Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Blood ; 115(18): 3686-94, 2010 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-20223923

RESUMEN

The mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase 1 (ERK1) and ERK2 are among the main signal transduction molecules, but little is known about their isoform-specific functions in vivo. We have examined the role of ERK1 in adult hematopoiesis with ERK1(-/-) mice. Loss of ERK1 resulted in an enhanced splenic erythropoiesis, characterized by an accumulation of erythroid progenitors in the spleen, without any effect on the other lineages or on bone marrow erythropoiesis. This result suggests that the ablation of ERK1 induces a splenic stress erythropoiesis phenotype. However, the mice display no anemia. Deletion of ERK1 did not affect erythropoietin (EPO) serum levels or EPO/EPO receptor signaling and was not compensated by ERK2. Splenic stress erythropoiesis response has been shown to require bone morphogenetic protein 4 (BMP4)-dependent signaling in vivo and to rely on the expansion of a resident specialized population of erythroid progenitors, termed stress erythroid burst-forming units (BFU-Es). A great expansion of stress BFU-Es and increased levels of BMP4 mRNA were found in ERK1(-/-) spleens. The ERK1(-/-) phenotype can be transferred by bone marrow cells. These findings show that ERK1 controls a BMP4-dependent step, regulating the steady state of splenic erythropoiesis.


Asunto(s)
Anemia/patología , Células Precursoras Eritroides/fisiología , Eritropoyesis/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Bazo/metabolismo , Anemia/inducido químicamente , Animales , Apoptosis , Western Blotting , Trasplante de Médula Ósea , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Ensayo de Unidades Formadoras de Colonias , Células Precursoras Eritroides/citología , Eritropoyetina/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Oxidantes/toxicidad , Fenilhidrazinas/toxicidad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Bazo/citología
2.
Blood ; 116(22): 4444-55, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20693433

RESUMEN

Edification of the human hematopoietic system during development is characterized by the production of waves of hematopoietic cells separated in time, formed in distinct embryonic sites (ie, yolk sac, truncal arteries including the aorta, and placenta). The embryonic liver is a major hematopoietic organ wherein hematopoietic stem cells (HSCs) expand, and the future, adult-type, hematopoietic cell hierarchy becomes established. We report herein the identification of a new, transient, and rare cell population in the human embryonic liver, which coexpresses VE-cadherin, an endothelial marker, CD45, a pan-hematopoietic marker, and CD34, a common endothelial and hematopoietic marker. This population displays an outstanding self-renewal, proliferation, and differentiation potential, as detected by in vitro and in vivo hematopoietic assays compared with its VE-cadherin negative counterpart. Based on VE-cadherin expression, our data demonstrate the existence of 2 phenotypically and functionally separable populations of multipotent HSCs in the human embryo, the VE-cadherin(+) one being more primitive than the VE-cadherin(-) one, and shed a new light on the hierarchical organization of the embryonic liver HSC compartment.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Sistema Hematopoyético/embriología , Hígado/citología , Hígado/embriología , Animales , Antígenos CD/genética , Antígenos CD34/metabolismo , Cadherinas/genética , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Expresión Génica , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético/citología , Humanos , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones SCID , Embarazo
3.
Haematologica ; 97(7): 975-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22271899

RESUMEN

CD105 is an auxiliary receptor for the transforming growth factor beta superfamily, highly expressed on proliferating endothelial cells and adult hematopoietic stem cells. Because CD105 mRNA expression was reported in the developing aortic region, we further characterized its expression profile in the aorta and examined the hematopoietic potential of CD105(+) cells. Aortic endothelial cells, intra-aortic hematopoietic cell clusters and the purified cell fraction enriched in progenitor/hematopoietic stem cell activity expressed CD105. Aortic hematopoietic short-term clonogenic progenitors were highly enriched in the CD105(intermediate) population whereas more immature long-term progenitors/hematopoietic stem cells are contained within the CD105(high) population. This places CD105 on the short list of molecules discriminating short-term versus long-term progenitors in the aorta. Furthermore, decreasing transforming growth factor beta signaling increases the number of clonogenic progenitors. This suggests that CD105 expression level defines a hierarchy among aortic hematopoietic cells allowing purification of clonogenic versus more immature hematopoietic progenitors, and that the transforming growth factor beta pathway plays a critical role in this process.


Asunto(s)
Antígenos CD/genética , Aorta/citología , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Receptores de Superficie Celular/genética , Animales , Antígenos CD/metabolismo , Aorta/metabolismo , Proliferación Celular , Embrión de Mamíferos , Endoglina , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Embarazo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
4.
Exp Hematol ; 44(4): 297-302.e1, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26733047

RESUMEN

Thrombopoietin (TPO) and its receptor Mpl (CD110) play a crucial role in the regulation of hematopoietic stem cells (HSCs). Functional study of Mpl-expressing HSCs has, however, been hampered by the lack of efficient monoclonal antibodies, explaining the very few data available on Mpl(+) HSCs during human embryonic development and after birth. Investigating the main monoclonal antibodies used so far to sort CD110(+) cells from cord blood (CB) and adult bone marrow (BM), we found that only the recent monoclonal antibody 1.6.1 engineered by Immunex Corporation was specific. Using in vitro functional assays, we found that this antibody can be used to sort a CD34(+)CD38(-)CD110(+) population enriched in hematopoietic progenitor stem cells, both in CB and in adult BM. In vivo injection into NSG mice further indicated that the CB CD34(+)CD38(-)CD110(+) population is highly enriched in HSCs compared with both CD34(+)CD38(-)CD110(-) and CD34(+)CD38(-) populations. Together our results validate MAb1.6.1 as an important tool, which has so far been lacking, in the HSC field.


Asunto(s)
Anticuerpos Monoclonales , Células de la Médula Ósea , Sangre Fetal/citología , Células Madre Hematopoyéticas/metabolismo , Receptores de Trombopoyetina/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos/inmunología , Antígenos de Superficie/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Humanos , Fenotipo , Receptores de Trombopoyetina/antagonistas & inhibidores , Receptores de Trombopoyetina/inmunología
5.
Exp Hematol ; 30(11): 1263-72, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12423679

RESUMEN

OBJECTIVE: p21(WAF1/Cip/kip) and p27(Kip1) are cyclin-dependant kinase inhibitors controlling cell-cycle exit and differentiation of numerous cell types. Among hematopoietic cells, megakaryocytes express high levels of p21, while in erythroid cells, p27(Kip1) is predominant. As p21 and p27 could display overlapping functions and as megakaryocytes and erythroid cells derive from a bipotent progenitor, we developed an in vivo model to determine the specific role of p21 in controlling the proliferation/differentiation balance of erythroid and megakaryocytic progenitors. METHODS: Transgenic mice that overexpressed p21 under the control of the human GPIIb promoter in early progenitors and along megakaryocytic differentiation were generated. Different subsets of hematopoietic progenitors (BFU and CFU) and primitive cells (CAFC, LTC-IC) were analyzed by methylcellulose assay. Phenotypic evolution and clonogenic properties of the lin(-) population were analyzed along erythroid and megakaryocytic differentiation. RESULTS: We observed p21 ectopic expression in early hematopoietic progenitors (lin(-)Sca(+)), megakaryocytes, and, to a lesser extent, erythroid cells. This expression induced an important decrease in the number of CFU-MK, BFU-E, CFU-E, primitive progenitors (CAFC day 35), and LTC-IC, but did not affect the maturation process of these cells and the blood cell count. CONCLUSIONS: We show that variation of p21 expression level changes the fate of hematopoietic cells by favoring either proliferation or differentiation pathways. This effect of p21 is exerted not only at the level of primitive progenitors but also in more mature progenitors. However, in vivo, a systemic compensation mechanism is most likely activated in response to variations of the flow of progenitor production.


Asunto(s)
Ciclinas/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Glicoproteína IIb de Membrana Plaquetaria/genética , Regiones Promotoras Genéticas , Animales , Recuento de Células Sanguíneas , Diferenciación Celular , División Celular , Linaje de la Célula , Ensayo de Unidades Formadoras de Colonias , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Ciclinas/genética , ADN Complementario/genética , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Regulación de la Expresión Génica , Genes Sintéticos , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Humanos , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Transgénicos , Proteínas Recombinantes de Fusión/fisiología
6.
Cell Stem Cell ; 12(1): 37-48, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23246483

RESUMEN

DNA double-strand breaks (DSBs) represent a serious threat for hematopoietic stem cells (HSCs). How cytokines and environmental signals integrate the DNA damage response and contribute to HSC-intrinsic DNA repair processes remains unknown. Thrombopoietin (TPO) and its receptor, Mpl, are critical factors supporting HSC self-renewal and expansion. Here, we uncover an unknown function for TPO-Mpl in the regulation of DNA damage response. We show that DNA repair following γ-irradiation (γ-IR) or the action of topoisomerase-II inhibitors is defective in Mpl(-/-) and in wild-type mouse or human hematopoietic stem and progenitor cells treated in the absence of TPO. TPO stimulates DNA repair in vitro and in vivo by increasing DNA-PK-dependent nonhomologous end-joining efficiency. This ensures HSC chromosomal integrity and limits their long-term injury in response to IR. This shows that niche factors can modulate the HSC DSB repair machinery and opens new avenues for administration of TPO agonists for minimizing radiotherapy-induced HSC injury and mutagenesis.


Asunto(s)
Daño del ADN/fisiología , Células Madre Hematopoyéticas/metabolismo , Mutagénesis/fisiología , Células Madre/metabolismo , Trombopoyetina/metabolismo , Animales , Ciclo Celular , Ensayo Cometa , Daño del ADN/genética , Femenino , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutagénesis/genética , Trombopoyetina/genética
7.
Int J Dev Biol ; 54(6-7): 1067-74, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20711984

RESUMEN

In a previous study, we underlined the functional role of the TPO receptor, Mpl, in the establishment of definitive mouse hematopoiesis, by demonstrating that the lack of Mpl led to a delayed production of definitive hematopoietic cells in the aorta-gonad-mesonephros (AGM) region, and resulted in the production of hematopoietic stem cells (HSCs) with an impaired activity at E11.5. In order to more accurately estimate the role of Mpl during generation of HSCs in the aorta, we performed an analysis of these AGMs at the time of the first HSC emergence (E10.5). Our results indicated that while Mpl-/- AGMs were found to contain more hematopoietic cells (HC) than C57Bl6 AGMs at E10.5, a defect in the expansion process of the HC/HSCs was detected in explant cultures of these AGMs, likely due to an increased apoptosis of these cells. To determine the molecular mechanisms by which invalidation of Mpl receptor affects the temporal distribution and expansion of HC/HSCs in the AGM, a study of the transcription level of of Mpl target genes was conducted. Expression of Runx1, a master transcription factor for the formation of hematopoietic progenitor (HP) cells and HSCs from the vasculature, as well as expression of Meis1 and HoxB4, known to play a role in self-renewal and expansion of HSCs, were found to be down regulated in E10.5 Mpl-/- AGMs. Our data indicate that Mpl is an active player during the first steps of definitive hematopoiesis establishment through direct regulation of the expression of transcription factors or genes important for the self-renewal, proliferation and apoptosis of HSCs.


Asunto(s)
Apoptosis/fisiología , Embrión de Mamíferos/irrigación sanguínea , Células Madre Hematopoyéticas/citología , Sistema Hematopoyético/embriología , Receptores de Trombopoyetina/fisiología , Animales , Antígenos CD34/metabolismo , Aorta/citología , Aorta/embriología , Apoptosis/genética , Recuento de Células , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Embrión de Mamíferos/metabolismo , Femenino , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Gónadas/irrigación sanguínea , Gónadas/citología , Gónadas/embriología , Células Madre Hematopoyéticas/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Masculino , Mesonefro/irrigación sanguínea , Mesonefro/citología , Mesonefro/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-2/genética , Receptores de Trombopoyetina/deficiencia , Receptores de Trombopoyetina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas de Cultivo de Tejidos
8.
Int J Dev Biol ; 54(6-7): 1165-73, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20711993

RESUMEN

Hematopoietic stem cells (HSCs) arise first in the third week of human ontogeny inside yolk sac developing blood vessels, and independently, from the wall of the embryonic aorta and vitelline arteries one week later. HSCs produced in the yolk sac and in the embryonic truncal arteries migrate to and transiently colonize the embryonic liver (EL), and thereafter the bone marrow (BM), their permanent site of residence. At the moment, the origin of human HSCs is still controversial; one of the main hypotheses being that they are generated by hemogenic endothelial cells (ECs). To prove definitively the endothelial origin of HSCs that arise within the human embryo, we previously purified ECs from either the yolk sac or the truncal arteries and reported that they were able to produce blood cells in vitro. We then found that some of the HSCs present in the human EL were co-expressing vascular endothelial (VE)-cadherin, an endothelial marker, CD45, a pan-hematopoietic marker, and CD34, a common endothelial and hematopoietic marker, and demonstrated that these HSCs bearing a dual hemato-endothelial phenotype were endowed with remarkably high self renewal and proliferative potentials. Furthermore, a transgenic mouse model based on the VE-cadherin cis-regulating elements that we engineered to trace the fate of the first VE-cadherin expressing cells allowed us to clearly demonstrate that a majority of adult BM HSCs derived from a VE-cadherin ancestor. Altogether our studies strongly suggest that at least a part of both the human and the murine hematopoietic systems arise from an endothelium-like ancestor.


Asunto(s)
Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Endotelio/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Adulto , Animales , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Cadherinas/metabolismo , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Endotelio/irrigación sanguínea , Endotelio/embriología , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Humanos , Inmunohistoquímica , Antígenos Comunes de Leucocito/metabolismo , Hígado/irrigación sanguínea , Hígado/citología , Hígado/embriología , Masculino , Ratones
9.
Exp Hematol ; 37(9): 1072-83, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19539688

RESUMEN

OBJECTIVE: Glycosaminoglycans (GAG) are major components of bone marrow extracellular matrix because they have the property to interact with cells and growth factors in hematopoietic niches. In this study, we investigated the effect of two different chemically defined GAG mimetics on mobilization of hematopoietic stem and progenitor cells (HSPCs) in mice peripheral blood. MATERIALS AND METHODS: Mobilization was achieved by intraperitoneal injection of GAG mimetics. Mobilized cells were characterized phenotypically by reverse transcription polymerase chain reaction and fluorescence-activated cell sorting analysis and functionally by colony-forming cell, cobblestone area-forming cell and long-term culture-initiating cell assays in vitro. Radioprotection assays were performed to confirm the functionality of primitive hematopoietic cells in vivo. Involvement of stromal-derived factor-1 (SDF-1) and matrix metalloproteinase-9 (MMP-9) were investigated. RESULTS: GAG mimetics treatment induces hyperleukocytosis and mobilization of HSPC. They synergize with the effects of granulocyte colony-stimulating factor or AMD3100 on hematopoietic progenitors mobilization. Reconstitution of lethally irradiated recipient mice with peripheral blood mononuclear cells from GAG mimetic-treated donor mice improves engraftment and survival. BiAcore studies indicate that the mimetics interact directly with SDF-1. In addition, GAG mimetics-induced mobilization is associated with increased levels of pro- and active MMP-9 from bone marrow cells and increased level of SDF-1 in peripheral blood. Finally, mobilization is partially inhibited by co-injection with anti-SDF-1 antibody. CONCLUSION: This study demonstrates that GAG mimetics induce efficient mobilization of HSPCs, associated with an activation of pro-MMP-9 and a modification in the SDF-1 concentration gradient between bone marrow and peripheral blood. We suggest that structural features of GAGs can modify the nature of mobilized cells.


Asunto(s)
Materiales Biomiméticos/farmacología , Quimiocina CXCL12/sangre , Glicosaminoglicanos/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/citología , Metaloproteinasa 9 de la Matriz/sangre , Animales , Fármacos Anti-VIH/agonistas , Fármacos Anti-VIH/farmacología , Bencilaminas , Médula Ósea/metabolismo , Ciclamas , Sinergismo Farmacológico , Glicosaminoglicanos/agonistas , Supervivencia de Injerto/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/agonistas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/agonistas , Compuestos Heterocíclicos/farmacología , Masculino , Ratones , Relación Estructura-Actividad , Trasplante Homólogo
10.
Development ; 134(16): 3031-40, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17634189

RESUMEN

Cytokine signaling pathways are important in promoting hematopoietic stem cell (HSC) self-renewal, proliferation and differentiation. Mpl receptor and its ligand, TPO, have been shown to play an essential role in the early steps of adult hematopoiesis. We previously demonstrated that the cytoplasmic domain of Mpl promotes hematopoietic commitment of embryonic stem cells in vitro, and postulated that Mpl could be important in the establishment of definitive hematopoiesis. To answer this question, we investigated the temporal expression of Mpl during mouse development by in situ hybridization. We found Mpl expression in the HSCs clusters emerging in the AGM region, and in the fetal liver (FL) as early as E10.5. Using Mpl(-/-) mice, the functional relevance of Mpl expression was tested by comparing the hematopoietic progenitor (HP) content, long-term hematopoietic reconstitution (LTR) abilities and HSC content of control and Mpl(-/-) embryos at different times of development. In the AGM, we observed delayed production of HSCs endowed with normal LTR but presenting a self-renewal defect. During FL development, we detected a decrease in HP and HSC potential associated with a defect in amplification and self-renewal/survival of the lin(-) AA4.1(+) Sca1(+) population of HSCs. These results underline the dual role of Mpl in the generation and expansion of HSCs during establishment of definitive hematopoiesis.


Asunto(s)
Hematopoyesis/genética , Receptores de Trombopoyetina/fisiología , Animales , Aorta/embriología , Aorta/metabolismo , Proliferación Celular , Supervivencia Celular , Embrión de Mamíferos , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo
11.
Blood ; 100(6): 2063-70, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12200367

RESUMEN

The Mpl receptor plays an important role at the level of adult hematopoietic stem cells, but little is known of its function in embryonic and fetal hematopoiesis. We investigated the signals sent by the MPL cytoplasmic domain in fetal liver hematopoietic progenitors and during embryonic stem (ES) cell hematopoietic commitment. Mpl was found to be expressed only from day 6 of ES cell differentiation into embryoid bodies. Therefore, we expressed Mpl in undifferentiated ES cells or in fetal progenitors and studied the effects on hematopoietic differentiation. To avoid the inadvertent effect of thrombopoietin, we used a chimeric receptor, PM-R, composed of the extracellular domain of the prolactin receptor (PRL-R) and the transmembrane and cytoplasmic domains of Mpl. This allowed activation of the receptor with a hormone that is not involved in hematopoietic differentiation and assessment of the specificity of responses to Mpl by comparing PM-R with another PRL-R chimeric receptor that includes the cytoplasmic domain of the erythropoietin receptor (EPO-R) ([PE-R]). We have shown that the cytoplasmic domain of the Mpl receptor transduces exclusive signals in fetal liver hematopoietic progenitors as compared with that of EPO-R and that it promotes hematopoietic commitment of ES cells. Our findings demonstrate for the first time the specific role of Mpl in early embryonic or fetal hematopoietic progenitors and stem cells.


Asunto(s)
Feto/citología , Células Madre Hematopoyéticas/citología , Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/fisiología , Receptores de Citocinas , Transducción de Señal , Animales , Diferenciación Celular/efectos de los fármacos , Citoplasma , Embrión de Mamíferos/citología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/metabolismo , Humanos , Hígado/citología , Ratones , Estructura Terciaria de Proteína/fisiología , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/farmacología , Conejos , Receptores de Eritropoyetina , Receptores de Prolactina/fisiología , Receptores de Trombopoyetina , Proteínas Recombinantes de Fusión/fisiología , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA