Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Cell Death Dis ; 7: e2308, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27468685

RESUMEN

Proteins involved in DNA double-strand break (DSB) repair localize within the promyelocytic leukemia nuclear bodies (PML-NBs), whose disruption is at the root of the acute promyelocytic leukemia (APL) pathogenesis. All-trans-retinoic acid (RA) treatment induces PML-RARα degradation, restores PML-NB functions, and causes terminal cell differentiation of APL blasts. However, the precise role of the APL-associated PML-RARα oncoprotein and PML-NB integrity in the DSB response in APL leukemogenesis and tumor suppression is still lacking. Primary leukemia blasts isolated from APL patients showed high phosphorylation levels of H2AX (γ-H2AX), an initial DSBs sensor. By addressing the consequences of ionizing radiation (IR)-induced DSB response in primary APL blasts and RA-responsive and -resistant myeloid cell lines carrying endogenous or ectopically expressed PML-RARα, before and after treatment with RA, we found that the disruption of PML-NBs is associated with delayed DSB response, as revealed by the impaired kinetic of disappearance of γ-H2AX and 53BP1 foci and activation of ATM and of its substrates H2AX, NBN, and CHK2. The disruption of PML-NB integrity by PML-RARα also affects the IR-induced DSB response in a preleukemic mouse model of APL in vivo. We propose the oncoprotein-dependent PML-NB disruption and DDR impairment as relevant early events in APL tumorigenesis.


Asunto(s)
Núcleo Celular/metabolismo , ADN/metabolismo , Regulación Leucémica de la Expresión Génica , Células Precursoras de Granulocitos/metabolismo , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de la radiación , Núcleo Celular/ultraestructura , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , ADN/genética , Roturas del ADN de Doble Cadena/efectos de la radiación , Modelos Animales de Enfermedad , Rayos gamma , Células Precursoras de Granulocitos/efectos de los fármacos , Células Precursoras de Granulocitos/patología , Células Precursoras de Granulocitos/efectos de la radiación , Histonas/genética , Histonas/metabolismo , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Transducción de Señal , Tretinoina/farmacología , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
2.
Leukemia ; 29(8): 1730-40, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25727291

RESUMEN

The mechanisms by which AML1/ETO (A/E) fusion protein induces leukemogenesis in acute myeloid leukemia (AML) without mutagenic events remain elusive. Here we show that interactions between A/E and hypoxia-inducible factor 1α (HIF1α) are sufficient to prime leukemia cells for subsequent aggressive growth. In agreement with this, HIF1α is highly expressed in A/E-positive AML patients and strongly predicts inferior outcomes, regardless of gene mutations. Co-expression of A/E and HIF1α in leukemia cells causes a higher cell proliferation rate in vitro and more serious leukemic status in mice. Mechanistically, A/E and HIF1α form a positive regulatory circuit and cooperate to transactivate DNMT3a gene leading to DNA hypermethylation. Pharmacological or genetic interventions in the A/E-HIF1α loop results in DNA hypomethylation, a re-expression of hypermethylated tumor-suppressor p15(INK4b) and the blockage of leukemia growth. Thus high HIF1α expression serves as a reliable marker, which identifies patients with a poor prognosis in an otherwise prognostically favorable AML group and represents an innovative therapeutic target in high-risk A/E-driven leukemia.


Asunto(s)
Transformación Celular Neoplásica/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Animales , Apoptosis , Western Blotting , Proliferación Celular , Inmunoprecipitación de Cromatina , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , ADN Metiltransferasa 3A , Citometría de Flujo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Técnicas para Inmunoenzimas , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Proteína 1 Compañera de Translocación de RUNX1 , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Death Differ ; 21(2): 290-301, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24141720

RESUMEN

MicroRNAs (miRNAs) regulate cell proliferation, differentiation and death during development and postnatal life. The expression level of mature miRNAs results from complex molecular mechanisms, including the transcriptional regulation of their genes. MiR-223 is a hematopoietic-specific miRNA participating in regulatory signaling networks involving lineage-specific transcription factors (TFs). However, the transcriptional mechanisms governing its expression levels and its functional role in lineage fate decision of human hematopoietic progenitors (HPCs) have not yet been clarified. We found that in CD34(+)HPCs undergoing unilineage differentiation/maturation, miR-223 is upregulated more than 10-fold during granulopoiesis, 3-fold during monocytopoiesis and maintained at low levels during erythropoiesis. Chromatin immunoprecipitation and promoter luciferase assays showed that the lineage-specific expression level of mature miR-223 is controlled by the coordinated binding of TFs to their DNA-responsive elements located in 'distal' and 'proximal' regulatory regions of the miR-223 gene, differentially regulating the transcription of two primary transcripts (pri-miRs). All this drives myeloid progenitor maturation into specific lineages. Accordingly, modulation of miR-223 activity in CD34(+)HPCs and myeloid cell lines significantly affects their differentiation/maturation into erythroid, granulocytic and monocytic/macrophagic lineages. MiR-223 overexpression increases granulopoiesis and impairs erythroid and monocytic/macrophagic differentiation. Its knockdown, meanwhile, impairs granulopoiesis and facilitates erythropoiesis and monocytic/macrophagic differentiation. Overall, our data reveal that transcriptional pathways acting on the differential regulation of two pri-miR transcripts results in the fine-tuning of a single mature miRNA expression level, which dictates the lineage fate decision of hematopoietic myeloid progenitors.


Asunto(s)
Linaje de la Célula/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Transcripción Genética/genética , Activación Transcripcional , Antígenos CD34/metabolismo , Humanos
4.
Cell Death Dis ; 4: e926, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24263100

RESUMEN

MicroRNAs are key regulators of many biological processes, including cell differentiation. These small RNAs exert their function assembled in the RNA-induced silencing complexes (RISCs), where members of Argonaute (Ago) family of proteins provide a unique platform for target recognition and gene silencing. Here, by using myeloid cell lines and primary blasts, we show that Ago2 has a key role in human monocytic cell fate determination and in LPS-induced inflammatory response of 1,25-dihydroxyvitamin D3 (D3)-treated myeloid cells. The silencing of Ago2 impairs the D3-dependent miR-17-5p/20a/106a, miR-125b and miR-155 downregulation, the accumulation of their translational targets AML1, VDR and C/EBPß and monocytic cell differentiation. Moreover, we show that Ago2 is recruited on miR-155 host gene promoter and on the upstream region of an overlapping antisense lncRNA, determining their epigenetic silencing, and miR-155 downregulation. These findings highlight Ago2 as a new factor in myeloid cell fate determination in acute myeloid leukemia cells.


Asunto(s)
Proteínas Argonautas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Argonautas/genética , Western Blotting , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Calcitriol/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células HL-60 , Humanos , Leucemia Mieloide Aguda/genética , MicroARNs/genética , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo
7.
Leukemia ; 22(8): 1503-18, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18548105

RESUMEN

During embryonic development and adult life, the plasticity and reversibility of modifications that affect the chromatin structure is important in the expression of genes involved in cell fate decisions and the maintenance of cell-differentiated state. Epigenetic changes in DNA and chromatin, which must occur to allow the accessibility of transcriptional factors at specific DNA-binding sites, are regarded as emerging major players for embryonic and hematopoietic stem cell (HSC) development and lineage differentiation. Epigenetic deregulation of gene expression, whether it be in conjunction with chromosomal alterations and gene mutations or not, is a newly recognized mechanism that leads to several diseases, including leukemia. The reversibility of epigenetic modifications makes DNA and chromatin changes attractive targets for therapeutic intervention. Here we review some of the epigenetic mechanisms that regulate gene expression in pluripotent embryonic and multipotent HSCs but may be deregulated in leukemia, and the clinical approaches designed to target the chromatin structure in leukemic cells.


Asunto(s)
Linaje de la Célula , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Células Madre Hematopoyéticas/metabolismo , Metilación de ADN , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Humanos , Proteínas del Grupo Polycomb , Proteínas Represoras/metabolismo
8.
Curr Top Microbiol Immunol ; 313: 73-84, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17217039

RESUMEN

Hematopoiesis is highly controlled by lineage-specific transcription factors that, by interacting with specific DNA sequences, directly activate or repress specific gene expression. These transcription factors have been found mutated or altered by chromosomal translocations associated with leukemias, indicating their role in the pathogenesis of these malignancies. The post-genomic era, however, has shown that transcription factors are not the only key regulators of gene expression. Epigenetic mechanisms such as DNA methylation, posttranslational modifications of histones, remodeling of nucleosomes, and expression of small regulatory RNAs all contribute to the regulation of gene expression and determination of cell and tissue specificity. Deregulation ofthese epigenetic mechanisms cooperates with genetic alterations to the establishment and progression of tumors. MicroRNAs (miRNAs) are negative regulators of the expression of genes involved in development, differentiation, proliferation, and apoptosis. Their expression appears to be tissue-specific and highly regulated according to the cell's developmental lineage and stage. Interestingly, miRNAs expressed in hematopoietic cells have been found mutated or altered by chromosomal translocations associated with leukemias. The expression levels of a specific miR-223 correlate with the differentiation fate of myeloid precursors. The activation of both pathways of transcriptional regulation by the myeloid lineage-specific transcription factor C/EBPalpha (CCAAT/enhancer-binding protein-alpha), and posttranscriptional regulation by miR-223 appears essential for granulocytic differentiation and clinical response of acute promyelocytic leukemia (APL) blasts to all-trans retinoic acid (ATRA). Together, this evidence underlies transcription factors, chromatin remodeling, and miRNAs as ultimate determinants for the correct organization of cell type-specific gene arrays and hematopoietic differentiation, therefore providing new targets for the diagnosis and treatment of leukemias.


Asunto(s)
Leucemia Promielocítica Aguda/fisiopatología , MicroARNs/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Hematopoyesis/genética , Hematopoyesis/fisiología , Humanos , Leucemia Promielocítica Aguda/genética , MicroARNs/genética , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-17381298

RESUMEN

The discovery of microRNAS (miRNAs) and of their mechanism of action has provided some very new clues on how gene expression is regulated. These studies established new concepts on how posttranscriptional control can fine-tune gene expression during differentiation and allowed the identification of new regulatory circuitries as well as factors involved therein. Because of the wealth of information available about the transcriptional and cellular networks involved in hematopoietic differentiation, the hematopoietic system is ideal for studying cell lineage specification. An interesting interplay between miRNAs and lineage-specific transcriptional factors has been found, and this can help us to understand how terminal differentiation is accomplished.


Asunto(s)
Hematopoyesis/genética , Hematopoyesis/fisiología , MicroARNs/genética , MicroARNs/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Humanos , Leucemia/genética , Leucemia/metabolismo , Modelos Biológicos , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Factores de Transcripción/metabolismo
10.
EMBO J ; 20(24): 6969-78, 2001 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-11742974

RESUMEN

Histone deacetylases (HDACs) play important roles in transcriptional regulation and pathogenesis of cancer. Thus, HDAC inhibitors are candidate drugs for differentiation therapy of cancer. Here, we show that the well-tolerated antiepileptic drug valproic acid is a powerful HDAC inhibitor. Valproic acid relieves HDAC-dependent transcriptional repression and causes hyperacetylation of histones in cultured cells and in vivo. Valproic acid inhibits HDAC activity in vitro, most probably by binding to the catalytic center of HDACs. Most importantly, valproic acid induces differentiation of carcinoma cells, transformed hematopoietic progenitor cells and leukemic blasts from acute myeloid leukemia patients. More over, tumor growth and metastasis formation are significantly reduced in animal experiments. Therefore, valproic acid might serve as an effective drug for cancer therapy.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Ácido Valproico/farmacología , Animales , Línea Celular Transformada , Cricetinae , Humanos , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Glucocorticoides/genética , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos
11.
Oncogene ; 20(24): 3110-5, 2001 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-11420727

RESUMEN

Recent discoveries have identified key molecular events in the pathogenesis of acute promyelocytic leukemia (APL), caused by chromosomal rearrangements of the transcription factor RAR (resulting in a fusion protein with the product of other cellular genes, such as PML). Oligomerization of RAR, through a self-association domain present in PML, imposes an altered interaction with transcriptional co-regulators (NCoR/SMRT). NCoR/SMRT are responsible for recruitment of histone deacetylases (HDACs), which is required for transcriptional repression of PML-RAR target genes, and for the transforming potential of the fusion protein. Oligomerization and altered recruitment of HDACs are also responsible for transformation by the fusion protein AML1-ETO, extending these mechanisms to other forms of acute myeloid leukemias (AMLs) and suggesting that HDAC is a common target for myeloid leukemias. Strikingly, AML1-ETO expression blocks retinoic acid (RA) signaling in hematopoietic cells, suggesting that interference with the RA pathway (genetically altered in APL) by HDAC recruitment may be a common theme in AMLs. Treatment of APLs with RA, and of other AMLs with RA plus HDAC inhibitors (HDACi), results in myeloid differentiation. Thus, activation of the RA signaling pathway and inhibition of HDAC activity might represent a general strategy for the differentiation treatment of myeloid leukemias.


Asunto(s)
Histona Desacetilasas/metabolismo , Enfermedad Aguda , Animales , Diferenciación Celular/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Inhibidores de Histona Desacetilasas , Humanos , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/enzimología , Leucemia Mieloide/patología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/enzimología , Leucemia Promielocítica Aguda/patología , Tretinoina/uso terapéutico
12.
Cancer Res ; 61(4): 1247-9, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11245412

RESUMEN

Remodeling of the chromatin template by inhibition of histone deacetylase (HDAC) activities represents a major goal for transcriptional therapy in neoplastic diseases. Recently, a number of specific and potent HDAC-inhibitors that modulate in vitro cell growth and differentiation have been developed. In this study we analyzed the effect of trichostatin A (TSA), a specific and potent HDAC-inhibitor, on mouse embryos developing in vivo. When administered i.p. to pregnant mice (at a concentration of 0.5-1 mg/kg) at postimplantation stages (embryonic day 8 to embryonic day 10), TSA was not toxic for the mother and did not cause any obvious malformation during somitogenesis or at later stages of development. Treated embryos were born at similar frequency and were indistinguishable from control animals, developed normally, and were fertile. Interestingly, embryos from TSA-treated mice killed during somitogenesis were modestly but consistently larger than control embryos and presented an increased (+2 to +6) number of somites. This correlated with an increased acetylation of histone H4, the number of somites expressing the myogenic factor Myf-5, and the expression of Notch, RARalpha2, and RARbeta2 mRNAs. These data indicate that the effects of TSA on transcription: (a) are not toxic for the mother; (b) transiently accelerated growth in mouse embryos without perturbing embryogenesis; and (c) do not result in teratogenesis, at least in rodents. Thus, TSA might represent a nontoxic and effective agent for the transcriptional therapy of neoplasia.


Asunto(s)
Desarrollo Embrionario y Fetal/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/toxicidad , Animales , Desarrollo Embrionario y Fetal/genética , Femenino , Crecimiento/efectos de los fármacos , Ratones , Embarazo
13.
Cancer Res ; 61(1): 2-7, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11196162

RESUMEN

Histone deacetylase (HDAC)-dependent transcriptional repression of the retinoic acid (RA)-signaling pathway underlies the differentiation block of acute promyelocytic leukemia. RA treatment relieves transcriptional repression and triggers differentiation of acute promyelocytic leukemia blasts, leading to disease remission. We report that transcriptional repression of RA signaling is a common mechanism in acute myeloid leukemias (AMLs). HDAC inhibitors restored RA-dependent transcriptional activation and triggered terminal differentiation of primary blasts from 23 AML patients. Accordingly, we show that AML1/ETO, the commonest AML-associated fusion protein, is an HDAC-dependent repressor of RA signaling. These findings relate alteration of the RA pathway to myeloid leukemogenesis and underscore the potential of transcriptional/differentiation therapy in AML.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Leucemia Mielomonocítica Aguda/enzimología , Leucemia Mielomonocítica Aguda/patología , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Acetilación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Inhibidores Enzimáticos/farmacología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/fisiología , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Aguda/genética , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1 , Transducción de Señal/fisiología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología , Tretinoina/fisiología , Células Tumorales Cultivadas
14.
Hematol J ; 2(6): 385-92, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11920278

RESUMEN

INTRODUCTION: Acute promyelocytic leukemia (APL) patients with t(15;17)(PML/RARalpha positive) achieve remission upon t-RA treatment, whereas patients with t(11;17)(PLZF/RARalpha positive) do not. Both APL translocation products bind to the histone deacetylase (HD)-recruiting nuclear co-repressor complex (HD-NCR) in a ligand-dependent manner through their RARalpha portion. Differently to PML/RARalpha, PLZF/RARalpha also binds the HD-NCR in a ligand-independent manner through the PLZF portion of the fusion protein (PLZF#), which seems to be crucial for the t-RA resistance of t(11;17) APL patients. MATERIALS AND METHODS: The t-RA sensitivity of U937 cells was tested by the nitro-blue tetrazolium reduction (NBT) assay and by analysis of t-RA-induced type II transglutaminase activity. The interaction between HD-NCR and PLZF/RARalpha was investigated by in vitro binding assays. RESULTS: (i) Deletions in PLZF# convert PLZF/RARalpha from a repressor to an activator of t-RA response in U937 cells; (ii) the effect of PLZF/RARalpha on t-RA-signaling is regulated by the POZ-domain and its down-stream regions of PLZF#; (iii) there are additional binding sites for HD-NCR in PLZF# and (iv) PLZF# not only directly binds but also regulates the binding of PLZF/RARalpha to the HD-NCR. CONCLUSIONS: At least two different mechanisms responsible for the aberrant recruitment of HD-NCR complexes by PLZF# are regulating the different t-RA-sensitivity of the PLZF/RARalpha and PML/RARalpha positive APL blasts: one is related to the direct binding of the different members of the HD-NCR complex to PLZF#; the other is an enforcing effect of PLZF# on the affinity of the PLZF/RARalpha fusion protein to the HD-NCR complex.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas Humanos Par 17 , Proteínas de Unión al ADN/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Translocación Genética , Proteínas de Unión al ADN/fisiología , Resistencia a Antineoplásicos , Histona Desacetilasas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel , Leucemia Promielocítica Aguda/etiología , Leucemia Promielocítica Aguda/genética , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/farmacología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/farmacología , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Unión Proteica , Proteínas Represoras/fisiología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/fisiología , Tretinoina/farmacología , Células U937 , Dedos de Zinc/genética
15.
Blood ; 96(9): 3200-8, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11050004

RESUMEN

Acute promyelocytic leukemia (APL) is characterized by a specific translocation, t(15;17), that fuses the promyelocytic leukemia (PML) gene with the RA receptor RARalpha. Pharmacologic doses of retinoic acid (RA) induce differentiation in human APL cells and complete clinical remissions. Unfortunately, APL cells develop resistance to RA in vitro and in vivo. Recently, mutations in PML/RARalpha have been described in APL cells from patients clinically resistant to RA therapy. The mutations cluster in 2 regions that are involved in forming the binding pocket for RA. These mutant PML/RARalpha proteins have been expressed in vitro, which shows that they cause a diversity of alterations in binding to ligand and to nuclear coregulators of transcription, leading to varying degrees of inhibition of retinoid-induced transcription. This contrasts with the nearly complete dominant negative activity of mutations in PML/RARalpha previously characterized in cell lines developing RA resistance in vitro. Current data from this study provide additional insight into the molecular mechanisms of resistance to RA and suggest that alterations in the ability of mutants to interact with coregulators can be determinant in the molecular mechanism of resistance to RA. In particular, ligand-induced binding to the coactivator ACTR correlated better with transcriptional activation of RA response elements than the ligand-induced release of the corepressor SMRT. The diversity of effects that are seen in patient-derived mutations may help explain the partial success to date of attempts to overcome this mechanism of resistance in patients by the clinical use of histone deacetylase inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Transcripción Genética , Tretinoina/uso terapéutico , Sustitución de Aminoácidos , Sitios de Unión , Cromosomas Humanos Par 15 , Cromosomas Humanos Par 17 , Clonación Molecular , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Modelos Moleculares , Mutación Puntual , Estructura Secundaria de Proteína , Translocación Genética , Células Tumorales Cultivadas
16.
Mol Cell ; 5(5): 811-20, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10882117

RESUMEN

RAR and AML1 transcription factors are found in leukemias as fusion proteins with PML and ETO, respectively. Association of PML-RAR and AML1-ETO with the nuclear corepressor (N-CoR)/histone deacetylase (HDAC) complex is required to block hematopoietic differentiation. We show that PML-RAR and AML1-ETO exist in vivo within high molecular weight (HMW) nuclear complexes, reflecting their oligomeric state. Oligomerization requires PML or ETO coiled-coil regions and is responsible for abnormal recruitment of N-CoR, transcriptional repression, and impaired differentiation of primary hematopoietic precursors. Fusion of RAR to a heterologous oligomerization domain recapitulated the properties of PML-RAR, indicating that oligomerization per se is sufficient to achieve transforming potential. These results show that oligomerization of a transcription factor, imposing an altered interaction with transcriptional coregulators, represents a novel mechanism of oncogenic activation.


Asunto(s)
Transformación Celular Neoplásica , Leucemia/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Factores de Transcripción/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Histona Desacetilasas/metabolismo , Humanos , Leucemia/etiología , Leucemia Mieloide/etiología , Leucemia Mieloide/genética , Leucemia Promielocítica Aguda/etiología , Leucemia Promielocítica Aguda/genética , Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Represoras/metabolismo , Elementos de Respuesta , Transcripción Genética , Tretinoina
17.
Oncogene ; 18(46): 6313-21, 1999 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-10597230

RESUMEN

Retinoic Acid (RA) treatment induces disease remission of Acute Promyelocytic Leukaemia (APL) patients by triggering terminal differentiation of neoplastic cells. RA-sensitivity in APL is mediated by its oncogenic protein, which results from the recombination of the PML and the RA receptor alpha (RAR alpha) genes (PML/RAR alpha fusion protein). Ectopic expression of PML/RAR alpha into haemopoietic cell lines results in increased response to RA-induced differentiation. By structure-function analysis of PML/RAR alpha-mediated RA-differentiation, we demonstrated that fusion of PML and RAR alpha sequences and integrity of the PML dimerization domain and of the RAR alpha DNA binding region are required for the effect of PML/RAR alpha on RA-differentiation. Indeed, direct fusion of the PML dimerization domain to the N- or C-terminal extremities of RAR alpha retained full biological activity. All the biologically active PML/RAR alpha mutants formed high molecular weight complexes in vivo. Functional analysis of mutations within the PML dimerization domain revealed that the capacity to form PML/RAR alpha homodimers, but not PML/RAR alpha-PML heterodimers, correlated with the RA-response. These results suggest that targeting of RAR alpha sequences by the PML dimerization domain and formation of nuclear PML/RAR alpha homodimeric complexes are crucial for the ability of PML/RAR alpha to mediate RA-response.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias/química , Proteínas de Fusión Oncogénica/química , Tretinoina/farmacología , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Dimerización , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Peso Molecular , Proteínas de Neoplasias/fisiología , Proteínas de Fusión Oncogénica/fisiología , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Células U937/efectos de los fármacos , Células U937/metabolismo , Dedos de Zinc
19.
Blood ; 93(5): 1477-81, 1999 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-10029573

RESUMEN

PML/RARalpha is the leukemogenetic protein of acute promyelocytic leukemia (APL). Treatment with retinoic acid (RA) induces degradation of PML/RARalpha, differentiation of leukaemic blasts, and disease remission. However, RA resistance arises during RA treatment of APL patients. To investigate the phenomenon of RA resistance in APL, we generated RA-resistant sublines from APL-derived NB4 cells. The NB4.007/6 RA-resistant subline does not express the PML/RARalpha protein, although its mRNA is detectable at levels comparable to those of the parental cell line. In vitro degradation assays showed that the half-life of PML/RARalpha is less than 30 minutes in NB4.007/6 and longer than 3 hours in NB4. Treatment of NB4.007/6 cells with the proteasome inhibitors LLnL and lactacystin partially restored PML/RARalpha protein expression and resulted in a partial release of the RA-resistant phenotype. Similarly, forced expression of PML/RARalpha, but not RARalpha, into the NB4/007.6 cells restored sensitivity to RA treatment to levels comparable to those of the NB4 cells. These results indicate that constitutive degradation of PML/RARalpha protein may lead to RA resistance and that PML/RARalpha expression is crucial to convey RA sensitivity to APL cells.


Asunto(s)
Antineoplásicos/farmacología , Cisteína Endopeptidasas/metabolismo , Resistencia a Antineoplásicos , Leucemia Promielocítica Aguda/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Tretinoina/farmacología , Humanos , Complejo de la Endopetidasa Proteasomal
20.
Leukemia ; 12(12): 1866-80, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9844917

RESUMEN

The Second International Symposium on Acute Promyelocytic Leukemia (APL) was held in Rome in 12-14 November 1997. Clinical and basic investigators had the opportunity to discuss in this meeting the important advances in the biology and treatment of this disease achieved in the last 4 years, since the First Roman Symposium was held in 1993. The first part of the meeting was dedicated to relevant aspects of laboratory research, and included the following topics: molecular mechanisms of leukemogenesis and of response/resistance to retinoids, biologic and therapeutic effects of new agents such as arsenicals and novel synthetic retinoids; characterization of APL heterogeneity at the morphological, cytogenetic and immunophenotypic level. The updated results of large cooperative clinical trials using variable combinations of all-trans retinoic acid (ATRA) and chemotherapy were presented by the respective group chairmen, and formed the 'core' part of the meeting. These studies, which in most cases integrated the molecular assessment of response to treatment, provided a stimulating framework for an intense debate on the most appropriate frontline treatment options to be adopted in the future. The last day was dedicated to special entities such as APL in the elderly and in the child, as well as the role of bone marrow transplantation. The prognostic value of molecular monitoring studies was also discussed in the final session of the meeting. In this article, we review the major advances and controversial issues in APL biology and treatment discussed in this symposium and emerging from very recent publications. We would like to credit the successful outcome of this meeting to the active and generous input of all invited speakers and to participants from all over the world who provided constructive and fruitful discussions.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia Promielocítica Aguda/terapia , Proteínas Nucleares , Tretinoina/uso terapéutico , Anciano , Trióxido de Arsénico , Arsenicales/uso terapéutico , Niño , Resistencia a Medicamentos , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunofenotipificación , Cariotipificación , Leucemia Promielocítica Aguda/diagnóstico , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasia Residual , Óxidos/uso terapéutico , Proteína de la Leucemia Promielocítica , Recurrencia , Investigación , Ciudad de Roma , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA