Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Pharmacol Res ; 197: 106980, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37944835

RESUMEN

The microRNAs (miRNAs) are potent regulators of tumorigenesis in various cancers, especially pancreatic cancer. The abnormal expression of miRNAs can be observed in tumor cells. Noteworthy, miRNAs could be transferred by exosomes as small extracellular vesicles in regulation of carcinogenesis. This research focused on exploring the roles and mechanisms of exosomal miR-484, derived from human bone marrow mesenchymal stem cells (hBMSCs), in the context of molecular interactions and regulation of mitochondrial metabolism. Exosomes were isolated for the examination of miR-484 expression. The impacts of hBMSCs-derived exosomal miR-484 on pancreatic cancer cells were studied using various assays. Evaluation of mitochondrial function and metabolism was performed. Wnt/MAPK pathway-related protein expression was assessed, and an in vivo tumor xenograft model was utilized to examine the functions. Our findings demonstrated a decreased miR-484 expression in pancreatic cancer cells. However, hBMSCs-derived exosomal miR-484 inhibited the proliferation and migration of these cells, while inducing apoptosis. Moreover, miR-484 led to an upsurge in reactive oxygen species production, a decrease in ATP levels, and a disruption in mitochondrial metabolism. In vivo analyses showed that hBMSCs-derived exosomal miR-484 lessened tumor size and weight, while also suppressing the expression of mitochondrial biomarkers. Further, there was a decline in ß-catenin and p-p38 protein levels both in vitro and in vivo. The addition of LiCl restored the disrupted mitochondrial metabolism. Conclusively, our results suggest that hBMSCs-derived exosomal miR-484 mitigates the malignant transformation and mitochondrial metabolism of pancreatic cancer by deactivating the Wnt/MAPK pathway.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/genética , MicroARNs/genética , Mitocondrias , Páncreas , Carcinogénesis , Transformación Celular Neoplásica , Neoplasias Pancreáticas
3.
Cancer Lett ; 530: 16-28, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35033590

RESUMEN

DNA damage repair is a major barrier for chemotherapy efficacy of pancreatic ductal adenocarcinoma (PDAC), including the efficacy of platinum-based and gemcitabine/nab-paclitaxel treatments. N6-methyladenosine modifications (m6A) have recently been reported to play a role in homologous recombination (HR) repair of DNA double strand breaks (DSBs); however, the mechanism of action remains unknown. Our previous work indicated that fisetin may be a promising anti-tumour agent that induces DNA damage. In this study, we reported that fisetin induced DSBs and suppressed HR repair through m6A modification in PDAC cells. The m6A writer ZC3H13 and PHF10, which is a subunit of the PBAF chromatin remodelling complex, were identified as the main molecules affected by fisetin treatment. To our knowledge, it's the first time that PHF10 was found and involved in the DNA damage response. PHF10 loss-of-function resulted in elevated recruitment of γH2AX, RAD51, and 53BP1 to DSB sites and decreased HR repair efficiency. Moreover, ZC3H13 knockdown downregulated the m6A methylation of PHF10 and decreased PHF10 translation in a YTHDF1-dependent manner. In conclusion, our study demonstrates that fisetin enhanced DSBs via ZC3Hl3-mediated m6A modification of PHF10, which may provide insight into novel therapeutic approaches for PDAC.


Asunto(s)
Adenosina/análogos & derivados , Daño del ADN/genética , Reparación del ADN/genética , Flavonoles/genética , Proteínas de Homeodominio/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Proteínas de Unión al ARN/genética , Adenosina/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Humanos , Recombinasa Rad51/genética , Reparación del ADN por Recombinación/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Neoplasias Pancreáticas
4.
iScience ; 23(8): 101431, 2020 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-32798974

RESUMEN

Immunotherapy in pancreatic ductal adenocarcinoma (PDAC) treatment faces serious challenges, due particularly to the poor immunogenicity. Cancer cell-derived small extracellular vesicles (sEVs) play important roles in damaging the immune system. However, the effects of pancreatic cancer-derived sEVs on T lymphocytes are unknown. Here we investigated changes in phenotypes and signal transduction pathways in sEVs-treated T lymphocytes. We identified the overexpression of immune checkpoint proteins PD-1, PD-L1, CTLA4, and Tim-3 and the enrichment of FOXP3+ Treg cluster in sEVs-treated T lymphocytes by CyTOF. Gene set enrichment analysis revealed that DNA damage response and metabolic pathways might be involved in sEVs-induced Tregs. ATM, AMPK, SIRT1, SIRT2, and SIRT6 were activated sequentially in sEVs-treated T lymphocytes and essential for sEVs-upregulated expressions of FOXO1A, FOXO3A, and FOXP3. Our study reveals the impact and mechanism of pancreatic cancer cell-derived sEVs on T lymphocytes and may provide insights into developing immunotherapy strategies for PDAC treatment.

5.
Oncol Lett ; 19(3): 2062-2070, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32194703

RESUMEN

Pancreatic cancer (PC) has a poor prognosis due to the lack of effective molecular biomarkers for early diagnosis. Recent studies have investigated the use of exosomal microRNAs (exmiRs) as diagnostic biomarkers in cancer. The present study examined exmiR-21, exmiR-10b and exmiR-212-3p expression in patients with PC and healthy individuals. The expression levels of exmiR-21, exmiR-10b and exmiR-212-3p were examined in the peripheral blood plasma of 36 patients with PC and 65 healthy controls, using tethered cationic lipoplex nanoparticle biochip. The levels of exmiR-21 in the plasma of 34 mice were also evaluated. The expression levels of exmiR-21 and exmiR-10b were significantly greater in patients with PC compared with the control group. The receiver operating characteristic (ROC) analysis indicated that exmiR-21 had better diagnostic performance (P=0.0003; AUC, 0.7171) compared with the other two exmiRs. The diagnostic value of exmiR-21 improved when combined with exmiR-10b (P<0.0001; AUC, 0.791). Furthermore, exmiR-21 was capable of distinguishing patients with early-stage PC from controls and advanced-stage PC (P<0.05, early stage vs. healthy; P<0.001, early stage vs. advanced stage). The results of the present study revealed that the plasma levels of exmiR-21 and exmiR-10b were upregulated in patients with PC. The ROC analyses indicated that exmiR-21 had the best diagnostic performance among the three exmiRs. Furthermore, exmiR-21 was capable of discriminating patients with early-stage PC from healthy controls. These findings indicate the potential of determining the expression of exmiR-21 from serum using a tethered cationic lipoplex nanoparticle biochip as a novel non-invasive strategy for the early diagnosis of PC.

6.
J Cancer ; 10(4): 911-917, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30854097

RESUMEN

Background: The oncogene DEK, which was originally identified as part of the protein product of the DEK-CAN fusion oncogene, has been shown to promote tumorigenesis in a variety of cancer cell types. However, little is known about the expression and role of DEK in pancreatic ductal adenocarcinoma (PDAC), which is one of the most refractory malignant tumors worldwide and has poor prognosis. Our study aimed to understand the role of DEK in the development and progression of pancreatic adenocarcinoma. Materials and methods: We used western blotting and immunohistochemistry to examine the expression of DEK in pancreatic adenocarcinoma cells and tissues. We analyzed the correlation between DEK expression and clinicopathological characteristics and prognosis in 163 pancreatic adenocarcinoma patients. Results: Protein levels of DEK in pancreatic adenocarcinoma tissues (76/136, 55.9%) were significantly higher than those in adjacent non-tumor tissues (16.2%, 22/136). A high expression level of DEK was associated with poor prognosis (P<0.001).In addition, the combination of CA19-9 and DEK expression (P<0.001) was a better prognostic indicator than CA19-9 expression alone (P=0.012). Conclusions: DEK may play a significant role as a valuable biomarker in the development and progression of pancreatic adenocarcinoma. The combination of DEK and CA19-9 improves the prognostic prediction in patients with pancreatic adenocarcinoma.

7.
Cell Death Dis ; 10(2): 142, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30760707

RESUMEN

Pancreatic cancer is one of the most aggressive tumors and patients have poor survival rates. Fisetin, a natural flavonoid, was recently reported to have antitumor effects in various cancer models. Autophagy is a conserved catabolic process that maintains cellular homoeostasis in response to stress, and together with apoptosis, determines cell fate. Herein, we examined the effect of fisetin on pancreatic cancer. We reveal that fisetin inhibits PANC-1 cell proliferation using a real-time cell analysis system. Moreover, the in vivo antitumor effect of fisetin was verified in pancreatic cancer using a luciferase-expressing murine xenograft pancreatic cancer model. We found that the AMPK/mTOR signaling pathway was enhanced after fisetin treatment; however, autophagy was not diminished by adding the AMPK inhibitor compound C. Thus, we hypothesized that an another autophagy regulating pathway existed. RNA-seq analysis revealed that the unfolded protein response pathway, which is activated by ER stress, was enriched. We also found that the stress-induced transcription factor p8 was increased in fisetin-treated PANC-1 cells, and that fisetin-induced autophagy was blocked by silencing p8. We revealed that p8-dependent autophagy was AMPK-independent, and that p8 regulated ATF6, ATF4, and PERK in response to ER stress via p53/PKC-α-mediated signaling. Furthermore, mitophagy was associated with Parkin and PINK1 in response to mitochondrial stress. Interestingly, ATF4 and ATF6 were increased in cells treated with fisetin and compound C. Moreover, inhibiting the AMPK/mTOR pathway with compound C may upregulate p8-dependent autophagy. Thus, there may be crosstalk between the AMPK/mTOR and p8-dependent pathways.


Asunto(s)
Autofagia/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Flavonoides/uso terapéutico , Mitocondrias/metabolismo , Neoplasias Pancreáticas/genética , Animales , Flavonoides/farmacología , Flavonoles , Humanos , Ratones , Neoplasias Pancreáticas/patología
8.
J Cancer ; 9(4): 650-659, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29556322

RESUMEN

Background: P38α is a ubiquitous protein kinase, which plays diverse roles in cancers. Surprisingly, P38α functions vary markedly in different cancers (e.g., cancer suppressor vs cancer promoter). However, there is no report on the expression of P38α, the family's most important member, in pancreatic ductal adenocarcinoma (PDAC) and its association with clinicoathological parameters and patients' prognosis. Materials and methods: We retrospectively analyzed 152 patients who underwent surgery and were pathologically diagnosed with PDAC from September 2013 to September 2015. We used immunohistochemistry to detect P38α expression in tumor and adjacent normal tissues. The significance of the association between P38α and clinicopathological parameters was evaluated using the χ² test and t tests. The Kaplan-Meier method was used to assess the association between P38α expression and preoperative carbohydrate antigen 19-9 (CA19-9) levels and patients' overall survival. The Cox regression model was used to analyze the association between clinicopathological parameters, P38α and preoperative CA19-9 levels, and prognosis. Statistical significance was defined as P < 0.05. Results: P38α was expressed in 63.16% tumor tissues of PDAC, which was significantly higher compared with the adjacent normal tissues (26.32%, P < 0.001). High expression of P38α was associated with patients' histological grade (P = 0.013), lymphatic metastasis (P = 0.025) and TNM stage (P = 0.048). The median survival time of the P38α-high group was 9.2 months, which was shorter compared with that of the P38α-low group (17.3 months, P = 0.011). The median survival time of the CA19-9 > 43.63 group was 11.1 months shorter than that of the CA19-9 < 43.63 group (24.8 months, P < 0.001). The Cox regression model revealed that age (P = 0.003), lymphatic invasion (P = 0.015), TNM stage (P = 0.003), histological grade (P < 0.001), preoperative CA19-9 (P = 0.049), and P38α expression (P = 0.008) were statistically significant independent risk factors affecting prognosis. Specifically, overall survival was 28.4 months in the P38α-low and CA19-9 < 43.63 groups, 16.3 months in the P38α-high or CA19-9 > 43.63 groups, and 9.7 months in the P38α-high and CA19-9 > 43.63 groups (P < 0.001). Conclusions: High expression of P38α was significantly associated with histological grade, lymphatic metastasis, TNM stage and prognosis in patients with PDAC. P38α and preoperative CA19-9 levels were independent risk factors affecting the prognosis of PDAC patients. High expression of p38α and preoperative carbohydrate antigen 19-9 indicate poor prognosis in patients with PDAC.

9.
J Cancer ; 9(3): 578-583, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29483963

RESUMEN

Recent studies show that CD36 plays a key role in the occurrence and development of tumors, especially in the metastasis of tumors. However, the expression and role of CD36 has not been reported in pancreatic cancer. This study is aimed to explore the expression of CD36 in pancreatic cancer and corresponding non-tumor normal tissues, and its correlation with clinicopathological features and prognosis of pancreatic cancer patients. By analyzing the chip results of database GSE16515, we found that there was significant differential expression of CD36 in pancreatic cancer and corresponding non-tumor normal tissues. In this study, western blot and immunohistochemistry were used to show that the expression of CD36 in pancreatic cancer cells and tissues is significantly lower than that in corresponding non-tumor normal tissues. By statistically analyzing clinical and pathological data, we found that low expression of CD36 predicts lower TNM staging and CA19-9 levels, but larger tumor size and poor survival prognosis. These findings indicated that CD36 can be used as a predictor of clinicopathological features and prognosis, but the contradiction is worthy of our further study.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA