Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1285743, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37901253

RESUMEN

B-cell acute lymphoblastic leukemia (B-ALL) stands as the primary contributor to childhood cancer-related mortality on a global scale. The development of the most conventional forms of this disease has been proposed to be conducted by two different steps influenced by different types of risk factors. The first step is led by a genetic insult that is presumably acquired before birth that transforms a healthy cell into a preleukemic one, which is maintained untransformed until the second step takes place. This necessary next step to leukemia development will be triggered by different risk factors to which children are exposed after birth. Murine models that recap the stepwise progression of B-ALL have been instrumental in identifying environmental and genetic factors that contribute to disease risk. Recent evidence from these models has demonstrated that specific environmental risk factors, such as common infections or gut microbiome dysbiosis, induce immune stress, driving the transformation of preleukemic cells, and harboring genetic alterations, into fully transformed leukemic cells. Such models serve as valuable tools for investigating the mechanisms underlying preleukemic events and can aid in the development of preventive approaches for leukemia in child. Here, we discuss the existing knowledge, learned from mouse models, of the impact of genetic and environmental risk factors on childhood B-ALL evolution and how B-ALL prevention could be reached by interfering with preleukemic cells.


Asunto(s)
Leucemia de Células B , Leucemia Linfocítica Crónica de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Humanos , Ratones , Animales , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Factores de Riesgo
2.
Nat Commun ; 14(1): 5159, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37620322

RESUMEN

The initial steps of B-cell acute lymphoblastic leukemia (B-ALL) development usually pass unnoticed in children. Several preclinical studies have shown that exposure to immune stressors triggers the transformation of preleukemic B cells to full-blown B-ALL, but how this takes place is still a longstanding and unsolved challenge. Here we show that dysregulation of innate immunity plays a driving role in the clonal evolution of pre-malignant Pax5+/- B-cell precursors toward leukemia. Transcriptional profiling reveals that Myd88 is downregulated in immune-stressed pre-malignant B-cell precursors and in leukemic cells. Genetic reduction of Myd88 expression leads to a significant increase in leukemia incidence in Pax5+/-Myd88+/- mice through an inflammation-dependent mechanism. Early induction of Myd88-independent Toll-like receptor 3 signaling results in a significant delay of leukemia development in Pax5+/- mice. Altogether, these findings identify a role for innate immunity dysregulation in leukemia, with important implications for understanding and therapeutic targeting of the preleukemic state in children.


Asunto(s)
Linfoma de Burkitt , Leucemia , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Animales , Ratones , Células Precursoras de Linfocitos B , Factor 88 de Diferenciación Mieloide/genética , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Inmunidad Innata , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética
3.
Int J Mol Sci ; 24(14)2023 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-37511301

RESUMEN

Cancer stem cells (CSCs) are now well-established as key players in tumor initiation, progression, and therapy resistance [...].


Asunto(s)
Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/patología , Células Madre Neoplásicas/patología
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.
Trends Cancer ; 8(11): 887-889, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35871053

RESUMEN

Preleukemic has been used to describe children with a propensity to develop B cell acute lymphoblastic leukemia (B-ALL). However, leukemia-predisposing mutations can also be present in differentiated cells unable to transform. We postulate that preleukemia should only be used when such mutations arise in progenitors capable of evolving to B-ALL.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Preleucemia , Niño , Humanos , Preleucemia/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Mutación
7.
Cancer Res ; 82(6): 1098-1109, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35131871

RESUMEN

Preventing development of childhood B-cell acute lymphoblastic leukemia (B-ALL), a disease with devastating effects, is a longstanding and unsolved challenge. Heterozygous germline alterations in the PAX5 gene can lead to B-ALL upon accumulation of secondary mutations affecting the JAK/STAT signaling pathway. Preclinical studies have shown that this malignant transformation occurs only under immune stress such as exposure to infectious pathogens. Here we show in Pax5+/- mice that transient, early-life administration of clinically relevant doses of ruxolitinib, a JAK1/2 inhibitor, significantly mitigates the risk of B-ALL following exposure to infection; 1 of 29 animals treated with ruxolitinib developed B-ALL versus 8 of 34 untreated mice. Ruxolitinib treatment preferentially targeted Pax5+/- versus wild-type B-cell progenitors and exerted unique effects on the Pax5+/- B-cell progenitor transcriptional program. These findings provide the first in vivo evidence for a potential strategy to prevent B-ALL development. SIGNIFICANCE: JAK/STAT inhibition suppresses tumorigenesis in a B-ALL-susceptible mouse model, presenting a novel approach to prevent B-ALL onset.


Asunto(s)
Quinasas Janus , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Humanos , Quinasas Janus/genética , Ratones , Factor de Transcripción PAX5/genética , Factores de Transcripción STAT , Transducción de Señal/genética
8.
Front Cell Dev Biol ; 9: 704591, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336858

RESUMEN

ETV6-RUNX1 is almost exclusively associated with childhood B-cell acute lymphoblastic leukemia (B-ALL), but the consequences of ETV6-RUNX1 expression on cell lineage decisions during B-cell leukemogenesis are completely unknown. Clinically silent ETV6-RUNX1 preleukemic clones are frequently found in neonatal cord blood, but few carriers develop B-ALL as a result of secondary genetic alterations. The understanding of the mechanisms underlying the first transforming steps could greatly advance the development of non-toxic prophylactic interventions. Using genetic lineage tracing, we examined the capacity of ETV6-RUNX1 to instruct a malignant phenotype in the hematopoietic lineage by cell-specific Cre-mediated activation of ETV6-RUNX1 from the endogenous Etv6 gene locus. Here we show that, while ETV6-RUNX1 has the propensity to trigger both T- and B-lymphoid malignancies, it is the second hit that determines tumor cell identity. To instigate leukemia, both oncogenic hits must place early in the development of hematopoietic/precursor cells, not in already committed B-cells. Depending on the nature of the second hit, the resulting B-ALLs presented distinct entities that were clearly separable based on their gene expression profiles. Our findings give a novel mechanistic insight into the early steps of ETV6-RUNX1+ B-ALL development and might have major implications for the potential development of ETV6-RUNX1+ B-ALL prevention strategies.

9.
Trends Immunol ; 42(5): 371-374, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33773925

RESUMEN

The prevalence of childhood B cell acute lymphoblastic leukemia (B-ALL) is increasing, particularly in developed countries. There is no clear explanation for this increment, but recent data suggest that, besides genetic predisposition, stress in the immune system (e.g., an infection) might have an important role in B-ALL leukemogenesis. Here, we speculate on how this knowledge might impact B-ALL prevention strategies.


Asunto(s)
Leucemia de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Predisposición Genética a la Enfermedad , Humanos
10.
Nat Rev Immunol ; 21(9): 570-581, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33558682

RESUMEN

B cell acute lymphoblastic leukaemia (B-ALL) is the most common form of childhood cancer. Although treatment has advanced remarkably in the past 50 years, it still fails in ~20% of patients. Recent studies revealed that more than 5% of healthy newborns carry preleukaemic clones that originate in utero, but only a small percentage of these carriers will progress to overt B-ALL. The drivers of progression are unclear, but B-ALL incidence seems to be increasing in parallel with the adoption of modern lifestyles. Emerging evidence shows that a major driver for the conversion from the preleukaemic state to the B-ALL state is exposure to immune stressors, such as infection. Here, we discuss our current understanding of the environmental triggers and genetic predispositions that may lead to B-ALL, highlighting lessons from epidemiology, the clinic and animal models, and identifying priority areas for future research.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras B/etiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Animales , Carcinogénesis/genética , Niño , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos , Infecciones/complicaciones , Microbiota/inmunología , Modelos Biológicos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo , Preleucemia/etiología , Preleucemia/genética , Preleucemia/inmunología , Factores de Riesgo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
11.
Blood ; 137(13): 1741-1753, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33024996

RESUMEN

Diffuse large B-cell lymphomas (DLBCLs) are clinically and genetically heterogeneous tumors. Deregulation of diverse biological processes specific to B cells, such as B-cell receptor (BCR) signaling and motility regulation, contribute to lymphomagenesis. Human germinal center associated lymphoma (HGAL) is a B-cell-specific adaptor protein controlling BCR signaling and B lymphocyte motility. In normal B cells, it is expressed in germinal center (GC) B lymphocytes and promptly downregulated upon further differentiation. The majority of DLBCL tumors, primarily GC B-cell types, but also activated types, express HGAL. To investigate the consequences of constitutive expression of HGAL in vivo, we generated mice that conditionally express human HGAL at different stages of hematopoietic development using 3 restricted Cre-mediated approaches to initiate expression of HGAL in hematopoietic stem cells, pro-B cells, or GC B cells. Following immune stimulation, we observed larger GCs in mice in which HGAL expression was initiated in GC B cells. All 3 mouse strains developed DLBCL at a frequency of 12% to 30% starting at age 13 months, leading to shorter survival. Immunohistochemical studies showed that all analyzed tumors were of the GC B-cell type. Exon sequencing revealed mutations reported in human DLBCL. Our data demonstrate that constitutive enforced expression of HGAL leads to DLBCL development.


Asunto(s)
Carcinogénesis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Linfoma de Células B Grandes Difuso/genética , Proteínas de Microfilamentos/genética , Animales , Carcinogénesis/patología , Línea Celular , Femenino , Mutación con Ganancia de Función , Regulación Neoplásica de la Expresión Génica , Centro Germinal/metabolismo , Centro Germinal/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Endogámicos C57BL
12.
Methods Mol Biol ; 2185: 361-372, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33165860

RESUMEN

Leukemia is a clonal malignant disease originated in a single cell and characterized by the accumulation of abnormal lymphoid cells. The nature of the leukemic stem cell (LSC) has been a subject of continuing discussion, given the fact that human disease is diagnosed at late stages and cannot be monitored during its natural evolution from its cell of origin. Animal models provide a means to determine the leukemic initiating cell and the causes of malignancy, and to develop new treatments. Recent findings in mice have shown that cancer stem cells can initially arise through a reprogramming-like mechanism when the oncogene expression is targeted to the mouse stem cell compartment (Garcia-Ramirez et al., EMBO J 37(14):298783, 2018; Martin-Lorenzo et al., Cancer Res 78 (10):2669-2679, 2018; Perez-Caro et al., EMBO J 28(1):8-20, 2009; Rodriguez-Hernandez et al., Cancer Res 77(16):4365-4377, 2017). If leukemia arises through reprogramming processes, then perhaps many of the oncogenes that initiate tumor formation might be dispensable for tumor progression and maintenance. Leukemia will be modeled in the mice only if we are able to target the right cancer-initiating cell with a precise given oncogene. In the last years, some examples have already started to appear in the literature showing that targeting oncogene expression to the stem cell compartment in model mice might be the correct way of reproducing the genotype-phenotype correlations found in human leukemias (Garcia-Ramirez et al., EMBO J 37(14):298783, 2018; Martin-Lorenzo et al., Cancer Res 78 (10):2669-2679, 2018; Perez-Caro et al., EMBO J 28(1):8-20, 2009; Rodriguez-Hernandez et al., Cancer Res 77(16):4365-4377, 2017). This chapter addresses how to generate LSCs by transgenesis in a way that makes the resulting animal models valuable tools to reproduce and understand leukemogenesis, and for the development of therapeutic applications like drug discovery or biomarker identification.


Asunto(s)
Transformación Celular Neoplásica , Técnicas de Reprogramación Celular , Regulación Leucémica de la Expresión Génica , Leucemia , Células Madre Neoplásicas/metabolismo , Oncogenes , Animales , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Ratones , Células Madre Neoplásicas/patología
14.
Sci Rep ; 10(1): 19189, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33154497

RESUMEN

PAX5 is one of the most frequently mutated genes in B-cell acute lymphoblastic leukemia (B-ALL), and children with inherited preleukemic PAX5 mutations are at a higher risk of developing the disease. Abnormal profiles of inflammatory markers have been detected in neonatal blood spot samples of children who later developed B-ALL. However, how inflammatory signals contribute to B-ALL development is unclear. Here, we demonstrate that Pax5 heterozygosis, in the presence of infections, results in the enhanced production of the inflammatory cytokine interleukin-6 (IL-6), which appears to act in an autocrine fashion to promote leukemia growth. Furthermore, in vivo genetic downregulation of IL-6 in these Pax5 heterozygous mice retards B-cell leukemogenesis, and in vivo pharmacologic inhibition of IL-6 with a neutralizing antibody in Pax5 mutant mice with B-ALL clears leukemic cells. Additionally, this novel IL-6 signaling paradigm identified in mice was also substantiated in humans. Altogether, our studies establish aberrant IL6 expression caused by Pax5 loss as a hallmark of Pax5-dependent B-ALL and the IL6 as a therapeutic vulnerability for B-ALL characterized by PAX5 loss.


Asunto(s)
Linfocitos B/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Factor de Transcripción PAX5/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Transducción de Señal/genética , Animales , Inflamación/genética , Interleucina-6/genética , Ratones , Ratones Noqueados , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética
15.
Blood ; 136(18): 2003-2017, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32911536

RESUMEN

The majority of childhood leukemias are precursor B-cell acute lymphoblastic leukemias (pB-ALLs) caused by a combination of prenatal genetic predispositions and oncogenic events occurring after birth. Although genetic predispositions are frequent in children (>1% to 5%), fewer than 1% of genetically predisposed carriers will develop pB-ALL. Although infectious stimuli are believed to play a major role in leukemogenesis, the critical determinants are not well defined. Here, by using murine models of pB-ALL, we show that microbiome disturbances incurred by antibiotic treatment early in life were sufficient to induce leukemia in genetically predisposed mice, even in the absence of infectious stimuli and independent of T cells. By using V4 and full-length 16S ribosomal RNA sequencing of a series of fecal samples, we found that genetic predisposition to pB-ALL (Pax5 heterozygosity or ETV6-RUNX1 fusion) shaped a distinct gut microbiome. Machine learning accurately (96.8%) predicted genetic predisposition using 40 of 3983 amplicon sequence variants as proxies for bacterial species. Transplantation of either wild-type (WT) or Pax5+/- hematopoietic bone marrow cells into WT recipient mice revealed that the microbiome is shaped and determined in a donor genotype-specific manner. Gas chromatography-mass spectrometry (GC-MS) analyses of sera from WT and Pax5+/- mice demonstrated the presence of a genotype-specific distinct metabolomic profile. Taken together, our data indicate that it is a lack of commensal microbiota rather than the presence of specific bacteria that promotes leukemia in genetically predisposed mice. Future large-scale longitudinal studies are required to determine whether targeted microbiome modification in children predisposed to pB-ALL could become a successful prevention strategy.


Asunto(s)
Susceptibilidad a Enfermedades , Disbiosis/complicaciones , Heces/microbiología , Microbioma Gastrointestinal , Leucemia Experimental/prevención & control , Factor de Transcripción PAX5/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/prevención & control , Animales , Femenino , Leucemia Experimental/genética , Leucemia Experimental/microbiología , Leucemia Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/microbiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología
17.
Blood Cancer Discov ; 1(3): 224-233, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33392513

RESUMEN

B-cells are an integral part of the adaptive immune system and regulate innate immunity. Derived from hematopoietic stem cells they mature through a series of cell fate decisions. Complex transcriptional circuits form and dissipate dynamically during these lineage restrictions. Genomic aberrations of involved transcription factors underlie various B-cell disorders. Acquired somatic aberrations are associated with cancer, whereas germline variations predispose to both malignant and non-malignant diseases. We review the opposing role of transcription factors during B-cell development in health and disease. We focus on early B-cell leukemia and discuss novel causative gene-environment cooperations and their implications for precision medicine.


Asunto(s)
Linfocitos B , Leucemia , Factores de Transcripción , Linfocitos B/fisiología , Diferenciación Celular/genética , Interacción Gen-Ambiente , Hematopoyesis , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia/genética , Leucemia/inmunología , Factores de Transcripción/genética
18.
Nat Commun ; 10(1): 5563, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804490

RESUMEN

The prerequisite to prevent childhood B-cell acute lymphoblastic leukemia (B-ALL) is to decipher its etiology. The current model suggests that infection triggers B-ALL development through induction of activation-induced cytidine deaminase (AID; also known as AICDA) in precursor B-cells. This evidence has been largely acquired through the use of ex vivo functional studies. However, whether this mechanism governs native non-transplant B-ALL development is unknown. Here we show that, surprisingly, AID genetic deletion does not affect B-ALL development in Pax5-haploinsufficient mice prone to B-ALL upon natural infection exposure. We next test the effect of premature AID expression from earliest pro-B-cell stages in B-cell transformation. The generation of AID off-target mutagenic activity in precursor B-cells does not promote B-ALL. Likewise, known drivers of human B-ALL are not preferentially targeted by AID. Overall these results suggest that infections promote B-ALL through AID-independent mechanisms, providing evidence for a new model of childhood B-ALL development.


Asunto(s)
Linfocitos B/metabolismo , Transformación Celular Neoplásica/metabolismo , Citidina Desaminasa/metabolismo , Infecciones/fisiopatología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Animales , Linfocitos B/patología , Transformación Celular Neoplásica/genética , Niño , Citidina Desaminasa/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Infecciones/genética , Estimación de Kaplan-Meier , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética
19.
Front Cell Dev Biol ; 7: 137, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31380372

RESUMEN

Leukemogenesis is considered to be a process by which a normal cell acquires new but aberrant identity in order to disseminate a malignant clonal population. Under this setting, the phenotype of the leukemic cells is identical to the leukemia-initiating cell in which the genetic insult is taking place. Thus, with some exceptions, B-cell and T-cell childhood leukemias are supposed to arise from B- or T-committed cells. In contrast, several recent studies have revealed that genetic alterations may act in a "hit-and-run" way in the cell-of-origin by imposing the tumor cell identity giving rise to either B-cell or T-cell leukemias. This novel mechanism of cell transformation is mediated by an epigenetic priming mechanism that is established by the initial genetic lesion. This initial hit might be unnecessary for the subsequent tumor evolution and conservation, being the epigenetic priming the engine for the tumor evolution.

20.
Bioelectromagnetics ; 40(5): 343-353, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31157932

RESUMEN

Exposure to extremely low-frequency magnetic fields (ELF-MFs) has been classified by the International Agency for Research on Cancer (IARC) as "possibly carcinogenic to humans," based on limited scientific evidence concerning childhood leukemia. This assessment emphasized the lack of appropriate animal models recapitulating the natural history of this disease. Childhood B-cell acute lymphoblastic leukemia (B-ALL) is the result of complex interactions between genetic susceptibility and exposure to exogenous agents. The most common chromosomal alteration is the ETV6-RUNX1 fusion gene, which confers a low risk of developing the malignancy by originating a preleukemic clone requiring secondary hits for full-blown disease to appear. To develop potential prophylactic interventions, we need to identify the environmental triggers of the second hit. Recently, we generated a B-ALL mouse model of the human ETV6-RUNX1+ preleukemic state. Here, we present the results from the ARIMMORA pilot study, obtained by exposing 34 Sca1-ETV6-RUNX1 mice (vs. 27 unexposed) to a 50 Hz magnetic field of 1.5 mT with both fundamental and harmonic content, with an on/off cycle of 10 min/5 min, for 20 h/day, from conception until 3 months of age. Mice were monitored until 2 years of age and peripheral blood was periodically analyzed by flow cytometry. One of the exposed mice developed B-ALL while none of the non-exposed did. Although the results are statistically non-significant due to the limited number of mice used in this pilot experiment, overall, the results show that the newly developed Sca1-ETV6-RUNX1 mouse can be successfully used for ELF-MF exposure studies about the etiology of childhood B-ALL. Bioelectromagnetics. 2019;40:343-353. © 2019 Bioelectromagnetics Society.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Campos Electromagnéticos/efectos adversos , Leucemia Experimental , Leucemia-Linfoma Linfoblástico de Células Precursoras , Proteínas Proto-Oncogénicas c-ets/genética , Ondas de Radio/efectos adversos , Proteínas Represoras/genética , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Humanos , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proyectos Piloto , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Represoras/metabolismo , Proteína ETS de Variante de Translocación 6
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA