Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
1.
Sci Rep ; 13(1): 22553, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38110522

RESUMO

The use of megakaryoblastic leukemia MEG-01 cells can help reveal the mechanisms of thrombopoiesis. However, conventional in vitro activation of platelet release from MEG-01 cells requires thrombopoietin, which is costly. Here, we aim to develop a more straightforward and affordable method. Synchronization of the MEG-01 cells was initially performed using serum-free culture, followed by spontaneous cell differentiation in the presence of serum. Different stages of megakaryoblast differentiation were classified based on cell morphology, DNA content, and cell cycle. The MEG-01 cells released platelet-like particles at a level comparable to that of the thrombopoietin-activated MEG-01 cells. The platelet-like particles were distinguishable from PLP-derived extracellular vesicles and could express P-selectin following ADP activation. Importantly, the platelet-like particles induced fibrin clotting in vitro using platelet-poor plasma. Therefore, this thrombopoietin-independent cell synchronization method is an effective and straightforward method for studying megakaryopoiesis and thrombopoiesis.


Assuntos
Megacariócitos , Trombopoetina , Megacariócitos/metabolismo , Trombopoetina/farmacologia , Trombopoetina/metabolismo , Células Progenitoras de Megacariócitos , Plaquetas , Trombopoese
2.
PLoS One ; 18(9): e0291662, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37729123

RESUMO

OBJECTIVES: In myelodysplastic syndromes (MDS), neoplastic myeloblast (CD34+CD13+CD33+ cells) numbers often increase over time, leading to secondary acute myeloid leukemia (AML). In recent studies, blasts in some MDS patients have been found to express a megakaryocyte-lineage molecule, CD41, and such patients show extremely poor prognosis. This is the first study to evaluate whether myeloblasts transition to CD41+ blasts over time and to investigate the detailed immunophenotypic features of CD41+ blasts in MDS. METHODS: We performed a retrospective cohort study, in which time-dependent changes in blast immunophenotypes were analyzed using multidimensional flow cytometry (MDF) in 74 patients with MDS and AML (which progressed from MDS). RESULTS: CD41+ blasts (at least 20% of CD34+ blasts expressing CD41) were detected in 12 patients. In five of these 12 patients, blasts were CD41+ from the first MDF analysis. In the other seven patients, myeloblasts (CD34+CD33+CD41- cells) transitioned to megakaryoblasts (CD34+CD41+ cells) over time, which was often accompanied by disease progression (including leukemic transformation). These CD41+ patients were more frequently observed among patients with monosomal and complex karyotypes. CD41+ blasts were negative for the erythroid antigen, CD235a, and positive for CD33 in all cases, but CD33 expression levels were lower in three cases when compared with CD34+CD41- blasts. Among the five CD41+ patients who underwent extensive immunophenotyping, CD41+ blasts all expressed CD61, but two cases had reduced CD42b expression, three had reduced/absent CD13 expression, and three also expressed CD7. CONCLUSIONS: Myeloblasts become megakaryoblastic over time in some MDS patients, and examining the megakaryocyte lineage (not only as a diagnostic work-up but also as follow-up) is needed to detect CD41+ MDS. The immunophenotypic features revealed in this study may have diagnostic relevance for CD41+ MDS patients.


Assuntos
Células Precursoras de Granulócitos , Síndromes Mielodisplásicas , Humanos , Imunofenotipagem , Células Progenitoras de Megacariócitos , Estudos Retrospectivos , Antígenos CD34
3.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3101-3110, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37162543

RESUMO

Acute megakaryocytic leukemia (AMKL) is a rare neoplasm caused by abnormal megakaryoblasts. Megakaryoblasts keep dividing and avoid undergoing polyploidization to escape maturation. Small-molecule probes inducing polyploidization of megakaryocytic leukemia cells accelerate the differentiation of megakaryocytes. This study aims to determine that Rho kinase (ROCK) inhibition on megakaryoblasts enhances polyploidization and the inhibition of ROCK1 by fasudil benefits AMKL mice. The study investigated fasudil on the megakaryoblast cells in vitro and in vivo. With the differentiation and apoptosis induction, fasudil was used to treat 6133/MPLW515L mice, and the differentiation level was evaluated. Fasudil could reduce proliferation and promote the polyploidization of megakaryoblasts. Meanwhile, fasudil reduced the disease burden of 6133/MPLW515L AMKL mice at a dose that is safe for healthy mice. Combination therapy of ROCK1 inhibitor fasudil and reported clinical AURKA inhibitor MLN8237 achieved a better antileukemia effect in vivo, which alleviated hepatosplenomegaly and promoted the differentiation of megakaryoblast cells. ROCK1 inhibitor fasudil is a good proliferation inhibitor and polyploidization inducer of megakaryoblast cells and might be a novel rationale for clinical AMKL treatment.


Assuntos
Leucemia Megacarioblástica Aguda , Megacariócitos , Animais , Camundongos , Megacariócitos/fisiologia , Leucemia Megacarioblástica Aguda/tratamento farmacológico , Leucemia Megacarioblástica Aguda/genética , Células Progenitoras de Megacariócitos , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/uso terapêutico , Quinases Associadas a rho
4.
Leuk Res ; 120: 106920, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35872339

RESUMO

Acute megakaryoblastic leukaemia (AMKL) is characterized by expansion of megakaryoblasts, which are hyper-proliferative cells that fail to undergo differentiation. Insight to the cell-cycle regulation revealed important events in early or late megakaryocytes (MKs) maturation; the cyclin-dependent kinases 4 and 6 (CDK4/6) have been reported to participate in the development of progenitor megakaryocytes, mainly by promoting cell cycle progression and DNA polyploidization. However, it remains unclear whether the continuous proliferation, but not differentiation, of megakaryoblasts is related to an aberrant regulation of CDK4/6 in AMKL. Here, we found that CDK4/6 were up regulated in patients with AMKL, and persistently maintained at a high level during the differentiation of abnormal megakaryocytes in vitro, according to a database and western blot. Additionally, AMKL cells were exceptionally reliant on the cell cycle regulators CDK4 or 6, as blocking their activity using an inhibitor or short hairpin RNA (shRNA) significantly reduced the proliferation of 6133/MPL megakaryocytes, reduced DNA polyploidy, induced apoptosis, decreased the level of phosphorylated retinoblastoma protein (p-Rb), and activation of caspase 3. Additionally, CDK4/6 inhibitors and shRNA reduced the numbers of leukemia cells in the liver and bone marrow (BM), alleviated hepatosplenomegaly, and prolonged the survival of AMKL-transplanted mice. These results suggested that blocking the activity of CDK4/6 may represent an effective approach to control megakaryoblasts in AMKL.


Assuntos
Leucemia Megacarioblástica Aguda , Animais , Ciclinas , DNA , Leucemia Megacarioblástica Aguda/tratamento farmacológico , Leucemia Megacarioblástica Aguda/genética , Células Progenitoras de Megacariócitos , Camundongos , RNA Interferente Pequeno
5.
Blood ; 139(22): 3233-3244, 2022 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-35108353

RESUMO

Fetal and neonatal megakaryocyte progenitors are hyperproliferative compared with adult progenitors and generate a large number of small, low-ploidy megakaryocytes. Historically, these developmental differences have been interpreted as "immaturity." However, more recent studies have demonstrated that the small, low-ploidy fetal and neonatal megakaryocytes have all the characteristics of adult polyploid megakaryocytes, including the presence of granules, a well-developed demarcation membrane system, and proplatelet formation. Thus, rather than immaturity, the features of fetal and neonatal megakaryopoiesis reflect a developmentally unique uncoupling of proliferation, polyploidization, and cytoplasmic maturation, which allows fetuses and neonates to populate their rapidly expanding bone marrow and blood volume. At the molecular level, the features of fetal and neonatal megakaryopoiesis are the result of a complex interplay of developmentally regulated pathways and environmental signals from the different hematopoietic niches. Over the past few years, studies have challenged traditional paradigms about the origin of the megakaryocyte lineage in both fetal and adult life, and the application of single-cell RNA sequencing has led to a better characterization of embryonic, fetal, and adult megakaryocytes. In particular, a growing body of data suggests that at all stages of development, the various functions of megakaryocytes are not fulfilled by the megakaryocyte population as a whole, but rather by distinct megakaryocyte subpopulations with dedicated roles. Finally, recent studies have provided novel insights into the mechanisms underlying developmental disorders of megakaryopoiesis, which either uniquely affect fetuses and neonates or have different clinical presentations in neonatal compared with adult life.


Assuntos
Megacariócitos , Trombopoese , Adulto , Medula Óssea , Feto , Humanos , Recém-Nascido , Células Progenitoras de Megacariócitos , Megacariócitos/metabolismo , Trombopoese/genética
6.
Stem Cell Res Ther ; 13(1): 54, 2022 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-35123563

RESUMO

BACKGROUND: Ex vivo production of induced megakaryocytes (MKs) and platelets from stem cells is an alternative approach for supplying transfusible platelets. However, it is difficult to generate large numbers of MKs and platelets from hematopoietic stem cells and progenitor cells (HSPCs). METHODS: To optimize the differentiation efficiency of megakaryocytic cells from HSPCs, we first employed a platelet factor 4 (PF4)-promoter reporter and high-throughput screening strategy to screen for small molecules. We also investigated the effects and possible mechanisms of candidate small molecules on megakaryocytic differentiation of human HSPCs. RESULTS: The small molecule Ricolinostat remarkably promoted the expression of PF4-promoter reporter in the megakaryocytic cell line. Notably, Ricolinostat significantly enhanced the cell fate commitment of MK progenitors (MkPs) from cord blood HSPCs and promoted the proliferation of MkPs based on cell surface marker detection, colony-forming unit-MK assay, and quantitative real-time PCR analyses. MkPs generated from Ricolinostat-induced HSPCs differentiated into mature MKs and platelets. Mechanistically, we found that Ricolinostat enhanced MkP fate mainly by inhibiting the secretion of IL-8 and decreasing the expression of the IL-8 receptor CXCR2. CONCLUSION: The addition of Ricolinostat to the culture medium promoted MkP differentiation from HSPCs and enhanced the proliferation of MkPs mainly by suppressing the IL-8/CXCR2 pathway. Our results can help the development of manufacturing protocols for the efficient generation of MKs and platelets from stem cells in vitro.


Assuntos
Ácidos Hidroxâmicos , Células Progenitoras de Megacariócitos , Diferenciação Celular , Células-Tronco Hematopoéticas , Humanos , Ácidos Hidroxâmicos/metabolismo , Ácidos Hidroxâmicos/farmacologia , Megacariócitos , Pirimidinas
7.
Blood Adv ; 6(1): 13-27, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34654056

RESUMO

Eltrombopag (ELT) is a thrombopoietic agent approved for immune thrombocytopenia and also a potent iron chelator. Here we found that ELT exhibited dose-dependent opposing effects on in vitro megakaryopoiesis: low concentrations (≤6 µM, ELT6) stimulated megakaryopoiesis, but high concentrations (30 µM, ELT30) suppressed megakaryocyte (MK) differentiation and proliferation. The suppressive effects of ELT30 were reproduced by other iron chelators, supporting iron chelation as a likely mechanism. During MK differentiation, committed MK progenitors (CD34+/CD41+ and CD34-/CD41+ cells) were significantly more sensitive than undifferentiated progenitors (CD34+/CD41- cells) to the suppressive effects of ELT30, which resulted from both decreased proliferation and increased apoptosis. The antiproliferative effects of ELT30 were reversed by increased iron in the culture, as were the proapoptotic effects when exposure to ELT30 was short. Because committed MK progenitors exhibited the highest proliferative rate and the highest sensitivity to iron chelation, we tested whether their iron status influenced their response to ELT during rapid cell expansion. In these studies, iron deficiency reduced the proliferation of CD41+ cells in response to all ELT concentrations. Severe iron deficiency also reduced the number of MKs generated in response to high thrombopoietin concentrations by ∼50%, compared with iron-replete cultures. Our findings support the hypothesis that although iron deficiency can stimulate certain cells and steps in megakaryopoiesis, it can also limit the proliferation of committed MK progenitors, with severity of iron deficiency and degree of thrombopoietic stimulation influencing the ultimate output. Further studies are needed to clarify how megakaryopoiesis, iron deficiency, and ELT stimulation are clinically interrelated.


Assuntos
Sangue Fetal , Células Progenitoras de Megacariócitos , Benzoatos , Diferenciação Celular , Hidrazinas , Ferro/farmacologia , Pirazóis
9.
Stem Cell Reports ; 16(12): 2861-2870, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34861163

RESUMO

Platelet transfusions are critical for severe thrombocytopenia but depend on blood donors. The shortage of donors and the potential of universal HLA-null platelet products have stimulated research on the ex vivo differentiation of human pluripotent stem cells (hPSCs) to platelets. We recently established expandable immortalized megakaryocyte cell lines (imMKCLs) from hPSCs by transducing MYC, BMI1, and BCL-XL (MBX). imMKCLs can act as cryopreservable master cells to supply platelet concentrates. However, the proliferation rates of the imMKCLs vary with the starting hPSC clone. In this study, we reveal from the gene expression profiles of several MKCL clones that the proliferation arrest is correlated with the expression levels of specific cyclin-dependent kinase inhibitors. Silencing CDKN1A and p53 with the overexpression of MBX was effective at stably inducing imMKCLs that generate functional platelets irrespective of the hPSC clone. Collectively, this improvement in generating imMKCLs should contribute to platelet industrialization and platelet biology.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inativação Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Plaquetas/metabolismo , Linhagem Celular , Proliferação de Células , Células Clonais , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Regulação para Cima , Proteína bcl-X/metabolismo
10.
J Vis Exp ; (171)2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-34096917

RESUMO

Bone marrow megakaryocytes are large polyploid cells that ensure the production of blood platelets. They arise from hematopoietic stem cells through megakaryopoiesis. The final stages of this process are complex and classically involve the bipotent Megakaryocyte-Erythrocyte Progenitors (MEP) and the unipotent Megakaryocyte Progenitors (MKp). These populations precede the formation of bona fide megakaryocytes and, as such, their isolation and characterization could allow for the robust and unbiased analysis of megakaryocyte formation. This protocol presents in detail the procedure to collect hematopoietic cells from mouse bone marrow, the enrichment of hematopoietic progenitors through magnetic depletion and finally a cell sorting strategy that yield highly purified MEP and MKp populations. First, bone marrow cells are collected from the femur, the tibia, and also the iliac crest, a bone that contains a high number of hematopoietic progenitors. The use of iliac crest bones drastically increases the total cell number obtained per mouse and thus contributes to a more ethical use of animals. A magnetic lineage depletion was optimized using 450 nm magnetic beads allowing a very efficient cell sorting by flow cytometry. Finally, the protocol presents the labeling and gating strategy for the sorting of the two highly purified megakaryocyte progenitor populations: MEP (Lin-Sca-1-c-Kit+CD16/32-CD150+CD9dim) and MKp (Lin- Sca-1-c-Kit+CD16/32-CD150+CD9bright). This technique is easy to implement and provides enough cellular material to perform i) molecular characterization for a deeper knowledge of their identity and biology, ii) in vitro differentiation assays, that will provide a better understanding of the mechanisms of maturation of megakaryocytes, or iii) in vitro models of interaction with their microenvironment.


Assuntos
Células Progenitoras de Megacariócitos , Megacariócitos , Animais , Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Células Progenitoras de Megacariócitos/citologia , Megacariócitos/citologia , Camundongos
11.
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
12.
Leuk Res ; 105: 106570, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33838549

RESUMO

Transient abnormal myelopoiesis (TAM) in neonates with Down syndrome, which spontaneously resolves within several weeks or months after birth, may represent a special form of leukemia developing in the fetal liver (FL). To explore the role of hepatoblasts, one of the major constituents of the FL hematopoietic microenvironment, in the pathogenesis of TAM, we investigated the influence of a human hepatoblastoma cell line, HUH-6, on the in vitro growth and differentiation of TAM blasts. In a coculture system with membrane filters, which hinders cell-to-cell contact between TAM blasts and HUH-6 cells, the growth and megakaryocytic differentiation of TAM blast progenitors were increased in the presence of HUH-6 cells. The culture supernatant of HUH-6 cells contained hematopoietic growth factors, including stem cell factor (SCF) and thrombopoietin (TPO). The neutralizing antibody against SCF abrogated the growth-stimulating activity of the culture supernatant of HUH-6 cells, demonstrating that, among the growth factors produced by HUH-6 cells, SCF may be the major growth stimulator and that TPO may be involved in megakaryocytic differentiation, rather than growth, of TAM blasts. This suggests that hepatoblasts function in the regulation of the growth and differentiation of TAM blasts in the FL through the production of hematopoietic growth factors, including SCF and TPO, and are involved in the leukemogenesis of TAM.


Assuntos
Síndrome de Down/patologia , Hepatoblastoma , Hepatócitos , Reação Leucemoide/patologia , Células Progenitoras de Megacariócitos , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Técnicas de Cocultura , Hepatoblastoma/metabolismo , Hepatoblastoma/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Leucemia Megacarioblástica Aguda/patologia , Células Progenitoras de Megacariócitos/metabolismo , Células Progenitoras de Megacariócitos/patologia , Mielopoese/fisiologia , Células-Tronco/metabolismo , Células-Tronco/patologia
13.
J Exp Med ; 218(4)2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33566111

RESUMO

In the embryo, the first hematopoietic cells derive from the yolk sac and are thought to be rapidly replaced by the progeny of hematopoietic stem cells. We used three lineage-tracing mouse models to show that, contrary to what was previously assumed, hematopoietic stem cells do not contribute significantly to erythrocyte production up until birth. Lineage tracing of yolk sac erythromyeloid progenitors, which generate tissue resident macrophages, identified highly proliferative erythroid progenitors that rapidly differentiate after intra-embryonic injection, persisting as the major contributors to the embryonic erythroid compartment. We show that erythrocyte progenitors of yolk sac origin require 10-fold lower concentrations of erythropoietin than their hematopoietic stem cell-derived counterparts for efficient erythrocyte production. We propose that, in a low erythropoietin environment in the fetal liver, yolk sac-derived erythrocyte progenitors efficiently outcompete hematopoietic stem cell progeny, which fails to generate megakaryocyte and erythrocyte progenitors.


Assuntos
Desenvolvimento Embrionário/genética , Eritrócitos/metabolismo , Eritropoese , Células Progenitoras de Megacariócitos/metabolismo , Saco Vitelino/fisiologia , Animais , Linhagem da Célula/genética , Eritropoetina/metabolismo , Feminino , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Proteínas Proto-Oncogênicas c-myb/deficiência , Proteínas Proto-Oncogênicas c-myb/genética
14.
Indian J Pathol Microbiol ; 63(3): 485-487, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32769348

RESUMO

Myeloma plasma cells vary from mature forms to immature, plasmablastic, and pleomorphic cells. Only a few cases of morphologic variant of plasma cell neoplasm have been reported, in which the plasma cell neoplasm presented with lymphoplasmacytic, megakaryocytic, plasmablastic, lymphocytosis-like, and variant hairy cell leukemia-like morphological features. A 66-year-old man sought medical attention with a previous 2-month history of lower back and chest pain. Magnetic resonance imaging (MRI) of the thoracic spine showed thoracic vertebral body shape and disc degeneration, and bone lesion. Blood work showed mild anemia (hemoglobin, 101 g/L; white blood cells, 6.98 × 109/L; platelets, 146 × 109/L.), hyperuricemia (UA 671 umol/L), and immunoglobulin G kappa [IgG(κ)] paraproteins. Bone marrow study revealed diffuse invasion by sheets of megakaryoblast-like cells. Flow cytometric analysis and bone marrow biopsy revealed plasma cell myeloma (PCM), and thoracic puncture biopsy indicated plasma cell neoplasms. Overall, the findings were in accordance with a PCM. To date, this is the first reported case of PCM with megakaryoblastic morphology mimicking acute leukemia. Recognizing the morphological variant of PCM is important in differentiating it from acute leukemia.


Assuntos
Leucemia Mieloide Aguda/diagnóstico , Células Progenitoras de Megacariócitos , Neoplasias de Plasmócitos/diagnóstico por imagem , Plasmócitos/patologia , Coluna Vertebral/diagnóstico por imagem , Doença Aguda , Idoso , Antineoplásicos/uso terapêutico , Biópsia , Medula Óssea/patologia , Diagnóstico Diferencial , Evolução Fatal , Humanos , Leucemia Mieloide Aguda/patologia , Imageamento por Ressonância Magnética , Masculino , Neoplasias de Plasmócitos/tratamento farmacológico , Coluna Vertebral/patologia
15.
Circ Res ; 127(9): 1182-1194, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32781905

RESUMO

RATIONALE: Mean platelet volume (MPV) and platelet count (PLT) are platelet measures that have been linked to cardiovascular disease (CVD) and mortality risk. Identifying protein biomarkers for these measures may yield insights into CVD mechanisms. OBJECTIVE: We aimed to identify causal protein biomarkers for MPV and PLT among 71 CVD-related plasma proteins measured in FHS (Framingham Heart Study) participants. METHODS AND RESULTS: We conducted integrative analyses of genetic variants associated with PLT/MPV with protein quantitative trait locus variants associated with plasma proteins followed by Mendelian randomization to infer causal relations of proteins for PLT/MPV. We also tested protein-PLT/MPV association in FHS participants. Using induced pluripotent stem cell-derived megakaryocyte clones that produce functional platelets, we conducted RNA-sequencing and analyzed expression differences between low- and high-platelet producing clones. We then performed small interfering RNA gene knockdown experiments targeting genes encoding proteins with putatively causal platelet effects in megakaryocyte clones to examine effects on platelet production. In protein-trait association analyses, ten proteins were associated with MPV and 31 with PLT. Mendelian randomization identified 4 putatively causal proteins for MPV and 4 for PLT. GP-5 (Glycoprotein V), GRN (granulin), and MCAM (melanoma cell adhesion molecule) were associated with PLT, while MPO (myeloperoxidase) showed significant association with MPV in both analyses. RNA-sequencing analysis results were directionally concordant with observed and Mendelian randomization-inferred associations for GP-5, GRN, and MCAM. In siRNA gene knockdown experiments, silencing GP-5, GRN, and MPO decreased PLTs. Genome-wide association study results suggest several of these may be linked to CVD risk. CONCLUSIONS: We identified 4 proteins that are causally linked to PLTs. These proteins may also have roles in the pathogenesis of CVD via a platelet/blood coagulation-based mechanism.


Assuntos
Doenças Cardiovasculares/genética , Granulinas , Volume Plaquetário Médio , Peroxidase , Contagem de Plaquetas , Glicoproteínas da Membrana de Plaquetas , Biomarcadores/sangue , Proteínas Sanguíneas/análise , Antígeno CD146/genética , Antígeno CD146/metabolismo , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/mortalidade , Diferenciação Celular , Feminino , Inativação Gênica , Estudo de Associação Genômica Ampla , Granulinas/genética , Granulinas/metabolismo , Humanos , Estudos Longitudinais , Masculino , Células Progenitoras de Megacariócitos , Megacariócitos/citologia , Análise da Randomização Mendeliana , Pessoa de Meia-Idade , Peroxidase/genética , Peroxidase/metabolismo , Fenótipo , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/metabolismo , Células-Tronco Pluripotentes , RNA Interferente Pequeno , Risco , Análise de Sequência de RNA
17.
Int J Mol Sci ; 21(3)2020 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-32024018

RESUMO

Megakaryopoiesis is the process during which megakaryoblasts differentiate to polyploid megakaryocytes that can subsequently shed thousands of platelets in the circulation. Megakaryocytes accumulate mRNA during their maturation, which is required for the correct spatio-temporal production of cytoskeletal proteins, membranes and platelet-specific granules, and for the subsequent shedding of thousands of platelets per cell. Gene expression profiling identified the RNA binding protein ATAXIN2 (ATXN2) as a putative novel regulator of megakaryopoiesis. ATXN2 expression is high in CD34+/CD41+ megakaryoblasts and sharply decreases upon maturation to megakaryocytes. ATXN2 associates with DDX6 suggesting that it may mediate repression of mRNA translation during early megakaryopoiesis. Comparative transcriptome and proteome analysis on megakaryoid cells (MEG-01) with differential ATXN2 expression identified ATXN2 dependent gene expression of mRNA and protein involved in processes linked to hemostasis. Mice deficient for Atxn2 did not display differences in bleeding times, but the expression of key surface receptors on platelets, such as ITGB3 (carries the CD61 antigen) and CD31 (PECAM1), was deregulated and platelet aggregation upon specific triggers was reduced.


Assuntos
Ataxina-2/genética , Perfilação da Expressão Gênica/métodos , Células Progenitoras de Megacariócitos/citologia , Animais , Antígenos CD34/genética , Ataxina-2/metabolismo , Diferenciação Celular , Linhagem Celular , RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Glicoproteína IIb da Membrana de Plaquetas/genética , Proteínas Proto-Oncogênicas/genética
18.
Sci Rep ; 10(1): 2277, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32042021

RESUMO

Megakaryocytes (MKs) play key roles in regulating bone metabolism. To test the roles of MK-secreted factors, we investigated whether MK and promegakaryocyte (pro-MK) conditioned media (CM) may affect bone formation and resorption. K562 cell lines were differentiated into mature MKs. Mouse bone marrow macrophages were differentiated into mature osteoclasts, and MC3T3-E1 cells were used for osteoblastic experiments. Bone formation was determined by a calvaria bone formation assay in vivo. Micro-CT analyses were performed in the femurs of ovariectomized female C57B/L6 and Balb/c nude mice after intravenous injections of MK or pro-MK CM. MK CM significantly reduced in vitro bone resorption, largely due to suppressed osteoclastic resorption activity. Compared with pro-MK CM, MK CM suppressed osteoblastic differentiation, but stimulated its proliferation, resulting in stimulation of calvaria bone formation. In ovariectomized mice, treatment with MK CM for 4 weeks significantly increased trabecular bone mass parameters, such as bone volume fraction and trabecular thickness, in nude mice, but not in C57B/L6 mice. In conclusion, MKs may secrete anti-resorptive and anabolic factors that affect bone tissue, providing a novel insight linking MKs and bone cells in a paracrine manner. New therapeutic agents against metabolic bone diseases may be developed from MK-secreted factors.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Megacariócitos/metabolismo , Osteogênese/efeitos dos fármacos , Comunicação Parácrina , Animais , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/etiologia , Diferenciação Celular/fisiologia , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Feminino , Fêmur/diagnóstico por imagem , Fêmur/fisiologia , Humanos , Injeções Intravenosas , Células K562 , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Células Progenitoras de Megacariócitos/metabolismo , Camundongos , Osteoclastos/fisiologia , Osteoporose Pós-Menopausa/diagnóstico por imagem , Osteoporose Pós-Menopausa/tratamento farmacológico , Osteoporose Pós-Menopausa/etiologia , Ovariectomia , Crânio/efeitos dos fármacos , Crânio/fisiologia , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA