Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood ; 129(14): 1901-1912, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28179275

RESUMO

Granulocyte colony-stimulating factor (G-CSF) is used clinically to treat leukopenia and to enforce hematopoietic stem cell (HSC) mobilization to the peripheral blood (PB). However, G-CSF is also produced in response to infection, and excessive exposure reduces HSC repopulation capacity. Previous work has shown that dormant HSCs contain all the long-term repopulation potential in the bone marrow (BM), and that as HSCs accumulate a divisional history, they progressively lose regenerative potential. As G-CSF treatment also induces HSC proliferation, we sought to examine whether G-CSF-mediated repopulation defects are a result of increased proliferative history. To do so, we used an established H2BGFP label retaining system to track HSC divisions in response to G-CSF. Our results show that dormant HSCs are preferentially mobilized to the PB on G-CSF treatment. We find that this mobilization does not result in H2BGFP label dilution of dormant HSCs, suggesting that G-CSF does not stimulate dormant HSC proliferation. Instead, we find that proliferation within the HSC compartment is restricted to CD41-expressing cells that function with short-term, and primarily myeloid, regenerative potential. Finally, we show CD41 expression is up-regulated within the BM HSC compartment in response to G-CSF treatment. This emergent CD41Hi HSC fraction demonstrates no observable engraftment potential, but directly matures into megakaryocytes when placed in culture. Together, our results demonstrate that dormant HSCs mobilize in response to G-CSF treatment without dividing, and that G-CSF-mediated proliferation is restricted to cells with limited regenerative potential found within the HSC compartment.


Assuntos
Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Animais , Proliferação de Células/genética , Regulação da Expressão Gênica/genética , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Transgênicos , Glicoproteína IIb da Membrana de Plaquetas/genética
2.
Exp Hematol ; 44(3): 194-206.e17, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26607595

RESUMO

Differentiation of hematopoietic stem-progenitor cells (HSPCs) into mature blood lineages results from the translation of extracellular signals into changes in the expression levels of transcription factors controlling cell fate decisions. Multiple transcription factor families are known to be involved in hematopoiesis. Although the T-box transcription factor family is known to be involved in the differentiation of multiple tissues, and expression of T-bet, a T-box family transcription factor, has been observed in HSPCs, T-box family transcription factors do not have a described role in HSPC differentiation. In the current study, we address the functional consequences of T-bet expression in mouse HSPCs. T-bet protein levels differed among HSPC subsets, with highest levels observed in megakaryo-erythroid progenitor cells (MEPs), the common precursor to megakaryocytes and erythrocytes. HSPCs from T-bet-deficient mice exhibited a defect in megakaryocytic differentiation when cultured in the presence of thrombopoietin. In contrast, erythroid differentiation in culture in the presence of erythropoietin was not substantially altered in T-bet-deficient HSPCs. Differences observed with respect to megakaryocyte number and maturity, as assessed by level of expression of CD41 and CD61, and megakaryocyte ploidy, in T-bet-deficient HSPCs were not associated with altered proliferation or survival in culture. Gene expression micro-array analysis of MEPs from T-bet-deficient mice exhibited diminished expression of multiple genes associated with the megakaryocyte lineage. These data advance our understanding of the transcriptional regulation of megakaryopoiesis by supporting a new role for T-bet in the differentiation of MEPs into megakaryocytes.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco Hematopoéticas/metabolismo , Megacariócitos/metabolismo , Proteínas com Domínio T/deficiência , Animais , Sobrevivência Celular , Eritrócitos/citologia , Eritrócitos/metabolismo , Células-Tronco Hematopoéticas/citologia , Integrina beta3/biossíntese , Integrina beta3/genética , Megacariócitos/citologia , Camundongos , Camundongos Mutantes , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/genética
3.
Dev Comp Immunol ; 53(1): 13-22, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26111997

RESUMO

The use of in vitro colony assays in mammals has contributed to identification of erythroid progenitor cells such as burst-forming unit-erythroid (BFU-E) and colony-forming unit-erythroid (CFU-E) progenitors, and serves to examine functions of erythropoietic growth factors like Erythropoietin (Epo) and Kit ligand. Here, we established an in vitro colony-forming assay capable of investigating erythropoiesis in carp (Cyprinus carpio), cloned and functionally characterized recombinant homologous molecules Epo and Kit ligand A (Kitla), and identified three distinct erythroid progenitor cells in carp. Recombinant carp Epo induced the formation of CFU-E-like and BFU-E-like erythroid colonies, expressing erythroid marker genes, ß-globin, epor and gata1. Recombinant carp Kitla alone induced limited colony formation, whereas a combination of Kitla and Epo dramatically enhanced erythroid colony formation and colony cell growth, as well as stimulated the formation of thrombocytic/erythroid colonies expressing not only erythroid markers but also thrombocytic markers, cd41 and c-mpl. Utilizing this colony assay to examine the distribution of distinct erythroid progenitor cells in carp, we demonstrated that carp head and trunk kidney play a primary role in erythropoiesis, while the spleen plays a secondary. Furthermore, we showed that presumably bi-potent thrombocytic/erythroid progenitor cells localize principally in the trunk kidney. Our results indicate that teleost fish possess mechanisms of Epo- and Kitla-dependent erythropoiesis similar to those in other vertebrates, and also help to demonstrate the diversity of erythropoietic sites among vertebrates.


Assuntos
Eritropoese/fisiologia , Eritropoetina/genética , Fator de Células-Tronco/genética , Células-Tronco/citologia , Animais , Carpas , Fator de Transcrição GATA1/biossíntese , Rim/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Receptores da Eritropoetina/biossíntese , Proteínas Recombinantes/genética , Baço/metabolismo , Trombopoetina/biossíntese , Globinas beta/biossíntese
4.
BMB Rep ; 48(12): 691-5, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26077028

RESUMO

We report that phytosphingosine, a sphingolipid found in many organisms and implicated in cellular signaling, promotes megakaryocytic differentiation of myeloid leukemia cells. Specifically, phytosphingosine induced several hallmark changes associated with megakaryopoiesis from K562 and HEL cells including cell cycle arrest, cell size increase and polyploidization. We also confirmed that cell type specific markers of megakaryocytes, CD41a and CD42b are induced by phytosphingosine. Phospholipids with highly similar structures were unable to induce similar changes, indicating that the activity of phytosphingosine is highly specific. Although phytosphingosine is known to activate p38 MAPK-mediated apoptosis, the signaling mechanisms involved in megakaryopoiesis appear to be distinct. In sum, we present another model for dissecting molecular details of megakaryocytic differentiation which in large part remains obscure.


Assuntos
Leucemia Mieloide/patologia , Megacariócitos/efeitos dos fármacos , Esfingosina/análogos & derivados , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Hematopoese , Humanos , Células K562 , Leucemia Mieloide/metabolismo , Megacariócitos/metabolismo , Megacariócitos/patologia , Complexo Glicoproteico GPIb-IX de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Transdução de Sinais , Esfingosina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Biochem Pharmacol ; 88(3): 364-71, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24518258

RESUMO

Integrins are heterodimeric adhesion receptors essential for adhesion of non-adherent cells to extracellular ligands such as extracellular matrix components. The affinity of integrins for ligands is regulated through a process termed integrin activation and de novo synthesis. Integrin activation is regulated by lipid raft components and the actin structure. However, there is little information on the relationship between integrin activation and its de novo synthesis. Cancerous mouse mast cells, mastocytoma P-815 cells (P-815 cells) are known to bind to fibronectin through de novo synthesis of integrin subtypes by prostaglandin (PG) E2 stimulation. The purpose of this study was to clarify the relationship between lipid raft components and the actin cytoskeleton, and PGE2-induced P-815 cells adhesion to fibronectin and the increase in surface expression and mRNA and protein levels of αvß3 and αIIbß3 integrins. Cholesterol inhibitor 6-O-α-maltosyl-ß cyclodextrin, glycosylphosphatidylinositol-anchored proteins inhibitor phosphatidylinositol-specific phospholipase C and actin inhibitor cytochalasin D inhibited PGE2-induced cell adhesion to fibronectin, but did not regulate the surface expression and mRNA and protein levels of αv and αIIb, and ß3 integrin subunits. In addition, inhibitor of integrin modulate protein CD47 had no effect on PGE2- and 8-Br-cAMP-induced cell adhesion. These results suggest that lipid raft components and the actin cytoskeleton are directly involved in increasing of adhesion activity of integrin αIIb, αv and ß3 subunits to fibronectin but not in stimulating of de novo synthesis of them in PGE2-stimulated P-815 cells. The modulation of lipid rafts and the actin structure is essential for P-815 cells adhesion to fibronectin.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Citoesqueleto de Actina/metabolismo , Dinoprostona/farmacologia , Fibronectinas/metabolismo , Integrinas/biossíntese , Microdomínios da Membrana/metabolismo , Animais , Antígeno CD47/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Colesterol/metabolismo , Integrina alfaV/biossíntese , Integrina alfaV/genética , Integrina beta3/biossíntese , Integrina beta3/genética , Integrinas/genética , Mastocitoma , Camundongos , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/genética , Ligação Proteica , Subunidades Proteicas/biossíntese , Subunidades Proteicas/genética , RNA Mensageiro/biossíntese
6.
Blood ; 121(5): 770-80, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23169780

RESUMO

UNLABELLED: Several studies have demonstrated that hematopoietic cells originate from endotheliumin early development; however, the phenotypic progression of progenitor cells during human embryonic hemogenesis is not well described. Here, we define the developmental hierarchy among intermediate populations of hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells (hESCs). We genetically modified hESCs to specifically demarcate acquisition of vascular (VE-cadherin) and hematopoietic (CD41a) cell fate and used this dual-reporting transgenic hESC line to observe endothelial to hematopoietic transition by real-time confocal microscopy. Live imaging and clonal analyses revealed a temporal bias in commitment of HPCs that recapitulates discrete waves of lineage differentiation noted during mammalian hemogenesis. Specifically, HPCs isolated at later time points showed reduced capacity to form erythroid/ megakaryocytic cells and exhibited a tendency toward myeloid fate that was enabled by expression of the Notch ligand Dll4 on hESC-derived vascular feeder cells. These data provide a framework for defining HPC lineage potential, elucidate a molecular contribution from the vascular niche in promoting hematopoietic lineage progression, and distinguish unique subpopulations of hemogenic endothelium during hESC differentiation. KEY POINTS: Live imaging of endothelial to hematopoietic conversion identifies distinct subpopulations of hESC-derived hemogenic endothelium. Expression of the Notch ligand DII4 on vascular ECs drives induction of myeloid fate from hESC-derived hematopoietic progenitors.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Transdução Genética , Antígenos CD/biossíntese , Antígenos CD/genética , Caderinas/biossíntese , Caderinas/genética , Técnicas de Cocultura , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Células Alimentadoras , Células-Tronco Hematopoéticas/citologia , Humanos , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/genética
7.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1209-20, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22505671

RESUMO

Although the lung expresses procoagulant proteins under inflammatory conditions, underlying mechanisms remain unclear. Here, we addressed lung endothelial expression of tissue factor (TF), which initiates the coagulation cascade and expression of which signifies development of a procoagulant phenotype in the vasculature. To establish the model of acid-induced acute lung injury (ALI), we intranasally instilled anesthetized mice with saline or acid. Then 2 h later, we isolated pulmonary vascular cells for flow cytometry and confocal microscopy to detect the leukocyte antigen, CD45 and the endothelial markers VE-cadherin and von Willebrand factor (vWf). Acid increased both the number of vWf-expressing cells as well as TF and P-selectin expressions on these cells. All of these effects were markedly inhibited by treating mice with antiplatelet serum, suggesting the involvement of platelets. The increased expressions of TF, vWf, and P-selectin in response to acid also occurred in platelets. Moreover, the effects were replicated in endothelial cells derived from isolated, blood-perfused lungs. However, the effect was inhibited completely in lungs perfused with platelet-depleted and, to a lesser extent, with leukocyte-depleted blood. Acid injury increased endothelial expressions of the platelet proteins, CD41 and CD42b, providing evidence that platelet proteins were transferred to the vascular surface. Reactive oxygen species (ROS) were implicated in these responses, in that the endothelial and platelet protein expressions were inhibited. We conclude that acid-induced ALI causes NOX2-mediated ROS generation that activates platelets, which then generate a procoagulant endothelial surface.


Assuntos
Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/induzido quimicamente , Plaquetas/metabolismo , Endotélio Vascular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Tromboplastina/biossíntese , Animais , Antígenos CD/biossíntese , Coagulação Sanguínea , Plaquetas/imunologia , Caderinas/biossíntese , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ácido Clorídrico/efeitos adversos , Ácido Clorídrico/toxicidade , Antígenos Comuns de Leucócito/biossíntese , Pulmão/imunologia , Pulmão/patologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NADPH Oxidase 2 , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Selectina-P/biossíntese , Selectina-P/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/imunologia , Complexo Glicoproteico GPIb-IX de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Tromboplastina/metabolismo , Fator de von Willebrand/biossíntese , Fator de von Willebrand/metabolismo
8.
J Exp Med ; 208(6): 1305-15, 2011 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-21624936

RESUMO

The aorta-gonad-mesonephros region plays an important role in hematopoietic stem cell (HSC) development during mouse embryogenesis. The vascular endothelial cadherin⁺ CD45⁺ (VE-cad⁺CD45⁺) population contains the major type of immature pre-HSCs capable of developing into long-term repopulating definitive HSCs. In this study, we developed a new coaggregation culture system, which supports maturation of a novel population of CD45-negative (VE-cad⁺CD45⁻CD41⁺) pre-HSCs into definitive HSCs. The appearance of these pre-HSCs precedes development of the VE-cad⁺CD45⁺ pre-HSCs (termed here type I and type II pre-HSCs, respectively), thus establishing a hierarchical directionality in the developing HSC lineage. By labeling the luminal surface of the dorsal aorta, we show that both type I and type II pre-HSCs are distributed broadly within the endothelial and subendothelial aortic layers, in contrast to mature definitive HSCs which localize to the aortic endothelial layer. In agreement with expression of CD41 in pre-HSCs, in vivo CD41-Cre-mediated genetic tagging occurs in embryonic pre-HSCs and persists in all lymphomyeloid lineages of the adult animal.


Assuntos
Células-Tronco Hematopoéticas/citologia , Antígenos Comuns de Leucócito/biossíntese , Animais , Aorta/metabolismo , Caderinas/metabolismo , Linhagem da Célula , Separação Celular , Endotélio Vascular/citologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Modelos Genéticos , Fenótipo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Transgenes
9.
Methods Mol Biol ; 750: 143-55, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21618089

RESUMO

The use of transgenic mice in which tissue or lineage-specific, cell-restricted promoters drive fluorescent reporters has recently been reported as a means to follow the in vivo migration of various hematopoietic cells during murine development. At present there is limited ability of these approaches to image the emergence of the first hematopoietic cell subsets due to lack of unique markers that define those hematopoietic cells. We have utilized whole embryo analysis via immunostaining and confocal laser-scanning microscopic (CLSM) imaging to define the emergence of the first hematopoietic elements in the yolk sac of the developing conceptus. The methods employed to examine yolk sac hematopoiesis may be applied to hematopoietic cell emergence in the embryo proper or fetal liver in the generation of a complete map of hematopoietic ontogeny.


Assuntos
Movimento Celular , Embrião de Mamíferos/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Glicoproteína IIb da Membrana de Plaquetas/análise , Imagem Corporal Total/métodos , Saco Vitelino/citologia , Animais , Linhagem da Célula , Embrião de Mamíferos/citologia , Fluorescência , Genes Reporter , Proteínas de Fluorescência Verde/análise , Imunoquímica , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Coloração e Rotulagem/métodos , Saco Vitelino/embriologia , Saco Vitelino/fisiologia
10.
Platelets ; 22(5): 390-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21385097

RESUMO

Besides thrombopoietin several additional factors (i.e neurotransmitters and receptors) are known to be involved in the regulation of megakaryopoiesis at different stages. Recently, we identified functional α7 nicotinic acetylcholine receptors (nAChRα7) on platelets and megakaryocytic precursors. In platelets nAChRα7 form functional Ca(2+) channels and are involved in fibrinogen receptor activation and aggregation. Here, we investigated the impact of nAChRα7 on the differentiation of the human megakaryoblastic cell line MEG-01. In vitro differentiation of MEG-01 cells was induced by the phorbol ester TPA for 5 days in the absence or presence of nicotine or the nAChRα7-selective antagonist methyllycaconitine (MLA), and this was monitored by the expression of the megakaryocytic antigens CD41 and CD61. In the presence of the cholinergic drugs (nicotine or MLA) CD41 and CD61 expression was significantly reduced, both at RNA and protein level. We postulate that the nAChRα7 receptor is involved in megakaryopoietic signal transduction and gene regulation. This could affect the generation of platelets in vivo and contribute to the development of novel therapeutic drugs that regulate platelet formation.


Assuntos
Aconitina/análogos & derivados , Diferenciação Celular/efeitos dos fármacos , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/metabolismo , Trombopoese/efeitos dos fármacos , Aconitina/farmacologia , Western Blotting , Diferenciação Celular/genética , Linhagem Celular , Colinérgicos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Integrina beta3/biossíntese , Integrina beta3/genética , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/metabolismo , Nicotina/farmacologia , Ésteres de Forbol/farmacologia , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Transdução de Sinais/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7
11.
Anticancer Res ; 31(2): 487-94, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21378328

RESUMO

It has been reported that human prostate-derived PC-3 cells that are CD44- and CD 41 (a2 B1)-positive are enriched in cancer stem cells. This study compared the effect of PC-3 cell proliferation under normoxia or hypoxia on the initial and subsequent expression of this doubly-labeled side-fraction. Despite the numerical advantage of attached normoxic cells, 48 h of culture under nitrogen, an environment containing minimal oxygen and CO(2) resulting in an elevated pH of the medium, was associated with a higher percentage, absolute and relative number of doubly-labeled (DL) hypoxic compared to normoxic cells. At 24 h, the reverse was found. When the pH was controlled with the use of 95% nitrogen and 5% carbon dioxide, the percentage and number of normoxic DL cells exceeded hypoxic ones at both 24 and 48 h. At 24 h, 2-deoxy-L-glucose or sodium arsenate reduced normoxic DL cell numbers more than hypoxic ones. The interplay between hypoxia, increased medium pH and the effect of inhibitors as they might influence therapy are considered.


Assuntos
Receptores de Hialuronatos/biossíntese , Células-Tronco Neoplásicas/patologia , Oxigênio/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Neoplasias da Próstata/patologia , Arseniatos/farmacologia , Contagem de Células , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Óxidos N-Cíclicos/farmacologia , Desoxiglucose/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Masculino , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/metabolismo
12.
Blood ; 117(19): 5088-91, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21415271

RESUMO

CD41 expression is associated with the earliest stages of mouse hematopoiesis. It is notably expressed on some cells of the intra-aortic hematopoietic clusters, an area where the first adult-repopulating hematopoietic stem cells (HSCs) are generated. Although it is generally accepted that CD41 expression marks the onset of primitive/definitive hematopoiesis, there are few published data concerning its expression on HSCs. It is as yet uncertain whether HSCs express CD41 throughout development, and if so, to what level. We performed a complete in vivo transplantation analysis with yolk sac, aorta, placenta, and fetal liver cells, sorted based on CD41 expression level. Our data show that the earliest emerging HSCs in the aorta express CD41 in a time-dependent manner. In contrast, placenta and liver HSCs are CD41⁻. Thus, differential and temporal expression of CD41 by HSCs in the distinct hematopoietic territories suggests a developmental/dynamic regulation of this marker throughout development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Animais , Aorta/embriologia , Aorta/metabolismo , Separação Celular , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Imuno-Histoquímica , Fígado/embriologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placenta/embriologia , Placenta/metabolismo , Gravidez , Saco Vitelino/embriologia , Saco Vitelino/metabolismo
13.
Blood ; 115(14): 2806-9, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20154212

RESUMO

Runx1 is required for the emergence of hematopoietic stem cells (HSCs) from hemogenic endothelium during embryogenesis. However, its role in the generation and maintenance of HSCs during adult hematopoiesis remains uncertain. Here, we present analysis of a zebrafish mutant line carrying a truncation mutation, W84X, in runx1. The runx1(W84X/W84X) embryos showed blockage in the initiation of definitive hematopoiesis, but some embryos were able to recover from a larval "bloodless" phase and develop to fertile adults with multilineage hematopoiesis. Using cd41-green fluorescent protein transgenic zebrafish and lineage tracing, we demonstrated that the runx1(W84X/W84X) embryos developed cd41(+) HSCs in the aorta-gonad-mesonephros region, which later migrated to the kidney, the site of adult hematopoiesis. Overall, our data suggest that in zebrafish adult HSCs can be formed without an intact runx1.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Mutação , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Células-Tronco Hematopoéticas/citologia , Mesonefro/citologia , Mesonefro/embriologia , Mesonefro/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
14.
Exp Hematol ; 37(10): 1238-1249.e5, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19619605

RESUMO

OBJECTIVE: Megakaryopoiesis and platelet formation is a multistep process through which hematopoietic progenitor cells develop into mature megakaryocytes (MKs) and form proplatelets. The present study investigates the regulation of different steps of megakaryopoiesis (i.e., differentiation, migration, and proplatelet formation) by extracellar signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) in two models of primary murine MKs derived from bone marrow (BM) cells and fetal liver (FL) cells. MATERIALS AND METHODS: A preparation of MKs was generated from BM obtained from femora and tibiae of C57BL6 mice. FL-derived MKs were obtained from the liver of mouse fetuses aged 13 to 15 days. RESULTS: For both cell populations, activation of MEK-ERK1/2 pathway by thrombopoietin was found to have a critical role in MK differentiation, regulating polyploidy and surface expression of CD34, GPIIb, and GPIb. The MEK-ERK1/2 pathway plays a major role in migration of BM-derived MKs toward a stromal-cell-derived factor 1alpha (SDF1alpha) gradient, whereas unexpectedly, FL-derived cells fail to migrate in response to the chemokine due to negligible expression of its receptor, CXCR4. The MEK-ERK1/2 pathway also plays a critical role in the generation of proplatelets. In contrast, p38MAPK pathway was not involved in any of these processes. CONCLUSION: This report demonstrates a critical role of MEK-ERK1/2 pathway in MK differentiation, motility, and proplatelet formation. This study highlights several differences between BM- and FL-derived MKs, which are discussed.


Assuntos
Plaquetas/citologia , Células da Medula Óssea/enzimologia , Quimiotaxia/fisiologia , Fígado/citologia , Sistema de Sinalização das MAP Quinases/fisiologia , Megacariócitos/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Trombopoese/fisiologia , Compostos de Anilina/farmacologia , Animais , Benzamidas/farmacologia , Células da Medula Óssea/citologia , Células Cultivadas/citologia , Células Cultivadas/enzimologia , Quimiocina CXCL12/farmacologia , Quimiotaxia/efeitos dos fármacos , Fígado/embriologia , Fígado/enzimologia , Megacariócitos/citologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Complexo Glicoproteico GPIb-IX de Plaquetas/biossíntese , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Glicoproteína IIb da Membrana de Plaquetas/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/análise , Trombopoetina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/análise
15.
Blood ; 114(10): 2181-92, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19584398

RESUMO

The role of miRNAs in regulating megakaryocyte differentiation was examined using bipotent K562 human leukemia cells. miR-34a is strongly up-regulated during phorbol ester-induced megakaryocyte differentiation, but not during hemin-induced erythrocyte differentiation. Enforced expression of miR-34a in K562 cells inhibits cell proliferation, induces cell-cycle arrest in G(1) phase, and promotes megakaryocyte differentiation as measured by CD41 induction. miR-34a expression is also up-regulated during thrombopoietin-induced differentiation of CD34(+) hematopoietic precursors, and its enforced expression in these cells significantly increases the number of megakaryocyte colonies. miR-34a directly regulates expression of MYB, facilitating megakaryocyte differentiation, and of CDK4 and CDK6, to inhibit the G(1)/S transition. However, these miR-34a target genes are down-regulated rapidly after inducing megakaryocyte differentiation before miR-34a is induced. This suggests that miR-34a is not responsible for the initial down-regulation but may contribute to maintaining their suppression later on. Previous studies have implicated miR-34a as a tumor suppressor gene whose transcription is activated by p53. However, in p53-null K562 cells, phorbol esters induce miR-34a expression independently of p53 by activating an alternative phorbol ester-responsive promoter to produce a longer pri-miR-34a transcript.


Assuntos
Diferenciação Celular/fisiologia , Fase G1/fisiologia , Megacariócitos/metabolismo , MicroRNAs/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/fisiologia , Antígenos CD34 , Carcinógenos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células K562 , Megacariócitos/citologia , MicroRNAs/genética , Ésteres de Forbol/farmacologia , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Regiões Promotoras Genéticas/fisiologia , Proteínas Proto-Oncogênicas c-myb/metabolismo , Trombopoetina/farmacologia , Proteína Supressora de Tumor p53/genética , Regulação para Cima/efeitos dos fármacos
16.
Life Sci ; 84(13-14): 458-67, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19302816

RESUMO

AIMS: Nm23-H1 is a suppressor of metastasis that has been implicated in the regulation of proliferation and differentiation of hematopoietic cells, although specific mechanisms for Nm23-H1 have not been well-characterized. Our study is designed to further elucidate the role of Nm23-H1 in the human chronic myeloid leukemia K562 cell line. MAIN METHODS: In this study we generated and selected two cell clone pools of human chronic myeloid leukemia K562 cells with up-regulated and down-regulated Nm23-H1 expression. KEY FINDINGS: Our data show that knockdown of Nm23-H1 decreased proliferation and increased the percentage of cells arrested in the G0/G1 phase of the cell cycle. Correspondingly, K562 cells overexpressing Nm23-H1 were more proliferative. After treatment of these two cell types with phorbol 12-myristate 13-acetate (PMA) for 48 h, cells with reduced Nm23-H1 expression had a higher percentage of 8N ploidy and higher expression of CD41 than K562 cells overexpressing Nm23-H1. A functional proteomics analysis identified ten proteins, including ANP32A, Cdc42GAP, and the isoform 2 of SET, whose expression levels were significantly altered by down-regulation of Nm23-H1. In addition, cells with decreased levels of Nm23-H1 had significantly reduced expression of Cdc42 independent of treatment with PMA. The interaction of the endogenous Nm23-H1 and Cdc42 proteins has been further validated by reciprocal immunoprecipitations. SIGNIFICANCE: We provide data that complement functional studies of Nm23-H1 in regulating hematopoietic cells, and address action mechanisms of Nm23-H1 that have not previously been reported.


Assuntos
Diferenciação Celular , Proliferação de Células , Nucleosídeo NM23 Difosfato Quinases/fisiologia , Proteômica , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo , Humanos , Células K562 , Nucleosídeo NM23 Difosfato Quinases/biossíntese , Nucleosídeo NM23 Difosfato Quinases/genética , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Proteômica/métodos , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Regulação para Cima , Proteína cdc42 de Ligação ao GTP/biossíntese
17.
Biol Pharm Bull ; 31(4): 569-73, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18379042

RESUMO

The mechanism of megakaryopoiesis, proplatelet formation (PPF) and platelet (PLT) production is not fully elucidated. Lactoferrin (LF) has been reported to have many biological functions including cell proliferation and differentiation, and the LF receptor is present on megakaryocytic cells. In the present study, we examined the effect of human LF (hLF) on PLT production from primary megakaryocytes (MKs). At first, we developed a PLT production system derived from human CD34+ cells by thrombopoietin (TPO) stimulation. Because the number of proplatelets, PLTs and CD41+ MKs was remarkably increased after day 5, we employed the TPO-induced CD34+ cells on day 5. Then, the effect of hLF on PLT production from human primary MKs was examined. In the range of 3-30 micrg/ml, hLF significantly inhibited PLT production up to about 60%. However, it did not significantly change the intensity of CD41 expression in MKs and the ploidy of MKs. In addition, it did not inhibit MK progenitors. These results suggest that LF directly inhibits PLT production from matured MKs, but does not inhibit megakaryopoiesis, including proliferation/maturation processes.


Assuntos
Lactoferrina/farmacologia , Megacariócitos/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Animais , Antígenos CD34/biossíntese , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Humanos , Indicadores e Reagentes , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Ploidias , Células-Tronco
18.
Stem Cells ; 26(7): 1732-42, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18436865

RESUMO

Although it has been proposed that the common myeloid progenitor gives rise to granulocyte/monocyte progenitors and megakaryocyte/erythroid progenitors (MEP), little is known about molecular switches that determine whether MEPs develop into either erythrocytes or megakaryocytes. We used the thrombopoietin receptor c-Mpl, as well as the megakaryocytic marker CD41, to optimize progenitor sorting procedures to further subfractionate the MEP (CD34(+)CD110(+)CD45RA(-)) into erythroid progenitors (CD34(+)CD110(+)CD45RA(-)CD41(-)) and megakaryocytic progenitors (CD34(+)CD110(+)CD45RA(-)CD41(+)) from peripheral blood. We have identified signal transducer and activator of transcription 5 (STAT5) as a critical denominator that determined lineage commitment between erythroid and megakaryocytic cell fates. Depletion of STAT5 from CD34(+) cells by a lentiviral RNAi approach in the presence of thrombopoietin and stem cell factor resulted in an increase in megakaryocytic progenitors (CFU-Mk), whereas erythroid progenitors (BFU-E) were decreased. Furthermore, an increase in cells expressing megakaryocytic markers CD41 and CD42b was observed in STAT5 RNAi cells, as was an increase in the percentage of polyploid cells. Reversely, overexpression of activated STAT5A(1*6) mutants severely impaired megakaryocyte development and induced a robust erythroid differentiation. Microarray and quantitative reverse transcription-polymerase chain reaction analysis revealed changes in expression of a number of genes, including GATA1, which was downmodulated by STAT5 RNAi and upregulated by activated STAT5.


Assuntos
Antígenos CD34/biossíntese , Regulação para Baixo , Eritropoese , Megacariócitos/citologia , Fator de Transcrição STAT5/biossíntese , Fator de Transcrição STAT5/fisiologia , Diferenciação Celular , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Megacariócitos/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Interferência de RNA , Receptores de Trombopoetina/metabolismo , Fator de Células-Tronco/metabolismo , Células-Tronco/citologia , Trombopoetina/metabolismo
19.
Am J Cardiovasc Drugs ; 7(6): 433-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18076210

RESUMO

BACKGROUND: Platelets play a crucial role in the pathogenesis of acute coronary syndromes (ACS). The efficacy of antiplatelet treatment is pivotal in the success of percutaneous coronary intervention (PCI) performed in patients with ACS. OBJECTIVE: The aim of the study was to investigate the effects of clopidogrel with or without abciximab on the expression of platelet surface receptors and platelet function in patients with ST-segment elevation myocardial infarction (STEMI) undergoing PCI. MATERIALS AND METHODS: Thirty patients with STEMI were included in the study. During acute primary coronary intervention, patients received aspirin (acetylsalicylic acid) and clopidogrel in a loading dose of 300mg. Clopidogrel was the only antiplatelet therapy used by nine patients (group B). Twenty-one patients (group A) received additional abciximab. Blood samples were collected and analyzed twice: before and up to 22 hours after administration of antiplatelet therapy. The platelet aggregation was established as primary platelet-related hemostasis (closure time [CT] assessed using the PFA100 system). The absolute number of platelet surface antigens as CD41a, CD42a, CD42b, CD61, and CD62P were determined by flow cytometry analysis. RESULTS: The study revealed a statistically significant increase in CT induced by adenosine diphosphate and adrenaline (epinephrine) +130 seconds (p < 0.0001) and +94 seconds (p < 0.0001), respectively, in group A patients post-therapy. While in group B the parameters of CT did not change after treatment. In addition, the absolute number of CD41a antigens (glycoprotein [GP] IIb/IIIa) increased significantly after treatment in group A. No significant changes were observed after treatment in the expression of CD62P (P-selectin) antigens in either treatment group. There was a significant reduction in the percentage of CD62P-positive platelets in group B after antiplatelet therapy. CONCLUSIONS: The absolute number of GP IIb/IIIa receptors increases and platelets are not activated up to 12 hours after cessation of abciximab therapy. Treatment of STEMI patients undergoing PCI with a loading dose of clopidogrel reduces the percentage of active platelets but does not influence the CT.


Assuntos
Anticorpos Monoclonais/farmacologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/biossíntese , Ticlopidina/análogos & derivados , Abciximab , Angioplastia Coronária com Balão , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Aspirina/administração & dosagem , Aspirina/farmacologia , Aspirina/uso terapêutico , Clopidogrel , Quimioterapia Combinada , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Integrina beta3/biossíntese , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Selectina-P/biossíntese , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/uso terapêutico , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Ticlopidina/administração & dosagem , Ticlopidina/farmacologia , Ticlopidina/uso terapêutico
20.
J Biol Chem ; 282(38): 27960-9, 2007 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-17644514

RESUMO

Microparticles from various cells are generated during inflammation. Platelet-derived microparticles (PMPs) harbor receptors that are not genuinely expressed by neutrophils. We tested whether or not functional glycoprotein IIb/IIIa (GPIIb/IIIa) receptors can be acquired by neutrophils via PMPs and whether these receptors participate in pro-inflammatory signaling. Surface expression was analyzed by flow cytometry and confocal microscopy. NF-kappaB activation was analyzed by Western blot experiments, electrophoretic mobility shift assays, and reverse transcription-PCR. Cell adhesion and spreading were estimated by myeloperoxidase assay and light microscopy. We found that PMPs transfer GPIIb/IIIa receptors to isolated and whole blood neutrophils via PMPs. We used specific antibodies in granulocyte macrophage colony-stimulating factor-treated neutrophils and observed that acquired GPIIb/IIIa receptors co-localized with beta2-integrins and cooperated in NF-kappaB activation. We show that Src and Syk non-receptor tyrosine kinases, as well as the actin cytoskeleton, control NF-kappaB activation. In contrast to NF-kappaB, acquisition of GPIIb/IIIa receptors was not necessary to induce adhesion to fibronectin or phosphatidylinositol 3-kinase/Akt signaling. When granulocyte macrophage colony-stimulating factor-stimulated neutrophils were incubated on fibronectin, strong NF-kappaB activation was observed, but only after loading with PMPs. Blocking either beta2-integrins or GPIIb/IIIa receptors abrogated this effect. Therapeutic GPIIb/IIIa inhibitors were similarly effective. The compounds also inhibited NF-kappaB-dependent tumor necrosis factor-alpha mRNA up-regulation. The data implicate GPIIb/IIIa receptors as new therapeutic targets in neutrophil-induced inflammation.


Assuntos
Plaquetas/metabolismo , Antígenos CD18/fisiologia , NF-kappa B/metabolismo , Neutrófilos/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Abciximab , Anticorpos Monoclonais/farmacologia , Plaquetas/efeitos dos fármacos , Eptifibatida , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/farmacologia , Integrina beta3/biossíntese , Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Tirofibana , Fator de Necrose Tumoral alfa/metabolismo , Tirosina/análogos & derivados , Tirosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA