Next Article in Journal
Relationship between Facial Color Changes and Psychological Problems Associated with Lower Back Pain
Previous Article in Journal
Effect of 9% Hydrofluoric Acid Gel Hot-Etching Surface Treatment on Shear Bond Strength of Resin Cements to Zirconia Ceramics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

miRNA Expression Associated with HbF in Saudi Sickle Cell Anemia

1
Department of Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
2
Department of Pathology, King Fahd Hospital of the University, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34445, Saudi Arabia
3
Ministry of Health, Qatif Central Hospital, Qatif 32654, Saudi Arabia
4
Department of Stem Cell Research, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
5
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
6
Department of Medicine, Division of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA 02118, USA
*
Author to whom correspondence should be addressed.
Medicina 2022, 58(10), 1470; https://doi.org/10.3390/medicina58101470
Submission received: 11 September 2022 / Revised: 8 October 2022 / Accepted: 12 October 2022 / Published: 17 October 2022
(This article belongs to the Section Pharmacology)

Abstract

:
Background and Objectives: Sickle cell anemia (SCA) is a hereditary monogenic disease due to a single β-globin gene mutation that codes for the production of sickle hemoglobin. Its phenotype is modulated by fetal hemoglobin (HbF), a product of γ-globin genes. Exploring the molecules that regulate γ-globin genes at both transcriptional and translational levels, including microRNA (miRNA), might help identify alternative therapeutic targets. Materials and Methods: Using next-generation sequencing we identified pre-miRNAs and mature miRNA expression signatures associated with different HbF levels in patients homozygous for the sickle hemoglobin gene. The involvement of identified miRNAs in potential SCD-related pathways was investigated with the DIANA TOOL and miRWalk 2.0 database. Results: miR-184 were most highly upregulated in reticulocytes. miR-3609 and miR-483-5p were most highly downregulated in sickle cell anemia with high HbF. miR-370-3p that regulates LIN28A, and miR-451a which is effective in modulating α- and β- globin levels were also significantly upregulated. miRNA targeted gene pathway interaction identified BCL7A, BCL2L1, LIN28A, KLF6, GATA6, solute carrier family genes and ZNF genes associated with erythropoiesis, cell cycle regulation, glycosphingolipid biosynthesis, cAMP, cGMP-PKG, mTOR, MAPK and PI3K-AKT signaling pathways and cancer pathways. Conclusions: miRNA signatures and their target genes identified novel miRNAs that could regulate fetal hemoglobin production and might be exploited therapeutically.

1. Introduction

Sickle cell anemia (SCA) is a result of homozygosity for a mutation in codon 6 of the β-globin gene (HBB; GAG→GTG) that leads to the production of sickle hemoglobin (HbS; glu7val; α2βS2). Fetal hemoglobin (HbF; α2γ2) encoded by HBG2 and HBG1 (HBG1/2) prevents the polymerization of deoxyHbS, the proximate cause of the phenotype of SCA, and therefore the major modulator of this disease [1,2].
Circulating hairpin and mature microRNAs (miRNAs) are small noncoding RNAs that modulate gene expression by inhibition of translation or stimulating degradation of gene transcripts. miRNAs are derived from distinct hairpin precursors (pre-miRNAs) and in contrast to mature miRNAs, profiling of pre-miRNAs is limited. These miRNA hairpin precursors, which fold into stem loop structures are ideal candidates for small molecule binding. A recent study reported knockdown of BCL11A using a miRNA-adapted short hairpin RNA for post-transcriptional silencing of BCL11A to treat SCD [3]. More than 1000 miRNAs are encoded within the human genome, and some were associated with HbF induction [4,5,6]. For example, BCL11A, a major suppressor HBG2/1 expression, is targeted by miR-210, Lin28B, miR-486-3p, miR-30a and Let-7 family [7,8,9,10]. Another HBG2/1 repressor, MYB, was targeted by miR-126 and miR-15a/-16-1 which increased HBG2/1 expression [11,12].
HbF levels vary 2 to 4-fold among the five common HBB haplotypes associated with the HbS gene. The Arab-Indian (AI) haplotype is associated with HbF that averages ~17% in adults compared with 5 to 7% in the Bantu, Benin and Cameroon haplotypes and 10% in the Senegal haplotype [2,13,14,15]. Even within the AI haplotype there is variation of HbF levels. The causes of high HbF in AI haplotype SCA are not fully explained [16]. To determine whether miRNAs have an association with HbF levels in AI haplotype SCA we explored the complete hairpin and mature miRNA expression profile of HbS homozygotes with “high” and “low” HbF levels.

2. Materials and Methods

2.1. Study Population and Samples

Participants aged from 3 to 42 years, 29 males (49%) and 30 females (51%), were clinically diagnosed by high performance liquid chromatography (HPLC) of hemoglobin. HbS homozygotes who were not having an acute vaso-occlusive episode or who were not currently on hydroxyurea, and normal hemoglobin controls were recruited from Qatif Central Hospital, Al-Qatif, Saudi Arabia. Blood samples (8–10 mL) were collected into EDTA tubes from 59 individuals that included 31 SCA patients and 28 healthy individuals without SCA. Blood counts were carried out by an automated clinical hematology analyzer (Sysmex, K-1000, Sysmex Corporation, Kobe, Japan), the percentage of HbS, HbA2 and HbF were measured by HPLC on the Bio-Rad Variant II Hb Testing System (Bio- Rad Laboratories, Hercules, CA, USA).
Ethical approval for the study was obtained from the Institutional Review Board Committee (IRB-2019-01-107) at Imam Abdulrahman bin Faisal University. The study was conducted according to the ethical principles of the Declaration of Helsinki and Good Clinical Practice Guidelines.

2.2. Magnetic Separation of CD71+ and CD235+ Cells

Peripheral blood mononuclear cells (PBMC) were isolated using density gradient Ficoll- Histopaque (Merck, St. Louis, MO, USA). PBMC were labeled with CD71 MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany), which were used for the positive selection of human reticulocytes. The cell suspension was loaded onto a MACS® Column which was placed in the magnetic field of a superMACS separator. The magnetically labeled CD71+ cells were retained on the column. After removal of the column from the magnetic field, the magnetically retained CD71+ cells were eluted as the positively selected cell fraction. Similarly, Glycophorin A+ (CD235+) cells were magnetically separated after labeling with Glycophorin-A MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany). CD235a (Glycophorin A) MicroBeads were used for the positive selection of human erythroid cells as single-pass transmembrane glycoprotein is expressed on mature erythrocytes. These isolated cells were fluorescently stained with CD71-FITC and CD235-APC antibodies and confirmed by flow cytometry (Guava, Millipore, Burlington, MA, USA). Both these cell types were further processed for miRNA isolation.

2.3. RNA Extraction and Quantification

Total RNA was isolated using miRNeasy Mini kit (Qiagen, Hilden, Germany) in accordance with the manufacturer’s instructions to obtain 50 µL of extracted sample. The RNA isolation protocol utilizes QIAzol Lysis reagent, chloroform and ethanol. The final elution was with 50 µL of RNase-free water and was immediately quantified using the Qubit® microRNA Assay Kits and the Qubit® 4.0 Fluorometer (Thermo Fisher Scientific, Waltham, MA, USA). The quantified RNA was stored at −80 °C until library preparation.

2.4. miRNA Library Construction and Sequencing

Aliquots of 100 ng of miRNAs, derived from the above-mentioned procedure, were used for the preparation of miRNA libraries. Sequencing-ready cDNA libraries using the Small RNASeq Library Prep kit for Illumina (Lexogen, Austria) were prepared according to the manufacturer’s instructions. Multiplexing indices were introduced during the PCR amplification step to distinguish each sample after the pooling phase, allowing multiplexing of up to 96 libraries [17]. The library product was cleaned and concentrated with a magnetic bead-based purification protocol [18]. Lastly, the sequencing step was performed with NGS technologies using Illumina platform NextSeq 500 and the NextSeq 500/550 High Output v2.5 kit (150 cycles).

2.5. Bioinformatic Analysis of the Raw Sequencing Data

FASTQ files were generated via Illumina bcl2fastq2 (Version 2.17.1.14-http://support.illumina.Com/downloads/bcl-2fastq-conversion-software-v217.html accessed on 16 June 2021) starting from the raw sequencing reads produced by Illumina NextSeq sequencer. MiRNAs were mapped on miRbase hairpins using SHRiMP software. Differential expression analysis for miRNAs was performed with the R package DESeq2 (Ver 1.18.1) (R software, Vienna, Austria). MiRNAs with more than 5 counts were retained for further analysis. The miRNA expression is considered differentially significant if the log2(disease sample/healthy control) ≥ 1 and a False Discovery Rate (FDR) ≤ 0.1. Hence, a minimum |Log2FC| of 1 and an FDR lower than 0.1 as thresholds to differentially expressed genes to maximize the sensitivity of this analysis and to perform a massive screening and identification of candidate miRNAs. For comparative analysis, we dichotomized HbF levels into low (6.35 ± 3.57%) and high (21.31 ± 6.73%) groups [15,19].

2.6. Target Gene and Interaction Analysis

The involvement of identified miRNAs in potential SCD-related pathways (predicted target genes and predicted pathways) was investigated with the DIANA TOOL and miRWalk 2.0 database. Significantly differentially expressed miRNAs in SCA patients were considered for the analyses conducted on the mirWalk ver-2.0 to predict potential targets of these miRNAs [20]. Analysis was preferred for only validated results, using miRTarBase filter option and were compared using other popular algorithms, such as miRDB or Target Scan. DIANA-mirPath was used to hierarchical targeted genes clusters-based pathway identification utilizing the exact significance (p-value threshold set at 0.05) after FDR correction (Benjamini and Hochberg) and exclusion of pathways with few targeted genes nodes.

3. Results

Subject characteristics are shown in Table 1. Mean age in SCA was 22.45 ± 16.36 yrs. compared with 33.43 ± 13.2 yrs. in controls. Sixteen patients had low percentage of HbF (6.35 ± 3.57%) and 15 had high HbF% (21.31 ± 6.73%).
miR-184 was the most upregulated miRNA in the reticulocytes of the high HbF compared with the Low HbF cohort (fold = 2.83; p = 2.90 × 10−03; Supplementary Table S1). miR-4508 (fold = 4.36; p = 7 × 10−08), miR-451a (fold = 4.25; p = 3.6 × 10−8), miR-144 (fold = 2.78; p = 1.1 × 10−4) were upregulated in high HbF in reticulocytes compared with erythrocytes. In the high HbF cohort, the significantly downregulated miRNAs were mir-4655 (fold = −4.11; p = 1.70 × 10−8), mir-12118 (fold = −3.46; p = 4.40 × 10−4), mir-3125 (fold = −4.11; p = 5.90 × 10−9) and mir-7703 (fold = −3.49; p = 1.20 × 10−3) (Supplementary Table S2). miR-3609 (fold = −2.05; p = 2.50 × 10−2) in reticulocytes and miR-483-5p (fold = −3.6; p = 1.20 × 10−2) in erythrocytes were the most downregulated miRNA of high HbF compared with low HbF groups. The Principal Component Analysis (PCA) plot illustrating genetic relatedness among hairpin and mature miRNA is shown in Figure 1.
In gene pathway interaction assay, BCL7A, BCL2L1, LIN28A, KLF6, GATA6, SLC4A8, SLC12A5, SLC30A4, SLC30A6, ZNF154, ZNF706, ZNF618, ZNF655, ZNF789, ZNF394 and others were identified. The overall biological and functional role of these miRNAs–target genes in KEGG pathways were interpreted using DIANA tool (mirPath v.3). Genes targeted by the miRNAs were significantly enriched in erythropoiesis, cell cycle regulation, cAMP, cGMP-PKG signaling pathway, glycosphingolipid biosynthesis, mTOR, MAPK and PI3K-AKT signaling pathways. The dysregulated miRNAs and their target genes and associated pathways are listed in Table 2, while node graphs generated for these dysregulated miRNA–target gene interactions are shown in Figure 2.

4. Discussion

We studied miRNAs in Saudi HbS homozygotes with the AI haplotype from the South-Eastern Province, where α thalassemia is found in more than half of all people and where consanguinity is high, to see if additional insight might be gained into the mechanisms underlying HBG2/1 regulation in this unique haplotype. miRNAs previously associated with HbF expression in diverse SCA patients, including Lin28B, miR-486-3p, Let-7, miR-146a, miR-15a/-16-1, miR-126, miR-150, miR-199-5p, miR-221/-222, miR-23a, miR27a, miR-96, miR-26b, miR-144, miR-451 and miR-210. Let-7, mir-3609, miR-370-3p, miR-451a, mir-19b, mir-26a, and mir-15b were also dysregulated in AI haplotype SCA [21,22,23]. miRNA-370-3p (p = 7.90 × 10−3) was significantly upregulated in erythrocytes of the low HbF cohort. Gene target analysis showed that this miRNA regulates LIN28A. Lee et al. [7] assessed the capability of LIN28B to repress the expression of let-7 miRNA and determined that LIN28 overexpression or let-7 miRNA downregulation results in a 19–40% elevation in HbF levels in SCA. In addition, Lin28A obstructs the pre-let-7 processing by Dicer via recruiting terminal uridylyl transferase 4 [24]. Hence, miR-370-3p could be a novel target to regulate LIN28 for elevated HbF production.
Similarly, miR-451a was upregulated in reticulocytes (p = 7.10 × 10−4). Expression of miRNA-451 is elevated during erythroid cell differentiation. Fang and Bartel [25] reported that miR-451, which derives from a hairpin with a suboptimal terminal loop and a suboptimal stem length, accumulates 40-fold higher levels when clustered with a helper hairpin. miR-451 was upregulated in reticulocytes by approximately 2.5-fold in the high HbF cohort. miRNA-451 forms a cluster with miRNA-144 and regulates the expression of many genes necessary for erythropoiesis. In 6 SCA patients with HbF 23.5 ± 2.1% and 6 patients with HbF 3.41 ± 1.0%, all with undetermined haplotypes, miRNA-144 expression in reticulocytes was upregulated 8-fold in low vs. high HbF patient groups [26]. miR-144 and its mature forms were significantly dysregulated in the reticulocytes of high HbF cohort compared with erythrocytes. Functional analysis of normal and sickle erythroid progenitors showed that the miRNA-144 antagomir can facilitate HbF production while increasing NRF2 expression [26]. Among eight adult patients with the Bantu HbS haplotype, 22 of 798 miRNAs were differentially expressed with 13 associated with genes that regulate HBG2/1 expression like BCL11A (miR-148b-3p, miR-32-5p, miR-340-5p, and miR-29c-3p), MYB (miR-105-5p), and KLF3 (miR-106b-5), and SP1 (miR-29b-3p, miR-625-5p, miR-324-5p, miR-125a-5p, miR-99b-5p, miR-374b-5p, and miR-145-5p) [23].
The zinc finger proteins, ZNF802 and ZNF410 are HbF gene repressors. Induced deficiency of ZNF802 resulted in an increase in HbF to 35.0  ±  3.5% [27]. Knockout of ZNF410 reduced Chromodomain Helicase DNA Binding Protein 4 (CHD4) levels by 60%, enough to substantially derepress HbF [28]. ZNF154, ZNF706, ZNF655, ZNF789, ZNF394 and ZNF618 were among the miRNA targets of the present study. Similarly, other than the well-studied BCL11A, BCL7A and BCL2L1 were also among the target genes in the present study. BCL2L1 has been associated with HbF gene expression in patients with SCA, where overexpression of BCL2L1 resulted in increased HBG2/1 expression fourfold and F-cells by 13% [29]. Likewise, other than the known KLF1, an interaction between KLF6, a new potential regulator of erythropoiesis and miR-2355-5p was identified, and its downregulation significantly raised γ-globin mRNA [30]. miR-181c-5p targeted KLF6, and the significantly downregulated miR-3609 targeted the binding site of KLF6 [31].
Hematopoietic stem cell differentiation is associated with activation of multiple signaling pathways including MAPK and PI3K [32,33,34]. We found novel miRNAs, miR-5787, miR-3609 and miR-483-5p associated with these signaling pathways might be potentially exploited as therapeutic targets. Histone deacetylase inhibitors like Apicidin and Tricostatin, increase H3 and H4 acetylation in the region adjacent to the HBG2/1 promoters [35] and induce γ-globin gene expression [36] by activating MAPK signaling pathway.

5. Conclusions

miRNA signatures and their target gene-pathway analysis identified novel miRNAs that could regulate fetal hemoglobin production and might be exploited therapeutically. Additional studies in erythroid cell lines and primary erythroid progenitor cells are needed to define how any of the identified miRNAs might modulate HBG2/1 expression and the accumulation of HbF and if some of the HbF-associated miRNAs can account for the high levels of HbF in AI haplotype SCA.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/medicina58101470/s1, Supplementary Table S1: List of upregulated miRNAs with more than 1.5-fold change with HbF level in SCA cohort; Supplementary Table S2: List of downregulated miRNAs with more than 1.5-fold change with HbF level in SCA cohort.

Author Contributions

C.C., C.V., A.A.-N., S.C., M.S.A., M.D., D.A. and A.A.-A. are involved in the design of the work, critically revising of protocol, patient recruitment, analysis, interpretation of data and drafting of the manuscript. C.C., S.C. and A.A.-A. are involved in the design of the work, laboratory work, analysis, interpretation of data and drafting of the manuscript. C.C., S.H.A. and M.H.S. are involved in the design of the work, critically revising of protocol, analysis, interpretation of data and drafting of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia, Grant Number-11-MED1422-46. This is a government agency.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board/Ethics Committee of Imam Abdulrahman bin Faisal University Ethical Committee (Reference IRB-2019-01-107).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to extend our gratitude to King Abdulaziz City for Science and Technology for sponsoring this project and for their continued support [Grant Number-11-MED1422-46]. We are also grateful to the patients, nurses, and technical staff at the hospital for their dedication and support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Piel, F.B.; Steinberg, M.H.; Rees, D.C. Sickle cell disease. N. Engl. J. Med. 2017, 376, 1561–1573. [Google Scholar] [CrossRef] [Green Version]
  2. Akinsheye, I.; Alsultan, A.; Solovieff, N.; Ngo, D.; Baldwin, C.T.; Sebastiani, P.; Chui, D.H.; Steinberg, M.H. Fetal hemoglobin in sickle cell anemia. Blood 2011, 118, 19–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Esrick, E.B.; Lehmann, L.E.; Biffi, A.; Achebe, M.; Brendel, C.; Ciuculescu, M.F.; Daley, H.; MacKinnon, B.; Morris, E.; Federico, A.; et al. Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease. N. Engl. J. Med. 2021, 384, 205–215. [Google Scholar] [CrossRef] [PubMed]
  4. Cyrus, C. The Role of miRNAs as Therapeutic Tools in Sickle Cell Disease. Medicina 2021, 57, 1106. [Google Scholar] [CrossRef] [PubMed]
  5. Gabbianelli, M.; Testa, U.; Morsilli, O.; Pelosi, E.; Saulle, E.; Petrucci, E.; Castelli, G.; Giovinazzi, S.; Mariani, G.; Fiori, M.E.; et al. Mechanism of human Hb switching: A possible role of the kit receptor/miR 221–222 complex. Haematologica 2010, 95, 1253–1260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Tayebi, B.; Abrishami, F.; Alizadeh, S.; Minayi, N.; Mohammadian, M.; Soleimani, M.; Dehghanifard, A.; Atwan, H.; Ajami, M.; Ajami, M. Modulation of microRNAs expression in hematopoietic stem cells treated with sodium butyrate in inducing fetal hemoglobin expression. Artif. Cells Nanomed. Biotechnol. 2017, 45, 146–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Lee, Y.T.; de Vasconcellos, J.F.; Yuan, J.; Byrnes, C.; Noh, S.J.; Meier, E.R.; Kim, K.S.; Rabel, A.; Kaushal, M.; Muljo, S.A.; et al. LIN28B-mediated expression of fetal hemoglobin and production of fetal-like erythrocytes from adult human erythroblasts ex vivo. Blood 2013, 122, 1034–1041. [Google Scholar] [CrossRef] [Green Version]
  8. Lulli, V.; Romania, P.; Morsilli, O.; Cianciulli, P.; Gabbianelli, M.; Testa, U.; Giuliani, A.; Marziali, G. MicroRNA-486-3p regulates γ-globin expression in human erythroid cells by directly modulating BCL11A. PLoS ONE 2013, 8, e60436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Gasparello, J.; Fabbri, E.; Bianchi, N.; Breveglieri, G.; Zuccato, C.; Borgatti, M.; Gambari, R.; Finotti, A. BCL11A mRNA Targeting by miR-210: A Possible Network Regulating γ-Globin Gene Expression. Int. J. Mol. Sci. 2017, 18, 2530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Gholampour, M.A.; Asadi, M.; Naderi, M.; Azarkeivan, A.; Soleimani, M.; Atashi, A. miR-30a regulates γ-globin expression in erythoid precursors of intermedia thalassemia through targeting BCL11A. Mol. Biol. Rep. 2020, 47, 3909–3918. [Google Scholar] [CrossRef] [PubMed]
  11. Grabher, C.; Payne, E.M.; Johnston, A.B.; Bolli, N.; Lechman, E.; Dick, J.E.; Kanki, J.P.; Look, A.T. Zebrafish microRNA-126 determines hematopoietic cell fate through c-Myb. Leukemia 2011, 25, 506–514. [Google Scholar] [CrossRef] [Green Version]
  12. Sankaran, V.G.; Menne, T.F.; Šćepanović, D.; Vergilio, J.-A.; Ji, P.; Kim, J.; Thiru, P.; Orkin, S.H.; Lander, E.S.; Lodish, H.F. MicroRNA-15a and-16-1 act via MYB to elevate fetal hemoglobin expression in human trisomy 13. Proc. Natl. Acad. Sci. USA 2011, 108, 1519–1524. [Google Scholar] [CrossRef] [Green Version]
  13. Habara, A.H.; Shaikho, E.M.; Steinberg, M.H. Fetal hemoglobin in sickle cell anemia: The Arab-Indian haplotype and new therapeutic agents. Am. J. Hematol. 2017, 92, 1233–1242. [Google Scholar] [CrossRef] [Green Version]
  14. Alnafie, A.N.; Alateeq, S.A.; Al-Muhanna, F.A.; Alsulaiman, A.M.; Alfarhan, M.; Buali, W.; Vatte, C.B.; Cyrus, C.; Keating, B.; Al-Ali, A.K.; et al. Exome sequencing in high and low fetal haemoglobin Arab-Indian haplotype sickle cell disease. Br. J. Haematol. 2021, 194, e61–e64. [Google Scholar] [CrossRef] [PubMed]
  15. Al-Ali, A.K.; Alsulaiman, A.; Alzahrani, A.J.; Obeid, O.T.; Vatte, C.B.; Cyrus, C.; Alnafie, A.N.; Alali, R.A.; Alfarhan, M.; Mozeleski, B.; et al. Prevalence and diversity of haplotypes of sickle cell disease in the Eastern Province of Saudi Arabia. Hemoglobin 2020, 44, 78–81. [Google Scholar] [CrossRef] [PubMed]
  16. Steinberg, M.H. Fetal Hemoglobin in Sickle Hemoglobinopathies: High HbF Genotypes and Phenotypes. J. Clin. Med. 2020, 9, 3782. [Google Scholar] [CrossRef] [PubMed]
  17. Benesova, S.; Kubista, M.; Valihrach, L. Small RNA-Sequencing: Approaches and Considerations for miRNA Analysis. Diagnostics 2021, 11, 964. [Google Scholar] [CrossRef]
  18. Lexogen User Guide. Available online: https://www.lexogen.com/wp-content/uploads/2020/02/052UG128V0110_Small-RNA-Seq-Library-Preparation-Kit_2020-01-22.pdf (accessed on 1 May 2021).
  19. Vathipadiekal, V.; Farrell, J.J.; Wang, S.; Edward, H.L.; Shappell, H.; Al-Rubaish, A.M.; Al-Muhanna, F.; Naserullah, Z.; Alsuliman, A.; Qutub, H.O.; et al. A candidate transacting modulator of fetal hemoglobin gene expression in the Arab-Indian haplotype of sickle cell anemia. Am. J. Hematol. 2016, 91, 1118–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Sticht, C.; De La Torre, C.; Parveen, A.; Gretz, N. miRWalk: An online resource for prediction of microRNA binding sites. PLoS ONE. 2018, 13, e0206239. [Google Scholar] [CrossRef]
  21. Chen, S.Y.; Wang, Y.; Telen, M.J.; Chi, J.T. The genomic analysis of erythrocyte microRNA expression in sickle cell diseases. PLoS ONE. 2008, 3, e2360. [Google Scholar] [CrossRef]
  22. Jain, S.; Kapetanaki, M.G.; Raghavachari, N.; Woodhouse, K.; Yu, G.; Barge, S.; Coronnello, C.; Benos, P.; Kato, G.; Kaminski, N.; et al. Expression of regulatory platelet microRNAs in patients with sickle cell disease. PLoS ONE. 2013, 8, e60932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mnika, K.; Mazandu, G.K.; Jonas, M.; Pule, G.D.; Chimusa, E.R.; Hanchard, N.A.; Wonkam, A. Hydroxyurea-induced miRNA expression in sickle cell disease patients in Africa. Front. Genet. 2019, 10, 509. [Google Scholar] [CrossRef] [Green Version]
  24. Piskounova, E.; Polytarchou, C.; Thornton, J.E.; LaPierre, R.J.; Pothoulakis, C.; Hagan, J.P.; Iliopoulos, D.; Gregory, R.I. Lin28A and Lin28B inhibit let-7 microRNA biogenesis by distinct mechanisms. Cell 2011, 147, 1066–1079. [Google Scholar] [CrossRef] [Green Version]
  25. Fang, W.; Bartel, D.P. MicroRNA Clustering Assists Processing of Suboptimal MicroRNA Hairpins through the Action of the ERH Protein. Mol. Cell 2020, 78, 289–302.e6. [Google Scholar] [CrossRef]
  26. Li, B.; Zhu, X.; Ward, C.M.; Starlard-Davenport, A.; Takezaki, M.; Berry, A.; Ward, A.; Wilder, C.; Neunert, C.; Kutlar, A.; et al. MIR-144-mediated NRF2 gene silencing inhibits fetal hemoglobin expression in sickle cell disease. Exp. Hematol. 2019, 70, 85–96.e5. [Google Scholar] [CrossRef] [PubMed]
  27. Wongborisuth, C.; Chumchuen, S.; Sripichai, O.; Anurathaphan, U.; Sathirapongsasuti, N.; Songdej, D.; Tangprasittipap, A.; Hongeng, S. Down-regulation of the transcriptional repressor ZNF802 (JAZF1) reactivates fetal hemoglobin in β0-thalassemia/HbE. Sci. Rep. 2022, 12, 4952. [Google Scholar] [CrossRef] [PubMed]
  28. Vinjamur, D.S.; Yao, Q.; Cole, M.A.; McGuckin, C.; Ren, C.; Zeng, J.; Hossain, M.; Luk, K.; Wolfe, S.A.; Pinello, L.; et al. ZNF410 represses fetal globin by singular control of CHD4. Nat. Genet. 2021, 53, 719–728. [Google Scholar] [CrossRef] [PubMed]
  29. Dai, Y.; Shaikho, E.M.; Perez, J.; Wilson, C.A.; Liu, L.Y.; White, M.R.; Farrell, J.J.; Chui, D.H.K.; Sebastiani, P.; Steinberg, M.H. BCL2L1 is associated with γ-globin gene expression. Blood Adv. 2019, 3, 2995–3001. [Google Scholar] [CrossRef] [PubMed]
  30. Cheng, Y.; Shang, X.; Chen, D.; Pang, D.; Zhao, C.; Xu, X. MicroRNA-2355-5p regulates γ-globin expression in human erythroid cells by inhibiting KLF6. Br. J. Haematol. 2021, 193, 401–405. [Google Scholar] [CrossRef]
  31. Fishilevich, S.; Nudel, R.; Rappaport, N.; Hadar, R.; Plaschkes, I.; Stein, T.I.; Rosen, N.; Kohn, A.; Twik, M.; Safran, M.; et al. GeneHancer: Genome-wide integration of enhancers and target genes in GeneCards. Database 2017, 2017, bax028. [Google Scholar] [CrossRef]
  32. Rahim, F.; Allahmoradi, H.; Salari, F.; Shahjahani, M.; Fard, A.D.; Hosseini, S.A.; Mousakhani, H. Evaluation of Signaling Pathways Involved in γ-Globin Gene Induction Using Fetal Hemoglobin Inducer Drugs. Int. J. Hematol. Oncol. Stem Cell Res. 2013, 7, 41–46. [Google Scholar] [PubMed]
  33. Nakao, T.; Geddis, A.E.; Fox, N.E.; Kaushansky, K. PI3K/Akt/FOXO3a pathway contributes to thrombopoietin-induced proliferation of primary megakaryocytes in vitro and in vivo via modulation of p27Kip1. Cell Cycle 2008, 7, 257–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kadri, Z.; Chretien, L.; Rooke, H.M.; Orkin, S.H.; Romeo, P.-H.; Mayeux, P.; Leboulch, P.; Chretien, S. Phosphatidylinositol 3-kinase/Akt induced by erythropoietin renders the erythroid differentiation factor GATA-1 competent for TIMP-1 gene transactivation. Mol. Cell Biol. 2005, 25, 7412–7422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kiefer, C.M.; Hou, C.; Little, J.A.; Dean, A. Epigenetics of beta-globin gene regulation. Mutat. Res. 2008, 647, 68–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sangerman, J.; Lee, M.S.; Yao, X.; Oteng, E.; Hsiao, C.H.; Li, W.; Zein, S.; Ofori-Acquah, S.F.; Pace, B.S. Mechanism for fetal hemoglobin induction by histone deacetylase inhibitors involves gamma-globin activation by CREB1 and ATF-2. Blood 2006, 108, 3590–3599. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Principal Component Analysis (PCA) plot illustrating genetic relatedness among hairpin and mature miRNA in study cohort.
Figure 1. Principal Component Analysis (PCA) plot illustrating genetic relatedness among hairpin and mature miRNA in study cohort.
Medicina 58 01470 g001
Figure 2. The node graphs of some dysregulated miRNA–target gene interaction associated with HbF level.
Figure 2. The node graphs of some dysregulated miRNA–target gene interaction associated with HbF level.
Medicina 58 01470 g002
Table 1. Baseline characteristics and hematological parameters.
Table 1. Baseline characteristics and hematological parameters.
Control HbS Homozygote Low HbFHigh HbFControl vs. SCALow HbF vs. High HbF
(N = 28)(N = 31)
Mean ± SDMean ± SDMean ± SDMean ± SDp Valuep Value
Age (Y)33.43 ± 13.222.45 ± 16.3617.34 ± 16.0527.25 ± 15.640.010.092
HbF (%)0.83 ± 0.3214.98 ± 9.346.35 ± 3.5721.31 ± 6.73<0.0001 <0.0001
HbA2 (%)3.54 ± 1.033.03 ± 0.52.91 ± 0.63.2 ± 0.290.120.193
RBC (103/mm3)5.79 ± 0.924.13 ± 0.743.9 ± 0.614.44 ± 0.83<0.00010.097
Hb (g/dL)12.04 ± 1.5810.65 ± 1.610.92 ± 1.4210.3 ± 1.820.090.366
Hematocrit (%)40.18 ± 5.9831.49 ± 5.2431.97 ± 4.0830.91 ± 6.56<0.00010.648
MCV (fL)69.92 ± 8.7278.17 ± 10.6782.86 ± 10.1472.55 ± 8.70.120.019
MCH (pg)21.06 ± 2.9726.42 ± 3.9528.01 ± 3.4324.51 ± 3.830.010.035
MCHC (g/dL)30.04 ± 1.3433.68 ± 1.5633.9 ± 1.2233.42 ± 1.93<0.00010.485
Platelets (103/mm3)393.2 ± 72.9271.7 ± 112.5229.33 ± 75.03322.6 ± 131.820.030.05
Table 2. Dysregulated miRNA, target gene interaction and associated pathways.
Table 2. Dysregulated miRNA, target gene interaction and associated pathways.
CohortDysregulated miRNATarget GenesTargeted Pathways
Low HbF vs. High HbF
upregulated
miR-184BIN3, LRRC8AArrhythmogenic right ventricular cardiomyopathy (ARVC)
Adherens junction
Adrenergic signaling in cardiomyocytes
Oocyte meiosis
Signaling pathways regulating pluripotency of stem cells
Thyroid hormone synthesis
Glioma
Choline metabolism in cancer
Pancreatic cancer
Hippo signaling pathway
GnRH signaling pathway
Oxytocin signaling pathway
cGMP-PKG signaling pathway
Mucin type O-Glycan biosynthesis
Glycosphingolipid biosynthesis-ganglio series
Acute myeloid leukemia
Axon guidance
Inflammatory mediator regulation of TRP channels
Insulin secretion
Salivary secretion
Fatty acid elongation
miR-3125BCL7A, BTF3L4, CCND2, GNL1, HDGFL3, HIF3A, HSP90AA1, IGF1R, LUC7L2, PDGFB, RPS6KA3, RPS6KA3, SFT2D2, SMG1, TRIM67, USB1
miR-3169TMCC1, TTL
miR-3668BTBD3
miR-3976BCL7A, KLHL8
miR-4261NFAT5
miR-4327FNDC3B, TMEM41B, TP53INP1
miR-4654ABI2, DCAF8, PMP22, PTP4A1, YWHAZ
miR-4754ARMCX6
miR-5787C10orf67, CBX6, CELF1, GTF3C1, IBA57, ILK, MAPK8IP3, MEMO1, MLX, NCKAP5L, PAK4, PNMA8B, PTGIS, RAP1GAP2, RGS6, SEC22C, SEPTIN14, TULP1
miR-6081ABL2, ASPH, BCL2L1, HEYL, HIF3A, HMGA1, NAB2, PLEKHA6, RPS28, UBE2D3
miR-662MCFD2
miR-7703CD226, CLOCK, FHL2, FNBP1L, GATA6, KCNN3, KIAA1958, LRPAP1, MRO, SLC12A5, SLC30A4, TEF, TFDP2, TSPAN31
miR-922CALU, CDK6, CMTM4, GABPA, GALNT1, GOLGA7, LRRFIP2, PAK2, PHC3, RNFT2, TMEM248, TRMT9B, UBE2H, ZBTB10, ZBTB44, ZNF154, ZNF706
Low HbF vs. High HbF
downregulated
miR-3609AKR7A2, BCL2L11, BTF3L4, BZW1, CCDC71L, FOXJ3, KCNK10, KIAA0513, KIAA1191, MDM4, MYLIP, NFAT5, NIBAN1, NKIRAS1, POU2F1, PTPN4, RAB11FIP1, RAP1B, RUFY2, SREK1IP1, TNKS2, WNK3, ZNF655, ZNF789, ZNF394Drug metabolism-cytochrome P450
Transcriptional misregulation in cancer
Ubiquitin mediated proteolysis
Lysine degradation
PI3K-Akt signaling pathway
Mucin type O-Glycan biosynthesis
Neurotrophin signaling pathway
miR-483-5pMAPK3
Control vs. High HbF
upregulated
miR-382-3pATP13A3, CCNT1M, KLHL29, METAP1, SLC4A8Glycosaminoglycan biosynthesis - heparan sulfate/heparin
Ubiquitin mediated proteolysis
Gap junction
Colorectal cancer
Basal cell carcinoma
Valine, leucine and isoleucine biosynthesis
mTOR signaling pathway
Tyrosine metabolism
Long-term potentiation
Endocytosis
TGF-beta signaling pathway
RNA degradation
Osteoclast differentiation
miR-451aVAPA
Control vs. High HbF downregulatedmiR-184CRISPLD2, EPB41L5, FBXO28, SF1Lysine degradation
Pentose phosphate pathway
Caffeine metabolism
Biosynthesis of unsaturated fatty acids
Taurine and hypotaurine metabolism
miR-542-3pASRGL1, EDEM3, GPATCH2L, GUCY1A2, HIPK3, MAP7, PLEKHM3, SAMD12, SLC30A6, VAPB, ZNF618
Control vs. Low HbF
upregulated
miR-134-5pANGPTL4Estrogen signaling pathway
Purine metabolism
Rap1 signaling pathway
Long-term potentiation
Proteoglycans in cancer
Glioma
Thyroid cancer
Acute myeloid leukemia
Prolactin signaling pathway
Non-small cell lung cancer
miR-181c-5pCPEB4, DDX3X, ETS1, KLF6, MECP2, MTX3, NAA50, OSBPL3, RCOR1, RPS6KA3, TRIM2
miR-370-3pCYB561D1, LIN28A, NF1, NSUN4, PARVB, TGFBR2
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cyrus, C.; Vatte, C.; Al-Nafie, A.; Chathoth, S.; Akhtar, M.S.; Darwish, M.; Almohazey, D.; AlDubayan, S.H.; Steinberg, M.H.; Al-Ali, A. miRNA Expression Associated with HbF in Saudi Sickle Cell Anemia. Medicina 2022, 58, 1470. https://doi.org/10.3390/medicina58101470

AMA Style

Cyrus C, Vatte C, Al-Nafie A, Chathoth S, Akhtar MS, Darwish M, Almohazey D, AlDubayan SH, Steinberg MH, Al-Ali A. miRNA Expression Associated with HbF in Saudi Sickle Cell Anemia. Medicina. 2022; 58(10):1470. https://doi.org/10.3390/medicina58101470

Chicago/Turabian Style

Cyrus, Cyril, Chittibabu Vatte, Awatif Al-Nafie, Shahanas Chathoth, Mohammed S. Akhtar, Mohammed Darwish, Dana Almohazey, Saud H. AlDubayan, Martin H. Steinberg, and Amein Al-Ali. 2022. "miRNA Expression Associated with HbF in Saudi Sickle Cell Anemia" Medicina 58, no. 10: 1470. https://doi.org/10.3390/medicina58101470

Article Metrics

Back to TopTop