Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.761
Filtrar
2.
PLoS One ; 19(6): e0305711, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38935795

RESUMEN

Chronic enteropathies are a common cause of morbidity in dogs and are associated with disruption of the normal gastrointestinal mucosal barrier. The objective of this prospective study was to determine the association between measures of gastrointestinal dysbiosis and plasma concentrations of glucagon-like peptide-2, a hormone responsible for normal mucosal structure, in dogs with chronic enteropathies. Fecal 16S V4 rRNA gene sequencing and quantitative PCR via the dysbiosis index was performed on 16 healthy controls and 18 dogs with chronic enteropathy prior to and 1 month after initiation of individualized therapy. Fasting and post-prandial plasma GLP-2 concentrations were measured via ELISA in healthy dogs and chronic enteropathy dogs at both time points. Alpha and beta diversity indices, as well as bacterial population abundances were compared between groups and time-points. Principal component analysis combined with least squares regression was used to identify taxa contributing to glucagon-like peptide-2 variance among groups. While the dysbiosis index did not differ between healthy dogs and dogs with chronic enteropathy, 16S V4 genomic sequencing identified 47 operational taxonomic units that differed between the groups, all but 2 of which resolved following chronic enteropathy treatment. Principal component analysis identified 6 families and 19 genera that contributed to differences in glucagon-like peptide-2 concentrations between groups. Dysbiosis associated with chronic enteropathies in dogs may contribute to the observed lower plasma glucagon-like peptide-2 concentrations. Further research into mechanisms of microbiota impact on the enteroendocrine system is needed. Association between glucagon-like peptide-2 secretion and microbiome indices may help to guide research into future treatment strategies for dogs with chronic enteropathy.


Asunto(s)
Enfermedades de los Perros , Disbiosis , Microbioma Gastrointestinal , Péptido 2 Similar al Glucagón , Perros , Animales , Péptido 2 Similar al Glucagón/sangre , Disbiosis/veterinaria , Disbiosis/microbiología , Disbiosis/sangre , Enfermedades de los Perros/microbiología , Enfermedades de los Perros/sangre , Femenino , Masculino , Enfermedad Crónica , Estudios Prospectivos , Heces/microbiología , ARN Ribosómico 16S/genética , Enfermedades Intestinales/veterinaria , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/sangre
3.
Life Sci ; 351: 122792, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38857657

RESUMEN

AIMS: Drug-induced enteropathy is often associated with the therapeutic use of certain glucuronidated drugs. One such drug is mycophenolic acid (MPA), a well-established immunosuppressant of which gastrointestinal adverse effects are a major concern. The role of bacterial ß-glucuronidase (ß-G) from the gut microbiota in MPA-induced enteropathy has recently been discovered. Bacterial ß-G hydrolyzes MPAG, the glucuronide metabolite of MPA excreted in the bile, leading to the digestive accumulation of MPA that would favor in turn these adverse events. We therefore hypothesized that taming bacterial ß-G activity might reduce MPA digestive exposure and prevent its toxicity. MAIN METHODS: By using a multiscale approach, we evaluated the effect of increasing concentrations of MPA on intestinal epithelial cells (Caco-2 cell line) viability, proliferation, and migration. Then, we investigated the inhibitory properties of amoxapine, a previously described bacterial ß-G inhibitor, by using molecular dynamics simulations, and evaluated its efficiency in blocking MPAG hydrolysis in an Escherichia coli-based ß-G activity assay. The pharmacological effect of amoxapine was evaluated in a mouse model. KEY FINDINGS: We observed that MPA impairs intestinal epithelial cell homeostasis. Amoxapine efficiently blocks the hydrolysis of MPAG to MPA and significantly reduces digestive exposure to MPA in mice. As a result, administration of amoxapine in MPA-treated mice significantly attenuated gastrointestinal lesions. SIGNIFICANCE: Collectively, these results suggest that the digestive accumulation of MPA is involved in the pathophysiology of MPA-gastrointestinal adverse effects. This study provides a proof-of-concept of the therapeutic potential of bacterial ß-G inhibitors in glucuronidated drug-induced enteropathy.


Asunto(s)
Biotransformación , Microbioma Gastrointestinal , Glucuronidasa , Glucurónidos , Ácido Micofenólico , Ácido Micofenólico/metabolismo , Ácido Micofenólico/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Glucuronidasa/metabolismo , Glucuronidasa/antagonistas & inhibidores , Humanos , Animales , Ratones , Glucurónidos/metabolismo , Células CACO-2 , Masculino , Inmunosupresores/farmacología , Inmunosupresores/toxicidad , Inmunosupresores/metabolismo , Enfermedades Intestinales/inducido químicamente , Enfermedades Intestinales/tratamiento farmacológico , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Proliferación Celular/efectos de los fármacos , Glicoproteínas
5.
Poult Sci ; 103(7): 103757, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38697006

RESUMEN

Stress is known to disrupt the intestinal barrier and induce intestinal dysfunction. A critical role for gonadotropin inhibitory hormone (GnIH) in stress has emerged. However, whether GnIH mediates stress-induced intestinal dysfunction remains unknown. The present study explored this question through in vivo and in vitro experiments in hens. Our in vivo experiments showed that continuous intraperitoneal injection of GnIH not only significantly increased the concentration of stress hormones in serum, but also significantly elevated the mRNA expression of glucocorticoid receptor (GR) in the duodenum and jejunum. Moreover, morphological and molecular analyses revealed that GnIH disrupted the physical and chemical barriers of the intestine and dramatically increased inflammatory factor levels in the intestine and serum of hens. Interestingly, the microbiomics results showed that GnIH altered the structure and composition of the gut flora in the cecum, revealing an increased abundance of harmful intestinal bacteria such as Desulfovibrionaceae. Similar results were found in in vitro studies in which the GnIH-induced intestinal mucosal barrier was disrupted, and inflammation increased in jejunal explants, although no significant difference was found in the expression of GR between the control and GnIH groups. Our results demonstrated that GnIH not only directly damaged intestinal barriers and elevated intestinal inflammation but also mediated stress and microflora imbalance-induced intestinal function disorder, suggesting that GnIH is a potential therapeutic target for gut dysfunction, stress-induced intestinal function disorder, and inflammatory bowel disease in animals and humans.


Asunto(s)
Pollos , Microbioma Gastrointestinal , Estrés Fisiológico , Animales , Pollos/fisiología , Femenino , Microbioma Gastrointestinal/fisiología , Hormonas Hipotalámicas/metabolismo , Hormonas Hipotalámicas/genética , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/fisiopatología , Proteínas Aviares/metabolismo , Proteínas Aviares/genética , Enfermedades Intestinales/veterinaria , Enfermedades Intestinales/microbiología
6.
J Histochem Cytochem ; 72(2): 71-78, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38189179

RESUMEN

Human intestinal spirochetosis (HIS) is a colorectal bacterial infection caused by the Brachyspira species. Griffonia simplicifolia-II (GS-II) is a lectin specific to terminal α/ßGlcNAc residues. Here, we investigated terminal ßGlcNAc residues in the context of HIS infection using GS-II-horseradish peroxidase staining and HIK1083 immunostaining specific to terminal αGlcNAc residues. Fourteen of 15 HIS cases were GS-II-positive on the bacterial body. No cases showed HIK1083 positivity. The percentage of bacterial bodies staining positively for GS-II based on comparison with anti-Treponema immunostaining was ≤30% in seven cases, 30-70% in two, and >70% in six. Of 15 HIS cases analyzed, none were comorbid with tubular adenomas, and three were comorbid with sessile serrated lesions (SSLs). To determine the species of spirochete infected, the B. aalborgi-specific or B. pilosicoli-specific NADPH oxidase genes were amplified by PCR. After direct sequencing of the PCR products, all nine cases in which PCR products were observed were found to be infected with B. aalborgi alone. These results indicate that the HIS bacterial body, especially of B. aalborgi, is characterized by terminal ßGlcNAc and also indicate that terminal ßGlcNAc on the HIS bacterial body is associated with HIS preference for SSLs.


Asunto(s)
Brachyspira , Enfermedades Intestinales , Infecciones por Spirochaetales , Humanos , Brachyspira/genética , Intestinos , Infecciones por Spirochaetales/microbiología , Infecciones por Spirochaetales/patología , Spirochaetales , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/patología
7.
Food Funct ; 15(3): 1265-1278, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38196314

RESUMEN

Nobiletin (NOB) exhibits significant biological activities and may be a potential dietary treatment for antibiotic-associated gut dysbiosis. In this study, mice were gavaged with 0.2 mL day-1 of 12.5 g L-1 cefuroxime (LFX) and 10 g L-1 levofloxacin (LVX) for a duration of 10 days, accompanied by 0.05% NOB to investigate the regulatory effect and potential mechanisms of NOB on antibiotic-induced intestinal microbiota disorder and intestinal barrier dysfunction. Our results indicated that dietary NOB improved the pathology of intestinal epithelial cells and the intestinal permeability by upregulating the expression of intestinal tight junction proteins (TJs) and the number of goblet cells. Furthermore, dietary NOB reduced the levels of serum lipopolysaccharide (LPS) and pro-inflammatory factors (TNF-α and IL-1ß), thereby facilitating the restoration of the intestinal mucosal barrier. Additionally, dietary NOB increased the abundance of beneficial bacteria f_Lachnospiraceae and regulated the metabolic disorders of short-chain fatty acids (SCFAs) and bile acids (BAs). Notably, NOB supplementation resulted in elevated levels of butyric acid and lithocholic acid (LCA), which contributed to the repair of the intestinal mucosal barrier function and the maintenance of intestinal homeostasis. Collectively, our results propose a healthy dietary strategy for the prevention or mitigation of antibiotic-associated gut dysbiosis by dietary NOB.


Asunto(s)
Flavonas , Microbioma Gastrointestinal , Enfermedades Intestinales , Animales , Ratones , Cefuroxima/efectos adversos , Levofloxacino/efectos adversos , Disbiosis/inducido químicamente , Enfermedades Intestinales/microbiología , Antibacterianos/efectos adversos
8.
Appl Microbiol Biotechnol ; 108(1): 139, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38229401

RESUMEN

Gut microorganism (GM) is an integral component of the host microbiome and health system. Abuse of antibiotics disrupts the equilibrium of the microbiome, affecting environmental pathogens and host-associated bacteria alike. However, relatively little research on Bacillus licheniformis alleviates the adverse effects of antibiotics. To test the effect of B. licheniformis as a probiotic supplement against the effects of antibiotics, cefalexin was applied, and the recovery from cefalexin-induced jejunal community disorder and intestinal barrier damage was investigated by pathology, real-time PCR (RT-PCR), and high-throughput sequencing (HTS). The result showed that A group (antibiotic treatment) significantly reduced body weight and decreased the length of jejunal intestinal villi and the villi to crypt (V/C) value, which also caused structural damage to the jejunal mucosa. Meanwhile, antibiotic treatment suppressed the mRNA expression of tight junction proteins ZO-1, claudin, occludin, and Ki67 and elevated MUC2 expression more than the other Groups (P < 0.05 and P < 0.01). However, T group (B. licheniformis supplements after antibiotic treatment) restored the expression of the above genes, and there was no statistically significant difference compared to the control group (P > 0.05). Moreover, the antibiotic treatment increased the relative abundance of 4 bacterial phyla affiliated with 16 bacterial genera in the jejunum community, including the dominant Firmicutes, Proteobacteria, and Cyanobacteria in the jejunum. B. licheniformis supplements after antibiotic treatment reduced the relative abundance of Bacteroidetes and Proteobacteria and increased the relative abundance of Firmicutes, Epsilonbacteraeota, Lactobacillus, and Candidatus Stoquefichus. This study uses mimic real-world exposure scenarios by considering the concentration and duration of exposure relevant to environmental antibiotic contamination levels. We described the post-antibiotic treatment with B. licheniformis could restore intestinal microbiome disorders and repair the intestinal barrier. KEY POINTS: • B. licheniformis post-antibiotics restore gut balance, repair barrier, and aid health • Antibiotics harm the gut barrier, alter structure, and raise disease risk • Long-term antibiotics affect the gut and increase disease susceptibility.


Asunto(s)
Bacillus licheniformis , Enfermedades Intestinales , Probióticos , Animales , Ratones , Bovinos , Antibacterianos/farmacología , Suplementos Dietéticos , Probióticos/farmacología , Enfermedades Intestinales/microbiología , Firmicutes/genética , Cefalexina
9.
Artículo en Inglés | MEDLINE | ID: mdl-37076331

RESUMEN

OBJECTIVES: To describe the clinical features, history and association with intestinal disease in central nervous system (CNS) S. bovis infections. METHODS: Four cases of S. bovis CNS infections from our institution are presented. Additionally a systematic literature review of articles published between 1975 and 2021 in PubMed/MEDLINE was conducted. RESULTS: 52 studies with 65 cases were found; five were excluded because of incomplete data. In total 64 cases were analyzed including our four cases: 55 with meningitis and 9 with intracranial focal infections. Both infections were frequently associated with underlying conditions (70.3%) such as immunosuppression (32.8%) or cancer (10.9%). In 23 cases a biotype was identified, with biotype II being the most frequent (69.6%) and S. pasteurianus the most common within this subgroup. Intestinal diseases were found in 60.9% of cases, most commonly neoplasms (41.0%) and Strongyloides infestation (30.8%). Overall mortality was 17.1%, with a higher rate in focal infection (44.4% vs 12.7%; p=0.001). CONCLUSIONS: CNS infections due to S. bovis are infrequent and the most common clinical form is meningitis. Compared with focal infections, meningitis had a more acute course, was less associated with endocarditis and had a lower mortality. Immunosuppression and intestinal disease were frequent in both infections.


Asunto(s)
Infecciones del Sistema Nervioso Central , Infecciones Estreptocócicas , Streptococcus bovis , Adulto , Humanos , Sistema Nervioso Central , Infecciones del Sistema Nervioso Central/microbiología , Infecciones del Sistema Nervioso Central/patología , Infección Focal/microbiología , Infección Focal/patología , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/patología , Meningitis/microbiología , Meningitis/patología , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/epidemiología , Streptococcus bovis/fisiología
11.
Avian Dis ; 67(2): 177-185, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37556297

RESUMEN

Focal duodenal necrosis (FDN) is a common intestinal disease of table egg layers. In this research we aimed to identify the bacteria commonly found in FDN lesions as seen with histopathological analysis. Fifty-nine ethanol-fixed duodenum samples were collected from egg layers on eight FDN-affected farms, and 42 samples had typical FDN lesions. Excision of bacteria-containing lesions using laser capture microdissection was performed, followed by 16S rRNA gene sequencing of extracted DNA for bacterial identification. Bacterial sequencing analysis revealed no consistent bacterial species identified from samples with FDN. However, analysis of the relative phylum abundance revealed differences in the duodenal microbiota between layers with FDN and healthy birds. There were differences in the abundance of Proteobacteria, Firmicutes, and Actinobacteria between FDN-positive and FDN-negative control samples compatible with intestinal dysbiosis. In addition, 10 duodenal samples with FDN lesions were collected for bacteriological analysis, yielding 47 colonies on tryptone soy agar, MacConkey agar, and blood agar plates. Using 16S rRNA gene PCR, 39/47 (53.8%) colonies were identified as Escherichia coli. PCR for E. coli virulence genes identified 21/39 (53.8%) E. coli isolates as avian pathogenic E. coli-like. PCR analysis for 19 E. coli virulence genes associated with intestinal disease strains including inflammatory bowel disease found 11/39 (28.2%) isolates containing more than 10 of these virulence genes. In conclusion, FDN appears to be a multifactorial inflammatory intestinal disease associated with intestinal dysbiosis, and Gram-negative bacteria including E. coli may contribute to the pathogenesis of this disease.


Microdisección por captura láser, análisis de cultivos y secuenciación bacteriana para evaluar la microbiota de la necrosis duodenal focal en aves de postura de huevo comercial. La necrosis duodenal focal (FDN) es una enfermedad intestinal común en las gallinas de postura de huevo comercial. En esta investigación, el objetivo fue identificar las bacterias que se encuentran comúnmente en las lesiones provocadas por la necrosis duodenal focal tal como se aprecian con el análisis histopatológico. Se recolectaron 59 muestras de duodeno fijadas con etanol de gallinas de postura de ocho granjas afectadas por necrosis duodenal focal, y 42 muestras tenían lesiones típicas de dicha enfermedad. Se realizó la escisión de las lesiones que contenían bacterias mediante microdisección por captura láser, seguida de la secuenciación del gene 16S rRNA del ADN extraído para la identificación bacteriana. El análisis de secuenciación bacteriana no reveló especies bacterianas consistentes identificadas a partir de muestras con necrosis duodenal focal. Sin embargo, el análisis de la abundancia relativa del phylum reveló diferencias en el microbiota duodenal entre gallinas de postura con necrosis duodenal focal y aves sanas. Hubo diferencias en la abundancia de Proteobacteria, Firmicutes y Actinobacteria entre las muestras controles positivas y negativas para la necrosis duodenal focal compatibles con disbiosis intestinal. Además, se recolectaron 10 muestras duodenales con lesiones de la necrosis duodenal focal para análisis bacteriológico, lo que produjo 47 colonias en placas de agar triptona soya, agar MacConkey y agar sangre. Utilizando un método de PCR para el gene 16S rRNA, 39/47 (53.8 %) colonias se identificaron como Escherichia coli. El método de PCR para genes de virulencia de E. coli identificó 21/39 (53.8 %) aislados de E. coli como similares a E. coli patogénica aviar. El análisis de PCR para 19 genes de virulencia de E. coli asociados con cepas que provocan enfermedades intestinales, incluida la enfermedad inflamatoria intestinal, detectó 11/39 (28.2 %) aislados que contenían más de 10 de estos genes de virulencia. En conclusión, la necrosis duodenal focal parece ser una enfermedad intestinal inflamatoria multifactorial asociada con disbiosis intestinal, y las bacterias Gramnegativas, incluida E. coli, pueden contribuir a la patogenia de esta enfermedad.


Asunto(s)
Enfermedades Intestinales , Microbiota , Enfermedades de las Aves de Corral , Animales , Escherichia coli/genética , Captura por Microdisección con Láser/veterinaria , ARN Ribosómico 16S/genética , Disbiosis/patología , Disbiosis/veterinaria , Agar , Enfermedades de las Aves de Corral/microbiología , Duodeno/microbiología , Bacterias , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/veterinaria , Aves , Necrosis/patología , Necrosis/veterinaria
12.
Epidemiol Infect ; 151: e109, 2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37313601

RESUMEN

Infectious intestinal disease (IID) studies conducted at different levels of the surveillance pyramid have found heterogeneity in the association of socioeconomic deprivation with illness. The aim of this study was to analyse the association between socioeconomic deprivation and incidence of IID by certain gastrointestinal pathogens reported to UKHSA. Data were extracted from 2015 to 2018 for Salmonella, Campylobacter, Shigella, Giardia species, and norovirus. Rates were calculated per 100,000 person-years by the index of multiple deprivation quintile, and an ecological analysis was conducted using univariant and multvariable regression models for each pathogen. Incidence of Campylobacter, and Giardia species decreased with increasing deprivation. Conversely, the incidence of norovirus, non-typhoidal Salmonella, Salmonella typhi/paratyphi, Shigella species increased with increasing deprivation. Multivariable analysis results showed that higher deprivation was significantly associated with higher odds of higher number of cases for Shigella flexneri, norovirus and S. typhi/paratyphi. Infections most associated with deprivation were those transmitted by person-to-person spread, and least associated were those transmitted by zoonotic contamination of the environment. Person-to-person transmission can be contained by implementing policies targeting over-crowding and poor hygiene. This approach is likely to be the most effective solution for the reduction of IID.


Asunto(s)
Infecciones Bacterianas , Enfermedades Intestinales , Humanos , Campylobacter , Incidencia , Enfermedades Intestinales/epidemiología , Enfermedades Intestinales/microbiología , Salmonella , Shigella , Factores Socioeconómicos , Reino Unido/epidemiología , Infecciones Bacterianas/epidemiología
13.
Food Funct ; 14(9): 4414-4429, 2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37097253

RESUMEN

5-Demethylnobiletin (5DN) is an important ingredient of citrus extract that is rich in polymethoxyflavones (PMFs). In this study, we systemically investigated the preventive effects of 5DN on antibiotic-associated intestinal disturbances. Experimental mice were gavaged 0.2 mL per day of the antibiotic cocktail (12.5 g L-1 cefuroxime and 10 g L-1 levofloxacin) for 10 days, accompanied by dietary 0.05% 5DN for 10 and 20 days. The results showed that the combination of cefuroxime and levofloxacin caused swelling of the cecum and injury to the colon tissue. Meanwhile, the balance of intestinal oxidative stress and the barrier function of mice was also damaged by the antibiotics through upregulation of the relative mRNA levels of superoxide dismutase 3 (SOD3), quinine oxidoreductase 1 (NQO1) and glutathione peroxidase 1 (GPX1), and downregulation of the relative protein levels of tight junction proteins (TJs). Moreover, antibiotic exposure led to disorder of the gut microbiota, particularly increased harmful bacteria (Proteobacteria) and decreased beneficial bacteria (Bacteroideta). However, dietary 5DN could reduce antibiotic-associated intestinal damage, evidenced by the results that 5DN alleviated gut oxidative damage and attenuated intestinal barrier injury via increasing the expression of TJs including occludin and zonula occluden1 (ZO1). Additionally, dietary 5DN modulated the composition of the gut microbiota in antibiotic-treated mice by increasing the relative levels of beneficial bacteria, such as Dubosiella and Lactobacillus. Moreover, PMFs increased the contents of isobutyric acid and butyric acid, which were almost eliminated by antibiotic exposure. In conclusion, 5DN could alleviate antibiotic-related imbalance of intestinal oxidative stress, barrier function damage, intestinal flora disorders and the reduction of short-chain fatty acids (SCFAs), which lays a foundation for exploring safer and more effective ways to prevent or mitigate antibiotic-associated intestinal damage.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Intestinales , Animales , Ratones , Antibacterianos/efectos adversos , Intestinos/microbiología , Cefuroxima/farmacología , Levofloxacino/farmacología , Disbiosis , Colon , Enfermedades Intestinales/microbiología , Ácido Butírico/farmacología , Bacterias/genética
14.
Front Immunol ; 13: 897395, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35911699

RESUMEN

Intestinal epithelial barrier injury disrupts immune homeostasis and leads to many intestinal disorders. Lactobacillus reuteri (L. reuteri) strains can influence immune system development and intestinal function. However, the underlying mechanisms of L. reuteri LR1 that regulate inflammatory response and intestinal integrity are still unknown. The present study aimed to determine the effects of LR1 on the ETEC K88-induced intestinal epithelial injury on the inflammatory response, intestinal epithelial barrier function, and the MLCK signal pathway and its underlying mechanism. Here, we showed that the 1 × 109 cfu/ml LR1 treatment for 4 h dramatically decreased interleukin-8 (IL-8) and IL-6 expression. Then, the data indicated that the 1 × 108 cfu/ml ETEC K88 treatment for 4 h dramatically enhanced IL-8, IL-6, and tumor necrosis factor-α (TNF-α) expression. Furthermore, scanning electron microscope (SEM) data indicated that pretreatment with LR1 inhibited the ETEC K88 that adhered on IPEC-J2 and alleviated the scratch injury of IPEC J2 cells. Moreover, LR1 pretreatment significantly reversed the declined transepithelial electrical resistance (TER) and tight junction protein level, and enhanced the induction by ETEC K88 treatment. Additionally, LR1 pretreatment dramatically declined IL-8, IL-17A, IL-6, and TNF-α levels compared with the ETEC K88 group. Then, ETEC K88-treated IPEC-J2 cells had a higher level of myosin light-chain kinase (MLCK), higher MLC levels, and a lower Rho-associated kinase (ROCK) level than the control group, while LR1 pretreatment significantly declined the MLCK and MLC expression and enhanced ROCK level in the ETEC K88-challenged IPEC-J2 cells. Mechanistically, depletion of MLCK significantly declined MLC expression in IPEC-J2 challenged with ETEC K88 compared to the si NC+ETEC K88 group. On the other hand, the TER of the si MLCK+ETEC K88 group was higher and the FD4 flux in the si MLCK+ETEC K88 group was lower compared with the si NC+ETEC K88 group. In addition, depletion of MLCK significantly enhanced Claudin-1 level and declined IL-8 and TNF-α levels in IPEC-J2 pretreated with LR1 followed by challenging with ETEC K88. In conclusion, our work indicated that L. reuteri LR1 can decline inflammatory response and improve intestinal epithelial barrier function through suppressing the MLCK signal pathway in the ETEC K88-challenged IPEC-J2.


Asunto(s)
Escherichia coli Enterotoxigénica , Infecciones por Escherichia coli , Mucosa Intestinal , Limosilactobacillus reuteri , Animales , Línea Celular , Escherichia coli Enterotoxigénica/inmunología , Infecciones por Escherichia coli/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Enfermedades Intestinales/microbiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiología , Limosilactobacillus reuteri/fisiología , Quinasa de Cadena Ligera de Miosina/metabolismo , Transducción de Señal , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
15.
PLoS One ; 17(6): e0269676, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35771750

RESUMEN

Symptom profiles have previously been identified for infectious intestinal disease (IID) which distinguish bacterial from viral organisms. However, there is evidence that the seasonality, severity, and duration of IID may differ between children, adults and elderly. A secondary data analysis was undertaken to explore whether symptom profiles for bacterial and viral IID vary across different age groups. Data from 844 cases of IID were divided into three age categories: <16 years, 16-65 years and >65 years. Multivariable logistic regression modelling was used to compare the significance of different symptoms across the three age groups. The odds of bacterial IID in children were increased by onset in the summer, diarrhoea in the absence of vomiting and fever. These symptoms were also associated with lower odds of a viral pathogen. In adults, diarrhoea but no vomiting, bloody diarrhoea and diarrhoea lasting more than 3 days were associated with increased odds of a bacterial organism, whilst onset in the winter or spring and a loss of appetite were associated with viral IID. In the elderly, diarrhoea in the absence of vomiting and diarrhoea lasting more than 3 days were associated with higher odds of bacterial IID and lower odds of a viral cause. Only diarrhoea in the absence of vomiting emerged as a key symptom across all three age groups. Variation in symptom profiles by age has implications for clinicians, public health specialists and epidemiologists who use symptoms to guide presumptive diagnoses in the absence of microbiological confirmation.


Asunto(s)
Infecciones Bacterianas , Enfermedades Intestinales , Infecciones Intraabdominales , Virosis , Adolescente , Adulto , Anciano , Niño , Diarrea/diagnóstico , Humanos , Enfermedades Intestinales/diagnóstico , Enfermedades Intestinales/microbiología , Vómitos
16.
Front Immunol ; 13: 822754, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154141

RESUMEN

Wild pigs usually showed high tolerance and resistance to several diseases in the wild environment, suggesting that the gut bacteria of wild pigs could be a good source for discovering potential probiotic strains. In our study, wild pig feces were sequenced and showed a higher relative abundance of the genus Lactobacillus (43.61% vs. 2.01%) than that in the domestic pig. A total of 11 lactic acid bacteria (LAB) strains including two L. rhamnosus, six L. mucosae, one L. fermentum, one L. delbrueckii, and one Enterococcus faecalis species were isolated. To investigate the synergistic effects of mixed probiotics strains, the mixture of 11 LAB strains from an intestinal ecology system was orally administrated in mice for 3 weeks, then the mice were challenged with Escherichia coli ATCC 25922 (2 × 109 CFU) and euthanized after challenge. Mice administrated with LAB strains showed higher (p < 0.05) LAB counts in feces and ileum. Moreover, alterations of specific bacterial genera occurred, including the higher (p < 0.05) relative abundance of Butyricicoccus and Clostridium IV and the lower (p < 0.05) abundance of Enterorhabdus in mice fed with mixed LAB strains. Mice challenged with Escherichia coli showed vacuolization of the liver, lower GSH in serum, and lower villus to the crypt proportion and Claudin-3 level in the gut. In contrast, administration of mixed LAB strains attenuated inflammation of the liver and gut, especially the lowered IL-6 and IL-1ß levels (p < 0.05) in the gut. Our study highlighted the importance of gut bacterial diversity and the immunomodulation effects of LAB strains mixture from wild pig in gut health.


Asunto(s)
Infecciones por Escherichia coli/terapia , Enfermedades Intestinales/terapia , Lactobacillales/fisiología , Probióticos/farmacología , Animales , Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad/efectos de los fármacos , Enfermedades Intestinales/inmunología , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lactobacillales/aislamiento & purificación , Masculino , Ratones , Ratones Endogámicos C57BL , Probióticos/uso terapéutico , Sus scrofa
17.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054790

RESUMEN

Intestinal homeostasis is a dynamic balance involving the interaction between the host intestinal mucosa, immune barrier, intestinal microecology, nutrients, and metabolites. Once homeostasis is out of balance, it will increase the risk of intestinal diseases and is also closely associated with some systemic diseases. Probiotics (Escherichia coli Nissle 1917, Akkermansia muciniphila, Clostridium butyricum, lactic acid bacteria and Bifidobacterium spp.), maintaining the gut homeostasis through direct interaction with the intestine, can also exist as a specific agent to prevent, alleviate, or cure intestinal-related diseases. With genetic engineering technology advancing, probiotics can also show targeted therapeutic properties. The aims of this review are to summarize the roles of potential native and engineered probiotics in oncology, inflammatory bowel disease, and obesity, discussing the therapeutic applications of these probiotics.


Asunto(s)
Diabetes Mellitus/microbiología , Enfermedades Intestinales/microbiología , Mucosa Intestinal/fisiología , Obesidad/microbiología , Probióticos/uso terapéutico , Akkermansia , Animales , Bifidobacterium , Clostridium butyricum , Diabetes Mellitus/terapia , Escherichia coli , Homeostasis , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Intestinales/terapia , Mucosa Intestinal/microbiología , Lactobacillales , Obesidad/terapia
18.
Dig Dis Sci ; 67(1): 121-133, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33569665

RESUMEN

BACKGROUND: Uric acid (UA) has anti- and pro-inflammatory properties. We previously revealed that elevated serum UA levels provide protection against murine small intestinal injury probably via luminal UA secreted in the small intestine. Luminal UA may act as an antioxidant, preventing microbiota vulnerability to oxidative stress. However, whether luminal UA is increased under hyperuricemia and plays a protective role in a dose-dependent manner as well as the mechanism by which luminal UA exerts its protective effects on enteropathy remains unknown. METHODS: Inosinic acid (IMP) (1000 mg/kg, i.p.) was administered to obtain high serum UA (HUA) and moderate serum UA (500 mg/kg IMP, i.p.) mice. UA concentrations and levels of oxidative stress markers in the serum and intestine were measured. Mice received indomethacin (20 mg/kg, i.p.) to evaluate the effects of UA on indomethacin-induced enteropathy. Reactive oxygen species (ROS) on the ileal mucosa were analyzed. The fecal microbiota of HUA mice was transplanted to investigate its effect on indomethacin-induced enteropathy. RESULTS: IMP increased luminal UA dose-dependently, with higher levels of luminal antioxidant markers. Indomethacin-induced enteropathy was significantly ameliorated in both UA-elevated groups, with decreased indomethacin-induced luminal ROS. The microbiota of HUA mice showed a significant increase in α-diversity and a significant difference in ß-diversity from the control. Fecal microbiota transplantation from HUA mice ameliorated indomethacin-induced enteropathy. CONCLUSIONS: The protective role of luminal UA in intestinal injury is likely exerted via oxidative stress elimination and microbiota composition modulation, preferably for gut immunity. Therefore, enhancing anaerobic conditions using antioxidants is a potential therapeutic target.


Asunto(s)
Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal , Indometacina/farmacología , Intestino Delgado , Ácido Úrico , Animales , Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Enfermedades Intestinales/inducido químicamente , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/terapia , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Ratones , Estrés Oxidativo/efectos de los fármacos , Factores Protectores , Especies Reactivas de Oxígeno/análisis , Resultado del Tratamiento , Ácido Úrico/sangre , Ácido Úrico/metabolismo
19.
Life Sci ; 289: 120243, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34922941

RESUMEN

Intestinal mucositis (IM) is a critical side-effect associated with antineoplastic therapy. Treatment available is only palliative and often not effective. However, alternative therapeutic strategies, such as probiotics, have attracted significant attention due to their immune-modulatory action in several diseases. Thus, the present study aims to elucidate the therapeutic potential of the probiotic strain Bifidobacterium longum 51A in a murine model of mucositis induced by irinotecan. Due to the scarcity of studies on dose-response and viability (probiotic vs paraprobiotic), we first evaluated which dose and cell viability would be most effective in treating mucositis. In this study, the oral pretreatment with viable B. longum 51A at a concentration of 1 × 109 CFU/mL reduced the daily disease activity index (p < 0.01), protected the intestinal architecture, preserved the length of the intestine (p < 0.05), and reduced intestinal permeability (p < 0.01), inflammation, and oxidative damage (p < 0.01) induced by irinotecan. Also, treatment with B. longum 51A increased the production of secretory immunoglobulin A (p < 0.05) in the intestinal fluid of mice with mucositis. Furthermore, B. longum 51A reversed the mucositis-induced increase in Enterobacteriaceae bacterial group in the gut (p < 0.01). In conclusion, these results showed that oral administration of B. longum 51A protects mice against intestinal damage caused by irinotecan, suggesting its use as a potential probiotic in therapy during mucositis.


Asunto(s)
Bifidobacterium longum , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Intestinales , Irinotecán/efectos adversos , Mucositis , Probióticos/farmacología , Animales , Femenino , Enfermedades Intestinales/inducido químicamente , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/terapia , Irinotecán/farmacología , Ratones , Ratones Endogámicos BALB C , Mucositis/inducido químicamente , Mucositis/microbiología , Mucositis/terapia
20.
Microbiol Spectr ; 9(3): e0065421, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34908474

RESUMEN

The present study aimed to explore the protective effects of exogenous catalase (CAT) from microorganisms against lipopolysaccharide (LPS)-induced intestinal injury and its molecular mechanism in weaned pigs. Fifty-four weaned pigs (21 days of age) were randomly allocated to CON, LPS, and LPS+CAT groups. The pigs in CON and LPS groups were fed a basal diet, whereas the pigs in LPS+CAT group fed the basal diet with 2,000 mg/kg CAT supplementation for 35 days. On day 36, six pigs were selected from each group, and LPS and LPS+CAT groups were administered with LPS (50 µg/kg body weight). Meanwhile, CON group was injected with an equivalent amount of sterile saline. Results showed that LPS administration damaged intestinal mucosa morphology and barrier. However, CAT supplementation alleviated the deleterious effects caused by LPS challenge through enhancing intestinal antioxidant capacity which was benefited to decrease proinflammatory cytokines concentrations and suppress enterocyte apoptosis. Besides, LPS-induced gut microbiota dysbiosis was significantly shifted by CAT through decreasing mainly Streptococcus and Escherichia-Shigella. Our study suggested that dietary supplemented with 2,000 mg/kg catalase was conducive to improve intestinal development and protect against LPS-induced intestinal mucosa injury via enhancing intestinal antioxidant capacity and altering microbiota composition in weaned pigs. IMPORTANCE Exogenous CAT derived from microorganisms has been widely used in food, medicine, and other industries. Recent study also found that exogenous CAT supplementation could improve growth performance and antioxidant capacity of weaned pigs. However, it is still unknown that whether dietary exogenous CAT supplementation can provide a defense against the oxidative stress-induced intestinal damage in weaned pigs. Our current study suggested that dietary supplemented with 2,000 mg/kg CAT was conducive to improve intestinal development and protect against LPS-induced intestinal mucosa injury via enhancing intestinal antioxidant capacity and altering microbiota composition in weaned pigs. Moreover, this study will also assist in developing of CAT produced by microorganisms to attenuate various oxidative stress-induced injury or diseases.


Asunto(s)
Antioxidantes/metabolismo , Catalasa/administración & dosificación , Proteínas Fúngicas/administración & dosificación , Enfermedades Intestinales/veterinaria , Intestinos/metabolismo , Penicillium chrysogenum/enzimología , Enfermedades de los Porcinos/tratamiento farmacológico , Animales , Suplementos Dietéticos/análisis , Terapia Enzimática , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Intestinales/tratamiento farmacológico , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Intestinos/efectos de los fármacos , Intestinos/lesiones , Intestinos/microbiología , Lipopolisacáridos/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Penicillium chrysogenum/química , Porcinos , Enfermedades de los Porcinos/etiología , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...