Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 541(7638): 541-545, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28068668

RESUMEN

Cell fate perturbations underlie many human diseases, including breast cancer. Unfortunately, the mechanisms by which breast cell fate are regulated are largely unknown. The mammary gland epithelium consists of differentiated luminal epithelial and basal myoepithelial cells, as well as undifferentiated stem cells and more restricted progenitors. Breast cancer originates from this epithelium, but the molecular mechanisms that underlie breast epithelial hierarchy remain ill-defined. Here, we use a high-content confocal image-based short hairpin RNA screen to identify tumour suppressors that regulate breast cell fate in primary human breast epithelial cells. We show that ablation of the large tumour suppressor kinases (LATS) 1 and 2 (refs 5, 6), which are part of the Hippo pathway, promotes the luminal phenotype and increases the number of bipotent and luminal progenitors, the proposed cells-of-origin of most human breast cancers. Mechanistically, we have identified a direct interaction between Hippo and oestrogen receptor-α (ERα) signalling. In the presence of LATS, ERα was targeted for ubiquitination and Ddb1-cullin4-associated-factor 1 (DCAF1)-dependent proteasomal degradation. Absence of LATS stabilized ERα and the Hippo effectors YAP and TAZ (hereafter YAP/TAZ), which together control breast cell fate through intrinsic and paracrine mechanisms. Our findings reveal a non-canonical (that is, YAP/TAZ-independent) effect of LATS in the regulation of human breast cell fate.


Asunto(s)
Mama/citología , Mama/enzimología , Diferenciación Celular , Linaje de la Célula , Receptor alfa de Estrógeno/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/agonistas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Mama/patología , Proteínas Portadoras/metabolismo , Células Cultivadas , Receptor alfa de Estrógeno/agonistas , Femenino , Genes Supresores de Tumor , Humanos , Fosfoproteínas/agonistas , Fosfoproteínas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteolisis , Transducción de Señal , Factores de Transcripción , Proteínas Supresoras de Tumor/deficiencia , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas , Proteínas Señalizadoras YAP
2.
Biochim Biophys Acta ; 1849(9): 1198-208, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26187004

RESUMEN

The human DMTF1 (DMP1) transcription factor, a DNA binding protein that interacts with cyclin D, is a positive regulator of the p14ARF (ARF) tumor suppressor. Our earlier studies have shown that three differentially spliced human DMP1 mRNAs, α, ß and γ, arise from the human gene. We now show that DMP1α, ß and γ isoforms differentially regulate ARF expression and promote distinct cellular functions. In contrast to DMP1α, DMP1ß and γ did not activate the ARF promoter, whereas only ß resulted in a dose-dependent inhibition of DMP1α-induced transactivation of the ARF promoter. Ectopic expression of DMP1ß reduced endogenous ARF mRNA levels in human fibroblasts. The DMP1ß- and γ-isoforms share domains necessary for the inhibitory function of the ß-isoform. That DMP1ß may interact with DMP1α to antagonize its function was shown in DNA binding assays and in cells by the close proximity of DMP1α/ß in the nucleus. Cells stably expressing DMP1ß, as well as shRNA targeting all DMP1 isoforms, disrupted cellular growth arrest induced by serum deprivation or in PMA-derived macrophages in the presence or absence of cellular p53. DMP1 mRNA levels in acute myeloid leukemia samples, as compared to granulocytes, were reduced. Treatment of acute promyelocytic leukemia patient samples with all-trans retinoic acid promoted differentiation to granulocytes and restored DMP1 transcripts to normal granulocyte levels. Our findings imply that DMP1α- and ß-ratios are tightly regulated in hematopoietic cells and DMP1ß antagonizes DMP1α transcriptional regulation of ARF resulting in the alteration of cellular control with a gain in proliferation.


Asunto(s)
Proliferación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Isoformas de Proteínas/fisiología , Factores de Transcripción/fisiología , Transcripción Genética/fisiología , Proteína p14ARF Supresora de Tumor/genética , Animales , Línea Celular , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Isoformas de Proteínas/genética , Empalme del ARN , ARN Mensajero/metabolismo , Factores de Transcripción/genética
3.
PLoS One ; 8(1): e51671, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23300950

RESUMEN

Emerging evidence suggests that some cancers contain a population of stem-like TICs (tumor-initiating cells) and eliminating TICs may offer a new strategy to develop successful anti-cancer therapies. As molecular mechanisms underlying the maintenance of the TIC pool are poorly understood, the development of TIC-specific therapeutics remains a major challenge. We first identified and characterized TICs and non-TICs isolated from a mouse breast cancer model. TICs displayed increased tumorigenic potential, self-renewal, heterogeneous differentiation, and bipotency. Gene expression analysis and immunostaining of TICs and non-TICs revealed that FGFR2 was preferentially expressed in TICs. Loss of FGFR2 impaired self-renewal of TICs, thus resulting in marked decreases in the TIC population and tumorigenic potential. Restoration of FGFR2 rescued the defects in TIC pool maintenance, bipotency, and breast tumor growth driven by FGFR2 knockdown. In addition, pharmacological inhibition of FGFR2 kinase activity led to a decrease in the TIC population which resulted in suppression of breast tumor growth. Moreover, human breast TICs isolated from patient tumor samples were found enriched in a FGFR2+ population that was sufficient to initiate tumor growth. Our data suggest that FGFR2 is essential in sustaining the breast TIC pool through promotion of self-renewal and maintenance of bipotent TICs, and raise the possibility of FGFR2 inhibition as a strategy for anti-cancer therapy by eradicating breast TICs.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Animales/metabolismo , Células Madre Neoplásicas/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Antígeno CD24/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Integrina beta1/metabolismo , Ratones , Ratones Endogámicos NOD , Microscopía Fluorescente , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Transducción de Señal
4.
PLoS One ; 7(2): e31226, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359577

RESUMEN

Tumor progenitor cells represent a population of drug-resistant cells that can survive conventional chemotherapy and lead to tumor relapse. However, little is known of the role of tumor progenitors in prostate cancer metastasis. The studies reported herein show that the CXCR4/CXCL12 axis, a key regulator of tumor dissemination, plays a role in the maintenance of prostate cancer stem-like cells. The CXCL4/CXCR12 pathway is activated in the CD44(+)/CD133(+) prostate progenitor population and affects differentiation potential, cell adhesion, clonal growth and tumorigenicity. Furthermore, prostate tumor xenograft studies in mice showed that a combination of the CXCR4 receptor antagonist AMD3100, which targets prostate cancer stem-like cells, and the conventional chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors as compared to monotherapy.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores CXCR4/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Bencilaminas , Adhesión Celular , Proliferación Celular , Quimiocina CXCL12/metabolismo , Ciclamas , Docetaxel , Compuestos Heterocíclicos/farmacología , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Taxoides/farmacología
5.
Mol Immunol ; 48(4): 714-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21094529

RESUMEN

C-type lectin domain family 5, member A (CLEC5A), also known as myeloid DNAX activation protein 12 (DAP12)-associating lectin-1 (MDL-1), is a cell surface receptor strongly associated with the activation and differentiation of myeloid cells. CLEC5A associates with its adaptor protein DAP12 to activate a signaling cascade resulting in activation of downstream kinases in inflammatory responses. Currently, little is known about the transcriptional regulation of CLEC5A. We identified CLEC5A as one of the most highly induced genes in a microarray gene profiling experiment of PU.1 restored myeloid PU.1-null cells. We further report that CLEC5A expression is significantly reduced in several myeloid differentiation models upon PU.1 inhibition during monocyte/macrophage or granulocyte differentiation. In addition, CLEC5A mRNA expression was significantly lower in primary acute myeloid leukemia (AML) patient samples than in macrophages and granulocytes from healthy donors. Moreover, we found activation of a CLEC5A promoter reporter by PU.1 as well as in vivo binding of PU.1 to the CLEC5A promoter. Our findings indicate that CLEC5A expression in monocyte/macrophage and granulocytes is regulated by PU.1.


Asunto(s)
Diferenciación Celular/genética , Lectinas Tipo C/genética , Células Mieloides/citología , Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Superficie Celular/genética , Transactivadores/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Lectinas Tipo C/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Datos de Secuencia Molecular , Monocitos/citología , Monocitos/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/metabolismo
6.
Clin Cancer Res ; 16(23): 5692-702, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21138868

RESUMEN

PURPOSE: The cancer stem cell hypothesis predicts that standard prostate cancer monotherapy eliminates bulk tumor cells but not a tumor-initiating cell population, eventually leading to relapse. Many studies have sought to determine the underlying differences between bulk tumor and cancer stem cells. EXPERIMENTAL DESIGN: Our previous data suggest that the PTEN/PI3K/AKT pathway is critical for the in vitro maintenance of CD133(+)/CD44(+) prostate cancer progenitors and, consequently, that targeting PI3K signaling may be beneficial in treatment of prostate cancer. RESULTS: Here, we show that inhibition of PI3K activity by the dual PI3K/mTOR inhibitor NVP-BEZ235 leads to a decrease in the population of CD133(+)/CD44(+) prostate cancer progenitor cells in vivo. Moreover, the combination of the PI3K/mTOR modulator NVP-BEZ235, which eliminates prostate cancer progenitor populations, and the chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors in a prostate cancer xenograft model than monotherapy. CONCLUSION: This combination treatment ultimately leads to the expansion of cancer progenitors with a PTEN E91D mutation, suggesting that the analysis of PTEN mutations could predict therapeutic response to the dual therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma/patología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Sistemas de Liberación de Medicamentos/métodos , Glicoproteínas/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Imidazoles/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosfohidrolasa PTEN/genética , Péptidos/metabolismo , Neoplasias de la Próstata/patología , Quinolinas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Proc Natl Acad Sci U S A ; 106(1): 268-73, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19116269

RESUMEN

Characterization of the molecular pathways that are required for the viability and maintenance of self-renewing tumor-initiating cells may ultimately lead to improved therapies for cancer. In this study, we show that a CD133(+)/CD44(+) population of cells enriched in prostate cancer progenitors (PCaPs) has tumor-initiating potential and that these progenitors can be expanded under nonadherent, serum-free, sphere-forming conditions. Cells grown under these conditions have increased in vitro clonogenic and in vivo tumorigenic potential. mRNA expression analysis of cells grown under sphere-forming conditions, compared with long-term monolayer cultures, revealed preferential activation of the PI3K/AKT signaling pathway. PI3K p110alpha and beta-protein levels were higher in cells grown under sphere-forming conditions, and phosphatase and tensin homolog (PTEN) knockdown by shRNA led to an increase in sphere formation as well as increased clonogenic and tumorigenic potential. Similarly, shRNA knockdown of FoxO3a led to an increase in tumorigenic potential. Consistent with these results, inhibition of PI3K activity by the dual PI3K/mTOR inhibitor NVP-BEZ235 led to growth inhibition of PCaPs. Taken together, our data strongly suggest that the PTEN/PI3K/Akt pathways are critical for prostate cancer stem-like cell maintenance and that targeting PI3K signaling may be beneficial in prostate cancer treatment by eliminating prostate cancer stem-like cells.


Asunto(s)
Células Madre Neoplásicas/patología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Antígeno AC133 , Antígenos CD , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Glicoproteínas , Humanos , Receptores de Hialuranos , Masculino , Péptidos , ARN Mensajero/análisis , ARN Neoplásico/análisis , Transducción de Señal/fisiología
8.
PLoS One ; 3(1): e1487, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18213395

RESUMEN

Specificity of protein ubiquitylation is conferred by E3 ubiquitin (Ub) ligases. We have annotated approximately 617 putative E3s and substrate-recognition subunits of E3 complexes encoded in the human genome. The limited knowledge of the function of members of the large E3 superfamily prompted us to generate genome-wide E3 cDNA and RNAi expression libraries designed for functional screening. An imaging-based screen using these libraries to identify E3s that regulate mitochondrial dynamics uncovered MULAN/FLJ12875, a RING finger protein whose ectopic expression and knockdown both interfered with mitochondrial trafficking and morphology. We found that MULAN is a mitochondrial protein - two transmembrane domains mediate its localization to the organelle's outer membrane. MULAN is oriented such that its E3-active, C-terminal RING finger is exposed to the cytosol, where it has access to other components of the Ub system. Both an intact RING finger and the correct subcellular localization were required for regulation of mitochondrial dynamics, suggesting that MULAN's downstream effectors are proteins that are either integral to, or associated with, mitochondria and that become modified with Ub. Interestingly, MULAN had previously been identified as an activator of NF-kappaB, thus providing a link between mitochondrial dynamics and mitochondria-to-nucleus signaling. These findings suggest the existence of a new, Ub-mediated mechanism responsible for integration of mitochondria into the cellular environment.


Asunto(s)
Genoma Humano , Mitocondrias/enzimología , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Humanos , Inmunohistoquímica , Ratones , Microscopía Confocal , Microscopía Inmunoelectrónica
9.
Proteomics ; 6(21): 5705-19, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17001604

RESUMEN

In the present study, we employed 2-DE to characterize the effect of the acute promyelocytic leukemia (APL)-specific PML-RARalpha fusion protein on the proteome. Differentially expressed proteins, a number of which are related to the cell cycle function, including oncoprotein18 (OP18), heat shock protein70, glucose-regulated protein75, and peptidyl-prolyl isomerase, were identified by MS. Subsequent bioinformatic pathway discovery revealed an integrated network constituting SMARCB1, MYC, and TP53-regulated pathways. The data from the DNA microarray and proteomic experiments demonstrated the correlation between the translocation and higher expression of OP18 at mRNA and protein levels. Transient cotransfection assay revealed that PML-RARalpha is a potent activator of OP18 promoter and this transcriptional activation is retinoic acid sensitive. PML-RARalpha induction also leads to decreased phosphorylation on Ser63 residue of OP18, which is okadaic acid sensitive suggesting the involvement of a phosphatase pathway. Overexpression of a constitutively phosphorylated Ser63 mutant of OP18 in PML-RARalpha expressing APL patient, PR9, and NB4 cells led to a G2/M-phase arrest in contrast to a phosphorylation-deficient Ser63 mutant and untransfected control. Taken together, our results demonstrate the significance of decreased Ser63 phosphorylation of OP18 in PML-RARalpha-mediated effects on cell cycle.


Asunto(s)
Leucemia Promielocítica Aguda/metabolismo , Proteínas de Fusión Oncogénica/análisis , Proteoma/análisis , Serina/metabolismo , Estatmina/metabolismo , Línea Celular , Células Clonales , Biología Computacional/métodos , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Genes Reporteros , Humanos , Leucemia Promielocítica Aguda/patología , Luciferasas/metabolismo , Espectrometría de Masas , Mutación , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Mapeo Peptídico , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Secundaria de Proteína , Proteómica/métodos , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Estatmina/química , Estatmina/genética , Transfección , Células U937 , Sulfato de Zinc
10.
Oncogene ; 22(36): 5646-57, 2003 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-12944913

RESUMEN

Overexpression of proto-oncogene c-jun and constitutive activation of the Jun N-terminal kinase (JNK) signaling pathway have been implicated in the leukemic transformation process. However, c-jun expression and the role of the JNK signaling pathway have not been investigated in primary acute myeloid leukemia (AML) cells with frequently observed balanced rearrangements such as t(8;21). In the present study, we report elevated c-jun mRNA expression in AML patient bone marrow cells with t(8;21), t(15;17) or inv(16), and a high correlation in mRNA expression levels of AML1-ETO and c-jun within t(8;21)-positive AML patient cells. In myeloid U937 cells, c-jun mRNA and protein expression increase upon inducible expression of AML1-ETO. AML1-ETO transactivates the human c-jun promoter through the proximal activator protein (AP-1) site by activating the JNK pathway. Overexpression of JNK-inhibitor JIP-1 and chemical JNK inhibitors reduce the transactivation capacity of AML1-ETO on the c-jun promoter and the proapoptotic function of AML1-ETO in U937 cells. An autocrine mechanism involving granulocyte-colony stimulating factor (G-CSF) and G-CSF receptor (G-CSF-R) might participate in AML1-ETO mediated JNK-signaling, because AML1-ETO induces G-CSF and G-CSF-R expression, and G-CSF-R-neutralizing antibodies reduce AML1-ETO-induced JNK phosphorylation. These data suggest a model in which AML1-ETO induces proto-oncogene c-jun expression via the proximal AP-1 site of the c-jun promoter in a JNK-dependent manner.


Asunto(s)
Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Genes jun , Leucemia Mieloide Aguda/genética , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/genética , Factor de Transcripción AP-1/fisiología , Factores de Transcripción/genética , Translocación Genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Factor Estimulante de Colonias de Granulocitos/fisiología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Fosforilación , Proto-Oncogenes Mas , Proteína 1 Compañera de Translocación de RUNX1 , Transducción de Señal , Activación Transcripcional , Células U937
11.
Oncogene ; 22(15): 2296-308, 2003 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-12700665

RESUMEN

In the present study, we investigated the mechanism of CD44 ligation with the anti-CD44 monoclonal antibody A3D8 to inhibit the proliferation of human acute myeloid leukemia (AML) cells. The effects of A3D8 on myeloid cells were associated with specific disruption of cell cycle events and induction of G0/G1 arrest. Induction of G0/G1 arrest was accompanied by an increase in the expression of p21, attenuation of pRb phosphorylation and associated with decreased Cdk2 and Cdk4 kinase activities. Since c-Jun is an important regulator of proliferation and cell cycle progression, we analysed its role in A3D8-mediated growth arrest. We observed that A3D8 treatment of AML patient blasts and HL60/U937 cells led to the downregulation of c-Jun expression at mRNA and protein level. Transient transfection studies showed the inhibition of c-jun promoter activity by A3D8, involving both AP-1 sites. Furthermore, A3D8 treatment caused a decrease in JNK protein expression and a decrease in the level of phosphorylated c-Jun. Ectopic overexpression of c-Jun in HL60 cells was able to induce proliferation and prevent the antiproliferative effects of A3D8. In summary, these data identify an important functional role of c-Jun in the induction of cell cycle arrest and proliferation arrest of myeloid leukemia cells because of the ligation of the cell surface adhesion receptor CD44 by anti-CD44 antibody. Moreover, targeting of G1 regulatory proteins and the resulting induction of G1 arrest by A3D8 may provide new insights into antiproliferative and differentiation therapy of AML.


Asunto(s)
Quinasas CDC2-CDC28 , Ciclo Celular/fisiología , Genes jun , Receptores de Hialuranos/fisiología , Leucemia Mieloide/metabolismo , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Proteínas Proto-Oncogénicas , Enfermedad Aguda , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/biosíntesis , Quinasas Ciclina-Dependientes/genética , Ciclinas/biosíntesis , Ciclinas/genética , Fase G1/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células HL-60/citología , Células HL-60/efectos de los fármacos , Células HL-60/metabolismo , Humanos , Receptores de Hialuranos/inmunología , Proteínas Quinasas JNK Activadas por Mitógenos , Leucemia Mieloide/patología , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Células U937/citología , Células U937/efectos de los fármacos , Células U937/metabolismo
12.
Expert Opin Ther Targets ; 6(4): 491-5, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12223063

RESUMEN

Recent results indicate that interactions of transcription factors with other nuclear proteins play an important role in stem cell development, lineage commitment and differentiation in the haematopoietic system, and the pathogenesis of myeloid leukaemias. High-throughput proteomics by mass spectrometric analysis of gel-separated proteins can identify multi-protein complexes and changes in the expression of multiple proteins simultaneously. This review describes an application of proteomic methods (2D gel electrophoresis (GE) and mass spectrometry (MS)), which can be used to identify regulated protein targets of transcription factors important in myeloid differentiation and leukaemia. This global high-throughput functional proteomics approach could lead to new insights into the network of protein-protein interactions and target proteins involved in myeloid stem cell development and leukaemia as well as provide new targets for rational pathogenesis-based therapies of leukaemia and cancer.


Asunto(s)
Leucemia/genética , Células Mieloides/fisiología , Proteínas de Neoplasias/fisiología , Células Madre/fisiología , Factores de Transcripción/fisiología , Proteínas Potenciadoras de Unión a CCAAT/análisis , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/análisis , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Electroforesis en Gel Bidimensional , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia/metabolismo , Proteínas de Neoplasias/análisis , Proteínas de Fusión Oncogénica/análisis , Proteínas de Fusión Oncogénica/fisiología , Péptidos/análisis , Proteómica , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/fisiología , Proteína 1 Compañera de Translocación de RUNX1 , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transactivadores/análisis , Transactivadores/fisiología , Factores de Transcripción/análisis
13.
Blood ; 100(2): 483-90, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12091339

RESUMEN

Several transcription factors have been implicated as playing a role in myelopoiesis. PU.1, an ets-family transcription factor, is required for the development of myeloid and lymphoid lineages, whereas the transcription factor CCAAT-enhancer binding protein family member C/EBPalpha is essential for granulocyte development. We present here the first evidence that C/EBPalpha blocks the function of PU.1. PU.1 and C/EBPalpha interact physically and colocalize in myeloid cells. As a consequence of this interaction, C/EBPalpha can inhibit the function of PU.1 to activate a minimal promoter containing only PU.1 DNA-binding sites. We further demonstrate that the leucine zipper in the DNA-binding domain of C/EBPalpha interacts with the beta3/beta4 region in the DNA-binding domain of PU.1 and as a result displaces the PU.1 coactivator c-Jun. Finally, C/EBPalpha blocks PU.1-induced dendritic cell development from CD34+ human cord blood cells. The functional blocking of PU.1 by C/EBPalpha could be the mechanism by which C/EBPalpha inhibits cell fates specified by PU.1 and directs cell development to the granulocyte lineage.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Granulocitos/citología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Animales , Sitios de Unión , Proteína alfa Potenciadora de Unión a CCAAT/farmacología , Línea Celular , Linaje de la Célula/efectos de los fármacos , Sangre Fetal/citología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ratones , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Receptor Cross-Talk , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...