Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.184
Filtrar
1.
Exp Hematol ; 106: 19-30, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34879257

RESUMO

Calcium (Ca2+) is an important second messenger molecule in the body, regulating cell cycle and fate. There is growing evidence that intracellular Ca2+ levels play functional roles in the total physiological process of erythroid differentiation, including the proliferation and differentiation of erythroid progenitor cells, terminal enucleation, and mature red blood cell aging and clearance. Moreover, recent research on the pathology of erythroid disorders has made great progress in the past decades, indicating that calcium ion hemostasis is closely related to ineffective erythropoiesis and increased sensitivity to stress factors. In this review, we summarized what is known about the functional roles of intracellular Ca2+ in erythropoiesis and erythrocyte-related diseases, with an emphasis on the regulation of the intracellular Ca2+ homeostasis during erythroid differentiation. An understanding of the regulation roles of Ca2+ homeostasis in erythroid differentiation will facilitate further studies and eventually molecular identification of the pathways involved in the pathological process of erythroid disorders, providing new therapeutic opportunities in erythrocyte-related disease.


Assuntos
Cálcio/metabolismo , Eritropoese , Animais , Cátions Bivalentes/metabolismo , Eritrócitos/citologia , Eritrócitos/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Humanos
2.
Genes (Basel) ; 12(8)2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34440315

RESUMO

Hydroxyurea (HU) causes nitric oxide (NO) bioactivation, acting as both a NO donor and a stimulator of NO synthase (NOS). To examine whether HU effects are NO mediated by chemical degradation or enzymatic induction, we studied human and mouse erythroid cells during proliferation, apoptosis, and differentiation. The HU and NO donor demonstrated persisted versus temporary inhibition of erythroid cell growth during differentiation, as observed by γ- and ß-globin gene expression. HU decreased the percentage of erythroleukemic K562 cells in the G2/M phase that was reversed by N-nitro l-arginine methyl ester hydrochloride (L-NAME). Besides activation of endothelial NOS, HU significantly increased apoptosis of K562 cells, again demonstrating NOS dependence. Administration of HU to mice significantly inhibited colony-forming unit-erythroid (CFU-E), mediated by NOS. Moreover, burst-forming-units-erythroid (BFU-E) and CFU-E ex vivo growth was inhibited by the administration of nitrate or nitrite to mice. Chronic in vivo NOS inhibition with L-NAME protected the bone marrow cellularity despite HU treatment of mice. NO metabolites and HU reduced the frequency of NOS-positive cells from CFU-E and BFU-E colonies that was reverted by NOS inhibition. HU regulation of the G2/M phase, apoptosis, differentiation, cellularity, and NOS immunoreactive cells was NOS dependent. Inhalation of NO therapy as well as strategies to increase endogenous NO production could replace or enhance HU activity.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Precursoras Eritroides/efeitos dos fármacos , Hidroxiureia/farmacologia , Óxido Nítrico Sintase/metabolismo , Animais , Células Precursoras Eritroides/citologia , Humanos , Células K562 , Camundongos , Doadores de Óxido Nítrico/farmacologia
3.
Cell Rep ; 36(6): 109507, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34380040

RESUMO

Survival or apoptosis is a binary decision in individual cells. However, at the cell-population level, a graded increase in survival of colony-forming unit-erythroid (CFU-E) cells is observed upon stimulation with erythropoietin (Epo). To identify components of Janus kinase 2/signal transducer and activator of transcription 5 (JAK2/STAT5) signal transduction that contribute to the graded population response, we extended a cell-population-level model calibrated with experimental data to study the behavior in single cells. The single-cell model shows that the high cell-to-cell variability in nuclear phosphorylated STAT5 is caused by variability in the amount of Epo receptor (EpoR):JAK2 complexes and of SHP1, as well as the extent of nuclear import because of the large variance in the cytoplasmic volume of CFU-E cells. 24-118 pSTAT5 molecules in the nucleus for 120 min are sufficient to ensure cell survival. Thus, variability in membrane-associated processes is sufficient to convert a switch-like behavior at the single-cell level to a graded population-level response.


Assuntos
Citoplasma/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Janus Quinase 2/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Animais , Calibragem , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Simulação por Computador , Eritropoetina/farmacologia , Camundongos Endogâmicos BALB C , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
4.
Genes (Basel) ; 12(7)2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34208866

RESUMO

Hematopoiesis is a convenient model to study how chromatin dynamics plays a decisive role in regulation of cell fate. During erythropoiesis a population of stem and progenitor cells becomes increasingly lineage restricted, giving rise to terminally differentiated progeny. The concerted action of transcription factors and epigenetic modifiers leads to a silencing of the multipotent transcriptome and activation of the transcriptional program that controls terminal differentiation. This article reviews some aspects of the biology of red blood cells production with the focus on the extensive chromatin reorganization during differentiation.


Assuntos
Diferenciação Celular , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Células Precursoras Eritroides/citologia , Eritropoese , Regulação da Expressão Gênica , Fatores de Transcrição/metabolismo , Animais , Genoma , Humanos , Fatores de Transcrição/genética
5.
Am J Hematol ; 96(9): 1064-1076, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34021930

RESUMO

Identification of stage-specific erythroid cells is critical for studies of normal and disordered human erythropoiesis. While immunophenotypic strategies have previously been developed to identify cells at each stage of terminal erythroid differentiation, erythroid progenitors are currently defined very broadly. Refined strategies to identify and characterize BFU-E and CFU-E subsets are critically needed. To address this unmet need, a flow cytometry-based technique was developed that combines the established surface markers CD34 and CD36 with CD117, CD71, and CD105. This combination allowed for the separation of erythroid progenitor cells into four discrete populations along a continuum of progressive maturation, with increasing cell size and decreasing nuclear/cytoplasmic ratio, proliferative capacity and stem cell factor responsiveness. This strategy was validated in uncultured, primary erythroid cells isolated from bone marrow of healthy individuals. Functional colony assays of these progenitor populations revealed enrichment of BFU-E only in the earliest population, transitioning to cells yielding BFU-E and CFU-E, then CFU-E only. Utilizing CD34/CD105 and GPA/CD105 profiles, all four progenitor stages and all five stages of terminal erythroid differentiation could be identified. Applying this immunophenotyping strategy to primary bone marrow cells from patients with myelodysplastic syndrome, identified defects in erythroid progenitors and in terminal erythroid differentiation. This novel immunophenotyping technique will be a valuable tool for studies of normal and perturbed human erythropoiesis. It will allow for the discovery of stage-specific molecular and functional insights into normal erythropoiesis as well as for identification and characterization of stage-specific defects in inherited and acquired disorders of erythropoiesis.


Assuntos
Células Eritroides/citologia , Células Precursoras Eritroides/citologia , Eritropoese , Antígenos CD/análise , Antígenos CD34/análise , Células da Medula Óssea/citologia , Células Cultivadas , Endoglina/análise , Citometria de Fluxo/métodos , Humanos , Imunofenotipagem/métodos
6.
Am J Hum Genet ; 108(4): 709-721, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33735615

RESUMO

The fetal-to-adult hemoglobin switch is regulated in a developmental stage-specific manner and reactivation of fetal hemoglobin (HbF) has therapeutic implications for treatment of ß-thalassemia and sickle cell anemia, two major global health problems. Although significant progress has been made in our understanding of the molecular mechanism of the fetal-to-adult hemoglobin switch, the mechanism of epigenetic regulation of HbF silencing remains to be fully defined. Here, we performed whole-genome bisulfite sequencing and RNA sequencing analysis of the bone marrow-derived GYPA+ erythroid cells from ß-thalassemia-affected individuals with widely varying levels of HbF groups (HbF ≥ 95th percentile or HbF ≤ 5th percentile) to screen epigenetic modulators of HbF and phenotypic diversity of ß-thalassemia. We identified an ETS2 repressor factor encoded by ERF, whose promoter hypermethylation and mRNA downregulation are associated with high HbF levels in ß-thalassemia. We further observed that hypermethylation of the ERF promoter mediated by enrichment of DNMT3A leads to demethylation of γ-globin genes and attenuation of binding of ERF on the HBG promoter and eventually re-activation of HbF in ß-thalassemia. We demonstrated that ERF depletion markedly increased HbF production in human CD34+ erythroid progenitor cells, HUDEP-2 cell lines, and transplanted NCG-Kit-V831M mice. ERF represses γ-globin expression by directly binding to two consensus motifs regulating γ-globin gene expression. Importantly, ERF depletion did not affect maturation of erythroid cells. Identification of alterations in DNA methylation of ERF as a modulator of HbF synthesis opens up therapeutic targets for ß-hemoglobinopathies.


Assuntos
Epigênese Genética , Perfilação da Expressão Gênica , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Talassemia beta/genética , gama-Globinas/genética , Animais , Antígenos CD34/metabolismo , Sequência de Bases , Sistemas CRISPR-Cas/genética , Diferenciação Celular , Linhagem Celular , Criança , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , DNA Metiltransferase 3A , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Feminino , Hemoglobina Fetal/genética , Edição de Genes , Humanos , Masculino , Camundongos , Regiões Promotoras Genéticas/genética , Reprodutibilidade dos Testes , Sulfitos , Sequenciamento Completo do Genoma , Talassemia beta/patologia
7.
Exp Hematol ; 97: 32-46.e35, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33675821

RESUMO

Oxygen is a critical noncellular component of the bone marrow microenvironment that plays an important role in the development of hematopoietic cell lineages. In this study, we investigated the impact of low oxygen (hypoxia) on ex vivo myeloerythroid differentiation of human cord blood-derived CD34+ hematopoietic stem and progenitor cells. We characterized the culture conditions to demonstrate that low oxygen inhibits cell proliferation and causes a metabolic shift in the stem and progenitor populations. We found that hypoxia promotes erythroid differentiation by supporting the development of progenitor populations. Hypoxia also increases the megakaryoerythroid potential of the common myeloid progenitors and the erythroid potential of megakaryoerythroid progenitors and significantly accelerates maturation of erythroid cells. Specifically, we determined that hypoxia promotes the loss of CD71 and the appearance of the erythroid markers CD235a and CD239. Further, evaluation of erythroid populations revealed a hypoxia-induced increase in proerythroblasts and in enucleation of CD235a+ cells. These results reveal the extensive role of hypoxia at multiple steps during erythroid development. Overall, our work establishes a valuable model for further investigations into the relationship between erythroid progenitors and/or erythroblast populations and their hypoxic microenvironment.


Assuntos
Eritroblastos/citologia , Células Eritroides/citologia , Células Precursoras Eritroides/citologia , Eritropoese , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Eritroblastos/metabolismo , Células Eritroides/metabolismo , Células Precursoras Eritroides/metabolismo , Humanos , Metaboloma
8.
PLoS One ; 16(3): e0247858, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33661998

RESUMO

Myeloproliferative neoplasms (MPNs) cause the over-production of blood cells such as erythrocytes (polycythemia vera) or platelets (essential thrombocytosis). JAK2 V617F is the most prevalent somatic mutation in many MPNs, but previous modeling of this mutation in mice relied on transgenic overexpression and resulted in diverse phenotypes that were in some cases attributed to expression level. CRISPR-Cas9 engineering offers new possibilities to model and potentially cure genetically encoded disorders via precise modification of the endogenous locus in primary cells. Here we develop "scarless" Cas9-based reagents to create and reverse the JAK2 V617F mutation in an immortalized human erythroid progenitor cell line (HUDEP-2), CD34+ adult human hematopoietic stem and progenitor cells (HSPCs), and immunophenotypic long-term hematopoietic stem cells (LT-HSCs). We find no overt in vitro increase in proliferation associated with an endogenous JAK2 V617F allele, but co-culture with wild type cells unmasks a competitive growth advantage provided by the mutation. Acquisition of the V617F allele also promotes terminal differentiation of erythroid progenitors, even in the absence of hematopoietic cytokine signaling. Taken together, these data are consistent with the gradually progressive manifestation of MPNs and reveals that endogenously acquired JAK2 V617F mutations may yield more subtle phenotypes as compared to transgenic overexpression models.


Assuntos
Edição de Genes , Transtornos Mieloproliferativos/genética , Sistemas CRISPR-Cas , Linhagem Celular , Técnicas de Cocultura , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Janus Quinase 2/genética
9.
J Cell Mol Med ; 25(5): 2377-2389, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33496386

RESUMO

The exact molecular mechanism underlying erythroblast enucleation has been a fundamental biological question for decades. In this study, we found that miR-144/451 critically regulated erythroid differentiation and enucleation. We further identified CAP1, a G-actin-binding protein, as a direct target of miR-144/451 in these processes. During terminal erythropoiesis, CAP1 expression declines along with gradually increased miR-144/451 levels. Enforced CAP1 up-regulation inhibits the formation of contractile actin rings in erythroblasts and prevents their terminal differentiation and enucleation. Our findings reveal a negative regulatory role of CAP1 in miR-144/451-mediated erythropoiesis and thus shed light on how microRNAs fine-tune terminal erythroid development through regulating actin dynamics.


Assuntos
Diferenciação Celular/genética , Células Precursoras Eritroides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Interferência de RNA , Serina Endopeptidases/genética , Regiões 3' não Traduzidas , Animais , Biomarcadores , Linhagem Celular , Células Cultivadas , Células Precursoras Eritroides/citologia , Eritropoese/genética , Imunofenotipagem , Camundongos
10.
Blood Cells Mol Dis ; 87: 102533, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33352376

RESUMO

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). The article has been retracted at the request of the editor. The journal was informed by Dr Xiangmin Xu and Dr Yongzhong Zhao that they were not involved in the study or research and that the article was submitted without their knowledge. As such this article represents a misuse of the scientific publishing system. The scientific community takes a very strong view on this matter and apologies are offered to readers of the journal that this was not detected during the submission process. All authors were informed of the article retraction however Dr Li and Dr Zeng did not respond to the enquiries.


Assuntos
Células Eritroides/citologia , Células Precursoras Eritroides/citologia , Eritropoese , Fatores de Transcrição Kruppel-Like/genética , alfa-Globinas/genética , Linhagem Celular , Epigênese Genética , Células Eritroides/metabolismo , Células Precursoras Eritroides/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Regiões Promotoras Genéticas , Ativação Transcricional
11.
Mol Cell ; 81(2): 239-254.e8, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33301730

RESUMO

Metazoan transcription factors typically regulate large numbers of genes. Here we identify via a CRISPR-Cas9 genetic screen ZNF410, a pentadactyl DNA-binding protein that in human erythroid cells directly activates only a single gene, the NuRD component CHD4. Specificity is conveyed by two highly evolutionarily conserved clusters of ZNF410 binding sites near the CHD4 gene with no counterparts elsewhere in the genome. Loss of ZNF410 in adult-type human erythroid cell culture systems and xenotransplantation settings diminishes CHD4 levels and derepresses the fetal hemoglobin genes. While previously known to be silenced by CHD4, the fetal globin genes are exposed here as among the most sensitive to reduced CHD4 levels.. In vitro DNA binding assays and crystallographic studies reveal the ZNF410-DNA binding mode. ZNF410 is a remarkably selective transcriptional activator in erythroid cells, and its perturbation might offer new opportunities for treatment of hemoglobinopathies.


Assuntos
DNA/genética , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Fatores de Transcrição/genética , Animais , Sítios de Ligação , Células COS , Sistemas CRISPR-Cas , Chlorocebus aethiops , DNA/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/transplante , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Hemoglobina Fetal/metabolismo , Feto , Edição de Genes , Células HEK293 , Xenoenxertos , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/química , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Camundongos , Modelos Moleculares , Células-Tronco Embrionárias Murinas/citologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Ativação Transcricional
12.
Blood Rev ; 46: 100740, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32798012

RESUMO

A characteristic feature of terminal erythropoiesis in mammals is extrusion of the highly condensed nucleus out of the cytoplasm. Other vertebrates, including fish, reptiles, amphibians, and birds, undergo nuclear condensation but do not enucleate. Enucleation provides mammals evolutionary advantages by gaining extra space for hemoglobin and being more flexible to migrate through capillaries. Nascent reticulocytes further mature into red blood cells through membrane and proteome remodeling and organelle clearance. Over the past decade, novel molecular mechanisms and signaling pathways have been uncovered that play important roles in chromatin condensation, enucleation, and reticulocyte maturation. These advances not only increase understanding of the physiology of erythropoiesis, but also facilitate efforts in generating in vitro red blood cells for various translational application. In the present review, recent studies in epigenetic modification and release of histones during chromatin condensation are highlighted. New insights in enucleation, including protein sorting, vesicle trafficking, transcriptional regulation, noncoding RNA, cytoskeleton remodeling, erythroblastic islands, and cytokinesis, are summarized. Moreover, organelle clearance and proteolysis mediated by ubiquitin-proteasome degradation during reticulocytes maturation is also examined. Perspectives for future directions in this rapidly evolving research area are also provided.


Assuntos
Diferenciação Celular , Eritrócitos , Eritropoese/fisiologia , Animais , Núcleo Celular , Cromatina/genética , Cromatina/metabolismo , Eritroblastos/citologia , Eritroblastos/metabolismo , Eritrócitos/citologia , Eritrócitos/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Regulação da Expressão Gênica , Humanos , Reticulócitos/citologia , Reticulócitos/metabolismo
14.
Circ Res ; 127(10): 1221-1232, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-32791884

RESUMO

RATIONALE: Endothelial cells are thought to emerge de novo from the mesoderm to form the entire circulatory system. Recently, erythro-myeloid progenitors (EMPs) have been proposed to be another remarkable developmental origin for blood vessels in multiple organs, including the hindbrain, liver, lung, and heart, as demonstrated by lineage tracing studies using different genetic tools. These observations challenge the current consensus that intraembryonic vessels are thought to expand solely by the proliferation of preexisting endothelial cells. Resolution of this controversy over the developmental origin of endothelial cells is crucial for developing future therapeutics for vessel-dependent organ repair and regeneration. OBJECTIVE: To examine the contribution of EMPs to intraembryonic endothelial cells. METHODS AND RESULTS: We first used a transgenic mouse expressing a tamoxifen-inducible Mer-iCre fusion protein driven by the Csf1r (colony stimulating factor 1 receptor) promoter. Genetic lineage tracing based on Csf1r-Mer-iCre-Mer showed no contribution of EMPs to brain endothelial cells identified by several markers. We also generated a knock-in mouse line by inserting an internal ribosome entry site-iCre cassette into the 3' untranslated region of Csf1r gene to further investigate the cellular fates of EMPs. Similarly, we did not find any Csf1r-ires-iCre traced endothelial cells in brain, liver, lung, or heart in development either. Additionally, we found that Kit (KIT proto-oncogene receptor tyrosine kinase) was expressed not only in EMPs but also in embryonic hindbrain endothelial cells. Therefore, Kit promoter-driven recombinase, such as Kit-CreER, is a flawed tool for lineage tracing when examining the contribution of EMPs to hindbrain endothelial cells. We also traced CD45 (protein tyrosine phosphatase receptor type C; Ptprc)+ circulating EMPs and did not find any CD45 lineage-derived endothelial cells during development. CONCLUSIONS: Our study suggested that EMPs are not the origin of intraembryonic endothelial cells.


Assuntos
Linhagem da Célula , Células Endoteliais/citologia , Células Precursoras Eritroides/citologia , Animais , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Coração Fetal/citologia , Fígado/citologia , Fígado/embriologia , Pulmão/citologia , Pulmão/embriologia , Macrófagos/citologia , Mesoderma/citologia , Camundongos , Rombencéfalo/citologia , Rombencéfalo/embriologia
15.
Exp Hematol ; 89: 1-12, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32798645

RESUMO

I am deeply honored to receive the International Society for Experimental Hematology (ISEH) 2020 Donald Metcalf Lecture Award. Although I am not a physician and have had no formal training in hematology, I have had the privilege of working with some of the top hematologists in the world, beginning in 1970 when Dr. David Nathan was a sabbatical visitor in my laboratory and introduced me to hematological diseases.  And I take this award to be given not just to me but to an exceptional group of MD and PhD trainees and visitors in my laboratory who have cloned and characterized many proteins and RNAs important for red cell development and function. Many of these projects involved taking exceptionally large risks in developing and employing novel experimental technologies. Unsurprisingly, all of these trainees have gone on to become leaders in hematology and, more broadly, in molecular cell biology and molecular medicine. To illustrate some of the challenges we have faced and the technologies we had to develop, I have chosen several of our multiyear projects to describe in some detail: elucidating the regulation of translation of α- and ß-globin mRNAs and the defect in beta thalassemia in the 1970s; cloning the Epo receptor and several red cell membrane proteins in the 1980s and 1990s; and more recently, determining the function of many microRNAs and long noncoding RNAs in red cell development. I summarize how we are currently utilizing single-cell transcriptomics (scRNAseq) to understand how dividing transit-amplifying burst-forming unit erythroid progenitors balance the need for more progenitor cells with the need for terminally differentiated erythroid cells, and to identify drugs potentially useful in treating Epo-resistant anemias such as Diamond Blackfan anemia. I hope that the lessons I learned in managing these diverse fellows and projects, initially without having grants to support them, will be helpful to others who would like to undertake ambitious and important lines of research in hematology.


Assuntos
Células Precursoras Eritroides/metabolismo , Hematologia/história , Biologia Molecular/história , Receptores da Eritropoetina/história , Talassemia beta/genética , Clonagem Molecular , Eritrócitos/metabolismo , Eritrócitos/patologia , Células Precursoras Eritroides/citologia , Eritropoese/genética , Expressão Gênica , História do Século XX , História do Século XXI , Humanos , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , alfa-Globinas/genética , alfa-Globinas/metabolismo , Globinas beta/genética , Globinas beta/metabolismo , Talassemia beta/metabolismo , Talassemia beta/patologia
16.
Exp Hematol ; 89: 43-54.e2, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32750404

RESUMO

Steady-state erythropoiesis generates new erythrocytes at a constant rate, and it has enormous productive capacity. This production is balanced by the removal of senescent erythrocytes by macrophages in the spleen and liver. Erythroid homeostasis is highly regulated to maintain sufficient erythrocytes for efficient oxygen delivery to the tissues, while avoiding viscosity problems associated with overproduction. However, there are times when this constant production of erythrocytes is inhibited or is inadequate; at these times, erythroid output is increased to compensate for the loss of production. In some cases, increased steady-state erythropoiesis can offset the loss of erythrocytes but, in response to inflammation caused by infection or tissue damage, steady-state erythropoiesis is inhibited. To maintain homeostasis under these conditions, an alternative stress erythropoiesis pathway is activated. Emerging data suggest that the bone morphogenetic protein 4 (BMP4)-dependent stress erythropoiesis pathway is integrated into the inflammatory response and generates a bolus of new erythrocytes that maintain homeostasis until steady-state erythropoiesis can resume. In this perspective, we define the mechanisms that generate new erythrocytes when steady-state erythropoiesis is impaired and discuss experimental models to study human stress erythropoiesis.


Assuntos
Proteína Morfogenética Óssea 4/genética , Eritrócitos/citologia , Células Precursoras Eritroides/citologia , Eritropoese/genética , Macrófagos/citologia , Estresse Fisiológico/genética , Animais , Proteína Morfogenética Óssea 4/imunologia , Senescência Celular/imunologia , Citocinas/genética , Citocinas/imunologia , Eritrócitos/imunologia , Células Precursoras Eritroides/imunologia , Eritropoese/imunologia , Regulação da Expressão Gênica , Humanos , Inflamação , Fígado/citologia , Fígado/imunologia , Macrófagos/imunologia , Camundongos , Modelos Biológicos , Fagocitose , Baço/citologia , Baço/imunologia , Estresse Fisiológico/imunologia
17.
Sci Rep ; 10(1): 11806, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32678227

RESUMO

The molecular mechanisms involved in the terminal differentiation of erythroblasts have been elucidated by comparing enucleation and cell division. Although various similarities and differences between erythroblast enucleation and cytokinesis have been reported, the mechanisms that control enucleation remain unclear. We previously reported that dynein and microtubule-organizing centers mediated the polarization of nuclei in human erythroblasts. Moreover, the accumulation of F-actin was noted during the enucleation of erythroblasts. Therefore, during enucleation, upstream effectors in the signal transduction pathway regulating dynein or actin, such as cell division control protein 42 homolog (Cdc42), may be crucial. We herein investigated the effects of the Cdc42 inhibitor, CASIN, on cytokinesis and enucleation in colony-forming units-erythroid (CFU-Es) and mature erythroblasts (day 10). CASIN blocked the proliferation of CFU-Es and their enucleation in a dose-dependent manner. Dynein adopted an island-like distribution in the cytoplasm of non-treated CFU-Es, but was concentrated near the nucleus as a dot and co-localized with γ-tubulin in CASIN-treated cells. CASIN blocked the accumulation of F-actin in CFU-Es and day 10 cells. These results demonstrated that Cdc42 plays an important role in cytokinesis, nuclear polarization and nuclear extrusion through a relationship with dynein and actin filament organization during the terminal differentiation of erythroblasts.


Assuntos
Actomiosina/metabolismo , Diferenciação Celular , Eritroblastos/citologia , Eritroblastos/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Biomarcadores , Diferenciação Celular/genética , Núcleo Celular/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Eritropoese/genética , Imunofluorescência , Expressão Gênica , Humanos , Imuno-Histoquímica , Proteína cdc42 de Ligação ao GTP/metabolismo
18.
Blood ; 136(9): 1044-1054, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32548608

RESUMO

Erythropoietin (EPO) provides the major survival signal to maturing erythroid precursors (EPs) and is essential for terminal erythropoiesis. Nonetheless, progenitor cells can irreversibly commit to an erythroid fate well before EPO acts, risking inefficiency if these progenitors are unneeded to maintain red blood cell (RBC) counts. We identified a new modular organization of erythropoiesis and, for the first time, demonstrate that the pre-EPO module is coupled to late EPO-dependent erythropoiesis by megakaryocyte (Mk) signals. Disrupting megakaryocytic transforming growth factor ß1 (Tgfb1) disorganized hematopoiesis by expanding the pre-EPO pool of progenitor cells and consequently triggering significant apoptosis of EPO-dependent EPs. Similarly, pharmacologic blockade of TGFß signaling in normal mice boosted the pre-EPO module, leading to apoptosis of EPO-sensitive EPs. Subsequent treatment with low-dose EPO triggered robust RBC production in both models. This work reveals modular regulation of erythropoiesis and offers a new strategy for overcoming chronic anemias.


Assuntos
Células Precursoras Eritroides/citologia , Eritropoese/fisiologia , Megacariócitos/citologia , Fator de Crescimento Transformador beta1/fisiologia , Animais , Apoptose/efeitos dos fármacos , Medula Óssea/patologia , Células Precursoras Eritroides/metabolismo , Eritropoetina/farmacologia , Técnicas de Inativação de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Imunofenotipagem , Células Progenitoras de Megacariócitos e Eritrócitos/citologia , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Quimera por Radiação , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/farmacologia
19.
Nat Cell Biol ; 22(6): 630-639, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32367048

RESUMO

How transplanted haematopoietic stem cells (HSCs) behave soon after they reside in a preconditioned host has not been studied due to technical limitations. Here, using single-cell RNA sequencing, we first obtained the transcriptome-based classifications of 28 haematopoietic cell types. We then applied them in conjunction with functional assays to track the dynamic changes of immunophenotypically purified HSCs in irradiated recipients within the first week after transplantation. Based on our transcriptional classifications, most homed HSCs in bone marrow and spleen became multipotent progenitors and, occasionally, some HSCs gave rise to megakaryocytic-erythroid or myeloid precursors. Parallel in vitro and in vivo functional experiments supported the paradigm of robust differentiation without substantial HSC expansion during the first week. Therefore, this study uncovers the previously inaccessible kinetics and fate choices of transplanted HSCs in myeloablated recipients at early stage, with implications for clinical applications of HSCs and other stem cells.


Assuntos
Diferenciação Celular , Células Precursoras Eritroides/citologia , Células-Tronco Hematopoéticas/citologia , Megacariócitos/citologia , Células Mieloides/citologia , Análise de Célula Única/métodos , Transcriptoma , Animais , Ciclo Celular , Linhagem da Célula , Células Precursoras Eritroides/metabolismo , Feminino , Células-Tronco Hematopoéticas/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo
20.
Blood ; 135(25): 2302-2315, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32384137

RESUMO

Erythropoiesis is a complex multistage process that involves differentiation of early erythroid progenitors to enucleated mature red blood cells, in which lineage-specific transcription factors play essential roles. Erythroid Krüppel-like factor (EKLF/KLF1) is a pleiotropic erythroid transcription factor that is required for the proper maturation of the erythroid cells, whose expression and activation are tightly controlled in a temporal and differentiation stage-specific manner. Here, we uncover a novel role of G-protein pathway suppressor 2 (GPS2), a subunit of the nuclear receptor corepressor/silencing mediator of retinoic acid and thyroid hormone receptor corepressor complex, in erythrocyte differentiation. Our study demonstrates that knockdown of GPS2 significantly suppresses erythroid differentiation of human CD34+ cells cultured in vitro and xenotransplanted in nonobese diabetic/severe combined immunodeficiency/interleukin-2 receptor γ-chain null mice. Moreover, global deletion of GPS2 in mice causes impaired erythropoiesis in the fetal liver and leads to severe anemia. Flow cytometric analysis and Wright-Giemsa staining show a defective differentiation at late stages of erythropoiesis in Gps2-/- embryos. Mechanistically, GPS2 interacts with EKLF and prevents proteasome-mediated degradation of EKLF, thereby increasing EKLF stability and transcriptional activity. Moreover, we identify the amino acids 191-230 region in EKLF protein, responsible for GPS2 binding, that is highly conserved in mammals and essential for EKLF protein stability. Collectively, our study uncovers a previously unknown role of GPS2 as a posttranslational regulator that enhances the stability of EKLF protein and thereby promotes erythroid differentiation.


Assuntos
Eritropoese/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Sequência Conservada , Células Precursoras Eritroides/citologia , Técnicas de Silenciamento de Genes , Transplante de Células-Tronco Hematopoéticas , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Fatores de Transcrição Kruppel-Like/química , Fígado/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Proteólise , Interferência de RNA , RNA Interferente Pequeno/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Transplante Heterólogo , Ubiquitinação , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...