Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Blood ; 140(26): 2805-2817, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36283106

ABSTRACT

Myelofibrosis (MF) is a disease associated with high unmet medical needs because allogeneic stem cell transplantation is not an option for most patients, and JAK inhibitors are generally effective for only 2 to 3 years and do not delay disease progression. MF is characterized by dysplastic megakaryocytic hyperplasia and progression to fulminant disease, which is associated with progressively increasing marrow fibrosis. Despite evidence that the inflammatory milieu in MF contributes to disease progression, the specific factors that promote megakaryocyte growth are poorly understood. Here, we analyzed changes in the cytokine profiles of MF mouse models before and after the development of fibrosis, coupled with the analysis of bone marrow populations using single-cell RNA sequencing. We found high interleukin 13 (IL-13) levels in the bone marrow of MF mice. IL-13 promoted the growth of mutant megakaryocytes and induced surface expression of transforming growth factor ß and collagen biosynthesis. Similarly, analysis of samples from patients with MF revealed elevated levels of IL-13 in the plasma and increased IL-13 receptor expression in marrow megakaryocytes. In vivo, IL-13 overexpression promoted disease progression, whereas reducing IL-13/IL-4 signaling reduced several features of the disease, including fibrosis. Finally, we observed an increase in the number of marrow T cells and mast cells, which are known sources of IL-13. Together, our data demonstrate that IL-13 is involved in disease progression in MF and that inhibition of the IL-13/IL-4 signaling pathway might serve as a novel therapeutic target to treat MF.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Primary Myelofibrosis , Mice , Animals , Interleukin-13/therapeutic use , Interleukin-4 , Neoplasms/complications , Myeloproliferative Disorders/complications , Primary Myelofibrosis/genetics , Signal Transduction/genetics , Fibrosis , Disease Progression
2.
Am J Blood Res ; 11(1): 84-92, 2021.
Article in English | MEDLINE | ID: mdl-33796394

ABSTRACT

Foetal spleen is described as a transient focus of haematopoiesis between the 3rd and 5th month of gestation: this function is however entirely replaced by the bone marrow before the end of pregnancy. This study identifies haematopoiesis in foetal spleen by exploring changes of echogenicity during its development throughout gestation. Two intervals of pregnancy were studied: Mid-Pregnancy (Mid-P, 19-23 weeks) and End-Pregnancy (End-P, 37-41 weeks). The foetal spleen was investigated in 80 pregnant women (41 vs 39). Due to quality criteria the comparison was made between 60 images (30 Mid-P vs 30 End-P). The acquisition of splenic parenchyma was followed by clustering segmentation. We identified two new parameters resulted from the clustering segmentation: Dark Ratio (DR) and Light Ratio (LR). These are related to splenic echogenicity expressing the percentage of dark and light signal in the clustered image, influenced by blood cellularity. The mean of DR value was different among the 2 groups (0.0631 vs 0.0483, P = 0.014), while LR did not show any significant differences. We conclude that DR may represent a reliable radiomic parameter in the determination of extramedullary haematopoiesis in the spleen.

3.
Blood ; 116(26): 6023-6, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20861460

ABSTRACT

Recently, Dawson et al identified a previously unrecognized nuclear role of JAK2 in the phosphorylation of histone H3 in hematopoietic cell lines. We searched nuclear JAK2 in total bone marrow (BM) cells and in 4 sorted BM cell populations (CD34(+), CD15(+), CD41(+), and CD71(+)) of 10 myeloproliferative neoplasia (MPN) patients with JAK2V617F mutation and 5 patients with wild-type JAK2 MPN. Confocal immunofluorescent images and Western blot analyses of nuclear and cytoplasmic fractions found nuclear JAK2 in CD34(+) cells of 10 of 10 JAK2-mutated patients but not in patients with wild-type JAK2. JAK2 was predominantly in the cytoplasmic fraction of differentiated granulocytic, megakaryocytic, or erythroid cells obtained from all patients. JAK2V617F up-regulates LMO2 in K562 and in JAK2V617F-positive CD34(+) cells. The selective JAK2 inhibitor AG490 normalizes the LMO2 levels in V617F-positive K562 and restores the cyto-plasmic localization of JAK2.


Subject(s)
Antigens, CD34/metabolism , Cell Nucleus/metabolism , Erythroid Cells/metabolism , Granulocytes/metabolism , Janus Kinase 2/genetics , Megakaryocytes/metabolism , Myeloproliferative Disorders/metabolism , Philadelphia Chromosome , Apoptosis , Blotting, Western , Bone Marrow/metabolism , Cells, Cultured , Cytoplasm/metabolism , Granulocytes/cytology , Humans , Janus Kinase 2/metabolism , K562 Cells , Megakaryocytes/cytology , Microscopy, Fluorescence , Mutation/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phosphorylation , Prognosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Cancer Res ; 69(4): 1633-42, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19208846

ABSTRACT

EVI1 is an oncogene inappropriately expressed in the bone marrow (BM) of approximately 10% of myelodysplastic syndrome (MDS) patients. This disease is characterized by severe anemia and multilineage myeloid dysplasia that are thought to be a major cause of mortality in MDS patients. We earlier reported on a mouse model that constitutive expression of EVI1 in the BM led to fatal anemia and myeloid dysplasia, as observed in MDS patients, and we subsequently showed that EVI1 interaction with GATA1 blocks proper erythropoiesis. Whereas this interaction could provide the basis for the erythroid defects in EVI1-positive MDS, it does not explain the alteration of myeloid differentiation. Here, we have examined the expression of several genes activated during terminal myelopoiesis in BM cells and identified a group of them that are altered by EVI1. A common feature of these genes is their regulation by the transcription factor PU.1. We report here that EVI1 interacts with PU.1 and represses the PU.1-dependent activation of a myeloid promoter. EVI1 does not seem to inhibit PU.1 binding to DNA, but rather to block its association with the coactivator c-Jun. After mapping the PU.1-EVI1 interaction sites, we show that an EVI1 point mutant, unable to bind PU.1, restores the activation of PU.1-regulated genes and allows a normal differentiation of BM progenitors in vitro.


Subject(s)
DNA-Binding Proteins/physiology , Myelodysplastic Syndromes/genetics , Myelopoiesis/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/physiology , Trans-Activators/genetics , Transcription Factors/physiology , 3T3 Cells , Anemia/genetics , Animals , Cell Differentiation , Cell Line , Chromatin/genetics , Colony-Forming Units Assay , DNA-Binding Proteins/genetics , Erythropoiesis/genetics , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , MDS1 and EVI1 Complex Locus Protein , Mice , Myelodysplastic Syndromes/pathology , Polymerase Chain Reaction , Proto-Oncogenes/genetics , Transcription Factors/genetics
6.
Ann Hematol ; 88(2): 151-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18709502

ABSTRACT

A large proportion of adult patients with acute myeloid leukemia (AML) relapse after treatment, and some of them are resistant to primary induction chemotherapy. Sixty-one patients from seven hematological centers with poor-risk AML, primary refractory (n = 16), or relapsed (n = 45) were treated with a salvage regimen, including fludarabine (2 days) and cytarabine (3 days) in a sequential continuous infusion, associated with liposomal daunorubicin (3 days) (FLAD). Complete response rate was 44% and 56% for refractory and relapsed patients, respectively, with an overall response rate of 52% (32 of 61). Twenty-two patients (36%) were resistant to the salvage therapy. Seven patients (12%) died early during chemotherapy, four of them because of sepsis. Nineteen patients in complete remission (CR) underwent a stem-cell transplant (SCT) procedure: five autologous, nine from a HL-A identical sibling, and five from HL-A matched unrelated donors. Post-treatment aplasia and mucositis were major toxicities. Twenty patients (62.5%) relapsed after this treatment in a median of 7.3 months; ten patients relapsed after a SCT procedure. Nine patients are alive and disease free; three of them were rescued after a further cytotoxic treatment. The FLAD regimen proved to be an effective and well-tolerated treatment, with acceptable toxicity in this group of high-risk patients. A better response rate was obtained in the subgroup of relapsed patients, compared to patients treated for refractory disease. More then half (five of nine) of long-surviving patients are those who were submitted to a transplant procedure; thus, the main indication for FLAD seems to be to try to induce a rapid CR with minimum toxicity in order to perform a transplant as soon as possible.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , Daunorubicin/administration & dosage , Daunorubicin/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Vidarabine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytarabine/administration & dosage , Cytarabine/adverse effects , Daunorubicin/adverse effects , Disease-Free Survival , Female , Humans , Infusions, Intravenous , Leukemia, Myeloid, Acute/surgery , Liposomes , Male , Middle Aged , Recurrence , Salvage Therapy , Stem Cell Transplantation , Survival Rate , Time Factors , Vidarabine/administration & dosage , Vidarabine/adverse effects , Vidarabine/therapeutic use
8.
Cancer Res ; 67(12): 5658-66, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575132

ABSTRACT

Recurring chromosomal translocations observed in human leukemia often result in the expression of fusion proteins that are DNA-binding transcription factors. These altered proteins acquire new dimerization properties that result in the assembly of inappropriate multimeric transcription complexes that deregulate hematopoietic programs and induce leukemogenesis. Recently, we reported that the fusion protein AML1/MDS1/EVI1 (AME), a product of a t(3;21)(q26;q22) associated with chronic myelogenous leukemia and acute myelogenous leukemia, displays a complex pattern of self-interaction. Here, we show that the 8th zinc finger motif of MDS1/EVI1 is an oligomerization domain involved not only in interaction of AME with itself but also in interactions with the parental proteins, RUNX1 and MDS1/EVI1, from which AME is generated. Because the 8th zinc finger motif is also present in the oncoprotein EVI1, we have evaluated the effects of the interaction between RUNX1 and EVI1 in vitro and in vivo. We found that in vitro, this interaction alters the ability of RUNX1 to bind to DNA and to regulate a reporter gene, whereas in vivo, the expression of the isolated 8th zinc finger motif of EVI1 is sufficient to block the granulocyte colony-stimulating factor-induced differentiation of 32Dcl3 cells, leading to cell death. As EVI1 is not detected in normal bone marrow cells, these data suggest that its inappropriate expression could contribute to hematopoietic transformation in part by a new mechanism that involves EVI1 association with key hematopoietic regulators, leading to their functional impairment.


Subject(s)
Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Leukemia/genetics , Proto-Oncogenes/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cloning, Molecular , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Transcription Factors/metabolism , Transfection , Zinc Fingers/physiology
9.
J Biol Chem ; 282(18): 13542-51, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17355962

ABSTRACT

RUNX1 (AML1, CBFalpha2, PEBP2alphaB) is a transcription factor essential for the establishment of the hematopoietic stem cell. It is generally thought that RUNX1 exists as a monomer that regulates hematopoietic differentiation by interacting with tissue-specific factors and its DNA consensus through its N terminus. RUNX1 is frequently altered in human leukemia by gene fusions or point mutations. In general, these alterations do not affect the N terminus of the protein, and it is unclear how they consistently lead to hematopoietic transformation and leukemia. Here we report that RUNX1 homodimerizes through a mechanism involving C terminus-C terminus interaction. This RUNX1-RUNX1 interaction regulates the activity of the protein in reporter gene assays and modulates its ability to induce hematopoietic differentiation of hematopoietic cell lines. The promoters of genes regulated by RUNX1 often contain multiple RUNX1 binding sites. This arrangement suggests that RUNX1 could homodimerize to bring and hold together distant chromatin sites and factors and that if the dimerization region is removed by gene fusions or is altered by point mutations, as observed in leukemia, the ability of RUNX1 to regulate differentiation could be impaired.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation , Animals , Binding Sites/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Dimerization , Gene Expression Regulation/genetics , HeLa Cells , Hematopoietic Stem Cells/metabolism , Humans , Leukemia/genetics , Leukemia/metabolism , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion , Point Mutation , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Response Elements
10.
Int J Cardiol ; 114(3): 403-4, 2007 Jan 18.
Article in English | MEDLINE | ID: mdl-16626820

ABSTRACT

Our retrospective experience underscores the ability of Doppler echocardiography to detect the cardiotoxicity of chemotherapy (functional and pericardial abnormalities, heart involvement) and points out the need for an accurate echocardiographic follow-up of hematologic patients.


Subject(s)
Antineoplastic Agents/adverse effects , Echocardiography, Doppler , Heart Diseases/chemically induced , Heart Diseases/diagnostic imaging , Hematologic Neoplasms/drug therapy , Female , Humans , Male , Middle Aged , Retrospective Studies
11.
Mol Cell Biol ; 26(20): 7658-66, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16954386

ABSTRACT

EVI1 is an aggressive nuclear oncoprotein deregulated by recurring chromosomal abnormalities in myelodysplastic syndrome (MDS). The expression of the corresponding gene is a very poor prognostic marker for MDS patients and is associated with severe defects of the erythroid lineage. We have recently shown that the constitutive expression of EVI1 in murine bone marrow results in a fatal disease with features characteristic of MDS, including anemia, dyserythropoiesis, and dysmegakaryopoiesis. These lineages are regulated by the DNA-binding transcription factor GATA1. EVI1 has two zinc finger domains containing seven motifs at the N terminus and three motifs at the C terminus. Supported by results of assays utilizing synthetic DNA promoters, it was earlier proposed that erythroid-lineage repression by EVI1 is based on the ability of this protein to compete with GATA1 for DNA-binding sites, resulting in repression of gene activation by GATA1. Here, however, we show that EVI1 is unable to bind to classic GATA1 sites. To understand the mechanism utilized by EVI1 to repress erythropoiesis, we used a combination of biochemical assays, mutation analyses, and in vitro bone marrow differentiation. The results indicate that EVI1 interacts directly with the GATA1 protein rather than the DNA sequence. We further show that this protein-protein interaction blocks efficient recognition or binding to DNA by GATA1. Point mutations that disrupt the geometry of two zinc fingers of EVI1 abolish the protein-protein interaction, leading to normal erythroid differentiation of normal murine bone marrow in vitro.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , DNA-Binding Proteins/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , GATA1 Transcription Factor/genetics , Humans , Immunoprecipitation , Mice , Point Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL