Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.281
Filter
1.
Dis Markers ; 2022: 7300593, 2022.
Article in English | MEDLINE | ID: mdl-35756492

ABSTRACT

Purpose: Oral squamous cell carcinoma (OSCC) is the sixth leading cause of cancer-related death worldwide and is characterized by metastasis and recurrence. We aimed to evaluate the expression of AKT1 and PLK1 in OSCC and identify their correlation with the clinical and histological features and prognosis of patients with OSCC. Methods: Tissue samples were collected from 70 patients with OSCC and 50 patients with normal oral mucosa. The expression levels of AKT1 and PLK1 in OSCC tissues and normal oral mucosa were detected by immunohistochemistry. The chi-square test was used to identify correlations between the expression levels of AKT1 and PLK1 with patients' clinicopathologic characteristics. Survival analysis was assessed by the Kaplan-Meier method. Spearman's rank correlation test was used to determine the relationships between AKT1 and PLK1 expressions. The bioinformatics database GEPIA was used to verify the experimental results. Results: The chi-square test and Fisher's exact test showed that the positive expression rate of AKT1 and PLK1 in OSCC tissue was significantly higher than that in the normal oral mucosa (P < 0.05). PLK1 expression levels were significantly correlated with tumor stage and size (P < 0.05). Kaplan-Meier analysis showed that the survival time of AKT1 and PLK1 with high expression was significantly shorter than that of patients with low expression (P < 0.05). Spearman's rank correlation test showed a strong correlation between AKT1 and PLK1 expression in OSCC tissue (R = 0.53; P < 0.05). GEPIA bioinformatics database analysis results show that the expression and overall survival of AKT1 and PLK1 analysis and the correlation analysis of AKT1 and PLK1 were consistent with experimental results. Conclusion: AKT1 and PLK1 expressions are associated with the occurrence and progression of OSCC and may be used as diagnostic and prognostic indicators of OSCC. There may be a correlation between AKT1 and PLK1 in OSCC tissue.


Subject(s)
Cell Cycle Proteins , Mouth Neoplasms , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins , Squamous Cell Carcinoma of Head and Neck , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/biosynthesis , Humans , Mouth Neoplasms/metabolism , Prognosis , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Squamous Cell Carcinoma of Head and Neck/metabolism , Polo-Like Kinase 1
2.
Pathol Res Pract ; 226: 153596, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34481213

ABSTRACT

Growth arrest specific 6 (GAS6) encodes a protein that serves as a ligand for AXL receptor tyrosine kinase and stimulates cell proliferation. Notably, an antisense RNA, namely GAS6-AS1 is transcribed from chromosome 13q34, near GAS6 gene. In vitro functional experiments have demonstrated that GAS6-AS1 can promote proliferation, migration and invasive properties of transformed cells through enhancing entry into S-phase. Notably, mechanistic investigations have shown that GAS6-AS1 can regulate expression of GAS6 at the transcriptional or translational stages through constructing a RNA-RNA duplex, thus enhancing expression of AXL and inducing AXL signaling. Both GAS6 and its antisense transcript contribute in the pathogenesis of human malignancies. In the current review, we provide a summary of studies that appraised the role of these genes in the carcinogenesis.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/physiology , Intercellular Signaling Peptides and Proteins/physiology , RNA, Antisense/physiology , Animals , Carcinogenesis/metabolism , Cell Proliferation/physiology , Humans , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Axl Receptor Tyrosine Kinase
3.
Medicine (Baltimore) ; 100(32): e26474, 2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34397869

ABSTRACT

ABSTRACT: This study is to identify potential biomarkers and therapeutic targets for lung adenocarcinoma (LUAD).GSE6044 and GSE118370 raw data from the Gene Expression Omnibus database were normalized with Robust Multichip Average. After merging these two datasets, the combat function of sva packages was used to eliminate batch effects. Then, limma packages were used to filtrate differentially expressed genes. We constructed protein-protein interaction relationships using STRING database and hub genes were identified based on connectivity degrees. The cBioportal database was used to explore the alterations of the hub genes. The promoter methylation of cyclin dependent kinase 1 (CDK1) and polo-like Kinase 1 (PLK1) and their association with tumor immune infiltration in patients with LUAD were investigated using DiseaseMeth version 2.0 and TIMER databases. The Cancer Genome Atlas-LUAD dataset was used to perform gene set enrichment analysis.We identified 10 hub genes, which were upregulated in LUAD, among which 8 were successfully verified in the Cancer Genome Atlas and Oncomine databases. Kaplan-Meier analysis indicated that the expressions of CDK1 and PLK1 in LUAD patients were associated with overall survival and disease-free survival. The methylation levels in the promoter regions of these 2 genes in LUAD patients were lower than those in normal lung tissues. Their expressions in LUAD were associated with tumor stages and relative abundance of tumor infiltrating immune cells, such as B cells, CD4+ T cells, and macrophages. Moreover, cell cycle, DNA replication, homologous recombination, mismatch repair, P53 signaling pathway, and small cell lung cancer signaling were significantly enriched in CDK1 and PLK1 high expression phenotype.CDK1 and PLK1 may be used as potential biomarkers and therapeutic targets for LUAD.


Subject(s)
Adenocarcinoma of Lung/genetics , CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Computational Biology/methods , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , CDC2 Protein Kinase/biosynthesis , Cell Cycle Proteins/biosynthesis , Databases, Genetic , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Prognosis , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , RNA, Neoplasm/genetics , Signal Transduction , Polo-Like Kinase 1
4.
Mol Hum Reprod ; 27(8)2021 08 07.
Article in English | MEDLINE | ID: mdl-34264319

ABSTRACT

Investigations of genes required in early mammalian development are complicated by protein deposits of maternal products, which continue to operate after the gene locus has been disrupted. This leads to delayed phenotypic manifestations and underestimation of the number of genes known to be needed during the embryonic phase of cellular totipotency. Here we expose a critical role of the gene Cops3 by showing that it protects genome integrity during the 2-cell stage of mouse development, in contrast to the previous functional assignment at postimplantation. This new role is mediated by a substantial deposit of protein (94th percentile of the proteome), divided between an exceptionally stable cortical rim, which is prevalent in oocytes, and an ancillary deposit in the embryonic nuclei. Since protein abundance and stability defeat prospects of DNA- or RNA-based gene inactivation in oocytes, we harnessed a classical method next to an emerging method for protein inactivation: antigen masking (for functional inhibition) versus TRIM21-mediated proteasomal degradation, also known as 'Trim away' (for physical removal). Both resulted in 2-cell embryo lethality, unlike the embryos receiving anti-green fluorescent protein. Comparisons between COPS3 protein-targeted and non-targeted embryos revealed large-scale transcriptome differences, which were most evident for genes associated with biological functions critical for RNA metabolism and for the preservation of genome integrity. The gene expression abnormalities associated with COPS3 inactivation were confirmed in situ by the occurrence of DNA endoreduplication and DNA strand breaks in 2-cell embryos. These results recruit Cops3 to the small family of genes that are necessary for early embryo survival. Overall, assigning genes with roles in embryogenesis may be less safe than assumed, if the protein products of these genes accumulate in oocytes: the inactivation of a gene at the protein level can expose an earlier phenotype than that identified by genetic techniques such as conventional gene silencing.


Subject(s)
Blastomeres/metabolism , COP9 Signalosome Complex/physiology , Embryonic Development , Oocytes/metabolism , Proto-Oncogene Proteins/physiology , Animals , Blastomeres/ultrastructure , COP9 Signalosome Complex/biosynthesis , COP9 Signalosome Complex/genetics , Cell Survival , DNA Breaks , Embryo Transfer , Embryonic Development/genetics , Endoreduplication , Female , Gene Expression Regulation, Developmental , Gene Ontology , Histones/biosynthesis , Histones/genetics , Luminescent Proteins/analysis , Mice , Microinjections , Oocytes/ultrastructure , Peptide Hydrolases/biosynthesis , Peptide Hydrolases/genetics , Pregnancy , Proteasome Endopeptidase Complex/metabolism , Proteome , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Recombinant Proteins/analysis , Ribonucleoproteins/physiology , Transcriptome , Zygote/metabolism , Red Fluorescent Protein
6.
Mol Hum Reprod ; 27(6)2021 05 29.
Article in English | MEDLINE | ID: mdl-33905521

ABSTRACT

Secreted frizzled-related protein-4 (SFRP4) belongs to a family of soluble ovarian-expressed proteins that participate in female reproduction, particularly in rodents. In humans, SFRP4 is highly expressed in cumulus cells (CCs). However, the mechanisms that stimulate SFRP4 in CCs have not been examined. We hypothesise that oocyte-secreted factors such as growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are involved in the regulation of SFRP4. Human CCs were collected from patients undergoing fertility treatments and treated with GDF9 or BMP15 or their combination in the presence of FSH or vehicle. FSH treatment significantly decreased SFRP4 mRNA levels when compared with nontreated cells. However, SFRP4 mRNA levels were increased significantly by GDF9 plus BMP15 in a concentration-dependent manner in the presence or absence of FSH. The combination of GDF9 plus BMP15 also increased SFRP4 protein levels and decreased the activity of the ß-catenin/T cell factor-responsive promoter significantly. GDF9 plus BMP15 inhibited steroidogenic acute regulatory protein and LH/hCG receptor stimulation by FSH, while treatment with SFRP4 blocked the stimulatory effect of FSH on these genes. The evidence demonstrates that GDF9 and BMP15 act in coordination to stimulate SFRP4 expression and suggests that SFRP4 mediates the anti-luteinising effects of the oocyte in human CCs.


Subject(s)
Bone Morphogenetic Protein 15/pharmacology , Cumulus Cells/drug effects , Growth Differentiation Factor 9/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Oocytes/physiology , Proto-Oncogene Proteins/biosynthesis , Bone Morphogenetic Protein 15/administration & dosage , Cells, Cultured , Cumulus Cells/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation/drug effects , Genes, Reporter , Growth Differentiation Factor 9/administration & dosage , Humans , Oocytes/chemistry , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Primary Cell Culture , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, LH/biosynthesis , Receptors, LH/genetics , Species Specificity
7.
J Heart Lung Transplant ; 40(6): 435-446, 2021 06.
Article in English | MEDLINE | ID: mdl-33846079

ABSTRACT

Cardiac Allograft Vasculopathy (CAV) is a leading contributor to late transplant rejection. Although implicated, the mechanisms by which bone marrow-derived cells promote CAV remain unclear. Emerging evidence implicates the cell surface receptor tyrosine kinase AXL to be elevated in rejecting human allografts. AXL protein is found on multiple cell types, including bone marrow-derived myeloid cells. The causal role of AXL from this compartment and during transplant is largely unknown. This is important because AXL is a key regulator of myeloid inflammation. Utilizing experimental chimeras deficient in the bone marrow-derived Axl gene, we report that Axl antagonizes cardiac allograft survival and promotes CAV. Flow cytometric and histologic analyses of Axl-deficient transplant recipients revealed reductions in both allograft immune cell accumulation and vascular intimal thickness. Co-culture experiments designed to identify cell-intrinsic functions of Axl uncovered complementary cell-proliferative pathways by which Axl promotes CAV-associated inflammation. Specifically, Axl-deficient myeloid cells were less efficient at increasing the replication of both antigen-specific T cells and vascular smooth muscle cells (VSMCs), the latter a key hallmark of CAV. For the latter, we discovered that Axl-was required to amass the VSMC mitogen Platelet-Derived Growth Factor. Taken together, our studies reveal a new role for myeloid Axl in the progression of CAV and mitogenic crosstalk. Inhibition of AXL-protein, in combination with current standards of care, is a candidate strategy to prolong cardiac allograft survival.


Subject(s)
Bone Marrow Cells/pathology , Gene Expression Regulation , Graft Rejection/genetics , Heart Transplantation/adverse effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adult , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Echocardiography , Flow Cytometry , Graft Rejection/diagnosis , Graft Rejection/metabolism , Graft Survival , Humans , Male , Mice , Mice, Inbred BALB C , Muscle, Smooth, Vascular/pathology , Myocytes, Cardiac/pathology , Myocytes, Smooth Muscle , Proto-Oncogene Proteins/biosynthesis , RNA/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Transplantation, Homologous , Axl Receptor Tyrosine Kinase
8.
Exp Neurol ; 341: 113688, 2021 07.
Article in English | MEDLINE | ID: mdl-33713655

ABSTRACT

Increasing numbers of patients with spontaneous subarachnoid hemorrhage(SAH) who recover from surgery and intensive care management still live with cognitive impairment after discharge, indicating the importance of white matter injury at the acute stage of SAH. In the present study, standard endovascular perforation was employed to establish an SAH mouse model, and a microRNA (miRNA) chip was used to analyze the changes in gene expression in white matter tissue after SAH. The data indicate that 17 miRNAs were downregulated, including miR-706, miR-669a-5p, miR-669p-5p, miR-7116-5p and miR-195a-3p, while 13 miRNAs were upregulated, including miR-6907-5p, miR-5135, miR-6982-5p, miR-668-5p, miR-8119. Strikingly, miR-706 was significantly downregulated with the highest fold change. Further experiments confirmed that miR-706 could alleviate white matter injury and improve neurological behavior, at least partially by inhibiting the PKCα/MST1/NF-κB pathway and the release of inflammatory cytokines. These results might provide a deeper understanding of the pathophysiological processes in white matter after SAH, as well as potential therapeutic strategies for the translational research.


Subject(s)
Hepatocyte Growth Factor/antagonists & inhibitors , MicroRNAs/biosynthesis , NF-kappa B/antagonists & inhibitors , Protein Kinase C-alpha/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Subarachnoid Hemorrhage/metabolism , White Matter/metabolism , Animals , Down-Regulation/physiology , Hepatocyte Growth Factor/biosynthesis , Male , Mice , Mice, Inbred C57BL , NF-kappa B/biosynthesis , Protein Kinase C-alpha/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Signal Transduction/physiology , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/prevention & control , White Matter/injuries , White Matter/pathology
9.
Anticancer Res ; 41(3): 1475-1483, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33788740

ABSTRACT

BACKGROUND/AIM: The oncoproteins murine double minute (MDM) 2 and MDM4 inactivate tumor-suppressor protein p53. Their mutual relationship with the prognosis of gastric cancer (GC) remains unknown. PATIENTS AND METHODS: Expression of MDM2, MDM4, and p53 in tumors of 241 patients with GC were evaluated immunohistochemically. Effects of overexpression of MDM4 on tumor-growth properties and sensitivity to cytotoxic drugs were investigated using NUGC4 human GC cell line. RESULTS: High expression of p53 was associated with poor overall survival in the whole population. Among 173 patients with low expression of p53 (implying nonmutation), high expression of MDM4 was an independent factor of poor prognosis in both stage I-III and IV, but of MDM2 was not. MDM4-transduced NUGC4 cells formed twice as many colonies and had a higher 50% inhibitory concentration for 5-fluorouracil and oxaliplatin than did the control cells. CONCLUSION: MDM4 expression is a factor conferring poor prognosis in patients with GC with low expression of p53 and may confer drug resistance.


Subject(s)
Cell Cycle Proteins/biosynthesis , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/biosynthesis , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cisplatin/administration & dosage , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Humans , Immunohistochemistry/methods , Male , Middle Aged , Multivariate Analysis , Oxaliplatin/administration & dosage , Stomach Neoplasms/diagnosis , Stomach Neoplasms/drug therapy
10.
Arch Biochem Biophys ; 701: 108789, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33548214

ABSTRACT

Calcium/calmodulin-dependent serine protein kinase (CASK), a member of membrane-associated guanylate kinase (MAGUK) super-family, is implicated in regulating cell proliferation, cytoskeletal remodeling, and cell metastasis. Our study aimed to investigate the effect of CASK on the malignant behaviors of pancreatic cancer cells and to determine the signaling pathway involved. CASK expression in pancreatic cancer tissues based on the TCGA database was analyzed using GEPIA online tool. The overall survival (OS) and disease-free survival (DFS) in patients with pancreatic cancer based on CASK expression was also analyzed using GEPIA. KEGG pathway enrichment analysis was used to show the association of 1522 CASK-related genes and signaling pathways. The expression of CASK, Notch1 and Hey1 was detected by Western blot. Cell proliferation, colony number, invasion, and apoptosis were detected by CCK-8, colony formation assay, Transwell invasion assay, and flow cytometry analysis, respectively. Results showed that CASK was upregulated in pancreatic cancer tissues and cells. Pancreatic cancer patients with high CASK expression showed shorter OS and DFS than patients with low CASK expression. KEGG pathway enrichment analysis proved that CASK and 1522 CASK-associated genes were primarily associated with the Notch pathway. CASK silencing inhibited cell proliferation, colony formation ability, and invasion and elicited apoptosis in pancreatic cancer cells. Additionally, we confirmed that CASK silencing inhibited the Notch pathway in pancreatic cancer cells. Overexpression of Notch1 resisted the anti-tumor functions of CASK knockdown in pancreatic cancer cells. In conclusion, CASK knockdown suppressed the malignant behaviors of pancreatic cancer cells by inactivating the Notch pathway.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Guanylate Kinases/biosynthesis , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptor, Notch1/metabolism , Signal Transduction , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Guanylate Kinases/genetics , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Receptor, Notch1/genetics
11.
J Neuroinflammation ; 18(1): 6, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33402183

ABSTRACT

BACKGROUND: Neonatal hypoxic-ischemic (HI) brain injury is a leading cause of acute mortality and chronic disability in newborns. Our previous studies demonstrated that HI insult significantly increased microRNA-210 (miR-210) in the brain of rat pups and inhibition of brain endogenous miR-210 by its inhibitor (LNA) provided neuroprotective effect in HI-induced brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. METHODS: We made a neonatal HI brain injury model in mouse pups of postnatal day 7 to uncover the mechanism of miR-210 in targeting the ten eleven translocation (TET) methylcytosine dioxygenase 2 that is a transcriptional suppressor of pro-inflammatory cytokine genes in the neonatal brain. TET2 silencing RNA was used to evaluate the role of TET2 in the neonatal HI-induced pro-inflammatory response and brain injury. MiR-210 mimic and inhibitor (LNA) were delivered into the brain of mouse pups to study the regulation of miR-210 on the expression of TET2. Luciferase reporter gene assay was performed to validate the direct binding of miR-210 to the 3' untranslated region of the TET2 transcript. Furthermore, BV2 mouse microglia cell line was employed to confirm the role of miR-210-TET2 axis in regulating pro-inflammatory response in microglia. Post-assays included chromatin immunoprecipitation (ChIP) assay, co-immunoprecipitation, RT-PCR, brain infarct assay, and neurobehavioral test. Student's t test or one-way ANOVA was used for statistical analysis. RESULTS: HI insult significantly upregulated miR-210, downregulated TET2 protein abundance, and increased NF-κB subunit p65 acetylation level and its DNA binding capacity to the interleukin 1 beta (IL-1ß) promoter in the brain of mouse pups. Inhibition of miR-210 rescued TET2 protein level from HI insult and miR-210 mimic decreased TET2 protein level in the brain of mouse pups, suggesting that TET2 is a functional target of miR-210. The co-immunoprecipitation was performed to reveal the role of TET2 in HI-induced inflammatory response in the neonatal brain. The result showed that TET2 interacted with NF-κB subunit p65 and histone deacetylase 3 (HDAC3), a co-repressor of gene transcription. Furthermore, TET2 knockdown increased transcriptional activity of acetyl-p65 on IL-1ß gene in the neonatal brain and enhanced HI-induced upregulation of acetyl-p65 level and pro-inflammatory cytokine expression. Of importance, TET2 knockdown exacerbated brain infarct size and neurological deficits and counteracted the neuroprotective effect of miR-210 inhibition. Finally, the in vitro results demonstrated that the miR-210-TET2 axis regulated pro-inflammatory response in BV2 mouse microglia cell line. CONCLUSIONS: The miR-210-TET2 axis regulates pro-inflammatory cytokine expression in microglia, contributing to neonatal HI brain injury.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Down-Regulation/physiology , Hypoxia-Ischemia, Brain/metabolism , Inflammation Mediators/metabolism , MicroRNAs/biosynthesis , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Animals , Animals, Newborn , Cell Line , Dioxygenases , Female , Hypoxia-Ischemia, Brain/pathology , Male , Mice
12.
Hematol Oncol ; 39(3): 364-379, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33497493

ABSTRACT

Wnt/Fzd signaling has been implicated in hematopoietic stem cell maintenance and in acute leukemia establishment. In our previous work, we described a recurrent rearrangement involving the WNT10B locus (WNT10BR ), characterized by the expression of WNT10BIVS1 transcript variant, in acute myeloid leukemia. To determine the occurrence of WNT10BR in T-cell acute lymphoblastic leukemia (T-ALL), we retrospectively analyzed an Italian cohort of patients (n = 20) and detected a high incidence (13/20) of WNT10BIVS1 expression. To address genes involved in WNT10B molecular response, we have designed a Wnt-targeted RNA sequencing panel. Identifying Wnt agonists and antagonists, it results that the expression of FZD6, LRP5, and PROM1 genes stands out in WNT10BIVS1 positive patients compared to negative ones. Using MOLT4 and MUTZ-2 as leukemic cell models, which are characterized by the expression of WNT10BIVS1 , we have observed that WNT10B drives major Wnt activation to the FZD6 receptor complex through receipt of ligand. Additionally, short hairpin RNAs (shRNAs)-mediated gene silencing and small molecule-mediated inhibition of WNTs secretion have been observed to interfere with the WNT10B/FZD6 interaction. We have therefore identified that WNT10BIVS1 knockdown, or pharmacological interference by the LGK974 porcupine (PORCN) inhibitor, reduces WNT10B/FZD6 protein complex formation and significantly impairs intracellular effectors and leukemic expansion. These results describe the molecular circuit induced by WNT10B and suggest WNT10B/FZD6 as a new target in the T-ALL treatment strategy.


Subject(s)
Frizzled Receptors/metabolism , Gene Expression Regulation, Leukemic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins/biosynthesis , Wnt Proteins/biosynthesis , Wnt Signaling Pathway , Acyltransferases/antagonists & inhibitors , Acyltransferases/genetics , Acyltransferases/metabolism , Female , Frizzled Receptors/genetics , HeLa Cells , Humans , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/genetics , Pyrazines/pharmacology , Pyridines/pharmacology , Wnt Proteins/genetics
13.
Gene ; 770: 145337, 2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33278551

ABSTRACT

BACKGROUND: The myeloma overexpressed gene (MYEOV) plays a critical role in tumorigenesis in a variety of cancers. However, little is known of the prognosis and immune infiltration associated with MYEOV in non-small cell lung cancer (NSCLC). METHODS: We used several databases (Oncomine, TCGA, and GEO) to analysis the expression, prognosis, and immune infiltration, associated with MYEOV in NSCLC. We also used RT-qPCR and immunohistochemistry to investigate the expression and prognosis of MYEOV in NSCLC. RESULTS: Compared with normal tissues, high MYEOV expression in NSCLC was observed in Oncomine database, and was validated in the TCGA database. High MYEOV expression was significantly associated with different subtypes of NSCLC. Moreover, high MYEOV expression was closely related with a poorer overall survival in NSCLC in TCGA cohort, and was validated in GEO database. Simultaneously, high expression of MYEOV correlates with clinical relevance of NSCLC. Specifically, MYEOV expression was negatively correlated with infiltrating levels of tumor purity and B cells in LUAD. MYEOV expression was negatively correlated with infiltrating levels of tumor purity, and positively associated with CD8 + T cells, CD4 + T cells, dendritic cells, and neutrophils in LUSC. GSEA also revealed that high MYEOV expression were enriched in certain cancer-specific pathways. In addition, RT-qPCR and immunohistochemistry showed MYEOV expression was higher in NSCLC compared to the normal tissues. Finally, high MYEOV expression was closely related with poorer overall survival of NSCLC in an independent validation cohort. CONCLUSION: Our analyses indicate that MYEOV can be used as a prognostic biomarker for determining prognosis and immune infiltration in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Proto-Oncogene Proteins , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Disease-Free Survival , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Survival Rate
14.
Int J Mol Med ; 47(1): 137-150, 2021 01.
Article in English | MEDLINE | ID: mdl-33236134

ABSTRACT

Overproduction of pro­inflammatory cytokines in the aged, which is called inflammaging, leads to the deterioration of periodontitis. Toll­like receptor 4 (TLR4) plays a role in the regulation of cellular senescence, and its expression increases with age. However, there has been limited research into the molecular mechanisms underlying the onset of periodontal inflammaging, and the interplay between TLR4 and inflammaging. In the present study, wild­type and TLR4 gene knockout mice were used to investigate the activation of the TLR4 pathway in mouse periodontitis and the expression of the nucleotide­binding and oligomerization domain­like receptor 3 (NLRP3) inflammasome, an upstream immune checkpoint during the development of inflammaging. Activation of TLR4 in a mouse model of periodontitis enhanced the expression of a senescence­associated secretory phenotype (SASP), which boosted the inflammaging process. Conversely, TLR4 activation downregulated the expression of B cell­specific Moloney murine leukemia virus integration site 1 (Bmi­1) and promoted the priming of NLRP3 inflammasome, both of which are regulators of SASP. Treating gingival fibroblasts with Bmi­1 inhibitor PTC209, it was demonstrated that TLR4 activated the NLRP3 pathway and the inflammaging process by suppressing Bmi­1. In addition, there was a significant reduction in the expression of Bmi­1 expression in the gingiva of patients with periodontitis compared with healthy controls. In conclusion, the present study demonstrated that TLR4 acted by inhibiting Bmi­1 to enhance the NLRP3 pathway and SASP factors. This cascade of reactions may contribute to the senescence of the periodontium.


Subject(s)
Gene Expression Regulation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Periodontitis/metabolism , Polycomb Repressive Complex 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Toll-Like Receptor 4/metabolism , Animals , Female , Inflammasomes/genetics , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Periodontitis/genetics , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , Toll-Like Receptor 4/genetics
15.
Dig Dis Sci ; 66(6): 1875-1884, 2021 06.
Article in English | MEDLINE | ID: mdl-32712782

ABSTRACT

BACKGROUND: The importance of long noncoding RNAs (lncRNAs) has been identified in human cancers, such as emerged as tumor facilitator or tumor suppressor. Small nucleolar RNA host gene 10 (SNHG10) has been reported as an oncogenic lncRNA in hepatocellular carcinoma. However, its functional role and underlying mechanism in gastric cancer (GC) need to be further explored. AIMS: Our study was conducted to investigate the function and molecular mechanism of SNHG10 in GC. METHODS: SNHG10 expression was detected by qRT-PCR. The effect of SNHG10 on GC cell growth was assessed by colony formation, EdU, JC-1, flow cytometry, and wound-healing assays. The interaction between SNHG10 and PBX3 was confirmed through ChIP and luciferase reporter assay. RIP and RNA pull down assays was used to define the binding of DEAD-box helicase 54 (DDX54) to SNHG10 or PBX homeobox 3 (PBX3). RESULTS: SNHG10 was expressed at a high level in GC cells. SNHG10 knockdown resulted in the inhibition on GC cell proliferation, migration but induced cell apoptosis. PBX3 could interact with SNHG10 promoter and thereby activate the expression of SNHG10. Subsequently, it was confirmed that SNHG10 positively modulated the expression of PBX3. Based on this, we found that DDX54 could bind to SNHG10 and PBX3, suggesting that SNHG10 maintained PBX3 mRNA stability through recruiting DDX54. Restoration assays indicated that PBX3 overexpression recovered SNHG10 silencing-induced inhibition on GC cell growth. CONCLUSIONS: SNHG10 facilitates cell growth by affecting DDX54-mediated PBX3 mRNA stability in GC.


Subject(s)
Cell Proliferation/physiology , DEAD-box RNA Helicases/biosynthesis , Feedback, Physiological/physiology , Homeodomain Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , RNA, Long Noncoding/biosynthesis , Stomach Neoplasms/metabolism , Adult , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , Female , Homeodomain Proteins/genetics , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
16.
Methods Mol Biol ; 2199: 127-149, 2021.
Article in English | MEDLINE | ID: mdl-33125648

ABSTRACT

The cell-free synthesis is an efficient strategy to produce in large scale protein samples for structural investigations. In vitro synthesis allows for significant reduction of production time, simplification of purification steps and enables production of both soluble and membrane proteins. The cell-free reaction is an open system and can be performed in presence of many additives such as cofactors, inhibitors, redox systems, chaperones, detergents, lipids, nanodisks, and surfactants to allow for the expression of toxic membrane proteins or intrinsically disordered proteins. In this chapter we present protocols to prepare E. coli S30 cellular extracts, T7 RNA polymerase, and their use for in vitro protein expression. Optimizations of the protocol are presented for preparation of protein samples enriched in deuterium, a prerequisite for the study of high-molecular-weight proteins by NMR spectroscopy. An efficient production of perdeuterated proteins is achieved together with a full protonation of all the amide NMR probes, without suffering from residual protonation on aliphatic carbons. Application to the production of the 468 kDa TET2 protein assembly for NMR investigations is presented.


Subject(s)
DNA-Binding Proteins , Deuterium/chemistry , Escherichia coli/chemistry , Isotope Labeling , Proto-Oncogene Proteins , Cell-Free System/chemistry , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dioxygenases , Humans , Nuclear Magnetic Resonance, Biomolecular , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
17.
Cell Cycle ; 19(23): 3317-3328, 2020 12.
Article in English | MEDLINE | ID: mdl-33218295

ABSTRACT

The colorectal cancer (CRC) dictates a common malignancy with high recurrence rate. Long non-coding RNAs (lncRNAs) belong to a class of regulatory factors involved in multiple cancers. In current work, we have uncovered a novel lncRNA named TSLC8. TSLC8 was dramatically downregulated in CRC samples and cell lines. Reintroduction of TSLC8 inhibited tumor sphere formation and viability in CRC cells. In vivo experiments further confirmed the tumor suppressive function of TSLC8. Ectopic TSLC8 expression elevates puma abundance whereas this effect is mediated by TSLC8-puma binding and stabilization. FOXO1 can transcriptionally induce TSLC8 expression. Epigenetic investigation suggested that TSLC8 locus was hypermethylated in CRC leading to diminished TSLC8 expression. Our current work has identified a novel tumor suppressive function of TSLC8, whose reduced expression may facilitate malignant phenotypes during CRC progression.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , RNA, Long Noncoding/biosynthesis , Animals , Apoptosis Regulatory Proteins/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/prevention & control , Female , HCT116 Cells , Humans , Mice , Mice, Nude , Proto-Oncogene Proteins/genetics , RNA, Long Noncoding/genetics , Xenograft Model Antitumor Assays/methods
18.
Best Pract Res Clin Haematol ; 33(3): 101194, 2020 09.
Article in English | MEDLINE | ID: mdl-33038988

ABSTRACT

BCR-ABL1 is in the center of chronic myeloid leukemia (CML) pathology, diagnosis and treatment, as confirmed by the success of tyrosine kinase inhibitor (TKI) therapy. However, additional mechanisms and events, many of which function independently of BCR-ABL1, play important roles, particularly in terms of leukemic stem cell (LSC) persistence, primary and secondary resistance, and disease progression. Promising therapeutic approaches aim to disrupt pathways which mediate LSC survival during successful TKI treatment, in the hope of improving long-term treatment-free-remission and perhaps provide a functional cure for some patients. Over the years through advances in sequencing technology frequent molecular aberrations in addition to BCR-ABL1 have been identified not only in advanced disease but also in chronic phase CML, often affecting epigenetic regulators such as ASXL1, DNMT3A and TET2. Analyses of serial samples have revealed various patterns of clonal evolution with some mutations preceding the BCR-ABL1 acquisition. Such mutations can be considered to be important co-factors in the pathogenesis of CML and could potentially influence therapeutic strategies in the future.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA-Binding Proteins , Epigenesis, Genetic , Fusion Proteins, bcr-abl , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Proto-Oncogene Proteins , Repressor Proteins , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Dioxygenases , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
19.
Cell Death Dis ; 11(9): 754, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934219

ABSTRACT

The identification of the essential role of cyclin-dependent kinases (CDKs) in the control of cell division has prompted the development of small-molecule CDK inhibitors as anticancer drugs. For many of these compounds, the precise mechanism of action in individual tumor types remains unclear as they simultaneously target different classes of CDKs - enzymes controlling the cell cycle progression as well as CDKs involved in the regulation of transcription. CDK inhibitors are also capable of activating p53 tumor suppressor in tumor cells retaining wild-type p53 gene by modulating MDM2 levels and activity. In the current study, we link, for the first time, CDK activity to the overexpression of the MDM4 (MDMX) oncogene in cancer cells. Small-molecule drugs targeting the CDK9 kinase, dinaciclib, flavopiridol, roscovitine, AT-7519, SNS-032, and DRB, diminished MDM4 levels and activated p53 in A375 melanoma and MCF7 breast carcinoma cells with only a limited effect on MDM2. These results suggest that MDM4, rather than MDM2, could be the primary transcriptional target of pharmacological CDK inhibitors in the p53 pathway. CDK9 inhibitor atuveciclib downregulated MDM4 and enhanced p53 activity induced by nutlin-3a, an inhibitor of p53-MDM2 interaction, and synergized with nutlin-3a in killing A375 melanoma cells. Furthermore, we found that human pluripotent stem cell lines express significant levels of MDM4, which are also maintained by CDK9 activity. In summary, we show that CDK9 activity is essential for the maintenance of high levels of MDM4 in human cells, and drugs targeting CDK9 might restore p53 tumor suppressor function in malignancies overexpressing MDM4.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 9/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Synergism , Humans , Imidazoles/pharmacology , MCF-7 Cells , Melanoma/genetics , Melanoma/pathology , Mice , Piperazines/pharmacology , Pluripotent Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Roscovitine/pharmacology , Sulfonamides/pharmacology , Transcription, Genetic , Transfection , Triazines/pharmacology
20.
Mol Reprod Dev ; 87(11): 1159-1172, 2020 11.
Article in English | MEDLINE | ID: mdl-32949181

ABSTRACT

Wnt/beta-catenin signaling may play an essential role in endometrial decidualization, placentation, and the establishment of pregnancy. We investigate here the possible roles, immunolocalizations, and synthesis of the Wnt3, Wnt7a, and beta-catenin proteins in the rat endometrium during the estrous cycle and early postimplantation period. Wnt3 and Wnt7a had a similar localization and dynamic expression relative to the endometrial stages. Wnt7a immunostaining was not limited only to the luminal epithelial cells, but also to strong stainings in the stromal and endothelial cells. Wnt3, Wnt7a, and beta-catenin were highly synthesized and colocalized at the trophoblast-decidual interface; and were more obvious in the primary decidual zone, the GTCs, and the ectoplacental cone. Beta-catenin was strongly localized at the borders of the mature decidual cells; however, Wnt3 and Wnt7a immunolocalizations were decreased in those cells. As such, the immunolocalization of Wnt3, Wnt7a, and beta-catenin shifted with decidualization and placentation. The expression level of Wnt3, Wnt7a, and beta-catenin messenger RNAs increased in early pregnancy, and especially between Days 8.5 and 9.5. The dramatic changes in the expression of Wnt3, Wnt7a, and beta-catenin observed during the early days of pregnancy and the estrous cycle may indicate their roles in decidualization, stromal cell proliferation, and trophoblast invasion.


Subject(s)
Endometrium/metabolism , Proto-Oncogene Proteins/physiology , Wnt Proteins/physiology , Wnt Signaling Pathway , Wnt3 Protein/physiology , beta Catenin/physiology , Animals , Decidua/cytology , Embryo, Mammalian/metabolism , Estrous Cycle/physiology , Female , Gene Expression Regulation, Developmental , Pregnancy , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Stromal Cells/metabolism , Trophoblasts/cytology , Wnt Proteins/biosynthesis , Wnt Proteins/genetics , Wnt3 Protein/biosynthesis , Wnt3 Protein/genetics , beta Catenin/biosynthesis , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...