Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Leukemia ; 26(4): 644-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21960247

ABSTRACT

Expression of c-Myb is required for normal hematopoiesis and for proliferation of myeloid leukemia blasts and a subset of T-cell leukemia, but its role in B-cell leukemogenesis is unknown. We tested the role of c-Myb in p190(BCR/ABL)-dependent B-cell leukemia in mice transplanted with p190(BCR/ABL)-transduced marrow cells with a c-Myb allele (Myb(f/d)) and in double transgenic p190(BCR/ABL)/Myb(w/d) mice. In both models, loss of a c-Myb allele caused a less aggressive B-cell leukemia. In p190(BCR/ABL)-expressing human B-cell leukemia lines, knockdown of c-Myb expression suppressed proliferation and colony formation. Compared with c-Myb(w/f) cells, expression of Bmi1, a regulator of stem cell proliferation and maintenance, was decreased in pre-B cells from Myb(w/d) p190(BCR/ABL) transgenic mice. Ectopic expression of a mutant c-Myb or Bmi1 enhanced the proliferation and colony formation of Myb(w/d) p190(BCR/ABL) B-cells; by contrast, Bmi1 downregulation inhibited colony formation of p190(BCR/ABL)-expressing murine B cells and human B-cell leukemia lines. Moreover, c-Myb interacted with a segment of the human Bmi1 promoter and enhanced its activity. In blasts from 19 Ph(1) adult acute lymphoblastic leukemia patients, levels of c-Myb and Bmi1 showed a positive correlation. Together, these findings support the existence of a c-Myb-Bmi1 transcription-regulatory pathway required for p190(BCR/ABL) leukemogenesis.


Subject(s)
Cell Transformation, Neoplastic , Fusion Proteins, bcr-abl/physiology , Leukemia, B-Cell/etiology , Mitogen-Activated Protein Kinase 7/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-myb/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Humans , Leukemia, B-Cell/pathology , Mice , Mice, Inbred C57BL , Polycomb Repressive Complex 1
2.
Oncogene ; 25(19): 2758-72, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16407849

ABSTRACT

The Myb proto-oncogene encodes a transcription factor (c-Myb) that is essential for normal hematopoiesis and is thought to regulate hematopoietic cell proliferation and differentiation by regulating expression of specific target genes. We identify the mouse erythroid-specific carbonic anhydrase I promoter (CAIe) as a target of c-Myb activity and demonstrate that Myb activity is critical for carbonic anhydrase I (CAI) expression in C19 MEL cells. CAI expression is downregulated when MEL cells differentiate in response to MEnT or treatment with N, N-hexamethylene bisacetamide (HMBA). Coexpression of GATA-1 with c-Myb results in synergistic activation of transcription from the CAIe promoter and both transcription factors interact with the CAIe promoter in vivo. We identify a novel 20 bp sequence in the CAIe promoter that is sufficient to mediate synergistic activation of the CAIe promoter by c-Myb and GATA-1. c-Myb and GATA-1 interact with this DNA sequence suggesting that c-Myb and GATA-1 may be contained in a complex that interacts with this region of the CAIe promoter. Forced expression of CAI delayed HMBA-induced differentiation of MEL cells and maintained them in a proliferating state. These data strongly suggest that CAI is a c-Myb target and is involved in regulating MEL cell proliferation and differentiation.


Subject(s)
Carbonic Anhydrase I/genetics , Cell Differentiation , Gene Expression Regulation, Neoplastic , Leukemia, Erythroblastic, Acute/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myb/metabolism , Acetamides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Carbonic Anhydrase I/metabolism , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Genes, Dominant , Leukemia, Erythroblastic, Acute/enzymology , Leukemia, Erythroblastic, Acute/pathology , Mice , Molecular Sequence Data , Tumor Cells, Cultured
3.
Leukemia ; 16(1): 127-34, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11840272

ABSTRACT

Multiple myeloma (MM) is a clonal neoplasm of plasma cells which offers an excellent model to study multistep molecular oncogenesis. In 20-25% of primary tumors and cell lines examined, cyclin D1 is overexpressed due to the translocation t(11;14)(q13;q32). We have characterized cyclin-dependent kinase inhibitor p15 (CDKN2B), p16 (CDKN2A) and p18 (CDKN2C) deletions in cyclin D1-expressing and non-expressing MM cell lines. p18 was found to be frequently deleted (38%); in some cases p18 deletions coexisted with hemizygous p16 deletion. To examine the function of p18 as a putative tumor suppressor in myeloma cells, a zinc-inducible p18 construct was stably transfected into KMS12, a MM cell line with biallelic p18 and monoallelic p16 deletions as well as cyclin D1 overexpression. Ectopic expression of p18 caused 40-45% growth suppression as determined by trypan blue exclusion and MTS assays. p18 induction also resulted in apoptosis, suggesting that inhibition of the cyclin D1/CDK/pRb pathway in these tumor cells could be a crucial step toward the induction of tumor regression via apoptotic cell death. This cell cycle pathway is thus frequently mutated and provides a potentially novel target for gene therapeutic or pharmacologic approaches to human myeloma.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins , Cyclin-Dependent Kinases , Gene Deletion , Genes, Tumor Suppressor , Multiple Myeloma/genetics , Neoplasm Proteins/genetics , Tumor Suppressor Proteins/genetics , Apoptosis/genetics , Cell Cycle , Cell Division , Cyclin D1/physiology , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p18 , Enzyme Inhibitors , Genotype , Humans , Lymphoma, Mantle-Cell/pathology , Multiple Myeloma/pathology , Neoplasm Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured , Tumor Suppressor Proteins/physiology
4.
Blood Cells Mol Dis ; 27(2): 429-36, 2001.
Article in English | MEDLINE | ID: mdl-11500057

ABSTRACT

Friend murine erythroleukemia (MEL) cells provide an early erythroid precursor model that can be induced to terminally differentiate in cell culture and has been used to study erythroid differentiation as well as multistage tumorigenesis. During the chemically induced differentiation of MEL cells, expression of the c-myb protooncogene is downregulated in a biphasic fashion and forced expression of c-myb is able to block the differentiation process, suggesting that c-myb activity may be limiting for differentiation in MEL cells. We have recently produced stable transfectants in the C19 MEL cell line that carry a dominant interfering myb allele (MEnT) under the control of an inducible mouse metallothionein I (MTH) promoter. Upon inducing expression of MEnT, transfected cells enter a differentiation program and begin to produce alpha-globin mRNA, assemble hemoglobin, and stop proliferating. Differential display was used to compare mRNA expression between parental C19 MEL cells induced to differentiate with hexamethylene bisacetamide (HMBA) and stable transfectants induced to differentiate via expression of MEnT to identify potential Myb target promoters. We identified six candidate cDNAs in this fashion and present evidence that two of these represent genes that are dependent on c-Myb activity for maximal expression in MEL cells.


Subject(s)
Genes, myb , Animals , Gene Expression Regulation, Neoplastic , Gene Targeting , Leukemia, Erythroblastic, Acute/genetics , Mice , Promoter Regions, Genetic , Tumor Cells, Cultured
5.
Blood ; 94(12): 4255-62, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10590070

ABSTRACT

Cyclin A1 differs from other cyclins in its highly restricted expression pattern. Besides its expression during spermatogenesis, cyclin A1 is also expressed in hematopoietic progenitor cells and in acute myeloid leukemia. We investigated mechanisms that might contribute to cyclin A1 expression in hematopoietic cells. Comparison of cyclin A1 and cyclin A promoter activity in adherent and myeloid leukemia cell lines showed that the cyclin A1 promoter is preferentially active in myeloid cell lines. This preferential activity was present in a small, 335-bp cyclin A1 promoter fragment that contained several potential c-myb binding sites. Coexpression of a c-myb expression vector with the cyclin A1 promoter constructs significantly increased the reporter activity in adherent CV-1 as well as in myeloid U937 cells. Gel-shift assays demonstrated that c-myb could bind to the cyclin A1 promoter at a binding site located near the transcription start site. Site-directed mutagenesis of this site decreased promoter transactivation by 50% in both KCL22 cells that express high levels of c-myb and in CV-1 cells that were transfected with c-myb. In addition, transfection of primary human embryonic fibroblasts with a c-myb expression vector led to induction of the endogenous cyclin A1 gene. Taken together, c-myb can directly transactivate the promoter of cyclin A1, and c-myb might be involved in the high-level expression of cyclin A1 observed in acute myeloid leukemia. These findings suggest that c-myb induces hematopoiesis-specific mechanisms of cell cycle regulation.


Subject(s)
Cyclin A/genetics , Gene Expression Regulation, Neoplastic , Genes, myb , Promoter Regions, Genetic , Base Sequence , Cloning, Molecular , Cyclin A1 , HeLa Cells , Humans , Molecular Sequence Data , Plasmids
6.
Genes Dev ; 13(9): 1073-8, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10323859

ABSTRACT

The c-Myb transcription factor is important for fetal hematopoiesis and has been proposed to mediate later stages of lymphocyte development. Using homozygous null c-Myb/Rag1 chimeric mice, we have determined that c-Myb plays an important role in the differentiation of macrophages and lymphocytes from precursor stem cells. We also determine that deletion of c-Myb leads to a complete block in early T cell development just before the oligopotent thymocyte matures into the definitive T cell precursor. These data indicate that c-Myb plays an important role at multiple stages of hematopoiesis and is required at an early stage of T cell development.


Subject(s)
Oncogenes , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Cell Differentiation/genetics , Genes, RAG-1 , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hyaluronan Receptors/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Receptors, Interleukin-2/metabolism , T-Lymphocytes/immunology
7.
Endocrinology ; 139(10): 4164-74, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9751496

ABSTRACT

Hormone-dependent breast cancer responds to primary therapies that block estrogen production or action, but tumor regrowth often occurs 12-18 months later. Additional hormonal treatments that further reduce estrogen synthesis or more effectively block its action cause additional remissions, but the mechanisms responsible for these secondary responses are not well understood. As a working hypothesis, we postulated that primary hormonal therapy induces adaptive changes, resulting in enhanced estrogen receptor (ER) expression and target gene activation and, further, that secondary treatment modalities interfere with these receptor-mediated transcriptional pathways. To test this hypothesis, we used an MCF-7 breast cancer model system involving deprivation of estradiol in culture for a prolonged period. These long-term estradiol-deprived (LTED) cells adapt by acquiring the ability to regrow in the absence of added estradiol. The experimental paradigm involved the comparison of wild-type cells with LTED cells. As endpoints, we directly assessed ER expression at the messenger RNA-, protein-, and ligand-binding levels and ER functionality by quantitating reporter gene activation and expression of endogenous estrogen target gene messenger RNA, as well as ER coactivator levels. Our data demonstrated an adaptive increase in ER expression and in basal ER functionality, as assessed by read-out of three different transfected reporters in LTED, as opposed to wild-type MCF-7 cells. Increased reporter gene read-out was dramatically inhibited by the pure antiestrogen ICI 182,780. As verification that endogenous (as well as transfected) estrogen target genes had enhanced transcription, we found that the basal levels of c-myb and c-myc message were substantially increased in LTED cells and could be inhibited by antiestrogen. Interestingly, the levels of c-myb and c-myc message in the LTED cells seemed to be increased out of proportion to the degree of ER reporter gene activation and were similar to those in wild-type cells maximally stimulated with estradiol. In addition, not all estrogen-responsive genes were activated, because transforming growth factor-alpha message level was not increased in LTED cells. Up-regulation of the steroid receptor coactivator SRC-1 did not seem to mediate the process of enhanced ER-induced transcription. Considering these observations together, we suggest that long-term estradiol deprivation causes adaptive processes that not only involve up-regulation of the ER but also influence the specificity and magnitude of activation of estrogen-responsive genes.


Subject(s)
Breast Neoplasms/metabolism , Estradiol/physiology , Receptors, Estrogen/physiology , Breast Neoplasms/pathology , Cell Division , Female , Genes, myc , Humans , Oncogenes , RNA, Messenger/analysis , Tumor Cells, Cultured
8.
J Biol Chem ; 271(37): 22697-705, 1996 Sep 13.
Article in English | MEDLINE | ID: mdl-8798443

ABSTRACT

The c-myb protooncogene encodes a highly conserved 75-89-kDa transcription factor that contains three functional domains, an amino-terminal DNA binding domain (DBD), a central acidic transactivation domain, and a carboxyl-terminal negative regulatory domain (NRD). Two acute transforming retroviruses, avian myeloblastosis virus and the E26 leukemia virus, transduced portions of c-myb and encode Myb proteins that are truncated in both the DBD and the NRD. Several conserved potential sites for phosphorylation by proline-directed serine/threonine protein kinases reside in or near the NRD, suggesting that phosphorylation might play a role in regulating c-Myb. We have previously demonstrated that serine 528, located in the NRD, is a target for p42(mapk) in vitro. Serine 528 is phosphorylated in vivo in several cell lines, and substitution of serine 528 to alanine (S528A) resulted in an increased ability of Myb to transactivate a synthetic promoter containing five copies of the mim-1A Myb-responsive element and a minimal herpes tk promoter. We have tested the ability of S528A Myb to transactivate a series of cellular target promoters and report that the serine to alanine substitution increased the ability of Myb to activate transcription from the CD34 promoter but not the c-myc or mim-1 promoters. This suggests that phosphorylation of serine 528 may differentially regulate c-Myb activity at different promoters. The DNA binding and multimerization activities of c-Myb appear to be unaffected by the S528A substitution, suggesting that phosphorylation of serine 528 may mediate its effect on the transcription transactivating activity of c-Myb by regulating interactions with other proteins.


Subject(s)
Gene Expression Regulation , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Alanine , Animals , Avian Myeloblastosis Virus , Binding Sites , Cell Line , Chlorocebus aethiops , DNA/metabolism , Mice , Molecular Weight , Phosphorylation , Proline/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myb , Serine , Structure-Activity Relationship , Trans-Activators/genetics , Transcription, Genetic , Transcriptional Activation
9.
Proc Natl Acad Sci U S A ; 92(14): 6429-33, 1995 Jul 03.
Article in English | MEDLINE | ID: mdl-7604007

ABSTRACT

The c-myb protooncogene encodes a highly conserved transcription factor that functions as both an activator and a repressor of transcription. The v-myb oncogenes of E26 leukemia virus and avian myeloblastosis virus encode proteins that are truncated at both the amino and the carboxyl terminus, deleting portions of the c-Myb DNA-binding and negative regulatory domains. This has led to speculation that the deleted regions contain important regulatory sequences. We previously reported that the 42-kDa mitogen-activated protein kinase (p42mapk) phosphorylates chicken and murine c-Myb at multiple sites in the negative regulatory domain in vitro, suggesting that phosphorylation might provide a mechanism to regulate c-Myb function. We now report that three tryptic phosphopeptides derived from in vitro phosphorylated c-Myb comigrate with three tryptic phosphopeptides derived from metabolically labeled c-Myb immunoprecipitated from murine erythroleukemia cells. At least two of these peptides are phosphorylated on serine-528. Replacement of serine-528 with alanine results in a 2- to 7-fold increase in the ability of c-Myb to transactivate a Myb-responsive promoter/reporter gene construct. These findings suggest that phosphorylation serves to regulate c-Myb activity and that loss of this phosphorylation site from the v-Myb proteins may contribute to their transforming potential.


Subject(s)
Oncogenes , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Avian Myeloblastosis Virus/genetics , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glutathione Transferase/biosynthesis , Glutathione Transferase/isolation & purification , Kidney , Leukemia Virus, Murine/genetics , Leukemia, Erythroblastic, Acute , Mice , Molecular Sequence Data , Peptide Mapping , Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/isolation & purification , Proto-Oncogene Proteins c-myb , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Regulatory Sequences, Nucleic Acid , Serine , Transcriptional Activation , Transfection , Tumor Cells, Cultured
10.
J Biol Chem ; 270(13): 7661-71, 1995 Mar 31.
Article in English | MEDLINE | ID: mdl-7706314

ABSTRACT

Expression of the c-myb proto-oncogene is primarily detected in normal tissue and tumor cell lines of immature hematopoietic origin, and the down-regulation of c-myb expression is associated with hematopoietic maturation. Cell lines that represent mature, differentiated hematopoietic cell types contain 10-100-fold less c-myb mRNA than immature hematopoietic cell types. Differences in steady-state c-myb mRNA levels appear to be primarily maintained by a conditional block to transcription elongation that occurs in the first intron of the gene. The block to transcription elongation has been mapped, using nuclear run-on analysis, to a region of DNA sequence that is highly conserved between mouse and man. Two sets of DNA-protein interactions, flanking the site of the block to transcription elongation, were detected that exhibited DNA-binding activities that strongly correlated with low steady-state c-myb mRNA levels. Several criteria demonstrated that members of the nuclear factor kappa B (NF-kappa B) family of transcription factors were involved in the DNA-protein interactions identified in these two sets. Surprisingly, cotransfection experiments demonstrated that coexpression of members of the NF-kappa B family, specifically p50 with p65 and p65 with c-Rel, transactivated a c-myb/chloramphenicol acetyltransferase reporter construct that contained 5'-flanking sequences, exon I, intron I, and exon II of the c-myb gene. Transactivation by these heterodimer combinations was dependent on regions of the c-myb first intron containing the NF-kappa B-binding sites. These findings suggest that NF-kappa B family members may be involved in either modifying the efficiency of transcription attenuation or acting as an enhancer-like activity to increase transcription initiation. Thus, the regulation of c-myb transcription may be quite complex, and members of the NF-kappa B family likely play an important role in this regulation.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Oncogenes , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogenes , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cell Line , Cell Nucleus/metabolism , Conserved Sequence , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Exons , Introns , Lymphoma, B-Cell , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myb , Restriction Mapping , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
11.
Oncogene ; 9(7): 1989-97, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7516062

ABSTRACT

Src family protein tyrosine kinases (PTKs) actively participate in signal transduction during lymphocyte activation. However, little is known about the roles of PTKs and their substrates in lymphocyte differentiation. To identify PTK substrates that may be differentially expressed during B lymphopoiesis, we screened a panel of murine B lymphoid tumor cell lines representing various developmental stages using monoclonal antibodies (MAbs) specific for pp60src substrates. A MAb specific for cortactin, a filamentous-actin binding pp60src substrate, immunoprecipitated proteins from murine plasma-cytoma cell lines but not from pre-B cell lymphoma or B cell lymphoma cell lines. We have cloned a murine cortactin cDNA which encodes a member of a family of proteins distinguished by amino-terminal repeat domains and carboxy-terminal Src Homology 3 domains. Two members of this family (cortactin and HS1) were differentially expressed in murine B lymphoid tumor cell lines; both were detected in plasmacytoma cell lines, however HS1 was additionally detected in pre-B lymphoma and B lymphoma cell lines. Cortactin RNA was detected in most murine tissues, but was not detected in B lymphocytes or plasma cells. We hypothesize that cortactin expression is associated with transformed plasma cells and not with the terminal differentiation of normal B lymphocytes to plasma cells.


Subject(s)
Lymphoma, B-Cell/metabolism , Microfilament Proteins/biosynthesis , 3T3 Cells , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , B-Lymphocytes/metabolism , Base Sequence , Blood Proteins/genetics , Blood Proteins/metabolism , Cloning, Molecular , Cortactin , DNA, Complementary , Mice , Mice, Inbred BALB C , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Molecular Sequence Data , Protein-Tyrosine Kinases/metabolism , RNA/genetics , RNA/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Tumor Cells, Cultured
12.
Oncogene ; 8(8): 2259-65, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8336948

ABSTRACT

The product of the c-myb proto-oncogene is a highly conserved transcription factor that has been shown to function as both a transactivator and repressor. The v-myb oncogenes of E26 leukemia virus and avian myeloblastosis virus (AMV) encode proteins truncated at both the amino and carboxy termini, deleting portions of the DNA-binding and negative regulatory domains present in c-Myb. Similar truncations of c-Myb alter its function, suggesting that the viral proteins lack important regulatory sequences. Interestingly, eight potential sites of phosphorylation by proline-directed protein kinases conserved between the avian, murine and human Myb proteins are clustered in or near the negative regulatory domain of c-Myb. The majority of these sites are deleted in both the E26 and AMV viral proteins. In this paper we show that one proline-directed protein kinase, p42mapk, phosphorylates bacterially synthesized avian and murine c-Myb but not AMV v-Myb in vitro. We find that p42mapk phosphorylates c-Myb on serine and threonine, but not on tyrosine. Furthermore, deletion analysis indicates that the sites of phosphorylation map to the C-terminal negative regulatory domain. We speculate that the inability of v-Myb to be phosphorylated by p42mapk may contribute to its oncogenic properties.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Animals , Mice , Mitogen-Activated Protein Kinase 1 , Mutation , Oncogene Proteins v-myb , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myb
13.
Mol Cell Biol ; 13(6): 3505-13, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8497265

ABSTRACT

The c-Myb protein is a transcription factor with an apparent but poorly defined role in hematopoietic cell growth and differentiation. The DNA binding and several transcriptional regulatory domains of the c-Myb protein have been defined by transient transfections into nonhematopoietic cell lines. Although the relationship between these domains and transformation has been studied, little is known about the function of these domains during hematopoietic maturation. Up-regulation of stably transfected c-myb in murine erythroleukemia (MEL) cells blocks terminal differentiation when MEL cells are induced to differentiate with N,N'-hexamethylene bisacetamide. To determine which functional domains of c-Myb are necessary and sufficient to block differentiation, mutated c-myb constructs under the control of a murine metallothionein promoter were transfected into C19 MEL cells, and stable clonal cell lines were established. The ability of Myb mutants to block differentiation paralleled their ability to transactivate transcription of a reporter gene containing Myb-responsive elements, by transient transfection into a lymphoid cell line. The smallest c-Myb mutant able to block differentiation consisted of the DNA binding domain juxtaposed to the transactivation domain. Therefore, the DNA binding domain and the transactivation domain are necessary and sufficient for c-Myb to block differentiation in MEL cells.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Oncogenes , Proto-Oncogene Proteins/metabolism , Acetamides/pharmacology , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Nucleus/metabolism , Friend murine leukemia virus/genetics , Gene Deletion , Leukemia, Erythroblastic, Acute , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides , Oligonucleotides, Antisense , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/isolation & purification , Proto-Oncogene Proteins c-myb , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Restriction Mapping , Transcriptional Activation , Transfection , Tumor Cells, Cultured
14.
J Immunol ; 148(3): 934-42, 1992 Feb 01.
Article in English | MEDLINE | ID: mdl-1730880

ABSTRACT

The expression of c-myb mRNA is differentially regulated in murine B lymphoid tumors such that B cell lymphomas and plasmacytomas contain significantly less c-myb mRNA than pre-B cell lymphomas. To examine the low level of c-myb mRNA expression in the murine B cell lymphoma cell line BCL1, nonessential amino acid starvation was used to block these cells in a G1 state. When BCL1 cells were released from this block, a 7- to 10-fold increase in c-myb mRNA was detected in late G1 and S phase cells relative to that detected in exponentially growing BCL1 cells. This increase was not inhibited by aphidicolin. To determine whether cell cycle regulation of c-myb mRNA expression occurred during exponential growth in both murine pre-B cell lymphoma and B cell lymphoma cell lines, elutriation was used to separate exponentially growing cell populations. An increase in c-myb mRNA expression was seen in late G1 and S phase fractions from B cell lymphoma cell lines. In contrast, c-myb mRNA levels remained constant in elutriation fractions isolated from pre-B cell lymphoma cell lines. Expression of c-myb mRNA was not detected in exponentially growing or in Go serum-stimulated murine fibroblasts. These results indicate that constitutive vs cell cycle regulation of c-myb mRNA expression is related to the state of differentiation in murine B lymphoid tumors and suggest that a switch in regulation may occur during normal B cell development.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, B-Cell/genetics , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins/genetics , Animals , B-Lymphocytes/physiology , Cell Cycle , Cell Separation , Genes , Genes, myc , Histones/genetics , Mice , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-myb , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Transcription, Genetic , Tumor Cells, Cultured
15.
J Immunol Methods ; 145(1-2): 137-42, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1765644

ABSTRACT

Certain sub-lines of the murine B cell lymphoma BCL1 can be maintained in vitro and respond to cytokines including IL-2 and IL-5. BCL1 cells, as well as other B lymphomas, are difficult to synchronize using conventional techniques such as thymidine block or DNA synthesis inhibition. We have found that BCL1 cells maintained in Dulbecco's minimum essential medium (DMEM) with non-essential amino acids (NEAA) can be readily synchronized by culture in DMEM lacking NEAA. Within 10-18 h of medium replacement, 98% of BCL1 cells are 2 N in DNA content, suggesting that these cells are arrested in G0/G1. This population of BCL1 cells is viable and can be stimulated to enter S phase by culture in media containing NEAA; however, arrested cells did not appear to return synchronously into the cell cycle on addition of NEAA. A transient increase in levels of c-fos and c-myc mRNA was not detected after arrested BCL1 cells were stimulated to enter S phase, suggesting that arrested cells are in the G1 phase of the cell cycle, rather than G0. This technique for obtaining G1 arrested B lymphoma cells may prove useful in the analysis of molecular events that occur in B cells as a function of cell cycle position.


Subject(s)
Lymphoma, B-Cell/pathology , Amino Acids/metabolism , Cell Cycle , Gene Expression , Genes, fos , Genes, myc , Humans , In Vitro Techniques , Isoleucine/deficiency , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Cells, Cultured
16.
Mol Cell Biol ; 10(2): 705-10, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2405253

ABSTRACT

During chemically induced differentiation of Friend virus-infected mouse erythroleukemia (MEL) cell lines, there is a biphasic down-regulation of the c-myb proto-oncogene. A plasmid containing a murine c-myb cDNA controlled by a mouse metallothionein I promoter was transfected into the C19 MEL cell line. For six transfected clones, it was found that expression of the exogenous c-myb mRNA could be up-regulated by the addition of 120 microM ZnCl2 and that the N,N'-hexamethylenebisacetamide-induced differentiation of these transfectants was inhibited in proportion to the level of exogenous c-myb mRNA expression. By adding or removing ZnCl2 at different times during the induction process, it was possible to show that up-regulation of exogenous c-myb limited to the first 2 days of induction had little or no effect on differentiation. In contrast, continuous expression of exogenous c-myb beginning at any time during the period of induction blocked further differentiation. These results suggest that during HMBA induction of MEL cells, the early down-regulation of c-myb mRNA is not necessary for terminal differentiation, whereas the down-regulation of c-myb at a later time is necessary.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Neoplastic , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Transfection , Zinc Compounds , Animals , Cell Differentiation/drug effects , Cell Line , Chlorides/pharmacology , Kinetics , Leukemia, Erythroblastic, Acute , Metallothionein/genetics , Mice , Plasmids , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myb , RNA, Neoplasm/genetics , Restriction Mapping , Zinc/pharmacology
20.
J Immunol ; 139(11): 3822-7, 1987 Dec 01.
Article in English | MEDLINE | ID: mdl-3316389

ABSTRACT

A series of murine B lymphoid tumor cell lines which are representative of the pre-B cell, immature and mature B cell, and plasma cell stages of B cell development have been examined for expression of c-myb proto-oncogene mRNA. The pre-B cell lymphoma cell lines express equivalent high steady state levels of c-myb mRNA. In contrast, the B cell lymphoma and plasmacytoma cell lines express steady state c-myb mRNA at levels which are 0.005 to 0.1 times that of the pre-B cell lymphoma lines. These results correlate high levels of c-myb mRNA expression with the pre-B cell stage of development. Subclones of the 1881 pre-B cell lymphoma which express K light chain and are surface IgM-positive as well as two types of hybrid B lymphoid cell lines have been used to demonstrate that surface immunoglobulin expression is not sufficient to result in the down-regulation of c-myb mRNA levels or changes in the expression N-myc mRNA, lambda 5 mRNA, or the BP-1 surface antigen which are markers of the pre-B cell stage of development. Thus, changes in the expression of genes which are independent of immunoglobulin expression are associated with transition from the pre-B cell to the immature B cell stage of development.


Subject(s)
B-Lymphocytes/pathology , Gene Expression Regulation , Lymphoma/pathology , Proto-Oncogene Proteins/biosynthesis , Tumor Cells, Cultured/pathology , Animals , Antibodies, Neoplasm/analysis , Cell Differentiation , Immunoglobulin M/analysis , Mice , Proto-Oncogene Proteins c-myb , RNA, Messenger/analysis , Receptors, Antigen, B-Cell/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...