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.
BMC Cancer ; 24(1): 638, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789960

RESUMEN

Emerging evidence suggests the dysregulation of long non-coding RNAs (lncRNAs) involved in pancreatic cancer (PC). However, the function of LINC00930 in PC has not been elaborated. In this study, we found that LINC00930 was significantly down-regulated in PC cell lines and tissues, and associated with tumor size, lymphatic metastasis, TNM stage and poor prognosis. According to the bioinformatics database, the downregulation of LINC00930 was a common event in PC associated with prognosis and EMT. Overexpression of LINC00930 inhibited the aggressive cancer phenotypes including proliferation, metastasis and epithelial-mesenchymal transition (EMT) of PC in vitro and in vivo. Bioinformatics and dual-luciferase reporter assay indicated that miR-6792-3p could directly bind to LINC00930. Additionally, the Zinc finger and BTB domain containing 16 (ZBTB16) was significantly declined in PC, which was predicted to be the downstream gene of miR-6792-3p. MiR-6792-3p mimic rescued the decreased proliferation, metastasis and EMT caused by ZBTB16 in PC cells. The LINC00930/miR-6792-3p/ZBTB16 axis was associated with the malignant progression and process of PC. The relative expression of LINC00930 was negatively correlated with the expression of miR-6792-3p and was closely linked with ZBTB16 levels in PC. LINC00930 might serve as a potential prognostic biomarker and therapeutic target for PC.


Asunto(s)
Proliferación Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , MicroARNs , Neoplasias Pancreáticas , ARN Largo no Codificante , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , MicroARNs/metabolismo , Proliferación Celular/genética , Animales , Ratones , Línea Celular Tumoral , Masculino , Femenino , Pronóstico , Persona de Mediana Edad , Movimiento Celular/genética
2.
Int J Biol Macromol ; 269(Pt 1): 131826, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679256

RESUMEN

The tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) is characterized by deposition of desmoplastic matrix (including collagen and hyaluronic acid). And the interactions between tumor-associated macrophages (TAMs) and tumor cells play a crucial role in progression of PDAC. Hence, the appropriate model of tumor cell-macrophage interaction within the unique PDAC TME is of significantly important. To this end, a 3D tumor niche based on dual-crosslinking gelatin methacrylate and hyaluronic acid methacrylate hydrogels was constructed to simulate the desmoplastic tumor matrix with matching compressive modulus and composition. The bionic 3D tumor niche creates an immunosuppressive microenvironment characterized by the downregulation of M1 markers and upregulation of M2 markers in TAMs. Mechanistically, RNA-seq analysis revealed that the PI3K-AKT signaling pathway might modulate the phenotypic balance and recruitment of macrophages through regulating SELE and VCAM-1. Furthermore, GO and GSEA revealed the biological process of leukocyte migration and the activation of cytokine-associated signaling were involved. Finally, the 3D tumor-macrophage niches with three different ratios were fabricated which displayed increased M2-like polarization and stemness. The utilization of the 3D tumor niche has the potential to provide a more accurate investigation of the interplay between PDAC tumor cells and macrophages within an in vivo setting.


Asunto(s)
Carcinoma Ductal Pancreático , Gelatina , Ácido Hialurónico , Metacrilatos , Microambiente Tumoral , Macrófagos Asociados a Tumores , Gelatina/química , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Humanos , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Metacrilatos/química , Metacrilatos/farmacología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Hidrogeles/química , Línea Celular Tumoral , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Transducción de Señal/efectos de los fármacos
4.
ACS Biomater Sci Eng ; 9(9): 5347-5360, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37561610

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) features high recurrence rates and intensified lethality, accompanied by stiffening of the extracellular matrix (ECM) microenvironment, which is mainly due to the deposition, remodeling, and cross-linking of collagen. Boosted stemness plays an essential role during occurrence and progression, which indicates a poor prognosis. Therefore, it is of great importance to understand the effect of the underlying interaction of matrix stiffness and stemness on PDAC. For this purpose, a methacrylated gelatin (GelMA) hydrogel with tunable stiffness was applied for incubating MIA PaCa-2 and PANC-1 cells. The results demonstrated that compared to the soft group (5% GelMA, w/v), the expression of stemness-related genes (SOX2, OCT4, and NANOG) in the stiff group (10% GelMA, w/v) displayed pronounced elevation as well as sphere formation. Intriguingly, we also observed that matrix stiffness regulated autophagy of PDAC, which played a momentous role in stemness promotion. In order to clarify the underlying relationship between matrix stiffness-mediated cell autophagy and stemness, rescue experiments with rapamycin and chloroquine were conducted with transmission electron microscopy, immunofluorescence staining, sphere formation, and qRT-PCR assays to evaluate the level of stemness and autophagy. For exploring the molecular mechanism in depth, RNA-seq and differential expression of miRNAs were carried out, which may sensor and respond to matrix stiffness during the regulation of stemness and autophagy. In conclusion, we validated that blocking autophagy repressed the stemness induced by matrix stiffness in PDAC and provided a potential therapeutic strategy for this aggressive cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Biomimética , Línea Celular Tumoral , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Autofagia/genética , Microambiente Tumoral , Neoplasias Pancreáticas
5.
Cancer Lett ; 512: 38-50, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33971282

RESUMEN

Pancreatic cancer (PC) is one of the most lethal malignant tumors and has the lowest survival rate due to early metastasis and drug resistance. Exosomes derived from bone marrow mesenchymal stem cells (BM-MSCs) have emerged as crucial regulators of the progression of various tumors. These vesicles contain abundant circRNAs that have important biological functions. This study aimed to elucidate the role of exosomal circRNAs in PC progression. In this study, we successfully isolated BM-MSCs from human bone marrow based on their surface marker expression and osteogenic and adipogenic differentiation potential. We found that BM-MSC-derived exosomes significantly reduced the invasion, migration, and proliferation of PC cells, as well as tumor stemness. According to whole-transcriptome resequencing and clustering heat map analysis, we identified the key molecule circ_0030167 and miR-338-5p, its downstream target. We revealed that circ_0030167 mainly regulates miR-338-5p, enhances Wif1 expression, and inhibits the Wnt8/ß-catenin pathway, thereby inhibiting the stemness of PC cells and tumor progression. Overall, BM-MSC exosomal circ_0030167 contributes to the progression and stemness of PC cells via the miR-338-5p/wif1/Wnt 8/ß-catenin axis. Our study provides a new perspective for the treatment of PC.


Asunto(s)
Células de la Médula Ósea/metabolismo , Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/terapia , ARN Circular/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Exosomas/patología , Humanos , MicroARNs/genética , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Circular/genética , ARN Circular/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
6.
J Cell Mol Med ; 25(7): 3654-3664, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33683827

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an invasive and aggressive cancer that remains a major threat to human health across the globe. Despite advances in cancer treatments and diagnosis, the prognosis of PDAC patients remains poor. New and more effective PDAC therapies are therefore urgently required. In this study, we identified a novel host factor, namely the LncRNA TP73-AS1, as overexpressed in PDAC tissues compared to adjacent healthy tissue samples. The overexpression of TP-73-AS1 was found to correlate with both PDAC stage and lymph node metastasis. To reveal its role in PDCA, we targeted TP73-AS1 using LnRNA inhibitors in a range of pancreatic cancer (PC) cell lines. We found that the inhibition of TP73-AS1 led to a loss of MMP14 expression in PC cells and significantly inhibited their migratory and invasive capacity. No effects of TP73-AS1 on cell survival or proliferation were observed. Mechanistically, we found that TP73-AS1 suppressed the expression of the known oncogenic miR-200a. Taken together, these data highlight the prognostic potential of TP73-AS1 for PC patients and highlight it as a potential anti-PDAC therapeutic target.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 14 de la Matriz/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/fisiología , Animales , Apoptosis , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Metástasis Linfática/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Pronóstico
7.
J Biomater Sci Polym Ed ; 32(2): 151-165, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32867627

RESUMEN

Islet transplantation is considered as one of the promising treatment options for curing diabetes. However, the extracellular matrix (ECM) is destroyed during the process of islet isolation and extraction, which leads to decreased islet activity in vitro. ECM-based biomaterials which used to reconstruct the microenvironment of cells have been applied in various fields. In this study, an electrospinning hybrid scaffolds with silk fibroin (SF) and pig pancreatic decellularized extracellular matrix (P-dECM) have been prepared to mimic the islet ECM in vivo. Furthermore, the activity and function of islet were evaluated in vitro. The microstructures, hydrophilia and the main components of scaffolds were characterized by SEM, contact angle analysis and immunohistochemical experiment. The toxicity of stents was assessed by MTT assay. Cell activity and function were estimated by the live-dead cell staining, immunofluorescence, glucose-stimulated insulin secretion assay and q-PCR. A nanofiber scaffold with good hydrophilicity, non-toxic and retention of key ECM components has been obtained, which can improve the survival and promote and function of islets. This scaffold can be a promising candidate for pancreatic tissue engineering and provides a new strategy for islet transplantation.


Asunto(s)
Fibroínas , Animales , Biomimética , Matriz Extracelular , Seda , Porcinos , Ingeniería de Tejidos , Andamios del Tejido
8.
Artif Organs ; 44(12): e532-e551, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32671848

RESUMEN

The development of pancreatic extracellular matrices enriched with insulin-secreting ß-cells is a promising tissue engineering approach to treat type 1 diabetes. However, its long-term therapeutic efficacy is restricted by the defensive mechanism of host immune response and the lack of developed vascularization as appropriate after transplantation. Platelet-rich plasma (PRP), as an autologous platelet concentrate, contains a large number of active factors that are essential for the cell viability, vascularization, and immune regulation. In this study, we have incorporated pancreatic extracellular matrix (PEM) with PRP to develop a three-dimensional (3D) injectable PEM-PRP hydrogel to coculture and transplant rat insulinoma cells (INS-1) and human umbilical vein endothelial cells (HUVECs). Results from this study demonstrated that PEM-PRP hydrogel mimicked the biochemical compositions of native extracellular matrices, and possessed the enhanced elastic modulus and resistance to enzymatic degradation that enabled biomaterials to maintain its volume and slowly degrade. Additionally, PEM-PRP hydrogel could release growth factors in a sustained manner. In vitro, PEM-PRP hydrogel significantly promoted the viability, insulin-secreting function, and insulin gene expression of gel encapsulated INS-1 cells. Moreover, HUVECs encapsulated in PEM-PRP hydrogel were found to constitute many lumen-like structures and exhibited high expression of proangiogenic genes. In vivo transplantation of PEM-PRP hydrogel encapsulated with INS-1 cells and HUVECs improved the viability of INS-1 cells, promoted vascularization, inhibited the host inflammatory response, and reversed hyperglycemia of diabetic rats. Our study suggests that the PEM-PRP hydrogel offers excellent biocompatibility and proangiogenic property, and may serve as an effective biomaterial platform to maintain the long-term survival and function of insulin-secreting ß cells.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Matriz Extracelular/trasplante , Hidrogeles/administración & dosificación , Plasma Rico en Plaquetas , Ingeniería de Tejidos/métodos , Animales , Materiales Biocompatibles , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Supervivencia Celular , Técnicas de Cocultivo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ensayo de Materiales , Páncreas/citología , Páncreas/metabolismo , Páncreas/patología , Ratas , Estreptozocina/administración & dosificación , Estreptozocina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...