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

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(8)2022 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-35457248

RESUMEN

Cisplatin-based chemotherapy causes intestinal mucositis, which causes patients immense suffering and hinders the process of cancer treatment. Dioscin is a natural steroid saponin that exhibits strong anti-inflammatory and immunomodulatory properties. Herein, we investigate the protective effect of dioscin on cisplatin induced mucositis in rats from the perspective of gut microbiota and intestinal barrier. We established a rat model of intestinal mucositis by tail vein injection of cisplatin, and concurrently treated with dioscin oral administration. Parameters, such as body weight, diarrheal incidence, and D-Lactate levels, were assessed in order to evaluate the effects of dioscin on intestinal mucositis in rats. Furthermore, biological samples were collected for microscopic gut microbiota, intestinal integrity, and immune inflammation analyses to elucidate the protective mechanisms of dioscin on intestinal mucositis. The results revealed that administration of dioscin significantly attenuated clinical manifestations, histological injury and inflammation in mucositis rats. Besides this, dioscin markedly inhibited the gut microbiota dysbiosis induced by cisplatin. Meanwhile, dioscin partially alleviated junctions between ileum epithelial cells and increased mucus secretion. Moreover, dioscin effectively inhibited the TLR4-MyD88-NF-κB signal transduction pathway and reduced the secretion of subsequent inflammatory mediators. These results suggested that dioscin effectively attenuated cisplatin-induced mucositis in part by modulating the gut microflora profile, maintaining ileum integrity and inhibiting the inflammatory response through the TLR4-MyD88-NF-κB pathway.


Asunto(s)
Microbioma Gastrointestinal , Mucositis , Animales , Cisplatino/farmacología , Diosgenina/análogos & derivados , Humanos , Inflamación/tratamiento farmacológico , Mucositis/inducido químicamente , Mucositis/tratamiento farmacológico , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Ratas , Transducción de Señal , Receptor Toll-Like 4/metabolismo
2.
Animals (Basel) ; 14(11)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38891683

RESUMEN

In order to explore the therapeutic effect of Resveratrol (Res)-activated Natural Killer (NK) cells on canine mammary tumors, this study employed a range of assays, including wound healing, colony formation, Transwell, flow cytometry, and Western blot experiments, to investigate the impact of Res-pretreated NK cells on canine mammary tumor cells in vitro. Additionally, a tumor-bearing mouse model was utilized to further analyze the therapeutic effects of Res-pretreated NK cells in vivo. The results showed that Res enhances the capacity of NK cells to induce apoptosis, pyroptosis, and ferroptosis in canine breast tumor cells, while also augmenting their influence on the migration, invasion, and epithelial-mesenchymal transition of these cells. Furthermore, pretreatment of NK cells with Res significantly amplified their inhibitory effect on breast tumor growth in vivo and promoted tumor tissue apoptosis. Additionally, Res enhanced the recruitment of NK cells to other immune cells in the body. In summary, Res has been shown to enhance the anti-breast-tumor effect of NK cells both in vitro and in vivo, offering a new avenue for optimizing immunotherapy for canine breast tumors.

3.
Theriogenology ; 217: 103-112, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38271764

RESUMEN

The development of cisplatin resistance is one of the major causes of mammary cancer treatment failure, and is associated with changes in Sox4 gene expression. To investigate the characteristic changes that occur in canine mammary gland tumor (CMGT) cells following the development of acquired cisplatin resistance, along with the relationship between these changes and the Sox4 gene. We constructed cisplatin-resistant cell line, CHMpCIS, from the cell line CHMp, which was isolated from the primary lesion of a malignant CMGT. The biological characteristics of these cells were examined by Western blot analysis, Transwell assays, and mammosphere formation assays. Compared to CHMp cells, CHMpCIS cells exhibited elevated cisplatin resistance, apoptotic escape ability, enhanced epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) features, in addition to over-activation of the Wnt/ß-catenin signaling pathway and increased Sox4 protein. In CMGT cases, CMGT tissues (CMGTT) expressed higher levels of Sox4 protein and mRNA compared to adjacent tissues (CAMGTT). We found that these changes were inhibited by silencing of Sox4 expression in CHMpCIS cells. Furthermore, activation of the Wnt/ß-catenin signaling pathway increased Sox4 expression levels through a positive feedback loop. These results suggested that CHMpCIS cells circumvented the damage caused by cisplatin through altering the expression of the Sox4 gene and activating the Wnt/ß-catenin pathway, thereby changing the cellular biological characteristics.


Asunto(s)
Cisplatino , beta Catenina , Animales , Perros , Cisplatino/farmacología , Línea Celular Tumoral , beta Catenina/genética , Factores de Transcripción , Vía de Señalización Wnt/genética , Proliferación Celular
4.
Chem Biol Interact ; 380: 110536, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37179038

RESUMEN

Cisplatin (CIS) is a widely used clinical chemotherapeutic agent for solid malignancies such as lung, testicular and ovarian cancers, but the development of nephrotoxicity has limited the use of this class of drugs. Some studies have shown that aspirin can reduce cisplatin-induced nephrotoxicity, but the mechanism of protection is not yet clear. By establishing a mouse model of cisplatin-induced acute kidney injury and a mouse model of aspirin combination, we observed a reduction in creatinine, blood urea nitrogen, and tissue damage, thus verifying that aspirin can alleviate cisplatin-induced acute kidney injury in mice. Aspirin was found to have a significant protective effect against cisplatin-induced acute kidney injury, as evidenced by the reduction in levels of ROS, NO, and MDA and the increase in T-AOC, CAT, SOD, and GSH. Furthermore, aspirin was observed to down-regulate the expression of pro-inflammatory factors TNF-α, NF-κB, IL-1ß, and IL-6 mRNA and proteins, increase the expression of BAX and Caspase3 as indicators of apoptosis, decrease the expression of Bcl-2, and improve the reduced expression of mtDNA, ATP content, ATPase activity and mitochondrial respiratory chain complex enzyme-related genes ND1, Atp5b, and SDHD. These findings suggest that the protective effects of aspirin are associated with its anti-inflammatory, antioxidant, anti-apoptotic properties, and its ability to maintain mitochondrial function, as demonstrated by the detection of AMPK-PGC-1α pathway-related genes. The results showed that the reduced expression of p-AMPK and mitochondrial production-related mRNA PGC-1α, NRF1, and TFAM in the kidney tissue of mice in the cisplatin group was alleviated by the effect of aspirin, indicating that aspirin could activate the p-AMPK, regulate mitochondrial production and alleviate cisplatin acute kidney injury through the AMPK-PGC-1α pathway. In summary, certain doses of aspirin protect the body from acute kidney injury by alleviating the cisplatin-induced inflammatory response oxidative stress, mitochondrial dysfunction, and apoptosis. Further studies have shown that the protective effect of aspirin is associated with AMPK-PGC-1α pathway activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Lesión Renal Aguda , Ratones , Animales , Proteínas Quinasas Activadas por AMP/metabolismo , Cisplatino/toxicidad , Cisplatino/metabolismo , Aspirina/farmacología , Transducción de Señal , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/prevención & control , Estrés Oxidativo , Apoptosis , ARN Mensajero/metabolismo , Riñón
5.
Food Funct ; 13(19): 10210-10224, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-36111853

RESUMEN

Intestinal injury is one of the main side-effects of cisplatin (CP) chemotherapy, severely limiting the clinical application of CP. Vitamin D3 is an essential nutrient for mammals and exists in a wide range of foods; it regulates immune function and reduces oxidative stress. However, the effect of vitamin D3 on CP-induced intestinal injury is not elucidated. This is the first study to investigate the relationship between ferroptosis and the protective effect of vitamin D3 on CP-induced intestinal injury. An animal model of CP-induced intestinal injury was established to evaluate the effect of vitamin D3 on CP-induced intestinal injury and elucidate the underlying mechanisms. We found that vitamin D3 alleviated intestinal barrier injury and the abnormal morphological structure in CP-induced intestinal injury mice. Vitamin D3 suppressed oxidative stress by increasing the antioxidant capacity, inhibiting the accumulation of ROS and MDA, and reducing intestinal inflammatory responses. Vitamin D3 also decreased excessive mitochondrial fission and increased mitochondrial ATPase activity by inhibiting ROS production, which further alleviated the accumulation of ROS. We also confirmed the involvement of ferroptosis in CP-induced intestinal injury in our animal model using ferrostatin-1 (Fer-1) intervention. Vitamin D3 decreased iron accumulation and reversed GPX4 and DHODH down-regulation. In conclusion, vitamin D3 protected against CP-induced intestinal injury by inhibiting ferroptosis and alleviating oxidative stress and ROS-mediated excessive mitochondrial fission, suggesting that it may be a novel and promising candidate to prevent CP-induced intestinal injury.


Asunto(s)
Cisplatino , Ferroptosis , Adenosina Trifosfatasas , Animales , Antioxidantes/farmacología , Colecalciferol/farmacología , Cisplatino/efectos adversos , Hierro/farmacología , Mamíferos , Ratones , Dinámicas Mitocondriales , Estrés Oxidativo , Especies Reactivas de Oxígeno
6.
Antioxidants (Basel) ; 11(12)2022 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-36552651

RESUMEN

Acute kidney injury (AKI) is a clinical syndrome with high morbidity and mortality worldwide, and there is currently no effective means to prevent it. Dioscin is naturally present in the dioscoreaceae plants and has antioxidant and anti-inflammatory effects. Here, we found that dioscin is protective against cisplatin-induced AKI. Pathological and ultrastructural observations revealed that dioscin reduced renal tissue lesions and mitochondrial damage. Furthermore, dioscin markedly suppressed reactive oxygen species and malondialdehyde levels in the kidneys of AKI rats and increased the contents of glutathione and catalase. In addition, dioscin dramatically reduced the number of apoptotic cells and the expression of pro-apoptotic proteins in rat kidneys and human renal tubular epithelial cells (HK2). Conversely, the protein levels of anti-ferroptosis including GPX4 and FSP1 in vivo and in vitro were significantly enhanced after dioscin treatment. Mechanistically, dioscin promotes the entry of Nrf2 into the nucleus and regulates the expression of downstream HO-1 to exert renal protection. However, the nephroprotective effect of dioscin was weakened after inhibiting Nrf2 in vitro and in vivo. In conclusion, dioscin exerts a reno-protective effect by decreasing renal oxidative injury, apoptosis and ferroptosis through the Nrf2/HO-1 signaling pathway, providing a new insight into AKI prevention.

7.
Int Immunopharmacol ; 111: 109111, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35933746

RESUMEN

Cisplatin is the most widely prescribed drug in chemotherapy, but its gastrointestinal toxicity reduces therapeutic efficacy. Oxidative stress and inflammation are considered to be the main pathogenesis of cisplatin-induced intestinal toxicity. Dioscin is a steroidal saponin with potential anti-cancer, antioxidant, and anti-inflammatory activities. In this study, we established a rat model of intestinal injury by tail vein injection of cisplatin, and intragastrically administered dioscin to evaluate its effect on intestinal injury. Biochemical markers, western blotting, qRT-PCR and histopathological staining were used to analyze intestinal injury according to various molecular mechanisms. The results revealed that dioscin significantly inhibited cisplatin-induced intestinal mucosal damage and decreased DAO levels in rats. Furthermore, dioscin activated the Nrf2/HO-1 pathway to increase the level of antioxidant enzymes and reduce the levels of MDA and H2O2. In addition, dioscin pretreatment significantly reduced ileum epithelial NLRP3 inflammasome formation and decreased the levels of inflammatory factors compared with the cisplatin group. In parallel, Nrf2 inhibitor ML385 blocked the therapeutic effect of dioscin in rat with cisplatin-induced intestinal toxicity. In terms of mechanisms, dioscin reversed cisplatin-induced up-regulation of MAPKs and up-regulated p-PI3K and p-AKT levels. Meanwhile, dioscin potently promoted Wnt3A/ß-catenin signaling to relieve cisplatin-induced proliferation inhibition. In conclusion, our study suggests that dioscin could ameliorate the cisplatin-induced intestinal toxicity by reducing oxidative stress and inflammation.


Asunto(s)
Diosgenina , Factor 2 Relacionado con NF-E2 , Animales , Antioxidantes/uso terapéutico , Cisplatino/toxicidad , Diosgenina/análogos & derivados , Diosgenina/farmacología , Diosgenina/uso terapéutico , Peróxido de Hidrógeno/farmacología , Inflamación/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Ratas
8.
Food Nutr Res ; 662022.
Artículo en Inglés | MEDLINE | ID: mdl-35844954

RESUMEN

Background: Troxerutin is a flavonoid compound and possesses potential anti-cancer, antioxidant, and anti-inflammatory activities. Besides, cisplatin is one of the most widely used therapeutic agents, but the clinical uses of cisplatin are often associated with multiple side effects, among which nephrotoxicity is more common. Objective and design: This study explored the protective effects of troxerutin (150 mg kg-1 day-1 for 14 days) against cisplatin-induced kidney injury and the potential mechanism using Wistar rats as an experimental mammalian model. Results: We discovered that troxerutin could significantly alleviate cisplatin-induced renal dysfunction, such as increased levels of blood urea nitrogen and creatinine (P < 0.01), as well as improved abnormal renal tissue microstructure and ultrastructure. Additionally, troxerutin significantly decreased malondialdehyde (MDA), hydrogen peroxide (H2O2), NO, inducible nitric oxide synthase (iNOS) levels (P < 0.01), p-NF-κB p65/NF-κB p65, TNF-α, Pro-IL-1ß, IL-6, B cell lymphoma-2 (Bcl-2)/Bcl-xl associated death promoter (Bad), Cytochrome C (Cyt C), Cleaved-caspase 9, Cleaved-caspase 3, and Cleaved-caspase 8 protein levels (P < 0.01) in the kidney tissues of cisplatin-treated rats; and increased superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), total antioxidant capacity (T-AOC) activities (P < 0.01), IL-10, Bcl-2 protein levels (P < 0.01). Conclusion: These results suggested that the underlying mechanism might be attributed to the regulation of Phosphoinositide 3 kinase/Protein kinase B (PI3K/AKT) pathway via enhancing MAP4 expression to attenuate cellular apoptosis, alleviating oxidative stress and inflammatory response.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA