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










Publication year range
1.
Cancer Manag Res ; 13: 1233-1243, 2021.
Article in English | MEDLINE | ID: mdl-33603469

ABSTRACT

PURPOSE: Red blood cell distribution width (RDW) has been considered as a potential indicator of the effects of treatment or as a prognostic indicator for various malignancies. Most chronic myeloid leukemia (CML) patients are in the chronic phase, but some have transformed to accelerated phase or blast phase (blast crisis). However, the clinical significance of RDW in CML remains limited. PATIENTS AND METHODS: In the present study, detailed clinical information and the RDW of 168 healthy people and 153 CML patients (106 patients for the training cohort and 47 patients for the validation cohort) were retrospectively assessed. RESULTS: Multivariate analysis demonstrated that patient age (OR, 1.081; 95CI% 1.039~1.125; p < 0.001), platelet counts (OR, 0.997; 95CI% 0.994~0.999; p = 0.001) and RDW at admission (OR,1.469; 95CI% 1.121~1.925; p = 0.005) were significantly associated with the patients with advanced phase. Among CML patients in the chronic phase, higher RDW was significantly associated with overall survival (OS; p = 0.0008) and the event-free survival (EFS; p = 0.0221) among CML patients with chronic phase, but not with Transformation-free survival (TFS; p = 0.0821). Furthermore, higher RDW was associated with higher mortality compared to patients with low RDW (CML-associated deaths; p < 0.0001). In addition, a decline in RDW is associated with the treatment of CML patients with tyrosine kinase inhibitors, especially at 6 and 12 months after the start of treatment. CONCLUSION: Higher RDW is a potential prognostic biomarker for chronic CML patients.

2.
Nat Commun ; 12(1): 51, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397955

ABSTRACT

Identifying novel drug targets to overcome resistance to tyrosine kinase inhibitors (TKIs) and eradicating leukemia stem/progenitor cells are required for the treatment of chronic myelogenous leukemia (CML). Here, we show that ubiquitin-specific peptidase 47 (USP47) is a potential target to overcome TKI resistance. Functional analysis shows that USP47 knockdown represses proliferation of CML cells sensitive or resistant to imatinib in vitro and in vivo. The knockout of Usp47 significantly inhibits BCR-ABL and BCR-ABLT315I-induced CML in mice with the reduction of Lin-Sca1+c-Kit+ CML stem/progenitor cells. Mechanistic studies show that stabilizing Y-box binding protein 1 contributes to USP47-mediated DNA damage repair in CML cells. Inhibiting USP47 by P22077 exerts cytotoxicity to CML cells with or without TKI resistance in vitro and in vivo. Moreover, P22077 eliminates leukemia stem/progenitor cells in CML mice. Together, targeting USP47 is a promising strategy to overcome TKI resistance and eradicate leukemia stem/progenitor cells in CML.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Cell Proliferation/drug effects , DNA Damage , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fusion Proteins, bcr-abl , Gene Expression Regulation, Leukemic/drug effects , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Stability/drug effects , Proteolysis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Xenograft Model Antitumor Assays , Y-Box-Binding Protein 1/metabolism , ras Proteins/metabolism
4.
FEBS Lett ; 589(15): 1981-7, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-25979172

ABSTRACT

HOTAIR is significantly overexpressed in various cancers and facilitates tumor invasion and metastasis. However, whether HOTAIR plays oncogenic roles in acute myeloid leukemia (AML) is still unknown. Here, we report that HOTAIR expression was obviously increased in leukemic cell lines and primary AML blasts. Clinically, AML patients with higher HOTAIR predicted worse clinical outcome compared with those with lower HOTAIR. Importantly, HOTAIR knockdown by small hairpin RNA inhibited cell growth, induced apoptosis, and decreased number of colony formation. Finally, HOTAIR modulated c-KIT expression by competitively binding miR-193a. Collectively, our data suggest that HOTAIR plays an important oncogenic role in AML and might serve as a marker for AML prognosis and a potential target for therapeutic intervention.


Subject(s)
Leukemia, Myeloid, Acute/genetics , MicroRNAs/physiology , Proto-Oncogene Proteins c-kit/genetics , RNA, Long Noncoding/physiology , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/pathology , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/genetics , Prognosis , RNA, Long Noncoding/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
5.
Asian Pac J Cancer Prev ; 16(6): 2219-25, 2015.
Article in English | MEDLINE | ID: mdl-25824741

ABSTRACT

SHP1 negatively regulates the Janus kinase 2/signal transducer and activator of transcription (JAK2/STAT) signaling pathway, which is constitutively activated in myeloproliferative neoplasms (MPNs) and leukemia. Promoter hypermethylation resulting in epigenetic inactivation of SHP1 has been reported in myelomas, leukemias and other cancers. However, whether SHP1 hypermethylation occurs in MPNs, especially in Chinese patients, has remained unclear. Here, we report that aberrant hypermethylation of SHP1 was observed in several leukemic cell lines and bone marrow mononuclear cells from MPN patients. About 51 of 118 (43.2%) MPN patients including 23 of 50 (46%) polycythaemia vera patients, 20 of 50 (40%) essential thrombocythaemia and 8 of 18 (44.4%) idiopathic myelofibrosis showed hypermethylation by methylation-specific polymerase chain reaction. However, SHP1 methylation was not measured in 20 healthy volunteers. Hypermethylation of SHP1 was found in MPN patients with both positive (34/81, 42%) and negative (17/37, 45.9%) JAK2V617F mutation. The levels of SHP1 mRNA were significantly lower in hypermethylated samples than unmethylated samples, suggesting SHP1 may be epigenetically inactivated in MPN patients. Furthermore, treatment with 5-aza-2'-deoxycytidine (AZA) in K562 cells showing hypermethylation of SHP1 led to progressive demethylation of SHP1, with consequently increased reexpression of SHP1. Meanwhile, phosphorylated JAK2 and STAT3 were progressively reduced. Finally, AZA increased the expression of SHP1 in primary MPN cells with hypermethylation of SHP1. Therefore, our data suggest that epigenetic inactivation of SHP1 contributes to the constitutive activation of JAK2/STAT signaling. Restoration of SHP1 expression by AZA may contribute to clinical treatment for MPN patients.


Subject(s)
DNA Methylation , Janus Kinase 2/genetics , Mutation/genetics , Myeloproliferative Disorders/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , STAT3 Transcription Factor/genetics , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Blotting, Western , Case-Control Studies , Decitabine , Epigenomics , Humans , Molecular Sequence Data , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Leuk Res ; 39(4): 471-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25666256

ABSTRACT

Constitutive activation of Janus kinase 2/signal transducers and activators of transcription (JAK2/STAT) signaling caused by JAK2V617F and other mutations is central to the pathogenesis of myeloproliferative neoplasm (MPN). Negative regulators such as suppressors of cytokine signaling (SOCS) inhibit activated JAK2/STAT signaling. However, whether silencing of negative regulators facilitates JAK2/STAT signaling is unclear. Here, we report that loss of miR-375 expression contributes to the constitutive activation of JAK2/STAT signaling. MiR-375 reduced JAK2 protein level and repressed the activity of a luciferase reporter by binding 3'-untranslated regions, which was abolished by the mutation of the predicted miR-375-binding site. Meanwhile, a significant inverse correlation between the expressions of miR-375 and JAK2 was found in multiple types of leukemic cell lines and bone marrow mononuclear cells from MPN patients, suggesting that JAK2 may be a miR-375 target gene. Furthermore, forced expression of miR-375 inhibited constitutive and inducible JAK2/STAT signaling, suppressed cell proliferation, and decreased colony formation in hematopoietic progenitors from MPN patients. Finally, histone deacetylation (HDAC) inhibitors restored miR-375 expression, which was much lower in patients with MPN compared with healthy volunteers. Collectively, our data suggest that the loss of miR-375 expression enhances the constitutive and persistent activation of JAK2/STAT signaling. Restoration of miR-375 expression might contribute to the clinical treatment for MPN patients.


