Activity

  • Welsh McCabe posted an update 4 days, 12 hours ago

    Gestational diabetes mellitus (GDM) is a serious life‑threatening disease that affects the mother and fetus. However, the pathogenesis of GDM is still unclear. microRNAs (miRs) play vital roles in the regulation of various cell functions. The present study aimed to investigate the effects of miR‑875‑5p and thioredoxin reductase 1 cytoplasmic (TXNRD1) in GDM rats and analyze the associated underlying mechanism. A GDM rat model was induced using an intraperitoneal injection of streptozotocin. miR‑875‑5p knockdown plasmids or TXNRD1 knockdown plasmids were injected into the rats via the caudal vein. miR‑875‑5p and TXNRD1 expression in the serum were detected using reverse transcription‑quantitative PCR (RT‑qPCR) or western blot (WB) analyses. The fasting blood‑glucose (FBG), fasting serum insulin, triglyceride and high density lipoprotein levels were detected by specific commercial kits. The inflammatory response and the induction of oxidative stress were analyzed by assessing the expression of associated markers via WB, RT‑qPCR or commercial kits. The pancreatic and placental injuries were detected by hematoxylin and eosin staining. The results indicated that miR‑875‑5p expression levels were downregulated, whereas TXNRD1 levels were upregulated in GDM rats compared with normal pregnancy rats. miR‑875‑5p significantly regulated TXNRD1 expression in GDM rats. miR‑875‑5p silencing notably reduced FBG and insulin resistance, which was accompanied by reduced expression levels of blood lipid and pro‑inflammatory markers as well as reduced oxidative stress. However, the effects of miR‑875‑5p could be reversed by TXNRD1 silencing. Therefore, the present study indicated that miR‑875‑5p regulated IR and inflammation by targeting TXNRD1 in GDM rats. miR‑875‑5p and TXNRD1 may be considered as potential targets for treating GDM.Semaphorin 4D (Sema4D) is highly expressed in a variety of tumors and is associated with high invasion, poor prognosis and poor therapeutic response. However, the expression and role of Sema4D in leukemia remains unclear. The present study investigated the expression of Sema4D in pediatric leukemia and its effects in leukemia cells. The results demonstrated that Sema4D protein was highly expressed in peripheral blood mononuclear cells of patients with pediatric leukemia, and high levels of soluble Sema4D were also observed in the plasma of these patients. Sema4D knockdown induced cell cycle arrest in G0/G1 phase, inhibited proliferation and promoted apoptosis in BALL‑1 cells, while Sema4D overexpression exhibited the opposite effect. In Jurkat cells, Sema4D knockdown inhibited proliferation and promoted apoptosis, while Sema4D overexpression decreased the abundance of the cells in the G0/G1 phase of the cell cycle and promoted proliferation. Sema4D overexpression also increased the migratory capacity of Jurkat cells and the invasive capacity of BALL‑1 cells. The phosphorylation level of PI3K was decreased in both Sema4D knocked‑down Jurkat and BALL‑1 cells, and the phosphorylation level of ERK was decreased in Sema4D knocked‑down BALL‑1 cells. The phosphorylation levels of PI3K, ERK and AKT were elevated in patients with pediatric leukemia, and were correlated to the increased Sema4D expression. Sema4D overexpression was associated with a shorter overall survival in patients with acute myeloid leukemia. Overall, the results of the present study indicated that Sema4D serves an important role in leukemia development by activating PI3K/AKT and ERK signaling, and it may be used as a potential target for the diagnosis and treatment of leukemia.Colorectal cancer (CRC) is a lethal and common malignancy worldwide. ICG-001 mouse Non‑coding (nc)RNAs have been shown to modulate tumor progression in several types of cancer. The present study aimed to investigate the role of hsa_circ_0000212 in CRC, as a sponge of microRNA (miR)‑491. The expression levels of miR‑491 and forkhead box P4 (FOXP4) were analyzed using data from The Cancer Genome Atlas. The association between miR‑491 and FOXP4 and the clinicopathological characteristics were also analyzed. A novel circular (circ)RNA, hsa_circ_0000212, was found to sponge miR‑491 based on bioinformatics analysis. The potential binding site between miR‑491 and FOXP4 or circ‑0000212 was validated using luciferase and RNA immunoprecipitation assays. The expression levels and distribution of circ‑0000212 was also determined. Cell Counting Kit‑8 and colony formation assays were performed to determine the role of miR‑491 or circ‑0000212 on the proliferation of the CRC cells. Decreased miR‑491 or increased FOXP4 expression levels were associated with the pathological stage in patients with CRC. In addition, miR‑491 inhibited cell proliferation by targeting FOXP4. circ‑0000212 was increased in CRC tissues and was predominantly localized in the cytoplasm. Furthermore, circ‑0000212 augmented viability of the CRC cells by sponging miR‑491 and modulating FOXP4. In conclusion, circ‑0000212 may serve as a novel tumor‑promoter and drug target in CRC.Subsequently to the publication of the above paper, the authors have realized that they should have credited a Professor René Csuk [Martin‑Luther‑Universität Halle‑Wittenberg, Halle (Saale), Germany] for the use of a compound that his group synthesized in the study. Therefore, the authors wish to include the following text in the Acknowledgements’ section of the Declarations ‘The authors are grateful to Professor Rene Csuk, Department of Organic Chemistry, Martin‑Luther University Halle‑Wittenberg, for providing us with the rhodamine B‑conjugated oleanolic acid derivative (RhodOA)’. All the named authors agree to this Corrigendum, and apologize to Professor Csuk for the upset and inconvenience caused. [the original article was published in Oncology Reports 44 1169‑1183, 2020; DOI 10.3892/or.2020.7666].To date, there is no effective therapy available for the treatment of castration‑resistant prostate cancer (CRPC), and patients generally succumb to the disease within 2 to 4 years. In the progression of CRPC, androgen receptor (AR) and its splice variants play critical roles. Hence, it is necessary to develop a drug to inhibit the expression and activity of the full‑length and splice variants of AR for the treatment of CRPC. Erastin, as the first discovered drug to induce ferroptosis, has been studied in various types of cancer. However, there are few studies focusing on the relationship between erastin and AR. In the present study, western blotting, and sulforhodamine B cell viability, glutathione, lipid peroxidation and reactive oxygen species assays were performed to verify the ferroptosis of CRPC cells; reverse transcription‑quantitative polymerase chain reaction, dual‑luciferase reporter, and lentiviral packaging and lentivirus‑infected cell assays were employed to evaluate how erastin affects AR. A mouse xenograft assay was used to determine the underlying mechanism in vivo.