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










Publication year range
1.
Int J Oncol ; 63(6)2023 Dec.
Article in English | MEDLINE | ID: mdl-37888731

ABSTRACT

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that, for the scratch wound assay experiments shown in Fig. 1 on p. 2413, the panels showing the '0 h' experiments for the respective incubations with VEGF or BC001 were apparently identical. The authors were able to re­examine their original data files, and realized that this figure had been inadverently assembled incorrectly. The revised version of Fig. 1, containing the correct data for the '0 h / BC001' panel, is shown below. Note that the revisions made to this figure do not affect the overall conclusions reported in the paper. The authors are grateful to the Editor of International Journal of Oncology for allowing them the opportunity to publish this Corrigendum, and apologize to the readership for any inconvenience caused. [International Journal of Oncology 45: 2411­2420, 2014; DOI: 10.3892/ijo.2014.2690].

2.
FASEB J ; 34(6): 8416-8427, 2020 06.
Article in English | MEDLINE | ID: mdl-32350948

ABSTRACT

During human erythroid maturation, Hsp70 translocates into the nucleus and protects GATA-1 from caspase-3 cleavage. Failure of Hsp70 to localize to the nucleus was found in Myelodysplastic syndrome (MDS) erythroblasts and can induce dyserythropoiesis, with arrest of maturation and death of erythroblasts. However, the mechanism of the nuclear trafficking of Hsp70 in erythroblasts remains unknown. Here, we found the hematopoietic transcriptional regulator, EDAG, to be a novel binding partner of Hsp70 that forms a protein complex with Hsp70 and GATA-1 during human normal erythroid differentiation. EDAG overexpression blocked the cytoplasmic translocation of Hsp70 induced by EPO deprivation, inhibited GATA-1 degradation, thereby promoting erythroid maturation in an Hsp70-dependent manner. Furthermore, in myelodysplastic syndrome (MDS) patients with dyserythropoiesis, EDAG is dramatically down-regulated, and forced expression of EDAG has been found to restore the localization of Hsp70 in the nucleus and elevate the protein level of GATA-1 to a significant extent. In addition, EDAG rescued the dyserythropoiesis of MDS patients by increasing erythroid differentiation and decreasing cell apoptosis. This study demonstrates the molecular mechanism of Hsp70 nuclear sustaining during erythroid maturation and establishes that EDAG might be a suitable therapeutic target for dyserythropoiesis in MDS patients.


Subject(s)
Cell Nucleus/metabolism , Erythroblasts/metabolism , Erythropoiesis/physiology , HSP70 Heat-Shock Proteins/metabolism , Myelodysplastic Syndromes/metabolism , Nuclear Proteins/metabolism , Apoptosis/physiology , Caspase 3/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cytoplasm/metabolism , Gene Expression Regulation/physiology , Hematologic Diseases/metabolism , Humans
3.
Oncol Rep ; 39(6): 2759-2768, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29620221

ABSTRACT

A novel small molecular compound, 4­ethyl­8­fluoro­hydroxy­9­methoxy­11­methyl­1,12­dihydro­4H­2­oxa­6,12a­diaza­dibenzo[b,h]fluorene­3,13­dione (CT­1042) exhibits potent antitumor activity against many tumor cells in vitro. However, the effects and underlying mechanisms of CT­1042 in non­small cell lung cancer (NSCLC) remain unclear. The present study was designed to determine the anticancer properties and underlying molecular mechanisms of CT­1042 in NCI­H460 NSCLC cells. A thiazolyl blue tetrazolium bromide assay (MTT) was performed to evaluate cell viability and flow cytometry was used to analyze apoptosis, mitochondrial membrane potential (MMP) and cell cycle. Real­time quantitative PCR and western blotting were conducted to determine relative mRNA and protein levels. A tumor xenograft experiment was performed to investigate the effects of CT­1042 on tumor growth in vivo. CT­1042 markedly inhibited the proliferation of twelve cancer cell lines, decreased MMP in subject cells and increased caspase­3 activity. Cell cycle analysis indicated that CT­1042 delayed the cell cycle progression during the G2/M phase in a dose­dependent manner. In addition, CT­1042 induced mitochondrial­mediated apoptosis by activating p53 and Bax, as well as inhibiting Bcl­2 and survivin. Finally, CT­1042 significantly suppressed NCI­H460 xenograft tumor growth in vivo, with low systemic toxicity. Collectively, these results revealed that CT­1042 has significant lung anticancer properties.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Fluorenes/administration & dosage , Inhibitor of Apoptosis Proteins/genetics , Lung Neoplasms/drug therapy , Repressor Proteins/genetics , Small Molecule Libraries/administration & dosage , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fluorenes/chemistry , Fluorenes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Repressor Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Survivin , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
Oncol Lett ; 13(6): 4762-4768, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28599477

ABSTRACT

5-Fluorouracil (5-FU) is one of the most important agents used to treat colorectal cancer. However, the therapeutic effect of 5-FU on colon cancer is limited. SM-1 is a novel type of proapoptotic agent that directly activates procaspase-3 to caspase-3, leading to apoptosis in human cancer cells. The aim of the present study was to evaluate the antitumor effects of 5-FU in combination with SM-1. The human colorectal cancer cell lines HCT116 and LoVo were cultured in the presence of SM-1 and 5-FU. The combination of SM-1 and 5-FU treatment exhibited increased proliferation inhibitory effects compared with 5-FU treatment alone in HCT116 and LoVo cells, as determined using an MTT assay. SM-1 significantly decreased the half-maximal inhibitory concentration of 5-FU from 8.07±0.49 to 2.55±0.41 µmol/l in HCT116 cells, and from 7.90±0.98 to 3.14±0.81 µmol/l in LoVo cells. Similarly, the apoptotic activity was increased to 47.95 and 35.19% in HCT116 and LoVo cells, respectively, as determined using Annexin V/propidium iodide staining and flow cytometry. The combination of SM-1 and 5-FU treatment led to significantly increased caspase-3 activity compared with either compound alone. The reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis revealed the downregulation of B-cell lymphoma 2 and Survivin, and the upregulation of apoptosis regulator Bcl-2-associated X protein and cleaved poly (ADP-ribose) polymerase in HCT116 and LoVo cells. In addition, RT-qPCR identified downregulation of X-linked inhibitor of apoptosis protein mRNA. 5-FU and SM-1 treatment in combination increased tumor proliferation inhibition in HCT116 and LoVo xenograft mouse models of colorectal cancer, compared with SM-1 or 5-FU treatment alone. SM-1 significantly enhanced the antitumor activity of 5-FU in colorectal cancer. These improved effects were due to increased activity of the apoptotic signaling pathway.

5.
PLoS One ; 10(2): e0116202, 2015.
Article in English | MEDLINE | ID: mdl-25658946

ABSTRACT

BACKGROUND: There is no highly effective chemotherapy for malignant gliomas to date. We found that dimethylaminomicheliolide (DMAMCL), a selective inhibitor of acute myeloid leukemia (AML) stem/progenitor cells, inhibited the growth of glioma cells. METHODS: The distribution of DMAMCL in brain was analyzed by an ultraperformance liquid chromatography-mass spectrometry (UPLC-MS/MS) system. The anti-tumor evaluations of DMAMCL in vitro were performed by MTT, FACS and RT-PCR. In vivo, the mixture of C6 cells and matrigel was injected into caudatum, and the anti-tumor activity of DMAMCL was evaluated by tumor growth and rat survival. The toxicity of DMAMCL was evaluated by body weight, daily food intake, hematological or serum biochemical analyses, and histological appearance of tissues. RESULTS: The IC50 values of DMAMCL against the C6 and U-87MG cell lines in vitro were 27.18 ± 1.89 µM and 20.58 ± 1.61 µM, respectively. DAMMCL down-regulated the anti-apoptosis gene Bcl-2 and increased apoptosis in C6 and U-87MG cells in a dose-dependent manner. In a C6 rat tumor model, daily administration of DMAMCL for 21 days reduced the burden of C6 tumors by 60% to 88% compared to controls, and more than doubled the mean lifespan of tumor-bearing rats. Distribution analysis showed that the DMAMCL concentration was higher in the brain than in plasma. Evaluations for toxicity revealed that oral administration of DMAMCL at 200 or 300 mg/kg once a day for 21 days did not result in toxicity. CONCLUSIONS: These results suggest that DMAMCL is highly promising for the treatment of glioma.


Subject(s)
Cell Proliferation/drug effects , Glioma/physiopathology , Sesquiterpenes, Guaiane/pharmacology , Analysis of Variance , Animals , Brain/metabolism , Cell Line , Chromatography, High Pressure Liquid , Flow Cytometry , Glioma/drug therapy , Inhibitory Concentration 50 , Molecular Structure , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sesquiterpenes, Guaiane/chemical synthesis , Sesquiterpenes, Guaiane/chemistry , Tandem Mass Spectrometry , Tetrazolium Salts , Thiazoles
6.
Int J Oncol ; 45(6): 2411-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25269419

ABSTRACT

The critical role of VEGFR2 in tumor neovascularization and progression has allowed the design of clinically beneficial therapies based on it. Here we show that BC001, a new fully human anti-VEGFR2 monoclonal antibody, inhibits VEGF-stimulated endothelial cell migration, tube formation, and effectively suppressed the transdifferentiation of cancer stem cells into endothelial cells in vitro. Since BC001 exhibited no activity against the mouse VEGFR2 and mouse based study was required to confirm its efficacy in vivo, BC101, the mouse analogue of BC001, was developed. BC101 significantly attenuated angiogenesis according to Matrigel plug assay and resulted in ~80% growth inhibition of mouse B16F10 homograft tumors relative to vehicle control. Similarly, human analogue BC001 suppressed the growth of human xenograft tumors HCT116 and BGC823. Furthermore, immunohistochemical results showed reduced expression of CD31, VEGFR2 and Ki-67, as well as increased expression of Caspase 3 in BC001-treated tumor, which indicated BC001 was able to significantly decrease microvessel density, suppress proliferation and promote apoptosis. These results demonstrate the fully human VEGFR2 monoclonal antibody BC001 can work as an effective inhibitor of tumor angiogenesis and tumor growth both in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Apoptosis/drug effects , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , HCT116 Cells , Humans , Mice , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/therapeutic use , Xenograft Model Antitumor Assays
7.
Anticancer Drugs ; 25(8): 857-67, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24714081

ABSTRACT

Immune adjuvants have been used in cancer biotherapies to stimulate immune response to tumor cells. Despite their potential as anticancer reagents, there are several impediments to their use in clinical applications. In this study, we aim to modify the existing tuftsin structure and evaluate its antitumor activity in preclinical models. We synthesized a novel tuftsin derivative, namely, the T peptide (TP), by linking four tuftsin peptides, which showed enhanced stability in vivo. We then evaluated its anticancer activity in a postoperative residual tumor model in mice, where we surgically removed most of the primary tumor from the host, a procedure mimicking clinically postoperative patients. Despite the limited effect in intact solid tumors, TP strongly inhibited relapsed growth of residual tumors in postsurgical mice. Surgical resection of tumors accelerated residual tumor growth, but TP slowed down this process significantly. Interestingly, TP showed similar effects in human xenograft residual models. As an immunomodulator, TP could synergize the functions of macrophages, thus inhibiting the growth of cocultured tumor cells in vitro. Furthermore, TP could shift the macrophages to the tumor-suppressive M1 type and mobilize them to produce elevated cytotoxic TNF-α and NO. As a result, TP effectively prolonged the survival time of tumor-resected mice. Using the postoperative residual tumor models, we provide a body of evidence showing the antitumor activity of TP, which causes no obvious toxicity. Our study highlights the potential of TP as a postoperative adjuvant in cancer therapies.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasm, Residual/drug therapy , Tuftsin/analogs & derivatives , Tuftsin/therapeutic use , Adjuvants, Immunologic/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Disease-Free Survival , Humans , Lysine/chemistry , Macrophages/drug effects , Macrophages/immunology , Mice , Neoplasm, Residual/immunology , Neoplasm, Residual/pathology , Tuftsin/chemistry , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
8.
Stem Cells ; 32(8): 2278-89, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24740910

ABSTRACT

Erythroid differentiation-associated gene (EDAG) has been considered to be a transcriptional regulator that controls hematopoietic cell differentiation, proliferation, and apoptosis. The role of EDAG in erythroid differentiation of primary erythroid progenitor cells and in vivo remains unknown. In this study, we found that EDAG is highly expressed in CMPs and MEPs and upregulated during the erythroid differentiation of CD34(+) cells following erythropoietin (EPO) treatment. Overexpression of EDAG induced erythroid differentiation of CD34(+) cells in vitro and in vivo using immunodeficient mice. Conversely, EDAG knockdown reduced erythroid differentiation in EPO-treated CD34(+) cells. Detailed mechanistic analysis suggested that EDAG forms complex with GATA1 and p300 and increases GATA1 acetylation and transcriptional activity by facilitating the interaction between GATA1 and p300. EDAG deletion mutants lacking the binding domain with GATA1 or p300 failed to enhance erythroid differentiation, suggesting that EDAG regulates erythroid differentiation partly through forming EDAG/GATA1/p300 complex. In the presence of the specific inhibitor of p300 acetyltransferase activity, C646, EDAG was unable to accelerate erythroid differentiation, indicating an involvement of p300 acetyltransferase activity in EDAG-induced erythroid differentiation. ChIP-PCR experiments confirmed that GATA1 and EDAG co-occupy GATA1-targeted genes in primary erythroid cells and in vivo. ChIP-seq was further performed to examine the global occupancy of EDAG during erythroid differentiation and a total of 7,133 enrichment peaks corresponding to 3,847 genes were identified. Merging EDAG ChIP-Seq and GATA1 ChIP-Seq datasets revealed that 782 genes overlapped. Microarray analysis suggested that EDAG knockdown selectively inhibits GATA1-activated target genes. These data provide novel insights into EDAG in regulation of erythroid differentiation.


Subject(s)
Cell Differentiation/physiology , E1A-Associated p300 Protein/metabolism , GATA1 Transcription Factor/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Nuclear Proteins/metabolism , Acetylation , Animals , Blotting, Western , Cell Separation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Female , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , Transcriptome
9.
Onco Targets Ther ; 6: 1153-60, 2013.
Article in English | MEDLINE | ID: mdl-24009426

ABSTRACT

Myeloid/lymphoid or mixed-lineage AF4 acute lymphoblastic leukemia (MLL-AF4 ALL) is a pediatric leukemia that occurs rarely in adults. MLL-AF4 ALL is typically characterized by the presence of chromosomal translocation (t(4;1l)(q21;q23)), leading to expression of MLL-AF4 fusion protein. Although MLL-AF4 fusion protein triggers a molecular pathogenesis and hematological presentations that are unique to leukemias, the precise role of this oncogene in leukemogenesis remains unclear. Previous studies have indicated that microRNAs (miRs) might modulate the expression of MLL-AF4 ALL fusion protein, thereby suggesting the involvement of miR in progression or suppression of MLL-AF4 ALL. We have previously demonstrated that miR-205 negatively regulates transcription of an MLL-AF4 luciferase reporter. Here, we report that exogenous expression of miR-205 in MLL-AF4 human cell lines (RS4;11 and MV4-11) inversely regulates the expression of MLL-AF4 at both messenger RNA (mRNA) and protein level. Furthermore, miR-205 significantly induced apoptosis in MLL-AF4 cells as evidenced by Annex in V staining using fluorescence-activated cell sorting (FACS) analysis. The proliferative capacity of leukemic cells was suppressed by miR-205. The addition of an miR-205 inhibitor was able to restore the observed effects. In conclusion, these findings demonstrate that miR-205 may have potential value as a novel therapeutic agent in the treatment of MLL-AF4 ALL.

10.
Mol Biol Rep ; 40(12): 6811-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24057258

ABSTRACT

The mixed-lineage leukemia (MLL)-AF4 fusion protein encoded by the chromosomal translocation t(4;11) predicts a poorer prognosis in acute lymphoblastic leukemia (ALL) than in other MLL-associated leukemias. However, the detailed mechanism underlying regulation of MLL-AF4 expression remains largely unknown. In this study, we showed that microRNA (miR)-142-3p was significantly downregulated in ALL patients expressing MLL-AF4. Upregulation of miR-142-3p decreased MLL-AF4 expression in the RS4;11 leukemic cell line, which suggests that MLL-AF4 is a direct target of miR-142-3p. Ectopic expression of miR-142-3p remarkably suppressed cell proliferation and induced apoptosis in RS4;11 cells expressing the MLL-AF4 fusion protein. We also found that exogenous expression of miR-142-3p strongly reduced the expression of MLL-AF4 target genes such as homeobox A (HOXA)9, HOXA7, and HOXA10 in RS4;11 cells. Taken together, our results indicate that miR-142-3p functions as a growth suppressor in MLL-AF4(+) ALL, and its suppressive effects are mediated primarily through repression of MLL-AF4 expression.


Subject(s)
MicroRNAs/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Oncogenes , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adolescent , Adult , Aged , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Down-Regulation/genetics , Female , Gene Expression Regulation, Leukemic , Humans , Male , MicroRNAs/metabolism , Middle Aged , Promoter Regions, Genetic/genetics , Young Adult
11.
Blood ; 121(3): 499-509, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23223432

ABSTRACT

t(8;21) is one of the most frequent chromosomal translocations occurring in acute myeloid leukemia (AML) and is considered the leukemia-initiating event. The biologic and clinical significance of microRNA dysregulation associated with AML1/ETO expressed in t(8;21) AML is unknown. Here, we show that AML1/ETO triggers the heterochromatic silencing of microRNA-193a (miR-193a) by binding at AML1-binding sites and recruiting chromatin-remodeling enzymes. Suppression of miR-193a expands the oncogenic activity of the fusion protein AML-ETO, because miR-193a represses the expression of multiple target genes, such as AML1/ETO, DNMT3a, HDAC3, KIT, CCND1, and MDM2 directly, and increases PTEN indirectly. Enhanced miR-193a levels induce G(1) arrest, apoptosis, and restore leukemic cell differentiation. Our study identifies miR-193a and PTEN as targets for AML1/ETO and provides evidence that links the epigenetic silencing of tumor suppressor genes miR-193a and PTEN to differentiation block of myeloid precursors. Our results indicated a feedback circuitry involving miR-193a and AML1/ETO/DNMTs/HDACs, cooperating with the PTEN/PI3K signaling pathway and contributing to leukemogenesis in vitro and in vivo, which can be successfully targeted by pharmacologic disruption of the AML1/ETO/DNMTs/HDACs complex or enhancement of miR-193a in t(8;21)-leukemias.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/genetics , Oncogene Proteins, Fusion/genetics , Signal Transduction/genetics , Animals , Core Binding Factor Alpha 2 Subunit/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Down-Regulation/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation, Leukemic/physiology , Gene Silencing/physiology , HL-60 Cells , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mice , Mice, Nude , Myelopoiesis/genetics , Neoplasm Transplantation , Oncogene Proteins, Fusion/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/physiology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RUNX1 Translocation Partner 1 Protein , U937 Cells
12.
FEBS J ; 279(16): 2848-62, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22712502

ABSTRACT

Erythroid differentiation-associated gene (EDAG) is a haematopoietic tissue-specific transcription regulator that plays a key role in maintaining the homeostasis of haematopoietic lineage commitment. In acute myeloid leukaemia (AML) patients, the high expression level of EDAG is associated with poor prognosis. NPM1 (nucleophosmin/B23), a ubiquitous nucleolar phosphoprotein, comprises a multifunctional protein that is involved in several cellular processes, including ribosome biogenesis, centrosome duplication, cell cycle progression, cell growth and transformation. Various studies have implicated NPM1 overexpression in promoting tumour cell proliferation, blocking the differentiation of leukaemia cells and resisting apoptosis. In the present study, using co-immunoprecipitation, we characterized EDAG as a physiological binding partner of NPM1; The N-terminal (amino acids 1-124) region of EDAG interacts with the N-terminal (amino acids 118-187) of NPM1. Under cycloheximide treatment, the stability of NPM1 protein was enhanced by EDAG overexpression, whereas knockdown of EDAG by lentivirus-mediated small interfering RNA resulted in an increased degradation rate of NPM1 in K562 cells. During 4ß-phorbol l2-myristate 13-acetate-induced K562 megakaryocytic differentiation, overexpression of EDAG prevented the down-regulation of NPM1 proteins, whereas knockdown of EDAG accelerated the down-regulation of NPM1. EDAG deletion mutant lacking the binding domain with NPM1 lost the ability to stabilize NPM1 protein. Furthermore, knockdown of EDAG in K562 cells led to increased cell apoptosis induced by imatinib, and re-expression of NPM1 attenuated the increased apoptosis. These results suggest that EDAG enhances the protein stability of NPM1 via binding to NPM1, which plays a critical role in the anti-apoptosis of leukaemia cells.


Subject(s)
Apoptosis/drug effects , Nuclear Proteins/metabolism , Benzamides , Cycloheximide/pharmacology , Down-Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Humans , Imatinib Mesylate , Immunoprecipitation , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Nucleophosmin , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Tetradecanoylphorbol Acetate/pharmacology
13.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 18(6): 1595-9, 2010 Dec.
Article in Chinese | MEDLINE | ID: mdl-21176377

ABSTRACT

In order to analyze the possible epigenetic regulation mechanism of mll-af4 gene expression and find the possible microRNA regulating mll-af4 gene expression, targetscan software was used to analyze potential microRNA target sites in 3'-UTR of mll-af4. 3'-UTR fragment of mll-af4 was amplified by PCR. PCR products were cloned into EcoR I/Pst I-digested pGL3-M reporter vector, placing the 3'-UTR with potential microRNA binding site downstream of coding sequence of luciferase. The construct was cotransfected in 293T cells with control plasmid or plasmids expressing microRNAs regulating mll-af4 potentially. Western blot and RT-PCR were used to detect the expression level of mll-af4 protein and mRNA in RS4; 11 cells after transfection of miR-142-3p, respectively. The results showed that the pGL3-AF4-3'UTR of luciferase reporter recombinant plasmid contain the 3'UTR sequence of mll-af4 gene with 1935 bp, 2104 bp and 1371 bp was constructed successfully and was confirmed by enzyme digestion and gene sequencing. The luciferase assay revealed that overexpression of miR-142 could reduce the luciferase activity from the reporter construct containing the mll-af4 3'-UTR significantly. Protein and mRNA expressions of mll-af4 were found to be downregulated by miR-142-3p. It is concluded that miR-142-3p regulates the expression of mll-af4 through target binding the mll-af4 gene 3'UTR site.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Binding Sites/genetics , Cell Culture Techniques/methods , Cell Line , Humans , Plasmids , Transfection
14.
Eur J Cancer ; 46(15): 2828-36, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20655737

ABSTRACT

BACKGROUND AND AIMS: Recently, some miRNAs have been reported to be connected closely with the development of human hepatocellular carcinoma. However, the functions of these miRNAs in HCC remain largely undefined. METHODS: The expression profiles of miR-193b were compared between HCC tissues and adjacent normal liver tissues using qRT-PCR method. This method was also be used to screen the potential target genes of miR-193b. A luciferase reporter assay was conducted to confirm target association. Finally, the functional effect of miR-193b in hepatoma cells was examined further. RESULTS: miR-193b was significantly down-regulated in most of the HCC tissues compared to the matching non-tumoural liver tissues. Furthermore, ectopic expression of miR-193b dramatically suppressed the ability of hepatoma cells to form colonies in vitro and to develop tumours in nude mice. CCND1 and ETS1 were revealed to be regulated by miR-193b directly. By regulating the expressions of these oncogenes, miR-193b induced cell cycle arrest and inhibited the invasion and migration of hepatoma cells. CONCLUSIONS: miR-193b may function as a tumour suppressor in the development of HCC by acting on multiple tumourigenic pathways.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/physiology , Adult , Aged , Cell Cycle , Cell Movement , Cell Transformation, Neoplastic/pathology , Cyclin D1/antagonists & inhibitors , Down-Regulation , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors
15.
BMC Cancer ; 10: 354, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20602797

ABSTRACT

BACKGROUND: In recent years, some miRNAs have been reported to be connected closely with the development of human hepatocellular carcinoma. In our previous studies, a set of miRNAs were revealed to be dysregulated in HCC tissues. However, the functions of these miRNAs in HCC remain largely undefined. METHODS: The expression profiles of miR-183 were compared between HCC tissues and adjacent normal liver tissues using qRT-PCR method. This method was used to screen the potential target genes of miR-183. A luciferase reporter assay was conducted to confirm target association. Finally, the functional effect of miR-183 in hepatoma cells was examined. RESULTS: Among the 25 HCC samples analyzed, microRNA-183 was significantly up-regulated (twofold to 367-fold) in 17 samples compared with the matching nontumoral liver tissues. Programmed cell death 4 (PDCD4) was identified as the target gene of miR-183. Moreover, PDCD4 is a proapoptotic molecule involved in TGF-beta1-induced apoptosis in human HCC cells, we found that miR-183 transfectants were resistant to apoptosis induced by TGF-beta1. CONCLUSIONS: We conclude that miR-183 can inhibit apoptosis in human HCC cells by repressing the PDCD4 expression, and miR-183 may play an important role in HCC development.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/physiology , RNA-Binding Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Aged , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , DNA Primers/chemistry , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Luciferases/metabolism , Male , Middle Aged , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Transforming Growth Factor beta1/genetics , Tumor Cells, Cultured
16.
J Cell Biochem ; 110(4): 866-74, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20564185

ABSTRACT

Erythroid differentiation-associated gene (EDAG), a hematopoietic tissue-specific transcription regulator, plays a key role in maintaining the homeostasis of hematopoietic lineage commitment. However, the mechanism and genes regulated by EDAG remain unknown. In this study, we showed that overexpression of EDAG in a myeloid cell line 32D led to an erythroid phenotype with increased number of benzidine-positive cells and up-regulation of erythroid specific surface marker TER119. The megakaryocytic specific marker CD61 was also induced significantly. Using a genome-wide microarray analysis and a twofold change cutoff, we identified 332 genes with reduced expression and 288 genes with increased expression. Among up-regulation genes, transcription factor GATA-1 and its target genes including EKLF, NF-E2, Gfi-1b, hemogen, SCL, hemoglobin alpha, beta and megakaryocytic gene GPIX were increased. Silencing of EDAG by RNA interference in K562 cells resulted in down-regulation of these genes. Taken together, EDAG functions as a positive regulator of erythroid/megakaryocytic differentiation in 32D cells associated with the induction of GATA-1 and its target genes.


Subject(s)
Erythrocytes/metabolism , GATA1 Transcription Factor/genetics , Megakaryocytes/metabolism , Nuclear Proteins/genetics , Cell Differentiation , Erythrocytes/cytology , Flow Cytometry , Gene Silencing , Humans , Interleukin-3/metabolism , K562 Cells , Megakaryocytes/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
17.
Biochim Biophys Acta ; 1779(10): 606-15, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18599389

ABSTRACT

EDAG, a hematopoietic tissue-specific protein, is involved in the regulation of proliferation, differentiation and apoptosis of hematopoietic cells. In this study, a dose-dependent inhibition of EDAG expression by PMA was observed in K562 cells. The responsive element for the PMA-induced inhibition was contained in the region between -211 and +32bp of the EDAG gene promoter. By oligonucleotide-directed mutagenesis, EMSA, ChIP and transient transfection assays, we found that two tandem repeat GATA-1 sites in the promoter of EDAG gene played an important role in the PMA-mediated down-regulation of the EDAG gene expression in K562 cells. The kinetics of EDAG expression during PMA induction showed that the levels of EDAG expression were down-regulated concomitantly with GATA-1 down-expression. Decreased GATA-1 expression by siRNA reduced expression of EDAG in K562 cells, and restored expression of GATA-1 significantly rescued EDAG expression from PMA-mediated suppression. Overexpression of EDAG in K562 cells inhibited the megakaryocytic differentiation induced by PMA which raised the interesting possibility that PMA induced K562 cells differentiation toward megakaryocytic phenotype through, at least in part, the inhibition of EDAG expression. In vivo analysis confirmed that EDAG was highly expressed in primitive progenitor cells and down-regulated in megakaryocytes which was consistent with the expression pattern of GATA-1. Furthermore, PKC and MAPK specific inhibitors treatment attenuated the down-regulation of EDAG induced by PMA. Taken together, these results suggest that the inhibition of the EDAG gene by PMA is mediated through down-regulation of transcription factor GATA-1 and involved the PKC/MAPK signaling pathway.


Subject(s)
GATA1 Transcription Factor/metabolism , Gene Silencing/drug effects , Nuclear Proteins/genetics , Tetradecanoylphorbol Acetate/pharmacology , Base Sequence , Binding Sites , Cell Differentiation/drug effects , Cells, Cultured , Dactinomycin/pharmacology , Down-Regulation/drug effects , GATA1 Transcription Factor/physiology , Humans , K562 Cells , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding/drug effects , RNA, Messenger/metabolism , Transfection
18.
FEBS Lett ; 579(17): 3849-54, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15978578

ABSTRACT

MicroRNAs (miRNAs) are short 20-25 nucleotides RNA molecules that have been shown to regulate gene expressions in a variety of eukaryotic systems. miRNAs are widespread in eukaryotes and several hundred of miRNAs have been identified, but still a lot of miRNAs have not been detected in various eukaryotic organisms. However, it is not an easy work to clone miRNAs by traditional methods. Here, we describe the identification of 27 miRNAs from a human fetal liver cDNA library by a novel cloning method. Low molecular weight RNA fraction (< or = 200 nt) from fetal liver tissue was extracted, and polyadenylated by poly(A) polymerase. A 5' RNA adaptor was ligated to poly(A)-tailed RNA using T4 RNA ligase. After reverse transcription, the cDNA was amplified by PCR with two adaptor primers. The PCR product with a size about 109 bp was recovered and cloned into T vector. After sequencing, database searching, and expression profiling, 5 novel miRNAs were discovered among other 22 known miRNAs in human fetal liver. These finding indicate that a large diverse population of miRNAs may function to regulate gene expression in hepatocyte.


Subject(s)
Cloning, Molecular/methods , Liver/embryology , Liver/metabolism , MicroRNAs/analysis , Cells, Cultured , Fetus/metabolism , Gene Library , Humans , Liver/chemistry , MicroRNAs/genetics
19.
Ai Zheng ; 23(11): 1238-43, 2004 Nov.
Article in Chinese | MEDLINE | ID: mdl-15522166

ABSTRACT

BACKGROUND & OBJECTIVE: Embryonic development associated gene 1 (EDAG-1), located at chromosome 9q22, is specially expressed in hematopoietic cells, and related to the regulation of hematopoietic system. This study was designed to explore relationship between pathogenesis of leukemia, lymphoma and EDAG-1 through analyzing the structure of EDAG-1 coding region, and its expression in these cell lines. METHODS: Fifteen leukemia and lymphoma cell lines, HEL, K562, HL-60, Namalwa, Raji, J111, Jurkat, HuT 78, MEG-01, U937, 6T-CEM, HPB-ALL, KG-1a, THP-1, and DAMI, were selected to observe the expression of EDAG-1 by reverse transcriptase-polymerase chain reaction (RT-PCR), EDAG-1 cDNA coding fragments (1.5 kb) were purified to construct the corresponding recombinant plasmid. Then, the plasmid was sequenced to analyze mutation of the coding region. The expression of EDAG-1 protein, and mRNA in these cell lines were detected by Western blot, and Northern blot; the rearrangement and amplification of EDAG-1 genome in these cell lines were detected by Southern blot. RESULTS: EDAG-1 mRNA and protein were highly expressed in erythroleukemia cell lines (K-562, HEL), megakaryoblast leukemia cell lines (DAMI, MEG-01), and T cell leukemia cell line (Jurkat), while no gene mutation was found in coding region, no amplification and rearrangement of genome was detected in these cell lines. EDAG-1 was absent in HL-60 cell line, and rearranged in HuT 78 cells. CONCLUSION: EDAG-1 may relate with pathogenesis of erythroleukemia and megakaryoblast leukemia; its coding region may have no relation with the mechanism of its activation.


Subject(s)
Burkitt Lymphoma/metabolism , Chromosomes, Human, Pair 9 , Leukemia, Erythroblastic, Acute/metabolism , Oncogenes , Proteins/metabolism , Burkitt Lymphoma/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Humans , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Mutation , Nuclear Proteins , Plasmids , Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
20.
J Hepatol ; 40(3): 391-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15123351

ABSTRACT

BACKGROUND/AIMS: Hepatocyte growth factor (HGF) regulates proliferation of hepatic stem cells. Transcription factor nuclear factor kappa B (NF-kappaB) has been demonstrated as a key mediator for cell growth regulation. We investigated the role of NF-kappaB in HGF-mediated cellular proliferation responses in a rat liver-derived hepatic stem-like cell line WB-F344. METHODS: Cell proliferation was determined by incorporation of [3H]thymidine. Phosphorylation of ERK1/2, p38 MAPK, Akt and IkappaBalpha by HGF stimulation was detected by Western blotting. NF-kappaB activation was determined by electrophoretic mobility shift assay and NF-kappaB-mediated SEAP reporter assay. NF-kappaB activation was inhibited by treatment with an IkappaBalpha dominant-negative vector or inhibitor BAY-11-7082. RESULTS: We found that stimulation of WB-F344 cells with HGF promoted cell proliferation and effectively protected WB-F344 cells from apoptosis induced by TNF-alpha. We also observed activation of ERK1/2, p38 MAPK, Akt and NF-kappaB signaling pathways by HGF in WB-F344 cells. HGF-induced cell proliferation was partly blocked by pre-treatment of the cells with inhibitors against MEK1 or p38 MAPK, and completely blocked using an inhibitor for NF-kappaB activity. Furthermore, it was demonstrated that IkappaB mutant that suppressed NF-kappaB activity completely blocked HGF-induced cell proliferation. CONCLUSIONS: NF-kappaB activity is required for HGF-induced proliferation in hepatic stem-like cell line WB-F344, and this activity requires ERK1/2 and p38 MAPK pathways.


Subject(s)
Hepatocyte Growth Factor/physiology , Hepatocytes/cytology , NF-kappa B/physiology , Stem Cells/cytology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Division/physiology , Cell Line , DNA/metabolism , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Transcription, Genetic/physiology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...