Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Blood ; 137(1): 89-102, 2021 01 07.
Article in English | MEDLINE | ID: mdl-32818241

ABSTRACT

The role of ribosome biogenesis in erythroid development is supported by the recognition of erythroid defects in ribosomopathies in both Diamond-Blackfan anemia and 5q- syndrome. Whether ribosome biogenesis exerts a regulatory function on normal erythroid development is still unknown. In the present study, a detailed characterization of ribosome biogenesis dynamics during human and murine erythropoiesis showed that ribosome biogenesis is abruptly interrupted by the decline in ribosomal DNA transcription and the collapse of ribosomal protein neosynthesis. Its premature arrest by the RNA Pol I inhibitor CX-5461 targeted the proliferation of immature erythroblasts. p53 was activated spontaneously or in response to CX-5461, concomitant to ribosome biogenesis arrest, and drove a transcriptional program in which genes involved in cell cycle-arrested, negative regulation of apoptosis, and DNA damage response were upregulated. RNA Pol I transcriptional stress resulted in nucleolar disruption and activation of the ATR-CHK1-p53 pathway. Our results imply that the timing of ribosome biogenesis extinction and p53 activation is crucial for erythroid development. In ribosomopathies in which ribosome availability is altered by unbalanced production of ribosomal proteins, the threshold downregulation of ribosome biogenesis could be prematurely reached and, together with pathological p53 activation, prevents a normal expansion of erythroid progenitors.


Subject(s)
Cell Differentiation/physiology , Erythroid Cells/cytology , Erythropoiesis/physiology , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Hematopoietic Stem Cells , Humans , Mice , Organelle Biogenesis
2.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37175558

ABSTRACT

One important environmental/health challenge is to determine, in a feasible way, the potential carcinogenic risk associated with environmental agents/exposures. Since a significant proportion of tumors have an environmental origin, detecting the potential carcinogenic risk of environmental agents is mandatory, as regulated by national and international agencies. The challenge mainly implies finding a way of how to overcome the inefficiencies of long-term trials with rodents when thousands of agents/exposures need to be tested. To such an end, the use of in vitro cell transformation assays (CTAs) was proposed, but the existing prevalidated CTAs do not cover the complexity associated with carcinogenesis processes and present serious limitations. To overcome such limitations, we propose to use a battery of assays covering most of the hallmarks of the carcinogenesis process. For the first time, we grouped such assays as early, intermediate, or advanced biomarkers which allow for the identification of the cells in the initiation, promotion or aggressive stages of tumorigenesis. Our proposal, as a novelty, points out that using a battery containing assays from all three groups can identify if a certain agent/exposure can pose a carcinogenic risk; furthermore, it can gather mechanistic insights into the mode of the action of a specific carcinogen. This structured battery could be very useful for any type of in vitro study, containing human cell lines aiming to detect the potential carcinogenic risks of environmental agents/exposures. In fact, here, we include examples in which these approaches were successfully applied. Finally, we provide a series of advantages that, we believe, contribute to the suitability of our proposed approach for the evaluation of exposure-induced carcinogenic effects and for the development of an alternative strategy for conducting an exposure risk assessment.


Subject(s)
Environmental Pollutants , Neoplasms , Humans , Carcinogens/toxicity , Environmental Pollutants/toxicity , Neoplasms/chemically induced , Environmental Exposure/adverse effects , Cell Transformation, Neoplastic/chemically induced
3.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555673

ABSTRACT

Prostate cancer is a major public health concern and one of the most prevalent forms of cancer worldwide. The definition of altered signaling pathways implicated in this complex disease is thus essential. In this context, abnormal expression of the receptor of Macrophage Colony-Stimulating Factor-1 (M-CSF or CSF-1) has been described in prostate cancer cells. Yet, outcomes of this expression remain unknown. Using mouse and human prostate cancer cell lines, this study has investigated the functionality of the wild-type CSF-1 receptor in prostate tumor cells and identified molecular mechanisms underlying its ligand-induced activation. Here, we showed that upon CSF-1 binding, the receptor autophosphorylates and activates multiple signaling pathways in prostate tumor cells. Biological experiments demonstrated that the CSF-1R/CSF-1 axis conferred significant advantages in cell growth and cell invasion in vitro. Mouse xenograft experiments showed that CSF-1R expression promoted the aggressiveness of prostate tumor cells. In particular, we demonstrated that the ligand-activated CSF-1R increased the expression of spp1 transcript encoding for osteopontin, a key player in cancer development and metastasis. Therefore, this study highlights that the CSF-1 receptor is fully functional in a prostate cancer cell and may be a potential therapeutic target for the treatment of prostate cancer.


Subject(s)
Osteopontin , Prostatic Neoplasms , Receptor, Macrophage Colony-Stimulating Factor , Animals , Humans , Male , Mice , Ligands , Macrophage Colony-Stimulating Factor/metabolism , Osteopontin/genetics , Prostatic Neoplasms/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism
4.
Blood ; 130(26): 2860-2871, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29138221

ABSTRACT

The BCR-ABL specific tyrosine kinase inhibitors (TKI) changed the outcome of chronic myeloid leukemia (CML), turning a life-threatening disease into a chronic illness. However, TKI are not yet curative, because most patients retain leukemic stem cells (LSC) and their progenitors in bone marrow and relapse following treatment cessation. At diagnosis, deregulation of the bone morphogenetic protein (BMP) pathway is involved in LSC and progenitor expansion. Here, we report that BMP pathway alterations persist in TKI-resistant patients. In comparison with patients in complete cytogenetic remission, TKI-resistant LSC and progenitors display high levels of BMPR1b expression and alterations of its cellular localization. In vitro treatment of immature chronic phase CML cells with TKI alone, or in combination with interferon-α, results in the preferential survival of BMPR1b+ cells. We demonstrated persistent and increasing BMP4 production by patients' mesenchymal cells with resistance. Patient follow-up revealed an increase of BMPR1b expression and in BMP4 expression in LSC from TKI-resistant patients in comparison with diagnosis, while remaining unchanged in sensitive patients. Both leukemic and nonleukemic cells exhibit higher BMP4 levels in the bone marrow of TKI-resistant patients. Exposure to BMP2/BMP4 does not alter BCR-ABL transcript expression but is accompanied by the overexpression of TWIST-1, a transcription factor highly expressed in resistant LSC. By modulating BMP4 or BMPR1b expression, we show that these elements are involved in TKI resistance. In summary, we reveal that persistence of BMP alterations and existence of an autocrine loop promote CML-primitive cells' TKI resistance.


Subject(s)
Autocrine Communication , Bone Morphogenetic Proteins/metabolism , Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/therapeutic use , Bone Morphogenetic Protein 4/analysis , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors, Type I/analysis , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/analysis , Humans , Neoplastic Stem Cells/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Twist-Related Protein 1/analysis , Twist-Related Protein 1/metabolism
5.
J Cell Sci ; 125(Pt 5): 1141-51, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22302993

ABSTRACT

We previously found that the NF-κB transcription factor is activated during the recovery period after heat shock; moreover, we demonstrated that NF-κB is essential for cell survival after heat shock by activating autophagy, a mechanism that probably helps the cell to cope with hyperthermic stress through clearance of damaged proteins. In this study, we analyze the involvement of NF-κB in basal and heat-stress-induced protein quality control, by comparing the level of multiubiquitylated and/or aggregated proteins, and proteasome and autophagic activity in NF-κB-competent and NF-κB-incompetent cells. We show that NF-κB has only a minor role in basal protein quality control, where it modulates autophagosome maturation. By contrast, NF-κB is shown to be a key player in protein quality control after hyperthermia. Indeed, NF-κB-incompetent cells show highly increased levels of multiubiquitylated and/or aggregated proteins and aggresome clearance defects; a phenotype that disappears when NF-κB activity is restored to normal. We demonstrate that during heat shock recovery NF-κB activates selective removal of misfolded or aggregated proteins--a process also called 'aggrephagy'--by controlling the expression of BAG3 and HSPB8 and by modulating the level of the BAG3-HspB8 complex. Thus NF-κB-mediated increase in the level of the BAG3-HspB8 complex leads to upregulation of aggrephagy and clearance of irreversibly damaged proteins and might increase cell survival in conditions of hyperthermia.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy/physiology , Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Survival , HeLa Cells , Humans , Molecular Chaperones , NF-kappa B/genetics , Protein Folding , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics , Ubiquitination
6.
Cancer Res Commun ; 3(9): 1966-1980, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37707389

ABSTRACT

An accurate estimate of patient survival at diagnosis is critical to plan efficient therapeutic options. A simple and multiapplication tool is needed to move forward the precision medicine era. Taking advantage of the broad and high CD10 expression in stem and cancers cells, we evaluated the molecular identity of aggressive cancer cells. We used epithelial primary cells and developed a breast cancer stem cell­based progressive model. The superiority of the early-transformed isolated molecular index was evaluated by large-scale analysis in solid cancers. BMP2-driven cell transformation increases CD10 expression which preserves stemness properties. Our model identified a unique set of 159 genes enriched in G2­M cell-cycle phases and spindle assembly complex. Using samples predisposed to transformation, we confirmed the value of an early neoplasia index associated to CD10 (ENI10) to discriminate premalignant status of a human tissue. Using a stratified Cox model, a large-scale analysis (>10,000 samples, The Cancer Genome Atlas Pan-Cancer) validated a strong risk gradient (HRs reaching HR = 5.15; 95% confidence interval: 4.00­6.64) for high ENI10 levels. Through different databases, Cox regression model analyses highlighted an association between ENI10 and poor progression-free intervals for more than 50% of cancer subtypes tested, and the potential of ENI10 to predict drug efficacy. The ENI10 index constitutes a robust tool to detect pretransformed tissues and identify high-risk patients at diagnosis. Owing to its biological link with refractory cancer stem cells, the ENI10 index constitutes a unique way of identifying effective treatments to improve clinical care. SIGNIFICANCE: We identified a molecular signature called ENI10 which, owing to its biological link with stem cell properties, predicts patient outcome and drugs efficiency in breast and several other cancers. ENI10 should allow early and optimized clinical management of a broad number of cancers, regardless of the stage of tumor progression.


Subject(s)
Neoplasms , Humans , Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Neprilysin
7.
Blood ; 116(23): 4795-805, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20733157

ABSTRACT

This study investigated the role of the ETS transcription factor Fli-1 in adult myelopoiesis using new transgenic mice allowing inducible Fli-1 gene deletion. Fli-1 deletion in adult induced mild thrombocytopenia associated with a drastic decrease in large mature megakaryocytes number. Bone marrow bipotent megakaryocytic-erythrocytic progenitors (MEPs) increased by 50% without increase in erythrocytic and megakaryocytic common myeloid progenitor progeny, suggesting increased production from upstream stem cells. These MEPs were almost unable to generate pure colonies containing large mature megakaryocytes, but generated the same total number of colonies mainly identifiable as erythroid colonies containing a reduced number of more differentiated cells. Cytological and fluorescence-activated cell sorting analyses of MEP progeny in semisolid and liquid cultures confirmed the drastic decrease in large mature megakaryocytes but revealed a surprisingly modest (50%) reduction of CD41-positive cells indicating the persistence of a megakaryocytic commitment potential. Symmetrical increase and decrease of monocytic and granulocytic progenitors were also observed in the progeny of purified granulocytic-monocytic progenitors and common myeloid progenitors. In summary, this study indicates that Fli-1 controls several lineages commitment decisions at the stem cell, MEP, and granulocytic-monocytic progenitor levels, stimulates the proliferation of committed erythrocytic progenitors at the expense of their differentiation, and is a major regulator of late stages of megakaryocytic differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Erythrocytes/cytology , Erythropoiesis/genetics , Megakaryocytes/cytology , Proto-Oncogene Protein c-fli-1/genetics , Animals , Blotting, Western , Cell Proliferation , Cell Separation , Flow Cytometry , Gene Deletion , Hematopoietic Stem Cells/cytology , Mice , Mice, Transgenic , Myeloid Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction
8.
Blood ; 115(17): 3463-71, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20154211

ABSTRACT

Precise spatiotemporal control of Gata1 expression is required in both early hematopoietic progenitors to determine erythroid/megakaryocyte versus granulocyte/monocyte lineage output and in the subsequent differentiation of erythroid cells and megakaryocytes. An enhancer element upstream of the mouse Gata1 IE (1st exon erythroid) promoter, mHS-3.5, can direct both erythroid and megakaryocytic expression. However, loss of this element ablates only megakaryocytes, implying that an additional element has erythroid specificity. Here, we identify a double DNaseI hypersensitive site, mHS-25/6, as having erythroid but not megakaryocytic activity in primary cells. It binds an activating transcription factor complex in erythroid cells where it also makes physical contact with the Gata1 promoter. Deletion of mHS-25/6 or mHS-3.5 in embryonic stem cells has only a modest effect on in vitro erythroid differentiation, whereas loss of both elements ablates both primitive and definitive erythropoiesis with an almost complete loss of Gata1 expression. Surprisingly, Gata2 expression was also concomitantly low, suggesting a more complex interaction between these 2 factors than currently envisaged. Thus, whereas mHS-3.5 alone is sufficient for megakaryocytic development, mHS-3.5 and mHS-25/6 collectively regulate erythroid Gata1 expression, demonstrating lineage-specific differences in Gata1 cis-element use important for development of these 2 cell types.


Subject(s)
Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/physiology , Erythroid Cells/metabolism , Erythropoiesis/physiology , GATA1 Transcription Factor/biosynthesis , Gene Expression Regulation/physiology , Megakaryocytes/metabolism , Animals , Embryonic Stem Cells/cytology , Erythroid Cells/cytology , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Megakaryocytes/cytology , Mice , Promoter Regions, Genetic/physiology , Sequence Deletion
9.
Front Cell Dev Biol ; 9: 787989, 2021.
Article in English | MEDLINE | ID: mdl-35047500

ABSTRACT

Understanding mechanisms of cancer development is mandatory for disease prevention and management. In healthy tissue, the microenvironment or niche governs stem cell fate by regulating the availability of soluble molecules, cell-cell contacts, cell-matrix interactions, and physical constraints. Gaining insight into the biology of the stem cell microenvironment is of utmost importance, since it plays a role at all stages of tumorigenesis, from (stem) cell transformation to tumor escape. In this context, BMPs (Bone Morphogenetic Proteins), are key mediators of stem cell regulation in both embryonic and adult organs such as hematopoietic, neural and epithelial tissues. BMPs directly regulate the niche and stem cells residing within. Among them, BMP2 and BMP4 emerged as master regulators of normal and tumorigenic processes. Recently, a number of studies unraveled important mechanisms that sustain cell transformation related to dysregulations of the BMP pathway in stem cells and their niche (including exposure to pollutants such as bisphenols). Furthermore, a direct link between BMP2/BMP4 binding to BMP type 1 receptors and the emergence and expansion of cancer stem cells was unveiled. In addition, a chronic exposure of normal stem cells to abnormal BMP signals contributes to the emergence of cancer stem cells, or to disease progression independently of the initial transforming event. In this review, we will illustrate how the regulation of stem cells and their microenvironment becomes dysfunctional in cancer via the hijacking of BMP signaling with main examples in myeloid leukemia and breast cancers.

10.
iScience ; 24(3): 102210, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33733070

ABSTRACT

Transcription factors are key players in a broad range of cellular processes such as cell-fate decision. Understanding how they act to control these processes is of critical importance for therapy purposes. FLI-1 controls several hematopoietic lineage differentiation including megakaryopoiesis and erythropoiesis. Its aberrant expression is often observed in cancer and is associated with poor prognosis. We showed that FLI-1 interacts with the LDB1 complex, which also plays critical roles in erythropoiesis and megakaryopoiesis. In this study, we aimed to unravel how FLI-1 and the LDB1 complex act together in murine erythroleukemia cells and in megakaryocyte. Combining omics techniques, we show that FLI-1 enables the recruitment of the LDB1 complex to regulatory sequences of megakaryocytic genes and to enhancers. We show as well for the first time that FLI-1 is able to modulate the 3D chromatin organization by promoting chromatin looping between enhancers and promoters most likely through the LDB1 complex.

11.
Blood ; 112(3): 576-84, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18523154

ABSTRACT

Previous observations suggested that functional antagonism between FLI-1 and EKLF might be involved in the commitment toward erythrocytic or megakaryocytic differentiation. We show here, using inducible shRNA expression, that EKLF knockdown in mouse erythroleukemia (MEL) cells decreases erythrocytic and increases megakaryocytic as well as Fli-1 gene expression. Chromatin immunoprecipitation analyses revealed that the increase in megakaryocytic gene expression is associated with a marked increase in RNA pol II and FLI-1 occupancy at their promoters, albeit FLI-1 protein levels are only minimally affected. Similarly, we show that human CD34(+) progenitors infected with shRNA lentivirus allowing EKLF knockdown generate an increased number of differentiated megakaryocytic cells associated with increased levels of megakaryocytic and Fli-1 gene transcripts. Single-cell progeny analysis of a cell population enriched in bipotent progenitors revealed that EKLF knockdown increases the number of megakaryocytic at the expense of erythrocytic colonies. Taken together, these data indicate that EKLF restricts megakaryocytic differentiation to the benefit of erythrocytic differentiation and suggest that this might be at least partially mediated by the inhibition of FLI-1 recruitment to megakaryocytic and Fli-1 gene promoters.


Subject(s)
Cell Differentiation , Erythrocytes/cytology , Kruppel-Like Transcription Factors/physiology , Megakaryocytes/cytology , Animals , Cell Line , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/genetics , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology
12.
Sci Rep ; 10(1): 20266, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33219251

ABSTRACT

Assessment of autophagy activity has historically been limited to investigations of fixed tissue or bulk cell populations. To address questions of heterogeneity and relate measurements to functional properties of viable cells isolated from primary tissue, we created a lentiviral (RFP-GFP-MAP1LC3B) vector that allows the autophagosome and autolysosome content of transduced cells to be monitored at the single-cell level. Use of this strategy to analyze purified subsets of normal human mammary cells showed that both the luminal progenitor-containing (LP) subset and the basal cells (BCs) display highly variable but overall similar autophagic flux activity despite differences suggested by measurements of the proteins responsible (i.e., LC3B, ATG7 and BECLIN1) in bulk lysates. Autophagosome content was also highly variable in the clonogenic cells within both the LPs and BCs, but the proliferative response of the BCs was more sensitive to autophagy inhibition. In addition, use of this vector showed cells with the lowest autophagosome content elicited the fastest tumor growth in 2 different models of human mammary tumorigenesis. These results illustrate the utility of this vector to define differences in the autophagy properties of individual cells in primary tissue and couple these with their responses to proliferative and oncogenic stimuli.


Subject(s)
Autophagy , Mammary Glands, Human/cytology , Single-Cell Analysis/methods , Cell Line, Transformed , Humans
13.
Cancers (Basel) ; 11(9)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31547326

ABSTRACT

Estrogens are major regulators of the mammary gland development, notably during puberty, via estrogen receptor (ER) activation, leading to the proliferation and differentiation of mammary cells. In addition to estrogens, the bone morphogenetic proteins (BMPs) family is involved in breast stem cell/progenitor commitment. However, these two pathways that synergistically contribute to the biology of the normal mammary gland have also been described to initiate and/or promote breast cancer development. In addition to intrinsic events, lifestyle habits and exposure to environmental cues are key risk factors for cancer in general, and especially for breast cancer. In the latter case, bisphenol A (BPA), an estrogen-mimetic compound, is a critical pollutant both in terms of the quantities released in our environment and of its known and speculated effects on mammary gland biology. In this review, we summarize the current knowledge on the actions of BMPs and estrogens in both normal mammary gland development and breast cancer initiation, dissemination, and resistance to treatment, focusing on the dysregulations of these processes by BPA but also by other bisphenols, including BPS and BPF, initially considered as safer alternatives to BPA.

14.
Mol Cell Biol ; 25(23): 10235-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16287841

ABSTRACT

Lineage specification and cellular maturation require coordinated regulation of gene expression programs. In large part, this is dependent on the activator and repressor functions of protein complexes associated with tissue-specific transcriptional regulators. In this study, we have used a proteomic approach to characterize multiprotein complexes containing the key hematopoietic regulator SCL in erythroid and megakaryocytic cell lines. One of the novel SCL-interacting proteins identified in both cell types is the transcriptional corepressor ETO-2. Interaction between endogenous proteins was confirmed in primary cells. We then showed that SCL complexes are shared but also significantly differ in the two cell types. Importantly, SCL/ETO-2 interacts with another corepressor, Gfi-1b, in red cells but not megakaryocytes. The SCL/ETO-2/Gfi-1b association is lost during erythroid differentiation of primary fetal liver cells. Genetic studies of erythroid cells show that ETO-2 exerts a repressor effect on SCL target genes. We suggest that, through its association with SCL, ETO-2 represses gene expression in the early stages of erythroid differentiation and that alleviation/modulation of the repressive state is then required for expression of genes necessary for terminal erythroid maturation to proceed.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Erythroid Cells/metabolism , Erythropoiesis , Megakaryocytes/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Erythroid Cells/cytology , Gene Expression Regulation , Mice , Mutation/genetics , Nuclear Proteins/genetics , Protein Binding , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid , Repressor Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/genetics , Transcription, Genetic/genetics
16.
Gene ; 296(1-2): 151-9, 2002 Aug 21.
Article in English | MEDLINE | ID: mdl-12383512

ABSTRACT

Mona/grb2 related adapter downstream of shc is a molecular adapter expressed in platelets, T lymphocytes and myelomonocytic cells. Using human hematopoietic cell lines, we have previously shown that lineage-specific Mona expression is achieved through the production of two transcripts (named 1A and 1B) differing by their 5' untranslated region (5'UTR). Thus, platelets and megakaryocytic cell lines K562 and HEL (Human Erythro-Leukemia) specifically express 1B messenger RNA (mRNA). We report here characterization of the (-2031/+72) genomic region relative to the putative transcription start site of 1B mRNA. We show this region is sufficient to ensure specific reporter gene expression in megakaryocytic cell lines, and that most promoter activity is contained in the (-225/+72) fragment. Electro-mobility shift assay and mutational analyses indicated that GATA-1 and a yet unidentified E-26 family member transcription factor are required for 1B (-2031/+72) promoter activity. Thus, Mona 1B promoter exhibits typical features of megakaryocyte-specific promoters.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Megakaryocytes/metabolism , Promoter Regions, Genetic/genetics , Base Sequence , Binding Sites/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Gene Expression Regulation/drug effects , Humans , Jurkat Cells , K562 Cells , Luciferases/genetics , Luciferases/metabolism , Megakaryocytes/pathology , Molecular Sequence Data , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Sequence Deletion , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , U937 Cells , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
17.
PLoS One ; 7(10): e46799, 2012.
Article in English | MEDLINE | ID: mdl-23056458

ABSTRACT

Clonal erythroleukemia developing in susceptible mice infected by Friend virus complex are associated with highly recurrent proviral insertions at one of three loci called Spi-1, Fli-1 or Fli-3, leading to deregulated expression of oncogenic Spi-1 or Fli-1 transcription factors or miR-17-92 miRNA cluster, respectively. Deregulated expression of each of these three oncogenes has been independently shown to contribute to cell proliferation of erythroleukemic clones. Previous studies showed a close relationship between Spi-1 and Fli-1, which belong to the same ETS family, Spi-1 activating fli-1 gene, and both Spi-1 and Fli-1 activating multiple common target genes involved in ribosome biogenesis. In this study, we demonstrated that Spi-1 and Fli-1 are also involved in direct miR-17-92 transcriptional activation through their binding to a conserved ETS binding site in its promoter. Moreover, we demonstrated that physiological re-expression of exogenous miR-17 and miR-20a are able to partially rescue the proliferation loss induced by Fli-1 knock-down and identified HBP1 as a target of these miRNA in erythroleukemic cells. These results establish that three of the most recurrently activated oncogenes in Friend erythroleukemia are actually involved in a same oncogenic network controlling cell proliferation. The putative contribution of a similar ETS-miR-17-92 network module in other normal or pathological proliferative contexts is discussed.


Subject(s)
Leukemia, Erythroblastic, Acute/metabolism , MicroRNAs/metabolism , Peptides/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Intercellular Signaling Peptides and Proteins , Leukemia, Erythroblastic, Acute/genetics , Mice , MicroRNAs/genetics , Peptides/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Protein c-fli-1/genetics
19.
Mol Cell Biol ; 29(10): 2852-64, 2009 May.
Article in English | MEDLINE | ID: mdl-19289502

ABSTRACT

Spi-1 and Fli-1 are ETS transcription factors recurrently deregulated in mouse erythroleukemia induced by Friend viruses. Since they share the same core DNA binding site, we investigated whether they may contribute to erythroleukemia by common mechanisms. Using inducible knockdown, we demonstrated that Fli-1 contributes to proliferation, survival, and differentiation arrest of erythroleukemic cells harboring an activated fli-1 locus. Similarly, we used inducible Fli-1 knockdown and either hexamethylenebisacetamide (HMBA)- or small interfering RNA-mediated Spi-1 knockdown to investigate their respective contributions in erythroleukemic cells harboring an activated spi-1 locus. In these cells, simple or double knockdown of both Spi-1 and Fli-1 additively contributed to induce proliferation arrest and differentiation. Transcriptome profiling revealed that virtually all transcripts affected by both Fli-1 knockdown and HMBA are affected in an additive manner. Among these additively downregulated transcripts, more than 20% encode proteins involved in ribosome biogenesis, and conserved ETS binding sites are present in their gene promoters. Through chromatin immunoprecipitation, we demonstrated the association of Spi-1 and Fli-1 on these promoters in Friend erythroleukemic cells. These data lead us to propose that the oncogenicity of Spi-1, Fli-1, and possibly other ETS transcription factors may involve their ability to stimulate ribosome biogenesis.


Subject(s)
Friend murine leukemia virus/metabolism , Leukemia, Erythroblastic, Acute , Peptides/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Ribosomes/metabolism , Tumor Cells, Cultured/physiology , Animals , Apoptosis/physiology , Cell Proliferation , Friend murine leukemia virus/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Intercellular Signaling Peptides and Proteins , Mice , Peptides/genetics , Phenotype , Proto-Oncogene Protein c-fli-1/genetics
20.
Proc Natl Acad Sci U S A ; 103(49): 18504-9, 2006 Dec 05.
Article in English | MEDLINE | ID: mdl-17132730

ABSTRACT

The Mediator complex forms the bridge between transcriptional activators and RNA polymerase II. Mediator subunit Med1/TRAP220 is a key component of Mediator originally found to associate with nuclear hormone receptors. Med1 deficiency causes lethality at embryonic day 11.5 because of defects in heart and placenta development. Here we show that Med1-deficient 10.5 days postcoitum embryos are anemic but have normal numbers of hematopoietic progenitor cells. Med1-deficient progenitor cells have a defect in forming erythroid burst-forming units (BFU-E) and colony-forming units (CFU-E), but not in forming myeloid colonies. At the molecular level, we demonstrate that Med1 interacts physically with the erythroid master regulator GATA-1. In transcription assays, Med1 deficiency leads to a defect in GATA-1-mediated transactivation. In chromatin immunoprecipitation experiments, we find Mediator components at GATA-1-occupied enhancer sites. Thus, we conclude that Mediator subunit Med1 acts as a pivotal coactivator for GATA-1 in erythroid development.


Subject(s)
Endodeoxyribonucleases/physiology , Erythropoiesis/physiology , GATA1 Transcription Factor/physiology , Protein Subunits/physiology , Transcription Factors/physiology , Animals , Cell Line , Embryonic Stem Cells/metabolism , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/genetics , Erythroid Precursor Cells/cytology , Mediator Complex Subunit 1 , Mice , Mice, Knockout , Proto-Oncogene Proteins c-kit/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL