Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
3.
Mol Ther Nucleic Acids ; 14: 171-183, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30594073

ABSTRACT

miR675 is highly expressed in several human tumor tissues and positively regulates cell progression. Herein, we demonstrate that miR675 promotes malignant transformation of human mesenchymal stem cells. Mechanistically, we reveal that miR675 enhances the expression of the polyubiquitin-binding protein p62. Intriguingly, P62 competes with SETD2 to bind histone H3 and then significantly reduces SETD2-binding capacity to substrate histone H3, triggering drastically the reduction of three methylation on histone H3 36th lysine (H3K36me3). Thereby, the H3K36me3-hMSH6-SKP2 triplex complex is significantly decreased. Notably, the ternary complex's occupancy capacity on chromosome is absolutely reduced, preventing it from DNA damage repair. By virtue of the reductive degradation ability of SKP2 for aging histone H3.3 bound to mismatch DNA, the aging histone H3.3 repair is delayed. Therefore, the mismatch DNA escapes from repair, triggering the abnormal expression of several cell cycle-related genes and causing the malignant transformation of mesenchymal stem cells. These observations strongly suggest understanding the novel functions of miR675 will help in the development of novel therapeutic approaches in a broad range of cancer types.

4.
J Mol Med (Berl) ; 96(10): 1119-1130, 2018 10.
Article in English | MEDLINE | ID: mdl-30140938

ABSTRACT

Both miR675 and pyruvate kinase M2 (PKM2) contribute to malignant progression of tumor, but its functions in liver cancer stem cells remain unclear. Herein, our findings indicate that miR675 plus PKM2 strongly promotes the growth of liver cancer stem cells. Mechanistically, miR675 plus PKM2 enhances the transcriptional activity of SUV39h2. On the other hand, the excessive SUV39h2 binds to more substrate histone H3, triggering an increase of tri-methylation of histone H3 on the ninth lysine. Furthermore, the tri-methylation of histone 3 on the ninth lysine (H3K9me3)-heterochromatin protein 1 alpha (HP1α) complex is increased when the complex occupancy ability on the C-myc promoter region is raised, recruiting CREB, P300, and RNApolII to the special position that results in C-myc high abundance. Therefore, miR675 plus PKM2 triggered the upregulation of C-myc by increasing the interaction between H3K9me3 and HP1α. Understanding the signaling pathways that miR675 plus PKM2 epigenetically possesses during the malignant transformation of liver cancer stem cells will contribute to more effective liver cancer therapies.


Subject(s)
Carrier Proteins , Embryonic Stem Cells , Liver Neoplasms , Membrane Proteins , MicroRNAs , Neoplastic Stem Cells , Thyroid Hormones , Animals , Cell Line , Cell Proliferation , Chromobox Protein Homolog 5 , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Thyroid Hormone-Binding Proteins
5.
Mol Ther Nucleic Acids ; 11: 367-381, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29858072

ABSTRACT

Inflammatory and autophagy-related gene P62 is highly expressed in most human tumor tissues. Herein, we demonstrate that P62 promotes human mesenchymal stem cells' malignant transformation via the cascade of P62-tumor necrosis factor alpha (TNF-α)-CUDR-CTCF-insulin growth factor II (IGFII)-H-Ras signaling. Mechanistically, we reveal P62 enhances IGFII transcriptional activity through forming IGFII promoter-enhancer chromatin loop and increasing METTL3 occupancy on IGFII 3' UTR and enhances H-Ras overexpression by harboring inflammation-related factors, e.g., TNFR1, CLYD, EGR1, NFκB, TLR4, and PPARγ. Furthermore, the P62 cooperates with TNF-α to promote malignant transformation of mesenchymal stem cells. These findings, for the first time, provide insight into the positive role that P62 plays in malignant transformation of mesenchymal stem cells and reveal a novel link between P62 and the inflammation factors in mesenchymal stem cells.

6.
Mol Ther Nucleic Acids ; 11: 494-507, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29858084

ABSTRACT

MicroRNAs are known to be involved in carcinogenesis. Recently, microRNA-372 (miR372) has been proven to play a substantial role in several human cancers, but its functions in liver cancer remain unclear. Herein, our results demonstrate that miR372 accelerates growth of liver cancer cells in vitro and in vivo. Mechanistically, miR372 enhances expression of Y-box-binding protein 1 (YB-1) by targeting for phosphatase and tensin homolog (PTEN) directly and consequently promotes phosphorylation of YB-1 via HULC looping dependent on ERK1/2 and PTEN. In particular, HULC knockdown or PTEN overexpression abrogated this miR372 action. Moreover, miR372 inhibits the degradation of ß-catenin dependent on phosphorylation of YB-1 and then enhances the expression and activity of pyruvate kinase M2 isoform (PKM2) by ß-catenin-LEF/TCF4 pathway. Furthermore, the loading of LEF/TCF4 on PKM2 promoter region was significantly increased in miR372 overexpressing Hep3B, and thus, glycolytic proton efflux rate (glycoPER) was significantly increased in rLV-miR372 group compared to the rLV group. Moreover, ß-catenin knockdown abrogates this function of miR372. Ultimately, miR372 promotes the expression of erbB-2 through PKM2-pH3T11-acetylation on histone H3 lysine 9 (H3K9Ac) pathway. Of significance, both YB-1 knockdown and erbB-2 knockdown abrogate oncogenic action of miR372. Our observations suggest that miR372 promotes liver cancer cell cycle progress by activating cyclin-dependent kinase 2 (CDK2)-cyclin E-P21/Cip1 complex through miR372-YB-1-ß-catenin-LEF/TCF4-PKM2-erbB-2 axis. This study elucidates a novel mechanism for miR372 in liver cancer cells and suggests that miR372 can be used as a novel therapeutic target of liver cancer.

7.
Mol Cancer ; 17(1): 94, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895332

ABSTRACT

BACKGROUND: Long noncoding RNA HULC is highly up-regulation in human hepatocellular carcinoma (HCC). However, the functions of HULC in hepatocarcinogenesis remains unclear. METHODS: RT-PCR, Western blotting, Chromatin immunoprecipitation (CHIP) assay, RNA Immunoprecipitation (RIP) and tumorignesis test in vitro and in vivo were performed. RESULTS: HULC is negatively associated with expression of PTEN or miR15a in patients of human liver cancer. Moreover, HULC accelerates malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, HULC increasesthe expression of P62 via decreasing mature miR15a. On the other hand, excessive HULC increases the expression of LC3 on the level of transcription and then activates LC3 through Sirt1 (a deacetylase). Notably, HULC enhanced the interplay between LC3 and ATG3. Furthermore, HULC also increases the expression of becline-1(autophagy related gene). Therefore, HULC increases the cellular autophagy by increasing LC3II dependent on Sirt1.Noteworthy, excessive HULC reduces the expression of PTEN, ß-catenin and enhances the expression of SAPK/JUNK, PKM2, CDK2, NOTCH1, C-Jun in liver cancer cells. Of significance, our observations also revealed that HULC inhibited PTEN through ubiquitin-proteasome system mediated by autophagy-P62.Ultimately,HULC activates AKT-PI3K-mTOR pathway through inhibiting PTEN in human liver cancer cells. CONCLUSIONS: This study elucidates a novel mechanism that lncRNA HULC produces a vital function during hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carrier Proteins/genetics , Liver Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Autophagy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Microtubule-Associated Proteins/genetics , Neoplasm Transplantation , RNA-Binding Proteins/genetics , Signal Transduction
8.
J Cell Mol Med ; 22(6): 3246-3258, 2018 06.
Article in English | MEDLINE | ID: mdl-29602239

ABSTRACT

Toll-like receptor 4 (TLR4) which acts as a receptor for lipopolysaccharide (LPS) has been reported to be involved in carcinogenesis. However, the regulatory mechanism of it has not been elucidated. Herein, we demonstrate that TLR4 promotes the malignant growth of liver cancer stem cells. Mechanistically, TLR4 promotes the expression of histone-lysine N-methyltransferase (SUV39 h2) and increases the formation of trimethyl histone H3 lysine 9-heterochromatin protein 1-telomere repeat binding factor 2 (H3K9me3-HP1-TRF2) complex at the telomeric locus under mediation by long non coding RNA urothelial cancer-associated 1 (CUDR). At the telomeric locus, this complex promotes binding of POT1, pPOT1, Exo1, pExo1, SNM1B and pSNM1B but prevents binding of CST/AAF to telomere, thus controlling telomere and maintaining telomere length. Furthermore, TLR4 enhances interaction between HP1α and DNA methyltransferase (DNMT3b), which limits RNA polymerase II deposition on the telomeric repeat-containing RNA (TERRA) promoter region and its elongation, thus inhibiting transcription of TERRA. Ultimately, TLR4 enhances the telomerase activity by reducing the interplay between telomerase reverse transcriptase catalytic subunit (TERT) and TERRA. More importantly, our results reveal that tri-complexes of HP1 isoforms (α, ß and γ) are required for the oncogenic action of TLR4. This study elucidates a novel protection mechanism of TLR4 in liver cancer stem cells and suggests that TLR4 can be used as a novel therapeutic target for liver cancer.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Liver Neoplasms/genetics , Telomerase/genetics , Toll-Like Receptor 4/genetics , Cell Line, Tumor , Chromobox Protein Homolog 5 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Isoforms/genetics , RNA, Long Noncoding/genetics , Telomere/genetics , Telomere Homeostasis/genetics , Telomeric Repeat Binding Protein 2/genetics , Transcription Factors/genetics , DNA Methyltransferase 3B
9.
Cell Death Dis ; 9(3): 253, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449541

ABSTRACT

Maternally expressed gene 3 (MEG3) encodes an lncRNA which is suggested to function as a tumor suppressor and has been showed to involve in a variety of cancers. Herein, our findings demonstrate that MEG3 inhibits the malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, MEG3 promotes the expression and maturition of miR122 which targets PKM2. Therefore, MEG3 decreases the expression and nuclear location of PKM2 dependent on miR122. Furthermore, MEG3 also inhibits CyclinD1 and C-Myc via PKM2 in liver cancer cells. On the other hand, MEG3 promotes ß-catenin degradation through ubiquitin-proteasome system dependent on PTEN. Strikingly, MEG3 inhibits ß-catenin activity through PKM2 reduction and PTEN increase. Significantly, we also found that excessive ß-catenin abrogated the effect of MEG3 in liver cancer. In conclusion, our study for the first time demonstrates that MEG3 acts as a tumor suppressor by negatively regulating the activity of the PKM2 and ß-catenin signaling pathway in hepatocarcinogenesis and could provide potential therapeutic targets for the treatment of liver cancer.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Liver Neoplasms/enzymology , Membrane Proteins/metabolism , PTEN Phosphohydrolase/metabolism , RNA, Long Noncoding/metabolism , Thyroid Hormones/metabolism , beta Catenin/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Enzyme Stability , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Membrane Proteins/genetics , Mice, Inbred BALB C , Mice, Nude , PTEN Phosphohydrolase/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA, Long Noncoding/genetics , Signal Transduction , Thyroid Hormones/genetics , Tumor Burden , beta Catenin/genetics , Thyroid Hormone-Binding Proteins
10.
Oncotarget ; 8(30): 49093-49109, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28467776

ABSTRACT

Changes in histone lysine methylation status have been observed during cancer formation. JMJD2A protein is a demethylase that is overexpressed in several tumors. Herein, our results demonstrate that JMJD2A accelerates malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, JMJD2A promoted the expression and mature of pre-miR372 epigenetically. Notably, miR372 blocks the editing of 13th exon-introns-14th exon and forms a novel transcript( JMJD2AΔ) of JMJD2A. In particular, JMJD2A inhibited P21(WAF1/Cip1) expression by decreasing H3K9me3 dependent on JMJD2AΔ. Thereby, JMJD2A could enhance Pim1 transcription by suppressing P21(WAF1/Cip1). Furthermore, through increasing the expression of Pim1, JMJD2A could facilitate the interaction among pRB, CDK2 and CyclinE which prompts the transcription and translation of oncogenic C-myc. Strikingly, JMJD2A may trigger the demethylation of Pim1. On the other hand, Pim1 knockdown and P21(WAF1/Cip1) overexpression fully abrogated the oncogenic function of JMJD2A. Our observations suggest that JMJD2A promotes liver cancer cell cycle progress through JMJD2A-miR372-JMJD2AΔ-P21WAF1/Cip1-Pim1-pRB-CDK2-CyclinE-C-myc axis. This study elucidates a novel mechanism for JMJD2A in liver cancer cells and suggests that JMJD2A can be used as a novel therapeutic targets of liver cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MicroRNAs/genetics , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Models, Animal , Epigenesis, Genetic , Fungal Proteins/metabolism , Heterografts , Humans , Liver Neoplasms/pathology , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Promoter Regions, Genetic , Retinoblastoma Binding Proteins/metabolism , Signal Transduction , Transcriptional Activation , Ubiquitin-Protein Ligases/metabolism
11.
Sci Rep ; 6: 36843, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27833137

ABSTRACT

Inflammatory cytokines and lncRNAs are closely associated with tumorigenesis. Herein, we reveal inflammatory cytokines IL6 cooperates with long noncoding RNA CUDR to trigger the malignant transformation of human embryonic stem cells-derived hepatocyte-like stem cells. Mechanistically, IL6 cooperates with CUDR to cause MELLT3 to interact with SUV39h1 mRNA3'UTR and promote SUV39h1 expression. Moreover, the excessive SUV39h1 also increases tri-methylation of histone H3 on nineth lysine (H3K9me3). Intriguingly, under inflammatory conditions, H3K9me3 promotes the excessive expression and phosphorylation of NF-κB, and in turn, phorsphorylated NF-κB promotes the expression and phosphorylation of Stat3. Furthermore, that the phosphorylated Stat3 loads onto the promoter region of miRs and lncRNAs. Ultimately, the abnormal expression of miRs and lncRNAs increased telomerase activity, telomere length and microsatellite instability (MSI), leading to malignant transformation of hepatocyte-like stem cells.


Subject(s)
Cell Transformation, Neoplastic , Interleukin-6/physiology , Liver Neoplasms, Experimental/pathology , RNA, Long Noncoding/physiology , Animals , Cell Line , Gene Expression Regulation, Neoplastic , Hepatocytes/pathology , Histone-Lysine N-Methyltransferase/metabolism , Humans , Liver/metabolism , Liver/pathology , Methyltransferases/metabolism , Mice, Inbred BALB C , MicroRNAs/metabolism , Microsatellite Instability , NF-kappa B/metabolism , Phosphorylation , Protein Processing, Post-Translational , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Stem Cells/pathology , Telomerase/metabolism , Telomere Homeostasis
12.
Sci Rep ; 6: 36045, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27782152

ABSTRACT

The dysregulation of lncRNAs has increasingly been linked to many human diseases, especially in cancers. Our results demonstrate HULC, MALAT1 and TRF2 are highly expressed in human hepatocellular carcinoma tissues, and HULC plus MALAT1 overexpression drastically promotes the growth of liver cancer stem cells. Mechanistically, both HULC and MALAT1 overexpression enhanced RNA polII, P300, CREPT to load on the promoter region of telomere repeat-binding factor 2(TRF2), triggering the overexpression, phosphorylation and SUMOylation of TRF2. Strikingly, the excessive TRF2 interacts with HULC or MALAT1 to form the complex that loads on the telomeric region, replacing the CST/AAF and recruiting POT1, pPOT1, ExoI, SNM1B, HP1 α. Accordingly, the telomere is greatly protected and enlonged. Furthermore, the excessive HULC plus MALAT1 reduced the methylation of the TERC promoter dependent on TRF2, increasing the TERC expression that causes the increase of interplay between TRET and TERC. Ultimately, the interaction between RFC and PCNA or between CDK2 and CyclinE, the telomerase activity and the microsatellite instability (MSI) are significantly increased in the liver cancer stem cells. Our demonstrations suggest that haploinsufficiency of HULC/MALAT1 plays an important role in malignant growth of liver cancer stem cell.


Subject(s)
Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , Telomeric Repeat Binding Protein 2/biosynthesis , Cell Line, Tumor , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Telomeric Repeat Binding Protein 2/genetics
13.
Oncotarget ; 7(31): 50131-50149, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27367027

ABSTRACT

Cancer stem cells are associated with tumor recurrence. IKK is a protein kinase that is composed of IKKα, IKKß, IKKγ. Herein, we demonstrate that IKKα plus IKKß promoted and IKKγ inhibited liver cancer stem cell growth in vitro and in vivo. Mechanistically, IKKα plus IKKß enhanced and IKKγ inhibited the interplay among HP1α, HP1ß and HP1γ that competes for the interaction among HP1α, SUZ12, HEZ2. Therefore, IKKα plus IKKß inhibited and IKKγ enhanced the activity of H3K27 methyltransferase SUZ12 and EZH2, which methylates H3K27 immediately sites on HOTAIR promoter region. Therefore, IKKα plus IKKß increased and IKKγ decreased the HOTAIR expression. Strikingly, IKKα plus IKKß decreases and IKKγ increases the HP1α interplays with DNA methyltransferase DNMT3b, which increases or decreases TERRA promoter DNA methylation. Thus IKKα plus IKKß reduces and IKKγ increases to recruit TRF1 and RNA polymerase II deposition and elongation on the TERRA promoter locus, which increases or decreases TERRA expression. Furthermore, IKKα plus IKKß decreases/increases and IKKγ increases/decreases the interplay between TERT and TRRRA/between TERT and TREC. Ultimately, IKKα plus IKKß increases and IKKγ decreases the telomerase activity. On the other hand, at the telomere locus, IKKα plus IKKß increases/drcreases and IKKγ decreases/increases TRF2, POT1, pPOT1, Exo1, pExo1, SNM1B, pSNM1B/CST-AAF binding, which keep active telomere regulatory genes and poised for telomere length. Strikingly, HOTAIR is required for IKKα plus IKKß and IKKγ to control telomerase activity and telomere length. These observations suggest that HOTAIR operates the action of IKKα, IKKß, IKKγ in liver cancer stem cells. This study provides a novel basis to elucidate the oncogenic action of IKKα, IKKß, IKKγ and prompts that IKKα, IKKß, IKKγ cooperate to HOTAR to be used as a novel therapeutic targets for liver cancer.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , I-kappa B Kinase/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/metabolism , Telomere/ultrastructure , Animals , Cell Movement , Chromobox Protein Homolog 5 , DNA Methylation , Disease Progression , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Inflammation , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local , Neoplasm Transplantation , Signal Transduction
14.
Oncotarget ; 7(41): 66525-66539, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27167190

ABSTRACT

P53 is frequently mutated in human tumors as a novel gain-of-function to promote tumor development. Although dimeric (M340Q/L344R) influences on tetramerisation on site-specific post-translational modifications of p53, it is not clear how dimeric (M340Q/L344R) plays a role during hepatocarcinogenesis. Herein, we reveal that P53 (N340Q/L344R) promotes hepatocarcinogenesis through upregulation of PKM2. Mechanistically, P53 (N340Q/L344R) forms complex with CUDR and the complex binds to the promoter regions of PKM2 which enhances the expression, phosphorylation of PKM2 and its polymer formation. Thereby, the polymer PKM2 (tetramer) binds to the eleventh threonine on histone H3 that increases the phosphorylation of the eleventh threonine on histone H3 (pH3T11). Furthermore, pH3T11 blocks HDAC3 binding to H3K9Ac that prevents H3K9Ac from deacetylation and stabilizes the H3K9Ac modification. On the other hand, it also decreased tri-methylation of histone H3 on the ninth lysine (H3K9me3) and increases one methylation of histone H3 on the ninth lysine (H3K9me1). Moreover, the combination of H3K9me1 and HP1 α forms more H3K9me3-HP1α complex which binds to the promoter region of Pim1, enhancing the expression of Pim1 that enhances the expression of TERT, oncogenic lncRNA HOTAIR and reduces the TERRA expression. Ultimately, P53 (N340Q/L344R) accerlerates the growth of liver cancer cells Hep3B by activating telomerase and prolonging telomere through the cascade of P53 (N340Q/L344R)-CUDR-PKM2-pH3T11- (H3K9me1-HP1α)-Pim1- (TERT-HOTAIR-TERRA). Understanding the novel functions of P53 (N340Q/L344R) will help in the development of new liver cancer therapeutic approaches that may be useful in a broad range of cancer types.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins c-pim-1/biosynthesis , RNA, Long Noncoding/genetics , Thyroid Hormones/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Chromobox Protein Homolog 5 , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Proto-Oncogene Proteins c-pim-1/genetics , Up-Regulation , Thyroid Hormone-Binding Proteins
15.
Mol Ther ; 24(2): 261-275, 2016 02.
Article in English | MEDLINE | ID: mdl-26581161

ABSTRACT

Long noncoding RNA CUDR plays an important role during tumorigenesis. Herein, we demonstrate that SET1A cooperates with CUDR to accelerate hepatocarcinogenesis and promote malignant transformation of hepatocyte-like stem cells. Mechanistically, CUDR enhances the phosphorylation of RB1, C-myc expression, and the interplay between the SET1A and pRB1. Notably, CUDR acts as a sponge cushion that shows a link between SET1A and pRB1, producing a activated pRB1-SET1A complex. On the other hand, the pRB1-SET1A complex may carry methyls(me) to occupy the position of H3K4, resulting in specific tri-methylation of forth lysine of histone H3 (H3K4me3). Thereby, the H3K4me3 loads on the TRF2 promoter region which causes the TRF2 overexpression. Ultimately, the excessive TRF2 binds to telomere repeat DNA, prolonging the telomere length. These findings provide the first demonstration that SET1A cooperates with CUDR to play a positive potential role during hepatocarcinogenesis and hepatocyte-like stem cells' malignant transformation epigenetically.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Liver Neoplasms/genetics , RNA, Long Noncoding/genetics , Stem Cells/pathology , Animals , Cell Transformation, Neoplastic/pathology , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Mice , Phosphorylation , Promoter Regions, Genetic , Retinoblastoma Protein/metabolism , Up-Regulation
16.
Oncotarget ; 6(38): 40775-98, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26513297

ABSTRACT

Cancer up-regulated drug resistant (CUDR) is a novel non-coding RNA gene. Herein, we demonstrate excessive CUDR cooperates with excessive CyclinD1 or PTEN depletion to accelerate liver cancer stem cells growth and liver stem cell malignant transformation in vitro and in vivo. Mechanistically, we reveal the decrease of PTEN in cells may lead to increase binding capacity of CUDR to CyclinD1. Therefore, CUDR-CyclinD1 complex loads onto the long noncoding RNA H19 promoter region that may lead to reduce the DNA methylation on H19 promoter region and then to enhance the H19 expression. Strikingly, the overexpression of H19 increases the binding of TERT to TERC and reduces the interplay between TERT with TERRA, thus enhancing the cell telomerase activity and extending the telomere length. On the other hand, insulator CTCF recruits the CUDR-CyclinD1 complx to form the composite CUDR-CyclinD1-insulator CTCF complex which occupancied on the C-myc gene promoter region, increasing the outcome of oncogene C-myc. Ultimately, excessive TERT and C-myc lead to liver cancer stem cell and hepatocyte-like stem cell malignant proliferation. To understand the novel functions of long noncoding RNA CUDR will help in the development of new liver cancer therapeutic and diagnostic approaches.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , Telomerase/metabolism , Animals , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromatin Immunoprecipitation , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , In Situ Hybridization , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Oncotarget ; 6(31): 31958-84, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26376677

ABSTRACT

microRNAs (miRNAs) are short non-coding RNAs that are involved in post-transcriptional regulation of gene expression in multicellular organisms by affecting both the stability and translation of mRNAs. miR675, embedded in H19's first exon, had been linked to the development of human cancers. Herein, we demonstrate miR675 overexpression promotes and silencing miR675 attenuated liver cancer cell growth in vitro and in vivo. Mechanistically, miR675 inhibits the heterochromatin1 isoform HP1α expression in human liver cancer cells which causes a dramatically decrease of the total histone H3 lysine 9 trimethylation (H3K9me3) , histone H3 lysine 27 trimethylation (H3K27me3) and a increase of histone H3 lysine 27 acetylation(H3K27Ac).Notably, a significant reduction of the H3K9me3 and H3K27me3 and the increment of H3K27Ac occupancy on the promoter region of EGR1 triggers EGR1 transcription, translation, sumoylation and activation which upregulates lincRNA H19. Strikingly, H19 may induce and activate tumor-specific pyruvate kinase M2 (PKM2) which is essential for the Warburg effect in its dimer and for gene expression in its teramer during tumorigenesis. Our results imply that miR675 is involved in the epigenetic regulation of H3K9me3, H3k27me3 and H3K27Ac for gene expression and function during hepatocarcinogenesis (e.g.C-myc,Pim1,Ras,CyclinD1,RB1).These findings sheds light on the significance of miR675-HP1α-EGR1-H19-PKM2 cascade signaling pathway in liver cancer.


Subject(s)
Early Growth Response Protein 1/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Acetylation , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Chromobox Protein Homolog 5 , Early Growth Response Protein 1/genetics , Epigenesis, Genetic/genetics , Humans , Immunoenzyme Techniques , Immunoprecipitation , In Situ Hybridization , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcriptional Activation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Oncotarget ; 6(29): 27847-64, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26172293

ABSTRACT

Long non-coding RNA HOTAIR predicts negative tumor prognosis and exhibits oncogenic activity. Herein, we demonstrate HOTAIR promotes human liver cancer stem cell malignant growth through downregulation of SETD2. Mechanistically, HOTAIR reduces the recuritment of the CREB, P300, RNA polII onto the SETD2 promoter region that inhibits SETD2 expression and its phosphorylation. Thereby, the SETD2 binding capacity to substrate histone H3 is weakened, triggering a reduction of trimethylation on histone H3 thirty-sixth lysine, and thereby the H3K36me3-hMSH2-hMSH6-SKP2 complex is also decreased. Strikingly, the complex occupancy on chromosome is depressed, preventing from mismatch DNA repair. While reducing the degradation capacity of Skp2 for aging histone H3 bound to damaged DNA, the aging histone repair is impaired. Furthermore, that the damaged DNA escaped to repair can causes microsatellite instability(MSI) and abnormal expression of cell cycle related genes that may trigger the hepatocarcinogenesis. This study provides evidence for HOTAIR to promote tumorigenesis via downregulating SETD2 in liver cancer stem cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/biosynthesis , Liver Neoplasms/genetics , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/genetics , Animals , Blotting, Western , Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , Cell Separation , Chromatin Immunoprecipitation , Down-Regulation , Female , Heterografts , Histone-Lysine N-Methyltransferase/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...