Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cell Sci ; 125(Pt 4): 993-1002, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22399799

RESUMEN

Gfi-1B is a transcriptional repressor essential for the regulation of erythropoiesis and megakaryopoiesis. Here we identify Gfi-1B p32, a Gfi-1B isoform, as essential for erythroid differentiation. Gfi-1B p32 is generated by alternative splicing and lacks the two first zinc finger domains of the protein. Selective knock down of Gfi-1B p32 compromises erythroid differentiation, whereas its ectopic expression induces erythropoiesis in the absence of erythropoietin. Gfi-1B p32 isoform binds to Gfi-1B target gene promoters and associates with the LSD1-CoREST repressor complex more efficiently than the major Gfi-1B p37 isoform. Furthermore, we show that Gfi-1B includes a KSKK motif in its SNAG domain, which recruits the repressor complex only when dimethylated on lysine 8. Mutation of lysine 8 prevents Gfi-1B p32-induced erythroid development. Our results thus highlight a key role for the alternatively spliced Gfi-1B p32 isoform in erythroid development.


Asunto(s)
Eritropoyesis , Histona Demetilasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Empalme Alternativo , Secuencias de Aminoácidos , Línea Celular , Proteínas Co-Represoras , Eritropoyesis/genética , Eritropoyetina , Regulación del Desarrollo de la Expresión Génica , Humanos , Lisina/metabolismo , Metilación , Peso Molecular , Regiones Promotoras Genéticas/genética , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Dedos de Zinc
2.
Exp Hematol ; 39(6): 629-42, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21420467

RESUMEN

OBJECTIVE: LYL-1 is a transcription factor containing a basic helix-loop-helix motif closely related to SCL/TAL-1, a regulator of erythroid differentiation. Because LYL-1 is expressed in erythroid cell populations, we addressed its role in erythropoiesis using knockin mice. MATERIALS AND METHODS: Erythropoiesis of LYL-1(-/-) mice was studied by progenitor assays, flow cytometry, reconstitution assays, and functional tests. Expression of LYL-1, SCL, and GATA-1 was assessed at messenger RNA level by quantitative reverse transcription polymerase chain reaction. RESULTS: LYL-1(-/-) mice displayed decreased erythropoiesis with a partial arrest in differentiation, and enhanced apoptosis associated with decreased Bcl-x(L) expression in the bone marrow (BM). In addition, LYL-1(-/-) BM cells were severely impaired in their abilities to reconstitute the erythroid lineage in competitive assays, suggesting a cell autonomous abnormality of erythropoiesis. In parallel, erythroid progenitor and precursor cells were significantly increased in the spleen of LYL-1(-/-) mice. Expression of LYL-1 was differentially regulated during maturation of erythroblasts and strikingly different between spleen- and BM-derived erythroblasts. Expression of LYL-1 decreased during erythroid differentiation in the spleen whereas it increased in the BM to reach the same level in mature erythroblasts as in the soleen. Loss of Lyl-1 expression was accompanied with an increase of SCL/TAL-1 and GATA-1 transcripts in spleen but not in BM-derived erythroblasts. Furthermore, phenylhydrazine-induced stress erythropoiesis was elevated in LYL-1(-/-) mice and mutant BM and spleen erythroid progenitors were hypersensitive to erythropoietin. CONCLUSIONS: Taken together, these results suggest that LYL-1 plays a definite role in erythropoiesis, albeit with different effects in BM specifically regulating basal erythropoiesis, and spleen, controlling stress-induced erythropoiesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Eritropoyesis/genética , Proteínas de Neoplasias/fisiología , Estrés Fisiológico , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cartilla de ADN , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Blood ; 115(14): 2784-95, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20124515

RESUMEN

Growth factor independence-1B (Gfi-1B) is a transcriptional repressor essential for erythropoiesis and megakaryopoiesis. Targeted gene disruption of GFI1B in mice leads to embryonic lethality resulting from failure to produce definitive erythrocytes, hindering the study of Gfi-1B function in adult hematopoiesis. We here show that, in humans, Gfi-1B controls the development of erythrocytes and megakaryocytes by regulating the proliferation and differentiation of bipotent erythro-megakaryocytic progenitors. We further identify in this cell population the type III transforming growth factor-beta receptor gene, TGFBR3, as a direct target of Gfi-1B. Knockdown of Gfi-1B results in altered transforming growth factor-beta (TGF-beta) signaling as shown by the increase in Smad2 phosphorylation and its inability to associate to the transcription intermediary factor 1-gamma (TIF1-gamma). Because the Smad2/TIF1-gamma complex is known to specifically regulate erythroid differentiation, we propose that, by repressing TGF-beta type III receptor (TbetaRIotaII) expression, Gfi-1B favors the Smad2/TIF1-gamma interaction downstream of TGF-beta signaling, allowing immature progenitors to differentiate toward the erythroid lineage.


Asunto(s)
Diferenciación Celular/fisiología , Eritrocitos/metabolismo , Megacariocitos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Proteínas Reguladoras de la Apoptosis/metabolismo , Proliferación Celular , Eritrocitos/citología , Eritropoyesis/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Células K562 , Células Progenitoras de Megacariocitos y Eritrocitos/citología , Células Progenitoras de Megacariocitos y Eritrocitos/metabolismo , Megacariocitos/citología , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Fosforilación/fisiología , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Trombopoyesis/fisiología
4.
Blood ; 115(3): 687-95, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965638

RESUMEN

Gfi-1B is a transcriptional repressor that is crucial for erythroid differentiation: inactivation of the GFI1B gene in mice leads to embryonic death due to failure to produce differentiated red cells. Accordingly, GFI1B expression is tightly regulated during erythropoiesis, but the mechanisms involved in such regulation remain partially understood. We here identify HMGB2, a high-mobility group HMG protein, as a key regulator of GFI1B transcription. HMGB2 binds to the GFI1B promoter in vivo and up-regulates its trans-activation most likely by enhancing the binding of Oct-1 and, to a lesser extent, of GATA-1 and NF-Y to the GFI1B promoter. HMGB2 expression increases during erythroid differentiation concomitantly to the increase of GfI1B transcription. Importantly, knockdown of HMGB2 in immature hematopoietic progenitor cells leads to decreased Gfi-1B expression and impairs their erythroid differentiation. We propose that HMGB2 potentiates GATA-1-dependent transcription of GFI1B by Oct-1 and thereby controls erythroid differentiation.


Asunto(s)
Diferenciación Celular/genética , Eritrocitos/fisiología , Proteína HMGB2/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA1/fisiología , Proteína HMGB2/antagonistas & inhibidores , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/farmacología , Proteínas Represoras/metabolismo , Activación Transcripcional/efectos de los fármacos
5.
Stem Cells ; 27(9): 2153-62, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19522008

RESUMEN

Growth Factor Independent-1B (Gfi-1B) is a transcriptional repressor that plays critical roles in the control of erythropoiesis and megakaryopoiesis. Gfi-1B expression was described to be repressed by an autoregulatory feedback control loop. Here, we show that Gfi-1 transcription is positively regulated early after induction of erythroid differentiation and remains highly active to late erythroblasts. Using chromatin immunoprecipitation assays in CD34+ cells from human cord blood, we found that Gfi-1 and GATA-2 in immature progenitors and then Gfi-1B and GATA-1 in erythroblasts are bound to the Gfi-1B promoter as well as to the promoter of c-myc, a known Gfi-1B target gene. Surprisingly, this Gfi-1/GATA-2-Gfi-1B/GATA-1 switch observed at erythroblast stages is associated to an increase in the Gfi-1B transcription whereas it triggers repression of c-myc transcription. Accordingly, analysis of chromatin modification patterns shows that HDAC, CoREST, and LSD1 are recruited to the c-myc promoter leading to appearance of repressive chromatin marks. In contrast, the Gfi-1B promoter remains associated with a transcriptionally active chromatin configuration as highlighted by an increase in histone H3 acetylation and concomitant release of the LSD1 and CoREST corepressors. The repressive function of Gfi-1B therefore depends on the nature of the proteins recruited to the target gene promoters and on chromatin modifications. We conclude that Gfi-1B behaves as a lineage-affiliated gene with an open chromatin configuration in multipotent progenitors and sustained activation as cells progress throughout erythroid differentiation.


Asunto(s)
Cromatina/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Western Blotting , Diferenciación Celular , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas Co-Represoras , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/metabolismo , Vectores Genéticos , Histona Demetilasas/metabolismo , Humanos , Lentivirus , Proteínas del Tejido Nervioso/metabolismo , Reacción en Cadena de la Polimerasa , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Transfección
6.
Blood ; 109(12): 5215-22, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17327410

RESUMEN

Control of intensity and duration of erythropoietin (Epo) signaling is necessary to tightly regulate red blood cell production. We have recently shown that the ubiquitin/proteasome system plays a major role in the control of Epo-R signaling. Indeed, after Epo stimulation, Epo-R is ubiquitinated and its intracellular part is degraded by the proteasome, preventing further signal transduction. The remaining part of the receptor and associated Epo are internalized and degraded by the lysosomes. We show that beta-Trcp is responsible for Epo-R ubiquitination and degradation. After Epo stimulation, beta-Trcp binds to the Epo-R. This binding, like Epo-R ubiquitination, requires Jak2 activation. The Epo-R contains a typical DSG binding sequence for beta-Trcp that is highly conserved among species. Interestingly, this sequence is located in a region of the Epo-R that is deleted in patients with familial polycythemia. Mutation of the serine residue of this motif to alanine (Epo-RS462A) abolished beta-Trcp binding, Epo-R ubiquitination, and degradation. Epo-RS462A activation was prolonged and BaF3 cells expressing this receptor are hypersensitive to Epo, suggesting that part of the hypersensitivity to Epo in familial polycythemia could be the result of the lack of beta-Trcp recruitment to the Epo-R.


Asunto(s)
Proliferación Celular , Receptores de Eritropoyetina/metabolismo , Ubiquitinas/metabolismo , Proteínas con Repetición de beta-Transducina/fisiología , Animales , Sitios de Unión , Línea Celular , Endocitosis , Humanos , Ratones , Unión Proteica
7.
Gene Expr Patterns ; 7(3): 215-26, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17112790

RESUMEN

The TAL-1/SCL and LYL-1 genes encode two closely related basic helix-loop-helix transcription factors involved in child T-acute lymphoblastic leukemia through chromosomal rearrangements and transcriptional deregulation. During ontogeny, Tal-1/SCL is required for hematopoietic cell generation, both in the yolk sac, where erythro-myeloid cells are first produced, then in the intra-embryonic compartment, where hematopoietic stem cells independently arise. We describe here the expression pattern of lyl-1 in mouse embryos from 7 to 14 days post coitus using in situ hybridization, as well as beta-Galactosidase (beta-Gal) expression in lyl-1-lacZ knock-in embryos, which express a C-terminally truncated Lyl-1 protein fused to the beta-Galactosidase (Lyl-1Delta/beta-Gal). In addition, we compare lyl-1 expression pattern with that of tal-1/scl. Similar to Tal-1/SCL, Lyl-1 mRNA expression occurs in the developing cardiovascular and hematopoietic systems. However, contrary to tal-1/scl, lyl-1 is not expressed in the developing nervous system. In lyl-1-lacZ knock-in heterozygous and homozygous embryos, beta-Gal expression completely correlates with Lyl-1 mRNA expression in the intra-embryonic compartment and is present: (1) in the developing hematopoietic system, precisely where hematopoietic stem cells emerge, and thereafter in the fetal liver; (2) in the developing vascular system; and (3) in the endocardium. In contrast, whereas Lyl-1 mRNA is expressed in yolk sac-derived endothelial and hematopoietic cells, Lyl-1Delta/beta-Gal is either absent or poorly expressed in these cell types, thus differing from Tal-1/SCL, which is highly expressed there at both mRNA and protein levels.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sistema Cardiovascular/embriología , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Sistema Hematopoyético/embriología , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas/genética , Animales , Desarrollo Embrionario , Femenino , Hibridación in Situ , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , Proteína 1 de la Leucemia Linfocítica T Aguda , beta-Galactosidasa/genética
8.
Blood ; 107(12): 4678-86, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16514064

RESUMEN

Hematopoietic stem cells (HSCs) arise, self-renew, or give rise to all hematopoietic lineages through the effects of transcription factors activated by signaling cascades. Lyl-1 encodes a transcription factor containing a basic helix-hoop-helix (bHLH) motif closely related to scl/tal, which controls numerous decisions in embryonic and adult hematopoiesis. We report here that Lyl-1 null mice are viable and display normal blood cell counts, except for a reduced number of B cells resulting from a partial block after the pro-B stage. Nevertheless, the deletion of Lyl-1 results in a diminution in the frequency of immature progenitors (Lin(-), CD34(-), sca-1(+), c-kit(+) [LSK], and LSK-side population [LSK-SP]) and in S(12) colony-forming unit (CFU-S(12)) and long-term culture-initiating cell (LTC-IC) content in embryonic day 14 fetal liver (E14 FL) and adult bone marrow (BM). More important, Lyl-1(-/-) E14 FL cells and BM are severely impaired in their competitive reconstituting abilities, especially with respect to B and T lineage reconstitution. Thus, ablation of Lyl-1 quantitatively and functionally affects HSCs, a cell population that transcribes Lyl-1 more actively than their differentiated progenies. Our results demonstrate for the first time that Lyl-1 functions are important for HSC properties and B-cell differentiation and that they are largely distinct from scl functions.


Asunto(s)
Linfocitos B/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Proteínas de Neoplasias/metabolismo , Animales , Linfocitos B/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Desarrollo Embrionario/fisiología , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/citología , Ratones , Proteínas de Neoplasias/deficiencia , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Linfocitos T/citología , Linfocitos T/fisiología , Transcripción Genética/fisiología
9.
EMBO J ; 25(2): 357-66, 2006 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-16407974

RESUMEN

The passage from proliferation to terminal differentiation is critical for normal development and is often perturbed in malignancies. To define the molecular mechanisms that govern this process during erythropoiesis, we have used tagging/proteomics approaches and characterized protein complexes nucleated by TAL-1/SCL, a basic helix-loop-helix transcription factor that specifies the erythrocytic lineage. In addition to known TAL-1 partners, GATA-1, E2A, HEB, LMO2 and Ldb1, we identify the ETO2 repressor as a novel component recruited to TAL-1 complexes through interaction with E2A/HEB. Ectopic expression and siRNA knockdown experiments in hematopoietic progenitor cells show that ETO2 actively represses erythroid TAL-1 target genes and governs the expansion of erythroid progenitors. At the onset of erythroid differentiation, a change in the stoichiometry of ETO2 within the TAL-1 complex activates the expression of known erythroid-specific TAL-1 target genes and of Gfi-1b and p21(Cip), encoding two essential regulators of erythroid cell proliferation. These results suggest that the dynamics of ETO2 recruitment within nuclear complexes couple cell proliferation to cell differentiation and determine the onset of terminal erythroid maturation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular , Eritropoyesis/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/fisiología , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Citometría de Flujo , Proteínas Fluorescentes Verdes , Células Madre Hematopoyéticas/metabolismo , Immunoblotting , Inmunoprecipitación , Ratones , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/metabolismo
10.
Blood ; 105(4): 1448-55, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15507521

RESUMEN

Growth factor independence-1B (Gfi-1B) is a transcription factor with a highly conserved transcriptional repressor snail-Gfi-1 (SNAG) domain and 6 zinc-finger domains at the N- and C-terminus, respectively. Disruption of the Gfi-1B gene is lethal in the embryo with failure to produce definitive enucleated erythrocytes. In this study, we analyzed the role of Gfi-1B in human erythropoiesis. We observed an increase of Gfi-1B expression during erythroid maturation of human primary progenitor cells. We studied the consequences of variations in Gfi-1B expression in 2 transformed cell lines (K562 and UT7 cells), as well as in primary CD36(+)/GPA(-) progenitors. A knock-down of Gfi-1B delayed the terminal differentiation of K562 and primary cells. Forced expression of Gfi-1B in UT7 and K562 cells led to an arrest of proliferation and an induction of erythroid differentiation. Enforced expression of Gfi-1B in primary cells at the colony-forming units-erythroid (CFU-E) stage led to a partial glycophorin A (GPA) induction after erythropoietin (EPO) withdrawal but failed to protect cells from apoptosis. Deletion of the SNAG repressor domain abolished Gfi-1B-induced erythroid maturation, strongly suggesting that Gfi-1B acts in the late stage of erythroid differentiation as a transcriptional repressor.


Asunto(s)
Diferenciación Celular/fisiología , Transformación Celular Neoplásica/patología , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/fisiología , Antígenos CD36/biosíntesis , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Regulación hacia Abajo/genética , Eritropoyetina/fisiología , Silenciador del Gen/fisiología , Humanos , Células K562 , Estructura Terciaria de Proteína/fisiología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/farmacología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transfección , Regulación hacia Arriba/genética
11.
Cell Signal ; 16(3): 355-63, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14687665

RESUMEN

In this report, we compared activation of NH2-terminal FLAG-labelled thrombopoietin receptor (Mpl) by anti-FLAG antibodies and by thrombopoietin (TPO). We found that anti-FLAG monoclonal antibodies M1 dimerize FLAG-labelled receptor and trigger proliferation of BaF3/FLAG-Mpl cells. In UT7/FLAG-Mpl cells, activation of the FLAG-Mpl receptor by low TPO concentrations triggered proliferation, while high concentrations triggered differentiation. Activation of FLAG-Mpl receptors in these cells by all tested concentrations of M1 antibodies induced proliferation but not differentiation. Low TPO concentrations induced similar to M1 antibodies level of Jak2, Stat3, Stat5 and Akt phosphorylation. In contrast, only TPO and not M1 antibodies activated Erks phosphorylation. Since the anti-FLAG antibodies do not react with the TPO binding site of the receptor, we hypothesize that they can trigger a distinct signal by dimerizing Mpl in a manner different from that induced by TPO.


Asunto(s)
Proteínas de la Leche , Proteínas de Neoplasias/metabolismo , Péptidos/química , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Péptidos/metabolismo , Transducción de Señal , Trombopoyetina/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Biomarcadores , Diferenciación Celular , División Celular , Línea Celular , Supervivencia Celular , Proteínas de Unión al ADN/metabolismo , Humanos , Janus Quinasa 2 , Megacariocitos/metabolismo , Ratones , Proteínas de Neoplasias/química , Oligopéptidos , Péptidos/inmunología , Fosforilación , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-akt , Receptores de Citocinas/química , Receptores de Trombopoyetina , Factor de Transcripción STAT3 , Factor de Transcripción STAT5 , Transactivadores/metabolismo
12.
Blood ; 103(3): 1059-68, 2004 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-14525786

RESUMEN

Acute myeloid leukemia (AML) is sustained by the extensive proliferation of leukemic stem and progenitor cells, which give rise to the population of leukemic blasts with defective differentiation and low proliferative capacity. We have recently shown that ligation of CD44, a cell surface molecule present on AML cells, with specific monoclonal antibodies (mAbs) inhibits their proliferation. However, its mechanism has not been investigated yet. Here, using the NB4 cell line as a model of proliferating human AML cells, and the A3D8 mAb to ligate CD44, we show for the first time that CD44 ligation stabilizes the cyclin-dependent kinase inhibitor p27(Kip1) (p27) protein, resulting in increased association with cyclin E/Cdk2 complexes and inhibition of their kinase activity. Moreover, using a p27 antisense vector, we provide direct evidence that p27 is the main mediator of cell growth arrest by CD44. CD44 ligation also leads to p27 accumulation in THP-1, KG1a, and HL60 cell lines and in primary leukemic cells, suggesting that this process is general in AML. Taken together, our present results suggest that CD44 is a new and efficient means to increase the expression of p27 in AML cells. Considering that elevated expression of p27 is a factor of good prognosis in AML, these results provide a new basis for developing CD44-targeted therapy in AML.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Receptores de Hialuranos/metabolismo , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Anticuerpos Monoclonales , Quinasas CDC2-CDC28/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Estabilidad de Medicamentos , Humanos , Ácido Hialurónico/farmacología , Técnicas In Vitro , Leucemia Mieloide Aguda/patología , Transfección , Proteínas Supresoras de Tumor/genética
13.
Blood ; 102(4): 1282-9, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12730112

RESUMEN

A defect in cell trafficking and chemotaxis plays an important role in the immune deficiency observed in Wiskott-Aldrich syndrome (WAS). In this report, we show that marrow cells from WAS protein (WASP)-deficient mice also have a defect in chemotaxis. Serial transplantation and competitive reconstitution experiments demonstrated that marrow cells, including hematopoietic progenitors and stem cells (HSCs), have decreased homing capacities that were associated with a defect in adhesion to collagen. During development, HSCs migrate from the liver to the marrow and the spleen, prompting us to ask if a defect in HSC homing during development may explain the skewed X-chromosome inactivation in WAS carriers. Preliminary evidence has shown that, in contrast to marrow progenitor cells, fetal liver progenitor cells from heterozygous females had a random X-chromosome inactivation. When fetal liver cells from WASP-carrier females were injected into irradiated recipients, a nonrandom inactivation of the X-chromosome was found at the level of hematopoietic progenitors and HSCs responsible for the short- and long-term hematopoietic reconstitution. Therefore, the mechanism of the skewed X-chromosomal inactivation observed in WAS carriers may be related to a migration defect of WASP-deficient HSCs.


Asunto(s)
Quimiotaxis/fisiología , Compensación de Dosificación (Genética) , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Proteínas/fisiología , Síndrome de Wiskott-Aldrich/genética , Actinas/antagonistas & inhibidores , Actinas/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Adhesión Celular/fisiología , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Colágeno/metabolismo , Citoesqueleto/metabolismo , Femenino , Fluorouracilo/farmacología , Glutatión Transferasa/metabolismo , Heterocigoto , Masculino , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas/metabolismo , Síndrome de Wiskott-Aldrich/mortalidad , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich , Proteína de Unión al GTP cdc42/metabolismo
14.
J Cell Sci ; 115(Pt 11): 2329-37, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12006617

RESUMEN

Thrombopoietin (TPO) plays a critical role in megakaryopoiesis through binding to its receptor Mpl. This involves activation of various intracellular signaling pathways, including phosphoinositide 3-kinase (PI3K) and the mitogen-activated protein kinase (MAPK) pathways. Their precise role in TPO-mediated proliferation, survival and differentiation is not fully understood. In the present study, we show that TPO induces different biological responses in Mpl-transduced BaF-3 cells, depending on the cell surface density of Mpl and the resulting activation level of signaling pathways. TPO mediates cell proliferation in cells expressing high levels of Mpl but only mediates survival without proliferation in cells expressing low levels of the receptor. By using the kinase inhibitors PD98059 and LY294002, we further showed that the activation level of the PI3K and MAPK p42/44 pathways is a determining factor for the proliferative effect. In cells expressing low levels of Mpl, the survival effect was strongly dependent on the activation level of the PI3K/AKT, but not the MAPK p42/44 pathway. Moreover, this effect was correlated with the phosphorylation level of BAD but not with the expression level of Bcl-X(L). However, PI3K pathway inhibition did not increase apoptosis when BaF-3 cells proliferated in response to TPO, indicating a compensating mechanism from other Mpl signaling pathways in this case.


Asunto(s)
División Celular/fisiología , Células Eucariotas/metabolismo , Proteínas de la Leche , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal/fisiología , Trombopoyetina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , División Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Células Eucariotas/citología , Células Eucariotas/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Ratones , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Receptores de Citocinas/efectos de los fármacos , Receptores de Citocinas/genética , Receptores de Trombopoyetina , Factor de Transcripción STAT5 , Transducción de Señal/efectos de los fármacos , Trombopoyetina/farmacología , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Proteína Letal Asociada a bcl
15.
Exp Hematol ; 30(2): 166-75, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11823052

RESUMEN

OBJECTIVE: Interaction of thrombopoietin (TPO) with its receptor c-Mpl is responsible for the formation of megakaryocytes and platelets. In humans, there are two major c-mpl molecules, MplP and MplK, which are generated by alternative splicing. In contrast to MplP, MplK has none of the intracellular sequences required for typical signal transduction but instead has a unique 27 amino acid sequence that is coded by intron 10. We tested to determine if MplK exerts a negative effect on TPO Mpl signal transduction by interfering with the normal homodimerization of MplP. MATERIALS AND METHODS: A cassette coding for MplK cDNA was introduced into parental and MplP-expressing BaF3 cells and TPO-mediated cell growth studied. RESULTS: Cells expressing MplK alone did not respond to TPO compared to cells that expressed MplP. When MplK was coexpressed with MplP on the cell surface of BaF3, no modification in cell growth was observed when compared to those expressing MplP alone. To determine if the normal homodimerization process was negatively influenced, two genetically engineered variants of c-Mpl, one lacking the box1 sequence and the other containing only the first nine amino acids of the intracellular domain, were introduced into MplP-expressing cells. In contrast to MplK, these mutants had a dominant negative effect on TPO-mediated cell growth. CONCLUSIONS: MplK does not influence TPO-mediated growth of Mpl-expressing cells. Our data suggest that the absence of a dominant negative effect of MplK most probably is due to the inability of MplK to dimerize with the MplP receptor.


Asunto(s)
Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Citocinas , Trombopoyetina/metabolismo , Animales , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , Ratones , Mutación , Unión Proteica , Conformación Proteica , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Receptores de Trombopoyetina , Transducción de Señal/efectos de los fármacos , Trombopoyetina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...