Differential gene expression by lithium chloride induction of adipose-derived stem cells into neural phenotype cells

Document Type : Original Article

Authors

1 Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran

2 Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, P.O.BOX.14115-331 Tehran, Iran

3 Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O.BOX.14115-331 Tehran, Iran

Abstract

Objective(s): Adipose-derived stem cells (ADSCs), with suitable and easy access, are multipotential cells that have the ability for differentiation into other mesodermal and transdifferentiate into neural phenotype cells. In this study, Lithium chloride (LiCl) was used for in vitro transdifferentiation of rat ADSCs into neuron-like cells (NLCs).
Materials and Methods: ADSCs were isolated from the rats’ perinephric region using Dulbecco΄s Modified Eagle΄s Medium (DMEM) with Fetal Bovine Serum (FBS), cultured for 3 passages, characterized by flowcytometry and differentiation into adipogenic and osteogenic phenotypes. The ADSCs were exposed to 0.1, 0.5, 1, 1.5, 2, 5, and 10 millimolar (mM) LiCl without serum for 24 hr. The optimum dose of LiCl was selected according the maximum viability of cells. The expression of neurofilament light chain (NfL), neurofilament high chain (NfH), and nestin was evaluated by immunocytochemistry. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to evaluate the amount of synaptophysin, neurogenin-1, neuroD1, NfL, NfH, and nestin genes’ expression in ADSCs and NLCs.
Results: The optimum dose of LiCl was 1 mM in 24 hr. The transdifferentiated ADSCs showed cytoplasmic extension with synapse-like formation. Synaptophysin, neurogenin-1, neuroD1, NfL, NfH, and nestin genes were significantly expressed more in NLCs than in ADSCs.
Conclusion: LiCl can induce ADSCs into neural phenotype cells with higher expression of neural and neuronal genes.

Keywords


2014;23:573-611.
2. Cao Q, Benton RL, Whittemore SR. Stem cell repair of central nervous system injury. J Neurosci Res 2002; 68:501-510.
3. Oh JS, Park IS, Kim KN, Yoon DH, Kim SH, Ha Y. Transplantation of an adipose stem cell cluster in a spinal cord injury. Neuroreport 2012; 23:277-282.
4. Lotfy A, Salama M, Zahran F, Jones E, Badawy A, Sobh M. Characterization of mesenchymal stem cells derived from rat bone marrow and adipose tissue: a comparative study. Int J Stem Cells 2014;7:135-142.
5. Cheung YT, Lau WK, Yu MS, Lai CS, Yeung SC, So KF, et al. Effects of all-trans-retinoic acid on human SH-SY5Y neuroblastoma as in vitro model in neurotoxicity research. Neurotoxicology 2009; 30:127-135.
6. Qiu Z, Mishra A, Li M, Farnsworth SL, Guerra B, Lanford RE, et al. Marmoset induced pluripotent stem cells: Robust neural differentiation following pretreatment with dimethyl sulfoxide. Stem Cell Res 2015; 15:141-150.
7. Mohanty R, Das SK, Patri M. Modulation of benzo[a]pyrene induced anxiolytic-like behavior by retinoic acid in zebrafish: Involvement of oxidative stress and antioxidant defense system. Neurotox Res 2017; 31:493-504.
8. Takeda K, Pokorski M, Sato Y, Oyamada Y, Okada Y. Respiratory toxicity of dimethyl sulfoxide. Adv Exp Med Biol 2016; 885:89-96.
9. Kim JS, Chang MY, Yu IT, Kim JH, Lee SH, Lee YS, et al. Lithium selectively increases neuronal differentiation of hippocampal neural progenitor cells both in vitro and in vivo. J Neurochem 2004; 89: 324-336.
10. Soleimani M, Ghasemi N. Lithium chloride can induce differentiation of human immortalized RenVm cells into dopaminergic neurons. Avicenna Journal of Medical Biotechnology 2017; 9:176-180.
11. Baghaban-Eslaminejad M, Talkhabi M, Zeynali B. Effect of Lithium chloride on proliferation and bone differentiation of rat marrow-derived mesenchymal stem cells in culture. Iranian Journal of Basic Medical Sciences 2008; 11:143-151.
12. Qi L, Tang Y, He W, Pan H, Jiang W, Wang L, et al. Lithium chloride promotes neuronal differentiation of rat neural stem cells and enhances neural regeneration in Parkinson’s disease model. Cytotechnology 2017; 69:277-287.
13. Abdel-Salam OM. Stem cell therapy for Alzheimer’s disease. CNS Neurol Disord Drug Targets 2011; 10:459-485.
14. Zhang D, Wang F, Zhai X, Li XH, He XJ. Lithium promotes recovery of neurological function after spinal cord injury by inducing autophagy. Neural Regen Res 2018; 13:2191-2199.
15. Hoffmann A, Sportelli V, Ziller M, Spengler D. From the psychiatrist’s couch to induced pluripotent stem cells: Bipolar disease in a dish. Int J Mol Sci 2018; 19:1-23.
16. Mohammadshirazi A, Sadrosadat H, Jaberi R, Zareikheirabadi M, Mirsadeghi S, Naghdabadi Z, et al. Combinational therapy of lithium and human neural stem cells in rat spinal cord contusion model. J Cell Physiol 2019; 234:20742-20754.
17. Liu Z, Li R, Jiang C, Zhao S, Li W, Tang X. The neuroprotective effect of lithium chloride on cognitive impairment through glycogen synthase kinase-3beta inhibition in intracerebral hemorrhage rats. Eur J Pharmacol 2018; 840:50-59.
18. Zhang J, He L, Yang Z, Li L, Cai W. Lithium chloride promotes proliferation of neural stem cells in vitro, possibly by triggering the Wnt signaling pathway. Anim Cells Syst (Seoul) 2019; 23:32-41.
19. Þorsteinsdóttir, Lillý S. Effects of lithium on neural stem cell proliferation in vitro [master’s thesis]. University of Iceland; 2013.
20. Alizadeh T, Tiraihi T. The effect of lithium chloride on the induction of bone marrow stromal cells into neural phenotype cells. Daneshvar 2009; 79:51-56.
21. Abdanipour A. Differentiation of mesenchymal stem cells of adipose tissue into pseudo-neuronal motility cells and their transplantation to rat spinal cord injury. Iran Biomed J 2011; 15:113-121.
22. Gao M, Lu P, Bednark B, Lynam D, Conner JM, Sakamoto J, et al. Templated agarose scaffolds for the support of motor axon regeneration into sites of complete spinal cord transection. Biomaterials 2013; 34:1529-1536.
23. Ferraz FB, Fernandez JH. Selection and validation of reference house-keeping genes in the J774A1 macrophage cell line for quantitative real-time PCR. Genet Mol Res 2016; 15:15017720-15017731.
24. Ghorbani S, Tiraihi T, Soleimani M. Differentiation of mesenchymal stem cells into neuron-like cells using composite 3D scaffold combined with valproic acid induction. J Biomater Appl 2018; 32:702-715.
25. Soleimani M, Ghasemi N. Lithium Chloride can Induce Differentiation of human immortalized renvm cells into dopaminergic neurons. Avicenna J Med Biotechnol 2017; 9:176-180.
26. Neradil J, Veselska R. Nestin as a marker of cancer stem cells. Cancer Sci 2015; 106:803-811.
27. Zammit V, Brincat MR, Cassar V, Muscat-Baron Y, Ayers D, Baron B. MiRNA influences in mesenchymal stem cell commitment to neuroblast lineage development. Noncoding RNA Res 2018; 3:232-242.
28. Li P, Du F, Yuelling LW, Lin T, Muradimova RE, Tricarico R, et al. A population of Nestin-expressing progenitors in the cerebellum exhibits increased tumorigenicity. Nat Neurosci 2013; 16:1737-1744.
29. Morrison SJ. Neuronal differentiation: proneural genes inhibit gliogenesis. Curr Biol 2001; 11:R349-351.
30. Evsen L, Sugahara S, Uchikawa M, Kondoh H, Wu DK. Progression of neurogenesis in the inner ear requires inhibition of Sox2 transcription by neurogenin1 and neurod1. J Neurosci 2013; 33:3879-3890.
31. Baker NE, Brown NL. All in the family: proneural bHLH genes and neuronal diversity. Development 2018; 145: dev159426-35
32. Ross SE, Greenberg ME, Stiles CD. Basic helix-loop-helix factors in cortical development. Neuron 2003; 39:13-25.
33. Fukuda S, Taga T. Cell fate determination regulated by a transcriptional signal network in the developing mouse brain. Anat Sci Int 2005; 80:12-18.
34. Iwai R, Tabata H, Inoue M, Nomura KI, Okamoto T, Ichihashi M, et al. A Prdm8 target gene Ebf3 regulates multipolar-to-bipolar transition in migrating neocortical cells. Biochem Biophys Res Commun 2018; 495:388-394.
35. Chen W, Zhang B, Xu S, Lin R, Wang W. Lentivirus carrying the NeuroD1 gene promotes the conversion from glial cells into neurons in a spinal cord injury model. Brain Res Bull 2017; 135:143-148.
36. Matsushita M, Nakatake Y, Arai I, Ibata K, Kohda K, Goparaju SK, et al. Neural differentiation of human embryonic stem cells induced by the transgene-mediated overexpression of single transcription factors. Biochem Biophys Res Commun 2017; 490:296-301.
37. Goparaju SK, Kohda K, Ibata K, Soma A, Nakatake Y, Akiyama T, et al. Rapid differentiation of human pluripotent stem cells into functional neurons by mRNAs encoding transcription factors. Sci Rep 2017; 7:42367-97.
38. Khalfallah O, Jarjat M, Davidovic L, Nottet N, Cestele S, Mantegazza M, et al. Depletion of the fragile X mental retardation protein in embryonic stem cells alters the kinetics of neurogenesis. Stem Cells 2017; 35:374-385.
39. McLeod CM, Mauck RL. On the origin and impact of mesenchymal stem cell heterogeneity: new insights and emerging tools for single cell analysis. Eur Cell Mater 2017; 34:217-231.
40. Mo M, Wang S, Zhou Y, Li H, Wu Y. Mesenchymal stem cell subpopulations: phenotype, property and therapeutic potential. Cell Mol Life Sci 2016; 73:3311-3321.
41. Yuan A, Rao MV, Veeranna, Nixon RA. Neurofilaments and neurofilament proteins in health and disease. Cold Spring Harb Perspect Biol 2017;9: a018309-33.
42. Han Q, Lin Q, Huang P, Chen M, Hu X, Fu H, et al. Microglia-derived IL-1beta contributes to axon development disorders and synaptic deficit through p38-MAPK signal pathway in septic neonatal rats. J Neuroinflammation 2017; 14:52-70.
43. Gordon SL, Harper CB, Smillie KJ, Cousin MA. A fine balance of synaptophysin levels underlies efficient retrieval of synaptobrevin II to synaptic vesicles. PLoS One 2016; 11:e0149457-69.
44. Shapiro LP, Parsons RG, Koleske AJ, Gourley SL. Differential expression of cytoskeletal regulatory factors in the adolescent prefrontal cortex: Implications for cortical development. J Neurosci Res 2017; 95:1123-1143.
45. Glantz LA, Gilmore JH, Hamer RM, Lieberman JA, Jarskog LF. Synaptophysin and postsynaptic density protein 95 in the human prefrontal cortex from mid-gestation into early adulthood. Neuroscience 2007; 149:582-591.
46. Kageyama R, Ishibashi M, Takebayashi K, Tomita K. bHLH transcription factors and mammalian neuronal differentiation. Int J Biochem Cell Biol 1997; 29:1389-1399.
47. Guan M, Zhang Y, Huang Q, He L, Fang Q, Zhang J, et al. Fetal bovine serum inhibits neomycin-induced apoptosis of hair cell-like HEI-OC-1 cells by maintaining mitochondrial function. Am J Transl Res 2019; 11:1343-1358.
48. Kwon D, Kim JS, Cha BH, Park KS, Han I, Bae H, et al. The effect of fetal bovine serum (FBS) on efficacy of cellular reprogramming for induced pluripotent stem cell (iPSC) generation. Cell Transplant 2016; 25:1025-1042.
49. Volonte C, Ciotti MT, Merlo D. LiCl promotes survival of GABAergic neurons from cerebellum and cerebral cortex: LiCl induces survival of GABAergic neurons. Neuroscience Letters 1994; 172:6-10.
50. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002; 13:4279-4295.
51. Pereira MRJ, Pinhatti VR, Silveira MDD, Matzenbacher CA, Freitas TRO, Silva JD, et al. Isolation and characterization of mesenchymal stem/stromal cells from Ctenomys minutus. Genet Mol Biol 2018; 41:870-877.
52. Kokai LE, Marra K, Rubin JP. Adipose stem cells: biology and clinical applications for tissue repair and regeneration. Transl Res 2014; 163:399-408.