Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Molecular Medicine Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1791-2997 Online ISSN: 1791-3004
Journal Cover
October-2025 Volume 32 Issue 4

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
October-2025 Volume 32 Issue 4

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML

  • Supplementary Files
    • Supplementary_Data1.pdf
    • Supplementary_Data2.pdf
Article Open Access

Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells

  • Authors:
    • Hikoto Sugiyama
    • Megumi Kikuchi
    • Mitsuru Chiba
    • Yoichiro Hosokawa
    • Satoru Monzen
  • View Affiliations / Copyright

    Affiliations: Department of Radiation Science, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036‑8564, Japan, Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036‑8564, Japan
    Copyright: © Sugiyama et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 280
    |
    Published online on: August 4, 2025
       https://doi.org/10.3892/mmr.2025.13645
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Patients with acute myeloid leukemia (AML) generally receive total body irradiation at a high‑dose rate for the ablation of bone marrow cells, including AML cells. However, in rare cases, radioresistant AML cells appear, interfering with the therapeutic effect. HL60 cells were used to model a radioresistant leukemia cell line that emerged from repeated radiation exposure (Res‑HL60). Notably, the mechanism through which microRNA (miRNA/miR) expression influences radioresistance in this model is unclear. In the current study, the expression profile of the miRNAs included in the small RNAs in Res‑HL60 was analyzed using an miRNA microarray. A total of 1,187 miRNAs were retained for analysis after normalization. Among them, 27 miRNAs (10 upregulated and 17 downregulated in Res‑HL60 cells compared with wild‑type‑HL60 cells) exhibited P<0.05 and fold change >1.5 or <0.66. Furthermore, the expression levels of five miRNAs were validated by reverse transcription‑quantitative PCR: miR‑146a‑5p (upregulated), and miR‑30c‑1‑3p, miR‑671‑5p, miR‑610 and miR‑3675‑5p (downregulated). To investigate the target mRNAs of these five miRNAs, OmicsNet (ver. 2.0) was used. A total of 27 mRNAs were identified as targets of these multiple miRNAs. Furthermore, Reactome analysis revealed enrichment in the following processes ‘Cell cycle, Mitotic’ (R‑HAS‑69278), ‘Apoptosis’ (R‑HAS‑109581) and ‘Immune system’ (R‑HAS‑168256), suggesting that these miRNAs regulate genes involved in these pathways. These findings indicated that the altered expression of five specific microRNAs in radioresistant AML cells may be associated with radioresistant conditions through the modulation of mRNA expression.

Introduction

Leukemia disrupts the homeostasis of the hematopoietic system and causes abnormal proliferation of blast cells (1). Acute myeloid leukemia (AML), including its M2 subtype, reduces survival in adults; therefore, establishing effective treatment strategies is critical for improving prognosis (2,3). Chemotherapy, such as the ‘7+3’ regimen, remains the standard first-line treatment for AML. However, in some relapsed or refractory cases, total body irradiation (TBI) is used as part of conditioning regimens before hematopoietic stem cell transplantation to eradicate residual leukemic cells (4,5). Repeated exposure to radiation in such settings may, in rare cases, contribute to the emergence of leukemic cells with reduced radiosensitivity, potentially impeding complete remission. Although the concept of ‘radiation-resistant AML’ is not widely recognized in clinical settings, where resistance is more commonly associated with chemotherapy, TBI has been implicated as a contributing factor in therapy-related AML (6). Furthermore, a phase II clinical trial evaluating treosulfan, fludarabine, and low-dose TBI in pediatric and young adult AML/MDS patients highlighted the clinical relevance and complex outcomes of TBI-based regimens, including relapse and non-relapse mortality (7). While radiation resistance is not a routine concern in AML treatment, these findings support the relevance of exploring the molecular basis of radiation response.

In the present study, we employed an in vitro AML model to investigate the acquisition of radioresistance. Specifically, we previously established a radioresistant AML cell line (Res-HL60) derived from HL60 cells, an AML M2 subtype cell line of human origin, and investigated the molecular mechanisms underlying radioresistance. Through repeated irradiation, the Res-HL60 cells acquired smaller morphology, increased proliferative potency, and showed elevated expression of CD38 cell surface antigen and several mRNAs (8–12). However, the upstream regulatory mechanism governing these gene expression changes remains unclear, necessitating further investigation.

MicroRNAs (miRNAs), a class of noncoding RNAs that regulate mRNA translation, have been implicated in cancer progression and cell behavior, and their expression patterns are increasingly recognized as diagnostic and prognostic biomarkers (13,14). Additionally, miRNAs have emerged as potential therapeutic targets in cancer. Despite this, the profile of miRNA expression in radioresistant AML cells such as Res-HL60 has not been previously reported. Given that chromosomal abnormalities, epigenetic dysregulation, and impaired miRNA biogenesis, potentially triggered by frequent radiation exposure, can all influence miRNA expression (15), investigating these changes may help clarify the molecular basis of acquired radioresistance. Therefore, in this study, we performed transcriptome analysis focusing on miRNA expression in Res-HL60 cells to explore mechanisms associated with radioresistance in AML.

Materials and methods

Leukemia cell line and culture

The human AML cell line HL60 (wild-type: Wt-HL60) was obtained from the RIKEN BioResource Center. The Res-HL60 cell line was generated by exposing Wi-HL60 cells to 4-Gy irradiation per week for 4 weeks. Wt-HL60 and Res-HL60 cells were cultured in RPMI-1640 medium (Thermo-Fisher Scientific, Inc.) supplemented with 10% heat-inactivated fetal bovine serum (Japan Bioserum, Inc.) and 1% penicillin/streptomycin (Thermo-Fisher Scientific, Inc.) in a humidified atmosphere containing 5% CO2 at 37°C. In this study, the control group was defined as non-irradiated Wt-HL60 cells cultured under these standard conditions (5% CO2 and 95% air). The characteristics of Res-HL60 cells, including higher proliferative capacity, smaller cell size and altered chemical reactivity, have been reported previously by our group (8–12). In the present study, a time-course analysis of morphological changes in Res-HL60 cells was conducted (Fig. 1). Cells were initially seeded at a density of 2×105 cells/ml on Day-1. X-ray irradiation was performed on Day 0. On Day 2 (i.e., the third day after initial seeding), cells were passaged and reseeded at 2×105 cells/ml. When the cell density remained below 2×105 cells/ml, only medium replacement was performed. No additional supplements (e.g., non-essential amino acids, sodium pyruvate) were added. To calculate the cumulative total viable cell number including all passages, the following formula was used: Cumulative cell number (/ml)=A × [(2×105) + B]/(2×105), where ‘A’ is the viable cell number (cells/ml) measured up to Day 2 (before passaging), and ‘B’ is the net increase in viable cell number (cells/ml) from Day 3 onward, relative to the reseeded density (2×105 cells/ml). B=0 for Days 0–2. This method allowed us to reflect the overall proliferation dynamics across reseeding and media replacement steps. The updated y-axis label in Fig. 2A and B reflects this definition. To confirm the identity and authenticity of the cell lines used in this study, short tandem repeat (STR) profiling was performed. The STR profiles of both Wt-HL60 and Res-HL60 showed a perfect match with the reference profile for HL60 (Cellosaurus ID: CVCL_0002, URL: http://www.cellosaurus.org/CVCL_0002), as provided by the RIKEN BioResource Center, indicating no evidence of contamination or misidentification. The STR profiles are provided in Fig. S1 and Table SI.

Characterization of Res-HL60 cell.
(A) Cellular morphology and subpopulation characteristics were
analyzed using flow cytometry. The horizontal and lateral axis
indicate the mean size of the cell (EV) and the intracellular
complexity (SS), respectively. (B) The EV histogram of each type of
cell. (C) The time course of the CD38 expression intensity and (D)
dot plots on the CD38-CD45 cell surface antigen after the exposure
of 4-Gy X-ray is shown. Non-irradiated HL60 cells cultured under
standard normoxic conditions (5% CO2 and 95% air) were
used as the control group in all analyses. Wt, wild type; Res,
resistant; EV, electronic volume; SS, side scatter.

Figure 1.

Characterization of Res-HL60 cell. (A) Cellular morphology and subpopulation characteristics were analyzed using flow cytometry. The horizontal and lateral axis indicate the mean size of the cell (EV) and the intracellular complexity (SS), respectively. (B) The EV histogram of each type of cell. (C) The time course of the CD38 expression intensity and (D) dot plots on the CD38-CD45 cell surface antigen after the exposure of 4-Gy X-ray is shown. Non-irradiated HL60 cells cultured under standard normoxic conditions (5% CO2 and 95% air) were used as the control group in all analyses. Wt, wild type; Res, resistant; EV, electronic volume; SS, side scatter.

Cell proliferation curve and DNA
fragmentation analysis. (A) Cumulative proliferation of Wt-HL60 and
Res-HL60 cells under non-irradiated conditions. Cells were seeded
at 2×105 cells/ml on Day-1 and cultured without
irradiation. On Day 2, the cells were passaged to maintain the same
seeding density. If cell density remained below this threshold,
only the medium was replaced without passaging. The cumulative
number of viable cells was calculated using the formula: Cumulative
cell number (/ml)=A × [(2×105) + B]/(2×105),
where ‘A’ is the viable cell number up to Day 2, and ‘B’ is the net
increase from Day 3 onward relative to the reseeding density. (B)
Cumulative proliferation of Wt-HL60 and Res-HL60 cells under 4-Gy
irradiated conditions. The same seeding and passaging protocol as
in (A) was followed, except that the cells were exposed to 4 Gy of
X-ray irradiation on Day 0. Cell proliferation was then monitored
to assess the impact of irradiation on the growth of wild-type and
resistant HL60 cells. Cumulative viable cell numbers were
calculated using the same formula as in (A). (C) Representative
images of the comet assay showing DNA damage 24 h after X-ray
exposure at doses of 1–4 Gy. (D) Quantification of DNA damage based
on tail moment calculated from the comet assay shown in (C). Data
are shown as the mean ± standard error of the mean from four
independent experiments. *P<0.05, **P<0.01 vs. Wt-HL60,
determined by unpaired Student's t-test. Wt, wild type; Res,
resistant.

Figure 2.

Cell proliferation curve and DNA fragmentation analysis. (A) Cumulative proliferation of Wt-HL60 and Res-HL60 cells under non-irradiated conditions. Cells were seeded at 2×105 cells/ml on Day-1 and cultured without irradiation. On Day 2, the cells were passaged to maintain the same seeding density. If cell density remained below this threshold, only the medium was replaced without passaging. The cumulative number of viable cells was calculated using the formula: Cumulative cell number (/ml)=A × [(2×105) + B]/(2×105), where ‘A’ is the viable cell number up to Day 2, and ‘B’ is the net increase from Day 3 onward relative to the reseeding density. (B) Cumulative proliferation of Wt-HL60 and Res-HL60 cells under 4-Gy irradiated conditions. The same seeding and passaging protocol as in (A) was followed, except that the cells were exposed to 4 Gy of X-ray irradiation on Day 0. Cell proliferation was then monitored to assess the impact of irradiation on the growth of wild-type and resistant HL60 cells. Cumulative viable cell numbers were calculated using the same formula as in (A). (C) Representative images of the comet assay showing DNA damage 24 h after X-ray exposure at doses of 1–4 Gy. (D) Quantification of DNA damage based on tail moment calculated from the comet assay shown in (C). Data are shown as the mean ± standard error of the mean from four independent experiments. *P<0.05, **P<0.01 vs. Wt-HL60, determined by unpaired Student's t-test. Wt, wild type; Res, resistant.

Irradiation

X-ray irradiation (150 kVp, 20 mA with 0.5-mm aluminum and 0.3-mm copper filters) was performed using an X-ray generator (MBR-1520R-3; Hitachi Medical Co., Ltd.) at a distance of 45 cm between the focus and target. The dose was monitored during irradiation using a thimble ionization chamber set next to the sample. The dose rate was 1 Gy/min. The cultured cells were exposed to X-rays as previously described (8–12).

Flow cytometry analysis

Flow cytometry was performed using Cell Lab Quanta™ Sc MPL system (Beckman Colter Immunotech) to assess cell size, granularity, and CD38 surface expression. Unlike conventional flow cytometers that use forward scatter to estimate cell size, the Cell Lab Quanta utilizes Electric Volume (EV), which is based on the Coulter principle, to directly measure cell size via changes in electrical resistance as cells pass through an aperture. This method allows for accurate assessment of cell volume independent of optical scattering. Granularity, representing internal cellular complexity, was evaluated based on side scatter (SSC) intensity, as is standard in flow cytometry. CD38 expression was also assessed due to its dual relevance as both a hematopoietic differentiation marker and a potential indicator of acquired radioresistance. CD38 is a well-characterized surface antigen known to be markedly upregulated during granulocytic differentiation of HL60 cells (16). Furthermore, our previous studies demonstrated sustained CD38 expression in Res-HL60 cells even in the absence of differentiation-inducing agents, suggesting a role in the radioresistant phenotype (8–12). Based on these findings, CD38 was utilized in this study both as a marker of differentiation and as a surrogate indicator of radiation resistance.

DNA fragmentation

The Comet Assay Single Cell Gel Electrophoresis Assay (cat. no. 4250-050-K; TREVIGEN, Inc., Gaithersburg, USA) was used to evaluate the DNA fragments of the cells irradiated with X-rays. One day after seeding in a 60-mmφ culture dish at a concentration of 1.0×105 cells/ml, the cells were irradiated with 1–4 Gy of X-rays, and the cells were collected after 24 h. The collected cells were fixed on glass slide (Matsunami Grass Inc., Osaka, Japan) using LM Agarose (TREVIGEN Inc.). The glass slide to which the sample was attached was immersed in an alkaline solution [200-mM NaOH, 1-mM ethylenediaminetetraacetic acid (EDTA)], and electrophoresis (21 V, 3 A, 30 min, 4°C) was performed. After washing the glass slide with distilled water and 70% EtOH and drying, it was stained with 100 µl of a staining solution containing a mixture of SYBR Green (TREVIGEN Inc.) and TE Buffer (10-mM Tris, 1-mM EDTA). After drying, the tail moment was observed using a fluorescence microscope (OLYMPUS Inc.). Images were taken using Comet Assay IV (Instem, Conshohocken, USA) for observation purposes. Tail moment was calculated using the formula: Tail Moment=Tail Length × % Tail DNA/100, using Comet Assay IV software. This calculation reflects both the extent and the intensity of DNA fragmentation in individual cells.

Extraction of total RNAs from cells

Total intracellular RNAs (four replicates performed in parallel) were extracted using ISOGEN II (Nippon Gene Co., Ltd.) according to the manufacturer's instructions. The purity and concentration of extracellular RNAs were assessed using a NanoDrop spectrophotometer (NanoDrop Technologies; Thermo-Fisher Scientific, Inc.). The RNA samples had 260/280-nm absorbance ratios of 1.8–2.0. Furthermore, the peaks of the small RNAs were confirmed using an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc.).

Microarray analysis of miRNAs

SurePrint G3 human miRNA 8×60-K Microarray (Agilent Technologies Inc.) was used to analyze intracellular microRNA expression. Cyanine 3 labeling was performed with 1 ng of intracellular microRNA using an miRNA Complete Labeling and Hybridization kit (Agilent Technologies Inc.), following the manufacturer's instructions. After hybridization and washing of the microarray slide, fluorescence image scanning was performed using a SureScan G 2600 D (Agilent Technologies Inc.). The expression data were processed using GeneSpring GX14.5 (Agilent Technologies, Inc.) to normalize the quantiles of all values in the respective microarrays. The obtained microarray data were registered with the Gene Expression Omnibus (GEO) (GSE285934).

Reverse transcription-quantitative PCR (RT-qPCR) analysis

First-strand cDNA was synthesized using Mir-X™ miRNA First-Strand Synthesis Kit (Takara Bio Inc., Otsu, Japan; Cat. Nos. 638313) according to the manufacturer's protocol (Protocol No. PT5001-1). MiRNA expression was assessed by qPCR using TB Green Advantage qPCR Premix (Takara Bio Inc., Cat. Nos. 639676) on a SmartCycler® II system (Takara Bio Inc.). The thermocycling conditions were as follows: 95°C for 10 sec, followed by 40 cycles of 95°C for 5 sec and 60°C for 20 sec. Relative miRNA expression levels were calculated using the 2−ΔΔCq method. U6 small nuclear RNA, included as an internal control primer set in the Mir-X™ First-Strand Synthesis Kit (Takara Bio Inc., Cat. Nos. 638313), was used for normalization. The exact primer sequences for U6 were not publicly disclosed, and direct inquiries to the manufacturer did not yield this information. However, the Mir-X™ system is widely used and has been independently validated in numerous peer-reviewed studies for reliable miRNA quantification. We adhered strictly to the manufacturer's standardized protocol to ensure data reproducibility and accuracy. For the analysis ΔCq was calculated as (Cq of target miRNA-Cq of U6). ΔΔCq was defined as (ΔCq of Res-HL60-ΔCq of Wt-HL60), and fold change was calculated as 2−ΔΔCq, with Wt-HL60 set as the baseline (fold change-1) (17). Sequences of the target miRNAs were obtained from the miRBase database (https://www.mirbase.org/). MiRNA-specific forward primers were synthesized by Eurofins Genomics Inc. and are listed in Table I. A universal reverse primer provided in the Mir-X™ kit was used for all miRNAs. The Mir-X™ qRT-PCR system employs a poly(A)-tailing strategy followed by reverse transcription with an adapter-anchored oligo(dT) primer. This approach selectively amplifies mature polyadenylated miRNAs, while excluding pre-miRNAs, long non-coding RNAs, and host transcripts lacking poly(A) tails. The specificity of the amplification was further supported by the observation of single, sharp melt curve peaks in all reactions.

Table I.

Sequences of miRNA primers for reverse transcription-quantitative PCR.

Table I.

Sequences of miRNA primers for reverse transcription-quantitative PCR.

Primer nameAccession no.Sequence, 5′-3′
hsa-miR-3612MIMAT0017989 AGGAGGCATCTTGAGAAATGGA
hsa-miR-324-5pMIMAT0000761 CGCATCCCCTAGGGCATTGGTG
hsa-miR-146a-5pMIMAT0000449 TGAGAACTGAATTCCATGGGTT
hsa-miR-1246MIMAT0005898 AATGGATTTTTGGAGCAGG
hsa-miR-3190-3pMIMAT0022839 TGTGGAAGGTAGACGGCCAGAGA
hsa-miR-3156-5pMIMAT0015030 AAAGATCTGGAAGTGGGAGACA
hsa-miR-124-3pMIMAT0000422 TAAGGCACGCGGTGAATGCCAA
hsa-miR-9-3pMIMAT0000442 ATAAAGCTAGATAACCGAAAGT
hsa-miR-324-3pMIMAT0000762 CCCACTGCCCCAGGTGCTGCTGG
hsa-let-7e-5pMIMAT0000066 TGAGGTAGGAGGTTGTATAGTT
hsa-miR-3669MIMAT0018092 TACGGAATATGTATACGGAATAT
hsa-miR-29b-3pMIMAT0000100 TAGCACCATTTGAAATCAGTGTT
hsa-miR-30c-1-3pMIMAT0004674 CTGGGAGAGGGTTGTTTACTCC
hsa-miR-671-5pMIMAT0003880 AGGAAGCCCTGGAGGGGCTGGAG
hsa-miR-425-5pMIMAT0003393 AATGACACGATCACTCCCGTTGA
hsa-miR-345-5pMIMAT0000772 GCTGACTCCTAGTCCAGGGCTC
hsa-miR-610MIMAT0003278 TGAGCTAAATGTGTGCTGGGA
hsa-miR-4260MIMAT0016881 CTTGGGGCATGGAGTCCCA
hsa-miR-376c-3pMIMAT0000720 AACATAGAGGAAATTCCACGT
hsa-miR-512-3pMIMAT0002823 AAGTGCTGTCATAGCTGAGGTC
hsa-miR-744-3pMIMAT0004946 CTGTTGCCACTAACCTCAACCT
hsa-miR-27b-3pMIMAT0000419 TTCACAGTGGCTAAGTTCTGC
hsa-miR-7-2-3pMIMAT0004554 CAACAAATCCCAGTCTACCTAA
hsa-miR-769-3pMIMAT0003887 CTGGGATCTCCGGGGTCTTGGTT
hsa-miR-3675-5pMIMAT0018098 TATGGGGCTTCTGTAGAGATTTC
hsa-miR-101-5pMIMAT0004513 CAGTTATCACAGTGCTGATGCT
hsa-miR-20b-5pMIMAT0001413 CAAAGTGCTCATAGTGCAGGTAG

[i] miR, microRNA.

Statistical analysis

Statistical analyses were performed using the Origin software package (Pro version 9.0; OriginLab Corporation, Northampton, MA, USA) and Microsoft Excel (Microsoft Office 365). To evaluate the statistical significance of differences between the control and experimental groups, an unpaired Student's t-test was used, as the data met assumptions for parametric testing. All results are presented as the mean ± SEM based on four independent experiments. P<0.05 was considered to indicate a statistically significant difference.

Results

Analysis of cell damage exposed to IR

The cell population exposed to 4-Gy radiation temporarily disappeared but reappeared on days 15–16 (Fig. 1A). However, it was confirmed that Res-HL60 had a decreased EV (Wt: 634.24, Res: 620.43) (Fig. 1B). Furthermore, the expression of the CD38 cell surface antigen in Res-HL60 cells was significantly increased after radiation compared with that in the wild-type cells (maximum on day 1: Res=7.55±0.53, Wt=3.54±0.89), confirming a similar previously reported response (Fig. 1C and D). In our previous study, we evaluated CD38 as a differentiation marker and found that its expression was consistently elevated in Res-HL60 cells (8). The Res-HL60 cells seeded at 1×105 cells/ml exhibited approximately 30-fold proliferation (3.1±0.4×105 cells/ml) on day 5, and the Wt-HL60 cells exhibited approximately 15-fold proliferation (1.6±0.1×105 cells/ml) (Fig. 2A). The 4-Gy irradiation condition of Res-HL60 cells was also maintained approximately 5-fold after exposure on day 4 (4.9±0.5×105 cells/ml); however, it remained at the level observed on day 0 in Wt-HL60 cells (1.9±0.1×105 cells/ml) (Fig. 2B). The extent of DNA damage caused by radiation exposure in these cells was determined using a comet assay (Fig. 2C). The tail moment of Res-HL60 cells irradiated with 1–4 Gy was similar to that observed in cells without irradiation, whereas the tail moment of Wt-HL60 cells exposed to irradiation was significantly increased (Fig. 2D).

Expression profiles of miRs

To clarify the miRNA expression profiles of Res-HL60 cells, total RNAs were extracted from these cells. Using an Agilent bioanalyzer, the extracted total RNAs were confirmed to have an RIN value (max-10) and rRNA 18s and 28s peaks, which reveal whether small RNAs, including miRNAs, are of sufficient concentration and quality for analysis (Fig. 3). Total RNAs extracted from each cell exhibited an RIN >9 in all samples. Furthermore, the peak of small RNAs, 18s, and 28s in the histogram was cleared, and these samples were considered to have good quality for RNA analysis (Table II).

Histogram of total RNA detection from
Res-HL60 and Wt-HL60 cells. Total RNA was detected in each cell
using an Agilent 2100 bioanalyzer. Wt, wild type; Res,
resistant.

Figure 3.

Histogram of total RNA detection from Res-HL60 and Wt-HL60 cells. Total RNA was detected in each cell using an Agilent 2100 bioanalyzer. Wt, wild type; Res, resistant.

Table II.

Concentration and quality of total RNA extracted from Res-HL60 cells and Wt-HL60 cells.

Table II.

Concentration and quality of total RNA extracted from Res-HL60 cells and Wt-HL60 cells.

Sample listConcentration of total RNA, µg/106 cellsRNA integrity numberRatio of rRNA, 28s/18s
Wt
  Replication 13.69.51.03
  Replication 25.69.50.98
  Replication 36.59.61.16
  Replication 44.49.10.84
  Mean ± SE5.0±0.69.4±0.11.0±0.1
Res
  Repliction 15.59.40.98
  Replication 24.39.61.18
  Replication 33.69.20.76
  Replication 44.19.61.13
  Mean ± SE4.4±0.39.5±0.11.0±0.1

[i] rRNA, ribosomal RNA; Wt, wild type; Res, resistant; SE, standard error.

After miRNA microarray analysis, data were extracted after quantile normalization, excluding under background, and collection of unsaturated signals (Fig. 4A). After processing, the signal values obtained using multiple types of detection probes were averaged for each miRNA name, yielding 1,187 miRNA data. Among the miRNAs that significantly increased or decreased in Res compared with Wt, 10 miRNAs had a fold change (Res/Wt) >1.5, and 17 miRNAs had a fold change (Res/Wt) <0.66 (Fig. 4B-D). For the 27 miRNAs that exhibited expression changes in the microarray analysis, a functional enrichment analysis using the Reactome Pathway Database in OmicsNet 2.0 was performed to estimate the associated mRNA expression pathways. Cellular functional changes were observed in the following pathways: ‘Immune System (R-HAS-168256)’, ‘Signaling by PGDF (R-HAS-186797)’, ‘Adaptive Immune System (R-HAS-1280218)’, ‘Signaling by EGFR in Cancer (R-HAS-1643713)’, ‘Apoptosis (R-HAS-109581)’, and ‘Cell Cycle, Mitotic (R-HAS-69278)’ for tumor growth and/or invasion (Fig. 5A).

miRNA microarray analysis. (A)
Statistical analysis for normalization, and the (B) relationship
between fold change and P-value. (C) Significant upregulated miRNAs
(P<0.05 by Student's t-test, >1.5 fold change), and (D)
significant downregulated miRNAs (P<0.05 by Student's t-test,
<0.66 fold change). The data are presented as the mean ±
standard error of the mean of four experiments. miRNA/miR,
microRNA; Wt, wild type; Res, resistant.

Figure 4.

miRNA microarray analysis. (A) Statistical analysis for normalization, and the (B) relationship between fold change and P-value. (C) Significant upregulated miRNAs (P<0.05 by Student's t-test, >1.5 fold change), and (D) significant downregulated miRNAs (P<0.05 by Student's t-test, <0.66 fold change). The data are presented as the mean ± standard error of the mean of four experiments. miRNA/miR, microRNA; Wt, wild type; Res, resistant.

Top 20 enriched Reactome pathways
associated with predicted target genes of differentially expressed
miRNAs. Pathway enrichment analysis was conducted using the
Reactome Pathway Database to categorize the predicted target genes
of significantly altered miRNAs into curated biological pathways.
Reactome pathway identifiers are shown in parentheses. Bar graphs
show the top 20 enriched pathways ranked by statistical
significance for the predicted target genes of (A) all 27
differentially expressed miRNAs, (B) 10 upregulated miRNAs and (C)
17 downregulated miRNAs. miRNA, microRNA.

Figure 5.

Top 20 enriched Reactome pathways associated with predicted target genes of differentially expressed miRNAs. Pathway enrichment analysis was conducted using the Reactome Pathway Database to categorize the predicted target genes of significantly altered miRNAs into curated biological pathways. Reactome pathway identifiers are shown in parentheses. Bar graphs show the top 20 enriched pathways ranked by statistical significance for the predicted target genes of (A) all 27 differentially expressed miRNAs, (B) 10 upregulated miRNAs and (C) 17 downregulated miRNAs. miRNA, microRNA.

Furthermore, when we focused on the functional analysis of the target mRNAs of the 10 upregulated miRNAs in Res-HL60 cells, these mRNAs were strongly related to the following functional pathways: ‘cell cycle’, ‘cell division’ and ‘apoptosis’ (Fig. 5B). In contrast, the target mRNAs of the 16 downregulated miRNAs in Res-HL60 cells were strongly related to the following functional pathways: ‘Signaling by PDGF’ and ‘Assembly of collagen fibrils and other multimeric structures (R-HAS-2022090)’ (Fig. 5C).

Validation of the expression of 27 miRNAs

The significant expression of the 27 miRNAs obtained from these microarray analysis signals was reconfirmed by RT-qPCR. Thus, the expression of miRNA-146a-5p was significantly increased, whereas the expression of miRNA-30c-1-3p, miRNA-671-5p, miRNA-610, and miRNA-3675-5p was significantly decreased (Fig. 6).

Validation analysis of the 27 focused
miRNAs by RT-qPCR. (A) Upregulated miRNA and (B) downregulated
miRNA group were validated by RT-qPCR. The quantitation by
fluorescence signal was performed using the ΔΔCq method. U6 small
nuclear RNA was used as the internal control. Fold change was
calculated by comparing Res-HL60 cells to Wt-HL60 cells after
normalization to U6 expression. Expression values in Wt-HL60 were
set to 1, and fold change values in Res-HL60 are shown relative to
this baseline (horizontal line). The data are presented as the mean
± standard error of the mean of four experiments. **P<0.01,
*P<0.05 by unpaired Student's t-test. RT-qPCR, reverse
transcription-quantitative PCR; miRNA/miR, microRNA; Wt, wild type;
Res, resistant.

Figure 6.

Validation analysis of the 27 focused miRNAs by RT-qPCR. (A) Upregulated miRNA and (B) downregulated miRNA group were validated by RT-qPCR. The quantitation by fluorescence signal was performed using the ΔΔCq method. U6 small nuclear RNA was used as the internal control. Fold change was calculated by comparing Res-HL60 cells to Wt-HL60 cells after normalization to U6 expression. Expression values in Wt-HL60 were set to 1, and fold change values in Res-HL60 are shown relative to this baseline (horizontal line). The data are presented as the mean ± standard error of the mean of four experiments. **P<0.01, *P<0.05 by unpaired Student's t-test. RT-qPCR, reverse transcription-quantitative PCR; miRNA/miR, microRNA; Wt, wild type; Res, resistant.

Analysis of bioinformatics: The mRNA networks controlled by the five reproduced miRNAs were analyzed using OmicsNet 2.0. In this network, 28 mRNAs were associated with two or more miRNAs (Fig. 7). Of these 28 target mRNAs, seven (i.e., ALG10B, CCDC6, CCND2, CPM, CYBRD1, NACC1, and UMPS) were targeted by miRNA-146a-5p, which is an upregulated miRNA, and the other four miRNAs, which are downregulated. Furthermore, three miRNAs (i.e., miRNA-610, miRNA-671-5p, and miRNA-30c-1-3p) targeted superoxide dismutase 2 (SOD2). In the Target Scan analysis, three miRNAs (miRNA-610, miRNA-671-5p, and miRNA-30c-1-3p) were predicted to bind to the 3′-UTR region of SOD2, which was predicted as the target mRNA (Fig. 8). Furthermore, two miRNAs (i.e., miRNA-671-5p and miRNA-30c-1-3p) were predicted to bind to the 3′-UTR region of MYH9 mRNA. The putative target sites of miR-610 and miR-30c-1-3p for CD38 mRNA and miR-3675-5p, miR-671-5p, and miR-30c-1-3p for ATM mRNA were located in the 3′-UTR region.

Predictive network analysis between
miRNA and its target mRNA using OmicsNet. OmicsNet was used to
predict the target genes. This study used the Reactome database of
OmicsNet to predict the target genes. Lines connect the miRNA
target genes. The figure shows green for miRNAs, blue for target
genes, and red for other related genes. miRNA/miR, microRNA.

Figure 7.

Predictive network analysis between miRNA and its target mRNA using OmicsNet. OmicsNet was used to predict the target genes. This study used the Reactome database of OmicsNet to predict the target genes. Lines connect the miRNA target genes. The figure shows green for miRNAs, blue for target genes, and red for other related genes. miRNA/miR, microRNA.

Target scan analysis in humans. The
images show a predictive region for adhesion by target mRNA on
3′UTR and miRNA. The data show (A) SOD2, (B) MYH9, (C) CD38 and (D)
ATM. miRNA/miR, microRNA; SOD2, superoxide dismutase 2.

Figure 8.

Target scan analysis in humans. The images show a predictive region for adhesion by target mRNA on 3′UTR and miRNA. The data show (A) SOD2, (B) MYH9, (C) CD38 and (D) ATM. miRNA/miR, microRNA; SOD2, superoxide dismutase 2.

Discussion

In this study, we analyzed the miRNA expression profile of a radioresistant AML cell model (Res-HL60) derived from the HL60 cell line, which belongs to the FAB M2 subtype of AML. We reconfirmed that Res-HL60 cells, which were established by repeated irradiation of HL60 cells, maintained the morphological characteristics previously described, even when remade (Fig. 1). The HL60 cell line, which is derived from a patient with AML, exhibits inherently low CD38 expression, as previously reported (18). We have previously evaluated CD38 as a differentiation marker and found that its expression was consistently elevated in Res-HL60 cells (8). In this study, we observed transient increases in CD38 expression immediately after irradiation. This upregulation has been reported to be associated with cellular activation and proliferation (19). CD38 was selected as the primary marker in this study because it is a well-established differentiation-associated antigen in the HL60 model system; its expression increases markedly during granulocytic differentiation, as demonstrated by Partida-Sánchez et al (16). In our Res-HL60 cells, CD38 consistently showed strong upregulation both under baseline conditions and immediately after irradiation. These observations, combined with CD38′s known roles in NAD+-dependent calcium signaling and immune modulation, made it a biologically relevant candidate for investigating radioresistant phenotypes in AML. Although Breitman et al broadly discussed the HL60 differentiation model, CD38-specific functional insights were established in later work (20). In this context, ‘cellular activation’ refers to an increased responsiveness of cells to external stimuli, such as radiation, typically accompanied by changes in surface markers, including CD38 upregulation. Although direct functional activation markers were not assessed in this study, similar CD38 upregulation has been correlated with cell activation and immune modulation in leukemia models. Our previously established Res-HL60 cells demonstrated enhanced proliferative capacity compared to Wt-HL60 cells, as evidenced by shortened population doubling times and increased viable cell counts (8–12). Although these proliferation characteristics were not re-evaluated in the present study, the phenotypic stability and CD38 expression patterns observed here are consistent with those findings.

Furthermore, DNA fragments did not increase in these cells due to radiation, and the response was confirmed to be similar to γ-H2AX, a marker for DNA double-strand breaks, and to previous DNA ladder analyses (9,21). In these damage responses in Res-HL60 cells, 27 miRNAs (10 upregulated and 17 downregulated) showed altered expression in the microarray analysis. Among them, five miRNAs (miR-146a-5p, miR-30c-1-3p, miR-671-5p, miR-610, and miR-3675-5p) consistently demonstrated significant changes in the same direction across both microarray and RT-qPCR validation experiments. This consistency suggests that these five miRNAs may play a regulatory role in modulating molecular pathways associated with the development of radiation resistance. These five miRNAs have been implicated in tumor proliferation, radiosensitivity, and hematopoiesis (22–30). miR-146a functions together with TRAF1 and IRAK1 as negative feedback factors for the two toll-like receptor pathway proteins (22). It is also involved in regulating the transcriptional activity of NF-κB, which is necessary for the differentiation of hematopoietic cells (23,24). In radiotherapy for meningioma, the expression of miR-30c-1-3p is suppressed when treatment resistance occurs (25). In another report, the suppression of miR-30c-1-3p expression was associated with the progression of prostate cancer (26). The overexpression of miR-671-5p has been shown to significantly increase apoptosis and DNA damage in human glioblastoma cells (27). In neuroblastoma cells, the overexpression of miR-610 induces apoptosis and reduces cell viability (28). miR-3651 promotes the proliferation of hepatocellular carcinoma and colorectal cancer cells (29,30). These reports from various cell types suggest that changes in miRNA expression in radioresistant AML are triggered by cell viability activity.

The analysis of linking target mRNA networks of five miRNAs with radioresistant AML is new information, and 11 target mRNAs of these miRs (i.e., ALG10B, CCDC6, CCND2, CPM, CYBRD1, NACC1, UMPS, SOD2, MYH9, CD38, and ATM) may regulate the expression of its associated proteins and transcription factors in a complex manner. SOD2 can scavenge active oxygen produced by radiation irradiation, and SOD2 has been reported to protect against cellular stress (31). Furthermore, MYH9 is involved in cell division, cell migration, and maintenance of cell morphology and acts as an antiapoptotic agent in head and neck cancers where MYH9 is highly expressed (32). Moreover, it also reduces intracellular reactive oxygen species levels through the activation of the MAPK signaling pathway and Nrf2/keap1 pathway and the promotion of the production of the antioxidant enzyme GCLC. It is also involved in radioresistance. This observation may also explain the morphological stability and resistance to radiation-induced apoptosis observed in Res-HL60 cells. To further investigate the clinical relevance of these target genes, we examined publicly available AML gene expression datasets from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). Yu et al have reported that MYH9 overexpression was significantly associated with poor prognosis in patients with AML based on TCGA-LAML data (33). This finding is also supported by GSE12417 (GEO), a dataset originally published by Metzeler et al (34), which includes cytogenetically normal AML cases. In a recent analysis by Zhai et al (35), MYH9 was among the genes linked to adverse outcomes in this cohort. Expression of ATM, although not frequently mutated in AML, has been implicated in treatment responses; low ATM expression enhances sensitivity to FLT3 inhibitors and may affect DNA damage signaling (36). SOD2 has not been directly studied in AML; however, it plays a key role in scavenging reactive oxygen species. A meta-analysis of pan-cancer TCGA datasets by Noh et al (31) reported that SOD2 upregulation is associated with treatment resistance, suggesting its potential role in radioresistant phenotypes. We note that original survival analyses were not performed in this study. Instead, we relied on published results from previous analyses of these public datasets. This approach was taken due to the experimental focus of our study on the HL60-derived model. However, we fully acknowledge the importance of such analyses in increasing the translational relevance of our findings. While we agree with the reviewer's suggestion to perform updated survival analyses using the most recent TCGA and GEO datasets, this was beyond the scope of the current in vitro study. Nonetheless, incorporating such analyses is a clear objective for future work. We plan to conduct comprehensive bioinformatics evaluations of MYH9, ATM, and SOD2 expression in larger patient cohorts using integrated clinical and molecular data. Furthermore, the use of radiation-resistant AML patient samples to validate these gene targets was not feasible in the present study due to sample unavailability. However, this represents a crucial next step. Future studies will aim to collect and analyze patient-derived AML samples, particularly from individuals with known radiation treatment history, to directly confirm the relevance of MYH9, ATM, and SOD2 in clinical radioresistance. These findings complement our in vitro results and support the relevance of SOD2, ATM, and MYH9 in AML radioresistance.

Most functions inferred from the Reactome analysis were related to anti-apoptotic processes and were associated with upregulated miR-146a-5p. Our original hypothesis was that miR-146a-5p promotes apoptosis by inhibiting antiapoptotic molecules, thereby enhancing radiosensitivity in Res-HL60 cells. However, the results of the inhibition of anti-apoptotic molecules did not support this hypothesis. Instead, these findings suggest that miR-146a-5p regulates an alternative pathway that could contribute to radiation resistance. Target-scan analysis revealed that the high expression of CD38 cell surface antigen protein, CD38 mRNA, and intracellular ATM is related to the regulation of the five miRNAs revealed here (Fig. 8). These miRNA regulations provide molecular biology information supporting previous cellular responses. This information enhances our understanding of the molecular mechanisms underlying radiation resistance in AML. To further elucidate the mechanisms linking CD38 expression and radiation resistance, our future studies will focus on dissecting the intracellular signaling pathways downstream of CD38. Specifically, we plan to investigate NAD+ metabolism and calcium signaling pathways, both of which are regulated by CD38 and are known to affect cell survival, DNA repair capacity, and oxidative stress responses. These pathways may provide crucial mechanistic links between CD38 expression and the observed radioresistant phenotype in AML cells. In addition, combining CD38 expression analysis with metabolic profiling may help clarify its functional relevance in therapeutic resistance. This approach may also reveal novel therapeutic targets to overcome resistance in CD38-expressing AML subtypes. Although CD38 was a useful marker in our initial evaluation, we recognized that other markers, such as CD34, CD123, and ALDH are also relevant in the context of leukemia stemness and therapy resistance. Exploring the role of these markers was not within the scope of this study. However, future studies incorporating a broader panel of surface markers are warranted to further elucidate the mechanisms of AML radioresistance.

One limitation of this study is the lack of a regulatory analysis for the expression of the five miRNAs in AML cells, including investigations of intracellular signaling pathways and knockdown of the 11 predicted target mRNAs. Another limitation is that all experiments were performed using a single cell line, HL60, and its derived radioresistant subline. Although HL60 is a widely used model for AML studies, it does not capture the full genetic and phenotypic heterogeneity of AML, which can significantly affect radiosensitivity and miRNA profiles. Therefore, caution should be exercised when generalizing these findings. Future studies should include additional AML cell lines representing different molecular subtypes, as well as patient-derived primary cells, to validate the observed miRNA and mRNA expression patterns. Furthermore, functional assays using miRNA mimics/inhibitors or siRNA-mediated knockdown of key targets such as MYH9, ATM, and SOD2 are necessary to directly confirm their roles in radiation resistance. Such validations will be crucial for establishing the broader applicability and clinical relevance of our findings. Clinically, the identified miRNAs and their targets (e.g., MYH9, ATM, SOD2) may serve as biomarkers for predicting radiotherapy resistance in AML. They may also provide novel therapeutic targets to overcome such resistance, enabling more personalized treatment strategies. Although our findings are currently preclinical, they lay important groundwork for future translational research aimed at improving AML patient outcomes.

In summary, further studies are warranted to clarify the role of these miRNA-mRNA networks in therapy-resistant leukemia. Future investigations employing integrative miRNA-mRNA analyses, as demonstrated here, may further elucidate the molecular basis of radiosensitivity in AML.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

This work was supported by JSPS KAKENHI, Grants-in-Aid for Scientific Research (B) (grant no. 21H02861/23K21419, to Satoru Monzen) and Takeda Science Foundation 2022 (to Satoru Monzen). The funders played no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Availability of data and materials

The sequencing data generated in the present study may be found in the GEO under accession number GSE285934 or at the following URL: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE285934. The other data generated in the present study may be requested from the corresponding author.

Authors' contributions

HS, YH and SM designed the study, prepared the manuscript draft and participated in the revision of the manuscript. HS, MK, MC and SM performed the biological analysis and collection of the experimental data. SM supervised the study, critically reviewed the manuscript and provided final approval for the version to be submitted and published. All authors have read and approved the final version of the manuscript. HS and SM confirm the authenticity of all the raw data.

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.

References

1 

Nemkov T, D'Alessandro A and Reisz JA: Metabolic underpinnings of leukemia pathology and treatment. Cancer Rep (Hoboken). 2:e11392019.PubMed/NCBI

2 

Shallis RM, Wang R, Davidoff A, Ma X and Zeidan AM: Epidemiology of acute myeloid leukemia: Recent progress and enduring challenges. Blood Rev. 36:70–87. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Wang ZY and Chen Z: Acute promyelocytic leukemia: From highly fatal to highly curable. Blood. 111:2505–2515. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Reske SN, Deisenhofer S, Glatting G, Zlatopolskiy BD, Morgenroth A, Vogg ATJ, Buck AK and Friesen C: 123I–ITdU-mediated nanoirradiation of DNA efficiently induces cell kill in HL60 leukemia cells and in doxorubicin-, beta-, or gamma-radiation-resistan cell lines. J Nucl Med. 48:1000–1007. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Godley LA and Larson RA: Therapy-related myeloid leukemia. Semin Oncol. 35:418–429. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Bhatia S: Therapy-related myelodysplasia and acute myeloid leukemia. Semin Oncol. 40:666–675. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Nemecek ER, Hilger RA, Adams A, Shaw BE, Kiefer D, Le-Rademacher J, Levine JE, Yanik G, Leung W, Talano JA, et al: Treosulfan, fludarabine, and low-dose total body irradiation for children and young adults with acute myeloid leukemia or myelodysplastic syndrome undergoing allogeneic hematopoietic cell transplantation: prospective phase II trial of the pediatric blood and marrow transplant consortium. Biol Blood Marrow Transplant. 24:1651–1656. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Monzen S, Takimura K, Kashiwakura I and Hosokawa Y: Acute promyelocytic leukemia mutated to radioresistance suppressed monocyte lineage differentiation by phorbol 12-myristate 13-acetate. Leuk Res. 37:1162–1169. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Hazawa M, Hosokawa Y, Monzen S, Yoshino H and Kashiwakura I: Regulation of DNA damage response and cell cycle in radiation-resistant HL60 myeloid leukemia cells. Oncol Rep. 28:55–61. 2012.PubMed/NCBI

10 

Monzen S, Chiba M, Ueno T, Morino Y, Terada K, Yamaya H and Hosokawa Y: A radioresistant fraction of acute promyelocytic leukemia cells exhibit CD38 cell-surface antigen and mRNA expression. Oncol Lett. 15:6709–6714. 2018.PubMed/NCBI

11 

Monzen S, Chiba M and Hosokawa Y: Genetic network profiles associated with established resistance to ionizing radiation in acute promyelocytic leukemia cells and their extracellular vesicles. Oncol Rep. 35:749–756. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Morino Y, Sugiyama H, Yamane K, Kikuchi M, Yamanaka T, Honda K and Monzen S: Additive antitumor effect of arsenic trioxide with exposure to ionizing radiation to human acute promyelocytic leukemia HL-60 cells. Oncol Rep. 52:1092024. View Article : Google Scholar : PubMed/NCBI

13 

He B, Zhao Z, Cai Q, Zhang Y, Zhang P, Shi S, Xie H, Peng X, Yin W, Tao Y and Wang X: miRNA-based biomarkers, therapies, and resistance in cancer. Int J Biol Sci. 16:2628–2647. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Sun Z, Shi K, Yang S, Liu J, Zhou Q, Wang G, Song J, Li Z, Zhang Z and Yuan W: Effect of exosomal miRNA on cancer biology and clinical applications. Mol Cancer. 17:1472018. View Article : Google Scholar : PubMed/NCBI

15 

Peng Y and Croce CM: The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 1:150042016. View Article : Google Scholar : PubMed/NCBI

16 

Partida-Sánchez S, Cockayne DA, Monard S, Jacobson EL, Oppenheimer N, Garvy B, Kusser K, Goodrich S, Howard M, Harmsen A, et al: Cyclic ADP-ribose production by CD38 regulates intracellular calcium release, extracellular calcium influx and chemotaxis in neutrophils and is required for bacterial clearance in vivo. Nat Med. 7:1209–1216. 2001. View Article : Google Scholar : PubMed/NCBI

17 

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. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Mouly E, Planquette C, Rousseau E and Delansorne R: Inecalcitol enhances daratumumab-induced antibody-dependent cell cytotoxicity towards multiple myeloma and acute myeloid leukemia cell lines. Blood. 132 (Suppl 1):S14472018. View Article : Google Scholar

19 

Zhong X and Ma H: Targeting CD38 for acute leukemia. Front Oncol. 12:10077832022. View Article : Google Scholar : PubMed/NCBI

20 

Breitman TR, Chen ZX and Takahashi N: Potential applications of cytodifferentiation therapy in hematologic malignancies. Semin Hematol. 31 (4 Suppl 5):S18–S25. 1994.

21 

Johansson P, Fasth A, Ek T and Hammarsten O: Validation of a flow cytometry-based detection of γ-H2AX, to measure DNA damage for clinical applications. Cytometry B Clin Cytom. 92:534–540. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Yuan W, Sun Q, Jiang Y, Zhang X, Chen L, Xie C, Qin F, Chen Y, Lv H, Chen W and Xiao Y: MiR-146a affects the alteration in myeloid differentiation induced by hydroquinone in human CD34+ hematopoietic progenitor cells and HL-60 cells. Toxicol Res (Camb). 5:848–858. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Vergani E, Dugo M, Cossa M, Frigerio S, Di Guardo L, Gallino G, Mattavelli I, Vergani B, Lalli L, Tamborini E, et al: miR-146a-5p impairs melanoma resistance to kinase inhibitors by targeting COX2 and regulating NFkB-mediated inflammatory mediators. Cell Commun Signal. 18:1562020. View Article : Google Scholar : PubMed/NCBI

24 

Tse AKW, Wan CK, Shen XL, Zhu GY, Cheung HY, Yang M and Fong WF: 1,25-dihydroxyvitamin D3 induces biphasic NF-kappaB responses during HL-60 leukemia cells differentiation through protein induction and PI3K/Akt-dependent phosphorylation/degradation of IkappaB. Exp Cell Res. 313:1722–1734. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Zhang X, Zhang G, Huang H, Li H, Lin S and Wang Y: Differentially expressed MicroRNAs in radioresistant and radiosensitive atypical meningioma: A clinical study in chinese patients. Front Oncol. 10:5012020. View Article : Google Scholar : PubMed/NCBI

26 

Chen W, Yao G and Zhou K: miR-103a-2-5p/miR-30c-1-3p inhibits the progression of prostate cancer resistance to androgen ablation therapy via targeting androgen receptor variant 7. J Cell Biochem. 120:14055–14064. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Lin JC, Kuo CY, Tsai JT and Liu WH: miR-671-5p inhibition by MSI1 promotes glioblastoma tumorigenesis via radioresistance, tumor motility and cancer stem-like cell properties. Biomedicines. 10:212021. View Article : Google Scholar : PubMed/NCBI

28 

Qiu X and Qin F: Retraction: FAM64A antagonizes tumor suppressive effects of miR-610 in neuroblastoma in vitro. J Neurosurg Sci. Apr 16–2021.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

29 

Liu Y, Hu L, Liu O, Ye J and Zhang J: miR-3651 participates in the growth cycle of hepatocellular carcinoma cells and promotes the malignant metastasis via the PI3K/AKT/mTOR signalling pathway. J Oncol. 2022:57449992022. View Article : Google Scholar : PubMed/NCBI

30 

Li C, Ding D, Gao Y and Li Y: MicroRNA-3651 promotes colorectal cancer cell proliferation through directly repressing T-box transcription factor 1. Int J Mol Med. 45:956–966. 2020.PubMed/NCBI

31 

Noh JK, Woo SR, Yun M, Lee MK, Kong M, Min S, Kim SI, Lee YC, Eun YG and Ko SG: SOD2- and NRF2-associated gene signature to predict radioresistance in head and neck cancer. Cancer Genomics Proteomics. 18:675–684. 2021. View Article : Google Scholar : PubMed/NCBI

32 

You GR, Chang JT, Li YL, Huang CW, Tsai YL, Fan KH, Kang CJ, Huang SF, Chang PH and Cheng AJ: MYH9 facilitates cell invasion and radioresistance in head and neck cancer via modulation of cellular ROS levels by activating the MAPK-Nrf2-GCLC pathway. Cells. 11:28552022. View Article : Google Scholar : PubMed/NCBI

33 

Yu M, Wang J, Zhu Z, Hu C, Ma Q, Li X, Yin X, Huang J, Zhang T, Ma Z, et al: Prognostic impact of MYH9 expression on patients with acute myeloid leukemia. Oncotarget. 8:156–163. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Metzeler KH, Hummel M, Bloomfield CD, Spiekermann K, Braess J, Sauerland MC, Heinecke A, Radmacher M, Marcucci G, Whitman SP, et al: An 86-probe-set gene-expression signature predicts survival in cytogenetically normal acute myeloid leukemia. Blood. 112:4193–4201. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Zhai Y, Shen H and Wei H: A comprehensive metabolism-related gene signature predicts the survival of patients with acute myeloid leukemia. Genes (Basel). 15:632023. View Article : Google Scholar : PubMed/NCBI

36 

Gregory MA, D'Alessandro A, Alvarez-Calderon F, Kim J, Nemkov T, Adane B, Rozhok AI, Kumar A, Kumar V, Pollyea DA, et al: ATM/G6PD-driven redox metabolism promotes FLT3 inhibitor resistance in acute myeloid leukemia. Proc Natl Acad Sci USA. 113:E6669–E6678. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Sugiyama H, Kikuchi M, Chiba M, Hosokawa Y and Monzen S: Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells. Mol Med Rep 32: 280, 2025.
APA
Sugiyama, H., Kikuchi, M., Chiba, M., Hosokawa, Y., & Monzen, S. (2025). Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells. Molecular Medicine Reports, 32, 280. https://doi.org/10.3892/mmr.2025.13645
MLA
Sugiyama, H., Kikuchi, M., Chiba, M., Hosokawa, Y., Monzen, S."Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells". Molecular Medicine Reports 32.4 (2025): 280.
Chicago
Sugiyama, H., Kikuchi, M., Chiba, M., Hosokawa, Y., Monzen, S."Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells". Molecular Medicine Reports 32, no. 4 (2025): 280. https://doi.org/10.3892/mmr.2025.13645
Copy and paste a formatted citation
x
Spandidos Publications style
Sugiyama H, Kikuchi M, Chiba M, Hosokawa Y and Monzen S: Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells. Mol Med Rep 32: 280, 2025.
APA
Sugiyama, H., Kikuchi, M., Chiba, M., Hosokawa, Y., & Monzen, S. (2025). Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells. Molecular Medicine Reports, 32, 280. https://doi.org/10.3892/mmr.2025.13645
MLA
Sugiyama, H., Kikuchi, M., Chiba, M., Hosokawa, Y., Monzen, S."Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells". Molecular Medicine Reports 32.4 (2025): 280.
Chicago
Sugiyama, H., Kikuchi, M., Chiba, M., Hosokawa, Y., Monzen, S."Relationship between specific microRNA expression and radioresistant conditions in HL60 acute myeloid leukemia cells". Molecular Medicine Reports 32, no. 4 (2025): 280. https://doi.org/10.3892/mmr.2025.13645
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team