Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Commun Biol ; 7(1): 753, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902349

ABSTRACT

Arginine methylation is catalyzed by protein arginine methyltransferases (PRMTs) and is involved in various cellular processes, including cancer development. PRMT2 expression is increased in several cancer types although its role in acute myeloid leukemia (AML) remains unknown. Here, we investigate the role of PRMT2 in a cohort of patients with AML, PRMT2 knockout AML cell lines as well as a Prmt2 knockout mouse model. In patients, low PRMT2 expressors are enriched for inflammatory signatures, including the NF-κB pathway, and show inferior survival. In keeping with a role for PRMT2 in control of inflammatory signaling, bone marrow-derived macrophages from Prmt2 KO mice display increased pro-inflammatory cytokine signaling upon LPS treatment. In PRMT2-depleted AML cell lines, aberrant inflammatory signaling has been linked to overproduction of IL6, resulting from a deregulation of the NF-κB signaling pathway, therefore leading to hyperactivation of STAT3. Together, these findings identify PRMT2 as a key regulator of inflammation in AML.


Subject(s)
Inflammation , Leukemia, Myeloid, Acute , Mice, Knockout , NF-kappa B , Protein-Arginine N-Methyltransferases , Signal Transduction , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Inflammation/metabolism , Inflammation/genetics , Intracellular Signaling Peptides and Proteins , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice, Inbred C57BL , NF-kappa B/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
2.
Biomedicines ; 10(12)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36551854

ABSTRACT

The human genome is composed of unique DNA sequences that encode proteins and unique sequence noncoding RNAs that are essential for normal development and cellular differentiation. The human genome also contains over 50% of genome sequences that are repeat in nature (tandem and interspersed repeats) that are now known to contribute dynamically to genetic diversity in populations, to be transcriptionally active under certain physiological conditions, and to be aberrantly active in disease states including cancer, where consequences are pleiotropic with impact on cancer cell phenotypes and on the tumor immune microenvironment. Repeat element-derived RNAs play unique roles in exogenous and endogenous cell signaling under normal and disease conditions. A key component of repeat element-derived transcript-dependent signaling occurs via triggering of innate immune receptor signaling that then feeds forward to inflammatory responses through interferon and NFκB signaling. It has recently been shown that cancer cells display abnormal transcriptional activity of repeat elements and that this is linked to either aggressive disease and treatment failure or to improved prognosis/treatment response, depending on cell context and the amplitude of the so-called 'viral mimicry' response that is engaged. 'Viral mimicry' refers to a cellular state of active antiviral response triggered by endogenous nucleic acids often derived from aberrantly transcribed endogenous retrotransposons and other repeat elements. In this paper, the literature regarding transcriptional activation of repeat elements and engagement of inflammatory signaling in normal (focusing on hematopoiesis) and cancer is reviewed with an emphasis on the role of innate immune receptor signaling, in particular by dsRNA receptors of the RIG-1 like receptor family and interferons/NFκB. How repeat element-derived RNA reprograms cell identity through RNA-guided chromatin state modulation is also discussed.

3.
Cancers (Basel) ; 14(21)2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36358861

ABSTRACT

Arginine methylation is a common post-translational modification affecting protein activity and the transcription of target genes when methylation occurs on histone tails. There are nine protein arginine methyltransferases (PRMTs) in mammals, divided into subgroups depending on the methylation they form on a molecule of arginine. During the formation and maturation of the different types of blood cells, PRMTs play a central role by controlling cell differentiation at the transcriptional level. PRMT enzymatic activity is necessary for many cellular processes in hematological malignancies, such as the activation of cell cycle and proliferation, inhibition of apoptosis, DNA repair processes, RNA splicing, and transcription by methylating histone tails' arginine. Chemical tools have been developed to inhibit the activity of PRMTs and have been tested in several models of hematological malignancies, including primary samples from patients, xenografts into immunodeficient mice, mouse models, and human cell lines. They show a significant effect by reducing cell viability and increasing the overall survival of mice. PRMT5 inhibitors have a strong therapeutic potential, as phase I clinical trials in hematological malignancies that use these molecules show promising results, thus, underlining PRMT inhibitors as useful therapeutic tools for cancer treatment in the future.

4.
Article in English | MEDLINE | ID: mdl-33608382

ABSTRACT

Diagnosis of B-cell chronic lymphocytic leukemia (B-CLL) is usually straightforward, involving clinical, immunophenotypic (Matutes score), and (immuno)genetic analyses (to refine patient prognosis for treatment). CLL cases with atypical presentation (e.g., Matutes ≤ 3) are also encountered, and for these diseases, biology and prognostic impact are less clear. Here we report the genomic characterization of a case of atypical B-CLL in a 70-yr-old male patient; B-CLL cells showed a Matutes score of 3, chromosomal translocation t(14;18)(q32;q21) (BCL2/IGH), mutated IGHV, deletion 17p, and mutations in BCL2, NOTCH1 (subclonal), and TP53 (subclonal). Quite strikingly, a novel PAX5 mutation that was predicted to be loss of function was also seen. Exome sequencing identified, in addition, a potentially actionable BRAF mutation, together with novel somatic mutations affecting the homeobox transcription factor NKX2-3, known to control B-lymphocyte development and homing, and the epigenetic regulator LRIF1, which is implicated in chromatin compaction and gene silencing. Neither NKX2-3 nor LRIF1 mutations, predicted to be loss of function, have previously been reported in B-CLL. Sequencing confirmed the presence of these mutations together with BCL2, NOTCH1, and BRAF mutations, with the t(14;18)(q32;q21) translocation, in the initial diagnostic sample obtained 12 yr prior. This is suggestive of a role for these novel mutations in B-CLL initiation and stable clonal evolution, including upon treatment withdrawal. This case extends the spectrum of atypical B-CLL with t(14;18)(q32;q21) and highlights the value of more global precision genomics for patient follow-up and treatment in these patients.


Subject(s)
Cell Cycle Proteins/metabolism , Epigenesis, Genetic , Homeodomain Proteins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , PAX5 Transcription Factor/genetics , Proto-Oncogene Proteins B-raf/metabolism , Transcription Factors/genetics , Aged , Cell Cycle Proteins/genetics , Clonal Evolution , Homeodomain Proteins/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Male , PAX5 Transcription Factor/metabolism , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, Notch1/genetics , Transcription Factors/metabolism , Translocation, Genetic , Tumor Suppressor Protein p53/genetics , Exome Sequencing
5.
Sci Rep ; 10(1): 16187, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32999332

ABSTRACT

Using a MLL-AF9 knock-in mouse model, we discovered that consumption of a high-fat diet (HFD) accelerates the risk of developing acute myeloid leukemia (AML). This regimen increases the clusterization of FLT3 within lipid rafts on the cell surface of primitive hematopoietic cells, which overactivates this receptor as well as the downstream JAK/STAT signaling known to enhance the transformation of MLL-AF9 knock-in cells. Treatment of mice on a HFD with Quizartinib, a potent inhibitor of FLT3 phosphorylation, inhibits the JAK3/STAT3, signaling and finally antagonizes the accelerated development of AML that occurred following the HFD regimen. We can therefore conclude that, on a mouse model of AML, a HFD enforces the FLT3 signaling pathway on primitive hematopoietic cells and, in turn, improves the oncogenic transformation of MLL-AF9 knock-in cells and the leukemia initiation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Diet, High-Fat , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Signal Transduction/physiology , fms-Like Tyrosine Kinase 3/metabolism , Animals , Benzothiazoles/pharmacology , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/drug effects , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Transgenic , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
6.
Oncogene ; 38(48): 7357-7365, 2019 11.
Article in English | MEDLINE | ID: mdl-31417180

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy characterized by an accumulation of immature T cells. Although patient outcomes have improved, novel targeted therapies are needed to reduce the intensity of chemotherapy and improve the prognosis of high-risk patients. Interleukin-7 (IL-7) modulates the survival and proliferation of normal and malignant T cells. Targeting the IL-7 signaling pathway is thus a potentially effective therapeutic strategy. To achieve such aim, it is essential to first understand how the IL-7 signaling pathway is activated. Although IL-7 production has been observed from multiple stromal tissues, T-ALL autocrine IL-7 secretion has not yet been described. Interestingly, using T-ALL cell lines, primary and patient-derived xenotransplanted (PDX) T-ALL cells, we demonstrate that T-ALL cells produce IL-7 whereas normal T cells do not. Finally, using knock down of IL7 gene in T-ALL cells, we describe to what extent IL-7 autocrine secretion is involved in the T-ALL cells propagation in bone marrow and how it affects the number of leukemia-initiating cells in PDX mice. Together, these results demonstrate how the autocrine production of the IL-7 cytokine mediated by T-ALL cells can be involved in the oncogenic development of T-ALL and offer novel insights into T-ALL spreading.


Subject(s)
Autocrine Communication , Bone Marrow/immunology , Interleukin-7/biosynthesis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , T-Lymphocytes/immunology , Animals , Apoptosis , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Proliferation , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Mol Med (Berl) ; 97(5): 633-645, 2019 05.
Article in English | MEDLINE | ID: mdl-30843084

ABSTRACT

Cohen syndrome (CS) is a rare genetic disorder due to mutations in VPS13B gene. Among various clinical and biological features, CS patients suffer from inconsistent neutropenia, which is associated with recurrent but minor infections. We demonstrate here that this neutropenia results from an exaggerate rate of neutrophil apoptosis. Besides this increased cell death, which occurs in the absence of any endoplasmic reticulum stress or defect in neutrophil elastase (ELANE) expression or localization, all neutrophil functions appeared to be normal. We showed a disorganization of the Golgi apparatus in CS neutrophils precursors, that correlates with an altered glycosylation of ICAM-1 in these cells, as evidenced by a migration shift of the protein. Furthermore, a striking decrease in the expression of SERPINB1 gene, which encodes a critical component of neutrophil survival, was detected in CS neutrophils. These abnormalities may account for the excessive apoptosis of neutrophils leading to neutropenia in CS. KEY MESSAGES: Cohen syndrome patients' neutrophils display normal morphology and functions. Cohen syndrome patients' neutrophils have an increased rate of spontaneous apoptosis compared to healthy donors' neutrophils. No ER stress or defective ELA2 expression or glycosylation was observed in Cohen syndrome patients' neutrophils. SerpinB1 expression is significantly decreased in Cohen syndrome neutrophils as well as in VPS13B-deficient cells.


Subject(s)
Apoptosis , Fingers/abnormalities , Intellectual Disability/genetics , Microcephaly/genetics , Muscle Hypotonia/genetics , Myopia/genetics , Neutropenia/genetics , Neutrophils/pathology , Obesity/genetics , Retinal Degeneration/genetics , Serpins/genetics , Adolescent , Adult , Child , Child, Preschool , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Down-Regulation , Female , Fingers/pathology , Humans , Intellectual Disability/complications , Intellectual Disability/pathology , Male , Microcephaly/complications , Microcephaly/pathology , Middle Aged , Muscle Hypotonia/complications , Muscle Hypotonia/pathology , Mutation , Myopia/complications , Myopia/pathology , Neutropenia/etiology , Neutropenia/pathology , Neutrophils/metabolism , Obesity/complications , Obesity/pathology , Retinal Degeneration/complications , Retinal Degeneration/pathology , Young Adult
8.
Nat Commun ; 10(1): 523, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30705272

ABSTRACT

Despite recent in vivo data demonstrating that high-fat diet (HFD)-induced obesity leads to major perturbations in murine hematopoietic stem cells (HSC), the direct role of a HFD is not yet completely understood. Here, we investigate the direct impact of a short-term HFD on HSC and hematopoiesis in C57BL/6J mice compared with standard diet-fed mice. We detect a loss of half of the most primitive HSC in the bone marrow (BM) cells of HFD-fed mice, which exhibit lower hematopoietic reconstitution potential after transplantation. Impaired maintenance of HSC is due to reduced dormancy after HFD feeding. We discover that a HFD disrupts the TGF-ß receptor within lipid rafts, associated to impaired Smad2/3-dependent TGF-ß signaling, as the main molecular mechanism of action. Finally, injecting HFD-fed mice with recombinant TGF-ß1 avoids the loss of HSC and alteration of the BM's ability to recover, underscoring the fact that a HFD affects TGF-ß signaling on HSC.


Subject(s)
Bone Marrow/drug effects , Bone Marrow/metabolism , Diet, High-Fat/adverse effects , Hematopoietic Stem Cells/metabolism , Membrane Microdomains/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/drug effects , Membrane Microdomains/drug effects , Mice , Mice, Inbred C57BL , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects
10.
Oncotarget ; 7(40): 64785-64797, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27579617

ABSTRACT

CD45 is a pan-leukocyte protein with tyrosine phosphatase activity involved in the regulation of signal transduction in hematopoiesis. Exploiting CD45 KO mice and lentiviral shRNA, we prove the crucial role that CD45 plays in acute myeloid leukemia (AML) development and maintenance. We discovered that CD45 does not colocalize with lipid rafts on murine and human non-transformed hematopoietic cells. Using a mouse model, we proved that CD45 positioning within lipid rafts is modified during their oncogenic transformation to AML. CD45 colocalized with lipid rafts on AML cells, which contributes to elevated GM-CSF signal intensity involved in proliferation of leukemic cells. We furthermore proved that the GM-CSF/Lyn/Stat3 pathway that contributes to growth of leukemic cells could be profoundly affected, by using a new plasma membrane disrupting agent, which rapidly delocalized CD45 away from lipid rafts. We provide evidence that this mechanism is also effective on human primary AML samples and xenograft transplantation. In conclusion, this study highlights the emerging evidence of the involvement of lipid rafts in oncogenic development of AML and the targeting of CD45 positioning among lipid rafts as a new strategy in the treatment of AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukocyte Common Antigens/metabolism , Membrane Microdomains/metabolism , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoiesis/genetics , Humans , Lentivirus/genetics , Leukemia, Myeloid, Acute/pathology , Leukocyte Common Antigens/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
11.
Exp Hematol ; 44(8): 727-739.e6, 2016 08.
Article in English | MEDLINE | ID: mdl-27130375

ABSTRACT

Trim33/Tif1γ (Trim33) is a member of the tripartite motif family. Using a conditional hematopoietic-specific Trim33 knock-out (Trim33(Δ/Δ)) mouse, we showed previously that Trim33 deficiency in hematopoietic stem cells leads to severe defects in hematopoiesis, resembling the main features of human chronic myelomonocytic leukemia. We also demonstrated that Trim33 is involved in hematopoietic aging through TGFß signaling. Nevertheless, how Trim33 contributes to the terminal stages of myeloid differentiation remains to be clarified. We reveal here the crucial role of Trim33 expression in the control of mature granulomonocytic differentiation. An important component of Trim33-deficient mice is the alteration of myeloid differentiation, as characterized by dysplastic features, abnormal granulocyte and monocyte maturation, and the expansion of CD11b(+)Ly6G(high)Ly6C(low) myeloid cells, which share some features with polymorphonuclear-myeloid-derived suppressor cells. Moreover, in Trim33(Δ/Δ) mice, we observed the alteration of CSF-1-mediated macrophage differentiation in association with the lack of Csf-1 receptor. Altogether, these results indicate that Trim33 deficiency leads to the expansion of a subset of myeloid cells characterizing the myelodysplastic/myeloproliferative neoplasm.


Subject(s)
Cell Differentiation/genetics , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Myelopoiesis/genetics , Transcription Factors/genetics , Animals , Apoptosis/genetics , Biomarkers , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Lineage , Cell Movement/genetics , Disease Models, Animal , Immunophenotyping , Mice , Mice, Knockout , Myeloid Cells , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Phenotype
14.
Proc Natl Acad Sci U S A ; 111(29): 10592-7, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002492

ABSTRACT

The hematopoietic system declines with age. Myeloid-biased differentiation and increased incidence of myeloid malignancies feature aging of hematopoietic stem cells (HSCs), but the mechanisms involved remain uncertain. Here, we report that 4-mo-old mice deleted for transcription intermediary factor 1γ (Tif1γ) in HSCs developed an accelerated aging phenotype. To reinforce this result, we also show that Tif1γ is down-regulated in HSCs during aging in 20-mo-old wild-type mice. We established that Tif1γ controls TGF-ß1 receptor (Tgfbr1) turnover. Compared with young HSCs, Tif1γ(-/-) and old HSCs are more sensitive to TGF-ß signaling. Importantly, we identified two populations of HSCs specifically discriminated by Tgfbr1 expression level and provided evidence of the capture of myeloid-biased (Tgfbr1(hi)) and myeloid-lymphoid-balanced (Tgfbr1(lo)) HSCs. In conclusion, our data provide a new paradigm for Tif1γ in regulating the balance between lymphoid- and myeloid-derived HSCs through TGF-ß signaling, leading to HSC aging.


Subject(s)
Cellular Senescence , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transcription Factors/metabolism , Aging/metabolism , Animals , Antigens, CD/metabolism , Cell Separation , Cellular Senescence/drug effects , Gene Expression Regulation/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Mice , Myeloid Cells/metabolism , Phenotype , Polyubiquitin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Signaling Lymphocytic Activation Molecule Family Member 1 , Transcription Factors/deficiency , Transcription Factors/genetics , Transforming Growth Factor beta1/pharmacology , Ubiquitination/drug effects
15.
Hum Mol Genet ; 23(9): 2391-9, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24334764

ABSTRACT

Cohen syndrome (CS) is a rare autosomal recessive disorder with multisytemic clinical features due to mutations in the VPS13B gene, which has recently been described encoding a mandatory membrane protein involved in Golgi integrity. As the Golgi complex is the place where glycosylation of newly synthesized proteins occurs, we hypothesized that VPS13B deficiency, responsible of Golgi apparatus disturbance, could lead to glycosylation defects and/or mysfunction of this organelle, and thus be a cause of the main clinical manifestations of CS. The glycosylation status of CS serum proteins showed a very unusual pattern of glycosylation characterized by a significant accumulation of agalactosylated fucosylated structures as well as asialylated fucosylated structures demonstrating a major defect of glycan maturation in CS. However, CS transferrin and α1-AT profiles, two liver-derived proteins, were normal. We also showed that intercellular cell adhesion molecule 1 and LAMP-2, two highly glycosylated cellular proteins, presented an altered migration profile on SDS-PAGE in peripheral blood mononuclear cells from CS patients. RNA interference against VPS13B confirmed these glycosylation defects. Experiments with Brefeldin A demonstrated that intracellular retrograde cell trafficking was normal in CS fibroblasts. Furthermore, early endosomes were almost absent in these cells and lysosomes were abnormally enlarged, suggesting a crucial role of VPS13B in endosomal-lysosomal trafficking. Our work provides evidence that CS is associated to a tissue-specific major defect of glycosylation and endosomal-lysosomal trafficking defect, suggesting that this could be a new key element to decipher the mechanisms of CS physiopathology.


Subject(s)
Fingers/abnormalities , Intellectual Disability/metabolism , Microcephaly/metabolism , Muscle Hypotonia/metabolism , Myopia/metabolism , Obesity/metabolism , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Developmental Disabilities/metabolism , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Glycosylation , Golgi Apparatus/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , RNA Interference , Retinal Degeneration , Transferrin/metabolism , Vesicular Transport Proteins/metabolism
16.
Biochim Biophys Acta ; 1833(12): 3054-3063, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994619

ABSTRACT

MOZ and MLL encoding a histone acetyltransferase and a histone methyltransferase, respectively, are targets for recurrent chromosomal translocations found in acute myeloblastic or lymphoblastic leukemia. We have previously shown that MOZ and MLL cooperate to activate HOXA9 gene expression in hematopoietic stem/progenitors cells. To dissect the mechanism of action of this complex, we decided to identify new proteins interacting with MOZ. We found that the scaffold protein Symplekin that supports the assembly of polyadenylation machinery was identified by mass spectrometry. Symplekin interacts and co-localizes with both MOZ and MLL in immature hematopoietic cells. Its inhibition leads to a decrease of the HOXA9 protein level but not of Hoxa9 mRNA and to an over-recruitment of MOZ and MLL onto the HOXA9 promoter. Altogether, our results highlight the role of Symplekin in transcription repression involving a regulatory network between MOZ, MLL and Symplekin.


Subject(s)
Hematopoietic System/cytology , Histone Acetyltransferases/metabolism , Homeodomain Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Nuclear Proteins/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Cell Line , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/genetics , Humans , Polyadenylation , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Biochim Biophys Acta ; 1833(8): 1936-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23602969

ABSTRACT

The differentiation of human peripheral blood monocytes into macrophages can be reproduced ex vivo by culturing the cells in the presence of colony-stimulating factor 1 (CSF1). Using microarray profiling to explore the role of microRNAs (miRNAs), we identified a dramatic decrease in the expression of the hematopoietic specific miR-142-3p. Up- and down-regulation of this miRNA in primary human monocytes altered CSF1-induced differentiation of monocytes, as demonstrated by changes in the expression of the cell surface markers CD16 and CD163. One of the genes whose expression is repressed by miR-142-3p encodes the transcription factor Early Growth Response 2 (Egr2). In turn, Egr2 associated with its co-repressor NGFI-A (Nerve Growth Factor-Induced gene-A) binding protein 2 (NAB2) binds to the pre-miR-142-3p promoter to negatively regulate its expression. Interestingly, the expression of miR-142-3p is abnormally low in monocytes from patients with the most proliferative forms of chronic myelomonocytic leukemia (CMML), and miR-142-3p re-expression in CMML dysplastic monocytes can improve their differentiation potential. Altogether, miR-142-3p which functions in a molecular circuitry with Egr2 is an actor of CSF1-induced differentiation of human monocytes whose expression could be altered in CMML.


Subject(s)
Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/physiology , MicroRNAs/genetics , Monocytes/drug effects , Monocytes/physiology , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , K562 Cells , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/metabolism , Leukemia, Myelomonocytic, Chronic/pathology , Macrophages/cytology , Macrophages/metabolism , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Monocytes/cytology , Monocytes/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Up-Regulation/drug effects
19.
Cell Mol Life Sci ; 69(17): 2853-61, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22415325

ABSTRACT

The relatively homogenous clinical features and poor prognosis of chronic myelomonocytic leukemia (CMML) are associated with a molecular heterogeneity, with various mutations impacting several convergent pathways. Due to the restricted understanding of the mechanism involved in leukemogenesis, CMML still appears as a diagnostic and therapeutic undertaking, and poor prognosis of leukemia. Contrary to chronic myelogenous leukemia, BCR-ABL1-positive, cytogenetic, and molecular abnormalities of CMML are not specific and not pathognomonic, confirming the different levels of heterogeneity of this disease. Various mutations can be associated with a common phenotype not distinct at the clinical level, further demonstrating that molecular probings are needed for choosing individual targeted therapies.


Subject(s)
Biomarkers, Tumor/genetics , Genetic Variation/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Humans , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...