Linlentz7136

Z Iurium Wiki

In 2016 WHO classification, EBV +DLBCL of the elderly was replaced by EBV+ DLBCL NOS. This is due to the fact that many young patients of EBV+ DLBCL were found in recent years.

In this study, we retrospectively analyzed clinical features and survival outcomes of EBV positive DLBCL patients in different age groups. All the patients treated at a single center.

When we use different ages (40, 50 and 60 years old) as cutoffs, the prevalence of EBV positive DLBCL was 12.0%, 12.3% and 13.0% in younger patients and 19.0%, 15.4% and 13.8% in elder patients respectively. Whatever the age cutoff was, EBV positive associated with unfavorable clinical prognosis in elder groups. When we use 40 and 50 years old as age cutoffs, poor impacts of EBV positive on overall survival and progression-free survival were observed only in elder patients, but not in younger patients. It should be noted that when we use 60 years old as age cutoff, the results were the opposite.

EBV+ DLBCL patients with age of 40 to 60 years old showed poorer prognostic features than EBV- DLBCL patients; however, patients in other age groups did not show evident differences in prognosis between EBV+ DLBCL patients and EBV- DLBCL patients. This finding was not reported before.

EBV+ DLBCL patients with age of 40 to 60 years old showed poorer prognostic features than EBV- DLBCL patients; however, patients in other age groups did not show evident differences in prognosis between EBV+ DLBCL patients and EBV- DLBCL patients. This finding was not reported before.

The aim of this study was to elucidate the role of FOXC2-AS1 in promoting the proliferative ability and inhibiting apoptosis of melanoma by silencing p15, thereafter regulating the progression of melanoma.

FOXC2-AS1 levels in melanoma patients with or without metastasis and those with the tumor in different stages were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Regulatory effects of FOXC2-AS1 on viability and apoptosis in melanoma cells were assessed, and subcellular distribution of FOXC2-AS1 was analyzed. Subsequently, the interactions of FOXC2-AS1 with EZH2 and SUZ12 were explored by RNA-Binding Protein Immunoprecipitation (RNA-RIP) assay. Through chromatin immunoprecipitation (ChIP) assay, the role of FOXC2-AS1 to regulate p15 transcription by recruiting EZH2 was verified. At last, regulatory effects of FOXC2-AS1/p15 axis on viability and apoptosis in melanoma cells were investigated.

It was found that FOXC2-AS1 was upregulated in melanoma tissues, especially those with metastasis or stage II-IV. Debio1143 Melanoma patients expressing high level of FOXC2-AS1 showed worse survival than those with low level. Knockdown of FOXC2-AS1 inhibited viability, and stimulated apoptosis in A375 and sk-mel-110 cells. Besides, P15 level was upregulated in melanoma cells transfected with si-FOXC2-AS1, and FOXC2-AS1 was mainly distributed in cytoplasm. RNA-RIP assay confirmed that FOXC2-AS1 was mainly enriched in anti-EZH2 and aniti-SUZ12. Knockdown of EZH2 could markedly upregulate protein level of p15 in melanoma cells. Furthermore, it was verified that FOXC2-AS1 inhibited p15 transcription via recruiting EZH2, and the knockdown of p15 could partially reverse the regulatory effects of FOXC2-AS1 on viability and apoptosis in melanoma.

FOXC2-AS1 stimulates proliferative ability in melanoma via silencing p15.

FOXC2-AS1 stimulates proliferative ability in melanoma via silencing p15.

Glioblastoma (GBM) is a deadly brain cancer that seriously threatens the lives of patients. Moreover, various microRNAs (miRNAs) have been found to be involved in the progression of GBM. The purpose of this study is to preliminarily elucidate the regulatory mechanism of miR-362 in GBM.

The abnormal expression of miR-362 and MAPK1 was detected by RT-qPCR or Western blot analysis in GBM tissues and cells. CCK-8 and transwell assays were performed to measure cell proliferation, migration and invasion. The relationship between miR-362 and MAPK1 was confirmed by luciferase reporter assay.

MiR-362 expression was reduced in GBM tissues and cells. The decreased expression of miR-362 predicted poor prognosis in GBM patients. Functionally, overexpression of miR-362 inhibited the proliferation and metastasis of GBM cells. In addition, miR-362 directly targets MAPK1. MAPK1 was negatively correlated with miR-362 expression in GBM. Moreover, MAPK1 was upregulated and served as a tumor promoter in GBM. More importantly, the upregulation of MAPK1 weakened the inhibitory effect of miR-362 on cell proliferation and metastasis in GBM.

MiR-362 restrains cell proliferation and metastasis in GBM by targeting MAPK1, indicating that miR-362 functions as a tumor suppressor in GBM.

MiR-362 restrains cell proliferation and metastasis in GBM by targeting MAPK1, indicating that miR-362 functions as a tumor suppressor in GBM.

Given that FK506 binding protein 51 (FKBP51) is upregulated in multiple cancers, we designed the present study to characterize its role as well as underlying regulatory mechanisms in glioma in the presence and absence of the chemotherapeutic carmustine (BCNU).

Through lentiviral overexpression and shRNA knockdown of FKBP51, we examined the effects on BT325 glioma cell proliferation, migration and invasion using quantitative reverse transcription PCR (qRT-PCR), CCK-8 assay, flow cytometry, and transwell assay.

The upregulation of FKBP51 resulted in significantly decreased BT325 cell proliferation and cell viability, cell cycle arrest, reduced BCNU chemosensitivity and AKT pathway inactivation. However, FKBP51-overexpressed BT325 cells showed enhanced migration and invasion, which was supported by corresponding increase in phosphorylated IKKα (p-IKKα), MMP-2, and MMP-9 levels, as well as increased NF-κB p65 nuclear translocation. By contrast, FKBP51-suppressed BT325 cells showed excessive proliferation and BCNU resistance due to increased p-AKT activation and attenuated migration and invasion.

We demonstrated that the effects of FKBP51 on BT325 glioma cell proliferation, migration, invasion and BCNU chemosensitization are modulated via the AKT and NF-κB pathways. Furthermore, our findings suggest the potential of FKBP51 as a prognostic glioma biomarker and an indicator of patient response to chemotherapy.

We demonstrated that the effects of FKBP51 on BT325 glioma cell proliferation, migration, invasion and BCNU chemosensitization are modulated via the AKT and NF-κB pathways. link2 Furthermore, our findings suggest the potential of FKBP51 as a prognostic glioma biomarker and an indicator of patient response to chemotherapy.

The purpose of this study was to detect the relative expression of long non-coding ribonucleic acid (lncRNA) in non-homologous end joining pathway 1 (LINP1) in papillary thyroid cancer (PTC) tissues and cells, and to investigate the molecular mechanisms of abnormal expression and biological function of LINP1.

The relative expression of LINP1 in PTC tissues and cells was detected via quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR), and the impact of small interfering (si)-LINP1 on the proliferative capacity of PTC cells was studied using Cell Counting Kit-8 (CCK-8) and colony formation assays. After the expression of LINP1 in PTC cells was interfered, flow cytometry was applied to determine the changes in cell cycle distribution and apoptosis rate. The transcription factors binding to the promoter region of LINP1 were predicted by bioinformatics. Next, qRT-PCR assay was adopted to measure the changes in LINP1 expression after interference in the expression of signal transducer and acs of STAT1 and LINP1 that the expression of molecular marker (Phosphorylation AMPK, p-AMPK) of the AMPK signaling pathway was altered but the expression of total AMPK did not change.

The transcription factor STAT1 promotes the expression of LINP1 in PTC, and highly expressed LINP1 facilitates the proliferation and inhibits the apoptosis of PTC by suppressing the AMPK signaling pathway.

The transcription factor STAT1 promotes the expression of LINP1 in PTC, and highly expressed LINP1 facilitates the proliferation and inhibits the apoptosis of PTC by suppressing the AMPK signaling pathway.

This study aims to explore the diagnostic and prognostic values of Lysophosphatidic acid receptor 5 (LPAR5) in non-small-cell lung cancer (NSCLC) and its regulatory effects on biological functions of NSCLC cells.

NSCLC and adjacent non-tumoral tissues were collected for analyzing differential levels of LPAR5 by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). link3 Clinical information of recruited NSCLC patients was collected for assessing the diagnostic and prognostic values of LPAR5. In vitro regulation of LPAR5 on proliferative and migratory potentials of H1299 and SPC-A1 cells was examined by Cell Counting Kit-8 (CCK-8) and transwell assay, respectively. In addition, in vivo regulation of LPAR5 on the growth rate of NSCLC in nude mice was detected by tumorigenicity assay. The interaction between LPAR5 and its downstream target MLLT11 was determined by rescue experiments.

LPAR5 was upregulated in NSCLC tissues than adjacent non-tumoral ones. High level of LPAR5 predicted higher rates of lymphatic metastasis and distant metastasis, as well as worse overall survival and progression-free survival in NSCLC. Knockdown of LPAR5 not only attenuated in vitro proliferative and migratory abilities in H1299 and SPC-A1 cells, but also slowed down in vivo growth of NSCLC in nude mice. MLLT11 was upregulated in NSCLC tissues, and displayed a positive correlation to LPAR5. Overexpression of MLLT11 was able to reverse the attenuated in vitro proliferative and migratory abilities, and the suppressed in vivo growth of NSCLC because of LPAR5 knockdown.

LPAR5 stimulates proliferative and migratory potentials in NSCLC by positively regulating MLLT11, which can be served as an effective diagnostic marker for early stage NSCLC.

LPAR5 stimulates proliferative and migratory potentials in NSCLC by positively regulating MLLT11, which can be served as an effective diagnostic marker for early stage NSCLC.

The purpose of this study was to investigate the role of microRNA-488-3p in the proliferation, invasion and migration of lung cancer cells and to further explore the potential regulatory mechanisms.

MicroRNA-488-3p expression in 46 pairs of tumor tissue and paracancerous tissue specimens collected from non-small cell lung cancer (NSCLC) patients were measured through quantitative real-time polymerase chain reaction (qRT-PCR) method, and the interplay between microRNA-488-3p expression and some clinical indicators of these subjects was also analyzed. In addition, microRNA-488-3p overexpression models were constructed in NSCLC cell lines, and then Cell Counting Kit-8 (CCK-8) test and transwell assays were carried out to evaluate the effect of microRNA-488-3p on the NSCLC cell functions. Furthermore, bioinformatics analysis and luciferase reporter gene assay were carried out to uncover the potential interaction between microRNA-488-3p and its downstream gene ADAM9.

QPCR results revealed that microRNA-488-3s in NSCLC patients, can inhibit the malignant progression of NSCLC cells by modulating ADAM9 expression.

It can be concluded that microRNA-488-3p, which is associated with the incidence of metastasis in NSCLC patients, can inhibit the malignant progression of NSCLC cells by modulating ADAM9 expression.

Autoři článku: Linlentz7136 (Nguyen Lillelund)