Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Microbiol ; 15: 1302998, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38292253

RESUMEN

Lactobacillus acidophilus (LA) is a common clinical probiotic that improves ulcerative colitis (UC) by restoring intestinal immune balance. However, the interaction of LA with the gut microbiota and its metabolites in the treatment of UC remains unknown. Therefore, this study seeks to elucidate whether the gut microbiota and its metabolites act as pivotal effectors in LA's therapeutic mechanisms and how precisely they modulate intestinal immunity. In this study, we verified that LA can obviously ameliorate the disease severity, and regulate intestinal immune disorders in UC mice. Subsequently, antibiotic (ABX)-mediated depletion of the gut microflora demonstrated that the therapeutic efficiency of LA was closely associated with gut microbiota. In addition, the results of metabolomics revealed that ursodeoxycholic acid (UDCA), a metabolite of intestinal flora, may be a potential effector molecule mediating therapeutic effects of LA. Indeed, we found that UDCA can improve the macro pathological characteristics of UC mice, and through a comprehensive set of in vivo and in vitro experiments, we discovered that UDCA exerts dual effects on immune regulation. Firstly, it promotes the differentiation of Treg cells, resulting in increased secretion of anti-inflammatory cytokines. Secondly, UDCA inhibits the polarization of M1 macrophages, effectively reducing the secretion of pro-inflammatory cytokines. Moreover, we found that UDCA regulation of immune response is directly related to the RapGap/PI3K-AKT/NF-κB signaling pathway. In conclusion, LA and its metabolite, UDCA, may treat UC by activating the RapGap/PI3K-AKT/NF-κB signaling pathway and modulating Treg cells and M1 macrophages. All in all, our findings highlight the potential of microbial metabolites in enhancing probiotic for UC treatment.

2.
Metab Brain Dis ; 38(7): 2327-2338, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37572229

RESUMEN

This report aimed to explore whether miR-188-5p regulated the pathological regulatory network of cerebral ischemia/reperfusion (I/R) injury. We simulated the cerebral I/R injury model with MACO/R and OGD/R treatments. Neuronal viability and apoptosis were assessed. The contents of miR-188-5p and Lin 28a were evaluated. The abundances of apoptosis-related proteins (Bax, Bcl-2, and cleaved caspase-3) and pro-inflammatory cytokines (TNF-α, IL-1ß, and IL-6) were measured. The interaction of miR-188-5p and Lin28a was confirmed. Lin28a silencing was supplemented to determine the delicate regulation of miR-188-5p. We revealed that miR-188-5p was upregulated and Lin28a was downregulated in I/R rats and OGD/R-induced cells. miR-188-5p silencing remarkably reduced the cerebral infarction volume, neurobehavioral score, brain edema, and Evans blue leakage. miR-188-5p silencing enhanced neuronal viability and alleviated apoptosis. The abundance of Bax and cleaved caspase-3 was reduced by miR-188-5p silencing, while Bcl-2 was augmented. miR-188-5p silencing impeded the contents of TNF-α, IL-1ß, and IL-6. miR-188-5p interacted with Lin28a and negatively regulated its expression. Interestingly, extra Lin28a silencing reversed apoptosis and the content of inflammatory cytokines. Our studies confirmed that miR-188-5p silencing alleviated neuronal apoptosis and inflammation by mediating the expression of Lin28a. The crosstalk of miR-188-5p and Lin28a offered a different direction for ischemic stroke therapy.


Asunto(s)
Isquemia Encefálica , MicroARNs , Daño por Reperfusión , Animales , Ratas , Apoptosis , Proteína X Asociada a bcl-2 , Isquemia Encefálica/metabolismo , Caspasa 3 , Citocinas/metabolismo , Glucosa , Infarto de la Arteria Cerebral Media/metabolismo , Interleucina-6 , MicroARNs/genética , MicroARNs/metabolismo , Daño por Reperfusión/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
Neurochem Res ; 48(11): 3457-3471, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37470906

RESUMEN

The objective of this research was to investigate the role of lncRNA MALAT1 and HSP90 in the regulation of neuronal necroptosis in mice with cerebral ischemia-reperfusion (CIR). We used male C57BL/6J mice to establish a middle cerebral artery occlusion (MCAO) model and conducted in vitro experiments using the HT-22 mouse hippocampal neuron cell line. The cellular localization of NeuN and MLKL, as well as the expression levels of neuronal necroptosis factors, MALAT1, and HSP90 were analyzed. Cell viability and necroptosis were assessed, and we also investigated the relationship between MALAT1 and HSP90. The results showed that MALAT1 expression increased after MCAO and oxygen-glucose deprivation/re-oxygenation (OGD/R) treatment in both cerebral tissues and cells compared with the control group. The levels of neuronal necroptosis factors and the co-localization of NeuN and MLKL were also increased in MCAO mice compared with the Sham group. MALAT1 was found to interact with HSP90, and inhibition of HSP90 expression led to decreased phosphorylation levels of neuronal necroptosis factors. Inhibition of MALAT1 expression resulted in decreased co-localization levels of NeuN and MLKL, decreased phosphorylation levels of neuronal necroptosis factors, and reduced necroptosis rate in cerebral tissues. Furthermore, inhibiting MALAT1 expression also led to a shorter half-life of HSP90, increased ubiquitination level, and decreased phosphorylation levels of neuronal necroptosis factors in cells. In conclusion, this study demonstrated that lncRNA MALAT1 promotes neuronal necroptosis in CIR mice by stabilizing HSP90.


Asunto(s)
Isquemia Encefálica , Proteínas HSP90 de Choque Térmico , ARN Largo no Codificante , Daño por Reperfusión , Animales , Masculino , Ratones , Apoptosis/genética , Isquemia Encefálica/metabolismo , Glucosa/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Necroptosis , Neuronas/metabolismo , Reperfusión , Daño por Reperfusión/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo
4.
Drug Des Devel Ther ; 17: 1371-1386, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37181826

RESUMEN

Purpose: This study aimed to investigate the underlying treatment mechanism of Radix Astragali (RA) in hyperuricemia from the perspective of microbiota and metabolomics. Methods: We used potassium oxyazinate (PO) to induce hyperuricemia mice, and we determined serum alanine aminotransferase/aspartate aminotransferase (ALT/AST), xanthine oxidase (XOD), creatinine (CRE), uric acid (UA), blood urea nitrogen (BUN) levels, liver XOD levels and assessed the kidney tissue histopathology. The therapeutic mechanism of RA in hyperuricemic mice was studied by 16S rRNA, metagenomic sequencing and metabolomics. Results: Our research showed that RA has therapeutic effect in hyperuricemia mice, such as slow the weight loss, repair kidney damage, and downregulate serum UA, XOD, CRE, ALT/AST, BUN, and liver XOD levels. RA restored the disturbance structure of the microbiota in hyperuricemia mice by increasing the relative abundances of beneficial bacteria (Lactobacillaceae and Lactobacillus murine) but decreasing the relative abundances of pathogenic bacteria (Prevotellaceae, Rikenellaceae and Bacteroidaceae). Meanwhile, we found that RA directly regulated the metabolic pathway (such as linoleic acid metabolism and glycerophospholipid metabolism) and indirectly regulated bile acid metabolism by mediating microbiota to ameliorate metabolic disorders. Subsequently, there was a robust correlation between specific microbiota, metabolites and the disease index. Conclusion: The ability of RA to protect mice against hyperuricemia is strongly linked to the microbiome-metabolite axis, which would provide evidence for RA as a medicine to prevent or treat hyperuricemia.


Asunto(s)
Medicamentos Herbarios Chinos , Hiperuricemia , Ratones , Animales , Hiperuricemia/tratamiento farmacológico , ARN Ribosómico 16S , Metagenómica , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Xantina Oxidasa/genética , Xantina Oxidasa/metabolismo
5.
Chem Biol Interact ; 376: 110449, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-36921834

RESUMEN

Clerodendranthus spicatus (Thunb.) C. Y. Wu, also known as kidney tea (KT), has been widely employed in kidney protection in Chinese Medicine. It has been reported that KT can lower uric acid (UA) and mitigate gout, while the mechanism remains to be elucidated. Given the close relationship between hyperuricemia (HUA), intestinal flora and host metabolism, this study aimed to explore the mechanism by which KT lowers UA from the perspective of the fecal microbiome and metabolome. Initially, mice were intraperitoneally injected with potassium oxonate to induce the HUA model. The results showed that KT markedly reduced the serum level of UA and impaired renal damage in HUA mice. Subsequently, the result of 16S rRNA gene sequencing analysis indicated that KT administration appeared a significant improvement in the structure of the intestinal flora, especially increased the abundances of Roseburia and Enterorhabdus, while decreased the abundances of Ileibacterium and UBA1819. Moreover, the levels of differential metabolites (including twenty-five in feces and eight in serum) identified by untargeted metabolomics returned to normal after KT intervention. Taken together, the mechanism of KT in alleviating HUA is related to the regulation of the intestinal flora and the remodeling of metabolic disorders, which will lay a theoretical foundation for KT as a UA-lowering drug.


Asunto(s)
Microbioma Gastrointestinal , Hiperuricemia , Ratones , Animales , Hiperuricemia/inducido químicamente , Hiperuricemia/tratamiento farmacológico , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Metaboloma , Riñón/metabolismo ,
6.
Eur J Pharmacol ; 942: 175504, 2023 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-36641101

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a clinical pathological syndrome of hepatic parenchymal cell steatosis caused by excessive lipid deposition, which is the chronic liver disease with the highest incidence in China. Asperuloside (ASP), a kind of iridoid compound, possesses natural pharmacological effects of anti-tumor, anti-inflammatory, antioxidant and anti-obesity. However, whether ASP can improve NAFLD remains unclear. PURPOSE: We aimed to investigate the effect of ASP on NAFLD mice induced by high-fat diet (HFD), and explore its mechanism in vivo and in vitro. METHODS: Pharmacodynamics of ASP was studied by HFD induction in NAFLD mice. HepG2 cells were induced by palmitic acid (PA) as cell model to investigate the effect of ASP on lipid deposition and inflammatory infiltration. Expression of Adenosine monophosphate - activated protein kinase (AMPK) signaling pathway and NOD-like receptor pyrin containing 3 (NLRP3) inflammasome were detected by Western blot and RT-PCR. Cytokines IL-1ß and TNF-α were detected by ELISA. RESULTS: ASP alleviated liver injury and inflammatory damage in mice with NAFLD. In addition, ASP improved lipid deposition as well as inflammatory response in HFD-induced NAFLD mice and PA-stimulated HepG2 cells. ASP ameliorated lipid deposition and inflammatory response by regulating the p-AMPK/SREBP-1c signaling pathway and NLRP3 inflammasome. CONCLUSION: Our results suggest that ASP improve lipid deposition and inflammatory infiltration in NAFLD mice via regulating the AMPK/SREBP-1c signaling pathway and NLRP3 inflammasome, which may be an effective candidate for the treatment of NAFLD.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Dieta Alta en Grasa/efectos adversos , Inflamasomas/metabolismo , Inflamación/metabolismo , Metabolismo de los Lípidos , Hígado , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ácido Palmítico/farmacología , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
7.
Neural Plast ; 2022: 3933252, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36338577

RESUMEN

Small extracellular vesicles (sEVs) secreted by mesenchymal stem cells (MSCs) have shown great therapeutic potential in cerebral ischemia-reperfusion injury (CIRI). In this study, we firstly performed a systematic review to evaluate the efficacy of MSCs-derived sEV for experimental cerebral ischemia/reperfusion injury. 24 studies were identified by searching 8 databases from January 2012 to August 2022. The methodological quality was assessed by using the SYRCLE 's risk of bias tool for animal studies. All the data were analyzed using RevMan 5.3 software. As a result, the score of study quality ranged from 3 to 9 in a total of ten points. Meta-analyses showed that MSCs-derived sEVs could effectively alleviate neurological impairment scores, reduced the volume of cerebral infarction and brain water content, and attenuated neuronal apoptosis. Additionally, the possible mechanisms of MSCs-derived sEVs for attenuating neuronal apoptosis were inhibiting microglia-mediated neuroinflammation. Thus, MSCs-derived sEVs might be regarded as a novel insight for cerebral ischemic stroke. However, further mechanistic studies, therapeutic safety, and clinical trials are required. Systematic review registration. PROSPERO CRD42022312227.


Asunto(s)
Isquemia Encefálica , Vesículas Extracelulares , Células Madre Mesenquimatosas , Daño por Reperfusión , Accidente Cerebrovascular , Animales , Roedores , Isquemia Encefálica/terapia , Accidente Cerebrovascular/terapia
8.
J Agric Food Chem ; 70(39): 12525-12534, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-36135333

RESUMEN

Patients with ulcerative colitis (UC) have been found to be frequently associated with secondary liver injury (SLI). In this study, we investigated the protective effect of GA on dextran sodium sulfate (DSS)-induced SLI in mice and its mechanism. The SLI was established by adding 4% DSS in the drinking water of mice, and the effects of GA (5, 20 mg/kg, p.o., once a day for 7 days) in hepatic tissues were analyzed. HepG2 cells were induced by lipopolysaccharide (LPS) to detect the effect of GA on ferroptosis and the underlying mechanism. Pathological damage was determined by H&E. Liver parameters (AST and ALT), antioxidant enzyme activities (MDA and SOD), and the level of Fe2+ in the liver were detected by kits. Cytokine levels (TNF-α, IL-1ß, and IL-6) and Gpx4 activity in the liver were detected by ELISA. Finally, the activation of nuclear factor erythroid 2-like 2 (Nrf2) was detected to explore the mechanism. The results indicated that GA significantly attenuated DSS-induced hepatic pathological damage, liver parameters, and cytokine levels and increased the antioxidant enzyme activities. Moreover, GA attenuated ferroptosis in DSS-induced liver injury and upregulated Gpx4 expression in DSS-induced mice. Mechanistic experiments revealed that GA activated Nrf2 in mice. Taken together, this study demonstrates that GA can alleviate ferroptosis in SLI in DSS-induced colitis mice, and its protective effects are associated with activating the Nrf2-Gpx4 signaling pathway.


Asunto(s)
Colitis , Agua Potable , Ferroptosis , Animales , Antioxidantes/farmacología , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/genética , Citocinas/metabolismo , Sulfato de Dextran/efectos adversos , Diarilheptanoides , Interleucina-6/farmacología , Lipopolisacáridos/efectos adversos , Hígado/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
Food Funct ; 13(5): 2985-2997, 2022 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-35195119

RESUMEN

As a disease caused by an impaired intestinal epithelial barrier, imbalanced flora, immune imbalance and genetic susceptibility, ulcerative colitis (UC) is becoming a health threat for all ages. Lactobacillus acidophilus (L. acidophilus), an attracting probiotic, has already been confirmed to improve immune dysfunction, stabilize intestinal microflora, and combat gut disorders. However, no studies have focused on the effects of different forms of L. acidophilus on UC, and its mechanism involved in the mitophagy/NLRP3 inflammasome pathway has not been reported. In this study, we found that compared with the heat-killed L. acidophilus and the culture supernatant of L. acidophilus, the live L. acidophilus (La) has the optimal therapeutic effect on UC rats. Furthermore, La evidently increased the contents of SCFAs, inhibited NLRP3 inflammasome and facilitated autophagy. SCFAs regulated by La balanced inflammation homeostasis and improved intestinal barrier dysfunctions in vitro and in vivo, which was achieved by activating the mitophagy/NLRP3 inflammasome pathway. Moreover, PCR analysis indicated that the aforementioned effects of SCFAs regulated by La may be due to the activation of G protein-coupled receptors. These findings provided guidance for the application of L. acidophilus in daily life and provided a new molecular target for interactions among L. acidophilus, its metabolites and host immunity.


Asunto(s)
Lactobacillus acidophilus , Probióticos/farmacología , Animales , Colitis Ulcerosa/prevención & control , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitofagia/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Probióticos/administración & dosificación , Probióticos/química , Ratas , Ratas Sprague-Dawley
10.
Eur J Pharmacol ; 883: 173358, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32710952

RESUMEN

Recently, it has reported that many inflammatory bowel disease (IBD) patients were contracted secondary liver injury. Monotropein (MON), an iridoid glycoside, is demonstrated to possess protective effects on acute colitis mice due to its anti-inflammatory activities. However, it was remained unknown whether MON could inhibit secondary liver injury caused by IBD. The aim of the present study was to investigate the protective roles and mechanisms of MON on secondary liver injury in chronic colitis mice model. In this study, 2% Dextran sodium sulfate (DSS) was used to induce mice model of chronic colitis. The results showed that MON attenuated DSS-induced hepatic pathological damage, liver parameters, infiltration of macrophages and cytokines levels. Furthermore, we found that MON attenuated liver injury through suppressing the activation of the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway and down-regulating the activity of NLRP3 (NOD-, LRR- and pyrin domain-containing 3) inflammasome. All the data indicated that MON may be an effective therapeutic reagent to attenuate secondary liver injury induced by chronic colitis.


Asunto(s)
Antiinflamatorios/farmacología , Colitis/tratamiento farmacológico , Inflamasomas/metabolismo , Iridoides/farmacología , Hepatopatías/prevención & control , Hígado/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Enfermedad Crónica , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Inflamasomas/genética , Hígado/metabolismo , Hígado/patología , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatopatías/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , FN-kappa B/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Transducción de Señal , Receptor Toll-Like 4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA