.g. iron and zinc), are thoroughly explored in relation to their impact on ISCs. Thus, we highlight the multifaceted mechanisms governing host-microbiota interactions in ISC homeostasis. Insights gained from this review provide strategies for the development of dietary or microbiota-based interventions to foster gut health.


Asunto(s)
Microbioma Gastrointestinal , Homeostasis , Interacciones Microbiota-Huesped , Mucosa Intestinal , Células Madre , Humanos , Microbioma Gastrointestinal/fisiología , Células Madre/metabolismo , Animales , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Bacterias/metabolismo , Bacterias/clasificación
9.
PLoS One ; 19(5): e0302522, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38758940

RESUMEN

Paddlefish has high economic and ecological value. In this study, microbial diversity and community structure in intestine, stomach, and mouth of paddlefish were detected using high-throughput sequencing. The results showed that the diversity and richness indices decreased along the digestive tract, and significantly lower proportion of those were observed in intestine. Firmicutes, Bacteroidetes and Proteobacteria were the dominant phyla. In top 10 phyla, there was no significant difference in mouth and stomach. But compared with intestine, there were significant differences in 8 of the 10 phyla, and Firmicutes and Bacteroidetes increased significantly, while Proteobacteria decreased significantly. There was no dominant genus in mouth and stomach, but Clostridium_sensu_stricto_1 and uncultured_bacterium_o_Bacteroidales was predominant in intestine. In conclusion, the species and abundance of microbiota in the mouth and stomach of paddlefish were mostly the same, but significantly different from those in intestine. Moreover, there was enrichment of the dominant bacteria in intestine.


Asunto(s)
Peces , Microbioma Gastrointestinal , Animales , Peces/microbiología , Tracto Gastrointestinal/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Boca/microbiología , Estómago/microbiología , Proteobacteria/aislamiento & purificación , Proteobacteria/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Intestinos/microbiología , Bacteroidetes/aislamiento & purificación , Bacteroidetes/genética , Firmicutes/aislamiento & purificación , Firmicutes/genética , Firmicutes/clasificación , ARN Ribosómico 16S/genética , Biodiversidad
10.
Cell Stem Cell ; 31(5): 591-592, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701755

RESUMEN

Recently in Cell Metabolism, Wei et al.1 unveiled a brain-to-gut pathway that conveys psychological stress to intestinal epithelial cells, leading to their dysfunction. This gut-brain axis involves a microbial metabolite, indole-3-acetate (IAA), as a niche signal that hampers mitochondrial respiration to skew intestinal stem cell (ISC) fate.


Asunto(s)
Células Madre , Células Madre/metabolismo , Células Madre/citología , Animales , Humanos , Intestinos/citología , Intestinos/microbiología , Estrés Fisiológico , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Diferenciación Celular , Mitocondrias/metabolismo
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 757-764, 2024 Apr 20.
Artículo en Chino | MEDLINE | ID: mdl-38708510

RESUMEN

OBJECTIVE: To explore the effect of intestinal nitrates on the growth of Klebsiella pneumoniae and its regulatory mechanisms. METHODS: K. pneumoniae strains with nitrate reductase narG and narZ single or double gene knockout or with NarXL gene knockout were constructed and observed for both aerobic and anaerobic growth in the presence of KNO3 using an automated bacterial growth analyzer and a spectrophotometer, respectively. The mRNA expressions of narG and narZ in K. pneumoniae in anaerobic cultures in the presence of KNO3 and the effect of the binary regulatory system NarXL on their expresisons were detected using qRT-PCR. Electrophoretic mobility shift assays (EMSA) and MST analysis were performed to explore the specific regulatory mechanisms of NarXL in sensing and utilizing nitrates. Competitive experiments were conducted to examine anaerobic growth advantages of narG and narZ gene knockout strains of K. pneumoniae in the presence of KNO3. RESULTS: The presence of KNO3 in anaerobic conditions, but not in aerobic conditions, promoted bacterial growth more effectively in the wild-type K. pneumoniae strain than in the narXL gene knockout strain. In anaerobic conditions, the narXL gene knockout strain showed significantly lowered mRNA expressions of narG and narZ (P < 0.0001). EMSA and MST experiments demonstrated that the NarXL regulator could directly bind to narG and narZ promoter regions. The wild-type K. pneumoniae strain in anaerobic cultures showed significantly increased expressions of narG and narZ mRNAs in the presence of KNO3 (P < 0.01), and narG gene knockout resulted in significantly attenuated anaerobic growth and competitive growth abilities of K. pneumoniae in the presence of KNO3 (P < 0.01). CONCLUSION: The binary regulatory system NarXL of K. pneumoniae can sense changes in intestinal nitrate concentration and directly regulate the expression of nitrate reductase genes narG and narZ to promote bacterial growth.


Asunto(s)
Klebsiella pneumoniae , Nitrato-Reductasa , Nitratos , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Klebsiella pneumoniae/efectos de los fármacos , Nitratos/metabolismo , Nitratos/farmacología , Nitrato-Reductasa/metabolismo , Nitrato-Reductasa/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Intestinos/microbiología , Regulación Bacteriana de la Expresión Génica , Anaerobiosis , Técnicas de Inactivación de Genes
12.
Food Funct ; 15(8): 4503-4514, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38567489

RESUMEN

Caloric restriction is an effective means of extending a healthy lifespan. Fasting mimicking diet (FMD) is a growing pattern of caloric restriction. We found that FMD significantly prolonged the lifespan of prematurely aging mice. In naturally aging mice, FMD improved cognitive and intestinal health. Through a series of behavioral experiments, we found that FMD relieved anxiety and enhanced cognition in aged mice. In the intestine, the FMD cycles enhanced the barrier function, reduced senescence markers, and maintained T cell naïve-memory balance in the lamina propria mucosa. To further explore the causes of immune alterations, we examined changes in the stool microbiota using 16S rRNA sequencing. We found that FMD remodeled gut bacterial composition and significantly expanded the abundance of Lactobacillus johnsonii. Our research revealed that FMD has in-depth investigative value as an anti-aging intervention for extending longevity and improving cognition, intestinal function, and gut microbiota composition.


Asunto(s)
Restricción Calórica , Cognición , Ayuno , Microbioma Gastrointestinal , Longevidad , Ratones Endogámicos C57BL , Animales , Ratones , Masculino , Envejecimiento , Intestinos/microbiología , Dieta
13.
World J Gastroenterol ; 30(15): 2096-2108, 2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38681982

RESUMEN

Probiotics are live microorganisms exerting beneficial effects on the host's health when administered in adequate amounts. Among the most popular and adequately studied probiotics are bacteria from the families Lactobacillaceae, Bifidobacteriaceae and yeasts. Most of them have been shown, both in vitro and in vivo studies of intestinal inflammation models, to provide favorable results by means of improving the gut microbiota composition, promoting the wound healing process and shaping the immunological responses. Chronic intestinal conditions, such as inflammatory bowel diseases (IBD), are characterized by an imbalance in microbiota composition, with decreased diversity, and by relapsing and persisting inflammation, which may lead to mucosal damage. Although the results of the clinical studies investigating the effect of probiotics on patients with IBD are still controversial, it is without doubt that these microorganisms and their metabolites, now named postbiotics, have a positive influence on both the host's microbiota and the immune system, and ultimately alter the topical tissue microenvironment. This influence is achieved through three axes: (1) By displacement of potential pathogens via competitive exclusion; (2) by offering protection to the host through the secretion of various defensive mediators; and (3) by supplying the host with essential nutrients. We will analyze and discuss almost all the in vitro and in vivo studies of the past 2 years dealing with the possible favorable effects of certain probiotic genus on gut immunological responses, highlighting which species are the most beneficial against intestinal inflammation.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Probióticos , Probióticos/uso terapéutico , Probióticos/administración & dosificación , Humanos , Microbioma Gastrointestinal/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Animales , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/inmunología , Intestinos/microbiología , Disbiosis/inmunología
14.
Microbiol Res ; 284: 127725, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38663233

RESUMEN

Increasing studies have focused on the relationship between Desulfovibrio bacteria (DSV) and host health in recent years. However, little is known about the mechanisms by which DSV affects host health and the strategies to accurately regulate DSV numbers. This review mainly presents the relationship between DSV and host health, potential modulatory strategies, and the potential mechanisms affecting host health. Evidence suggests that DSV can both promote host health and induce the occurrence and development of disease, and these effects are closely related to its metabolites (e.g., H2S and short-chain fatty acids) and biofilm. DSV abundance in the intestine is influenced by probiotics, prebiotics, diet, lifestyle, and drugs.


Asunto(s)
Biopelículas , Desulfovibrio , Microbioma Gastrointestinal , Probióticos , Desulfovibrio/metabolismo , Desulfovibrio/fisiología , Humanos , Microbioma Gastrointestinal/fisiología , Biopelículas/crecimiento & desarrollo , Intestinos/microbiología , Prebióticos , Animales , Ácidos Grasos Volátiles/metabolismo , Sulfuro de Hidrógeno/metabolismo , Dieta
15.
J Microbiol Biotechnol ; 34(4): 828-837, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38668685

RESUMEN

Vancomycin (VAN) and metronidazole (MTR) remain the current drugs of choice for the treatment of non-severe Clostridioides difficile infection (CDI); however, while their co-administration has appeared in clinical treatment, the efficacy varies greatly and the mechanism is unknown. In this study, a CDI mouse model was constructed to evaluate the therapeutic effects of VAN and MTR alone or in combination. For a perspective on the intestinal ecology, 16S rRNA amplicon sequencing and non-targeted metabolomics techniques were used to investigate changes in the fecal microbiota and metabolome of mice under the co-administration treatment. As a result, the survival rate of mice under co-administration was not dramatically different compared to that of single antibiotics, and the former caused intestinal tissue hyperplasia and edema. Co-administration also significantly enhanced the activity of amino acid metabolic pathways represented by phenylalanine, arginine, proline, and histidine, decreased the level of deoxycholic acid (DCA), and downregulated the abundance of beneficial microbes, such as Bifidobacterium and Akkermansia. VAN plays a dominant role in microbiota regulation in co-administration. In addition, co-administration reduced or increased the relative abundance of antibiotic-sensitive bacteria, including beneficial and harmful microbes, without a difference. Taken together, there are some risks associated with the co-administration of VAN and MTR, and this combination mode should be used with caution in CDI treatment.


Asunto(s)
Antibacterianos , Clostridioides difficile , Infecciones por Clostridium , Modelos Animales de Enfermedad , Quimioterapia Combinada , Heces , Microbioma Gastrointestinal , Metronidazol , ARN Ribosómico 16S , Vancomicina , Animales , Metronidazol/administración & dosificación , Vancomicina/administración & dosificación , Vancomicina/farmacología , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/genética , ARN Ribosómico 16S/genética , Heces/microbiología , Intestinos/microbiología , Intestinos/efectos de los fármacos , Masculino , Bacterias/clasificación , Bacterias/genética , Bacterias/efectos de los fármacos , Metaboloma/efectos de los fármacos
16.
J Nutr Biochem ; 129: 109637, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38574828

RESUMEN

Adequate dietary L-tryptophan (Trp) governs intestinal homeostasis in piglets. However, the defensive role of Trp in the diet against enterotoxigenic Escherichia coli F4 (K88) in pigs is still poorly understood. Here, sixty (6.15 ± 1.52 kg, 24-day-old, Duroc × Landrace × Yorkshire) weaned piglets were used for an E. coli F4 attack test in a 2 × 2 factorial design. The growth (ADG, ADFI, GH), immune factors (IL-10, IgA, IgG, IgM), Trp metabolite 5-HT, intestinal morphology (jejunal and colonic VH), mRNA expression of ß-defensins (jejunal BD-127, BD-119, ileal BD-1, BD-127), and abundance of beneficial microorganisms in the colon (Prevotella 9, Lactobacillus, Phascolarctobacterium, Faecalibacterium) were higher in the piglets in the HT (High Trp) and HTK (High Trp, K88) groups than in the LT (Low Trp) and LTK (Low Trp, K88) groups (P<.05), while FCR, diarrhea rate, diarrhea index, serum Trp, Kyn, IDO, D-LA, ET, and abundance of harmful microorganisms in the colon (Spirochaetes, Fusobacteria, Prevotella, Christensenellaceae R7) were lower in the HT and HTK groups than in the LT and LTK groups (P<.05). High Trp reduced the expression of virulence genes (K88 and LT) after E. coli F4 attack (P<.05). The IL-6, TNF-α was lower in the HTK group than in the LT, LTK group (P<.05). In short, a diet containing 0.35% Trp protected piglets from enterotoxigenic E. coli F4 (K88) via Trp metabolism promoting BD expression in the intestinal mucosa, which improved growth and intestinal health.


Asunto(s)
Escherichia coli Enterotoxigénica , Triptófano , Destete , beta-Defensinas , Animales , Triptófano/metabolismo , Porcinos , beta-Defensinas/metabolismo , Infecciones por Escherichia coli/veterinaria , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/prevención & control , Intestinos/microbiología , Alimentación Animal , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Dieta/veterinaria
17.
Cell Host Microbe ; 32(5): 661-675.e10, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38657606

RESUMEN

The intestine and liver are thought to metabolize dietary nutrients and regulate host nutrient homeostasis. Here, we find that the gut microbiota also reshapes the host amino acid (aa) landscape via efficiently metabolizing intestinal aa. To identify the responsible microbes/genes, we developed a metabolomics-based assay to screen 104 commensals and identified candidates that efficiently utilize aa. Using genetics, we identified multiple responsible metabolic genes in phylogenetically diverse microbes. By colonizing germ-free mice with the wild-type strain and their isogenic mutant deficient in individual aa-metabolizing genes, we found that these genes regulate the availability of gut and circulatory aa. Notably, microbiota genes for branched-chain amino acids (BCAAs) and tryptophan metabolism indirectly affect host glucose homeostasis via peripheral serotonin. Collectively, at single-gene level, this work characterizes a microbiota-encoded metabolic activity that affects host nutrient homeostasis and provides a roadmap to interrogate microbiota-dependent activity to improve human health.


Asunto(s)
Aminoácidos de Cadena Ramificada , Aminoácidos , Microbioma Gastrointestinal , Homeostasis , Triptófano , Animales , Microbioma Gastrointestinal/fisiología , Ratones , Aminoácidos/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Triptófano/metabolismo , Ratones Endogámicos C57BL , Nutrientes/metabolismo , Intestinos/microbiología , Humanos , Metabolómica , Glucosa/metabolismo , Serotonina/metabolismo , Vida Libre de Gérmenes , Bacterias/metabolismo , Bacterias/genética , Bacterias/clasificación , Masculino
18.
ACS Biomater Sci Eng ; 10(5): 3387-3400, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38656158

RESUMEN

Given the worldwide problem posed by enteric pathogens, the discovery of safe and efficient intestinal adjuvants combined with novel antigen delivery techniques is essential to the design of mucosal vaccines. In this work, we designed poly (lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs) to codeliver all-trans retinoic acid (atRA), novel antigens, and CpG. To address the insolubility of the intestinal adjuvant atRA, we utilized PLGA to encapsulate atRA and form a "nanocapsid" with polydopamine. By leveraging polydopamine, we adsorbed the water-soluble antigens and the TLR9 agonist CpG onto the NPs' surface, resulting in the pathogen-mimicking PLPCa NPs. In this study, the novel fusion protein (HBf), consisting of the Mycobacterium avium subspecies paratuberculosis antigens HBHA, Ag85B, and Bfra, was coloaded onto the NPs. In vitro, PLPCa NPs were shown to promote the activation and maturation of bone marrow-derived dendritic cells. Additionally, we found that PLPCa NPs created an immune-rich microenvironment at the injection site following intramuscular administration. From the results, the PLPCa NPs induced strong IgA levels in the gut in addition to enhancing powerful systemic immune responses. Consequently, significant declines in the bacterial burden and inflammatory score were noted in PLPCa NPs-treated mice. In summary, PLPCa can serve as a novel and safe vaccine delivery platform against gut pathogens, such as paratuberculosis, capable of activating both systemic and intestinal immunity.


Asunto(s)
Nanopartículas , Paratuberculosis , Animales , Nanopartículas/química , Paratuberculosis/inmunología , Paratuberculosis/prevención & control , Ratones , Tretinoina/química , Tretinoina/farmacología , Mycobacterium avium subsp. paratuberculosis/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/química , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Intestinos/inmunología , Intestinos/microbiología , Ratones Endogámicos C57BL , Femenino , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/administración & dosificación , Vacunas Bacterianas/inmunología , Ratones Endogámicos BALB C
19.
J Anim Sci ; 1022024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38629856

RESUMEN

Frequent incidence of postweaning enterotoxigenic Escherichia coli (ETEC) diarrhea in the swine industry contributes to high mortality rates and associated economic losses. In this study, a combination of butyric, caprylic, and capric fatty acid monoglycerides was investigated to promote intestinal integrity and host defenses in weanling pigs infected with ETEC. A total of 160 pigs were allotted to treatment groups based on weight and sex. Throughout the 17-d study, three treatment groups were maintained: sham-inoculated pigs fed a control diet (uninfected control [UC], n = 40), ETEC-inoculated pigs fed the same control diet (infected control [IC], n = 60), and ETEC-inoculated pigs fed the control diet supplemented with monoglycerides included at 0.3% of the diet (infected supplemented [MG], n = 60). After a 7-d acclimation period, pigs were orally inoculated on each of three consecutive days with either 3 mL of a sham-control (saline) or live ETEC culture (3 × 109 colony-forming units/mL). The first day of inoculations was designated as 0 d postinoculation (DPI), and all study outcomes reference this time point. Fecal, tissue, and blood samples were collected from 48 individual pigs (UC, n = 12; IC, n = 18; MG, n = 18) on 5 and 10 DPI for analysis of dry matter (DM), bacterial enumeration, inflammatory markers, and intestinal permeability. ETEC-inoculated pigs in both the IC and MG groups exhibited clear signs of infection including lower (P < 0.05) gain:feed and fecal DM, indicative of excess water in the feces, and elevated (P < 0.05) rectal temperatures, total bacteria, total E. coli, and total F18 ETEC during the peak-infection period (5 DPI). Reduced (P < 0.05) expression of the occludin, tumor necrosis factor α, and vascular endothelial growth factor A genes was observed in both ETEC-inoculated groups at the 5 DPI time point. There were no meaningful differences between treatments for any of the outcomes measured at 10 DPI. Overall, all significant changes were the result of the ETEC infection, not monoglyceride supplementation.


Infection caused by the bacterium known as enterotoxigenic Escherichia coli (ETEC) is a common disruptor of weaned pigs' health, leading to economic losses for the producers. To determine if nutritional supplementation could help protect against these losses, weaned pigs were assigned to one of three treatments: 1) uninfected and fed a standard nursery pig diet, 2) infected with ETEC and fed the same standard diet, or 3) infected with ETEC and fed the standard diet supplemented with a combination of butyric, caprylic, and capric fatty acid monoglycerides. Growth performance was tracked throughout the 17-d study and health outcomes were measured at the peak and resolution of ETEC infection. At the peak-infection time point, pigs that were infected with ETEC had lower fecal moisture content, greater fecal bacterial concentrations, and elevated body temperatures compared with uninfected pigs. Additionally, infection reduced expression of genes related to inflammation, angiogenesis, and the intestinal barrier during the peak-infection period. Overall, all significant changes were the result of the ETEC infection, and there were no meaningful differences observed between the different treatments.


Asunto(s)
Alimentación Animal , Suplementos Dietéticos , Escherichia coli Enterotoxigénica , Infecciones por Escherichia coli , Monoglicéridos , Enfermedades de los Porcinos , Animales , Porcinos , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/prevención & control , Infecciones por Escherichia coli/veterinaria , Infecciones por Escherichia coli/prevención & control , Escherichia coli Enterotoxigénica/fisiología , Masculino , Femenino , Alimentación Animal/análisis , Dieta/veterinaria , Intestinos/microbiología , Diarrea/veterinaria , Diarrea/microbiología , Heces/microbiología , Destete
20.
BMC Biol ; 22(1): 76, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38581018

RESUMEN

BACKGROUND: The gut microbiota, vital for host health, influences metabolism, immune function, and development. Understanding the dynamic processes of bacterial accumulation within the gut is crucial, as it is closely related to immune responses, antibiotic resistance, and colorectal cancer. We investigated Escherichia coli behavior and distribution in zebrafish larval intestines, focusing on the gut microenvironment. RESULTS: We discovered that E. coli spread was considerably suppressed within the intestinal folds, leading to a strong physical accumulation in the folds. Moreover, a higher concentration of E. coli on the dorsal side than on the ventral side was observed. Our in vitro microfluidic experiments and theoretical analysis revealed that the overall distribution of E. coli in the intestines was established by a combination of physical factor and bacterial taxis. CONCLUSIONS: Our findings provide valuable insight into how the intestinal microenvironment affects bacterial motility and accumulation, enhancing our understanding of the behavioral and ecological dynamics of the intestinal microbiota.


Asunto(s)
Microbioma Gastrointestinal , Intestinos , Animales , Intestinos/microbiología , Escherichia coli/fisiología , Factores Biológicos , Pez Cebra/fisiología , Microbioma Gastrointestinal/fisiología , Bacterias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA