Components of reverse-transcription reaction.

Components of reverse-transcription reaction.

Source publication
Article
Full-text available
Lysine-deficient protein kinase-1 (WNK1) is critical for both embryonic angiogenesis and tumor-induced angiogenesis. However, the downstream effectors of WNK1 during these processes remain ambiguous. In this study, we identified that oxidative stress responsive 1b (osr1b) is upregulated in endothelial cells in both embryonic and tumor-induced angio...

Context in source publication

Context 1
... complementary DNA (cDNA) was reverse-transcribed from total RNA by iScript TM Synthesis Kit (BIO-RAD, Hercules, CA, USA). The components are listed as Table 3 below: Reaction was set at 25 • C for 5 min, 46 • C for 20 min, 95 • C for 1 min, and soak at 4 • C. After the RT reaction, cDNA was diluted (100-fold to 20-fold) for QPCR. ...

Citations

... 28 WNK1-mediated phosphorylation of the native substrate OSR1 was inhibited by WNK463 with an IC 50 of 41 nM in biochemical assays, and of 106 nM if in HEK293 cells following induction of osmotic stress. 28 In subsequent cell culture studies, WNK463 was mostly applied using 1 μM, [49][50][51][52][53][54][55] or sometimes in lower (100-625 nM), 56,57 or higher (4-10 μM) concentrations. 58,59 Off-targets were examined using a concentration of 10 μM in a screening assay with 444 kinases. ...
Article
Full-text available
With no lysine/K kinases (WNKs) promote vasocontraction and vascular smooth muscle cell proliferation. In the prostate, smooth muscle contraction and growth may be critical for the development and medical treatment of voiding symptoms in benign prostatic hyperplasia. Here, we examined the effects of isoform‐specific WNK silencing and of the WNK inhibitor WNK463 on growth‐related functions and contraction in prostate stromal cells, and in human prostate tissues. Impacts of WNK silencing by transfection of cultured stromal cells with isoform‐specific siRNAs were qualitatively and quantitatively similar for each WNK isoform. Effects of silencing were largest on cell death (3–5 fold increase in annexin V‐positive/7‐AAD‐positive cells), on proliferation rate, Ki‐67 mRNA expression and actin organization (reduced around two‐thirds). Contraction in matrix contraction assays and viability were reduced to a lower degree (approximately half), but again to a similar extent for each WNK isoform. Effects of silencing were quantitatively and qualitatively reproduced by 10 μM WNK463, while 1 μM still induced cell death and breakdown in actin organization, without affecting proliferation or viability. Using 500 nM and 10 μM, WNK463 partly inhibited neurogenic and U46619‐induced contractions of human prostate tissues (around half), while inhibition of α1‐adrenergic contractions (around half) was limited to 10 μM. All four WNK isoforms suppress cell death and promote proliferation in prostate stromal cells. WNK‐driven contraction of stromal cells appears possible, even though to a limited extent. Outcomes of isoform‐specific WNK silencing can be fully reproduced by WNK463, including inhibition of smooth muscle contraction in human prostate tissues, but require high concentrations.
... Its activation triggers a domino effect, inevitably culminating in apoptosis (Bao et al., 2020). P53 is a vital component in cancer prevention, which is activated under stressful conditions, promoting apoptosis (Hou et al., 2022). It influences the expression of apoptosis-associated genes by amplifying pro-apoptotic gene expression while curtailing anti-apoptotic gene activity (Hao et al., 2021). ...
... in the screening and identification of new members of the MAP kinase (MAP2K) family. WNK1 was early thought to be a blood pressure regulator and can regulate angiogenesis and induce cell migration in a range of cancer types 43 . In recent years, more and more evidence shows that WNK1 is a key kinase involved in many types of cancer, promoting cell proliferation and inducing cancer through anti-apoptosis and promoting survival function. ...
Article
Full-text available
In the advanced stage of cancer, the pain caused by bone metastasis is unbearable, but the mechanism of bone cancer pain (BCP) is very complicated and remains unclear. In this study, we used 4T1 mouse breast cancer cells to establish a bone cancer pain model to study the mechanism of BCP. Then the paw withdrawal mechanical threshold (PWMT) and the hematoxylin-eosin staining were used to reflect the erosion of cancer cells on tibia tissue. We also determined the role of proinflammatory factors (TNF-α, IL-17, etc.) in BCP by the enzyme-linked immunosorbent assay in mouse serum. When GSK690693, a new Akt inhibitor, was given and the absence of intermediate signal dominated by Akt is found, pain may be relieved by blocking the transmission of pain signal and raising the PWMT. In addition, we also found that GSK690693 inhibited the phosphorylation of Akt protein, resulting in a significant decrease in with-nolysinekinases 1 (WNK1) expression in the spinal cord tissue. In the BCP model, we confirmed that GSK690693 has a relieving effect on BCP, which may play an analgesic effect through PI3K-WNK1 signal pathway. At the same time, there is a close relationship between inflammatory factors and PI3K-WNK1 signal pathway. The PI3K/Akt pathway in the dorsal horn of the mouse spinal cord activates the downstream WNK1 protein, which promotes the release of inflammatory cytokines, which leads to the formation of BCP in mice. Inhibition of Akt can reduce the levels of IL-17 and TNF-α, cut off the downstream WNK1 protein signal receiving pathway, increase the PWMT and relieve BCP in mice. To clarify the analgesic target of BCP, to provide reference and theoretical support for the clinical effective treatment of BCP and the development of new high-efficiency analgesics.
... Two downstream effectors of WNK1-mediated angiogenesis are the oxidative stress responsive 1 (OSR1) and the protein phosphatase 2A (PPP2R1A) proteins. Up-regulation of WNK1 and OSR1 in endothelial cells favors the process of neoangiogenesis, and the axis WNK1-ORSR1-PPP2R1A was demonstrated to play a critical role in both endothelial (HUVEC) and hepatoma (HepG2) cells during angiogenesis and cell migration [42]. The zebrafish model was used in a very recent study to demonstrate how the co-administration of oligo-fucoidan could improve the anti-cancer efficacy of WNK463 (a WNK1 inhibitor) and rafoxanide (an OSR1 inhibitor) in advanced hepatocellular carcinoma [42] ( Table 1). ...
... Up-regulation of WNK1 and OSR1 in endothelial cells favors the process of neoangiogenesis, and the axis WNK1-ORSR1-PPP2R1A was demonstrated to play a critical role in both endothelial (HUVEC) and hepatoma (HepG2) cells during angiogenesis and cell migration [42]. The zebrafish model was used in a very recent study to demonstrate how the co-administration of oligo-fucoidan could improve the anti-cancer efficacy of WNK463 (a WNK1 inhibitor) and rafoxanide (an OSR1 inhibitor) in advanced hepatocellular carcinoma [42] ( Table 1). The main mechanism responsible for the anticancer activity of fucoidan in association with WNK463 and rafoxanide has been ascribed to its ability to counteract the inflammatory response evoked by the aforementioned inhibitors [42]. ...
... The zebrafish model was used in a very recent study to demonstrate how the co-administration of oligo-fucoidan could improve the anti-cancer efficacy of WNK463 (a WNK1 inhibitor) and rafoxanide (an OSR1 inhibitor) in advanced hepatocellular carcinoma [42] ( Table 1). The main mechanism responsible for the anticancer activity of fucoidan in association with WNK463 and rafoxanide has been ascribed to its ability to counteract the inflammatory response evoked by the aforementioned inhibitors [42]. However, it is possible to hypothesize that the antiangiogenic and antimetastatic activity of fucoidan may contribute to the recorded anticancer effects. ...
Article
Full-text available
Angiogenesis and metastasis represent two challenging targets to combat cancer development in the later stages of its progression. Numerous studies have indicated the important role of natural products in blocking tumor angiogenesis signaling pathways in several advanced tumors. In recent years, the marine polysaccharides fucoidans emerged as promising anticancer compounds showing potent antitumor activity in both in vitro and in vivo models of different types of cancers. The objective of this review is to focus on the antiangiogenic and antimetastatic activities of fucoidans with special emphasis on preclinical studies. Independently from their source, fucoidans inhibit several angiogenic regulators, primarily vascular endothelial growth factor (VEGF). A glance towards fucoidans' ongoing clinical trials and pharmacokinetic profile is provided to present the main challenges that still need to be addressed for their bench-to-bedside translation.
Article
Developing anticancer drugs is a complex and time-consuming process. The inability of current laboratory models to reflect important aspects of the tumor in vivo limits anticancer medication research. Zebrafish is a rapid, semi-automated in vivo screening platform that enables the use of non-invasive imaging methods to monitor morphology, survival, developmental status, response to drugs, locomotion, or other behaviors. Zebrafish models are widely used in drug discovery and development for anticancer drugs, especially in conjunction with live imaging techniques. Herein, we concentrated on the use of zebrafish live imaging in anticancer therapeutic research, including drug screening, efficacy assessment, toxicity assessment, and mechanism studies. Zebrafish live imaging techniques have been used in numerous studies, but this is the first time that these techniques have been comprehensively summarized and compared side by side. Finally, we discuss the hypothesis of Zebrafish Composite Model, which may provide future directions for zebrafish imaging in the field of cancer research.
Article
Full-text available
BACKGROUND B56ε is a regulatory subunit of the serine/threonine protein phosphatase 2A, which is abnormally expressed in tumors and regulates various tumor cell functions. At present, the application of B56ε in pan-cancer lacks a comprehensive analysis, and its role and mechanism in hepatocellular carcinoma (HCC) are still unclear. AIM To analyze B56ε in pan-cancer, and explore its role and mechanism in HCC. METHODS The Cancer Genome Atlas, Genotype-Tissue Expression, Gene Expression Profiling Interactive Analysis, and Tumor Immune Estimation Resource databases were used to analyze B56ε expression, prognostic mutations, somatic copy number alterations, and tumor immune characteristics in 33 tumors. The relationships between B56ε expression levels and drug sensitivity, immunotherapy, immune checkpoints, and human leukocyte antigen (HLA)-related genes were further analyzed. Gene Set Enrichment Analysis (GSEA) was performed to reveal the role of B56ε in HCC. The Cell Counting Kit-8, plate cloning, wound healing, and transwell assays were conducted to assess the effects of B56ε interference on the malignant behavior of HCC cells. RESULTS In most tumors, B56ε expression was upregulated, and high B56ε expression was a risk factor for adrenocortical cancer, HCC, pancreatic adenocarcinoma, and pheochromocytoma and paraganglioma (all P < 0.05). B56ε expression levels were correlated with a variety of immune cells, such as T helper 17 cells, B cells, and macrophages. There was a positive correlation between B56ε expression levels with immune checkpoint genes and HLA-related genes (all P < 0.05). The expression of B56ε was negatively correlated with the sensitivity of most chemotherapy drugs, but a small number showed a positive correlation (all P < 0.05). GSEA analysis showed that B56ε expression was related to the cancer pathway, p53 downstream pathway, and interleukin-mediated signaling in HCC. Knockdown of B56ε expression in HCC cells inhibited the proliferation, migration, and invasion capacity of tumor cells. CONCLUSION B56ε is associated with the microenvironment, immune evasion, and immune cell infiltration of multiple tumors. B56ε plays an important role in HCC progression, supporting it as a prognostic marker and potential therapeutic target for HCC.
Article
Lenvatinib is a clinically effective multikinase inhibitor approved for first‐line therapy of advanced hepatocellular carcinoma (HCC). Although resistance against lenvatinib often emerges and limits its antitumor activity, the underlying molecular mechanisms involved in endogenous and acquired resistance remain elusive. In this study, we identified focal adhesion kinase (FAK) as a critical contributor to lenvatinib resistance in HCC. The elevated expression and phosphorylation of FAK were observed in both acquired and endogenous lenvatinib‐resistant (LR) HCC cells. Furthermore, inhibition of FAK reversed lenvatinib resistance in vitro and in vivo. Mechanistically, FAK promoted lenvatinib resistance through regulating lysine‐deficient kinase 1 (WNK1). Phosphorylation of WNK1 was significantly increased in LR‐HCC cells. Further, WNK1 inhibitor WNK463 resensitized either established or endogenous LR‐HCC cells to lenvatinib treatment. In addition, overexpression of WNK1 desensitized parental HCC cells to lenvatinib treatment. Conclusively, our results establish a crucial role and novel mechanism of FAK in lenvatinib resistance and suggest that targeting the FAK/WNK1 axis is a promising therapeutic strategy in HCC patients showing lenvatinib resistance.