Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 16(6): 1598-1613, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34019813

RESUMO

Age-related morbidity is associated with a decline in hematopoietic stem cell (HSC) function, but the mechanisms of HSC aging remain unclear. We performed heterochronic HSC transplants followed by quantitative analysis of cell reconstitution. Although young HSCs outperformed old HSCs in young recipients, young HSCs unexpectedly failed to outcompete the old HSCs of aged recipients. Interestingly, despite substantial enrichment of megakaryocyte progenitors (MkPs) in old mice in situ and reported platelet (Plt) priming with age, transplanted old HSCs were deficient in reconstitution of all lineages, including MkPs and Plts. We therefore performed functional analysis of young and old MkPs. Surprisingly, old MkPs displayed unmistakably greater regenerative capacity compared with young MkPs. Transcriptome analysis revealed putative molecular regulators of old MkP expansion. Collectively, these data demonstrated that aging affects HSCs and megakaryopoiesis in fundamentally different ways: whereas old HSCs functionally decline, MkPs gain expansion capacity upon aging.


Assuntos
Envelhecimento/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Células Progenitoras de Megacariócitos/fisiologia , Trombopoese , Transcriptoma , Animais , Linhagem da Célula , Feminino , Transplante de Células-Tronco Hematopoéticas/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Biochem Biophys Res Commun ; 510(3): 456-461, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30732856

RESUMO

Platelet, apart from its classic role of homeostasis, serves also as a crucial immune cell component that contributes to the aggravation of atherosclerosis. It has been reported that myocardial infarction (MI) triggers leukocytosis in the bone marrow and spleen, which accelerates post-MI atherosclerosis. However, it remains unclear whether thrombopoiesis is also enhanced after MI. Here, using flow cytometry and bone marrow whole-mount immunofluorescence staining combined with three-dimensional (3D) reconstruction, we for the first time demonstrated an enhanced thrombopoiesis and megakaryopoiesis in a mouse model of coronary artery ligation as a mimic of MI. We showed that MI leads to increasing number of peripheral platelets, as well as elevating number and larger size of bone marrow MKs. We also observed more proplatelets and fragmented MKs, and a closer spatial distribution of MK populations to the bone marrow vascular niche after MI. This study provides direct evidence that MI induces bone marrow megakaryocyte proliferation, maturation and platelet production. It opens a new scope that targeting platelet production might become a novel therapeutic approach for attenuating post-MI atherosclerosis.


Assuntos
Plaquetas/fisiologia , Megacariócitos/citologia , Infarto do Miocárdio/fisiopatologia , Trombopoese , Animais , Proliferação de Células , Masculino , Células Progenitoras de Megacariócitos/fisiologia , Megacariócitos/fisiologia , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia
3.
Thromb Haemost ; 119(5): 726-734, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30759486

RESUMO

Chronic treatment with aspirin in healthy volunteers (HVs) is associated with recovery of adenosine diphosphate (ADP)-induced platelet activation. The purinergic P2Y1 receptor exerts its effects via a Gq-protein, which is the same biochemical pathway activated by thromboxane-A2 receptor. We hypothesized that recovery of ADP-induced platelet activation could be attributed to increased P2Y1 expression induced by chronic aspirin exposure. We performed a multi-phase investigation which embraced both in vitro and in vivo experiments conducted in (1) human megakaryoblastic DAMI cells, (2) human megakaryocytic progenitor cell cultures, (3) platelets obtained from HVs treated with aspirin and (4) platelets obtained from aspirin-treated patients. DAMI cells treated with aspirin or WY14643 (PPARα agonist) had a significant up-regulation of P2Y1 mRNA, which was shown to be a PPARα-dependent process. In human megakaryocytic progenitors, in the presence of aspirin or WY14643, P2Y1 mRNA expression was higher than in mock culture. P2Y1 expression increased in platelets obtained from HVs treated with aspirin for 8 weeks. Platelets obtained from patients who were on aspirin for more than 2 months had increased P2Y1 expression and ADP-induced aggregation compared with patients on aspirin treatment for less than a month. Overall, our results suggest that aspirin induces genomic changes in megakaryocytes leading to P2Y1 up-regulation and that PPARα is the nuclear receptor involved in this regulation. Since P2Y1 is coupled to the same Gq-protein of thromboxane-A2 receptor, platelet adaptation in response to pharmacological inhibition seems not to be receptor specific, but may involve other receptors with the same biochemical pathway.


Assuntos
Aspirina/uso terapêutico , Plaquetas/fisiologia , Células Progenitoras de Megacariócitos/fisiologia , Inibidores da Agregação Plaquetária/uso terapêutico , Receptores Purinérgicos P2Y1/metabolismo , Difosfato de Adenosina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , PPAR alfa/agonistas , Ativação Plaquetária , Agregação Plaquetária , Pirimidinas/farmacologia , Receptores Purinérgicos P2Y1/genética
4.
Exp Hematol ; 44(8): 664-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26773569

RESUMO

Although the synergy between erythropoietin and thrombopoietin has previously been pointed out, the clonal demonstration of a human bipotent erythroid/megakaryocytic progenitor (MEP) was first published in Experimental Hematology (Papayannopoulou T, Brice M, Farrer D, Kaushansky K. Exp Hematol. 1996;24:660-669) and later in the same year in Blood (Debili N, Coulombel L, Croisille L, et al. Blood. 1996;88:1284-1296). This demonstration, and the fact that both bipotent and monopotent erythroid or megakaryocytic progenitors co-express markers of both lineages and respond to both lineage-specific transcription factors, has provided a background for the extensive use of MEP assessment by fluorescence-activated cell sorting in many subsequent studies. Beyond this, the demonstration of shared regulatory elements and the presence of single mutations affecting both lineages have inspired further studies to decipher how the shift in transcription factor networks occurs from one lineage to the other. Furthermore, in addition to shared effects, erythropoietin and thrombopoietin have additional independent effects. Most notable for thrombopoietin is its effect on hematopoietic stem cells illustrated by in vitro and in vivo approaches.


Assuntos
Células Precursoras Eritroides/fisiologia , Eritropoetina/fisiologia , Células Progenitoras de Megacariócitos/fisiologia , Trombopoetina/fisiologia , Animais , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Humanos , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Fenótipo , Trombopoetina/farmacologia
5.
Cell Stem Cell ; 17(4): 422-34, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26299573

RESUMO

Infections are associated with extensive platelet consumption, representing a high risk for health. However, the mechanism coordinating the rapid regeneration of the platelet pool during such stress conditions remains unclear. Here, we report that the phenotypic hematopoietic stem cell (HSC) compartment contains stem-like megakaryocyte-committed progenitors (SL-MkPs), a cell population that shares many features with multipotent HSCs and serves as a lineage-restricted emergency pool for inflammatory insults. During homeostasis, SL-MkPs are maintained in a primed but quiescent state, thus contributing little to steady-state megakaryopoiesis. Even though lineage-specific megakaryocyte transcripts are expressed, protein synthesis is suppressed. In response to acute inflammation, SL-MkPs become activated, resulting in megakaryocyte protein production from pre-existing transcripts and a maturation of SL-MkPs and other megakaryocyte progenitors. This results in an efficient replenishment of platelets that are lost during inflammatory insult. Thus, our study reveals an emergency machinery that counteracts life-threatening platelet depletions during acute inflammation.


Assuntos
Plaquetas/patologia , Inflamação/patologia , Células Progenitoras de Megacariócitos/patologia , Trombopoese , Animais , Plaquetas/fisiologia , Linhagem da Célula , Proliferação de Células , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Células Progenitoras de Megacariócitos/fisiologia , Camundongos
6.
Radiat Res ; 184(3): 235-48, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26284423

RESUMO

Although radiation therapy is an important cancer treatment modality, patients may experience adverse effects. The use of a radiation-effect modulator may help improve the outcome and health-related quality of life (HRQOL) of patients undergoing radiation therapy either by enhancing tumor cell killing or by protecting normal tissues. Historically, the successful translation of radiation-effect modulators to the clinic has been hindered due to the lack of focused collaboration between academia, pharmaceutical companies and the clinic, along with limited availability of support for such ventures. The U.S. Government has been developing medical countermeasures against accidental and intentional radiation exposures to mitigate the risk and/or severity of acute radiation syndrome (ARS) and the delayed effects of acute radiation exposures (DEARE), and there is now a drug development pipeline established. Some of these medical countermeasures could potentially be repurposed for improving the outcome of radiation therapy and HRQOL of cancer patients. With the objective of developing radiation-effect modulators to improve radiotherapy, the Small Business Innovation Research (SBIR) Development Center at the National Cancer Institute (NCI), supported by the Radiation Research Program (RRP), provided funding to companies from 2011 to 2014 through the SBIR contracts mechanism. Although radiation-effect modulators collectively refer to radioprotectors, radiomitigators and radiosensitizers, the focus of this article is on radioprotection and mitigation of radiation injury. This specific SBIR contract opportunity strengthened existing partnerships and facilitated new collaborations between academia and industry. In this commentary, we assess the impact of this funding opportunity, outline the review process, highlight the organ/site-specific disease needs in the clinic for the development of radiation-effect modulators, provide a general understanding of a framework for gathering preclinical and clinical evidence to obtain regulatory approval and provide a basis for broader venture capital needs and support from pharmaceutical companies to fully capitalize on the advances made thus far in this field.


Assuntos
Neoplasias/radioterapia , Proteção Radiológica , Terapia Genética , Genisteína/uso terapêutico , Humanos , Células Progenitoras de Megacariócitos/fisiologia , Neoplasias/psicologia , Fragmentos de Peptídeos/uso terapêutico , Qualidade de Vida , Radioterapia/efeitos adversos , Empresa de Pequeno Porte , Trombina/uso terapêutico , Receptor 5 Toll-Like/agonistas
7.
Thromb Haemost ; 114(5): 969-81, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26202144

RESUMO

The function of the mammalian target of rapamycin (mTOR) is upregulated in response to cell stimulation with growing and differentiating factors. Active mTOR controls cell proliferation, differentiation and death. Since mTOR associates with different proteins to form two functional macromolecular complexes, we aimed to investigate the role of the mTOR1 and mTOR2 complexes in MEG-01 cell physiology in response to thrombopoietin (TPO). By using mTOR antagonists and overexpressing FKBP38, we have explored the role of both mTOR complexes in proliferation, apoptosis, maturation-like mechanisms, endoplasmic reticulum-stress and the intracellular location of both active mTOR complexes during MEG-01 cell stimulation with TPO. The results demonstrate that mTOR1 and mTOR2 complexes play different roles in the physiology of MEG-01 cells and in the maturation-like mechanisms; hence, these findings might help to understand the mechanism underlying generation of platelets.


Assuntos
Plaquetas/fisiologia , Células Progenitoras de Megacariócitos/fisiologia , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Apoptose/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Naftiridinas/farmacologia , Sirolimo/farmacologia , Proteína 1A de Ligação a Tacrolimo/genética , Proteína 1A de Ligação a Tacrolimo/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo , Trombopoetina/metabolismo , Transgenes/genética
8.
Blood ; 124(2): 277-86, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-24735964

RESUMO

Megakaryocyte (MK) development in the bone marrow progresses spatially from the endosteal niche, which promotes MK progenitor proliferation, to the sinusoidal vascular niche, the site of terminal maturation and thrombopoiesis. The chemokine stromal cell-derived factor-1 (SDF-1), signaling through CXCR4, is implicated in the maturational chemotaxis of MKs toward sinusoidal vessels. Here, we demonstrate that both IV administration of SDF-1 and stabilization of endogenous SDF-1 acutely increase MK-vasculature association and thrombopoiesis with no change in MK number. In the setting of radiation injury, we find dynamic fluctuations in marrow SDF-1 distribution that spatially and temporally correlate with variations in MK niche occupancy. Stabilization of altered SDF-1 gradients directly affects MK location. Importantly, these SDF-1-mediated changes have functional consequences for platelet production, as the movement of MKs away from the vasculature decreases circulating platelets, while MK association with the vasculature increases circulating platelets. Finally, we demonstrate that manipulation of SDF-1 gradients can improve radiation-induced thrombocytopenia in a manner additive with earlier TPO treatment. Taken together, our data support the concept that SDF-1 regulates the spatial distribution of MKs in the marrow and consequently circulating platelet numbers. This knowledge of the microenvironmental regulation of the MK lineage could lead to improved therapeutic strategies for thrombocytopenia.


Assuntos
Movimento Celular , Quimiocina CXCL12/fisiologia , Megacariócitos/citologia , Megacariócitos/fisiologia , Lesões Experimentais por Radiação , Nicho de Células-Tronco/genética , Trombopoese/genética , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Células da Medula Óssea/efeitos da radiação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/efeitos da radiação , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Movimento Celular/efeitos da radiação , Células Cultivadas , Quimiocina CXCL12/administração & dosagem , Feminino , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Células Progenitoras de Megacariócitos/fisiologia , Células Progenitoras de Megacariócitos/efeitos da radiação , Megacariócitos/efeitos dos fármacos , Megacariócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Lesões Experimentais por Radiação/genética , Lesões Experimentais por Radiação/patologia , Receptores CXCR4/administração & dosagem , Receptores CXCR4/metabolismo , Nicho de Células-Tronco/efeitos dos fármacos , Nicho de Células-Tronco/efeitos da radiação , Trombopoese/efeitos dos fármacos , Trombopoese/efeitos da radiação
9.
Genes Cells ; 18(10): 886-98, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23890289

RESUMO

GATA1 is a transcription factor essential for erythropoiesis and megakaryopoiesis. It has been found that Gata1 gene knockdown heterozygous female (Gata1(G1.05/+)) mice spontaneously develop erythroblastic leukemias. In this study, we have generated a novel Gata1 knockdown erythroblastic cell line, designated GAK14, from the leukemia cells in the Gata1(G1.05/+) mice. Although GAK14 cells maintain immature phenotype on OP9 stromal cells in the presence of erythropoietin and stem cell factor, the cells produce Gr-1-, Mac1-, B220-, CD3e- or CD49b-positive hematopoietic cells when co-cultured with DAS104-8 feeder cells. However, GAK14 cells did not produce erythroid and megakaryocytic lineages, perhaps due to the absence of GATA1. Indeed, GAK14 cells became capable of differentiating into mature erythroid cells when complemented with full-length GATA1 and co-cultured with fetal liver-derived FLS5 stromal cells. This differentiation potential was impaired when GATA1 lacking the N-terminal domain was complemented. The N-terminal domain is known to contribute to the pathogenesis of transient abnormal myelopoiesis and acute megakaryoblastic leukemia related to Down syndrome. These results thus showed that GAK14 cells will serve as a powerful tool for dissecting domain function of GATA1 and that the GATA1 N-terminal domain is essential for the erythroid differentiation of GAK14 cells.


Assuntos
Aminoácidos/genética , Linhagem Celular Tumoral , Células Precursoras Eritroides/fisiologia , Eritropoese , Fator de Transcrição GATA1/química , Fator de Transcrição GATA1/metabolismo , Leucemia Eritroblástica Aguda , Animais , Linhagem da Célula , Técnicas de Cocultura , Células Precursoras Eritroides/metabolismo , Feminino , Fator de Transcrição GATA1/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Células Progenitoras de Megacariócitos/fisiologia , Camundongos , Mielopoese , Estrutura Terciária de Proteína
10.
Blood ; 122(9): 1649-57, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23794065

RESUMO

C1galt1 is essential for synthesis of the core 1 structure of mucin-type O-glycans. To clarify the physiological role of O-glycans in adult hematopoiesis, we exploited the interferon-inducible Mx1-Cre transgene to conditionally ablate the C1galt(flox) allele (Mx1-C1). Mx1-C1 mice exhibit severe thrombocytopenia, giant platelets, and prolonged bleeding times. Both the number and DNA ploidy of megakaryocytes in Mx1-C1 bone marrow were similar to those in wild-type (WT) mice. However, there were few proplatelets in Mx1-C1 primary megakaryocytes. Conversely, bone marrow transplanted from Mx1-C1 to WT and splenectomized Mx1-C1 mice gave rise to observations similar to those described above. The expression of GPIbα messenger RNA was unchanged in Mx1-C1 bone marrow, whereas flow cytometric and western blot analyses using megakaryocytes and platelets revealed that the expression of GPIbα protein was significantly reduced in Mx1-C1 mice. Moreover, circulating Mx1-C1 platelets exhibited an increase in the number of microtubule coils, despite normal levels of α- and ß-tubulin. Our observations suggest that O-glycan is required for terminal megakaryocyte differentiation and platelet production and that the decrease in GPIbα in cells lacking O-glycan might be caused by increased proteolysis.


Assuntos
Diferenciação Celular/genética , Galactosiltransferases/genética , Megacariócitos/fisiologia , Trombocitopenia/genética , Animais , Células Cultivadas , Feminino , Galactosiltransferases/fisiologia , Técnicas de Transferência de Genes , Masculino , Células Progenitoras de Megacariócitos/metabolismo , Células Progenitoras de Megacariócitos/fisiologia , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Trombocitopenia/patologia , Trombocitopenia/fisiopatologia , Trombopoese/genética
11.
Blood ; 121(20): 4205-20, 2013 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-23509158

RESUMO

The SH2 domain-containing protein-tyrosine phosphatases Shp1 and Shp2 have been implicated in regulating signaling from a variety of platelet and megakaryocyte receptors. In this study, we investigate the functions of Shp1 and Shp2 in megakaryocytes and platelets. Megakaryocyte/platelet (MP)-specific deletion of Shp1 in mice resulted in platelets being less responsive to collagen-related peptide due to reduced GPVI expression and signaling via the Src family kinase (SFK)-Syk-PLCγ2 pathway, and fibrinogen due to reduced SFK activity. By contrast, deletion of Shp2 in the MP lineage resulted in macrothrombocytopenia and platelets being hyper-responsive to anti-CLEC-2 antibody and fibrinogen. Shp1- and Shp2-deficient megakaryocytes had partial blocks at 2N/4N ploidy; however, only the latter exhibited reduced proplatelet formation, thrombopoietin, and integrin signaling. Mice deficient in both Shp1 and Shp2 were severely macrothrombocytopenic and had reduced platelet surface glycoprotein expression, including GPVI, αIIbß3, and GPIbα. Megakaryocytes from these mice were blocked at 2N/4N ploidy and did not survive ex vivo. Deletion of the immunoreceptor tyrosine-based inhibition motif-containing receptor G6b-B in the MP lineage phenocopied multiple features of Shp1/2-deficient mice, suggesting G6b-B is a critical regulator of Shp1 and Shp2. This study establishes Shp1 and Shp2 as major regulators of megakaryocyte development, platelet production, and function.


Assuntos
Plaquetas/fisiologia , Deleção de Genes , Megacariócitos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Trombopoese/genética , Animais , Plaquetas/metabolismo , Células Cultivadas , Células Progenitoras de Megacariócitos/metabolismo , Células Progenitoras de Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/fisiologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/fisiologia , Trombopoese/fisiologia
12.
Oncogene ; 32(20): 2565-75, 2013 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22777353

RESUMO

The coordinated recruitment of epigenetic regulators of gene expression by transcription factors such as RUNX1 (AML1, acute myeloid leukemia 1) is crucial for hematopoietic differentiation. Here, we identify protein arginine methyltransferase 6 (PRMT6) as a central functional component of a RUNX1 corepressor complex containing Sin3a and HDAC1 in human hematopoietic progenitor cells. PRMT6 is recruited by RUNX1 and mediates asymmetric histone H3 arginine-2 dimethylation (H3R2me2a) at megakaryocytic genes in progenitor cells. H3R2me2a keeps RUNX1 target genes in an intermediate state with concomitant H3K27me3 and H3K4me2 but not H3K4me3. Upon megakaryocytic differentiation PRMT6 binding is lost, the H3R2me2a mark decreases and a coactivator complex containing WDR5/MLL and p300/pCAF is recruited. This leads to an increase of H3K4me3 and H3K9ac, which result in augmented gene expression. Our results provide novel mechanistic insight into how RUNX1 activity in hematopoietic progenitor cells maintains differentiation genes in a suppressed state but poised for rapid transcriptional activation.


Assuntos
Arginina/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Células K562 , Células Progenitoras de Megacariócitos/fisiologia , Megacariócitos/metabolismo , Metilação , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Proteína-Arginina N-Metiltransferases/genética , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
13.
Cytokine ; 55(2): 181-7, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21555225

RESUMO

OBJECTIVE: There have been various reports on the roles of CXC receptors (CXCR) in modulation of hematopoiesis. In the present study, we investigated the effects of CXCR1 and/or CXCR2 inhibition on expansion and differentiation of umbilical cord blood (UCB) CD133(+) cells into megakaryocytic progenitors. MATERIALS AND METHODS: Purified UCB CD133(+) cells were cultured in a serum-free liquid culture either in the presence or absence of neutralizing anti-CXCR1 and/or anti-CXCR2 antibodies in combination with a conventional cytokine cocktail for up to 14days. Expression of megakaryocytic lineage markers (CD41 and CD61) and determination of ploidy level were determined by flowcytometry. In addition, colony-forming unit assay was performed using CD133(+) cultures in serum-free collagen-based medium containing the cytokine cocktail plus neutralizing CXCR1 and -R2 antibodies. Colony forming unit-megakaryocyte (CFU-MKs) and non-MKs were counted after immunocytochemistry staining on day 12. RESULTS: We show that while simultaneous inhibition of both CXCR1 and -R2 causes a significant reduction in the fold expansion of UCB CD133(+) cells, it also leads to an increase in percentages of CD61(+), CD41(+), and CFU-MK populations. CONCLUSION: CXCR1 and CXCR2 play significant roles in the suppression of megakaryopoiesis. We demonstrate that blocking of this suppressive effect by a simultaneous inhibition of both receptors can enhance the differentiation of UCB CD133(+) cells into megakayocytic progenitors.


Assuntos
Antígenos CD/imunologia , Diferenciação Celular/imunologia , Sangue Fetal/citologia , Glicoproteínas/imunologia , Células Progenitoras de Megacariócitos/fisiologia , Peptídeos/imunologia , Receptores de Interleucina-8A/imunologia , Receptores de Interleucina-8B/imunologia , Antígeno AC133 , Animais , Biomarcadores/metabolismo , Separação Celular , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Ploidias , Gravidez
14.
Okajimas Folia Anat Jpn ; 87(3): 151-4, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21174945

RESUMO

Thrombopoietin is the cytokine involved in megakaryopoiesis and its receptor (c-Mpl) is considered to regulate development of megakaryocyte. In this research, to elucidate the underlying mechanisms of c-mpl gene expression in megakaryoblastic cells, we investigated the effect of a protein kinase C (PKC) on c-mpl promoter activity in a time-dependent manner. PKC is a member of a family of serine/threonine protein kinases in the cytosol involved in cell growth and differentiation. Phorbol 12-myristate 13-acetate (PMA) is known as PKC activator, significantly enhanced the c-mpl promoter activity and PKC inhibitor, 2-methylpiperazine dihydrochloride (H-7) suppressed the up-regulation of PMA-induced promoter activity and this effect decreased in a time-dependent manner. These results clearly suggest that in megakaryoblastic cells, PKC plays the crucial role in the initiation of up-regulation of PMA-induced c-mpl promoter activity.


Assuntos
Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/fisiologia , Proteína Quinase C/metabolismo , Receptores de Trombopoetina/genética , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Carcinógenos/farmacologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Humanos , Regiões Promotoras Genéticas/fisiologia , Acetato de Tetradecanoilforbol/farmacologia
15.
Platelets ; 21(8): 648-57, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20942599

RESUMO

Both, the activity of transcription factors as well as epigenetic alterations in defined DNA regions regulate cellular differentiation processes. Hence, neuronal differentiation from neural progenitor cells is promoted by the transcription factor all trans retinoic acid (ATRA) and the histone deacetylase inhibitor valproic acid (VPA). VPA has also been shown to be involved in differentiation of tumor cells and to greatly improve the reprogramming of human somatic cells to induced pluripotent stem cells. Here we have investigated the impact of ATRA and VPA on the differentiation of megakaryoctes and platelets from the megakaryocyte progenitor cell line MEG-01. Our results show that treatment with ATRA (10⁻¹¹ M) and VPA (2 × 10⁻³ M) induces megakaryopoiesis of MEG-01 cells as estimated by polyploidy, formation of characteristic proplatelets and elevated expression of the megakaryocytic markers CD41 and CD61. The resulting megakaryocytes stayed viable for more than 3 weeks and shed platelet-like particles positive for CD41, CD61 and CD42b into the supernatant. Platelet-like particles responded to thrombin receptor activating peptide (TRAP-6) with increased externalization of P-selectin. Thus, ATRA and VPA proved to be efficient agents for the gentle induction of megakaryopoiesis and thrombopoiesis of MEG-01 cells providing the possibility to study molecular events underlying megakaryopoiesis and human platelet production over longer time periods.


Assuntos
Plaquetas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Progenitoras de Megacariócitos , Megacariócitos , Tretinoína/farmacologia , Ácido Valproico/farmacologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Inibidores de Histona Desacetilases/farmacologia , Humanos , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Células Progenitoras de Megacariócitos/fisiologia , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/fisiologia , Fragmentos de Peptídeos/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Transcrição/metabolismo
16.
Blood ; 114(1): 153-6, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19339696

RESUMO

Survivin is a member of the chromosome passenger complex, which plays an important role in chromosome alignment, separation, and cytokinesis. Although survivin is required for the proliferation and survival of hematopoietic stem and progenitor cells, the extent to which it is necessary for endomitosis of megakaryocytes remains controversial. To determine whether survivin is required for polyploidization, we analyzed mice with a megakaryocyte-specific deletion. PF4-Cre/survivin(fl/fl) mice harbored normal platelet counts with megakaryocytes that reached ploidy states comparable with those of control littermates. The CD41(+) cells within these animals showed little excision but increased annexin V staining, implying that survivin is required for survival of megakaryocyte progenitors in vivo. In contrast, megakaryocytes in which survivin was excised ex vivo showed robust excision and an increased degree of polyploidization. These results demonstrate that survivin is necessary for survival of megakaryocyte progenitors, but is not required for polyploidization of committed megakaryocytes.


Assuntos
Megacariócitos/citologia , Megacariócitos/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Mitose/fisiologia , Animais , Diferenciação Celular , Sobrevivência Celular , Técnicas In Vitro , Proteínas Inibidoras de Apoptose , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/fisiologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Fator Plaquetário 4/genética , Poliploidia , Regiões Promotoras Genéticas , Proteínas Repressoras , Survivina
17.
Blood ; 113(7): 1535-42, 2009 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19029443

RESUMO

Quebec platelet disorder (QPD) is an inherited bleeding disorder associated with increased urokinase plasminogen activator (uPA) in platelets but not in plasma, intraplatelet plasmin generation, and alpha-granule protein degradation. These abnormalities led us to investigate uPA expression by QPD CD34(+) progenitors, cultured megakaryocytes, and platelets, and whether uPA was stored in QPD alpha-granules. Although QPD CD34(+) progenitors expressed normal amounts of uPA, their differentiation into megakaryocytes abnormally increased expression of the uPA gene but not the flanking genes for vinculin or calcium/calmodulin-dependent protein kinase IIgamma on chromosome 10. The increased uPA production by cultured QPD megakaryocytes mirrored their production of alpha-granule proteins, which was normal. uPA was localized to QPD alpha-granules and it showed extensive colocalization with alpha-granule proteins in both cultured QPD megakaryocytes and platelets, and with plasminogen in QPD platelets. In QPD megakaryocytes, cultured without or with plasma as a source of plasminogen, alpha-granule proteins were stored undegraded and this was associated with much less uPA-plasminogen colocalization than in QPD platelets. Our studies indicate that the overexpression of uPA in QPD emerges with megakaryocyte differentiation, without altering the expression of flanking genes, and that uPA is costored with alpha-granule proteins prior to their proteolysis in QPD.


Assuntos
Transtornos Plaquetários/patologia , Transtornos Plaquetários/fisiopatologia , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Antígenos CD34/metabolismo , Plaquetas/citologia , Plaquetas/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Expressão Gênica/fisiologia , Humanos , Plasminogênio/metabolismo , Trombopoese/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
18.
Blood ; 113(7): 1543-6, 2009 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-18988861

RESUMO

Quebec platelet disorder (QPD) is an autosomal dominant disorder with high penetrance that is associated with increased risks for bleeding. The hallmark of QPD is a gain-of-function defect in fibrinolysis due to increased platelet content of urokinase plasminogen activator (uPA) without systemic fibrinolysis. We hypothesized that increased expression of uPA by differentiating QPD megakaryocytes is linked to PLAU. Genetic marker analyses indicated that QPD was significantly linked to a 2-Mb region on chromosome 10q containing PLAU with a maximum multipoint logarithm of the odds (LOD) score of +11 between markers D10S1432 and D10S1136. Analysis of PLAU by sequencing and Southern blotting excluded mutations within PLAU and its known regulatory elements as the cause of QPD. Analyses of uPA mRNA indicated that QPD distinctly increased transcript levels of the linked PLAU allele with megakaryocyte differentiation. These findings implicate a mutation in an uncharacterized cis element near PLAU as the cause of QPD.


Assuntos
Transtornos Plaquetários/genética , Escore Lod , Células Progenitoras de Megacariócitos/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Alelos , Diferenciação Celular/fisiologia , Cromossomos Humanos Par 10 , Expressão Gênica , Teste de Complementação Genética , Humanos , Células Progenitoras de Megacariócitos/citologia
19.
PLoS One ; 3(10): e3565, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18958162

RESUMO

BACKGROUND: The SH2-containing-5'inositol phosphatase-1 (SHIP) influences signals downstream of cytokine/chemokine receptors that play a role in megakaryocytopoiesis, including thrombopoietin, stromal-cell-derived-Factor-1/CXCL-12 and interleukin-3. We hypothesize that SHIP might control megakaryocytopoiesis through effects on proliferation of megakaryocyte progenitors (MKP) and megakaryocytes (MK). METHODOLOGY AND PRINCIPAL FINDINGS: Herein, we report the megakaryocytic phenotype and MK functional assays of hematopoietic organs of two strains of SHIP deficient mice with deletion of the SHIP promoter/first exon or the inositol phosphatase domain. Both SHIP deficient strains exhibit a profound increase in MKP numbers in bone marrow (BM), spleen and blood as analyzed by flow cytometry (Lin(-)c-Kit+CD41+) and functional assays (CFU-MK). SHIP deficient MKP display increased phosphorylation of Signal Transducers and Activators of Transcription 3 (STAT-3), protein kinase B (PKB/AKT) and extracellular signal-regulated kinases (ERKs). Despite increased MKP content, total body number of mature MK (Lin(-)c-kit(-)CD41+) are not significantly changed as SHIP deficient BM contains reduced MK while spleen MK numbers are increased. Reduction of CXCR-4 expression in SHIP deficient MK may influence MK localization to the spleen instead of the BM. Endomitosis, process involved in MK maturation, was preserved in SHIP deficient MK. Circulating platelets and red blood cells are also reduced in SHIP deficient mice. CONCLUSIONS/SIGNIFICANCE: SHIP may play an important role in regulation of essential signaling pathways that control early megakaryocytopoiesis in vivo.


Assuntos
Diferenciação Celular/genética , Células Progenitoras de Megacariócitos/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Células Sanguíneas/metabolismo , Células Sanguíneas/fisiologia , Medula Óssea/metabolismo , Medula Óssea/fisiologia , Movimento Celular/genética , Células Cultivadas , Inositol Polifosfato 5-Fosfatases , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Receptores CXCR4/metabolismo , Baço/metabolismo , Baço/fisiologia , Trombopoese/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA