401
|
Diederichs S, Bäumer N, Ji P, Metzelder SK, Idos GE, Cauvet T, Wang W, Möller M, Pierschalski S, Gromoll J, Schrader MG, Koeffler HP, Berdel WE, Serve H, Müller-Tidow C. Identification of interaction partners and substrates of the cyclin A1-CDK2 complex. J Biol Chem 2004; 279:33727-41. [PMID: 15159402 DOI: 10.1074/jbc.m401708200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The CDK2-associated cyclin A1 is essential for spermatogenesis and contributes to leukemogenesis. The detailed molecular functions of cyclin A1 remain unclear, since the molecular networks involving cyclin A1-CDK2 have not been elucidated. Here, we identified novel cyclin A1/CDK2 interaction partners in a yeast triple-hybrid approach. Several novel proteins (INCA1, KARCA1, and PROCA1) as well as the known proteins GPS2 (G-protein pathway suppressor 2), Ku70, receptor for activated protein kinase C1/guanine nucleotide-binding protein beta-2-like-1, and mRNA-binding motif protein 4 were identified as interaction partners. These proteins link the cyclin A1-CDK2 complex to diverse cellular processes such as DNA repair, signaling, and splicing. Interactions were confirmed by GST pull-down assays and co-immunoprecipitation. We cloned and characterized the most frequently isolated unknown gene, which we named INCA1 (inhibitor of CDK interacting with cyclin A1). The nuclear INCA1 protein is evolutionarily conserved and lacks homology to any known gene. This novel protein and two other interacting partners served as substrates for the cyclin A1-CDK2 kinase complex. Cyclin A1 and all interaction partners were highly expressed in testis with varying degrees of tissue specificity. The highest expression levels were observed at different time points during testis maturation, whereas expression levels in germ cell cancers and infertile testes decreased. Taken together, we identified testicular interaction partners of the cyclin A1-CDK2 complex and studied their expression pattern in normal organs, testis development, and testicular malignancies. Thereby, we establish a new basis for future functional analyses of cyclin A1. We provide evidence that the cyclin A1-CDK2 complex plays a role in several signaling pathways important for cell cycle control and meiosis.
Collapse
|
402
|
Müller-Tidow C, Steffen B, Cauvet T, Tickenbrock L, Ji P, Diederichs S, Sargin B, Köhler G, Stelljes M, Puccetti E, Ruthardt M, deVos S, Hiebert SW, Koeffler HP, Berdel WE, Serve H. Translocation products in acute myeloid leukemia activate the Wnt signaling pathway in hematopoietic cells. Mol Cell Biol 2004; 24:2890-904. [PMID: 15024077 PMCID: PMC371102 DOI: 10.1128/mcb.24.7.2890-2904.2004] [Citation(s) in RCA: 238] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The acute myeloid leukemia (AML)-associated translocation products AML1-ETO, PML-retinoic acid receptor alpha (RARalpha), and PLZF-RARalpha encode aberrant transcription factors. Several lines of evidence suggest similar pathogenetic mechanisms for these fusion proteins. We used high-density oligonucleotide arrays to identify shared target genes in inducibly transfected U937 cells expressing AML1-ETO, PML-RARalpha, or PLZF-RARalpha. All three fusion proteins significantly repressed the expression of 38 genes and induced the expression of 14 genes. Several of the regulated genes were associated with Wnt signaling. One of these, plakoglobin (gamma-catenin), was induced on the mRNA and protein level by all three fusion proteins. In addition, primary AML blasts carrying one of the fusion proteins significantly overexpressed plakoglobin. The plakoglobin promoter was cloned and shown to be induced by AML1-ETO, with promoter activation depending on the corepressor and histone deacetylase binding domains. The induction of plakoglobin by AML fusion proteins led to downstream signaling and transactivation of TCF- and LEF-dependent promoters, including the c-myc promoter, which was found to be bound by plakoglobin in vivo after AML1-ETO expression. beta-Catenin protein levels and TCF and LEF target genes such as c-myc and cyclin D1 were found to be induced by the fusion proteins. On the functional level, a dominant negative TCF inhibited colony growth of AML1-ETO-positive Kasumi cells, whereas plakoglobin transfection into myeloid 32D cells enhanced proliferation and clonal growth. Injection of plakoglobin-expressing 32D cells into syngeneic mice accelerated the development of leukemia. Transduction of plakoglobin into primitive murine hematopoietic progenitor cells preserved the immature phenotype during colony growth, suggesting enhanced self-renewal. These data provide evidence that activation of Wnt signaling is a common feature of several balanced translocations in AML.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Core Binding Factor Alpha 2 Subunit
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Desmoplakins
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Hematopoietic Stem Cells/physiology
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Lymphoid Enhancer-Binding Factor 1
- Mice
- Mice, Inbred C57BL
- Neoplasm Proteins/metabolism
- Oligonucleotide Array Sequence Analysis
- Oncogene Proteins, Fusion/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins/metabolism
- RUNX1 Translocation Partner 1 Protein
- Signal Transduction/physiology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transplantation, Isogeneic
- Wnt Proteins
- Zebrafish Proteins
- gamma Catenin
Collapse
|
403
|
Coletta RD, Christensen K, Reichenberger KJ, Lamb J, Micomonaco D, Huang L, Wolf DM, Müller-Tidow C, Golub TR, Kawakami K, Ford HL. The Six1 homeoprotein stimulates tumorigenesis by reactivation of cyclin A1. Proc Natl Acad Sci U S A 2004; 101:6478-83. [PMID: 15123840 PMCID: PMC404070 DOI: 10.1073/pnas.0401139101] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Homeobox genes constitute a large family of transcription factors that are essential during normal development and are often dysregulated in cancer. However, the molecular mechanisms by which homeobox genes influence cancer remain largely unknown. Here we show that the tissue-restricted cyclin A1 is a transcriptional target of the Six1 homeoprotein. Both genes are expressed in the embryonic but not the terminally differentiated mammary gland, and Six1-knockout mice show a dramatic reduction of cyclin A1 in the embryonic mammary gland. In addition, both genes are reexpressed in breast cancers. Six1 overexpression increases cyclin A1 mRNA levels and activity, cell proliferation, and tumor volume, whereas Six1 down-regulation decreases cyclin A1 mRNA levels and proliferation. Overexpression of Six1 in wild-type mouse embryonic fibroblasts, but not in knockout variants lacking the cyclin A1 gene, induces cell proliferation. Furthermore, inhibition of cyclin A1 in Six1-overexpressing mammary carcinoma cells decreases proliferation. Together these results demonstrate that cyclin A1 is required for the proliferative effect of Six1. We conclude that Six1 overexpression reinstates an embryonic pathway of proliferation in breast cancer by up-regulating cyclin A1.
Collapse
|
404
|
Yang G, Khalaf W, van de Locht L, Jansen JH, van der Reijden BA, Müller-Tidow C, Delwel HR, Serve H, Clapp DW, Hiebert SW. Epigenetic regulation of tumor suppressors in t(8:21)-containing AML. Ann Hematol 2004; 83:329-30. [PMID: 15052372 DOI: 10.1007/s00277-003-0841-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2003] [Accepted: 12/15/2003] [Indexed: 11/29/2022]
Abstract
The t(8;21) is perhaps the most frequent chromosomal translocation associated with acute myeloid leukemia. The translocation creates a fusion protein that consists of the DNA binding domain of the RUNX1 transcription factor fused to the MTG8 transcriptional co-repressor to create a potent transcriptional repressor. Here, we discuss the possibility that the t(8;21) fusion protein represses tumor suppressors that regulate the RAS signaling pathway and the p53 oncogenic checkpoint.
Collapse
MESH Headings
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 8/genetics
- Core Binding Factor Alpha 2 Subunit
- DNA-Binding Proteins/genetics
- Humans
- Leukemia, Myeloid, Acute/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/physiology
- Proto-Oncogene Proteins/genetics
- RUNX1 Translocation Partner 1 Protein
- Signal Transduction
- Transcription Factors/genetics
- Transcription, Genetic
- Translocation, Genetic
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/physiology
- ras Proteins/genetics
- ras Proteins/metabolism
Collapse
|
405
|
Yasmeen A, Berdel WE, Serve H, Müller-Tidow C. E- and A-type cyclins as markers for cancer diagnosis and prognosis. Expert Rev Mol Diagn 2004; 3:617-33. [PMID: 14510182 DOI: 10.1586/14737159.3.5.617] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cyclin-dependent kinase (CDK)2 interacting cyclins perform essential functions for DNA replication and cellular proliferation. The human genome encodes two E-type cyclins (E and E2) and two A-type cyclins (A1 and A2). Dysregulation of the CDK2-bound cyclins plays an important role in the pathogenesis of cancer. Cyclin A2 is associated with cellular proliferation and can be used for molecular diagnostics as a proliferation marker. In addition, cyclin A2 expression is associated with a poor prognosis in several types of cancer. Cyclin A1 is a tissue-specific cyclin that is highly expressed in acute myeloid leukemia and in testicular cancer. High levels of cyclin E expression are found in many types of cancer. Overexpression of cyclin E at the mRNA level can be based on gene amplification and transcriptional mechanisms. In addition, proteolytically cleaved forms of cyclin E that show oncogenic functions have been described. Cyclin E plays a critical role for G1/S transition. Its overexpression is not only associated with proliferation but rather indicates a more malignant phenotype which is likely to be linked to the induction of chromosomal instability. These biological functions of cyclin E relate to a poor prognosis when high cyclin E levels are found. The link between cyclin E and poor prognosis is well established in breast and lung cancer but is likely to be observed in other cancers as well. The second E-type cyclin, cyclin E2, has been shown to be overexpressed in breast cancers although the potential role as a diagnostic or prognostic marker is unknown. This review provides an overview of the potential of cyclins E and A as markers for diagnosis and prognosis in human cancer.
Collapse
|
406
|
Müller-Tidow C, Metzelder SK, Buerger H, Packeisen J, Ganser A, Heil G, Kügler K, Adigüzel G, Schwäble J, Steffen B, Ludwig WD, Heinecke A, Büchner T, Berdel WE, Serve H. Expression of the p14ARF tumor suppressor predicts survival in acute myeloid leukemia. Leukemia 2004; 18:720-6. [PMID: 14973498 DOI: 10.1038/sj.leu.2403296] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cell cycle aberrations are associated with therapy outcome in many types of cancer. We analyzed mRNA expression levels of 18 cell cycle-related genes in bone marrow samples from 78 acute myeloid leukemia (AML) patients and six controls using high-throughput quantitative RT-PCR. Samples of AML patients contained significantly increased mRNA expression levels of the mdm2 and c-myc oncogenes. Also, the average expression levels of p14ARF and p16INK4A were higher in patient samples compared to controls. Leukemic blasts and control bone marrow samples did not differ significantly in the expression levels of proliferation-associated genes such as cyclin A2 and pcna. When single genes were analyzed for prognostic significance in Kaplan-Meier and Cox regression analyses, a low p14ARF level emerged as a strong and independent predictor for poor survival (P=0.04 and 0.029). Subsequently, p14ARF mRNA levels were analyzed in a second, independent patient population (n=57). Again, low p14ARF levels were associated with a worse outcome. Finally, immunohistochemistry analysis of AML tissue arrays confirmed the widespread expression of c-myc and p14ARF in AML on the protein level. Taken together, the expression of the p53 regulators mdm2 and p14ARF are altered in AML, and low p14ARF levels indicate a poor prognosis.
Collapse
|
407
|
Tiesmeier J, Czwalinna A, Müller-Tidow C, Krauter J, Serve H, Heil G, Ganser A, Verbeek W. Evidence for allelic evolution of C/EBPalpha mutations in acute myeloid leukaemia. Br J Haematol 2003; 123:413-9. [PMID: 14616999 DOI: 10.1046/j.1365-2141.2003.04618.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transcription factor CCAAT/enhancer binding protein alpha (C/EBPalpha) is mutated in 6-10% of patients with acute myeloid leukaemia (AML). Recently, we reported the emergence of an N-terminal C/EBPalpha mutation after chemotherapy in a patient with secondary AML. The clone carrying the mutation became the dominant clone at relapse. This observation prompted us to compare the C/EBPalpha mutational status of 26 de novo non-core binding factor AML patients at diagnosis and at relapse after induction and consolidation chemotherapy. Four mutations in the C/EBPalpha gene were identified in two out of 26 patients. In both these cases, a biallelic mutation was present at diagnosis and at relapse: an amino-terminal frameshift mutation and a mutation of the fork/leucine finger 1 region. In patient 1, the amino-terminal frameshift mutation was duplicated and found on both alleles at relapse. In patient 2, the amino-terminal frameshift mutation and a mutation in the fork region were found either alone or combined on the same allele, suggesting a subclone formation. None of the patients without a C/EBPalpha mutation at diagnosis showed a mutation at relapse. This is the first report of an evolution of the C/EBPalpha gene between diagnosis and relapse in AML.
Collapse
|
408
|
Müller-Tidow C, Steffen B, Berdel WE, Serve H. Redirection of oncoproteins to kill cancer cells. Cell Cycle 2003; 2:531-3. [PMID: 14504468 DOI: 10.4161/cc.2.6.585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Oncogenic activity is often associated with altered intracellular protein localization and with specific gains of function, e.g., kinase activity. We propose, that intracellular redirection of oncogenic proteins towards novel targets can be used to specifically kill tumor cells. For example, an oncogenic kinase could be redirected to activate an apoptosis inducing protein. This redirection approach offers the advantages of high specificity (the oncogene is restricted to tumor cells) and potentially high activity since it makes use of the intrinsic functions of the oncogene. Also, the oncogene is not only functionally inhibited but it is turned against the cancer cell. Activity and specificity of the redirection therapy approach were demonstrated in AML1-ETO positive leukemia cells. A recombinant protein redirected the transcriptionally inhibitory AML1-ETO protein to essential proliferation-associated promoters. As a result, leukemia cells were inhibited with high effectivity and specificity. This approach can be utilized to target a wide variety of human cancers.
Collapse
|
409
|
Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, Thomas M, Berdel WE, Serve H, Müller-Tidow C. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene 2003; 22:8031-41. [PMID: 12970751 DOI: 10.1038/sj.onc.1206928] [Citation(s) in RCA: 1753] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Early-stage non-small cell lung cancer (NSCLC) can be cured by surgical resection, but a substantial fraction of patients ultimately dies due to distant metastasis. In this study, we used subtractive hybridization to identify gene expression differences in stage I NSCLC tumors that either did or did not metastasize in the course of disease. Individual clones (n=225) were sequenced and quantitative RT-PCR verified overexpression in metastasizing samples. Several of the identified genes (eIF4A1, thymosin beta4 and a novel transcript named MALAT-1) were demonstrated to be significantly associated with metastasis in NSCLC patients (n=70). The genes' association with metastasis was stage- and histology specific. The Kaplan-Meier analyses identified MALAT-1 and thymosin beta4 as prognostic parameters for patient survival in stage I NSCLC. The novel MALAT-1 transcript is a noncoding RNA of more than 8000 nt expressed from chromosome 11q13. It is highly expressed in lung, pancreas and other healthy organs as well as in NSCLC. MALAT-1 expressed sequences are conserved across several species indicating its potentially important function. Taken together, these data contribute to the identification of early-stage NSCLC patients that are at high risk to develop metastasis. The identification of MALAT-1 emphasizes the potential role of noncoding RNAs in human cancer.
Collapse
|
410
|
Steffen B, Serve H, Berdel WE, Agrawal S, Linggi B, Büchner T, Hiebert SW, Müller-Tidow C. Specific protein redirection as a transcriptional therapy approach for t(8;21) leukemia. Proc Natl Acad Sci U S A 2003; 100:8448-53. [PMID: 12819347 PMCID: PMC166249 DOI: 10.1073/pnas.1330293100] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Important progress has been achieved in the knowledge about the pathogenesis of cancer. However, despite these advances, the therapeutic strategies are still limited. Leukemias are often characterized by specific balanced translocations, with the t(8;21) balanced translocation being the most frequent chromosomal aberration in acute myeloid leukemia (AML). This translocation produces the AML1-ETO fusion protein, which binds to AML1 target promoter sequences. Transcriptional repression of AML1-dependent genes by AML1-ETO and associated corepressors represents the pathogenetic mechanisms of t(8;21). Here, we show that targeting of AML1-ETO to essential, MYB-dependent gene promoters induces t(8;21)-restricted cell death. We constructed a chimeric protein that contained the MYB DNA-binding domain and the AML1-binding domain of myeloid Elf-1-like factor (MEF). This protein associated with AML1-ETO and directed the complex to MYB-responsive promoters in vitro and in vivo. In the presence of AML1-ETO, the chimeric protein repressed the activity of MYB-responsive promoters, rapidly induced apoptosis, and specifically inhibited colony growth. All these effects occurred only in AML1-ETO-positive cells, whereas no adverse effects were observed in cells not expressing AML1-ETO. Taken together, this study demonstrates that redirection of oncogenic proteins can be used as a strategy to dramatically influence their cellular effects, with the ultimate goal to design highly specific therapies for cancer.
Collapse
MESH Headings
- Acute Disease
- Animals
- Apoptosis/physiology
- Binding Sites
- COS Cells
- 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
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Genes, myb
- Humans
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/pathology
- Leukemia, Myeloid/therapy
- Macromolecular Substances
- Mice
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Oncogene Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/physiology
- Promoter Regions, Genetic/genetics
- Protein Binding
- Protein Structure, Tertiary
- Proto-Oncogene Proteins c-kit
- Proto-Oncogene Proteins c-myb/chemistry
- Proto-Oncogene Proteins c-myb/physiology
- RUNX1 Translocation Partner 1 Protein
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/physiology
- Substrate Specificity
- Transcription Factors/chemistry
- Transcription Factors/genetics
- Transcription Factors/physiology
- Transcription, Genetic
- Translocation, Genetic
- Tumor Cells, Cultured
- Tumor Stem Cell Assay
Collapse
|
411
|
Müller-Tidow C, Kiehl M, Sindermann JR, Probst M, Banger N, Zühlsdorf M, Chou TC, Berdel WE, Serve H, Koch OM. Synergistic growth inhibitory effects of interferon-alpha and lovastatin on bcr-abl positive leukemic cells. Int J Oncol 2003; 23:151-8. [PMID: 12792788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
The chimeric bcr-abl tyrosine kinase is of crucial pathogenic importance in chronic myeloid leukemia (CML). As shown, bcr-abl activates the ras pathway by phosphorylation of adapter proteins such as Grb-2 and Crkl. Functional inhibition of p21ras might partially inhibit the mitogenic signaling by bcr-abl. By depletion of cellular mevalonate pools, p21ras proteins can be rendered non-functional as a result of deficient post-translational protein farnesylation. We investigated the pharmacologic effect of mevalonate depletion by lovastatin in conjunction with interferon-alpha 2b (INF-alpha 2b) in bcr-abl positive K562 cells. At various concentrations, both drugs synergistically reduced cell proliferation of CML line K562 in a liquid culture system as well as clonal growth of colony forming units in a patient with newly diagnosed CML. Lovastatin and IFN-alpha 2b in combination led to cell cycle arrest and resulted in significant reduction of phosphorylation on tyrosine, serine, and threonine protein residues. IFN-alpha 2b alone showed little effect on protein phosphorylation but strongly enhanced lovastatin driven loss of phosphorylation. Subsequently, DNA fragmentation occurred in 50% of cells. In conclusion, exposure to IFN-alpha 2b and lovastatin synergistically inhibited proliferation of bcr-abl positive cells and resulted in loss of protein phosphorylation and subsequent apoptosis in K562 cells. Our in vitro model suggests further investigations are required of the potential value of HMG-CoA reductase inhibitors as adjunct to therapy of CML with interferon.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Apoptosis
- Cell Cycle
- Cell Division
- Cell Line, Tumor
- DNA Fragmentation
- Dose-Response Relationship, Drug
- Flow Cytometry
- Fusion Proteins, bcr-abl/biosynthesis
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Interferon alpha-2
- Interferon-alpha/therapeutic use
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Lovastatin/therapeutic use
- Mevalonic Acid/metabolism
- Phosphorylation
- Protein Prenylation
- Protein Processing, Post-Translational
- Proto-Oncogene Proteins p21(ras)/metabolism
- Recombinant Proteins
- Serine/chemistry
- Threonine/chemistry
- Time Factors
- Tyrosine/chemistry
Collapse
|
412
|
Mizuki M, Schwable J, Steur C, Choudhary C, Agrawal S, Sargin B, Steffen B, Matsumura I, Kanakura Y, Böhmer FD, Müller-Tidow C, Berdel WE, Serve H. Suppression of myeloid transcription factors and induction of STAT response genes by AML-specific Flt3 mutations. Blood 2003; 101:3164-73. [PMID: 12468433 DOI: 10.1182/blood-2002-06-1677] [Citation(s) in RCA: 233] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The receptor tyrosine kinase Flt3 is expressed and functionally important in early myeloid progenitor cells and in the majority of acute myeloid leukemia (AML) blasts. Internal tandem duplications (ITDs) in the juxtamembrane domain of the receptor occur in 25% of AML cases. Previously, we have shown that these mutations activate the receptor and induce leukemic transformation. In this study, we performed genome-wide parallel expression analyses of 32Dcl3 cells stably transfected with either wild-type or 3 different ITD isoforms of Flt3. Comparison of microarray expression analyses revealed that 767 of 6586 genes differed in expression between FLT3-WT- and FLT3-ITD-expressing cell lines. The target genes of mutationally activated Flt3 resembled more closely those of the interleukin 3 (IL-3) receptor than those of ligand-activated Flt3. The serine-threonine kinase Pim-2 was up-regulated on the mRNA and the protein level in Flt3-ITD-expressing cells. Further experiments indicated that Pim-2 function was important for clonal growth of 32D cells. Several genes repressed by the mutations were found to be involved in myeloid gene regulation. Pu.1 and C/EBPalpha, both induced by ligand-activation of wild-type Flt3, were suppressed in their expression and function by the Flt3 mutations. In conclusion, internal tandem duplication mutations of Flt3 activate transcriptional programs that partially mimic IL-3 activity. Interestingly, other parts of the transcriptional program involve novel, IL-3-independent pathways that antagonize differentiation-inducing effects of wild-type Flt3. The identification of the transcriptional program induced by ITD mutations should ease the development of specific therapies.
Collapse
|
413
|
Müller-Tidow C, Readhead C, Cohen AH, Asotra K, Idos G, Diederichs S, Cauvet T, Yang R, Berdel WE, Serve H, Koeffler HP. Successive increases in human cyclin A1 promoter activity during spermatogenesis in transgenic mice. Int J Mol Med 2003; 11:311-5. [PMID: 12579332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
The human cyclin A1 gene is highly expressed in pachytene spermatocytes and is essential for spermatogenesis. To analyze mechanisms of cyclin A1 gene expression in vivo, we cloned a 1.3 kb fragment of the promoter upstream of the cDNA of enhanced green fluorescent protein (EGFP). Four lines of transgenic mice were generated that carried the transgene. Cyclin A1 promoter activity in the organs of the transgenic mice was analyzed using fluorescence microscopy and flow cytometry. Expression of EGFP was seen in male germ cells of all four murine lines. Spermatogonia at the basal membrane expressed low levels of EGFP, but bright green fluorescence was present in spermatocytes entering meiosis. Interestingly, a further sharp increase in EGFP expression was found in spermatocytes approximately at the stage of the first meiotic division. EGFP levels stayed high thereafter and EGFP was present in mature spermatozoa. A portion of c-kit expressing cells in the testis also expressed EGFP indicating cyclin A1 promoter activity in a subpopulation of spermatogonia. These data suggest that cyclin A1 is active not only in pachytene spermatocytes but also in earlier phases of spermatogenesis.
Collapse
|
414
|
Müller-Tidow C, Readhead C, Cohen A, Asotra K, Idos G, Diederichs S, Cauvet T, Yang R, Berdel W, Serve H, Koeffler H. Successive increases in human cyclin A1 promoter activity during spermatogenesis in transgenic mice. Int J Mol Med 2003. [DOI: 10.3892/ijmm.11.3.311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
415
|
Müller-Tidow C, Diederichs S, Schrader MG, Vogt U, Miller K, Berdel WE, Serve H. Cyclin A1 is highly expressed in aggressive testicular germ cell tumors. Cancer Lett 2003; 190:89-95. [PMID: 12536081 DOI: 10.1016/s0304-3835(02)00582-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cyclin A1 is a tissue-specific A-type cyclin that is essential for spermatogenesis. Overexpression of cyclin A1 was found in acute myeloid leukemia and cyclin A1 induced leukemia in a transgenic mouse model. We used quantitative real-time reverse transcription-polymerase chain reaction to analyze cyclin A1 expression in solid tumors. Cyclin A1 expression was very low in breast cancer, non-small cell lung cancer and in cervical carcinoma. However, substantial expression of cyclin A1 was found in testicular and ovarian cancer and in endometrial cancer. In testis specimens, cyclin A1 expression was much higher in testicular tumors compared to Sertoli cell only syndrome that lacks spermatogenesis. Compared to normal spermatogenesis, testicular cancers expressed on average lower levels of cyclin A1. Among the different histological subtypes of testicular tumors, embryonal cell carcinomas and immature teratomas expressed the highest levels of cyclin A1. The cyclin A1 levels in these tumors were similar to those seen in normal testis. Seminomas and yolk sac tumors expressed intermediate levels, whereas cyclin A1 expression was very low in mature teratomas. These findings indicate that cyclin A1 is expressed in selected solid tumors. Its known oncogenic function and the high expression levels in aggressive testicular tumors suggest a role for cyclin A1 in germ cell tumorigenesis.
Collapse
|
416
|
Readhead C, Müller-Tidow C. Genes associated with the development of the male germ line. Reprod Biomed Online 2003; 4 Suppl 1:52-7. [PMID: 12470336 DOI: 10.1016/s1472-6483(12)60012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The development of the mammalian germ line has been well studied, from the designation of primordial germ cells and their migration in the embryo to their progression through gametogenesis. The pattern of germ cell development, as established through classical studies, is now being overlaid with molecular, genetic and epigenetic data. Eventually, proteonomics will lead to a deeper understanding of the function of these genes. Through knowledge of germ cell gene expression patterns, it is now possible to develop transgenic molecular tools for the isolation of germ cells at different stages of development. By linking stage-specific germ cell promoter regions to the green fluorescent protein (GFP) reporter gene it is possible to tag these cells genetically for histological identification and cell sorting. Our long-term goal is to develop male germ cells as stem cells for therapeutic purposes. It is hoped that this goal will be achieved by purifying germ cells at different stages in development and gaining a deeper understanding of them by studying their gene expression patterns, potency and plasticity, both in vivo and in vitro.
Collapse
|
417
|
Müller-Tidow C, Steur C, Mizuki M, Schwäble J, Brandts C, Berdel WE, Serve H. [Mutations of growth factor receptor Flt3 in acute myeloid leukemia: transformation of myeloid cells by Ras-dependent and Ras-independent mechanisms]. Dtsch Med Wochenschr 2002; 127:2195-200. [PMID: 12397548 DOI: 10.1055/s-2002-34942] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND AND OBJECTIVE The tyrosine kinase receptor Flt3 mediates important functions in early hematopoietic progenitors. Recently mutations of a growth factor receptor have been identified in about 30 % of patients with acute myeloid leukemia (AML). These mutations are associated with a poor prognosis. In-vitro and animal data show their involvement in leukemic transformation. Experiments analyzing the effects of these mutations on signal transduction and gene expression patterns of myeloid cells allow for the classification of this receptor as an oncogene. Furthermore, they help to define the receptor and its signaling intermediates as therapeutic targets. METHODS In order to analyze the signaling properties of mutated FLT3 receptors, we isolated the receptor mRNA from two patients with AML. Wild-type and mutant Flt3 isoforms were expressed in 32D cells that were subsequently analyzed for proliferation, survival, activation of signaling intermediates and gene expression levels. Also, the effects of of Ras-, MAP-Kinase and PI3-Kinase inhibition were analyzed. RESULTS The expression of mutated Flt3 (Flt3-ITD) induced factor-independent proliferation and survival in the myeloid progenitor cell line 32D. Flt3-ITD activated Ras- and PI3-kinase-dependent signaling pathways, as well as STAT5 and STAT3. Activation of STAT proteins was followed by the induction of known STAT target genes like SOCS2, SOCS3 and CIS. Inhibition of Ras-dependent signal transduction by a dominant negative Ras construct inhibited some, but not all biological effects of Flt3-ITD. Similar results were obtained by chemical inhibition of the MAP kinases. In contrast, inhibition of PI3 kinase activity inhibited growth factor-independent growth and apoptosis resistance of 32D cells. CONCLUSIONS Inhibition of Ras-dependent signaling pathways is not sufficient to abrogate the functional consequences of Flt3-mutations in myeloid cells. Therefore, therapeutic intervention by Ras-Inhibitors may not be sufficient to treat Flt3-driven disease.
Collapse
|
418
|
Schrader M, Müller-Tidow C, Ravnik S, Müller M, Schulze W, Diederichs S, Serve H, Miller K. Cyclin A1 and gametogenesis in fertile and infertile patients: a potential new molecular diagnostic marker. Hum Reprod 2002; 17:2338-43. [PMID: 12202422 DOI: 10.1093/humrep/17.9.2338] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The study aim was to evaluate cyclin A1 mRNA expression levels as a potential molecular diagnostic parameter in the work-up of testicular tissue from fertile versus infertile patients. METHODS Cyclin A1 expression was quantified in 55 cryopreserved testicular tissue specimens by fluorescence real-time RT-PCR. A conventional histological work-up was performed concomitantly in all tissue specimens with additional semi-thin sectioning in all cases of non-obstructive azoospermia (n = 12), maturation arrest (n = 17) and Sertoli cell-only syndrome (SCOS; n = 9). RESULTS The mean (+/- SD) normalized cyclin A1 expression (N(CyclinA1)) was 3.82 +/- 2.23 relative gene expression (RGE) in tissue specimens with normal spermatogenesis, and 0.625 +/- 0.221 RGE in those with maturation arrest at the level of early spermatids. Only minimal N(CyclinA1) was detected in tissue specimens with spermatogonia only or maturation arrest at the level of primary spermatocytes (0.005 +/- 0.008). Cyclin A1 expression was absent in the majority of SCOS specimens (0.002 +/- 0.002). CONCLUSIONS These investigations suggested that cyclin A1 expression is altered in cases of spermatogenic disorders. Moreover, the level of cyclin A1 mRNA expression correlates with gametogenic disorders and seems well suited for a molecular-diagnostic classification supplementing the histopathological evaluation of spermatogenic disorders.
Collapse
|
419
|
Schrader M, Ravnik S, Müller-Tidow C, Müller M, Straub B, Diedrichs S, Serve H, Miller K. Quantification of cyclin A1 and glyceraldehyde-3-phosphate dehydrogenase expression in testicular biopsies of infertile patients by fluorescence real-time RT-PCR. INTERNATIONAL JOURNAL OF ANDROLOGY 2002; 25:202-9. [PMID: 12121569 DOI: 10.1046/j.1365-2605.2002.00343.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The objective of this study was to establish a classification of meiotic disorders by detection of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and cyclin A1 mRNA in patients presenting with non-obstructive azoospermia. GAPDH and cyclin A1 expression levels were detected in 37 histologically classified testicular tissue specimens by one-line RT-PCR. Levels of cyclin A1 expression (NCyclinA1) were high in tissue specimens with full spermatogenesis [3.519 +/- 3.141 (mean +/- SD)] but only minimal in those without haploid germ cells. A lack of cyclin A1 expression was seen in most sertoli-cell-only syndrome (SCOS) specimens. The criteria for an ideal internal standard were only partially met by GAPDH, as we detected decreased expression in tissue samples with maturation arrest [(268.2 +/- 106.2 relative gene expression (mean +/- SD)] and SCOS (201.7 +/- 95.2) compared to those with normal histological findings (325.1 +/- 129.3). It was concluded that the level of cyclin A1 mRNA expression predicts meiotic disorders during spermatogenesis. GAPDH is not an ideal internal standard for specific gene expression in testicular tissue specimens.
Collapse
|
420
|
Padró T, Bieker R, Ruiz S, Steins M, Retzlaff S, Bürger H, Büchner T, Kessler T, Herrera F, Kienast J, Müller-Tidow C, Serve H, Berdel WE, Mesters RM. Overexpression of vascular endothelial growth factor (VEGF) and its cellular receptor KDR (VEGFR-2) in the bone marrow of patients with acute myeloid leukemia. Leukemia 2002; 16:1302-10. [PMID: 12094254 DOI: 10.1038/sj.leu.2402534] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2001] [Accepted: 02/18/2002] [Indexed: 11/09/2022]
Abstract
Vascular endothelial growth factor (VEGF) and its cellular receptor VEGFR-2 have been implicated as the main endothelial pathway required for tumor neovascularization. However, the importance of the VEGF/VEGFR-2 system for angiogenesis in hematologic malignancies such as AML remains to be elucidated. In 32 patients with newly diagnosed untreated AML, we observed by immunohistochemical analysis of bone marrow biopsies significantly higher levels of VEGF and VEGFR-2 expression than in 10 control patients (P <0.001). In contrast, VEGFR-1 staining levels in AML patients were in the same range as in the controls. Expression of VEGF and VEGFR-2 was significantly higher in patients with a high degree of microvessel density compared to those with a low degree (VEGF: P =0.024; VEGFR-2: P =0.040) and correlated well with bone marrow microvessel density (r(s)=0.566 and 0.609, respectively; P <0.001). Furthermore, in patients who achieved a complete remission following induction chemotherapy VEGFR-2 staining levels decreased into the normal range. In conclusion, our results provide evidence for increased expression of VEGF/VEGFR-2 of leukemic blasts and correlation with angiogenesis in the bone marrow of AML patients. Thus, VEGF/VEGFR-2 might constitute promising targets for antiangiogenic and antileukemic treatment strategies in AML.
Collapse
MESH Headings
- Acute Disease
- Adult
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bone Marrow Cells/metabolism
- Bone Marrow Cells/pathology
- Endothelial Growth Factors/biosynthesis
- Endothelial Growth Factors/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Lymphokines/biosynthesis
- Lymphokines/genetics
- Middle Aged
- Neovascularization, Pathologic
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/genetics
- Receptors, Growth Factor/biosynthesis
- Receptors, Growth Factor/genetics
- Receptors, Mitogen/biosynthesis
- Receptors, Mitogen/genetics
- Receptors, Vascular Endothelial Growth Factor
- Remission Induction
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
|
421
|
Linggi B, Müller-Tidow C, van de Locht L, Hu M, Nip J, Serve H, Berdel WE, van der Reijden B, Quelle DE, Rowley JD, Cleveland J, Jansen JH, Pandolfi PP, Hiebert SW. The t(8;21) fusion protein, AML1 ETO, specifically represses the transcription of the p14(ARF) tumor suppressor in acute myeloid leukemia. Nat Med 2002; 8:743-50. [PMID: 12091906 DOI: 10.1038/nm726] [Citation(s) in RCA: 213] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The t(8;21) is one of the most frequent chromosomal translocations associated with acute leukemia. This translocation creates a fusion protein consisting of the acute myeloid leukemia-1 transcription factor and the eight-twenty-one corepressor (AML1 ETO), which represses transcription through AML1 (RUNX1) DNA binding sites and immortalizes hematopoietic progenitor cells. We have identified the p14(ARF) tumor suppressor, a mediator of the p53 oncogene checkpoint, as a direct transcriptional target of AML1 ETO. AML1 ETO repressed the p14(ARF) promoter and reduced endogenous levels of p14(ARF) expression in multiple cell types. In contrast, AML1 stimulated p14(ARF) expression and induced phenotypes consistent with cellular senescence. Chromatin immunoprecipitation assays demonstrated that AML1 ETO was specifically bound to the p14(ARF) promoter. In acute myeloid leukemia samples containing the t(8;21), levels of p14(ARF) mRNA were markedly lower when compared with other acute myeloid leukemias lacking this translocation. Repression of p14(ARF) may explain why p53 is not mutated in t(8;21)-containing leukemias and suggests that p14(ARF) is an important tumor suppressor in a large number of human leukemias.
Collapse
MESH Headings
- Antigens, CD/analysis
- CD4 Antigens/analysis
- Chromosomes, Human, Pair 21
- Chromosomes, Human, Pair 8
- Core Binding Factor Alpha 2 Subunit
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Genes, Tumor Suppressor
- Hematopoietic Stem Cells/pathology
- Humans
- K562 Cells
- Leukemia, Myeloid, Acute/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Plasmids
- RUNX1 Translocation Partner 1 Protein
- Repressor Proteins/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic/physiology
- Translocation, Genetic
- Tumor Cells, Cultured
- Tumor Suppressor Protein p14ARF/genetics
Collapse
|
422
|
Wang WB, Böing S, Zhou XQ, Ji P, Dong Y, Yao Q, Müller-Tidow C. Identification of metastasis-associated genes in early stage non-small cell lung cancer by subtractive hybridization. SHENG WU HUA XUE YU SHENG WU WU LI XUE BAO ACTA BIOCHIMICA ET BIOPHYSICA SINICA 2002; 34:273-8. [PMID: 12019437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of death and a substantial fraction of patients with surgically resected disease ultimately dies due to distant metastasis. To identify gene expression differences in early stage adenocarcinoma that either did or did not metastasize within a 5-year period, we employed a subtractive hybridization strategy of pooled RNA from primary adenocarcinomas (stage I) of the lung. Individual clones (n=225) of the subtracted cDNA library were sequenced. Further analyses of mRNA expression levels in a cohort of 70 NSCLC patients (stage I to IIIA) showed that the metastasis association of the identified genes was stage and histology specific. Cox regression analyses identified two genes (EIF4A1, MALA1) to be independent prognostic parameters for patients' survival in stage I and II disease. These findings could help to identify early-stage NSCLC patients at high risk for the development of distant metastasis.
Collapse
|
423
|
Müller-Tidow C, Kügler K, Diederichs S, Klümpen S, Möller M, Vogt U, Metzger R, Schneider PM, Berdel WE, Serve H. Loss of expression of HDAC-recruiting methyl-CpG-binding domain proteins in human cancer. Br J Cancer 2001; 85:1168-74. [PMID: 11710831 PMCID: PMC2375156 DOI: 10.1054/bjoc.2001.2041] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dysregulation of CpG-methylation is a common feature of many human cancers and tumour suppressor genes can be silenced by hypermethylation. Recently, 2 methyl-CpG-binding domain proteins have been linked to gene inactivation by their ability to recruit co-repressors and HDAC-activity to methylated gene promoters. Here, we have analysed mRNA expression of these genes, MeCP2 and MBD2, in a wide variety of primary human tumours. In solid tumours, expression levels of MBD2 (57/71) and MeCP2 (64/71) were significantly reduced in the majority of primary tumours as detected by quantitative real-time RT-PCR. Western blot analyses of MeCP2 in matched tumour-normal samples of patients with non-small-cell lung cancer (NSCLC) indicated reduced protein in a significant percentage of patients. In acute myelogenous leukaemia (n = 26), expression levels were only slightly reduced and did not differ between samples analysed at diagnosis or at the time of relapse. In early-stage NSCLC (n = 70) expression of MeCP2 and MBD2 was significantly lower in squamous cell carcinoma than in adenocarcinoma or large cell carcinoma (P = 0.03 and P = 0.01). To further elucidate the mechanisms of gene regulation, we analysed MeCP2 and MBD2 regulation during haematopoietic differentiation. No significant changes in MeCP2 or MBD2 expression were found when NB4 cells were differentiated toward granulocytes suggesting that neither differentiation nor cell cycle status were relevant for the reduced expression of these genes in human cancer. In conclusion, the significant loss of MeCP2 and MBD2 expression in human cancers suggests a potential role of this phenomenon in the development of solid human tumours.
Collapse
|
424
|
Müller-Tidow C, Wang W, Idos GE, Diederichs S, Yang R, Readhead C, Berdel WE, Serve H, Saville M, Watson R, Koeffler HP. Cyclin A1 directly interacts with B-myb and cyclin A1/cdk2 phosphorylate B-myb at functionally important serine and threonine residues: tissue-specific regulation of B-myb function. Blood 2001; 97:2091-7. [PMID: 11264176 DOI: 10.1182/blood.v97.7.2091] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cyclin A1 is tissue-specifically expressed during spermatogenesis, but it is also highly expressed in acute myeloid leukemia (AML). Its pathogenetic role in AML and in the cell cycle of leukemic blasts is unknown. B-myb is essential for G1/S transition and has been shown to be phosphorylated by the cyclin A2/cdk2 complex. Here it is demonstrated that cyclin A1 interacts with the C-terminal portion of B-myb as shown by glutathione S-transferase (GST) precipitation. This interaction is confined to cyclin A1 because binding could not be detected between cyclin A2 and B-myb. Also, cdk2 was not pulled down by GST-B-myb from U937 lysates. In addition, co-immunoprecipitation of cyclin A1 and B-myb in leukemic cells evidenced protein interaction in vivo. Baculovirus-expressed cyclin A1/cdk2 complexes were able to phosphorylate human as well as murine B-myb in vitro. Tryptic phosphopeptide mapping revealed that cyclin A1/cdk2 complexes phosphorylated the C-terminal part of B-myb at several sites including threonine 447, 490, and 497 and serine 581. These phosphorylation sites have been demonstrated to be important for the enhancement of B-myb transcriptional activity. Further studies showed that cyclin A1 cooperated with B-myb to transactivate myb binding site containing promoters including the promoter of the human cyclin A1 gene. Taken together, the data suggest that cyclin A1 is a tissue-specific regulator of B-myb function and activates B-myb in leukemic blasts. (Blood. 2001;97:2091-2097)
Collapse
|
425
|
Müller-Tidow C, Bornemann C, Diederichs S, Westermann A, Klümpen S, Zuo P, Wang W, Berdel WE, Serve H. Analyses of the genomic methylation status of the human cyclin A1 promoter by a novel real-time PCR-based methodology. FEBS Lett 2001; 490:75-8. [PMID: 11172814 DOI: 10.1016/s0014-5793(01)02128-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The role of CpG methylation in the regulation of tissue-specific gene expression is highly controversial. Cyclin A1 is a tissue-specifically expressed gene that is strongly methylated in non-expressing tumor cell lines. We have established a novel real-time PCR method to quantitate genomic CpG methylation of the cyclin A1 promoter. Genomic DNA samples from different human organs were treated with bisulfite and amplified with methylation-specific primers and with primers amplifying methylated as well as non-methylated DNA. PCR product quantitation was obtained by using a fluorogenic probe labeled with FAM and TAMRA. These analyses demonstrated that the human cyclin A1 promoter was methylated in kidney, colon, spleen, testis, and small intestine, but not in brain, liver, pancreas, or heart. Expression of cyclin A1 was predominantly found in testis. Low level expression of cyclin A1 was present in spleen, prostate, leukocytes, colon, and thymus. Taken together, our data provide evidence that CpG methylation patterns of the human cyclin A1 promoter in human organs do not generally correlate with cyclin A1 gene expression in vivo.
Collapse
|