Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Ther ; 28(2): 572-586, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31732298

RESUMO

MicroRNA24-2 (miR24-2) is associated with human tumorigenesis; however, its molecular mechanisms are poorly understood. Herein, our findings demonstrate that miR24-2 promotes the proliferation ability in vitro and the tumorigenic ability in vivo in human liver cancer stem cells (hLCSCs). Mechanically, the miR24-2 targets for 3' UTR (2,627-2,648) of protein arginine methyltransferase 7 (PRMT7) inhibit the translational ability of prmt7 gene. Moreover, miR24-2 inhibits the di-/tri-methylation of histone H4 arginine 3 by reducing PRMT7 and then promotes the expression of Nanog via long noncoding RNA HULC. Notably, miR24-2 inhibits histone deacetylase HDAC3 through miR675, which promotes the acetylation of histone H4 at lysine 16. Subsequently, miR24-2 enhances the interaction between LC3 and ATG4 dependent on PI3K and triggers cellular autophagy. Strikingly, miR24-2 inhibits the degradation of pyruvate kinase M1 via autophagosome-P62 in hLCSCs. Furthermore, miR24-2 enhances the activity of Src by promoting the binding of PKM1 to the Src promoter regions in hLCSCs. In particular, our results also indicate that src gene determines the oncogenic functions of miR24-2. These results provided a valuable theoretical basis for the discovery of liver cancer therapeutic targets and diagnosis markers based on miR24-2.


Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Quinases da Família src/genética , Acetilação , Autofagia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Proteínas de Membrana/metabolismo , Metilação , Proteína Homeobox Nanog/genética , Proteína-Arginina N-Metiltransferases/genética , Interferência de RNA , Hormônios Tireóideos/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
2.
J Cell Mol Med ; 24(5): 2772-2790, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32030886

RESUMO

Several microRNAs are associated with carcinogenesis and tumour progression. Herein, our observations suggest both miR24-2 and Pim1 are up-regulated in human liver cancers, and miR24-2 accelerates growth of liver cancer cells in vitro and in vivo. Mechanistically, miR24-2 increases the expression of N6-adenosine-methyltransferase METTL3 and thereafter promotes the expression of miR6079 via RNA methylation modification. Furthermore, miR6079 targets JMJD2A and then increased the tri-methylation of histone H3 on the ninth lysine (H3K9me3). Therefore, miR24-2 inhibits JMJD2A by increasing miR6079 and then increases H3K9me3. Strikingly, miR24-2 increases the expression of Pim1 dependent on H3K9me3 and METTL3. Notably, our findings suggest that miR24-2 alters several related genes (pHistone H3, SUZ12, SUV39H1, Nanog, MEKK4, pTyr) and accelerates progression of liver cancer cells through Pim1 activation. In particular, Pim1 is required for the oncogenic action of miR24-2 in liver cancer. This study elucidates a novel mechanism for miR24-2 in liver cancer and suggests that miR24-2 may be used as novel therapeutic targets of liver cancer.


Assuntos
Progressão da Doença , Histonas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Lisina/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Masculino , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Modelos Biológicos , Oncogenes , Proteínas Proto-Oncogênicas c-pim-1/genética
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(6): 808-814, 2019 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-31880110

RESUMO

OBJECTIVE: To evaluate the correlation of Fast-track extubation ultrasound score (FTEUS) and clinical multi-organ information indicators in post-cardiac surgery patients. METHODS: prospectively recruit post-cardiac surgery patients who were about to extubating from Febuary 2019 to September 2019. A fast-track extubation ultrasound score protocol (FTE-USP) was developed on the basis of the conventional fast-track extubation standard precisely and individualized. Cardiac, pulmonary and inferior vena cava ultrasound examinations were performed by specially trained observers, video data were saved, FTE-USP was used for scoring, Kendall consistency coefficient was used to meature the interobserver consistency. The correlation between the FTEUS and the patients' clinical indicators was evaluated. RESULTS: A total of 207 patients were recruited in the study, including 89 males and 118 females, aged (54.63±11.80) years. The FTEUS was performed at bedside with a mean time of (8.23±2.08) min, Kendall consistency coefficient is 0.941. With the increase of the total score of FTEUS, the incidence of clinical adverse events increased (especially the arrhythmia), and there were significant changes in liver, kidney, heart, lung and other organ function indicators, among which serum creatinine level, serum cystatin C level, serum NT-pro-brain natriuretic peptide, length of stay in intensive care unit, non-invasive mechanical ventilation time after extubation, and incidence of arrhythmia were positively correlated with FTEUS (P < 0.05).With FTEUS increased to 5 points, the incidence of arrhythmia (14/24, 58.3%), cardiopulmonary resuscitation (2/24, 8.3%) and weaning failure (2/24, 8.3%) increased. CONCLUSION: FTE-USP integrates multi-organ informations, can be performed quickly at the bedside and alerts adverse events. It has the potential to be applied to assist clinical decision-making in post-cardiac surgery patients before extubation.


Assuntos
Extubação , Procedimentos Cirúrgicos Cardíacos , Adulto , Idoso , Feminino , Humanos , Intubação Intratraqueal , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Fatores de Tempo
5.
Mol Cancer ; 17(1): 94, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29895332

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/patologia , Proteínas de Transporte/genética , Neoplasias Hepáticas/patologia , MicroRNAs/genética , RNA Longo não Codificante/genética , Animais , Autofagia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Transplante de Neoplasias , Proteínas de Ligação a RNA/genética , Transdução de Sinais
6.
Mol Ther ; 24(2): 261-275, 2016 02.
Artigo em Inglês | MEDLINE | ID: mdl-26581161

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/genética , Epigênese Genética , Histona-Lisina N-Metiltransferase/genética , Neoplasias Hepáticas/genética , RNA Longo não Codificante/genética , Células-Tronco/patologia , Animais , Transformação Celular Neoplásica/patologia , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Proteína do Retinoblastoma/metabolismo , Regulação para Cima
7.
Mol Ther ; 23(12): 1843-53, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26347501

RESUMO

Long noncoding RNA cancer upregulated drug resistant (CUDR) is overexpressed in many tumors and promotes tumorigenesis. Herein, we demonstrate CUDR could enhance the human embryonic stem cells (ESC) differentiation into hepatocyte-like cells by reducing trimethylation on histone H3 twenty-seventh lysine (H3K27me3). On the other hand, excessive CUDR triggers hepatocyte-like cells malignant transformation. Mechanistically, we identify CUDR causes highly upregulated in liver cancer (HULC) and ß-catenin abnormal expression by inhibiting HULC promoter methylation and promoting ß-catenin promoter-enhancer chromatin looping formation mediated by CUDR-ccctc-binding factor (CTCF) complex, which recruits more RNA polII and P300. Strikingly, HULC and ß-catenin activity are crucial for CUDR oncogenic function. These findings provide the first demonstration that CUDR plays a positive potential role in liver cancer stem cell through the cascade of CUDR-HULC/CUDR-ß-catenin signaling, and offer insights into a novel link between long noncoding RNA (lncRNA) and the epigenetic modification in cancer stem cells.


Assuntos
Diferenciação Celular , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , RNA Longo não Codificante/genética , beta Catenina/metabolismo , Fator de Ligação a CCCTC , Linhagem Celular Tumoral , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Neoplasias Hepáticas/terapia , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Regulação para Cima , beta Catenina/genética
8.
Biochem Mol Biol Educ ; 52(3): 291-298, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38189805

RESUMO

The laboratory practice "Primary culture and directional differentiation of rat bone marrow mesenchymal stem cells (BMSCs)" is part of a required course for sophomore medical students at Tongji university, which has been conducted since 2012. Blended learning has been widely applied in medical courses. Based on a student-centered teaching philosophy, we reconstructed a comprehensive stem cell laboratory module with blended learning in 2021, aiming to facilitate students in enhancing their understanding of the multi-lineage differentiation potential of stem cells and improve their experimental skills, self-directed learning ability, and innovative thinking. First, we constructed in-depth online study resources, including videos demonstrating laboratory procedures, a PowerPoint slide deck, and published literature on student self-learning before class. In class, students performed a primary culture of BMSCs, freely chose among adipogenic, osteogenic, or chondrogenic differentiation, and used cytochemical or immunofluorescence staining for identification. After class, the extracurricular part involved performing quantitative polymerase chain reaction to examine the expression of multi-lineage differentiation marker genes, which was designed as an elective. After 2 years of practice, positive feedback was obtained from both students and faculty members who achieved, the learning goal as expected. The reconstructed stem cell laboratory module provides comprehensive practice opportunities for students. Students have a better understanding of BMSC at the molecular, cellular, and functional levels and have improved their experimental skills, which forms a basis for scientific research for medical students. Introducing blended learning into other medical laboratory practices thus seems valuable.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais , Estudantes de Medicina , Humanos , Ratos , Animais , Células-Tronco Mesenquimais/citologia , Universidades , Aprendizagem , Laboratórios , Educação de Graduação em Medicina/métodos
10.
Mol Ther Nucleic Acids ; 23: 310-323, 2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33425489

RESUMO

Circular RNA (CircRNA) is a newly identified special class of non-coding RNA (ncRNA) that plays an important regulatory role in the progression of certain diseases. Herein, our results indicate that CircMEG3 is downregulated expression and negatively correlated with the expression of telomerase-related gene Cbf5 in human liver cancer. Moreover, CircMEG3 inhibits the growth of human liver cancer stem cells in vivo and in vitro. CircMEG3 inhibits the expression of m6A methyltransferase METTL3 dependent on HULC. Moreover, CircMEG3 inhibits the expression of Cbf5, a component of telomere synthetase H/ACA ribonucleoprotein (RNP; catalyst RNA pseudouracil modification) through METTL3 dependent on HULC. Thereby, CircMEG3 inhibits telomerase activity and shortens telomere lifespan dependent on HULC and Cbf5 in human liver cancer stem cell. Strikingly, increased Cbf5 abrogates the ability of CircMEG3 to inhibit malignant differentiation of human liver cancer stem cells. In summary, these observations provide important basic information for finding effective liver cancer therapeutic targets.

11.
Mol Ther Oncolytics ; 17: 471-483, 2020 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-32490171

RESUMO

miR-155 is associated with the promotion of tumorigenesis. Herein, we indicate that abnormal miR-155 was negatively correlated with the expression of P21WAF1/Cip1. Our results suggest that miR-155 alters the transcriptome and inhibits the expression of H3F3A in liver cancer cells. Therefore, miR-155 inhibits the methylation modification of histone H3 on the 27th lysine. Notably, on the one hand, miR-155-dependent CTCF loops cause the CDK2 interacting with cyclin E in liver cancer cells; on the other hand, miR-155 promotes the phosphorylation modification of CDK2 by inhibiting H3F3A. Subsequently, miR-155 competitively blocks the binding of RNA polymerase II (RNA Pol II) to the P21WAF1/CIP1 promoter by increasing the phosphorylation of CDK2, inhibiting the transcription and translation of P21WAF1/CIP1. Strikingly, excessive P21WAF1/CIP1 abolishes the cancerous function of miR-155. In conclusion, miR-155 can play a positive role in the development of liver cancer and influence a series of gene expression through epigenetic regulation.

12.
Stem Cell Res Ther ; 11(1): 8, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31900225

RESUMO

BACKGROUND: The functions of HULC have been demonstrated in several cancers. However, its mechanism has not been elucidated in human liver cancer stem cells. METHODS: Liver cancer stem cells were isolated from Huh7 cells; gene infection and tumorigenesis test in vitro and in vivo were performed. RESULTS: We demonstrate that HULC promotes growth of liver cancer stem cells in vitro and in vivo. Mechanistically, HULC enhances the expression of Sirt1 dependent on miR675 and then induces the cellular autophagy through Sirt1. HULC enhances CyclinD1 and thereby increases pRB and inhibited P21 WAF1/CIP 1 via autophagy-miR675-PKM2 pathway in human liver cancer stem cells. Ultimately, our results demonstrate that CyclinD1 is required for the oncogenic functions of HULC in liver cancer stem cells. CONCLUSIONS: It reveals the key molecular signaling pathways for HULC and provides important basic information for finding effective tumor therapeutic targets based on HULC.


Assuntos
Proteínas de Transporte/metabolismo , Ciclina D1/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/patologia , RNA Longo não Codificante/metabolismo , Hormônios Tireóideos/metabolismo , Animais , Autofagia/fisiologia , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Transfecção , Regulação para Cima , Proteínas de Ligação a Hormônio da Tireoide
13.
Mol Ther Nucleic Acids ; 14: 171-183, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30594073

RESUMO

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.

14.
Cell Death Dis ; 9(3): 253, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29449541

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Proliferação de Células , Neoplasias Hepáticas/enzimologia , Proteínas de Membrana/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , RNA Longo não Codificante/metabolismo , Hormônios Tireóideos/metabolismo , beta Catenina/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Estabilidade Enzimática , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , PTEN Fosfo-Hidrolase/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , RNA Longo não Codificante/genética , Transdução de Sinais , Hormônios Tireóideos/genética , Carga Tumoral , beta Catenina/genética , Proteínas de Ligação a Hormônio da Tireoide
15.
Mol Ther Nucleic Acids ; 11: 367-381, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29858072

RESUMO

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.

16.
Mol Ther Nucleic Acids ; 11: 494-507, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29858084

RESUMO

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.

17.
Oncotarget ; 8(30): 49093-49109, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-28467776

RESUMO

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.


Assuntos
Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroRNAs/genética , Animais , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Modelos Animais de Doenças , Epigênese Genética , Proteínas Fúngicas/metabolismo , Xenoenxertos , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Regiões Promotoras Genéticas , Proteínas de Ligação a Retinoblastoma/metabolismo , Transdução de Sinais , Ativação Transcricional , Ubiquitina-Proteína Ligases/metabolismo
18.
Oncotarget ; 7(31): 50131-50149, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27367027

RESUMO

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.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Quinase I-kappa B/metabolismo , Neoplasias Hepáticas/metabolismo , Células-Tronco Neoplásicas/metabolismo , RNA Longo não Codificante/metabolismo , Telômero/ultraestrutura , Animais , Movimento Celular , Homólogo 5 da Proteína Cromobox , Metilação de DNA , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Recidiva Local de Neoplasia , Transplante de Neoplasias , Transdução de Sinais
19.
Sci Rep ; 6: 36843, 2016 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-27833137

RESUMO

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.


Assuntos
Transformação Celular Neoplásica , Interleucina-6/fisiologia , Neoplasias Hepáticas Experimentais/patologia , RNA Longo não Codificante/fisiologia , Animais , Linhagem Celular , Regulação Neoplásica da Expressão Gênica , Hepatócitos/patologia , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Metiltransferases/metabolismo , Camundongos Endogâmicos BALB C , MicroRNAs/metabolismo , Instabilidade de Microssatélites , NF-kappa B/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Células-Tronco/patologia , Telomerase/metabolismo , Homeostase do Telômero
20.
Oncotarget ; 6(38): 40775-98, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26513297

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/patologia , Transformação Celular Neoplásica/patologia , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Longo não Codificante/genética , Telomerase/metabolismo , Animais , Apoptose , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Imunoprecipitação da Cromatina , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoprecipitação , Hibridização In Situ , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telomerase/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA