Open Access

Effects of miR‑210‑3p on the erythroid differentiation of K562 cells under hypoxia

  • Authors:
    • Caiyan Hu
    • Yupeng Yan
    • Chengbing Fu
    • Jin Ding
    • Tiantian Li
    • Shuqiong Wang
    • Liu Fang
  • View Affiliations

  • Published online on: June 7, 2021     https://doi.org/10.3892/mmr.2021.12202
  • Article Number: 563
  • Copyright: © Hu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

GATA binding protein 1 (GATA‑1) is one of the most important hematopoietic transcription factors in the production of blood cells, such as platelets, eosinophils, mast cells and erythrocytes. GATA‑1 regulates the participation of microRNA (miRNAs/miRs) in erythroid differentiation under normoxia. However, GATA‑1 expression and the regulation of miR‑210‑3p in the context of erythroid differentiation under hypoxia remain unknown. The present study examined the expression levels of GATA‑1 and miR‑210‑3p in the model of erythroid differentiation in K562 cells under hypoxia, and determined the effects of GATA‑1, miR‑210‑3p and SMAD2 on erythroid differentiation through lentivirus transfection experiments. The present study detected increased GATA‑1 expression under hypoxia. Moreover, miR‑210‑3p was identified as a positive regulator of erythroid differentiation, which was upregulated both during erythroid differentiation and in GATA‑1 overexpression experiments under hypoxia. Importantly, in the K562 cell model of erythroid differentiation under hypoxia, miR‑210‑3p was upregulated in a GATA‑1‑dependent manner. Using a double luciferase reporter assay, miR‑210‑3p was identified as a downstream target of GATA‑1‑mediated regulation of erythropoiesis. Gain‑ or loss‑of‑function analysis of miR‑210‑3p identified its importance in erythroid differentiation. Furthermore, it was found that SMAD2 may be a downstream target gene for miR‑210‑3p. Bioinformatics predictions suggested that SMAD2 mediated miR‑210‑3p‑induced regulation of erythroid differentiation. Collectively, the present study provides novel insights into the miRNA regulation of erythroid differentiation.

Introduction

Erythropoiesis is a multistep process that produces erythroid cells, and it is regulated by transcription factors and oxygen concentration (1). The body oxygen level is one of the most critical factors affecting erythropoiesis. For example, low oxygen levels trigger erythropoiesis via erythropoietin release (2). Moreover, the oxygen level in the bone marrow microenvironment mediates the interaction between erythroid progenitors and stromal cells, further promoting erythroid differentiation (2). Changes in the oxygen level also alter the expression levels of globin genes, such as γ-globin (35).

GATA binding protein 1 (GATA-1), a member of the GATA transcription factor family, is expressed in hematopoietic progenitor cells, megakaryocytes, eosinophil granulocyte cells, testicular mast cells and testicular cells (6). GATA-1 is essential for normal erythroid cell proliferation and development (7,8). Under normoxia, GATA-1 modulates erythroid cells by inducing the expression of various erythroid differentiation- and maturation-related genes, such as zinc finger protein, FOG family member 1 and erythroid Krüppel-like factor (9,10). Accumulating evidence has indicated that microRNA (miRNA/miR) is a downstream target of GATA-1. For instance, GATA-1 activates miR-451a and completes a regulatory circuit that modulates erythroid maturation (11). However, the mechanism underlying GATA-1 regulation of erythroid differentiation under hypoxia remains unknown.

miRNAs are members of a large class of non-coding RNAs that control gene expression and regulate a wide array of biological processes. miRNAs target mRNAs and induce translational repression or mRNA degradation (12). Several miRNAs, including miR-451a and miR-210-3p, profoundly alter the erythroid phenotype by regulating early cell maturation and proliferation, the expression level of fetal γ-globin genes and enucleation (13). miR-210-3p, known as ‘hypoxamiR,’ contributes to the cellular adaptation to hypoxia (14). Moreover, miR-210-3p is associated with an elevated expression level of fetal γ-globin in mithramycin-induced K562 cells (13). miR-210-3p expression is also reportedly elevated during murine fetal liver erythroid cell differentiation in vitro (15). Thus, miR-210-3p may mediate erythropoiesis during hypoxia.

K562 cells, a myelogenous leukemia cell line derived from the highly undifferentiated progenitor of the erythrocytic and megakaryocytic lineages (16), have the potential for megakaryocyte and erythroid cell differentiation, thereby providing an excellent model system for investigating cellular differentiation-related mechanisms (17).

The present study used a K562 cellular erythroid differentiation model under hypoxia to investigate the expression level of GATA-1 and the mechanism underlying its effects on erythroid cell differentiation and miR-210-3p expression regulation, which ultimately affects SMAD2 expression.

Materials and methods

Cell lines and lentivirus vectors

K562 cells were purchased from The Cell Bank of Type Culture Collection of Chinese Academy of Sciences and were maintained in RPMI-1640 medium (Hyclone; Cytiva) supplemented with 10% FBS (Zhejiang Tianhang Biotechnology Co., Ltd.) and 1% penicillin/streptomycin solution (Beijing Solarbio Science & Technology Co., Ltd.), at 37°C and 5% CO2. Lentivirus vectors (LV-GATA1, cat. no. 26211-1; LV-GATA1-RNAi, cat. no. 18817-1; LV-hsa-mir-210, cat. no. 41113-2; LV-hsa- miR-210-3p-inhibition, cat. no. 5212-1; LV-SMAD2-RNAi, cat. no. 15901-1) and transfection reagent (HitransG A&P, cat. no. REVG003-1) were purchased from Shanghai GeneChem Co., Ltd.

Experimental grouping

K562 cells were divided into a normoxic group (21% O2, 5% CO2, 37°C, saturation humidity) and a hypoxic group (1% O2, 5% CO2, 94% N2, 37°C, saturation humidity). Hemin was added (40 mM/l; 37°C; Beijing Solarbio Science & Technology Co., Ltd.), and incubation was performed for 96 h.

Western blot analysis

K562 cells were lysed with mammalian cell lysis buffer (Nanjing KeyGen Biotech Co., Ltd.) containing protease and phosphatase (both Nanjing KeyGen Biotech Co., Ltd.) inhibitors. The BCA protein determination method was used to detect the amount of protein. The total protein content was extracted with a 10% Tris-HCl gradient gel (Bio-Rad Laboratories, Inc.) and transferred onto PVDF membranes, which was blocked using 5% non-fat milk in TBS/Tween-20 (0.1%) for 2 h at room temperature. The membrane was then probed with antibodies for GATA-1 (monoclonal rabbit anti-human; 1:10,000; cat. no. ab181544; Abcam), SMAD2 (monoclonal rabbit anti-human; 1:10,000; cat. no. ab40855; Abcam) and α-tubulin (monoclonal mouse anti-human; 1:10,000; cat. no. ab7291; Abcam) and incubated overnight at 4°C. The following day, the PVDF membrane was taken out of the refrigerator, reheated at room temperature for 1 h and washed in TBS/Tween-20 (0.1%). The secondary antibodies (Goat anti-mouse IgG, HRP conjugate, cat. no. SA00001-1; Goat Anti-Rabbit IgG, HRP conjugate, cat. no. SA00001-2; ProteinTech Group, Inc.) were added and incubated at room temperature for 1 h, followed by washing with TBS/Tween-20 (0.1%). The visualization reagent (cat. no. 34094; Thermo Fisher Scientific, Inc.) was prepared according to the instructions of the kit. Quantity One software (4.6.2; Bio-Rad Laboratories, Inc.) was used for density analysis of protein bands.

RNA isolation and reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the cells harvested using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. RNA was quantified using an Ultra-microspectrophotometer (Nanodrop 2000; Thermo Fisher Scientific, Inc.) at 260 nm. cDNA was synthesized using a reverse transcriptase kit (60 min at 42°C and then 5 min at 70°C; cat. no. K1691; Thermo Fisher Scientific, Inc.) from 1 µg total RNA. For mRNAs, RT-qPCR was performed using the ABI 7500 real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) and the QuantiNova SYBR Green PCR kit (Qiagen Sciences, Inc.), according to the manufacturer's instructions (95°C, 2 min for pre-degeneration; followed by 40 cycles at 95°C for 5 sec and 60°C for 30 sec). The relative quantification of the transcripts was performed using the 2−ΔΔCq method (18). The primer sequences used were as follows: γ-globin forward, 5′-GCAGCTTGTCACAGTGCAGTTC-3′ and reverse, 5′-TGGCAAGAAGGTGCTGACTTC-3′; and β-actin forward, 5′-CCTGGCACCCAGCACAAT-3′ and reverse, 5′-GCTGATCCACATCTGCTGGAA-3′.

miRNA isolation and RT-qPCR

miRNA was isolated using a miRcute miRNA isolation kit (Tiangen Biotech Co., Ltd.) and transcribed into cDNA, according to the manufacturer's instructions (42°C for 60 min, 95°C for 3 min; cat. no. KR211; Tiangen Biotech Co., Ltd.). RT-qPCR was performed using ABI 7500 real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) and a miRcute Plus miRNA qPCR Detection kit (FP411-02, Tiangen Biotech Co., Ltd.), according to the manufacturer's instructions (initial denaturation at 95°C for 15 min; followed by 40 cycles at 94°C for 20 sec and 60°C for 34 sec). The relative quantification of the transcripts was performed using the 2−ΔΔCq method (18). Primers for internal reference U6 (cat. no. CD201-0145) and miR-210-3p (cat. no. CD201-0293) were purchased from Tiangen Biotech Co., Ltd.

Benzidine staining

A total of 1×106 cells were collected into a 1.5-ml EP tube with 500 µl PBS, following which 15 µl 0.4% benzidine solution and 10 µl 3% H2O2 were added and the suspension was incubated for 3 min at room temperature. Then, 1 µl 5% sodium nitroprusside solution was added. Then, 10 min later at room temperature, 10 µl suspension was collected for observation (magnification, ×200) using an inverted microscope (Olympus Corporation). The positive cells were blue-black in color, and 200 cells were counted to calculate the positive cell rate.

Wright's-Giemsa staining

A 10-µl cell suspension was coated on the slide. After drying at room temperature, the staining solution A (Wright's-Giemsa Staining Solution) was added for 1 min and the staining solution B (PBS) was added and mixed for 8–10 min at room temperature. After rinsing the excess dye with running water, the cells were observed under an inverted microscope (magnification, ×1,000; Olympus Corporation). In total, 200 cells were counted in each preparation, and the proportion of cells with increased volume, nuclear migration and nuclear shrinkage was determined.

Lentivirus transfection

For the GATA-1 overexpression assay, K562 cells were transfected with either the GV358 vector with a full-length GATA-1 [multiplicity of infection (MOI)=30] or with an empty vector, and a transfection reagent was added into medium. For the GATA-1 knockdown assay, K562 cells were transfected with either a GV248 vector with the sequence 5′-ACAGAGCATGGCCTCCAGA-3′ (MOI=80) or with an empty vector, and a transfection reagent was added into medium. FLAG was inserted into the lentivirus vector of GATA-1 overexpression, and the high-molecular-weight bands in western blotting results were FLAG protein bands. After adding lentivirus reagent in the cell medium (operates at room temperature), the culture was maintained in the incubator at 37°C with 5% CO2. After 96 h, the cells were observed using an inverted fluorescence microscope (magnification, ×100; Olympus Corporation). Transfection efficiency was determined using western blotting.

For the miR-210-3p overexpression and knockdown assay, K562 cells were transfected with either a GV309 vector with a full-length cDNA of miR-210-3p (MOI=30) or a GV280 vector with a reverse complementary sequence (5′-TCAGCCGCTGTCACACGCACAG-3′; MOI=50), and a transfection reagent was added into medium. K562 cells transfected with lentiviruses with the sequence, 5′-TTCTCCGAACGTGTCACGT-3′ (MOI=30), were used as negative controls (NC) for both reactions. After adding lentivirus reagent in the cell medium (operates at room temperature), the culture was continued in the incubator at 37°C with 5% CO2. After 96 h, the cells were observed using an inverted fluorescence microscope (magnification, ×100; Olympus Corporation). Transfection efficiency was confirmed via RT-qPCR.

For the SMAD2 inhibition assay, K562 cells were transfected with either a GV248 vector with the sequence 5′-CGATTAGATGAGCTTGAGAAA-3′ (MOI=80) or with an empty vector, and a transfection reagent was added into medium. After adding lentivirus reagent in the cell medium (operates at room temperature), the culture was continued in the incubator at 37°C with 5% CO2. After 96 h, the cells were observed using an inverted fluorescence microscope (magnification, ×100; Olympus Corporation). Transfection efficiency was confirmed using western blotting.

Flow cytometry analysis of CD235a

K562 cells were harvested at the indicated time points (induced by hemin for 0 and 96 h) and washed twice at 4°C with PBS. Cells were incubated with APC-Cy7-conjugated anti-CD235a antibodies for 15 min (cat. no. 349116; BioLegend, Inc.). Flow cytometry was conducted in K562 cells using the fluorescence-activated cell sorter Beckman CytoFLEX flow cytometer (Beckman Coulter, Inc.) to analyze the primary erythroid cell surface marker CD235a.

Luciferase reporter assay

Cells in the logarithmic growth stage were resuspended, counted, inoculated in a 24-well culture plate (~105 cells, depending on the size of the cells) and cultured at 37°C in a 5% CO2 incubator until the degree of cell fusion reached ~60%. PGL3 plasmid (Shanghai GeneChem Co., Ltd.) containing firefly and adrenal luciferase reporter genes was transfected into cells at room temperature using the X-tremeGENE HP transfection reagent (cat. no. 06366236001; Roche Diagnostics) for 24–48 h to observe the fluorescent marker gene expression on the plasmid and to determine the transfection efficiency, or cells were transfected for 48 h to detect luciferase activity using a Dual-Luciferase® Reporter Assay system (cat. no. E2910; Promega Corporation), according to the manufacturer's instructions (1921).

miRNA target prediction

The downstream target genes of miR-210-3p were predicted by using databases such as miRBase (miRbase, http://www.mirbase.org/) and TargetScan (TargetScanHuman 7.2, http://www.targetscan.org/vert_72/). Finally, from a literature review, the target genes related to erythroid development were screened and sequence matching analysis was carried out.

Statistical analysis

SPSS 19.0 software (IBM, Corp.) was used for data processing. Normally distributed data are presented as mean ± SD from three independent experiments. One-way ANOVA was used to compare multiple groups. When the variances were homogeneous, the Tukey's test was used for post hoc analysis. When the variances were uneven, Tamhane's T2 test was used for post hoc analysis. An unpaired Student's t-test was used to compare two groups. P<0.05 was considered to indicate a statistically significant difference. Experiments was repeated for three times.

Results

K562 cells successfully differentiate into erythroid cells under hypoxia

An erythroid cell differentiation model under hypoxia was established using hemin-induced K562 cells. Benzidine staining was performed to identify hemoglobin-containing cells as an indicator of erythrocyte differentiation. The results demonstrated that hemin treatment significantly increased the proportion of benzidine-positive K562 cells under hypoxia (Fig. 1A and B). Moreover, under hypoxia, a 2.6-fold elevation of γ-globin expression was observed at 96 h compared with the level at 0 h (Fig. 1C). The results also indicated that the 96-h hemin treatment increased the proportion of CD235a+ K562 cells compared with the 0-h treatment group (Fig. 1D and E). Thus, it was found that K562 cells successfully differentiated into erythroid cells under hypoxia.

GATA-1 expression is upregulated in hemin-induced K562 cells under hypoxia

To assess the influence of hypoxia on GATA-1 expression during erythroid cell differentiation, GATA-1 expression levels were determined in hemin-treated K562 cells using western blotting. The results demonstrated that the expression level of GATA-1 was significantly increased in the hypoxia group compared with that in the normoxia group (Fig. 2).

GATA-1 promotes the erythroid differentiation of K562 cells under hypoxia

To further validate the role of GATA-1 in erythroid differentiation under hypoxia, the corresponding erythroid differentiation index was detected after GATA-1 overexpression or knockdown. Under fluorescence microscope, the cells showed green fluorescence, indicating successful lentivirus transfection (Fig. 3A and B). The results of western blot analysis of GATA-1 protein expression showed that its expression level was higher in the overexpression group compared with that of the NC group (Fig. 3C); In addition, the results of western blot analysis of GATA-1 protein expression indicated that the relative expression level in the knockdown group was significantly lower compared with that of the NC group (Fig. 3D).

An elevation in the percentage of benzidine-positive cells (Fig. 4A; left panel) and γ-globin expression (Fig. 4B; left panel) was observed at 96 h in GATA-1-overexpressing K562 cells compared with the NC group. Additionally, the percentage of CD235a+ (Fig. 4C; left panel) in GATA-1-overexpressing K562 cells showed an expected increase at 96 h under hypoxia. Wright's-Giemsa staining also demonstrated that the proportion of cells with a larger cell volume and a smaller nucleus accumulated at one side was significantly higher in the GATA-1-overexpressing group compared with that in the NC group (Fig. 4D; upper panel). Conversely, transfection with the GV248 vector (knockdown group) inhibited the expression of GATA-1 in K562 cells, as indicated by the decrease in benzidine-positive cells (Fig. 4A; right panel) and repressed γ-globin accumulation (Fig. 4B; right panel) compared with the phenotype observed in the NC group. Concurrently, the reduced percentage of CD235a+ cells (Fig. 4C; right panel), as well as the proportion of cells with an increased volume and a lopsided nucleus were significantly lower in the GATA-1-knockdown group compared with in the NC group; however, no significant difference was observed with hypoxia (Fig. 4D; bottom panel). These results indicated that GATA-1 promoted erythroid differentiation in K562 cells under hypoxia.

miR-210-3p is a direct target gene of GATA-1 in erythroid differentiation

qPCR was conducted to validate the expression level of miR-210-3p in K562 cells during erythroid differentiation under hypoxia. The expression level of miR-210-3p in the hypoxia group was higher compared with that in the normoxia group (Fig. 5A). To further determine the direct relevance of miR-210-3p upregulation in response to GATA-1 activation during erythroid cell differentiation, qPCR was conducted to analyze miR-210-3p expression in K562 cells after GATA-1 overexpression or knockdown. The results demonstrated that miR-210-3p showed a significant increase in GATA-1-overexpressing K562 cells under hypoxia (Fig. 5B). Moreover, miR-210-3p was downregulated after the GATA-1 knockdown (Fig. 5C), which revealed the mechanism via which GATA-1 could activate miR-210-3p during erythroid cell differentiation under hypoxia.

To confirm these findings, a dual-luciferase reporter assay was performed to determine the binding site of miR-210-3p and GATA-1. The binding site prediction of the miR-210-3p promoter region is shown in Fig. 5D. The reporter assays revealed a GATA-1-dependent activation of the miR-210-3p promoter. Notably, mutations of the GATA-1-binding site abolished this upregulation, as evidenced by the luciferase activity assay (Fig. 5E).

miR-210-3p promotes the erythroid differentiation of K562 cells under hypoxia

To examine the role of miR-210-3p in erythroid cell differentiation under hypoxia, a miR-210-3p-overexpressing and knockdown lentivirus was transfected into K562 cells. Under fluorescence microscope, the cells showed green fluorescence, indicating successful lentivirus transfection (Fig. 6A and B), and RT-qPCR was performed to assess the transfection efficiency (Fig. 6C and D). The effect of NC lentivirus on the expression level of miR-210-3p was also detected via RT-qPCR. The results demonstrated that, compared with the blank control group, the NC lentivirus did not affect the expression level of miR-210-3p itself, which was convenient for conducting subsequent experiments (Fig. 6E).

The benzidine staining results demonstrated that miR-210-3p overexpression increased the proportion of benzidine-positive cells after 96 h of hemin treatment in K562 cells under hypoxia (Fig. 7A; left panel). Furthermore, a 1.7-fold increase in γ-globin expression was observed at 96 h in miR-210-3p-overexpressing K562 cells compared with that in the NC group under hypoxia (Fig. 7B; left panel). It was found that miR-210-3p overexpression increased the percentage of CD235a+ cells compared with that in the NC group under hypoxia (Fig. 7C; left panel). Wright's-Giemsa staining also showed that miR-210-3p served a vital role in erythroid cell differentiation by increasing the cell volume, nuclear shrinkage and incidence of a lopsided nucleus under hypoxic conditions (Fig. 7D; left panel). In agreement with the gain-of-function data, a significant decrease was observed in the results of the erythroid cell markers γ-globin and CD235a, combined with the benzidine staining, under normoxia, which demonstrated that loss of miR-210-3p function impaired erythroid cell maturation (Fig. 7; right panels of A-D). Thus, it was suggested miR-210-3p promoted erythroid cell differentiation.

SMAD2 is a potential downstream target gene for miR-210-3p

As miRNAs function by translationally repressing their targets (22), the current study aimed to examine the targets of miR-210-3p. To this end, TargetScan was used to predict the downstream target genes of miR-210-3p (Fig. 8A). When further establishing the interaction between miR-210-3p and SMAD2, a significant decrease in SMAD2 expression was observed in the presence of lentivirus-mediated miR-210-3p overexpression in K562 cells under hypoxia compared with the NC group, in which no effect was observed (Fig. 8B). Conversely, SMAD2 expression was increased after the endogenous knockdown of miR-210-3p by the inhibition lentivirus under normoxia (Fig. 8C).

SMAD2 acts as a negative regulator of erythroid cell differentiation under hypoxia

To date, the function of SMAD2 in erythropoiesis remains unknown. Thus, to investigate its biological role in erythroid cell differentiation under hypoxia, a loss-of-function experiment was conducted using lentivirus-mediated SMAD2-inhibition in K562 cells. Under fluorescence microscope, the cells showed green fluorescence, indicating successful lentivirus transfection, western blotting was performed to assess transfection efficiency (Fig. 9A). The percentage of CD235a+ cells (Fig. 9B) and benzidine-positive cells (Fig. 9C) was increased in SMAD2 inhibition lentivirus-transfected K562 cells under normoxic and hypoxic conditions. Furthermore, Wright's-Giemsa staining revealed that, under normoxic and hypoxic conditions, the percentage of cells with larger volumes, lopsided nucleus and nuclear shrinkage was higher compared with that in the NC group (Fig. 9D). As expected, SMAD2 knockdown increased the expression level of γ-globin in K562 cells at 96 h under normoxic and hypoxia (Fig. 9E). The aforementioned results suggested that the increase was more significant under hypoxia. Therefore, it was indicated SMAD2 negatively regulated erythroid cell differentiation under hypoxia.

SMAD2 does not bind to miR-210-3p directly

To further determine the regulatory relationship between miR-210-3p and SMAD2, direct binding sites between miR-210-3p and SMAD2 were detected using a dual-luciferase assay. The results demonstrated that the relative luciferase activity of miR-210-3p-overexpressing vectors combined with SMAD2 wild-type vectors was not significantly different compared with that of SMAD2 mutant vectors. Moreover, miR-210-3p could not bind directly to the 3′ untranslated region (UTR) of SMAD2 (Fig. 9F).

Discussion

Erythropoiesis, the process of erythroid cell production, is controlled by several factors, including oxygen levels (23). Hypoxia occurs at high-altitude areas and in several physiological and pathological processes, such as rapid tissue growth and acute and chronic ischemia (24). Recently, studies have been performed from the perspective that various geographical and specific environmental differences may influence erythroid cell differentiation. In fact, it has been reported that ‘special environments’ (e.g., high-altitude areas) significantly influence various physiological functions of the human body, including erythroid cell differentiation (25). The effect of high-altitude hypoxia on erythroid cell development continues to gain increased attention from researchers. Of note, hypoxia may affect the core regulatory factors of healthy erythroid cell differentiation (26).

GATA-1 regulates numerous erythroid cell differentiation-specific genes by binding to its target protein via double zinc finger domains. By activating target genes, GATA-1 helps to establish and maintain erythroid phenotypes (27). Moreover, it has been reported that the maturation of GATA-1-deficient erythroid progenitor cells was inhibited, and apoptosis was induced. However, the erythroid cells matured when GATA-1 activity was restored in GATA-1 knockout erythroid lines (28). GATA-1 can modulate erythroid cell differentiation by modulating critical miRNAs, and the mechanism underlying gene regulation via these post-transcriptional inhibitors is being gradually revealed by experimental observations (11). Previous studies have confirmed that miRNAs are essential regulators of all stages of hematopoiesis and hematopoietic disorders (29,30). Some miRNAs reportedly prevent the differentiation of early-stage progenitor cells or regulate the terminal stages of hematopoietic development (31).

In the present study, an erythroid differentiation model of K562 cells under hypoxia was used to investigate the effects and functional relationship of GATA-1 and miR-210-3p on erythroid differentiation and elucidate the possible regulatory mechanism of erythroid differentiation under hypoxia. The expression level of GATA-1 protein in the K562 cell erythroid differentiation model was significantly higher compared with that in the normoxic group. Additionally, the current study evaluated the GATA-1-mediated promotion of erythroid development in K562 cells during hypoxia through gain- and loss-of-function experiments. The results demonstrated that GATA-1 promoted erythroid cell differentiation under hypoxic conditions. Other regulatory pathways under hypoxia, however, may also influence erythroid cell differentiation (32). Hypoxia can upregulate the expression level of GATA-1 and accelerate erythroid cell differentiation; this may be why some indicators of erythroid differentiation after inhibiting GATA-1 under hypoxia showed no significant difference compared with the NC group.

The present study demonstrated that the expression level of miR-210-3p was associated with the upregulation and downregulation of GATA-1 during erythroid differentiation of K562 cells under hypoxia. A dual-luciferase assay was used to verify the relationship between GATA-1 and miR-210-3p. The results demonstrated that the transcription factor could bind to the wild-type miR-210-3p vector, increasing the level of fluorescence expression, thus suggesting the presence of a direct binding site between GATA-1 and the miR-210-3p promoter.

miR-210-3p reportedly mediates hypoxia-induced K562 and erythroid progenitor cell differentiation (33). miR-210-3p also participates in the regulation of erythrocytic maturation, proliferation (13) and γ-globin gene expression in early erythrocytes. Previous research has reported that miR-210-3p enhanced CD34+ erythroid progenitor cell differentiation (34). Moreover, a notable increase in miR-210-3p expression during erythroid differentiation of a murine fetal liver cell culture has been observed (15), and the induction of erythropoiesis after phenylhydrazine-induced hemolytic anemia increased miR-210-3p levels (15). Based on these results, it was suggested that miR-210-3p could affect erythroid differentiation under hypoxia. Therefore, the current study aimed to upregulate and to inhibit miR-210-3p expression to detect the corresponding erythroid differentiation indexes. It was found that miR-210-3p positively regulated erythroid cell differentiation under hypoxia.

To determine the possible underlying mechanism, a previous study investigated miR-210-3p using miRBase/Targetscan/KEGG (Kyoto Encyclopedia of Genes and Genomes) (35) pathway analyses using bioinformatics software and identified SMAD2, which is involved in the proliferation (36), apoptosis (37) and differentiation (38) of several types of cells, as the possible downstream target gene involved in the regulation of erythroid differentiation. SMAD2, which exerts an inhibitory influence under normal steady-state conditions, has emerged as an important regulator of erythropoiesis (3942). Additionally, overactivation or dysregulation of SMAD2 signaling has been implicated in diseases characterized by impaired erythroid cell differentiation (4346). Histological examination has shown that SMAD2 was activated in hematopoietic progenitor cells and participated in the regulation of TGF-β-mediated proliferation and erythroid differentiation (43). Accumulating evidence has also suggested that luspatercept-mediated inhibition of SMAD2 signaling promotes erythroid differentiation (47). Moreover, inhibition of SMAD2 increased the level of hepatocyte growth factor, an effective angiogenic factor, in patients with squamous cell carcinoma (48,49). Taken together, these results indicate that SMAD2 could regulate the development of erythrocytes. It has also been reported that miR-210-3p has a direct binding site for members of the SMAD family, which regulates its expression (50). Therefore, we hypothesized that miR-210-3p may serve a role in promoting erythroid cell differentiation by inhibiting the gene expression of SMAD2.

To verify whether SMAD2 was directly regulated by miR-210-3p, a dual-luciferase assay was conducted. However, it was found that miR-210-3p did not bind directly to the 3′UTR of SMAD2, and it was identified that it negatively affected SMAD2 expression, as determined detecting SMAD2 protein expression after the overexpression and knockdown of miR-210-3p expression. Further analysis demonstrated that SMAD2 knockdown enhanced erythroid cell differentiation. A previous study revealed that miR-210-3p could inhibit the activity of the TGF signaling pathway during osteoblast differentiation (51). However, SMAD-1/5/8 competitive inhibition may interfere with SMAD2/3 binding to Co-SMAD (52), thereby accelerating osteoblast differentiation (53). Therefore, we hypothesized the existence of other sequences in the 3′UTR of SMAD2 that could directly bind to miR-210-3p. Alternatively, miR-210-3p may not bind directly to the 3′UTR of SMAD2 but may indirectly inhibit SMAD2 by suppressing the activity of the TGF signaling pathway, thus facilitating erythroid differentiation.

In conclusion, the present data suggested that, under hypoxia, GATA-1 overexpression significantly promoted erythroid differentiation, possibly by modulating miR-210-3p expression. Furthermore, the expression level of miR-210-3p increased with the degree of differentiation as it regulated several erythroid differentiation-related genes. Thus, with increasing information regarding miRNA profiles and transcription factor regulation, integrating these data may improve the current understanding of the molecular mechanisms underlying human adaptation and pathophysiology under hypoxic conditions.

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (grant no. 81760333), the Basic Research (Application) Project of the QingHai Science and Technology Department (grant no. 2017-ZJ-722) and the Scientific Research Fund for Young Scientist (grant no. 2019-kty-3). The funding sources helped in the design of the study. First, when applying for funding for a project, members of the foundation reviewed the feasibility of the project and proposed suggestions for modification. Second, the foundation conducted regular inspections as the project progressed.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

CH and LF conceptualized and designed this study. CH and LF are responsible for confirming the authenticity of the raw data. CY, CF, YY and JD acquired the data. CH, CF, JD, TL and SW analyzed and interpreted the data. CH drafted the manuscript, and CH, YY and LF edited the original draft. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

miRNAs/miRs

microRNAs

NC

negative control

RT-qPCR

reverse transcription-quantitative PCR

GATA-1

GATA binding protein 1

References

1 

Kerenyi MA and Orkin SH: Networking erythropoiesis. J Exp Med. 207:2537–2541. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Haase VH: Hypoxic regulation of erythropoiesis and iron metabolism. Am J Physiol Ren Physiol. 299:F1–F13. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Narayan AD, Ersek A, Campbell TA, Colón DM, Pixley JS and Zanjani ED: The effect of hypoxia and stem cell source on haemoglobin switching. Br J Haematol. 128:562–570. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Rogers HM, Yu X, Wen J, Smith R, Fibach E and Noguchi CT: Hypoxia alters progression of the erythroid program. Exp Hematol. 36:17–27. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Vlaski M, Lafarge X, Chevaleyre J, Duchez P, Boiron JM and Ivanovic Z: Low oxygen concentration as a general physiologic regulator of erythropoiesis beyond the EPO-related downstream tuning and a tool for the optimization of red blood cell production ex vivo. Exp Hematol. 37:573–584. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Ferreira R, Ohneda K, Yamamoto M and Philipsen S: GATA1 Function, a paradigm for transcription factors in hematopoiesis. Mol Cell Biol. 25:1215–1227. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Yu M, Riva L, Xie H, Schindler Y, Moran TB, Cheng Y, Yu D, Hardison R, Weiss MJ, Orkin SH, et al: Insights into GATA-1-mediated gene activation versus repression via genome-wide chromatin occupancy analysis. Mol Cell. 36:682–695. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Scherzer CR, Grass JA, Liao Z, Pepivani I, Zheng B, Eklund AC, Ney PA, Ng J, McGoldrick M, Mollenhauer B, et al: GATA transcription factors directly regulate the Parkinson's disease-linked gene alpha-synuclein. Proc Natl Acad Sci USA. 105:10907–10912. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Crispino JD: GATA1 in normal and malignant hematopoiesis. Semin Cell Dev Biol. 16:137–147. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Cantor AB and Orkin SH: Transcriptional regulation of erythropoiesis: an affair involving multiple partners. Oncogene. 21:3368–3376. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Dore LC, Amigo JD, Dos Santos CO, Zhang Z, Gai X, Tobias JW, Yu D, Klein AM, Dorman C, Wu W, et al: A GATA-1-regulated microRNA locus essential for erythropoiesis. Proc Natl Acad Sci USA. 105:3333–3338. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Nilsen TW: Mechanisms of microRNA-mediated gene regulation in animal cells. Trends Genet. 5:243–249. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Bianchi N, Zuccato C, Finotti A, Lampronti I, Borgatti M and Gambari R: Involvement of miRNA in erythroid differentiation. Epigenomics. 4:51–65. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Huang X, Le QT and Giaccia AJ: MiR-210-micromanager of the hypoxia pathway. Trends Mol Med. 16:230–237. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Kosaka N, Sugiura K, Yamamoto Y, Yoshioka Y, Miyazaki H, Komatsu N, Ochiya T and Kato T: Identification of erythropoietin-induced microRNAs in haematopoietic cells during erythroid differentiation. Br J Haematol. 142:293–300. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Rowley PT, Ohlsson-Wilhelm BM, Farley BA and LaBella S: Inducers of erythroid differentiation in K562 human leukemia cells. Exp Hematol. 9:32–37. 1981.PubMed/NCBI

17 

Gahmberg CG and Andersson LC: K562-a human leukemia cell line with erythroid features. Semin Hematol. 18:72–77. 1981.PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Mazan-Mamczarz K and Gartenhaus RB: Role of microRNA deregulation in the pathogenesis of diffuse large B-cell lymphoma (DLBCL). Leuk Res. 37:1420–1428. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, Ivey KN, Bruneau BG, Stainier DY and Srivastava D: miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 15:272–284. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Abend JR, Uldrick T and Ziegelbauer JM: Regulation of tumor necrosis factor-like weak inducer of apoptosis receptor protein (TWEAKR) expression by kaposi's sarcoma-associated herpesvirus MicroRNA prevents tweak-induced apoptosis and inflammatory cytokine expression. J Virol. 84:12139–12151. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Shivdasani RA: MicroRNAs: Regulators of gene expression and cell differentiation. Blood. 108:3646–3653. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Bapat A, Schippel N, Shi X, Jasbi P, Gu H, Kala M, Sertil A and Sharma S: Hypoxia promotes erythroid differentiation through the development of progenitors and proerythroblasts. Exp Hematol. 97:32–46.e35. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Kaelin WG Jr and Ratcliffe PJ: Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol Cell. 30:393–402. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Windsor JS and Rodway GW: Heights and haematology: The story of haemoglobin at altitude. Postgrad Med J. 83:148–151. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Noguchi CT and Rogers H: Hypoxia alters progression of the erythroid program. Exp Hematol. 2:163. 2007.

27 

Welch JJ, Watts JA, Vakoc CR, Yao Y, Wang H, Hardison RC, Blobel GA, Chodosh LA and Weiss MJ: Global regulation of erythroid gene expression by transcription factor GATA-1. Blood. 10:3136–3147. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Wu J, Zhou LQ, Yu W, Zhao ZG, Xie XM, Wang WT, Xiong J, Li M, Xue Z, Wang X, et al: PML4 facilitates erythroid differentiation by enhancing the transcriptional activity of GATA-1. Blood. 123:261–270. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Doss JF, Corcoran DL, Jima D, Telen MJ, Dave SS and Chi JT: A comprehensive joint analysis of the long and short RNA transcriptomes of human erythrocytes. BMC Genomics. 16:952. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Rasmussen KD, Simmini S, Abreugoodger C, Bartonicek N, Di Giacomo M, Bilbao-Cortes D, Horos R, Von Lindern M, Enright AJ and O'Carroll D: The miR-144/451 locus is required for erythroid homeostasis. J Exp Med. 207:1351–1358. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Undi RB, Kandi R and Gutti RK: MicroRNAs as haematopoiesis regulators. Adv Hematol. 2013:6957542013. View Article : Google Scholar : PubMed/NCBI

32 

Xie Y, Li W, Feng J, Wu T and Li J: MicroRNA-363 and GATA-1 are regulated by HIF-1α in K562 cells under hypoxia. Mol Med Rep. 14:2503–2510. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Raghuwanshi S, Karnati HK, Sarvothaman S, Gutti U, Saladi RGV, Tummala PR and Gutti RK: microRNAs: Key players in hematopoiesis. Adv Exp Med Bio. 887:171–211. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Sarakul O, Vattanaviboon P, Tanaka Y, Fucharoen S, Abe Y, Svasti S and Umemura T: Enhanced erythroid cell differentiation in hypoxic condition is in part contributed by miR-210. Blood Cells Mol Dis. 51:98–103. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Zhu Y, Wang D, Wang F, Li T, Dong L, Liu H, Ma Y, Jiang F, Yin H, Yan W, et al: A comprehensive analysis of GATA-1-regulated miRNAs reveals miR-23a to be a positive modulator of erythropoiesis. Nucleic Acids Res. 41:4129–4143. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Liu B, Sun J, Lei X, Zhu Z, Pei C and Qin L: MicroRNA-486-5p suppresses TGF-β2-induced proliferation, invasion and epithelial-mesenchymal transition of lens epithelial cells by targeting Smad2. J Biosci. 42:575–584. 2017. View Article : Google Scholar : PubMed/NCBI

37 

AlMegbel AM and Shuler CF: SMAD2 overexpression rescues the TGF-β3 null mutant mice cleft palate by increased apoptosis. Differentiation. 111:60–69. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Cheung KS, Sposito N, Stumpf PS, Wilson DI, Sanchez-Elsner T and Oreffo ROC: MicroRNA-146a regulates human foetal femur derived skeletal stem cell differentiation by down-regulating SMAD2 and SMAD3. PLoS One. 9:e980632014. View Article : Google Scholar : PubMed/NCBI

39 

Söderberg SS, Karlsson G and Karlsson S: Complex and context dependent regulation of hematopoiesis by TGF-β superfamily signaling. Ann N Y Acad Sci. 1176:55–69. 2009. View Article : Google Scholar

40 

Blank U and Karlsson S: The role of Smad signaling in hematopoiesis and translational hematology. Leukemia. 25:1379–1388. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Shav-Tal Y and Zipori D: The role of activin A in regulation of hemopoiesis. Stem Cells. 20:493–500. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Shiozaki M, Sakai R, Tabuchi M, Nakamura T, Sugino K, Sugino H and Eto Y: Evidence for the participation of endogenous activin A/erythroid differentiation factor in the regulation of erythropoiesis. Proc Natl Acad Sci USA. 89:1553–1556. 1992. View Article : Google Scholar : PubMed/NCBI

43 

Zhou L, Nguyen AN, Sohal D, Ma JY, Pahanish P, Gundabolu K, Hayman J, Chubak A, Mo Y, Bhagat TD, et al: Inhibition of the TGF-β receptor I kinase promotes hematopoiesis in MDS. Blood. 112:3434–3443. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Dussiot M, Maciel TT, Fricot A, Chartier C, Negre O, Veiga J, Grapton D, Paubelle E, Payen E, Beuzard Y, et al: An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia. Nat Med. 20:398–407. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Suragani RN, Cawley SM, Li R, Wallner S, Alexander MJ, Mulivor AW, Gardenghi S, Rivella S, Grinberg AV, Pearsall RS and Kumar R: Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine β-thalassemia. Blood. 123:3864–3872. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Suragani RN, Cadena SM, Cawley SM, Sako D, Mitchell D, Li R, Davies MV, Alexander MJ, Devine M, Loveday KS, et al: Transforming growth factor-β superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med. 20:408–414. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Martinez PA, Li R, Ramanathan HN, Bhasin M, Persall RS, Kumar R and Suragani RNVS: Smad2/3-pathway ligand trap luspatercept enhances erythroid differentiation in murine β-thalassaemia by increasing GATA-1 availability. J Cell Mol Med. 24:6162–6177. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Wu F, Weigel KJ, Zhou H and Wang XJ: Paradoxical roles of TGF-β signaling in suppressing and promoting squamous cell carcinoma. Acta Biochim Biophys Sin (Shanghai). 50:730. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Tannehill-Gregg SH, Kusewitt DF, Rosol TJ and Weinstein M: The roles of Smad2 and Smad3 in the development of chemically induced skin tumors in mice. Vet Pathol. 41:278–282. 2004. View Article : Google Scholar : PubMed/NCBI

50 

Phuah NH, Azmi MN, Awang K and Nagoor NH: Down-regulation of microRNA-210 confers sensitivity towards 1′s-1′-acetoxychavicol acetate (ACA) in cervical cancer cells by targeting SMAD4. Mol Cells. 40:291–298. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Mizuno Y, Tokuzawa Y, Ninomiya Y, Yagi K, Yatsuka-Kanesaki Y, Suda T, Fukuda T, Katagiri T, Kondoh Y, Amemiya T, et al: miR-210 promotes osteoblastic differentiation through inhibition of AcvR1b. FEBS Lett. 583:2263–2268. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Miyazono K, Maeda S and Imamura T: BMP receptor signaling: Transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev. 16:251–263. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Maeda S, Hayashi M, Komiya S, Imamura T and Miyazono K: Endogenous TGF-beta signaling suppresses maturation of osteoblastic mesenchymal cells. EMBO J. 23:552–563. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2021
Volume 24 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hu C, Yan Y, Fu C, Ding J, Li T, Wang S and Fang L: Effects of miR‑210‑3p on the erythroid differentiation of K562 cells under hypoxia. Mol Med Rep 24: 563, 2021
APA
Hu, C., Yan, Y., Fu, C., Ding, J., Li, T., Wang, S., & Fang, L. (2021). Effects of miR‑210‑3p on the erythroid differentiation of K562 cells under hypoxia. Molecular Medicine Reports, 24, 563. https://doi.org/10.3892/mmr.2021.12202
MLA
Hu, C., Yan, Y., Fu, C., Ding, J., Li, T., Wang, S., Fang, L."Effects of miR‑210‑3p on the erythroid differentiation of K562 cells under hypoxia". Molecular Medicine Reports 24.2 (2021): 563.
Chicago
Hu, C., Yan, Y., Fu, C., Ding, J., Li, T., Wang, S., Fang, L."Effects of miR‑210‑3p on the erythroid differentiation of K562 cells under hypoxia". Molecular Medicine Reports 24, no. 2 (2021): 563. https://doi.org/10.3892/mmr.2021.12202