Subject(s)
Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Janus Kinase 2/metabolism , MicroRNAs/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , STAT Transcription Factors/metabolism , Apoptosis , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Humans , Janus Kinase 2/genetics , Mutation/genetics , Myeloproliferative Disorders/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/genetics , Signal Transduction , Tumor Cells, Cultured
7.
J Cell Biochem ; 116(2): 287-98, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25187418

ABSTRACT

Honokiol, a constituent of Magnolia officinalis, has been reported to possess potent anti-cancer activity through targeting multiple signaling pathways in numerous malignancies including acute myeloid leukemia (AML). However, the underlying mechanisms remain to be defined. Here, we report that honokiol effectively decreased enzyme activity of histone deacetylases (HDACs) and reduced the protein expression of class I HDACs in leukemic cells. Moreover, treatment with proteasome inhibitor MG132 prevented honokiol-induced degradation of class I HDACs. Importantly, honokiol increased the levels of p21/waf1 and Bax via triggering acetylation of histone in the regions of p21/waf1 and Bax promoter. Honokiol induced apoptosis, decreased activity of HDACs, and significantly inhibited the clonogenic activity of hematopoietic progenitors in bone marrow mononuclear cells from patients with AML. However, honokiol did not decrease the activity of HDACs and induce apoptosis in normal hematopoietic progenitors from unbilicial cord blood. Finally, honokiol dramatically reduced tumorigenicity in a xenograft leukemia model. Collectively, our findings demonstrate that honokiol has anti-leukemia activity through inhibiting HDACs. Thus, being a relative non-toxic agent, honokiol may serve as a novel natural agent for cancer prevention and therapy in leukemia.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Cycle Checkpoints/drug effects , Histone Deacetylases/metabolism , Leukemia, Myeloid/drug therapy , Lignans/pharmacology , Acute Disease , Adult , Aged , Animals , Biocatalysis/drug effects , Blotting, Western , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drugs, Chinese Herbal/pharmacology , Female , Humans , K562 Cells , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Male , Mice, Nude , Middle Aged , Proteasome Endopeptidase Complex/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
8.
Leuk Lymphoma ; 55(2): 349-57, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23550990

ABSTRACT

The single nucleotide polymorphism (SNP) rs16754 of the WT1 gene has been described as a possible prognostic marker in patients with acute myeloid leukemia (AML). However, the results in this field are not reproducible in different cohorts. In this study, we investigated WT1 mutations, expression levels and SNP rs16754 in a cohort of 122 adult patients with AML. As the major allele (65.6%) in a Chinese population, WT1(GG) was associated with younger age (≤ 60) and lower percentage of blasts than WT1(GA/AA). Meanwhile, improved overall survival (OS, p = 0.035) and disease-free survival (DFS, p = 0.021) were observed in WT1(GG) compared with WT1(GA/AA). We then found that WT1 mutation, occurring in 8% of patients with AML, did not predict clinical outcome. Finally, WT1 levels were higher in patients with WT1(GG) than in those with WT1(GA/AA). However, high levels of WT1 (> median) predicted worse OS (p = 0.015) and DFS (p = 0.034) than low levels of WT1 (≤ median). However, further studies are required to elucidate the mechanism of why WT1(GG), which was associated with higher median expression of WT1 that predicts worse OS and DFS compared to low expression of WT1, predicted better OS and DFS compared with WT1(GA/AA). In summary, WT1 rs16754 and WT1 expression have a significant impact on clinical outcome in patients with AML.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid/genetics , Mutation , Polymorphism, Single Nucleotide , WT1 Proteins/genetics , Acute Disease , Adolescent , Adult , Aged , Asian People/genetics , China , Female , Gene Frequency , Genotype , Humans , Induction Chemotherapy/methods , Kaplan-Meier Estimate , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/ethnology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/ethnology , Leukemia, Promyelocytic, Acute/genetics , Male , Middle Aged , Multivariate Analysis , Outcome Assessment, Health Care/statistics & numerical data , Proportional Hazards Models , Remission Induction , Young Adult
9.
Mol Cell Biochem ; 383(1-2): 137-48, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23867991

ABSTRACT

Histone deacetylases (HDACs) inhibitor is a promising new approach to the treatment of lung cancer therapy via inhibiting cell growth and inducing apoptosis. miR-15a and miR-16-1 are important tumor suppressors through modulating B cell lymphoma 2 (Bcl-2), Cyclin D1, D2, and others. However, whether HDACs inhibitor modulates the expression of miR-15a/16-1 in lung cancer is still unknown. The purpose of our study was to identify a new miRNA-mediated mechanism which plays an important role in the anti-cancer effects of HDACs inhibitor. We found HDACs inhibitors trichostatin A (TSA) and sodium butyrate upregulated the expression of miR-15a/16-1, residing in the host tumor suppressor Dleu2 gene, through increasing the histone acetylation in the region of Dleu2/miR-15a/16-1 promoter in lung cancer cells. Moreover, among class Ι HDACs subtypes, only knockdown of HDAC3 by specific siRNA increased the hyperacetylation of Dleu2/miR-15a/16-1 promoter region and finally resulted in the upregulation of miR-15a/16-1. Furthermore, overexpression of miR-15a/16-1, which were always deleted or downregulated in lung cancer cells, effectively suppressed cell growth and reduced colony formation. Finally, TSA reduced the expression of Bcl-2, an important survival protein in lung cancer cells, partly through upregulation of miR-15a/16-1. Therefore, this offers a therapeutic strategy that lung cancer patients who exhibit low level of miR-15a/16-1 or high activity of HDACs may benefit from HDACs inhibitor-based therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , MicroRNAs/genetics , Tumor Suppressor Proteins/genetics , Acetylation/drug effects , Apoptosis/drug effects , Butyric Acid/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Histones/metabolism , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Long Noncoding , Transferases , Tumor Stem Cell Assay , Tumor Suppressor Proteins/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
10.
Carcinogenesis ; 34(7): 1442-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23430957

ABSTRACT

Suppressors of cytokine signaling, SOCS1 and SOCS3, are important negative regulators of Janus kinase 2/signal transducers and activators of transcription signaling, which is constitutively activated in myeloproliferative neoplasms (MPNs) and leukemia. Curcumin has been shown to possess anticancer activity through different mechanisms. However, whether curcumin can regulate the expression of SOCS1 and SOCS3 is still unknown. Here, we found that curcumin elevated the expression of SOCS1 and SOCS3 via triggering acetylation of histone in the regions of SOCS1 and SOCS3 promoter in K562 and HEL cells. As a novel histone deacetylases (HDACs) inhibitor, curcumin inhibited HDAC enzyme activities and decreased the levels of HDAC1, 3 and 8 but not HDAC2. Knockdown of HDAC8 by small interfering RNA markedly elevated the expression of SOCS1 and SOCS3. Moreover, ectopic expression of HDAC8 decreased the levels of SOCS1 and SOCS3. Thus, HDAC8 plays an important role in the modulation of SOCS1 and SOCS3 by curcumin. Also, trichostatin A (TSA), an inhibitor of HDACs, increased the levels of SOCS1 and SOCS3. Furthermore, curcumin increased the transcript levels of SOCS1 and SOCS3 and significantly inhibited the clonogenic activity of hematopoietic progenitors from patients with MPNs. Finally, curcumin markedly inhibited HDAC activities and decreased HDAC8 levels in primary MPN cells. Taken together, our data uncover a regulatory mechanism of SOCS1 and SOCS3 through inhibition of HDAC activity (especially HDAC8) by curcumin. Thus, being a relative non-toxic agent, curcumin may offer a therapeutic advantage in the clinical treatment for MPNs.


Subject(s)
Bone Marrow Neoplasms/metabolism , Curcumin/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Myeloproliferative Disorders/pathology , Suppressor of Cytokine Signaling Proteins/metabolism , Acetylation , Bone Marrow Neoplasms/enzymology , Bone Marrow Neoplasms/genetics , Chromatin Immunoprecipitation , Enzyme Activation , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , K562 Cells , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics , Primary Cell Culture , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
11.
Exp Hematol ; 41(3): 261-70.e4, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23111066

ABSTRACT

Constitutive activation of Janus kinase 2/signal transducers and activators of transcription (JAK2/STAT) signaling has an important role in the oncogenesis of myeloproliferative neoplasms (MPNs) and leukemia. Histone deacetylases (HDACs) inhibitors have been reported to possess anticancer activity through different mechanisms. However, whether HDACs inhibitors suppress JAK2/STAT signaling in MPNs is still unknown. In this study, we show that the HDAC inhibitor sodium butyrate (SB) inhibited JAK2/STAT signaling and increased the expression of suppressors of cytokine signaling 1 (SOCS1) and SOCS3, both of which are the potent feedback inhibitors of JAK2/STAT signaling. SB upregulated the expression of SOCS1 and SOCS3 by triggering the promoter-associated histone acetylation of SOCS1 and SOCS3 in K562 and HEL cell lines. Importantly, we found that upon knockdown of each class I HDACs, only knockdown of HDAC8 resulted in the increased expression of SOCS1 and SOCS3. Moreover, overexpression of SOCS1 and SOCS3 significantly inhibited cell growth and suppressed JAK2/STAT signaling in K562 and HEL cells. Furthermore, SB increased the transcript levels of SOCS1 and SOCS3 and inhibited the clonogenic activity of hematopoietic progenitors from patients with MPNs. Taken together, these data establish a new anticancer mechanism that SB inhibits JAK2/STAT signaling through HDAC8-mediated upregulation of SOCS1 and SOCS3. Thus, HDACs inhibitors may have therapeutic potential for the treatment of MPNs.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Janus Kinase 2/metabolism , Repressor Proteins/metabolism , STAT Transcription Factors/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Acetylation/drug effects , Blotting, Western , Butyrates/pharmacology , Cell Line, Tumor , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/metabolism , Histone Deacetylases/genetics , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Janus Kinase 2/genetics , K562 Cells , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , RNA Interference , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Up-Regulation/drug effects
12.
J Exp Clin Cancer Res ; 31: 27, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22449094

ABSTRACT

BACKGROUND: Pure curcumin has been reported to down-regulate the expression of WT1 in leukemic cells. However, the molecular mechanism underlying the down-regulation of WT1 by curcumin is not completely delineated. The purpose of this present study is to identify a new miRNA-mediated mechanism which plays an important role in the anti-proliferation effects of curcumin in leukemic cells. METHODS: K562 and HL-60 cells were treated with different concentrations of curcumin for 24 and 48 hours, the level of miR-15a/16-1 and WT1 were detected by qRT-PCR and Western blotting. WT1 expression and cell proliferation were detected by Western blotting and CCK-8, after curcumin treated-K562 and HL-60 cells were transfected with anti-miR-15a/16-1 oligonucleotides. RESULTS: We found that pure curcumin upregulated the expression of miR-15a/16-1 and downregulated the expression of WT1 in leukemic cells and primary acute myeloid leukemia (AML) cells. Overexpression of miR-15a/16-1 deduced the protein level of WT1 in leukemic cells, but downregulation of WT1 by siRNA-WT1 could not increase the expression of miR-15a/16-1 in leukemic cells. These results reveal that curcumin induced-upregulation of miR-15a/16-1 is an early event upstream to downregulation of WT1. Furthermore, anti-miR-15a/16-1 oligonucleotides (AMO) partly reversed the downregulation of WT1 induced by pure curcumin in leukemic cells and AMO promoted the growth of curcumin treated-K562 and HL-60 cells. CONCLUSION: Thus, these data suggest for the first time that pure curcumin downregulated the expression of WT1 partly by upregulating the expression of miR-15a/16-1 in leukemic cells. miR-15a/16-1 mediated WT1 downregulation plays an important role in the anti-proliferation effect of curcumin in leukemic cells.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Leukemia/genetics , MicroRNAs/genetics , WT1 Proteins/genetics , Cell Proliferation/drug effects , HL-60 Cells , Humans , K562 Cells , Oligonucleotides/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
13.
J Exp Clin Cancer Res ; 30: 110, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22133358

ABSTRACT

BACKGROUND: miR-15a and miR-16-1(miR-15a/16-1) have been implicated as tumor suppressors in chronic lymphocytic leukemia, multiple myeloma, and acute myeloid leukemic cells. However the mechanism of inhibiting the proliferation of leukemic cells is poorly understood. METHODS: K562 and HL-60 cells were transfected with pRS-15/16 or pRS-E, cell growth were measured by CCK-8 assay and direct cell count. Meanwhile WT1 protein and mRNA level were measured by Western blotting and quantitative real-time PCR. RESULTS: In this study we found that over-expression of miR-15a/16-1 significantly inhibited K562 and HL-60 cells proliferation. Enforced expression of miR-15a/16-1 in K562 and HL-60 cells significantly reduced the protein level of WT1 but not affected the mRNA level. However enforced expression of miR-15a/16-1 can not reduce the activity of a luciferase reporter carrying the 3'-untranslated region(3'UTR) of WT1. Silencing of WT1 by specific siRNA suppressed leukemic cells proliferation resembling that of miR-15a/16-1 over-expression. Anti-miR-15a/16-1 oligonucleotides (AMO) reversed the expression of WT1 in K562 and HL-60 cells. Finally, we found a significant inverse correlation between miR-15a or miR-16-1 expression and WT1 protein levels in primary acute myeloid leukemia (AML) blasts and normal controls. CONCLUSIONS: These data suggest that miR-15a/16-1 may function as a tumor suppressor to regulate leukemic cell proliferation potentially by down-regulating the WT1 oncogene. However WT1 is not directly targeted by miR-15a/16-1 through miRNA-mRNA base pairing, therefore more study are required to understand the mechanism by which miR-15a/16-1 downregulate WT1.


Subject(s)
Down-Regulation , Leukemia/genetics , Leukemia/metabolism , MicroRNAs/metabolism , WT1 Proteins/metabolism , 3' Untranslated Regions , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , HL-60 Cells , Humans , K562 Cells , Oligonucleotides, Antisense , RNA, Messenger/metabolism
14.
Leuk Lymphoma ; 52(12): 2365-71, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21838537

ABSTRACT

miR-15a and miR-16-1 (miR-15a/16-1) have been implicated in apoptosis, cell cycle regulation and chemosensitivity of tumor cells, but little is known about the role of miR-15a/16-1 in the differentiation of leukemic cells. In this study, we found that the expression level of miR-15a/16-1 was up-regulated by all-trans retinoic acid (ATRA) treatment in NB4, HL-60 and U937 cell lines and primary leukemic cells. Overexpression of miR-15a/16-1 could not directly drive cells to undergo differentiation but enhanced ATRA-induced differentiation in NB4 and U937 cells. Up-regulation of miR-15a/16-1 was also observed in 36 patients with acute myeloid leukemia (AML) who achieved a complete remission (CR), and two of them showed sharp down-regulation of miR-15a/16-1 when they had a molecular relapse. These data indicate that miR-15a/16-1 plays an important role in the ATRA-induced differentiation of leukemic and primary AML cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Leukemia/genetics , MicroRNAs/metabolism , Tretinoin/pharmacology , Up-Regulation/drug effects , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , Leukemia/drug therapy , Tretinoin/therapeutic use , U937 Cells
15.
Cell Biol Int ; 35(9): 891-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21524276

ABSTRACT

TIEG1 (TGF-ß inducible early gene 1) plays a significant role in regulating cell proliferation and apoptosis in various cell types. Previous studies have shown a close relationship between the expression level of TIEG1 and various cancers, including breast, prostate, colorectal and pancreatic cancer. In this study, we up-regulated the gene expression of TIEG1 in SW1990 pancreatic cancer cell line by a lentivirus transfection system and investigated its potential as a therapeutic target for pancreatic cancer. The results showed that lentivirus-mediated overexpression of TIEG1 gene inhibited human pancreatic cancer SW1990 cell proliferation and caused the cell cycle arrest at the G1-phase in vitro. SW1990 cells transduced with lenti-TIEG1 showed significant inhibition of colony formation and cancer cell growth in 3-D culture model. Moreover, overexpression of TIEG1 gene significantly slowed the growth of SW1990 xenografts in nude mice. Taken together, these data provided evidence that overexpression of TIEG1 gene by a lentivirus transfection system led to suppressed human pancreatic cancer cell growth and might therefore be a feasible approach in the clinical management of pancreatic cancer.


Subject(s)
Lentivirus/genetics , Pancreatic Neoplasms/pathology , Transforming Growth Factor beta/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , RNA, Messenger/metabolism , Transfection , Transforming Growth Factor beta/metabolism
16.
Biochem Biophys Res Commun ; 403(2): 203-8, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21056550

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that regulate target gene expression through translation repression or messenger RNA degradation. MiR-15a and 16-1 form a cluster at the chromosomal region 13q14, which is frequently deleted or down-regulated in chronic lymphocytic leukemia. Arsenic trioxide (As(2)O(3), ATO) has been successfully applied to treat acute promyelocytic leukemia (APL). Its combination with other drugs presented therapeutic activities in hematologic and solid tumors. Here we investigated the potential synergy between miR-15a/16-1 and ATO on Bcr-Abl positive leukemic K562 cells. In this study, we found that combination of miR-15a/16-1 and ATO synergistically induced growth inhibition and apoptosis in K562 cells. The apoptosis, at least in part, through regulating mitochondrial function including the release of cytochrome c and loss of mitochondrial transmembrane potential, also activation of caspase-3 and degradation of poly-adenosine diphosphate-ribose polymerase. However, the expression of Bcr-Abl was not affected by ATO and/or miR-15a/16-1. Moreover, apoptotic synergy between miR-15a/16-1 and ATO was observed in Bcr-Abl negative leukemic cell lines and primary leukemic cells. Taken together, these findings suggested that the combined regiment of miR-15a/16-1 and ATO might be a potential therapeutic remedy for the treatment of leukemia.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis , Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , MicroRNAs/therapeutic use , Oxides/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Arsenic Trioxide , Drug Synergism , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Mitochondria/drug effects
17.
Blood ; 105(9): 3714-21, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15671440

ABSTRACT

As a promising new class of anticancer drugs, camptothecins have advanced to the forefront of several areas of therapeutic and developmental chemotherapy. In the present study, we report that NSC606985, a rarely studied camptothecin analog, induces apoptosis in acute myeloid leukemia (AML) cells NB4 and U937 and inhibits the proliferation without cell death in breakpoint cluster region-Abelson murine leukemia (bcr-abl) kinase-carrying leukemic K562 cells. For apoptosis induction or growth arrest, nanomolar concentrations of NSC606985 are sufficient. At such low concentrations, this agent also significantly inhibits the clonogenic activity of hematopoietic progenitors from patients with AML. For apoptosis induction, NSC606985 rapidly induces the proteolytic activation of protein kinase Cdelta (PKCdelta) with loss of mitochondrial transmembrane potential (DeltaPsim) and caspase-3 activation. Cotreatment with rottlerin, a PKCdelta-specific inhibitor, completely blocks NSC606985-induced mitochondrial DeltaPsim loss and caspase-3 activation, while the inhibition of caspase-3 by z-DEVD-fluoromethyl ketone (Z-DEVD-fmk) only partially attenuates PKCdelta activation and apoptosis. These data indicate that NSC606985-induced PKCdelta activation is an early event upstream to mitochondrial DeltaPsim loss and caspase-3 activation, while activated caspase-3 has an amplifying effect on PKCdelta proteolysis. In addition, NSC606985-induced apoptosis by PKCdelta also involves caspase-3-independent mechanisms. Taken together, our results suggest that NSC606985 is a potential agent for the treatment of AML.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Camptothecin/analogs & derivatives , Leukemia/pathology , Protein Kinase C/metabolism , Camptothecin/pharmacology , Camptothecin/therapeutic use , Caspase 3 , Caspases , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Intracellular Membranes/physiology , Leukemia/drug therapy , Membrane Potentials , Mitochondria/physiology , Mitochondria/ultrastructure , Protein Kinase C-delta
18.
Article in Chinese | MEDLINE | ID: mdl-15256145

ABSTRACT

OBJECTIVE: To evaluate the protective effect of amifostine on hydroquinone-induced apoptosis of bone marrow mononuclear cells in vitro. METHODS: The mononuclear cells were separated and divided into four groups: blank control, amifostine group, hydroquinone group, amifostine + hydroquinone group. The cell apoptotic rate was examined in separated group at different time point, and apoptosis was detected by HT stain, then cell morphology was observed under fluorescent microscope and DNA fragments was tested by agarose gel electrophoresis. In addition, apoptotic and necrotic rate was detected by flow cytometer. RESULTS: After 10 hour culture, DNA ladder was detected in the hydroquinone group, but not in other groups. The apoptotic rate was not significantly different between amifostine group and blank control group at different culture time (P > 0.05). After 8 - 12 hour culture, the apoptotic rate in amifostine + hydroquinone group was significantly lower than that in the group of hydroquinone alone (P < 0.01). After 18 - 48 hour culture, the necrotic rate in amifostine + hydroquinone group was lower than that in the group of hydroquinone alone (P < 0.05). CONCLUSION: Amifostine can protect cell from hydroguinone-induced bone marrow damage through inhibition on cell apoptosis, and decrease in cell necrosis.


Subject(s)
Amifostine/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/cytology , Hydroquinones/antagonists & inhibitors , Cells, Cultured , Humans , Leukocytes, Mononuclear/cytology , Protective Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...