Open Access

Peptide nucleic acids targeting β-globin mRNAs selectively inhibit hemoglobin production in murine erythroleukemia cells

  • Authors:
    • Giulia Montagner
    • Chiara Gemmo
    • Enrica Fabbri
    • Alex Manicardi
    • Igea Accardo
    • Nicoletta Bianchi
    • Alessia Finotti
    • Giulia Breveglieri
    • Francesca Salvatori
    • Monica Borgatti
    • Ilaria Lampronti
    • Alberto Bresciani
    • Sergio Altamura
    • Roberto Corradini
    • Roberto Gambari
  • View Affiliations

  • Published online on: November 14, 2014     https://doi.org/10.3892/ijmm.2014.2005
  • Pages: 51-58
  • Copyright: © Montagner et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

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


Abstract

In the treatment of hemoglobinopathies, amending altered hemoglobins and/or globins produced in excess is an important part of therapeutic strategies and the selective inhibition of globin production may be clinically beneficial. Therefore the development of drug-based methods for the selective inhibition of globin accumulation is required. In this study, we employed peptide nucleic acids (PNAs) to alter globin gene expression. The main conclusion of the present study was that PNAs designed to target adult murine β-globin mRNA inhibit hemoglobin accumulation and erythroid differentiation of murine erythroleukemia (MEL) cells with high efficiency and fair selectivity. No major effects were observed on cell proliferation. Our study supports the concept that PNAs may be used to target mRNAs that, similar to globin mRNAs, are expressed at very high levels in differentiating erythroid cells. Our data suggest that PNAs inhibit the excess production of globins involved in the pathophysiology of hemoglobinopathies.

Introduction

The majority of the molecular biology based approaches developed for the experimental therapy of thalassemia and sickle-cell anemia (SCA) are based on the induction of defective globin (β-globin in β-thalassemia) (15) or (also in association with this strategy) the induction of fetal hemoglobin (HbF) (610). The de novo production of adult hemoglobin (HbA) can be achieved in β0-thalassemias by gene therapy (14) and gene correction, by homologous recombination (11,12) and/or by the treatment of erythroid cells with molecules causing read-through (13). The induction of HbF can be obtained by using low molecular weight drugs causing the induction of the γ-globin gene (68,1417), artificial promoters (18,19), decoy molecules targeting transcription factors involved in the transcriptional repression of γ-globin genes (MYB, KLF-1 and BCL-11A) (20,21), or microRNAs targeting mRNAs coding for these repressors (data are available for microRNAs miR-15a, miR-16-1, miR-486-3p and miR-23a/27a) (2224). On the other hand, clinical complications in SCA and β-thalassemia are also related to the production of defective proteins (β-globin in SCA) (2527) or to the accumulation of free globins which are not organized in a functional tetramer (such as in the case of free α-globins in β-thalassemia) (28,29). It is well known that sickle hemoglobin (HbS) has peculiar biochemical properties, leading to polymerization when deoxygenated. HbS polymerization is associated with a reduction in cell ions and water content (cell dehydration) and increased red cell density, which further accelerates HbS polymerization. Pathophysiological studies have indicated that the dense, dehydrated red cells may play a central role in acute and chronic clinical manifestations of sickle-cell disease, in which intravascular sickling in capillaries and small vessels leads to vaso-occlusion and impaired blood flow in a variety of organs and tissues (30). Therefore, the development of drug-based methods for the inhibition of the accumulation of defective hemoglobins (HbS in SCA) or globin produced in excess (α-globins in β-thalassemia) is required (31,32).

In this field of investigation, peptide nucleic acids (PNAs) may be of great interest (33). PNAs are DNA analogues in which the sugarphosphate backbone is replaced by N-(2-aminoethyl)glycine units (33). These very interesting molecules were described for the first time by Nielsen et al (34) and, despite a radical structural change with respect to DNA and RNA, they are capable of sequence-specific and efficient hybridization with complementary DNA and RNA, forming Watson-Crick double helices (35). In addition, they are able to generate triple helix formation with double-stranded DNA and perform strand invasion (34). Accordingly, PNA-based analogues have been proposed as antisense molecules targeting mRNAs and microRNAs, triple-helix forming molecules targeting eukaryotic gene promoters, artificial promoters and decoy molecules targeting transcription factors (36). To the best of our knowledge, PNAs have not yet been employed to inhibit the expression of globin genes in erythroid cells. Thus, the aim of this study was to verify whether PNAs targeting globin mRNAs can be used to modulate globin gene expression and to reduce the level of a given type of globin. For this purpose, we produced one PNA targeting murine adult β-globin mRNAs and another recognizing the human γ-globin and β-globin mRNAs. These PNAs were tested on relevant target erythroid cell lines, such as the murine erythroleukemia (MEL) cell line. Erythroid differentiation and the high production of hemoglobins were induced by treatment with dimethylsulfoxide (DMSO) and hexamethylene bisacetamide (HMBA) (3739).

Materials and methods

Synthesis and characterization of PNAs

The synthesis of the two PNAs was performed using standard automated Fmoc-based chemistry with HBTU/DIPEA coupling on a ChemMatrix resin loaded with Fmoc-Gly-OH as first monomer (loading 0.2 mmol/g, 5 μmol scale), on a Syro II peptide synthesizer, using commercially available monomers (Link Technologies, Bellshill, UK); Fmoc-Arg (Pbf)-OH (Sigma-Aldrich, St. Louis, MO, USA) was used for octaarginine synthesis. PNA purification was performed by reversed-phase high-performance liquid chromatography (RP-HPLC) with UV detection at 260 nm using a semi-prep column C18 (10 μm, 300×7.7 mm, Xterra Waters, 300 Å), eluting with water containing 0.1% TFA (eluent A) and acetonitrile containing 0.1% TFA (eluent B); elution gradient: from 100% A to 50% B in 30 min, flow: 4 ml/min. The purity and identity of the purified PNA were examined by ultra-performance liquid chromatography tandem mass-spectrometry (UPLC-MS; Waters Acquity equipped with ESI-Q analizer) using an Acquity UPLC BEH C18; 2.1×50 MM, 1.7 μm column. Anti-M-βglob-PNA: yield, 6%; electrospray ionization mass spectrometry (ESI-MS): m/z found (calculated): 1267.3 (1267.3) [MH55+], 1056.2 (1056.2) [MH66+], 905.4 (905.5) [MH77+], 792.2 (792.4) [MH88+], 704.5 (704.5) [MH99+]; calculated MW: 6331.39. Anti-H-γglob-PNA: yield: 8%; ESI-MS: m/z found (calculated): 1346.0 (1345.4) [MH44+]; 1076.8 (1076.5) [MH55+], 897.7 (897.3) [MH66+], 769.5 (769.2) [MH77+], 673.3 (673.2) [MH88+], 598.8 (598.5) [MH99+], 539.0 (538.8) [MH1010+]; calculated MW: 5377.54.

MEL and K562 cell lines and culture conditions

MEL cells (3739) were cultured in humidified atmosphere of 5% CO2/air in RPMI-1640 medium (Sigma-Aldrich) supplemented with 10% fetal bovine serum (FBS; Biowest, Nuaillé, France), 50 U/ml penicillin and 50 μg/ml streptomycin (39). DMSO and HMBA were from Sigma-Aldrich. Stock solutions of HMBA were stored at −20°C in the dark and diluted immediately before use. Treatment of the MEL cells with HMBA and DMSO was carried out by adding the appropriate drug concentrations at the beginning of the cultures (30,000 cells/ml were seeded). To determine the effects of the treatments on the proliferation of the MEL cells, cell growth was monitored by determining the cell number/ml using a Z1 Coulter Counter (Coulter Electronics, Hialeah, FL, USA). Erythroid differentiated MEL cells containing hemoglobin were detected by specific reaction with a benzidine/hydrogen peroxide solution (0.2% in 5 M glacial acetic acid and 10% H2O2). The K562-D5 cell line was employed as it produces, in addition to hemoglobin (Hb) Gower1 and hemoglobin (Hb) Portland, HbF and HbA (40).

RNA extraction

The cells were isolated by centrifugation at 1,500 rpm for 10 min at 4°C, washed with phosphate-buffered saline (PBS) and lysed with TRI-reagent™ (Sigma-Aldrich) according to the manufacturer’s instructions. The isolated RNA was washed once with cold 75% ethanol, dried and dissolved in nuclease-free pure water prior to use.

Quantitative reverse transcription polymerase chain reaction (RT-qPCR)

For gene expression analysis, 500 ng of total RNA were reverse transcribed using random hexamers. Quantitative PCR assays were carried out using gene-specific double-quenched probes containing a 5′-FAM fluorophore, a 3′-IBFQ quencher and an internal ZEN quencher. The nucleotide sequences used for the RT-qPCR analysis of mouse globin mRNAs were α-globin forward, 5′-CTG ACC TCC AAG TAC CGT TAA G-3′ and reverse primer, 5′-GCT TCT TCC TAC TCA GGC TTT AT-3′ and α-globin probe, 5′-/56-FAM/TCT CTC CCT/ZEN/TGC ACC TGT ACC TCT/3IABkFQ/-3′; β-globin forward, 5′-GGA AAG GTG AAC TCC GAT GAA-3′ and reverse primer, 5′-TGA TAG CAG AGG CAG AGG ATA G-3′ and β-globin probe, 5′-/56-FAM/CCT TGG ACC/ZEN/CAG CGG TAC TTT GAT/3IABkFQ/-3′. The primers and probes used to assay mouse globins were purchased from Integrated DNA Technologies (IDT; Coralville, IA, USA). The relative expression was calculated using the comparative cycle threshold method and the endogenous control mouse gene, GAPDH, was used as a reference gene (PrimeTime Mm.PT.39a.1, IDT).

HPLC analysis

The cells were harvested, washed once with PBS and the pellets were lysed in lysis buffer (sodium dodecyl sulphate 0.01%). After spinning for 1 min in a microcentrifuge, the supernatant was collected and stored at 4°C. Hemoglobins in the lysates were separated by cation-exchange HPLC (Pharmacia LKB Gradient Pump 2249, VWM 2141), using a Synchropak CM300 (250×4.6 mm) column (Eichrom Technologies, Inc., Darien, IL, USA) and BisTris (30 mM) buffer. Standard HbA and HbF (Sigma-Aldrich, Milwaukee, WI, USA; Helena Laboratories, Beaumont, TX, USA) solutions were used as a reference.

Bioinformatics analysis

The secondary structure of the mouse βmajor- and βminor-globin mRNA sequences, 5′ untranslated region (UTR), coding sequence (CDS) and 3′UTR, was predicted using the program available online TBI ViennaRNA Web Services (http://rna.tbi.univie.ac.at). The mouse β-globin reference sequences analyzed were obtained from the NCBI website.

Statistical analysis

The results are expressed as the means ± standard error of the mean (SEM). Comparisons between groups were made using a paired Student’s t-test and one-way analysis of variance (ANOVA). Statistical significance was defined with p<0.01.

Results

Design of PNAs

The location of the binding sites and the sequences of the PNAs employed in this study are presented in Fig. 1. As clearly appreciable from the analysis of the sequences recognized, the anti-M-βglob-PNA is able to hybridize to a region of both mouse βmajor- and βminor-globin mRNAs exhibiting similar predicted secondary structures (Fig. 1B and C). This feature allows us to study the effects of the PNAs simply analyzing the proportion of erythroid differentiated MEL cells.

These cells, upon the induction of erythroid differentiation with HMBA or DMSO produce almost exclusively Hbmajor2β2major) and Hbminor2β2minor). Moreover, these PNAs display 4 to 8 mismatches with human β-globin (8 mismatches), γ-globin (3 mismatches) and ɛ-globin (4 mismatches) (Fig. 1A, bottom panel). This allows us to verify possible non-specific inhibitory effects when used on K562 cell clones subjected to erythroid differentiation which produce, upon treatment with mithramycin (MTH), high levels of Hb Gower 1 (ζ2ɛ2), Hb Portland (ζ2γ2), HbF (α2γ2) and HbA (α2β2) (40). Conversely, as control PNA molecules we used anti-H-γglob-PNA recognizing the evolutionarily homologue human γ-globin mRNAs and displaying 3 mismatches with the murine βmajor and βminor mRNAs (Fig. 1A). Both anti-M-βglob-PNA and anti-H-γglob-PNA were linked with a R8 polyarginine peptide, since it has been reported in several studies that the uptake of PNAs by target cells is difficult (41,42). R8 was employed in order to maximize PNA uptake, as reported by our group for several PNAs, which, without exception, are internalized with very high efficiency to target cells (4345).

Effects of anti-M-βglob-PNA on the growth of MEL cells

Fig. 2 shows the kinetics of differentiation (Fig. 2A) and cell growth (Fig. 2B) obtained when the MEL cells are treated with 2% and 2.5 mM DMSO and HMBA, respectively. The high level of induction (>80% after 3 or 4 days in all the experiments performed) confirms that this cellular system is excellent to determine inhibitors of the expression of adult β-globin genes, since, unlike other erythroid cellular model systems (such as human K562 cells), these cells mostly produce the adult-type Hbmajor and Hbminor hemoglobins (39). As shown in Fig. 2C, the addition of anti-M-βglob-PNA did not alter the proliferation rate of these cells, formally demonstrating no cytotoxic effects of this PNA on the MEL cells, either in the absence of differentiation inducers (data not shown) or in the presence of DMSO or HMBA.

Anti-M-βglob-PNA inhibits the erythroid differentiation of MEL cells induced by DMSO and HMBA

The effects of anti-M-βglob-PNA on the erythroid differentiation of MEL cells were first analyzed by benzidine staining of the treated cells. The results of this experiment are presented in Fig. 3, which clearly illustrates that the MEL cells treated with DMSO (Fig. 3G and H) and HMBA (Fig. 3C and D) do not efficiently differentiate in the presence of anti-M-βglob-PNA. Fig. 3A–C and E–G shows representative experiments, while Fig. 3D and H shows the summary of 3 independent experiments, confirming a decrease in the proportion of benzidine-positive (hemoglobin-containing) PNA-treated cells.

Effects of anti-M-βglob-PNA on hemoglobin and β-globin mRNA accumulation in MEL cells treated with HMBA

The effects of anti-M-βglob-PNA on hemoglobin and β-globin mRNA accumulation were examined in the HMBA-treated cells by HPLC (for hemoglobin analysis) and RT-qPCR (for the analysis of β-globin mRNA). The results are presented in Fig. 4, which clearly illustrates that anti-M-βglob-PNA inhibited the accumulation of both Hbmajor and Hbminor hemoglobins fully in agreement with the data shown in Fig. 3. Of note, anti-M-βglob-PNA also inhibited, to a certain extent, β-globin mRNA, suggesting that the inhibition of hemoglobin production may be also associated with the lower stability of β-globin mRNA (data not shown).

Specificity of the effects of anti-M-βglob-PNA

The effects of anti-M-βglob-PNA on erythroid differentiation were also examined using human K562 cells subjected to erythroid differentiation by treatment with mithramycin (MTH). As shown in Fig. 5 no inhibitory effects of anti-M-βglob-PNA were observed on the MTH-stimulated K562-D5 cell clones, indicating high levels of specificity of the inhibitory effects of the anti-M-βglob-PNA. Furthermore, control experiments were also performed using the anti-H-γglob-PNA on HMBA- and DMSO-treated MEL cells. Of note, no inhibitory effects were observed using the anti-H-γglob-PNA, suggesting that the effects of treatment of the erythroid cells with PNAs were sequence-specific (Fig. 6).

Discussion

In the treatment of SCA, HbS appears to be an important therapeutic target, since its polymerization is responsible for the sickling of SCA red-blood cells and important adverse clinical parameters (2527). For instance, intravascular sickling in capillaries and small vessels leads to vaso-occlusion and impaired blood flow in a variety of organs and tissues (30). These conclusions are sustained by strong evidence suggesting that the induction of HbF following the treatment of SCA patients with hydroxyurea (HU) appears to be beneficial, due to the intrinsic anti-sickling properties of HbF (46,47). In any case, the inhibition of β-globin may be beneficial, allowing further reduction of sickling properties. Therefore the proof-of-principle of PNA-based methods enabling the effective inhibition of the accumulation of β-globin is of interest and of potential biomedical application.

Acknowledgements

This study was supported by a grant from the Italian Ministry of University and Research (MIUR) and from TOP-THAL (R&S project for PMI Lazio-Cup code F57I-12000010007). R.G. received a grant from Fondazione Cariparo (Cassa di Risparmio di Padova e Rovigo), CIB, from UE FP7 THALAMOSS Project (THALAssaemia Modular Stratification System for personalized therapy of β-thalassemia; 306201), from Telethon GGP10124 and from AIRC IG13575. This study was also supported by Associazione Veneta per la Lotta alla Talassemia (AVLT), Rovigo.

References

1 

Cavazzana-Calvo M, Payen E, Negre O, Wang G, Hehir K, Fusil F, Down J, Denaro M, Brady T, Westerman K, Cavallesco R, Gillet-Legrand B, Caccavelli L, Sgarra R, Maouche-Chrétien L, Bernaudin F, Girot R, Dorazio R, Mulder GJ, Polack A, Bank A, Soulier J, Larghero J, Kabbara N, Dalle B, Gourmel B, Socie G, Chrétien S, Cartier N, Aubourg P, Fischer A, Cornetta K, Galacteros F, Beuzard Y, Gluckman E, Bushman F, Hacein-Bey-Abina S and Leboulch P: Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia. Nature. 467:318–322. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Kaiser J: Gene therapy. Beta-thalassemia treatment succeeds, with a caveat. Science. 326:1468–1469. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Dong A, Rivella S and Breda L: Gene therapy for hemoglobinopathies: progress and challenges. Transl Res. 161:293–306. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Breda L, Casu C, Gardenghi S, Bianchi N, Cartegni L, Narla M, Yazdanbakhsh K, Musso M, Manwani D, Little J, Gardner LB, Kleinert DA, Prus E, Fibach E, Grady RW, Giardina PJ, Gambari R and Rivella S: Therapeutic hemoglobin levels after gene transfer in β-thalassemia mice and in hematopoietic cells of β-thalassemia and sickle cells disease patients. PLoS One. 7:e323452012. View Article : Google Scholar

5 

Zou J, Mali P, Huang X, Dowey SN and Cheng L: Site-specific gene correction of a point mutation in human iPS cells derived from an adult patient with sickle cell disease. Blood. 118:4599–4608. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Gambari R and Fibach E: Medicinal chemistry of fetal hemoglobin inducers for treatment of beta-thalassemia. Curr Med Chem. 14:199–212. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Reid ME, El Beshlawy A, Inati A, Kutlar A, Abboud MR, Haynes J Jr, Ward R, Sharon B, Taher AT, Smith W, Manwani D and Ghalie RG: A double-blind, placebo-controlled phase II study of the efficacy and safety of 2,2-dimethylbutyrate (HQK-1001), an oral fetal globin inducer, in sickle cell disease. Am J Hematol. 89:709–713. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Perrine SP, Pace BS and Faller DV: Targeted fetal hemoglobin induction for treatment of beta hemoglobinopathies. Hematol Oncol Clin North Am. 28:233–248. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Breda L, Rivella S, Zuccato C and Gambari R: Combining gene therapy and fetal hemoglobin induction for treatment of β-thalassemia. Expert Rev Hematol. 6:255–264. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Zuccato C, Breda L, Salvatori F, Breveglieri G, Gardenghi S, Bianchi N, Brognara E, Lampronti I, Borgatti M, Rivella S and Gambari R: A combined approach for β-thalassemia based on gene therapy-mediated adult hemoglobin (HbA) production and fetal hemoglobin (HbF) induction. Ann Hematol. 91:1201–1213. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Voit RA, Hendel A, Pruett-Miller SM and Porteus MH: Nuclease-mediated gene editing by homologous recombination of the human globin locus. Nucleic Acids Res. 42:1365–1378. 2014. View Article : Google Scholar :

12 

Ma N, Liao B, Zhang H, Wang L, Shan Y, Xue Y, Huang K, Chen S, Zhou X, Chen Y, Pei D and Pan G: Transcription activator-like effector nuclease (TALEN)-mediated gene correction in integration-free β-thalassemia induced pluripotent stem cells. J Biol Chem. 288:34671–34679. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Salvatori F, Breveglieri G, Zuccato C, Finotti A, Bianchi N, Borgatti M, Feriotto G, Destro F, Canella A, Brognara E, Lampronti I, Breda L, Rivella S and Gambari R: Production of beta-globin and adult hemoglobin following G418 treatment of erythroid precursor cells from homozygous beta039 thalassemia patients. Am J Hematol. 84:720–728. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Ansari SH, Shamsi TS, Ashraf M, Perveen K, Farzana T, Bohray M, Erum S and Mehboob T: Efficacy of hydroxyurea in providing transfusion independence in β-thalassemia. J Pediatr Hematol Oncol. 33:339–343. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Fibach E, Bianchi N, Borgatti M, Prus E and Gambari R: Mithramycin induces fetal hemoglobin production in normal and thalassemic human erythroid precursor cells. Blood. 102:1276–1281. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Salvador A, Brognara E, Vedaldi D, Castagliuolo I, Brun P, Zuccato C, Lampronti I and Gambari R: Induction of erythroid differentiation and increased globin mRNA production with furocoumarins and their photoproducts. J Photochem Photobiol B. 121:57–66. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Fibach E, Bianchi N, Borgatti M, Zuccato C, Finotti A, Lampronti I, Prus E, Mischiati C and Gambari R: Effects of rapamycin on accumulation of alpha-, beta- and gamma-globin mRNAs in erythroid precursor cells from beta-thalassaemia patients. Eur J Haematol. 77:437–441. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Gräslund T, Li X, Magnenat L, Popkov M and Barbas CF III: Exploring strategies for the design of artificial transcription factors: targeting sites proximal to known regulatory regions for the induction of gamma-globin expression and the treatment of sickle cell disease. J Biol Chem. 280:3707–3714. 2005. View Article : Google Scholar

19 

Wilber A, Tschulena U, Hargrove PW, Kim YS, Persons DA, Barbas CF III and Nienhuis AW: A zinc-finger transcriptional activator designed to interact with the gamma-globin gene promoters enhances fetal hemoglobin production in primary human adult erythroblasts. Blood. 115:3033–3041. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Xu XS, Hong X and Wang G: Induction of endogenous gamma-globin gene expression with decoy oligonucleotide targeting Oct-1 transcription factor consensus sequence. J Hematol Oncol. 2:152009. View Article : Google Scholar : PubMed/NCBI

21 

Gambari R: Alternative options for DNA-based experimental therapy of β-thalassemia. Expert Opin Biol Ther. 12:443–462. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Sankaran VG, Menne TF, Šćepanović D, Vergilio JA, Ji P, Kim J, Thiru P, Orkin SH, Lander ES and Lodish HF: MicroRNA-15a and -16-1 act via MYB to elevate fetal hemoglobin expression in human trisomy 13. Proc Natl Acad Sci USA. 108:1519–1524. 2001. View Article : Google Scholar

23 

Lulli V, Romania P, Morsilli O, Cianciulli P, Gabbianelli M, Testa U, Giuliani A and Marziali G: MicroRNA-486-3p regulates γ-globin expression in human erythroid cells by directly modulating BCL11A. PLoS One. 8:e604362013. View Article : Google Scholar

24 

Ma Y, Wang B, Jiang F, Wang D, Liu H, Yan Y, Dong H, Wang F, Gong B, Zhu Y, Dong L, Yin H, Zhang Z, Zhao H, Wu Z, Zhang J, Zhou J and Yu J: A feedback loop consisting of microRNA 23a/27a and the β-like globin suppressors KLF3 and SP1 regulates globin gene expression. Mol Cell Biol. 33:3994–4007. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Kaul DK, Fabry ME and Nagel RL: The pathophysiology of vascular obstruction in the sickle syndromes. Blood Rev. 10:29–44. 1996. View Article : Google Scholar : PubMed/NCBI

26 

Rodgers GP: Overview of pathophysiology and rationale for treatment of sickle cell anemia. Semin Hematol. 34:2–7. 1997.PubMed/NCBI

27 

Vekilov PG: Sickle-cell haemoglobin polymerization: is it the primary pathogenic event of sickle-cell anaemia? Br J Haematol. 139:173–184. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Vasseur C, Pissard S, Domingues-Hamdi E, Marden MC, Galactéros F and Baudin-Creuza V: Evaluation of the free α-hemoglobin pool in red blood cells: a new test providing a scale of β-thalassemia severity. Am J Hematol. 86:199–202. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Rivella S: Do not super-excess me! Blood. 119:5064–5065. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Kassim AA and DeBaun MR: Sickle cell disease, vasculopathy, and therapeutics. Annu Rev Med. 64:451–466. 2013. View Article : Google Scholar

31 

Raghupathy R and Billett HH: Promising therapies in sickle cell disease. Cardiovasc Hematol Disord Drug Targets. 9:1–8. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Steinberg MH: Pathophysiologically based drug treatment of sickle cell disease. Trends Pharmacol Sci. 27:204–210. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Gambari R: Peptide nucleic acids: a review on recent patents and technology transfer. Expert Opin Ther Pat. 24:267–294. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Nielsen PE, Egholm M, Berg RH and Buchardt O: Sequence-selective recognition of DNA by strand displacement with a thymine-substituted polyamide. Science. 254:1497–1500. 1991. View Article : Google Scholar : PubMed/NCBI

35 

Egholm M, Buchardt O, Christensen L, Behrens C, Freier SM, Driver DA, Berg RH, Kim SK, Norden B and Nielsen PE: PNA hybridizes to complementary oligonucleotides obeying the Watson-Crick hydrogen-bonding rules. Nature. 365:566–568. 1993. View Article : Google Scholar : PubMed/NCBI

36 

Gambari R: Peptide-nucleic acids (PNAs): a tool for the development of gene expression modifiers. Curr Pharm Des. 7:1839–1862. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Gambari R, Terada M, Bank A, Rifkind RA and Marks PA: Synthesis of globin mRNA in relation to the cell cycle during induced murine erythroleukemia differentiation. Proc Natl Acad Sci USA. 75:3801–3804. 1978. View Article : Google Scholar : PubMed/NCBI

38 

Gambari R, Marks PA and Rifkind RA: Murine erythroleukemia cell differentiation: relationship of globin gene expression and of prolongation of G1 to inducer effects during G1/early S. Proc Natl Acad Sci USA. 76:4511–4515. 1979. View Article : Google Scholar : PubMed/NCBI

39 

Rutherford TR and Harrison PR: Globin synthesis and erythroid differentiation in a Friend cell variant deficient in heme synthesis. Proc Natl Acad Sci USA. 76:5660–5664. 1979. View Article : Google Scholar : PubMed/NCBI

40 

Salvatori F, Cantale V, Breveglieri G, Zuccato C, Finotti A, Bianchi N, Borgatti M, Feriotto G, Destro F, Canella A, Breda L, Rivella S and Gambari R: Development of K562 cell clones expressing beta-globin mRNA carrying the beta039 thalassaemia mutation for the screening of correctors of stop-codon mutations. Biotechnol Appl Biochem. 54:41–52. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Fabbri E, Manicardi A, Tedeschi T, Sforza S, Bianchi N, Brognara E, Finotti A, Breveglieri G, Borgatti M, Corradini R, Marchelli R and Gambari R: Modulation of the biological activity of microRNA-210 with peptide nucleic acids (PNAs). ChemMedChem. 6:2192–2202. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Nastruzzi C, Cortesi R, Esposito E, Gambari R, Borgatti M, Bianchi N, Feriotto G and Mischiati C: Liposomes as carriers for DNA-PNA hybrids. J Control Release. 68:237–249. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Borgatti M, Breda L, Cortesi R, Nastruzzi C, Romanelli A, Saviano M, Bianchi N, Mischiati C, Pedone C and Gambari R: Cationic liposomes as delivery systems for double-stranded PNA-DNA chimeras exhibiting decoy activity against NF-kappaB transcription factors. Biochem Pharmacol. 64:609–616. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Brognara E, Fabbri E, Bianchi N, Finotti A, Corradini R and Gambari R: Molecular methods for validation of the biological activity of peptide nucleic acids targeting microRNAs. Methods Mol Biol. 1095:165–176. 2014. View Article : Google Scholar

45 

Brognara E, Fabbri E, Aimi F, Manicardi A, Bianchi N, Finotti A, Breveglieri G, Borgatti M, Corradini R, Marchelli R and Gambari R: Peptide nucleic acids targeting miR-221 modulate p27Kip1 expression in breast cancer MDA-MB-231 cells. Int J Oncol. 41:2119–2127. 2012.PubMed/NCBI

46 

Bhatnagar P, Keefer JR, Casella JF, Barron-Casella EA, Bean CJ, Hooper CW, Payne AB, Arking DE and Debaun MR: Association between baseline fetal hemoglobin levels and incidence of severe vaso-occlusive pain episodes in children with sickle cell anemia. Pediatr Blood Cancer. 60:E125–E127. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Rodrigue CM, Arous N, Bachir D, Smith-Ravin J, Romeo PH, Galacteros F and Garel MC: Resveratrol, a natural dietary phytoalexin, possesses similar properties to hydroxyurea towards erythroid differentiation. Br J Haematol. 113:500–507. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 35 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Montagner G, Gemmo C, Fabbri E, Manicardi A, Accardo I, Bianchi N, Finotti A, Breveglieri G, Salvatori F, Borgatti M, Borgatti M, et al: Peptide nucleic acids targeting β-globin mRNAs selectively inhibit hemoglobin production in murine erythroleukemia cells. Int J Mol Med 35: 51-58, 2015
APA
Montagner, G., Gemmo, C., Fabbri, E., Manicardi, A., Accardo, I., Bianchi, N. ... Gambari, R. (2015). Peptide nucleic acids targeting β-globin mRNAs selectively inhibit hemoglobin production in murine erythroleukemia cells. International Journal of Molecular Medicine, 35, 51-58. https://doi.org/10.3892/ijmm.2014.2005
MLA
Montagner, G., Gemmo, C., Fabbri, E., Manicardi, A., Accardo, I., Bianchi, N., Finotti, A., Breveglieri, G., Salvatori, F., Borgatti, M., Lampronti, I., Bresciani, A., Altamura, S., Corradini, R., Gambari, R."Peptide nucleic acids targeting β-globin mRNAs selectively inhibit hemoglobin production in murine erythroleukemia cells". International Journal of Molecular Medicine 35.1 (2015): 51-58.
Chicago
Montagner, G., Gemmo, C., Fabbri, E., Manicardi, A., Accardo, I., Bianchi, N., Finotti, A., Breveglieri, G., Salvatori, F., Borgatti, M., Lampronti, I., Bresciani, A., Altamura, S., Corradini, R., Gambari, R."Peptide nucleic acids targeting β-globin mRNAs selectively inhibit hemoglobin production in murine erythroleukemia cells". International Journal of Molecular Medicine 35, no. 1 (2015): 51-58. https://doi.org/10.3892/ijmm.2014.2005