• 제목/요약/키워드: histone deacetylase 8

검색결과 35건 처리시간 0.019초

Effects of Trichostatin A and 5-aza-2'deoxycytidine on Nuclear Reprogramming in Pig Cloned Embryos

  • Lee, Sung Hyun;Xu, Yong-Nan;Heo, Young-Tae;Cui, Xiang-Shun;Kim, Nam-Hyung
    • Reproductive and Developmental Biology
    • /
    • 제37권4호
    • /
    • pp.269-279
    • /
    • 2013
  • Low efficiency of somatic cell nuclear transfer (SCNT) is attributed to incomplete reprogramming of transfered nuclei into oocytes. Trichostatin A (TSA), histone deacetylase inhibitor and 5-aza-2'deoxycytidine (5-aza-dC), DNA methylation inhibitor has been used to enhance nuclear reprogramming following SCNT. However, it was not known molecular mechanism by which TSA and 5-aza-dC improve preimplantation embryo and fetal development following SCNT. The present study investigates embryo viability and gene expression of cloned porcine preimplantation embryos in the presence and absence of TSA and 5-aza-dC as compared to embryos produced by parthenogenetic activation. Our results indicated that TSA treatment significantly improved development. However 5-aza-dC did not improve development. Presence of TSA and 5-aza-dC significantly improved total cell number, and also decreased the apoptotic and autophagic index. Three apoptotic-related genes, Bak, Bcl-xL, and Caspase 3 (Casp3), and three autophagic-related genes, ATG6, ATG8, and lysosomal-associated membrane protein 2 (LAMP2), were measured by real time RT-PCR. TSA and 5-aza-dC treatment resulted in high expression of anti-apoptotic gene Bcl-xL and low pro-apoptotic gene Bak expression compared to untreated NT embryos or parthenotes. Furthermore, LC3 protein expression was lower in NT-TSA and NT-5-aza-dC embryos than those of NT and parthenotes. In addition, TSA and 5-aza-dC treated embryos displayed a global acetylated histone H3 at lysine 9 and methylated DNA H3 at lysine 9 profile similar to the parthenogenetic blastocysts. Finally, we determined that several DNA methyltransferase genes Dnmt1, Dnmt3a and Dnmt3b. NT blastocysts showed higher levels Dnmt1 than those of the TSA and 5-aza-dC blastocysts. Dnmt3a is lower in 5-aza-dC than NT, NTTSA and parthenotes. However, Dnmt3b is higher in 5-aza-dC than NT and NTTSA. These results suggest that TSA and 5-aza-dC positively regulates nuclear reprogramming which result in modulation of apoptosis and autophagy related gene expression and then reduce apoptosis and autophagy. In addition, TSA and 5-aza-dC affects the acetylated and methylated status of the H3K9.

비소세포성 폐암에서 인슐린 양 성장 인자 결합 단백질-3의 발현 조절 기전 (Regulatory Mechanism of Insulin-Like Growth Factor Binding Protein-3 in Non-Small Cell Lung Cancer)

  • 장윤수;이호영;김영삼;김형중;장준;안철민;김성규;김세규
    • Tuberculosis and Respiratory Diseases
    • /
    • 제56권5호
    • /
    • pp.465-484
    • /
    • 2004
  • 배 경 : 인슐린 양 성장 인자(IGF) 결합 단백질-3(IGFBP-3)은 IGF와 결합하여 IGF의 세포 분열 촉진 및 항세포 고사 기전을 억제할 뿐 아니라 IGF와는 독립적으로 세포고사를 유도함으로써 비소세포성 폐암 세포주의 성장을 억제한다. 방 법 : 본 연구에서 저자들은 IGFBP-3 promoter의 hyper-methylation이 IGFBP-3 단백 발현에 어떠한 역할을 하는가를 연구하였다. 또한 비소세포성 폐암 세포주에서 methylation된 IGFBP-3 promoter에서 유전자 발현을 억제하는 기전을 연구하였다. 결 과 : 본 연구에 사용된 15 종의 비소세포성 폐암 세포주 중 7종 (46.7%)에서 IGFBP-3 promoter의 methylation 이 관찰되었으며, 23명의 I기 환자 검체 중 16 (69.7%), 9명의 II기 환자 검체중 7 (77.8%), 5명의 IIIA 환자 검체중 4 (80%), 6명의 IIIB 환자 검체중 4 (66.7 %), 그리고 6 명의 IV기 환자검체중 6명 모두에서 (100%) promoter 의 methylation 이 관찰되었다. 이 비소세포성 폐암 세포주에서 promoter methylation 상태는 IGFBP-3 단백 및 mRNA 발현양상과 잘 일치하였으며, IGFBP-3의 발현이 억제되었던 비소세포성 폐암 세포주들 중 일부의 세포에서 demethylating 약제인 5'-aza-2'-deoxycytidine (5'-aza-dC) 처리 후 그 발현이 회복되었다. IGFBP-3 promoter 활성도에 중요한 역할을 하는 Sp-1/Sp-3 결합 요소는 IGFBP-3 단백 발현이 억제된 비소세포성 폐암 세포주에서 methylation되어 있었으며, 이 요소의 methylation 은 Sp-1 전사 인자의 결합을 억제하였다. ChIP assay 결과에서 IGFBP-3 promoter의 methylation 상태는 Sp-1/Sp-3 결합 요소에 Sp-1, methyl-CpG binding protein-2 (MeCP2), 그리고 histone deacetylase (HDAC)의 결합에 영향을 주며, 이는 5'-aza-dC 처리에 의하여 역전 되었다. Sp-1/Sp-3 결합 요소를 포함하고 있는 IGFBP-3 promoter의 in vitro methylation은 promoter activity를 현저히 감소시켰으며 이는 MeCP2 단백을 동시에 발현 시켰을 때 더욱 억제되며 5'-aza-dC 처리시 회복되었다. 결 론 : 이러한 결과들은 IGFBP-3 promoter의 methylation이 IGFBP-3 발현을 억제하는 하나의 기전이며, HDAC의 모집을 유도함으로서 MeCP2가 IGFBP-3 발현 억제에 중요한 역할을 함을 보이는 것이다. 이런 현상은 비소세포성 폐암에서 진단 당시의 진행된 병기와도 관계가 있어 IGFBP-3 promoter의 methylation 상태가 비소세포성 폐암의 발암 기전 및 진행에 중요한 역할을 하고 있음을 보이고 있으며, 나아가 조기 진단 및 암 예방영역에서 하나의 생물학적 지표로도 사용될 수 있을 것으로 생각된다.

HDAC 저해제에 의한 인체 백혈병 U937 세포의 apoptosis 유발에 미치는 Bcl-2의 영향 (Effects of Bcl-2 Overexpressing on the Apoptotic Cell Death Induced by HDAC Inhibitors in Human Leukemic U937 Cells)

  • 이준혁;허만규;박동일;최병태;최영현
    • 생명과학회지
    • /
    • 제17권4호
    • /
    • pp.552-560
    • /
    • 2007
  • 본 연구에서는 인체백혈병세포 U937에서 HDAC 저해제에 의한 증식억제, 세포주기 교란 및 apoptosis 유도에 미치는 Bcl-2 유전자의 영향에 관하여 조사하였다. 이를 위하여 U937/vector 및 U937/Bcl-2 세포주를 대상으로 대표적인 HDAC 저해제인 TSA 및 Na-B 처리에 의한 세포 증식 및 생존율에 미치는 영향을 조사한 결과, TSA에 의한 U937 세포의 증식억제 및 생존율의 감소는 Bcl-2의 과발현에 의하여 차단되는 효과를 보였으나, Na-B는 U937/vector 및 U937/Bcl-2세포사이에 큰 변화를 보이지는 않았다. 세포주기 교란효과에서 Na-B는 TSA에 비하여 유의적인 차이를 보이지 못하였으며, 이는 TSA에 의한 apoptosis가 U937/Bcl-2 세포에서는 억제되었으나, Na-B에 의한 apoptosis는 Bcl-2의 과발현에 의하여 차단되지 못한 것과 연관성이 있는 결과였다. 또한 TSA에 의한 apoptosis 유발의 Bcl-2에 의한 차단 효과는 TSA에 의하여 활성화된 caspase의 활성 억제, Bcl-2 발현 자체의 완화 등 apoptosis 조절 인자들의 발현 및 활성 변화에 기인 된 것임을 알 수 있었다.

MHY2251, a New SIRT1 Inhibitor, Induces Apoptosis via JNK/p53 Pathway in HCT116 Human Colorectal Cancer Cells

  • Yong Jung Kang;Young Hoon Kwon;Jung Yoon Jang;Jun Ho Lee;Sanggwon Lee;Yujin Park;Hyung Ryong Moon;Hae Young Chung;Nam Deuk Kim
    • Biomolecules & Therapeutics
    • /
    • 제31권1호
    • /
    • pp.73-81
    • /
    • 2023
  • Sirtuins (SIRTs) belong to the nicotinamide adenine dinucleotide (NAD+)-dependent class III histone deacetylase family. They are key regulators of cellular and physiological processes, such as cell survival, senescence, differentiation, DNA damage and stress response, cellular metabolism, and aging. SIRTs also influence carcinogenesis, making them potential targets for anticancer therapeutic strategies. In this study, we investigated the anticancer properties and underlying molecular mechanisms of a novel SIRT1 inhibitor, MHY2251, in human colorectal cancer (CRC) cells. MHY2251 reduced the viability of various human CRC cell lines, especially those with wild-type TP53. MHY2251 inhibited SIRT1 activity and SIRT1/2 protein expression, while promoting p53 acetylation, which is a target of SIRT1 in HCT116 cells. MHY2251 treatment triggered apoptosis in HCT116 cells. It increased the percentage of late apoptotic cells and the sub-G1 fraction (as detected by flow cytometric analysis) and induced DNA fragmentation. In addition, MHY2251 upregulated the expression of FasL and Fas, altered the ratio of Bax/Bcl-2, downregulated the levels of pro-caspase-8, -9, and -3 proteins, and induced subsequent poly(ADP-ribose) polymerase cleavage. The induction of apoptosis by MHY2251 was related to the activation of the caspase cascade, which was significantly attenuated by pre-treatment with Z-VAD-FMK, a pan-caspase inhibitor. Furthermore, MHY2251 stimulated the phosphorylation of c-Jun N-terminal kinase (JNK), and MHY2251-triggered apoptosis was blocked by pre-treatment with SP600125, a JNK inhibitor. This finding indicated the specific involvement of JNK in MHY2251-induced apoptosis. MHY2251 shows considerable potential as a therapeutic agent for targeting human CRC via the inhibition of SIRT1 and activation of JNK/p53 pathway.

Effect of Trichostatin A on Anti HepG2 Liver Carcinoma Cells: Inhibition of HDAC Activity and Activation of Wnt/β-Catenin Signaling

  • Shi, Qing-Qiang;Zuo, Guo-Wei;Feng, Zi-Qiang;Zhao, Lv-Cui;Luo, Lian;You, Zhi-Mei;Li, Dang-Yang;Xia, Jing;Li, Jing;Chen, Di-Long
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제15권18호
    • /
    • pp.7849-7855
    • /
    • 2014
  • Purpose: To investigate the effect of deacetylase inhibitory trichostatin A (TSA) on anti HepG2 liver carcinoma cells and explore the underlying mechanisms. Materials and Methods: HepG2 cells exposed to different concentrations of TSA for 24, 48, or 72h were examined for cell growth inhibition using CCK8, changes in cell cycle distribution with flow cytometry, cell apoptosis with annexin V-FTIC/PI double staining, and cell morphology changes under an inverted microscope. Expression of ${\beta}$-catenin, HDAC1, HDAC3, H3K9, CyclinD1 and Bax proteins was tested by Western blotting. Gene expression for ${\beta}$-catenin, HDAC1and HDAC3 was tested by q-PCR. ${\beta}$-catenin and H3K9 proteins were also tested by immunofluorescence. Activity of Renilla luciferase (pTCF/LEF-luc) was assessed using the Luciferase Reporter Assay system reagent. The activity of total HDACs was detected with a HDACs colorimetric kit. Results: Exposure to TSA caused significant dose-and time-dependent inhibition of HepG2 cell proliferation (p<0.05) and resulted in increased cell percentages in G0/G1 and G2/M phases and decrease in the S phase. The apoptotic index in the control group was $6.22{\pm}0.25%$, which increased to $7.17{\pm}0.20%$ and $18.1{\pm}0.42%$ in the treatment group. Exposure to 250 and 500nmol/L TSA also caused cell morphology changes with numerous floating cells. Expression of ${\beta}$-catenin, H3K9and Bax proteins was significantly increased, expression levels of CyclinD1, HDAC1, HDAC3 were decreased. Expression of ${\beta}$-catenin at the genetic level was significantly increased, with no significant difference in HDAC1and HDAC3 genes. In the cytoplasm, expression of ${\beta}$-catenin fluorescence protein was not obvious changed and in the nucleus, small amounts of green fluorescence were observed. H3K9 fluorescence protein were increased. Expression levels of the transcription factor TCF werealso increased in HepG2 cells following induction by TSA, whikle the activity of total HDACs was decreased. Conclusions: TSA inhibits HDAC activity, promotes histone acetylation, and activates Wnt/${\beta}$-catenin signaling to inhibit proliferation of HepG2 cell, arrest cell cycling and induce apoptosis.