Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
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
2.
Thromb J ; 22(1): 74, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39123189

RESUMEN

BACKGROUND: Proper control of the lineage bias of megakaryocytic and erythroid progenitor cells (MEPs) is of significant importance, the disorder of which will lead to abnormalities in the number and function of platelets and erythrocytes. Unfortunately, the signaling pathways regulating MEP differentiation largely remain to be elucidated. This study aimed to analyze the role and the underlying molecular mechanism of miR-1915-3p in megakaryocytic and erythroid differentiation. METHODS: We utilized miRNA mimics and miRNA sponge to alter the expression of miR-1915-3p in megakaryocytic and/or erythroid potential cells; siRNA and overexpression plasmid to change the expression of SOCS4, a potential target of miR-1915-3p. The expression of relevant surface markers was detected by flow cytometry. We scanned for miR-1915-3p target genes by mRNA expression profiling and bioinformatic analysis, and confirmed the targeting by dual-luciferase reporter assay, western blot and gain- and lost-of-function studies. One-way ANOVA and t-test were used to analyze the statistical significance. RESULTS: In this study, overexpression or knockdown of miR-1915-3p inhibited or promoted erythroid differentiation, respectively. Accordingly, we scanned for miR-1915-3p target genes and confirmed that SOCS4 is one of the direct targets of miR-1915-3p. An attentive examination of the endogenous expression of SOCS4 during megakaryocytic and erythroid differentiation suggested the involvement of SOCS4 in erythroid/megakaryocytic lineage determination. SOCS4 knockdown lessened erythroid surface markers expression, as well as improved megakaryocytic differentiation, similar to the effects of miR-1915-3p overexpression. While SOCS4 overexpression resulted in reversed effects. SOCS4 overexpression in miR-1915-3p upregulated cells rescued the effect of miR-1915-3p. CONCLUSIONS: miR-1915-3p acts as a negative regulator of erythropoiesis, and positively in thrombopoiesis. SOCS4 is one of the key mediators of miR-1915-3p during the differentiation of MEPs.

3.
Hemoglobin ; 48(1): 4-14, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38419555

RESUMEN

Long noncoding RNAs (lncRNAs) are important because they are involved in a variety of life activities and have many downstream targets. Moreover, there is also increasing evidence that some lncRNAs play important roles in the expression and regulation of γ-globin genes. In our previous study, we analyzed genetic material from nucleated red blood cells (NRBCs) extracted from premature and full-term umbilical cord blood samples. Through RNA sequencing (RNA-Seq) analysis, lncRNA H19 emerged as a differentially expressed transcript between the two blood types. While this discovery provided insight into H19, previous studies had not investigated its effect on the γ-globin gene. Therefore, the focus of our study was to explore the impact of H19 on the γ-globin gene. In this study, we discovered that overexpressing H19 led to a decrease in HBG mRNA levels during erythroid differentiation in K562 cells. Conversely, in CD34+ hematopoietic stem cells and human umbilical cord blood-derived erythroid progenitor (HUDEP-2) cells, HBG expression increased. Additionally, we observed that H19 was primarily located in the nucleus of K562 cells, while in HUDEP-2 cells, H19 was present predominantly in the cytoplasm. These findings suggest a significant upregulation of HBG due to H19 overexpression. Notably, cytoplasmic localization in HUDEP-2 cells hints at its potential role as a competing endogenous RNA (ceRNA), regulating γ-globin expression by targeting microRNA/mRNA interactions.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , gamma-Globinas/genética , gamma-Globinas/metabolismo , Regulación hacia Arriba , ARN Mensajero/genética , Expresión Génica
4.
Rinsho Ketsueki ; 65(3): 183-187, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-38569864

RESUMEN

The transcription factor GATA-1 is essential for erythroid differentiation. Recently, FAM210B, which encodes a mitochondrial inner membrane protein, has been identified as a novel target of GATA-1. To clarify the role of FAM210B, we depleted endogenous FAM210B in human iPS-derived erythroid progenitor (HiDEP-1) cells, and found that erythroid differentiation was more pronounced in the FAM210B depleted cells. Comprehensive metabolite analysis revealed a decline in mitochondrial function accompanied by increased lactate production, indicative of anaerobic glycolysis. Mass spectrometry revealed that FAM210B could interact with multiple subunits of mitochondrial ATP synthases, such as subunit alpha (ATP5A) and beta (ATP5B). Our results suggested that FAM210B contributes prominently to erythroid differentiation by regulating mitochondrial energy metabolism. This review will discuss the potential association between mitochondrial metabolism and erythropoiesis.


Asunto(s)
Factor de Transcripción GATA1 , Mitocondrias , Humanos , Mitocondrias/metabolismo , Células Precursoras Eritroides/metabolismo , Diferenciación Celular/fisiología , Eritropoyesis/fisiología
5.
Molecules ; 29(1)2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38202591

RESUMEN

Induction of fetal hemoglobin (HbF) is highly beneficial for patients carrying ß-thalassemia, and novel HbF inducers are highly needed. Here, we describe a new class of promising HbF inducers characterized by an isoxazole chemical skeleton and obtained through modification of two natural molecules, geldanamycin and radicicol. After preliminary biological assays based on benzidine staining and RT-qPCR conducted on human erythroleukemic K562 cells, we employed erythroid precursors cells (ErPCs) isolated from ß-thalassemic patients. ErPCs weretreated with appropriate concentrations of isoxazole derivatives. The accumulation of globin mRNAs was studied by RT-qPCR, and hemoglobin production by HPLC. We demonstrated the high efficacy of isozaxoles in inducing HbF. Most of these derivatives displayed an activity similar to that observed using known HbF inducers, such as hydroxyurea (HU) or rapamycin; some of the analyzed compounds were able to induce HbF with more efficiency than HU. All the compounds were active in reducing the excess of free α-globin in treated ErPCs. All the compounds displayed a lack of genotoxicity. These novel isoxazoles deserve further pre-clinical study aimed at verifying whether they are suitable for the development of therapeutic protocols for ß-thalassemia.


Asunto(s)
Hemoglobina Fetal , Talasemia beta , Humanos , Hemoglobina Fetal/genética , Células Precursoras Eritroides , Talasemia beta/tratamiento farmacológico , Bioensayo , Hidroxiurea/farmacología , Isoxazoles
6.
Toxicology ; 505: 153843, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38801936

RESUMEN

Benzene, a widely used industrial chemical, has been clarified to cause hematotoxicity. Our previous study suggested that miR-451a may play a role in benzene-induced impairment of erythroid differentiation. However, the mechanism underlying remains unclear. In this study, we explored the role of miR-451a and its underlying mechanisms in hydroquinone (HQ)-induced suppression of erythroid differentiation in K562 cells. 0, 1.0, 2.5, 5.0, 10.0, and 50 µM HQ treatment of K562 cells resulted in a dose-dependent inhibition of erythroid differentiation, as well as the expression of miR-451a. Bioinformatics analysis was conducted to predict potential target genes of miR-451a and dual-luciferase reporter assays confirmed that miR-451a can directly bind to the 3'-UTR regions of BATF, SETD5, and ARHGEF3 mRNAs. We further demonstrated that over-expression or down-regulation of miR-451a altered the expression of BATF, SETD5, and ARHGEF3, and also modified erythroid differentiation. In addition, BATF, SETD5, and ARHGEF3 were verified to play a role in HQ-induced inhibition of erythroid differentiation in this study. Knockdown of SETD5 and ARHGEF3 reversed HQ-induced suppression of erythroid differentiation while knockdown of BATF had the opposite effect. On the other hand, we also identified c-Jun as a potential transcriptional regulator of miR-451a. Forced expression of c-Jun increased miR-451a expression and reversed the inhibition of erythroid differentiation induced by HQ, whereas knockdown of c-Jun had the opposite effect. And the binding site of c-Jun and miR-451a was verified by dual-luciferase reporter assay. Collectively, our findings indicate that miR-451a and its downstream targets BATF, SETD5, and ARHGEF3 are involved in HQ-induced erythroid differentiation disorder, and c-Jun regulates miR-451a as a transcriptional regulator in this process.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Diferenciación Celular , MicroARNs , Factores de Intercambio de Guanina Nucleótido Rho , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Diferenciación Celular/efectos de los fármacos , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células K562 , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Metiltransferasas/genética , Metiltransferasas/metabolismo
7.
Artículo en Inglés | MEDLINE | ID: mdl-38573002

RESUMEN

Aims: Erythropoiesis is controlled by several factors, including oxygen level under different circumstances. However, the role of hypoxia in erythroid differentiation and the underlying mechanisms are poorly understood. We studied the effect and mechanism of hypoxia on erythroid differentiation of K562 cells and observed the effect of hypoxia on early erythropoiesis of zebrafish. Results: Compared with normal oxygen culture, both hemin-induced erythroid differentiation of K562 cells and the early erythropoiesis of zebrafish were inhibited under hypoxic treatment conditions. Hypoxia-inducible factor 1 alpha (HIF1α) plays a major role in the response to hypoxia. Here, we obtained a stable HIF1α knockout K562 cell line using the CRISPR-Cas9 technology and further demonstrated that HIF1α knockout promoted hemin-induced erythroid differentiation of K562 cells under hypoxia. We demonstrated an HIF1-mediated induction of the nuclear factor interleukin-3 (NFIL3) regulated in K562 cells under hypoxia. Interestingly, a gradual decrease in NFIL3 expression was detected during erythroid differentiation of erythropoietin-induced CD34+ hematopoietic stem/progenitor cells (HSPCs) and hemin-induced K562 cells. Notably, erythroid differentiation was inhibited by enforced expression of NFIL3 under normoxia and was promoted by the knockdown of NFIL3 under hypoxia in hemin-treated K562 cells. In addition, a target of NFIL3, pim-1 proto-oncogene, serine/threonine kinase (PIM1), was obtained by RNA microarray after NFIL3 knockdown. PIM1 can rescue the inhibitory effect of NFIL3 on hemin-induced erythroid differentiation of K562 cells. Innovation and Conclusion: Our findings demonstrate that the HIF1α-NFIL3-PIM1 signaling axis plays an important role in erythroid differentiation under hypoxia. These results will provide useful clues for preventing the damage of acute hypoxia to erythropoiesis.

8.
Hum Cell ; 37(3): 648-665, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38388899

RESUMEN

Human myeloid leukemia cells (such as K562) could be used for the study of erythropoiesis, and mature erythroid markers and globins could be induced during leukemia cell differentiation; however, the pathways involved are different compared with those of hematopoietic stem cells (HSCs).We identified the differentially expressed genes (DEGs) of K562 cells and HSCs associated with stem cells and erythroid differentiation. Furthermore, we showed that hemin-induced differentiation of K562 cells could be induced by serum starvation or treatment with the tyrosine kinase inhibitor saracatinib. However, erythroid differentiation of HSCs was inhibited by the deprivation of the important serum component erythropoietin (EPO) or treatment with saracatinib. Finally, we found that the mRNA expression of K562 cells and HSCs was different during saracatinib-treated erythroid differentiation, and the DEGs of K562 cells and HSCs associated with tyrosine-protein kinase were identified.These findings elucidated the cellular phenomenon of saracatinib induction during erythroid differentiation of K562 cells and HSCs, and the potential mechanism is the different mRNA expression profile of tyrosine-protein kinase in K562 cells and HSCs.


Asunto(s)
Benzodioxoles , Eritropoyesis , Hemina , Quinazolinas , Humanos , Hemina/farmacología , Células K562 , Eritropoyesis/genética , Diferenciación Celular/genética , Células Madre Hematopoyéticas , ARN Mensajero , Tirosina , Proteínas Quinasas
9.
Int J Hematol ; 120(2): 157-166, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38814500

RESUMEN

G protein pathway suppressor 2 (GPS2) has been shown to play a pivotal role in human and mouse definitive erythropoiesis in an EKLF-dependent manner. However, whether GPS2 affects human primitive erythropoiesis is still unknown. This study demonstrated that GPS2 positively regulates erythroid differentiation in K562 cells, which have a primitive erythroid phenotype. Overexpression of GPS2 promoted hemin-induced hemoglobin synthesis in K562 cells as assessed by the increased percentage of benzidine-positive cells and the deeper red coloration of the cell pellets. In contrast, knockdown of GPS2 inhibited hemin-induced erythroid differentiation of K562 cells. GPS2 overexpression also enhanced erythroid differentiation of K562 cells induced by cytosine arabinoside (Ara-C). GPS2 induced hemoglobin synthesis by increasing the expression of globin and ALAS2 genes, either under steady state or upon hemin treatment. Promotion of erythroid differentiation of K562 cells by GPS2 mainly relies on NCOR1, as knockdown of NCOR1 or lack of the NCOR1-binding domain of GPS2 potently diminished the promotive effect. Thus, our study revealed a previously unknown role of GPS2 in regulating human primitive erythropoiesis in K562 cells.


Asunto(s)
Diferenciación Celular , Eritropoyesis , Hemina , Leucemia Eritroblástica Aguda , Co-Represor 1 de Receptor Nuclear , Humanos , Células K562 , Eritropoyesis/genética , Leucemia Eritroblástica Aguda/patología , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 1 de Receptor Nuclear/genética , Hemina/farmacología , Células Eritroides/metabolismo , Células Eritroides/citología , 5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , Hemoglobinas/metabolismo , Técnicas de Silenciamiento del Gen , Péptidos y Proteínas de Señalización Intracelular
10.
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
11.
Cell Rep ; 43(7): 114378, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38889007

RESUMEN

The Myb proto-oncogene encodes the transcription factor c-MYB, which is critical for hematopoiesis. Distant enhancers of Myb form a hub of interactions with the Myb promoter. We identified a long non-coding RNA (Myrlin) originating from the -81-kb murine Myb enhancer. Myrlin and Myb are coordinately regulated during erythroid differentiation. Myrlin TSS deletion using CRISPR-Cas9 reduced Myrlin and Myb expression and LDB1 complex occupancy at the Myb enhancers, compromising enhancer contacts and reducing RNA Pol II occupancy in the locus. In contrast, CRISPRi silencing of Myrlin left LDB1 and the Myb enhancer hub unperturbed, although Myrlin and Myb expressions were downregulated, decoupling transcription and chromatin looping. Myrlin interacts with the KMT2A/MLL1 complex. Myrlin CRISPRi compromised KMT2A occupancy in the Myb locus, decreasing CDK9 and RNA Pol II binding and resulting in Pol II pausing in the Myb first exon/intron. Thus, Myrlin directly participates in activating Myb transcription by recruiting KMT2A.


Asunto(s)
Elementos de Facilitación Genéticos , N-Metiltransferasa de Histona-Lisina , Proteína de la Leucemia Mieloide-Linfoide , Proteínas Proto-Oncogénicas c-myb , Transcripción Genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Proto-Oncogénicas c-myb/genética , Animales , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Ratones , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Elementos de Facilitación Genéticos/genética , ARN Polimerasa II/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , Quinasa 9 Dependiente de la Ciclina/metabolismo , Quinasa 9 Dependiente de la Ciclina/genética , Proto-Oncogenes Mas , Unión Proteica , Diferenciación Celular/genética , ARN Potenciadores
12.
Basic & Clinical Medicine ; (12): 963-969, 2017.
Artículo en Zh | WPRIM | ID: wpr-612011

RESUMEN

Objective To study the effects of interferon γ(IFN-γ) on terminal erythroid differentiation.Methods RT-qPCR was used to detect the expression of IFN-γ receptors during erythroid differentiation of K562 induced by hemin.Both hemin-induced K562 cells and human umbilical cord CD34+ cell derived primary erythroid cells were treated with IFN-γ.Erythroid differentiation of the cells was evaluated using RT-qPCR to detect the mRNA level of erythroid specific surface markers CD71 and CD235a,and benzidine staining assay was applied to explore the change of hemoglobin expression.Results The expression of IFN-γ receptors in K562 cells decreased and climbed up again after reaching the lowest point at 48 h of hemin induction.IFN-γ treatment increased CD71 and CD235a expression in both hemin-induced K562 cells and the later stage (E15D) primary erythroid cells.Benzidine staining showing increased globin protein expression in hemin-induced K562 cells after IFN-γstimulation.Furthermore,our results indicated that IFN-γ promoted hemin-induced K562 erythroid differentiation in a time dependent manner.Mechanistically,the results showed that IFN-γ treatment stimulated the expression of erythroid transcription factors NFE2,which was critically for erythroid maturation.Conclusions IFN-γaccelerates terminal erythroid differentiation in hemin-induced K562 cells and human umbilical cord CD34+ derived primary erythroid cells.

13.
Appl. cancer res ; 37: 1-4, 2017. tab, ilus
Artículo en Inglés | LILACS, Inca | ID: biblio-914823

RESUMEN

Background: Tet methylcytosine dioxygenase 2 (TET2) is frequently mutated and/or downregulated in myeloid neoplasm, including myelodysplastic syndromes. Despite the extensive studies, the specific contribution of TET2 in disease phenotype of myeloid neoplasms is not fully elucidated. Recent findings have grown attention on the role of TET2 in normal and malignant erythropoiesis. Methods: In the present study, we investigated TET2 mRNA levels by quantitative PCR during erythropoietin-induced erythroid differentiation CD34+ cells from healthy donor and myelodysplastic syndrome patients. Statistical analyses were performed using the ANOVA and Bonferroni post hoc test and a p-value <0.05 was considered statically significant. Results: TET2 expression is upregulated during erythroid differentiation of CD34+ cells from healthy donor and myelodysplastic syndrome patients. Conclusions: Our findings corroborate that TET2 is involved in the erythrocyte differentiation (AU)


Asunto(s)
Masculino , Femenino , Anciano , Síndromes Mielodisplásicos , Antígenos CD34 , Eritropoyesis
14.
Basic & Clinical Medicine ; (12): 1167-1170, 2015.
Artículo en Zh | WPRIM | ID: wpr-479333

RESUMEN

Objective To explore the effects of ZNF330 on erythroid differentiation of K562 cells and underlying mechanism .Methods Realtime PCR was performed to detect the expression of ZNF 330 in K562 cells induced by hemin .After CD34 +cells being infected by the recombination lentivirus ZNF 330-RNAi, Realtime PCR was applied to detect the expression of CD235a and γ-globin.The luciferase report assay was performed to examine if ZNF 330 could act as a trans-acting factor in 293T/17 cells.Co-Immunoprecipitation (Co-IP) was applied in 293T/17 cells to detect the interaction between ZNF 330 and ZNF408 which was involved in mRNA degradation .Results The ex-pression of ZNF330 was up-regulated after hemin treatment .The expression of CD235a andγ-globin decreased after inhibition expression of ZNF 330 had no effect on report gene .Co-Ip in two ways confirmed the direct binding be-tween ZNF330 and ZNF408 .Conclusions ZNF330 can promote erythroid differentiation , and a possible mecha-nism is that ZNF330 inhibits the function of ZNF 408 , a factor that is involved in mRNA degradation , through the interaction between the two proteins .

15.
Basic & Clinical Medicine ; (12): 167-173, 2015.
Artículo en Zh | WPRIM | ID: wpr-480679

RESUMEN

Objective To study the effects of microRNA-34a-5p on erythroid differentiation of K562 cells.Methods K562 cells were transfected with the microRNA-34a-5p mimics and antisense inhibitors specifically targeting mi-croRNA-34a-5p, respectively.The effects of over-expression or knocking-down of microRNA-34a-5p were exam-ined by Quantitative RT-PCR.Flow cytometry was performed to detect specific surface marker of erythroid cells . The benzidine staining assay was used to access the differentiation of K 562 cells.Western blot was performed to de-tect miRNA targets.Results microRNA-34a-5p was down-regulated at the early stage of K562 erythroid differenti-ation.Over-expression of microRNA-34a-5p in K562 cells attenuates erythroid differentiation , in contrast, inhibi-tion of microRNA-34a-5p accelerates erythroid pheotypes in K562 cells.c-MYB was found to be the direct target of microRNA-34 a-5 p in erythroid cells .Conclusions microRNA-34 a-5 p regulates early erythroid differentiation of K562 cells via repressing c-MYB.

16.
Artículo en Zh | WPRIM | ID: wpr-840294

RESUMEN

Objective: To investigate the role of SHP1 gene in inducing apoptosis and erythroid differentiation in human erythromyeloblastoid leukemia cell line K562. Methods: The full length cDNA of SHP1 gene was cloned by RT-PCR and was subcloned into mammalian expression vector pcDNA3.0. The cDNA sequence of the cloned gene was validated by enzyme digestion and DNA sequencing. Then the recombinant plasmid was used to transfect K562 cells via lipofectin. The apoptosis of K562 cells was examined by Hoechst 33258 staining assay and Annexin V/PI double-labeled assay; the differentiation of K562 cells was observed by benzidine staining and expression of glycophorin A (GPA). Results: RT-PCR and Western blotting analysis showed expression of SHP1 in K562 cells after transfection with pcDAN3-SHP1 plasmid. Apoptotic cells were detected in the K562 cells 48 h after treatment with pcDNA3-SHP1, with the apoptosis rate being 16.84%, which was significantly higher than that in cells transfected with pcDNA3.0 (6.23% , P=0.000). The positive rate of benzidine staining was 14.67% and the positive rate of GPA expression was 19.38% in cells treated with pcNDA3-SHP1, both were significantly different from those in the cells transfected with pcDNA3.0 (P=0.005). Conclusion: Over-expression of SHP1 can effectively induce apoptosis and erythroid differentiation in K562 cells.

17.
Artículo en Inglés | WPRIM | ID: wpr-720789

RESUMEN

BACKGROUND: The Polycomb-group gene Bmi-1 is known to be a molecular regulator of self-renewal of normal and leukemic stem cells and be involved in various aspects of cellular proliferation, differentiation, and survival. METHODS: This study evaluated the effects of overexpression of Bmi-1 on human cord blood CD34+ cells. Bmi-1 was introduced into CD34+ cells through lentivirus transduction. Bmi-1 expressing CD34+ cells were applied to colony forming assay, stromal co-culture, and cytokine-stimulatied culture. RESULTS: Ectopic expression of Bmi-1 resulted in the increased number of erythroid colonies in primary and secondary colony forming assay in an erythropoietin dependent manner. In stromal co-culture, Bmi-1-expressing postnatal hematopoietic stem cells seemed to lose the ability of self-renewal, as determined by week 5 cobblestone area-forming cell assay and by week 5 secondary colony assay. In cytokine-stimulated suspension culture of Bmi-1-transduced CD34+ cells, we observed increased erythropoiesis marked by Glycophorin A expression. CONCLUSION: Our data suggest that ectopic expression of Bmi-1 in human hematopoietic stem/progenitor cells may result in the differentiation to the erythroid lineage rather than promoting self-renewal.


Asunto(s)
Humanos , Proliferación Celular , Técnicas de Cocultivo , Eritropoyesis , Eritropoyetina , Sangre Fetal , Glicoforinas , Células Madre Hematopoyéticas , Lentivirus , Células Madre
18.
Artículo en Ko | WPRIM | ID: wpr-720633

RESUMEN

BACKGROUND: The K562 erythroleukemia cell line was used to study the molecular mechanisms regulating lineage commitment of hematopoietic cells. There are numerous similarities between the erythroid or megakaryocytic lineages. In this study, we examined role of the region -269~-240 of gamma-globin gene promoter in fetal hemoglobin expression during either erythroid or megakaryocytic differentiation. METHODS: K562 cells were cultured and treated with differentiation inducers. Hemoglobin content was scored by benzidine staining, and hemoglobin F was stained by acid elution technique. To determine whether transcription factor binding to the gamma-globin gene promoter is critical to lineage determination, DNA-protein interaction of gamma-globin gene promoter was examined under both uninduced and induced conditions of K562 cells using gel mobility shift assay and southwestern blot analysis. RESULTS: Phorbol 12-myristate 13-acetate (PMA) induced a megakaryocytic differentiation, but suppressed erythroid differentiation. On the other hand, hydroxyurea (HU), hemin, n-butanol, and sodium butyrate (NaB) induced the expression of erythroid phenotypes. Parallel to hemoglobinization, increase in gamma-globin mRNA was observed in HU- and hemin-treated K562 cells. Gel mobility shift assay and southwestern blot analysis revealed that binding of a erythroid-specific protein (p120) to the region -269~-240 of gamma-globin gene promoter occurred with treatment of erythroid differentiation inducers and did not occur with treatment of PMA. CONCLUSION: These results suggest that erythroid differentiation inducers may act via DNA- protein interaction at the gamma-globin gene promoter region to induce erythroid differentiation.


Asunto(s)
1-Butanol , Southwestern Blotting , Ácido Butírico , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Hemoglobina Fetal , gamma-Globinas , Mano , Hemina , Hidroxiurea , Células K562 , Leucemia Eritroblástica Aguda , Fenotipo , Regiones Promotoras Genéticas , ARN Mensajero , Factores de Transcripción
19.
Artículo en Inglés | WPRIM | ID: wpr-56734

RESUMEN

The chronic myelogenous leukemic K562 cell line carrying Bcr-Abl tyrosine kinase is considered as pluripotent hematopoietic progenitor cells expressing markers for erythroid, granulocytic, monocytic, and megakaryocytic lineages. Here we investigated the signaling modulations required for induction of erythroid differentiation of K562 cells. When the K562 cells were treated with herbimycin A (an inhibitor of protein tyrosine kinase), ras antisense oligonucleotide, and PD98059 (a specific inhibitor of MEK), inhibition of ERK/MAPK activity and cell growth, and induction of erythroid differentiation were observed. The ras mutant, pZIPRas61leu-transfected cells, K562-Ras61leu, have shown a markedly decreased cell proliferation rate with approximately 2-fold doubling time, compared with the parental K562 cells, and about 60% of these cells have shown the phenotype of erythroid differentiation. In addition, herbimycin A inhibited the growth rate and increased the erythroid differentiation, but did not affect the elevated activity of ERK/MAPK in the K562-Ras61leu cells. On the other hand, effects of PD98059 on the growth and differentiation of K562-Ras61leu cells were biphasic. At low concentration of PD98059, which inhibited the elevated activity of ERK/MAPK to the level of parental cells, the growth rate increased and the erythroid differentiation decreased slightly, and at high concentration of PD98059, which inhibited the elevated activity of ERK/MAPK below that of the parental cells, the growth rate turned down and the erythroid differentiation was restored to the untreated control level. Taken together, these results suggest that an appropriate activity of ERK/MAPK is required to maintain the rapid growth and transformed phenotype of K562 cells.


Asunto(s)
Humanos , Androstadienos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina , Diferenciación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Precursoras Eritroides/fisiología , Células Precursoras Eritroides/citología , Eritropoyesis , Flavonas/farmacología , Células K562 , Leucemia Mieloide/patología , Oligonucleótidos Antisentido/farmacología , Quinonas/farmacología , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda