Korsholmkaufman0444
However, unlike TNF-α, EGF expression did not result in the upregulation of inflammatory molecules, including intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and monocyte chemoattractant protein-1. These results indicate that EGF preferentially upregulated the protective mediator A20 over proinflammatory factors. selleck compound To our knowledge, the present study is the first to demonstrate that EGF induced A20 expression by activating the MEK1/MSK1/p-p65 (Ser276) signaling pathway without causing an apparent inflammatory response. These results may further extend our understanding of liver inflammation and tumor development.
Nonalcoholic fatty liver disease (NAFLD) is a progressive liver disease, which may develop into end-stage liver disease and endanger human life. miR-122-5p may be related to the progression of NAFLD disease, but the specific regulation mechanism is still unknown. It is helpful for us to optimize the prevention or treatment strategy of NAFLD.
Real-time PCR was applied to test miR-122-5p and KIF5B in serum, rat liver tissue induced by high fat diet (HFD), and primary hepatocytes exposed to oleic acid ester and palmitate (FFA) of NAFLD patients. The role of miR-122-5p on inflammatory factors (MCP-1, TNF-α, IL-10) and liver injury markers (AST, ALT) in vivo and in vitro was analyzed.
miR-122-5p and KIF5B were both highly expressed in NAFLD patients' serum, rat liver tissue and primary hepatocytes, while KIF5B was low expressed. miR-122-5p expression enhanced with the increase of HFD feeding time. The dual luciferase reporter gene assay system confirmed that there was a targeting relationship between miR-122-5p and KIF5B, indicating that KIF5B and protein level were evidently up-regulated in primary hepatocytes. Down-regulation of miR-122-5p was helpful to improve the liver weight/body weight ratio (liver index) level of rats, as well as the levels of triglyceride (TG), inflammatory factors and liver injury markers in liver tissues in vivo and in vitro. Phosphorylation of AMPK/AKT pathway-related proteins and fat metabolism-related factors in rat liver tissues and cells in primary hepatocytes were notably reduced, while down-regulation of miR-122-5p was helpful to restore activation of the pathway and increase the level of fat metabolism-related factors.
Decrease of miR-122-5p can target and enhance KIF5B, which can be applied for treating NAFLD.
Decrease of miR-122-5p can target and enhance KIF5B, which can be applied for treating NAFLD.Cushing disease has a very high mortality rate and glucocorticoid resistance caused by GR down-regulation is one major reason of mortality. Although HIF1α signaling and GR signaling are involved in the pathogenesis of pituitary adenomas, it's unclear whether and how these two essential pathways could cross-talk with each other. Here, we performed a comprehensive study to investigate the reciprocal effects of HIF1α and GR on each other in AtT20 cell lines and explored the potential therapeutic effect of HIF1α inhibitor in in-vivo mouse model. We find that hypoxia up-regulated the promoter activity, mRNA and protein levels of GR and the induced GR protein was localized in cytosol. On the other hand, GR activation by its agonist DEX increased HIF1α protein through post-transcriptional mechanism. However, hypoxia and DEX show differential synergistic effects on HIF1α and GR. In hypoxia-DEX condition, HIF1α protein was further up-regulated but mainly localized in cytosol while GR was trapped and degraded in cytosol via UPS pathway. Further Co-IP experiments demonstrate that DNA binding domain of GR can interact with PASb domain of HIF1α. In a in-vivo mouse model of Cushing's disease, HIF1α inhibitor reduced HIF1α and GR protein levels, reduced tumor size and lowered the plasma concentrations of ACTH and corticosterone. In summary, we find that a novel HIF1α-GR crosstalk contributes to the pathogenesis of pituitary adenomas and HIF1α inhibitor shows potential therapeutic effects for Cushing's disease.
The aim was to research the POU2F1 related genes and mechanism during the progress of immune escape of lung cancer.
Lung cancer cell lines (H1993, HCC827, A549, H2228, H3122 and H1975) and Human normal lung epithelial cell line (BEAS-2B) were involved in this study. Overexpression or knockdown of POU2F1 was processed in lung cancer cells. POU2F1, PD-L1 and CRK expression in cells were detected by WB and RT-PCR. Flow cytometry and immunofluorescence was used to detect PD-L1 expression on the cell surface. Luciferase reporter detected the promoter activity of CRK. C57BL/6 mice models with knocked down of of POU2F1 were constructed. After tumor formation, anti-PD-1 was administered to detect tumor suppressing ability. IHC assay showed the number of intratumoral CD3+, CD8+, GranzB+ T cells.
POU2F1 and PD-L1 were positively correlated in lung cancer cell lines. Overexpression of POU2F1 promoted the expression level of PD-L1 in lung cancer cells. POU2F1 transcription activated the expression of CRK, and further promoted the expression of PD-L1. Knockdown of POU2F1 promoted the efficacy of Anti-PD-1. In addition, tumor growth ability decreased after POU2F1 was knocked down. Cytotoxic effector cytokines levels, tumor suppressive chemokines and interleukin increased, while IL17a level decreased when POU2F1 was knocked down.
POU2F1 activates the expression of CRK, further promotes the expression of PD-L1, and finally improves the immune escape in lung cancer.
POU2F1 activates the expression of CRK, further promotes the expression of PD-L1, and finally improves the immune escape in lung cancer.Berberine (BBR) confers potential cardioprotective effects. However, the relevant mechanisms underlying its regulation of cardiomyocyte survival following hypoxia/reoxygenation (H/R) treatment remain unknown. The present study investigated whether BBR could protect H/R by suppressing apoptosis and explored how TGF-β/Smad4 signaling pathway influenced H/R in vitro. Two cardiomyocyte cell lines-AC16 and H9c2- were treated with H/R and BBR. The survival and apoptosis of these two cell lines were assessed using the MTT and BrdU assays and western blotting (WB) and flow cytometry. Mitochondrial reactive oxygen species (ROS) and caspase (Cas)-3, Cas-8, and Cas-9 activation were evaluated using enzyme-linked immunosorbent assay as well as WB. Compared to the control group, H/R resulted in notable cell apoptosis, whereas BBR treatment evidently counteracted the process. BBR also markedly suppressed H/R-triggered excessive mitochondrial ROS generation and inhibited Smad4 expression. Overexpressing Smad4 in BBR-treated H/R-exposed cardiomyocytes reversed the effect of BBR treatment on apoptosis. Therefore, BBR protects H/R-treated cardiomyocytes from apoptosis by inhibiting the TGF-β/Smad4 signaling pathway.Spinal cord injury (SCI) is an insult to the spinal cord resulting in a change, either temporary or permanent, in its normal motor or sensory function, but the mechanism of neuron loss after spinal cord injury is still unclear. Long non-coding RNAs (lncRNAs) can play an important role in regulating cell physiological activities through competitively binding to miRNAs. However, there is still a lack of research on the effect of lncRNAs on SCI. In this study, we selected SCI gene expression data and miRNA expression data from the NCBI database for differential expression analysis, and predicted miRNA target genes. Subsequently, biological analysis of gene expression and miRNA changes was performed on a rat SCI model. The results showed that the expression level of lncRNA-MEG increased significantly in rat SCI model. Subsequently, we found that lncRNA-MEG can promote the expression level of PDCD4 by inhibiting miR-21-5p, which leads to neuronal cell apoptosis. Furthermore, knocking down of lncRNA-MEG with shRNA can reverse the effect of miR-21-5p and inhibit the effect of PDCD4 to reduce the expression of apoptosis-related proteins. Furthermore, we found lncRNA-MEG can regulate PDCD4 expression through miR-21-5p by targeting 3'UTR of PDCD4 in the OGD cell model. In summary, we first discovered lncRNA-MEG regulates neuronal cell apoptosis through miR-21-5p by targeting PDCD4 in SCI.Osteoarthritis (OA) is a progressively degenerative disease of joints. In vitro culture of chondrocytes results in dedifferentiation, which is characterized by the development of fibroblast phenotypes, reduced ability to produce cartilage extracellular matrix (ECM) and increase the expression of molecular markers Col1a1, Col10a1 and Runx2. Redifferentiation of chondrocytes is indicated by increased expression of the molecular markers Col2a1, Aggrecan and Sox9. In the current study, we investigated the effects of allogeneic rabbit adipose-derived mesenchymal stem cells (ADSCs) on articular chondrocytes, and explored the therapeutic effect of ADSCs on damaged articular cartilage at different stages in a rabbit OA model. In vitro, the proliferation and migration of primary articular chondrocytes were enhanced by cocultured rabbit ADSCs, and the expression of redifferentiation markers in chondrocytes cocultured with ADSCs was increased at both the mRNA and protein levels, while the expression of dedifferentiation markers was decreased. In vivo, the rabbit model of OA was established by anterior cruciate ligament transection (ACLT) with complete medial meniscectomy (MMx). Two weeks after surgery, ADSCs were used for OA rabbit treatment. Intra-articular injection of ADSCs gradually alleviated articular cartilage destruction, decreased Osteoarthritis Research Society International (OARSI) and Mankin scores, and reduced MMP13 expression at different stages in the rabbit model of OA. During the experiment, allogeneic ADSCs did not cause any adverse events. The current study demonstrates the effects and molecular mechanisms of ADSCs on articular chondrocytes and provides a favorable reference for clinical OA treatment with mesenchymal stem cells (MSCs) derived from adipose tissue.MiR-543 and Numb are associated with various malignancies, including prostate cancer (PCa). However, whether miR-543 regulates Numb in PCa development remains unclear. In this study, we identified Numb as a direct target of miR-543. The role of miR-543 was examined both in vitro and in vivo. The in vivo effects of miR-543 were investigated using tumor transplantation experiments and a lung metastasis model. The in vitro effects of miR-543 on proliferation, migration, invasion, and cancer stem-like cell (CSC)-associated properties were also examined. The binding sites of Numb were predicted using bioinformatics tools and confirmed by luciferase and rescue assays. QRT-PCR and western blot analyses were used to detect target expression levels. Expression levels of both miR-543 and Numb were manipulated in CD44+ and CD44-PCa cells followed by a series of functional assays. The results demonstrated that miR-543 promoted PCa growth and metastasis both in vivo and in vitro. Luciferase reporter assays, qRT-PCR, and western blot analyses revealed Numb as a direct target of miR-543. The function of miR-543 was abolished by Numb, as shown in rescue experiments. Moreover, miR-543 was verified to promote CSC properties, whereas Numb elicited the opposite effects. MiR-543 also influenced the expression of several stem-like factors, including Dll4, NF-κB, c-myc, and Oct-4, and the Numb/p53 signaling pathway. Taken together, these results demonstrate that miR-543 plays an oncogenic role by negatively controlling Numb, revealing the existence of an miR-543/Numb/p53 regulatory pathway in PCa tumorigenesis and development.