Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.269
Filtrar
Más filtros

Publication year range
1.
Blood ; 144(8): 809-821, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-38875504

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Animales , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Infecciones por Citomegalovirus , Preleucemia/genética , Preleucemia/patología , Ratones Endogámicos C57BL , Animales Recién Nacidos , Diploidia
2.
Blood ; 143(22): 2245-2255, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38498034

RESUMEN

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.


Asunto(s)
Progresión de la Enfermedad , Humanos , Preleucemia/patología , Preleucemia/genética , Síndromes Mielodisplásicos/patología , Síndromes Mielodisplásicos/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/genética , Animales , Lesiones Precancerosas/patología , Lesiones Precancerosas/genética , Mutación , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/metabolismo
3.
Blood ; 137(7): 945-958, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33254233

RESUMEN

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.


Asunto(s)
Eritropoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Hemo/biosíntesis , Isocitrato Deshidrogenasa/genética , Mutación Missense , Mutación Puntual , Preleucemia/genética , Acilcoenzima A/biosíntesis , Acilcoenzima A/deficiencia , Anemia/genética , Animales , Médula Ósea/patología , Eritroblastos/metabolismo , Técnicas de Sustitución del Gen , Glutaratos/metabolismo , Hemo/deficiencia , Hemo-Oxigenasa 1/metabolismo , Isocitrato Deshidrogenasa/fisiología , Complejo Cetoglutarato Deshidrogenasa/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Mielopoyesis/genética , Preleucemia/metabolismo , Preleucemia/patología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Esplenomegalia/etiología , Trombocitopenia/genética
4.
Blood ; 138(21): 2066-2092, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34111240

RESUMEN

t(4;11) MLL-AF4 acute leukemia is one of the most aggressive malignancies in the infant and pediatric population, yet we have little information on the molecular mechanisms responsible for disease progression. This impairs the development of therapeutic regimens that can address the aggressive phenotype and lineage plasticity of MLL-AF4-driven leukemogenesis. This study highlights novel mechanisms of disease development by focusing on 2 microRNAs (miRNAs) upregulated in leukemic blasts from primary patient samples: miR-130b and miR-128a. We show that miR-130b and miR-128a are downstream targets of MLL-AF4 and can individually drive the transition from a pre-leukemic stage to an acute leukemia in an entirely murine Mll-AF4 in vivo model. They are also required to maintain the disease phenotype. Interestingly, miR-130b overexpression led to a mixed/B-cell precursor (BCP)/myeloid leukemia, propagated by the lymphoid-primed multipotent progenitor (LMPP) population, whereas miR-128a overexpression resulted in a pro-B acute lymphoblastic leukemia (ALL), maintained by a highly expanded Il7r+c-Kit+ blast population. Molecular and phenotypic changes induced by these two miRNAs fully recapitulate the human disease, including central nervous system infiltration and activation of an MLL-AF4 expression signature. Furthermore, we identified 2 downstream targets of these miRNAs, NR2F6 and SGMS1, which in extensive validation studies are confirmed as novel tumor suppressors of MLL-AF4+ leukemia. Our integrative approach thus provides a platform for the identification of essential co-drivers of MLL-rearranged leukemias, in which the preleukemia to leukemia transition and lineage plasticity can be dissected and new therapeutic approaches can be tested.


Asunto(s)
Leucemia Mieloide Aguda/genética , MicroARNs/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Femenino , Regulación Leucémica de la Expresión Génica , Humanos , Masculino , Ratones , Preleucemia/genética , Factores de Elongación Transcripcional/genética , Translocación Genética
5.
Int J Mol Sci ; 23(14)2022 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-35886910

RESUMEN

Leukemia is the most usual childhood cancer, and B-cell acute lymphoblastic leukemia (B-ALL) is its most common presentation. It has been proposed that pediatric leukemogenesis occurs through a "multi-step" or "multi-hit" mechanism that includes both in utero and postnatal steps. Many childhood leukemia-initiating events, such as chromosomal translocations, originate in utero, and studies so far suggest that these "first-hits" occur at a far higher frequency than the incidence of childhood leukemia itself. The reason why only a small percentage of the children born with such preleukemic "hits" will develop full-blown leukemia is still a mystery. In order to better understand childhood leukemia, mouse modeling is essential, but only if the multistage process of leukemia can be recapitulated in the model. Therefore, mouse models naturally reproducing the "multi-step" process of childhood B-ALL will be essential to identify environmental or other factors that are directly linked to increased risk of disease.


Asunto(s)
Síndromes Mielodisplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Preleucemia , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Preleucemia/genética , Translocación Genética
6.
J Intern Med ; 289(5): 636-649, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33511694

RESUMEN

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.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia/patología , Preleucemia/patología , Biomarcadores de Tumor , Médula Ósea/patología , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Variación Genética , Hematopoyesis , Humanos , Leucemia/genética , Leucemia/metabolismo , Mutación , Fenotipo , Preleucemia/genética , Preleucemia/metabolismo , Microambiente Tumoral
7.
Ann Hematol ; 99(10): 2329-2338, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32821971

RESUMEN

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.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Hematopoyéticas/patología , Células Mieloides/patología , Mielopoyesis/genética , Neutropenia/congénito , Preleucemia/genética , Receptores del Factor Estimulante de Colonias/genética , Animales , División Celular , Ensayo de Unidades Formadoras de Colonias , Síndromes Congénitos de Insuficiencia de la Médula Ósea/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Perfilación de la Expresión Génica , Inmunidad Innata , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutropenia/genética , Neutropenia/patología , Preleucemia/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores del Factor Estimulante de Colonias/fisiología , Proteínas Recombinantes/genética , Organismos Libres de Patógenos Específicos
8.
Int J Mol Sci ; 21(9)2020 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-32344823

RESUMEN

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.


Asunto(s)
Médula Ósea/patología , Regulación de la Expresión Génica/genética , Hidroxibutirato Deshidrogenasa/fisiología , Leucemia Mieloide Aguda/enzimología , Síndromes Mielodisplásicos/enzimología , Preleucemia/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Médula Ósea/metabolismo , Puntos de Control del Ciclo Celular/genética , Diferenciación Celular/genética , Femenino , Ferritinas/sangre , Regulación Leucémica de la Expresión Génica , Humanos , Hidroxibutirato Deshidrogenasa/biosíntesis , Hidroxibutirato Deshidrogenasa/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Lipocalina 2/biosíntesis , Lipocalina 2/genética , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/sangre , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Preleucemia/genética , Preleucemia/patología , Pronóstico , Supervivencia sin Progresión , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , ARN Interferente Pequeño/genética , Células THP-1 , Adulto Joven
9.
Genes Chromosomes Cancer ; 58(12): 828-838, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30939217

RESUMEN

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.


Asunto(s)
Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/patología , Evolución Clonal , Células Clonales/patología , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Leucemia Mieloide Aguda/patología , Síndromes Mielodisplásicos/patología , Trastornos Mieloproliferativos/genética , Preleucemia/genética , Preleucemia/patología
11.
Haematologica ; 103(12): 1945-1955, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30337364

RESUMEN

The bone marrow microenvironment, also known as the bone marrow niche, is a complex network of cell types and acellular factors that supports normal hematopoiesis. For many years, leukemia was believed to be caused by a series of genetic hits to hematopoietic stem and progenitor cells, which transform them to preleukemic, and eventually to leukemic, cells. Recent discoveries suggest that genetic alterations in bone marrow niche cells, particularly in osteogenic cells, may also cause myeloid leukemia in mouse models. The osteogenic niche, which consists of osteoprogenitors, preosteoblasts, mature osteoblasts, osteocytes and osteoclasts, has been shown to play a critical role in the maintenance and expansion of hematopoietic stem and progenitor cells as well as in their oncogenic transformation into leukemia stem/initiating cells. We have recently shown that acute myeloid leukemia cells induce osteogenic differentiation in mesenchymal stromal cells to gain a growth advantage. In this review, we discuss the role of the osteogenic niche in the maintenance of hematopoietic stem and progenitor cells, as well as in their transformation into leukemia cells. We also discuss the signaling pathways that regulate osteogenic niche-hematopoietic stem and progenitor cells or osteogenic niche-leukemic stem/initiating cell interactions in the bone marrow, together with novel approaches for therapeutically targeting these interactions.


Asunto(s)
Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Leucemia/genética , Osteogénesis/genética , Preleucemia/genética , Nicho de Células Madre , Animales , Transformación Celular Neoplásica/genética , Regulación de la Expresión Génica , Humanos , Transducción de Señal/genética
12.
Ann Hematol ; 97(7): 1219-1227, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29492600

RESUMEN

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.


Asunto(s)
Linfocitos B/patología , Linfocitosis/patología , Linfoma de Células B/patología , Agammaglobulinemia/patología , Anciano , Anciano de 80 o más Años , Antígenos CD5/análisis , Células Clonales/patología , Diagnóstico Diferencial , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Reordenamiento Génico de Cadena Pesada de Linfocito B , Humanos , Inmunofenotipificación , Leucemia Linfocítica Crónica de Células B/patología , Linfocitosis/clasificación , Linfocitosis/diagnóstico , Masculino , Persona de Mediana Edad , Gammopatía Monoclonal de Relevancia Indeterminada/complicaciones , Paraproteinemias/patología , Paraproteínas/análisis , Preleucemia/patología , Pronóstico , Receptores de IgE/análisis , Estudios Retrospectivos
13.
Proc Natl Acad Sci U S A ; 111(7): 2548-53, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550281

RESUMEN

Cancer is widely characterized by the sequential acquisition of genetic lesions in a single lineage of cells. Our previous studies have shown that, in acute myeloid leukemia (AML), mutation acquisition occurs in functionally normal hematopoietic stem cells (HSCs). These preleukemic HSCs harbor some, but not all, of the mutations found in the leukemic cells. We report here the identification of patterns of mutation acquisition in human AML. Our findings support a model in which mutations in "landscaping" genes, involved in global chromatin changes such as DNA methylation, histone modification, and chromatin looping, occur early in the evolution of AML, whereas mutations in "proliferative" genes occur late. Additionally, we analyze the persistence of preleukemic mutations in patients in remission and find CD34+ progenitor cells and various mature cells that harbor preleukemic mutations. These findings indicate that preleukemic HSCs can survive induction chemotherapy, identifying these cells as a reservoir for the reevolution of relapsed disease. Finally, through the study of several cases of relapsed AML, we demonstrate various evolutionary patterns for the generation of relapsed disease and show that some of these patterns are consistent with involvement of preleukemic HSCs. These findings provide key insights into the monitoring of minimal residual disease and the identification of therapeutic targets in human AML.


Asunto(s)
Evolución Clonal/genética , Epigénesis Genética/genética , Células Madre Hematopoyéticas/química , Leucemia Mieloide Aguda/genética , Modelos Biológicos , Preleucemia/genética , Antígenos CD34/metabolismo , Secuencia de Bases , Exoma/genética , Citometría de Flujo , Genotipo , Humanos , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia de ADN
14.
Blood ; 124(3): 426-36, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24894773

RESUMEN

Acute myeloid leukemia (AML) results from the activity of driver mutations that deregulate proliferation and survival of hematopoietic stem cells (HSCs). The fusion protein CBFß-SMMHC impairs differentiation in hematopoietic stem and progenitor cells and induces AML in cooperation with other mutations. However, the combined function of CBFß-SMMHC and cooperating mutations in preleukemic expansion is not known. Here, we used Nras(LSL-G12D); Cbfb(56M) knock-in mice to show that allelic expression of oncogenic Nras(G12D) and Cbfß-SMMHC increases survival of preleukemic short-term HSCs and myeloid progenitor cells and maintains the differentiation block induced by the fusion protein. Nras(G12D) and Cbfß-SMMHC synergize to induce leukemia in mice in a cell-autonomous manner, with a shorter median latency and higher leukemia-initiating cell activity than that of mice expressing Cbfß-SMMHC. Furthermore, Nras(LSL-G12D); Cbfb(56M) leukemic cells were sensitive to pharmacologic inhibition of the MEK/ERK signaling pathway, increasing apoptosis and Bim protein levels. These studies demonstrate that Cbfß-SMMHC and Nras(G12D) promote the survival of preleukemic myeloid progenitors primed for leukemia by activation of the MEK/ERK/Bim axis, and define Nras(LSL-G12D); Cbfb(56M) mice as a valuable genetic model for the study of inversion(16) AML-targeted therapies.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Oncogénicas/metabolismo , Preleucemia/metabolismo , Preleucemia/patología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Supervivencia Celular , Técnicas de Sustitución del Gen , Leucemia Experimental/etiología , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/genética , Mutación Missense , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Oncogénicas/genética , Proteínas de Fusión Oncogénica/genética , Preleucemia/genética , Proteínas Proto-Oncogénicas/metabolismo
15.
Curr Opin Hematol ; 22(2): 77-84, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25575035

RESUMEN

PURPOSE OF REVIEW: In the present review, we will define the preleukemic state. We aim at increasing awareness and research in the field of preleukemia that will nurture targeted therapy for the earlier steps of leukemia evolution. RECENT FINDINGS: Emerging evidence supports the role of hematopoietic stem/progenitor cells carrying recurrent leukemia-related mutations as the cell of origin of both myeloid and lymphoid malignancies. The preleukemic stem cells can maintain at least to some extent their functionality; however, they have increased fitness endowed by the preleukemic mutations that lead to clonal expansion. SUMMARY: The latent preleukemic period before overt leukemia presents can take years, and the majority of carriers will never develop leukemia in their lifetime. The preleukemic state is not rare, with greater than 1% of individuals having acquired one or more of the recognized preleukemic lesions. The high frequency of such abnormalities in the population may be the cost of growing old; however, another view could be that in order to survive to old age, the hematopoietic system must adapt to create robust hematopoietic stem/progenitor cells with an increased fitness and clonal expansion. Hence, leukemia does not necessarily start as a disease, but rather as a need, with the normally functioning preleukemic hematopoietic stem cells trying to maintain health for years but in time succumbing to their own acquired virtues.


Asunto(s)
Preleucemia/diagnóstico , Preleucemia/etiología , Animales , Progresión de la Enfermedad , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/etiología , Neoplasias Hematológicas/terapia , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Leucemia/diagnóstico , Leucemia/etiología , Leucemia/terapia , Preleucemia/terapia
17.
Blood ; 121(21): 4377-87, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23482930

RESUMEN

Transient abnormal myelopoiesis (TAM) is a clonal preleukemic disorder that progresses to myeloid leukemia of Down syndrome (ML-DS) through the accumulation of genetic alterations. To investigate the mechanism of leukemogenesis in this disorder, a xenograft model of TAM was established using NOD/Shi-scid, interleukin (IL)-2Rγ(null) mice. Serial engraftment after transplantation of cells from a TAM patient who developed ML-DS a year later demonstrated their self-renewal capacity. A GATA1 mutation and no copy number alterations (CNAs) were detected in the primary patient sample by conventional genomic sequencing and CNA profiling. However, in serial transplantations, engrafted TAM-derived cells showed the emergence of divergent subclones with another GATA1 mutation and various CNAs, including a 16q deletion and 1q gain, which are clinically associated with ML-DS. Detailed genomic analysis identified minor subclones with a 16q deletion or this distinct GATA1 mutation in the primary patient sample. These results suggest that genetically heterogeneous subclones with varying leukemia-initiating potential already exist in the neonatal TAM phase, and ML-DS may develop from a pool of such minor clones through clonal selection. Our xenograft model of TAM may provide unique insight into the evolutionary process of leukemia.


Asunto(s)
Evolución Clonal/fisiología , Síndrome de Down/sangre , Síndrome de Down/complicaciones , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Reacción Leucemoide/genética , Reacción Leucemoide/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Síndrome de Down/genética , Síndrome de Down/patología , Síndrome de Down/terapia , Recambio Total de Sangre , Femenino , Factor de Transcripción GATA1/genética , Humanos , Recién Nacido , Leucemia Mieloide/terapia , Reacción Leucemoide/terapia , Masculino , Ratones , Ratones Noqueados , Ratones SCID , Preleucemia/genética , Preleucemia/patología , Preleucemia/terapia , Trasplante Heterólogo
18.
Blood ; 122(24): 3908-17, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24021668

RESUMEN

Transient abnormal myelopoiesis (TAM), a preleukemic disorder unique to neonates with Down syndrome (DS), may transform to childhood acute myeloid leukemia (ML-DS). Acquired GATA1 mutations are present in both TAM and ML-DS. Current definitions of TAM specify neither the percentage of blasts nor the role of GATA1 mutation analysis. To define TAM, we prospectively analyzed clinical findings, blood counts and smears, and GATA1 mutation status in 200 DS neonates. All DS neonates had multiple blood count and smear abnormalities. Surprisingly, 195 of 200 (97.5%) had circulating blasts. GATA1 mutations were detected by Sanger sequencing/denaturing high performance liquid chromatography (Ss/DHPLC) in 17 of 200 (8.5%), all with blasts >10%. Furthermore low-abundance GATA1 mutant clones were detected by targeted next-generation resequencing (NGS) in 18 of 88 (20.4%; sensitivity ∼0.3%) DS neonates without Ss/DHPLC-detectable GATA1 mutations. No clinical or hematologic features distinguished these 18 neonates. We suggest the term "silent TAM" for neonates with DS with GATA1 mutations detectable only by NGS. To identify all babies at risk of ML-DS, we suggest GATA1 mutation and blood count and smear analyses should be performed in DS neonates. Ss/DPHLC can be used for initial screening, but where GATA1 mutations are undetectable by Ss/DHPLC, NGS-based methods can identify neonates with small GATA1 mutant clones.


Asunto(s)
Células Clonales/metabolismo , Síndrome de Down/genética , Mutación , Enfermedad Aguda , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromatografía Líquida de Alta Presión/métodos , Células Clonales/patología , Análisis Mutacional de ADN/métodos , Síndrome de Down/sangre , Factor de Transcripción GATA1 , Pruebas Genéticas/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Recién Nacido , Leucemia Mieloide/sangre , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/genética , Mielopoyesis/genética , Tamizaje Neonatal/métodos , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Preleucemia/sangre , Preleucemia/diagnóstico , Preleucemia/genética , Reproducibilidad de los Resultados , Factores de Riesgo , Sensibilidad y Especificidad
19.
Blood ; 121(1): 178-87, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23152544

RESUMEN

The origin of aberrant DNA methylation in cancer remains largely unknown. In the present study, we elucidated the DNA methylome in primary acute promyelocytic leukemia (APL) and the role of promyelocytic leukemia-retinoic acid receptor α (PML-RARα) in establishing these patterns. Cells from APL patients showed increased genome-wide DNA methylation with higher variability than healthy CD34(+) cells, promyelocytes, and remission BM cells. A core set of differentially methylated regions in APL was identified. Age at diagnosis, Sanz score, and Flt3-mutation status characterized methylation subtypes. Transcription factor-binding sites (eg, the c-myc-binding sites) were associated with low methylation. However, SUZ12- and REST-binding sites identified in embryonic stem cells were preferentially DNA hypermethylated in APL cells. Unexpectedly, PML-RARα-binding sites were also protected from aberrant DNA methylation in APL cells. Consistent with this, myeloid cells from preleukemic PML-RARα knock-in mice did not show altered DNA methylation and the expression of PML-RARα in hematopoietic progenitor cells prevented differentiation without affecting DNA methylation. Treatment of APL blasts with all-trans retinoic acid also did not result in immediate DNA methylation changes. The results of the present study suggest that aberrant DNA methylation is associated with leukemia phenotype but is not required for PML-RARα-mediated initiation of leukemogenesis.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/genética , Regulación Leucémica de la Expresión Génica , Leucemia Promielocítica Aguda/genética , Factores de Transcripción/metabolismo , Animales , Transformación Celular Neoplásica/genética , Cromosomas Humanos/ultraestructura , Islas de CpG , ADN de Neoplasias/metabolismo , Progresión de la Enfermedad , Técnicas de Sustitución del Gen , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/fisiología , Fenotipo , Complejo Represivo Polycomb 2/metabolismo , Preleucemia/genética , Proteínas Recombinantes de Fusión/fisiología , Proteínas Represoras/metabolismo , Tretinoina/uso terapéutico
20.
J Biol Chem ; 288(25): 18219-27, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23673656

RESUMEN

Acute T-cell lymphoblastic leukemia/lymphoma (T-ALL) is an aggressive hematopoietic malignancy affecting both children and adults. Previous studies of T-ALL mouse models induced by different genetic mutations have provided highly diverse results on the issues of T-cell leukemia/lymphoma-initiating cells (T-LICs) and potential mechanisms contributing to T-LIC transformation. Here, we show that oncogenic Kras (Kras G12D) expressed from its endogenous locus is a potent inducer of T-ALL even in a less sensitized BALB/c background. Notch1 mutations, including exon 34 mutations and recently characterized type 1 and 2 deletions, are detected in 100% of Kras G12D-induced T-ALL tumors. Although these mutations are not detected at the pre-leukemia stage, incremental up-regulation of NOTCH1 surface expression is observed at the pre-leukemia and leukemia stages. As secondary genetic hits in the Kras G12D model, Notch1 mutations target CD8(+) T-cells but not hematopoietic stem cells to further promote T-ALL progression. Pre-leukemia T-cells without detectable Notch1 mutations do not induce T-ALL in secondary recipient mice compared with T-ALL tumor cells with Notch1 mutations. We found huge variations in T-LIC frequency and immunophenotypes of cells enriched for T-LICs. Unlike Pten deficiency-induced T-ALL, oncogenic Kras-initiated T-ALL is not associated with up-regulation of the Wnt/ß-catenin pathway. Our results suggest that up-regulation of NOTCH1 signaling, through either overexpression of surface NOTCH1 or acquired gain-of-function mutations, is involved in both T-ALL initiation and progression. Notch1 mutations and Kras G12D contribute cooperatively to leukemogenic transformation of normal T-cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Transformación Celular Neoplásica/genética , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptor Notch1/genética , Adulto , Animales , Trasplante de Médula Ósea , Transformación Celular Neoplásica/metabolismo , Citometría de Flujo , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/cirugía , Preleucemia/genética , Preleucemia/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda