Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 17(8): e1009693, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34351909

RESUMEN

The ubiquitin-proteasome system plays important roles in various biological processes as it degrades the majority of cellular proteins. Adequate proteasomal degradation of crucial transcription regulators ensures the proper development of neutrophils. The ubiquitin E3 ligase of Growth factor independent 1 (GFI1), a key transcription repressor governing terminal granulopoiesis, remains obscure. Here we report that the deficiency of the ring finger protein Interferon regulatory factor 2 binding protein 2a (Irf2bp2a) leads to an impairment of neutrophils differentiation in zebrafish. Mechanistically, Irf2bp2a functions as a ubiquitin E3 ligase targeting Gfi1aa for proteasomal degradation. Moreover, irf2bp2a gene is repressed by Gfi1aa, thus forming a negative feedback loop between Irf2bp2a and Gfi1aa during neutrophils maturation. Different levels of GFI1 may turn it into a tumor suppressor or an oncogene in malignant myelopoiesis. Therefore, discovery of certain drug targets GFI1 for proteasomal degradation by IRF2BP2 might be an effective anti-cancer strategy.


Asunto(s)
Proteínas de Unión al ADN/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Proteínas de Unión al ADN/metabolismo , Retroalimentación Fisiológica , Femenino , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Células HEK293 , Células HL-60 , Humanos , Leucopoyesis , Masculino , Proteolisis , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
2.
Blood ; 137(18): 2429-2437, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33651885

RESUMEN

The retinoic acid receptors (RARA, RARB, and RARG) are ligand-regulated nuclear receptors that act as transcriptional switches. These master genes drew significant interest in the 1990s because of their key roles in embryogenesis and involvement in a rare malignancy, acute promyelocytic leukemia (APL), in which the RARA (and very rarely, RARG or RARB) genes are rearranged, underscoring the central role of deregulated retinoid signaling in leukemogenesis. Several recent provocative observations have revived interest in the roles of retinoids in non-APL acute myeloid leukemia (AML), as well as in normal hematopoietic differentiation. We review the role of retinoids in hematopoiesis, as well as in the treatment of non-APL AMLs. From this perspective, broader uses of retinoids in the management of hematopoietic tumors are discussed.


Asunto(s)
Hematopoyesis , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/metabolismo , Retinoides/uso terapéutico , Animales , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología
3.
Blood ; 133(13): 1495-1506, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30674471

RESUMEN

Acute promyelocytic leukemia (APL) is often associated with activating FLT3 signaling mutations. These are highly related to hyperleukocytosis, a major adverse risk factor with chemotherapy-based regimens. APL is a model for oncogene-targeted therapies: all-trans retinoic acid (ATRA) and arsenic both target and degrade its ProMyelocytic Leukemia/Retinoic Acid Receptor α (PML/RARA) driver. The combined ATRA/arsenic regimen now cures virtually all patients with standard-risk APL. Although FLT3-internal tandem duplication (ITD) was an adverse risk factor for historical ATRA/chemotherapy regimens, the molecular bases for this effect remain unknown. Using mouse APL models, we unexpectedly demonstrate that FLT3-ITD severely blunts ATRA response. Remarkably, although the transcriptional output of initial ATRA response is unaffected, ATRA-induced PML/RARA degradation is blunted, as is PML nuclear body reformation and activation of P53 signaling. Critically, the combination of ATRA and arsenic fully rescues therapeutic response in FLT3-ITD APLs, restoring PML/RARA degradation, PML nuclear body reformation, P53 activation, and APL eradication. Moreover, arsenic targeting of normal PML also contributes to APL response in vivo. These unexpected results explain the less favorable outcome of FLT3-ITD APLs with ATRA-based regimens, and stress the key role of PML nuclear bodies in APL eradication by the ATRA/arsenic combination.


Asunto(s)
Antineoplásicos/uso terapéutico , Arsénico/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Tretinoina/uso terapéutico , Tirosina Quinasa 3 Similar a fms/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Femenino , Leucemia Promielocítica Aguda/genética , Ratones Endogámicos C57BL , Mutación
4.
Haematologica ; 106(5): 1443-1456, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33567810

RESUMEN

Adult T cell leukemia/lymphoma (ATL) is associated to chronic human T cell leukemia virus type 1 (HTLV-1) infection and carries a poor prognosis. Arsenic trioxide (AS) and interferon-alpha (IFNα) together selectively trigger Tax viral oncoprotein degradation and cure Tax-driven murine ATL. AS/IFNα/zidovudine treatment achieves a high response rate in patients with chronic ATL. Interleukin 10 (IL-10) is an immuno-suppressive cytokine whose expression is activated by Tax. Here we show that, in ATL, AS/IFNα-induced abrogation of leukemia initiating cell activity requires IL-10 expression shutoff. Loss of IL-10 secretion drives production of inflammatory cytokines by the microenvironment, followed by innate immunity-mediated clearance of Taxdriven leukemic cells. Accordingly, anti-IL-10 monoclonal antibodies significantly increased the efficiency of AS/IFNα therapy. These results emphasize the sequential targeting of malignant ATL cells and their immune microenvironment in leukemia initiating cell (LIC) eradication and provide a strong rational to test AS/IFNα/anti-IL10 combination in ATL.


Asunto(s)
Virus Linfotrópico T Tipo 1 Humano , Leucemia-Linfoma de Células T del Adulto , Adulto , Animales , Humanos , Inmunidad Innata , Interferón-alfa , Interleucina-10/genética , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Ratones , Microambiente Tumoral
5.
Haematologica ; 106(12): 3090-3099, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34047175

RESUMEN

Retinoic acid (RA) was proposed to increase survival of chemotherapy- treated patients with nucleophosmin-1 (NPM-1c)-mutated acute myeloid leukemia. We reported that, ex vivo, RA triggers NPM-1c degradation, P53 activation and growth arrest. PML organizes domains that control senescence or proteolysis. Here, we demonstrate that PML is required to initiate RA-driven NPM-1c degradation, P53 activation and cell death. Mechanistically, RA enhances PML basal expression through inhibition of activated Pin1, prior to NPM-1c degradation. Such PML induction drives P53 activation, favoring blast response to chemotherapy or arsenic in vivo. This RA/PML/P53 cascade could mechanistically explain RA-facilitated chemotherapy response in patients with NPM-1c mutated acute myeloid leukemia.


Asunto(s)
Leucemia Mieloide Aguda , Leucemia Promielocítica Aguda , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Tretinoina/farmacología , Tretinoina/uso terapéutico , Proteína p53 Supresora de Tumor/genética
6.
Retrovirology ; 17(1): 5, 2020 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-32199462

RESUMEN

BACKGROUND: Adult T-cell leukemia-lymphoma (ATL) is an aggressive mature lymphoid proliferation associated with poor prognosis. Standard of care includes chemotherapy and/or the combination of zidovudine and interferon-alpha. However, most patients experience relapse less than 6 months after diagnosis. Allogeneic stem cell transplantation is the only curative treatment, but is only feasible in a minority of cases. We previously showed in a mouse model that Arsenic trioxide (As2O3) targets ATL leukemia initiating cells. RESULTS: As2O3 consolidation was given in 9 patients with ATL (lymphoma n = 4; acute n = 2; and indolent n = 3), who were in complete (n = 4) and partial (n = 3) remission, in stable (n = 1) and in progressive (n = 1) disease. Patients received up to 8 weeks of As2O3 at the dose of 0.15 mg/kg/day intravenously in combination with zidovudine and interferon-alpha. One patient in progression died rapidly. Of the remaining eight patients, three with indolent ATL subtype showed overall survivals of 48, 53 and 97 months, and duration of response to As2O3 of 22, 25 and 73 months. The other 5 patients with aggressive ATL subtype had median OS of 36 months and a median duration of response of 10 months. Side effects were mostly hematological and cutaneous (one grade 3) and reversible with dose reduction of AZT/IFN and/or As2O3 discontinuation. The virus integration analysis revealed the regression of the predominant malignant clone in one patient with a chronic subtype. CONCLUSION: These results suggest that consolidation with As2O3 could be an option for patients with ATL in response after induction therapy and who are not eligible for allogeneic stem cell transplantation.


Asunto(s)
Trióxido de Arsénico/uso terapéutico , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Administración Intravenosa , Adulto , Trióxido de Arsénico/administración & dosificación , Quimioterapia Combinada , Femenino , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Humanos , Interferón-alfa/uso terapéutico , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Zidovudina/uso terapéutico
7.
Haematologica ; 105(2): 325-337, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31123027

RESUMEN

Aproper choice of neutrophil-macrophage progenitor cell fate is essential for the generation of adequate myeloid subpopulations during embryonic development and in adulthood. The network governing neutrophil-macrophage progenitor cell fate has several key determinants, such as myeloid master regulators CCAAT enhancer binding protein alpha (C/EBPα) and spleen focus forming virus proviral integration oncogene (PU.1). Nevertheless, more regulators remain to be identified and characterized. To ensure balanced commitment of neutrophil-macrophage progenitors toward each lineage, the interplay among these determinants is not only synergistic, but also antagonistic. Depletion of interferon regulatory factor 2 binding protein 2b (Irf2bp2b), a well-known negative transcription regulator, results in a bias in neutrophil-macrophage progenitor cell fate in favor of macrophages at the expense of neutrophils during the stage of definitive myelopoiesis in zebrafish embryos. Mechanistic studies indicate that Irf2bp2b acts as a downstream target of C/EBPα, repressing PU.1 expression, and that SUMOylation confers the repressive function of Irf2bp2b. Thus, Irf2bp2b is a novel determinant in the choice of fate of neutrophil-macrophage progenitor cells.


Asunto(s)
Proteínas de Unión al ADN/genética , Mielopoyesis , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Pez Cebra , Animales , Proteínas Portadoras , Diferenciación Celular , Macrófagos , Neutrófilos
9.
FASEB J ; 32(9): 4930-4940, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29634367

RESUMEN

RING finger protein 4 (RNF4) is a multifunctional small ubiquitin-related modifier (SUMO)-targeted ubiquitin E3 ligase (STUbL) ubiquitously expressed in all tissues, and which mainly participates in DNA repair and in chromatin and transcriptional regulation. Although RNF4 has been implicated in hematopoietic disorders, its ontogenic role during hematopoietic development remains undiscovered. We generated a zebrafish rnf4 knockout line by using transcription activator-like effector nucleases technology to address the impact of rnf4 during hematopoiesis. Rnf4-deficient zebrafish embryos exhibited sharply decreased neutrophils numbers during both primitive and definitive hematopoiesis. Mechanistic studies revealed that repression of the key granulocytic activator, CCAAT/enhancer-binding protein α ( c/ebpα), via promoter hypermethylation by SUMOylated DNA methyltransferase 1 (DNMT1) was the main cause of impaired granulopoiesis in rnf4-deficient zebrafish. In addition, for the first time, we identified DNMT1 as a potential new STUbL substrate of RNF4, with knockdown of dnmt1 largely restoring primitive and definitive granulopoiesis in rnf4-deficient zebrafish. Collectively, RNF4 is indispensable for zebrafish granulopoiesis through regulation of the DNMT1-C/EBPα functional axis.-Wang, L., Liu, X., Wang, H., Yuan, H., Chen, S., Chen, Z., de The, H., Zhou, J., Zhu, J. RNF4 regulates zebrafish granulopoiesis through the DNMT1-C/EBPα axis.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Regulación de la Expresión Génica/genética , Mutación/genética , Proteínas Nucleares/genética , Proteína SUMO-1/metabolismo , Factores de Transcripción/genética , Proteínas de Pez Cebra/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , Reparación del ADN/genética , Hematopoyesis/genética , Humanos , Proteína SUMO-1/genética , Sumoilación/genética , Sumoilación/fisiología , Factores de Transcripción/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
10.
Proc Natl Acad Sci U S A ; 112(46): 14278-83, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578773

RESUMEN

Promyelocytic leukemia protein (PML) nuclear bodies (NBs) recruit multiple partners, including p53 and many of its regulators. NBs are believed to facilitate several posttranslational modifications and are key regulators of senescence. PML, the organizer of NBs, is expressed as a number of splice variants that all efficiently recruit p53 partners. However, overexpression of only one of them, PML IV, triggers p53-driven senescence. Here, we show that PML IV specifically binds ARF, a key p53 regulator. Similar to ARF, PML IV enhances global SUMO-1 conjugation, particularly that of p53, resulting in p53 stabilization and activation. ARF interacts with and stabilizes the NB-associated UBC9 SUMO-conjugating enzyme, possibly explaining PML IV-enhanced SUMOylation. These results unexpectedly link two key tumor suppressors, highlighting their convergence for global control of SUMO conjugation, p53 activation, and senescence induction.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas Nucleares/metabolismo , Proteína SUMO-1/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica , Estabilidad Proteica , Proteína SUMO-1/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
11.
Blood ; 125(3): 474-82, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25395419

RESUMEN

The human T-cell lymphotropic virus type I (HTLV-1) Tax transactivator initiates transformation in adult T-cell leukemia/lymphoma (ATL), a highly aggressive chemotherapy-resistant malignancy. The arsenic/interferon combination, which triggers degradation of the Tax oncoprotein, selectively induces apoptosis of ATL cell lines and has significant clinical activity in Tax-driven murine ATL or human patients. However, the role of Tax loss in ATL response is disputed, and the molecular mechanisms driving degradation remain elusive. Here we demonstrate that ATL-derived or HTLV-1-transformed cells are dependent on continuous Tax expression, suggesting that Tax degradation underlies clinical responses to the arsenic/interferon combination. The latter enforces promyelocytic leukemia protein (PML) nuclear body (NB) formation and partner protein recruitment. In arsenic/interferon-treated HTLV-1 transformed or ATL cells, Tax is recruited onto NBs and undergoes PML-dependent hyper-sumoylation by small ubiquitin-like modifier (SUMO)2/3 but not SUMO1, ubiquitination by RNF4, and proteasome-dependent degradation. Thus, the arsenic/interferon combination clears ATL through degradation of its Tax driver, and this regimen could have broader therapeutic value by promoting degradation of other pathogenic sumoylated proteins.


Asunto(s)
Arsenicales/farmacología , Productos del Gen tax/metabolismo , Interferones/farmacología , Leucemia-Linfoma de Células T del Adulto/virología , Proteínas Nucleares/metabolismo , Proteolisis/efectos de los fármacos , Proteína SUMO-1/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Transformación Celular Viral/efectos de los fármacos , Quimioterapia Combinada , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HeLa , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Inmunoprecipitación , Leucemia-Linfoma de Células T del Adulto/genética , Proteína de la Leucemia Promielocítica , Reactivos de Sulfhidrilo/farmacología , Sumoilación/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
12.
Blood ; 125(22): 3447-54, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25800051

RESUMEN

Nucleophosmin-1 (NPM1) is the most frequently mutated gene in acute myeloid leukemia (AML). Addition of retinoic acid (RA) to chemotherapy was proposed to improve survival of some of these patients. Here, we found that RA or arsenic trioxide synergistically induce proteasomal degradation of mutant NPM1 in AML cell lines or primary samples, leading to differentiation and apoptosis. NPM1 mutation not only delocalizes NPM1 from the nucleolus, but it also disorganizes promyelocytic leukemia (PML) nuclear bodies. Combined RA/arsenic treatment significantly reduced bone marrow blasts in 3 patients and restored the subnuclear localization of both NPM1 and PML. These findings could explain the proposed benefit of adding RA to chemotherapy in NPM1 mutant AMLs, and warrant a broader clinical evaluation of regimen comprising a RA/arsenic combination.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , Óxidos/farmacología , Proteolisis/efectos de los fármacos , Tretinoina/farmacología , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Trióxido de Arsénico , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas Mutantes/efectos de los fármacos , Proteínas Mutantes/metabolismo , Mutación , Nucleofosmina , Células Tumorales Cultivadas
14.
Biochim Biophys Acta Gen Subj ; 1861(7): 1691-1701, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28285006

RESUMEN

BACKGROUND: SUMOylation is a critical regulatory protein modification in eukaryotic cells and plays a pivotal role in cardiac development and disease. Several cardiac transcription factors are modified by SUMO, but little is known about the impact of SUMOylation on their function during cardiac development. METHODS: We used a zebrafish model to address the impact of SUMOylation on GATA5, an essential transcription factor in zebrafish cardiac development. GATA5 SUMOylation was probed by western blot, the subcellular localization and transcriptional activity of GATA5 mutants were examined by immunostaining and luciferase reporter assay. The in vivo function of GATA5 SUMOylation was evaluated by gata5 mutants mRNA microinjection and in situ hybridization in gata5 morphants and ubc9 mutants. RESULTS: Firstly, we identified GATA5 as a SUMO substrate, and lysine 324 (K324) and lysine 360 (K360) as two major modification sites. Conversion of lysine to arginine at these two sites did not affect subcellular localization, but did affect the transcriptional activity of GATA5. Secondly, in vivo experiments demonstrated that the wild type (WT) and K324R mutant of gata5 could rescue impaired cardiac precursor differentiation, while the K360R mutant of gata5 drastically lost this potency in gata5 morphant. Furthermore, in SUMOylation-deficient ubc9 mutants, the abnormal expression pattern displayed by the early markers of cardiac development (nkx2.5 and mef2cb) could be restored using a sumo-gata5 fusion, but not with a WT gata5. CONCLUSION: GATA5 SUMOylation is indispensable for early zebrafish cardiac development. GENERAL SIGNIFICANCE: Our studies highlight the potential importance of transcription factor SUMOylation in cardiac development.


Asunto(s)
Factor de Transcripción GATA5/fisiología , Corazón/embriología , Sumoilación , Pez Cebra/embriología , Transporte Activo de Núcleo Celular , Animales , Diferenciación Celular , Células Cultivadas
16.
Blood ; 124(25): 3772-80, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25258343

RESUMEN

PML/RARA, a potent transcriptional inhibitor of nuclear receptor signaling, represses myeloid differentiation genes and drives acute promyelocytic leukemia (APL). Association of the retinoid X receptor-α (RXRA) coreceptor to PML/RARA is required for transformation, with RXRA promoting its efficient DNA binding. APL is exquisitely sensitive to retinoic acid (RA) and arsenic trioxide (arsenic), which both trigger cell differentiation in vivo. Whereas RA elicits transcriptional activation of PML/RARA targets, how arsenic triggers differentiation remains unclear. Here we demonstrate that extinction of PML/RARA triggers terminal differentiation in vivo. Similarly, ablation of retinoid X receptors loosens PML/RARA DNA binding, inducing terminal differentiation of APL cells ex vivo or in vivo. RXRA sumoylation directly contributes to PML/RARA-dependent transformation ex vivo, presumably by enhancing transcriptional repression. Thus, APL differentiation is a default program triggered by clearance of PML/RARA-bound promoters, rather than obligatory active transcriptional activation, explaining how arsenic elicits APL maturation through PML/RARA degradation.


Asunto(s)
Diferenciación Celular/genética , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas/genética , Animales , Antineoplásicos/farmacología , Trióxido de Arsénico , Arsenicales/farmacología , Células COS , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Cultivadas , Chlorocebus aethiops , Perfilación de la Expresión Génica , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Desnudos , Ratones Transgénicos , Proteínas de Fusión Oncogénica/metabolismo , Óxidos/farmacología , Unión Proteica , Interferencia de ARN , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sumoilación , Activación Transcripcional/genética , Tretinoina/farmacología
17.
Proc Natl Acad Sci U S A ; 110(9): 3495-500, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23382200

RESUMEN

The refractoriness of acute promyelocytic leukemia (APL) with t(11;17)(q23;q21) to all-trans retinoic acid (ATRA)-based therapy concerns clinicians and intrigues basic researchers. By using a murine leukemic model carrying both promyelocytic leukemia zinc finger/retinoic acid receptor-α (PLZF/RARα) and RARα/PLZF fusion genes, we discovered that 8-chlorophenylthio adenosine-3', 5'-cyclic monophosphate (8-CPT-cAMP) enhances cellular differentiation and improves gene trans-activation by ATRA in leukemic blasts. Mechanistically, in combination with ATRA, 8-CPT-cAMP activates PKA, causing phosphorylation of PLZF/RARα at Ser765 and resulting in increased dissociation of the silencing mediator for retinoic acid and thyroid hormone receptors/nuclear receptor corepressor from PLZF/RARα. This process results in changes of local chromatin and transcriptional reactivation of the retinoic acid pathway in leukemic cells. Meanwhile, 8-CPT-cAMP also potentiated ATRA-induced degradation of PLZF/RARα through its Ser765 phosphorylation. In vivo treatment of the t(11;17) APL mouse model demonstrated that 8-CPT-cAMP could significantly improve the therapeutic effect of ATRA by targeting a leukemia-initiating cell activity. This combined therapy, which induces enhanced differentiation and oncoprotein degradation, may benefit t(11;17) APL patients.


Asunto(s)
Diferenciación Celular , AMP Cíclico/análogos & derivados , Leucemia Promielocítica Aguda/tratamiento farmacológico , Proteínas de Fusión Oncogénica/metabolismo , Proteolisis , Tionucleótidos/uso terapéutico , Translocación Genética , Tretinoina/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 17/genética , AMP Cíclico/farmacología , AMP Cíclico/uso terapéutico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Co-Represor 2 de Receptor Nuclear/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Tionucleótidos/farmacología , Translocación Genética/efectos de los fármacos , Tretinoina/farmacología
18.
Breast Cancer Res Treat ; 149(1): 81-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25503779

RESUMEN

Several publications have suggested that histone deacetylase inhibitors (HDACis) could reverse the repression of estrogen receptor alpha (ERα) in triple-negative breast cancer (TNBC) cell lines, leading to the induction of a functional protein. Using different HDACis, vorinostat, panobinostat, and abexinostat, we therefore investigated this hypothesis in various human TNBC cell lines and patient-derived xenografts (PDXs). We used three human TNBC cell lines and three PDXs. We analyzed the in vitro toxicity of the compounds, their effects on the hormone receptors and hormone-related genes and protein expression both in vitro and in vivo models. We then explored intra-tumor histone H3 acetylation under abexinostat in xenograft models. Despite major cytotoxicity of all tested HDAC inhibitors and repression of deactylation-dependent CCND1 gene, neither ERα nor ERß, ESR1 or ESR2 genes respectively, were re-expressed in vitro. In vivo, after administration of abexinostat for three consecutive days, we did not observe any induction of ESR1 or ESR1-related genes and ERα protein expression by RT-qPCR and immunohistochemical methods in PDXs. This observation was concomitant to the fact that in vivo administration of abexinostat increased intra-tumor histone H3 acetylation. These observations do not allow us to confirm previous studies which suggested that HDACis are able to convert ER-negative (ER-) tumors to ER-positive (ER+) tumors, and that a combination of HDAC inhibitors and hormone therapy could be proposed in the management of TNBC patients.


Asunto(s)
Ciclina D1/biosíntesis , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/biosíntesis , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Benzofuranos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Histonas/genética , Humanos , Ácidos Hidroxámicos/administración & dosificación , Indoles/administración & dosificación , Panobinostat , Receptor ErbB-2/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Blood ; 122(12): 2008-10, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23894153

RESUMEN

Rarely in the field of cancer treatment did we experience as many surprises as with acute promyelocytic leukemia (APL). Yet, the latest clinical trial reported by Lo-Coco et al in the New England Journal of Medicine is a practice-changing study, as it reports a very favorable outcome of virtually all enrolled low-intermediate risk patients with APL without any DNA-damaging chemotherapy. Although predicted from previous small pilot studies, these elegant and stringently controlled results open a new era in leukemia therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Trióxido de Arsénico , Arsenicales/administración & dosificación , Humanos , Óxidos/administración & dosificación , Tretinoina/administración & dosificación
20.
Haematologica ; 100(8): 1064-75, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26088929

RESUMEN

Acute promyelocytic leukemia is an aggressive malignancy characterized by the accumulation of promyelocytes in the bone marrow. PML/RARA is the primary abnormality implicated in this pathology, but the mechanisms by which this chimeric fusion protein initiates disease are incompletely understood. Identifying PML/RARA targets in vivo is critical for comprehending the road to pathogenesis. Utilizing a novel sorting strategy, we isolated highly purified promyelocyte populations from normal and young preleukemic animals, carried out microarray and methylation profiling analyses, and compared the results from the two groups of animals. Surprisingly, in the absence of secondary lesions, PML/RARA had an overall limited impact on both the transcriptome and methylome. Of interest, we did identify down-regulation of secondary and tertiary granule genes as the first step engaging the myeloid maturation block. Although initially not sufficient to arrest terminal granulopoiesis in vivo, such alterations set the stage for the later, complete differentiation block seen in leukemia. Further, gene set enrichment analysis revealed that PML/RARA promyelocytes exhibit a subtle increase in expression of cell cycle genes, and we show that this leads to both increased proliferation of these cells and expansion of the promyelocyte compartment. Importantly, this proliferation signature was absent from the poorly leukemogenic p50/RARA fusion model, implying a critical role for PML in the altered cell-cycle kinetics and ability to initiate leukemia. Thus, our findings challenge the predominant model in the field and we propose that PML/RARA initiates leukemia by subtly shifting cell fate decisions within the promyelocyte compartment.


Asunto(s)
Metilación de ADN , Células Precursoras de Granulocitos/metabolismo , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Transcripción Genética , Animales , Antígenos CD34/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Células Precursoras de Granulocitos/patología , Humanos , Inmunofenotipificación , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones , Ratones Transgénicos , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA