Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.613
Filtrar
1.
Front Immunol ; 15: 1394501, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38774883

RESUMEN

Extracellular vesicles (EVs) are cell-derived membrane-surrounded vesicles that carry bioactive molecules. Among EVs, outer membrane vesicles (OMVs), specifically produced by Gram-negative bacteria, have been extensively characterized and their potential as vaccines, adjuvants or immunotherapeutic agents, broadly explored in mammals. Nonetheless, Gram-positive bacteria can also produce bilayered spherical structures from 20 to 400 nm involved in pathogenesis, antibiotic resistance, nutrient uptake and nucleic acid transfer. However, information regarding their immunomodulatory potential is very scarce, both in mammals and fish. In the current study, we have produced EVs from the Gram-positive probiotic Bacillus subtilis and evaluated their immunomodulatory capacities using a rainbow trout intestinal epithelial cell line (RTgutGC) and splenic leukocytes. B. subtilis EVs significantly up-regulated the transcription of several pro-inflammatory and antimicrobial genes in both RTgutGC cells and splenocytes, while also up-regulating many genes associated with B cell differentiation in the later. In concordance, B. subtilis EVs increased the number of IgM-secreting cells in splenocyte cultures, while at the same time increased the MHC II surface levels and antigen-processing capacities of splenic IgM+ B cells. Interestingly, some of these experiments were repeated comparing the effects of B. subtilis EVs to EVs obtained from another Bacillus species, Bacillus megaterium, identifying important differences. The data presented provides evidence of the immunomodulatory capacities of Gram-positive EVs, pointing to the potential of B. subtilis EVs as adjuvants or immunostimulants for aquaculture.


Asunto(s)
Bacillus subtilis , Vesículas Extracelulares , Leucocitos , Oncorhynchus mykiss , Bazo , Animales , Bacillus subtilis/inmunología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Oncorhynchus mykiss/inmunología , Oncorhynchus mykiss/microbiología , Bazo/inmunología , Bazo/citología , Leucocitos/inmunología , Leucocitos/metabolismo , Probióticos/farmacología , Línea Celular , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Inmunomodulación , Intestinos/inmunología
2.
PLoS One ; 19(5): e0301477, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38768108

RESUMEN

Food allergy is widely recognized as a significant health issue, having escalated into a global epidemic, subsequently giving rise to the development of numerous additional complications. Currently, the sole efficient method to curb the progression of allergy is through the implementation of an elimination diet. The increasing number of newly identified allergens makes it harder to completely remove or avoid them effectively. The immunoreactivity of proteins of bacterial origin remains an unexplored topic. Despite the substantial consumption of microbial proteins in our diets, the immunologic mechanisms they might induce require thorough validation. This stands as the primary objective of this study. The primary objective of this study was to evaluate the effects of bacterial proteins on the intestinal barrier and immune system parameters during hypersensitivity induction in both developing and mature organisms. The secondary objective was to evaluate the role of lipids in the immunoreactivity programming of these bacterial proteins. Notably, in this complex, comprehensively designed in vitro, in vivo, and ex vivo trial, the immunoreactivity of various bacterial proteins will be examined. In summary, the proposed study intends to address the knowledge gaps regarding the effects of Lactobacillus microbial proteins on inflammation, apoptosis, autophagy, and intestinal barrier integrity in a single study.


Asunto(s)
Proteínas Bacterianas , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/inmunología , Lípidos , Leche/microbiología , Leche/inmunología , Ratones , Lactobacillales/metabolismo , Lactobacillales/inmunología , Hipersensibilidad a los Alimentos/inmunología , Hipersensibilidad a los Alimentos/microbiología , Femenino , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología
3.
Food Res Int ; 186: 114322, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729712

RESUMEN

Lactobacillus delbrueckii subsp. lactis CIDCA 133 is a health-promoting bacterium that can alleviate gut inflammation and improve the epithelial barrier in a mouse model of mucositis. Despite these beneficial effects, the protective potential of this strain in other inflammation models, such as inflammatory bowel disease, remains unexplored. Herein, we examined for the first time the efficacy of Lactobacillus delbrueckii CIDCA 133 incorporated into a fermented milk formulation in the recovery of inflammation, epithelial damage, and restoration of gut microbiota in mice with dextran sulfate sodium-induced colitis. Oral administration of Lactobacillus delbrueckii CIDCA 133 fermented milk relieved colitis by decreasing levels of inflammatory factors (myeloperoxidase, N-acetyl-ß-D-glucosaminidase, toll-like receptor 2, nuclear factor-κB, interleukins 10 and 6, and tumor necrosis factor), secretory immunoglobulin A levels, and intestinal paracellular permeability. This immunobiotic also modulated the expression of tight junction proteins (zonulin and occludin) and the activation of short-chain fatty acids-related receptors (G-protein coupled receptors 43 and 109A). Colonic protection was effectively associated with acetate production and restoration of gut microbiota composition. Treatment with Lactobacillus delbrueckii CIDCA 133 fermented milk increased the abundance of Firmicutes members (Lactobacillus genus) while decreasing the abundance of Proteobacteria (Helicobacter genus) and Bacteroidetes members (Bacteroides genus). These promising outcomes influenced the mice's mucosal healing, colon length, body weight, and disease activity index, demonstrating that this immunobiotic could be explored as an alternative approach for managing inflammatory bowel disease.


Asunto(s)
Colitis , Productos Lácteos Cultivados , Sulfato de Dextran , Microbioma Gastrointestinal , Lactobacillus delbrueckii , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Colitis/microbiología , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/tratamiento farmacológico , Lactobacillus delbrueckii/metabolismo , Productos Lácteos Cultivados/microbiología , Ratones , Probióticos/uso terapéutico , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Inflamación , Colon/microbiología , Colon/metabolismo , Lactobacillus
4.
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
5.
Nutrients ; 16(9)2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38732577

RESUMEN

BACKGROUND: Cadmium (Cd) is an environmental contaminant that poses risks to human and animal health. Selenium (Se), a beneficial element, alleviates the detrimental consequences of colitis and Cd toxicity. Se is found in food products as both inorganic Se (sodium selenite) and organic Se (typically Se-enriched yeast). Nano-selenium (nano-Se; a novel form of Se produced through the bioreduction of Se species) has recently garnered considerable interest, although its effects against Cd-induced enterotoxicity are poorly understood. The aim of this study was to investigate the impact of nano-selenium on mitigating cadmium toxicity and safeguarding the integrity of the intestinal barrier. METHODS: For a total of two cycles, we subjected 6-week-old C57 mice to chronic colitis by exposing them to Cd and nano-selenium for two weeks, followed by DSS water for one week. RESULTS: The application of nano-selenium mitigated the intensity of colitis and alleviated inflammation in the colon. Nano-selenium enhanced the diversity of the intestinal flora, elevated the concentration of short-chain fatty acids (SCFAs) in feces, and improved the integrity of the intestinal barrier. CONCLUSIONS: In summary, nano-Se may reduce intestinal inflammation by regulating the growth of intestinal microorganisms and protecting the intestinal barrier.


Asunto(s)
Cadmio , Colitis , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Selenio , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Selenio/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Masculino , Enfermedad Crónica , Modelos Animales de Enfermedad , Nanopartículas , Ácidos Grasos Volátiles/metabolismo , Heces/microbiología , Sulfato de Dextran , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología
6.
Nutrients ; 16(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732614

RESUMEN

The incidence of ulcerative colitis (UC) is increasing annually, and UC has a serious impact on patients' lives. Polysaccharides have gained attention as potential drug candidates for treating ulcerative colitis (UC) in recent years. Huaier (Trametes robiniophila Murr) is a fungus that has been used clinically for more than 1000 years, and its bioactive polysaccharide components have been reported to possess immunomodulatory effects, antitumour potential, and renoprotective effects. In this study, we aimed to examine the protective effects and mechanisms of Huaier polysaccharide (HP) against UC. Based on the H2O2-induced oxidative stress model in HT-29 cells and the dextran sulphate sodium salt (DSS)-induced UC model, we demonstrated that Huaier polysaccharides significantly alleviated DSS-induced colitis (weight loss, elevated disease activity index (DAI) scores, and colonic shortening). In addition, HP inhibited oxidative stress and inflammation and alleviated DSS-induced intestinal barrier damage. It also significantly promoted the expression of the mucin Muc2. Furthermore, HP reduced the abundance of harmful bacteria Escherichia-Shigella and promoted the abundance of beneficial bacteria Muribaculaceae_unclassified, Anaerotruncus, and Ruminococcaceae_unclassified to regulate the intestinal flora disturbance caused by DSS. Nontargeted metabolomics revealed that HP intervention would modulate metabolism by promoting levels of 3-hydroxybutyric acid, phosphatidylcholine (PC), and phosphatidylethanolamine (PE). These results demonstrated that HP had the ability to mitigate DSS-induced UC by suppressing oxidative stress and inflammation, maintaining the intestinal barrier, and modulating the intestinal flora. These findings will expand our knowledge of how HP functions and offer a theoretical foundation for using HP as a potential prebiotic to prevent UC.


Asunto(s)
Sulfato de Dextran , Microbioma Gastrointestinal , Estrés Oxidativo , Polisacáridos , Microbioma Gastrointestinal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Humanos , Polisacáridos/farmacología , Ratones , Masculino , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/microbiología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Células HT29 , Ratones Endogámicos C57BL , Colitis/inducido químicamente , Colitis/tratamiento farmacológico
7.
Sci Rep ; 14(1): 10702, 2024 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729976

RESUMEN

Coccidiosis, an intestinal disease caused by Eimeria parasites, is responsible for major losses in the poultry industry by impacting chicken health. The gut microbiota is associated with health factors, such as nutrient exchange and immune system modulation, requiring understanding on the effects of Eimeria infection on the gut microbiota. This study aimed to determine the effects of Eimeria acervulina infection on the luminal and mucosal microbiota of the cecum (CeL and CeM) and ileum (IlL and IlM) at multiple time points (days 3, 5, 7, 10, and 14) post-infection. E. acervulina infection decreased evenness in CeL microbiota at day 10, increased richness in CeM microbiota at day 3 before decreasing richness at day 14, and decreased richness in IlL microbiota from day 3 to 10. CeL, CeM, and IlL microbiota differed between infected and control birds based on beta diversity at varying time points. Infection reduced relative abundance of bacterial taxa and some predicted metabolic pathways known for short-chain fatty acid production in CeL, CeM, and IlL microbiota, but further understanding of metabolic function is required. Despite E. acervulina primarily targeting the duodenum, our findings demonstrate the infection can impact bacterial diversity and abundance in the cecal and ileal microbiota.


Asunto(s)
Ciego , Pollos , Coccidiosis , Eimeria , Microbioma Gastrointestinal , Íleon , Enfermedades de las Aves de Corral , Animales , Pollos/microbiología , Pollos/parasitología , Ciego/microbiología , Ciego/parasitología , Eimeria/fisiología , Íleon/microbiología , Íleon/parasitología , Coccidiosis/veterinaria , Coccidiosis/parasitología , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/parasitología , Mucosa Intestinal/microbiología , Mucosa Intestinal/parasitología
8.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731952

RESUMEN

Porphyromonas gingivalis (Pg), a Gram-negative oral pathogen, promotes and accelerates periodontitis-associated gut disorders. Intestinal epithelial barrier dysfunction is crucial in the pathogenesis of intestinal and systemic diseases. In this study, we sought to elucidate the protective role of cinnamaldehyde (CNM, an activator of Nrf2) against P. gingivalis (W83) and Pg-derived lipopolysaccharide (Pg-LPS) induced intestinal epithelial barrier dysfunction via antioxidative mechanisms in IEC-6 cells. IEC-6 (ATCC, CRL-1592) cells were pretreated with or without CNM (100 µM), in the presence or absence of P. gingivalis (strain W83, 109 MOI) or Pg-LPS (1, 10, and 100 µg/mL), respectively, between 0-72 h time points by adopting a co-culture method. Intestinal barrier function, cytokine secretion, and intestinal oxidative stress protein markers were analyzed. P. gingivalis or Pg-LPS significantly (p < 0.05) increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels expressing oxidative stress damage. Pg-LPS, as well as Pg alone, induces inflammatory cytokines via TLR-4 signaling. Furthermore, infection reduced Nrf2 and NAD(P)H quinone dehydrogenase 1 (NQO1). Interestingly, inducible nitric oxide synthase (iNOS) protein expression significantly (p < 0.05) increased with Pg-LPS or Pg infection, with elevated levels of nitric oxide (NO). CNM treatment suppressed both Pg- and Pg-LPS-induced intestinal oxidative stress damage by reducing ROS, MDA, and NO production. Furthermore, CNM treatment significantly upregulated the expression of tight junction proteins via increasing the phosphorylation levels of PI3K/Akt/Nrf2 suppressing inflammatory cytokines. CNM protected against Pg infection-induced intestinal epithelial barrier dysfunction by activating the PI3K/Akt-mediated Nrf2 signaling pathway in IEC-6 cells.


Asunto(s)
Acroleína , Mucosa Intestinal , Factor 2 Relacionado con NF-E2 , Óxido Nítrico , Fosfatidilinositol 3-Quinasas , Porphyromonas gingivalis , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Factor 2 Relacionado con NF-E2/metabolismo , Acroleína/análogos & derivados , Acroleína/farmacología , Animales , Transducción de Señal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Porphyromonas gingivalis/patogenicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Óxido Nítrico/metabolismo , Línea Celular , Lipopolisacáridos , Estrés Oxidativo/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Citocinas/metabolismo
9.
J Interferon Cytokine Res ; 44(5): 208-220, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38691831

RESUMEN

Intestinal damage and secondary bacterial translocation are caused by the inflammatory response induced by sepsis. Tongfu Lifei (TLF) decoction has a protective effect on sepsis-related gastrointestinal function injury. However, the relation between gut microbiota, immune barrier, and sepsis under the treatment of TLF have not been well clarified yet. Here, rats were subjected to cecal ligation and puncture (CLP) to create a sepsis model. Subsequently, the TLF decoction was given to CLP rats by gavage, fecal microbiota transplantation (FMT), and antibiotic were used as positive control. TLF suppressed the inflammatory response and improved the pathological changes in the intestines of CLP rats. Besides, TLF promoted the balance of the percentage of the Th17 and Treg cells. Intestinal barrier function was also improved by TLF through enhancing ZO-1, and Occludin and Claudin 1 expression, preventing the secondary translocation of other gut microbiota. TLF dramatically boosted the gut microbiota's alpha- and beta-diversity in CLP rats. Moreover, it increased the relative abundance of anti-inflammatory gut microbiota and changed the progress of the glucose metabolism. In short, TLF regulated the gut microbiota to balance the ratio of Th17/Treg cells, reducing the inflammation in serum and intestinal mucosal injury in rats.


Asunto(s)
Medicamentos Herbarios Chinos , Microbioma Gastrointestinal , Mucosa Intestinal , Sepsis , Linfocitos T Reguladores , Células Th17 , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Sepsis/inmunología , Sepsis/tratamiento farmacológico , Sepsis/complicaciones , Células Th17/inmunología , Células Th17/efectos de los fármacos , Ratas , Medicamentos Herbarios Chinos/farmacología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/microbiología , Masculino , Ratas Sprague-Dawley
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.
Front Cell Infect Microbiol ; 14: 1346087, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736751

RESUMEN

Epithelial cells form a resilient barrier and orchestrate defensive and reparative mechanisms to maintain tissue stability. This review focuses on gut and airway epithelia, which are positioned where the body interfaces with the outside world. We review the many signaling pathways and mechanisms by which epithelial cells at the interface respond to invading pathogens to mount an innate immune response and initiate adaptive immunity and communicate with other cells, including resident microbiota, to heal damaged tissue and maintain homeostasis. We compare and contrast how airway and gut epithelial cells detect pathogens, release antimicrobial effectors, collaborate with macrophages, Tregs and epithelial stem cells to mount an immune response and orchestrate tissue repair. We also describe advanced research models for studying epithelial communication and behaviors during inflammation, tissue injury and disease.


Asunto(s)
Homeostasis , Inmunidad Innata , Mucosa Intestinal , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Animales , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/inmunología , Células Epiteliales/microbiología , Transducción de Señal , Inmunidad Adaptativa , Macrófagos/inmunología , Macrófagos/microbiología , Interacciones Huésped-Patógeno
12.
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
13.
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
14.
Sci Rep ; 14(1): 11479, 2024 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769412

RESUMEN

Salmonella enterica serovar Dublin (S. Dublin) is an important enteric pathogen affecting cattle and poses increasing public health risks. Understanding the pathophysiology and host-pathogen interactions of S. Dublin infection are critical for developing effective control strategies, yet studies are hindered by the lack of physiologically relevant in vitro models. This study aimed to generate a robust ileal monolayer derived from adult bovine organoids, validate its feasibility as an in vitro infection model with S. Dublin, and evaluate the epithelial response to infection. A stable, confluent monolayer with a functional epithelial barrier was established under optimized culture conditions. The model's applicability for studying S. Dublin infection was confirmed by documenting intracellular bacterial invasion and replication, impacts on epithelial integrity, and a specific inflammatory response, providing insights into the pathogen-epithelium interactions. The study underscores the utility of organoid-derived monolayers in advancing our understanding of enteric infections in livestock and highlights implications for therapeutic strategy development and preventive measures, with potential applications extending to both veterinary and human medicine. The established bovine ileal monolayer offers a novel and physiologically relevant in vitro platform for investigating enteric pathogen-host interactions, particularly for pathogens like S. Dublin.


Asunto(s)
Interacciones Huésped-Patógeno , Íleon , Organoides , Salmonelosis Animal , Animales , Bovinos , Organoides/microbiología , Íleon/microbiología , Íleon/patología , Salmonelosis Animal/microbiología , Salmonella enterica/patogenicidad , Salmonella enterica/fisiología , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/microbiología , Enfermedades de los Bovinos/microbiología
15.
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
16.
Arch Microbiol ; 206(6): 265, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38761195

RESUMEN

Acute pancreatitis frequently causes intestinal barrier damage, which aggravates pancreatitis. Although Clostridium butyricum exerts anti-inflammatory and protective effects on the intestinal barrier during acute pancreatitis, the underlying mechanism is unclear. The G protein-coupled receptors 109 A (GPR109A) and adenosine monophosphate-activated protein kinase (AMPK)/ peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) signaling pathways can potentially influence the integrity of the intestinal barrier. Our study generated acute pancreatitis mouse models via intraperitoneal injection of cerulein and lipopolysaccharides. After intervention with Clostridium butyricum, the model mice showed reduced small intestinal and colonic intestinal barrier damage, dysbiosis amelioration, and increased GPR109A/AMPK/PGC-1α expression. In conclusion, Clostridium butyricum could improve pancreatic and intestinal inflammation and pancreatic injury, and relieve acute pancreatitis-induced intestinal barrier damage in the small intestine and colon, which may be associated with GPR109A/AMPK/PGC-1α.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Clostridium butyricum , Modelos Animales de Enfermedad , Pancreatitis , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptores Acoplados a Proteínas G , Animales , Clostridium butyricum/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Ratones , Pancreatitis/metabolismo , Pancreatitis/microbiología , Pancreatitis/patología , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones Endogámicos C57BL , Masculino , Transducción de Señal , Regulación hacia Arriba
17.
Nat Commun ; 15(1): 4051, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744839

RESUMEN

Intestinal homeostasis is maintained by the response of gut-associated lymphoid tissue to bacteria transported across the follicle associated epithelium into the subepithelial dome. The initial response to antigens and how bacteria are handled is incompletely understood. By iterative application of spatial transcriptomics and multiplexed single-cell technologies, we identify that the double negative 2 subset of B cells, previously associated with autoimmune diseases, is present in the subepithelial dome in health. We show that in this location double negative 2 B cells interact with dendritic cells co-expressing the lupus autoantigens DNASE1L3 and C1q and microbicides. We observe that in humans, but not in mice, dendritic cells expressing DNASE1L3 are associated with sampled bacteria but not DNA derived from apoptotic cells. We propose that fundamental features of autoimmune diseases are microbiota-associated, interacting components of normal intestinal immunity.


Asunto(s)
Linfocitos B , Células Dendríticas , Endodesoxirribonucleasas , Microbioma Gastrointestinal , Animales , Humanos , Ratones , Linfocitos B/inmunología , Linfocitos B/metabolismo , Microbioma Gastrointestinal/inmunología , Endodesoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Femenino , Ratones Endogámicos C57BL , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Masculino
18.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38719750

RESUMEN

Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans-MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms.


Asunto(s)
Candida albicans , Enfermedad Celíaca , Homeostasis , Mastocitos , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/metabolismo , Humanos , Candida albicans/patogenicidad , Candida albicans/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Microbioma Gastrointestinal/inmunología , Disbiosis/inmunología , Candidiasis/inmunología , Candidiasis/microbiología , Animales , Candida/patogenicidad , Candida/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo
19.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732126

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) is a critical public health concern due to its role in severe gastrointestinal illnesses in humans, including hemorrhagic colitis and the life-threatening hemolytic uremic syndrome. While highly pathogenic to humans, cattle, the main reservoir for EHEC, often remain asymptomatic carriers, complicating efforts to control its spread. Our study introduces a novel method to investigate EHEC using organoid-derived monolayers from adult bovine ileum and rectum. These polarized epithelial monolayers were exposed to EHEC for four hours, allowing us to perform comparative analyses between the ileal and rectal tissues. Our findings mirrored in vivo observations, showing a higher colonization rate in the rectum compared with the ileum (44.0% vs. 16.5%, p < 0.05). Both tissues exhibited an inflammatory response with increased expression levels of TNF-a (p < 0.05) and a more pronounced increase of IL-8 in the rectum (p < 0.01). Additionally, the impact of EHEC on the mucus barrier varied across these gastrointestinal regions. Innovative visualization techniques helped us study the ultrastructure of mucus, revealing a net-like mucin glycoprotein organization. While further cellular differentiation could enhance model accuracy, our research significantly deepens understanding of EHEC pathogenesis in cattle and informs strategies for the preventative measures and therapeutic interventions.


Asunto(s)
Escherichia coli Enterohemorrágica , Íleon , Organoides , Recto , Animales , Bovinos , Íleon/microbiología , Íleon/metabolismo , Íleon/ultraestructura , Recto/microbiología , Escherichia coli Enterohemorrágica/patogenicidad , Organoides/metabolismo , Organoides/microbiología , Moco/metabolismo , Infecciones por Escherichia coli/microbiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestructura
20.
PLoS One ; 19(4): e0301822, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38603764

RESUMEN

The human gut is a complex environment where the microbiota and its metabolites play a crucial role in the maintenance of a healthy state. The aim of the present work is the reconstruction of a new in vitro minimal human gut microbiota resembling the microbe-microbe networking comprising the principal phyla (Bacillota, Bacteroidota, Pseudomonadota, and Actinomycetota), to comprehend the intestinal ecosystem complexity. In the reductionist model, we mimicked the administration of Maitake extract as prebiotic and a probiotic formulation (three strains belonging to Lactobacillus and Bifidobacterium genera), evaluating the modulation of strain levels, the release of beneficial metabolites, and their health-promoting effects on human cell lines of the intestinal environment. The administration of Maitake and the selected probiotic strains generated a positive modulation of the in vitro bacterial community by qPCR analyses, evidencing the prominence of beneficial strains (Lactiplantibacillus plantarum and Bifidobacterium animalis subsp. lactis) after 48 hours. The bacterial community growths were associated with the production of metabolites over time through GC-MSD analyses such as lactate, butyrate, and propionate. Their effects on the host were evaluated on cell lines of the intestinal epithelium and the immune system, evidencing positive antioxidant (upregulation of SOD1 and NQO1 genes in HT-29 cell line) and anti-inflammatory effects (production of IL-10 from all the PBMCs). Therefore, the results highlighted a positive modulation induced by the synergic activities of probiotics and Maitake, inducing a tolerogenic microenvironment.


Asunto(s)
Bifidobacterium animalis , Microbioma Gastrointestinal , Grifola , Probióticos , Humanos , Ecosistema , Mucosa Intestinal/microbiología , Lactobacillus/fisiología , Probióticos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...