Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Thromb Haemost ; 15(11): 2188-2197, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28834196

RESUMEN

Essentials The Royal disease (RD) is a form of hemophilia B predicted to be caused by a splicing mutation. We generated an iPSC-based model of the disease allowing mechanistic studies at the RNA level. F9 mRNA analysis in iPSC-derived hepatocyte-like cells showed the predicted abnormal splicing. Mutated F9 mRNA level was very low but we also found traces of wild type transcripts. SUMMARY: Background The royal disease is a form of hemophilia B (HB) that affected many descendants of Queen Victoria in the 19th and 20th centuries. It was found to be caused by the mutation F9 c.278-3A>G. Objective To generate a physiological cell model of the disease and to study F9 expression at the RNA level. Methods Using fibroblasts from skin biopsies of a previously identified hemophilic patient bearing the F9 c.278-3A>G mutation and his mother, we generated induced pluripotent stem cells (iPSCs). Both the patient's and mother's iPSCs were differentiated into hepatocyte-like cells (HLCs) and their F9 mRNA was analyzed using next-generation sequencing (NGS). Results and Conclusion We demonstrated the previously predicted aberrant splicing of the F9 transcript as a result of an intronic nucleotide substitution leading to a frameshift and the generation of a premature termination codon (PTC). The F9 mRNA level in the patient's HLCs was significantly reduced compared with that of his mother, suggesting that mutated transcripts undergo nonsense-mediated decay (NMD), a cellular mechanism that degrades PTC-containing mRNAs. We also detected small proportions of correctly spliced transcripts in the patient's HLCs, which, combined with genetic variability in splicing and NMD machineries, could partially explain some clinical variability among affected members of the European royal families who had lifespans above the average. This work allowed the demonstration of the pathologic consequences of an intronic mutation in the F9 gene and represents the first bona fide cellular model of HB allowing the study of rare mutations at the RNA level.


Asunto(s)
Factor IX/genética , Hemofilia B/genética , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , ARN Mensajero/genética , Adolescente , Empalme Alternativo , Diferenciación Celular , Línea Celular , Factor IX/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Hemofilia B/sangre , Hemofilia B/diagnóstico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Fenotipo , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN
2.
Br J Surg ; 103(9): 1209-19, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27256140

RESUMEN

BACKGROUND: Portal vein embolization (PVE) is used routinely to prevent postoperative liver failure as a result of anticipated insufficient future liver remnant volume following resection. The authors have recently developed a technique for temporary PVE. The aim of this study was to assess the effect of repeated reversible PVE on hepatocyte proliferation and subsequent liver hypertrophy in rodents. METHODS: Four treatments were compared (n = 21 rats per group): single reversible PVE, two PVEs separated by 14 days, partial portal vein ligation or sham procedure. The feasibility and tolerance of the procedure were assessed. Volumetric imaging by CT was used to estimate the evolution of liver volumes. After death, the weight of liver lobes was measured and hepatocyte proliferation evaluated by immunostaining. RESULTS: Embolization of portal branches corresponding to 70 per cent of total portal flow was performed successfully in all animals. Repeated PVE induced additional hepatocyte proliferation. Repeated embolization resulted in superior hepatocyte proliferation in the non-occluded segments compared with portal vein ligation (31·1 versus 22·2 per cent; P = 0·003). The non-occluded to total liver volume ratio was higher in the repeated PVE group than in the single PVE and sham groups (P = 0·050 and P = 0·001 respectively). CONCLUSION: Repeated reversible PVE successfully induced additional hepatocyte proliferation and subsequent liver hypertrophy. Surgical relevance Portal vein embolization (PVE) is used routinely to prevent postoperative liver failure as a result of anticipated insufficient future liver remnant volume following resection. In the present study, a technique of repeated temporary PVE was developed in a rat model; this induced additional hepatocyte proliferation and an increase in liver volume compared with single embolization. This novel approach might help induce major hypertrophy of the future remnant liver, which could increase the rate of patients amenable to major liver resections.


Asunto(s)
Embolización Terapéutica/métodos , Hepatectomía , Regeneración Hepática , Hígado/crecimiento & desarrollo , Vena Porta , Animales , Estudios de Factibilidad , Ligadura , Hígado/diagnóstico por imagen , Hígado/cirugía , Masculino , Tamaño de los Órganos , Vena Porta/cirugía , Ratas , Ratas Wistar , Tomografía Computarizada por Rayos X
3.
J Thromb Haemost ; 14(7): 1470-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27148783

RESUMEN

UNLABELLED: Essentials A signaling role of glycoprotein (GP)Ibß is postulated but not formally demonstrated in platelets. Lentiviral-mediated rescue in knock-out mice can be used to evaluate GPIbß function in vivo. Transduction of the native subunit corrected the main defects associated with GPIb-IX deficiency Deletion of intracellular 159-170 segment increased thrombosis, 150-160 removal increased bleeding. SUMMARY: Background The platelet glycoprotein (GP)Ib-V-IX complex is required for normal hemostasis and megakaryopoiesis. A role in GPIb-dependent responses has been ascribed to the less well characterized GPIbß subunit using a specific antibody and GPIb-IX transfected cells. Objectives Our aim was to evaluate, in vivo, the role of the GPIbß in hemostasis and thrombosis. Methods GPIbß(null) Sca-1(+) progenitors transduced with viral particles harboring hGPIbß were transplanted into lethally irradiated GPIbß(-/-) recipient mice. Results hGPIbß transplanted into the bone marrow of GPIbß(null) mice rescued GPIb-IX expression in 97% of circulating platelets. These platelets efficiently bound von Willebrand factor (VWF) and extended filopodia on a VWF matrix, demonstrating the restoration of GPIb-dependent adhesive and signaling properties. These mice exhibited less severe macrothrombocytopenia and had normal tail bleeding times as compared with GPIbß(null) mice. This strategy was employed to manipulate and evaluate the role of the GPIbß intracellular domain. Removal of the membrane proximal segment (Δ(150-160) ) decreased GPIb-IX expression by 43%, confirming its involvement in receptor assembly and biosynthesis, and resulted in increased bleeding times and decreased thrombosis in a mechanical injury model in the aorta. On the other hand, deletion of the C-flanking 159-170 segment allowed normal GPIb-IX expression, VWF-dependent responses and bleeding times, but resulted in enhanced arterial thrombosis. Conclusion This pointed to a repressor role of GPIbß in thrombus formation in vivo that was not predicted in studies of heterologous cells. These results highlight the utility of this lentiviral strategy for the structure-function evaluation of GPIb-IX in platelets.


Asunto(s)
Síndrome de Bernard-Soulier/genética , Técnicas de Transferencia de Gen , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Animales , Aorta/metabolismo , Tiempo de Sangría , Plaquetas/metabolismo , Células de la Médula Ósea/citología , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Vectores Genéticos , Células HEK293 , Hemorragia , Hemostasis , Humanos , Lentivirus , Masculino , Megacariocitos/citología , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Dominios Proteicos , Trombosis/metabolismo , Transgenes , Factor de von Willebrand/metabolismo
4.
Leukemia ; 22(10): 1864-73, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18615109

RESUMEN

Spontaneous apoptosis of bone marrow erythroid precursors accounts for the anemia that characterizes most low-grade myelodysplastic syndromes (MDS). We have shown that death of these precursors involved the Fas-dependent activation of caspase-8. To explore the pathway leading from caspase-8 activation to apoptosis, we transduced MDS bone marrow CD34(+) cells with a lentivirus encoding wild-type (WT) or endoplasmic reticulum (ER)-targeted Bcl-2 protein before inducing their erythroid differentiation. Both WT-Bcl-2 and ER-targeted Bcl-2 prevented spontaneous and Fas-dependent apoptosis in MDS erythroid precursors. ER-targeted Bcl-2 inhibited mitochondrial membrane depolarization and cytochrome c release in MDS erythroid precursors undergoing apoptosis, indicating a role for the ER in the death pathway, upstream of the mitochondria. MDS erythroid precursors demonstrated elevated ER Ca(2+) stores and these stores remained unaffected by ER-targeted Bcl-2. The ER-associated protein Bcl-2-associated protein (BAP) 31 was cleaved by caspase-8 in MDS erythroid precursors undergoing apoptosis. The protective effect of ER-targeted Bcl-2 toward spontaneous and Fas-induced apoptosis correlated with inhibition of BAP31 cleavage. A protective effect of erythropoietin against Fas-induced BAP31 cleavage and apoptosis was observed. We propose that apoptosis of MDS erythroid precursors involves the ER, downstream of Fas and upstream of the mitochondria, through the cleavage of the ER-associated BAP31 protein.


Asunto(s)
Apoptosis , Retículo Endoplásmico/fisiología , Células Precursoras Eritroides/fisiología , Síndromes Mielodisplásicos/sangre , Receptor fas/fisiología , Anemia/etiología , Calcio/metabolismo , Inhibidores de Caspasas , Células Precursoras Eritroides/química , Eritropoyetina/uso terapéutico , Humanos , Proteínas de la Membrana/metabolismo , Mitocondrias/fisiología , Síndromes Mielodisplásicos/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteínas Proto-Oncogénicas c-bcl-2/fisiología
5.
J Thromb Haemost ; 4(2): 426-35, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16420576

RESUMEN

BACKGROUND AND AIM: As platelets are able to endocytose human immunodeficiency virus (HIV), we have investigated the fate of lentiviruses when endocytosed by human platelets and megakaryocytes (MK), and have characterized a specific receptor directly involved in this function. METHODS: Genetically modified (non-replicative) lentiviruses with an HIV envelope (HIV-e) or with a vesicular stomatitis virus protein G envelope (VSV-e) were alternatively used and their interaction with platelets and MK analyzed by electron microscopy (EM) and immunoEM. RESULTS: When incubated with platelets, HIV-e and VSV-e lentiviruses were internalized in specific endocytic vesicles and trafficked to the surface connected canalicular system (SCCS). Double immunolabeling for the viral P24 core protein and alpha-granule markers showed that lentiviruses were degraded in the SCCS after contact with alpha-granule proteins. In culture MK, lentiviruses were found in endocytic vesicles and accumulated in acid phosphatase-containing multivesicular bodies (MVB). The expression of the pathogen receptor dendritic cell-specific ICAM-grabbing non-integrin (DC-SIGN) was then demonstrated in platelets by flow cytometry, immunoEM and Western blot. Anti-DC-SIGN antibodies decreased HIV-e lentivirus internalization by platelets, showing that the receptor is functional. Specific signals for DC-SIGN protein and mRNA were also found in MK. CONCLUSION: This study indicates that platelets and MK can internalize lentiviruses in a pathway, which either provide a shelter to lentiviral particles or alternatively disrupts viral integrity. The receptor DC-SIGN is involved in this function.


Asunto(s)
Plaquetas/metabolismo , Plaquetas/virología , Moléculas de Adhesión Celular/sangre , Lectinas Tipo C/sangre , Lentivirus/patogenicidad , Megacariocitos/metabolismo , Megacariocitos/virología , Receptores de Superficie Celular/sangre , Anticuerpos Monoclonales , Secuencia de Bases , Plaquetas/ultraestructura , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , ADN Complementario/genética , Endocitosis , Expresión Génica , Genes env , Vectores Genéticos , VIH-1/genética , Células HeLa , Humanos , Técnicas In Vitro , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Lentivirus/genética , Megacariocitos/ultraestructura , Microscopía Electrónica , ARN Mensajero/sangre , ARN Mensajero/genética , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores Virales/sangre , Receptores Virales/genética , Virus de la Estomatitis Vesicular Indiana/genética
6.
J Thromb Haemost ; 2(12): 2231-40, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15613031

RESUMEN

The storage mechanism of endogenous secretory proteins in megakaryocyte alpha-granules is poorly understood. We have elected to study the granule storage of platelet factor 4 (PF4), a well-known platelet alpha-granule protein. The reporter protein green fluorescent protein (GFP), PF4, or PF4 fused to GFP (PF4-GFP), were transfected in the well-characterized mouse pituitary AtT20 cell line, and in the megakaryocytic leukemic DAMI cell line. These proteins were also transduced using a lentiviral vector, in human CD34+ cells differentiated into megakaryocytes in vitro. Intracellular localization of expressed proteins, and colocalization studies were achieved by laser scanning confocal microscopy and immuno-electronmicroscopy. In preliminary experiments, GFP, a non-secretory protein (no signal peptide), localized in the cytoplasm, while PF4-GFP colocalized with adrenocorticotropin hormone (ACTH)-containing granules in AtT20 cells. In the megakaryocytic DAMI cell line and in human megakaryocytes differentiated in vitro, PF4-GFP localized in alpha-granules along with the alpha granular protein von Willebrand factor (VWF). The signal peptide of PF4 was not sufficient to specify alpha-granule storage of PF4, since when PF4 signal peptide was fused to GFP (SP4-GFP), GFP was not stored into granules in spite of its efficient translocation to the ER-Golgi constitutive secretory pathway. We conclude that the PF4 storage pathway in alpha-granules is not a default pathway, but rather a regular granule storage pathway probably requiring specific sorting mechanisms. In addition PF4-GFP appears as an appropriate probe with which to analyze alpha-granule biogenesis and its alterations in the congenital defect gray platelet syndrome.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Factor Plaquetario 4/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Animales , Antígenos CD34/biosíntesis , Trastornos de las Plaquetas Sanguíneas/sangre , Trastornos de las Plaquetas Sanguíneas/congénito , Western Blotting , Línea Celular , Línea Celular Tumoral , Citoplasma/metabolismo , Cartilla de ADN/química , Sangre Fetal/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Inmunoprecipitación , Lentivirus/genética , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Microscopía Confocal , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Señales de Clasificación de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Trombina/metabolismo , Transfección , Factor de von Willebrand/metabolismo
7.
J Hematother Stem Cell Res ; 11(2): 327-36, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11983104

RESUMEN

Hematopoietic stem cells (HSC) are subject to great interest because of their medical importance and their biological properties. Therefore, the possibility of genetically modifying human HSC is a major concern in several inherited pathologies. In this study, we aimed to demonstrate that a murine oncoretroviral vector can transduce multipotential cord blood (CB) stem cells. Sorted CB CD34(+)CD38(low) cells were transduced with a Moloney-based MFG retroviral vector containing the coding sequence of the murine CD2 (mCD2). CD34(+)mCD2(+) cells were sorted by flow cytometry and cultured either in bulk or at one cell per well in culture conditions that allow differentiation along lymphoid (T, B, and NK) and myeloid (M) lineages. Phenotypic analysis of cells generated in culture showed that CD34(+)mCD2(+) cells could give rise to all lymphoid and myeloid progeny, indicating that the MFG/mCD2 vector had transduced progenitors of all tested lineages. Moreover, clonal cultures of 660 CD34(+)mCD2(+) cells showed that approximately 5% of these cells were able to generate both myeloid and lymphoid (B + NK) progenies; for 25% of them, this included the production of lymphoid T cells. We also demonstrate that transduced CD34(+)CD38(low) CB cells with lymphoid and myeloid potentials were capable of engraftment into the bone marrow (BM) of nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice during several months. These results show that MFG retroviral vectors can transduce multipotent (T, B, NK, M) human hematopoietic progenitors with in vivo repopulating activity.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Células Madre Multipotentes/metabolismo , Transducción Genética/métodos , Animales , Antígenos CD34 , Células de la Médula Ósea , Antígenos CD2/genética , Movimiento Celular , Genes Reporteros , Vectores Genéticos , Supervivencia de Injerto , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones SCID , Transducción Genética/normas
8.
Gene Ther ; 8(3): 190-8, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11313790

RESUMEN

Retroviral vectors have become the primary tool for gene delivery into hematopoietic cells, including T lymphocytes. Lentiviral vectors offer an advantage over Moloney murine leukemia virus (MuLV) vectors because of their ability to translocate across an intact nuclear membrane and integrate into the genome of nonproliferating cells. We have recently demonstrated that a central strand displacement event, controlled by the central polypurine tract (cPPT) and the central termination sequence (CTS), results in the formation of a central DNA flap which acts as a cis-determinant of HIV-1 genome nuclear import. Here, we show that insertion of this DNA determinant in a classical lentiviral vector resulted in a significantly higher level of transduction in activated T cells (51 +/- 12.7% versus 15 +/- 1.4%). CD4(+) and CD8(+) T cells were transduced at equivalent levels. Importantly, freshly isolated T cells stimulated only during the 12-h transduction period could be efficiently transduced with this new flap-containing lentiviral vector, but not with the parental lentiviral vector nor an MuLV vector. Transgene expression in the flap-containing lentiviral vector, under the control of either an internal cytomegalovirus or the elongation factor-1 alpha (EF1 alpha) promoter, was significant and expression remained elevated in resting T cells. Thus, this system allows stable expression of transgenes in T lymphocytes following a short ex vivo transduction protocol.


Asunto(s)
ADN Viral/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus/genética , Linfocitos T/inmunología , Adulto , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Técnicas de Cultivo de Célula , División Celular/genética , Humanos , Activación de Linfocitos/genética , Virus de la Leucemia Murina de Moloney/genética , Transducción Genética , Transgenes
9.
Mol Ther ; 3(4): 438-48, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11319904

RESUMEN

The recent development of lentivirus-derived vectors is an important breakthrough in gene transfer technology because these vectors allow transduction of nondividing cells such as hematopoietic stem cells (HSC), due to an active nuclear import of reverse-transcribed vector DNA. We recently demonstrated that addition of the central DNA flap of HIV-1 to an HIV-derived lentiviral vector strikingly increases transduction of CD34(+) cells. We now describe improvements of the transduction protocol designed to preserve HSC properties and two modifications of the previously described TRIP-CMV vector. First, deletion of the enhancer/promoter of the 3' LTR in the TRIP-CMV vector resulted in a safer vector (TRIPDeltaU3-CMV) with conserved transduction efficiency and increased EGFP transgene expression. Second, the original internal CMV promoter was replaced with the promoter for the ubiquitously expressed elongation factor 1alpha (EF1alpha). This promoter substitution resulted in a significantly more homogeneous expression of the EGFP transgene in all hematopoietic cell types, including CD34(+)-derived T lymphocytes, in which the CMV promoter was inactive, and NOD/SCID mouse repopulating cells. We thus present here an HIV-derived lentiviral vector, TRIPDeltaU3-EF1alpha, which can very efficiently transduce human cord blood HSC and results in high long-term transgene expression in CD34(+)-derived T, B, NK, and myeloid hematopoietic cells.


Asunto(s)
Antígenos CD34/biosíntesis , Sangre Fetal/metabolismo , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Linfocitos T/metabolismo , Transgenes , Animales , Citometría de Flujo , Vectores Genéticos , VIH/genética , Humanos , Ratones , Ratones SCID , Factor 1 de Elongación Peptídica/genética , Plásmidos/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Transcripción Genética , Transducción Genética
10.
Blood ; 96(13): 4103-10, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11110680

RESUMEN

Gene transfer in human hematopoietic stem cells (HSCs) has great potential for both gene therapy and the understanding of hematopoiesis. As HSCs have extensive proliferative capacities, stable gene transfer should include genomic integration of the transgene. Lentiviral vectors are now preferred to oncoretroviral vectors especially because they integrate in nondividing cells such as HSCs, thereby avoiding the use of prolonged cytokine stimulation. Human immunodeficiency virus type-1 (HIV-1) has evolved a complex reverse transcription strategy including a central strand displacement event controlled in cis by the central polypurine tract (cPPT) and the central termination sequence (CTS). This creates, at the center of HIV-1 linear DNA molecules, a 99-nucleotide-long plus-strand overlap, the DNA flap, which acts as a cis-determinant of HIV-1 genome nuclear import. The reinsertion of the DNA flap sequence in an HIV-derived lentiviral vector promotes a striking increase of gene transduction efficiency in human CD34(+) hematopoietic cells, and the complementation of the nuclear import defect present in the parental vector accounts for this result. In a short ex vivo protocol, the flap-containing vector allows efficient transduction of the whole hierarchy of human HSCs including both slow-dividing or nondividing HSCs that have multiple lymphoid and myeloid potentials and primitive cells with long-term engraftment ability in nonobese diabetic/severe combined immunodeficiency mice (NOD/SCID).


Asunto(s)
Núcleo Celular/metabolismo , ADN Complementario/metabolismo , ADN Viral/metabolismo , Vectores Genéticos/genética , VIH-1/genética , Células Madre Hematopoyéticas/metabolismo , Transfección/métodos , Adulto , Animales , Transporte Biológico , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/virología , División Celular , ADN Viral/química , Sangre Fetal/citología , Vectores Genéticos/metabolismo , Supervivencia de Injerto , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/virología , Humanos , Recién Nacido , Ratones , Ratones Endogámicos NOD , Ratones SCID , Reacción en Cadena de la Polimerasa , Especificidad de la Especie , Relación Estructura-Actividad , Trasplante Heterólogo
11.
Biochem Biophys Res Commun ; 276(2): 803-12, 2000 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-11027551

RESUMEN

To elucidate the role of helix-loop-helix (HLH) Id proteins in hematopoietic differentiation, we used a model of embryonic stem (ES) cell differentiation in vitro which gives access not only to hematopoietic myeloid progenitor cells but also to the more primitive blast colony-forming cell (BL-CFC), the in vitro equivalent of the hemangioblast that gives rise to blast cell colonies in the presence of VEGF. We first demonstrated that ES cell-derived blast cell colonies could be used as a model to study hematopoietic differentiation and maturation. We next established the expression profile of Id genes in this model. Transcripts of the four Id genes were present in ES cells. Id1, Id3 and Id4 gene expression was down-regulated during the development of blast cell colonies while that of Id2 was maintained. Thus, Id1, Id3, and Id4 proteins are candidates for being negative regulators of hematopoiesis in the model of hematopoietic ES cell differentiation in vitro.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Hematopoyesis/genética , Proteínas Represoras , Factores de Transcripción/genética , Animales , Línea Celular , Senescencia Celular , Embrión de Mamíferos/citología , Secuencias Hélice-Asa-Hélice , Células Madre Hematopoyéticas/citología , Humanos , Inmunofenotipificación , Proteína 1 Inhibidora de la Diferenciación , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA