Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Infect Immun ; 87(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30455199

RESUMEN

Granulocyte colony-stimulating factor receptor (G-CSFR), encoded by the CSF3R gene, represents a major regulator of neutrophil production and function in mammals, with inactivating extracellular mutations identified in a cohort of neutropenia patients unresponsive to G-CSF treatment. This study sought to elucidate the role of the zebrafish G-CSFR by generating mutants harboring these inactivating extracellular mutations using genome editing. Zebrafish csf3r mutants possessed significantly decreased numbers of neutrophils from embryonic to adult stages, which were also functionally compromised, did not respond to G-CSF, and displayed enhanced susceptibility to bacterial infection. The study has identified an important role for the zebrafish G-CSFR in maintaining the number and functionality of neutrophils throughout the life span and created a bona fide zebrafish model of nonresponsive neutropenia.


Asunto(s)
Neutropenia/fisiopatología , Neutrófilos/fisiología , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Edición Génica , Factor Estimulante de Colonias de Granulocitos , Células Mieloides/citología , Neutropenia/patología , Neutrófilos/citología , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Pez Cebra/embriología
2.
Cytokine ; 78: 69-78, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26687628

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is a pleiotropic cytokine best known for its role in promoting the generation and function of neutrophils. G-CSF is also found to be involved in macrophage generation and immune regulation; however, its in vivo role in immune homeostasis is largely unknown. Here, we examined the role of G-CSF in dextran sulfate sodium (DSS)-induced acute colitis using G-CSF receptor-deficient (G-CSFR(-/-)) mice. Mice were administered with 1.5% DSS in drinking water for 5days, and the severity of colitis was measured for the next 5days. GCSFR(-/-) mice were more susceptible to DSS-induced colitis than G-CSFR(+/+) or G-CSFR(-/+) mice. G-CSFR(-/-) mice harbored less F4/80(+) macrophages, but a similar number of neutrophils, in the intestine. In vitro, bone marrow-derived macrophages prepared in the presence of both G-CSF and macrophage colony-stimulating factor (M-CSF) (G-BMDM) expressed higher levels of regulatory macrophage markers such as programmed death ligand 2 (PDL2), CD71 and CD206, but not in arginase I, transforming growth factor (TGF)-ß, Ym1 (chitinase-like 3) and FIZZ1 (found in inflammatory zone 1), and lower levels of inducible nitric oxide synthase (iNOS), CD80 and CD86 than bone marrow-derived macrophages prepared in the presence of M-CSF alone (BMDM), in response to interleukin (IL)-4/IL-13 and lipopolysaccharide (LPS)/interferon (IFN)-γ, respectively. Adoptive transfer of G-BMDM, but not BMDM, protected G-CSFR(-/-) mice from DSS-induced colitis, and suppressed expression of tumor necrosis factor (TNF)-α, IL-1ß and iNOS in the intestine. These results suggest that G-CSF plays an important role in preventing colitis, likely through populating immune regulatory macrophages in the intestine.


Asunto(s)
Colitis/inmunología , Colitis/prevención & control , Factor Estimulante de Colonias de Granulocitos/fisiología , Homeostasis , Intestinos/inmunología , Macrófagos/fisiología , Traslado Adoptivo , Animales , Células Cultivadas , Colitis/inducido químicamente , Sulfato de Dextran , Interleucina-13/inmunología , Interleucina-1beta/metabolismo , Intestinos/citología , Intestinos/fisiología , Lipopolisacáridos/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
Bull Math Biol ; 78(12): 2304-2357, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27324993

RESUMEN

We develop a physiological model of granulopoiesis which includes explicit modelling of the kinetics of the cytokine granulocyte colony-stimulating factor (G-CSF) incorporating both the freely circulating concentration and the concentration of the cytokine bound to mature neutrophils. G-CSF concentrations are used to directly regulate neutrophil production, with the rate of differentiation of stem cells to neutrophil precursors, the effective proliferation rate in mitosis, the maturation time, and the release rate from the mature marrow reservoir into circulation all dependent on the level of G-CSF in the system. The dependence of the maturation time on the cytokine concentration introduces a state-dependent delay into our differential equation model, and we show how this is derived from an age-structured partial differential equation model of the mitosis and maturation and also detail the derivation of the rest of our model. The model and its estimated parameters are shown to successfully predict the neutrophil and G-CSF responses to a variety of treatment scenarios, including the combined administration of chemotherapy and exogenous G-CSF. This concomitant treatment was reproduced without any additional fitting to characterize drug-drug interactions.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/fisiología , Hematopoyesis/fisiología , Neutrófilos/citología , Neutrófilos/fisiología , Animales , Retroalimentación Fisiológica , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacocinética , Hematopoyesis/efectos de los fármacos , Humanos , Conceptos Matemáticos , Ratones , Ratones Noqueados , Modelos Biológicos , Neutrófilos/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología
4.
J Exp Med ; 189(4): 683-92, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9989983

RESUMEN

In approximately 20% of cases of severe congenital neutropenia (SCN), mutations are found in the gene encoding the granulocyte colony-stimulating factor receptor (G-CSF-R). These mutations introduce premature stop codons, which result in truncation of 82-98 COOH-terminal amino acids of the receptor. SCN patients who develop secondary myelodysplastic syndrome and acute myeloid leukemia almost invariably acquired a GCSFR mutation, suggesting that this genetic alteration represents a key step in leukemogenesis. Here we show that an equivalent mutation targeted in mice (gcsfr-Delta715) results in the selective expansion of the G-CSF- responsive progenitor (G-CFC) compartment in the bone marrow. In addition, in vivo treatment of gcsfr-Delta715 mice with G-CSF results in increased production of neutrophils leading to a sustained neutrophilia. This hyperproliferative response to G-CSF is accompanied by prolonged activation of signal transducer and activator of transcription (STAT) complexes and extended cell surface expression of mutant receptors due to defective internalization. In view of the continuous G-CSF treatment of SCN patients, these data provide insight into why progenitor cells expressing truncated receptors clonally expand in vivo, and why these cells may be targets for additional genetic events leading to leukemia.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Síndromes de Inmunodeficiencia/inmunología , Leucemia Mieloide/genética , Activación de Linfocitos/efectos de los fármacos , Proteínas de la Leche , Neutropenia/genética , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Enfermedad Aguda , Animales , Médula Ósea/patología , Diferenciación Celular/genética , División Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Ensayo de Unidades Formadoras de Colonias , Contraindicaciones , Proteínas de Unión al ADN/metabolismo , Endocitosis , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Células Madre Hematopoyéticas/patología , Humanos , Síndromes de Inmunodeficiencia/genética , Cinética , Leucemia Mieloide/inducido químicamente , Ratones , Ratones Transgénicos , Neutropenia/congénito , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología , Factor de Transcripción STAT1 , Factor de Transcripción STAT5 , Eliminación de Secuencia , Organismos Libres de Patógenos Específicos , Transactivadores/metabolismo
5.
J Exp Med ; 188(6): 1173-84, 1998 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-9743535

RESUMEN

Cytokines stimulate granulopoiesis through signaling via receptors whose expression is controlled by lineage-specific transcription factors. Previously, we demonstrated that granulocyte colony-stimulating factor (G-CSF) receptor mRNA was undetectable and granulocyte maturation blocked in CCAAT enhancer binding protein alpha (C/EBPalpha)-deficient mice. This phenotype is distinct from that of G-CSF receptor-/- mice, suggesting that other genes are likely to be adversely affected by loss of C/EBPalpha. Here we demonstrate loss of interleukin 6 (IL-6) receptor and IL-6-responsive colony-forming units (CFU-IL6) in C/EBPalpha-/- mice. The observed failure of granulopoiesis could be rescued by the addition of soluble IL-6 receptor and IL-6 or by retroviral transduction of G-CSF receptors, demonstrating that loss of both of these receptors contributes to the absolute block in granulocyte maturation observed in C/EBPalpha-deficient hematopoietic cells. The results of these and other studies suggest that additional C/EBPalpha target genes, possibly other cytokine receptors, are also important for the block in granulocyte differentiation observed in vivo in C/EBPalpha-deficient mice.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Granulocitos/fisiología , Hematopoyesis , Proteínas Nucleares/fisiología , Receptores de Factor Estimulante de Colonias de Granulocito/biosíntesis , Receptores de Interleucina-6/biosíntesis , Factores de Transcripción/fisiología , Regulación hacia Arriba/fisiología , Animales , Proteínas Potenciadoras de Unión a CCAAT , Diferenciación Celular/genética , Ensayo de Unidades Formadoras de Colonias , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Sinergismo Farmacológico , Elementos de Facilitación Genéticos , Feto , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-6/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/fisiología , Ratones , Ratones Noqueados , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Receptores de Factor Estimulante de Colonias de Granulocito/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Receptores de Interleucina-6/antagonistas & inhibidores , Receptores de Interleucina-6/deficiencia , Receptores de Interleucina-6/genética , Solubilidad , Factores de Transcripción/genética , Regulación hacia Arriba/genética
6.
J Clin Invest ; 102(3): 483-92, 1998 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9691084

RESUMEN

The role of mutations of the granulocyte colony-stimulating factor receptor (G-CSFR) in the pathogenesis of severe congenital neutropenia (SCN) and the subsequent development of acute myeloid leukemia (AML) is controversial. Mice carrying a targeted mutation of their G-CSFR that reproduces the mutation found in a patient with SCN and AML have been generated. The mutant G-CSFR allele is expressed in a myeloid-specific fashion at levels comparable to the wild-type allele. Mice heterozygous or homozygous for this mutation have normal levels of circulating neutrophils and no evidence for a block in myeloid maturation, indicating that resting granulopoiesis is normal. However, in response to G-CSF treatment, these mice demonstrate a significantly greater fold increase in the level of circulating neutrophils. This effect appears to be due to increased neutrophil production as the absolute number of G-CSF-responsive progenitors in the bone marrow and their proliferation in response to G-CSF is increased. Furthermore, the in vitro survival and G-CSF-dependent suppression of apoptosis of mutant neutrophils are normal. Despite this evidence for a hyperproliferative response to G-CSF, no cases of AML have been detected to date. These data demonstrate that the G-CSFR mutation found in patients with SCN is not sufficient to induce an SCN phenotype or AML in mice.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Hematopoyesis/efectos de los fármacos , Neutropenia/genética , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Enfermedad Aguda , Animales , Apoptosis/efectos de los fármacos , Médula Ósea/patología , División Celular/efectos de los fármacos , Susceptibilidad a Enfermedades , Femenino , Genotipo , Humanos , Leucemia Mieloide/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndromes Mielodisplásicos/etiología , Neutropenia/congénito , Neutropenia/patología , Neutrófilos/patología , Receptores de Factor Estimulante de Colonias de Granulocito/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocito/genética
7.
Nat Commun ; 6: 6745, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25865621

RESUMEN

Duchenne muscular dystrophy (DMD) is a chronic and life-threatening disease that is initially supported by muscle regeneration but eventually shows satellite cell exhaustion and muscular dysfunction. The life-long maintenance of skeletal muscle homoeostasis requires the satellite stem cell pool to be preserved. Asymmetric cell division plays a pivotal role in the maintenance of the satellite cell pool. Here we show that granulocyte colony-stimulating factor receptor (G-CSFR) is asymmetrically expressed in activated satellite cells. G-CSF positively affects the satellite cell population during multiple stages of differentiation in ex vivo cultured fibres. G-CSF could be important in developing an effective therapy for DMD based on its potential to modulate the supply of multiple stages of regenerated myocytes. This study shows that the G-CSF-G-CSFR axis is fundamentally important for long-term muscle regeneration, functional maintenance and lifespan extension in mouse models of DMD with varying severities.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Regeneración/efectos de los fármacos , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/metabolismo , Longevidad/efectos de los fármacos , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Células Musculares/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Proteína MioD/genética , Proteína MioD/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Cultivo Primario de Células , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Regeneración/genética , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Transducción de Señal , Células Madre/metabolismo , Células Madre/patología
8.
Placenta ; 22(6): 609-12, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11440552

RESUMEN

Administration of granulocyte colony stimulating factor (G-CSF), a haematopoietic growth factor, to pregnant rats increases neutrophil production in the pups. The mechanism for the placental transfer is unknown, but it has been speculated to involve the placental G-CSF receptor (G-CSFR). The purpose of this study was to test that hypothesis. Pregnant mice were treated with a single subcutaneous dose of 50 microg/kg recombinant human G-CSF (rhG-CSF). Mice with an intact G-CSFR ("wild type", WT) and those with a homozygous deletion in the G-CSFR gene (G-CSFR deficient, "knock-out", KO) were studied. At intervals after injection, fetuses were delivered and maternal blood, amniotic fluid (AF) and fetal blood collected. G-CSF concentrations were measured using an enzyme linked immunosorbent assay specific for human G-CSF. Thirty minutes after injection, G-CSF was measurable in the AF (167+/-50 versus 445+/-217 pg/ml, mean+/-sem, WT versus KO) and fetal plasma (774+/-673 versus 427+/-121 pg/ml, WT versus KO). Peak concentrations occurred 2 h after injection in WT dams (572 542+/-41 262 pg/ml) and 4 h in KO dams (616 100+/-96 300 pg/ml). Therefore, in mice, a functional G-CSFR is not essential for the transfer of rhG-CSF from pregnant dams to their fetuses.


Asunto(s)
Sangre Fetal/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Placenta/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología , Líquido Amniótico/metabolismo , Animales , Femenino , Eliminación de Gen , Factor Estimulante de Colonias de Granulocitos/sangre , Humanos , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Proteínas Recombinantes/metabolismo
9.
Leuk Res ; 24(1): 11-7, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10634640

RESUMEN

The CBFA2 gene on chromosome band 21q22 is one of the most commonly translocated genes in leukemia. As with other translocations, those involving CBFA2 are associated with specific disease phenotypes. Only one of the different translocations involving CBFA2, the t(12;21), has been associated with a non-myeloid lineage. Several different CBFA2 fusion transcripts were expressed in the myeloid 32Dcl3 cell line, and show that unlike the myeloid specific fusion transcripts, the lymphoid specific ETV6/CBFA2 transcript is not compatible with myeloid cell differentiation. It is shown that myeloid cells expressing the ETV6/CBFA2 transcript undergo apoptosis in response to a G-CSF differentiation signal. The molecular differences in the cells we studied are characterized using Western blot analysis to show that t(12;21) expressing cells fail to express the G-CSF receptor.


Asunto(s)
Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 21/genética , Proteínas de Unión al ADN , Leucemia Mieloide/genética , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Factores de Transcripción/genética , Translocación Genética , Enfermedad Aguda , Apoptosis/efectos de los fármacos , Western Blotting , Diferenciación Celular , División Celular , Linaje de la Célula , Cromosomas Humanos Par 12/ultraestructura , Cromosomas Humanos Par 21/ultraestructura , Subunidad alfa 2 del Factor de Unión al Sitio Principal , ADN Complementario/genética , Citometría de Flujo , Regulación Leucémica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Leucemia Mieloide/patología , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/química , Peroxidasa/análisis , Estructura Terciaria de Proteína , Proteína 1 Compañera de Translocación de RUNX1 , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Eliminación de Secuencia , Transcripción Genética
10.
Ann N Y Acad Sci ; 938: 305-20; discussion 320-1, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11458519

RESUMEN

Mutations in the genes of hematopoietic growth factor receptors as a cause of congenital cytopenia, such as congenital amegakaryocytic thrombocytopenia (CAMT) or severe congenital neutropenia (CN), are discussed. There are striking differences in the relevance of receptor mutations in these diseases. CAMT is a rare disease characterized by severe hypomegakaryocytic thrombocytopenia during the first years of life that develops into pancytopenia in later childhood. In patients with CAMT, we found inherited mutations in c-mpl, the gene coding for the thrombopoietin receptor, in 8 out of 8 cases. The type of mutation seems to correlate with the clinical course seen in the patients. Functional studies demonstrated defective thrombopoietin (TPO) reactivity in hematopoietic progenitor cells and platelets in CAMT patients. CN is a group of hematopoietic disorders characterized by profound, absolute neutropenia due to a maturation arrest of myeloid progenitor cells. About 10% of all patients develop secondary MDS/leukemia. The malignant progression is associated with acquired nonsense mutations within the G-CSF receptor gene that lead to the truncation of the carboxy-terminal cytoplasmic domain of the receptor protein involved in maturation of myeloid progenitor cells. This seems to be one important step in leukemogenesis in CN patients. CAMT is caused by inherited mutations in c-mpl, the gene for the thrombopoietin receptor, which lead to reduced or absent reactivity to TPO. In contrast, mutations in the G-CSF receptor in CN are acquired and are most probably connected with progression of the neutropenia into MDS/leukemia as a result of a loss of differentiation signaling.


Asunto(s)
Mutación , Proteínas de Neoplasias , Neutropenia/genética , Proteínas Proto-Oncogénicas/genética , Receptores de Citocinas , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Trombocitopenia/genética , Animales , Médula Ósea/patología , Diferenciación Celular , Transformación Celular Neoplásica/genética , Codón sin Sentido , Análisis Mutacional de ADN , Progresión de la Enfermedad , Factor Estimulante de Colonias de Granulocitos/deficiencia , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/fisiología , Humanos , Leucemia Mieloide/etiología , Megacariocitos/patología , Ratones , Ratones Noqueados , Mutación Missense , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/patología , Neutropenia/congénito , Pancitopenia/etiología , Pancitopenia/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Trombopoyetina , Transducción de Señal/genética , Síndrome , Trombocitopenia/congénito , Trombocitopenia/patología , Trombopoyetina/fisiología
11.
Hematol Oncol Clin North Am ; 27(1): 61-73, viii, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23351988

RESUMEN

Following activation by their cognate ligands, cytokine receptors undergo intracellular routing toward lysosomes, where they are degraded. This review focuses on the signaling function of the G-CSFR in relation to the dynamics of endosomal routing of the G-CSFR. Mechanisms involving receptor lysine ubiquitination and redox-controlled phosphatase activities are discussed. Specific attention is paid to the consequences of G-CSFR mutations, acquired in patients with severe congenital neutropenias who receive G-CSF therapy, particularly in the context of leukemic transformation, a major clinical complication of the disease.


Asunto(s)
Neutropenia/congénito , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Transducción de Señal , Animales , Transformación Celular Neoplásica/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Progresión de la Enfermedad , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Leucemia/etiología , Lisosomas/metabolismo , Mutación , Neutropenia/tratamiento farmacológico , Neutropenia/genética , Neutropenia/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Ubiquitinación
12.
Hematol Oncol Clin North Am ; 27(1): 75-88, viii, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23351989

RESUMEN

Several signaling systems downstream of G-CSFR have been identified that are defective or hyperactivated in myeloid cells of patients with congenital neutropenia: severely reduced expression of myeloid-specific transcription factors LEF-1 and C/EBPα, severely reduced expression and functions of HCLS1 protein, severely reduced expression of neutrophil elastase protein, dramatic compensatory up-regulation of the NAMPT/NAD(+)/SIRT pathway leading to continuous activation of emergency granulopoiesis via the transcription factor C/EBPß, and hyperactivation of STAT5 protein by tyrosine phosphorylation.


Asunto(s)
Neutropenia/congénito , Neutropenia/genética , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Sanguíneas/genética , Proteína alfa Potenciadora de Unión a CCAAT/genética , Diferenciación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Regulación de la Expresión Génica , Humanos , Elastasa de Leucocito/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Mutación , Células Mieloides/metabolismo , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Mielopoyesis/genética , Neutropenia/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia
13.
J Exp Med ; 208(2): 251-60, 2011 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-21282380

RESUMEN

Granulocyte colony-stimulating factor (G-CSF), the prototypical mobilizing cytokine, induces hematopoietic stem and progenitor cell (HSPC) mobilization from the bone marrow in a cell-nonautonomous fashion. This process is mediated, in part, through suppression of osteoblasts and disruption of CXCR4/CXCL12 signaling. The cellular targets of G-CSF that initiate the mobilization cascade have not been identified. We use mixed G-CSF receptor (G-CSFR)-deficient bone marrow chimeras to show that G-CSF-induced mobilization of HSPCs correlates poorly with the number of wild-type neutrophils. We generated transgenic mice in which expression of the G-CSFR is restricted to cells of the monocytic lineage. G-CSF-induced HSPC mobilization, osteoblast suppression, and inhibition of CXCL12 expression in the bone marrow of these transgenic mice are intact, demonstrating that G-CSFR signals in monocytic cells are sufficient to induce HSPC mobilization. Moreover, G-CSF treatment of wild-type mice is associated with marked loss of monocytic cells in the bone marrow. Finally, we show that bone marrow macrophages produce factors that support the growth and/or survival of osteoblasts in vitro. Together, these data suggest a model in which G-CSFR signals in bone marrow monocytic cells inhibit the production of trophic factors required for osteoblast lineage cell maintenance, ultimately leading to HSPC mobilization.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/metabolismo , Movilización de Célula Madre Hematopoyética/métodos , Monocitos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Análisis de Varianza , Animales , Quimiocina CXCL12/metabolismo , Quimera/metabolismo , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Factor Estimulante de Colonias de Granulocitos/farmacología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Monocitos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Cell Stem Cell ; 6(3): 227-37, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20207226

RESUMEN

During a screen for humoral factors that promote cardiomyocyte differentiation from embryonic stem cells (ESCs), we found marked elevation of granulocyte colony-stimulating factor receptor (G-CSFR) mRNA in developing cardiomyocytes. We confirmed that both G-CSFR and G-CSF were specifically expressed in embryonic mouse heart at the midgestational stage, and expression levels were maintained throughout embryogenesis. Intrauterine G-CSF administration induced embryonic cardiomyocyte proliferation and caused hyperplasia. In contrast, approximately 50% of csf3r(-/-) mice died during late embryogenesis because of the thinning of atrioventricular walls. ESC-derived developing cardiomyocytes also strongly expressed G-CSFR. When extrinsic G-CSF was administered to the ESC- and human iPSC-derived cardiomyocytes, it markedly augmented their proliferation. Moreover, G-CSF-neutralizing antibody inhibited their proliferation. These findings indicated that G-CSF is critically involved in cardiomyocyte proliferation during development, and may be used to boost the yield of cardiomyocytes from ESCs for their potential application to regenerative medicine.


Asunto(s)
Proliferación Celular , Células Madre Embrionarias/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Corazón/embriología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo
15.
J Bone Miner Res ; 23(11): 1765-74, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18597629

RESUMEN

Long-term treatment of mice or humans with granulocyte colony-stimulating factor (G-CSF) is associated with a clinically significant osteopenia characterized by increased osteoclast activity and number. In addition, recent reports have observed a decrease in number of mature osteoblasts during G-CSF administration. However, neither the extent of G-CSF's suppressive effect on the osteoblast compartment nor its mechanisms are well understood. Herein, we show that short-term G-CSF treatment in mice leads to decreased numbers of endosteal and trabecular osteoblasts. The effect is specific to mature osteoblasts, because bone-lining cells, osteocytes, and periosteal osteoblasts are unaffected. G-CSF treatment accelerates osteoblast turnover in the bone marrow by inducing osteoblast apoptosis. In addition, whereas G-CSF treatment sharply increases osteoprogenitor number, differentiation of mature osteoblasts is impaired. Bone marrow transplantation studies show that G-CSF acts through a hematopoietic intermediary to suppress osteoblasts. Finally, G-CSF treatment, through suppression of mature osteoblasts, also leads to a marked decrease in osteoprotegerin expression in the bone marrow, whereas expression of RANKL remains relatively constant, suggesting a novel mechanism contributing to the increased osteoclastogenesis seen with long-term G-CSF treatment. In sum, these findings suggest that the hematopoietic system may play a novel role in regulating osteoblast differentiation and apoptosis during G-CSF treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Animales , Recuento de Células , Linaje de la Célula/efectos de los fármacos , Quimera , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Sistema Hematopoyético/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteoprotegerina/metabolismo , Periostio/citología , Ligando RANK/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia
16.
Blood ; 109(8): 3235-43, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17185469

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) regulates the production, maturation, and function of neutrophils. Its expression is often induced during infection, resulting in high concentrations of G-CSF in inflammatory exudates and in the blood, suggesting that it may regulate both local and systemic neutrophil responses. Herein, we characterize the neutrophil response in G-CSFR(-/-) mice following intratracheal injection with Pseudomonas aeruginosa-laden agarose beads, modeling the pulmonary infection observed in many patients with cystic fibrosis. G-CSFR(-/-) mice are markedly susceptible to bronchopulmonary P aeruginosa infection, exhibiting decreased survival and bacterial clearance as well as extensive damage to lung tissue. The systemic neutrophil response was mediated primarily by enhanced neutrophil release from the bone marrow rather than increased neutrophil production and was attenuated in G-CSFR(-/-) mice. Despite normal to increased local production of inflammatory chemokines, neutrophil accumulation into the infected lung of G-CSFR(-/-) mice was markedly reduced. Moreover, the percentage of apoptotic neutrophils in the lung was elevated, suggesting that G-CSF signals may play an important role in regulating neutrophil survival at the inflammatory site. Collectively, these data provide new evidence that G-CSF signals play important but specific roles in the regulation of the systemic and local neutrophil response following infection.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Neumonía Bacteriana/metabolismo , Infecciones por Pseudomonas/metabolismo , Animales , Apoptosis/genética , Médula Ósea/metabolismo , Médula Ósea/patología , Supervivencia Celular/genética , Quimiocinas/metabolismo , Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Fibrosis Quística/microbiología , Fibrosis Quística/patología , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Infiltración Neutrófila/genética , Neutrófilos/patología , Neumonía Bacteriana/etiología , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo
17.
Hematol Cell Ther ; 39(2): 102-4, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9168308

RESUMEN

Kostmann Syndrome is defined as a chronic neutropenia, dating from early childhood, characterized typically by a granulopoeisis impairment at the promyelocyte stage. The origin is not yet understood. G-CSF receptor anomaly -the intra-cellular carboxy terminal region- was noted in a few patients (6 out of 54), initially in two patients who later developed secondary leukemia. More follow-up, with other patients, led us to consider the mutation of the G-CSF receptor sometimes as a transient event, not systematically resulting in malignancy. This finding directs research toward intra-cellular signaling pathway in a pathology that raises questions both of granulopoeisis and leukemogenesis.


Asunto(s)
Neutropenia/sangre , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Cromosomas Humanos Par 7 , Enfermedad Crónica , Humanos , Neutropenia/etiología , Neutropenia/genética , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Síndrome
18.
Br J Haematol ; 89(4): 771-9, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7539624

RESUMEN

The expression of the granulocyte colony-stimulating factor (G-CSF) receptor in childhood Burkitt's lymphoma (BL) cells, and the mitogenic effect of G-CSF on these cells, was studied in a panel of 13 Epstein-Barr virus (EBV) positive and negative BL cell lines derived from nine children. G-CSF receptor mRNA expression was investigated by Northern blot analysis and reverse transcriptase polymerase chain reaction (RT-PCR). Binding of G-CSF to BL cell lines was measured by chemical crosslinking of 125I-G-CSF, and proliferation by thymidine incorporation. Inducibility of the G-CSF receptor was studied by stimulation with interleukin-1 beta, tumour necrosis factor-alpha, Staphylococcus aureus Cowan A, anti-human IgM, phorbol myristate acetate, calcium ionophore A23187, and by infection in vitro by immortalizing and non-immortalizing strains of EBV. BL cell lines, unstimulated or stimulated by biological reagents or EBV infection, did not bind radioionated G-CSF in crosslinking experiments. No stimulation by recombinant human G-CSF was observed in 3H-thymidine incorporation assays. No G-CSF receptor mRNA was detected by Northern blot analysis or RT-PCR in BL cell lines. It is concluded that G-CSF plays no direct stimulatory role in the growth of these malignant B-cells, making a deleterious influence of G-CSF in the clinical treatment situation unlikely.


Asunto(s)
Linfoma de Burkitt/metabolismo , Factor Estimulante de Colonias de Granulocitos/farmacología , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Adolescente , Secuencia de Bases , Northern Blotting , Niño , Reactivos de Enlaces Cruzados , Humanos , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Células Tumorales Cultivadas
19.
Immunity ; 5(5): 491-501, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8934575

RESUMEN

We have generated mice carrying a homozygous null mutation in the granulocyte colony-stimulating factor receptor (G-CSFR) gene. G-CSFR-deficient mice have decreased numbers of phenotypically normal circulating neutrophils. Hematopoietic progenitors are decreased in the bone marrow, and the expansion and terminal differentiation of these progenitors into granulocytes is impaired. Neutrophils isolated from G-CSFR-deficient mice have an increased susceptibility to apoptosis, suggesting that the G-CSFR may also regulate neutrophil survival. These data confirm a role for the G-CSFR as a major regulator of granulopoiesis in vivo and provide evidence that the G-CSFR may regulate granulopoiesis by several mechanisms. However, the data also suggest that G-CSFR-independent mechanisms of granulopoiesis must exist.


Asunto(s)
Apoptosis/inmunología , Neutrófilos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Animales , División Celular/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Ratones , Ratones Mutantes , Mutagénesis Sitio-Dirigida/inmunología , Neutrófilos/inmunología
20.
Blood ; 101(8): 2990-5, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12672695

RESUMEN

Severe congenital neutropenia (SCN) is a hematopoietic disorder characterized by neutropenia in peripheral blood and maturation arrest of neutrophil precursors in bone marrow. Patients with SCN may evolve to have myelodysplastic syndrome or acute myelocytic leukemia. In approximately 20% of SCN cases, a truncation mutation is found in the cytoplasmic region of the granulocyte colony-stimulating factor receptor (G-CSFR). We then generated mice carrying murine wild-type G-CSFR and its mutants equivalent to truncations at amino acids 718 and 731 in human G-CSFR, those were reported to be related to leukemic transformation of SCN. Although numbers of peripheral white blood cells, red blood cells, and platelets did not differ among mutant and wild-type G-CSFR transgenic (Tg) mice, both of the mutant receptor Tg mice had one third of peripheral neutrophil cell counts compared with wild-type receptor Tg mice. The mutant receptor Tg mice also showed impaired resistance to the infection with Staphylococcus aureus. Moreover, bone marrow of these Tg mice had an increased percentage of immature myeloid cells, a feature of SCN. This maturation arrest was also observed in in vitro cultures of bone marrow cells of truncated G-CSFR Tg mice under G-CSF stimulation. In addition, clonal culture of bone marrow cells of the truncated G-CSFR Tg mice showed the hypersensitivity to G-CSF in myeloid progenitors. Our Tg mice may be useful in the analysis of the role of truncated G-CSFR in SCN pathobiology.


Asunto(s)
Neutropenia/genética , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología , Enfermedad Aguda , Animales , Médula Ósea/patología , Diferenciación Celular , Células Cultivadas/patología , Células Clonales/patología , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Leucemia Mieloide/etiología , Recuento de Leucocitos , Ratones , Ratones Transgénicos , Síndromes Mielodisplásicos/etiología , Células Mieloides/patología , Neutropenia/congénito , Neutropenia/patología , Receptores de Factor Estimulante de Colonias de Granulocito/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Eliminación de Secuencia , Infecciones Estafilocócicas/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA