Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 81(3): 530-545.e5, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33382982

RESUMEN

Transcription factors regulate gene networks controlling normal hematopoiesis and are frequently deregulated in acute myeloid leukemia (AML). Critical to our understanding of the mechanism of cellular transformation by oncogenic transcription factors is the ability to define their direct gene targets. However, gene network cascades can change within minutes to hours, making it difficult to distinguish direct from secondary or compensatory transcriptional changes by traditional methodologies. To overcome this limitation, we devised cell models in which the AML1-ETO protein could be quickly degraded upon addition of a small molecule. The rapid kinetics of AML1-ETO removal, when combined with analysis of transcriptional output by nascent transcript analysis and genome-wide AML1-ETO binding by CUT&RUN, enabled the identification of direct gene targets that constitute a core AML1-ETO regulatory network. Moreover, derepression of this gene network was associated with RUNX1 DNA binding and triggered a transcription cascade ultimately resulting in myeloid differentiation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , ARN Neoplásico/biosíntesis , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Transcripción Genética , Acetilación , Sitios de Unión , Unión Competitiva , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Autorrenovación de las Células , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Sangre Fetal/citología , Regulación Leucémica de la Expresión Génica , Redes Reguladoras de Genes , Células HEK293 , Células Madre Hematopoyéticas/patología , Histonas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Proteínas de Fusión Oncogénica/genética , Unión Proteica , Proteolisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Neoplásico/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Tiempo , Transcriptoma
2.
Proc Natl Acad Sci U S A ; 119(42): e2213718119, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36215477

RESUMEN

Transcription factors (TFs) play critical roles in hematopoiesis, and their aberrant expression can lead to various types of leukemia. The t(8;21) leukemogenic fusion protein AML1-ETO (AE) is the most common fusion protein in acute myeloid leukemia and can enhance hematopoietic stem cell renewal while blocking differentiation. A key question in understanding AE-mediated leukemia is what determines the choice of AE to activate self-renewal genes or repress differentiation genes. Toward the resolution of this problem, we earlier showed that AE resides in the stable AETFC complex and that its components colocalize on up- or down-regulated target genes and are essential for leukemogenesis. In the current study, using biochemical and genomic approaches, we show that AE-containing complexes are heterogeneous, and that assembly of the larger AETFC (containing AE, CBFß, HEB, E2A, LYL1, LMO2, and LDB1) requires LYL1. Furthermore, we provide strong evidence that the LYL1-containing AETFC preferentially binds to active enhancers and promotes AE-dependent gene activation. Moreover, we show that coactivator CARM1 interacts with AETFC and facilitates gene activation by AETFC. Collectively, this study describes a role of oncoprotein LYL1 in AETFC assembly and gene activation by recruiting CARM1 to chromatin for AML cell survival.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Adaptadoras de Señalización CARD , Cromatina , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Guanilato Ciclasa , Humanos , Proteínas con Homeodominio LIM/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína-Arginina N-Metiltransferasas , Activación Transcripcional
3.
FASEB J ; 36(10): e22562, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36125067

RESUMEN

Oncoprotein AML1-ETO (AE) derived from t(8;21)(q22;q22) translocation is typically present in a portion of French-American-British-M2 subtype of acute myeloid leukemia (AML). Although these patients have relatively favorable prognoses, substantial numbers of them would relapse after conventional therapy. Here, we explored whether reinforcing the endogenous differentiation potential of t(8;21) AML cells would diminish the associated malignancy. In doing so, we noticed an expansion of immature erythroid blasts featured in both AML1-ETO9a (AE9a) and AE plus c-KIT (N822K) (AK) murine leukemic models. Interestingly, in the AE9a murine model, a spontaneous step-wise erythroid differentiation path, as characterized by the differential expression of CD43/c-Kit and the upregulation of several key erythroid transcription factors (TFs), accompanied the decline or loss of leukemia-initiating potential. Notably, overexpression of one of the key erythroid TFs, Ldb1, potently disrupted the repopulation of AE9a leukemic cells in vivo, suggesting a new promising intervention strategy of t(8;21) AML through enforcing their erythroid differentiation.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica , Animales , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas con Dominio LIM , Proteínas con Homeodominio LIM , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/genética , Translocación Genética
4.
Genes Dev ; 29(20): 2123-39, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26494788

RESUMEN

RUNX1-RUNX1T1 (formerly AML1-ETO), a transcription factor generated by the t(8;21) translocation in acute myeloid leukemia (AML), dictates a leukemic program by increasing self-renewal and inhibiting differentiation. Here we demonstrate that the histone demethylase JMJD1C functions as a coactivator for RUNX1-RUNX1T1 and is required for its transcriptional program. JMJD1C is directly recruited by RUNX1-RUNX1T1 to its target genes and regulates their expression by maintaining low H3K9 dimethyl (H3K9me2) levels. Analyses in JMJD1C knockout mice also establish a JMJD1C requirement for RUNX1-RUNX1T1's ability to increase proliferation. We also show a critical role for JMJD1C in the survival of multiple human AML cell lines, suggesting that it is required for leukemic programs in different AML cell types through its association with key transcription factors.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia Mieloide Aguda/fisiopatología , Oxidorreductasas N-Desmetilantes/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Leucemia Mieloide Aguda/genética , Ratones Noqueados , Oxidorreductasas N-Desmetilantes/genética , Transporte de Proteínas/genética
5.
Pharmacol Res ; 185: 106503, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36241000

RESUMEN

Acute myeloid leukemia (AML) is featured with poor prognosis and high mortality, because chemo-resistance, nonspecific distribution and dose-limiting toxicity lead to a high rate of relapse and a very low 5-year survival percentage of less than 25%. CXCR4 is a highly expressed chemokine receptor in multiple types of AML cells and closely associated with the drug resistance and relapse. In this work, we integrate a chemically synthesized CXCR4 antagonistic peptide and doxorubicin using DSPE-mPEG2000 micelles (referred to as M-E5-Dox) that is applied to a very challenging refractory AML mouse model as well as human AML cell lines. Results showed that M-E5-Dox can effectively bind to the CXCR4-expressing AML cells, downregulating the signaling proteins mediated by CXCR4/CXCL12 axis and increasing the cellular uptake of Dox. Importantly, M-E5-Dox remarkably decreases the leukemic cells in the peripheral blood and bone marrow, as well as their infiltration in the spleen and liver of the AML mice, which in turn prolongs the survival significantly. Meanwhile, M-E5-Dox did not increase the cardiotoxicity of Dox. In conclusion, M-E5-Dox harnesses the functions of CXCR4 specific binding and CXCR4 antagonism of the peptide and the tumor cell killing capacity of Dox, which displays significant therapeutic effects and promising translational potentials for the treatment of refractory AML.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Ratones , Animales , Leucemia Mieloide Aguda/metabolismo , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Transducción de Señal , Péptidos/farmacología , Recurrencia , Receptores CXCR4
6.
Proc Natl Acad Sci U S A ; 116(3): 890-899, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30593567

RESUMEN

The AML1-ETO fusion protein, generated by the t(8;21) chromosomal translocation, is causally involved in nearly 20% of acute myeloid leukemia (AML) cases. In leukemic cells, AML1-ETO resides in and functions through a stable protein complex, AML1-ETO-containing transcription factor complex (AETFC), that contains multiple transcription (co)factors. Among these AETFC components, HEB and E2A, two members of the ubiquitously expressed E proteins, directly interact with AML1-ETO, confer new DNA-binding capacity to AETFC, and are essential for leukemogenesis. However, the third E protein, E2-2, is specifically silenced in AML1-ETO-expressing leukemic cells, suggesting E2-2 as a negative factor of leukemogenesis. Indeed, ectopic expression of E2-2 selectively inhibits the growth of AML1-ETO-expressing leukemic cells, and this inhibition requires the bHLH DNA-binding domain. RNA-seq and ChIP-seq analyses reveal that, despite some overlap, the three E proteins differentially regulate many target genes. In particular, studies show that E2-2 both redistributes AETFC to, and activates, some genes associated with dendritic cell differentiation and represses MYC target genes. In AML patients, the expression of E2-2 is relatively lower in the t(8;21) subtype, and an E2-2 target gene, THPO, is identified as a potential predictor of relapse. In a mouse model of human t(8;21) leukemia, E2-2 suppression accelerates leukemogenesis. Taken together, these results reveal that, in contrast to HEB and E2A, which facilitate AML1-ETO-mediated leukemogenesis, E2-2 compromises the function of AETFC and negatively regulates leukemogenesis. The three E proteins thus define a heterogeneity of AETFC, which improves our understanding of the precise mechanism of leukemogenesis and assists development of diagnostic/therapeutic strategies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/etiología , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/metabolismo , Recurrencia
7.
Int J Mol Sci ; 23(17)2022 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-36077220

RESUMEN

Topoisomerase inhibitors are widely used in cancer chemotherapy. However, one of the potential long-term adverse effects of such therapy is acute leukemia. A key feature of such therapy-induced acute myeloid leukemia (t-AML) is recurrent chromosomal translocations involving AML1 (RUNX1) or MLL (KMT2A) genes. The formation of chromosomal translocation depends on the spatial proximity of translocation partners and the mobility of the DNA ends. It is unclear which of these two factors might be decisive for recurrent t-AML translocations. Here, we used fluorescence in situ hybridization (FISH) and chromosome conformation capture followed by sequencing (4C-seq) to investigate double-strand DNA break formation and the mobility of broken ends upon etoposide treatment, as well as contacts between translocation partner genes. We detected the separation of the parts of the broken AML1 gene, as well as the increased mobility of these separated parts. 4C-seq analysis showed no evident contacts of AML1 and MLL with loci, implicated in recurrent t-AML translocations, either before or after etoposide treatment. We suggest that separation of the break ends and their increased non-targeted mobility-but not spatial predisposition of the rearrangement partners-plays a major role in the formation of these translocations.


Asunto(s)
Leucemia Mieloide Aguda , Translocación Genética , ADN , Roturas del ADN de Doble Cadena , Etopósido/efectos adversos , Humanos , Hibridación Fluorescente in Situ , Leucemia Mieloide Aguda/genética , Inhibidores de Topoisomerasa II/efectos adversos
8.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-36613512

RESUMEN

The AML1-ETO (RUNX1-RUNX1T1) fusion gene created by the chromosome translocation t(8;21) (q21;q22) is one of the essential contributors to leukemogenesis. Only a few studies in the literature have focused on fusion gene-derived circular RNAs (f-circRNAs). Here, we report several AML1-ETO-related fusion circular RNAs (F-CircAEs) in AML1-ETO-positive cell lines and primary patient blasts. Functional studies demonstrate that the over-expression of F-CircAE in NIH3T3 cells promotes cell proliferation in vitro and in vivo. F-CircAE expression enhances the colony formation ability of c-Kit+ hematopoietic stem and progenitor cells (HSPCs). Meanwhile, the knockdown of endogenous F-CircAEs can inhibit the proliferation and colony formation ability of AML1-ETO-positive Kasumi-1 cells. Intriguingly, bioinformatic analysis revealed that the glycolysis pathway is down-regulated in F-CircAE-knockdown Kasumi-1 cells and up-regulated in F-CircAE over-expressed NIH3T3 cells. Further studies show that F-CircAE binds to the glycolytic protein ENO-1, up-regulates the expression level of glycolytic enzymes, and enhances lactate production. In summary, our study demonstrates that F-CircAE may exert biological activities on the growth of AML1-ETO leukemia cells by regulating the glycolysis pathway. Determining the role of F-CircAEs in AML1-ETO leukemia can lead to great strides in understanding its pathogenesis, thus providing new diagnostic markers and therapeutic targets.


Asunto(s)
Leucemia Mieloide Aguda , ARN Circular , Ratones , Animales , Humanos , ARN Circular/genética , Células 3T3 NIH , Proteína 1 Compañera de Translocación de RUNX1/genética , Leucemia Mieloide Aguda/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proliferación Celular/genética , Proteínas de Fusión Oncogénica/metabolismo , Cromosomas Humanos Par 21/metabolismo , Translocación Genética
9.
J Biol Chem ; 295(13): 4212-4223, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32071087

RESUMEN

In up to 15% of acute myeloid leukemias (AMLs), a recurring chromosomal translocation, termed t(8;21), generates the AML1-eight-twenty-one (ETO) leukemia fusion protein, which contains the DNA-binding domain of Runt-related transcription factor 1 (RUNX1) and almost all of ETO. RUNX1 and the AML1-ETO fusion protein are coexpressed in t(8;21) AML cells and antagonize each other's gene-regulatory functions. AML1-ETO represses transcription of RUNX1 target genes by competitively displacing RUNX1 and recruiting corepressors such as histone deacetylase 3 (HDAC3). Recent studies have shown that AML1-ETO and RUNX1 co-occupy the binding sites of AML1-ETO-activated genes. How this joined binding allows RUNX1 to antagonize AML1-ETO-mediated transcriptional activation is unclear. Here we show that RUNX1 functions as a bona fide repressor of transcription activated by AML1-ETO. Mechanistically, we show that RUNX1 is a component of the HDAC3 corepressor complex and that HDAC3 preferentially binds to RUNX1 rather than to AML1-ETO in t(8;21) AML cells. Studying the regulation of interleukin-8 (IL8), a newly identified AML1-ETO-activated gene, we demonstrate that RUNX1 and HDAC3 collaboratively repress AML1-ETO-dependent transcription, a finding further supported by results of genome-wide analyses of AML1-ETO-activated genes. These and other results from the genome-wide studies also have important implications for the mechanistic understanding of gene-specific coactivator and corepressor functions across the AML1-ETO/RUNX1 cistrome.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Histona Desacetilasas/genética , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Humanos , Interleucina-8/genética , Leucemia Mieloide Aguda/patología , Regiones Promotoras Genéticas , Activación Transcripcional/genética , Translocación Genética/genética
10.
Ann Hematol ; 100(10): 2593-2601, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34312685

RESUMEN

This study aims to explore the effect of the ITI (interferon alpha-1b, thalidomide, and interleukin-2) regimen on the AML1-ETO fusion gene in patients with t(8;21) acute myeloid leukemia (AML) who were in hematologic remission but positive for the AML1-ETO fusion gene. From September 2014 to November 2020; 20 patients with AML (15 from The Affiliated Cancer Hospital of Zhengzhou University, 4 from The First Affiliated Hospital; and College of Clinical Medicine of Henan University of Science and Technology, and 1 from Anyang District Hospital) with hematological remission but AML1-ETO fusion gene positivity were treated with different doses of the ITI regimen to monitor changes in AML1-ETO fusion gene levels. Twenty patients were treated with a routine dose of the ITI regimen, including 13 males and 7 females. The median patient age was 38 (14-70 years). The fusion gene was negative in 10 patients after 1 (0.5 ~ 8.6) month, significantly decreased in 4 patients after 2.8 (1 ~ 6) months, increased in 4 patients, and unchanged in 2 patients. The 4 patients with elevated levels of the fusion gene were treated with an increased dose of the ITI regimen, and all four patients became negative, for a total effective rate of 90%. The ITI regimen reduces AML1-ETO fusion gene levels in patients with AML who are in hematologic remission but are fusion gene-positive. Improvement was observed in patients' response to a higher dose administration, and patients tolerated the treatment well.


Asunto(s)
Antineoplásicos/uso terapéutico , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Factores Inmunológicos/uso terapéutico , Interferón-alfa/uso terapéutico , Interleucina-2/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Talidomida/uso terapéutico , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Persona de Mediana Edad , Fusión de Oncogenes/efectos de los fármacos , Adulto Joven
11.
Adv Exp Med Biol ; 1287: 9-30, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33034023

RESUMEN

The Notch signal transduction cascade requires cell-to-cell contact and results in the proteolytic processing of the Notch receptor and subsequent assembly of a transcriptional coactivator complex containing the Notch intracellular domain (NICD) and transcription factor RBPJ. In the absence of a Notch signal, RBPJ remains at Notch target genes and dampens transcriptional output. Like in other signaling pathways, RBPJ is able to switch from activation to repression by associating with corepressor complexes containing several chromatin-modifying enzymes. Here, we focus on the recent advances concerning RBPJ-corepressor functions, especially in regard to chromatin regulation. We put this into the context of one of the best-studied model systems for Notch, blood cell development. Alterations in the RBPJ-corepressor functions can contribute to the development of leukemia, especially in the case of acute myeloid leukemia (AML). The versatile role of transcription factor RBPJ in regulating pivotal target genes like c-MYC and HES1 may contribute to the better understanding of the development of leukemia.


Asunto(s)
Regulación de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Receptores Notch/metabolismo , Cromatina/genética , Cromatina/metabolismo , Humanos , Transducción de Señal
12.
Vnitr Lek ; 67(E-5): 9-12, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35459386

RESUMEN

Acute myeloid leukemia (AML) is a highly heterogeneous subtype of leukemia, accounting for 25 % of childhood leukemias. By the presence of genetic mutations in hematopoietic/ progenitor stem cells, the bone marrow produces a large number of abnormal undifferentiated leukocytes (blasts), which significantly impairs the proper differentiation of cells. AML is induced by two interventions. Chromosomal translocation during hematopoiesis of intrauterine development is the first intervention. This creates preleukemic fusion genes (PFG), which can later be transformed by a second intervention (point genetic mutation - deletion, insertion ) into a functional malignant clone. Characteristic AML fusion genes include AML1-ETO, PML-RARA or MLL-AF9, which in turn produce hybrid proteins with altered function. Several studies suggest that these PFGs are considered an important prognostic tool in disease assessment. While the incidence of PFG characteristic of acute lymphoblastic leukemia (ALL) has been relatively well studied by several research groups and has been estimated at 1 to 5% in the umbilical cord blood of healthy neonates, PFG relevant to AML are still not sufficiently clarified.


Asunto(s)
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Recién Nacido , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Pronóstico
13.
Immunology ; 160(1): 24-37, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32022254

RESUMEN

The transcription factor Foxp3 controls the differentiation and function of regulatory T-cells (Treg). Studies in the past decades identified numerous Foxp3-interacting protein partners. However, it is still not clear how Foxp3 produces the Treg-type transcriptomic landscape through cooperating with its partners. Here I show the current understanding of how Foxp3 transcription factor complexes regulate the differentiation, maintenance and functional maturation of Treg. Importantly, T-cell receptor (TCR) signalling plays central roles in Treg differentiation and Foxp3-mediated gene regulation. Differentiating Treg will have recognized their cognate antigens and received TCR signals before initiating Foxp3 transcription, which is triggered by TCR-induced transcription factors including NFAT, AP-1 and NF-κB. Once expressed, Foxp3 seizes TCR signal-induced transcriptional and epigenetic mechanisms through interacting with AML1/Runx1 and NFAT. Thus, Foxp3 modifies gene expression dynamics of TCR-induced genes, which constitute cardinal mechanisms for Treg-mediated immune suppression. Next, I discuss the following key topics, proposing new mechanistic models for Foxp3-mediated gene regulation: (i) how Foxp3 transcription is induced and maintained by the Foxp3-inducing enhanceosome and the Foxp3 autoregulatory transcription factor complex; (ii) molecular mechanisms for effector Treg differentiation (i.e. Treg maturation); (iii) how Foxp3 activates or represses its target genes through recruiting coactivators and corepressors; (iv) the 'decision-making' Foxp3-containing transcription factor complex for Th17 and Treg differentiation; and (v) the roles of post-translational modification in Foxp3 regulation. Thus, this article provides cutting-edge understanding of molecular biology of Foxp3 and Treg, integrating findings by biochemical and genomic studies.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Modelos Biológicos , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Epigénesis Genética/inmunología , Humanos , Activación de Linfocitos , Ratones , Procesamiento Proteico-Postraduccional/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Transcripción Genética/inmunología
14.
Clin Sci (Lond) ; 134(23): 3079-3091, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33201243

RESUMEN

The AML1-ETO oncoprotein, which results from t(8;21) translocation, is considered an initial event of t(8;21) acute myeloid leukemia (AML). However, the precise mechanisms of the oncogenic activity of AML1-ETO is yet to be fully determined. The present study demonstrates that AML1-ETO triggers the heterochromatic silencing of microRNA-564 (miR564) by binding at the AML1 binding site along the miR564 promoter region and recruiting chromatin-remodeling enzymes. Suppression of miR564 enhances the oncogenic activity of the AML1-ETO oncoprotein by directly inhibiting the expression of CCND1 and the DNMT3A genes. Ectopic expression of miR564 can induce retardation of G1/S transition, reperform differentiation, promote apoptosis, as well as inhibit the proliferation and colony formation of AML1-ETO+ leukemia cells in vitro. Enhanced miR564 levels can significantly inhibit the tumor proliferation of t(8;21)AML in vivo. We first identify an unexpected and important epigenetic circuitry of AML1-ETO/miR564/CCND1/DNMT3A that contributes to the leukemogenesis in vitro/vivo of AML1-ETO+ leukemia, indicating that miR564 enhancement could provide a potential therapeutic method for AML1-ETO+ leukemia.


Asunto(s)
Carcinogénesis/genética , Epigénesis Genética , Silenciador del Gen , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , MicroARNs/metabolismo , Translocación Genética , Animales , Apoptosis/genética , Secuencia de Bases , Carcinogénesis/patología , Puntos de Control del Ciclo Celular/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Ciclina D1/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , ADN Metiltransferasa 3A , Femenino , Regulación Leucémica de la Expresión Génica , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Proteínas de Fusión Oncogénica/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Regulación hacia Arriba/genética
15.
Rinsho Ketsueki ; 61(6): 687-696, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-32624544

RESUMEN

The RUNX1 gene is a critical transcription factor for the generation and maintenance of hematopoietic stem cells. RUNX1 is also one of the most frequently mutated gene in sporadic leukemias. Heterozygous loss-of-function mutations of the RUNX1 gene in the germline cause a rare autosomal dominant disorder called familial platelet disorder with propensity to acute myelogenous leukemia (FPD/AML). Besides the preexisting platelet disorder in FPD/AML patients, AML also develops in 20-60% of affected individuals. Since its discovery by the Gilliland group in 1999, RUNX1 mutation in the germline has been extensively investigated in the field. The past two decades of research have taught us three important lessons: 1) patients with FPD/AML display atypical symptoms and they have a widened clinical spectrum of FPD, such as eczema and syndromic thrombocytopenia, 2) the elucidation of variant of uncertain significance (VUS) of RUNX1 have revealed their role in epigenetic functions and involvement in the Fanconi anemia DNA repair pathway, and 3) non-coding mutations of RUNX1 also causes FPD/AML. In three distinct familial cases, an enhancer for RUNX1, eR1, was either lost or disconnected from the promoter through genetic deletion or chromosomal translocation abnormalities. This experience, with congenital mutations of RUNX1, will be very useful for future research for a series of other leukemia-causing germline mutations that have been recently identified.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda , Células Germinativas , Mutación de Línea Germinal , Humanos , Leucemia Mieloide Aguda/genética , Mutación , Linaje
16.
J Cell Mol Med ; 23(8): 5246-5258, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31119862

RESUMEN

AML1-ETO, the most common fusion oncoprotein by t (8;21) in acute myeloid leukaemia (AML), enhances hematopoietic self-renewal and leukemogenesis. However, currently no specific therapies have been reported for t (8;21) AML patients as AML1-ETO is still intractable as a pharmacological target. For this purpose, leukaemia cells and AML1-ETO-induced murine leukaemia model were used to investigate the degradation of AML1-ETO by melatonin (MLT), synthesized and secreted by the pineal gland. MLT remarkedly decreased AML1-ETO protein in leukemic cells. Meanwhile, MLT induced apoptosis, decreased proliferation and reduced colony formation. Furthermore, MLT reduced the expansion of human leukemic cells and extended the overall survival in U937T-AML1-ETO-xenografted NSG mice. Most importantly, MLT reduced the infiltration of leukaemia blasts, decreased the frequency of leukaemia stem cells (LSCs) and prolonged the overall survival in AML1-ETO-induced murine leukaemia. Mechanistically, MLT increased the expression of miR-193a, which inhibited AML1-ETO expression via targeting its putative binding sites. Furthermore, MLT decreased the expression of ß-catenin, which is required for the self-renewal of LSC and is the downstream of AML1-ETO. Thus, MLT presents anti-self-renewal of LSC through miR-193a-AML1-ETO-ß-catenin axis. In conclusion, MLT might be a potential treatment for t (8;21) leukaemia by targeting AML1-ETO oncoprotein.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia/tratamiento farmacológico , Melatonina/farmacología , MicroARNs/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/antagonistas & inhibidores , Modelos Animales de Enfermedad , Humanos , Leucemia/genética , Leucemia/patología , Ratones , MicroARNs/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1/antagonistas & inhibidores , Translocación Genética/efectos de los fármacos , beta Catenina/genética
17.
J Cell Biochem ; 120(5): 7309-7322, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30417424

RESUMEN

Inhibitors of the bromodomain and extraterminal domain family (BETi) offer a new approach to treat hematological malignancies, with leukemias containing mixed lineage leukemia rearrangements being especially sensitive due to a reliance on the regulation of transcription elongation. We explored the mechanism of action of BETi in cells expressing the t(8;21), and show that these compounds reduced the size of acute myeloid leukemia cells, triggered a rapid but reversible G0 /G1 arrest, and with time, cause cell death. Meta-analysis of PRO-seq data identified ribosomal genes, which are regulated by MYC, were downregulated within 3 hours of addition of the BETi. This reduction of MYC regulated metabolic genes coincided with the loss of mitochondrial respiration and large reductions in the glycolytic rate. In addition, gene expression analysis showed that transcription of BCL2 was rapidly affected by BETi but this did not cause dramatic increases in cell death. Cell cycle arrest, lowered metabolic activity, and reduced BCL2 levels suggested that a second compound was needed to push these cells over the apoptotic threshold. Indeed, low doses of the BCL2 inhibitor, venetoclax, in combination with the BETi was a potent combination in t(8;21) containing cells. Thus, BET inhibitors that affect MYC and BCL2 expression should be considered for combination therapy with venetoclax.

18.
Int J Mol Sci ; 20(2)2019 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-30654457

RESUMEN

Oncogenic fusion protein RUNX1-ETO is the product of the t(8;21) translocation, responsible for the most common cytogenetic subtype of acute myeloid leukemia. RUNX1, a critical transcription factor in hematopoietic development, is fused with almost the entire ETO sequence with the ability to recruit a wide range of repressors. Past efforts in providing a comprehensive picture of the genome-wide localization and the target genes of RUNX1-ETO have been inconclusive in understanding the underlying mechanism by which it deregulates native RUNX1. In this review; we dissect the current data on the epigenetic impact of RUNX1 and RUNX1-ETO. Both share similarities however, in recent years, research focused on epigenetic factors to explain their differences. RUNX1-ETO impairs DNA repair mechanisms which compromises genomic stability and favors a mutator phenotype. Among an increasing pool of mutated factors, regulators of DNA methylation are frequently found in t(8;21) AML. Together with the alteration of both, histone markers and distal enhancer regulation, RUNX1-ETO might specifically disrupt normal chromatin structure. Epigenetic studies on the fusion protein uncovered new mechanisms contributing to leukemogenesis and hopefully will translate into clinical applications.


Asunto(s)
Epigénesis Genética , Inestabilidad Genómica , Leucemia/genética , Proteínas de Fusión Oncogénica/genética , Animales , Cromatina/metabolismo , Metilación de ADN/genética , Humanos , Proteínas de Fusión Oncogénica/química
19.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(1): 109-114, 2019 Jan.
Artículo en Zh | MEDLINE | ID: mdl-31037912

RESUMEN

OBJECTIVE: To investigate the mRNA expression of galectin-3 and its clinical significance in acute myeloid leukemia (AML) patients carrying AML1/ETOfusion gene. METHODS: RQ-PCR method was used to detect the expression of galectin-3 mRNA in bone marrow mononuclear cells of 53 AML patients with AML1/ETO+, ELISA was used to detect the expression of galectin-3 protein in peripheral blood, and the correlations of galectin-3 expression with clinical and laboratory features and outcomes were analyzed. RESULTS: The mRNA and protein levels of galectin-3 were significantly higher in newly diagnosed AML1/ETO+ AML patients compared with the control ( P<0.001). Galectin-3 mRNA and protein expressions were positively correlated (r=0.732, P<0.001). Galectin-3 protein was significantly decreased during the period of complete remission (CR)( P<0.001). The mRNA expression of galectin-3 was negatively correlated with the count of white blood cells ( P=0.014), and positively correlated with CD34 expression and additional cytogenetic aberrations (ACA) ( P=0.001, P=0.026). There was no significant difference in CR, partial remission (PR), induction death (early mortality) between galectin-3 high-expression group and low-expression group ( P>0.05), but there was significant difference in recurrence rate between the two groups ( P=0.029). The median overall survival (OS) rate and disease-free survival (DFS) rate were shortened in the high-expression group ( P=0.007, P=0.015) and the cumulative incidence of relapse was increased ( P=0.045), but there was no significant difference in the cumulative incidence of CM(155mm]mortality ( P>0.05). Cox regression analysis suggested galectin-3 mRNA level an independent indicator of OS and DFS in AML1/ETO+ AML patients. CONCLUSION: Bone marrow galectin-3 mRNA level may be an important reference index for evaluating the prognosis and guiding the treatment of AML1/ETO+ AML patients.


Asunto(s)
Leucemia Mieloide Aguda , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Galectina 3 , Humanos , Proteínas de Fusión Oncogénica , Proteína 1 Compañera de Translocación de RUNX1
20.
J Cell Biochem ; 119(4): 3706-3715, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29236325

RESUMEN

The AML1/ETO onco-fusion protein is crucial for the genesis of t(8;21) acute myeloid leukemia (AML) and is well documented as a transcriptional repressor through dominant-negative effect. However, little is known about the transactivation mechanism of AML1/ETO. Through large cohort of patient's expression level data analysis and a series of experimental validation, we report here that AML1/ETO transactivates c-KIT expression through directly binding to and mediating the long-range interaction between the promoter and intronic enhancer regions of c-KIT. Gene expression analyses verify that c-KIT expression is significantly high in t(8;21) AML. Further ChIP-seq analysis and motif scanning identify two regulatory regions located in the promoter and intronic enhancer region of c-KIT, respectively. Both regions are enriched by co-factors of AML1/ETO, such as AML1, CEBPe, c-Jun, and c-Fos. Further luciferase reporter assays show that AML1/ETO trans-activates c-KIT promoter activity through directly recognizing the AML1 motif and the co-existence of co-factors. The induction of c-KIT promoter activity is reinforced with the existence of intronic enhancer region. Furthermore, ChIP-3C-qPCR assays verify that AML1/ETO mediates the formation of DNA-looping between the c-KIT promoter and intronic enhancer region through the long-range interaction. Collectively, our data uncover a novel transcriptional activity mechanism of AML1/ETO and enrich our knowledge of the onco-fusion protein mediated transcription regulation.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Regiones Promotoras Genéticas/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Humanos , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/genética , Interferencia de ARN , Proteína 1 Compañera de Translocación de RUNX1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA