Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 401
Filtrar
1.
Adv Mater ; 34(19): e2200905, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35294781

RESUMO

Due to the adverse effects of erythropoietin (EPO) on cancer patient survival, it is necessary to develop new agents that can be used to efficiently manage and treat cancer-related anemia. In this study, novel distinctive carbon dots, J-CDs, derived from jujube are designed, synthesized, and characterized. Based on the obtained results, this material comprises sp2 and sp3 carbon atoms, as well as oxygen/nitrogen-based groups, and it specifically promotes the proliferation of erythroid cells by stimulating the self-renewal of erythroid progenitor cells in vitro and in vivo. Moreover, J-CDs have no discernible effects on tumor proliferation and metastasis, unlike EPO. Transcriptome profiling suggests that J-CDs upregulate the molecules involved in hypoxia response, and they also significantly increase the phosphorylation levels of STAT5, the major transducer of signals for erythroid progenitor cell proliferation. Overall, this study demonstrates that J-CDs effectively promote erythrocyte production without affecting tumor proliferation and metastasis; thus, they may be promising agents for the treatment of cancer-related anemia.


Assuntos
Anemia , Eritropoetina , Neoplasias , Anemia/tratamento farmacológico , Anemia/patologia , Carbono/farmacologia , Carbono/uso terapêutico , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/fisiologia , Eritropoese/fisiologia , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Humanos , Neoplasias/complicações , Neoplasias/tratamento farmacológico
2.
Viral Immunol ; 33(8): 539-549, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32412895

RESUMO

Human parvovirus B19 (B19), a human pathogen of the erythroparvovirus genus, is responsible for a variety of diseases. B19 cause less symptoms in healthy individuals, also cause acute and chronic anemia in immunodeficiency patients. Transient aplastic crisis and pure red cell aplasia are two kinds of anemic hemogram, respectively, in acute and chronic B19 infection phase, especially occurring in patients with a shortened red cell survival or with immunodeficiency. In addition, B19-infected pregnant women may cause hydrops fetalis or fetal loss. B19 possesses high affinity to bone marrow and fetal liver due to its extremely restricted cytotoxicity to erythroid progenitor cells (EPCs) mediated by viral proteins. The nonstructural protein NS1 is considered to be the major pathogenic factor, which has been shown to inhibit the differentiation and maturation of EPCs through inducing viral DNA damage responses and cell cycle arrest. The time phase property of NS1 activity during DNA replication and conformity to transient change of hemogram are suggestive of its role in regulating differentiation of hematopoietic cells, which is not completely understood. In this review, we summarized the bridge between B19 NS1 and Notch signaling pathway or transcriptional factors GATA, which play an important role in erythroid cell proliferation and differentiation, to provide a new insight of the potential mechanism of B19-induced differential inhibition of EPCs.


Assuntos
Diferenciação Celular , Células Precursoras Eritroides/fisiologia , Células Precursoras Eritroides/virologia , Parvovirus B19 Humano/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Animais , Replicação do DNA , Feminino , Humanos , Camundongos , Infecções por Parvoviridae/virologia , Gravidez , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Replicação Viral
3.
Mol Biol Rep ; 47(5): 3909-3918, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32406020

RESUMO

Patients with ß-thalassemia suffer from a lack or absence of the beta-globin chain of normal hemoglobin (Hb). Therefore, an increase in fetal Hb (HbF) levels could improve the clinical status of these patients. Downregulation of BCL11A, a key regulatory transcription factor, could ameliorate the clinical status of thalassemic patients by increasing HbF levels. miR-30a expression and its relationship with the BCL11A gene in erythroid precursors was explored in patients with ß-thalassemia. The relevance of miR-30a to clinical parameters was also investigated. We evaluated the expressions of miR-30a, BCL11A, and γ-globin genes by quantitative real-time PCR (qRT-PCR) on isolated erythroid precursors from peripheral blood samples of ß-thalassemia intermedia (TI) patients and in bone marrow samples from healthy individuals as controls. The correlation between miR-30a expression and clinical indices that included HbF levels, ferritin, and the frequency of blood transfusions were assessed. We observed increased expression of miR-30a in conjunction with decreased BCL11A expression and elevated γ-globin and HbF levels. Patients with elevated miR-30a expression had a higher percentage of HbF and a lower level of ferritin. In addition, we observed that overexpression of miR-30a in erythroid precursor cells led to reduced BCL11A expression and was associated with elevated γ-globin expression. Our findings showed the importance of miR-30a in BCL11A and HbF regulation, and in the clinical status of patients with ß-thalassemia.


Assuntos
MicroRNAs/genética , Proteínas Repressoras/metabolismo , Talassemia beta/genética , Adulto , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/fisiologia , Feminino , Hemoglobina Fetal/genética , Regulação da Expressão Gênica , Humanos , Masculino , MicroRNAs/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Globinas beta/genética , Talassemia beta/metabolismo , gama-Globinas/genética , gama-Globinas/metabolismo
4.
Exp Hematol ; 80: 42-54.e4, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31756359

RESUMO

In contrast to steady-state erythropoiesis, which generates new erythrocytes at a constant rate, stress erythropoiesis rapidly produces a large bolus of new erythrocytes in response to anemic stress. In this study, we illustrate that Yes-associated protein (Yap1) promotes the rapid expansion of a transit-amplifying population of stress erythroid progenitors in vivo and in vitro. Yap1-mutated erythroid progenitors failed to proliferate in the spleen after transplantation into lethally irradiated recipient mice. Additionally, loss of Yap1 impaired the growth of actively proliferating erythroid progenitors in vitro. This role in proliferation is supported by gene expression profiles showing that transiently amplifying stress erythroid progenitors express high levels of genes associated with Yap1 activity and genes induced by Yap1. Furthermore, Yap1 promotes the proliferation of stress erythroid progenitors in part by regulating the expression of key glutamine-metabolizing enzymes. Thus, Yap1 acts as an erythroid regulator that coordinates the metabolic status with the proliferation of erythroid progenitors to promote stress erythropoiesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ciclo Celular/fisiologia , Células Precursoras Eritroides/fisiologia , Eritropoese/fisiologia , Regeneração/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Alelos , Animais , Divisão Celular , Células Cultivadas , Indução Enzimática , Células Precursoras Eritroides/citologia , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/biossíntese , Quimera por Radiação , Tolerância a Radiação , Proteínas Recombinantes/metabolismo , Baço/citologia , Estresse Fisiológico/genética , Fatores de Transcrição/genética , Proteínas de Sinalização YAP
5.
Am J Hematol ; 94(9): 963-974, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31148215

RESUMO

Malaria pathogenesis is caused by the replication of Plasmodium parasites within the red blood cells (RBCs) of the vertebrate host. This selective pressure has favored the evolution of protective polymorphisms in erythrocyte proteins, a subset of which serve as cognate receptors for parasite invasion ligands. Recently, the generation of RBCs from immortalized hematopoietic stem cells (HSCs) has offered a more tractable system for genetic manipulation and long-term in vitro culture, enabling elucidation of the functional determinants of host susceptibility in vitro. Here we report the generation of an immortalized erythroid progenitor cell line (EJ cells) from as few as 100 000 peripheral blood mononuclear cells. It offers a robust method for the creation of customized model systems from small volumes of peripheral blood. The EJ cell differentiation mirrored erythropoiesis of primary HSCs, yielding orthochromatic erythroblasts and enucleated RBCs after eight days (ejRBCs). The ejRBCs supported invasion by both P. vivax and P. falciparum. To demonstrate the genetic tractability of this system, we used CRISPR/Cas9 to disrupt the Duffy Antigen/Receptor for Chemokines (DARC) gene, which encodes the canonical receptor of P. vivax in humans. Invasion of P. vivax into this DARC-knockout cell line was strongly inhibited providing direct genetic evidence that P. vivax requires DARC for RBC invasion. Further, genetic complementation of DARC restored P. vivax invasion. Taken together, the peripheral blood immortalization method presented here offers the capacity to generate biologically representative model systems for studies of blood-stage malaria invasion from the peripheral blood of donors harboring unique genetic backgrounds, or rare polymorphisms.


Assuntos
Células Precursoras Eritroides , Malária Falciparum , Malária Vivax , Modelos Biológicos , Células-Tronco de Sangue Periférico , Plasmodium falciparum/metabolismo , Plasmodium vivax/metabolismo , Linhagem Celular Transformada , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/parasitologia , Células Precursoras Eritroides/fisiologia , Humanos , Malária Falciparum/metabolismo , Malária Falciparum/patologia , Malária Vivax/metabolismo , Malária Vivax/patologia , Células-Tronco de Sangue Periférico/metabolismo , Células-Tronco de Sangue Periférico/parasitologia , Células-Tronco de Sangue Periférico/patologia
6.
Dev Biol ; 442(1): 138-154, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30016639

RESUMO

Macrophages are well characterized as immune cells. However, in recent years, a multitude of non-immune functions have emerged many of which play essential roles in a variety of developmental processes (Wynn et al., 2013; DeFalco et al., 2014). In adult animals, macrophages are derived from circulating monocytes originating in the bone marrow, but much of the tissue-resident population arise from erythro-myeloid progenitors (EMPs) in the extra-embryonic yolk sac, appearing around the same time as primitive erythroblasts (Schulz et al., 2012; Kierdorf et al., 2013; McGrath et al., 2015; Gomez Perdiguero et al., 2015; Mass et al., 2016). Of particular interest to our group, macrophages have been shown to act as pro-angiogenic regulators during development (Wynn et al., 2013; DeFalco et al., 2014; Hsu et al., 2015), but there is still much to learn about these early cells. The goal of the present study was to isolate and expand progenitors of yolk-sac-derived Embryonic Macrophages (EMs) in vitro to generate a new platform for mechanistic studies of EM differentiation. To accomplish this goal, we isolated pure (>98%) EGFP+ populations by flow cytometry from embryonic day 9.5 (E9.5) Csf1r-EGFP+/tg mice, then evaluated the angiogenic potential of EMs relative to Bone Marrow-Derived Macrophages (BMDMs). We found that EMs expressed more pro-angiogenic and less pro-inflammatory macrophage markers than BMDMs. EMs also promoted more endothelial cell (EC) cord formation in vitro, as compared to BMDMs in a manner that required direct cell-to-cell contact. Importantly, EMs preferentially matured into microglia when co-cultured with mouse Neural Stem/Progenitor Cells (NSPCs). In conclusion, we have established a protocol to isolate and propagate EMs in vitro, have further defined specialized properties of yolk-sac-derived macrophages, and have identified EM-EC and EM-NSPC interactions as key inducers of EC tube formation and microglial cell maturation, respectively.


Assuntos
Células Precursoras Eritroides/fisiologia , Macrófagos/fisiologia , Células Progenitoras Mieloides/fisiologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Técnicas de Cocultura/métodos , Citometria de Fluxo/métodos , Células-Tronco Hematopoéticas/fisiologia , Macrófagos/citologia , Camundongos/embriologia , Fenótipo , Saco Vitelino/citologia
7.
J Vis Exp ; (142)2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30596387

RESUMO

Erythroid cell commitment and differentiation proceed through activation of a lineage-restricted transcriptional network orchestrated by a group of cell fate determining and maturing factors. We previously set out to define the minimal set of factors necessary for instructing red blood cell development using direct lineage reprogramming of fibroblasts into induced erythroid progenitors/precursors (iEPs). We showed that overexpression of Gata1, Tal1, Lmo2, and c-Myc (GTLM) can rapidly convert murine and human fibroblasts directly to iEPs that resemble bona fide erythroid cells in terms of morphology, phenotype, and gene expression. We intend that iEPs will provide an invaluable tool to study erythropoiesis and cell fate regulation. Here we describe the stepwise process of converting murine tail tip fibroblasts into iEPs via transcription factor-driven direct lineage reprogramming (DLR). In this example, we perform the reprogramming in fibroblasts from erythroid lineage-tracing mice that express the yellow fluorescent protein (YFP) under the control of the erythropoietin receptor gene (EpoR) promoter, enabling visualization of erythroid cell fate induction upon reprogramming. Following this protocol, fibroblasts can be reprogrammed into iEPs within five to eight days. While improvements can still be made to the process, we show that GTLM-mediated reprogramming is a rapid and direct process, yielding cells with properties of bona fide erythroid progenitor and precursor cells.


Assuntos
Células Precursoras Eritroides/fisiologia , Eritropoese/fisiologia , Fibroblastos/fisiologia , Regulação da Expressão Gênica , Engenharia Genética , Receptores da Eritropoetina/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula , Eritropoese/genética , Redes Reguladoras de Genes , Humanos , Camundongos , Regiões Promotoras Genéticas , Receptores da Eritropoetina/genética , Fatores de Transcrição/genética
8.
J Theor Biol ; 437: 286-298, 2018 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-29102644

RESUMO

Production of red blood cells involves growth-factor mediated regulation of erythroid progenitor apoptosis and self-renewal. During severe anemia, characterized by a strong fall of the hematocrit followed by a recovery phase, these controls allow a fast recovery of the hematocrit and survival of the organism. Using a mathematical model of stress erythropoiesis and an ad hoc numerical method, we investigate the respective roles of anemia-inducing phenylhydrazine injections and physiological regulation on the organism's recovery. By explicitly modeling the experimental protocol, we show that it mostly characterizes the fall of the hematocrit following the anemia and its severeness, while physiological process regulation mainly controls the recovery. We confront our model and our conclusions to similar experiments inducing anemia and show the model's ability to reproduce several protocols of phenylhydrazine-induced anemia. In particular, we establish a link between phenylhydrazine effect and the severeness of the anemia.


Assuntos
Algoritmos , Anemia/fisiopatologia , Eritropoese/fisiologia , Modelos Biológicos , Anemia/sangue , Anemia/induzido quimicamente , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Autorrenovação Celular , Simulação por Computador , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/fisiologia , Hematócrito , Camundongos , Fenil-Hidrazinas , Fatores de Tempo
9.
Anat Rec (Hoboken) ; 300(11): 1993-1999, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28681554

RESUMO

Cell suspensions of head kidney and spleen of black scorpionfish (Scorpaena porcus L.) have been studied using flow cytometry and light microscopy. On the basis of forward scatter (FS) and side scatter (SS) distribution and light microscopy, two main types of cells in the hemopoietic organs were identified: "small cells" (5.1-8.9 µm) and "large cells" (10.8-15.3 µm). Subpopulation of small cells was formed by thrombocytes, lymphocytes, and elements with functionally inactive nuclei. Euchromatin level in their nuclei was low and acidophilic cytoplasm corresponded to negligible nucleic acids content. No proliferative activity was observed using SYBR Green I fluorescence analysis. Morphological characteristics of these cells coincided with colony forming units of mammals. Large cells in head kidney consisted of two subpopulations of cells differing in granularity and DNA content. Proliferating blast cells and differentiating cells of all hemopoietic lines were identified among them. Macrophages and apoptotic cells were also detected in head kidney. In spleen large cells cluster mainly included aged red blood cells with extended lengthwise axis. Blast cells and differentiating elements in spleen were not observed. Anat Rec, 2017. © 2017 Wiley Periodicals, Inc. Anat Rec, 300:1993-1999, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Células Precursoras Eritroides/fisiologia , Hematopoese/fisiologia , Rim/fisiologia , Perciformes/fisiologia , Baço/fisiologia , Animais , Proliferação de Células/fisiologia , Feminino , Citometria de Fluxo , Rim/citologia , Macrófagos/fisiologia , Masculino , Microscopia , Modelos Animais , Baço/citologia
10.
J Biol Chem ; 292(31): 12735-12743, 2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28615441

RESUMO

Cellular iron homeostasis is maintained by iron and heme transport proteins that work in concert with ferrireductases, ferroxidases, and chaperones to direct the movement of iron into, within, and out of cells. Systemic iron homeostasis is regulated by the liver-derived peptide hormone, hepcidin. The interface between cellular and systemic iron homeostasis is readily observed in the highly dynamic iron handling of four main cell types: duodenal enterocytes, erythrocyte precursors, macrophages, and hepatocytes. This review provides an overview of how these cell types handle iron, highlighting how iron and heme transporters mediate the exchange and distribution of body iron in health and disease.


Assuntos
Homeostase , Ferro/fisiologia , Modelos Biológicos , Animais , Duodeno/citologia , Duodeno/fisiologia , Enterócitos/fisiologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/fisiologia , Eritropoese , Heme/efeitos adversos , Heme/metabolismo , Hepatócitos/fisiologia , Hepcidinas/fisiologia , Humanos , Absorção Intestinal , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Ferro/sangue , Ferro da Dieta/efeitos adversos , Ferro da Dieta/metabolismo , Macrófagos/imunologia , Macrófagos/fisiologia
11.
Biochem Pharmacol ; 136: 32-39, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28377277

RESUMO

Parvovirus B19 (B19V) infection is restricted to erythroid progenitor cells (EPCs) of the human bone marrow, leading to transient arrest of erythropoiesis and severe complications mainly in subjects with underlying hematological disorders or with immune system deficits. Currently, there are no specific antiviral drugs for B19V treatment, but identification of compounds inhibiting B19V replication can be pursued by a drug repositioning strategy. In this frame, the present study investigates the activity of hydroxyurea (HU), the only disease-modifying therapy approved for sickle cell disease (SCD), towards B19V replication in the two relevant cellular systems, the UT7/EpoS1 cell line and EPCs. Results demonstrate that HU inhibits B19V replication with EC50 values of 96.2µM and 147.1µM in UT7/EpoS1 and EPCs, respectively, providing experimental evidence of the antiviral activity of HU towards B19V replication, and confirming the efficacy of a drug discovery process by drug repositioning strategy. The antiviral activity occurs in vitro at concentrations lower than those affecting cellular DNA replication and viability, and at levels measured in plasma samples of SCD patients undergoing HU therapy. HU might determine a dual beneficial effect on SCD patients, not only for the treatment of the disease but also towards a virus responsible for severe complications.


Assuntos
Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/fisiologia , Hidroxiureia/farmacologia , Parvovirus B19 Humano/fisiologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia , Antivirais/farmacologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Células Precursoras Eritroides/virologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/fisiologia
12.
Cell Rep ; 16(5): 1470-1484, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27452463

RESUMO

Mass spectrometry-based proteomics now enables the absolute quantification of thousands of proteins in individual cell types. We used this technology to analyze the dynamic proteome changes occurring during human erythropoiesis. We quantified the absolute expression of 6,130 proteins during erythroid differentiation from late burst-forming units-erythroid (BFU-Es) to orthochromatic erythroblasts. A modest correlation between mRNA and protein expression was observed. We identified several proteins with unexpected expression patterns in erythroid cells, highlighting a breakpoint in the erythroid differentiation process at the basophilic stage. We also quantified the distribution of proteins between reticulocytes and pyrenocytes after enucleation. These analyses identified proteins that are actively sorted either with the reticulocyte or the pyrenocyte. Our study provides the absolute quantification of protein expression during a complex cellular differentiation process in humans, and it establishes a framework for future studies of disordered erythropoiesis.


Assuntos
Eritropoese/fisiologia , Proteoma/metabolismo , Diferenciação Celular , Células Cultivadas , Eritroblastos/metabolismo , Eritroblastos/fisiologia , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/fisiologia , Humanos , Proteômica/métodos , RNA Mensageiro/metabolismo
13.
Sci Rep ; 6: 28789, 2016 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-27352931

RESUMO

ASXL1 mutations are found in a spectrum of myeloid malignancies with poor prognosis. Recently, we reported that Asxl1(+/-) mice develop myelodysplastic syndrome (MDS) or MDS and myeloproliferative neoplasms (MPN) overlapping diseases (MDS/MPN). Although defective erythroid maturation and anemia are associated with the prognosis of patients with MDS or MDS/MPN, the role of ASXL1 in erythropoiesis remains unclear. Here, we showed that chronic myelomonocytic leukemia (CMML) patients with ASXL1 mutations exhibited more severe anemia with a significantly increased proportion of bone marrow (BM) early stage erythroblasts and reduced enucleated erythrocytes compared to CMML patients with WT ASXL1. Knockdown of ASXL1 in cord blood CD34(+) cells reduced erythropoiesis and impaired erythrocyte enucleation. Consistently, the BM and spleens of VavCre(+);Asxl1(f/f) (Asxl1(∆/∆)) mice had less numbers of erythroid progenitors than Asxl1(f/f) controls. Asxl1(∆/∆) mice also had an increased percentage of erythroblasts and a reduced erythrocyte enucleation in their BM compared to littermate controls. Furthermore, Asxl1(∆/∆) erythroblasts revealed altered expression of genes involved in erythroid development and homeostasis, which was associated with lower levels of H3K27me3 and H3K4me3. Our study unveils a key role for ASXL1 in erythropoiesis and indicates that ASXL1 loss hinders erythroid development/maturation, which could be of prognostic value for MDS/MPN patients.


Assuntos
Células Precursoras Eritroides/fisiologia , Eritropoese , Proteínas Repressoras/fisiologia , Animais , Apoptose , Diferenciação Celular , Epigênese Genética , Histonas/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
14.
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
15.
Sci China Life Sci ; 58(12): 1270-5, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26588913

RESUMO

Erythropoiesis is a process during which multipotential hematopoietic stem cells proliferate, differentiate and eventually form mature erythrocytes. Interestingly, unlike most cell types, an important feature of erythropoiesis is that following each mitosis the daughter cells are morphologically and functionally different from the parent cell from which they are derived, demonstrating the need to study erythropoiesis in a stage-specific manner. This has been impossible until recently due to lack of methods for isolating erythroid cells at each distinct developmental stage. This review summarizes recent advances in the development of methods for isolating both murine and human erythroid cells and their applications. These methods provide powerful means for studying normal and impaired erythropoiesis associated with hematological disorders.


Assuntos
Separação Celular/métodos , Eritrócitos/fisiologia , Células Precursoras Eritroides/fisiologia , Eritropoese/fisiologia , Citometria de Fluxo/métodos , Anemia/classificação , Anemia/fisiopatologia , Animais , Diferenciação Celular/fisiologia , Eritrócitos/citologia , Células Precursoras Eritroides/citologia , Humanos , Camundongos
16.
J Clin Invest ; 125(10): 3965-80, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26389678

RESUMO

Erythropoiesis is an important response to certain types of stress, including hypoxia, hemorrhage, bone marrow suppression, and anemia, that result in inadequate tissue oxygenation. This stress-induced erythropoiesis is distinct from basal red blood cell generation; however, neither the cellular nor the molecular factors that regulate this process are fully understood. Here, we report that type 1 conventional dendritic cells (cDC1s), which are defined by expression of CD8α in the mouse and XCR1 and CLEC9 in humans, are critical for induction of erythropoiesis in response to stress. Specifically, using murine models, we determined that engagement of a stress sensor, CD24, on cDC1s upregulates expression of the Kit ligand stem cell factor on these cells. The increased expression of stem cell factor resulted in Kit-mediated proliferative expansion of early erythroid progenitors and, ultimately, transient reticulocytosis in the circulation. Moreover, this stress response was triggered in part by alarmin recognition and was blunted in CD24 sensor- and CD8α+ DC-deficient animals. The contribution of the cDC1 subset to the initiation of stress erythropoiesis was distinct from the well-recognized role of macrophages in supporting late erythroid maturation. Together, these findings offer insight into the mechanism of stress erythropoiesis and into disorders of erythrocyte generation associated with stress.


Assuntos
Células Dendríticas/fisiologia , Eritropoese/fisiologia , Estresse Fisiológico/fisiologia , Alarminas/fisiologia , Animais , Antígeno CD24/fisiologia , Antígenos CD8/análise , Cisplatino/toxicidade , Ensaio de Unidades Formadoras de Colônias , Células Dendríticas/classificação , Células Precursoras Eritroides/fisiologia , Feminino , Perfilação da Expressão Gênica , Proteína HMGB1/toxicidade , Transplante de Células-Tronco Hematopoéticas , Xenoenxertos , Humanos , Hipóxia/fisiopatologia , Mesilato de Imatinib/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Flebotomia/efeitos adversos , Quimera por Radiação , Proteínas Recombinantes/toxicidade , Esplenectomia/efeitos adversos , Fator de Células-Tronco/biossíntese , Fator de Células-Tronco/genética
17.
Dev Growth Differ ; 57(6): 466-473, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26098172

RESUMO

Long noncoding RNAs (LncRNAs) are longer than 200 nucleotide noncoding RNAs without apparent functional coding capacity that function as regulators of cell growth and development. In recent years, increasing evidence implicates the involvement of LncRNAs in erythropoiesis. shlnc-EC6 is a LncRNA associated with erythroid differentiation but the mechanism remains undefined. In this study, we found that knockdown of shlnc-EC6 in purified mouse fetal liver erythroid progenitor and hematopoietic stem cells (FLEPHSCs) significantly blocked erythroid enucleation. We also showed that Rac1 was negatively regulated by shlnc-EC6 at the posttranscriptional level via specific binding to sites within the 3'UTR of Rac1 mRNA. Moreover, we found that knockdown of shlnc-EC6 led to upregulation of Rac1, followed by the activation of the downstream protein PIP5K, and subsequently resulted in the inhibition of enucleation in cultured mouse fetal erythroblasts. Thus, our findings suggest that shlnc-EC6 acts as a novel modulator to regulate mouse erythropoiesis via Rac1/PIP5K signaling pathway.


Assuntos
Núcleo Celular/genética , Células Eritroides/metabolismo , Células Precursoras Eritroides/fisiologia , Eritropoese/genética , RNA Longo não Codificante/fisiologia , RNA Interferente Pequeno/fisiologia , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos , Eritrócitos/fisiologia , Células Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , RNA Longo não Codificante/farmacologia , RNA Interferente Pequeno/farmacologia
18.
Leukemia ; 29(11): 2221-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25987256

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital erythroid hypoplasia caused by haploinsufficiency of genes encoding ribosomal proteins (RPs). Perturbed ribosome biogenesis in DBA has been shown to induce a p53-mediated ribosomal stress response. However, the mechanisms of p53 activation and its relevance for the erythroid defect remain elusive. Previous studies have indicated that activation of p53 is caused by the inhibition of mouse double minute 2 (Mdm2), the main negative regulator of p53, by the 5S ribonucleoprotein particle (RNP). Meanwhile, it is not clear whether this mechanism solely mediates the p53-dependent component found in DBA. To approach this question, we crossed our mouse model for RPS19-deficient DBA with Mdm2(C305F) knock-in mice that have a disrupted 5S RNP-Mdm2 interaction. Upon induction of the Rps19 deficiency, Mdm2(C305F) reversed the p53 response and improved expansion of hematopoietic progenitors in vitro, and ameliorated the anemia in vivo. Unexpectedly, disruption of the 5S RNP-Mdm2 interaction also led to selective defect in erythropoiesis. Our findings highlight the sensitivity of erythroid progenitor cells to aberrations in p53 homeostasis mediated by the 5S RNP-Mdm2 interaction. Finally, we provide evidence indicating that physiological activation of the 5S RNP-Mdm2-p53 pathway may contribute to functional decline of the hematopoietic system in a cell-autonomous manner over time.


Assuntos
Anemia de Diamond-Blackfan/etiologia , Células Precursoras Eritroides/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Ribonucleoproteínas/fisiologia , Animais , Modelos Animais de Doenças , Doxiciclina/farmacologia , Eritropoese , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 5S/fisiologia , Proteínas Ribossômicas/fisiologia , Transdução de Sinais , Proteína Supressora de Tumor p53/fisiologia
19.
J Infect Dis ; 212(7): 1070-81, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25805750

RESUMO

Human parvovirus B19 (B19V) is a common pathogen in microvascular disease and cardiomyopathy, owing to infection of endothelial cells. B19V replication, however, is almost restricted to erythroid progenitor cells (ErPCs). Endothelial regeneration attributable to bone marrow-derived circulating angiogenic cells (CACs) is a prerequisite for organ function. Because of many similarities of ErPCs and CACs, we hypothesized that B19V is a perpetrator of impaired endogenous endothelial regeneration. B19V DNA and messenger RNA from endomyocardial biopsy specimens, bone marrow specimens, and circulating progenitor cells were quantified by polymerase chain reaction analysis. The highest B19V DNA concentrations were found in CD34(+)KDR(+) cells from 17 patients with chronic B19V-associated cardiomyopathy. B19V replication intermediates could be detected in nearly half of the patients. Furthermore, chronic B19V infection was associated with impaired endothelial regenerative capacity. B19V infection of CACs in vitro resulted in expression of transcripts encoding B19V proteins. The capsid protein VP1 was identified as a novel inducer of apoptosis, as were nonstructural proteins. Inhibition studies identified so-called death receptor signaling with activation of caspase-8 and caspase-10 to be responsible for apoptosis induction. B19V causally impaired endothelial regeneration with spreading of B19V in CACs in an animal model in vivo. We thus conclude that B19V infection and damage to CACs result in dysfunctional endogenous vascular repair, supporting the emergence of primary bone marrow disease with secondary end-organ damage.


Assuntos
Apoptose , Cardiomiopatias/complicações , Eritema Infeccioso/virologia , Células Precursoras Eritroides/virologia , Parvovirus B19 Humano/fisiologia , Adulto , Idoso , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Estudos de Casos e Controles , Caspase 10/genética , Caspase 10/metabolismo , Linhagem Celular , Células Endoteliais/fisiologia , Células Endoteliais/virologia , Células Precursoras Eritroides/fisiologia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Parvovirus B19 Humano/genética , Regeneração , Transdução de Sinais , Replicação Viral
20.
Bull Exp Biol Med ; 158(4): 417-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25711660

RESUMO

The involvement of PI3K, ERK and p38-dependent signaling system in the regulation of functional activity of erythroid precursors after blood loss (30% of circulating volume) was studied. We demonstrated the important role of PI3K and p38 in the suppression of differentiation of erythroid precursors the contribution of p38 to stimulation of mitotic activity of erythroid CFU, which maintains the growth potential of the precursors at the optimal physiological level. The classical MAPK/ERK-kinase pathway does not determine the proliferative and differentiation status of erythroid CFU.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Precursoras Eritroides/fisiologia , Hemorragia/fisiopatologia , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Ensaio de Unidades Formadoras de Colônias , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estatísticas não Paramétricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...