Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35055188

RESUMEN

In a previous study, we uncovered three immune-responsive patterns of gut microbes using an in vitro mesenteric lymph node cell suspension model, abbreviated as the MLN model hereafter. We used Akkermansia muciniphila and Clostridium butyricum as the first group directly inducing an immune response, Bifidobacterium sp. and Bacteroides sp. as the second group evoking an immune response with the help of stimuli (anti-CD3 and anti-CD28 antibodies), and Lactobacillus sp. as the third group blunting the immune response with or without stimuli. Our group previously clarified the immune-activation characteristics of A. muciniphila and linked its in vivo immune induction effect in GF and SPF mice under homeostasis. In the present study, we supplemented the characteristics of C. butyricum and B. bifidum in the in vitro MLN model and addressed the specific elements of the model. Finally, we used an in vivo TNBS-challenge model to show the functional differences between these species with different response patterns in vitro. The results showed that C. butyricum and B. bifidum evoked an immune response in vitro in a dose-dependent and strain-unique manner. Although TLR2, rather than TLR4, is indispensable for immune activation in the present in vitro model, it may not involve interaction between TLR2 and bacterial ligands. Like the PBMC model, the present in vitro MLN model is highly dependent on cell resources and should be given more attention when used to conduct a quantitative comparison. Finally, a mixture of two strong immunogenic strains, A. muciniphila and C. butyricum, significantly increased the mortality of TNBS-challenged (2,4,6-trinitrobenzene sulfonic acid, TNBS) mice, indicating a possible link between the in vitro MLN model and in vivo functional evaluation. However, more evidence is needed to clarify the associations and underlying mechanisms.


Asunto(s)
Bifidobacterium/inmunología , Clostridium butyricum/inmunología , Ganglios Linfáticos/citología , Ácido Trinitrobencenosulfónico/efectos adversos , Animales , Técnicas de Cocultivo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Masculino , Mesenterio , Ratones , Modelos Biológicos , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
2.
Front Immunol ; 12: 771826, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899723

RESUMEN

Clostridium butyricum (CB) can enhance antioxidant capacity and alleviate oxidative damage, but the molecular mechanism by which this occurs remains unclear. This study used enterotoxigenic Escherichia coli (ETEC) K88 as a pathogenic model, and the p62-Keap1-Nrf2 signaling pathway and intestinal microbiota as the starting point to explore the mechanism through which CB alleviates oxidative damage. After pretreatment with CB for 15 d, mice were challenged with ETEC K88 for 24 h. The results suggest that CB pretreatment can dramatically reduce crypt depth (CD) and significantly increase villus height (VH) and VH/CD in the jejunum of ETEC K88-infected mice and relieve morphological lesions of the liver and jejunum. Additionally, compared with ETEC-infected group, pretreatment with 4.4×106 CFU/mL CB can significantly reduce malondialdehyde (MDA) level and dramatically increase superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels in the serum. This pretreatment can also greatly increase the mRNA expression levels of tight junction proteins and genes related to the p62-Keap1-Nrf2 signaling pathway in the liver and jejunum in ETEC K88-infected mice. Meanwhile, 16S rDNA amplicon sequencing revealed that Clostridium disporicum was significantly enriched after ETEC K88 challenge relative to the control group, while Lactobacillus was significantly enriched after 4.4×106 CFU/mL CB treatment. Furthermore, 4.4×106 CFU/mL CB pretreatment increased the short-chain fatty acid (SCFA) contents in the cecum of ETEC K88-infected mice. Moreover, we found that Lachnoclostridium, Roseburia, Lactobacillus, Terrisporobacter, Akkermansia, and Bacteroides are closely related to SCFA contents and oxidative indicators. Taken together, 4.4×106 CFU/mL CB pretreatment can alleviate ETEC K88-induced oxidative damage through activating the p62-Keap1-Nrf2 signaling pathway and remodeling the cecal microbiota community in mice.


Asunto(s)
Antibiosis/inmunología , Infecciones Bacterianas/inmunología , Ciego/microbiología , Clostridium butyricum/inmunología , Escherichia coli Enterotoxigénica/inmunología , Estrés Oxidativo/inmunología , Proteínas/inmunología , Animales , Antibiosis/fisiología , Infecciones Bacterianas/genética , Infecciones Bacterianas/microbiología , Ciego/metabolismo , Clostridium butyricum/fisiología , Escherichia coli Enterotoxigénica/fisiología , Regulación de la Expresión Génica/inmunología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Hemo-Oxigenasa 1/metabolismo , Yeyuno/inmunología , Yeyuno/metabolismo , Yeyuno/microbiología , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/inmunología , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Masculino , Ratones , Microbiota/genética , Microbiota/inmunología , Microbiota/fisiología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/inmunología , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/inmunología , Proteína Sequestosoma-1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Superóxido Dismutasa/genética , Superóxido Dismutasa/inmunología , Superóxido Dismutasa/metabolismo , Porcinos
3.
Gut Microbes ; 13(1): 1-28, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33874858

RESUMEN

Clostridium butyricum is a butyrate-producing human gut symbiont that has been safely used as a probiotic for decades. C. butyricum strains have been investigated for potential protective or ameliorative effects in a wide range of human diseases, including gut-acquired infection, intestinal injury, irritable bowel syndrome, inflammatory bowel disease, neurodegenerative disease, metabolic disease, and colorectal cancer. In this review we summarize the studies on C. butyricum supplementation with special attention to proposed mechanisms for the associated health benefits and the supporting experimental evidence. These mechanisms center on molecular signals (especially butyrate) as well as immunological signals in the digestive system that cascade well beyond the gut to the liver, adipose tissue, brain, and more. The safety of probiotic C. butyricum strains appears well-established. We identify areas where additional human randomized controlled trials would provide valuable further data related to the strains' utility as an intervention.


Asunto(s)
Butiratos/metabolismo , Clostridium butyricum/inmunología , Clostridium butyricum/metabolismo , Inmunidad , Probióticos , Animales , Suplementos Dietéticos , Interacciones Microbiota-Huesped , Humanos , Inflamación/inmunología , Inflamación/microbiología , Síndrome del Colon Irritable/inmunología , Síndrome del Colon Irritable/microbiología , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/microbiología , Neoplasias/inmunología , Neoplasias/microbiología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/microbiología , Simbiosis
4.
Int J Mol Sci ; 21(2)2020 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-31941102

RESUMEN

Previous studies have suggested that immune system development and weaning stress are closely related to the maturation of gut microbiota. The early-life period is a "window of opportunity" for microbial colonization, which potentially has a critical impact on the development of the immune system. Fecal microbiota transplantation (FMT) and probiotics are often used to regulate gut microbial colonization. This study aims to test whether early intervention with FMT using fecal microbiota from gestation sows combined with Clostridium butyricum and Saccharomyces boulardii (FMT-CS) administration could promote the maturation of gut microbiota and development of immune system in piglets. Piglets were assigned to control (n = 84) and FMT-CS treatment (n = 106), which were treated with placebo and bacterial suspension during the first three days after birth, respectively. By 16S rRNA gene sequencing, we found that FMT-CS increased the α-diversity and reduced the unweighted UniFrac distances of the OTU community. Besides, FMT-CS increased the relative abundance of beneficial bacteria, while decreasing that of opportunistic pathogens. FMT-CS also enhanced the relative abundance of genes related to cofactors and vitamin, energy, and amino acid metabolisms during the early-life period. ELISA analysis revealed that FMT-CS gave rise to the plasma concentrations of IL-23, IL-17, and IL-22, as well as the plasma levels of anti-M.hyo and anti-PCV2 antibodies. Furthermore, the FMT-CS-treated piglets showed decreases in inflammation levels and oxidative stress injury, and improvement of intestinal barrier function after weaning as well. Taken together, our results suggest that early-life intervention with FMT-CS could promote the development of innate and adaptive immune system and vaccine efficacy, and subsequently alleviate weaning stress through promoting the maturation of gut microbiota in piglets.


Asunto(s)
Clostridium butyricum/inmunología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/inmunología , Probióticos/farmacología , Saccharomyces boulardii/inmunología , Estrés Fisiológico , Animales , Animales Recién Nacidos , Citocinas/inmunología , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/inmunología , Porcinos , Destete
5.
Sci Rep ; 9(1): 18002, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31784669

RESUMEN

It is known that the day-night shift-rotation has a negative impact on the immune system. The underlying mechanism remains to be further investigated. Probiotics have regulatory effects on immune functions. This study aims to investigate the role of probiotic ingestion in preventing the DEC205+ dendritic cell (decDC) dysfunction in day-night shift-engaging nurses. In this study, blood samples were collected from day-night shift-rotating nurses who took or did not take yogurt (containing C. Butyricum) during the night shift (NS). decDC functions were evaluated with pertinent immunological approaches. We observed that the immune tolerogenic functions and interleukin (IL)-10 expression were impaired in decDCs of nurses after NS. HDAC11 was detected in decDCs that was markedly up regulated after NS. The HDAC11 levels were negatively correlated with the immune tolerogenic functions in decDCs. Ingestion of probiotic-containing yogurt during NS efficiently suppressed Bmal1 and HDAC11 levels as well as up regulated the immune regulatory functions in decDCs. In conclusion, NS has a negative impact on decDC immune tolerogenic functions, which can be prevented by ingesting probiotics-containing yogurt during NS.


Asunto(s)
Células Dendríticas/inmunología , Enfermeras y Enfermeros , Probióticos/administración & dosificación , Horario de Trabajo por Turnos/efectos adversos , Yogur/microbiología , Adulto , Antígenos CD/metabolismo , Ritmo Circadiano/inmunología , Clostridium butyricum/inmunología , Células Dendríticas/metabolismo , Femenino , Histona Desacetilasas/metabolismo , Humanos , Interleucina-10/metabolismo , Lectinas Tipo C/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Receptores de Superficie Celular/metabolismo
6.
World J Gastroenterol ; 25(36): 5469-5482, 2019 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-31576093

RESUMEN

BACKGROUND: Irritable bowel syndrome (IBS) is one of the most common functional gas-troenterological diseases characterized by abnormal visceral sensitivity and low-grade inflammation. The role of Clostridium butyricum (C. butyricum) in reducing intestinal low-grade inflammation via immune pathways has been well defined. However, the detailed mechanisms of the effects of C. butyricum on intestinal mucosal immunity, especially on immune cells of the lamina propria, remain unclear. Dendritic cells (DCs), which are important immune cells, secrete proinflammatory cytokines (IL-1ß, IL-6, and others) and express T cell immuno-globulin and mucin domain-3 (TIM3), promoting proliferation and activation of DCs, and mediating Th1 and Th17 inflammatory responses. AIM: To investigate the role of DCs in the development of IBS in a rat model and to understand the regulation of DCs after C. butyricum intervention. METHODS: An IBS animal model was established using C57BL/6 mice, and C. butyricum was continuously administered via the intragastric route to simulate different intestinal immune states. Intestinal visceral hypersensitivity and histopathology were assessed using the abdominal withdrawal reflex (AWR) test and hematoxylin & eosin (H&E) staining, respectively. The expression of proinflammatory cytokines (IL-1ß and IL-6) and TIM3 was analyzed by Western blot analysis and real-time PCR. Flow cytometry was applied to analyze the quantity, function, and membrane molecule TIM3 of the lamina propria dendritic cells (LPDCs). The regulatory effect of C. butyricum was verified in bone marrow-derived dendritic cells by in vitro experiments. RESULTS: The secretion of proinflammatory cytokines (IL-1ß and IL-6) in mice with IBS was significantly increased compared with that of the control group, which suggested that the intestinal mucosa in mice with IBS was in a low-grade inflammatory state. The expression of CD11C+CD80+ and CD11c+TIM3+ in intestinal LPDCs in mice with IBS increased significantly. Meanwhile, the cytokines (IL-1ß and IL-6) were significantly reduced after the intervention with probiotic C. butyricum. The amount and function of LPDCs and the TIM3 on the surface of the LPDCs were decreased with the alleviation of the intestinal inflammatory response. CONCLUSION: The results suggest that C. butyricum regulates the amount and functional status of LPDCs in the intestinal mucosa of mice with IBS, and therefore modulates the local immune response in the intestine.


Asunto(s)
Clostridium butyricum/inmunología , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Síndrome del Colon Irritable/terapia , Probióticos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Humanos , Inmunidad Mucosa , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Síndrome del Colon Irritable/inducido químicamente , Síndrome del Colon Irritable/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento , Ácido Trinitrobencenosulfónico/toxicidad
7.
Front Immunol ; 10: 1662, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31428083

RESUMEN

Gut microbiota has been proposed as an important environmental factor which can intervene and modulate central nervous system autoimmunity. Here, we altered the composition of gut flora with Clostridium butyricum and norfloxacin in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. We found that appropriate C. butyricum (5.0 × 106 CFU/mL intragastrically daily, staring at weaning period of age) and norfloxacin (5 mg/kg intragastrically daily, 1 week prior to EAE induction) treatment could both ameliorate EAE although there are obvious differences in gut microbiota composition between these two interventions. C. butyricum increased while norfloxacin decreased the abundance and diversity of the gut microbiota in EAE mice, and both of the treatments decreased firmicutes/bacteroidetes ratio. In the genus level, C. butyricum treatment increased the abundance of Prevotella while Akkermansia and Allobaculum increased in norfloxacin treatment. Moreover, both interventions reduced Desulfovibroneceae and Ruminococcus species. Although there was discrepancy in the gut microbiota composition with the two interventions, C. butyricum and norfloxacin treatment both reduced Th17 response and increased Treg response in the gastrointestinal tract and extra-gastrointestinal organ systems in EAE mice. And the reduced activity of p38 mitogen-activated kinase and c-Jun N-terminal kinase signaling in spinal cord could be observed in the two interventions. The results suggested that manipulation of gut microbiota interventions should take factors such as timing, duration, and dosage into consideration. The discrepancy in the gut microbiota composition and the similar protective T cells response of C. butyricum and norfloxacin implies that achieving intestinal microecology balance by promoting and/or inhibiting the gut microbiota contribute to the well-being of immune response in EAE mice.


Asunto(s)
Clostridium butyricum , Encefalomielitis Autoinmune Experimental/terapia , Microbioma Gastrointestinal/inmunología , Norfloxacino/uso terapéutico , Probióticos/uso terapéutico , Animales , Bacteroidaceae/efectos de los fármacos , Bacteroidaceae/aislamiento & purificación , Clostridium butyricum/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Firmicutes/efectos de los fármacos , Firmicutes/aislamiento & purificación , Microbioma Gastrointestinal/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/análisis , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/análisis , Norfloxacino/farmacología , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Organismos Libres de Patógenos Específicos , Médula Espinal/enzimología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/análisis
8.
J Dig Dis ; 20(3): 143-151, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30663229

RESUMEN

OBJECTIVE: This study aimed to determine the effect of corticotropin-releasing factor (CRF) on regulating the NOD-like receptor pyrin domain-containing protein 6 (NLRP6)-inflammatory cytokine axis in a murine model of irritable bowel syndrome (IBS). METHODS: C57BL/6 mice were subjected to water avoidance stress (WAS) for 1 h per day for 10 days, and the abdominal withdrawal reflex (AWR) and colonic inflammation were assessed. We also measured the levels of CRF, NLRP6 inflammasome components, myeloperoxidase, D-lactate, interleukin (IL)-1ß, and IL-18. In vitro experiments with Caco-2 cell line were also performed. In addition, we assessed the effect of Clostridium butyricum (C. butyricum) on IBS mice. RESULTS: IBS mice exhibited visceral hypersensitivity and inflammation, accompanied by increases in CRF, myeloperoxidase, D-lactate, IL-1ß, and IL-18 levels, but a decrease in NLRP6 expression. In vitro data showed that CRF suppressed NLRP6, but induced IL-1ß and IL-18 levels, in Caco-2 cells. C. butyricum restored CRF levels and maintained the NLRP6-inflammatory cytokine axis in IBS mice. CONCLUSIONS: CRF induces the NLRP6-inflammatory cytokine axis in IBS mice. C. butyricum could be beneficial in controlling IBS.


Asunto(s)
Clostridium butyricum/inmunología , Hormona Liberadora de Corticotropina/inmunología , Citocinas/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Síndrome del Colon Irritable/inmunología , Animales , Células CACO-2 , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Síndrome del Colon Irritable/microbiología , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Transducción de Señal
9.
J Gastroenterol Hepatol ; 32(9): 1640-1648, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28109017

RESUMEN

BACKGROUND AND AIM: Enterohepatic immunologic derangement is associated with non-alcoholic steatohepatitis. Here, we investigated whether Clostridium butyricum B1 (CB) would be an effective immune-targeted substance to attenuate steatohepatitis in mice. METHODS: Thirty mice were randomized into a control group fed with common forage, a high-fat diet (HFD) group fed an HFD for 16 weeks, and an HFD + CB group treated with CB for the latter 8 weeks. Inflammation-associated or metabolism-associated genes in the liver or epididymal fat tissue were quantified; intrahepatic and intestinal immune factors were detected. Further short-chain fatty acids in the cecal contents or liver were measured, and differentiations of T cells in vitro were analyzed. RESULTS: Characteristics of non-alcoholic steatohepatitis in the HFD group were obvious and were significantly attenuated in the HFD + CB group. The messenger RNA levels of monocyte chemotactic protein-1 and tumor necrosis factor-α in the liver and epididymal fat tissue were increased in the HFD group compared with the control group and were downregulated in the HFD + CB group. Intrahepatic and intestinal interferon-γ and interleukin (IL)-17 were significantly increased, whereas forkhead box P3, IL-4, and IL-22 were significantly decreased in the HFD group compared with the control group. However, these intrahepatic or intestinal immune changes were reversed after CB intervention. Furthermore, butyrate in the cecal content and liver of the HFD + CB group was significantly elevated. An in vitro investigation showed that sodium butyrate promoted CD4+ T cell differentiation into Th2, Th22, or Treg, whereas it inhibited CD4+ T cell differentiation into Th1 or Th17 under a cytokine milieu, which was mimicked by Trichostatin A. CONCLUSION: Clostridium butyricum B1 could attenuate HFD-induced steatohepatitis in mice partially through butyrate-induced enterohepatic immunoregulation.


Asunto(s)
Clostridium butyricum/inmunología , Dieta Alta en Grasa/efectos adversos , Hígado Graso/etiología , Hígado Graso/terapia , Intestinos/inmunología , Hígado/inmunología , Animales , Butiratos/metabolismo , Linfocitos T CD4-Positivos , Diferenciación Celular , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Ácidos Grasos Volátiles/metabolismo , Hígado Graso/inmunología , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
10.
Microb Pathog ; 100: 112-118, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27622346

RESUMEN

BACKGROUND & AIMS: Probiotics is recognized to promote growth performance and immune function via balancing the intestinal microflora. Live clostridium butyricum and bifidobacterium combined powder (LCBBCP) has been widely to treat intestinal dysbacteriosis in newborns in China. This study was undertaken to investigate the effects of the combined probiotics on the expression of B and T lymphocyte attenuator (BTLA) on CD4+ T cells and the differentiation of lymphocyte subsets in late preterm infants. METHODS: Eighty eligible late preterm infants were equally randomized into LCBBCP therapy group (oral LCBBCP dissolved in formula milk before intake) and control group (treated with simple formula milk for preterm infants) by random digit table. Flow cytometry was used to determine the expression level of BTLA on CD4+ T cells and the percentage of individual subpopulation of lymphocytes in peripheral-blood mononuclear cells (PBMCs) obtained from the late preterm infants in both groups. RESULTS: BTLA protein expression on CD4+ T cells showed no significant change in LCBBCP therapy group before and after intervention, yet was rapidly and significantly down-regulated in the controls. The percentage of increased CD4+ T cells, decreased CD8+ T cells and increased ratio of CD4+/CD8+ T cell proportion were seen in both groups after treatment, yet the increasing or decreasing extent in LCBBCP therapy group was more obvious than in control group. The proportion of NK cells and B lymphocytes remained no significant difference between the two groups before and after therapy. CONCLUSIONS: LCBBCP appears capable of facilitating the activation, proliferation and differentiation of T lymphocytes, which is beneficial to improving immunity in late preterm infants. The continuous high expression of BTLA on CD4+ T cells in LCBBCP therapy group may be involved in the inhibiting of excessive activation of T lymphocytes. Our findings may lay a basis for further clinical evaluation of the efficacies and wider clinical recommendation of probiotics containing live clostridium butyricum and bifidobacterium for late preterm infants.


Asunto(s)
Bifidobacterium/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Diferenciación Celular , Clostridium butyricum/inmunología , Probióticos/administración & dosificación , Receptores Inmunológicos/análisis , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/fisiología , Proliferación Celular , China , Citometría de Flujo , Humanos , Recién Nacido , Recien Nacido Prematuro
11.
PLoS One ; 11(4): e0154373, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27123997

RESUMEN

Obesity is associated with a cluster of metabolic disorders and systemic low-grade inflammation involving multiple organs. Recent findings have suggested that intestine is a key organ altered in response to high fat diet (HFD) feeding. Probiotics mainly lactobacillus strains have earlier been implicated in alleviating metabolic disorders. Here we aimed to examine the effects of a naturally occurring butyrate-producing probiotic clostridium butyricum CGMCC0313.1 (CB0313.1) in limiting the development of HFD-induced obesity. Mice treated with CB0313.1 exhibited reduced lipid accumulation in liver and serum, lower circulating insulin levels and improved glucose tolerance and insulin sensitivity. Furthermore, CB0313.1 administration reversed the HFD-induced colonic inflammation as evidenced by reduced tumor necrosis factor (TNF)-α level and increases the interleukin (IL)-10 and IL-22 levels in colon tissue. Additionally to colonic inflammation, CB0313.1 also reduced the colon permeability by upregulating the tight junction (TJ) proteins (claudin-1 and occludin) and contributed to a decreased circulating endotoxin level. In colon content, CB0313.1 administration restored the reduced production of butyrate and other short chain fatty acids (SCFAs) caused by HFD feeding. In adipose tissue, lower transcriptional levels of pro-inflammatory TNF-α, IL-6, IL-1ß and monocyte chemotactic protein (MCP)-1 in adipose tissue were observed in CB0313.1-treated mice. Collectively, our data demonstrated that CB0313.1, targeting colon inflammation and permeability, ameliorated HFD-induced obesity, insulin resistance as well as adipose inflammation.


Asunto(s)
Clostridium butyricum/inmunología , Colon/efectos de los fármacos , Ácidos Grasos Volátiles/sangre , Resistencia a la Insulina/inmunología , Obesidad/dietoterapia , Probióticos/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Claudina-1/genética , Claudina-1/inmunología , Colon/inmunología , Colon/metabolismo , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Endotoxinas/sangre , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Homeostasis/efectos de los fármacos , Homeostasis/inmunología , Inmunidad Innata , Insulina/sangre , Interleucina-10/genética , Interleucina-10/inmunología , Interleucinas/genética , Interleucinas/inmunología , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/inmunología , Obesidad/patología , Ocludina/genética , Ocludina/inmunología , Permeabilidad , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Interleucina-22
12.
Cell Mol Immunol ; 13(1): 110-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25683610

RESUMEN

The disruption of epithelial barrier integrity is an important factor in the pathogenesis of various immune disorders. However, the restitution of the compromised barrier functions is difficult. This study investigates the regulation of TWIK-related potassium channel-1 (Trek1) in the restitution of intestinal epithelial barrier functions. The human colon epithelial cell line T84 was cultured in monolayers and used to observe epithelial barrier functions in vitro. An intestinal allergy mouse model was created. Cytokine levels were determined by enzyme-linked immunosorbent assay and western blotting. The results showed that Trek1 deficiency induced T84 monolayer barrier disruption. Allergic responses markedly suppressed the expression of Trek1 in the intestinal epithelia via activating the mitogen-activated protein kinase pathways and increasing the expression of histone deacetylase-1. The inhibition of histone deacetylase-1 by sodium butyrate or the administration of a butyrate-producing probiotic (Clostridium butyricum) restored the intestinal epithelial barrier functions and markedly enhanced the effect of antigen-specific immunotherapy. The data suggest that Trek1 is required for the maintenance of intestinal epithelial barrier integrity. Allergic responses induce an insufficiency of Trek1 expression in the intestinal epithelia. Trek1 expression facilitates the restoration of intestinal epithelial barrier functions in an allergic environment.


Asunto(s)
Ácido Butírico/farmacología , Clostridium butyricum/inmunología , Células Epiteliales/inmunología , Hipersensibilidad/terapia , Mucosa Intestinal/inmunología , Canales de Potasio de Dominio Poro en Tándem/inmunología , Probióticos/farmacología , Animales , Línea Celular , Citocinas/genética , Citocinas/inmunología , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Regulación de la Expresión Génica , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/inmunología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Mastocitos/patología , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , Ovalbúmina , Canales de Potasio de Dominio Poro en Tándem/agonistas , Canales de Potasio de Dominio Poro en Tándem/genética , Transducción de Señal
13.
Immunity ; 43(1): 65-79, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26141582

RESUMEN

Colonization with a mixture of Clostridium species has been shown to induce accumulation of induced regulatory T (iTreg) cells in the colon. Transforming growth factor-ß (TGF-ß) is an essential factor for iTreg cell induction; however, the relationship between Clostridium species and TGF-ß remains to be clarified. Here we demonstrated that a gram-positive probiotic bacterial strain, Clostridium butyricum (C. butyricum), promoted iTreg cell generation in the intestine through induction of TGF-ß1 from lamina propria dendritic cells (LPDCs). C. butyricum-mediated TGF-ß1 induction was mainly Toll-like receptor 2 (TLR2) dependent, and the ERK-AP-1 kinase pathway played an important role. In addition, the autocrine TGF-ß-Smad3 transcription factor signal was necessary for robust TGF-ß expression in DCs, whereas Smad2 negatively regulated TGF-ß expression. Smad2-deficient DCs expressed higher concentrations of TGF-ß and were tolerogenic for colitis models. This study reveals a novel mechanism of TGF-ß induction by Clostridia through a cooperation between TLR2-AP-1 and TGF-ß-Smad signaling pathways.


Asunto(s)
Clostridium butyricum/inmunología , Células Dendríticas/inmunología , Proteína Smad2/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/biosíntesis , Animales , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/microbiología , Colitis/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Regiones Promotoras Genéticas/genética , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 2/inmunología , Factor de Transcripción AP-1/inmunología , Factor de Crecimiento Transformador beta1/genética
14.
J Innate Immun ; 7(6): 601-11, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25998580

RESUMEN

Monocytes (Mos) play an important role in the pathogenesis of intestinal mucosal inflammation. This study aims to investigate the mechanism by which the intestinal epithelial cell-derived thrombospondin 1 (TSP1) modulates Mo properties and regulates intestinal inflammatory responses. In this study, the production of TSP1 by intestinal epithelial cells was evaluated by quantitative real-time PCR and Western blotting. The properties of Mos were analyzed by flow cytometry. A mouse model of colitis was created to assess the role of epithelium-derived TSP1 in the suppression of intestinal inflammation. The results demonstrated that mouse intestinal epithelial cells (IECs) expressed TSP1, which was markedly upregulated by butyrate or feeding with Clostridium butyricum. Coculture of the butyrate-primed IECs and Mos or exposure of Mos to TSP1 in the culture induced the expression of transforming growth factor (TGF)-ß in Mos. These TGF-ß+ Mos had tolerogenic properties that could promote generation of inducible regulatory T cells. Adoptive transfer with TSP1-primed Mos, or feeding C. butyricum could prevent experimental colitis in mice. In summary, C. butyricum induces intestinal epithelial cells to produce TSP1 and induces TGF-ß+ Mos, which further suppress experimental colitis in mice. The results implicate that the administration of C. butyricum or butyrate may have the potential to ameliorate chronic intestinal inflammation through inducing immunosuppressive Mos.


Asunto(s)
Colitis/inmunología , Mucosa Intestinal/inmunología , Monocitos/inmunología , Trombospondina 1/inmunología , Animales , Butiratos/toxicidad , Clostridium butyricum/inmunología , Colitis/genética , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados , Monocitos/patología , Trombospondina 1/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
15.
Cell Host Microbe ; 13(6): 711-22, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23768495

RESUMEN

Imbalance in gut bacterial composition provokes host proinflammatory responses causing diseases such as colitis. Colonization with a mixture of Clostridium species from clusters IV and XIVa was shown to suppress colitis through the induction of IL-10-producing regulatory T (Treg) cells. We demonstrate that a distinct Clostridium strain from cluster I, Clostridium butyricum (CB), prevents acute experimental colitis in mice through induction of IL-10, an anti-inflammatory cytokine. However, while CB treatment had no effect on IL-10 production by T cells, IL-10-producing F4/80(+)CD11b(+)CD11c(int) macrophages accumulated in the inflamed mucosa after CB treatment. CB directly triggered IL-10 production by intestinal macrophages in inflamed mucosa via the TLR2/MyD88 pathway. The colitis-preventing effect of CB was negated in macrophage-specific IL-10-deficient mice, suggesting that induction of IL-10 by intestinal macrophages is crucial for the probiotic action of CB. Collectively, CB promotes IL-10 production by intestinal macrophages in inflamed mucosa, thereby preventing experimental colitis in mice.


Asunto(s)
Infecciones por Clostridium/inmunología , Clostridium butyricum/inmunología , Colitis/inmunología , Interleucina-10/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Animales , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/prevención & control , Colitis/microbiología , Colitis/prevención & control , Modelos Animales de Enfermedad , Mucosa Intestinal/inmunología , Ratones , Ratones Noqueados
16.
Jpn J Infect Dis ; 66(1): 46-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23429085

RESUMEN

A mouse-human chimeric antibody that can neutralize botulinum neurotoxin serotype E (BoNT/E) was developed. Variable regions of heavy and light chains obtained using a mouse hybridoma clone (E9-4) cDNA, which was selected on the basis of neutralizing activity against BoNT/E, were fused with the upstream regions of the constant counterparts of human kappa light and gamma 1 heavy chain genes, respectively. CHO-DG44 cells were transfected with these plasmids and a mouse-human chimeric antibody (EC94) was purified to examine binding and neutralizing activity against BoNT/E. EC94 exhibited the same levels of binding activities against BoNT/E as those of a parent mouse monoclonal antibody and neutralized more than 4,000 LD(50)/mg antibody. This chimeric antibody seems to be a useful candidate for infant botulism in which the use of passive immunotherapy is not planned so as to avoid serious events such as anaphylactic shock. We designed shuffling chimeric antibodies with replacement of V(H) or V(L) of EC94 with that of a chimeric antibody (AC24) that possessed neutralizing activity against BoNT/A. These shuffling antibodies did not exhibit neutralizing activity against either BoNT/E or BoNT/A.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Neutralizantes/biosíntesis , Toxinas Botulínicas/inmunología , Botulismo/prevención & control , Clostridium botulinum tipo E/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Botulismo/inmunología , Células Cultivadas , Quimera , Clostridium botulinum/inmunología , Clostridium botulinum/patogenicidad , Clostridium butyricum/inmunología , Clostridium butyricum/patogenicidad , Humanos , Hibridomas , Cadenas gamma de Inmunoglobulina/genética , Cadenas gamma de Inmunoglobulina/inmunología , Cadenas kappa de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Neurotoxinas/inmunología , Pruebas de Neutralización , Proteínas Recombinantes de Fusión
17.
BMC Microbiol ; 12: 100, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22681958

RESUMEN

BACKGROUND: Clostridium butyricum has become increasingly important in preventing and treating intestinal inflammation. In the intestine it may increase the resistance of the gut to pathogen invasion via inducing the secretion of anti-inflammatory cytokines. Interleukin 10 (IL-10) plays a central role in preventing certain inflammatory diseases by down-regulating inflammatory cascades. In a previous study, we observed that the level of IL-10 mRNA was modulated by C. butyricum. The aim of this study was to investigate whether C. butyricum achieves its beneficial effects through IL-10. RESULTS: We treated HT-29 cells with anti-IL-10 (IL-10 antibody) or siIL-10 (IL-10 small interfering RNA) to disrupt IL-10. In both cases, the effects of C. butyricum-induced NF-κB activation and IL-8 expression were enhanced. We also found that neutralization or knockdown of IL-10 could induce apoptosis and necrosis of HT-29 cells treated with C. butyricum compared with control cells. CONCLUSIONS: These findings show that IL-10 serves an important role in C. butyricum-mediated immune protection, and in host recognition of C. butyricum.


Asunto(s)
Clostridium butyricum/inmunología , Clostridium butyricum/patogenicidad , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Interleucina-10/inmunología , Apoptosis , Células HT29 , Humanos
18.
Mol Cell Biochem ; 361(1-2): 31-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21956671

RESUMEN

Oral administration of Clostridium butyricum as probiotic is increasingly gaining importance in the treatment of diarrhea and the improvement of animal performance. However, the mechanisms of host cell receptor recognition of C. butyricum and the downstream immune signaling pathways leading to these benefits remain unclear. The objective of this study was to analyze the mechanisms involved in C. butyricum induction of the toll-like receptor (TLR) signaling. Knockdown of myeloid differentiation primary response protein 88 (MyD88) expression using small interfering RNA in this manner did not affect C. butyricum-induced elevated levels of nuclear factor κB (NF-κB), interleukin-8 (IL-8), IL-6, and tumor necrosis factor alpha (TNF-α), suggesting a MyD88-independent route to TLR signaling transduction. However, a significant reduction in the levels of NF-κB, IL-8, IL-6, and TNF-α was evident in the absence of TLR2 expression, implying the need for TLR2 in C. butyricum recognition. Hence, C. butyricum activates TLR2-mediated MyD88-independent signaling pathway in human epithelial cells, which adds to our understanding of the molecular mechanisms of this probiotic action on gut epithelium.


Asunto(s)
Clostridium butyricum/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Citocinas/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Quinasa I-kappa B/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Factor 88 de Diferenciación Mieloide/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Probióticos , Proteolisis , Interferencia de ARN , Receptor Toll-Like 2/genética
19.
J Med Microbiol ; 60(Pt 11): 1617-1625, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21700738

RESUMEN

In contrast to most modern pharmaceuticals, probiotics are used in many parts of the world with little or no research data on the complex system of interactions that each strain may elicit in the human body. Research on probiotics has recently become more significant, as probiotics have begun to be prescribed by clinicians as an alternative for some gut infections, especially when antibiotics are contraindicated. This study attempted to elucidate the inhibitory interaction between the Japanese probiotic strain Clostridium butyricum MIYAIRI 588 (CBM588) and the hospital pathogen Clostridium difficile, which is responsible for a large proportion of antibiotic-associated diarrhoea and colitis. CBM588 has previously shown effectiveness against C. difficile in vivo, and here it was found that the toxicity of C. difficile in in vitro co-culture with CBM588 was greatly decreased or absent. This was dependent on the inoculation ratio and was not accounted for by the small degree of growth and mRNA inhibition observed. CBM588 and its cell-free supernatant also had no effect on toxin already secreted into the culture medium, and culture of the two strains separated by a semi-permeable membrane resulted in loss of the inhibition. Therefore, it was concluded that the detoxification probably occurred by the inhibition of toxin protein production and that this required close proximity or contact between the two species. The low-pH conditions caused by organic acid secretion were also observed to have inhibitory effects on C. difficile growth, metabolism and toxicity.


Asunto(s)
Clostridioides difficile/crecimiento & desarrollo , Clostridium butyricum/inmunología , Enterotoxinas/inmunología , Probióticos/farmacología , Animales , Western Blotting , Supervivencia Celular/inmunología , Chlorocebus aethiops , Clostridioides difficile/inmunología , Técnicas de Cocultivo , Enterotoxinas/genética , Humanos , ARN Bacteriano/química , ARN Bacteriano/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero
20.
Pediatr Infect Dis J ; 29(2): 135-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20135748

RESUMEN

BACKGROUND: Acute infectious diarrhea is a major cause of childhood morbidity and economic burden for families. We evaluate the clinical, microbiologic, and immunologic effects of probiotics in acute infectious diarrhea. METHODS: Children (n = 304) aged 3 months to 6 years hospitalized for acute diarrhea were randomized to receive Bio-three (a mixture of Bacillus mesentericus, Enterococcus faecalis, and Clostridium butyricum) or placebo orally 3 times daily for 7 days. Fecal samples were homogenized for bacterial culture and blood cells were isolated for cell culture and cytokine analysis. This study is registered (NCT00463190). RESULTS: The mean duration of diarrhea after start of therapy was 60.1 hours in the probiotics group versus 86.3 hours in the placebo group (P = 0.003). Hospital stay was shorter in the probiotics group than in the placebo group (P = 0.009). Counts of Bifidobacteria and Lactobacillus species were elevated in stool culture of the probiotics (Bio-three) group. IL-10 was increased in the serum and supernatants of cell culture in the probiotics group, and tumor necrosis factor-alpha values were down-regulated. Interferon- gamma and IL-12 were mildly elevated in the probiotics group, compared with the placebo group. CONCLUSIONS: This probiotics mixture reduced the severity of diarrhea and length of hospital stay in children with acute diarrhea. In addition to restoring beneficial intestinal flora, probiotics may enhance host protective immunity such as down-regulation of pro-inflammatory cytokines and up-regulation of anti-inflammatory cytokines.


Asunto(s)
Diarrea/terapia , Probióticos/uso terapéutico , Antibiosis , Niño , Preescolar , Clostridium butyricum/crecimiento & desarrollo , Clostridium butyricum/inmunología , Citocinas/análisis , Enterococcus faecalis/crecimiento & desarrollo , Enterococcus faecalis/inmunología , Heces/química , Heces/microbiología , Femenino , Humanos , Lactante , Lactobacillus/crecimiento & desarrollo , Lactobacillus/inmunología , Tiempo de Internación , Masculino , Placebos/administración & dosificación , Probióticos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...