Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Cell Int ; 24(1): 72, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347567

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most lethal primary brain tumor in adult, characterized by highly aggressive and infiltrative growth. The current therapeutic management of GBM includes surgical resection followed by ionizing radiations and chemotherapy. Complex and dynamic interplay between tumor cells and tumor microenvironment drives the progression and contributes to therapeutic resistance. Extracellular vesicles (EVs) play a crucial role in the intercellular communication by delivering bioactive molecules in the surrounding milieu modulating tumor microenvironment. METHODS: In this study, we isolated by ultracentrifugation EVs from GBM stem-like cell (GSC) lines and human microvascular endothelial cells (HMVECs) exposed or not to ionizing irradiation. After counting and characterization, we evaluated the effects of exposure of GSCs to EVs isolated from endothelial cells and vice versa. The RNA content of EVs isolated from GSC lines and HMVECs exposed or not to ionizing irradiation, was analyzed by RNA-Seq. Periostin (POSTN) and Filamin-B (FLNB) emerged in gene set enrichment analysis as the most interesting transcripts enriched after irradiation in endothelial cell-derived EVs and GSC-derived EVs, respectively. POSTN and FLNB expression was modulated and the effects were analyzed by in vitro assays. RESULTS: We confirmed that ionizing radiations increased EV secretion by GSCs and normal endothelial cells, affected the contents of and response to cellular secreted EVs. Particularly, GSC-derived EVs decreased radiation-induced senescence and promoted migration in HMVECs whereas, endothelial cell-derived EVs promoted tumorigenic properties and endothelial differentiation of GSCs. RNA-Seq analysis of EV content, identified FLNB and POSTN as transcripts enriched in EVs isolated after irradiation from GSCs and HMVECs, respectively. Assays performed on POSTN overexpressing GSCs confirmed the ability of POSTN to mimic the effects of endothelial cell-derived EVs on GSC migration and clonogenic abilities and transdifferentiation potential. Functional assays performed on HMVECs after silencing of FLNB supported its role as mediator of the effects of GSC-derived EVs on senescence and migration. CONCLUSION: In this study, we identified POSTN and FLNB as potential mediators of the effects of EVs on GSC and HMVEC behavior confirming that EVs play a crucial role in the intercellular communication by delivering bioactive molecules in the surrounding milieu modulating tumor microenvironment.

2.
Front Oncol ; 12: 867886, 2022.
Article in English | MEDLINE | ID: mdl-35814429

ABSTRACT

MiR-378a-3p plays a critical role in carcinogenesis acting as a tumor suppressor, promoting apoptosis and cell cycle arrest and reducing invasion and drug resistance in several human cancers, including colorectal cancer (CRC), where its expression is significantly associated with histological classification and prognosis. In this study, we investigated the biological and cellular processes affected by miR-378a-3p in the context of CRC carcinogenesis. In agreement with the literature, miR-378a-3p is downregulated in our cohort of CRC patients as well as, in 15 patient-derived colorectal cancer stem-like cell (CRC-SC) lines and 8 CRC cell lines, compared to normal mucosae. Restoration of miR-378a-3p restrains tumorigenic properties of CRC and CRC-SC lines, as well as, significantly reduces tumor growth in two CRC-SC xenograft mouse models. We reported that miR-378a-3p modulates the expression of the lncRNAs MALAT1 and NEAT1. Their expression is inversely correlated with that of miR-378a-3p in patient-derived CRC-SC lines. Silencing of miR-378a-3p targets, MALAT1 and NEAT1, significantly impairs tumorigenic properties of CRC-SCs, supporting the critical role of miR-378a-3p in CRC carcinogenesis as a tumor-suppressor factor by establishing a finely tuned crosstalk with lncRNAs MALAT1 and NEAT1.

3.
Int J Mol Sci ; 21(10)2020 May 20.
Article in English | MEDLINE | ID: mdl-32443824

ABSTRACT

Glioblastoma (GBM) is the most aggressive and prevalent form of a human brain tumor in adults. Several data have demonstrated the implication of microRNAs (miRNAs) in tumorigenicity of GBM stem-like cells (GSCs). The regulatory functions of miRNAs in GSCs have emerged as potential therapeutic candidates for glioma treatment. The current study aimed at investigating the function of miR-370-3p in glioma progression, as aberrant expression of miR-370-3p, is involved in various human cancers, including glioma. Analyzing our collection of GBM samples and patient-derived GSC lines, we found the expression of miR-370-3p significantly downregulated compared to normal brain tissues and normal neural stem cells. Restoration of miR-370-3p expression in GSCs significantly decreased proliferation, migration, and clonogenic abilities of GSCs, in vitro, and tumor growth in vivo. Gene expression analysis performed on miR-370-3p transduced GSCs, identified several transcripts involved in Epithelial to Mesenchymal Transition (EMT), and Hypoxia signaling pathways. Among the genes downregulated by the restored expression of miR-370-3p, we found the EMT-inducer high-mobility group AT-hook 2 (HMGA2), the master transcriptional regulator of the adaptive response to hypoxia, Hypoxia-inducible factor (HIF)1A, and the long non-coding RNAs (lncRNAs) Nuclear Enriched Abundant Transcript (NEAT)1. NEAT1 acts as an oncogene in a series of human cancers including gliomas, where it is regulated by the Epidermal Growth Factor Receptor (EGFR) pathways, and contributes to tumor growth and invasion. Noteworthy, the expression levels of miR-370-3p and NEAT1 were inversely related in both GBM tumor specimens and GSCs, and a dual-luciferase reporter assay proved the direct binding between miR-370-3p and the lncRNAs NEAT1. Our results identify a critical role of miR-370-3p in the regulation of GBM development, indicating that miR-370-3p acts as a tumor-suppressor factor inhibiting glioma cell growth, migration and invasion by targeting the lncRNAs NEAT1, HMGA2, and HIF1A, thus, providing a potential candidate for GBM patient treatment.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MicroRNAs/metabolism , Neural Stem Cells/metabolism , Adult , Animals , Brain Neoplasms/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , HEK293 Cells , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor Cells, Cultured
4.
Neuro Oncol ; 22(12): 1771-1784, 2020 12 18.
Article in English | MEDLINE | ID: mdl-32459347

ABSTRACT

BACKGROUND: Glioblastoma (GBM) stemlike cells (GSCs) are thought to be responsible for the maintenance and aggressiveness of GBM, the most common primary brain tumor in adults. This study aims at elucidating the involvement of deregulations within the imprinted delta-like homolog 1 gene‒type III iodothyronine deiodinase gene (DLK-DIO3) region on chromosome 14q32 in GBM pathogenesis. METHODS: Real-time PCR analyses were performed on GSCs and GBM tissues. Methylation analyses, gene expression, and reverse-phase protein array profiles were used to investigate the tumor suppressor function of the maternally expressed 3 gene (MEG3). RESULTS: Loss of expression of genes and noncoding RNAs within the DLK1-DIO3 region was observed in GSCs and GBM tissues compared with normal brain. This downregulation is mainly mediated by epigenetic silencing. Kaplan-Meier analysis indicated that low expression of MEG3 and MEG8 long noncoding (lnc)RNAs significantly correlated with short survival in GBM patients. MEG3 restoration impairs tumorigenic abilities of GSCs in vitro by inhibiting cell growth, migration, and colony formation and decreases in vivo tumor growth, reducing infiltrative growth. These effects were associated with modulation of genes involved in cell adhesion and epithelial-to-mesenchymal transition (EMT). CONCLUSION: In GBM, MEG3 acts as a tumor suppressor mainly regulating cell adhesion, EMT, and cell proliferation, thus providing a potential candidate for novel GBM therapies.


Subject(s)
Glioblastoma , RNA, Long Noncoding , Adult , Calcium-Binding Proteins , Cell Proliferation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genomic Imprinting , Glioblastoma/genetics , Humans , Membrane Proteins/genetics , RNA, Long Noncoding/genetics
5.
Mol Oncol ; 11(9): 1115-1129, 2017 09.
Article in English | MEDLINE | ID: mdl-28248456

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and malignant primary brain tumor in adults, characterized by aggressive growth, limited response to therapy, and inexorable recurrence. Because of the extremely unfavorable prognosis of GBM, it is important to develop more effective diagnostic and therapeutic strategies based on biologically and clinically relevant patient stratification systems. Analyzing a collection of patient-derived GBM stem-like cells (GSCs) by gene expression profiling, nuclear magnetic resonance spectroscopy, and signal transduction pathway activation, we identified two GSC clusters characterized by different clinical features. Due to the widely documented role played by microRNAs (miRNAs) in the tumorigenesis process, in this study we explored whether these two GBM patient subtypes could also be discriminated by different miRNA signatures. Global miRNA expression pattern was analyzed by oblique principal component analysis and principal component analysis. By a combined inferential strategy on PCA results, we identified a reduced set of three miRNAs - miR-23a, miR-27a, and miR-9* (miR-9-3p) - able to discriminate the proneural- and mesenchymal-like GSC phenotypes as well as mesenchymal and proneural subtypes of primary GBM included in The Cancer Genome Atlas (TCGA) data set. Kaplan-Meier analysis showed a significant correlation between the selected miRNAs and overall survival in 429 GBM specimens from TCGA-identifying patients who had an unfavorable outcome. The survival prognostic capability of the three-miRNA signatures could have important implications for the understanding of the biology of GBM subtypes and could be useful in patient stratification to facilitate interpretation of results from clinical trials.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Profiling , Glioblastoma/genetics , MicroRNAs/genetics , Adult , Aged , Aged, 80 and over , Cluster Analysis , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Humans , Kaplan-Meier Estimate , Male , Mesoderm/pathology , MicroRNAs/metabolism , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Principal Component Analysis , Prognosis , Proton Magnetic Resonance Spectroscopy , Treatment Outcome
6.
Oncotarget ; 6(35): 37241-56, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26437223

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and fatal malignant adult primary brain tumor. Currently, the overall prognosis for GBM patients remains poor despite advances in neurosurgery and adjuvant treatments. MicroRNAs (miRNAs) contribute to the pathogenesis of various types of tumor, including GBM. In this study we analyzed the expression of a panel of miRNAs, which are known to be differentially expressed by the brain and GBM tumor, in a collection of patient-derived GBM stem-like cells (GSCs). Notably, the average expression level of miR-135b, was the most downregulated compared to its normal counterpart, suggesting a potential role as anti-oncogene.Restoration of miR-135b in GSCs significantly decreased proliferation, migration and clonogenic abilities. More importantly, miR-135b restoration was able to significantly reduce brain infiltration in mouse models of GBM obtained by intracerebral injection of GSC lines. We identified ADAM12 and confirmed SMAD5 and GSK3ß as miR-135b targets and potential mediators of its effects. The whole transcriptome analysis ascertained that the expression of miR-135b downmodulated additional genes driving key pathways in GBM survival and infiltration capabilities.Our results identify a critical role of miR-135b in the regulation of GBM development, suggesting that miR-135b might act as a tumor-suppressor factor and thus providing a potential candidate for the treatment of GBM patients.


Subject(s)
Cell Movement , Cell Proliferation , Cell Self Renewal , Glioblastoma/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM12 Protein , Animals , Apoptosis , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genotype , Glioblastoma/genetics , Glioblastoma/pathology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heterografts , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Phenotype , Signal Transduction , Smad5 Protein/genetics , Smad5 Protein/metabolism , Time Factors , Transfection
8.
PLoS One ; 8(4): e60436, 2013.
Article in English | MEDLINE | ID: mdl-23593217

ABSTRACT

MicroRNAs (miRNAs) play key roles in modulating a variety of cellular processes through repression of mRNAs target. The functional relevance of microRNAs has been proven in normal and malignant hematopoiesis. While analyzing miRNAs expression profile in unilineage serum-free liquid suspension unilineage cultures of peripheral blood CD34(+) hematopoietic progenitor cells (HPCs) through the erythroid, megakaryocytic, granulocytic and monocytic pathways, we identified miR-486-3p as mainly expressed within the erythroid lineage. We showed that miR-486-3p regulates BCL11A expression by binding to the extra-long isoform of BCL11A 3'UTR. Overexpression of miR-486-3p in erythroid cells resulted in reduced BCL11A protein levels, associated to increased expression of γ-globin gene, whereas inhibition of physiological miR-486-3p levels increased BCL11A and, consequently, reduced γ-globin expression. Thus, miR-486-3p regulating BCL11A expression might contributes to fetal hemoglobin (HbF) modulation and arise the question as to what extent this miRNA might contribute to different HbF levels observed among ß-thalassemia patients. Erythroid cells, differentiated from PB CD34(+) cells of a small cohort of patients affected by major or intermedia ß-thalassemia, showed miR-486-3p levels significantly higher than those observed in normal counterpart. Importantly, in these patients, miR-486-3p expression correlates with increased HbF synthesis. Thus, our data indicate that miR-486-3p might contribute to different HbF levels observed among thalassemic patients and, possibly, to the clinical severity of the disease.


Subject(s)
Carrier Proteins/genetics , Erythroid Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Nuclear Proteins/genetics , gamma-Globins/genetics , Alternative Splicing , Antigens, CD34/metabolism , Base Pairing , Base Sequence , Cell Line , Cell Lineage/genetics , Cells, Cultured , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Knockout Techniques , Genotype , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Models, Biological , RNA Isoforms , Repressor Proteins , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
9.
BMC Syst Biol ; 4: 85, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20553595

ABSTRACT

BACKGROUND: The differentiation process, proceeding from stem cells towards the different committed cell types, can be considered as a trajectory towards an attractor of a dynamical process. This view, taking into consideration the transcriptome and miRNome dynamics considered as a whole, instead of looking at few 'master genes' driving the system, offers a novel perspective on this phenomenon. We investigated the 'differentiation trajectories' of the hematopoietic system considering a genome-wide scenario. RESULTS: We developed serum-free liquid suspension unilineage cultures of cord blood (CB) CD34+ hematopoietic progenitor cells through erythroid (E), megakaryocytic (MK), granulocytic (G) and monocytic (Mo) pathways. These cultures recapitulate physiological hematopoiesis, allowing the analysis of almost pure unilineage precursors starting from initial differentiation of HPCs until terminal maturation. By analyzing the expression profile of protein coding genes and microRNAs in unilineage CB E, MK, G and Mo cultures, at sequential stages of differentiation and maturation, we observed a coordinated, fully interconnected and scalable character of cell population behaviour in both transcriptome and miRNome spaces reminiscent of an attractor-like dynamics. MiRNome and transcriptome space differed for a still not terminally committed behaviour of microRNAs. CONCLUSIONS: Consistent with their roles, the transcriptome system can be considered as the state space of a cell population, while the continuously evolving miRNA space corresponds to the tuning system necessary to reach the attractor. The behaviour of miRNA machinery could be of great relevance not only for the promise of reversing the differentiated state but even for tumor biology.


Subject(s)
Cell Differentiation/physiology , Genome/genetics , Hematopoietic Stem Cells/physiology , Models, Biological , Signal Transduction/physiology , Antigens, CD34/metabolism , Cell Culture Techniques , Cell Differentiation/genetics , Cell Lineage , Computational Biology/methods , Fetal Blood/cytology , Flow Cytometry , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics
10.
Haematologica ; 95(10): 1633-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20435626

ABSTRACT

BACKGROUND: Ets-1 is a widely expressed transcription factor implicated in several biological processes including hematopoiesis, where it contributes to the regulation of cellular differentiation. The functions of Ets-1 are regulated by transcription factors as well as by phosphorylation events: phosphorylation of threonine 38 activates Ets-1, whereas phosphorylation of a cluster of serines within exon VII reduces DNA binding activity. This study focuses on the role of Ets-1 during granulocytic differentiation of NB4 promyelocytic and HL60 myeloblastic leukemia cell lines induced by all-trans retinoic acid. DESIGN AND METHODS: Ets-1 expression was measured by real-time reverse transcriptase polymerase chain reaction and western blotting. The role of Ets-1 during all-trans retinoic acid-induced differentiation was analyzed by using a transdominant negative molecule or small interfering RNA. RESULTS: NB4 and HL60 cell lines expressed high levels of p51 Ets-1, while the splice variant isoform that lacks exon VII (p42) was almost undetectable. The addition of all-trans retinoic acid reduced p51 Ets-1 levels and induced inhibitory phosphorylation of the remaining protein. Expression of Ets-1 was also reduced during dimethylsulfoxide-induced differentiation and during granulocytic differentiation of human CD34(+) hematopoietic progenitor cells but not in NB4.R2 and HL60R cells resistant to all-trans retinoic acid. In line with these observations, transduction of a transdominant negative molecule of Ets-1, which inhibited DNA binding and transcriptional activity of the wild-type Ets-1, significantly increased chemical-induced differentiation. Consistently, Ets-1 knockdown by small interfering RNA increased the number of mature neutrophils upon addition of all-trans retinoic acid. Interestingly, p51 Ets-1 over-expression was frequently observed in CD34(+) hematopoietic progenitor cells derived from patients with acute myeloid leukemia, as compared to its expression in normal CD34(+) cells. CONCLUSIONS: Our results indicated that a decreased expression of Ets-1 protein generalizes to granulocytic differentiation and may represent a crucial event for granulocytic maturation.


Subject(s)
Cell Differentiation/drug effects , Gene Silencing/drug effects , Granulocytes/drug effects , Proto-Oncogene Protein c-ets-1/genetics , Cell Line, Tumor , Granulocytes/cytology , HL-60 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Leukemia/pathology , Leukemia, Promyelocytic, Acute , Proto-Oncogene Protein c-ets-1/physiology , RNA, Small Interfering/pharmacology
11.
Haematologica ; 94(4): 479-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19278969

ABSTRACT

BACKGROUND: MicroRNAs are small non-coding RNAs that regulate gene expression through mRNA degradation or translational inhibition. MicroRNAs are emerging as key regulators of normal hematopoiesis and hematologic malignancies. Several miRNAs are differentially expressed during hematopoiesis and their specific expression regulates key functional proteins involved in hematopoietic lineage differentiation. This study focused on the functional role of microRNA-223 (miR-223) on erythroid differentiation. DESIGN AND METHODS: Purified cord blood CD34+ hematopoietic progenitor cells were grown in strictly controlled conditions in the presence of saturating dosage of erythropoietin to selectively induce erythroid differentiation. The effects of enforced expression of miR-223 in unilin-eage erythroid cultures were evaluated in liquid phase culture experiments and clonogenic studies. RESULTS: In unilineage erythroid culture of cord blood CD34+ hematopoietic progenitor cells miR-223 is down-regulated, whereas LMO2, an essential protein for erythroid differentiation, is up-regulated. Functional studies showed that enforced expression of miR-223 reduces the mRNA and protein levels of LMO2, by binding to LMO2 3' UTR, and impairs differentiation of erythroid cells. Accordingly, knockdown of LMO2 by short interfering RNA mimics the action of miR-223. Furthermore, hematopoietic progenitor cells transduced with miR-223 showed a significant reduction of their erythroid clonogenic capacity, suggesting that downmodulation of this miRNA is required for erythroid progenitor recruitment and commitment. CONCLUSIONS: These results show that the decline of miR-223 is an important event for erythroid differentiation that leads to the expansion of erythroblast cells at least partially mediated by unblocking LMO2 protein expression.


Subject(s)
DNA-Binding Proteins/genetics , Erythropoiesis , Metalloproteins/genetics , MicroRNAs/physiology , Adaptor Proteins, Signal Transducing , Cell Differentiation , Erythroid Cells , Fetal Blood , Gene Expression Regulation , Humans , LIM Domain Proteins , Proto-Oncogene Proteins
12.
Br J Haematol ; 143(4): 570-80, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18950466

ABSTRACT

MicroRNAs (miRNAs) control basic biological functions and are emerging as key regulators of haematopoiesis. This study focused on the functional role of MIRN155 on megakaryocytic (MK) differentiation of human cord blood CD34+ haematopoietic progenitor cells (HPCs). MIRN155, abundantly expressed in early HPCs, decreases sharply during MK differentiation. Functional studies showed that enforced expression of MIRN155 impairs proliferation and differentiation of MK cells. Furthermore, HPCs transfected with MIRN155 showed a significant reduction of their MK clonogenic capacity, suggesting that down-modulation of this miRNA favours MK progenitor differentiation. Consistent with this observation, MIRN155 downregulates, by directly binding to their 3'-UTR, the expression of Ets-1 and Meis1, two transcription factors with well-known functions in MK cells. These results show that the decline of MIRN155 is required for MK proliferation and differentiation at progenitors and precursors level and indicate that sustained expression of MIRN155 inhibits megakaryopoiesis.


Subject(s)
Homeodomain Proteins/genetics , Megakaryocytes/cytology , MicroRNAs/physiology , Neoplasm Proteins/genetics , Proto-Oncogene Protein c-ets-1/genetics , Thrombopoiesis/genetics , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Down-Regulation , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Humans , K562 Cells , MicroRNAs/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein , Proto-Oncogene Protein c-ets-1/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Thrombopoiesis/physiology , Transfection
13.
Biochem J ; 377(Pt 2): 367-78, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14505489

ABSTRACT

IRFs [IFN (interferon) regulatory factors] constitute a family of transcription factors involved in IFN signalling and in the development and differentiation of the immune system. IRF-2 has generally been described as an antagonist of IRF-1-mediated transcription of IFN and IFN-inducible genes; however, it has been recently identified as a transcriptional activator of some genes, such as those encoding histone H4, VCAM-1 (vascular cell adhesion molecule-1) and Fas ligand. Biologically, IRF-2 plays an important role in cell growth regulation and has been shown to be a potential oncogene. Studies in knock-out mice have also implicated IRF-2 in the differentiation and functionality of haematopoietic cells. Here we show that IRF-2 expression in a myeloid progenitor cell line leads to reprogramming of these cells towards the megakaryocytic lineage and enables them to respond to thrombopoietin, as assessed by cell morphology and expression of specific differentiation markers. Up-regulation of transcription factors involved in the development of the megakaryocytic lineage, such as GATA-1, GATA-2, FOG-1 (friend of GATA-1) and NF-E2 (nuclear factor-erythroid-2), and transcriptional stimulation of the thrombopoietin receptor were also demonstrated. Our results provide evidence for a key role for IRF-2 in the induction of a programme of megakaryocytic differentiation, and reveal a remarkable functional diversity of this transcription factor in the regulation of cellular responses.


Subject(s)
DNA-Binding Proteins/physiology , Megakaryocytes/metabolism , Repressor Proteins , Apoptosis , Cell Differentiation , Cell Division , Cell Line , Cell Lineage , Humans , Interferon Regulatory Factor-2 , Megakaryocytes/cytology , Myeloid Progenitor Cells/physiology , Transcription Factors/metabolism , Transcriptional Activation
14.
Oncogene ; 21(52): 7933-44, 2002 Nov 14.
Article in English | MEDLINE | ID: mdl-12439744

ABSTRACT

High expression of transferrin receptor (TfR) on the membrane of erythroid cells accounts for the high level of iron required to sustain heme synthesis. Several studies indicate that during erythroid differentiation TfR expression is highly dependent on transcriptional regulation. In this study we characterized the minimal region able to confer transcriptional regulation during erythroid differentiation in Friend leukemia cells (FLC). This region of 120 bp, upstream the transcription start site, contains an overlapping consensus recognition sequence for AP1/CREB/ATF transcription factors and for proteins of the Ets family and a GC rich region. Here, we report that both the Ets and the Ap1/CRE like sites are essential for promoter activity during erythroid differentiation. We showed that Ets-1 binds to the EBS-TfR and its binding activity decreases in FLC induced to differentiate and during normal erythroid differentiation. Consistent with this, FLC constitutively expressing Ets-1 show a decrease in TfR gene expression, globin mRNA and hemoglobin synthesis. We conclude that Ets-1 binding activity is modulated during erythroid maturation and that a deregulated expression of this transcription factor interferes with terminal erythroid differentiation.


Subject(s)
Cell Differentiation/physiology , Erythrocytes/chemistry , Gene Expression Regulation/physiology , Proto-Oncogene Proteins/physiology , Receptors, Transferrin/genetics , Transcription Factors/physiology , Transcription, Genetic/physiology , Base Sequence , Cells, Cultured , DNA , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Transcription Factors/metabolism , Tumor Cells, Cultured
15.
Blood ; 100(9): 3203-8, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12384418

ABSTRACT

Postnatal CD34(+) cells expressing vascular endothelial growth factor receptor 2 (KDR) generate hematopoietic or endothelial progeny in different in vitro and in vivo assays. Hypothetically, CD34(+)KDR(+) cells may comprise hemangioblasts bipotent for both lineages. This hypothesis is consistent with 2 series of experiments. In the first series, in clonogenic culture permissive for hematopoietic and endothelial cell growth, CD34(+)KDR(+) cells generate large hemato-endothelial (Hem-End) colonies (5% of seeded cells), whereas CD34(+)KDR(-) cells do not. Limiting-dilution analysis indicates that Hem-End colonies are clonally generated by single hemangioblasts. Sibling cells generated by a hemangioblast, replated in unicellular culture, produce either hematopoietic or Hem-End colonies, depending on the specific culture conditions. Identification of endothelial cells was based on the expression of VE-cadherin and endothelial markers and with lack of CD45 and hematopoietic molecules, as evaluated by immunofluorescence, immunocytochemistry, and reverse transcription-polymerase chain reaction. Furthermore, endothelial cells were functionally identified using low-density lipoprotein (LDL) uptake and tube-formation assays. In the second series, to evaluate the self-renewal capacity of hemangioblasts, single CD34(+)KDR(+) cells were grown in 3-month extended long-term culture (ELTC) through 3 serial culture rounds-that is, blast cells generated in unicellular ELTC were reseeded for a subsequent round of unicellular ELTC. After 9 months, 10% blasts from tertiary ELTC functioned as hemangioblasts and generated macroscopic Hem-End colonies in clonogenic culture. These studies identified postnatal hemangioblasts in a CD34(+)KDR(+) cell subset, endowed with long-term proliferative potential and bilineage differentiation capacity. Although exceedingly rare, hemangioblasts may represent the lifetime source/reservoir for primitive hematopoietic and endothelial progenitors.


Subject(s)
Antigens, CD34/analysis , Cadherins/analysis , Endothelium, Vascular/cytology , Stem Cells/classification , Vascular Endothelial Growth Factor Receptor-2/analysis , Adult , Age Factors , Antigens, CD , Antigens, Differentiation/analysis , Bone Marrow Cells/classification , Cell Culture Techniques/methods , Cell Division , Cell Lineage , Cells, Cultured/cytology , Colony-Forming Units Assay , Culture Media, Serum-Free , Fetal Blood/cytology , Humans , Infant, Newborn , Leukocyte Common Antigens/analysis , Lipoproteins, LDL/metabolism , Stem Cells/chemistry
16.
J Exp Med ; 195(10): 1359-70, 2002 May 20.
Article in English | MEDLINE | ID: mdl-12021315

ABSTRACT

Transcription of the human immunodeficiency virus (HIV)-1 is controlled by the cooperation of virally encoded and host regulatory proteins. The Tat protein is essential for viral replication, however, expression of Tat after virus entry requires HIV-1 promoter activation. A sequence in the 5' HIV-1 LTR, containing a binding site for transcription factors of the interferon regulatory factors (IRF) family has been suggested to be critical for HIV-1 transcription and replication. Here we show that IRF-1 activates HIV-1 LTR transcription in a dose-dependent fashion and in the absence of Tat. This has biological significance since IRF-1 is produced early upon virus entry, both in cell lines and in primary CD4+ T cells, and before expression of Tat. IRF-1 also cooperates with Tat in amplifying virus gene transcription and replication. This cooperation depends upon a physical interaction that is blocked by overexpression of IRF-8, the natural repressor of IRF-1, and, in turn is released by overexpression of IRF-1. These data suggest a key role of IRF-1 in the early phase of viral replication and/or during viral reactivation from latency, when viral transactivators are absent or present at very low levels, and suggest that the interplay between IRF-1 and IRF-8 may play a key role in virus latency.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , HIV-1/growth & development , Phosphoproteins/metabolism , Transcription Factors/metabolism , Virus Replication , Cell Line , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Gene Products, tat/metabolism , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Interferon Regulatory Factors , Jurkat Cells , Phosphoproteins/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcriptional Activation , tat Gene Products, Human Immunodeficiency Virus
17.
Cell Signal ; 14(6): 537-45, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11897494

ABSTRACT

Macrophage cell lines exhibit different responses to IFN-gamma depending on their maturation stage. We investigated the mechanisms underlying the differential IFN-gamma responsiveness in the less mature P388.D1 and in mature RAW264.7 cells. A reduction in the binding activity of the signal transducer and activator of transcription-1 (STAT-1) to different STAT binding elements (SBEs) was observed in P388.D1. This reduced binding activity was not due to an impaired STAT-1 activation. Studies on the expression of a negative regulator of cytokine signalling, protein-inhibiting activated STAT-1 (PIAS-1), showed that this protein was expressed constitutively at high levels in P388.D1. Forced expression of a PIAS-1 homologue, the Gu binding protein (GBP), inhibited the STAT-1-mediated gene activation in RAW264.7 cells, whereas a construct expressing the 5' portion of GBP in the antisense orientation reverts the IFN-gamma-resistant phenotype of P388.D1. Thus, our results indicate that PIAS-1 may account for the differential IFN-gamma responsiveness in macrophage cell lines at different stages of maturation.


Subject(s)
Interferon-gamma/antagonists & inhibitors , Macrophages/immunology , Proteins/physiology , Animals , Cell Differentiation , Cell Line , DNA, Antisense/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Interferon-gamma/pharmacology , Macrophages/drug effects , Mice , Protein Inhibitors of Activated STAT , Proteins/genetics , RNA, Messenger/biosynthesis , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism
18.
Blood Cells Mol Dis ; 29(3): 553-61, 2002.
Article in English | MEDLINE | ID: mdl-12547252

ABSTRACT

Members of the Ets gene family are known to be expressed in the hematopoietic tissue and some of them play a pivotal role in normal hematopoietic cell development. Ets-1 gene expression was analyzed in Friend Leukemia Cells (FLC) induced to erythroid differentiation by DMSO. We show that the level of Ets-1 protein and its binding activity decreases in FLC along erythroid differentiation of primary human progenitors. The same behavior was observed during normal erythroid differentiation. Moreover, FLC constitutively expressing Ets-1 show a decrease in TfR gene expression, globin mRNA and hemoglobin synthesis. These data indicate that a decrease in Ets-1 binding activity is required for a normal erythroid maturation and that a deregulated expression of this transcription factor may interfere with terminal erythroid differentiation.


Subject(s)
Cell Differentiation/physiology , Erythroid Precursor Cells/physiology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Humans , K562 Cells , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets
SELECTION OF CITATIONS
SEARCH DETAIL
...