Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Cell Mol Biol (Noisy-le-grand) ; 66(3): 72-78, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32538750

RESUMEN

This study aimed to explore the expression of lncRNA-metastasis associated lung adenocarcinoma transcript 1 (lncRNA-MALAT1) in breast cancer (BC) patients and its influences on the prognosis of the patients. A total of 120 BC patients admitted to our hospital were enrolled as a BC group, of which 58 patients at I/II stage were treated with breast-conserving surgery as an operation group, and the other 62 patients at III/IV stage were treated with neoadjuvant chemotherapy combined with breast-conserving surgery as a combination group. Meantime, 100 healthy people in physical examination during the same period were enrolled as a normal group. The expression of serum lncRNA-MALAT1 in the subjects was determined, and the expression in BC patients and its influences on the patients were analyzed. LncRNA-MALAT1 was over-expressed in patients from the BC group, and the area-under-the-curve (AUC) of it for diagnosing BC was 0.911. After treatment, the expression of lncRNA-MALAT1 in the operation group and the combination group significantly decreased, and the expression of it in patients with good prognosis was greatly lower than that in patients with poor prognosis. The AUC of lncRNA-MALAT1 for predicting poor prognosis was 0.838, and TNM staging, pathological differentiation, tumor diameter, and lncRNA-MALAT1 were independent prognostic factors for poor prognosis of the patients. Furthermore, low expression of lncRNA-MALAT1 was associated with a relatively high 5-year overall survival (OS) of BC patients. The expression of lncRNA-MALAT1 was up-regulated in BC patients, while it was down-regulated in BC patients treated with breast-conserving surgery combined with neo-adjuvant chemotherapy, so lncRNA-MALAT1 can be used as a potential indicator for early diagnosis and prognosis prediction of BC patients.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Adulto , Anciano , Femenino , Humanos , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Modelos de Riesgos Proporcionales , ARN Largo no Codificante/metabolismo , Factores de Riesgo , Análisis de Supervivencia
2.
J Inflamm Res ; 17: 2513-2530, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38699595

RESUMEN

Purpose: Explore the therapeutic effects and regulatory mechanism of Qingyi Decoction (QYD) on severe acute pancreatitis (SAP) associated acute lung injury (ALI). Methods: We identified the constituents absorbed into the blood of QYD based on a network pharmacological strategy. The differentially expressed genes from the GEO database were screened to identify the critical targets of QYD treatment of SAP-ALI. The SAP-ALI rat model was constructed.Some methods were used to evaluate the efficacy and mechanism of QYD in treating SAP-ALI. LPS-stimulated pulmonary microvascular endothelial cell injury simulated the SAP-induced pulmonary endothelial injury model. We further observed the therapeutic effect of QYD and CDK5 plasmid transfection on endothelial cell injury. Results: 18 constituents were absorbed into the blood, and 764 targets were identified from QYD, 25 of which were considered core targets for treating SAP-ALI. CDK5 was identified as the most critical gene. The results of differential expression analysis showed that the mRNA expression level of CDK5 in the blood of SAP patients was significantly up-regulated compared with that of healthy people. Animal experiments have demonstrated that QYD can alleviate pancreatic and lung injury inflammatory response and reduce the upregulation of CDK5 in lung tissue. QYD or CDK5 inhibitors could decrease the expression of NFAT5 and GEF-H1, and increase the expression of ACE-tub in SAP rat lung tissue. Cell experiments proved that QYD could inhibit the expression of TNF-α and IL-6 induced by LPS. Immunofluorescence results suggested that QYD could alleviate the cytoskeleton damage of endothelial cells, and the mechanism might be related to the inhibition of CDK5-mediated activation of NFAT5, GEF-H1, and ACE-tub. Conclusion: CDK5 has been identified as a critical target for pulmonary endothelial injury of SAP-ALI. QYD may partially alleviate microtubule disassembly by targeting the CDK5/NFAT5/GEF-H1 signaling pathway, thus relieving SAP-induced pulmonary microvascular endothelial cell injury.

3.
Clin Transl Oncol ; 25(6): 1629-1640, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36630025

RESUMEN

PURPOSE: Breast cancer is one of the leading causes of tumor death worldwide in female, and the five-year overall survival of breast cancer patients remains poor. It is an urgent need to seek novel target for its treatment. Synaptotagmin 13 (SYT13) is a synaptic vesicle transporting protein that regulates the malignant phenotypes of various cancers. However, its role in breast cancer is still unclear. The current study aimed to investigate the effects of SYT13 on the progression of breast cancer. METHODS: Twenty-five pairs of breast cancer tissues and non-tumor tissues were obtained to assess the expression of SYT13. We manually modified the expression of SYT13 in MCF-7 and MDA-MB-231 cells. CCK-8 assay, EdU staining, and cell cycle analysis were carried out to measure the proliferated ability of cells. Annexin V/PI and TUNEL assays were used to detect the apoptotic ability of cells. Wound healing and transwell assays were employed to evaluate the migrated and invasive ability of breast cancer cells. RESULTS: The results revealed that the mRNA and protein levels of SYT13 were higher in breast cancer tissues and cell lines. Knockdown of SYT13 inhibited the cell proliferation and induced cell cycle arrest in G1 phase of MCF-7 cells by downregulating cyclin D1 and CDK4, as well as upregulating p21. The migration and invasion of MCF-7 cells were repressed by the loss of SYT13 via the gain of E-cadherin and the loss of vimentin. Overexpression of SYT13 in MDA-MB-231 cells led to the opposite effects. Silencing of SYT13 induced the apoptosis ability of MCF-7 cells by the upregulation of bax and the downregulation of bcl-2. Moreover, we found that SYT13 depletion suppressed the FAK/AKT signaling pathway. PF573228 (a FAK inhibitor) and MK2206 (an AKT inhibitor) reversed the SYT13 overexpression-induced promotion of proliferation, migration, and invasion of MDA-MB-231 cells. CONCLUSION: The results indicated that SYT13 promoted the malignant phenotypes of breast cancer cells by the activation of FAK/AKT signaling pathway.


Asunto(s)
Neoplasias de la Mama , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Sinaptotagminas , Femenino , Humanos , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Células MCF-7 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sinaptotagminas/genética , Sinaptotagminas/metabolismo
4.
Hum Cell ; 36(4): 1501-1515, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37165255

RESUMEN

E3 ubiquitin ligase Zinc and Ring Finger 2 (ZNRF2) has been demonstrated to be engaged in the development of multiple cancers. Nevertheless, the function of ZNRF2 in breast cancer (BC) still unclear. In this work, we firstly analyzed the differentially expressed genes in BC by bioinformatics and found that ZNRF2 was highly expressed in BC. Consistently, we further confirmed that ZNRF2 was upregulated in BC tissues compared with adjacent normal tissues, and this was positively correlated with the poor prognosis and the higher pathological grades of patients with BC. Functional assays performed on HCC1937 and MCF-7 cells indicated that silencing of ZNRF2 suppressed cell proliferation, as evidenced by the decrease in the expression of cyclin A, PCNA and cyclin D1. Flow cytometry and Hoechst staining showed that knockdown of ZNRF2 induced cell apoptosis, which was verified by the upregulation of apoptosis genes such as Bax, cleaved PARP and Bim. ZNRF2 knockdown also inhibited in vivo tumor growth. But, instead, ZNRF2-overexpressed BC cells exhibited obvious malignant phenotypes. Additionally, we observed that cAMP response element binding protein 1 (CREB1) directly bound to the promoter sequence of ZNRF2 and thus activating its transcription, suggesting that ZNRF2 is transcriptionally regulated by CREB1. Additionally, ZNRF2 knockdown could reverse the proliferation-promoting action of CREB1 on BC cells, Hence, this study demonstrated that ZNRF2 might serve as a prospective therapeutic target for BC.


Asunto(s)
MicroARNs , Neoplasias , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Zinc , Línea Celular Tumoral , Oncogenes/genética , MicroARNs/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Apoptosis/genética , Movimiento Celular/genética , Neoplasias/genética
5.
Int J Nanomedicine ; 18: 6743-6761, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38026528

RESUMEN

Background: Numerous preclinical investigations have exhibited the beneficial impact of emodin (EMO) on the management of severe acute pancreatitis (SAP)-associated acute lung injury (ALI). However, the potential of EMO to mitigate organ damage through the modulation of exosome (Exo)-specific miRNA expression profiles remains unclear. Methods: The SAP rat model was established by retrograde injection of 5% sodium taurocholate into the pancreatic bile duct. Rats received intragastric administration of EMO at 2 h and 12 h post-modeling. Plasma and bronchoalveolar lavage fluid (BALF)-derived exosomes were isolated and purified from SAP rats treated with EMO. The therapeutic effects of these Exos in SAP rats were assessed using hematoxylin-eosin staining and measurement of inflammatory factor levels. MicroRNA (miRNA) sequencing was conducted on plasma and BALF-derived Exos, and rescue experiments were performed to investigate the function of NOVEL miR-29a-3p in the treatment of SAP using EMO. Results: EMO exhibits ameliorative effects on pancreatic and lung injury and inflammation in rats with SAP. Plasma/BALF-derived Exos from EMO-treated SAP rats also have therapeutic effects on SAP rats. The miRNA expression profile of plasma and BALF-derived Exos in SAP rats underwent significant changes upon exposure to EMO. In particular, 34 differentially expressed miRNAs (DEmiRNAs) were identified when comparing BALF-SAP+EMO-Exo and BALF-SAP-Exo. 39 DEmiRNAs were identified when comparing plasma-SAP+EMO-Exo to plasma-SAP-Exo. We found that SAP rats treated with Exos derived from BALF exhibited a more potent therapeutic response than those treated with Exos derived from plasma. EMO may rely on NOVEL-rno-miR-29a-3p expression to prevent pulmonary injury in SAP rats. Conclusion: The mechanism of action of EMO is observed to have a significant impact on the miRNA expression profile of Exos derived from plasma and BALF in SAP rats. NOVEL-rno-miR-29a-3p, which is specific to Exos, and is derived from BALF, may play a crucial role in the therapeutic efficacy of EMO.


Asunto(s)
Lesión Pulmonar Aguda , Emodina , Exosomas , MicroARNs , Pancreatitis , Ratas , Animales , Pancreatitis/inducido químicamente , Pancreatitis/tratamiento farmacológico , Emodina/farmacología , Enfermedad Aguda , Exosomas/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , MicroARNs/genética , MicroARNs/metabolismo
6.
J Healthc Eng ; 2022: 7146338, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35186236

RESUMEN

Breast cancer remains a dangerous disease, and delving the molecular mechanism of breast cancer is still necessary. To illustrate the role of miR-511-5p, TCGA database was used to excavate the abundance of miR-511-5p, and the miR-511-5p level was measured in the pathological tissues and tumor cell lines. Moreover, the targets of miR-511-5p were identified with miRDIP and GEPIA and then were used for functional enrichment analysis. Besides, the targets of miR-511-5p were analyzed with the protein-protein interaction (PPI) network for the hub nodes, and then the expression levels of the hub nodes were visualized with the GEPIA database. The results showed that miR-511-5p was significantly downregulated in multiple types of tumor samples in the online database, and the downregulated miR-511-5p was also found in pathological tissues and tumor cell lines. Moreover, 48 genes were identified as the potential targets of miR-511-5p by miRDIP and GEPIA databases and enriched in cell cycle, PI3K/AKT, and P53 pathways. Besides, seven genes including BRCA1, FN1, CCNE1, CCND1, CHEK1, BUB3, and CDC25A were identified as the hub nodes by the PPI network, and CCNE1 and CHEK1 were confirmed to be related with the prognostic survival of the patients with breast cancer. In conclusion, the proofs in this study suggest that reduced miR-511-5p was a biomarker event for breast cancer, and CCNE1 and CHEK1 served as potential targets of miR-511-5p to involve the progression of breast cancer.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Biología Computacional , Femenino , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasas , Pronóstico
7.
Clin. transl. oncol. (Print) ; 25(6): 1629-1640, jun. 2023. graf
Artículo en Inglés | IBECS (España) | ID: ibc-221195

RESUMEN

Purpose Breast cancer is one of the leading causes of tumor death worldwide in female, and the five-year overall survival of breast cancer patients remains poor. It is an urgent need to seek novel target for its treatment. Synaptotagmin 13 (SYT13) is a synaptic vesicle transporting protein that regulates the malignant phenotypes of various cancers. However, its role in breast cancer is still unclear. The current study aimed to investigate the effects of SYT13 on the progression of breast cancer. Methods Twenty-five pairs of breast cancer tissues and non-tumor tissues were obtained to assess the expression of SYT13. We manually modified the expression of SYT13 in MCF-7 and MDA-MB-231 cells. CCK-8 assay, EdU staining, and cell cycle analysis were carried out to measure the proliferated ability of cells. Annexin V/PI and TUNEL assays were used to detect the apoptotic ability of cells. Wound healing and transwell assays were employed to evaluate the migrated and invasive ability of breast cancer cells. Results The results revealed that the mRNA and protein levels of SYT13 were higher in breast cancer tissues and cell lines. Knockdown of SYT13 inhibited the cell proliferation and induced cell cycle arrest in G1 phase of MCF-7 cells by downregulating cyclin D1 and CDK4, as well as upregulating p21. The migration and invasion of MCF-7 cells were repressed by the loss of SYT13 via the gain of E-cadherin and the loss of vimentin. Overexpression of SYT13 in MDA-MB-231 cells led to the opposite effects. Silencing of SYT13 induced the apoptosis ability of MCF-7 cells by the upregulation of bax and the downregulation of bcl-2. Moreover, we found that SYT13 depletion suppressed the FAK/AKT signaling pathway. PF573228 (a FAK inhibitor) and MK2206 (an AKT inhibitor) reversed the SYT13 overexpression-induced promotion of proliferation, migration, and invasion of MDA-MB-231 cells (AU)


Asunto(s)
Humanos , Femenino , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Células MCF-7 , Transducción de Señal , Sinaptotagminas/genética , Sinaptotagminas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA