Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 394
Filtrer
1.
Blood ; 144(8): 809-821, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-38875504

RÉSUMÉ

ABSTRACT: Epidemiological studies report opposing influences of infection on childhood B-cell acute lymphoblastic leukemia (B-ALL). Although infections in the first year of life appear to exert the largest impact on leukemia risk, the effect of early pathogen exposure on the fetal preleukemia cells (PLC) that lead to B-ALL has yet to be reported. Using cytomegalovirus (CMV) infection as a model early-life infection, we show that virus exposure within 1 week of birth induces profound depletion of transplanted E2A-PBX1 and hyperdiploid B-ALL cells in wild-type recipients and in situ-generated PLC in Eµ-ret mice. The age-dependent depletion of PLC results from an elevated STAT4-mediated cytokine response in neonates, with high levels of interleukin (IL)-12p40-driven interferon (IFN)-γ production inducing PLC death. Similar PLC depletion can be achieved in adult mice by impairing viral clearance. These findings provide mechanistic support for potential inhibitory effects of early-life infection on B-ALL progression and could inform novel therapeutic or preventive strategies.


Sujet(s)
Modèles animaux de maladie humaine , Leucémie-lymphome lymphoblastique à précurseurs B , Animaux , Souris , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Infections à cytomégalovirus , Préleucémie/génétique , Préleucémie/anatomopathologie , Souris de lignée C57BL , Animaux nouveau-nés , Diploïdie
2.
Blood ; 143(22): 2245-2255, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38498034

RÉSUMÉ

ABSTRACT: The spectrum of myeloid disorders ranges from aplastic bone marrow failure characterized by an empty bone marrow completely lacking in hematopoiesis to acute myeloid leukemia in which the marrow space is replaced by undifferentiated leukemic blasts. Recent advances in the capacity to sequence bulk tumor population as well as at a single-cell level has provided significant insight into the stepwise process of transformation to acute myeloid leukemia. Using models of progression in the context of germ line predisposition (trisomy 21, GATA2 deficiency, and SAMD9/9L syndrome), premalignant states (clonal hematopoiesis and clonal cytopenia of unknown significance), and myelodysplastic syndrome, we review the mechanisms of progression focusing on the hierarchy of clonal mutation and potential roles of transcription factor alterations, splicing factor mutations, and the bone marrow environment in progression to acute myeloid leukemia. Despite major advances in our understanding, preventing the progression of these disorders or treating them at the acute leukemia phase remains a major area of unmet medical need.


Sujet(s)
Évolution de la maladie , Humains , Préleucémie/anatomopathologie , Préleucémie/génétique , Syndromes myélodysplasiques/anatomopathologie , Syndromes myélodysplasiques/génétique , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/génétique , Animaux , États précancéreux/anatomopathologie , États précancéreux/génétique , Mutation , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Transformation cellulaire néoplasique/métabolisme
3.
Fukushima J Med Sci ; 70(1): 11-24, 2024 Jan 27.
Article de Anglais | MEDLINE | ID: mdl-37952978

RÉSUMÉ

Acute myeloid leukemia (AML) arises from preleukemic conditions. We have investigated the pathogenesis of typical preleukemia, myeloproliferative neoplasms, and clonal hematopoiesis. Hematopoietic stem cells in both preleukemic conditions harbor recurrent driver mutations; additional mutation provokes further malignant transformation, leading to AML onset. Although genetic alterations are defined as the main cause of malignant transformation, non-genetic factors are also involved in disease progression. In this review, we focus on a non-histone chromatin protein, high mobility group AT-hook2 (HMGA2), and a physiological p53 inhibitor, murine double minute X (MDMX). HMGA2 is mainly overexpressed by dysregulation of microRNAs or mutations in polycomb components, and provokes expansion of preleukemic clones through stem cell signature disruption. MDMX is overexpressed by altered splicing balance in myeloid malignancies. MDMX induces leukemic transformation from preleukemia via suppression of p53 and p53-independent activation of WNT/ß-catenin signaling. We also discuss how these non-genetic factors can be targeted for leukemia prevention therapy.


Sujet(s)
Leucémie aigüe myéloïde , Préleucémie , Animaux , Souris , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/anatomopathologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Mutation , Préleucémie/génétique , Préleucémie/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
4.
Sci Adv ; 8(31): eabn4886, 2022 08 05.
Article de Anglais | MEDLINE | ID: mdl-35921412

RÉSUMÉ

Transcriptional variability facilitates stochastic cell diversification and can in turn underpin adaptation to stress or injury. We hypothesize that it may analogously facilitate progression of premalignancy to cancer. To investigate this, we initiated preleukemia in mouse cells with enhanced transcriptional variability due to conditional disruption of the histone lysine acetyltransferase gene Kat2a. By combining single-cell RNA sequencing of preleukemia with functional analysis of transformation, we show that Kat2a loss results in global variegation of cell identity and accumulation of preleukemic cells. Leukemia progression is subsequently facilitated by destabilization of ribosome biogenesis and protein synthesis, which confer a transient transformation advantage. The contribution of transcriptional variability to early cancer evolution reflects a generic role in promoting cell fate transitions, which, in the case of well-adapted malignancies, contrastingly differentiates and depletes cancer stem cells. That is, transcriptional variability confers forward momentum to cell fate systems, with differential multistage impact throughout cancer evolution.


Sujet(s)
Leucémies , Préleucémie , Animaux , Différenciation cellulaire , Leucémies/génétique , Souris , Préleucémie/génétique , Préleucémie/anatomopathologie , Biosynthèse des protéines
5.
Science ; 373(6551)2021 07 09.
Article de Anglais | MEDLINE | ID: mdl-34244384

RÉSUMÉ

Children with Down syndrome have a 150-fold increased risk of developing myeloid leukemia, but the mechanism of predisposition is unclear. Because Down syndrome leukemogenesis initiates during fetal development, we characterized the cellular and developmental context of preleukemic initiation and leukemic progression using gene editing in human disomic and trisomic fetal hematopoietic cells and xenotransplantation. GATA binding protein 1 (GATA1) mutations caused transient preleukemia when introduced into trisomy 21 long-term hematopoietic stem cells, where a subset of chromosome 21 microRNAs affected predisposition to preleukemia. By contrast, progression to leukemia was independent of trisomy 21 and originated in various stem and progenitor cells through additional mutations in cohesin genes. CD117+/KIT proto-oncogene (KIT) cells mediated the propagation of preleukemia and leukemia, and KIT inhibition targeted preleukemic stem cells.


Sujet(s)
Protéines du cycle cellulaire/génétique , Syndrome de Down/génétique , Facteur de transcription GATA-1/génétique , Cellules souches hématopoïétiques/physiologie , Leucémie myéloïde/génétique , Préleucémie/génétique , Animaux , Antigènes CD34/analyse , Protéines du cycle cellulaire/métabolisme , Lignage cellulaire , Prolifération cellulaire , Transformation cellulaire néoplasique , Protéines chromosomiques nonhistones/génétique , Chromosomes humains de la paire 21/génétique , Chromosomes humains de la paire 21/métabolisme , Modèles animaux de maladie humaine , Évolution de la maladie , Syndrome de Down/complications , Femelle , Facteur de transcription GATA-1/métabolisme , Hématopoïèse , Transplantation de cellules souches hématopoïétiques , Hétérogreffes , Humains , Leucémie myéloïde/métabolisme , Leucémie myéloïde/anatomopathologie , Foie/embryologie , Mâle , Mégacaryocytes/physiologie , Souris , microARN/génétique , microARN/métabolisme , Mutation , Préleucémie/métabolisme , Préleucémie/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Proto-oncogène Mas , Protéines proto-oncogènes c-kit/analyse , Protéines proto-oncogènes c-kit/antagonistes et inhibiteurs ,
6.
J Intern Med ; 289(5): 636-649, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33511694

RÉSUMÉ

Haematopoietic stem and progenitor cells (HSPCs) are defined as unspecialized cells that give rise to more differentiated cells. In a similar way, leukaemic stem and progenitor cells (LSPCs) are defined as unspecialized leukaemic cells, which can give rise to more differentiated cells. Leukaemic cells carry leukaemic mutations/variants and have clear differentiation abnormalities. Pre-leukaemic HSPCs (PreL-HSPCs) carry pre-leukaemic mutations/variants (pLMs) and are capable of producing mature functional cells, which will carry the same variants. Under the roof of LSPCs, one can find a broad range of cell types genetic and disease phenotypes. Present-day knowledge suggests that this phenotypic heterogeneity is the result of interactions between the cell of origin, the genetic background and the microenvironment background. The combination of these attributes will define the LSPC phenotype, frequency, differentiation capacity and evolutionary trajectory. Importantly, as LSPCs are leukaemia-initiating cells that sustain clinical remission and are the source of relapse, an improved understanding of LSPCs phenotype would offer better clinical opportunities for the treatment and hopefully prevention of human leukaemia. The current review will focus on LSPCs attributes in the context of human haematologic malignancies.


Sujet(s)
Cellules souches hématopoïétiques/anatomopathologie , Leucémies/anatomopathologie , Préleucémie/anatomopathologie , Marqueurs biologiques tumoraux , Moelle osseuse/anatomopathologie , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Variation génétique , Hématopoïèse , Humains , Leucémies/génétique , Leucémies/métabolisme , Mutation , Phénotype , Préleucémie/génétique , Préleucémie/métabolisme , Microenvironnement tumoral
7.
Blood ; 137(7): 945-958, 2021 02 18.
Article de Anglais | MEDLINE | ID: mdl-33254233

RÉSUMÉ

Isocitrate dehydrogenase (IDH) mutations are common genetic alterations in myeloid disorders, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Epigenetic changes, including abnormal histone and DNA methylation, have been implicated in the pathogenic build-up of hematopoietic progenitors, but it is still unclear whether and how IDH mutations themselves affect hematopoiesis. Here, we show that IDH1-mutant mice develop myeloid dysplasia in that these animals exhibit anemia, ineffective erythropoiesis, and increased immature progenitors and erythroblasts. In erythroid cells of these mice, D-2-hydroxyglutarate, an aberrant metabolite produced by the mutant IDH1 enzyme, inhibits oxoglutarate dehydrogenase activity and diminishes succinyl-coenzyme A (CoA) production. This succinyl-CoA deficiency attenuates heme biosynthesis in IDH1-mutant hematopoietic cells, thus blocking erythroid differentiation at the late erythroblast stage and the erythroid commitment of hematopoietic stem cells, while the exogenous succinyl-CoA or 5-ALA rescues erythropoiesis in IDH1-mutant erythroid cells. Heme deficiency also impairs heme oxygenase-1 expression, which reduces levels of important heme catabolites such as biliverdin and bilirubin. These deficits result in accumulation of excessive reactive oxygen species that induce the cell death of IDH1-mutant erythroid cells. Our results clearly show the essential role of IDH1 in normal erythropoiesis and describe how its mutation leads to myeloid disorders. These data thus have important implications for the devising of new treatments for IDH-mutant tumors.


Sujet(s)
Érythropoïèse/génétique , Cellules souches hématopoïétiques/métabolisme , Hème/biosynthèse , Isocitrate dehydrogenases/génétique , Mutation faux-sens , Mutation ponctuelle , Préleucémie/génétique , Acyl coenzyme A/biosynthèse , Acyl coenzyme A/déficit , Anémie/génétique , Animaux , Moelle osseuse/anatomopathologie , Érythroblastes/métabolisme , Techniques de knock-in de gènes , Glutarates/métabolisme , Hème/déficit , Heme oxygenase-1/métabolisme , Isocitrate dehydrogenases/physiologie , Ketoglutarate dehydrogenase complex/antagonistes et inhibiteurs , Protéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Cellules myéloïdes/anatomopathologie , Myélopoïèse/génétique , Préleucémie/métabolisme , Préleucémie/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Protéines recombinantes/métabolisme , Splénomégalie/étiologie , Thrombopénie/génétique
9.
Ann Ist Super Sanita ; 56(3): 336-350, 2020.
Article de Anglais | MEDLINE | ID: mdl-32959800

RÉSUMÉ

In normal dividing tissues, cell homeostasis is maintained by rare cellular elements, the stem cells, that have the unique property of self-renewal and differentiation to generate a population of functionally mature tissue elements. Recent studies carried out in the last three decades support the existence of stem cells also in tumors, the so-called cancer stem cells. Cancer stem cells have the property of initiating and maintaining tumor growth, are able to self-renew and to differentiate, are the main drivers of intra- and inter-tumoral heterogeneity and the main cellular mediators of drug resistance, leading to tumor recurrence and metastasis. Cancer stem cells can be identified in many tumors according to specific immunophenotypic features, but cancer stemness cannot be defined as a fixed property, due to cancer plasticity. For these properties, cancer stem cells represent attractive targets for developing new anti-cancer therapies and there is supporting evidence that the combination of conventional anticancer therapies with drugs targeting cancer stem cells could lead to cancer eradication. Ongoing studies in some tumors strongly support the clinical utility of developing efficient strategies of cancer stem cell targeting.


Sujet(s)
Thérapie moléculaire ciblée , Cellules souches tumorales/effets des médicaments et des substances chimiques , Carcinogenèse , Tests de cancérogénicité , Essais cliniques comme sujet , Clones cellulaires/anatomopathologie , Humains , Leucémies/traitement médicamenteux , Leucémies/anatomopathologie , Métastase tumorale , Protéines tumorales/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Préleucémie/traitement médicamenteux , Préleucémie/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques
10.
Ann Hematol ; 99(10): 2329-2338, 2020 Oct.
Article de Anglais | MEDLINE | ID: mdl-32821971

RÉSUMÉ

Patients with the pre-leukemia bone marrow failure syndrome called severe congenital neutropenia (CN) have an approximately 15% risk of developing acute myeloid leukemia (AML; called here CN/AML). Most CN/AML patients co-acquire CSF3R and RUNX1 mutations, which play cooperative roles in the development of AML. To establish an in vitro model of leukemogenesis, we utilized bone marrow lin- cells from transgenic C57BL/6-d715 Csf3r mice expressing a CN patient-mimicking truncated CSF3R mutation. We transduced these cells with vectors encoding RUNX1 wild type (WT) or RUNX1 mutant proteins carrying the R139G or R174L mutations. Cells transduced with these RUNX1 mutants showed diminished in vitro myeloid differentiation and elevated replating capacity, compared with those expressing WT RUNX1. mRNA expression analysis showed that cells transduced with the RUNX1 mutants exhibited hyperactivation of inflammatory signaling and innate immunity pathways, including IL-6, TLR, NF-kappaB, IFN, and TREM1 signaling. These data suggest that the expression of mutated RUNX1 in a CSF3R-mutated background may activate the pro-inflammatory cell state and inhibit myeloid differentiation.


Sujet(s)
Insuffisances médullaires congénitales/génétique , Sous-unité alpha 2 du facteur CBF/génétique , Cellules souches hématopoïétiques/anatomopathologie , Cellules myéloïdes/anatomopathologie , Myélopoïèse/génétique , Neutropénie/congénital , Préleucémie/génétique , Récepteurs aux facteurs de croissance hématopoïétique/génétique , Animaux , Division cellulaire , Test clonogénique , Insuffisances médullaires congénitales/anatomopathologie , Sous-unité alpha 2 du facteur CBF/physiologie , Analyse de profil d'expression de gènes , Immunité innée , Inflammation , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Neutropénie/génétique , Neutropénie/anatomopathologie , Préleucémie/anatomopathologie , ARN messager/biosynthèse , ARN messager/génétique , Récepteurs aux facteurs de croissance hématopoïétique/physiologie , Protéines recombinantes/génétique , Organismes exempts d'organismes pathogènes spécifiques
11.
Int J Mol Sci ; 21(9)2020 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-32344823

RÉSUMÉ

Iron overload is related to leukemia transformation in myelodysplastic syndrome (MDS) patients. Siderophores help to transport iron. Type 2-hydroxybutyrate dehydrogenase (BDH2) is a rate-limiting factor in the biogenesis of siderophores. Using qRT-PCR, we analyze BDH2mRNA expression in the bone marrow (BM) of 187 MDS patients, 119 de novo acute myeloid leukemia (AML) patients, and 43 lymphoma patients with normal BM. Elevated BDH2mRNA expression in BM is observed in MDS patients (n = 187 vs. 43, normal BM; P = 0.009), and this is related to ferritin levels. Patients with higher BDH2 expression show a greater risk of leukemia progression (15.25% vs. 3.77%, lower expression; P = 0.017) and shorter leukemia-free-survival (medium LFS, 9 years vs. 7 years; P = 0.024), as do patients with a ferritin level ≥350 ng/mL. Additionally, we investigate the mechanisms related to the prognostic ability of BDH2 by using BDH2-KD THP1. The cell cycle analysis, surface markers, and special stain studies indicate that BDH2-KD induces differentiation and decreases the growth rate of THP1 cells, which is associated with the retardation of the cell cycle. Moreover, many genes, including genes related to mitochondrial catabolism, oncogenes, tumor suppressor genes, and genes related to cell differentiation and proliferation influence BDH2-KD THP1 cells. Herein, we demonstrate that BDH2 is involved in cell cycle arrest and the inhibition of differentiation in malignant cells. Furthermore, the high BDH2 expression in MDS patients could be suggestive of a poor prognostic factor. This study provides a foundation for further research on the roles of BDH2 and iron metabolism in the pathogenesis of MDS.


Sujet(s)
Moelle osseuse/anatomopathologie , Régulation de l'expression des gènes/génétique , Hydroxybutyrate dehydrogenase/physiologie , Leucémie aigüe myéloïde/enzymologie , Syndromes myélodysplasiques/enzymologie , Préleucémie/enzymologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Apoptose/génétique , Moelle osseuse/métabolisme , Points de contrôle du cycle cellulaire/génétique , Différenciation cellulaire/génétique , Femelle , Ferritines/sang , Régulation de l'expression des gènes dans la leucémie , Humains , Hydroxybutyrate dehydrogenase/biosynthèse , Hydroxybutyrate dehydrogenase/génétique , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Lipocaline-2/biosynthèse , Lipocaline-2/génétique , Mâle , Adulte d'âge moyen , Syndromes myélodysplasiques/sang , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/anatomopathologie , Protéines tumorales/biosynthèse , Protéines tumorales/sang , Protéines tumorales/génétique , Protéines tumorales/physiologie , Préleucémie/génétique , Préleucémie/anatomopathologie , Pronostic , Survie sans progression , Interférence par ARN , ARN messager/biosynthèse , ARN tumoral/biosynthèse , Petit ARN interférent/génétique , Cellules THP-1 , Jeune adulte
12.
Science ; 367(6477): 586-590, 2020 01 31.
Article de Anglais | MEDLINE | ID: mdl-32001657

RÉSUMÉ

The initiating mutations that contribute to cancer development are sometimes present in premalignant cells. Whether therapies targeting these mutations can eradicate premalignant cells is unclear. Acute myeloid leukemia (AML) is an attractive system for investigating the effect of preventative treatment because this disease is often preceded by a premalignant state (clonal hematopoiesis or myelodysplastic syndrome). In Npm1c/Dnmt3a mutant knock-in mice, a model of AML development, leukemia is preceded by a period of extended myeloid progenitor cell proliferation and self-renewal. We found that this self-renewal can be reversed by oral administration of a small molecule (VTP-50469) that targets the MLL1-Menin chromatin complex. These preclinical results support the hypothesis that individuals at high risk of developing AML might benefit from targeted epigenetic therapy in a preventative setting.


Sujet(s)
Thérapie génétique/méthodes , Leucémie expérimentale/prévention et contrôle , Leucémie aigüe myéloïde/prévention et contrôle , Protéines nucléaires/génétique , Préleucémie/thérapie , Animaux , Chromatine/métabolisme , DNA (cytosine-5-)-methyltransferase/génétique , DNA methyltransferase 3A , Techniques de knock-in de gènes , Histone-lysine N-methyltransferase/métabolisme , Leucémie expérimentale/génétique , Leucémie aigüe myéloïde/génétique , Souris , Souris de lignée C57BL , Souches mutantes de souris , Mutation , Progéniteurs myéloïdes/anatomopathologie , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Nucléophosmine , Préleucémie/génétique , Préleucémie/anatomopathologie , Protéines proto-oncogènes/métabolisme
13.
Exp Hematol ; 76: 49-59, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31381950

RÉSUMÉ

T(4;11) MLL-AF4 acute leukemia is one of the most aggressive malignancies in infant and pediatric populations. Epidemiological and functional studies have highlighted the influence of an overstimulation of the immune system on leukemia development. This study aimed at assessing if the cell-of-origin of t(4;11) MLL-AF4 acute leukemia is sensitive to a viral or bacterial mimic and if maternal immune activation can lead to a full-blown leukemia. To answer this, we used the Mll-AF4 pre-leukemia mouse model that initiates the expression of Mll-AF4 in the first definitive hematopoietic cells formed during embryonic development. We observed an increase in proliferation upon hematopoietic differentiation of fetal liver Mll-AF4+ Lineage-Sca1+ckit+ (LSK) cells exposed to the immune stimulants, poly(I:C) or LPS/lipopolysaccharide. This was accompanied by increased expression of a subset of MLL-AF4 signature genes and members of the Toll-like receptor signaling pathways in fetal liver Mll-AF4+ LSK exposed to poly(I:C), suggesting that the cell-of-origin responds to inflammatory stimuli. Maternal immune activation using a single dose of poly(I:C) did not lead to the development of leukemia in Mll-AF4+ and control offspring. Instead, aging MLL-AF4+ mice showed an increased proportion of T-lymphoid cells in the spleen, lost their B-lymphoid bias, and had decreased frequencies of hematopoietic stem and multipotent progenitor cells. Overall, this study suggests that the fetal liver Mll-AF4+ LSK cells are sensitive to direct exposure to inflammatory stimuli, especially poly(I:C); however, maternal immune activation induced by a single exposure to poly(I:C) is not sufficient to initiate MLL-AF4 leukemogenesis.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Inflammation/génétique , Protéine de la leucémie myéloïde-lymphoïde/analyse , Protéines de fusion oncogènes/analyse , Poly I-C/pharmacologie , Préleucémie/anatomopathologie , Effets différés de l'exposition prénatale à des facteurs de risque , Adjuvants immunologiques/toxicité , Animaux , Apoptose/effets des médicaments et des substances chimiques , Transformation cellulaire néoplasique/effets des médicaments et des substances chimiques , Endotoxines/pharmacologie , Femelle , Cellules souches hématopoïétiques/immunologie , Inflammation/induit chimiquement , Foie/cytologie , Foie/embryologie , Lymphocytes/cytologie , Lymphocytes/effets des médicaments et des substances chimiques , Souris , Souris transgéniques , Cellules myéloïdes/cytologie , Cellules myéloïdes/effets des médicaments et des substances chimiques , Poly I-C/toxicité , Grossesse , Transcriptome
14.
Genes Chromosomes Cancer ; 58(12): 828-838, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-30939217

RÉSUMÉ

Myeloid neoplasms including myelodysplastic syndromes and acute myeloid leukemia (AML) originate from hematopoietic stem cells through sequential acquisition of genetic and epigenetic alterations that ultimately cause the disease-specific phenotype of impaired differentiation and increased proliferation. It has become clear that preleukemic clonal hematopoiesis (CH), characterized by an expansion of stem and progenitor cells that carry somatic mutations but are still capable of normal differentiation, can precede the development of clinically overt myeloid neoplasia by many years. CH commonly develops in the aging hematopoietic system, yet progression to myelodysplasia or AML is rare. The discovery that myeloid neoplasms frequently develop from premalignant precursor conditions that are detectable in many healthy individuals has important consequences for the diagnosis, and potentially for the treatment of these disorders. In this review, we summarize the current knowledge on CH as a precursor of myeloid cancers and the implications of CH-related gene mutations in the diagnostic workup of patients with suspected myelodysplastic syndrome. We will discuss the risk of progression associated with CH in healthy persons and in patients undergoing chemotherapy for a non-hematologic cancer, and the significance of CH in autologous and allogeneic stem cell transplantation. Finally, we will review the significance of preleukemic clones in AML and their persistence in patients who achieve a remission after chemotherapeutic treatment.


Sujet(s)
Leucémie aigüe myéloïde/génétique , Syndromes myélodysplasiques/génétique , Syndromes myéloprolifératifs/anatomopathologie , Évolution clonale , Clones cellulaires/anatomopathologie , Hématopoïèse/génétique , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques , Humains , Leucémie aigüe myéloïde/anatomopathologie , Syndromes myélodysplasiques/anatomopathologie , Syndromes myéloprolifératifs/génétique , Préleucémie/génétique , Préleucémie/anatomopathologie
15.
Exp Hematol ; 64: 33-44.e5, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-29751067

RÉSUMÉ

Recent advances in next-generation sequencing have identified novel mutations and revealed complex genetic architectures in human hematological malignancies. Moving forward, new methods to quickly generate animal models that recapitulate the complex genetics of human hematological disorders are needed to transform the genetic information to new therapies. Here, we used a ribonucleoprotein-based CRISPR/Cas9 system to model human clonal hematopoiesis of indeterminate potential and acute myeloid leukemia (AML). We edited multiple genes recurrently mutated in hematological disorders, including those encoding epigenetic regulators, transcriptional regulators, and signaling components in murine hematopoietic stem/progenitor cells. Tracking the clonal dynamics by sequencing the indels induced by CRISPR/Cas9 revealed clonal expansion in some recipient mice that progressed to AML initiated by leukemia-initiating cells. Our results establish that the CRISPR/Cas9-mediated multiplex mutagenesis can be used to engineer a variety of murine models of hematological malignancies with complex genetic architectures seen in human disease.


Sujet(s)
Systèmes CRISPR-Cas , Clones cellulaires/anatomopathologie , Édition de gène/méthodes , Cellules souches hématopoïétiques/anatomopathologie , Leucémie aigüe myéloïde/anatomopathologie , Cellules souches tumorales/anatomopathologie , Animaux , Transplantation de moelle osseuse , Protéine-9 associée à CRISPR , Modèles animaux de maladie humaine , Évolution de la maladie , Femelle , Gènes tumoraux , Humains , Mutation de type INDEL , Leucémie aigüe myéloïde/génétique , Mâle , Souris , Souris de lignée C57BL , Mutation , Préleucémie/génétique , Préleucémie/anatomopathologie , Ribonucléoprotéines/génétique , Organismes exempts d'organismes pathogènes spécifiques
16.
Cancer Res ; 78(10): 2669-2679, 2018 05 15.
Article de Anglais | MEDLINE | ID: mdl-29490943

RÉSUMÉ

Preleukemic clones carrying BCR-ABLp190 oncogenic lesions are found in neonatal cord blood, where the majority of preleukemic carriers do not convert into precursor B-cell acute lymphoblastic leukemia (pB-ALL). However, the critical question of how these preleukemic cells transform into pB-ALL remains undefined. Here, we model a BCR-ABLp190 preleukemic state and show that limiting BCR-ABLp190 expression to hematopoietic stem/progenitor cells (HS/PC) in mice (Sca1-BCR-ABLp190) causes pB-ALL at low penetrance, which resembles the human disease. pB-ALL blast cells were BCR-ABL-negative and transcriptionally similar to pro-B/pre-B cells, suggesting disease onset upon reduced Pax5 functionality. Consistent with this, double Sca1-BCR-ABLp190+Pax5+/- mice developed pB-ALL with shorter latencies, 90% incidence, and accumulation of genomic alterations in the remaining wild-type Pax5 allele. Mechanistically, the Pax5-deficient leukemic pro-B cells exhibited a metabolic switch toward increased glucose utilization and energy metabolism. Transcriptome analysis revealed that metabolic genes (IDH1, G6PC3, GAPDH, PGK1, MYC, ENO1, ACO1) were upregulated in Pax5-deficient leukemic cells, and a similar metabolic signature could be observed in human leukemia. Our studies unveil the first in vivo evidence that the combination between Sca1-BCR-ABLp190 and metabolic reprogramming imposed by reduced Pax5 expression is sufficient for pB-ALL development. These findings might help to prevent conversion of BCR-ABLp190 preleukemic cells.Significance: Loss of Pax5 drives metabolic reprogramming, which together with Sca1-restricted BCR-ABL expression enables leukemic transformation. Cancer Res; 78(10); 2669-79. ©2018 AACR.


Sujet(s)
Protéines de fusion bcr-abl/métabolisme , Régulation de l'expression des gènes dans la leucémie/génétique , Protéine activatrice spécifique des lymphocytes B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Animaux , Lymphocytes B/métabolisme , Lignée cellulaire , Métabolisme énergétique/génétique , Protéines de fusion bcr-abl/génétique , Glucose/métabolisme , Cellules HEK293 , Humains , Souris , Souris de lignée C57BL , Souris de lignée CBA , Souris transgéniques , Protéine activatrice spécifique des lymphocytes B/métabolisme , Préleucémie/anatomopathologie
17.
Ann Hematol ; 97(7): 1219-1227, 2018 Jul.
Article de Anglais | MEDLINE | ID: mdl-29492600

RÉSUMÉ

High-count monoclonal B cell lymphocytosis (MBL) with a chronic lymphocytic leukemia (CLL) phenotype is a well-known entity, featuring 1-4% annual risk of progression towards CLL requiring treatment. Lymphoma-like MBL (L-MBL), on the other hand, remains poorly defined and data regarding outcome are lacking. We retrospectively evaluated 33 L-MBL cases within our hospital population and compared them to 95 subjects with CLL-like MBL (C-MBL). Diagnoses of L-MBL were based on asymptomatic B cell clones with Matutes score < 3, B cells < 5.0 × 103/µl, and negative computerized tomography scans. We found that median B cell counts were considerably lower compared to C-MBL (0.6 vs 2.3 × 103/µl) and remained stable over time. Based on immunophenotyping and immunogenetic profiling, most L-MBL clones did not correspond to known lymphoma entities. A strikingly high occurrence of paraproteinemia (48%), hypogammaglobulinemia (45%), and biclonality (21%) was seen; these incidences being significantly higher than in C-MBL (17, 21, and 5%, respectively). Unrelated monoclonal gammopathy of undetermined significance was a frequent feature, as the light chain type of 5/12 paraproteins detected was different from the clonal surface immunoglobulin. After 46-month median follow-up, 2/24 patients (8%) had progressed towards indolent lymphoma requiring no treatment. In contrast, 41% of C-MBL cases evolved to CLL and 17% required treatment. We conclude that clinical L-MBL is characterized by pronounced immune dysregulation and very slow or absent progression, clearly separating it from its CLL-like counterpart.


Sujet(s)
Lymphocytes B/anatomopathologie , Hyperlymphocytose/anatomopathologie , Lymphome B/anatomopathologie , Agammaglobulinémie/anatomopathologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes CD5/analyse , Clones cellulaires/anatomopathologie , Diagnostic différentiel , Évolution de la maladie , Femelle , Études de suivi , Réarrangement des gènes des chaines lourdes des lymphocytes B , Humains , Immunophénotypage , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Hyperlymphocytose/classification , Hyperlymphocytose/diagnostic , Mâle , Adulte d'âge moyen , Gammapathie monoclonale de signification indéterminée/complications , Paraprotéinémies/anatomopathologie , Paraprotéines/analyse , Préleucémie/anatomopathologie , Pronostic , Récepteurs aux IgE/analyse , Études rétrospectives
20.
Leukemia ; 31(2): 423-433, 2017 02.
Article de Anglais | MEDLINE | ID: mdl-27568523

RÉSUMÉ

Fanconi anemia (FA) is an inherited bone marrow failure syndrome with extremely high risk of leukemic transformation. Here we investigate the relationship between DNA damage response (DDR) and leukemogenesis using the Fanca knockout mouse model. We found that chronic exposure of the Fanca-/- hematopoietic stem cells to DNA crosslinking agent mitomycin C in vivo leads to diminished DDR, and the emergence/expansion of pre-leukemia stem cells (pre-LSCs). Surprisingly, although genetic correction of Fanca deficiency in the pre-LSCs restores DDR and reduces genomic instability, but fails to prevent pre-LSC expansion or delay leukemia development in irradiated recipients. Furthermore, we identified transcription program underlying dysregulated DDR and cell migration, myeloid proliferation, and immune response in the Fanca-/- pre-LSCs. Forced expression of the downregulated DNA repair genes, Rad51c or Trp53i13, in the Fanca-/- pre-LSCs partially rescues DDR but has no effect on leukemia, whereas shRNA knockdown of the upregulated immune receptor genes Trem1 or Pilrb improves leukemia-related survival, but not DDR or genomic instability. Furthermore, Trem1 cooperates with diminished DDR in vivo to promote Fanca-/- pre-LSC expansion and leukemia development. Our study implicates diminishing DDR as a root cause of FA leukemogenesis, which subsequently collaborates with other signaling pathways for leukemogenic transformation.


Sujet(s)
Altération de l'ADN , Glycoprotéines membranaires/métabolisme , Cellules souches tumorales/métabolisme , Préleucémie/génétique , Préleucémie/métabolisme , Récepteurs immunologiques/métabolisme , Animaux , Marqueurs biologiques , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Analyse de regroupements , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Anémie de Fanconi/anatomopathologie , Protéine du groupe de complémentation A de l'anémie de Fanconi/génétique , Expression des gènes , Analyse de profil d'expression de gènes , Techniques de knock-down de gènes , Instabilité du génome , Génotype , Immunomodulation/génétique , Mâle , Souris , Souris knockout , Cellules souches tumorales/anatomopathologie , Préleucémie/mortalité , Préleucémie/anatomopathologie , ARN messager/génétique , ARN messager/métabolisme , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE