Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Genet ; 19(2): 199-202, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9620782

ABSTRACT

Telomeric DNA of mammalian chromosomes consists of several kilobase-pairs of tandemly repeated sequences with a terminal 3' overhang in single-stranded form. Maintaining the integrity of these repeats is essential for cell survival; telomere attrition is associated with chromosome instability and cell senescence, whereas stabilization of telomere length correlates with the immortalization of somatic cells. Telomere elongation is carried out by telomerase, an RNA-dependent DNA polymerase which adds single-stranded TAGGGT repeats to the 3' ends of chromosomes. While proteins that associate with single-stranded telomeric repeats can influence tract lengths in yeast, equivalent factors have not yet been identified in vertebrates. Here, it is shown that the heterogeneous nuclear ribonucleoprotein A1 participates in telomere biogenesis. A mouse cell line deficient in A1 expression harbours telomeres that are shorter than those of a related cell line expressing normal levels of A1. Restoring A1 expression in A1-deficient cells increases telomere length. Telomere elongation is also observed upon introduction of exogenous UP1, the amino-terminal fragment of A1. While both A1 and UP1 bind to vertebrate single-stranded telomeric repeats directly and with specificity in vitro, only UP1 can recover telomerase activity from a cell lysate. These findings establish A1/UP1 as the first single-stranded DNA binding protein involved in mammalian telomere biogenesis and suggest possible mechanisms by which UP1 may modulate telomere length.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Repetitive Sequences, Nucleic Acid , Ribonucleoproteins/metabolism , Telomerase/metabolism , Telomere/metabolism , Animals , Cells, Cultured , DNA Helicases/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoproteins , Mice , Thymus Hormones/metabolism
2.
Oncogene ; 37(3): 323-331, 2018 01 18.
Article in English | MEDLINE | ID: mdl-28925397

ABSTRACT

Coactosin-like protein (CLP, or Cotl1), is an F-actin-binding protein, whose role in cancer is largely unknown. Here we show that CLP/Cotl1 is highly expressed in a rat epithelial breast cancer cell line (FE1.3) compared with its mesenchymal counterpart (FE1.2). Knockdown of CLP/Cotl1 in FE1.3 cells increased cell proliferation, whereas its overexpression in FE1.2 cells inhibited proliferation in culture and reduced tumor growth in xenograft assays in mice. Mechanistically, we identified two major pathways through which CLP/Cotl1 exerts its suppressive effects. First, CLP/Cotl1 re-expression in FE1.2 and in human MCF7 breast cancer cells induced expression of the growth-suppressor gene interleukin-24 (IL-24), which independently of p53 upregulates the tumor-suppressor genes p53 apoptosis effector related to PMP-22 (PERP) and p21cip1. Second, overexpression of CLP/Cotl1 potentiated the growth-suppressive effect of transforming growth factor-ß1 (TGFß1), leading to downregulation of TGFß-responsive genes vascular growth factor A/B (VEGFA/VEGFB), hypoxia inducing factor 1α (HIF-1α) and trombospondin 1 (TSP1), which mediate various hallmarks of cancer progression including angiogenesis, invasion and metastasis. CLP/Cotl1 inhibited TGFß signaling via a non-canonical signaling involving IL-24-instigated inhibition of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) phosphorylation and subsequent post-transcriptional downregulation of SMAD2 and SMAD4. We also showed that CLP/COTL1 expression sensitizes breast cancer cells to chemotherapeutic drugs, and this was further enhanced by addition of exogenous TGFß1. CLP/Cotl1 expression is lost in many human malignancies including prostate, uterine and breast cancers. Thus, our results uncover a novel tumor-suppressor role for CLP/Cotl1 and identify the downstream effectors interleukin 24 (IL-24)/PERP and IL-24/MAPK/ERK/TGFß as potential targets for precision therapy.


Subject(s)
Breast Neoplasms/pathology , Carrier Proteins/metabolism , Microfilament Proteins/metabolism , Animals , Carrier Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Knockdown Techniques , Genes, Tumor Suppressor , HEK293 Cells , Humans , Interleukins/metabolism , MAP Kinase Signaling System , MCF-7 Cells , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Microfilament Proteins/genetics , RNA, Small Interfering/metabolism , Rats , Transforming Growth Factor beta1/metabolism , Xenograft Model Antitumor Assays
3.
Mol Cell Biol ; 12(10): 4449-55, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1406633

ABSTRACT

A large number of novel cellular proto-oncogenes have been identified and cloned by analysis of common integration sites in retrovirally induced malignancies. In the multistage erythroleukemias induced by the various strains of Friend leukemia virus, the analysis of proviral-integration events has led to the identification of two genes, Fli-1 and Spi-1, both novel members of the ets oncogene family of transcription factors. In this report, we describe the identification of another integration site, designated Fli-2 (Friend leukemia virus integration-2), in an erythroleukemia cell line induced by Friend murine leukemia virus (F-MuLV). Rearrangements at the Fli-2 locus were found in two erythroleukemia cell lines independently induced by F-MuLV and one leukemic cell line derived from the spleen of a mouse infected with the polycythemia strain of Friend leukemia virus. The deduced amino acid sequence of a cDNA corresponding to a transcript originating from genomic DNA adjacent to Fli-2 is identical to that of the human heterogeneous nuclear ribonucleoprotein A1 gene, a member of the gene family of RNA-binding proteins involved in RNA splicing. In one erythroleukemia cell line, A1 expression was undetectable as a result of F-MuLV integration in one allele and loss of the other allele. These results suggest that perturbations in RNA splicing mechanisms may contribute to malignant transformation and provide direct evidence that the A1 protein is not required for cell growth.


Subject(s)
Cell Division/genetics , Friend murine leukemia virus/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Ribonucleoproteins/genetics , Virus Integration , Amino Acid Sequence , Animals , Base Sequence , Cell Division/physiology , DNA , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Leukemia, Erythroblastic, Acute , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Molecular Sequence Data , RNA Splicing , Restriction Mapping , Ribonucleoproteins/physiology , Transcription, Genetic , Tumor Cells, Cultured
4.
Mol Cell Biol ; 21(1): 73-80, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11113182

ABSTRACT

In previous studies, we identified a common site of retroviral integration designated Fli-2 in Friend murine leukemia virus (F-MuLV)-induced erythroleukemia cell lines. Insertion of F-MuLV at the Fli-2 locus, which was associated with the loss of the second allele, resulted in the inactivation of the erythroid cell- and megakaryocyte-specific gene p45(NFE2). Frequent disruption of p45(NFE2) due to proviral insertion suggests a role for this transcription factor in the progression of Friend virus-induced erythroleukemias. To assess this possibility, erythroleukemia was induced by F-MuLV in p45(NFE2) mutant mice. Since p45(NFE2) homozygous mice mostly die at birth, erythroleukemia was induced in +/- and +/+ mice. We demonstrate that +/- mice succumb to the disease moderately but significantly faster than +/+ mice. In addition, the spleens of +/- mice were significantly larger than those of +/+ mice. Of the 37 tumors generated from the +/- and +/+ mice, 10 gave rise to cell lines, all of which were derived from +/- mice. Establishment in culture was associated with the loss of the remaining wild-type p45(NFE2) allele in 9 of 10 of these cell lines. The loss of a functional p45(NFE2) in these cell lines was associated with a marked reduction in globin gene expression. Expression of wild-type p45(NFE2) in the nonproducer erythroleukemic cells resulted in reduced cell growth and restored the expression of globin genes. Similarly, the expression of p45(NFE2) in these cells also slows tumor growth in vivo. These results indicate that p45(NFE2) functions as an inhibitor of erythroid cell growth and that perturbation of its expression contributes to the progression of Friend erythroleukemia.


Subject(s)
DNA-Binding Proteins/metabolism , Friend murine leukemia virus/physiology , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Erythroblastic, Acute/virology , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Division , Clone Cells/metabolism , Clone Cells/pathology , Clone Cells/virology , DNA-Binding Proteins/genetics , Disease Progression , Erythroid-Specific DNA-Binding Factors , Gene Deletion , Gene Expression Regulation, Neoplastic , Genotype , Globins/genetics , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , NF-E2 Transcription Factor, p45 Subunit , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured
5.
Mol Cell Biol ; 12(6): 2681-9, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1375325

ABSTRACT

A novel protein kinase, the Esk kinase, has been isolated from an embryonal carcinoma (EC) cell line by using an expression cloning strategy. Sequence analysis of two independent cDNA clones (2.97 and 2.85 kb) suggested the presence of two Esk isoforms in EC cells. The esk-1 cDNA sequence predicted an 857-amino-acid protein kinase with a putative membrane-spanning domain, while the esk-2 cDNA predicted an 831-amino-acid kinase which lacked this domain. In adult mouse cells, esk mRNA levels were highest in tissues possessing a high proliferation rate or a sizeable stem cell compartment, suggesting that the Esk kinase may play some role in the control of cell proliferation or differentiation. As anticipated from the screening procedure, bacterial expression of the Esk kinase reacted with antiphosphotyrosine antibodies on immunoblots. Furthermore, in in vitro kinase assays, the Esk kinase was shown to phosphorylate both itself and the exogenous substrate myelin basic protein on serine, threonine, and tyrosine residues, confirming that the Esk kinase is a novel member of the serine/threonine/tyrosine family of protein kinases.


Subject(s)
Protein Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Compartmentation , Cell Membrane/enzymology , Cloning, Molecular , Cytoplasm/enzymology , DNA/genetics , Gene Expression , In Vitro Techniques , Mice , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoserine/metabolism , Phosphothreonine/metabolism , Phosphotyrosine , Protein Kinases/metabolism , RNA, Messenger/genetics , Sequence Alignment , Substrate Specificity , Tumor Cells, Cultured , Tyrosine/analogs & derivatives , Tyrosine/metabolism
6.
Mol Cell Biol ; 19(6): 4452-64, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330185

ABSTRACT

Erythropoietin (Epo) is a major regulator of erythropoiesis that alters the survival, proliferation, and differentiation of erythroid progenitor cells. The mechanism by which these events are regulated has not yet been determined. Using HB60, a newly established erythroblastic cell line, we show here that Epo-induced terminal erythroid differentiation is associated with a transient downregulation in the expression of the Ets-related transcription factor Fli-1. Constitutive expression of Fli-1 in HB60 cells, similar to retroviral insertional activation of Fli-1 observed in Friend murine leukemia virus (F-MuLV)-induced erythroleukemia, blocks Epo-induced differentiation while promoting Epo-induced proliferation. These results suggest that Fli-1 modulates the response of erythroid cells to Epo. To understand the mechanism by which Fli-1 regulates erythropoiesis, we searched for downstream target genes whose expression is regulated by this transcription factor. Here we show that the retinoblastoma (Rb) gene, which was previously shown to be involved in the development of mature erythrocytes, contains a Fli-1 consensus binding site within its promoter. Fli-1 binds to this cryptic Ets consensus site within the Rb promoter and transcriptionally represses Rb expression. Both the expression level and the phosphorylation status of Rb are consistent with the response of HB60 cells to Epo-induced terminal differentiation. We suggest that the negative regulation of Rb by Fli-1 could be one of the critical determinants in erythroid progenitor cell differentiation that is specifically deregulated during F-MuLV-induced erythroleukemia.


Subject(s)
DNA-Binding Proteins/physiology , Erythroid Precursor Cells/physiology , Erythropoietin/physiology , Genes, Retinoblastoma/genetics , Proto-Oncogene Proteins , Trans-Activators/physiology , Transcription, Genetic , Animals , Blotting, Northern , Cell Cycle , Cell Differentiation , Cell Division , Chromatin/metabolism , Electrophoresis, Polyacrylamide Gel , Immunoblotting , Mice , Mice, Inbred BALB C , Models, Genetic , Neoplasms, Experimental , Oligonucleotides, Antisense , Precipitin Tests , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1 , Proto-Oncogene Proteins c-kit/physiology , Recombinant Fusion Proteins , Stem Cell Factor/physiology , Time Factors , Transfection , Tumor Cells, Cultured
7.
Adv Protein Chem Struct Biol ; 107: 223-273, 2017.
Article in English | MEDLINE | ID: mdl-28215225

ABSTRACT

Multiplatform genomic analyses have identified 93 frequently altered genes in breast cancer. Of these, as many as 49 genes are directly or indirectly involved in transcription. These include constitutive and inducible DNA-binding transcription factors (DB-TFs, 13 genes), corepressors/coactivators (14 genes), epigenetic (10), and mediator/splicing/rRNA (3) factors. At least nine additional genes are immediate upstream regulators of transcriptional cofactors. G:profiler analysis reveals that these alterations affect cell cycle, development/differentiation, steroid hormone, and chromatin modification pathways. A notable observation is that DB-TFs that mediate major oncogenic signaling (e.g., WNT, receptor tyrosine kinase (RTK), NOTCH, and HIPPO), which switch from default repression (signal OFF) to transcriptional activation (signal ON), are not altered in breast cancer. Instead, corepressors (e.g., pRb for E2F1 downstream of various proliferation signals) or upstream factors (e.g., APC and AXIN for TCF, downstream of canonical WNT signaling) are lost, or coactivators (e.g., NOTCH1/2 for CSL/RBPJk) are induced. In contrast, constitutive (MYC, TBX3) and signal-induced (TP53, FOXA1) DB-TFs that do not mediate default repression are directly altered in breast cancer. Some of these TFs have been implicated in the establishment of super-enhancers and positive transcriptional elongation. In addition, oncogenic transcription is induced by mutations affecting regulatory elements or chromatin conformation that create new TF-binding sites in promoters and enhancers of oncogenic genes to promote tumorigenesis. Here we review these diverse oncogenic alterations in TFs in BC and discuss implications for therapy.


Subject(s)
Breast Neoplasms/metabolism , Genome , Transcription Factors/metabolism , Breast Neoplasms/genetics , Female , Humans , Oncogenes , Transcription Factors/genetics , Transcription, Genetic
8.
Cancer Res ; 47(24 Pt 1): 6590-4, 1987 Dec 15.
Article in English | MEDLINE | ID: mdl-2824038

ABSTRACT

Clonality of radiation leukemia virus (RadLV)-induced thymic lymphomas was determined by detection of rearrangements in the genetic locus coding for the beta chain of the T-cell receptor (T beta). Unique T beta rearrangements were detected in four of six lymphomas. Two of the T beta-rearranged thymic lymphomas and two in which such a rearrangement was not detected had a biallelic deletion of C beta 1. With an anti-RadLV monoclonal antibody it was found that 1-2 days after virus inoculation more than one-third of the cells in the thymus were infected by the virus. The frequency of virus-positive cells gradually declined and persisted at 1-2% until the appearance of a clonal lymphoma at which time virtually all the cells in the thymus were virus positive. Transfer of thymocytes from a single, preleukemic mouse 21 days post-virus inoculation into several adoptive recipients resulted in donor-type thymic lymphomas in the majority of the mice. T beta rearrangement analysis revealed that these lymphomas were clonal and derived from different potentially leukemic (preleukemic) cells in the thymus of the donor mouse. Eleven of 15 lymphomas had a biallelic deletion of C beta 1. These results suggest that clonal, RadLV-induced thymomas are selected from an oligoclonal, RadLV-infected preleukemic T-cell population.


Subject(s)
Deoxyribonucleases, Type II Site-Specific , Leukemia, Experimental/pathology , Preleukemia/pathology , Tumor Virus Infections/pathology , Alleles , Animals , Antibodies, Monoclonal , Antibodies, Viral/analysis , Chromosome Deletion , Clone Cells/analysis , DNA Restriction Enzymes/metabolism , Deoxyribonuclease EcoRI , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Retroviridae/immunology , T-Lymphocytes/immunology , T-Lymphocytes/ultrastructure
9.
Cancer Res ; 55(5): 1139-45, 1995 Mar 01.
Article in English | MEDLINE | ID: mdl-7867000

ABSTRACT

Expression of resistance to cis-diamminedichloroplatinum(II) (CDDP), one of the most effective chemotherapeutic drugs used to treat a variety of malignancies, remains a serious obstacle for improving cancer treatment. To study possible genetic mechanisms underlying the development of CDDP resistance, we have adopted the approach of retroviral insertional mutagenesis. An early-stage CDDP-sensitive human melanoma cell line, WM35, was infected with a defective amphotropic murine retrovirus (murine stem cell virus), and the pooled cells were subsequently selected for CDDP-resistant variants. Nine CDDP-resistant clones independently derived from murine stem cell virus-infected WM35 cells were analyzed and it was found that five of these clones acquired an identical retroviral integration site, designated as CDDP resistance locus 1 (CRL-1), as revealed by isolation of retroviral flanking sequences. Furthermore, using the flanking sequence as probe, we have detected a 3.5-4.0-kilobase message, the expression of which is strongly increased in clones carrying a rearranged CRL-1 locus. These results strongly suggest that overexpression of CRL-1 confers resistance to CDDP in these clones. In addition, the present study indicates that retroviral insertional mutagenesis represents a potential strategy to identify genes responsible for CDDP resistance and possibly other chemotherapeutic drugs as well.


Subject(s)
Cisplatin/pharmacology , Melanoma/drug therapy , Melanoma/genetics , Mutagenesis, Insertional , Proviruses/genetics , Retroviridae/genetics , Cloning, Molecular , Drug Resistance , Humans , Melanoma/virology , Nucleic Acid Hybridization , Retroviridae Infections/genetics , Transcription, Genetic , Tumor Cells, Cultured , Virus Integration
10.
Cancer Res ; 56(13): 3075-86, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8674065

ABSTRACT

The vast majority of primary human cutaneous melanomas undergo a slow and gradual progression from a clinically indolent, curable radial growth phase (RGP) to a malignant vertical growth phase. We sought to develop a way of isolating genetically related malignant variants from a benign RGP human melanoma, called WM35. The parent and variants were then used as a model system to examine to what extent the expression of clinically and biologically relevant phenotypic features characteristic of advanced melanomas are associated with (and thus perhaps causative of) such a malignant conversion. Such a model system could also be used as a means of eventually identifying genetic alterations and cellular changes involved in the malignant switch in melanoma progression. To develop such a model, we subjected WM35 cells to retroviral insertional mutagenesis, which was followed by selection for progressive growth of solid tumors in nude mice. Highly aggressive and phenotypically stable tumorigenic variants were derived which contained at least four integrated proviruses. In contrast to the parental WM35 cells, these cell lines expressed several phenotypic features characteristic of naturally derived, advanced-stage malignant melanoma cells. Thus, in addition to tumor-forming ability in nude mice, the variants were growth factor and anchorage independent, overexpressed the MUC18 adhesion molecule, and lost responsiveness to the growth-inhibitory effect of several cytokines, including interleukin 6, transforming growth factor beta, interleukin 1beta, and tumor necrosis factor-alpha. Tumorigenicity and "multicytokine resistance" were dominant traits since in somatic cell hybrids between the parental cells and a tumorigenic subline no suppressive effect of the former cell population was observed. These findings suggest that one or more dominantly acting genetic alterations might be involved in this progression of RGP melanoma cells. The identity of such alterations remains to be determined.


Subject(s)
Antigens, CD , Melanoma/genetics , Melanoma/pathology , Neural Cell Adhesion Molecules , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , CD146 Antigen , Cell Division/physiology , Cell Transformation, Neoplastic , Chromosome Aberrations , Cytokines/pharmacology , Drug Resistance, Multiple , Humans , Hybrid Cells , Karyotyping , Melanoma/virology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Staging , Oncogenes , Phenotype , Retroviridae Infections/genetics , Skin Neoplasms/virology , Tumor Cells, Cultured
11.
Cancer Res ; 58(14): 2919-22, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9679945

ABSTRACT

Familial adenomatous polyposis is a dominantly inherited colon cancer syndrome associated with germ-line mutations in the APC tumor suppressor gene. An APC gene sequence alteration, the I1307K allele, occurs in 6% of the Ashkenazi Jewish population and is reported to double the risk for colorectal cancer. We screened a population of 190 Ashkenazi women who were diagnosed with epithelial ovarian carcinoma for the I1307K variant and measured the effect of this allele on the risk for cancer development in their first-degree relatives. We identified the I1307K allele in 7.9% (15 of 190) of our ovarian cancer cases. The average age of ovarian cancer diagnosis in carriers of the I1307K allele (57.5 years) was not statistically different than the age for noncarriers (56.4 years; P = 0.70). Among the 1087 first-degree relatives, there were 23 cases of colorectal cancer; 3 of 100 relatives of probands with the I1307K allele (3.0%) had a history of colorectal cancer versus 20 of 987 relatives of probands without the I1307K allele (2.1%; relative risk, 1.48; 95% confidence interval, 0.45-4.88; P = 0.462). Relatives of the I1307K carriers had a risk of 38.0% for developing any cancer to age 80, similar to the risk for relatives of noncarriers of the I1307K allele (42.1%; P = 0.86). The average age of diagnosis of cancer of any type was not different between relatives of carriers (59.0 years) and noncarriers (60.4 years). In the Ashkenazi Jewish population, the I1307K allele is unlikely to increase the risk of ovarian cancer or of cancer in general.


Subject(s)
Biomarkers, Tumor/genetics , Genes, APC/genetics , Jews , Ovarian Neoplasms/genetics , Colorectal Neoplasms/genetics , Female , Humans , Middle Aged , Ovarian Neoplasms/ethnology , Pedigree , Risk Factors
12.
Oncogene ; 19(55): 6482-9, 2000 Dec 18.
Article in English | MEDLINE | ID: mdl-11175364

ABSTRACT

Aberrant expression of the Fli-1 transcription factor following genetic mutation has been recognized as a seminal event in the initiation of certain types of malignant transformation. Indeed, the etiology of a number of virally induced leukemias, including Friend virus-induced erythroleukemia, has been associated with Fli-1 overexpression. The clinical relevance of Fli-1 becomes apparent in human Ewing's sarcoma in which Fli-1 is the target of a characteristic chromosomal translocation. As such, Fli-1 has generated considerable interest over the past several years for its role in malignant transformation and tumor progression. This review will present a synopsis of the current research on Fli-1 with emphasis on its function in malignant transformation. Moreover, the possible role of Fli-1 in cellular proliferation, differentiation and survival, as well as the recent development of transgenic and knock-out mice to investigate the function of Fli-1 will be discussed. Finally, the significance of identifying target genes that are regulated by Fli-1 and their role in cellular function will be reviewed.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/physiology , Proto-Oncogene Proteins , Trans-Activators/physiology , 3T3 Cells , Adult , Animals , Apoptosis/genetics , Bone Neoplasms/genetics , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , Friend murine leukemia virus/genetics , Gene Duplication , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Helix-Loop-Helix Motifs , Hematopoiesis/genetics , Humans , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/virology , Mice , Mice, Knockout , Multigene Family , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Oncogene Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Organ Specificity , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Sarcoma, Ewing/genetics , Sequence Deletion , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/physiology
13.
Oncogene ; 3(2): 179-85, 1988 Aug.
Article in English | MEDLINE | ID: mdl-2842714

ABSTRACT

The p53 gene is rearranged in a high proportion of erythroleukemic cell lines derived from the spleens of mice infected with Friend leukemia virus. These rearrangements result in either the synthesis of a truncated protein or the inactivation of the p53 gene. Here we have molecularly characterized the rearrangements in two murine erythroleukemic cell lines induced by Friend leukemia virus, DP20-1 and CB3, that contain a rearranged p53 gene and fail to express p53 protein. The rearrangement in the DP20-1 cell line is due to the insertion of Friend spleen focus-forming provirus (SFFV) in the 3' end of the p53 gene in intron sequences between exons 9 and 10. Transfection of molecular clones of this SFFV provirus into NIH3T3 cells results in the generation of infectious virus as determined by its ability, in the presence of helper virus, to induce rapid splenomegaly and polycythemia when injected into adult DBA/2J mice. Insertion of SFFV in DP20-1 cells resulted in the expression of an aberrant 2.9 kb RNA species. Analysis of a molecular clone of the rearranged p53 gene in a second cell line, CB3, revealed that the p53 gene in this clone has sustained a large deletion within the p53 gene resulting in the loss of coding sequences between exons 4 and 8. The 5' end of the deletion originates within exon 4 and extends 3' to within the eighth intron. The significance of these findings with regard to the multi-stage nature of Friend virus induced erythroleukemia is discussed.


Subject(s)
Chromosome Deletion , Leukemia Virus, Murine/genetics , Leukemia, Erythroblastic, Acute/genetics , Neoplasm Proteins/genetics , Oncogenes , Phosphoproteins/genetics , Spleen Focus-Forming Viruses/genetics , Animals , Base Sequence , Friend murine leukemia virus , Leukemia, Erythroblastic, Acute/etiology , Mice , Mice, Inbred DBA , Molecular Sequence Data , Recombination, Genetic , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53
14.
Oncogene ; 8(10): 2721-9, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8378083

ABSTRACT

The erythroleukemias induced by Friend murine leukemia virus (F-MuLV) result from the accumulation of a number of genetic changes, including activation of the Fli-1 proto-oncogene and inactivation of the p53 tumor suppressor gene. We have determined the temporal order of mutation of the genes involved in this multistage malignancy, by serial in vivo transplantation of F-MuLV induced primary erythroleukemias into syngenic Balb/c mice. These primary tumors are capable of growing when transplanted into syngenic mice, but die after several days of in vitro culture. From the transplanted tumors grown in syngenic mice, erythropoietin-dependent cell lines were established in culture that are clonally related to cells in the primary tumors. We show that retroviral insertional activation of the Fli-1 ets family member is the first detectable genetic event in F-MuLV induced primary erythroleukemias. Mutations in the p53 gene were observed in the Epo-dependent cell lines but not in the transplanted erythroleukemias used to establish these cell lines in culture. These data suggest that activation of Fli-1 plays an important role in the early stages of F-MuLV-induced leukemia, perhaps by altering the self-renewal probabilities of erythroid progenitor cells and that p53 mutations immortalize these cells, enabling them to grow in vitro in the presence of Epo.


Subject(s)
DNA, Neoplasm/genetics , Friend murine leukemia virus , Gene Expression Regulation , Gene Rearrangement/genetics , Genes, p53/genetics , Leukemia, Erythroblastic, Acute/genetics , Point Mutation/genetics , Proto-Oncogenes/genetics , Animals , Cell Division , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Tumor Cells, Cultured
15.
Oncogene ; 18(40): 5525-34, 1999 Sep 30.
Article in English | MEDLINE | ID: mdl-10523829

ABSTRACT

Erythroleukemias induced by Friend Murine Leukemia Virus (F-MuLV) involve the insertional activation of the proto-oncogene Fli-1, and the inactivation of the p53 tumor suppressor gene. While the activation of Fli-1 is an early, primary transforming event, p53 mutations are correlated with the immortalization of erythroleukemic cells in culture. In this study we have further analysed the role of p53 loss in F-MuLV induced erythroleukemias by examining the progression of this disease in p53 deficient mice. We found that p53-/- mice succumb to the disease more rapidly than p53+/+ littermates. Additionally, of the 112 tumors generated, 19 gave rise to immortal cell lines, eight of which were derived from p53-/- mice, and ten of which were from p53+/- mice. The ability of these primary tumor cells to grow in culture was associated with the complete loss of wild-type p53 in these cell lines. However, cells from many of the tumors induced in p53-/- hosts did not survive in vitro. These results suggest that the loss of p53 does not directly immortalize tumor cells. Instead, we have evidence to suggest that the loss of p53 promotes the accumulation of mutations that are required for survival in culture and that are capable of accelerating tumor progression in vivo. Indeed, mutations causing expression of the growth factor gene erythropoietin (Epo), were detected in two of seven Epo-independent cell lines from p53 deficient primary erythroleukemias. Moreover, the mechanism of activation of the Epo gene in one of these two Epo-independent cell lines involved genomic rearrangement, that is a hallmark of genetic instability. We propose that, in F-MuLV induced-erythroleukemias, p53 loss may encourage the accumulation of further mutations, subsequently conferring a growth advantage and immortality to the transformed erythroblasts.


Subject(s)
Erythroid Precursor Cells/metabolism , Friend murine leukemia virus/physiology , Gene Deletion , Gene Expression Regulation, Leukemic , Genes, p53 , Leukemia, Erythroblastic, Acute/genetics , Mutagenesis/genetics , Proto-Oncogene Proteins , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , Animals , Animals, Newborn , Blood Proteins/biosynthesis , Blood Proteins/genetics , Cell Division , Cellular Senescence , DNA-Binding Proteins/physiology , Disease Progression , Erythroid Precursor Cells/virology , Erythropoietin/pharmacology , Female , Leukemia, Erythroblastic, Acute/virology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Proto-Oncogene Protein c-fli-1 , Retroviridae Infections/virology , Trans-Activators/physiology , Tumor Cells, Cultured/transplantation , Tumor Suppressor Protein p53/deficiency , Tumor Virus Infections/virology
16.
Oncogene ; 8(6): 1621-30, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8502483

ABSTRACT

The late stages of the erythroleukemias induced by either the replication-defective Friend spleen focus-forming virus (SFFV) or the Friend murine leukemia virus (F-MuLV) are associated with the insertional activation of one of two members (Spi-1 or Fli-1) of the Ets protooncogene family of transcriptional factors. Fli-1 is not rearranged or activated in the erythroleukemias induced by SFFV, and similarly Spi-1 is not rearranged or activated in the leukemic cell clones induced by F-MuLV. This strict specificity of integration sites suggests that Fli-1 and Spi-1 may be functionally distinct and transactivate different downstream genes during the progression of multistage Friend erythroleukemia. In this study, we show that the Fli-1 protein, like other Ets proteins, has DNA-binding activity and can act as a sequence-specific transcriptional activator. We also show that the Fli-1 and Spi-1 proteins are functionally distinct in that they recognize and transactivate through distinct DNA binding sites. Furthermore, we have identified an octanucleotide core sequence that is required in vitro for optimal binding of Fli-1 to the Drosophila E74 target and the promoter sequence of the human GPIIB gene.


Subject(s)
DNA-Binding Proteins/genetics , Friend murine leukemia virus/genetics , Leukemia, Erythroblastic, Acute/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogenes , Sarcoma, Ewing/genetics , Spleen Focus-Forming Viruses/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , DNA-Binding Proteins/metabolism , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/microbiology , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides , Platelet Membrane Glycoproteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Mas , Proto-Oncogene Protein c-fli-1 , Proto-Oncogene Proteins c-ets , Sequence Homology, Nucleic Acid , Trans-Activators/metabolism , Transcription Factors/metabolism , Transfection
17.
Oncogene ; 13(11): 2447-57, 1996 Dec 05.
Article in English | MEDLINE | ID: mdl-8957087

ABSTRACT

We have studied TGF-beta mediated G1 arrest in WM35, an early stage human melanoma cell line. These cells have lost p15INK4B expression through loss of one chromosome 9 and rearrangement of the other. In asynchronously growing WM35, TGF-beta caused reductions in cyclin D1, cyclin A and cdk4 proteins and their associated kinase activities and an increase in both p21Cip1/WAF1 and p27Kip1. These findings were confirmed in cells released from quiescence in the presence of TGF-beta, in which TGF-beta inhibited or delayed the reduction in the cdk inhibitors that normally occurs in late G1. In contrast to observations in other cell types, there was an increased association of both p21Cip1/WAF1 and p27Kip1 with cyclin D1/cdk4 and with cyclin E/cdk2 during TGF-beta mediated arrest of asynchronously growing cells. Upregulation of p21Cip1/WAF1 preceded that of p27Kip1. Furthermore, p21Cip1/WAF1 and p27Kip1 were not present in the same cdk complexes but bound distinct populations of target cdk molecules. Both p21Cip1/WAF1 and p27Kip1 immunoprecipitates from asynchronously growing cells contained active kinase complexes. These KIP-associated kinase activities were reduced in TGF-beta arrested cells. It has been proposed that in TGF-beta arrested epithelial cells, up-regulation of p15INK4B and of p15INK4B binding to cdk4 serves to destabilize the association of p27Kip1 with cyclin D1/cdk4, promoting p27Kip1 binding and inhibition of cyclin E/cdk2. Our findings demonstrate that this is not a universal mechanism of G1 arrest by TGF-beta. In TGF-beta arrested WM35, which lack p15INK4B, the increased p21Cip1/WAF1 may serve a similar function to that of p15INK4B: initiating kinase inhibition and providing an additional mechanism to supplement the effect of p27Kip1 on G1 cyclin/cdks.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Chromosomes, Human, Pair 9/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Cyclins/metabolism , G1 Phase/drug effects , Melanoma/physiopathology , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins , Carrier Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/antagonists & inhibitors , G1 Phase/physiology , Gene Rearrangement , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Phosphorylation , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured , Up-Regulation
18.
Oncogene ; 12(7): 1405-15, 1996 Apr 04.
Article in English | MEDLINE | ID: mdl-8622856

ABSTRACT

Retroviral insertional activation of Fli-1 is the first detectable genetic alteration associated with F-MuLV-induced primary erythroleukemias, while mutations within p53 are only observed in Epo-dependent (ED) cell lines derived from syngeneic mice serially transplanted with F-MuLV-induced primary erythroleukemias. In this study we have determined the mechanism of growth factor independence in several Epo-independent (EI) cell lines established from adult mice previously injected with ED-erythroleukemia cell lines or serially transplanted primary tumor cells. Here we have shown constitutive expression of the Epo gene in 12 of 15 (80%) EI-erythroleukemia cell lines. Among these 12 cell lines, eight were shown to possess clonal rearrangement of the Epo gene which could be detected in the tumors used to establish the majority of these EI-cell lines. Analysis of the pattern of proviral integration revealed that the activation of the Epo gene in these cell lines is independent of retroviral insertional mutagenesis, but apparently the result of genomic rearrangements. Furthermore, the acquisition of growth factor independence by these leukemic cells confers a selective growth advantage in vivo and is associated with enhanced tumorigenicity. Together these observations suggest that the activation of the Epo gene in the large majority of these F-MuLV-induced erythroleukemic cell lines establishes an autocrine loop resulting in the constitutive activation of the Epo receptor signal transduction pathway, thereby conferring a growth and survival advantage in vito and in vitro.


Subject(s)
Erythropoietin/genetics , Friend murine leukemia virus/pathogenicity , Gene Expression , Leukemia, Erythroblastic, Acute/genetics , Animals , Antibodies/immunology , Base Sequence , DNA Primers , Erythropoietin/immunology , Erythropoietin/metabolism , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Erythroblastic, Acute/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Neutralization Tests
19.
Oncogene ; 19(19): 2296-304, 2000 May 04.
Article in English | MEDLINE | ID: mdl-10822380

ABSTRACT

We have recently isolated the erythroleukemic cell line, HB60-5, that proliferates in the presence of erythropoietin (Epo) and stem cell factor (SCF), but undergoes terminal differentiation in the presence of Epo alone. Ectopic expression of the ets related transcription factor Fli-1 in these cells resulted in the establishment of the Epo-dependent cell line HB60-ED that proliferates in the presence of Epo. In this study, we utilized these two cell lines to examine the signal transduction pathways that are activated in response to Epo and SCF stimulation. We demonstrate that Epo, but not SCF, phosphorylates STAT-5 in both HB60-5 and HB60-ED cells. Interestingly, SCF activates the Shc/ras pathway in HB60-5 cells while Epo does not. However, both Epo and SCF are capable of activating the Shc/ras pathway in HB60ED cells. Furthermore, enforced expression of gp55 in HB60-5 cells by means of infection with the Spleen Focus Forming virus-P (SFFV-P), confers Epo independent growth, which is associated with the up-regulation of Fli-1. Activation of the Shc/ras pathway is readily detected in gp55 expressing cells in response to both Epo and SCF, and is associated with a block in STAT-5B tyrosine phosphorylation. These results suggest that STAT-5 activation, in the absence of Shc/ras activation, plays a role in erythroid differentiation. Moreover, Fli-1 is capable of switching Epo-induced differentiation to Epo-induced proliferation, suggesting that this ets factor regulated genes whose products modulate the Epo-Epo-R signal transduction pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Erythropoietin/metabolism , Friend murine leukemia virus , Leukemia, Erythroblastic, Acute/metabolism , Milk Proteins , Proto-Oncogene Proteins , Signal Transduction , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , DNA-Binding Proteins/metabolism , Erythropoiesis/physiology , Erythropoietin/pharmacology , Humans , Leukemia, Erythroblastic, Acute/drug therapy , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteins/metabolism , Proto-Oncogene Protein c-fli-1 , Receptors, Erythropoietin/metabolism , STAT5 Transcription Factor , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology , Trans-Activators/metabolism , Tumor Cells, Cultured , Viral Envelope Proteins/metabolism , ras Proteins/metabolism
20.
Oncogene ; 19(3): 395-402, 2000 Jan 20.
Article in English | MEDLINE | ID: mdl-10656687

ABSTRACT

A major obstacle in the systemic treatment of advanced malignant melanoma is its intrinsic resistance to conventionally used chemotherapeutic agents. In order to investigate the mechanisms of this intrinsic resistance, we have previously utilized retroviral insertional mutagenesis on an early-stage, drug sensitive human melanoma cell line (WM35) to establish mutated cell lines that exhibited increased resistance to cis-diammi-nedichloroplatinum(II) (CDDP). Here, we demonstrate that this increased resistance to CDDP is mediated by the over-expression of tyrosinase-related protein-2 (TYRP2), an enzyme that normally functions in the biosynthesis of the pigment, melanin. Northern and Western blot analyses revealed that the expression of TYRP2 in the virally-derived cell lines as well as in a panel of human melanoma cell lines positively correlated with their levels of resistance to CDDP. Furthermore, enforced expression of TYRP2 in WM35 cells by transfection elevated their resistance to CDDP. The increased CDDP resistance in the virally-derived clones and TYRP2 transfectants was accompanied by a reduction in CDDP-induced apoptosis. Interestingly, the virally-derived CDDP-resistant clones also showed cross resistance to carboplatin and methotrexate, but not taxol, suggesting that TYRP2 over-expression may confer resistance specifically to DNA damaging agents. Overall, these results demonstrate a novel mechanism of drug resistance in human melanoma cells that is mediated by the over-expression of TYRP2. Since TYRP2 is expressed only in cells of melanocytic lineage, this may represent the first report of a lineage-specific mechanism of drug resistance. In summary, these findings suggest a significant role for TYRP2 in the intrinsic drug resistance phenotype of human melanoma cells and may have important implications in the development of chemosensitization strategies for the clinical management of this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Intramolecular Oxidoreductases/physiology , Melanoma/drug therapy , Apoptosis/drug effects , Carboplatin/pharmacology , Cisplatin/therapeutic use , Humans , Methotrexate/pharmacology , Paclitaxel/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL