Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 15(3): 294-303, 2017 03.
Article in English | MEDLINE | ID: mdl-28011885

ABSTRACT

PRL-3 (PTP4A3), a metastasis-associated phosphatase, is also upregulated in patients with acute myeloid leukemia (AML) and is associated with poor prognosis, but the underlying molecular mechanism is unknown. Here, constitutive expression of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo Furthermore, PRL-3 phosphatase activity dependently upregulates LIN28B, a stem cell reprogramming factor, which in turn represses the let-7 mRNA family, inducing a stem cell-like transcriptional program. Notably, elevated levels of LIN28B protein independently associate with worse survival in AML patients. Thus, these results establish a novel signaling axis involving PRL-3/LIN28B/let-7, which confers stem cell-like properties to leukemia cells that is important for leukemogenesis.Implications: The current study offers a rationale for targeting PRL-3 as a therapeutic approach for a subset of AML patients with poor prognosis. Mol Cancer Res; 15(3); 294-303. ©2016 AACR.


Subject(s)
Carcinogenesis/genetics , Leukemia, Myeloid, Acute/metabolism , Protein Tyrosine Phosphatases/metabolism , RNA-Binding Proteins/metabolism , Stem Cells/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HL-60 Cells , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Transfection
2.
Exp Hematol ; 42(12): 1041-52.e1-2, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25139404

ABSTRACT

Overexpression of protein-tyrosine phosphatase of regenerating liver 3 (PRL-3) has been identified in about 50% of patients with acute myeloid leukemia (AML). The mechanism of regulation of PRL-3 remains obscure. Signal transducer and activator of transcription 3 (STAT3), a latent transcriptional factor, has also been often found to be activated in AML. We first identified STAT3-consensus-binding sites in the promoter of PRL-3 genes. Then we experimentally validated the direct binding and transcriptional activation. We applied shRNA-mediated knockdown and overexpression approaches in STAT3(-/-) liver cells and leukemic cells to validate the functional regulation of PRL-3 by STAT3. A STAT3 core signature, derived through data mining from publicly available gene expression data, was employed to correlate PRL-3 expression in large AML patient samples. We discovered that STAT3 binds to the -201 to -210 region of PRL-3, which was conserved between human and mouse. Importantly, PRL-3 protein was significantly reduced in mouse STAT3-knockout liver cells compared with STAT3-wild type counterparts, and ectopic expression of STAT3 in these cells led to a pronounced increase in PRL-3 protein. We demonstrated that STAT3 functionally regulated PRL-3, and STAT3 core signature was enriched in AML with high PRL-3 expression. Targeting either STAT3 or PRL-3 reduced leukemic cell viability. Silencing PRL-3 impaired invasiveness and induced leukemic cell differentiation. In conclusion, PRL-3 was transcriptionally regulated by STAT3. The STAT3/PRL-3 regulatory loop contributes to the pathogenesis of AML, and it might represent an attractive therapeutic target for antileukemic therapy.


Subject(s)
Immediate-Early Proteins/physiology , Leukemia, Myeloid, Acute/enzymology , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , STAT3 Transcription Factor/physiology , Animals , Binding Sites , Cell Differentiation , Cell Line, Tumor , Conserved Sequence , DNA Mutational Analysis , DNA, Neoplasm/genetics , Gene Dosage , Gene Expression Regulation, Leukemic , Genes, Reporter , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Liver/metabolism , Mice , Mutagenesis, Site-Directed , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Tyrosine Phosphatases/biosynthesis , Protein Tyrosine Phosphatases/genetics , RNA Interference , RNA, Small Interfering/pharmacology , STAT3 Transcription Factor/deficiency , Signal Transduction , Species Specificity , Transfection
3.
Cancer Res ; 74(11): 3043-53, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24686170

ABSTRACT

PRL-3, an oncogenic dual-specificity phosphatase, is overexpressed in 50% of acute myelogenous leukemia (AML) and associated with poor survival. We found that stable expression of PRL-3 confers cytokine independence and growth advantage of AML cells. However, how PRL-3 mediates these functions in AML is not known. To comprehensively screen for PRL3-regulated proteins in AML, we performed SILAC-based quantitative proteomics analysis and discovered 398 significantly perturbed proteins after PRL-3 overexpression. We show that Leo1, a component of RNA polymerase II-associated factor (PAF) complex, is a novel and important mediator of PRL-3 oncogenic activities in AML. We described a novel mechanism where elevated PRL-3 protein increases JMJD2C histone demethylase occupancy on Leo1 promoter, thereby reducing the H3K9me3 repressive signals and promoting Leo1 gene expression. Furthermore, PRL-3 and Leo1 levels were positively associated in AML patient samples (N=24; P<0.01). On the other hand, inhibition of Leo1 reverses PRL-3 oncogenic phenotypes in AML. Loss of Leo1 leads to destabilization of the PAF complex and downregulation of SOX2 and SOX4, potent oncogenes in myeloid transformation. In conclusion, we identify an important and novel mechanism by which PRL-3 mediates its oncogenic function in AML.


Subject(s)
Carcinogenesis/genetics , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oncogenes , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Down-Regulation/genetics , Female , Gene Expression Regulation, Leukemic , HEK293 Cells , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Up-Regulation
4.
Oncotarget ; 4(7): 1008-18, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23900284

ABSTRACT

Multiple myeloma (MM) is characterized by recurrent chromosomal translocations. MMSET, identified by its fusion to the IgH locus in t(4;14) MM, is universally overexpressed in t(4;14) MM. In order to identify cell surface biomarkers associated with t(4;14) MM for small molecule or antibody based therapies, we knocked down MMSET expression with shRNA and generated a cell line pair from KMS11, a t(4;14) MM cell line. We used quantitative mass spectrometry to identify plasma membrane proteins associated with MMSET overexpression. Using this approach, 50 cell surface proteins were identified as differentially expressed between KMS11 and KMS11/shMMSET. Western blot and flow cytometry analysis indicated SLAMF7 was over-expressed in t(4;14) MM cell lines and down-regulated by MMSET shRNAs. SLAMF7 expression was also confirmed in primary t(4;14) MM samples by flow cytometry analysis. Quantitative RT-PCR and ChIP analysis indicated MMSET might regulate the transcription level of SLAMF7 and be an important functional element for SLAMF7 promoter activity. Furthermore, SLAMF7 shRNA could induce G1 arrest or apoptosis and reduce clonogenetic capacity in t(4;14) MM cells. Overall, these results illustrated SLAMF7 might be a novel cell surface protein associated with t(4;14) MM. It is potential to develop t(4;14) MM targeted therapy by SLAMF7 antibody mediated drug delivery.


Subject(s)
Biomarkers, Tumor/metabolism , Histone-Lysine N-Methyltransferase/biosynthesis , Multiple Myeloma/metabolism , Repressor Proteins/biosynthesis , Cell Line, Tumor , Cell Membrane/genetics , Cell Membrane/metabolism , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 4 , Down-Regulation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Multiple Myeloma/genetics , Proteomics/methods , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family , Transfection , Translocation, Genetic
5.
Oncotarget ; 4(8): 1230-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23985559

ABSTRACT

Multiple myeloma is the abnormal clonal expansion of post germinal B cells in the bone marrow. It was previously reported that clonogenic myeloma cells are CD138-. Human MM cell lines RPMI8226 and NCI H929 contained 2-5% of CD138- population. In this study, we showed that CD138- cells have increased ALDH1 activity, a hallmark of normal and neoplastic stem cells. CD138-ALDH+ cells were more clonogenic than CD138+ALDH- cells and only CD138- cells differentiated into CD138+ populations. In vivo tumor initiation and clonogenic potentials of the CD138- population was confirmed using NOG mice. We derived a gene expression signature from functionally validated and enriched CD138- clonogenic population from MM cell lines and validated these in patient samples. This data showed that CD138- cells had an enriched expression of genes that are expressed in normal and malignant stem cells. Differentially expressed genes included components of the polycomb repressor complex (PRC) and their targets. Inhibition of PRC by DZNep showed differential effect on CD138- and CD138+ populations. The 'stemness' signature derived from clonogenic CD138- cells overlap significantly with signatures of common progenitor cells, hematopoietic stem cells, and Leukemic stem cells and is associated with poorer survival in different clinical datasets.


Subject(s)
Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , Aldehyde Dehydrogenase/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Clone Cells , Culture Media , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Neoplastic Stem Cells/metabolism , Survival Analysis , Syndecan-1/deficiency , Syndecan-1/metabolism , Tumor Cells, Cultured
6.
Blood ; 121(22): 4512-20, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23529930

ABSTRACT

The role of enhancer of zeste homolog 2 (EZH2) in cancer is complex and may vary depending on the cellular context. We found that EZH2 is aberrantly overexpressed in the majority of natural killer/T-cell lymphoma (NKTL), an aggressive lymphoid malignancy with very poor prognosis. We show that EZH2 upregulation is mediated by MYC-induced repression of its regulatory micro RNAs and EZH2 exerts oncogenic properties in NKTL. Ectopic expression of EZH2 in both primary NK cells and NKTL cell lines leads to a significant growth advantage. Conversely, knock-down of EZH2 in NKTL cell lines results in cell growth inhibition. Intriguingly, ectopic EZH2 mutant deficient for histone methyltransferase activity is also able to confer growth advantage and rescue growth inhibition on endogenous EZH2 depletion in NKTL cells, indicating an oncogenic role of EZH2 independent of its gene-silencing activity. Mechanistically, we show that EZH2 directly promotes the transcription of cyclin D1 and this effect is independent of its enzymatic activity. Furthermore, depletion of EZH2 using a PRC2 inhibitor 3-deazaneplanocin A significantly inhibits growth of NK tumor cells. Therefore, our study uncovers an oncogenic role of EZH2 independent of its methyltransferase activity in NKTL and suggests that targeting EZH2 may have therapeutic usefulness in this lymphoma.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Killer Cells, Natural/physiology , Lymphoma, T-Cell/physiopathology , Polycomb Repressive Complex 2/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation, Neoplastic , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Humans , Killer Cells, Natural/cytology , Lymphoma, T-Cell/pathology , Male , MicroRNAs/metabolism , Middle Aged , Mutagenesis/physiology , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/physiology , Up-Regulation/physiology , Young Adult
7.
Mol Cancer ; 11: 72, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22995644

ABSTRACT

BACKGROUND: Resistance to tyrosine kinase inhibitors (TKIs) remains a challenge in management of patients with chronic myeloid leukemia (CML). A better understanding of the BCR-ABL signalling network may lead to better therapy. FINDINGS: Here we report the discovery of a novel downstream target of BCR-ABL signalling, PRL-3 (PTP4A3), an oncogenic tyrosine phosphatase. Analysis of CML cancer cell lines and CML patient samples reveals the upregulation of PRL-3. Inhibition of BCR-ABL signalling either by Imatinib or by RNAi silencing BCR-ABL reduces PRL-3 and increases cleavage of PARP. In contrast, the amount of PRL-3 protein remains constant or even increased in response to Imatinib treatment in drug resistant cells expressing P210 T315I. Finally, analysis with specific shRNA shows PRL-3 involvement in the proliferation and self-renewal of CML cells. CONCLUSIONS: These data support a role for PRL-3 in BCR-ABL signalling and CML biology and may be a potential therapeutic target downstream of BCR-ABL in TKI resistant mutant cells.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Neoplasm Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression , Gene Expression Regulation, Leukemic , Gene Silencing , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplasm Metastasis/genetics , Neoplasm Proteins/antagonists & inhibitors , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Pyrimidines/pharmacology , STAT Transcription Factors/metabolism , Signal Transduction/drug effects
8.
PLoS One ; 6(6): e21583, 2011.
Article in English | MEDLINE | ID: mdl-21720561

ABSTRACT

The 3-Deazaneplanocin A (DZNep), one of S-adenosylhomocysteine (AdoHcy) hydrolase inhibitors, has shown antitumor activities in a broad range of solid tumors and acute myeloid leukemia. Here, we examined its effects on multiple myeloma (MM) cells and found that, at 500 nM, it potently inhibited growth and induced apoptosis in 2 of 8 MM cell lines. RNA from un-treated and DZNep treated cells was profiled by Affymetrix HG-U133 Plus 2.0 microarray and genes with a significant change in gene expression were determined by significance analysis of microarray (SAM) testing. ALOX5 was the most down-regulated gene (5.8-fold) in sensitive cells and was expressed at low level in resistant cells. The results were corroborated by quantitative RT-PCR. Western-blot analysis indicated ALOX5 was highly expressed only in sensitive cell line H929 and greatly decreased upon DZNep treatment. Ectopic expression of ALOX5 reduced sensitivity to DZNep in H929 cells. Furthermore, down-regulation of ALOX5 by RNA interference could also induce apoptosis in H929. Gene expression analysis on MM patient dataset indicated ALOX5 expression was significantly higher in MM patients compared to normal plasma cells. We also found that Bcl-2 was overexpressed in DZNep insensitive cells, and cotreatment with DZNep and ABT-737, a Bcl-2 family inhibitor, synergistically inhibited growth and induced apoptosis of DZNep insensitive MM cells. Taken together, this study shows one of mechanisms of the DZNep efficacy on MM correlates with its ability to down-regulate the ALOX5 levels. In addition, DZNep insensitivity might be associated with overexpression of Bcl-2, and the combination of ABT-737 and DZNep could synergistically induced apoptosis. These results suggest that DZNep may be exploited therapeutically for a subset of MM.


Subject(s)
Adenosine/analogs & derivatives , Apoptosis/drug effects , Multiple Myeloma/pathology , Adenosine/pharmacology , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Biphenyl Compounds/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Screening Assays, Antitumor , Drug Synergism , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Nitrophenols/pharmacology , Piperazines/pharmacology , Polycomb Repressive Complex 2 , Sulfonamides/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Blood ; 118(10): 2830-9, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21734239

ABSTRACT

Recent studies have shown that 3-Deazaneplanocin A (DZNep), a histone methyltransferase inhibitor, disrupts polycomb-repressive complex 2 (PRC2), and preferentially induces apoptosis in cancer cells, including acute myeloid leukemia (AML). However, the underlying molecular mechanisms are not well understood. The present study demonstrates that DZNep induces robust apoptosis in AML cell lines, primary cells, and targets CD34(+)CD38(-) leukemia stem cell (LSC)-enriched subpopulations. Using RNA interference (RNAi), gene expression profiling, and ChIP, we identified that TXNIP, a major redox control molecule, plays a crucial role in DZNep-induced apoptosis. We show that disruption of PRC2, either by DZNep treatment or EZH2 knockdown, reactivates TXNIP, inhibits thioredoxin activity, and increases reactive oxygen species (ROS), leading to apoptosis. Furthermore, we show that TXNIP is down-regulated in AML and is a direct target of PRC2-mediated gene silencing. Consistent with the ROS accumulation on DZNep treatment, we also see a signature of endoplasmic reticulum (ER) stress-regulated genes, commonly associated with cell survival, down-regulated by DZNep. Taken together, we uncover a novel molecular mechanism of DZNep-mediated apoptosis and propose that EZH2 may be a potential new target for epigenetic treatment in AML.


Subject(s)
Adenosine/analogs & derivatives , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Adenosine/pharmacology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Proliferation , Chromatin Immunoprecipitation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Epigenomics , Female , Gene Expression Profiling , Gene Silencing , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Cells, Cultured
10.
PLoS One ; 6(5): e19798, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21589872

ABSTRACT

Combination with other small molecule drugs represents a promising strategy to improve therapeutic efficacy of FLT3 inhibitors in the clinic. We demonstrated that combining ABT-869, a FLT3 inhibitor, with SAHA, a HDAC inhibitor, led to synergistic killing of the AML cells with FLT3 mutations and suppression of colony formation. We identified a core gene signature that is uniquely induced by the combination treatment in 2 different leukemia cell lines. Among these, we showed that downregulation of PTP4A3 (PRL-3) played a role in this synergism. PRL-3 is downstream of FLT3 signaling and ectopic expression of PRL-3 conferred therapeutic resistance through upregulation of STAT (signal transducers and activators of transcription) pathway activity and anti-apoptotic Mcl-1 protein. PRL-3 interacts with HDAC4 and SAHA downregulates PRL-3 via a proteasome dependent pathway. In addition, PRL-3 protein was identified in 47% of AML cases, but was absent in myeloid cells in normal bone marrows. Our results suggest such combination therapies may significantly improve the therapeutic efficacy of FLT3 inhibitors. PRL-3 plays a potential pathological role in AML and it might be a useful therapeutic target in AML, and warrant clinical investigation.


Subject(s)
Leukemia, Myeloid, Acute/therapy , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Signal Transduction , fms-Like Tyrosine Kinase 3/metabolism , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/pathology , Neoplasm Metastasis
11.
Biotechnol Lett ; 33(2): 221-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20882314

ABSTRACT

PURPOSE OF WORK: mutation of the p53 gene is the most common genetic alteration in human cancers. Our study proposes to rationally design a p53 antisense oligonucleotide (ASO) repository, which contains a series of ASOs containing single nucleotide differences to discriminate between each mutant and wild type (WT) p53. The Sfold software was used to predict target-accessibility and we designed an initial series of antisense oligonucleotides (ASO) that target the p53 mutants A161T, R175H and R249S. Western-blot analysis indicated that ASOs strongly inhibited the expression of p53 mutants in a panel of human tumor cell lines (SNU-449, SK-BR-3 and PLC/PRF/5) while having little effect on the expression of WT p53 (HepG2 cells). In three cancer lines harboring each of the p53 mutations, mutant-specific ASO treatment led to a dose-dependent inhibition of cell growth, cell viability, colony formation and invasion, and expression of mutant p53-dependent survival proteins. Our preliminary results indicate that a single nucleotide difference in ASOs can discriminate between mutant and WT p53. These observations support the hypothesis that a p53 ASO repository can be a potentially valuable tool to knock down oncogenic mutant p53 and warrant the testing of a p53 ASO repository in in vivo settings.


Subject(s)
Antineoplastic Agents/metabolism , Mutant Proteins/antagonists & inhibitors , Mutation, Missense , Oligodeoxyribonucleotides, Antisense/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Mutant Proteins/genetics , Oligodeoxyribonucleotides, Antisense/genetics , Tumor Suppressor Protein p53/genetics
12.
J Proteome Res ; 9(7): 3671-9, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20515073

ABSTRACT

Gastric cancer has one of the highest morbidities and mortalities worldwide. Early detection is key measure to improve the outcome of gastric cancer patients. In our efforts to identify potential markers for gastric cancer detection, we coupled xenotransplantation mouse model with a plasma proteomic approach. MKN45 gastric cancer cells were subcutaneously injected into nude mice and plasma samples from mice bearing different sizes of tumors were collected and subjected to iTRAQ and mass spectrometry analysis. ITIH3 protein was found to be more highly expressed in plasma of tumor bearing mice compared to control. Subsequent screening of ITIH3 expression in 167 clinical plasma samples, including 83 cancer-free subjects and 84 gastric cancer patients, revealed higher ITIH3 level in the plasma of gastric cancer patients. A receiver operating characteristics (ROC) curve estimated a maximal sensitivity of 96% at 66% specificity for ITIH3 in gastric cancer detection. In addition, plasma from early stage gastric cancer patient has significantly (p < 0.001) higher level of ITIH3 compared to that from noncancer subject. Our data suggest that ITIH3 may be a useful biomarker for early detection of gastric cancer.


Subject(s)
Alpha-Globulins/metabolism , Biomarkers, Tumor/metabolism , Proteome/metabolism , Stomach Neoplasms/metabolism , Alpha-Globulins/analysis , Animals , Biomarkers, Tumor/analysis , Blood Proteins/analysis , Blotting, Western , Cell Line, Tumor , Densitometry , Histocytochemistry , Humans , Isotope Labeling , Mass Spectrometry , Mice , Mice, Inbred BALB C , Proteome/analysis , ROC Curve , Reproducibility of Results , Stomach Neoplasms/diagnosis , Transplantation, Heterologous
13.
J Proteomics ; 73(8): 1632-40, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20399916

ABSTRACT

There is no suitable diagnostic and prognostic biomarker for gastric cancer. The biggest hurdles in biomarker discovery are (i) the low abundance of cancer cell-specific proteins that limits their detection and (ii) complex inter-patient variations that complicate the discovery process. To circumvent these issues, we conducted proteomics on the plasma of gastric cancer mouse xenograft and attempted to identify proteins released by cancer cells. MKN45 gastric cancer cells were subcutaneously implanted into immune-incompetent nude mice. Plasma samples collected from mice with different tumor sizes (low, mid and high tumor loads) were subjected to iTRAQ and mass spectrometric analyses. Detection of human APOA1 in mouse plasma was verified and its expression level was shown to be lower in mice with large tumors compared to those with small tumors. Studies on a panel of about 14 gastric cancer cell lines supported the notion that APOA1 in mouse plasma was of human gastric cancer cell origin. While the clinical utility of APOA1 remains to be ascertained with a larger scale study, the current work supported the feasibility of using mouse xenograft model for gastric cancer biomarker discovery.


Subject(s)
Apolipoprotein A-I/blood , Biomarkers, Tumor/blood , Stomach Neoplasms/metabolism , Xenograft Model Antitumor Assays , Amino Acid Sequence , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Proteomics/methods , Sequence Alignment , Stomach Neoplasms/genetics
14.
Blood ; 113(17): 4052-62, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19144991

ABSTRACT

To further investigate potential mechanisms of resistance to FLT3 inhibitors, we developed a resistant cell line by long-term culture of MV4-11 cells with ABT-869, designated as MV4-11-R. Gene profiling reveals up-regulation of FLT3LG (FLT3 ligand) and BIRC5 (survivin), but down-regulation of SOCS1, SOCS2, and SOCS3 in MV4-11-R cells. Hypermethylation of these SOCS genes leads to their transcriptional silencing. Survivin is directly regulated by STAT3. Stimulation of the parental MV4-11 cells with FLT3 ligand increases the expression of survivin and phosphorylated protein STAT1, STAT3, STAT5. Targeting survivin by short-hairpin RNA (shRNA) in MV4-11-R cells induces apoptosis and augments ABT-869-mediated cytotoxicity. Overexpression of survivin protects MV4-11 from apoptosis. Subtoxic dose of indirubin derivative (IDR) E804 resensitizes MV4-11-R to ABT-869 treatment by inhibiting STAT signaling activity and abolishing survivin expression. Combining IDR E804 with ABT-869 shows potent in vivo efficacy in the MV4-11-R xenograft model. Taken together, these results demonstrate that enhanced activation of STAT pathways and overexpression of survivin are important mechanisms of resistance to ABT-869, suggesting that the STAT pathways and survivin could be potential targets for reducing resistance developed in patients receiving FLT3 inhibitors.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Microtubule-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Epoxy Compounds/pharmacology , Female , Humans , Indazoles/pharmacology , Inhibitor of Apoptosis Proteins , Leukemia, Myeloid, Acute/genetics , Ligands , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/genetics , Phenylurea Compounds/pharmacology , STAT3 Transcription Factor/genetics , Sesquiterpenes/pharmacology , Substrate Specificity , Survivin , Up-Regulation/genetics , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...