Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Int J Cancer ; 155(3): 582-594, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38380807

The proto-oncogene MYCN expression marked a cancer stem-like cell population in hepatocellular carcinoma (HCC) and served as a therapeutic target of acyclic retinoid (ACR), an orally administered vitamin A derivative that has demonstrated promising efficacy and safety in reducing HCC recurrence. This study investigated the role of MYCN as a predictive biomarker for therapeutic response to ACR and prognosis of HCC. MYCN gene expression in HCC was analyzed in the Cancer Genome Atlas and a Taiwanese cohort (N = 118). Serum MYCN protein levels were assessed in healthy controls (N = 15), patients with HCC (N = 116), pre- and post-surgical patients with HCC (N = 20), and a subset of patients from a phase 3 clinical trial of ACR (N = 68, NCT01640808). The results showed increased MYCN gene expression in HCC tumors, which positively correlated with HCC recurrence in non-cirrhotic or single-tumor patients. Serum MYCN protein levels were higher in patients with HCC, decreased after surgical resection of HCC, and were associated with liver functional reserve and fibrosis markers, as well as long-term HCC prognosis (>4 years). Subgroup analysis of a phase 3 clinical trial of ACR identified serum MYCN as the risk factor most strongly associated with HCC recurrence. Patients with HCC with higher serum MYCN levels after a 4-week treatment of ACR exhibited a significantly higher risk of recurrence (hazard ratio 3.27; p = .022). In conclusion, serum MYCN holds promise for biomarker-based precision medicine for the prevention of HCC, long-term prognosis of early-stage HCC, and identification of high-response subgroups for ACR-based treatment.


Biomarkers, Tumor , Carcinoma, Hepatocellular , Liver Neoplasms , N-Myc Proto-Oncogene Protein , Neoplasm Recurrence, Local , Proto-Oncogene Mas , Adult , Aged , Female , Humans , Male , Middle Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/blood , Liver Neoplasms/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/prevention & control , Liver Neoplasms/pathology , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Recurrence, Local/prevention & control , Neoplasm Recurrence, Local/blood , Prognosis
2.
Cell Death Dis ; 14(6): 358, 2023 06 13.
Article En | MEDLINE | ID: mdl-37308486

Transglutaminase 2 (TG2) is a multifunctional protein that promotes or suppresses tumorigenesis, depending on intracellular location and conformational structure. Acyclic retinoid (ACR) is an orally administered vitamin A derivative that prevents hepatocellular carcinoma (HCC) recurrence by targeting liver cancer stem cells (CSCs). In this study, we examined the subcellular location-dependent effects of ACR on TG2 activity at a structural level and characterized the functional role of TG2 and its downstream molecular mechanism in the selective depletion of liver CSCs. A binding assay with high-performance magnetic nanobeads and structural dynamic analysis with native gel electrophoresis and size-exclusion chromatography-coupled multi-angle light scattering or small-angle X-ray scattering showed that ACR binds directly to TG2, induces oligomer formation of TG2, and inhibits the transamidase activity of cytoplasmic TG2 in HCC cells. The loss-of-function of TG2 suppressed the expression of stemness-related genes, spheroid proliferation and selectively induced cell death in an EpCAM+ liver CSC subpopulation in HCC cells. Proteome analysis revealed that TG2 inhibition suppressed the gene and protein expression of exostosin glycosyltransferase 1 (EXT1) and heparan sulfate biosynthesis in HCC cells. In contrast, high levels of ACR increased intracellular Ca2+ concentrations along with an increase in apoptotic cells, which probably contributed to the enhanced transamidase activity of nuclear TG2. This study demonstrates that ACR could act as a novel TG2 inhibitor; TG2-mediated EXT1 signaling is a promising therapeutic target in the prevention of HCC by disrupting liver CSCs.


Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Protein Glutamine gamma Glutamyltransferase 2 , Neoplastic Stem Cells , Glycosyltransferases
3.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article En | MEDLINE | ID: mdl-34769072

Inhaled nebulized interferon (IFN)-α and IFN-ß have been shown to be effective in the management of coronavirus disease 2019 (COVID-19). We aimed to construct a virus-free rapid detection system for high-throughput screening of IFN-like compounds that induce viral RNA degradation and suppress the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We prepared a SARS-CoV-2 subreplicon RNA expression vector which contained the SARS-CoV-2 5'-UTR, the partial sequence of ORF1a, luciferase, nucleocapsid, ORF10, and 3'-UTR under the control of the cytomegalovirus promoter. The expression vector was transfected into Calu-3 cells and treated with IFN-α and the IFNAR2 agonist CDM-3008 (RO8191) for 3 days. SARS-CoV-2 subreplicon RNA degradation was subsequently evaluated based on luciferase levels. IFN-α and CDM-3008 suppressed SARS-CoV-2 subreplicon RNA in a dose-dependent manner, with IC50 values of 193 IU/mL and 2.54 µM, respectively. HeLa cells stably expressing SARS-CoV-2 subreplicon RNA were prepared and treated with the IFN-α and pan-JAK inhibitor Pyridone 6 or siRNA-targeting ISG20. IFN-α activity was canceled with Pyridone 6. The knockdown of ISG20 partially canceled IFN-α activity. Collectively, we constructed a virus-free rapid detection system to measure SARS-CoV-2 RNA suppression. Our data suggest that the SARS-CoV-2 subreplicon RNA was degraded by IFN-α-induced ISG20 exonuclease activity.


Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Interferon-alpha/pharmacology , RNA, Viral/metabolism , SARS-CoV-2/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Exoribonucleases/genetics , Genetic Vectors , HeLa Cells , Humans , Interferon-alpha/administration & dosage , Luciferases/genetics , Luciferases/metabolism , Naphthyridines/administration & dosage , Naphthyridines/pharmacology , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacology , RNA, Viral/drug effects , Replicon
4.
Cell Death Discov ; 7(1): 130, 2021 Jun 02.
Article En | MEDLINE | ID: mdl-34078875

Chronic hepatitis B virus (HBV) infections remain a health burden affecting ~250 million people worldwide. Thus far, available interferon-alpha (IFNα)-based therapies have shown unsatisfactory cure rates, and alternative therapeutic molecules are still required. However, their development has been hampered because accessible cell models supporting relevant HBV replication and appropriate antiviral activity are lacking. Strategies that reverse epigenetic alterations offer a unique opportunity for cell reprogramming, which is valuable for restoring altered cellular functions in human cell lines. This work aimed to investigate the feasibility of converting HepG2 cells that stably overexpress the HBV entry receptor (sodium/taurocholate cotransporting polypeptide, NTCP) toward IFNα-responsive cells using epigenetic reprogramming. Herein, we showed that an epigenetic regimen with non-cytotoxic doses of the demethylating compound 5-azacytidine restored the anti-HBV action of IFNα in epigenetically reprogrammed HepG2-NTCP-C4 cells, named REP-HepG2-NTCP cells. Thus, a significant inhibition in HBV DNA levels was measured in REP-HepG2-NTCP cells after IFNα treatment. This inhibitory effect was associated with the enhancement of IFNα-mediated induction of critical interferon-stimulated genes (ISGs), which was limited in non-reprogrammed cells. In particular, our data indicated that re-expression of 2'-5'-oligoadenylate synthetase 1 (OAS1) and interferon regulatory factor 9 (IRF9) was the result of an epigenetically driven unmasking of these genes in reprogrammed cells. At last, we evaluated the therapeutic potential of the IFN analog CDM-3008 in REP-HepG2-NTCP cells and demonstrated the efficiency of this chemical compound in triggering ISG induction and HBV inhibition. In summary, this study shows that epigenetic reprogramming promotes the IFNα response in HBV-infected cells and is potentially attractive for cell-based experimental screening of IFN-like compounds.

5.
Front Oncol ; 10: 618515, 2020.
Article En | MEDLINE | ID: mdl-33937011

Upregulated MYCN gene expression is restricted to specialized cell populations such as EpCAM+ cancer stem cells in liver cancer, regardless of DNA amplification and mutation. Here, we reviewed the role of MYCN gene expression in liver homeostasis, regeneration, and tumorigenesis, and discussed the potential non-genomic mechanisms involved in controlling MYCN gene expression in liver cancer, with a focus on inflammation-mediated signal transduction and microRNA-associated post-transcriptional regulation. We concluded that dynamic MYCN gene expression is an integrated consequence of multiple signals in the tumor microenvironment, including tumor growth-promoting signals, lipid desaturation-mediated endoplasmic reticulum stress adaptation signals, and tumor suppressive miRNAs, making it a potential predictive biomarker of tumor stemness and plasticity. Therefore, understanding and tracing the dynamic changes and functions of MYCN gene expression will shed light on the origin of liver tumorigenesis at the cellular level and the development of novel therapeutic and diagnostic strategies for liver cancer treatment.

6.
Cancer Sci ; 111(3): 869-880, 2020 Mar.
Article En | MEDLINE | ID: mdl-31883160

Primary hepatic tumors mainly include hepatocellular carcinoma (HCC), which is one of the most frequent causes of cancer-related deaths worldwide. Thus far, HCC prognosis has remained extremely poor given the lack of effective treatments. Numerous studies have described the roles played by microRNAs (miRNAs) in cancer progression and the potential of these small noncoding RNAs for diagnostic or therapeutic applications. The current consensus supports the idea that direct repression of a wide range of oncogenes by a single key miRNA could critically affect the malignant properties of cancer cells in a synergistic manner. In this study, we aimed to investigate the oncogenes controlled by miR-493-5p, a major tumor suppressor miRNA that inactivates miR-483-3p oncomir in hepatic cancer cells. Using global gene expression analysis, we highlighted a set of candidate genes potentially regulated by miR-493-5p. In particular, the canonical MYCN protooncogene (MYCN) appeared to be an attractive target of miR-493-5p given its significant inhibition through 3'-UTR targeting in miR-493-5p-rescued HCC cells. We showed that MYCN was overexpressed in liver cancer cell lines and clinical samples from HCC patients. Notably, MYCN expression levels were inversely correlated with miR-493-5p in tumor tissues. We confirmed that MYCN knockdown mimicked the anticancer effect of miR-493-5p by inhibiting HCC cell growth and invasion, whereas MYCN rescue hindered miR-493-5p activity. In summary, miR-493-5p is a pivotal miRNA that modulates various oncogenes after its reexpression in liver cancer cells, suggesting that tumor suppressor miRNAs with a large spectrum of action could provide valuable tools for miRNA replacement therapies.


Carcinoma, Hepatocellular/genetics , Cell Proliferation/genetics , Liver Neoplasms/genetics , N-Myc Proto-Oncogene Protein/genetics , Oncogenes/genetics , 3' Untranslated Regions/genetics , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor/physiology , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , MicroRNAs , Middle Aged , Prognosis , Proto-Oncogenes/genetics
7.
Stem Cells ; 38(4): 504-515, 2020 04.
Article En | MEDLINE | ID: mdl-31828873

The role of microRNAs (miRNAs) during mouse early development, especially in endoderm germ layer formation, is largely unknown. Here, via miRNA profiling during endoderm differentiation, we discovered that miR-124a negatively regulates endoderm lineage commitment in mouse embryonic stem cells (mESCs). To further investigate the functional role of miR-124a in early stages of differentiation, transfection of embryoid bodies with miR-124a mimic was performed. We showed that overexpression of miR-124a inhibits endoderm differentiation in vitro through targeting the 3'-untranslated region (UTR) of Sox17 and Gata6, revealing the existence of interplay between miR-124a and the Sox17/Gata6 transcription factors in hepato-specific gene regulation. In addition, we presented a feasible in vivo system that utilizes teratoma and gene expression profiling from microarray to quantitatively evaluate the functional role of miRNA in lineage specification. We demonstrated that ectopic expression of miR-124a in teratomas by intratumor delivery of miR-124a mimic and Atelocollagen, significantly suppressed endoderm and mesoderm lineage differentiation while augmenting the differentiation into ectoderm lineage. Collectively, our findings suggest that miR-124a plays a significant role in mESCs lineage commitment.


GATA6 Transcription Factor/metabolism , MicroRNAs/metabolism , Mouse Embryonic Stem Cells/metabolism , SOXF Transcription Factors/metabolism , Animals , Cell Lineage , Endoderm , Mice , Transfection
8.
Cancer Sci ; 111(2): 395-405, 2020 Feb.
Article En | MEDLINE | ID: mdl-31854049

The onset of breast cancer among young patients is a major issue in cancer etiology. Our previous study has shown that poor prognosis in young women with breast cancer is associated with lower expression of the microRNA miR-1285-5p. In this study, we showed that the expression of miR-1285-5p is lower in tumor tissues than in normal tissues. Accumulating evidence suggests that miR-1285-5p plays critical roles in various types of cancers. However, the functional role of miR-1285-5p in breast cancer remains to be elucidated. Here, we showed the tumor-suppressive role of miR-1285-5p and detailed its mechanism of action in breast cancer. Overexpression of miR-1285-5p significantly inhibited cell proliferation in breast cancer cells regardless of the tumor subtype. Among the target genes of miR-1285-5p, we found that transmembrane protein 194A (TMEM194A) was directly regulated by miR-1285-5p. Notably, separation of centrosomes from the nuclear envelope was observed upon knockdown of TMEM194A or overexpression of miR-1285-5p. In conclusion, our findings show that miR-1285-5p is a tumor suppressor via TMEM194A inhibition in breast cancer.


Breast Neoplasms/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , ran GTP-Binding Protein/genetics , 3' Untranslated Regions , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Centrosome/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Nuclear Proteins/metabolism , ran GTP-Binding Protein/metabolism
9.
Cell Death Dis ; 10(8): 553, 2019 07 18.
Article En | MEDLINE | ID: mdl-31320614

Numerous studies have described the critical role played by microRNAs (miRNAs) in cancer progression and the potential of these small non-coding RNAs for diagnostic or therapeutic applications. However, the mechanisms responsible for the altered expression of miRNAs in malignant cells remain poorly understood. Herein, via epigenetic unmasking, we identified a group of miRNAs located in the imprinted delta like non-canonical Notch ligand 1 (DLK1)-maternally expressed 3 (MEG3) locus that were repressed in hepatic tumor cells. Notably, miR-493-5p epigenetic silencing was correlated with hypermethylation of the MEG3 differentially regulated region (DMR) in liver cancer cell lines and tumor tissues from patients. Experimental rescue of miR-493-5p promoted an anti-cancer response by hindering hepatocellular carcinoma (HCC) cell growth in vitro and tumor progression in vivo. We found that miR-493-5p mediated part of its tumor-suppressor activity by abrogating overexpression of insulin-like growth factor 2 (IGF2) and the IGF2-derived intronic oncomir miR-483-3p in HCC cells characterized by IGF2 loss of imprinting (LOI). In summary, this study describes an unknown miRNA-dependent regulatory mechanism between two distinct imprinted loci and a possible therapeutic window for liver cancer patients exhibiting IGF2-miR-483 LOI and amplification.


Carcinoma, Hepatocellular/genetics , Genomic Imprinting/genetics , Insulin-Like Growth Factor II/genetics , Liver Neoplasms/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Humans , Insulin-Like Growth Factor II/metabolism , Introns , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Transplantation, Heterologous
10.
Mol Ther ; 26(7): 1840-1854, 2018 07 05.
Article En | MEDLINE | ID: mdl-29759938

Primary liver tumors are mainly represented by hepatocellular carcinoma (HCC), one of the most aggressive and resistant forms of cancer. Liver tumorigenesis is characterized by an accumulation of epigenetic abnormalities, leading to gene extinction and loss of hepatocyte differentiation. The aim of this work was to investigate the feasibility of converting liver cancer cells toward a less aggressive and differentiated phenotype using a process called epigenetic reconditioning. Here, we showed that an epigenetic regimen with non-cytotoxic doses of the demethylating compound 5-azacytidine (5-AZA) promoted an anti-cancer response by inhibiting HCC cell tumorigenicity. Furthermore, epigenetic reconditioning improved sorafenib response. Remarkably, epigenetic treatment was associated with a significant restoration of differentiation, as attested by the increased expression of characteristic hepatocyte markers in reconditioned cells. In particular, we showed that reexpression of these epigenetically silenced liver genes following 5-AZA treatment or after knockdown of DNA methyltransferase 1 (DNMT1) was the result of regional CpG demethylation. Lastly, we confirmed the efficacy of HCC differentiation therapy by epigenetic reconditioning using an in vivo tumor growth model. In summary, this work demonstrates that epigenetic reconditioning using the demethylating compound 5-AZA shows therapeutic significance for liver cancer and is potentially attractive for the treatment of solid tumors.


Carcinogenesis/genetics , Cell Differentiation/genetics , Epigenesis, Genetic/genetics , Liver Neoplasms/genetics , Animals , Azacitidine/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , DNA Methylation/drug effects , DNA Methylation/genetics , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Liver/drug effects , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Mice, Nude
11.
Cell Death Dis ; 9(5): 468, 2018 05 01.
Article En | MEDLINE | ID: mdl-29700299

Curative management of pancreatic adenocarcinoma is limited because this malignancy remains resistant to most chemotherapeutic drugs. Strategies that reverse epigenetic alterations offer a unique opportunity for cancer cell reprogramming, which is valuable for development of new treatments. The aim of this work was to reprogram pancreatic ductal adenocarcinoma (PDAC) cells toward a less aggressive and drug-responsive phenotype. The process applied is called "epigenetic reprogramming". To evaluate the efficiency of PDAC epigenetic reprogramming, we assessed tumor growth and drug response in PANC-1 cells after exposure to non-cytotoxic doses of the demethylating agent 5-azacytidine (5-AZA). Here, we showed that an epigenetic regimen using 5-AZA promoted an anti-cancer response by inhibiting PDAC tumor growth in vivo after the engraftment of treated cells. Remarkably, the subsequent addition of gemcitabine (GEM) to the 5-AZA-mediated reprogramming resulted in a marked growth inhibition effect in GEM-resistant pancreatic cancer cells. We observed that various characteristic peptides expressed in the pancreas, which included the antiproliferative hormone somatostatin (SST) and the SST receptor 2 (SSTR2), were significantly upregulated in the epigenetically reprogrammed PDAC cells. The inhibitory effect of octreotide (OCT), an SST analog, was tested on PDAC cells and found to be improved after cell reprogramming. Furthermore, we found that SST gene expression restoration following 5-AZA treatment or following knockdown of the DNA methyltransferase (DNMT) 1 enzyme was associated with the reversion of SST epigenetic silencing through regional CpG demethylation. Lastly, we confirmed the efficacy of 5-AZA-based epigenetic reprogramming in vivo using a PDAC tumor growth model. In conclusion, this study demonstrates that epigenetic reprogramming using the demethylating compound 5-AZA shows anti-cancer effects in PANC-1 cells and is potentially attractive for the treatment of solid tumors.


Azacitidine/pharmacology , Carcinoma, Pancreatic Ductal , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
Int Immunol ; 29(1): 11-19, 2017 01 01.
Article En | MEDLINE | ID: mdl-28184439

One of the pathological hallmarks of Alzheimer's disease (AD) is the presence of extracellular plaques resulting from the accumulation of beta-amyloid peptide (Aß). To date, a definitive cure for this disease is still lacking as the currently approved drugs used are mainly symptomatic treatments. The revolutionary discovery of extracellular vesicles (EVs) has shed new light on the development of disease-modifying treatments for AD, owing to their potential in delivering the therapeutic agents to the brain. The feasibility of harnessing EVs for clinical applications is highly dependent on the donor cell, which determines the intrinsic properties of EVs. The merit of mesenchymal stem cells (MSCs) as therapeutic delivery vehicles, and the proven therapeutic effects of the EVs derived from these cells, make researchers esteem MSCs as ideal producers of EVs. Therefore, MSC-derived EVs (MSC-EVs) emerge to be an appealing therapeutic delivery approach for the treatment of AD. Here, we discuss perspectives on the therapeutic strategies using MSC-EVs to treat AD and the associated challenges in clinical application.


Alzheimer Disease/therapy , Cell-Derived Microparticles/transplantation , Mesenchymal Stem Cells , Alzheimer Disease/immunology , Animals , Humans
14.
FEBS Lett ; 589(24 Pt B): 4071-8, 2015 Dec 21.
Article En | MEDLINE | ID: mdl-26632510

Extracellular vesicles (EVs) contain microRNAs (miRNAs). However, the exact molecular mechanisms of the recruitment of miRNAs in EVs are not well characterized. Based on proteomic analysis, we identified that silencing of Annexin A2 (ANXA2) significantly decreased the amount of miRNAs in EVs. In addition, microarray analysis revealed that ANXA2 regulated the loading of miRNAs into EVs in a sequence independent manner. Lastly, immunoprecipitation analysis confirmed that ANXA2 could bind miRNAs in EVs in the presence of Ca(2+). These observations demonstrate that ANXA2 plays an important role in the packaging process of miRNAs into EVs.


Annexin A2/metabolism , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , Up-Regulation , Annexin A2/antagonists & inhibitors , Annexin A2/genetics , Annexin A5/antagonists & inhibitors , Annexin A5/genetics , Annexin A5/metabolism , Biological Transport , Calcium Signaling , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , MicroRNAs/chemistry , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Particle Size , Proteomics/methods , RNA Interference , RNA, Neoplasm/chemistry , RNA, Small Interfering , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
15.
Sci Rep ; 5: 14697, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26423775

Certain dietary agents, such as natural products, have been reported to show anti-cancer effects. However, the underlying mechanisms of these substances in human cancer remain unclear. We recently found that resveratrol exerts an anti-cancer effect by upregulating tumour-suppressor microRNAs (miRNAs). In the current study, we aimed to identify new dietary products that have the ability to activate tumour-suppressor miRNAs and that therefore may serve as novel tools for the prevention and treatment of human cancers. We describe the generation and use of an original screening system based on a luciferase-based reporter vector for monitoring miR-200c tumour-suppressor activity. By screening a library containing 139 natural substances, three natural compounds - enoxolone, magnolol and palmatine chloride - were identified as being capable of inducing miR-200c expression in breast cancer cells at 10 µM. Moreover, these molecules suppressed the invasiveness of breast cancer cells in vitro. Next, we identified a molecular pathway by which the increased expression of miR-200c induced by natural substances led to ZEB1 inhibition and E-cadherin induction. These results indicate that our method is a valuable tool for a fast identification of natural molecules that exhibit tumour-suppressor activity in human cancer through miRNA activation.


Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Dietary Supplements , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , MicroRNAs/genetics , Berberine Alkaloids/pharmacology , Biphenyl Compounds/pharmacology , Cadherins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/genetics , Glycyrrhetinic Acid/pharmacology , Homeodomain Proteins/genetics , Humans , Lignans/pharmacology , Phenotype , Small Molecule Libraries , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1
16.
J Cell Physiol ; 229(7): 903-15, 2014 Jul.
Article En | MEDLINE | ID: mdl-24501087

The extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein (MAP) kinase pathway has been involved in the positive and negative regulation of cell proliferation. Upon mitogen stimulation, ERK1/ERK2 activation is necessary for G1- to S-phase progression whereas when hyperactived, this pathway could elicit cell cycle arrest. The mechanisms involved are not fully elucidated but a kinase-independent function of ERK1/2 has been evidenced in the MAPK-induced growth arrest. Here, we show that p70S6K, a central regulator of protein biosynthesis, is essential for the cell cycle arrest induced by overactivation of ERK1/2. Indeed, whereas MEK1 silencing inhibits cell cycle progression, we demonstrate that active mutant form of MEK1 or MEK2 triggers a G1 phase arrest by stimulating an activation of p70S6K by ERK1/2 kinases. Silencing of ERK1/2 activity by shRNA efficiently suppresses p70S6K phosphorylation on Thr421/Ser424 and S6 phosphorylation on Ser240/244 as well as p21 expression, but these effects can be partially reversed by the expression of kinase-dead mutant form of ERK1 or ERK2. In addition, we demonstrate that the kinase p70S6K modulates neither the p21 gene transcription nor the stability of the protein but enhances the translation of the p21 mRNA. In conclusion, our data emphasizes the importance of the translational regulation of p21 by the MEK1/2-ERK1/2-p70S6K pathway to negatively control the cell cycle progression.


MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , MAP Kinase Signaling System/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Cell Cycle Checkpoints/genetics , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , G1 Phase Cell Cycle Checkpoints/genetics , Hep G2 Cells , Humans , MAP Kinase Kinase 1/biosynthesis , MAP Kinase Kinase 2/biosynthesis , Phosphorylation , Protein Biosynthesis , RNA, Small Interfering , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis , Signal Transduction
17.
Hepatology ; 58(3): 1153-65, 2013 Sep.
Article En | MEDLINE | ID: mdl-23532995

UNLABELLED: MicroRNAs (miRNAs) are evolutionary conserved small RNAs that post-transcriptionally regulate the expression of target genes. To date, the role of miRNAs in liver development is not fully understood. By using an experimental model that allows the induced and controlled differentiation of mouse fetal hepatoblasts (MFHs) into mature hepatocytes, we identified miR-148a as a hepatospecific miRNA highly expressed in adult liver. The main finding of this study revealed that miR-148a was critical for hepatic differentiation through the direct targeting of DNA methyltransferase (DNMT) 1, a major enzyme responsible for epigenetic silencing, thereby allowing the promotion of the "adult liver" phenotype. It was also confirmed that the reduction of DNMT1 by RNA interference significantly promoted the expression of the major hepatic biomarkers. In addition to the essential role of miR-148a in hepatocyte maturation, we identified its beneficial effect through the repression of hepatocellular carcinoma (HCC) cell malignancy. miR-148a expression was frequently down-regulated in biopsies of HCC patients as well as in mouse and human HCC cell lines. Overexpressing miR-148a led to an enhancement of albumin production and a drastic inhibition of the invasive properties of HCC cells, whereas miR-148a silencing had the opposite consequences. Finally, we showed that miR-148a exerted its tumor-suppressive effect by regulating the c-Met oncogene, regardless of the DNMT1 expression level. CONCLUSION: miR-148a is essential for the physiology of the liver because it promotes the hepatospecific phenotype and acts as a tumor suppressor. Most important, this report is the first to demonstrate a functional role for a specific miRNA in liver development through regulation of the DNMT1 enzyme.


Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Hepatocytes/pathology , Liver Neoplasms/pathology , MicroRNAs/physiology , Phenotype , Albumins/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Cell Differentiation , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Disease Models, Animal , Down-Regulation/physiology , Humans , Liver Neoplasms/metabolism , Mice , Neoplasm Invasiveness/pathology
18.
Front Biosci (Landmark Ed) ; 18(1): 199-223, 2013 01 01.
Article En | MEDLINE | ID: mdl-23276918

Primary liver tumors are mainly represented by hepatocellular hepatocarcinoma (HCC), one of the most aggressive and resistant forms of cancer. Numerous studies have reported the key role of microRNAs (miRNAs) in development, cell proliferation, apoptosis, and tumor biology. The alteration of cancer-related miRNA expression can be associated with tumorigenesis. In HCC, deregulated miRNAs frequently act as oncogenes or altered tumor suppressors. Distinct subtypes of hepatic cancer can also be related to an aberrant expression of particular miRNAs, arguing for the significance of using miRNAs as tumor biomarkers in order to refine the HCC grading assessment. In this article, we review the latest reports regarding miRNA profiling and the potential of small RNAs in HCC diagnosis. The relevance of cancer-related miRNA signatures for the prognosis and better understanding of liver cancer outcome is then considered.


Carcinoma, Hepatocellular/diagnosis , Liver Neoplasms/diagnosis , MicroRNAs , Apoptosis/genetics , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Exosomes/metabolism , Fatty Liver/complications , Fatty Liver, Alcoholic/complications , Hepatitis B, Chronic/complications , Hepatitis C, Chronic/complications , Humans , Liver Cirrhosis/complications , Liver Neoplasms/etiology , MicroRNAs/biosynthesis , Non-alcoholic Fatty Liver Disease , Prognosis
19.
Histol Histopathol ; 28(4): 437-51, 2013 04.
Article En | MEDLINE | ID: mdl-23224781

MicroRNAs (miRNAs) have emerged as key factors involved in several biological processes, including development, differentiation, cell proliferation, and tumorigenesis. In hepatocellular carcinoma (HCC), miRNAs frequently present aberrant expression profiles, which make them potentially attractive for diagnostic or prognostic applications. Currently, accumulating evidence is indicating the role of miRNAs as tumor suppressors or oncogenes in hepatic malignancies. In particular, comprehensive studies have made possible a better understanding of HCC behavior, such as tumor growth, response to therapies, metastatic potential, or recurrence, regarding the altered expression of cancer-related miRNAs. Based on these findings, efforts are under way to define new markers for liver cancer in both invasive (hepatic biopsy or tumor resection) and non-invasive (circulating miRNAs in blood serum) ways. Due to their implication in the control of various cell processes altered in HCC, cancer-related miRNAs also offer encouraging perspectives for the development of innovative cancer therapies. In this article, we review the importance of miRNA deregulation in HCC progression and the role of these small non-coding RNAs as tumor suppressors and oncogenes. The significance of miRNAs in HCC diagnosis and miRNA-based therapeutic strategies is then discussed.


Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Liver Neoplasms/genetics , MicroRNAs/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , Genetic Therapy , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , MicroRNAs/therapeutic use , Prognosis
20.
Methods Mol Biol ; 826: 33-47, 2012.
Article En | MEDLINE | ID: mdl-22167638

During development, liver precursors constitute a valuable source of pluripotent stem cells that present the ability to differentiate into both a hepatic and biliary lineage. In the present chapter, we report an experimental procedure developed by our group to isolate mouse fetal hepatoblasts (MFHs) with high purity. The method is based on a selective harvesting of the hepatic parenchymal cells from fetuses (E 14.5), followed by the sorting of E-cadherin(+) progenitors through the use of magnetic beads and specific antibodies. This protocol allows the isolation of bipotent liver stem cells expressing both hepatic and biliary markers. Primary cultures of purified MFHs can be maintained under proliferation until confluence, leading to promotion of the differentiation process in the presence of hepatotrophic factors. By using a quantitative real-time polymerase chain reaction approach, we show the hepatospecific phenotype and the progressive maturation of MFHs, delineating early (α-fetoprotein), mid (albumin), and late (glucose-6-phosphatase) hepatic markers. Consequently, the model appears to be a valuable cell system for the study of molecular and cellular aspects occurring in hepatic differentiation.


Cell Culture Techniques/methods , Cell Separation/methods , Fetus/cytology , Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Albumins/metabolism , Animals , Biomarkers/metabolism , Cell Proliferation , Glucose-6-Phosphatase/metabolism , Mice , Real-Time Polymerase Chain Reaction , alpha-Fetoproteins/metabolism
...