Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Biosystems ; 231: 104961, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37392989

ABSTRACT

Primary myelofibrosis is an untreatable age-related disorder of haematopoiesis in which a break in the crosstalk between progenitor Haematopoietic Stem Cells (HSCs) and neighbouring mesenchymal stem cells causes HSCs to rapidly proliferate and migrate out of the bone marrow. Around 90% of patients harbour mutations in driver genes that all converge to overactivate haematopoietic JAK-STAT signalling, which is thought to be critical for disease progression, as well as microenvironment modification induced by chronic inflammation. The trigger to the initial event is unknown but dysregulated thrombopoietin (TPO) and Toll-Like Receptor (TLR) signalling are hypothesised to initiate chronic inflammation which then disrupts stem cell crosstalk. Using a systems biology approach, we have constructed an intercellular logical model that captures JAK-STAT signalling and key crosstalk channels between haematopoietic and mesenchymal stem cells. The aim of the model is to decipher how TPO and TLR stimulation can perturb the bone marrow microenvironment and dysregulate stem cell crosstalk. The model predicted conditions in which the disease was averted and established for both wildtype and ectopically JAK mutated simulations. The presence of TPO and TLR are both required to disturb stem cell crosstalk and result in the disease for wildtype. TLR signalling alone was sufficient to perturb the crosstalk and drive disease progression for JAK mutated simulations. Furthermore, the model predicts probabilities of disease onset for wildtype simulations that match clinical data. These predictions might explain why patients who test negative for the JAK mutation can still be diagnosed with PMF, in which continual exposure to TPO and TLR receptor activation may trigger the initial inflammatory event that perturbs the bone marrow microenvironment and induce disease onset.

3.
Leukemia ; 29(10): 1993-2002, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25925206

ABSTRACT

The BMI1 polycomb protein regulates self-renewal, proliferation and survival of cancer-initiating cells essentially through epigenetic repression of the CDKN2A tumor suppressor locus. We demonstrate here for the first time that BMI1 also prevents autophagy in chronic myeloid leukemia (CML) cell lines, to support their proliferation and clonogenic activity. Using chromatin immunoprecipitation, we identified CCNG2/cyclin G2 (CCNG2) as a direct BMI1 target. BMI1 downregulation in CD34+ CML cells by PTC-209 pharmacological treatment or shBMI1 transduction triggered CCNG2 expression and decreased clonogenic activity. Also, ectopic expression of CCNG2 in CD34+ CML cells strongly decreased their clonogenicity. CCNG2 was shown to act by disrupting the phosphatase 2A complex, which activates a PKCζ-AMPK-JNK-ERK pathway that engages autophagy. We observed that BMI1 and CCNG2 levels evolved inversely during the progression of CML towards an acute deadly phase, and therefore hypothesized that BMI1 could support acute transformation of CML through the silencing of a CCNG2-mediated tumor-suppressive autophagy response.


Subject(s)
Autophagy , Cell Proliferation , Cyclin G2/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Polycomb Repressive Complex 1/metabolism , Apoptosis , Blotting, Western , Chromatin Immunoprecipitation , Cyclin G2/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured
5.
J Exp Med ; 193(12): 1361-71, 2001 Jun 18.
Article in English | MEDLINE | ID: mdl-11413191

ABSTRACT

Promyelocytic leukemia (PML) is the organizer of nuclear matrix domains, PML nuclear bodies (NBs), with a proposed role in apoptosis control. In acute promyelocytic leukemia, PML/retinoic acid receptor (RAR) alpha expression disrupts NBs, but therapies such as retinoic acid or arsenic trioxide (As2O3) restore them. PML is conjugated by the ubiquitin-related peptide SUMO-1, a process enhanced by As2O3 and proposed to target PML to the nuclear matrix. We demonstrate that As2O3 triggers the proteasome-dependent degradation of PML and PML/RARalpha and that this process requires a specific sumolation site in PML, K160. PML sumolation is dispensable for its As2O3-induced matrix targeting and formation of primary nuclear aggregates, but is required for the formation of secondary shell-like NBs. Interestingly, only these mature NBs harbor 11S proteasome components, which are further recruited upon As2O3 exposure. Proteasome recruitment by sumolated PML only likely accounts for the failure of PML-K160R to be degraded. Therefore, studying the basis of As2O3-induced PML/RARalpha degradation we show that PML sumolation directly or indirectly promotes its catabolism, suggesting that mature NBs could be sites of intranuclear proteolysis and opening new insights into NB alterations found in viral infections or transformation.


Subject(s)
Adenosine Triphosphatases/metabolism , Arsenicals/pharmacology , Endopeptidases , Neoplasm Proteins/metabolism , Nuclear Matrix/metabolism , Nuclear Proteins , Oxides/pharmacology , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Ubiquitins/metabolism , Amino Acid Motifs , Animals , Arsenic Trioxide , CHO Cells , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Cricetinae , Mice , Models, Biological , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Promyelocytic Leukemia Protein , Proteasome Endopeptidase Complex , Protein Isoforms/chemistry , Protein Transport , Retinoic Acid Receptor alpha , SUMO-1 Protein , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Proteins
6.
J Biol Chem ; 275(37): 28785-92, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10825164

ABSTRACT

Polycomb group (PcG) proteins were first described in Drosophila as factors responsible for maintaining the transcriptionally repressed state of Hox/homeotic genes in a stable and heritable manner throughout development. A growing number of vertebrate genes related to the Drosophila PcG proteins have recently been identified, including two Polycomb orthologues, Pc2 and M33. PcG proteins form multiprotein complexes, termed PcG bodies, that are thought to repress transcription by altering chromatin structure. Here we report the identification and characterization of HPC3 (human Polycomb 3), a novel PcG protein isolated in a yeast two-hybrid screen using human RING1 as bait. HPC3 shows strong sequence similarity to Drosophila Pc and also to vertebrate Pc2 and M33, particularly within the chromodomain and C-box. Previous studies indicate that M33 and human Pc2 (HPC2) can interact with RING1, and we show here that HPC3 also binds to RING1. This interaction is dependent upon the HPC3 C-box but, only partially on the RING finger of RING1. In contrast to HPC2, HPC3 interactions with RING1 are only observed in vivo with covalently modified forms of RING1. HPC3 also colocalizes with other PcG proteins in human PcG bodies. Consistent with its role as a PcG member, HPC3 is able to act as a long range transcriptional silencer when targeted to a reporter gene by a heterologous DNA-binding domain. Taken together, these data suggest that HPC3 is part of a large multiprotein complex that also contains other PcG proteins and is involved in repression of transcriptional activity.


Subject(s)
DNA-Binding Proteins/chemistry , Drosophila Proteins , Nuclear Proteins/chemistry , Proto-Oncogene Proteins/chemistry , Repressor Proteins/chemistry , Amino Acid Sequence , Binding Sites , DNA, Complementary/isolation & purification , Humans , Molecular Sequence Data , Polycomb Repressive Complex 1 , Repressor Proteins/genetics , Repressor Proteins/physiology , Sequence Homology
7.
Leukemia ; 14(2): 255-61, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10673742

ABSTRACT

The fusion protein PML/RARA, associated with acute promyelocytic leukemia behaves as an abnormal retinoic acid (RA) receptor with altered transactivation properties but is still inducible by RA. The chimeric protein is thought to promote leukemogenesis but also paradoxically to mediate the sensitivity to ATRA of APL cells. This has been supported by works reporting that in vitro ATRA resistance is characterized by defects in the RARA/E-domain of PML/RARA. In the present report, we identified a new mutation in the E domain of PML/RARA which is associated with a RA-resistant subline of NB4 cells; NB4-R2. This mutation, identical to the Gln411 mutation found in HL60-R, changes the amino acid Gln903 to an in-phase stop codon, generating a truncated form of PML/RARA which has lost 52 amino acids at its C-terminal end. We have studied the effect of the truncated PML/RARA protein on PML NB formation and RARA and PML/RARA transcriptional activity. We show here that the fusion mutant exerts a dominant negative effect on wild-type PML, PML/RARA and RARA transcription activity. These findings highlight the important role of the RARA E-domain of PML/RARA in mediating RA sensitivity in APL cells.


Subject(s)
Leukemia, Promyelocytic, Acute/genetics , Mutation , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Receptors, Retinoic Acid/genetics , Transcription, Genetic , Tretinoin/metabolism , Codon/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/genetics , HeLa Cells , Humans , Luciferases/analysis , Microscopy, Confocal , Plasmids , Polymerase Chain Reaction , Retinoic Acid Receptor alpha , Sequence Analysis, RNA
8.
Leukemia ; 13(12): 1982-92, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10602419

ABSTRACT

JEM-1 is a novel gene whose mRNA expression in acute promyelocytic leukemia (APL) is induced by retinoid treatments. The gene product, a 45 kDa basic nuclear factor containing a leucine repeat, was transiently expressed in HeLa or COS-7 cells and immunocharacterized within the nuclei in fine punctuated structures which increase in size after cell transfection. Jem-1 was not expressed in the nucleoli. Experimental deletion of peptide domains of Jem-1 (JemDelta331-400 and Jem DeltaL179-206) showed that its C-terminal sequence (Thr331 --> Leu400) is required for nuclear translocation, while the leucine repeat domain (Arg179 --> Glu206) has no influence on subcellular localization. The Jem-1 protein was not detected in the PML-containing nuclear bodies or in speckled structures containing the splicing factor SC-35. In contrast it was localized in the nucleus in structures containing activator protein-1 (AP-1). DNA mobility shift assays showed that the in vitro translated Jem protein interacts neither with the DNA binding site of AP-1, nor directly with in vitro co-translated c-Fos or/and c-Jun proteins bound to this specific sequence. Interestingly, Jem-1-1 increased substantially the transcriptional activity of c-Jun (three-fold) and more strongly that of ectopically co-expressed c-Fos and c-Jun (five- to six-fold), as measured by a CAT reporter gene driven by a heterologous promoter containing the AP-1 binding site of the human collagenase gene. These synergistic effects were strongly Jem-1 dose-dependent. However, Jem-1 alone showed no activity on the collagenase promoter. A deletion of the leucine repeat of Jem-1 (Arg179 --> Glu206) did not diminish the enhancer capacity of Jem-1 on AP-1 activity. In contrast, the enhanced AP-1 activity was abrogated when Jem-1 was deleted of its C-terminus (Thr331 --> Leu400). We conclude that the 45 kDa nuclear product of the JEM-1 gene has features of a novel transcription cofactor, which is enhancing AP-1 activity without directly interacting with c-Jun or c-Fos proteins. Possible implications of these findings for APL cell maturation are discussed.


Subject(s)
Cell Nucleus/chemistry , Nuclear Proteins/analysis , Transcription Factor AP-1/physiology , Transcription Factors/analysis , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , COS Cells , HeLa Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Transcription Factors/genetics , Transcription Factors/physiology
9.
Br J Haematol ; 105(1): 198-207, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10233384

ABSTRACT

Acute promyelocytic leukaemia (APL) is characterized by the t(15;17) leading to the formation of PML-RARalpha and RARalpha-PML fusion genes; this rearrangement has been considered both diagnostic for, and restricted to, this subtype of acute myeloid leukaemia (AML FAB M3). We describe two cases of AML with the t(15;17) associated with a PML/RARalpha rearrangement which lacked typical APL morphology, classified as FAB M1 and M2 respectively. In both cases morphological review revealed small populations of cells which exhibited some features associated with APL. In the case classified as M1, PML immunofluorescence studies revealed the classic microparticulate nuclear staining pattern as observed in typical cases of APL with the t(15;17). Similarly, blasts from this case were found to be sensitive to ATRA in vitro as determined by NBT reduction test and by normalization of the PML nuclear body staining pattern. To determine the frequency of PML/RARalpha rearrangements in FAB subtypes other than M3, 530 patients from the MRC AML trials were screened using nested RT-PCR. Only one individual, initially classified as M5 with a normal karyotype, was found to have a PML/RARalpha rearrangement. The diagnosis was revised to M3 variant on subsequent morphological review. In conclusion, this study demonstrates that, in rare cases, the t(15;17) is not restricted to patients with M3 morphology as defined by current FAB criteria. Therefore, although we consider cytogenetic analysis of newly diagnosed cases of AML to be mandatory, our data suggests that routine molecular screening for PML/RARalpha rearrangements is not justified and should be reserved for those cases displaying features which may be suspicious of APL even if such cells comprise only a minority of the total population.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 17/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Promyelocytic, Acute/diagnosis , Translocation, Genetic/genetics , Adolescent , Adult , Cell Transformation, Neoplastic , Female , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Genetic Testing/methods , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Promyelocytic, Acute/genetics , Male
10.
J Cell Sci ; 112 ( Pt 3): 381-93, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9885291

ABSTRACT

PML is a nuclear phosphoprotein that was first identified as part of a translocated chromosomal fusion product associated with acute promyelocytic leukaemia (APL). PML localises to distinct nuclear multi-protein complexes termed ND10, Kr bodies, PML nuclear bodies and PML oncogenic domains (PODs), which are disrupted in APL and are the targets for immediate early viral proteins, although little is known about their function. In a yeast two-hybrid screen, we first identified a ubiquitin-like protein named PIC1 (now known as SUMO-1), which interacts and co-localises with PML in vivo. More recent studies have now shown that SUMO-1 covalently modifies a number of target proteins including PML, RanGAP1 and IkappaBalpha and is proposed to play a role in either targeting modified proteins and/or inhibiting their degradation. The precise molecular role for the SUMO-1 modification of PML is unclear, and the specific lysine residues within PML that are targeted for modification and the PML sub-domains necessary for mediating the modification in vivo are unknown. Here we show that SUMO-1 covalently modifies PML both in vivo and in vitro and that the modification is mediated either directly or indirectly by the interaction of UBC9 with PML through the RING finger domain. Using site-specific mutagenesis, we have identified the primary PML-SUMO-1 modification site as being part of the nuclear localisation signal (Lys487 or Lys490). However SUMO-1 modification is not essential for PML nuclear localisation as only nuclear PML is modified. The sequence of the modification site fits into a consensus sequence for SUMO-1 modification and we have identified several other nuclear proteins which could also be targets for SUMO-1. We show that SUMO-1 modification appears to be dependant on the correct subcellular compartmentalisation of target proteins. We also find that the APL-associated fusion protein PML-RARA is efficiently modified in vitro, resulting in a specific and SUMO-1-dependent degradation of PML-RARA. Our results provide significant insights into the role of SUMO-1 modification of PML in both normal cells and the APL disease state.


Subject(s)
Ligases/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes , Ubiquitins/pharmacology , Consensus Sequence , Fluorescent Antibody Technique , Humans , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Neoplasm Proteins/analysis , Nuclear Localization Signals , Nuclear Matrix/metabolism , Promyelocytic Leukemia Protein , Recombinant Fusion Proteins , SUMO-1 Protein , Sequence Alignment , Transcription Factors/analysis , Translocation, Genetic , Tumor Cells, Cultured , Tumor Suppressor Proteins
11.
Leukemia ; 12(11): 1733-40, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9823948

ABSTRACT

The JEM-1 gene, recently identified in acute promyelocytic leukemia (APL) cells, codes for a novel nuclear factor (Duprez et al Oncogene 1997; 14: 1563-1570). JEM-1 is kept silent in the APL cell line NB4, but up-regulated (3 kb transcript) during cell maturation. Here, we show that retinoic acid (RA)-induced JEM-1 expression is biphasic (peaks at 6 h and 48 h) and associated with the later stages of maturation. Retinoids, which cooperates with cAMP to induce maturation, also cooperates with cAMP to up-regulate JEM-1, either in maturation-responsive NB4 cells or in NB4-R1 resistant subclones. APL patients showed a low, yet variable, level of JEM-1 mRNA in bone marrow. RA treatment induced an increase in the level of JEM-1 mRNA, as detected by a semi-quantitative PCR. This increase can result from both gene up-regulation or replacement of leukemia cells by differentiated ones. Analysis of JEM-1 expression patterns in normal and tumor cells revealed that JEM-1 expression was ubiquitous. Cell lines derived from monocytic and erythroid leukemias, expressed low and high amounts of JEM-1 mRNA, respectively. Using a JEM cDNA probe, distinct profiles of expression and different transcript sizes (4 kb, 3 kb and 2 kb) were also identified in tumour and normal non-hematopoietic tissues, while interestingly only the 3kb transcript was up-regulated in NB4 cells. This work identifies JEM-1 as a novel ubiquitous gene whose expression is low in APL cells, but can be restored by RA treatment, concomitant with cell maturation.


Subject(s)
Leukemia, Promyelocytic, Acute/genetics , Nuclear Proteins/genetics , RNA, Messenger/genetics , Retinoids/pharmacology , Transcription Factors/genetics , Adult , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cells, Cultured , Chromosomes, Human, Pair 1 , DNA Primers , Fetus , Humans , Leukemia, Promyelocytic, Acute/pathology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Tumor Cells, Cultured
12.
J Cell Sci ; 111 ( Pt 10): 1319-29, 1998 May.
Article in English | MEDLINE | ID: mdl-9570750

ABSTRACT

The ret finger protein (rfp) is a member of the B-box zinc finger gene family many of which may function in growth regulation and in the appropriate context become oncogenic. Members of this family are nuclear proteins that possess a characteristic tripartite motif consisting of the RING and B-box zinc binding domains and a coiled-coil domain. The promyelocytic leukemia gene (PML), another B-box family member, produces a protein product that is detected within punctate nuclear structures called PML nuclear bodies (NBs) or PML oncogenic domains (PODs). These NBs are complex structures that consist of a number of different proteins many of which have yet to be identified. In the disease acute promyelocytic leukemia (APL) a fusion protein, PML-RARA, is produced through the t(15:17) translocation. In APL the morphology of the NBs is altered. We report that rfp co-localizes with PML in a subset of the PML NBs and that it interacts directly with PML. This interaction is mediated through the rfp B-box and the distal two coils. In contrast, homomultimerization of rfp preferentially involves the B-box and the proximal coil. The association of rfp with the PML NBs is altered by mutations that affect rfp/PML interaction and in NB4 cells that are derived from APL patients. When treated with retinoic acid, rfp reassociates with the NBs in a pattern similar to non APL cells. Additionally, we found that rfp colocalizes with PML-RARA protein produced in APL patients. These results suggest that rfp, along with the other known/unknown components of PML NBs, have an important role in regulating cellular growth and differentiation.


Subject(s)
Cell Nucleus/chemistry , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Leukemia, Promyelocytic, Acute/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Cell Differentiation/physiology , Cell Division/physiology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fibrosarcoma , Humans , Molecular Sequence Data , Mutagenesis/physiology , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Binding/physiology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transcription Factors/analysis , Transcription Factors/genetics , Transfection , Tretinoin/pharmacology , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/physiology , Tumor Suppressor Proteins , Zinc Fingers/genetics
13.
J Cell Sci ; 110 ( Pt 18): 2197-205, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9378769

ABSTRACT

The human protein PML, was first identified as part of a fusion protein with retinoic acid receptor alpha as found in the chromosomal translocation which gives rise to acute promyelocytic leukaemia. PML is normally localised to large matrix-associated nuclear domains (known as ND10, Kr bodies, PODS or PML NBs) which comprise several multi-protein complexes. Within the PML protein, there are a number of identified zinc-binding domains, one of which called the RING finger is found in a large family of diverse and unrelated proteins. Here, we report the effect of site-directed mutations within the context of the whole PML protein, of amino acids found on the surface of the PML RING finger domain and PML NB formation in vivo. Mutations of a small region of the RING finger domain surface affect the size and numbers of PML NBs in a mouse fibroblast expression assay, resulting in fewer but larger exogenous PML NBs. Mutations of other surface RING residues, however, do not affect exogenous PML NB formation. Furthermore, all of the PML RING mutants co-localise to both endogenous and exogenous wild-type PML NBs. These data identify a specific region of the PML RING finger domain which is directly involved in correct PML NB formation. They also provide evidence to suggest that the PML RING finger is involved in mediating PML-PML oligomeric interactions, as part of a mechanism leading to the assembly of the PML NB complex.


Subject(s)
Neoplasm Proteins , Nuclear Matrix/chemistry , Nuclear Proteins , Transcription Factors/chemistry , Transcription Factors/genetics , Zinc Fingers/genetics , 3T3 Cells/physiology , Animals , Glutamic Acid , HeLa Cells , Humans , Lysine , Mice , Mutagenesis, Site-Directed/physiology , Promyelocytic Leukemia Protein , Protein Structure, Tertiary , Static Electricity , Transfection , Tumor Suppressor Proteins
14.
Oncogene ; 14(13): 1563-70, 1997 Apr 03.
Article in English | MEDLINE | ID: mdl-9129147

ABSTRACT

Retinoid-induced proliferation causing hyperleukocytosis is a severe complication of retinoid therapy in t(15;17) acute promyelocytic leukaemia. The molecular basis of this phenomenon is unknown. It is possible that the transiently enhanced cell proliferation results from RA-induction of growth regulatory genes. Using Differential Display of cDNAs from NB4 cells we have identified Jem, a novel gene transcript which is upregulated by retinoids during the early proliferative response in maturating cells but not in resistant cells. A 2.7 kb cDNA was cloned and sequenced. The open reading frame contains a 400 amino acid sequence corresponding to a novel 45 kDa basic protein (pI 8.9). The JEM DNA sequence is detected by FISH on human chromosome 1 at q24. The Jem peptide sequence shows a 'leucine-zipper' dimerisation motif with limited homology to Fos/Jun and ATF/CREB proteins and several putative phosphorylation sites. An atypical basic region may correspond to an unknown DNA-binding domain. The C-terminal end of Jem spans a long stretch featuring a PEST motif. After transfection into COS cells, the Jem protein shows a ponctuated nuclear localisation. We hypothesise that this novel nuclear factor may act as a transcription factor, or a coregulator, involved in either cell growth control and/or maturation.


Subject(s)
Gene Expression Regulation, Neoplastic , Leucine Zippers , Leukemia, Promyelocytic, Acute/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Tretinoin/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , COS Cells , Cell Nucleus/chemistry , Chromosome Mapping , Chromosomes, Human, Pair 1 , Cloning, Molecular , DNA, Complementary , Dimerization , Humans , In Situ Hybridization, Fluorescence , Leukemia, Promyelocytic, Acute/metabolism , Molecular Sequence Data , Nuclear Proteins/analysis , Nuclear Proteins/chemistry , Phosphorylation , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Transcription Factors/analysis , Transcription Factors/chemistry , Transcription, Genetic , Transfection
15.
Oncogene ; 14(3): 349-57, 1997 Jan 23.
Article in English | MEDLINE | ID: mdl-9018121

ABSTRACT

The human TEL gene is involved in several 12p13 chromosomal abnormalities present in various human hematological malignancies, the most frequent being the t(12;21)(p13;q22), specific for childhood acute lymphoblastic leukemia. The predicted product of TEL harbours an amino acid region similar to the ETS DNA binding domain. We now report the isolation of the murine TEL cDNA and the characterization of the human TEL proteins. Human and murine TEL proteins are particularly homologous within their aminoterminal regions and their ETS domains. TEL proteins are nuclear and display specific DNA binding activity toward classical ETS binding sites. In addition, we show that TEL mRNAs initiate translation at either of the two first inframe ATGs (codon 1 and 43) to encode 50 kDa and 57 kDa TEL proteins. In vivo, each of these primary translational products is modified by multiple phosphorylation events.


Subject(s)
DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Repressor Proteins , Transcription Factors/genetics , Animals , Base Sequence , Blotting, Western , COS Cells , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , Cloning, Molecular , DNA/metabolism , DNA, Complementary/isolation & purification , DNA-Binding Proteins/isolation & purification , Humans , In Situ Hybridization, Fluorescence , Leukemia, B-Cell/genetics , Mice , Molecular Sequence Data , Nuclear Proteins/isolation & purification , Phosphoproteins/isolation & purification , Phosphorylation , Proto-Oncogene Proteins c-ets , RNA, Messenger/isolation & purification , Transcription Factors/isolation & purification , Translocation, Genetic , Tumor Cells, Cultured , ETS Translocation Variant 6 Protein
16.
Blood ; 90(12): 4876-85, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9389704

ABSTRACT

Acute promyelocytic leukemia (APL) is typified by the reciprocal translocation, t(15; 17)(q22; q21), leading to the formation of PML-RARalpha and RARalpha-PML fusion genes. We have characterized 7 cases of morphologic APL found to lack the t(15; 17) on conventional cytogenetic assessment. In 6 of 7 cases, cryptic PML-RARalpha rearrangements were identified by reverse transcriptase-polymerase chain reaction and fluorescent in situ hybridization (FISH); whereas, in the remaining patient, APL was associated with the variant translocation, t(11; 17)(q23; q12-21), leading to the formation of PLZF-RARalpha and RARalpha-PLZF fusion genes. In each of the cases with cryptic PML-RARalpha rearrangements, PML-RARalpha transcripts were detected in the absence of RARalpha-PML, consistent with the concept that PML-RARalpha is the critical oncogenic fusion protein. In 4 of these cases with evaluable metaphase spreads, the occurrence of a nonreciprocal translocation was confirmed by FISH with sole formation of the PML-RARalpha fusion gene; in 3 cases with morphologically normal chromosomes 15 and 17, RARalpha was inserted into PML on 15q, whereas in the remaining patient the PML-RARalpha fusion arose due to insertion of 15q-derived material including PML into RARalpha on 17q. Immunofluorescence studies were performed using antibodies raised against PML and PIC 1, a ubiquitin-homology domain protein previously identified as an interaction partner of PML. In acute myeloid leukemia (AML) of subtypes other than M3, PIC 1 was localized to the nuclear membrane and colocalized with PML within discrete nuclear bodies. In APL cases with cryptic PML-RARalpha rearrangements, the characteristic microparticulate pattern of PML staining was detected with partial colocalization with PIC 1, indicative of disruption of the nuclear bodies; whereas in t(11; 17)-associated APL, PML and PIC 1 remained colocalized within discrete nuclear bodies, as observed in non-APL cases. Although deregulation of the putative growth suppressor PML and delocalization of other nuclear body constituents have been advocated to play a key role in the development of t(15; 17)-associated APL, the present study shows that disruption of PML nuclear bodies per se is not a prerequisite for the pathogenesis of APL.


Subject(s)
Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 17 , Gene Rearrangement , Leukemia, Promyelocytic, Acute/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Translocation, Genetic , Fluorescent Antibody Technique , Humans , In Situ Hybridization, Fluorescence
17.
Oncogene ; 12(11): 2443-50, 1996 Jun 06.
Article in English | MEDLINE | ID: mdl-8649786

ABSTRACT

The expression of retinoid receptors (RXRalpha, RARalpha and the chimeric form PML-RARalpha) was analysed both at the mRNA and protein level in the maturation sensitive NB4 and resistant NB4-R1 cell lines of t(15;17) promyelocytic leukemia (APL). All-trans RA and cAMP which show synergistic activity in inducing maturation of NB4 cells and maturation triggering of the RA 'primed' NB4-R1 resistant cells, distinctly modulate RXRalpha, RARalpha and PML-RARalpha mRNA. In the NB4 and NB4-R1 cells, RXRalpha mRNA was downregulated by RA, but only in RA-primed NB4-R1 cells a release from RXRalpha mRNA downregulation was obtained by cAMP treatment. RXRalpha protein (53 kDa) was decreased to the western-blot detection limit (97.5%) by RA in NB4 cells, but in NB4-R1 cells although it was frankly decreased (85%), the signal for RXRalpha protein remained very significant. More importantly, while cAMP slightly upregulated RXRalpha protein in RA-treated NB4 cells, it caused an increase of RXRalpha protein in RA-treated NB4-R1 cells bringing RXRalpha to the initial control level. RXRalpha partners in heterodimers (PML-RARalpha, RARalpha) were also analysed. In contrast to RXRalpha, RARalpha and PML-RARalpha mRNA were not modulated by RA and/or cAMP, while significant changes were observed at the protein levels. A putatively phosphorylated form of RARalpha (52 kDa) decreased during maturation of NB4 cells, but was unchanged in resistant NB4-R1 cells. Conversely, while PML-RARalpha remained stable during RA-induced NB4 maturation, RA treatment which failed to induce maturation of NB4-R1 cells significantly down-regulated the chimeric receptor (120 kDa). These differences most likely results from translational and post-translational regulation. This work reveals complex pattern of subtle changes at the protein level distinguishing RA-sensitive and RA-resistant cells. Our data show that the RA-cAMP synergistic effect on NB4 cell maturation and cooperation in triggering maturation of RA-primed NB4-R1 cells operate changes in the RXR/PML-RARalpha ratio which are both favouring RXRalpha. In both cell lines, variations of PML-RARalpha and RXRalpha may result in a decrease in the formation of the PML-RARalpha/RXRalpha heterodimers which are supposed involved in the block of maturation. This may prove crucial to embark cells on maturation.


Subject(s)
Cyclic AMP/pharmacology , Leukemia, Promyelocytic, Acute/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , RNA, Messenger/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , Cell Cycle , Cell Nucleus/metabolism , Down-Regulation , Humans , Leukemia, Promyelocytic, Acute/pathology , Retinoic Acid Receptor alpha , Retinoid X Receptors , Tumor Cells, Cultured
18.
Oncogene ; 12(11): 2451-9, 1996 Jun 06.
Article in English | MEDLINE | ID: mdl-8649787

ABSTRACT

We analysed the expression of retinoid receptors and PML in relation to the morphology of PML-containing nuclear bodies (PODs) in maturation sensitive (NB4) and resistant subclones (NB4-R1 and R2) of the promyelocytic NB4 cell line. The basal level of RARalpha, RXRalpha and PML mRNA and protein were roughly the same in the three cell lines. While NB4 and NB4-R1 cells express comparable amounts of PM-RARalpha mRNA and 120 kDa protein, NB4-R2 cells despite normal mRNA levels the 120 kDa protein was not detectable. In NB4-R2 cells however, two novel PML-related entities of 65 kDa and 85 kDA were detected with a anti-PML antibody, in addition to the two PML isoforms of 78 and 97 kDa found in any NB4 cells. Despite the 120 kDa PML-RARalpha defect, NB4-R2 cells show micropunctuated nuclear bodies typical of APL cells. Contrasting with NB4 cells, neither NB4-R1 cells which express PML-RARalpha, nor NB4-R2 cells lacking the 120 kDa PML-RARalpha reorganised nuclear bodies (PODs) in response to RA. Importantly, in RA-primed NB4-R1 cells, a secondary event triggered by cAMP restored PODs, concomitant to maturation. This indicates that the recovery of nuclear bodies in APL is dissociated from the early action of RA in cell maturation. Finally, the key finding of this work is that cAMP signalling ultimately determines the recovery of nuclear bodies associated to cell maturation.


Subject(s)
Cell Nucleus/drug effects , Cyclic AMP/pharmacology , Leukemia, Promyelocytic, Acute/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins , Oncogene Proteins, Fusion/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , Cell Cycle/drug effects , Cell Nucleus/metabolism , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 17 , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Promyelocytic Leukemia Protein , RNA, Messenger/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors , Translocation, Genetic , Tumor Cells, Cultured , Tumor Suppressor Proteins
19.
Proc Natl Acad Sci U S A ; 91(18): 8428-32, 1994 Aug 30.
Article in English | MEDLINE | ID: mdl-7915840

ABSTRACT

In t(15;17) acute promyelocytic leukemia, all-trans retinoic acid (RA) induces leukemic cell maturation in vitro and remission in acute promyelocytic leukemia patients, but in vivo treatments invariably lead to relapse with resistance to RA. NB4, a maturation-inducible cell line, and NB4-RAr sublines (R1 and R2) displaying no maturation in the presence of RA have been isolated from a patient in relapse. We show that resistance to maturation is not a mere unresponsiveness to RA: rather, R1 "resistant" cells do respond to RA (1 microM) by sustained growth, become competent to undergo terminal maturation, and up-regulate CD11c/CD18 integrins. Interestingly, maturation of "resistant" cells, rendered competent by RA, can be achieved by cAMP-elevating agents (prostaglandin E, isoproterenol, cholera toxin, or phosphodiesterase inhibitor) or stable agonistic cAMP analogs such as (SP)-8-chloroadenosine cyclic 3',5'-phosphorothioate. This shows that activation of cAMP-dependent protein kinase (cA kinase) can override the RA resistance and suggests interdependent RA and cAMP signaling pathways in acute promyelocytic leukemia maturation. No such cooperation was observed in the R2 resistant cells, though their cA-kinase was functional. (RP)-8-Chloroadenosine cyclic 3',5'-phosphorothioate, which by displacing endogenous cAMP inhibits the basal cA-kinase activity, decreased the response of sensitive cells to RA. This raises the possibility that cA-kinase plays a key role in the maturation also of RA-sensitive cells. Our results define two discrete steps in the maturation process: an RA-dependent priming step that maintains proliferation while cells become competent to undergo maturation in response to retinoids and a cAMP-dependent step that triggers RA-primed cells to undergo terminal maturation. Uncoupling RA and cAMP action might cause the so-called "resistance."


Subject(s)
Cell Differentiation/drug effects , Leukemia, Promyelocytic, Acute/pathology , Tretinoin/pharmacology , Antigens, CD/metabolism , CD11 Antigens , CD18 Antigens , Cyclic AMP/physiology , Drug Resistance , Humans , In Vitro Techniques , Integrins/metabolism , Signal Transduction , Tumor Cells, Cultured
20.
J Biol Chem ; 268(11): 8332-40, 1993 Apr 15.
Article in English | MEDLINE | ID: mdl-8385140

ABSTRACT

cAMP induced rapid apoptosis (> 90% cell death in 6 h) of non-growth-arrested rat leukemia IPC-81 cells. A cell clone selected for cAMP resistance had a normally functioning apoptotic machinery whose triggering required about 30-fold higher cellular cAMP than in the parent cells. The cAMP subresponsiveness was due to a heterozygous point mutation (Ala336-->Asp) in the RI subunit of cAMP-dependent protein kinase I. In fact, apoptosis correlated with intracellular cAMP binding to the subresponsive RI. The mutated alanine is invariantly present in cyclic nucleotide kinases, but of unknown function. The mutation decreased the cAMP affinity to site B by increasing the cAMP dissociation rate 500x. The ability of site B to discriminate adenine-modified cAMP analogues was affected, suggesting that Ala336 faced the adenine moiety of cAMP. That the heterozygously expressed RID336 was a dominant suppressor of apoptosis was explained by a higher expression of R than C subunits in the mutant cells by preferential expression of the mutant form of RI, and by the ability of mutant RI to exert dominant negative control of activation of wild type cAMP kinase at moderate cAMP levels. Apoptosis was induced at a similar cAMP level in cells treated with cholera toxin or other cAMP elevating agents, indicating that cAMP kinase was essential for toxin action.


Subject(s)
Alanine , Apoptosis/physiology , Aspartic Acid , Cyclic AMP/pharmacology , Point Mutation , Protein Kinases/genetics , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Base Sequence , Binding Sites , Blotting, Northern , Blotting, Southern , Cell Nucleus/metabolism , Clone Cells , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Drug Resistance/physiology , Enzyme Activation , Heterozygote , Kinetics , Leukemia, Experimental , Macromolecular Substances , Molecular Sequence Data , Oligonucleotide Probes , Polymerase Chain Reaction/methods , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Rats , Restriction Mapping , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...