Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pineal Res ; 76(1): e12929, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38047407

RESUMEN

Cholestatic liver disease is characterized by disturbances in the intestinal microbiota and excessive accumulation of toxic bile acids (BA) in the liver. Melatonin (MT) can improve liver diseases. However, the underlying mechanism remains unclear. This study aimed to explore the mechanism of MT on hepatic BA synthesis, liver injury, and fibrosis in 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-fed and Mdr2-/- mice. MT significantly improved hepatic injury and fibrosis with a significant decrease in hepatic BA accumulation in DDC-fed and Mdr2-/- mice. MT reprogramed gut microbiota and augmented fecal bile salt hydrolase activity, which was related to increasing intestinal BA deconjugation and fecal BA excretion in both DDC-fed and Mdr2-/- mice. MT significantly activated the intestinal farnesoid X receptor (FXR)/fibroblast growth factor 15 (FGF-15) axis and subsequently inhibited hepatic BA synthesis in DDC-fed and Mdr2-/- mice. MT failed to improve DDC-induced liver fibrosis and BA synthesis in antibiotic-treated mice. Furthermore, MT provided protection against DDC-induced liver injury and fibrosis in fecal microbiota transplantation mice. MT did not decrease liver injury and fibrosis in DDC-fed intestinal epithelial cell-specific FXR knockout mice, suggesting that the intestinal FXR mediated the anti-fibrosis effect of MT. In conclusion, MT ameliorates cholestatic liver diseases by remodeling gut microbiota and activating intestinal FXR/FGF-15 axis-mediated inhibition of hepatic BA synthesis and promotion of BA excretion in mice.


Asunto(s)
Colestasis , Hepatopatías , Melatonina , Ratones , Animales , Melatonina/farmacología , Melatonina/metabolismo , Hígado/metabolismo , Colestasis/tratamiento farmacológico , Colestasis/metabolismo , Colestasis/patología , Hepatopatías/metabolismo , Hepatopatías/patología , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/farmacología , Ratones Noqueados , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Ratones Endogámicos C57BL
2.
Food Chem Toxicol ; 176: 113751, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37030333

RESUMEN

Aflatoxin B1 (AFB1) is one of major pollutant in food and feed worldwide. The purpose of this study is to investigate the mechanism of AFB1-induced liver injury. Our results showed that AFB1 caused hepatic bile duct proliferation, oxidative stress, inflammation and liver injury in mice. AFB1 exposure induced gut microbiota dysbiosis and reduced fecal bile salt hydrolase (BSH) activity. AFB1 exposure promoted hepatic bile acid (BA) synthesis and changed intestinal BA metabolism, especially increased intestinal conjugated bile acids levels. AFB1 exposure inhibited intestinal farnesoid X receptor (FXR)/fibroblast growth factor 15 (FGF-15) signaling. Furthermore, the mice received fecal microbiota transplantation from AFB1-treated mice induced liver injury, reduced intestinal FXR signaling and increased hepatic BA synthesis. Finally, the intestine-restricted FXR agonist treatment decreased hepatic BA synthesis, ROS level, inflammation and liver injury in AFB1-treated mice. This study suggests that modifying the gut microbiota, altering intestinal BA metabolism and/or activating intestinal FXR/FGF-15 signaling may be of value for the treatment of AFB1-induced liver disease.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Microbioma Gastrointestinal , Ratones , Animales , Aflatoxina B1/toxicidad , Aflatoxina B1/metabolismo , Ácidos y Sales Biliares/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Inflamación/metabolismo , Ratones Endogámicos C57BL
3.
J Agric Food Chem ; 71(1): 867-876, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36579420

RESUMEN

Aflatoxin B1 (AFB1) is a widespread mycotoxin in food and feed. Although the liver is the main target organ of AFB1, the intestine is the first exposure organ to AFB1. However, the mechanism by which AFB1 induced intestinal barrier dysfunction via regulating the farnesoid X receptor (FXR)-mediated myosin light chain kinase (MLCK) signaling pathway has rarely been studied. In vivo, AFB1 exposure significantly decreased the small intestine length and increased the intestinal permeability. Meanwhile, AFB1 exposure markedly suppressed the protein expressions of FXR, ZO-1, occludin, and claudin-1 and enhanced the protein expression of MLCK. In vitro, AFB1 exposure induced intestinal barrier dysfunction by the elevation in the FITC-Dextran 4 kDa flux and inhibition in the transepithelial electrical resistance in a dose-dependent manner. In addition, AFB1 exposure downregulated the mRNA and protein expressions of FXR, ZO-1, occludin, and claudin-1, redistributed the ZO-1 protein, and enhanced the protein expressions of MLCK and p-MLC. However, fexaramine (Fex, FXR agonist) pretreatment markedly reversed the AFB1-induced FXR activity reduction, MLCK protein activation, and intestinal barrier impairment in vitro and in vivo. Moreover, pretreatment with the inhibition of MLCK with ML-7 significantly alleviated the AFB1-induced intestinal barrier dysfunction and tight junction disruption in vitro. In conclusion, AFB1 induced intestinal barrier impairment via regulating the FXR-mediated MLCK signaling pathway in vitro and in vivo and provided novel insights to prevent mycotoxin poisoning in the intestine.


Asunto(s)
Enfermedades Intestinales , Quinasa de Cadena Ligera de Miosina , Animales , Ratones , Aflatoxina B1/toxicidad , Aflatoxina B1/metabolismo , Células CACO-2 , Claudina-1/genética , Claudina-1/metabolismo , Células Epiteliales/metabolismo , Enfermedades Intestinales/metabolismo , Mucosa Intestinal/metabolismo , Cadenas Ligeras de Miosina , Quinasa de Cadena Ligera de Miosina/genética , Ocludina/genética , Ocludina/metabolismo , Transducción de Señal , Uniones Estrechas/metabolismo
4.
Sci Total Environ ; 849: 157861, 2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-35934034

RESUMEN

Cadmium (Cd) is a widely prevalent environmental pollutant that accumulates in the liver and induces liver injury. The mechanism of Cd-induced liver injury remains elusive. Our study aimed to clarify the mechanism by which changes in the gut microbiota contribute to Cd-induced liver injury. Here, a murine model of liver injury induced by chronic Cd exposure was used. Liver injury was assessed by biochemistry and histopathology. Expression profiles of genes involved in bile acid (BA) homeostasis, inflammation and injury were assessed via Realtime-PCR and Western-blot. 16S rRNA gene sequencing and mass spectrometry-based metabolomics were used to investigate changes in the gut microbiota and its metabolites in the regulation of Cd-induced liver injury. Here, we showed that Cd exposure induced hepatic ductular proliferation, hepatocellular damage and inflammatory infiltration in mice. Cd exposure induced gut microbiota dysbiosis and reduced the fecal bile salt hydrolase activity leading to an increase of tauro-ß-muricholic acid levels in the intestine. Cd exposure decreased intestine FXR/FGF-15 signaling and promoted hepatic BA synthesis. Furthermore, the mice receiving fecal microbiota transplantation from Cd-treated mice showed reduced intestinal FXR/FGF-15 signaling, increased hepatic BA synthesis, and liver injury. However, the depletion of the commensal microbiota by antibiotics failed to change these indices in Cd-treated mice. Finally, the administration of the intestine-restricted FXR agonist fexaramine attenuated the liver injury, improved the intestinal barrier, and decreased hepatic BA synthesis in the Cd-treated mice. Our study identified a new mechanism of Cd-induced liver injury. Cd-induced gut microbiota dysbiosis, decreased feces BSH activity, and increased intestinal T-ßMCA levels led to an inhibition of intestinal FXR/FGF-15 signaling and an increase in hepatic BA synthesis, ultimately facilitating the development of hepatic ductular proliferation, inflammation, and injury in mice. This study expands our understanding of the health hazards caused by environmental Cd pollution.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Contaminantes Ambientales , Microbioma Gastrointestinal , Animales , Antibacterianos/metabolismo , Ácidos y Sales Biliares/metabolismo , Cadmio/metabolismo , Cadmio/toxicidad , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Disbiosis/inducido químicamente , Contaminantes Ambientales/metabolismo , Microbioma Gastrointestinal/fisiología , Inflamación , Intestinos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S , Receptores Citoplasmáticos y Nucleares/metabolismo
5.
J Pineal Res ; 73(2): e12812, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35652241

RESUMEN

Aflatoxin B1 (AFB1) is a widespread contaminant in foods and feedstuffs, and its target organ is the liver. Melatonin (MT) has been shown to alleviate inflammation in organs and remodel gut microbiota in animals and humans. However, the underlying mechanism by which MT alleviates AFB1-induced liver injury remains unclear. In the present study, MT pretreatment markedly increased the expression of intestinal tight junction proteins (ZO-1, Occludin, and Claudin-1), decreased intestinal permeability, reduced production of gut-derived Lipopolysaccharide (LPS) and remodeled gut microbiota, ultimately alleviated AFB1-induced liver injury in mice. Interestingly, MT pretreatment failed to exert beneficial effects on the intestine and liver in antibiotic-treated mice. Meanwhile, MT pretreatment significantly increased the farnesoid X receptor (FXR) protein expression of ileum, and decreased the TLR4/NF-κB signaling pathway-related messenger RNA (mRNA) and proteins (TLR4, MyD88, p-p65, and p-IκBα) expression in livers of AFB1-exposed mice. Subsequently, pretreatment by Gly-ß-MCA, an intestine-selective FXR inhibitor, blocked the alleviating effect of MT on liver injury through increasing the liver-specific expression of TLR4/NF-κB signaling pathway-related mRNA and proteins (TLR4, MyD88, p-p65, and p-IκBα). In conclusion, MT pretreatment ameliorated AFB1-induced liver injury and the potential mechanism may be related to regulate gut microbiota/intestinal FXR/liver TLR4 signaling axis, which provides a strong evidence for the protection of gut-derived liver inflammation.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Microbioma Gastrointestinal , Melatonina , Aflatoxina B1/toxicidad , Animales , Humanos , Inflamación , Hígado/metabolismo , Melatonina/farmacología , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , ARN Mensajero , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
6.
Sci Total Environ ; 832: 155027, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35381244

RESUMEN

Deoxynivalenol (DON) and arsenic (As) are widespread environmental contaminants, which are frequently found in human and animal food products. The intestine is a common target of As and DON when they are digested. Numerous studies mainly evaluate the individual effects whereas their combined toxicity has rarely been elucidated. Hence, this study was to assess the effect of low dose of NaAsO2 on DON-induced intestinal damage and explore the underling mechanism in mice and IPEC-J2 cells. The results showed that low dose of NaAsO2 exacerbated DON-induced intestinal impairment by increasing intestinal permeability and decreasing the abundance of tight junction proteins (ZO-1, Occludin, Claudin-1). Further, low dose of NaAsO2 enhanced the AhR signaling pathway and autophagy-related mRNA/protein expressions induced by DON. Interestingly, FICZ, an AhR activator, instead of CH223191, an AhR inhibitor, could alleviate toxicity of the low dose of NaAsO2 in the mice and IPEC-J2 cells. Compared to the WT IPEC-J2 cells, the intestinal barrier damage was more serious in LC3B-/- IPEC-J2 cells induced by low dose of NaAsO2 combination with DON. Collectively, our study demonstrated that low dose of NaAsO2 exacerbated DON-induced intestinal barrier impairment in vivo and in vitro. The present study also demonstrated that activation of AhR-mediated autophagy might be a self-protection mechanism. Hence, AhR and autophagy might be novel therapeutic targets to prevent or alleviate NaAsO2 combined with DON-induced intestinal barrier impairment.


Asunto(s)
Arsénico , Animales , Arsénico/toxicidad , Autofagia , Línea Celular , Ratones , Receptores de Hidrocarburo de Aril/genética , Tricotecenos
7.
Toxicology ; 457: 152802, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33905761

RESUMEN

Ochratoxin A (OTA) and fumonisin B1 (FB1), two of the most toxicologically important mycotoxins, often coexist in a variety of foodstuff and feed in humans and animals. Because of the low content of FB1 in foodstuff and feed, alone harmfulness of FB1 is often ignored. However, it is unknown whether the lower dosage of FB1 aggravates the toxicity of other mycotoxins. In this article, we aimed to investigate the effects of the lower dosage of FB1 on OTA-induced nephrotoxicity and apoptosis, and its underlying mechanism in porcine kidney cells (PK-15). Our current study showed that the non-cytotoxic concentration of FB1 (8 µM) could enhance OTA(5 µM)-induced nephrocytotoxicity and the expression of pro-apoptosis-associated genes in PK-15 cells. We also observed that the production of reactive oxygen species (ROS) was increased. However, the expression of pro-apoptosis-associated genes were down-regulated when the N-acetylcysteine (NAC), a ROS scavenger, was used in our experiment. Besides, we found that the combined toxins could increase the protein expression of p-JNK instead of p-p38 and p-ERK. Pretreatment with SP600125, a JNK inhibitor, could significantly block the promotion effects of FB1 on OTA-induced nephrocytotoxicity and apoptosis. The protein expression of p-JNK was also inhibited and the promotion effects of FB1 were significantly alleviated when NAC was used. In conclusion, the non-cytotoxic dosage of FB1 could aggravate the nephrocytotoxicity and apoptosis caused by OTA via ROS-dependent JNK/MAPK signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Fumonisinas/toxicidad , Riñón/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ocratoxinas/toxicidad , Especies Reactivas de Oxígeno , Animales , Apoptosis/fisiología , Línea Celular , Fumonisinas/administración & dosificación , Riñón/metabolismo , Riñón/patología , Ocratoxinas/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Porcinos
8.
Zhongguo Dang Dai Er Ke Za Zhi ; 21(10): 955-959, 2019 Oct.
Artículo en Chino | MEDLINE | ID: mdl-31642426

RESUMEN

OBJECTIVE: To study the clinical effect and safety of double filtration plasmapheresis (DFPP) combined with double pulse therapy with methylprednisolone (MP) and cyclophosphamide (CTX) in the treatment of children with severe Henoch-Schönlein purpura nephritis (HSPN). METHODS: A total of 60 children with severe HSPN who were admitted to the hospital from January 2014 to March 2018 were enrolled and were randomly divided into an observation group and a control group (n=30 each). In addition to routine treatment, the children in the control group were given MP+CTX pulse therapy. Those in the observation group were given DFPP treatment in addition to the treatment in the control group, with three courses of treatment in total. After three courses of treatment, the two groups were compared in terms of 24-hour urinary protein, urinary microproteins, renal function parameters, adverse reactions, and clinical outcome. RESULTS: After three courses of treatment, the observation group had significantly greater reductions in 24-hour urinary protein, urinary albumin, urinary immunoglobulin G, urinary ß2-microglobulin, serum creatinine, and blood urea nitrogen than the control group (P<0.05). After the treatment ended, the observation group had a significantly shorter time to achieve remission than the control group (P<0.05). No serious adverse reactions, such as hemorrhagic cystitis, thrombocytopenia, and hemolysis, were observed, and there was no significant difference in the overall incidence rate of adverse reactions between the two groups (P>0.05). CONCLUSIONS: Compared with MP+CTX pulse therapy alone in the treatment of severe HSPN in children, DFPP combined with MP+CTX pulse therapy can further alleviate renal injury and improve clinical outcome and does not increase the incidence rate of adverse reactions.


Asunto(s)
Vasculitis por IgA , Nefritis , Niño , Glucocorticoides , Humanos , Inmunosupresores , Plasmaféresis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA