Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cardiovasc Drugs Ther ; 27(2): 125-32, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23344929

RESUMEN

PURPOSE: Although amiodarone is recognized as the most effective anti-arrhythmic drug available, it has negative hemodynamic effects. Nano-sized liposomes can accumulate in and selectively deliver drugs to ischemic/reperfused (I/R) myocardium, which may augment drug effects and reduce side effects. We investigated the effects of liposomal amiodarone on lethal arrhythmias and hemodynamic parameters in an ischemia/reperfusion rat model. METHODS AND RESULTS: We prepared liposomal amiodarone (mean diameter: 113 ± 8 nm) by a thin-film method. The left coronary artery of experimental rats was occluded for 5 min followed by reperfusion. Ex vivo fluorescent imaging revealed that intravenously administered fluorescent-labeled nano-sized beads accumulated in the I/R myocardium. Amiodarone was measurable in samples from the I/R myocardium when liposomal amiodarone, but not amiodarone, was administered. Although the intravenous administration of amiodarone (3 mg/kg) or liposomal amiodarone (3 mg/kg) reduced heart rate and systolic blood pressure compared with saline, the decrease in heart rate or systolic blood pressure caused by liposomal amiodarone was smaller compared with a corresponding dose of free amiodarone. The intravenous administration of liposomal amiodarone (3 mg/kg), but not free amiodarone (3 mg/kg), 5 min before ischemia showed a significantly reduced duration of lethal arrhythmias (18 ± 9 s) and mortality (0 %) during the reperfusion period compared with saline (195 ± 42 s, 71 %, respectively). CONCLUSIONS: Targeting the delivery of liposomal amiodarone to ischemic/reperfused myocardium reduces the mortality due to lethal arrhythmia and the negative hemodynamic changes caused by amiodarone. Nano-size liposomes may be a promising drug delivery system for targeting I/R myocardium with cardioprotective agents.


Asunto(s)
Amiodarona/administración & dosificación , Antiarrítmicos/administración & dosificación , Arritmias Cardíacas/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Amiodarona/sangre , Amiodarona/farmacocinética , Animales , Antiarrítmicos/sangre , Antiarrítmicos/farmacocinética , Arritmias Cardíacas/sangre , Arritmias Cardíacas/fisiopatología , Modelos Animales de Enfermedad , Hemodinámica/efectos de los fármacos , Liposomas , Masculino , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/fisiopatología , Nanopartículas/administración & dosificación , Ratas , Ratas Wistar
2.
J Control Release ; 160(2): 274-80, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22020380

RESUMEN

Increased expression of heparin-binding epidermal growth factor-like growth factor (HB-EGF) is frequently observed in certain cancers such as ovarian and breast cancers, and this protein is a desirable target for drug delivery by a drug delivery system (DDS). In the present study, we developed novel immunoliposomes targeting HB-EGF for cancer therapy. The immunoliposomes significantly associated with Vero-H cells overexpressing HB-EGF compared with their binding to wild-type Vero cells, whereas liposomes without modification by the antibody did not associate with either type of cells. Moreover, enhanced uptake of the immunoliposomes into Vero-H cells was observed as well as that into MDA-MB-231 human breast cancer cells, which are known to highly express HB-EGF. These results suggest that HB-EGF mediates the binding and uptake of the immunoliposomes in HB-EGF-expressing cells. Next, we determined the therapeutic effect of these immunoliposomes encapsulating an anticancer drug on tumor-bearing mice. For this purpose, we prepared doxorubicin (DOX)-encapsulated immunoliposomes and injected them intravenously into mice bearing MDA-MB-231 cancer cells. As a result, these DOX-encapsulated immunoliposomes suppressed not only tumor progression but also tumor regression. In conclusion, our results indicate that anti-HB-EGF antibody-modified liposomes could be a useful DDS carrier for the treatment of HB-EGF-expressing cancers.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Inmunoglobulina G , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Animales , Antibióticos Antineoplásicos/uso terapéutico , Western Blotting , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Doxorrubicina/uso terapéutico , Femenino , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Liposomas , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Clin Invest ; 119(4): 852-64, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19287095

RESUMEN

Acute megakaryoblastic leukemia (AMKL) is a form of acute myeloid leukemia (AML) associated with a poor prognosis. The genetics and pathophysiology of AMKL are not well understood. We generated a knockin mouse model of the one twenty-two-megakaryocytic acute leukemia (OTT-MAL) fusion oncogene that results from the t(1;22)(p13;q13) translocation specifically associated with a subtype of pediatric AMKL. We report here that OTT-MAL expression deregulated transcriptional activity of the canonical Notch signaling pathway transcription factor recombination signal binding protein for immunoglobulin kappa J region (RBPJ) and caused abnormal fetal megakaryopoiesis. Furthermore, cooperation between OTT-MAL and an activating mutation of the thrombopoietin receptor myeloproliferative leukemia virus oncogene (MPL) efficiently induced a short-latency AMKL that recapitulated all the features of human AMKL, including megakaryoblast hyperproliferation and maturation block, thrombocytopenia, organomegaly, and extensive fibrosis. Our results establish that concomitant activation of RBPJ (Notch signaling) and MPL (cytokine signaling) transforms cells of the megakaryocytic lineage and suggest that specific targeting of these pathways could be of therapeutic value for human AMKL.


Asunto(s)
Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Leucemia Megacarioblástica Aguda/genética , Fusión de Oncogenes , Proteínas de Fusión Oncogénica/genética , Animales , Modelos Animales de Enfermedad , Hematopoyesis , Humanos , Leucemia Megacarioblástica Aguda/etiología , Leucemia Megacarioblástica Aguda/metabolismo , Leucemia Megacarioblástica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Receptores Notch/metabolismo , Receptores de Trombopoyetina/genética , Transducción de Señal , Transcripción Genética
4.
Diabetes Metab Res Rev ; 24(7): 585-90, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18727153

RESUMEN

BACKGROUND: In this study, we carried out bone marrow and foetal liver cell transplantation to determine if these cells could differentiate into pancreatic beta-cells or promote regeneration. METHODS: To exclude an artificial or immunological influence for induction of diabetes to recipients, Akita mice, which develop diabetes spontaneously,were used. In addition, we used mice harbouring the transgenic green fluorescent protein (GFP) reporter for insulin 1 gene as donors to mark donor-derived beta-cells. RESULTS: All transplanted Akita mice after intravenous injection showed full donor chimerism in peripheral blood analysis. Their diabetic state represented by blood glucose levels did not change after transplantation. In spite of examination of more than 200 islets in each group, we could not find GFP-positive cells in any of the recipients. CONCLUSIONS: Bone marrow cells or foetal liver cells do not differentiate to new pancreatic beta-cells or promote regeneration in Akita mice, a non-chemical or non-immune model of diabetes.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Células , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Islotes Pancreáticos/fisiología , Hígado/citología , Hígado/embriología , Animales , Modelos Animales de Enfermedad , Ratones , Regeneración
5.
J Exp Med ; 205(3): 611-24, 2008 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-18299398

RESUMEN

The zinc finger transcription factor GATA-1 requires direct physical interaction with the cofactor friend of GATA-1 (FOG-1) for its essential role in erythroid and megakaryocytic development. We show that in the mast cell lineage, GATA-1 functions completely independent of FOG proteins. Moreover, we demonstrate that FOG-1 antagonizes the fate choice of multipotential progenitor cells for the mast cell lineage, and that its down-regulation is a prerequisite for mast cell development. Remarkably, ectopic expression of FOG-1 in committed mast cell progenitors redirects them into the erythroid, megakaryocytic, and granulocytic lineages. These lineage switches correlate with transcriptional down-regulation of GATA-2, an essential mast cell GATA factor, via switching of GATA-1 for GATA-2 at a key enhancer element upstream of the GATA-2 gene. These findings illustrate combinatorial control of cell fate identity by a transcription factor and its cofactor, and highlight the role of transcriptional networks in lineage determination. They also provide evidence for lineage instability during early stages of hematopoietic lineage commitment.


Asunto(s)
Factor de Transcripción GATA1/antagonistas & inhibidores , Factor de Transcripción GATA2/antagonistas & inhibidores , Mastocitos/citología , Mastocitos/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factor de Transcripción GATA1/deficiencia , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/deficiencia , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Saco Vitelino/citología , Saco Vitelino/metabolismo
6.
Science ; 316(5826): 860-6, 2007 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-17495164

RESUMEN

Hematopoietic stem cells in the bone marrow give rise to lymphoid progenitors, which subsequently differentiate into B and T lymphocytes. Here we show that the proto-oncogene LRF plays an essential role in the B versus T lymphoid cell-fate decision. We demonstrate that LRF is key for instructing early lymphoid progenitors in mice to develop into B lineage cells by repressing T cell-instructive signals produced by the cell-fate signal protein, Notch. We propose a new model for lymphoid lineage commitment, in which LRF acts as a master regulator of the cell's determination of B versus T lineage.


Asunto(s)
Linfocitos B/citología , Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/citología , Linfopoyesis , Proto-Oncogenes , Receptores Notch/metabolismo , Linfocitos T/citología , Factores de Transcripción/genética , Animales , Linfocitos B/fisiología , Células de la Médula Ósea/citología , Linaje de la Célula , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Eliminación de Gen , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Transducción de Señal , Linfocitos T/fisiología , Timo/citología , Factores de Transcripción/fisiología
7.
Proc Natl Acad Sci U S A ; 104(14): 6001-6, 2007 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-17376872

RESUMEN

OTT1(RBM15) was originally described as a 5' translocation partner of the MAL(MKL1) gene in t(1,22)(p13;q13) infant acute mega karyocytic leukemia. OTT1 has no established physiological function, but it shares homology with the spen/Mint/SHARP family of proteins defined by three amino-terminal RNA recognition motifs and a carboxyl-terminal SPOC (Spen paralog and ortholog carboxyl-terminal) domain believed to act as a transcriptional repressor. To define the role of OTT1 in hematopoiesis and help elucidate the mechanism of t(1,22) acute megakaryocytic leukemia pathogenesis, a conditional allele of Ott1 was generated in mice. Deletion of Ott1 in adult mice caused a loss of peripheral B cells due to a block in pro/pre-B differentiation. There is myeloid and megakaryocytic expansion in spleen and bone marrow, an increase in the Lin(-)Sca-1(+)c-Kit(+) compartment that includes hematopoietic stem cells, and a shift in progenitor fate toward granulocyte differentiation. These data show a requirement for Ott1 in B lymphopoiesis, and inhibitory roles in the myeloid, megakaryocytic, and progenitor compartments. The ability of Ott1 to affect hematopoietic cell fate and expansion in multiple lineages is a novel attribute for a spen family member and delineates Ott1 from other known effectors of hematopoietic development. It is plausible that dysregulation of Ott1-dependent hematopoietic developmental pathways, in particular those affecting the megakaryocyte lineage, may contribute to OTT1-MAL-mediated leukemogenesis.


Asunto(s)
Proteínas de Drosophila/fisiología , Hematopoyesis/fisiología , Sistema Hematopoyético/fisiología , Proteínas de Unión al ARN/fisiología , Animales , Linfocitos B/fisiología , Células de la Médula Ósea/citología , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Proteínas de Drosophila/genética , Leucemia Megacarioblástica Aguda/etiología , Ratones , Ratones Mutantes , Ratones Transgénicos , Proteínas de Unión al ARN/genética , Bazo/citología
8.
Cell Stem Cell ; 1(4): 416-27, 2007 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-18371378

RESUMEN

A hierarchical hematopoietic development with myeloid versus lymphoid bifurcation has been proposed downstream of the multipotent progenitor (MPP) stage, based on prospective isolation of progenitors capable of generating only myeloerythroid cells (common myeloid progenitor, CMP) or only lymphocytes (common lymphoid progenitor, CLP). By utilizing GATA-1 and PU.1 transcription factor reporters, here we identified progenitor populations that are precursors for either CMPs or CLPs. Two independent populations expressing either GATA-1 or PU.1 resided within the CD34(+)Sca-1(+)c-Kit(+) MPP fraction. The GATA-1(+) MPP displayed potent myeloerythroid potential without giving rise to lymphocytes, whereas the PU.1(+) MPP showed granulocyte/monocyte/lymphoid-restricted progenitor activity without megakaryocyte/erythroid differentiation. Furthermore, GATA-1(+) and PU.1(+) MPPs possessed huge expansion potential and differentiated into the original CMPs and CLPs, respectively. Thus, the reciprocal activation of GATA-1 and PU.1 primarily organizes the hematopoietic lineage fate decision to form the earliest hematopoietic branchpoint that comprises isolatable myeloerythroid and myelolymphoid progenitor populations.


Asunto(s)
Linaje de la Célula , Células Precursoras Eritroides/citología , Factor de Transcripción GATA1/genética , Células Madre Hematopoyéticas/citología , Células Progenitoras Linfoides/citología , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Regulación hacia Arriba/genética , Animales , Antígenos CD34/metabolismo , Ataxina-1 , Ataxinas , Diferenciación Celular , Factor de Transcripción GATA1/metabolismo , Genes Reporteros , Linfocitos/citología , Megacariocitos/citología , Ratones , Modelos Biológicos , Células Madre Multipotentes/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo
9.
Genes Dev ; 20(21): 3010-21, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17079688

RESUMEN

The mechanism of lineage specification in multipotent stem cells has not been fully understood. We recently isolated progenitors with the eosinophil, basophil, or mast cell lineage potential, all of which originate from granulocyte/monocyte progenitors (GMPs). By using these prospectively purified progenitors, we show here that the expression timing of GATA-2 and CCAAT enhancer-binding protein alpha (C/EBPalpha) can differentially control their lineage commitment. The expression of GATA-2 instructed C/EBPalpha-expressing GMPs to commit exclusively into the eosinophil lineage, while it induced basophil and/or mast cell lineage commitment if C/EBPalpha was suppressed at the GMP stage. Furthermore, simply by switching the order of C/EBPalpha and GATA-2 transduction, even lymphoid-committed progenitors recaptured these developmental processes to be reprogrammed into each of these lineages. We propose that the order of expression of key transcription factors is critical for their interplay to selectively drive lineage specification programs, by which stem cells could generate multiple lineage cells in a hierarchical manner.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Factor de Transcripción GATA2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Factor de Transcripción GATA2/genética , Granulocitos/citología , Granulocitos/metabolismo , Células Madre Hematopoyéticas/citología , Mastocitos/citología , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo
10.
Mol Cell Biol ; 26(21): 8052-60, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16954383

RESUMEN

The proliferation of neutrophil granulocyte lineage is driven largely by granulocyte colony-stimulating factor (G-CSF) acting via the G-CSF receptors. In this study, we show that mice lacking cyclin D3, a component of the core cell cycle machinery, are refractory to stimulation by the G-CSF. Consequently, cyclin D3-null mice display deficient maturation of granulocytes in the bone marrow and have reduced levels of neutrophil granulocytes in their peripheral blood. The mutant mice are unable to mount a normal response to bacterial challenge and succumb to microbial infections. In contrast, the expansion of hematopoietic stem cells and lineage-committed myeloid progenitors proceeds relatively normally in mice lacking cyclin D3, revealing that the requirement for cyclin D3 function operates at later stages of neutrophil development. Importantly, we verified that this requirement is specific to cyclin D3, as mice lacking other G(1) cyclins (D1, D2, E1, or E2) display normal granulocyte counts. Our analyses revealed that in the bone marrow cells of wild-type mice, activation of the G-CSF receptor leads to upregulation of cyclin D3. Collectively, these results demonstrate that cyclin D3 is an essential cell cycle recipient of G-CSF signaling, and they provide a molecular link of how G-CSF-dependent signaling triggers cell proliferation.


Asunto(s)
Proliferación Celular , Ciclinas/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Neutrófilos/fisiología , Animales , Infecciones Bacterianas , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Ciclo Celular/fisiología , Linaje de la Célula , Ciclina D3 , Ciclinas/genética , Susceptibilidad a Enfermedades/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutropenia , Neutrófilos/citología , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Transducción de Señal/fisiología
11.
Blood ; 108(5): 1708-15, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16675706

RESUMEN

Most patients with acute promyelocytic leukemia (APL) express PML-RAR alpha, the fusion product of t(15;17)(q22;q11.2). Transgenic mice expressing PML-RAR alpha develop APL with long latency, low penetrance, and acquired cytogenetic abnormalities. Based on observations that 4% to 10% of APL patients harbor oncogenic ras mutations, we coexpressed oncogenic K-ras from its endogenous promoter with PML-RAR alpha to generate a short-latency, highly penetrant mouse model of APL. The APL disease was characterized by splenomegaly, leukocytosis, extramedullary hematopoiesis (EMH) in spleen and liver with an increased proportion of immature myeloperoxidase-expressing myeloid forms; transplantability to secondary recipients; and lack of cytogenetic abnormalities. Bone marrow cells showed enhanced self-renewal in vitro. This model establishes a role for oncogenic ras in leukemia pathogenesis and thus validates the oncogenic RAS signaling pathway as a potential target for therapeutic inhibition in leukemia patients. This mouse model should be useful for investigating signaling pathways that promote self-renewal in APL and for testing the in vivo efficacy of RAS signaling pathway inhibitors in conjunction with other targeted therapies such as ATRA (all trans retinoic acid) and arsenic trioxide.


Asunto(s)
Genes ras , Leucemia Promielocítica Aguda/fisiopatología , Proteínas de Fusión Oncogénica/genética , Animales , Médula Ósea/patología , Citometría de Flujo , Humanos , Cariotipificación , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/mortalidad , Leucemia Promielocítica Aguda/patología , Hígado/patología , Ratones , Ratones Transgénicos , Bazo/patología , Análisis de Supervivencia
12.
Proc Natl Acad Sci U S A ; 102(50): 18105-10, 2005 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-16330751

RESUMEN

Basophils and mast cells, which are selectively endowed with the high-affinity IgE receptor and mediate a range of adaptive and innate immune responses, have an unknown developmental relationship. Here, by evaluating the expression of the beta7 integrin, a molecule that is required for selective homing of mast cell progenitors (MCPs) to the periphery, we identified bipotent progenitors that are capable of differentiating into either cell type in the mouse spleen. These basophil/mast cell progenitors (BMCPs) gave rise to basophils and mast cells at the single-cell level and reconstituted both mucosal and connective tissue mast cells. We also identified the basophil progenitor (BaP) and the MCP in the bone marrow and the gastrointestinal mucosa, respectively. We further show that the granulocyte-related transcription factor CCAAT/enhancer-binding protein alpha (C/EBPalpha) plays a primary role in the fate decision of BMCPs, being expressed in BaPs but not in MCPs. Thus, circulating basophils and tissue mast cells share a common developmental stage at which their fate decision might be controlled principally by C/EBPalpha.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Mastocitos/citología , Animales , Células de la Médula Ósea , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Cadenas beta de Integrinas/metabolismo , Mucosa Intestinal/citología , Mastocitos/inmunología , Ratones , Ratones Mutantes , Ovalbúmina/inmunología , Bazo/citología , Trichinella/inmunología
13.
J Exp Med ; 201(12): 1891-7, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15955840

RESUMEN

Eosinophil lineage-committed progenitors (EoPs) are phenotypically isolatable in the steady-state murine bone marrow. Purified granulocyte/monocyte progenitors (GMPs) gave rise to eosinophils as well as neutrophils and monocytes at the single cell level. Within the short-term culture of GMPs, the eosinophil potential was found exclusively in cells activating the transgenic reporter for GATA-1, a transcription factor capable of instructing eosinophil lineage commitment. These GATA-1-activating cells possessed an IL-5Ralpha(+)CD34(+)c-Kit(lo) phenotype. Normal bone marrow cells also contained IL-5Ralpha(+)CD34(+)c-Kit(lo) EoPs that gave rise exclusively to eosinophils. EoPs significantly increased in number in response to helminth infection, suggesting that the EoP stage is physiologically involved in eosinophil production in vivo. EoPs expressed eosinophil-related genes, such as the eosinophil peroxidase and the major basic protein, but did not express basophil/mast cell-related mast cell proteases. The enforced retroviral expression of IL-5Ralpha in GMPs did not enhance the frequency of eosinophil lineage read-outs, whereas IL-5Ralpha(+) GMPs displayed normal neutrophil/monocyte differentiation in the presence of IL-5 alone. Thus, IL-5Ralpha might be expressed specifically at the EoP stage as a result of commitment into the eosinophil lineage. The newly identified EoPs could be the cellular target in the treatment of a variety of disorders mediated by eosinophils.


Asunto(s)
Células de la Médula Ósea/citología , Linaje de la Célula/fisiología , Eosinófilos/citología , Células Precursoras de Granulocitos/citología , Animales , Antígenos CD34/metabolismo , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Factores de Unión al ADN Específico de las Células Eritroides , Citometría de Flujo , Factor de Transcripción GATA1 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-5 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo , Transducción Genética
14.
Blood ; 106(5): 1590-600, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15914556

RESUMEN

The PU.1 transcription factor is a key regulator of hematopoietic development, but its role at each hematopoietic stage remains unclear. In particular, the expression of PU.1 in hematopoietic stem cells (HSCs) could simply represent "priming" of genes related to downstream myelolymphoid lineages. By using a conditional PU.1 knock-out model, we here show that HSCs express PU.1, and its constitutive expression is necessary for maintenance of the HSC pool in the bone marrow. Bone marrow HSCs disrupted with PU.1 in situ could not maintain hematopoiesis and were outcompeted by normal HSCs. PU.1-deficient HSCs also failed to generate the earliest myeloid and lymphoid progenitors. PU.1 disruption in granulocyte/monocyte-committed progenitors blocked their maturation but not proliferation, resulting in myeloblast colony formation. PU.1 disruption in common lymphoid progenitors, however, did not prevent their B-cell maturation. In vivo disruption of PU.1 in mature B cells by the CD19-Cre locus did not affect B-cell maturation, and PU.1-deficient mature B cells displayed normal proliferation in response to mitogenic signals including the cross-linking of surface immunoglobulin M (IgM). Thus, PU.1 plays indispensable and distinct roles in hematopoietic development through supporting HSC self-renewal as well as commitment and maturation of myeloid and lymphoid lineages.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Animales , Médula Ósea/metabolismo , Células Cultivadas , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Hígado/embriología , Hígado/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Transactivadores/deficiencia , Transactivadores/genética
15.
Blood ; 106(2): 494-504, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15784726

RESUMEN

Homozygous loss of function of Runx1 (Runt-related transcription factor 1 gene) during murine development results in an embryonic lethal phenotype characterized by a complete lack of definitive hematopoiesis. In light of recent reports of disparate requirements for hematopoietic transcription factors during development as opposed to adult hematopoiesis, we used a conditional gene-targeting strategy to effect the loss of Runx1 function in adult mice. In contrast with the critical role of Runx1 during development, Runx1 was not essential for hematopoiesis in the adult hematopoietic compartment, though a number of significant hematopoietic abnormalities were observed. Runx1 excision had lineage-specific effects on B- and T-cell maturation and pronounced inhibition of common lymphocyte progenitor production. Runx1 excision also resulted in inefficient platelet production. Of note, Runx1-deficient mice developed a mild myeloproliferative phenotype characterized by an increase in peripheral blood neutrophils, an increase in myeloid progenitor populations, and extramedullary hematopoiesis composed of maturing myeloid and erythroid elements. These findings indicate that Runx1 deficiency has markedly different consequences during development compared with adult hematopoiesis, and they provide insight into the phenotypic manifestations of Runx1 deficiency in hematopoietic malignancies.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Hematopoyesis/fisiología , Proteínas Proto-Oncogénicas/deficiencia , Factores de Transcripción/deficiencia , Animales , Secuencia de Bases , Subunidad alfa 2 del Factor de Unión al Sitio Principal , ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Hematopoyesis/genética , Tejido Linfoide/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Fenotipo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
16.
Cancer Cell ; 7(2): 179-91, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15710330

RESUMEN

The SH2-containing tyrosine phosphatase Shp2 (PTPN11) is required for growth factor and cytokine signaling. Germline Shp2 mutations cause Noonan Syndrome (NS), which is associated with increased risk of juvenile myelomonocytic leukemia (JMML). Somatic Shp2 mutations occur in sporadic JMML and other leukemias. We found that Shp2 mutants associated with sporadic leukemias transform murine bone marrow cells, whereas NS mutants are less potent in this assay. Transformation requires multiple domains within Shp2 and the Shp2 binding protein Gab2, and is associated with hyperactivation of the Erk, Akt, and Stat5 pathways. Mutant Shp2-transduced BM causes a fatal JMML-like disorder or, less commonly, lymphoproliferation. Shp2 mutants also cause myeloproliferation in Drosophila. Mek or Tor inhibitors potently inhibit transformation, suggesting new approaches to JMML therapy.


Asunto(s)
Leucemia/genética , Mutación , Proteínas Tirosina Fosfatasas/genética , Alelos , Animales , Animales Modificados Genéticamente , Células de la Médula Ósea/citología , Proliferación Celular , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Drosophila , Drosophila melanogaster , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Interleucina-3/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Leucemia/metabolismo , Ratones , Modelos Genéticos , Neoplasias Experimentales , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Retroviridae/genética , Transducción de Señal , Factores de Tiempo
17.
Immunity ; 21(6): 853-63, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15589173

RESUMEN

The transcription factor C/EBP alpha is required for granulopoiesis and frequently disrupted in human acute myeloid leukemia (AML). Here, we show disruption of C/EBP alpha blocks the transition from the common myeloid to the granulocyte/monocyte progenitor but is not required beyond this stage for terminal granulocyte maturation. C/EBP alpha-deficient hematopoietic stem cells (HSCs) have increased expression of Bmi-1 and enhanced competitive repopulating activity. Bone marrow in adult C/EBP alpha-deficient mice was filled with myeloblasts, similar to human AML, supporting the notion that disruption of C/EBP alpha cooperates with other events in the development of leukemia. Therefore, C/EBP alpha is not only essential for granulocyte development but, in addition, is a regulator of hematopoietic stem cell activity.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/deficiencia , Diferenciación Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Envejecimiento/fisiología , Animales , Recuento de Células Sanguíneas , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Feto/metabolismo , Eliminación de Gen , Granulocitos/citología , Granulocitos/metabolismo , Hematopoyesis , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Hígado/citología , Hígado/metabolismo , Ratones , Ratones Noqueados
18.
Cancer Cell ; 6(6): 587-96, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15607963

RESUMEN

To better understand the origin of leukemic stem cells, we tested the hypothesis that all leukemia oncogenes could transform committed myeloid progenitor cells lacking the capacity for self-renewal, as has recently been reported for MLL-ENL. Flow-sorted populations of common myeloid progenitors and granulocyte-monocyte progenitors were transduced with the oncogenes MOZ-TIF2 and BCR-ABL, respectively. MOZ-TIF2-transduced progenitors could be serially replated in methylcellulose cultures and continuously propagated in liquid culture, and resulted in an acute myeloid leukemia in vivo that could be serially transplanted. In contrast, BCR-ABL transduction conferred none of these properties to hematopoietic progenitors. These data demonstrate that some, but not all, leukemia oncogenes can confer properties of leukemic stem cells to hematopoietic progenitors destined to undergo apoptotic cell death.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes abl/fisiología , Células Madre Hematopoyéticas/patología , Proteínas de Fusión Oncogénica/fisiología , Enfermedad Aguda , Animales , Southern Blotting , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular/genética , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/patología , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Genes abl/genética , Células Precursoras de Granulocitos/metabolismo , Células Precursoras de Granulocitos/patología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Interleucina-3/farmacología , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas de Fusión Oncogénica/genética
19.
Immunity ; 21(1): 43-53, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15345219

RESUMEN

The developmental origin of type I interferon (IFN)-producing plasmacytoid dendritic cells (PDCs) is controversial. In particular, the rearrangement of immunoglobulin heavy chain (IgH) genes in murine PDCs and the expression of pre-T cell receptor alpha (pTalpha) gene by human PDCs were proposed as evidence for their "lymphoid" origin. Here we demonstrate that PDCs capable of IFN production develop efficiently from both myeloid- and lymphoid-committed progenitors. Rearranged IgH genes as well as RAG transcripts were found in both myeloid- and lymphoid-derived PDCs. The human pTalpha transgenic reporter was activated in both myeloid- and lymphoid-derived PDCs at a level comparable to pre-T cells. PDCs were the only cell population that activated murine RAG1 knockin and human pTalpha transgenic reporters outside the lymphoid lineage. These results highlight a unique developmental program of PDCs that distinguishes them from other cell types including conventional dendritic cells.


Asunto(s)
Células de la Médula Ósea/fisiología , Células Dendríticas/fisiología , Células Madre Hematopoyéticas/fisiología , Células Plasmáticas/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Reordenamiento Génico de Cadena Pesada de Linfocito B , Proteínas de Homeodominio/metabolismo , Humanos , Interferón Tipo I/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta
20.
Cell ; 118(4): 477-91, 2004 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-15315760

RESUMEN

D-type cyclins (cyclins D1, D2, and D3) are regarded as essential links between cell environment and the core cell cycle machinery. We tested the requirement for D-cyclins in mouse development and in proliferation by generating mice lacking all D-cyclins. We found that these cyclin D1(-/-)D2(-/-)D3(-/-) mice develop until mid/late gestation and die due to heart abnormalities combined with a severe anemia. Our analyses revealed that the D-cyclins are critically required for the expansion of hematopoietic stem cells. In contrast, cyclin D-deficient fibroblasts proliferate nearly normally but show increased requirement for mitogenic stimulation in cell cycle re-entry. We found that the proliferation of cyclin D1(-/-)D2(-/-)D3(-/-) cells is resistant to the inhibition by p16(INK4a), but it critically depends on CDK2. Lastly, we found that cells lacking D-cyclins display reduced susceptibility to the oncogenic transformation. Our results reveal the presence of alternative mechanisms that allow cell cycle progression in a cyclin D-independent fashion.


Asunto(s)
Ciclinas/fisiología , Regulación del Desarrollo de la Expresión Génica , Animales , Northern Blotting , Western Blotting , Quinasas CDC2-CDC28/metabolismo , Ciclo Celular , División Celular , Transformación Celular Neoplásica , Ciclina A/metabolismo , Ciclina D1/genética , Ciclina D2 , Ciclina D3 , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Embrión de Mamíferos/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Metilcelulosa/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Fenotipo , Unión Proteica , Células Madre/citología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...