101
|
Harris PJ, Speranza G, Dansky Ullmann C. Targeting embryonic signaling pathways in cancer therapy. Expert Opin Ther Targets 2012; 16:131-45. [DOI: 10.1517/14728222.2011.645808] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
102
|
Tian W, Han X, Yan M, Xu Y, Duggineni S, Lin N, Luo G, Li YM, Han X, Huang Z, An J. Structure-based discovery of a novel inhibitor targeting the β-catenin/Tcf4 interaction. Biochemistry 2012; 51:724-31. [PMID: 22224445 DOI: 10.1021/bi201428h] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Overactivation or overexpression of β-catenin in the Wnt (wingless) signaling pathway plays an important role in tumorigenesis. Interaction of β-catenin with T-cell factor (Tcf) DNA binding proteins is a key step in the activation of the proliferative genes in response to upstream signals of this Wnt/β-catenin pathway. Recently, we identified a new small molecule inhibitor, named BC21 (C(32)H(36)Cl(2)Cu(2)N(2)O(2)), which effectively inhibits the binding of β-catenin with Tcf4-derived peptide and suppresses β-catenin/Tcf4 driven reporter gene activity. This inhibitor decreases the viability of β-catenin overexpressing HCT116 colon cancer cells that harbor the β-catenin mutation, and more significantly, it inhibits the clonogenic activity of these cells. Down-regulation of c-Myc and cyclin D1 expression, the two important effectors of the Wnt/β-catenin signaling, is confirmed by treating HCT116 cells with BC21. This compound represents a new and modifiable potential anticancer candidate that targets β-catenin/Tcf-4 interaction.
Collapse
Affiliation(s)
- Wang Tian
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Kajiguchi T, Katsumi A, Tanizaki R, Kiyoi H, Naoe T. Y654 of β-catenin is essential for FLT3/ITD-related tyrosine phosphorylation and nuclear localization of β-catenin. Eur J Haematol 2012; 88:314-20. [PMID: 22126602 DOI: 10.1111/j.1600-0609.2011.01738.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
β-Catenin plays a dual role as a key effecter in the regulation of adherens junctions as well as a transcriptional co-activator. Tyrosine phosphorylation of β-catenin affects the cell adhesion, migration, and gene transcription in many types of human cancer cells, including acute myeloid leukemia cells with FLT3 internal tandem duplication (FLT3/ITD-AML). Here, we investigated the relationship between three tyrosine residues (Y86, Y142, and Y654) in β-catenin and oncogenic FLT3/ITD kinase. In the experiments using COS-7 cells expressing FLT3/ITD and Wt or mutant β-catenin, FLT3/ITD phosphorylated Y654, and this residue was essential for β-catenin's nuclear localization by FLT3/ITD. Promoter-reporter assays demonstrated that Y654 phosphorylation of β-catenin was closely related to TCF transcriptional activity. In vitro kinase assays, using recombinant FLT3 and biotinylated β-catenin peptide including Y654 showed that FLT3 directly phosphorylated Y654 of β-catenin. These results explain how FLT3/ITD affects the tyrosine phosphorylation, nuclear localization, and transcriptional activity of β-catenin. Targeting Y654 phosphorylation may lead to the development of novel approaches to therapy for FLT3/ITD-AML.
Collapse
Affiliation(s)
- Tomohiro Kajiguchi
- Department of Infectious Diseases, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya,
| | | | | | | | | |
Collapse
|
104
|
Fan R, Zhang LY, Wang H, Yang B, Han T, Zhao XL, Wang W, Wang XQ, Lin GW. Methylation of the CpG Island Near SOX7 Gene Promoter Is Correlated with the Poor Prognosis of Patients with Myelodysplastic Syndrome. TOHOKU J EXP MED 2012; 227:119-128. [DOI: 10.1620/tjem.227.119] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Affiliation(s)
- Rong Fan
- Department of Hematology, Huashan Hospital of Fudan University
| | | | - Hong Wang
- Department of Hematology, The First Affiliated Hospital of Soochow University
| | - Bo Yang
- Fudan-Cinpathogen Clinical & Molecular Research Center
| | - Tao Han
- Fudan-Cinpathogen Clinical & Molecular Research Center
| | - Xiao-Li Zhao
- Department of Hematology, Huashan Hospital of Fudan University
| | - Wei Wang
- Department of Hematology, Huashan Hospital of Fudan University
| | - Xiao-Qin Wang
- Department of Hematology, Huashan Hospital of Fudan University
| | - Guo-Wei Lin
- Department of Hematology, Huashan Hospital of Fudan University
| |
Collapse
|
105
|
Overexpression of LEF1 predicts unfavorable outcome in adult patients with B-precursor acute lymphoblastic leukemia. Blood 2011; 118:6362-7. [DOI: 10.1182/blood-2011-04-350850] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Abstract
Aberrant activation of the Wnt pathway plays a pathogenetic role in various tumors and has been associated with adverse outcome in acute lymphoblastic leukemia (ALL). LEF1, a key mediator of Wnt signaling, has been linked to leukemic transformation, and recurrent mutations of LEF1 have been identified in pediatric T-ALL. Here we evaluated the prognostic significance of LEF1 expression in B-precursor ALL patients. LEF1 expression was determined by quantitative real-time RT-PCR in 282 adult B-precursor ALL patients treated on 06/99 and 07/03 GMALL trials. Patients were grouped into quartiles (Q1-Q4) according to LEF1 expression levels (LEF1 high, Q4; n = 71; LEF1 low, Q1-Q3; n = 211). Patients with high LEF1 expression had a significantly shorter relapse-free survival (RFS) compared with low LEF1 expressers (5-year RFS: LEF1 high, 27%; LEF1 low, 47%; P = .05). Importantly, high LEF1 expression was also associated with inferior RFS in standard-risk patients and was independently predictive for RFS (P = .02) in multivariate analyses for this subgroup. Thus, high LEF1 expression identifies B-precursor ALL patients with inferior RFS, supporting a pathogenetic role of Wnt signaling in ALL. Standard-risk patients with high LEF1 expression might benefit from early treatment modifications and new molecular therapies, including agents targeting the Wnt pathway.
Collapse
|
106
|
Jiang J, Griffin JD. Wnt/β-catenin Pathway Modulates the Sensitivity of the Mutant FLT3 Receptor Kinase Inhibitors in a GSK-3β Dependent Manner. Genes Cancer 2011; 1:164-76. [PMID: 21779446 DOI: 10.1177/1947601910362446] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The FLT3 tyrosine kinase receptor is involved in both hematopoiesis and hematological malignancies. The Wnt/β-catenin pathway has been shown to participate in the self-renewal of hematopoietic stem cells and to cooperate with the mutant FLT3 receptors in leukemic transformation. However, the detailed biological impact of such a constitutively activated Wnt pathway remains to be further explored. Here, the authors report that activating mutations of FLT3 constitutively activate β-catenin by inhibition of GSK-3β in a PI3 kinase pathway-dependent manner. Ectopic expression of a dominant negative form of GSK-3β in FLT3-ITD-expressing cells activated β-catenin and blocked the downregulation of the TCF/β-catenin transcriptional activity induced by inhibition of FLT3 kinase. Furthermore, inhibition of cell proliferation and colony formation induced by such suppression of FLT3 kinase activity could be partially reversed by knockdown of GSK-3β and restored by knockdown of either TCF4 or β-catenin. Moreover, exogenous activation of the Wnt pathway also attenuated such inhibitory effect. These findings indicate that the potencies of the inhibitors of FLT3 kinase activity could be modulated by the activity of the Wnt/β-catenin pathway in the cells harboring FLT3-ITD mutations, and FLT3-ITDs signal through GSK-3β to activate β-catenin that this is likely to directly contribute to the leukemic phenotype.
Collapse
Affiliation(s)
- Jingrui Jiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | |
Collapse
|
107
|
Zheng F, Li H, Du W, Huang S. Role of hERG1 K(+) channels in leukemia cells as a positive regulator in SDF-1a-induced proliferation. ACTA ACUST UNITED AC 2011; 16:177-84. [PMID: 21669058 DOI: 10.1179/102453311x12940641878000] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Previous work from our laboratory has confirmed that human ether-à-go-go-related gene 1 (hERG1) K(+) channels are constitutively expressed in leukemia cells and enhanced cell proliferation. More importantly, it has shown that stromal cell-derived factor-1a (SDF-1a) significantly increases hERG1 K(+) tail current and a specific hERG1 K(+) channels inhibitor significantly blocks SDF-1a-induced migration of leukemic cells. In this study, we investigated a possible regulatory effect of hERG1 K(+) channels upon SDF-1a-mediated cell proliferation as a mean to uncover new molecular events involved in bone marrow microenvironment and leukemogenesis. RT-PCR showed that SDF-1a enhanced hERG1 expression in a dose-dependent manner. Cell proliferation assay illustrated that SDF-1a promoted cell proliferation in a dose-dependent manner, whereas this effect was impaired by E-4031. In addition, E-4031 inhibited SDF-1a-stimulated leukemic cell proliferation by inducing G(0)/G(1) arrest. Interestingly, E-4031 promoted SDF-1a-induced apoptosis in HL-60 and leukemic blasts, which markedly impaired the protection effect of SDF-1a in AML. Moreover, SDF-1a increased the expression of Wnt/beta-catenin target genes, including beta-catenin, cyclin-D1, and c-myc; however, this manner was abolished by blockage with the hERG1 K(+) channels. Taken together, our results provide evidence of a novel mechanism involved in the proliferative effects of SDF-1a and highlight hERG1 K(+) channels as a therapeutic target for leukemia treatment and prevention.
Collapse
Affiliation(s)
- Fang Zheng
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | |
Collapse
|
108
|
Abstract
Most adult patients with acute myeloid leukemia (AML) die from their disease. Relapses are frequent even after aggressive multiagent chemotherapy and allogeneic stem cell transplantation. AML is a biologically heterogeneous disease, characterized by frequent cytogenetic abnormalities and an increasing spectrum of genetic mutations and molecular aberrations. Laboratory data suggest that AML originates from a rare population of cells, termed leukemic stem cells (LSCs) or leukemia-initiating cells, which are capable of self-renewal, proliferation and differentiation. These cells may persist after treatment and are probably responsible for disease relapse. This review will describe bench and translational research in LSCs and discuss how the data should be used to change the direction of developmental therapeutics and clinical trials in AML.
Collapse
Affiliation(s)
- Gail J Roboz
- Weill Medical College of Cornell University, The New York Presbyterian Hospital, 520 East 70th Street, New York, NY 10021, USA.
| | | |
Collapse
|
109
|
Wu X, Gao H, Ke W, Hager M, Xiao S, Freeman MR, Zhu Z. VentX trans-activates p53 and p16ink4a to regulate cellular senescence. J Biol Chem 2011; 286:12693-701. [PMID: 21325273 DOI: 10.1074/jbc.m110.206078] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cell senescence is a process of irreversible arrest of cell proliferation and plays an important role in tumor suppression. Recent studies showed that Wnt inhibition is a trigger of cellular senescence. Using methods of reverse genetics, we recently identified VentX, a human homolog of the vertebrate Xenopus Vent family of homeobox genes, as a novel Wnt repressor and a putative tumor suppressor in lymphocytic leukemia. Here, we show that VentX is a direct transcriptional activator of p53-p21 and p16ink4a-Rb tumor suppression pathways. Ectopic expression of VentX in cancer cells caused an irreversible cell cycle arrest with a typical senescence-like phenotype. Conversely, inhibition of VentX expression by RNA interference ameliorated chemotherapeutic agent-induced senescence in lymphocytic leukemia cells. The results of our study further reveal the mechanisms underlying tumor suppression function of VentX and suggest a role of VentX as a potential target in cancer prevention and treatment.
Collapse
Affiliation(s)
- Xiaoming Wu
- Gastroenterology Division, the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
110
|
Liu YC, Lai WC, Chuang KA, Shen YJ, Hu WS, Ho CH, Chen YB, Hsu MF, Hsu HC, Lieu CH. Blockade of JAK2 activity suppressed accumulation of β-catenin in leukemic cells. J Cell Biochem 2011; 111:402-11. [PMID: 20503246 DOI: 10.1002/jcb.22714] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The Wnt/β-catenin pathway has been implicated in leukemogenesis. We found β-catenin abnormally accumulated in both human acute T cell leukemia Jurkat cells and human erythroleukemia HEL cells. β-Catenin can be significantly down-regulated by the Janus kinase 2 specific inhibitor AG490 in these two cells. AG490 also reduces the luciferase activity of a reporter plasmid driven by LEF/β-catenin promoter. Similar results were observed in HEL cells infected with lentivirus containing shRNA against JAK2 gene. After treatment with 50 µM AG490 or shRNA, the mRNA expression levels of β-catenin, APC, Axin, β-Trcp, GSK3α, and GSK3β were up-regulated within 12-16 h. However, only the protein levels of GSK3β and β-Trcp were found to have increased relative to untreated cells. Knockdown experiments revealed that the AG490-induced inhibition of β-catenin can be attenuated by shRNA targeting β-TrCP. Taken together; these results suggest that β-Trcp plays a key role in the cross-talk between JAK/STAT and Wnt/β-catenin signaling in leukemia cells.
Collapse
Affiliation(s)
- Ya-Chen Liu
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Lyssiotis CA, Lairson LL, Boitano AE, Wurdak H, Zhu S, Schultz PG. Chemical Control of Stem Cell Fate and Developmental Potential. Angew Chem Int Ed Engl 2010; 50:200-42. [DOI: 10.1002/anie.201004284] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Costas A. Lyssiotis
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Luke L. Lairson
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Anthony E. Boitano
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Heiko Wurdak
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Shoutian Zhu
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Peter G. Schultz
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| |
Collapse
|
112
|
Lyssiotis CA, Lairson LL, Boitano AE, Wurdak H, Zhu S, Schultz PG. Chemische Kontrolle des Schicksals und Entwicklungspotenzials von Stammzellen. Angew Chem Int Ed Engl 2010. [DOI: 10.1002/ange.201004284] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Costas A. Lyssiotis
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Luke L. Lairson
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Anthony E. Boitano
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Heiko Wurdak
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Shoutian Zhu
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Peter G. Schultz
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| |
Collapse
|
113
|
Griffiths EA, Gore SD, Hooker C, McDevitt MA, Karp JE, Smith BD, Mohammad HP, Ye Y, Herman JG, Carraway HE. Acute myeloid leukemia is characterized by Wnt pathway inhibitor promoter hypermethylation. Leuk Lymphoma 2010; 51:1711-9. [PMID: 20795789 DOI: 10.3109/10428194.2010.496505] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Nuclear localization of non-phosphorylated, active beta-catenin is a measure of Wnt pathway activation and is associated with adverse outcome in patients with acute myeloid leukemia (AML). While genetic alterations of the Wnt pathway are infrequent in AML, inhibitors of this pathway are silenced by promoter methylation in other malignanices. Leukemia cell lines were examined for Wnt pathway inhibitor methylation and total beta-catenin levels, and had frequent methylation of Wnt inhibitors and upregulated beta-catenin by Western blot and immunofluorescence. One hundred sixty-nine AML samples were examined for methylation of Wnt inhibitor genes. Diagnostic samples from 72 patients with normal cytogenetics who received standard high-dose induction chemotherapy were evaluated for associations between methylation and event-free or overall survival. Extensive methylation of Wnt pathway inhibitor genes was observed in cell lines, and 89% of primary AML samples had at least one methylated gene: DKK1 (16%), DKK3 (8%), RUNX3 (27%), sFRP1 (34%), sFRP2 (66%), sFRP4 (9%), sFRP5 (54%), SOX17 (29%), and WIF1 (32%). In contrast to epithelial tumors, methylation of APC (2%) and RASSF1A (0%) was rare. In patients with AML with normal cytogenetics, sFRP2 and sFRP5 methylation at the time of diagnosis was associated with an increased risk of relapse, and sFRP2 methylation was associated with an increased risk for death. In patients with AML: (a) there is a high frequency of Wnt pathway inhibitor methylation; (b) Wnt pathway inhibitor methylation is distinct from that observed in epithelial malignancies; and (c) methylation of sFRP2 and sFRP5 may predict adverse clinical outcome in patients with normal karyotype AML.
Collapse
Affiliation(s)
- Elizabeth A Griffiths
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Siapati EK, Papadaki M, Kozaou Z, Rouka E, Michali E, Savvidou I, Gogos D, Kyriakou D, Anagnostopoulos NI, Vassilopoulos G. Proliferation and bone marrow engraftment of AML blasts is dependent on β-catenin signalling. Br J Haematol 2010; 152:164-74. [PMID: 21118196 DOI: 10.1111/j.1365-2141.2010.08471.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
B-catenin is the central effector molecule of the canonical Wnt signalling pathway, which controls self-renewal of haematopoietic stem cells. Deregulation of this pathway occurs in various malignancies including myeloid leukaemias. The present study examined the functional outcome of stable β-catenin down-regulation through lentivirus-mediated expression of short hairpin RNA (shRNA). Reduction of the β-catenin levels in acute myeloid leukaemia (AML) cell lines and patient samples decelerated their in vitro proliferation ability without affecting cell viability. Transplantation of leukaemic cells with control or reduced levels of β-catenin in non-obese diabetic severe combined immunodeficient animals indicated that, while the immediate homing of the cells was unaffected, the bone marrow engraftment was directly dependent on β-catenin levels. Subsequent examination of bone sections revealed that β-catenin was implicated in the localization of AML to the endosteum. Examination of adhesion molecule expression before and after transplantation, revealed down-regulation of CD44 expression, accompanied by reduced in vitro adhesion. Gene expression analysis disclosed the presence of an autocrine compensatory mechanism, which responds to the reduced β-catenin levels by altering the expression of positive and negative pathway regulators. In conclusion, our study showed that β-catenin comprises an integral part of AML cell proliferation, cell cycle progression, and adhesion, and influences disease establishment in vivo.
Collapse
Affiliation(s)
- Elena K Siapati
- Biomedical Research Foundation of the Academy of Athens, Greece.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Chuang KA, Lieu CH, Tsai WJ, Wu MH, Chen YC, Liao JF, Wang CC, Kuo YC. Evaluation of anti-Wnt/β-catenin signaling agents by pGL4-TOP transfected stable cells with a luciferase reporter system. Braz J Med Biol Res 2010; 43:931-41. [PMID: 20835687 DOI: 10.1590/s0100-879x2010007500091] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Accepted: 08/19/2010] [Indexed: 01/05/2023] Open
Abstract
Refractory and relapsed leukemia is a major problem during cancer therapy, which is due to the aberrant activation of Wnt/β-catenin signaling pathway. Activation of this pathway is promoted by wingless (Wnt) proteins and induces co-activator β-catenin binding to lymphoid enhancer factor (LEF)/T-cell factor protein (TCF). To provide a convenient system for the screening of anti-Wnt/β-catenin agents, we designed a bi-functional pGL4-TOP reporter plasmid that contained 3X β-catenin/LEF/TCF binding sites and a selectable marker. After transfection and hygromycin B selection, HEK 293-TOP and Jurkat-TOP stable clones were established. The luciferase activity in the stable clone was enhanced by the recombinant Wnt-3A (rWnt-3A; 100-400 ng/mL) and GSK3β inhibitor (2'Z,3'E)-6-bromoindirubin-3'-oxime (BIO; 5 µM) but was inhibited by aspirin (5 mM). Using this reporter model, we found that norcantharidin (NCTD; 100 µM) reduced 80% of rWnt-3A-induced luciferase activity. Furthermore, 50 µM NCTD inhibited 38% of BIO-induced luciferase activity in Jurkat-TOP stable cells. Employing ³H-thymidine uptake assay and Western blot analysis, we confirmed that NCTD (50 µM) significantly inhibited proliferation of Jurkat cells by 64%, which are the dominant β-catenin signaling cells and decreased β-catenin protein in a concentration-dependent manner. Thus, we established a stable HEK 293-TOP clone and successfully used it to identify the Wnt/β-catenin signaling inhibitor NCTD.
Collapse
Affiliation(s)
- K A Chuang
- Department of Biotechnology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Transactivation of lifeguard (LFG) by Akt-/LEF-1 pathway in MCF-7 and MDA-MB 231 human breast cancer cells. Apoptosis 2010; 15:814-21. [PMID: 20336373 DOI: 10.1007/s10495-010-0493-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lifeguard (LFG) has been identified as a molecule that uniquely inhibits death mediated by Fas. The molecular function of human LFG and its regulation in carcinogenesis is uncertain. In our study, we investigated the potential regulation of LFG expression by Akt/LEF-1 pathway. The Glycogen synthase kinase-3 (GSK3) can be regulated by different signaling pathways including those mediated by protein kinase Akt. Inhibition of GSK3beta subunits activity results in the stabilisation of the beta-catenin protein and its accumulation in the nucleus, where it associates with members of the TCF/LEF-1 family of transcription factors to mediate gene transcription. In Western blots, RT-PCR and by small interfering RNA directed against LEF-1, we demonstrated that LFG expression correlates with GSK3beta and LEF-1 activation. Moreover, we showed that LFG mRNA was down-regulated after transfection with siRNA against LEF-1 in MDA-MB-231 cells. Our results therefore identify LFG as a target of the Akt/LEF-1 pathway in MDA-MB-231 breast tumour cells, a regulation which could play a key role in breast tumour progression.
Collapse
|
117
|
Reins J, Mossner M, Neumann M, Platzbecker U, Schumann C, Thiel E, Hofmann WK. Transcriptional down-regulation of the Wnt antagonist SFRP1 in haematopoietic cells of patients with different risk types of MDS. Leuk Res 2010; 34:1610-6. [PMID: 20471677 DOI: 10.1016/j.leukres.2010.04.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 03/27/2010] [Accepted: 04/21/2010] [Indexed: 10/19/2022]
Abstract
Secreted frizzled related protein 1 (SFRP1) is an extracellular antagonist of the Wnt signalling pathway that plays an important role in the pathogenesis of solid tumours and haematopoietic malignancies. SFRP1 has been observed to be transcriptionally down-regulated due to hypermethylation in acute and chronic leukaemia, but so far not in myelodysplastic syndrome (MDS). Moreover, it has been shown that the epigenetic inactivation of SFRP1 correlates with an overexpression of the Wnt receptor Frizzled 3 (Fzd3) in acute leukaemia. Using real-time quantitative reverse transcription polymerase chain reaction (RT-PCR) we examined mRNA expression of SFRP1 and Fzd3 in bone marrow cells derived from 121 patients with different risk types of MDS, acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL). We employed pyrosequencing to quantify promoter DNA methylation in MDS and acute leukaemia. We detected significant lower mRNA transcription of SFRP1 in MDS compared to healthy individuals. However, DNA sequence mutations or frequent elevated DNA methylation levels of the SFRP1 promoter could not be observed in MDS but in AML and ALL as previously reported. The expression levels of Fzd3 were up-regulated in both acute leukaemia and MDS. Our data show a significant transcriptional down-regulation of SFRP1 as a common event in AML, ALL and - as demonstrated for the first time - in MDS. An inactivation of SFRP1 and the transcriptional up-regulation of Fzd3 seem to be associated with an activation of the Wnt signalling pathway in these haematopoietic diseases.
Collapse
MESH Headings
- Acute Disease
- Chronic Disease
- DNA Methylation
- Down-Regulation
- Female
- Frizzled Receptors/metabolism
- Gene Expression Regulation
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Intercellular Signaling Peptides and Proteins/biosynthesis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Membrane Proteins/biosynthesis
- Myelodysplastic Syndromes/metabolism
- Myelodysplastic Syndromes/pathology
- Promoter Regions, Genetic
- Receptors, G-Protein-Coupled/metabolism
- Risk Factors
- Transcription, Genetic
- Wnt Proteins/antagonists & inhibitors
Collapse
Affiliation(s)
- Jana Reins
- Department of Hematology and Oncology, Charité Campus Benjamin Franklin, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
118
|
Liu X, Wang L, Zhao S, Ji X, Luo Y, Ling F. β-Catenin overexpression in malignant glioma and its role in proliferation and apoptosis in glioblastma cells. Med Oncol 2010; 28:608-14. [PMID: 20300972 DOI: 10.1007/s12032-010-9476-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Accepted: 02/25/2010] [Indexed: 11/30/2022]
Abstract
β-Catenin, a core component of Wnt/β-catenin signaling, has been shown to be a crucial factor in a broad range of tumors, while its role in glioma is not well understood. In this study, the expression of β-catenin in astrocytic glioma tissues with different grade and human normal cerebral tissues was examined using reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. We found a higher expression level of β-catenin in astrocytic glioma patients with high grade in comparison with the normal controls. Additionally, siRNA was transfected into human U251 glioblastoma cells by liposome after the design of siRNA was confirmed to effectively inhibit the expression of β-catenin by RT-PCR. Compared to the control siRNA group, siRNA-mediated knockdown of β-catenin in human U251 cells inhibited cell proliferation, resulted in cell apoptosis, and arrested cell cycle in G₀/G₁. Additionally, downregulation of β-catenin decreased the expression level of cyclin D1, c-Myc and c-jun. Taken together, these results indicate that overexpression of β-catenin may be an important contributing factor to glioma progression.
Collapse
Affiliation(s)
- Xiangrong Liu
- Cerebrovascular Diseases Research Institute, Key Laboratory of Neurodegenerative Diseases of Ministry of Education and Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 100053, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
119
|
Martín V, Valencia A, Agirre X, Cervera J, San Jose-Eneriz E, Vilas-Zornoza A, Rodriguez-Otero P, Sanz MA, Herrera C, Torres A, Prosper F, Román-Gómez J. Epigenetic regulation of the non-canonical Wnt pathway in acute myeloid leukemia. Cancer Sci 2010; 101:425-32. [PMID: 19874313 PMCID: PMC11159284 DOI: 10.1111/j.1349-7006.2009.01413.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024] Open
Abstract
Wnt5a is a member of the Wnt family of proteins that signals through the non-canonical Wnt/Ca(2+)pathway to suppress cyclin D1. Deregulation of this pathway has been found in animal models suggesting that it acts as tumour suppressor in acute myeloid leukemia (AML). Although DNA methylation is the main mechanism of regulation of the canonical Wnt pathway in AML, the role of WNT5A abnormalities has never been evaluated in this clinical setting. The methylation status of WNT5A promoter-exon 1 was analyzed by methylation-specific PCR and sequencing in eleven AML-derived cell lines and 252 AML patients. We observed WNT5A hypermethylation in seven cell lines and in 43% (107/252) of AML patients. WNT5A methylation was associated with decreased WNT5A expression (P < 0.001) that was restored after exposure to 5-Aza-2'-deoxycytidine. Moreover, WNT5A hypermethylation correlated with upregulation of CYCLIN D1 expression (P < 0.001). Relapse (15%vs 37%, P < 0.001) and mortality (61%vs 79%, P = 0.004) rates were lower for patients in the non-methylated group. Disease-free survival and overall survival at 6 and 7 years, respectively, were 60% and 27% for unmethylated patients and 20% and 0% for hypermethylated patients (P = 0.0001 and P = 0.04, respectively). Interestingly, significant differences were also observed when the analysis was carried out according to cytogenetic risk groups. We demonstrate that WNT5A, a putative tumor suppressor gene in AML, is silenced by methylation in this disease and that this epigenetic event is associated with upregulation of CYCLIN D1 expression and confers poor prognosis in patients with AML.
Collapse
Affiliation(s)
- Vanesa Martín
- Hematology Department, Cellular Therapy Area, Reina Sofia Hospital, Maimonides Institute for Biomedical Research, Cordoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Jesse S, Koenig A, Ellenrieder V, Menke A. Lef-1 isoforms regulate different target genes and reduce cellular adhesion. Int J Cancer 2010; 126:1109-20. [PMID: 19653274 DOI: 10.1002/ijc.24802] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The lymphoid enhancer factor 1 (Lef-1) belongs to the nuclear transducers of canonical Wnt-signalling in embryogenesis and cancer. Lef-1 acts, in cooperation with beta-catenin, as a context-dependent transcriptional activator or repressor, thereby influencing multiple cellular functions such as proliferation, differentiation and migration. Here we report that an increased Lef-1 expression in human pancreatic cancer correlates with advanced tumour stages. In pancreatic tumours, two different transcripts of Lef-1 have been detected in various stages, as demonstrated by RT-PCR analysis. One transcript was identified as the full length Lef-1 (Lef-1 FL), whereas the second, shorter transcript lacked exon VI (Lef-1 Deltaexon VI) compared to the published sequence. Comparative analysis of these two Lef-1 variants revealed that they exhibit different cellular effects after transient expression in pancreatic carcinoma cells. Forced expression of Lef-1 Deltaexon VI inhibited E-cadherin expression in a beta-catenin-independent way. Increased amounts of Lef-1 Deltaexon VI resulted in reduced cellular aggregation and increased cell migration. Expression of Lef-1 FL, but not the newly identified Lef-1 Deltaexon VI, induced the expression of the cell cycle regulating proteins c-myc and cyclin D1 in cooperation with beta-catenin and it enhanced cell proliferation. Our findings indicate that expression of alternatively spliced Lef-1 isoforms is involved in the determination of proliferative or migratory characteristics of pancreatic carcinoma cells.
Collapse
Affiliation(s)
- Sarah Jesse
- Department of Internal Medicine I, University of Ulm, D-89081 Ulm, Germany
| | | | | | | |
Collapse
|
121
|
Gao H, Le Y, Wu X, Silberstein LE, Giese RW, Zhu Z. VentX, a novel lymphoid-enhancing factor/T-cell factor-associated transcription repressor, is a putative tumor suppressor. Cancer Res 2009; 70:202-11. [PMID: 20028861 DOI: 10.1158/0008-5472.can-09-2668] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lymphoid-enhancing factor/T-cell factors (LEF1/TCF) are a high-mobility group of transcriptional factors that play essential roles in cell fate determination during early embryogenesis and ontogenesis. Aberrant activations of LEF1/TCF-mediated transcription have been implicated in a variety of malignancies. Our recent studies on vertebrate embryogenesis identified Xom, a homeobox protein of the bone morphogenetic protein 4 pathway, as a novel LEF/TCF-associated transcriptional modulator. Here, we report that VentX, a human Xom homologue, is a LEF/TCF-associated inhibitor of canonical Wnt/beta-catenin signaling and a negative regulator of cell proliferation. VentX is predominantly expressed in hematopoietic cells, and its expression is significantly downregulated in chronic lymphocytic leukemia. Altered expression of VentX is associated with corresponding changes of LEF/TCF target oncogenes such as cyclin D1, suggesting a potential role of VentX in the clinical behavior of hematopoietic malignancies.
Collapse
Affiliation(s)
- Hong Gao
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
122
|
Foss B, Tronstad KJ, Bruserud Ø. Connexin-based signaling in acute myelogenous leukemia (AML). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:1-8. [PMID: 19883623 DOI: 10.1016/j.bbamem.2009.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 09/15/2009] [Accepted: 10/26/2009] [Indexed: 10/20/2022]
Abstract
Normal and malignant hematopoiesis are regulated by intercellular communication in the hematopoietic microenvironments, and both soluble mediators as well as direct cell-cell contact play important functional roles. Gap junctions are complex membrane structures that transfer molecules between neighboring cells and thereby alter intracellular signaling and metabolism. The gap junction building blocks, the connexins, are also involved in gap junction-independent intercellular communication by forming hemichannels that transfer substances between the intra- and extracellular spaces. Connexins are furthermore involved in cell regulation as single molecules by modulating intracellular pathways and possibly gene transcription. The role of connexins in leukemogenesis and leukemic cell functions are not well characterized. In this review, we describe the known effects of gap junctions and connexins in acute myelogenous leukemia and the diverse potential of connexins in acute myelogenous leukemia chemosensitivity, intracellular signaling and cell death regulation.
Collapse
Affiliation(s)
- Brynjar Foss
- Department of Health Studies, University of Stavanger, Stavanger, Norway.
| | | | | |
Collapse
|
123
|
Targeting the WNT/beta-catenin/TCF/LEF1 axis in solid and haematological cancers: Multiplicity of therapeutic options. Eur J Cancer 2009; 45:2759-67. [PMID: 19729298 DOI: 10.1016/j.ejca.2009.08.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 07/30/2009] [Accepted: 08/04/2009] [Indexed: 11/21/2022]
Abstract
Among aberrantly regulated signalling pathways in cancer the WNT/beta-catenin pathway plays an outstanding role, since it was shown to be critically involved in a wide range of neoplasias. While the underlying mechanisms vary, overexpression of WNTs was found to mediate active signalling in some of these diseases. Other cancers show a mutation in pathway members further downstream, such as APC, Axin or beta-catenin, leading to aberrant signalling activation. Another mechanism initiating activation of WNT/beta-catenin signalling is the silencing of expression of negative WNT/beta-catenin regulators, such as DKK and WIF1, by, for example, promoter hypermethylation. All these mechanisms result in a common consequence, the activation of TCF/LEF1 transcription factors and subsequent target gene expression. Several target genes are known to be key players in tumourigenesis, such as c-myc, cyclin D1 or survivin. The variety of possible underlying mechanisms leading to beta-catenin/TCF/LEF1 activation offers multiple options to target the aberrantly activated pathway in order to prevent target gene expression and/or their gene products to exert their tumourigenic function. Here, we summarise the physiological role of WNT/beta-catenin signalling and the consequences of its aberrant activation during tumourigenesis. Furthermore, we discuss the possible strategies to target this pathway and their potential importance in cancer treatment.
Collapse
|
124
|
Nteliopoulos G, Marley SB, Gordon MY. Influence of PI-3K/Akt pathway on Wnt signalling in regulating myeloid progenitor cell proliferation. Evidence for a role of autocrine/paracrine Wnt regulation. Br J Haematol 2009; 146:637-51. [DOI: 10.1111/j.1365-2141.2009.07823.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
125
|
Wang Z, Havasi A, Gall JM, Mao H, Schwartz JH, Borkan SC. Beta-catenin promotes survival of renal epithelial cells by inhibiting Bax. J Am Soc Nephrol 2009; 20:1919-28. [PMID: 19696224 DOI: 10.1681/asn.2009030253] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Ischemia activates Bax, a proapoptotic BCL2 protein, as well as the prosurvival beta-catenin/Wnt signaling pathway. To test the hypothesis that beta-catenin/Wnt signaling regulates Bax-mediated apoptosis after induction of metabolic stress, which occurs during renal ischemia, we infected immortalized and primary proximal tubular epithelial cells with adenovirus to express either constitutively active or dominant negative beta-catenin constructs. Constitutively active beta-catenin significantly decreased apoptosis and improved cell survival after metabolic stress. Furthermore, active beta-catenin decreased Bax activation, oligomerization, and translocation to mitochondria, and reduced both organelle membrane injury and apoptosis. Dominant negative beta-catenin had the opposite effects. Because Akt regulates Bax, we examined the effects of the beta-catenin mutants on Akt expression and activation. Constitutively active beta-catenin increased Akt-1 expression and activation before and after stress, and treatment with a phosphatidylinositol-3 kinase inhibitor antagonized the protective effects of beta-catenin on Akt activation, Bax inhibition, and cell survival. In addition, beta-catenin significantly increased the rate of phosphorylation at Bax serine(184), an Akt-specific target. Taken together, these results suggest that beta-catenin/Wnt signaling promotes survival of renal epithelial cells after metabolic stress, in part by inhibiting Bax in a phosphatidylinositol-3 kinase/Akt-dependent manner.
Collapse
Affiliation(s)
- Zhiyong Wang
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
126
|
Masson K, Rönnstrand L. Oncogenic signaling from the hematopoietic growth factor receptors c-Kit and Flt3. Cell Signal 2009; 21:1717-26. [PMID: 19540337 DOI: 10.1016/j.cellsig.2009.06.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 06/04/2009] [Accepted: 06/09/2009] [Indexed: 01/01/2023]
Abstract
Signal transduction in response to growth factors is a strictly controlled process with networks of feedback systems, highly selective interactions and finely tuned on-and-off switches. In the context of cancer, detailed signaling studies have resulted in the development of some of the most frequently used means of therapy, with several well established examples such as the small molecule inhibitors imatinib and dasatinib in the treatment of chronic myeloid leukemia. Impaired function of receptor tyrosine kinases is implicated in various types of tumors, and much effort is put into mapping the many interactions and downstream pathways. Here we discuss the hematopoietic growth factor receptors c-Kit and Flt3 and their downstream signaling in normal as well as malignant cells. Both receptors are members of the same family of tyrosine kinases and crucial mediators of stem-and progenitor-cell proliferation and survival in response to ligand stimuli from the surrounding microenvironment. Gain-of-function mutations/alterations render the receptors constitutively and ligand-independently activated, resulting in aberrant signaling which is a crucial driving force in tumorigenesis. Frequently found mutations in c-Kit and Flt3 are point mutations of aspartic acid 816 and 835 respectively, in the activation loop of the kinase domains. Several other point mutations have been identified, but in the case of Flt3, the most common alterations are internal tandem duplications (ITDs) in the juxtamembrane region, reported in approximately 30% of patients with acute myeloid leukemia (AML). During the last couple of years, the increasing understanding of c-Kit and Flt3 signaling has also revealed the complexity of these receptor systems. The impact of gain-of-function mutations of c-Kit and Flt3 in different malignancies is well established and shown to be of clinical relevance in both prognosis and therapy. Many inhibitors of both c-Kit or Flt3 or of their downstream substrates are in clinical trials with encouraging results, and targeted therapy using a combination of such inhibitors is considered a promising approach for future treatments.
Collapse
Affiliation(s)
- Kristina Masson
- Experimental Clinical Chemistry, Wallenberg Laboratory, Department of Laboratory Medicine, Malmö University Hospital, Lund University, 20502 Malmö, Sweden
| | | |
Collapse
|
127
|
Valencia A, Román-Gómez J, Cervera J, Such E, Barragán E, Bolufer P, Moscardó F, Sanz GF, Sanz MA. Wnt signaling pathway is epigenetically regulated by methylation of Wnt antagonists in acute myeloid leukemia. Leukemia 2009; 23:1658-66. [PMID: 19387464 DOI: 10.1038/leu.2009.86] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Activation of the Wnt signaling pathway has been implicated recently in the pathogenesis of leukemia. We studied the function of epigenetic regulation of the Wnt pathway and its prognostic relevance in acute myelogenous leukemia (AML). We used a methylation-specific polymerase chain reaction approach to analyze the promoter methylation status of a panel of Wnt antagonists including sFRP1, sFRP2, sFRP4, sFRP5, DKK1 and DKK3. Aberrant methylation of Wnt antagonists was detected in four AML cell lines and in up to 64% of AML marrow samples. Treatment of the cell lines with 5-aza-2'-deoxycytidine induced reexpression of methylated Wnt antagonists and inactivation of the Wnt pathway by downregulating the Wnt pathway genes cyclin D1, TCF1 and LEF1 and reducing nuclear localization of beta-catenin. In a subgroup of patients 60 years and younger with newly diagnosed AML and intermediate-risk cytogenetics, abnormal methylation of Wnt antagonists was associated with decreased 4-year relapse-free survival (28 vs 61%, respectively, P=0.03). Our results indicate a function of the epigenetic regulation of the Wnt pathway in predicting relapse in a subgroup of AML patients.
Collapse
Affiliation(s)
- A Valencia
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Chen CC, Gau JP, You JY, Lee KD, Yu YB, Lu CH, Lin JT, Lan C, Lo WH, Liu JM, Yang CF. Prognostic significance of beta-catenin and topoisomerase IIalpha in de novo acute myeloid leukemia. Am J Hematol 2009; 84:87-92. [PMID: 19127593 DOI: 10.1002/ajh.21334] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The Wnt/beta-catenin signaling is important for controlling self-renewal of hematopoietic stem cells and its constitutive activation has recently been documented in a significant proportion of acute myeloid leukemia (AML) cases. Topoisomerase IIalpha (Topo IIalpha) is a marker of cell proliferation and a crucial target for anthracycline cytotoxicity, the mainstay of management employed in AML. We retrospectively investigated the prognostic roles of beta-catenin and topo IIalpha in a cohort of 59 patients with newly diagnosed AML by immunohistochemistry. Aberrant beta-catenin expression was demonstrated in 13 patients (22%), and it was more likely to occur in those with unfavorable karyotypes. Advanced age and poor performance status adversely influenced the achievement of complete remission, while neither aberrant beta-catenin expression nor enhanced topo IIalpha activity did. On multivariate survival analysis, four factors independently predicted a shortened overall survival: aberrant beta-catenin expression, high topo IIalpha activity, poor-risk cytogenetics, and presence of at least one comorbidity factor. Our results suggest that both beta-catenin and topo IIalpha independently predicted an adverse prognosis and might serve as new markers for risk stratification in AML patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, Neoplasm/analysis
- Antimetabolites, Antineoplastic/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/analysis
- Cohort Studies
- Comorbidity
- Cytarabine/therapeutic use
- DNA Topoisomerases, Type II/analysis
- DNA-Binding Proteins/analysis
- Female
- Humans
- Karyotyping
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Male
- Middle Aged
- Neoplasm Proteins/analysis
- Prognosis
- Retrospective Studies
- Risk Factors
- Survival Analysis
- Wnt Proteins/physiology
- beta Catenin/analysis
Collapse
Affiliation(s)
- Chih-Cheng Chen
- Division of Hematology Oncology, Department of Medicine, Chang Gung Memorial Hospital-Chiayi, Chiayi, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Minke KS, Staib P, Puetter A, Gehrke I, Gandhirajan RK, Schlösser A, Schmitt EK, Hallek M, Kreuzer KA. Small molecule inhibitors of WNT signaling effectively induce apoptosis in acute myeloid leukemia cells. Eur J Haematol 2008; 82:165-75. [PMID: 19067737 DOI: 10.1111/j.1600-0609.2008.01188.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In a significant proportion of acute myeloid leukemia (AML) cases the canonical WNT pathway is upregulated and targeting the WNT/LEF1 signaling cascade in AML may be a promising approach to develop new treatments for this entity. Recently two compounds (CGP049090 and PFK115-584) have been identified, which specifically inhibit complexation of beta-catenin (CTNNB1) and lymphoid enhancer-binding factor 1 (LEF1) leading to transcriptional inactivation of LEF1 in colon carcinoma cell lines. To evaluate the effect of WNT inhibition utilizing theses compounds with regard to their effectivity in AML we treated the AML cell lines Kasumi-1 and HL-60, primary AML blasts and healthy peripheral blood mononuclear cells (PBMCs) with varying concentrations of both substances. Treatment with both compounds for 24 h resulted in a significant killing of AML cell lines and primary AML blasts with 50% effective concentration doses (EC(50)) within the submicromolar range. PBMCs were not significantly affected as indicated by EC(50)-values 100-fold higher than for AML cells. Cell kill was mediated by apoptosis as indicated by induction of caspases 3 and 7 and cleavage of poly(ADP-ribose) polymerase (PARP) upon treatment. Furthermore, we could show that both compounds substantially decrease expression of CTNNB1/LEF1 target genes c-myc, cyclin D1 and survivin, proofing the specificity of the substances. This was shown in both, AML cell lines and most of the tested primary samples. Our data demonstrate that targeting this pathway seems to be an innovative approach in the treatment of AML.
Collapse
|
130
|
Abstract
Recently, many malignancies have been demonstrated to be modeled on a loose developmental hierarchy. At the apex of these hierarchies sit so-called cancer stem cells or cancer-initiating cells, which are wholly responsible for the continued growth and propagation of the tumor. The first such cancer stem cells were described in acute myeloid leukemia (AML). The discovery of these cells also has important clinical implications. Following treatment, the majority of tumors, including leukemias, initially respond. However, relapse is common and often fatal. A likely explanation for this is that leukemia stem cells are relatively insensitive to current therapies and that tumor bulk reduction reflects the death of leukemic blasts that lack tumor initiation potential. This review will focus on what is known of the molecular and cellular biology of the leukemia stem cell and the leukemia stem cell niche in AML and then will identify molecular pathways critical for leukemia stem cells. Finally, we will identify current and prospective therapeutic targets to facilitate eradication of leukemia stem cells. It is hoped that, in defining the biology of cancer stem cells and how they differ from their adult tissue stem cell counterpart, we should identify therapeutic targets to improve treatment outcomes in leukemia and other malignant diseases.
Collapse
Affiliation(s)
- Wai-In Chan
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | | |
Collapse
|
131
|
Jost E, Schmid J, Wilop S, Schubert C, Suzuki H, Herman JG, Osieka R, Galm O. Epigenetic inactivation of secreted Frizzled-related proteins in acute myeloid leukaemia. Br J Haematol 2008; 142:745-53. [PMID: 18537968 DOI: 10.1111/j.1365-2141.2008.07242.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Wnt signalling pathway has a key function in stem cell maintenance and differentiation of haematopoietic progenitors. Secreted Frizzled-related protein genes (SFRPs), functioning as Wnt signalling antagonists, have been found to be downregulated by promoter hypermethylation in many tumours. To analyse epigenetic dysregulation of SFRPs in acute myeloid leukaemia (AML), we examined the promoter methylation status of SFRP1, -2, -4 and -5 in AML cell lines by methylation-specific polymerase chain reaction (MSP). Aberrant CpG island methylation was found for all four SFRP genes. By real-time reverse transcription-PCR, corresponding transcriptional silencing for SFRP1 and -2 was demonstrated and treatment of cell lines with 5-aza-2'-deoxycytidine resulted in re-expression. The methylation status of the SFRP genes was analysed in 100 specimens obtained from AML patients at diagnosis. The frequencies of aberrant methylation among the patient samples were 29% for SFRP1, 19% for SFRP2, 0% for SFRP4 and 9% for SFRP5. For SFRP2, a correlation between promoter hypermethylation and transcriptional downregulation was found in primary AML samples. Among AML cases with a favourable karyotype, hypermethylation of SFRP genes was restricted to patients with core binding factor (CBF) leukaemia, and aberrant methylation of the SFRP2 promoter was an adverse risk factor for survival in CBF leukaemia.
Collapse
Affiliation(s)
- E Jost
- Medizinische Klinik IV, Universitaetsklinikum Aachen, RWTH Aachen, Aachen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Si J, Collins SJ. Activated Ca2+/calmodulin-dependent protein kinase IIgamma is a critical regulator of myeloid leukemia cell proliferation. Cancer Res 2008; 68:3733-42. [PMID: 18483256 PMCID: PMC2443690 DOI: 10.1158/0008-5472.can-07-2509] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ca(2+) signaling is an important component of signal transduction pathways regulating B and T lymphocyte proliferation, but the functional role of Ca(2+) signaling in regulating myeloid leukemia cell proliferation has been largely unexplored. We observe that the activated (autophosphorylated) Ca(2+)/calmodulin-dependent protein kinase IIgamma (CaMKIIgamma) is invariably present in myeloid leukemia cell lines as well as in the majority of primary acute myelogenous leukemia patient samples. In contrast, myeloid leukemia cells induced to terminally differentiate or undergo growth arrest display a marked reduction in this CaMKIIgamma autophosphorylation. In cells harboring the bcr-abl oncogene, the activation (autophosphorylation) of CaMKIIgamma is regulated by this oncogene. Moreover, inhibition of CaMKIIgamma activity with pharmacologic agents, dominant-negative constructs, or short hairpin RNAs inhibits the proliferation of myeloid leukemia cells, and this is associated with the inactivation/down-regulation of multiple critical signal transduction networks involving the mitogen-activated protein kinase, Janus-activated kinase/signal transducers and activators of transcription (Jak/Stat), and glycogen synthase kinase (GSK3beta)/beta-catenin pathways. In myeloid leukemia cells, CaMKIIgamma directly phosphorylates Stat3 and enhances its transcriptional activity. Thus, CaMKIIgamma is a critical regulator of multiple signaling networks regulating the proliferation of myeloid leukemia cells. Inhibiting CaMKIIgamma may represent a novel approach in the targeted therapy of myeloid leukemia.
Collapse
Affiliation(s)
- Jutong Si
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | |
Collapse
|
133
|
Xu J, Suzuki M, Niwa Y, Hiraga J, Nagasaka T, Ito M, Nakamura S, Tomita A, Abe A, Kiyoi H, Kinoshita T, Naoe T. Clinical significance of nuclear non-phosphorylated beta-catenin in acute myeloid leukaemia and myelodysplastic syndrome. Br J Haematol 2008; 140:394-401. [PMID: 18217891 PMCID: PMC2253692 DOI: 10.1111/j.1365-2141.2007.06914.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Wnt signaling activates the canonical pathway and induces the accumulation of non-phosphorylated beta-catenin (NPBC) in the nucleus. Although this pathway plays an important role in the maintenance of haematopoietic stem cells as well as in oncogenesis, the significance of nuclear NPBC remains unclear in malignant haematopoiesis. This study examined the expression of nuclear NPBC in bone marrow specimens from 54 and 44 patients with de novo acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS), respectively. On immunohistochemistry with an anti-NPBC antibody, the nuclei were positively stained in 22 and 18 of AML and MDS specimens, respectively. Staining of nuclear NPBC was associated with AML subtypes (M6 and M7), low complete remission (CR) rate, and poor prognosis. Nuclear NPBC was also associated with a high score when using the International Prognostic Scoring System (IPSS) for MDS and with −7/−7q and complex karyotypes. These findings suggest that in situ detection of nuclear NPBC by immunohistochemistry could provide new insights into the pathogenesis and prognosis of AML and MDS.
Collapse
Affiliation(s)
- Jinglan Xu
- Department of Haematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Kajiguchi T, Lee S, Lee MJ, Trepel JB, Neckers L. KIT regulates tyrosine phosphorylation and nuclear localization of beta-catenin in mast cell leukemia. Leuk Res 2008; 32:761-70. [PMID: 17949810 PMCID: PMC2682210 DOI: 10.1016/j.leukres.2007.08.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 07/06/2007] [Accepted: 08/08/2007] [Indexed: 12/20/2022]
Abstract
Gain-of-function mutations in the proto-oncogene c-kit that induce constitutive kinase activity of its product, KIT protein, are characteristic of human mast cell disease and are believed to play a central role in mast cell leukemia oncogenesis, proliferation and survival. Nuclear overexpression of the Wnt effector beta-catenin and deregulated beta-catenin nuclear signaling can promote malignant transformation in solid tumors and hematologic malignancies. However, a role for beta-catenin in mast cell leukemia has not been described. Nuclear accumulation of beta-catenin is upregulated by its tyrosine phosphorylation, a process that can be exacerbated by deregulated expression of oncogenic tyrosine kinases. Here, we investigated the relationship between activated KIT and beta-catenin signaling in mast cell leukemia. Beta-catenin was tyrosine-phosphorylated in cells with KIT activated by either gain-of-function mutation or incubation with the KIT ligand stem cell factor. Beta-catenin tyrosine phosphorylation depended on KIT activity but not on PI3K-AKT activation. Tyrosine phosphorylation of beta-catenin was associated with its nuclear localization and enhanced transcription of target genes c-myc and cyclin D1. Endogenous KIT and beta-catenin were found to associate in mast cell leukemia cells, and in vitro kinase assay demonstrated that active KIT phosphorylates tyrosine residues of beta-catenin directly. Aberrant beta-catenin-driven transcription caused by deregulated KIT may represent a significant new target for treatment of mast cell leukemia.
Collapse
Affiliation(s)
- Tomohiro Kajiguchi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sunmin Lee
- Medical Oncology Branch; National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Min-Jung Lee
- Medical Oncology Branch; National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jane B. Trepel
- Medical Oncology Branch; National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Len Neckers
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
135
|
Wnt-expressing rat embryonic fibroblasts suppress Apo2L/TRAIL-induced apoptosis of human leukemia cells. Apoptosis 2008; 13:573-87. [PMID: 18347988 DOI: 10.1007/s10495-008-0191-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Accepted: 02/18/2008] [Indexed: 12/27/2022]
Abstract
Wnt signaling enhances cell proliferation and the maintenance of hematopoietic cells. In contrast, cytotoxic ligand Apo2L/TRAIL induces the apoptosis of various transformed cells. We observed that co-culture of human pre-B leukemia cells KM3 and REH with Wnt1- or Wnt3a-producing rat embryonic fibroblasts efficiently suppressed Apo2L/TRAIL-induced apoptosis of the lymphoid cells. This suppression occurs at the early stages of the Apo2L/TRAIL apoptotic cascade and, interestingly, the activation of the Wnt pathway alone in human leukemia cells is not sufficient for their full anti-apoptotic protection. We hypothesize that a stimulus emanating specifically from Wnt1- or Wnt3a-expressing rat fibroblasts is responsible for the observed resistance to Apo2L/TRAIL. This anti-apoptotic signaling was significantly hampered by the inhibition of the MEK1/ERK1/2 or NFkappaB pathways in KM3 and REH cells. Our results imply that paracrine Wnt-related signals could be important for the survival of pre-B cell-derived malignancies.
Collapse
|
136
|
Nemeth MJ, Bodine DM. Regulation of hematopoiesis and the hematopoietic stem cell niche by Wnt signaling pathways. Cell Res 2008; 17:746-58. [PMID: 17768401 DOI: 10.1038/cr.2007.69] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare population of cells that are responsible for life-long generation of blood cells of all lineages. In order to maintain their numbers, HSCs must establish a balance between the opposing cell fates of self-renewal (in which the ability to function as HSCs is retained) and initiation of hematopoietic differentiation. Multiple signaling pathways have been implicated in the regulation of HSC cell fate. One such set of pathways are those activated by the Wnt family of ligands. Wnt signaling pathways play a crucial role during embryogenesis and deregulation of these pathways has been implicated in the formation of solid tumors. Wnt signaling also plays a role in the regulation of stem cells from multiple tissues, such as embryonic, epidermal, and intestinal stem cells. However, the function of Wnt signaling in HSC biology is still controversial. In this review, we will discuss the basic characteristics of the adult HSC and its regulatory microenvironment, the "niche", focusing on the regulation of the HSC and its niche by the Wnt signaling pathways.
Collapse
Affiliation(s)
- Michael J Nemeth
- Hematopoiesis Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, Bethesda, MD 20892-4442, USA.
| | | |
Collapse
|
137
|
Suzuki R, Onizuka M, Kojima M, Shimada M, Fukagawa S, Tsuboi K, Kobayashi H, Shintani A, Ogawa Y, Kawada H, Hotta T, Ando K. Preferential hypermethylation of the Dickkopf-1 promoter in core-binding factor leukaemia. Br J Haematol 2007; 138:624-31. [PMID: 17686056 DOI: 10.1111/j.1365-2141.2007.06702.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The Dickkopf-1 (DKK1) gene product is an extracellular Wnt inhibitor. Hypermethylation of the DKK1 promoter results in transcriptional silencing and may play an important role in cancer development. Here, we investigated hypermethylation of the DKK1 promoter in patients with acute myeloid leukaemia (AML), especially core-binding factor (CBF) leukaemia. The methylation status of DKK1 was analysed using methylation-specific polymerase chain reaction in 47 patients with AML. DKK1 methylation was found in 14 (29.8%) patients, and more frequently in those with CBF leukaemia (6 of 12 patients), than in those with acute promyelocytic leukaemia (APL) (0 of 6 patients) (P = 0.03). In contrast, Wnt inhibitory factor-1 methylation was found in APL (4 of 6 patients) but not in CBF leukaemia (0 of 12 patients) (P = 0.001). Multivariate analyses suggested that DKK1 methylation was a risk factor for poorer overall survival. Sequential analysis using four paired samples obtained at diagnosis and relapse suggested that DKK1 methylation was involved in the progression of leukaemia. Therefore, DKK1 methylation may be involved in leukaemogenesis, especially in CBF leukaemia, and may be a useful prognostic marker in AML.
Collapse
Affiliation(s)
- Rikio Suzuki
- Department of Haematology/Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Yi F, Merrill BJ. Stem cells and TCF proteins: a role for beta-catenin--independent functions. ACTA ACUST UNITED AC 2007; 3:39-48. [PMID: 17873380 DOI: 10.1007/s12015-007-0003-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/18/2022]
Abstract
The Wnt signal transduction pathway has been shown to stimulate stem cell self renewal and has been shown to cause cancer in humans. One interesting aspect of Wnt signaling is that it utilizes downstream DNA-binding transcription factors, called Tcf proteins, which can activate transcription of target genes in the presence of a Wnt signal and repress the expression of target genes in the absence of a Wnt signal. Since Tcf proteins are present in Wnt-stimulated and unstimulated stem cells, understanding how Tcf proteins regulate target gene expression in each state offers the potential to understand how stem cells regulate their self-renewal, differentiation, and proliferation. In this article, we will review recent work elucidating the roles Tcf-protein interactions in the context of stem cells and cancer.
Collapse
Affiliation(s)
- Fei Yi
- Molecular Biology Research Building, Department of Biochemistry and Molecular Genetics, University of Illinois, 900 S. Ashland Ave., Chicago, IL 60607, USA
| | | |
Collapse
|
139
|
Boultwood J, Pellagatti A, Cattan H, Lawrie CH, Giagounidis A, Malcovati L, Porta MGD, Jädersten M, Killick S, Fidler C, Cazzola M, Hellström-Lindberg E, Wainscoat JS. Gene expression profiling of CD34+cells in patients with the 5q− syndrome. Br J Haematol 2007; 139:578-89. [DOI: 10.1111/j.1365-2141.2007.06833.x] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
140
|
Tonks A, Pearn L, Musson M, Gilkes A, Mills KI, Burnett AK, Darley RL. Transcriptional dysregulation mediated by RUNX1-RUNX1T1 in normal human progenitor cells and in acute myeloid leukaemia. Leukemia 2007; 21:2495-505. [PMID: 17898786 DOI: 10.1038/sj.leu.2404961] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The t(8;21)(q22;q22) occurs frequently in acute myelogenous leukaemia and gives rise to the transcription factor fusion protein, RUNX1-RUNX1T1 (also known as AML1-ETO). To identify the genes dysregulated by the aberrant transcriptional activity of RUNX1-RUNX1T1, we used microarrays to determine the effect of this mutation on gene expression in human progenitor cells and during subsequent development. Gene signatures of these developmental subsets were very dissimilar indicating that effects of RUNX1-RUNX1T1 are highly context dependent. We focused on gene changes associated with the granulocytic lineage and identified a clinically relevant subset of these by comparison with 235 leukaemia patient transcriptional signatures. We confirmed the overexpression of a number of significant genes (Sox4, IL-17BR, CD200 and gamma-catenin). Further, we show that overexpression of CD200 and gamma-catenin is also associated with the inv(16) abnormality which like RUNX1-RUNX1T1 disrupts core binding factor activity. We investigated the functional significance of CD200 and gamma-catenin overexpression in normal human progenitor cells. The effect of IL17 on growth was also assessed. Individually, none of these changes were sufficient to recapitulate the effects of RUNX1-RUNX1T1 on normal development. These data provide the most comprehensive and pertinent assessment of the effect of RUNX1-RUNX1T1 on gene expression and demonstrate the highly context-dependent effects of this fusion gene.
Collapse
MESH Headings
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Cell Line, Tumor/metabolism
- Cell Lineage
- Cells, Cultured/metabolism
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 21/ultrastructure
- Chromosomes, Human, Pair 8/genetics
- Chromosomes, Human, Pair 8/ultrastructure
- Core Binding Factor Alpha 2 Subunit/physiology
- Desmoplakins/genetics
- Desmoplakins/physiology
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic/genetics
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- High Mobility Group Proteins/biosynthesis
- High Mobility Group Proteins/genetics
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Oncogene Proteins, Fusion/physiology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- RUNX1 Translocation Partner 1 Protein
- Receptors, Interleukin-17/biosynthesis
- Receptors, Interleukin-17/genetics
- Recombinant Fusion Proteins/physiology
- SOXC Transcription Factors
- Trans-Activators/biosynthesis
- Trans-Activators/genetics
- Transcription, Genetic/genetics
- Translocation, Genetic
- gamma Catenin/genetics
- gamma Catenin/physiology
Collapse
Affiliation(s)
- A Tonks
- Department of Haematology, School of Medicine, Cardiff University, Cardiff, UK.
| | | | | | | | | | | | | |
Collapse
|
141
|
Nemeth MJ, Topol L, Anderson SM, Yang Y, Bodine DM. Wnt5a inhibits canonical Wnt signaling in hematopoietic stem cells and enhances repopulation. Proc Natl Acad Sci U S A 2007; 104:15436-41. [PMID: 17881570 PMCID: PMC1986571 DOI: 10.1073/pnas.0704747104] [Citation(s) in RCA: 226] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanisms that regulate hematopoietic stem cell (HSC) fate decisions between proliferation and multilineage differentiation are unclear. Members of the Wnt family of ligands that activate the canonical Wnt signaling pathway, which utilizes beta-catenin to relay the signal, have been demonstrated to regulate HSC function. In this study, we examined the role of noncanonical Wnt signaling in regulating HSC fate. We observed that noncanonical Wnt5a inhibited Wnt3a-mediated canonical Wnt signaling in HSCs and suppressed Wnt3a-mediated alterations in gene expression associated with HSC differentiation, such as increased expression of myc. Wnt5a increased short- and long-term HSC repopulation by maintaining HSCs in a quiescent G(0) state. From these data, we propose that Wnt5a regulates hematopoiesis by the antagonism of the canonical Wnt pathway, resulting in a pool of quiescent HSCs.
Collapse
Affiliation(s)
| | - Lilia Topol
- Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD 20892-4442
| | | | - Yingzi Yang
- Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD 20892-4442
| | - David M. Bodine
- *Genetics and Molecular Biology Branch and
- To whom correspondence should be addressed at:
Genetics and Molecular Biology Branch, National Human Genome Research Institute, 49 Convent Drive, Room 4A04, Bethesda, MD 20892-4442. E-mail:
| |
Collapse
|
142
|
Kajiguchi T, Chung EJ, Lee S, Stine A, Kiyoi H, Naoe T, Levis MJ, Neckers L, Trepel JB. FLT3 regulates beta-catenin tyrosine phosphorylation, nuclear localization, and transcriptional activity in acute myeloid leukemia cells. Leukemia 2007; 21:2476-84. [PMID: 17851558 DOI: 10.1038/sj.leu.2404923] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Deregulated accumulation of nuclear beta-catenin enhances transcription of beta-catenin target genes and promotes malignant transformation. Recently, acute myeloid leukemia (AML) cells with activating mutations of FMS-like tyrosine kinase-3 (FLT3) were reported to display elevated beta-catenin-dependent nuclear signaling. Tyrosine phosphorylation of beta-catenin has been shown to promote its nuclear localization. Here, we examined the causal relationship between FLT3 activity and beta-catenin nuclear localization. Compared to cells with wild-type FLT3 (FLT3-WT), cells with the FLT3 internal tandem duplication (FLT3-ITD) and tyrosine kinase domain mutation (FLT3-TKD) had elevated levels of tyrosine-phosphorylated beta-catenin. Although beta-catenin was localized mainly in the cytoplasm in FLT3-WT cells, it was primarily nuclear in FLT3-ITD cells. Treatment with FLT3 kinase inhibitors or FLT3 silencing with RNAi decreased beta-catenin tyrosine phosphorylation and nuclear localization. Conversely, treatment of FLT3-WT cells with FLT3 ligand increased tyrosine phosphorylation and nuclear accumulation of beta-catenin. Endogenous beta-catenin co-immunoprecipitated with endogenous activated FLT3, and recombinant activated FLT3 directly phosphorylated recombinant beta-catenin. Finally, FLT3 inhibitor decreased tyrosine phosphorylation of beta-catenin in leukemia cells obtained from FLT3-ITD-positive AML patients. These data demonstrate that FLT3 activation induces beta-catenin tyrosine phosphorylation and nuclear localization, and thus suggest a mechanism for the association of FLT3 activation and beta-catenin oncogeneic signaling in AML.
Collapse
Affiliation(s)
- T Kajiguchi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Rawat VPS, Thoene S, Naidu VM, Arseni N, Heilmeier B, Metzeler K, Petropoulos K, Deshpande A, Quintanilla-Martinez L, Bohlander SK, Spiekermann K, Hiddemann W, Feuring-Buske M, Buske C. Overexpression of CDX2 perturbs HOX gene expression in murine progenitors depending on its N-terminal domain and is closely correlated with deregulated HOX gene expression in human acute myeloid leukemia. Blood 2007; 111:309-19. [PMID: 17855634 DOI: 10.1182/blood-2007-04-085407] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The mechanisms underlying deregulation of HOX gene expression in AML are poorly understood. The ParaHox gene CDX2 was shown to act as positive upstream regulator of several HOX genes. In this study, constitutive expression of Cdx2 caused perturbation of leukemogenic Hox genes such as Hoxa10 and Hoxb8 in murine hematopoietic progenitors. Deletion of the N-terminal domain of Cdx2 abrogated its ability to perturb Hox gene expression and to cause acute myeloid leukemia (AML) in mice. In contrast inactivation of the putative Pbx interacting site of Cdx2 did not change the leukemogenic potential of the gene. In an analysis of 115 patients with AML, expression levels of CDX2 were closely correlated with deregulated HOX gene expression. Patients with normal karyotype showed a 14-fold higher expression of CDX2 and deregulated HOX gene expression compared with patients with chromosomal translocations such as t(8:21) or t(15;17). All patients with AML with normal karyotype tested were negative for CDX1 and CDX4 expression. These data link the leukemogenic potential of Cdx2 to its ability to dysregulate Hox genes. They furthermore correlate the level of CDX2 expression with HOX gene expression in human AML and support a potential role of CDX2 in the development of human AML with aberrant Hox gene expression.
Collapse
|
144
|
Gebeshuber CA, Sladecek S, Grunert S. Beta-catenin/LEF-1 signalling in breast cancer--central players activated by a plethora of inputs. Cells Tissues Organs 2007; 185:51-60. [PMID: 17587808 DOI: 10.1159/000101303] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although the role of Wnt signalling in breast cancer is far from being fully understood, in the last years its importance has been reported frequently. Besides stimulation by canonical Wnt signalling, the downstream effectors beta-catenin and the transcriptional modulators of the T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) family can also be activated by other inputs including the TGF-beta pathway. Wnt and TGF-beta signalling are both major signal transduction pathways, which provide important cues during development and tumor progression. However, particularly TGF-beta has a complicated influence on oncogenesis, which ranges from suppressive to promoting activity. Signalling pathways activated in parallel with TGF-beta might determine the oncogenic influence, and therefore place signals cooperating with TGF-beta into the limelight. During early development Wnt and TGF-beta signalling collaborate extensively. Here we provide an overview of the known interactions of Wnt with TGF-beta signalling in development and metastasis, particularly in breast cancer. We want to focus on the Wnt-activated transcription factor complex beta-catenin/LEF-1, its upstream activators, its downstream targets and consequences on the cellular level in response to beta-catenin/LEF-1 activation.
Collapse
|
145
|
Abstract
Recent evidence has demonstrated the existence of a small subset of the tumour mass that is wholly responsible for the sustained growth and propagation of the tumour. This cancer stem cell (CSC) compartment is also likely to be responsible both for disease relapse and the resistance to therapy that often accompanies relapse. The evidence for CSCs in various malignancies is presented. The failure of existing therapeutics to eradicate CSCs suggests that they are relatively resistant to present cancer treatments. This resistance may reflect the preservation of normal stem cell protective mechanisms, such as an increased expression of drug efflux pumps or alterations in apoptotic, cell cycle and DNA repair mechanisms. Targeting these mechanisms, and taking advantage of potential differences in the biology of normal stem cells and CSCs, such as differences in surface phenotype, self renewal/quiescence and stem cell-niche interactions are discussed and preliminary preclinical or clinical data are presented. Finally, the authors give their opinion of the direction in which one must travel to successfully target the CSC and improve treatment outcomes in malignant disease.
Collapse
Affiliation(s)
- Brynn T Kvinlaug
- University of Cambridge, Department of Haematology, Cambridge Institute for Medical Research, Cambridge, CB2 2XY, UK
| | | |
Collapse
|
146
|
Mikesch JH, Steffen B, Berdel WE, Serve H, Müller-Tidow C. The emerging role of Wnt signaling in the pathogenesis of acute myeloid leukemia. Leukemia 2007; 21:1638-47. [PMID: 17554387 DOI: 10.1038/sj.leu.2404732] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Wnt signaling plays an important role in stem cell self-renewal and proliferation. Aberrant activation of Wnt signaling and its downstream targets are intimately linked with several types of cancer with colon cancer being the best-studied example. However, recent results also suggest an important role of Wnt signaling in normal as well as leukemic hematopoietic stem cells. Aberrant activation of Wnt signaling and downstream effectors has been demonstrated in acute myeloid leukemia. Here, mutant receptor tyrosine kinases, such as Flt3 and chimeric transcription factors such as promyelocytic leukemia-retinoic acid receptor-alpha and acute myeloid leukemia1-ETO, induce downstream Wnt signaling events. These findings suggest that the Wnt signaling pathway is an important target in several leukemogenic pathways and may provide a novel opportunity for targeting leukemic stem cells.
Collapse
Affiliation(s)
- J-H Mikesch
- Department of Medicine, Hematology and Oncology, University of Muenster, Münster, Germany
| | | | | | | | | |
Collapse
|
147
|
Chong ZZ, Li F, Maiese K. Cellular demise and inflammatory microglial activation during beta-amyloid toxicity are governed by Wnt1 and canonical signaling pathways. Cell Signal 2007; 19:1150-62. [PMID: 17289346 PMCID: PMC1913492 DOI: 10.1016/j.cellsig.2006.12.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 12/06/2006] [Accepted: 12/14/2006] [Indexed: 11/24/2022]
Abstract
Initially described as a modulator of embryogenesis for a number of organ systems, Wnt1 has recently been linked to the development of several neurodegenerative disorders, none being of greater significance than Alzheimer's disease. We therefore examined the ability of Wnt1 to oversee vital pathways responsible for cell survival during beta-amyloid (Abeta1-42) exposure. Here we show that Wnt1 is critical for protection in the SH-SY5Y neuronal cell line against genomic DNA degradation, membrane phosphatidylserine (PS) exposure, and microglial activation, since these neuroprotective attributes of Wnt1 are lost during gene silencing of Wnt1 protein expression. Intimately tied to Wnt1 protection is the presence and activation of Akt1. Pharmacological inhibition of the PI 3-K pathway or gene silencing of Akt1 expression can abrogate the protective capacity of Wnt1. Closely aligned with Wnt1 and Akt1 are the integrated canonical pathways of synthase kinase-3beta (GSK-3beta) and beta-catenin. Through Akt1 dependent pathways, Wnt1 phosphorylates GSK-3beta and maintains beta-catenin integrity to insure its translocation from the cytoplasm to the nucleus to block apoptosis. Our work outlines a highly novel role for Wnt1 and its integration with Akt1, GSK-3beta, and beta-catenin to foster neuronal cell survival and repress inflammatory microglial activation that can identify new avenues of therapy against neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Faqi Li
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201
- Departments of Neurology and Anatomy & Cell Biology, Center for Molecular Medicine and Genetics, Institute of Environmental Health Sciences, Wayne State University School of Medicine, Detroit, Michigan 48201
| |
Collapse
|
148
|
McLornan DP, McMullin MF, Johnston P, Longley DB. Molecular mechanisms of drug resistance in acute myeloid leukaemia. Expert Opin Drug Metab Toxicol 2007; 3:363-77. [PMID: 17539744 DOI: 10.1517/17425255.3.3.363] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Resistance to chemotherapy in acute myeloid leukaemia is a major obstacle to a successful outcome for many patients. Often, there is resistance against a broad range of drugs due to multiple, simultaneously active processes. These mechanisms include effects on drug influx and efflux, drug activation/inactivation, DNA repair mechanisms, altered response of end targets, an altered haematopoietic microenvironment and dysfunctional apoptotic pathways. This article reviews the factors that determine leukaemic cell chemosensitivity and discusses the potential for rationally guided therapy.
Collapse
Affiliation(s)
- Donal P McLornan
- Medical Research Council Clinical Research Fellow, Queen's University Belfast, Centre for Cancer Research and Cell Biology, BT7 1NN, Northern Ireland, UK
| | | | | | | |
Collapse
|
149
|
Scholl C, Bansal D, Döhner K, Eiwen K, Huntly BJ, Lee BH, Rücker FG, Schlenk RF, Bullinger L, Döhner H, Gilliland DG, Fröhling S. The homeobox gene CDX2 is aberrantly expressed in most cases of acute myeloid leukemia and promotes leukemogenesis. J Clin Invest 2007; 117:1037-48. [PMID: 17347684 PMCID: PMC1810574 DOI: 10.1172/jci30182] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 01/12/2007] [Indexed: 12/21/2022] Open
Abstract
The homeobox transcription factor CDX2 plays an important role in embryonic development and regulates the proliferation and differentiation of intestinal epithelial cells in the adult. We have found that CDX2 is expressed in leukemic cells of 90% of patients with acute myeloid leukemia (AML) but not in hematopoietic stem and progenitor cells derived from normal individuals. Stable knockdown of CDX2 expression by RNA interference inhibited the proliferation of various human AML cell lines and strongly reduced their clonogenic potential in vitro. Primary murine hematopoietic progenitor cells transduced with Cdx2 acquired serial replating activity, were able to be continuously propagated in liquid culture, generated fully penetrant and transplantable AML in BM transplant recipients, and displayed dysregulated expression of Hox family members in vitro and in vivo. These results demonstrate that aberrant expression of the developmental regulatory gene CDX2 in the adult hematopoietic compartment is a frequent event in the pathogenesis of AML; suggest a role for CDX2 as part of a common effector pathway that promotes the proliferative capacity and self-renewal potential of myeloid progenitor cells; and support the hypothesis that CDX2 is responsible, in part, for the altered HOX gene expression that is observed in most cases of AML.
Collapse
Affiliation(s)
- Claudia Scholl
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Dimple Bansal
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Konstanze Döhner
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Karina Eiwen
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Brian J.P. Huntly
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Benjamin H. Lee
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Frank G. Rücker
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Richard F. Schlenk
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Lars Bullinger
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Hartmut Döhner
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - D. Gary Gilliland
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Stefan Fröhling
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
150
|
Abstract
This review addresses the scope of influence of mRNA decay on cellular functions and its potential role in normal and malignant hematopoiesis. Evidence is emerging that leukemic oncogenes and hematopoietic cytokines interact with mRNA decay pathways. These pathways can co-regulate functionally related genes through specific motifs in the 3'-untranslated region of targeted transcripts. The steps that link external stimuli to transcript turnover are not fully understood, but include subcellular relocalization or post-transcriptional modification of specific transcript-stabilizing or -destabilizing proteins. Improper functioning of these regulators of mRNA turnover can impede normal cellular differentiation or promote cancers. By delineating how subsets of transcripts decay in synchrony during normal hematopoiesis, it may be possible to determine whether this post-transcriptional regulatory pathway is hijacked in leukemogenesis.
Collapse
Affiliation(s)
- R A Steinman
- University of Pittsburgh Cancer Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|