Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 79
Filtrer
1.
PLoS One ; 19(1): e0295641, 2024.
Article de Anglais | MEDLINE | ID: mdl-38215076

RÉSUMÉ

Brain metastasis leads to increased mortality and is a major site of relapse for several cancers, yet the molecular mechanisms of brain metastasis are not well understood. In this study, we established and characterized a new leukemic cell line, FIA10, that metastasizes into the central nervous system (CNS) following injection into the tail vein of syngeneic mice. Mice injected with FIA10 cells developed neurological symptoms such as loss of balance, tremor, ataxic gait and seizures, leading to death within 3 months. Histopathology coupled with PCR analysis clearly showed infiltration of leukemic FIA10 cells into the brain parenchyma of diseased mice, with little involvement of bone marrow, peripheral blood and other organs. To define pathways that contribute to CNS metastasis, global transcriptome and proteome analysis was performed on FIA10 cells and compared with that of the parental stem cell line FDCP-Mix and the related FIA18 cells, which give rise to myeloid leukemia without CNS involvement. 188 expressed genes (RNA level) and 189 proteins were upregulated (log2 ratio FIA10/FIA18 ≥ 1) and 120 mRNAs and 177 proteins were downregulated (log2 ratio FIA10/FIA18 ≤ 1) in FIA10 cells compared with FIA18 cells. Major upregulated pathways in FIA10 cells revealed by biofunctional analyses involved immune response components, adhesion molecules and enzymes implicated in extracellular matrix remodeling, opening and crossing the blood-brain barrier (BBB), molecules supporting migration within the brain parenchyma, alterations in metabolism necessary for growth within the brain microenvironment, and regulators for these functions. Downregulated RNA and protein included several tumor suppressors and DNA repair enzymes. In line with the function of FIA10 cells to specifically infiltrate the brain, FIA10 cells have acquired a phenotype that permits crossing the BBB and adapting to the brain microenvironment thereby escaping immune surveillance. These data and our model system FIA10 will be valuable resources to study the occurrence of brain metastases and may help in the development of potential therapies against brain invasion.


Sujet(s)
Tumeurs du cerveau , Tumeurs du système nerveux central , Souris , Animaux , Transcriptome , Protéomique , Encéphale/métabolisme , Barrière hémato-encéphalique/métabolisme , Tumeurs du système nerveux central/anatomopathologie , Tumeurs du cerveau/anatomopathologie , Analyse de profil d'expression de gènes , ARN/métabolisme , Lignée cellulaire , Microenvironnement tumoral
2.
Oncogene ; 40(44): 6258-6272, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34556815

RÉSUMÉ

Neuroblastoma (NB) has a low frequency of recurrent mutations compared to other cancers, which hinders the development of targeted therapies and novel risk stratification strategies. Multikinase inhibitors have shown potential in treating high-risk NB, but their efficacy is likely impaired by the cancer cells' ability to adapt to these drugs through the employment of alternative signaling pathways. Based on the expression of 48 growth factor-related genes in 1189 NB tumors, we have developed a model for NB patient survival prediction. This model discriminates between stage 4 NB tumors with favorable outcomes (>80% overall survival) and very poor outcomes (<10%) independently from MYCN-amplification status. Using signaling pathway analysis and gene set enrichment methods in 60 NB patients with known therapy response, we identified signaling pathways, including EPO, NGF, and HGF, upregulated in patients with no or partial response. In a therapeutic setting, we showed that among six selected growth factors, EPO, and NGF showed the most pronounced protective effects in vitro against several promising anti-NB multikinase inhibitors: imatinib, dasatinib, crizotinib, cabozantinib, and axitinib. Mechanistically kinase inhibitors potentiated NB cells to stronger ERK activation by EPO and NGF. The protective action of these growth factors strongly correlated with ERK activation and was ERK-dependent. ERK inhibitors combined with anticancer drugs, especially with dasatinib, showed a synergistic effect on NB cell death. Consideration of growth factor signaling activity benefits NB outcome prediction and tailoring therapy regimens to treat NB.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Érythropoïétine/génétique , Facteur de croissance nerveuse/génétique , Neuroblastome/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mutation , Stadification tumorale , Neuroblastome/traitement médicamenteux , Neuroblastome/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Analyse de survie
3.
Viruses ; 12(5)2020 05 25.
Article de Anglais | MEDLINE | ID: mdl-32466195

RÉSUMÉ

HIV-1 infection is a complex, multi-step process involving not only viral, but also multiple cellular factors. To date, drug discovery methods have primarily focused on the inhibition of single viral proteins. We present an efficient and unbiased approach, compatible with biosafety level 1 (BSL-1) conditions, to identify inhibitors of HIV-1 reverse transcription, intracellular trafficking, nuclear entry and genome integration. Starting with a fluorescent assay setup, we systematically improved the screening methodology in terms of stability, efficiency and pharmacological relevance. Stability and throughput were optimized by switching to a luciferase-based readout. BSL-1 compliance was achieved without sacrificing pharmacological relevance by using lentiviral particles pseudo-typed with the mouse ecotropic envelope protein to transduce human PM1 T cells gene-modified to express the corresponding murine receptor. The cellular assay was used to screen 26,048 compounds selected for maximum diversity from a 200,640-compound in-house library. This yielded z' values greater than 0.8 with a hit rate of 3.3% and a confirmation rate of 50%. We selected 93 hits and enriched the collection with 279 similar compounds from the in-house library to identify promising structural features. The most active compounds were validated using orthogonal assay formats. The similarity of the compound profiles across the different platforms demonstrated that the reported lentiviral assay system is a robust and versatile tool for the identification of novel HIV-1 inhibitors.


Sujet(s)
Évaluation préclinique de médicament/méthodes , Vecteurs génétiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Tests de criblage à haut débit/méthodes , Lentivirus/génétique , Animaux , Agents antiVIH/pharmacologie , Lignée cellulaire , Confinement de risques biologiques , Développement de médicament , Découverte de médicament , Cellules HEK293 , Humains , Souris , Protéines de l'enveloppe virale , Virion
4.
Leukemia ; 33(1): 99-109, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-29907810

RÉSUMÉ

Primary myelofibrosis (PMF) is a hematopoietic stem cell (HSC) disease, characterized by aberrant differentiation of all myeloid lineages and profound disruption of the bone marrow niche. PMF samples carry several mutations, but their cell origin and hierarchy in regulating the different waves of clonal and aberrant myeloproliferation from the prime HSC compartment is poorly understood. Genotyping of >2000 colonies from CD133+HSC and progenitors from PMF patients confirmed the complex genetic heterogeneity within the neoplastic population. Notably, mutations in chromatin regulators ASXL1 and/or EZH2 were identified as the first genetic lesions, preceding both JAK2-V617F and CALR mutations, and are thus drivers of clonal myelopoiesis in a PMF subset. HSC from PMF patients with double ASXL1/EZH2 mutations exhibited significantly higher engraftment in immunodeficient mice than those from patients without histone modifier mutations. EZH2 mutations correlate with aberrant erythropoiesis in PMF patients, exemplified by impaired maturation and cell cycle arrest of erythroid progenitors. These data underscore the importance of post-transcriptional modifiers of histones in neoplastic stem cells, whose clonal growth sustains aberrant myelopoiesis and expansion of pre-leukemic clones in PMF.


Sujet(s)
Transformation cellulaire néoplasique/anatomopathologie , Évolution clonale , Protéine-2 homologue de l'activateur de Zeste/génétique , Érythropoïèse , Cellules souches hématopoïétiques/anatomopathologie , Mutation , Myélofibrose primitive/anatomopathologie , Protéines de répression/génétique , Animaux , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Cellules souches hématopoïétiques/métabolisme , Humains , Souris , Souris de lignée NOD , Souris SCID , Myélofibrose primitive/génétique , Myélofibrose primitive/métabolisme , Cellules cancéreuses en culture
5.
Arch Toxicol ; 92(6): 2119-2135, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29589053

RÉSUMÉ

A remaining expression of the transcription factor Wilms tumor 1 (WT1) after cytotoxic chemotherapy indicates remaining leukemic clones in patients. We determined the regulation and relevance of WT1 in leukemic cells exposed to replicative stress and DNA damage. To induce these conditions, we used the clinically relevant chemotherapeutics hydroxyurea and doxorubicin. We additionally treated cells with the pro-apoptotic kinase inhibitor staurosporine. Our data show that these agents promote apoptosis to a variable extent in a panel of 12 leukemic cell lines and that caspases cleave WT1 during apoptosis. A chemical inhibition of caspases as well as an overexpression of mitochondrial, anti-apoptotic BCL2 family proteins significantly reduces the processing of WT1 and cell death in hydroxyurea-sensitive acute promyelocytic leukemia cells. Although the reduction of WT1 correlates with the pharmacological efficiency of chemotherapeutics in various leukemic cells, the elimination of WT1 by different strategies of RNA interference (RNAi) does not lead to changes in the cell cycle of chronic myeloid leukemia K562 cells. RNAi against WT1 does also not increase the extent of apoptosis and the accumulation of γH2AX in K562 cells exposed to hydroxyurea. Likewise, a targeted genetic depletion of WT1 in primary oviduct cells does not increase the levels of γH2AX. Our findings position WT1 as a downstream target of the apoptotic process that occurs in response to cytotoxic forms of replicative stress and DNA damage.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Altération de l'ADN , Doxorubicine/pharmacologie , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques , Hydroxy-urée/pharmacologie , Protéines WT1/métabolisme , Animaux , Apoptose/génétique , Caspases/métabolisme , Cycle cellulaire/effets des médicaments et des substances chimiques , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Réplication de l'ADN/effets des médicaments et des substances chimiques , Trompes utérines/effets des médicaments et des substances chimiques , Femelle , Humains , Cellules K562 , Souris knockout , Culture de cellules primaires , Protéines WT1/génétique
6.
Oncotarget ; 8(34): 56991-57002, 2017 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-28915648

RÉSUMÉ

One of the most common chromosomal translocations in acute myeloid leukemia is t(8;21)(q22;q22), which results in the appearance of abnormal transcripts encoding for the fusion protein RUNX1-ETO. Therefore, this oncoprotein is considered to be a pertinent and promising target for treating t(8;21) leukemia. Previously, we have shown that downregulation of RUNX1-ETO leads to activation of intracellular signaling pathways enhancing cell survival and determined that the protein ERK2 can mediate activation of most of these pathways. Here we used a combination of oridonin (natural tetracycline diterpenoid), which has been shown to exhibit anti-RUNX1-ETO activity, and ERK2 kinase inhibitors. We found that treatment of leukemic t(8;21)-positive Kasumi-1 cells with oridonin cause decrease of phosphorylated ERK1/2. Treatment of these cells with ERK2 inhibitors makes them more sensitive to RUNX1-ETO inhibition with oridonin. Therefore we postulate that simultaneous inhibition of RUNX1-ETO and ERK2 cause synergistic effect on survival of leukemic cells.

7.
Biol Blood Marrow Transplant ; 23(7): 1095-1101, 2017 Jul.
Article de Anglais | MEDLINE | ID: mdl-28389256

RÉSUMÉ

Molecular genetics may influence outcome for patients with myelofibrosis. To determine the impact of molecular genetics on outcome after allogeneic stem cell transplantation, we screened 169 patients with primary myelofibrosis (n = 110), post-essential thrombocythemia/polycythemia vera myelofibrosis (n = 46), and myelofibrosis in transformation (n = 13) for mutations in 16 frequently mutated genes. The most frequent mutation was JAK2V617F (n = 101), followed by ASXL1 (n = 49), calreticulin (n = 34), SRSF2 (n = 16), TET2 (n = 10), U2AF1 (n = 11), EZH2 (n = 7), MPL (n = 6), IDH2 (n = 5), IDH1 (n = 4), and CBL (n = 1). The cumulative incidence of nonrelapse mortality (NRM) at 1 year was 21% and of relapse at 5 years 25%. The 5-year rates progression-free (PFS) and overall survival (OS) were and 56%, respectively. In a multivariate analysis CALR mutation was an independent factor for lower NRM (HR, .415; P = .05), improved PFS (HR, .393; P = .01), and OS (HR, .448; P = .03). ASXL1 and IDH2 mutations were independent risk factors for lower PFS (HR, 1.53 [P = .008], and HR, 5.451 [P = .002], respectively), whereas no impact was observed for "triple negative" patients. Molecular genetics, especially CALR, IDH2, and ASXL1 mutations, may thus be useful to predict outcome independently from known clinical risk factors after allogeneic stem cell transplantation for myelofibrosis.


Sujet(s)
Transplantation de cellules souches hématopoïétiques/méthodes , Biologie moléculaire/méthodes , Myélofibrose primitive/génétique , Conditionnement pour greffe/méthodes , Transplantation homologue/méthodes , Adolescent , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte
8.
J Exp Med ; 214(3): 737-752, 2017 03 06.
Article de Anglais | MEDLINE | ID: mdl-28213513

RÉSUMÉ

Acute myeloid leukemia (AML) is induced by the cooperative action of deregulated genes that perturb self-renewal, proliferation, and differentiation. Internal tandem duplications (ITDs) in the FLT3 receptor tyrosine kinase are common mutations in AML, confer poor prognosis, and stimulate myeloproliferation. AML patient samples with FLT3-ITD express high levels of RUNX1, a transcription factor with known tumor-suppressor function. In this study, to understand this paradox, we investigated the impact of RUNX1 and FLT3-ITD coexpression. FLT3-ITD directly impacts on RUNX1 activity, whereby up-regulated and phosphorylated RUNX1 cooperates with FLT3-ITD to induce AML. Inactivating RUNX1 in tumors releases the differentiation block and down-regulates genes controlling ribosome biogenesis. We identified Hhex as a direct target of RUNX1 and FLT3-ITD stimulation and confirmed high HHEX expression in FLT3-ITD AMLs. HHEX could replace RUNX1 in cooperating with FLT3-ITD to induce AML. These results establish and elucidate the unanticipated oncogenic function of RUNX1 in AML. We predict that blocking RUNX1 activity will greatly enhance current therapeutic approaches using FLT3 inhibitors.


Sujet(s)
Sous-unité alpha 2 du facteur CBF/physiologie , Leucémie aigüe myéloïde/étiologie , Tyrosine kinase-3 de type fms/physiologie , Animaux , Cellules cultivées , Sous-unité alpha 2 du facteur CBF/antagonistes et inhibiteurs , Protéines à homéodomaine/génétique , Humains , Souris , Maturation post-traductionnelle des protéines , Facteurs de transcription/génétique
9.
Blood ; 127(26): 3369-81, 2016 06 30.
Article de Anglais | MEDLINE | ID: mdl-27076172

RÉSUMÉ

Disrupting mutations of the RUNX1 gene are found in 10% of patients with myelodysplasia (MDS) and 30% of patients with acute myeloid leukemia (AML). Previous studies have revealed an increase in hematopoietic stem cells (HSCs) and multipotent progenitor (MPP) cells in conditional Runx1-knockout (KO) mice, but the molecular mechanism is unresolved. We investigated the myeloid progenitor (MP) compartment in KO mice, arguing that disruptions at the HSC/MPP level may be amplified in downstream cells. We demonstrate that the MP compartment is increased by more than fivefold in Runx1 KO mice, with a prominent skewing toward megakaryocyte (Meg) progenitors. Runx1-deficient granulocyte-macrophage progenitors are characterized by increased cloning capacity, impaired development into mature cells, and HSC and Meg transcription signatures. An HSC/MPP subpopulation expressing Meg markers was also increased in Runx1-deficient mice. Rescue experiments coupled with transcriptome analysis and Runx1 DNA-binding assays demonstrated that granulocytic/monocytic (G/M) commitment is marked by Runx1 suppression of genes encoding adherence and motility proteins (Tek, Jam3, Plxnc1, Pcdh7, and Selp) that support HSC-Meg interactions with the BM niche. In vitro assays confirmed that enforced Tek expression in HSCs/MPPs increases Meg output. Interestingly, besides this key repressor function of Runx1 to control lineage decisions and cell numbers in progenitors, our study also revealed a critical activating function in erythroblast differentiation, in addition to its known importance in Meg and G/M maturation. Thus both repressor and activator functions of Runx1 at multiple hematopoietic stages and lineages likely contribute to the tumor suppressor activity in MDS and AML.


Sujet(s)
Sous-unité alpha 2 du facteur CBF/métabolisme , Régulation de l'expression des gènes , Cellules souches hématopoïétiques/métabolisme , Mégacaryocytes/métabolisme , Transcription génétique , Protéines suppresseurs de tumeurs/métabolisme , Animaux , Sous-unité alpha 2 du facteur CBF/génétique , Cellules souches hématopoïétiques/anatomopathologie , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Mégacaryocytes/anatomopathologie , Souris , Souris knockout , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/métabolisme , Syndromes myélodysplasiques/anatomopathologie , Protéines suppresseurs de tumeurs/génétique
10.
Blood ; 127(5): 572-81, 2016 Feb 04.
Article de Anglais | MEDLINE | ID: mdl-26660426

RÉSUMÉ

The sequential activation of distinct developmental gene networks governs the ultimate identity of a cell, but the mechanisms involved in initiating downstream programs are incompletely understood. The pre-B-cell receptor (pre-BCR) is an important checkpoint of B-cell development and is essential for a pre-B cell to traverse into an immature B cell. Here, we show that activation of myocyte enhancer factor 2 (Mef2) transcription factors (TFs) by the pre-BCR is necessary for initiating the subsequent genetic network. We demonstrate that B-cell development is blocked at the pre-B-cell stage in mice deficient for Mef2c and Mef2d TFs and that pre-BCR signaling enhances the transcriptional activity of Mef2c/d through phosphorylation by the Erk5 mitogen-activating kinase. This activation is instrumental in inducing Krüppel-like factor 2 and several immediate early genes of the AP1 and Egr family. Finally, we show that Mef2 proteins cooperate with the products of their target genes (Irf4 and Egr2) to induce secondary waves of transcriptional regulation. Our findings uncover a novel role for Mef2c/d in coordinating the transcriptional network that promotes early B-cell development.


Sujet(s)
Lymphocytes B/métabolisme , Précurseurs lymphoïdes B/métabolisme , Animaux , Lymphocytes B/cytologie , Lignée cellulaire , Régulation de l'expression des gènes , Techniques de knock-out de gènes , Réseaux de régulation génique , Facteurs de transcription MEF2/génétique , Facteurs de transcription MEF2/métabolisme , Souris , Souris knockout , Mitogen-Activated Protein Kinase 7/métabolisme , Phosphorylation , Précurseurs lymphoïdes B/cytologie , Transduction du signal , Activation de la transcription
11.
Blood ; 125(23): 3570-9, 2015 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-25911237

RÉSUMÉ

The activity of antagonizing transcription factors represents a mechanistic paradigm of bidirectional lineage-fate control during hematopoiesis. At the megakaryocytic/erythroid bifurcation, the cross-antagonism of krueppel-like factor 1 (KLF1) and friend leukemia integration 1 (FLI1) has such a decisive role. However, how this antagonism is resolved during lineage specification is poorly understood. We found that runt-related transcription factor 1 (RUNX1) inhibits erythroid differentiation of murine megakaryocytic/erythroid progenitors and primary human CD34(+) progenitor cells. We show that RUNX1 represses the erythroid gene expression program during megakaryocytic differentiation by epigenetic repression of the erythroid master regulator KLF1. RUNX1 binding to the KLF1 locus is increased during megakaryocytic differentiation and counterbalances the activating role of T-cell acute lymphocytic leukemia 1 (TAL1). We found that corepressor recruitment by RUNX1 contributes to a block of the KLF1-dependent erythroid gene expression program. Our data indicate that the repressive function of RUNX1 influences the balance between erythroid and megakaryocytic differentiation by shifting the balance between KLF1 and FLI1 in the direction of FLI1. Taken together, we show that RUNX1 is a key player within a network of transcription factors that represses the erythroid gene expression program.


Sujet(s)
Différenciation cellulaire/physiologie , Sous-unité alpha 2 du facteur CBF/métabolisme , Régulation de l'expression des gènes/physiologie , Mégacaryocytes/métabolisme , Thrombopoïèse/physiologie , Antigènes CD34/génétique , Antigènes CD34/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Sous-unité alpha 2 du facteur CBF/génétique , Précurseurs érythroïdes/cytologie , Précurseurs érythroïdes/métabolisme , Érythropoïèse/physiologie , Humains , Cellules K562 , Facteurs de transcription Krüppel-like/génétique , Facteurs de transcription Krüppel-like/métabolisme , Progéniteurs mégacaryocytaires/cytologie , Progéniteurs mégacaryocytaires/métabolisme , Mégacaryocytes/cytologie , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Protéine-1 de la lleucémie lymphoïde aiguë à cellules T
12.
Haematologica ; 100(6): 768-79, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25724578

RÉSUMÉ

Primary myelofibrosis is a myeloproliferative neoplasm characterized by bone marrow fibrosis, megakaryocyte atypia, extramedullary hematopoiesis, and transformation to acute myeloid leukemia. To date the stem cell that undergoes the spatial and temporal chain of events during the development of this disease has not been identified. Here we describe a CD133(+) stem cell population that drives the pathogenesis of primary myelofibrosis. Patient-derived circulating CD133(+) but not CD34(+)CD133(-) cells, with a variable burden for JAK2 (V617F) mutation, had multipotent cloning capacity in vitro. CD133(+) cells engrafted for up to 10 months in immunocompromised mice and differentiated into JAK2-V617F(+) myeloid but not lymphoid progenitors. We observed the persistence of human, atypical JAK2-V617F(+) megakaryocytes, the initiation of a prefibrotic state, bone marrow/splenic fibrosis and transition to acute myeloid leukemia. Leukemic cells arose from a subset of CD133(+) cells harboring EZH2 (D265H) but lacking a secondary JAK2 (V617F) mutation, consistent with the hypothesis that deregulation of EZH2 activity drives clonal growth and increases the risk of acute myeloid leukemia. This is the first characterization of a patient-derived stem cell population that drives disease resembling both chronic and acute phases of primary myelofibrosis in mice. These results reveal the importance of the CD133 antigen in deciphering the neoplastic clone in primary myelofibrosis and indicate a new therapeutic target for myeloproliferative neoplasms.


Sujet(s)
Antigènes CD/sang , Marqueurs biologiques tumoraux/sang , Glycoprotéines/sang , Cellules souches hématopoïétiques/métabolisme , Peptides/sang , Myélofibrose primitive/sang , Myélofibrose primitive/diagnostic , Antigène AC133 , Adulte , Sujet âgé , Animaux , Femelle , Humains , Agranulocytes/métabolisme , Mâle , Souris , Souris de lignée NOD , Souris SCID , Souris transgéniques , Adulte d'âge moyen
13.
J Virol ; 89(8): 4700-4, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25673711

RÉSUMÉ

The development of treatments for Ebola virus disease (EVD) has been hampered by the lack of small-animal models that mimick human disease. Here we show that mice with transplanted human hematopoetic stem cells reproduce features typical of EVD. Infection with Ebola virus was associated with viremia, cell damage, liver steatosis, signs of hemorrhage, and high lethality. Our study provides a small-animal model with human components for the development of EVD therapies.


Sujet(s)
Modèles animaux de maladie humaine , Ebolavirus/immunologie , Transplantation de cellules souches hématopoïétiques/méthodes , Fièvre hémorragique à virus Ebola/immunologie , Fièvre hémorragique à virus Ebola/transmission , Hétérogreffes/immunologie , Souris de lignée NOD , Animaux , Stéatose hépatique/anatomopathologie , Hémorragie/anatomopathologie , Fièvre hémorragique à virus Ebola/anatomopathologie , Humains , Estimation de Kaplan-Meier , Souris , Microscopie de fluorescence , Virémie/anatomopathologie
14.
Proc Natl Acad Sci U S A ; 111(23): 8595-600, 2014 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-24912157

RÉSUMÉ

The compound immunodeficiencies in nonobese diabetic (NOD) inbred mice homozygous for the Prkdc(scid) and Il2rg(null) alleles (NSG mice) permit engraftment of a wide-range of primary human cells, enabling sophisticated modeling of human disease. In studies designed to define neoplastic stem cells of primary myelofibrosis (PMF), a myeloproliferative neoplasm characterized by profound disruption of the hematopoietic microenvironment, we observed a high frequency of acute myeloid leukemia (AML) in NSG mice. AML was of mouse origin, confined to PMF-xenografted mice, and contained multiple clonal integrations of ecotropic murine leukemia virus (E-MuLV). Significantly, MuLV replication was not only observed in diseased mice, but also in nontreated NSG controls. Furthermore, in addition to the single ecotropic endogenous retrovirus (eERV) located on chromosome 11 (Emv30) in the NOD genome, multiple de novo germ-line eERV integrations were observed in mice from each of four independent NSG mouse colonies. Analysis confirmed that E-MuLV originated from the Emv30 provirus and that recombination events were not necessary for virus replication or AML induction. Pathogenicity is thus likely attributable to PMF-mediated paracrine stimulation of mouse myeloid cells, which serve as targets for retroviral infection and transformation, as evidenced by integration into the Evi1 locus, a hotspot for retroviral-induced myeloid leukemia. This study thus corroborates a role of paracrine stimulation in PMF disease progression, underlines the importance of target cell type and numbers in MuLV-induced disease, and mandates awareness of replicating MuLV in NOD immunodeficient mice, which can significantly influence experimental results and their interpretation.


Sujet(s)
Rétrovirus endogènes/génétique , Leucémie expérimentale/génétique , Leucémie aigüe myéloïde/génétique , Myélofibrose primitive/génétique , Sujet âgé , Animaux , Technique de Southern , Femelle , Humains , Sous-unité gamma commune aux récepteurs des interleukines/génétique , Sous-unité gamma commune aux récepteurs des interleukines/métabolisme , Virus de la leucémie murine/génétique , Leucémie expérimentale/anatomopathologie , Leucémie expérimentale/virologie , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/virologie , Mâle , Souris , Souris de lignée C57BL , Souris de lignée NOD , Souris knockout , Souris SCID , Souris transgéniques , Adulte d'âge moyen , Données de séquences moléculaires , Myélofibrose primitive/anatomopathologie , Myélofibrose primitive/virologie , Provirus/génétique , Transplantation hétérologue , Intégration virale/génétique , Réplication virale/génétique , Jeune adulte
15.
Blood ; 123(14): 2229-37, 2014 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-24523240

RÉSUMÉ

Severe congenital neutropenia (CN) is a preleukemic bone marrow failure syndrome with a 20% risk of evolving into leukemia or myelodysplastic syndrome (MDS). Patterns of acquisition of leukemia-associated mutations were investigated using next-generation deep-sequencing in 31 CN patients who developed leukemia or MDS. Twenty (64.5%) of the 31 patients had mutations in RUNX1. A majority of patients with RUNX1 mutations (80.5%) also had acquired CSF3R mutations. In contrast to their high frequency in CN patients who developed leukemia or MDS, RUNX1 mutations were found in only 9 of 307 (2.9%) patients with de novo pediatric acute myeloid leukemia. A sequential analysis at stages prior to overt leukemia revealed RUNX1 mutations to be late events in leukemic transformation. Single-cell analyses in 2 patients showed that RUNX1 and CSF3R mutations were present in the same malignant clone. Functional studies demonstrated elevated granulocyte colony-stimulating factor (G-CSF)-induced proliferation with diminished myeloid differentiation of hematopoietic CD34(+) cells coexpressing mutated forms of RUNX1 and CSF3R. The high frequency of cooperating RUNX1 and CSF3R mutations in CN patients suggests a novel molecular pathway of leukemogenesis: mutations in the hematopoietic cytokine receptor (G-CSFR) in combination with the second mutations in the downstream hematopoietic transcription fator (RUNX1). The detection of both RUNX1 and CSF3R mutations could be used as a marker for identifying CN patients with a high risk of progressing to leukemia or MDS.


Sujet(s)
Transformation cellulaire néoplasique/génétique , Sous-unité alpha 2 du facteur CBF/génétique , Leucémie myéloïde/génétique , Mutation , Neutropénie/congénital , Récepteurs aux facteurs de croissance hématopoïétique/génétique , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Insuffisances médullaires congénitales , Analyse cytogénétique , Femelle , Humains , Mâle , Neutropénie/génétique , Neutropénie/anatomopathologie , Transduction du signal/génétique , Jeune adulte
16.
J Exp Med ; 210(11): 2289-304, 2013 Oct 21.
Article de Anglais | MEDLINE | ID: mdl-24081948

RÉSUMÉ

Differentiation arrest is a hallmark of acute leukemia. Genomic alterations in B cell differentiation factors such as PAX5, IKZF1, and EBF-1 have been identified in more than half of all cases of childhood B precursor acute lymphoblastic leukemia (ALL). Here, we describe a perturbed epigenetic and transcriptional regulation of ZNF423 in ALL as a novel mechanism interfering with B cell differentiation. Hypomethylation of ZNF423 regulatory sequences and BMP2 signaling result in transactivation of ZNF423α and a novel ZNF423ß-isoform encoding a nucleosome remodeling and histone deacetylase complex-interacting domain. Aberrant ZNF423 inhibits the transactivation of EBF-1 target genes and leads to B cell maturation arrest in vivo. Importantly, ZNF423 expression is associated with poor outcome of ETV6-RUNX1-negative B precursor ALL patients. Our work demonstrates that ALL is more than a genetic disease and that epigenetics may uncover novel mechanisms of disease with prognostic implications.


Sujet(s)
Lymphocytes B/anatomopathologie , Différenciation cellulaire , Sous-unité alpha 2 du facteur CBF/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de fusion oncogènes/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Animaux , Lymphocytes B/métabolisme , Protéine morphogénétique osseuse de type 2/métabolisme , Différenciation cellulaire/génétique , Lignage cellulaire/génétique , Méthylation de l'ADN/génétique , Protéines de liaison à l'ADN/composition chimique , Protéines de liaison à l'ADN/génétique , Survie sans rechute , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes dans la leucémie , Cellules souches hématopoïétiques/métabolisme , Complexe Mi-2/NuRD/métabolisme , Souris , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Pronostic , Liaison aux protéines/génétique , Isoformes de protéines/métabolisme , Structure tertiaire des protéines , Protéines , Transduction du signal/génétique , Protéines Smad/métabolisme , Transactivateurs/génétique , Activation de la transcription/génétique , Régulation positive/génétique
17.
Blood ; 122(3): 413-23, 2013 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-23704093

RÉSUMÉ

The t(12;21) chromosomal translocation, targeting the gene encoding the RUNX1 transcription factor, is observed in 25% of pediatric acute lymphoblastic leukemia (ALL) and is an initiating event in the disease. To elucidate the mechanism by which RUNX1 disruption initiates leukemogenesis, we investigated its normal role in murine B-cell development. This study revealed 2 critical functions of Runx1: (1) to promote survival and development of progenitors specified to the B-cell lineage, a function that can be substituted by ectopic Bcl2 expression, and (2) to enable the developmental transition through the pre-B stage triggered by the pre-B-cell antigen receptor (pre-BCR). Gene expression analysis and genomewide Runx1 occupancy studies support the hypothesis that Runx1 reinforces the transcription factor network governing early B-cell survival and development and specifically regulates genes encoding members of the Lyn kinase subfamily (key integrators of interleukin-7 and pre-BCR signaling) and the stage-specific transcription factors SpiB and Aiolos (critical downstream effectors of pre-BCR signaling). Interrogation of expression databases of 257 ALL samples demonstrated the specific down-regulation of the SPIB and IKZF3 genes (the latter encoding AIOLOS) in t(12;21) ALL, providing novel insight into the mechanism by which the translocation blocks B-cell development and promotes leukemia.


Sujet(s)
Lymphocytes B/cytologie , Lymphocytes B/immunologie , Sous-unité alpha 2 du facteur CBF/métabolisme , Animaux , Apoptose/génétique , Sites de fixation , Différenciation cellulaire/immunologie , Lignage cellulaire/génétique , Lignage cellulaire/immunologie , Prolifération cellulaire , Survie cellulaire/génétique , Survie cellulaire/immunologie , Chromosomes humains de la paire 12/génétique , Chromosomes humains de la paire 21/génétique , Sous-unité alpha 2 du facteur CBF/déficit , Éléments activateurs (génétique)/génétique , Délétion de gène , Régulation de l'expression des gènes au cours du développement , Régulation de l'expression des gènes dans la leucémie , Ciblage de gène , Génome/génétique , Humains , Facteur de transcription Ikaros , Souris , Souris de lignée C57BL , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Liaison aux protéines/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Transactivateurs/génétique , Transactivateurs/métabolisme , Translocation génétique
18.
Cell Rep ; 3(5): 1617-28, 2013 May 30.
Article de Anglais | MEDLINE | ID: mdl-23623495

RÉSUMÉ

Dendritic cells (DCs) are essential regulators of immune responses; however, transcriptional mechanisms that establish DC lineage commitment are poorly defined. Here, we report that the PU.1 transcription factor induces specific remodeling of the higher-order chromatin structure at the interferon regulatory factor 8 (Irf8) gene to initiate DC fate choice. An Irf8 reporter mouse enabled us to pinpoint an initial progenitor stage at which DCs separate from other myeloid lineages in the bone marrow. In the absence of Irf8, this progenitor undergoes DC-to-neutrophil reprogramming, indicating that DC commitment requires an active, Irf8-dependent escape from alternative myeloid lineage potential. Mechanistically, myeloid Irf8 expression depends on high PU.1 levels, resulting in local chromosomal looping and activation of a lineage- and developmental-stage-specific cis-enhancer. These data delineate PU.1 as a concentration-dependent rheostat of myeloid lineage selection by controlling long-distance contacts between regulatory elements and suggest that specific higher-order chromatin remodeling at the Irf8 gene determines DC differentiation.


Sujet(s)
Assemblage et désassemblage de la chromatine , Chromatine/métabolisme , Cellules dendritiques/cytologie , Facteurs de régulation d'interféron/génétique , Protéines proto-oncogènes/métabolisme , Transactivateurs/métabolisme , Animaux , Séquence nucléotidique , Cellules de la moelle osseuse/cytologie , Différenciation cellulaire , Lignage cellulaire , Cellules cultivées , Cellules dendritiques/métabolisme , Humains , Facteurs de régulation d'interféron/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Cellules myéloïdes/cytologie , Régions promotrices (génétique) , Protéines proto-oncogènes/composition chimique , Transactivateurs/composition chimique
19.
AIDS Res Ther ; 10(1): 1, 2013 Jan 03.
Article de Anglais | MEDLINE | ID: mdl-23286882

RÉSUMÉ

BACKGROUND: Despite progress in the development of combined antiretroviral therapies (cART), HIV infection remains a significant challenge for human health. Current problems of cART include multi-drug-resistant virus variants, long-term toxicity and enormous treatment costs. Therefore, the identification of novel effective drugs is urgently needed. METHODS: We developed a straightforward screening approach for simultaneously evaluating the sensitivity of multiple HIV gag-pol mutants to antiviral drugs in one assay. Our technique is based on multi-colour lentiviral self-inactivating (SIN) LeGO vector technology. RESULTS: We demonstrated the successful use of this approach for screening compounds against up to four HIV gag-pol variants (wild-type and three mutants) simultaneously. Importantly, the technique was adapted to Biosafety Level 1 conditions by utilising ecotropic pseudotypes. This allowed upscaling to a large-scale screening protocol exploited by pharmaceutical companies in a successful proof-of-concept experiment. CONCLUSIONS: The technology developed here facilitates fast screening for anti-HIV activity of individual agents from large compound libraries. Although drugs targeting gag-pol variants were used here, our approach permits screening compounds that target several different, key cellular and viral functions of the HIV life-cycle. The modular principle of the method also allows the easy exchange of various mutations in HIV sequences. In conclusion, the methodology presented here provides a valuable new approach for the identification of novel anti-HIV drugs.

20.
J Gen Virol ; 94(Pt 2): 453-463, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23136363

RÉSUMÉ

Prion diseases are fatal neurodegenerative disorders. An important step in disease pathophysiology is the conversion of cellular prion protein (PrP(C)) to disease-associated misfolded conformers (PrP(Sc)). These misfolded PrP variants are a common component of prion infectivity and are detectable in diseased brain and lymphoreticular organs such as spleen. In the latter, PrP(Sc) is thought to replicate mainly in follicular dendritic cells within spleen follicles. Although the presence of PrP(Sc) is a hallmark for prion disease and serves as a main diagnostic criterion, in certain instances the amount of PrP(Sc) does not correlate well with neurotoxicity or prion infectivity. Therefore, it has been proposed that prions might be a mixture of different conformers and aggregates with differing properties. This study investigated the impact of disruption of spleen architecture by neoplasia on the abundance of different PrP species in spleens of prion-infected mice. Although follicular integrity was completely disturbed, titres of prion infectivity in neoplastic spleens were not significantly altered, yet no protease-resistant PrP(Sc) was detectable. Instead, unique protease-sensitive prion species could be detected in neoplastic spleens. These results indicate the dissociation of PrP(Sc) and prion infectivity and showed the presence of non-PrP(Sc) PrP species in spleen with divergent biochemical properties that become apparent after tissue architecture disruption.


Sujet(s)
Peptide hydrolases/métabolisme , Maladies à prions/complications , Prions/isolement et purification , Prions/métabolisme , Rate/anatomopathologie , Tumeurs spléniques/complications , Animaux , Modèles animaux de maladie humaine , Souris , Maladies à prions/anatomopathologie , Protéines prion , Prions/classification , Tumeurs spléniques/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...