Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Cell Biochem ; 119(10): 8346-8358, 2018 11.
Article in English | MEDLINE | ID: mdl-29893431

ABSTRACT

This study aims to explore the effects of miR-539 on osteoblast proliferation and differentiation and osteoclast apoptosis in a rat model of osteoporosis, and its mechanism involving the regulation of the AXIN1-mediated wingless-Int (Wnt) signaling pathway. A rat model of osteoporosis was successfully established by ovariectomy. With osteoblasts and osteoclasts of rats not receiving ovariectomy in the sham group as control, those of osteoporotic rats were treated with miR-539 inhibitor, miR-539 mimic, and AXIN1 shRNA. The expression of miR-53, AXIN1, the Wnt pathway related-genes, apoptosis related-genes, and osteogenic markers were measured by RT-qPCR and Western blot analysis, respectively. Alkaline phosphatase (ALP) activity in osteoblast and tartrate-resistant acid phosphatase (TRAP) activity in osteoclasts were determined after cell transfection. Osteoblast and osteoclast viability was assayed by CCK-8 assay. Cell cycle and apoptosis of osteoblasts and osteoclasts were detected by flow cytometry. Lastly, alizarin red S staining was used to detect mineralized nodules of osteoblasts. Firstly, we determined that miR-539 was down-regulated in osteoblast and osteoclast of osteoporotic rats and AXIN1 was negatively regulated by miR-539. Additionally, overexpression of miR-539 increased the expressions of ß-catenin, LEF1, c-myc, cyclin D1, RUNX2, BGP, BMP-2 in osteoblast as well as ß-catenin, RhoA, caspase-3, and Bcl-2 in osteoclasts. Finally, overexpression of miR-539 elevated ALP activity, proliferation, and mineralized nodules in osteoblast and osteoclast apoptosis, with reduced TRAP activity in osteoclasts. Our results demonstrate that miR-539 promotes osteoblast proliferation and differentiation as well as osteoclast apoptosis through the AXIN1-dependent Wnt signaling pathway in osteoporotic rats.


Subject(s)
Axin Protein/genetics , MicroRNAs/genetics , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/genetics , Wnt Signaling Pathway , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Antagomirs/genetics , Antagomirs/metabolism , Apoptosis/genetics , Axin Protein/antagonists & inhibitors , Axin Protein/metabolism , Base Sequence , Bone Density , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation , Germ-Free Life , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Molecular Mimicry , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Osteoblasts/cytology , Osteoclasts/cytology , Osteoporosis/etiology , Osteoporosis/metabolism , Osteoporosis/pathology , Ovariectomy/adverse effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Tartrate-Resistant Acid Phosphatase/genetics , Tartrate-Resistant Acid Phosphatase/metabolism
2.
Cell Physiol Biochem ; 51(1): 470-486, 2018.
Article in English | MEDLINE | ID: mdl-30453289

ABSTRACT

BACKGROUND/AIMS: MicroRNA-197 (miR-197) has been shown to play roles in epithelialmesenchymal transition (EMT) and metastasis. The Wnt/ß-catenin pathway is associated with EMT, but whether miR-197 regulatesWnt/ß-catenin remains unclear. This study was to demonstrate the role of miR-197 on the Wnt/ß-catenin pathway in hepatocellular carcinoma (HCC). METHODS: Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to detect the expression of miR-197 in 105 HCC specimens and 15 HCC cell lines. We tested the predicted target gene of miR-197 using a genetic report system. The role of miR-197 in HCC cell invasion and migration (wound healingand cell invasion and migrationby Transwell assays) and in an HCC xenograft modelwas analyzed. RESULTS: Using a miRNA microarray analysis of HCC specimens and compared with non-metastatic HCC, miR-197 was identified as one of the most upregulated miRNAs in metastatic HCC. miR-197 expression was positively associated with the invasiveness of HCC cell lines. Metastatic HCC cells with high miR-197 expression had Wnt/ß-catenin signaling activation. High levels of miR-197 expression also promoted EMT and invasionHCC cells in vitro and in vivo. miR-197 directly targeted Axin-2, Naked cuticle 1 (NKD1), and Dickkopf-related protein 2 (DKK2), leading to inhibition of Wnt/ß-catenin signaling. High miR-197 expression was found in HCC specimens from patients with portal vein metastasis;high miR-197 expression correlated to the expression of Axin2, NKD1, and DKK2. CONCLUSION: miR-197 promotes HCC invasion and metastasis by activating Wnt/ß-catenin signaling. miR-197 could possibly be used as a prognostic marker and therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/metabolism , Wnt Signaling Pathway , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing , Animals , Antagomirs/metabolism , Antagomirs/therapeutic use , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Axin Protein/metabolism , Calcium-Binding Proteins , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/metabolism
3.
Biochem Biophys Res Commun ; 496(2): 302-308, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29307822

ABSTRACT

It is well known that colon cancer stemness and invasiveness are the main reasons for tumor recurrence and metastasis. MicroRNAs dysregulation can disrupt the balance of cell signaling and growth processes, resulting in cancer proliferation, invasion and metastasis, chemoresistance and so on. In this study, we used colon cancer cell lines HCT-116 and SW-480 to investigate the effects of miR-3120-5p on stemness and invasiveness of colon cancer. We found that the population of CD133 + and Lgr5+ stem cells in both cell lines expressed miR-3120-5p highly, and introducing miR-3120-5p into both cell lines increased the population of cancer stem cells, as measured by flow cytometry, qRT-PCR and sphere formation assays. Transwell assay, Gelatin zymography assay and Western blot assays further revealed that miR-3120-5p promotes colon cancer cells invasive ability. By the target prediction algorithm TargetScan, we found Axin2 is a potential target for miR-3120-5p, and luciferase reporter assay demonstrated that miR-3120-5p reduces Axin2 expression. Transfection of siRNA against Axin2 into colon cancer cells promoted the stemness and invasion of colon cancer cells. Furthermore, Axin2 overexpression partially reversed the promotion of stemness and invasiveness caused by miR-3120-5p in colon cancer cells. Together, all the results demonstrated miR-3120-5p promotes stemness and invasiveness of colon cancer cells through direct targeting of Axin2. They suggest that antago-miR-3120-5p plays important roles on treatment strategy for colon cancer.


Subject(s)
Axin Protein/genetics , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/metabolism , AC133 Antigen/genetics , AC133 Antigen/metabolism , Antagomirs/genetics , Antagomirs/metabolism , Axin Protein/antagonists & inhibitors , Axin Protein/metabolism , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Genes, Reporter , HCT116 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Spheroids, Cellular/pathology
4.
Proc Natl Acad Sci U S A ; 110(50): 20224-9, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277854

ABSTRACT

Wnt signaling is one of the key oncogenic pathways in multiple cancers, and targeting this pathway is an attractive therapeutic approach. However, therapeutic success has been limited because of the lack of therapeutic agents for targets in the Wnt pathway and the lack of a defined patient population that would be sensitive to a Wnt inhibitor. We developed a screen for small molecules that block Wnt secretion. This effort led to the discovery of LGK974, a potent and specific small-molecule Porcupine (PORCN) inhibitor. PORCN is a membrane-bound O-acyltransferase that is required for and dedicated to palmitoylation of Wnt ligands, a necessary step in the processing of Wnt ligand secretion. We show that LGK974 potently inhibits Wnt signaling in vitro and in vivo, including reduction of the Wnt-dependent LRP6 phosphorylation and the expression of Wnt target genes, such as AXIN2. LGK974 is potent and efficacious in multiple tumor models at well-tolerated doses in vivo, including murine and rat mechanistic breast cancer models driven by MMTV-Wnt1 and a human head and neck squamous cell carcinoma model (HN30). We also show that head and neck cancer cell lines with loss-of-function mutations in the Notch signaling pathway have a high response rate to LGK974. Together, these findings provide both a strategy and tools for targeting Wnt-driven cancers through the inhibition of PORCN.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Pyrazines/pharmacology , Pyridines/pharmacology , Wnt Signaling Pathway/drug effects , Acyltransferases , Animals , Axin Protein/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Cloning, Molecular , High-Throughput Screening Assays , Humans , Mice , Mutagenesis , Phosphorylation/drug effects , Pyrazines/therapeutic use , Pyridines/therapeutic use , Radioligand Assay , Rats , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Nat Chem Biol ; 9(9): 579-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23892894

ABSTRACT

The Wnt/ß-catenin signaling pathway has a crucial role in embryonic development, stem cell maintenance and human disease. By screening a synthetic chemical library of lycorine derivatives, we identified 4-ethyl-5-methyl-5,6-dihydro-[1,3]dioxolo[4,5-j]phenanthridine (HLY78) as an activator of the Wnt/ß-catenin signaling pathway, which acts in a Wnt ligand-dependent manner. HLY78 targets the DIX domain of Axin and potentiates the Axin-LRP6 association, thus promoting LRP6 phosphorylation and Wnt signaling transduction. Moreover, we identified the critical residues on Axin for HLY78 binding and showed that HLY78 may weaken the autoinhibition of Axin. In addition, HLY78 acts synergistically with Wnt in the embryonic development of zebrafish and increases the expression of the conserved hematopoietic stem cell (HSC) markers, runx1 and cmyb, in zebrafish embryos. Collectively, our study not only provides new insights into the regulation of the Wnt/ß-catenin signaling pathway by a Wnt-specific small molecule but also will facilitate therapeutic applications, such as HSC expansion.


Subject(s)
Axin Protein/metabolism , Benzodioxoles/pharmacology , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Phenanthridines/pharmacology , Small Molecule Libraries/pharmacology , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , Animals , Axin Protein/antagonists & inhibitors , Axin Protein/chemistry , Benzodioxoles/chemistry , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-5/chemistry , Low Density Lipoprotein Receptor-Related Protein-6/chemistry , Phenanthridines/chemistry , Protein Binding/drug effects , Small Molecule Libraries/chemistry , Zebrafish/embryology , Zebrafish/metabolism
6.
Bioorg Med Chem Lett ; 23(20): 5511-4, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24018191

ABSTRACT

Based on the Wnt inhibitors as potential targets in the development of anticancer agents, natural compounds were evaluated for ß-catenin-mediated transcriptional activity. A natural lignan hydnocarpin isolated from Lonicera japonica was considered a potential inhibitor for Wnt/ß-catenin signalings. The anti-proliferative activity of hydnocarpin was also found to be associated with the suppression of Wnt/ß-catenin-mediated signaling pathway in human colon cancer cells. These data suggest that hydnocarpin might be a novel Wnt inhibitor and has a potential of signaling regulator in ß-catenin-mediated signaling pathways.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Flavonolignans/chemistry , Lignans/chemistry , Wnt Proteins/metabolism , beta Catenin/metabolism , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/toxicity , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Axin Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Flavonolignans/isolation & purification , Flavonolignans/toxicity , Humans , Lonicera/chemistry , Lonicera/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Wnt Signaling Pathway/drug effects
7.
Eur Rev Med Pharmacol Sci ; 22(15): 4828-4836, 2018 08.
Article in English | MEDLINE | ID: mdl-30070321

ABSTRACT

OBJECTIVE: Previous investigations have shown that miR-183 is upregulated in bladder cancer (BC); however, its biological significance is not fully investigated. The goal of the current study is to analyze the function of miR-183 in BC development and progression. PATIENTS AND METHODS: 23 pairs of BC tumor and adjacent tissues were analyzed for miR-183 and c-Myc expression using Real-time polymerase chain reaction (PCR). MiR-183 expression was modulated by transfection of miR-183 or miR-183 inhibitor (miR-183-in). Protein expression of AXIN2, c-Myc and Cyclin D1 was determined by western blot. Cell growth activity and apoptotic potential were evaluated by cell viability assay and flow cytometry assay, respectively. Luciferase activity assay was conducted to determine whether AXIN2 is a direct target of miR-183. RESULTS: The expression of miR-183 is upregulated in BC tissues and cell lines, and is positively correlated with the expression of the Wnt target gene, c-Myc. MiR-183 positively regulated Wnt signaling activity by directly suppressing its negative feedback regulator, AXIN2. Overexpression of miR-183 promoted cell growth and inhibited apoptosis. Inhibition of miR-183 attenuated cell growth and enhanced apoptosis. The effect of miR-183 on cell growth and apoptosis can be abolished by knockdown of AXIN2. CONCLUSIONS: MiR-183 functions as an oncomiR in BC and upregulates Wnt signaling activity by directly suppressing AXIN2 expression.


Subject(s)
Apoptosis/physiology , Axin Protein/biosynthesis , Cell Proliferation/physiology , MicroRNAs/biosynthesis , Urinary Bladder Neoplasms/metabolism , Wnt Signaling Pathway/physiology , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Cell Line, Tumor , Humans , MicroRNAs/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
8.
J Hypertens ; 34(5): 877-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26882042

ABSTRACT

OBJECTIVE: Pulmonary arterial hypertension (PAH) is characterized by increased vascular tone, altered vasoreactivity and vascular remodeling induced by smooth muscle cell proliferation. Similarities exist between cancer and PAH. Aberrant expression of the tumor suppressor protein is closely associated with PAH. Here, we tested the hypothesis that a tumor suppressor-axis inhibition protein 2 (Axin2) deficiency leads to PAH. METHODS AND RESULTS: We measured right ventricular systolic pressure in Axin2 knockout mice and assessed the expression of Axin2 in patients. We found that Axin2 expression level was decreased in both mice exposed to chronic hypoxia and patients with PAH in remodeled pulmonary arterioles. Axin2 knockout mice showed elevated mean right ventricular systolic pressure and enhanced contraction in response to phenylephrine. An increase in the cross-sectional area of the vessels was occupied by the vessel wall, indicating pulmonary vascular remodeling. Furthermore, knocking down Axin2 with small interfering RNA inhibited apoptosis of pulmonary arterial smooth muscle cells (PASMCs). This inhibition was significantly abolished by ß-catenin inhibitors, indicating that Axin2 through ß-catenin increased vascular wall by inhibiting the apoptosis of PASMCs. Importantly, overexpression of Axin2 attenuates the development of hypoxia-induced PAH in mice. CONCLUSION: Taken together, our study, for the first time, established that Axin2 plays a key role in the progression of PAH. We identified Axin2 as a novel mediator of pulmonary vasoconstriction and PASMC growth in hypoxia-mediated PAH. Our results suggest that downregulation of Axin2 in the pulmonary vasculature may be an underlying mechanism in the development of hypoxia-induced PAH.


Subject(s)
Axin Protein/metabolism , Hypertension, Pulmonary/physiopathology , beta Catenin/metabolism , Animals , Axin Protein/antagonists & inhibitors , Axin Protein/blood , Disease Models, Animal , Down-Regulation , Female , Humans , Hypertension, Pulmonary/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/metabolism , Signal Transduction
9.
FEBS Lett ; 590(1): 185-92, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26763102

ABSTRACT

Mineralizing osteoblasts (MOBs) can release exosomes, although the functional significance remains unclear. In the present study, we demonstrate that exosomes derived from mineralizing pre-osteoblast MC3T3-E1 cells can promote bone marrow stromal cell (ST2) differentiation to osteoblasts. We reveal that MOB-derived exosomes significantly influence miRNA profiles in recipient ST2 cells, and these changes tend to activate the Wnt signaling pathway by inhibiting Axin1 expression and increasing ß-catenin expression. We also suggest that MOB derived-exosomes partly induce the variation in miRNA expression in recipient ST2 cells by exosomal miRNA transfer. These findings suggest an exosome-mediated mode of cell-to-cell communication in the osteogenic microenvironment, and also indicate the potential of MOB exosomes in bone tissue engineering.


Subject(s)
Calcification, Physiologic , Exosomes/metabolism , Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Osteoblasts/metabolism , Osteogenesis , 3T3 Cells , Animals , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Axin Protein/metabolism , Biomarkers/metabolism , Cell Communication , Cell Line , Cellular Microenvironment , Computational Biology , Expert Systems , Gene Expression Profiling , Mesenchymal Stem Cells/cytology , Mice , Oligonucleotide Array Sequence Analysis , Osteoblasts/cytology , Wnt Signaling Pathway , beta Catenin/agonists , beta Catenin/genetics , beta Catenin/metabolism
10.
Oncol Rep ; 34(4): 1988-94, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26252180

ABSTRACT

MicroRNA-374a (miR-374a) is involved in the progress of various types of cancer, and may indicate a poor prognosis. However, its role in esophageal cancer remains to be determined. In the present study, the role of miR-374a in esophageal cancers and cancer cell growth was examined using miR-374a overexpression and underexpression models. The results showed that miR-374a was markedly increased in esophageal cancer cell lines and tumor samples from patients with esophageal cancer. In esophageal cancer Eca109 cells, the ectopic overexpression of miR-374a promoted cell growth. Additionally, cell growth was reduced by miR­374a inhibition. The mechanisms underlying the promotive role were examined and it was found that miR-374a significantly decreased the expression and transcription activity of axis inhibition protein 2 (Axin2). Axin2, a tumor suppressor, exhibited a marked inhibitory effect on Eca109 cell growth. The results identified a new role of miR-374a in esophageal cancer involving Axin2 suppression.


Subject(s)
Axin Protein/biosynthesis , Cell Proliferation/genetics , Esophageal Neoplasms/genetics , MicroRNAs/biosynthesis , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Cell Line, Tumor , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , MicroRNAs/genetics
11.
Protein Cell ; 5(3): 186-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24474204

ABSTRACT

The Wnt signaling pathway plays crucial roles during embryonic development, whose aberration is implicated in a variety of human cancers. Axin, a key component of canonical Wnt pathway, plays dual roles in modulating Wnt signaling: on one hand, Axin scaffolds the "ß-catenin destruction complex" to promote ß-catenin degradation and therefore inhibits the Wnt signal transduction; on the other hand, Axin interacts with LRP5/6 and facilitates the recruitment of GSK3 to the plasma membrane to promote LRP5/6 phosphorylation and Wnt signaling. The differential assemblies of Axin with these two distinct complexes have to be tightly controlled for appropriate transduction of the "on" or "off" Wnt signal. So far, there are multiple mechanisms revealed in the regulation of Axin activity, such as post-transcriptional modulation, homo/hetero-polymerization and auto-inhibition. These mechanisms may work cooperatively to modulate the function of Axin, thereby playing an important role in controlling the canonical Wnt signaling. In this review, we will focus on the recent progresses regarding the regulation of Axin function in canonical Wnt signaling.


Subject(s)
Axin Protein/metabolism , Wnt Signaling Pathway , Animals , Axin Protein/antagonists & inhibitors , Axin Protein/chemistry , Epigenesis, Genetic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Protein Processing, Post-Translational , Wnt Signaling Pathway/genetics
12.
PLoS One ; 8(7): e69593, 2013.
Article in English | MEDLINE | ID: mdl-23869245

ABSTRACT

While the TRAIL pathway represents a promising therapeutic target in melanoma, resistance to TRAIL-mediated apoptosis remains a barrier to its successful adoption. Since the Wnt/ß-catenin pathway has been implicated in facilitating melanoma cell apoptosis, we investigated the effect of Wnt/ß-catenin signaling on regulating the responses of melanoma cells to TRAIL. Co-treatment of melanoma cell lines with WNT3A-conditioned media and recombinant TRAIL significantly enhanced apoptosis compared to treatment with TRAIL alone. This apoptosis correlates with increased abundance of the pro-apoptotic proteins BCL2L11 and BBC3, and with decreased abundance of the anti-apoptotic regulator Mcl1. We then confirmed the involvement of the Wnt/ß-catenin signaling pathway by demonstrating that siRNA-mediated knockdown of an intracellular ß-catenin antagonist, AXIN1, or treating cells with an inhibitor of GSK-3 also enhanced melanoma cell sensitivity to TRAIL. These studies describe a novel regulation of TRAIL sensitivity in melanoma by Wnt/ß-catenin signaling, and suggest that strategies to enhance Wnt/ß-catenin signaling in combination with TRAIL agonists warrant further investigation.


Subject(s)
Apoptosis , Melanoma/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Wnt Signaling Pathway , Wnt3A Protein/pharmacology , beta Catenin/metabolism , Apoptosis Regulatory Proteins/metabolism , Axin Protein/antagonists & inhibitors , Bcl-2-Like Protein 11 , Cell Culture Techniques , Cell Line, Tumor , Culture Media, Conditioned , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Melanoma/pathology , Membrane Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , RNA Interference , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
13.
Cell Cycle ; 12(10): 1578-87, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23624843

ABSTRACT

p53 is a bona fide tumor suppressor gene whose loss of function marks the most common genetic alteration in human malignancy. Although the causal link between loss of p53 function and tumorigenesis has been clearly demonstrated, the mechanistic links by which loss of p53 potentiates oncogenic signaling are not fully understood. Recent evidence indicates that the microRNA-34 (miR-34) family, a transcriptional target of the p53, directly suppresses a set of canonical Wnt genes and Snail, resulting in p53-mediated suppression of Wnt signaling and the EMT process. In this study, we report that p53 regulates GSK-3ß nuclear localization via miR-34-mediated suppression of Axin2 in colorectal cancer. Exogenous miR-34a decreases Axin2 UTR-reporter activity through multiple binding sites within the 5' and 3' UTR of Axin2. Suppression of Axin2 by p53 or miR-34 increases nuclear GSK-3ß abundance and leads to decreased Snail expression in colorectal cancer cells. Conversely, expression of the non-coding UTR of Axin2 causes depletion of endogenous miR-34 via the miR-sponge effect together with increased Axin2 function, supporting that the RNA-RNA interactions with Axin2 transcripts act as an endogenous decoy for miR-34. Further, RNA transcripts of miR-34 target were correlated with Axin2 in clinical data set of colorectal cancer patients. Although the biological relevance of nuclear GSK-3 level has not been fully studied, our results demonstrate that the tumor suppressor p53/miR-34 axis plays a role in regulating nuclear GSK-3 levels and Wnt signaling through the non-coding UTR of Axin2 in colorectal cancer.


Subject(s)
Axin Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , MicroRNAs/metabolism , Tumor Suppressor Protein p53/metabolism , 3' Untranslated Regions , 5' Untranslated Regions , Antibiotics, Antineoplastic/pharmacology , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Binding Sites , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Transcription, Genetic , Wnt Signaling Pathway
14.
Cell Metab ; 18(4): 546-55, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24093678

ABSTRACT

The AMP-activated protein kinase (AMPK) is a master regulator of metabolic homeostasis by sensing cellular energy status. AMPK is mainly activated via phosphorylation by LKB1 when cellular AMP/ADP levels are increased. However, how AMP/ADP brings about AMPK phosphorylation remains unclear. Here, we show that it is AMP, but not ADP, that drives AXIN to directly tether LKB1 to phosphorylate AMPK. The complex formation of AXIN-AMPK-LKB1 is greatly enhanced in glucose-starved or AICAR-treated cells and in cell-free systems supplemented with exogenous AMP. Depletion of AXIN abrogated starvation-induced AMPK-LKB1 colocalization. Importantly, adenovirus-based knockdown of AXIN in the mouse liver impaired AMPK activation and caused exacerbated fatty liver after starvation, underscoring an essential role of AXIN in AMPK activation. These findings demonstrate an initiating role of AMP and demonstrate that AXIN directly transmits AMP binding of AMPK to its activation by LKB1, uncovering the mechanistic route for AMP to elicit AMPK activation by LKB1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/pharmacology , Axin Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , Acetyl-CoA Carboxylase/metabolism , Adenosine Monophosphate/metabolism , Animals , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Cell Line , Cell-Free System , Enzyme Activation , HEK293 Cells , Humans , Lipid Metabolism/physiology , Liver/cytology , Liver/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
15.
Cell Cycle ; 11(20): 3724-30, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22895053

ABSTRACT

The limitations of revolutionary new mutation-specific inhibitors of BRAF(V600E) include the universal recurrence seen in melanoma patients treated with this novel class of drugs. Recently, our lab showed that simultaneous activation of the Wnt/ß-catenin signaling pathway and targeted inhibition of BRAF(V600E) by PLX4720 synergistically induces apoptosis across a spectrum of BRAF(V600E) melanoma cell lines. As a follow-up to that study, treatment of BRAF-mutant and NRAS-mutant melanoma lines with WNT3A and the MEK inhibitor AZD6244 also induces apoptosis. The susceptibility of BRAF-mutant lines and NRAS-mutant lines to apoptosis correlates with negative regulation of Wnt/ß-catenin signaling by ERK/MAPK signaling and dynamic decreases in abundance of the downstream scaffolding protein, AXIN1. Apoptosis-resistant NRAS-mutant lines can sensitize to AZD6244 by pretreatment with AXIN1 siRNA, similar to what we previously reported in BRAF-mutant cell lines. Taken together, these findings indicate that NRAS-mutant melanoma share with BRAF-mutant melanoma the potential to regulate apoptosis upon MEK inhibition through WNT3A and dynamic regulation of cellular AXIN1. Understanding the cellular context that makes melanoma cells susceptible to this combination treatment will contribute to the study and development of novel therapeutic combinations that may lead to more durable responses.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Indoles/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Sulfonamides/pharmacology , Wnt3A Protein/pharmacology , Apoptosis/drug effects , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Axin Protein/metabolism , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mutation , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , beta Catenin/agonists , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL