Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nutrients ; 14(10)2022 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-35631220

RESUMEN

Gut microbiota dysbiosis is strongly associated with psychiatric disorders and inflammatory bowel disease (IBD). Herein, we examined whether the fecal microbiota of IBD patients with depression (IBDD) and their gut microbiota culture (iGm) could cause depression and colitis in mice and anti-inflammatory probiotics could mitigate depression in iGm-transplanted or immobilization stress (IS)-exposed mice. Fecal microbiota transplantation (FMT) from IBDD patients, which exhibited Enterobacteriaceae-rich gut microbiota, and its gut microbiota culture (iGm) increased depression-like behaviors in mice. Their treatments heightened the blood lipopolysaccharide (LPS) level and colonic IL-1ß and IL-6 expression. However, FMT from healthy volunteers or sulfasalazine treatment alleviated cGm-induced depressive-like behaviors and hippocampal and colonic inflammation in mice. Moreover, oral administration of Lactobacillus plantarum NK151, Bifidobacterium longum NK173, and Bifidobacterium bifidum NK175, which inhibited LPS-induced IL-6 expression in macrophages, alleviated cGm-induced depression-like behaviors, hippocampal NF-κB+Iba1+ cell numbers and IL-1ß and IL-6 expression, blood LPS, IL-6, and creatinine levels, and colonic NF-κB+CD11c+ number and IL-1ß and IL-6 expression in mice. Treatment with NK151, NK173, or NK175 mitigated immobilization stress (IS)-induced depressive-like behaviors, neuroinflammation, and gut inflammation in mice. NK151, NK173, or NK175 also decreased IS-induced blood LPS, IL-6, and creatinine levels. The transplantation of Enterobacteriaceae-rich gut microbiota can cause depression and colitis, as IS exposure, and anti-inflammatory NK151, NK173, and NK175, may alleviate stress-induced fatigue, depression, and colitis by regulating the expression of proinflammatory and anti-inflammatory cytokines through the suppression of gut bacterial LPS.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Probióticos , Animales , Antiinflamatorios , Colitis/inducido químicamente , Colitis/microbiología , Colitis/terapia , Creatinina , Depresión/psicología , Depresión/terapia , Humanos , Inmunoglobulina M , Inflamación/inducido químicamente , Inflamación/terapia , Interleucina-6 , Lipopolisacáridos , Ratones , FN-kappa B/metabolismo
2.
Sci Rep ; 12(1): 9389, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672451

RESUMEN

Gut dysbiosis is closely associated with the outbreak of inflammatory bowel disease (IBD) and psychiatric disorder. The Enterobacteriaceae population was higher in the feces of patients with inflammatory bowel disease (IBD-F) than in those of healthy control volunteers (HC-F). The Enterococcaceae and Lactobacillaceae populations were higher in the feces of IBD patients with depression (IBD/D+-F) vs. the feces of IBD patients without depression (IBD/D--F). Therefore, we examined the effects of Klebsiella oxytoca, Escherichia coli, Cronobacter sakazakii, Enterococcus faecium, and Pediococcus acidolactici overpopulated in IBD/D+-F and their byproducts LPS and exopolysaccharide (EPS) on the occurrence of depression and colitis in mice. Oral gavages of Klebsiella oxytoca, Escherichia coli, and Cronobacter sakazakii belonging to Enterobacteriaceae, singly or together, caused dose-dependently colitis and depression-like behaviors in germ-free and specific-pathogen-free mice. Although Enterococcus faecium and Pediococcus acidolactici did not significantly cause colitis and depression-like behaviors, they significantly deteriorated Klebsiella oxytoca- or Escherichia coli-induced colitis, neuroinflammation, and anxiety/depression-like behaviors and increased blood LPS, corticosterone, and IL-6 levels. The EPSs from Enterococcus faecium and Pediococcus acidolactici also worsened Klebsiella oxytoca LPS-induced colitis, neuroinflammation, and depression-like behaviors in mice and increased the translocation of fluorescein isothiocyanate-conjugated LPS into the hippocampus. However, Bifidobacterium longum, which was lower in IBD/D+-F vs. IBD/D--F, or its EPS suppressed them. In conclusion, Enterococcus faecium and Pediococcus acidolactici, known as a probiotic strain, and their EPSs may be a risk factor for the outbreak of depression and IBD.


Asunto(s)
Colitis , Enterococcus faecium , Enfermedades Inflamatorias del Intestino , Pediococcus acidilactici , Animales , Colitis/inducido químicamente , Colitis/microbiología , Depresión/psicología , Enterobacteriaceae , Escherichia coli , Humanos , Lipopolisacáridos , Ratones
3.
Sci Rep ; 11(1): 20406, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34650107

RESUMEN

Gut dysbiosis is closely associated with the occurrence of inflammatory bowel disease (IBD) and psychiatric disorder. Here, to understand the difference of gut microbiota composition and physiological effect between IBD patients with (IBD/D+) or without depression (IBD/D-), we analyzed the fecal microbiota composition of patients with IBD with (/D+) or without depression (/D-) and healthy volunteers (HVs) and examined the effects of these fecal microbiota transplantations (FMTs) on the occurrence of systemic inflammation and anxiety/depression in mice. FMTs from patients with IBD/D+ or IBD/D- caused IBD-like colitis in the transplanted mice: they increased the myeloperoxidase activity, IL-1ß and IL-6 expression, and NF-κB+/CD11c+ cell population in the colon. Transplantation of the IBD/D+ patient feces (IBD/D+-F) caused IBD-like colitis more strongly than that of IBD/D--F. FMTs from patients with IBD/D+ also caused anxiety-/depression-like behaviors, increased the NF-κB+/Iba1+ and lipopolysaccharide (LPS)+/Iba1+ cell populations, and decreased the BDNF+/NeuN+ cell population in the hippocampus. They increased LPS levels in the blood. FMTs from patients with IBD/D- caused anxiety-like, but not depression-like, behaviors. α-/ß-diversities and composition of gut microbiota in IBD-F were different from those of HV feces (HV-F). The Enterobacteriaceae and Enterococcaceae populations and LPS levels were higher in the IBD-F than in the HV-F. The Enterococcaceae population was higher in IBD/D+-F vs. IBD/D--F. However, the transplantation of HV-F into mice previously transplanted with IBD/D+-F significantly reduced depression-like behaviors, NF-κB+/Iba1+ and LPS+/Iba1+ cell populations in the hippocampus, LPS levels in the feces and blood, and IL-1ß expression in the colon. These findings suggest that the outbreak of depression/anxiety may be dependent on the systemic inflammation with a leaky gut through the gut dysbiosis-attributable overproduction of bacterial LPS and suppression of tight junction protein expression in patients with IBD.


Asunto(s)
Depresión/microbiología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiología , Inmunidad , Enfermedades Inflamatorias del Intestino/microbiología , Adulto , Animales , Ansiedad/etiología , Colitis/etiología , Colon/metabolismo , Corticosterona/sangre , Depresión/complicaciones , Depresión/etiología , Trasplante de Microbiota Fecal/efectos adversos , Hipocampo/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Interleucina-6/sangre , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Peroxidasa/metabolismo
4.
Sci Rep ; 11(1): 20659, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667205

RESUMEN

Gut lactobacilli and bifidobacteria on the immune homeostasis. Therefore, to understand the mechanism in vivo, we selected human fecal Lactobacillus rhamnosus NK210 and Bifidobacterium longum NK219, which strongly suppressed the IFN-γ to IL-10 expression (IIE) ratio in lipopolysaccharide-stimulated macrophages. Thereafter, we examined their effects on the endotoxin, antibiotics, or antitumor drug-stimulated immune imbalance in mice. Intraperitoneal injection of lipopolysaccharide and oral gavage of ampicillin increased IFN-γ and TNF-α expression in the spleen, colon, and hippocampus, while IL-10 expression decreased. However, intraperitoneal injection of cyclophosphamide suppressed IFN-γ, TNF-α, and IL-10 expression. LPS exposure induced splenic natural killer cell cytotoxicity against YAC-1 cells (sNK-C) and peritoneal macrophage phagocytosis against Candida albicans (pMA-P) activities, while cyclophosphamide and ampicillin treatments suppressed sNK-C and pMA-P activities. However, LPS, ampicillin, cyclophosphamide all increased IIE and TNF-α to IL-10 expression (TIE) ratios. Oral administration of NK210 and/or NK219 significantly reduced LPS-induced sNK-C, pMA-P, and IFN-γ expression, while cyclophosphamide- or ampicillin-suppressed sNK-C and pMA-P activities, cyclophosphamide-suppressed IFN-γ, TNF-α, and IL-10 expression, and ampicillin-suppressed IL-10 expression increased. Nevertheless, they suppressed LPS-, ampicillin-, or cyclophosphamide-induced IIE and TIE ratios, cognitive impairment, and gut dysbiosis. In particular, NK219, but not NK210, increased the IIE expression ratio in vitro and in vivo, and enhanced sNK-C and pMA-P activities in normal control mice, while cognitive function and gut microbiota composition were not significantly affected. These findings suggest that NK210, Lactobacillus sp, and NK219, Bifidobacterium additively or synergistically alleviate gut dysbiosis, inflammation, and cognitive impairment with immune imbalance by controlling IIE and TIE ratios.


Asunto(s)
Bifidobacterium longum/metabolismo , Disbiosis/terapia , Lacticaseibacillus rhamnosus/metabolismo , Animales , Bifidobacterium/metabolismo , Bifidobacterium longum/patogenicidad , Disfunción Cognitiva/microbiología , Disfunción Cognitiva/terapia , Colitis/microbiología , Colitis/terapia , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Inflamación/metabolismo , Interferón gamma/antagonistas & inhibidores , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Lactobacillus/metabolismo , Lacticaseibacillus rhamnosus/patogenicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Probióticos/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
5.
Food Funct ; 12(21): 10750-10763, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34608923

RESUMEN

Bacterial lipopolysaccharide (LPS) is a risk factor for the outbreak of Alzheimer's disease. Therefore, we isolated Lactobacillus plantarum NK151 and Bifidobacterium longum NK173 from a human fecal bacteria collection, which inhibited Escherichia coli LPS production, and examined their effects on the Escherichia coli K1- or LPS-induced cognitive impairment in mice. Oral gavage of NK151, NK173, or their (4 : 1) mixture (NKm) significantly alleviated Escherichia coli K1-induced cognitive impairment-like behaviors in the Y-maze and novel object recognition tasks. Their treatments decreased IL-1ß, IL-6, and TNF-α expression and NF-κB+/Iba1+ and LPS+/Iba1+ cell populations in the hippocampus, while the brain-derived neurotrophic factor (BDNF)+/neuronal nuclei (NeuN)+ cell population and BDNF to proBNDF expression increased. They suppressed LPS-induced cognition impairment-like behaviors and neuroinflammation marker levels in the hippocampus. Treatment with them reduced Escherichia coli K1- or LPS-induced LPS and apolipoprotein E levels in the blood and inflammatory marker levels in the colon. Furthermore, treatment with them modulated fecal Proteobacteria, Bacteroidetes, and Verrucomicrobia populations. Of these gut bacteria, Bacteroidaceae, Odoribacteraceae, Lactobacillaceae, Bifidobacteriaceae, Rikenellaceae, Helicobacteraceae, and Deferribacteraceae are correlated with cognitive function and blood and fecal LPS levels. These findings suggest that NK151 and NK173 may alleviate cognitive impairment with colitis by upregulating NF-κB-mediated BDNF expression through the suppression of fecal and blood bacterial LPS levels.


Asunto(s)
Bifidobacterium longum , Disfunción Cognitiva/prevención & control , Microbioma Gastrointestinal/fisiología , Lactobacillus plantarum , Lipopolisacáridos/metabolismo , Probióticos/farmacología , Animales , Disfunción Cognitiva/inducido químicamente , Microbioma Gastrointestinal/efectos de los fármacos , Genoma Bacteriano , Masculino , Ratones , ARN Bacteriano/genética , ARN Ribosómico 16S/genética , Organismos Libres de Patógenos Específicos , Secuenciación Completa del Genoma
6.
Front Immunol ; 11: 273, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32158447

RESUMEN

To understand the roles of human gut bacteria in the occurrence of neuropsychiatric disorders, we isolated inflammatory Escherichia coli K1 and anti-inflammatory Lactobacillus mucosae from healthy human feces and examined their effects on the occurrence of altered microbiota, cognitive decline, and depression in mice. Oral gavage of Escherichia coli K1 caused colitis, cognitive decline, and depression in mice in the elevated plus maze, tail suspension, and forced swimming tasks. However, NK41 treatment reduced K1-induced cognitive decline and anxiety/depression. Furthermore, NK41 treatment increased K1-suppressed brain-derived neurotrophic factor (BDNF) expression and BDNF+/NeuN+ cell population and suppressed K1-induced NF-κB activation and LPS+/Iba1+ and NF-κB+/Iba1+ (microglial) cell populations in the hippocampus. NK41 treatment also suppressed K1-induced TNF-α and LPS levels in the blood and TNF-α expression, myeloperoxidase activity, NF-κB+/CD11c+ and CD11b+/CD11c+ cell populations in the colon. Furthermore, NK41 treatment decreased K1-induced colonic MUC2 expression, gut Proteobacteria population, and fecal LPS levels and modified the bacterial abundance related to polysaccharide breaking and biosynthesis. In conclusion, the overgrowth of inflammatory bacteria such as Escherichia coli in the gastrointestinal tract can cause neuropsychiatric disorders with gut microbiota alteration and the superiority of anti-inflammatory bacteria such as Lactobacillus mucosae can alleviate neuropsychiatric disorders with the attenuation of altered microbiota.


Asunto(s)
Escherichia coli/fisiología , Microbioma Gastrointestinal/fisiología , Lactobacillus/fisiología , Interacciones Microbianas/fisiología , Animales , Ansiedad , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Disfunción Cognitiva , Depresión , Escherichia coli/inmunología , Heces/microbiología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Inflamación , Lactobacillus/inmunología , Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Nutrients ; 11(4)2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30979031

RESUMEN

The gut dysbiosis by stressors such as immobilization deteriorates psychiatric disorders through microbiota-gut-brain axis activation. To understand whether probiotics could simultaneously alleviate anxiety/depression and colitis, we examined their effects on immobilization stress (IS)-induced anxiety/depression and colitis in mice. The probiotics Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98 were isolated from healthy human feces. Mice with anxiety/depression and colitis were prepared by IS treatment. NK33 and NK98 potently suppressed NF-κB activation in lipopolysaccharide (LPS)-induced BV-2 cells. Treatment with NK33 and/or NK98, which were orally gavaged in mice before or after IS treatment, significantly suppressed the occurrence and development of anxiety/depression, infiltration of Iba1⁺ and LPS⁺/CD11b⁺ cells (activated microglia) into the hippocampus, and corticosterone, IL-6, and LPS levels in the blood. Furthermore, they induced hippocampal BDNF expression while NF-κB activation was suppressed. NK33 and/or NK98 treatments suppressed IS-induced colon shortening, myeloperoxidase activity, infiltration of CD11b⁺/CD11c⁺ cells, and IL-6 expression in the colon. Their treatments also suppressed the IS-induced fecal Proteobacteria population and excessive LPS production. They also induced BDNF expression in LPS-induced SH-SY5Y cells in vitro. In conclusion, NK33 and NK98 synergistically alleviated the occurrence and development of anxiety/depression and colitis through the regulation of gut immune responses and microbiota composition.


Asunto(s)
Ansiedad/terapia , Bifidobacterium adolescentis , Colitis/terapia , Depresión/terapia , Limosilactobacillus reuteri , Estrés Psicológico/complicaciones , Animales , Ansiedad/prevención & control , Ansiedad/psicología , Línea Celular , Colitis/psicología , Depresión/prevención & control , Depresión/psicología , Disbiosis/psicología , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía , FN-kappa B/metabolismo , Neuroblastoma , Probióticos/administración & dosificación , Restricción Física , Estrés Psicológico/etiología , Células Tumorales Cultivadas
8.
Neurosci Lett ; 698: 51-57, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30615977

RESUMEN

The excessive intake of a high-fat diet (HFD) leads to obesity, including metabolic syndromes, disturbs gut microbiota composition, causes colitis, and increases the plasma concentration of lipopolysaccharide (LPS). In the present study, we examined the role of gut microbiota in the occurrence of HFD-induced psychiatric disorders in mice. C57BL/6 J male mice fed a HFD for 9 weeks were led to obesity; their memory impairment was assessed by the Y-maze and novel object recognition test, and anxiety-like behaviors by the elevated plus maze. The intake of a HFD suppressed brain-derived neurotrophic factor (BDNF) expression in the hippocampus and increased blood TNF-α and LPS levels. HFD treatment more potently increased NF-κB activation and Iba1+ (microglial) cell populations in the hippocampus. Furthermore, HFD feeding increased TNF-α expression, myeloperoxidase activity, and CD11b+/CD11c+ cell (macrophages and dendritic cells) populations in the colon and altered gut microbiota composition including increases in the Proteobacteria population, and increases in fecal LPS levels. The stool lysates of HFD-treated mice suppressed BDNF expression and CREB phosphorylation in SH-SY5Y cells and increased NF-κB activation in BV-2 microglial cells compared to those of low-fat diet-treated mice while these effects were attenuated by treatment with anti-LPS antibody. These findings suggest that excessive intake of HFD can simultaneously cause obesity and psychiatric disorders by suppressing hippocampal BDNF expression with the disturbance of gut microbiota composition, particularly the increase in Proteobacteria population and LPS production.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal , Trastornos Mentales/microbiología , Proteobacteria/fisiología , Animales , Ansiedad/etiología , Ansiedad/microbiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/microbiología , Enfermedades del Colon/etiología , Enfermedades del Colon/microbiología , Heces/microbiología , Hipocampo/metabolismo , Inflamación/etiología , Inflamación/microbiología , Trastornos de la Memoria/etiología , Trastornos de la Memoria/microbiología , Trastornos Mentales/etiología , Trastornos Mentales/psicología , Ratones , Ratones Endogámicos C57BL , FN-kappa B , Obesidad/etiología , Obesidad/microbiología , Organismos Libres de Patógenos Específicos
9.
Int Immunopharmacol ; 73: 246-253, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31112869

RESUMEN

The flower of Pueraria lobata (family Fabaceae) has been clinically used in traditional Chinese medicine to counteract symptoms associated with drinking alcohol and liver injury and to alleviate inflammatory diseases. Its major constituent kakkalide is metabolized to irisolidone by gut microbiota. This research study was undertaken to understand the anti-colitis mechanism of kakkalide and irisolidone in vitro and in vivo. Kakkalide and its metabolite irisolidone inhibited lipopolysaccharide (LPS)-stimulated NF-κB activation and TNF-α expression in macrophages. They also inhibited LPS-induced phosphorylation of IRAK1 and TAK1 and activation of NF-κB by inhibiting the binding of Alexa Fluor 488-conjugated LPS in vitro. Orally administered irisolidone or kakkalide alleviated colon shortening and myeloperoxidase activity in mice with 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis. Their treatments also protected epithelial cell disruption and infiltration of CD11b+/CD11c+ cells in the colon. Furthermore, they suppressed TNBS-induced expression of M1 macrophage markers TNF-α, CD80, CD86, and Arg2 expression while the expression of M2 macrophage markers Arg1, CD163, CD206, and IL-10 was induced. They also suppressed the fecal Proteobacteria population. Overall, the anti-colitic effects of irisolidone were superior to those of kakkalide. Kakkalide and its metabolite irisolidone inhibited inflammation in vitro and in vivo by inhibiting LPS binding to toll-like receptor 4 and gut proteobacteria population.


Asunto(s)
Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Flavonoides/uso terapéutico , Glicósidos/uso terapéutico , Isoflavonas/uso terapéutico , Animales , Antiinflamatorios/farmacología , Células Cultivadas , Colitis/inducido químicamente , Colitis/inmunología , Flavonoides/farmacología , Glicósidos/farmacología , Isoflavonas/farmacología , Lipopolisacáridos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Proteobacteria , Receptor Toll-Like 4/inmunología , Ácido Trinitrobencenosulfónico , Factor de Necrosis Tumoral alfa/inmunología
10.
Mol Nutr Food Res ; 63(6): e1800978, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30636176

RESUMEN

SCOPE: Long-term feeding of a high-fat diet (HFD) causes gastrointestinal inflammation and gut microbiota disturbance, leading to the increased occurrence of obesity and anxiety. In the present study, the effects of heat-labile Lactobacillus sakei OK67, tyndallized OK67 (tOK67), and heat-stable Lactobacillus sakei PK16 on HFD-induced obesity and anxiety in mice are examined. METHODS AND RESULTS: Obesity is induced in mice by feeding with HFD. Oral administration of live OK67, tOK67, or PK16 reduces HFD-induced body and liver weights and blood triglyceride, total cholesterol, corticosterone, and lipopolysaccharide levels. These treatments also suppress HFD-induced NF-κB activation and increased HFD-suppressed AMP-activated protein kinase (AMPK) activation and SIRT-1 expression in the liver. OK67 or PK16 treatment alleviates HFD-induced anxiety-like behaviors and increases BDNF expression and NF-κB activation in the hippocampus. Moreover, OK67 or PK16 treatment suppresses HFD-induced colitis and suppresses the Proteobacteria population and fecal lipopolysaccharide levels in mice. OK67 or PK16 treatment inhibits NF-κB activation and induced AMPK activation and SIRT-1 expression in lipopolysaccharide-stimulated Caco-2 cells. Overall, the antiobesity and anxiolytic effects of live OK67 are more potent than those of tOK67. CONCLUSION: Lactobacillus sakei can alleviate HFD-induced obesity, colitis, and anxiety by regulating gut microbiota-mediated AMPK and NF-κB activation and SIRT-1 expression.


Asunto(s)
Ansiedad/dietoterapia , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal , Latilactobacillus sakei , Obesidad/dietoterapia , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Fármacos Antiobesidad/farmacología , Ansiedad/etiología , Células CACO-2 , Colitis/dietoterapia , Colitis/etiología , Activación Enzimática , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Obesidad/etiología , Probióticos/farmacología , Sirtuina 1/metabolismo
11.
Sci Rep ; 8(1): 13897, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-30224732

RESUMEN

The present study aimed to understand the crosstalk between anxiety and gut microbiota. Exposure of mice to immobilization stress (IS) led to anxiety-like behaviors, increased corticosterone and tumor necrosis factor-α levels in the blood, increased nuclear factor (NF)-κB activation and microglia/monocyte populations in the hippocampus, and suppressed brain-derived neurotrophic factor (BDNF) expression in the hippocampus. Furthermore, IS exposure increased NF-κB activation and monocyte population in the colon and increased Proteobacteria and Escherichia coli populations in the gut microbiota and fecal and blood lipopolysaccharide (LPS) levels while decreasing the lactobacilli population. Oral administration of the fecal microbiota of mice treated with IS (FIS) or E. coli led to the increased NF-κB activation and monocyte population in the colon. These treatments increased blood corticosterone and LPS levels and anxiety-like behaviors, decreased BDNF expression, and induced NF-κB activation and microglia/monocyte populations in the hippocampus. Intraperitoneal injection of LPS purified from E. coli also led to anxiety and colitis in mice. Oral administration of commensal lactobacilli, particularly Lactobacillus johnsonii, attenuated IS- or E. coli-induced colitis and anxiety-like behaviors and biomarkers. These findings suggest that exposure to stressors can increase Proteobacteria populations and fecal LPS levels and cause gastrointestinal inflammation, resulting in the deterioration of anxiety through NF-κB activation. However, the amelioration of gastrointestinal inflammation by treatment with probiotics including L. johnsonii can alleviate anxiety.


Asunto(s)
Ansiedad/etiología , Escherichia coli/crecimiento & desarrollo , Microbioma Gastrointestinal , Inmovilización/psicología , FN-kappa B/metabolismo , Estrés Fisiológico , Animales , Ansiedad/microbiología , Colitis/etiología , Colitis/microbiología , Colitis/terapia , Trasplante de Microbiota Fecal , Masculino , Ratones , Ratones Endogámicos C57BL , Probióticos/administración & dosificación
13.
Mucosal Immunol ; 11(5): 1386-1397, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29867078

RESUMEN

The aim of the present study was to determine whether there is the mechanistic connection between antibacterial-dependent gut microbiota disturbance and anxiety. First, exposure of mice to ampicillin caused anxiety and colitis and increased the population of Proteobacteria, particularly Klebsiella oxytoca, in gut microbiota and fecal and blood lipopolysaccharide levels, while decreasing lactobacilli population including Lactobacillus reuteri. Next, treatments with fecal microbiota of ampicillin-treated mouse (FAP), K. oxytoca, or lipopolysaccharide isolated from K. oxytoca (KL) induced anxiety and colitis in mice and increased blood corticosterone, IL-6, and lipopolysaccharide levels. Moreover, these treatments also increased the recruitment of microglia (Iba1+), monocytes (CD11b+/CD45+), and dendritic cells (CD11b+/CD11c+) to the hippocampus, as well as the population of apoptotic neuron cells (caspase-3+/NeuN+) in the brain. Furthermore, ampicillin, K. oxytoca, and KL induced NF-κB activation and IL-1ß and TNF-α expression in the colon and brain as well as increased gut membrane permeability. Finally, oral administration of L. reuteri alleviated ampicillin-induced anxiety and colitis. These results suggest that ampicillin exposure can cause anxiety through neuro-inflammation which can be induced by monocyte/macrophage-activated gastrointestinal inflammation and elevated Proteobacteria population including K. oxytoca, while treatment with lactobacilli suppresses it.


Asunto(s)
Antibacterianos/efectos adversos , Ansiedad/inducido químicamente , Ansiedad/microbiología , Colon/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Ampicilina/efectos adversos , Animales , Ansiedad/sangre , Corticosterona/sangre , Células Dendríticas/efectos de los fármacos , Heces/microbiología , Hipocampo/efectos de los fármacos , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/microbiología , Interleucina-6/sangre , Lipopolisacáridos/sangre , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Monocitos/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteobacteria/efectos de los fármacos
15.
Am J Chin Med ; 46(8): 1879-1897, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30518233

RESUMEN

A variety of products have been developed with red ginseng (RG, the steamed roots of Panax ginseng Meyer). To clarify the immunomodulating effects of water-extracted RG (wRG), 50% ethanol-extracted RG (eRG), enzyme-treated eRG (ERG) and probiotic-fermented eRG (FRG), we examined their immunopotentiating and immunosuppressive effects in mice with cyclophosphamide (CP)-induced immunosuppression (CI) or 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis (TC). Oral administration of RG in CI mice significantly increased blood IFN- γ levels. Treatment with RG also increased the tumoricidal effects of CI mouse splenic cytotoxic T (Tc) and NK cells against YAC-1 cells. Treatment with RGs, in particular FRG and wRG, significantly increased Th1 cell differentiation. Treatment with RG except wRG increased Treg cell differentiation. However, wRG alone increased IL-6 and IL-17 expression in the colon of CI mice. Furthermore, RG alleviated colitis in TC mice. FRG most potently suppressed TNBS-induced colon shortening, NF- κ B activation and TNF- α and IL-17 expression and increased IL-10 expression. RGs inhibited TNF- α expression and increased IL-10 expression in lipopolysaccharide-stimulated primary macrophages in vitro while the differentiation of splenic T cells into type 1 T (Th1) and regulatory T (Treg) cells was increased by FRG in vitro. In conclusion, FRG can alleviate immunosuppression and inflammation by inhibiting macrophage activation and regulating Th1 and Treg cell differentiation.


Asunto(s)
Adyuvantes Inmunológicos , Diferenciación Celular/efectos de los fármacos , Colitis/tratamiento farmacológico , Ciclofosfamida/antagonistas & inhibidores , Fermentación , Inmunosupresores/antagonistas & inhibidores , Activación de Macrófagos/efectos de los fármacos , Panax/química , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Linfocitos T/fisiología , Ácido Trinitrobencenosulfónico/efectos adversos , Administración Oral , Animales , Células Cultivadas , Colitis/inducido químicamente , Colitis/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos BALB C , Extractos Vegetales/administración & dosificación , Extractos Vegetales/aislamiento & purificación
16.
Int Immunopharmacol ; 45: 90-97, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28189973

RESUMEN

The roots of Abrus precatorius (AP, Fabaceae) have traditionally been used in Vietnam and China for the treatment of inflammatory diseases such as stomatitis, asthma, bronchitis, and hepatitis. Therefore, in this study, we isolated 4-methoxylonchocarpin (ML), an anti-inflammatory compound present in AP, and studied its anti-inflammatory effects in mice with 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis. In lipopolysaccharide (LPS)-stimulated macrophages, ML was found to inhibit nuclear factor (NF)-κB activation and tumor necrosis factor (TNF) and interleukin (IL)-6 expression by inhibiting LPS binding to Toll-like receptor 4 (TLR4) in vitro. Oral administration of ML in mice with TNBS-induced colitis suppressed colon shortening and colonic myeloperoxidase activity. ML treatment significantly inhibited the activation of nuclear factor (NF)-κB and phosphorylation of transforming growth factor ß-activated kinase 1 in the colon. Treatment with ML also inhibited TNBS-induced expression of IL-1ß, IL-17A, and TNF. While ML reduced the TNBS-induced expression of M1 macrophage markers such as arginase-2 and TNF, it was found to increase the expression of M2 macrophage markers such as arginase-1 and IL-10. In conclusion, oral administration of ML attenuated colitis in mice by inhibiting the binding of LPS to TLR4 on immune cells and increasing the polarization of M1 macrophages to M2 macrophages.


Asunto(s)
Abrus/inmunología , Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Colon/efectos de los fármacos , Flavonas/uso terapéutico , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Animales , Antiinflamatorios/química , Diferenciación Celular , Células Cultivadas , Colitis/inducido químicamente , Colon/inmunología , Flavonas/química , Inflamación/inducido químicamente , Interleucina-6 , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Ácido Trinitrobencenosulfónico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA