Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 492
Filtrar
1.
Mol Med Rep ; 29(6)2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38695236

RESUMEN

During hematopoiesis, megakaryocytic erythroid progenitors (MEPs) differentiate into megakaryocytic or erythroid lineages in response to specific transcriptional factors, yet the regulatory mechanism remains to be elucidated. Using the MEP­like cell line HEL western blotting, RT­qPCR, lentivirus­mediated downregulation, flow cytometry as well as chromatin immunoprecipitation (ChIp) assay demonstrated that the E26 transformation­specific (ETS) transcription factor friend leukemia integration factor 1 (Fli­1) inhibits erythroid differentiation. The present study using these methods showed that while FLI1­mediated downregulation of GATA binding protein 1 (GATA1) suppresses erythropoiesis, its direct transcriptional induction of GATA2 promotes megakaryocytic differentiation. GATA1 is also involved in megakaryocytic differentiation through regulation of GATA2. By contrast to FLI1, the ETS member erythroblast transformation­specific­related gene (ERG) negatively controls GATA2 and its overexpression through exogenous transfection blocks megakaryocytic differentiation. In addition, FLI1 regulates expression of LIM Domain Binding 1 (LDB1) during erythroid and megakaryocytic commitment, whereas shRNA­mediated depletion of LDB1 downregulates FLI1 and GATA2 but increases GATA1 expression. In agreement, LDB1 ablation using shRNA lentivirus expression blocks megakaryocytic differentiation and modestly suppresses erythroid maturation. These results suggested that a certain threshold level of LDB1 expression enables FLI1 to block erythroid differentiation. Overall, FLI1 controlled the commitment of MEP to either erythroid or megakaryocytic lineage through an intricate regulation of GATA1/GATA2, LDB1 and ERG, exposing multiple targets for cell fate commitment and therapeutic intervention.


Asunto(s)
Diferenciación Celular , Células Eritroides , Megacariocitos , Humanos , Diferenciación Celular/genética , Línea Celular , Células Eritroides/metabolismo , Células Eritroides/citología , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA2/metabolismo , Factor de Transcripción GATA2/genética , Regulación de la Expresión Génica , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Megacariocitos/metabolismo , Megacariocitos/citología , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Regulador Transcripcional ERG/metabolismo , Regulador Transcripcional ERG/genética
2.
J Cell Mol Med ; 28(9): e18308, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38683131

RESUMEN

Destruction of erythropoiesis process leads to various diseases, including thrombocytopenia, anaemia, and leukaemia. miR-429-CT10 regulation of kinase-like (CRKL) axis involved in development, progression and metastasis of cancers. However, the exact role of miR-429-CRKL axis in leukaemic cell differentiation are still unknown. The current work aimed to uncover the effect of miR-429-CRKL axis on erythropoiesis. In the present study, CRKL upregulation was negatively correlated with miR-429 downregulation in both chronic myeloid leukaemia (CML) patient and CR patient samples. Moreover, CRKL expression level was significantly decreased while miR-429 expression level was increased during the erythroid differentiation of K562 cells following hemin treatment. Functional investigations revealed that overexpression and knockdown of CRKL was remarkably effective in suppressing and promoting hemin-induced erythroid differentiation of K562 cells, whereas, miR-429 exhibited opposite effects to CRKL. Mechanistically, miR-429 regulates erythroid differentiation of K562 cells by downregulating CRKL via selectively targeting CRKL-3'-untranslated region (UTR) through Raf/MEK/ERK pathway. Conversely, CRKII had no effect on erythroid differentiation of K562 cells. Taken together, our data demonstrated that CRKL (but not CRKII) and miR-429 contribute to development, progression and erythropoiesis of CML, miR-429-CRKL axis regulates erythropoiesis of K562 cells via Raf/MEK/ERK pathway, providing novel insights into effective diagnosis and therapy for CML patients.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Diferenciación Celular , Células Eritroides , Hemina , Leucemia Mielógena Crónica BCR-ABL Positiva , MicroARNs , Proteínas Proto-Oncogénicas c-crk , Humanos , Regiones no Traducidas 3' , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/efectos de los fármacos , Células Eritroides/metabolismo , Células Eritroides/efectos de los fármacos , Células Eritroides/patología , Células Eritroides/citología , Eritropoyesis/genética , Eritropoyesis/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Hemina/farmacología , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Proteínas Proto-Oncogénicas c-crk/genética
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(6): 494-500, 2022 Jun.
Artículo en Chino | MEDLINE | ID: mdl-35732606

RESUMEN

Objective To investigate the role of endothelial PAS domain-containing protein 1 (EPAS1) gene encoded hypoxia-inducible factor 2α (HIF-2α) in erythroid differentiation of K562 human erythroleukemia cells under hypoxic conditions. Methods K562 cells were treated with 40 µmol/L of hemin and 0.1 ng/mL of cytarabine for erythroid differentiation. After normoxic and hypoxic(50 mL/L O2) incubation, the ratio of CD235a+CD71+ cells was detected by flow cytometry. The percentage of hemoglobin-positive cells was detected by benzidine staining. The level of cell proliferation was detected by CCK-8 assay. The mRNA and protein levels of EPAS1, insulin receptor substrate 2 (IRS2) and γ-globin were detected by real-time quantitative PCR and Wester blot analysis. Besides, the changes in the erythroid differentiation of K562 cells were evaluated after knockdown of EPAS1. Results Hypoxia promoted the erythroid differentiation of K562 cells and upregulated the expression of EPAS1. After EPAS1 kncokdown, the ratio of CD235a+CD71+ cells and hemoglobin-positive cells decreased, and the expressions of IRS2 and γ-globin declined significantly. Conclusion Hypoxia can significantly up-regulate the expression of EPAS1 in K562 cells and promote the erythroid differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Hipoxia de la Célula , Células Eritroides , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Eritroides/citología , Hemoglobinas , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Células K562 , gamma-Globinas/genética
4.
Blood ; 139(3): 439-451, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-34614145

RESUMEN

The hormone erythroferrone (ERFE) is produced by erythroid cells in response to hemorrhage, hypoxia, or other erythropoietic stimuli, and it suppresses the hepatic production of the iron-regulatory hormone hepcidin, thereby mobilizing iron for erythropoiesis. Suppression of hepcidin by ERFE is believed to be mediated by interference with paracrine bone morphogenetic protein (BMP) signaling that regulates hepcidin transcription in hepatocytes. In anemias with ineffective erythropoiesis, ERFE is pathologically overproduced, but its contribution to the clinical manifestations of these anemias is not well understood. We generated 3 lines of transgenic mice with graded erythroid overexpression of ERFE and found that they developed dose-dependent iron overload, impaired hepatic BMP signaling, and relative hepcidin deficiency. These findings add to the evidence that ERFE is a mediator of iron overload in conditions in which ERFE is overproduced, including anemias with ineffective erythropoiesis. At the highest levels of ERFE overexpression, the mice manifested decreased perinatal survival, impaired growth, small hypofunctional kidneys, decreased gonadal fat depots, and neurobehavioral abnormalities, all consistent with impaired organ-specific BMP signaling during development. Neutralizing excessive ERFE in congenital anemias with ineffective erythropoiesis may not only prevent iron overload but may have additional benefits for growth and development.


Asunto(s)
Citocinas/metabolismo , Discapacidades del Desarrollo/metabolismo , Células Eritroides/metabolismo , Sobrecarga de Hierro/metabolismo , Proteínas Musculares/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Citocinas/genética , Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/genética , Células Eritroides/citología , Femenino , Hepcidinas/metabolismo , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Musculares/genética , Transducción de Señal , Regulación hacia Arriba
5.
Leukemia ; 36(3): 847-855, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34743190

RESUMEN

Although a glycosylphosphatidylinositol-anchored protein (GPI-AP) CD109 serves as a TGF-ß co-receptor and inhibits TGF-ß signaling in keratinocytes, the role of CD109 on hematopoietic stem progenitor cells (HSPCs) remains unknown. We studied the effect of CD109 knockout (KO) or knockdown (KD) on TF-1, a myeloid leukemia cell line that expresses CD109, and primary human HSPCs. CD109-KO or KD TF-1 cells underwent erythroid differentiation in the presence of TGF-ß. CD109 was more abundantly expressed in hematopoietic stem cells (HSCs) than in multipotent progenitors and HSPCs of human bone marrow (BM) and cord blood but was not detected in mouse HSCs. Erythroid differentiation was induced by TGF-ß to a greater extent in CD109-KD cord blood or iPS cell-derived megakaryocyte-erythrocyte progenitor cells (MEPs) than in wild-type MEPs. When we analyzed the phenotype of peripheral blood MEPs of patients with paroxysmal nocturnal hemoglobinuria who had both GPI(+) and GPI(-) CD34+ cells, the CD36 expression was more evident in CD109- MEPs than CD109+ MEPs. In summary, CD109 suppresses TGF-ß signaling in HSPCs, and the lack of CD109 may increase the sensitivity of PIGA-mutated HSPCs to TGF-ß, thus leading to the preferential commitment of erythroid progenitor cells to mature red blood cells in immune-mediated BM failure.


Asunto(s)
Antígenos CD/metabolismo , Células Eritroides/citología , Células Madre Hematopoyéticas/citología , Proteínas de Neoplasias/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Células Eritroides/metabolismo , Eritropoyesis , Proteínas Ligadas a GPI/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos
6.
Nat Commun ; 12(1): 7019, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857757

RESUMEN

Yolk sac (YS) hematopoiesis is critical for the survival of the embryo and a major source of tissue-resident macrophages that persist into adulthood. Yet, the transcriptional and epigenetic regulation of YS hematopoiesis remains poorly characterized. Here we report that the epigenetic regulator Ezh2 is essential for YS hematopoiesis but dispensable for subsequent aorta-gonad-mesonephros (AGM) blood development. Loss of EZH2 activity in hemogenic endothelium (HE) leads to the generation of phenotypically intact but functionally deficient erythro-myeloid progenitors (EMPs), while the generation of primitive erythroid cells is not affected. EZH2 activity is critical for the generation of functional EMPs at the onset of the endothelial-to-hematopoietic transition but subsequently dispensable. We identify a lack of Wnt signaling downregulation as the primary reason for the production of non-functional EMPs. Together, our findings demonstrate a critical and stage-specific role of Ezh2 in modulating Wnt signaling during the generation of EMPs from YS HE.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Células Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Células Progenitoras Mieloides/metabolismo , Proteínas de Transporte Vesicular/genética , Saco Vitelino/metabolismo , Animales , Diferenciación Celular , Embrión de Mamíferos , Proteína Potenciadora del Homólogo Zeste 2/deficiencia , Epigénesis Genética , Células Eritroides/citología , Femenino , Feto , Genes Reporteros , Hematopoyesis/genética , Hígado/citología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Células Progenitoras Mieloides/patología , Cultivo Primario de Células , Proteínas de Transporte Vesicular/metabolismo , Vía de Señalización Wnt , Saco Vitelino/citología , Saco Vitelino/crecimiento & desarrollo , Proteína Fluorescente Roja
7.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768865

RESUMEN

NKL homeobox genes encode transcription factors that impact normal development and hematopoietic malignancies if deregulated. Recently, we established an NKL-code that describes the physiological expression pattern of eleven NKL homeobox genes in the course of hematopoiesis, allowing evaluation of aberrantly activated NKL genes in leukemia/lymphoma. Here, we identify ectopic expression of NKL homeobox gene NKX2-4 in an erythroblastic acute myeloid leukemia (AML) cell line OCI-M2 and describe investigation of its activating factors and target genes. Comparative expression profiling data of AML cell lines revealed in OCI-M2 an aberrantly activated program for endothelial development including master factor ETV2 and the additional endothelial signature genes HEY1, IRF6, and SOX7. Corresponding siRNA-mediated knockdown experiments showed their role in activating NKX2-4 expression. Furthermore, the ETV2 locus at 19p13 was genomically amplified, possibly underlying its aberrant expression. Target gene analyses of NKX2-4 revealed activated ETV2, HEY1, and SIX5 and suppressed FLI1. Comparative expression profiling analysis of public datasets for AML patients and primary megakaryocyte-erythroid progenitor cells showed conspicuous similarities to NKX2-4 activating factors and the target genes we identified, supporting the clinical relevance of our findings and developmental disturbance by NKX2-4. Finally, identification and target gene analysis of aberrantly expressed NKX2-3 in AML patients and a megakaryoblastic AML cell line ELF-153 showed activation of FLI1, contrasting with OCI-M2. FLI1 encodes a master factor for myelopoiesis, driving megakaryocytic differentiation and suppressing erythroid differentiation, thus representing a basic developmental target of these homeo-oncogenes. Taken together, we have identified aberrantly activated NKL homeobox genes NKX2-3 and NKX2-4 in AML, deregulating genes involved in megakaryocytic and erythroid differentiation processes, and thereby contributing to the formation of specific AML subtypes.


Asunto(s)
Células Eritroides/citología , Proteínas de Homeodominio/genética , Leucemia Eritroblástica Aguda/genética , Megacariocitos/citología , Factores de Transcripción/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Endotelio/citología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Humanos , Factores Reguladores del Interferón/genética , Leucemia Eritroblástica Aguda/patología , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Transcripción SOXF/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
8.
Cells ; 10(11)2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34831239

RESUMEN

MicroRNAs (miRNAs) are small non-coding RNAs, which play an important role in various cellular and developmental processes. The study of miRNAs in erythropoiesis is crucial to uncover the cellular pathways that are modulated during the different stages of erythroid differentiation. Using erythroid cells derived from human CD34+ hematopoietic stem and progenitor cells (HSPCs)and small RNA sequencing, our study unravels the various miRNAs involved in critical cellular pathways in erythroid maturation. We analyzed the occupancy of erythroid transcription factors and chromatin accessibility in the promoter and enhancer regions of the differentially expressed miRNAs to integrate miRNAs in the transcriptional circuitry of erythropoiesis. Analysis of the targets of the differentially expressed miRNAs revealed novel pathways in erythroid differentiation. Finally, we described the application of Clustered regularly interspaced short palindromic repeats-Cas9 (CRISPR-Cas9) based editing of miRNAs to study their function in human erythropoiesis.


Asunto(s)
Eritropoyesis/genética , MicroARNs/genética , Adulto , Sistemas CRISPR-Cas/genética , Diferenciación Celular/genética , Línea Celular , Cromatina/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Edición Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , MicroARNs/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
9.
Nature ; 598(7880): 327-331, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34588693

RESUMEN

Haematopoiesis in the bone marrow (BM) maintains blood and immune cell production throughout postnatal life. Haematopoiesis first emerges in human BM at 11-12 weeks after conception1,2, yet almost nothing is known about how fetal BM (FBM) evolves to meet the highly specialized needs of the fetus and newborn. Here we detail the development of FBM, including stroma, using multi-omic assessment of mRNA and multiplexed protein epitope expression. We find that the full blood and immune cell repertoire is established in FBM in a short time window of 6-7 weeks early in the second trimester. FBM promotes rapid and extensive diversification of myeloid cells, with granulocytes, eosinophils and dendritic cell subsets emerging for the first time. The substantial expansion of B lymphocytes in FBM contrasts with fetal liver at the same gestational age. Haematopoietic progenitors from fetal liver, FBM and cord blood exhibit transcriptional and functional differences that contribute to tissue-specific identity and cellular diversification. Endothelial cell types form distinct vascular structures that we show are regionally compartmentalized within FBM. Finally, we reveal selective disruption of B lymphocyte, erythroid and myeloid development owing to a cell-intrinsic differentiation bias as well as extrinsic regulation through an altered microenvironment in Down syndrome (trisomy 21).


Asunto(s)
Células de la Médula Ósea/citología , Médula Ósea , Síndrome de Down/sangre , Síndrome de Down/inmunología , Feto/citología , Hematopoyesis , Sistema Inmunológico/citología , Linfocitos B/citología , Células Dendríticas/citología , Síndrome de Down/metabolismo , Síndrome de Down/patología , Células Endoteliales/patología , Eosinófilos/citología , Células Eritroides/citología , Granulocitos/citología , Humanos , Inmunidad , Células Mieloides/citología , Células del Estroma/citología
10.
Physiol Res ; 70(5): 701-707, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-34505521

RESUMEN

The present nuclear and cell body diameter measurements demonstrated size differences of the approximate cell space estimate occupied by the cell nucleus during the cell differentiation in lymphocytic, granulocytic and erythroid cell lineages. These lineages were used as convenient models because all differentiation steps were easily identified and accessible in diagnostic peripheral blood or bone marrow smears of blood donors (BDs), patients suffering from chronic lymphocytic leukemia (CLL), patients with chronic myeloid leukemia (CML) and refractory anemia (RA) of the myelodysplastic syndrome (MDS). The cell space occupied by the nucleus was constant and did not change during the cell differentiation in the lymphocytic cell lineages of BDs and CLL patients despite the decreased cell size. In contrary, the cell space occupied by the nucleus markedly decreased in differentiating cells of granulocytic and erythroid lineages of patients suffering from CML. In the erythroid cell lineage in patients with RA of MDS the small reduction of the cell space occupied by the nucleus during the differentiation was not significant. The measurements also indicated that in progenitor cells of all studied cell lineages nuclei occupied more than 70 % of the cell space. Thus, the nucleus-cytoplasmic morphological and functional equilibrium appeared to be characteristic for each differentiation step and each specific cell lineage.


Asunto(s)
Diferenciación Celular , Núcleo Celular , Células Eritroides/citología , Granulocitos/citología , Linfocitos/citología , Anemia Refractaria/patología , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología
11.
FASEB J ; 35(10): e21915, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34496088

RESUMEN

During development, erythroid cells are generated by two waves of hematopoiesis. In zebrafish, primitive erythropoiesis takes place in the intermediate cell mass region, and definitive erythropoiesis arises from the aorta-gonad mesonephros. TALE-homeoproteins Meis1 and Pbx1 function upstream of GATA1 to specify the erythroid lineage. Embryos lacking Meis1 or Pbx1 have weak gata1 expression and fail to produce primitive erythrocytes. Nevertheless, the underlying mechanism of how Meis1 and Pbx1 mediate gata1 transcription in erythrocytes remains unclear. Here we show that Hif1α acts downstream of Meis1 to mediate gata1 expression in zebrafish embryos. Inhibition of Meis1 expression resulted in suppression of hif1a expression and abrogated primitive erythropoiesis, while injection with in vitro-synthesized hif1α mRNA rescued gata1 transcription in Meis1 morphants and recovered their erythropoiesis. Ablation of Hif1α expression either by morpholino knockdown or Crispr-Cas9 knockout suppressed gata1 transcription and abrogated primitive erythropoiesis. Results of chromatin immunoprecipitation assays showed that Hif1α associates with hypoxia-response elements located in the 3'-flanking region of gata1 during development, suggesting that Hif1α regulates gata1 expression in vivo. Together, our results indicate that Meis1, Hif1α, and GATA1 indeed comprise a hierarchical regulatory network in which Hif1α acts downstream of Meis1 to activate gata1 transcription through direct interactions with its cis-acting elements in primitive erythrocytes.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyesis , Factor de Transcripción GATA1/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Inmunoprecipitación de Cromatina , Eritrocitos/citología , Eritrocitos/metabolismo , Células Eritroides/citología , Eritropoyesis/genética , Factor de Transcripción GATA1/genética , Regulación del Desarrollo de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/deficiencia , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B/deficiencia , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Transcripción Genética , Pez Cebra/sangre , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
12.
Blood Cells Mol Dis ; 91: 102594, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34520986

RESUMEN

Cytokines/chemokines regulate hematopoiesis, most having multiple cell actions. Numerous but not all chemokine family members act as negative regulators of hematopoietic progenitor cell (HPC) proliferation, but very little is known about such effects of the chemokine, CXCL15/Lungkine. We found that CXCL15/Lungkine-/- mice have greatly increased cycling of multi cytokine-stimulated bone marrow and spleen hematopoietic progenitor cells (HPCs: CFU-GM, BFU-E, and CFU-GEMM) and CXCL15 is expressed in many bone marrow progenitor and other cell types. This suggests that CXCL15/Lungkine acts as a negative regulator of the cell cycling of these HPCs in vivo. Recombinant murine CXCL15/Lungkine, decreased numbers of functional HPCs during cytokine-enhanced ex-vivo culture of lineage negative mouse bone marrow cells. Moreover, CXCL15/Lungkine, through S-Phase specific actions, was able to suppress in vitro colony formation of normal wildtype mouse bone marrow CFU-GM, CFU-G, CFU-M, BFU-E, and CFU-GEMM. This clearly identifies the negative regulatory activity of CXCL15/Lungkine on proliferation of multiple types of mouse HPCs.


Asunto(s)
Quimiocinas CXC/metabolismo , Células Eritroides/citología , Granulocitos/citología , Macrófagos/citología , Células Madre/citología , Animales , Proliferación Celular , Células Cultivadas , Células Eritroides/metabolismo , Granulocitos/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Puntos de Control de la Fase S del Ciclo Celular , Células Madre/metabolismo
13.
J Biol Chem ; 297(3): 101051, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34364872

RESUMEN

The asymmetric cell division of stem or progenitor cells generates daughter cells with distinct fates that balance proliferation and differentiation. Asymmetric segregation of Notch signaling regulatory protein Numb plays a crucial role in cell diversification. However, the molecular mechanism remains unclear. Here, we examined the unequal distribution of Numb in the daughter cells of murine erythroleukemia cells (MELCs) that undergo DMSO-induced erythroid differentiation. In contrast to the cytoplasmic localization of Numb during uninduced cell division, Numb is concentrated at the cell boundary in interphase, near the one-spindle pole in metaphase, and is unequally distributed to one daughter cell in anaphase in induced cells. The inheritance of Numb guides this daughter cell toward erythroid differentiation while the other cell remains a progenitor cell. Mitotic spindle orientation, critical for distribution of cell fate determinants, requires complex communication between the spindle microtubules and the cell cortex mediated by the NuMA-LGN-dynein/dynactin complex. Depletion of each individual member of the complex randomizes the position of Numb relative to the mitotic spindle. Gene replacement confirms that multifunctional erythrocyte protein 4.1R (4.1R) functions as a member of the NuMA-LGN-dynein/dynactin complex and is necessary for regulating spindle orientation, in which interaction between 4.1R and NuMA plays an important role. These results suggest that mispositioning of Numb is the result of spindle misorientation. Finally, disruption of the 4.1R-NuMA-LGN complex increases Notch signaling and decreases the erythroblast population. Together, our results identify a critical role for 4.1R in regulating the asymmetric segregation of Numb to mediate erythropoiesis.


Asunto(s)
División Celular Asimétrica , Células Eritroides/citología , Células Eritroides/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Complejo Dinactina/genética , Complejo Dinactina/metabolismo , Dineínas/genética , Dineínas/metabolismo , Proteínas de la Membrana/genética , Ratones , Proteínas de Microfilamentos/genética , Mitosis , Proteínas del Tejido Nervioso/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Huso Acromático/genética , Huso Acromático/metabolismo
14.
Nucleic Acids Res ; 49(17): 9783-9798, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34450641

RESUMEN

The activity of hematopoietic factor GATA-1 is modulated through p300/CBP-mediated acetylation and FOG-1 mediated indirect interaction with HDAC1/2 containing NuRD complex. Although GATA-1 acetylation is implicated in GATA-1 activation, the role of deacetylation is not studied. Here, we found that the FOG-1/NuRD does not deacetylate GATA-1. However, HDAC1/2 can directly bind and deacetylate GATA-1. Two arginine residues within the GATA-1 linker region mediates direct interaction with HDAC1. The arginine to alanine mutation (2RA) blocks GATA-1 deacetylation and fails to induce erythroid differentiation. Gene expression profiling and ChIP-seq analysis further demonstrate the importance of GATA-1 deacetylation for gene activation and chromatin recruitment. GATA-12RA knock-in (KI) mice suffer mild anemia and thrombocytopenia with accumulation of immature erythrocytes and megakaryocytes in bone marrow and spleen. Single cell RNA-seq analysis of Lin- cKit+ (LK) cells further reveal a profound change in cell subpopulations and signature gene expression patterns in HSC, myeloid progenitors, and erythroid/megakaryocyte clusters in KI mice. Thus, GATA-1 deacetylation and its interaction with HDAC1 modulates GATA-1 chromatin binding and transcriptional activity that control erythroid/megakaryocyte commitment and differentiation.


Asunto(s)
Cromatina/metabolismo , Factor de Transcripción GATA1/metabolismo , Hematopoyesis/genética , Histona Desacetilasa 1/metabolismo , Transcripción Genética , Anemia/genética , Animales , Sitios de Unión , Células Eritroides/citología , Células Eritroides/metabolismo , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/fisiología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Histona Desacetilasa 1/fisiología , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Trombocitopenia/genética
15.
Sci Rep ; 11(1): 17129, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34429458

RESUMEN

Production of red blood cells relies on proper mitochondrial function, both for their increased energy demands during differentiation and for proper heme and iron homeostasis. Mutations in genes regulating mitochondrial function have been reported in patients with anemia, yet their pathophysiological role often remains unclear. PGC1ß is a critical coactivator of mitochondrial biogenesis, with increased expression during terminal erythroid differentiation. The role of PGC1ß has however mainly been studied in skeletal muscle, adipose and hepatic tissues, and its function in erythropoiesis remains largely unknown. Here we show that perturbed PGC1ß expression in human hematopoietic stem/progenitor cells from both bone marrow and cord blood results in impaired formation of early erythroid progenitors and delayed terminal erythroid differentiation in vitro, with accumulations of polychromatic erythroblasts, similar to MDS-related refractory anemia. Reduced levels of PGC1ß resulted in deregulated expression of iron, heme and globin related genes in polychromatic erythroblasts, and reduced hemoglobin content in the more mature bone marrow derived reticulocytes. Furthermore, PGC1ß knock-down resulted in disturbed cell cycle exit with accumulation of erythroblasts in S-phase and enhanced expression of G1-S regulating genes, with smaller reticulocytes as a result. Taken together, we demonstrate that PGC1ß is directly involved in production of hemoglobin and regulation of G1-S transition and is ultimately required for proper terminal erythroid differentiation.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyesis , Proteínas de Unión al ARN/metabolismo , Ciclo Celular , Células Cultivadas , Células Eritroides/citología , Hemoglobinas/metabolismo , Humanos , Proteínas de Unión al ARN/genética
16.
Blood ; 138(18): 1691-1704, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34324630

RESUMEN

Histone H3 lysine 4 methylation (H3K4Me) is most often associated with chromatin activation, and removing H3K4 methyl groups has been shown to be coincident with gene repression. H3K4Me demethylase KDM1a/LSD1 is a therapeutic target for multiple diseases, including for the potential treatment of ß-globinopathies (sickle cell disease and ß-thalassemia), because it is a component of γ-globin repressor complexes, and LSD1 inactivation leads to robust induction of the fetal globin genes. The effects of LSD1 inhibition in definitive erythropoiesis are not well characterized, so we examined the consequences of conditional inactivation of Lsd1 in adult red blood cells using a new Gata1creERT2 bacterial artificial chromosome transgene. Erythroid-specific loss of Lsd1 activity in mice led to a block in erythroid progenitor differentiation and to the expansion of granulocyte-monocyte progenitor-like cells, converting hematopoietic differentiation potential from an erythroid fate to a myeloid fate. The analogous phenotype was also observed in human hematopoietic stem and progenitor cells, coincident with the induction of myeloid transcription factors (eg, PU.1 and CEBPα). Finally, blocking the activity of the transcription factor PU.1 or RUNX1 at the same time as LSD1 inhibition rescued myeloid lineage conversion to an erythroid phenotype. These data show that LSD1 promotes erythropoiesis by repressing myeloid cell fate in adult erythroid progenitors and that inhibition of the myeloid-differentiation pathway reverses the lineage switch induced by LSD1 inactivation.


Asunto(s)
Células Eritroides/citología , Eritropoyesis , Histona Demetilasas/metabolismo , Células Mieloides/citología , Animales , Línea Celular , Células Cultivadas , Células Eritroides/metabolismo , Eliminación de Gen , Histona Demetilasas/genética , Humanos , Ratones , Células Mieloides/metabolismo
17.
Genome Biol ; 22(1): 197, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34225769

RESUMEN

BACKGROUND: Single-cell technologies are transforming biomedical research, including the recent demonstration that unspliced pre-mRNA present in single-cell RNA-Seq permits prediction of future expression states. Here we apply this RNA velocity concept to an extended timecourse dataset covering mouse gastrulation and early organogenesis. RESULTS: Intriguingly, RNA velocity correctly identifies epiblast cells as the starting point, but several trajectory predictions at later stages are inconsistent with both real-time ordering and existing knowledge. The most striking discrepancy concerns red blood cell maturation, with velocity-inferred trajectories opposing the true differentiation path. Investigating the underlying causes reveals a group of genes with a coordinated step-change in transcription, thus violating the assumptions behind current velocity analysis suites, which do not accommodate time-dependent changes in expression dynamics. Using scRNA-Seq analysis of chimeric mouse embryos lacking the major erythroid regulator Gata1, we show that genes with the step-changes in expression dynamics during erythroid differentiation fail to be upregulated in the mutant cells, thus underscoring the coordination of modulating transcription rate along a differentiation trajectory. In addition to the expected block in erythroid maturation, the Gata1-chimera dataset reveals induction of PU.1 and expansion of megakaryocyte progenitors. Finally, we show that erythropoiesis in human fetal liver is similarly characterized by a coordinated step-change in gene expression. CONCLUSIONS: By identifying a limitation of the current velocity framework coupled with in vivo analysis of mutant cells, we reveal a coordinated step-change in gene expression kinetics during erythropoiesis, with likely implications for many other differentiation processes.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyesis/genética , Factor de Transcripción GATA1/genética , Regulación del Desarrollo de la Expresión Génica , Organogénesis/genética , Animales , Diferenciación Celular , Conjuntos de Datos como Asunto , Embrión de Mamíferos , Células Eritroides/citología , Feto , Factor de Transcripción GATA1/deficiencia , Gástrula/crecimiento & desarrollo , Gástrula/metabolismo , Humanos , Cinética , Hígado/citología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Análisis de la Célula Individual , Transactivadores/genética , Transactivadores/metabolismo , Activación Transcripcional
18.
Mol Cell Biol ; 41(9): e0066820, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34180713

RESUMEN

We previously demonstrated that the two paralogous RNA-binding proteins PCBP1 and PCBP2 are individually essential for mouse development: Pcbp1-null embryos are peri-implantation lethal, while Pcbp2-null embryos lose viability at midgestation. Midgestation Pcbp2-/- embryos revealed a complex phenotype that included loss of certain hematopoietic determinants. Whether PCBP2 directly contributes to erythropoietic differentiation and whether PCBP1 has a role in this process remained undetermined. Here, we selectively inactivated the genes encoding these two RNA-binding proteins during differentiation of the erythroid lineage in the developing mouse embryo. Individual inactivation of either locus failed to impact viability or blood formation. However, combined inactivation of the two loci resulted in midgestational repression of erythroid/hematopoietic gene expression, loss of blood formation, and fetal demise. Orthogonal ex vivo analyses of primary erythroid progenitors selectively depleted of these two RNA-binding proteins revealed that they mediate a combination of overlapping and isoform-specific impacts on hematopoietic lineage transcriptome, impacting both mRNA representation and exon splicing. These data lead us to conclude that PCBP1 and PCBP2 mediate functions critical to differentiation of the erythroid lineage.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Eritropoyesis , Proteínas de Unión al ARN/metabolismo , Envejecimiento/fisiología , Animales , Linaje de la Célula , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/metabolismo , Células Eritroides/citología , Exones/genética , Sitios Genéticos , Células Madre Hematopoyéticas/metabolismo , Ratones , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Transcriptoma/genética
19.
Mol Med Rep ; 24(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34109429

RESUMEN

GATA binding protein 1 (GATA­1) is one of the most important hematopoietic transcription factors in the production of blood cells, such as platelets, eosinophils, mast cells and erythrocytes. GATA­1 regulates the participation of microRNA (miRNAs/miRs) in erythroid differentiation under normoxia. However, GATA­1 expression and the regulation of miR­210­3p in the context of erythroid differentiation under hypoxia remain unknown. The present study examined the expression levels of GATA­1 and miR­210­3p in the model of erythroid differentiation in K562 cells under hypoxia, and determined the effects of GATA­1, miR­210­3p and SMAD2 on erythroid differentiation through lentivirus transfection experiments. The present study detected increased GATA­1 expression under hypoxia. Moreover, miR­210­3p was identified as a positive regulator of erythroid differentiation, which was upregulated both during erythroid differentiation and in GATA­1 overexpression experiments under hypoxia. Importantly, in the K562 cell model of erythroid differentiation under hypoxia, miR­210­3p was upregulated in a GATA­1­dependent manner. Using a double luciferase reporter assay, miR­210­3p was identified as a downstream target of GATA­1­mediated regulation of erythropoiesis. Gain­ or loss­of­function analysis of miR­210­3p identified its importance in erythroid differentiation. Furthermore, it was found that SMAD2 may be a downstream target gene for miR­210­3p. Bioinformatics predictions suggested that SMAD2 mediated miR­210­3p­induced regulation of erythroid differentiation. Collectively, the present study provides novel insights into the miRNA regulation of erythroid differentiation.


Asunto(s)
Hipoxia de la Célula/genética , Células Eritroides/metabolismo , MicroARNs/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Eritroides/citología , Células Eritroides/efectos de los fármacos , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Técnicas de Silenciamiento del Gen , Hemina/farmacología , Humanos , Células K562 , MicroARNs/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Regulación hacia Arriba/genética
20.
Blood ; 138(18): 1740-1756, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34075391

RESUMEN

The terminal maturation of human erythroblasts requires significant changes in gene expression in the context of dramatic nuclear condensation. Defects in this process are associated with inherited anemias and myelodysplastic syndromes. The progressively dense appearance of the condensing nucleus in maturing erythroblasts led to the assumption that heterochromatin accumulation underlies this process, but despite extensive study, the precise mechanisms underlying this essential biologic process remain elusive. To delineate the epigenetic changes associated with the terminal maturation of human erythroblasts, we performed mass spectrometry of histone posttranslational modifications combined with chromatin immunoprecipitation coupled with high-throughput sequencing, Assay for Transposase Accessible Chromatin, and RNA sequencing. Our studies revealed that the terminal maturation of human erythroblasts is associated with a dramatic decline in histone marks associated with active transcription elongation, without accumulation of heterochromatin. Chromatin structure and gene expression were instead correlated with dynamic changes in occupancy of elongation competent RNA polymerase II, suggesting that terminal erythroid maturation is controlled largely at the level of transcription. We further demonstrate that RNA polymerase II "pausing" is highly correlated with transcriptional repression, with elongation competent RNA polymerase II becoming a scare resource in late-stage erythroblasts, allocated to erythroid-specific genes. Functional studies confirmed an essential role for maturation stage-specific regulation of RNA polymerase II activity during erythroid maturation and demonstrate a critical role for HEXIM1 in the regulation of gene expression and RNA polymerase II activity in maturing erythroblasts. Taken together, our findings reveal important insights into the mechanisms that regulate terminal erythroid maturation and provide a novel paradigm for understanding normal and perturbed erythropoiesis.


Asunto(s)
Eritroblastos/metabolismo , Células Eritroides/metabolismo , ARN Polimerasa II/metabolismo , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Eritroblastos/citología , Células Eritroides/citología , Eritropoyesis , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , ARN Polimerasa II/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA