Generic placeholder image

Current Molecular Medicine

Editor-in-Chief

ISSN (Print): 1566-5240
ISSN (Online): 1875-5666

Research Article

Adipose Tissue-Mesenchymal Stem Cells Caused to Change the Methylation Status of hTERT Gene Promoter CpG Islands of Molt-4 Leukemia Cells as Cell-based Therapy

Author(s): Ezzatollah Fathi*, Soheila Montazersaheb, Somayeh Vandghanooni, Raheleh Farahzadi and Ilja Vietor

Volume 23, Issue 3, 2023

Published on: 01 March, 2022

Page: [266 - 274] Pages: 9

DOI: 10.2174/1566524022666220118103136

Price: $65

conference banner
Abstract

Background: DNA methylation was considered as prognostic information in some hematological malignancies. Previous studies have reported the in vitro and in vivo biology role of mesenchymal stem cells (MSCs) on leukemic cells. The aim of this study was to investigate the effect of MSCs on the promoter methylation status of hTERT as a catalytic subunit of telomerase enzyme.

Methods: In the experimental study, the Molt-4 leukemic cells were co-cultured with MSCs for 7 days. At the end of the co-culture period, the Molt-4 cells were collected, DNA and protein were extracted. Then methylation specific-PCR and western blotting were done for evaluating the hTERT gene promoter methylation status and cyclin D1 and hTERT protein expression, respectively. In the following, the flow cytometry was done for cell cycle distribution assay.

Results: It was found that MSCs resulted in a significant decrease in the cyclin D1 and hTERT protein expression levels. Also, MSCs caused changes in the methylation status of the CpG islands in the hTERT gene promoter region. The following results showed that MSCs caused a significant increase in the number of cells at G0/G1 phase and arrest the G0/G1 phase as well as decrease in the cell proliferation of Molt-4 cells.

Conclusion: It is concluded that co-culture of MSCs with Molt-4 cells could be involved in changing the methylation status of hTERT gene promoter, cell cycle and hTERT protein expression; it could be potentially beneficial for further investigations regarding the cell transplantation and cell-based therapy.

Keywords: Mesenchymal stem cells, hTERT gene promoter methylation status, Cell cycle distribution, Cyclin D1, Cell-based therapy, Telomerase.

[1]
Zhang HM, Zhang LS. Influence of human bone marrow mesenchymal stem cells on proliferation of chronic myeloid leukemia cells. Chin J Cancer 2009; 28(1): 29-32.
[PMID: 19448412]
[2]
Lin HD, Fong CY, Biswas A, Choolani M, Bongso A. Human umbilical cord wharton’s jelly stem cell conditioned medium induces tumoricidal effects on lymphoma cells through hydrogen peroxide mediation. J Cell Biochem 2016; 117(9): 2045-55.
[http://dx.doi.org/10.1002/jcb.25501] [PMID: 27392313]
[3]
Zimmerlin L, Donnenberg AD, Rubin JP, Basse P, Landreneau RJ, Donnenberg VS. Regenerative therapy and cancer: in vitro and in vivo studies of the interaction between adipose-derived stem cells and breast cancer cells from clinical isolates. Tissue Eng Part A 2011; 17(1-2): 93-106.
[http://dx.doi.org/10.1089/ten.tea.2010.0248] [PMID: 20673000]
[4]
Papaccio F, Paino F, Regad T, Papaccio G, Desiderio V, Tirino V. Concise review: cancer cells, cancer stem cells, and mesenchymal stem cells: Influence in cancer development. Stem Cells Transl Med 2017; 6(12): 2115-25.
[http://dx.doi.org/10.1002/sctm.17-0138] [PMID: 29072369]
[5]
Calorini L, Bianchini F. Environmental control of invasiveness and metastatic dissemination of tumor cells: The role of tumor cell-host cell interactions. Cell Commun Signal 2010; 8(1): 24-34.
[http://dx.doi.org/10.1186/1478-811X-8-24] [PMID: 20822533]
[6]
Farahzadi R, Fathi E, Mesbah-Namin SA, Zarghami N. Zinc sulfate contributes to promote telomere length extension via increasing telomerase gene expression, telomerase activity and change in the TERT gene promoter CpG island methylation status of human adipose-derived mesenchymal stem cells. PLoS One 2017; 12(11): e0188052.
[http://dx.doi.org/10.1371/journal.pone.0188052] [PMID: 29145503]
[7]
Farahzadi R, Fathi E, Mesbah-Namin SA, Zarghami N. Anti-aging protective effect of L-carnitine as clinical agent in regenerative medicine through increasing telomerase activity and change in the hTERT promoter CpG island methylation status of adipose tissue-derived mesenchymal stem cells. Tissue Cell 2018; 54: 105-13.
[http://dx.doi.org/10.1016/j.tice.2018.08.012] [PMID: 30309499]
[8]
Azouz A, Wu YL, Hillion J, et al. Epigenetic plasticity of hTERT gene promoter determines retinoid capacity to repress telomerase in maturation-resistant acute promyelocytic leukemia cells. Leukemia 2010; 24(3): 613-22.
[http://dx.doi.org/10.1038/leu.2009.283] [PMID: 20072159]
[9]
Montazersaheb S, Kazemi M, Nabat E, Nielsen PE, Hejazi MS. Downregulation of TdT expression through splicing modulation by antisense peptide nucleic acid (PNA). Curr Pharm Biotechnol 2019; 20(2): 168-78.
[http://dx.doi.org/10.2174/1389201020666190206202650] [PMID: 30727883]
[10]
Fathi E, Valipour B, Farahzadi R. Targeting the proliferation inhibition of chronic myeloid leukemia cells by bone marrow derived-mesenchymal stem cells via ERK pathway as a therapeutic strategy. Acta Med Iran 2020; 58(5): 199-206.
[http://dx.doi.org/10.18502/acta.v58i5.3952]
[11]
Adibkia K, Ehsani A, Jodaei A, Fathi E, Farahzadi R, Barzegar-Jalali M. Silver nanoparticles induce the cardiomyogenic differentiation of bone marrow derived mesenchymal stem cells via telomere length extension. Beilstein J Nanotechnol 2021; 12(1): 786-97.
[http://dx.doi.org/10.3762/bjnano.12.62] [PMID: 34395152]
[12]
Fathi E, Vandghanooni S, Montazersaheb S, Farahzadi R. Mesenchymal stem cells promote caspase-3 expression of SH-SY5Y neuroblastoma cells via reducing telomerase activity and telomere length. Iran J Basic Med Sci 2021; 24(11): 1583-9.
[13]
Heidari HR, Fathi E, Montazersaheb S, et al. Mesenchymal stem cells cause telomere length reduction of molt-4 cells via Caspase-3, BAD and P53 apoptotic pathway. Int J Mol Cell Med 2021; 10(2): 113-22.
[PMID: 34703795]
[14]
Fathi E, Vietor I. Mesenchymal stem cells promote caspase expression in Molt-4 leukemia cells via GSK-3α/β and ERK1/2 signaling pathways as a therapeutic strategy. Curr Gene Ther 2021; 21(1): 81-8.
[http://dx.doi.org/10.2174/1566523220666201005111126] [PMID: 33019931]
[15]
Tarhriz V, Eyvazi S, Musavi M, et al. Transient induction of Cdk9 in the early stage of differentiation is critical for myogenesis. J Cell Biochem 2019; 120(11): 18854-61.
[http://dx.doi.org/10.1002/jcb.29204] [PMID: 31257635]
[16]
Fathi E, Farahzadi R, Vietor I, Javanmardi S. Cardiac differentiation of bone-marrow-resident c-kit+ stem cells by L-carnitine increases through secretion of VEGF, IL6, IGF-1, and TGF- β as clinical agents in cardiac regeneration. J Biosci 2020; 45(1): 1-11.
[http://dx.doi.org/10.1007/s12038-020-00063-0] [PMID: 32713855]
[17]
Mansouri E, Tarhriz V, Yousefi V, Dilmaghani A. Intercalation and release of an anti-inflammatory drug into designed three-dimensionally layered double hydroxide nanostructure via calcination–reconstruction route. Adsorption 2020; 26: 835-42.
[http://dx.doi.org/10.1007/s10450-020-00217-4]
[18]
Fathi E, Farahzadi R, Valipour B. Alginate/gelatin encapsulation promotes NK cells differentiation potential of bone marrow resident C-kit+ hematopoietic stem cells. Int J Biol Macromol 2021; 177: 317-27.
[http://dx.doi.org/10.1016/j.ijbiomac.2021.02.131] [PMID: 33621568]
[19]
Devereux TR, Horikawa I, Anna CH, Annab LA, Afshari CA, Barrett JC. DNA methylation analysis of the promoter region of the human telomerase reverse transcriptase (hTERT) gene. Cancer Res 1999; 59(24): 6087-90.
[PMID: 10626795]
[20]
Gholizadeh-Ghaleh Aziz S, Fathi E, Rahmati-Yamchi M, Akbarzadeh A, Fardyazar Z, Pashaiasl M. An update clinical application of amniotic fluid-derived stem cells (AFSCs) in cancer cell therapy and tissue engineering. Artif Cells Nanomed Biotechnol 2017; 45(4): 765-74.
[http://dx.doi.org/10.1080/21691401.2016.1216857] [PMID: 27684534]
[21]
Tran C, Damaser MS. Stem cells as drug delivery methods: application of stem cell secretome for regeneration. Adv Drug Deliv Rev 2015; 82-83: 1-11.
[http://dx.doi.org/10.1016/j.addr.2014.10.007] [PMID: 25451858]
[22]
Alipour M, Firouzi N, Aghazadeh Z, et al. The osteogenic differentiation of human dental pulp stem cells in alginate-gelatin/Nano-hydroxyapatite microcapsules. BMC Biotechnol 2021; 21(1): 6.
[http://dx.doi.org/10.1186/s12896-020-00666-3] [PMID: 33430842]
[23]
Stellavato A, La Noce M, Corsuto L, et al. Hybrid complexes of high and low molecular weight hyaluronans highly enhance HASCs differentiation: implication for facial bioremodelling. Cell Physiol Biochem 2017; 44(3): 1078-92.
[http://dx.doi.org/10.1159/000485414] [PMID: 29179206]
[24]
Hassan Famian M, Montazer Saheb S, Montaseri A. Conditioned medium of wharton’s jelly derived stem cells can enhance the cartilage specific genes expression by chondrocytes in monolayer and mass culture systems. Adv Pharm Bull 2017; 7(1): 123-30.
[http://dx.doi.org/10.15171/apb.2017.016] [PMID: 28507946]
[25]
Karantalis V, DiFede DL, Gerstenblith G, et al. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The Prospective Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trial. Circ Res 2014; 114(8): 1302-10.
[http://dx.doi.org/10.1161/CIRCRESAHA.114.303180] [PMID: 24565698]
[26]
Cousin B, Ravet E, Poglio S, et al. Adult stromal cells derived from human adipose tissue provoke pancreatic cancer cell death both in vitro and in vivo. PLoS One 2009; 4(7): e6278.
[http://dx.doi.org/10.1371/journal.pone.0006278] [PMID: 19609435]
[27]
Cetintas VB, Aktug H, Oltulu F, Keskinoglu A, Del BE, Castello DT. The effects of mesenchymal stem cells on lymphoblastic leukemia cell proliferation. J BUON 2014; 19(4): 1006-17.
[28]
Yang C, Lei D, Ouyang W, et al. Conditioned media from human adipose tissue-derived mesenchymal stem cells and umbilical cord-derived mesenchymal stem cells efficiently induced the apoptosis and differentiation in human glioma cell lines in vitro. BioMed Res Int 2014; 2014(109389): 109389.
[http://dx.doi.org/10.1155/2014/109389] [PMID: 24971310]
[29]
Borssén M, Cullman I, Norén-Nyström U, et al. hTERT promoter methylation and telomere length in childhood acute lymphoblastic leukemia: Associations with immunophenotype and cytogenetic subgroup. Exp Hematol 2011; 39(12): 1144-51.
[http://dx.doi.org/10.1016/j.exphem.2011.08.014] [PMID: 21914494]
[30]
Galm O, Herman JG, Baylin SB. The fundamental role of epigenetics in hematopoietic malignancies. Blood Rev 2006; 20(1): 1-13.
[http://dx.doi.org/10.1016/j.blre.2005.01.006] [PMID: 16426940]
[31]
Roman-Gomez J, Jimenez-Velasco A, Agirre X, et al. CpG island methylator phenotype redefines the prognostic effect of t(12;21) in childhood acute lymphoblastic leukemia. Clin Cancer Res 2006; 12(16): 4845-50.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-2592] [PMID: 16914570]
[32]
Shin KH, Kang MK, Dicterow E, Park NH. Hypermethylation of the hTERT promoter inhibits the expression of telomerase activity in normal oral fibroblasts and senescent normal oral keratinocytes. Br J Cancer 2003; 89(8): 1473-8.
[http://dx.doi.org/10.1038/sj.bjc.6601291] [PMID: 14562019]
[33]
Guilleret I, Benhattar J. Unusual distribution of DNA methylation within the hTERT CpG island in tissues and cell lines. Biochem Biophys Res Commun 2004; 325(3): 1037-43.
[http://dx.doi.org/10.1016/j.bbrc.2004.10.137] [PMID: 15541393]
[34]
Bechter OE, Eisterer W, Dlaska M, Kühr T, Thaler J. CpG island methylation of the hTERT promoter is associated with lower telomerase activity in B-cell lymphocytic leukemia. Exp Hematol 2002; 30(1): 26-33.
[http://dx.doi.org/10.1016/S0301-472X(01)00760-3] [PMID: 11823034]
[35]
Wick M, Zubov D, Hagen G. Genomic organization and promoter characterization of the gene encoding the human telomerase reverse transcriptase (hTERT). Gene 1999; 232(1): 97-106.
[http://dx.doi.org/10.1016/S0378-1119(99)00108-0] [PMID: 10333526]
[36]
Renaud S, Loukinov D, Abdullaev Z, et al. Dual role of DNA methylation inside and outside of CTCF-binding regions in the transcriptional regulation of the telomerase hTERT gene. Nucleic Acids Res 2007; 35(4): 1245-56.
[http://dx.doi.org/10.1093/nar/gkl1125] [PMID: 17267411]
[37]
Ramlee MK, Wang J, Toh WX, Li S. Transcription regulation of the human telomerase reverse transcriptase (hTERT) gene. Genes (Basel) 2016; 7(8): 50-93.
[http://dx.doi.org/10.3390/genes7080050] [PMID: 27548225]
[38]
Deng W-G, Jayachandran G, Wu G, Xu K, Roth JA, Ji L. Tumor-specific activation of human telomerase reverses transcriptase promoter activity by activating enhancer-binding protein-2β in human lung cancer cells. J Biol Chem 2007; 282(36): 26460-70.
[http://dx.doi.org/10.1074/jbc.M610579200] [PMID: 17630431]
[39]
Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL. Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 2011; 11(8): 558-72.
[http://dx.doi.org/10.1038/nrc3090] [PMID: 21734724]
[40]
Li J, Huang X, Xie X, Wang J, Duan M. Human telomerase reverse transcriptase regulates cyclin D1 and G1/S phase transition in laryngeal squamous carcinoma. Acta Otolaryngol 2011; 131(5): 546-51.
[http://dx.doi.org/10.3109/00016489.2011.557393] [PMID: 21492065]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy