DOI QR코드

DOI QR Code

Kir4.1 is coexpressed with stemness markers in activated astrocytes in the injured brain and a Kir4.1 inhibitor BaCl2 negatively regulates neurosphere formation in culture

  • Kwon, Jae-Kyung (Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine) ;
  • Choi, Dong-Joo (Department of Pharmacology, Ajou University School of Medicine) ;
  • Yang, Haijie (Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine) ;
  • Ko, Dong Wan (Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine) ;
  • Jou, Ilo (Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine) ;
  • Park, Sang Myun (Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine) ;
  • Joe, Eun-Hye (Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine)
  • Received : 2021.05.09
  • Accepted : 2021.09.07
  • Published : 2021.11.01

Abstract

Astrocytes are activated in response to brain damage. Here, we found that expression of Kir4.1, a major potassium channel in astrocytes, is increased in activated astrocytes in the injured brain together with upregulation of the neural stem cell markers, Sox2 and Nestin. Expression of Kir4.1 was also increased together with that of Nestin and Sox2 in neurospheres formed from dissociated P7 mouse brains. Using the Kir4.1 blocker BaCl2 to determine whether Kir4.1 is involved in acquisition of stemness, we found that inhibition of Kir4.1 activity caused a concentration-dependent increase in sphere size and Sox2 levels, but had little effect on Nestin levels. Moreover, induction of differentiation of cultured neural stem cells by withdrawing epidermal growth factor and fibroblast growth factor from the culture medium caused a sharp initial increase in Kir4.1 expression followed by a decrease, whereas Sox2 and Nestin levels continuously decreased. Inhibition of Kir4.1 had no effect on expression levels of Sox2 or Nestin, or the astrocyte and neuron markers glial fibrillary acidic protein and β-tubulin III, respectively. Taken together, these results indicate that Kir4.1 may control gain of stemness but not differentiation of stem cells.

Keywords

Acknowledgement

This work was supported by grants funded by the Korean government (NRF-2017R1A2B3011281 and NRF-2016M3C7A1905072), and a grant from KOSEF through the Chronic Inflammatory Disease Research Center at Ajou University (NRF-2012R1A5A048183) to EJ.

References

  1. Kofuji P, Ceelen P, Zahs KR, Surbeck LW, Lester HA, Newman EA. Genetic inactivation of an inwardly rectifying potassium channel (Kir4.1 subunit) in mice: phenotypic impact in retina. J Neurosci. 2000;20:5733-5740. https://doi.org/10.1523/jneurosci.20-15-05733.2000
  2. Poopalasundaram S, Knott C, Shamotienko OG, Foran PG, Dolly JO, Ghiani CA, Gallo V, Wilkin GP. Glial heterogeneity in expression of the inwardly rectifying K+ channel, Kir4.1, in adult rat CNS. Glia. 2000;30:362-372. https://doi.org/10.1002/(SICI)1098-1136(200006)30:4<362::AID-GLIA50>3.0.CO;2-4
  3. Yakushigawa H, Tokunaga Y, Inanobe A, Kani K, Kurachi Y, Maeda T. A novel junction-like membrane complex in the optic nerve astrocyte of the Japanese macaque with a possible relation to a potassium ion channel. Anat Rec. 1998;250:465-474. https://doi.org/10.1002/(SICI)1097-0185(199804)250:4<465::AID-AR10>3.0.CO;2-M
  4. Nagelhus EA, Mathiisen TM, Ottersen OP. Aquaporin-4 in the central nervous system: cellular and subcellular distribution and coexpression with KIR4.1. Neuroscience. 2004;129:905-913. https://doi.org/10.1016/j.neuroscience.2004.08.053
  5. Djukic B, Casper KB, Philpot BD, Chin LS, McCarthy KD. Conditional knock-out of Kir4.1 leads to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced shortterm synaptic potentiation. J Neurosci. 2007;27:11354-11365. https://doi.org/10.1523/JNEUROSCI.0723-07.2007
  6. Cui Y, Yang Y, Ni Z, Dong Y, Cai G, Foncelle A, Ma S, Sang K, Tang S, Li Y, Shen Y, Berry H, Wu S, Hu H. Astroglial Kir4.1 in the lateral habenula drives neuronal bursts in depression. Nature. 2018;554:323-327. https://doi.org/10.1038/nature25752
  7. Tong X, Ao Y, Faas GC, Nwaobi SE, Xu J, Haustein MD, Anderson MA, Mody I, Olsen ML, Sofroniew MV, Khakh BS. Astrocyte Kir4.1 ion channel deficits contribute to neuronal dysfunction in Huntington's disease model mice. Nat Neurosci. 2014;17:694-703. https://doi.org/10.1038/nn.3691
  8. Srivastava R, Aslam M, Kalluri SR, Schirmer L, Buck D, Tackenberg B, Rothhammer V, Chan A, Gold R, Berthele A, Bennett JL, Korn T, Hemmer B. Potassium channel KIR4.1 as an immune target in multiple sclerosis. N Engl J Med. 2012;367:115-123. https://doi.org/10.1056/NEJMoa1110740
  9. Urrego D, Tomczak AP, Zahed F, Stuhmer W, Pardo LA. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130094. https://doi.org/10.1098/rstb.2013.0094
  10. Yasuda T, Bartlett PF, Adams DJ. Kir and Kv channels regulate electrical properties and proliferation of adult neural precursor cells. Mol Cell Neurosci. 2008;37:284-297. https://doi.org/10.1016/j.mcn.2007.10.003
  11. Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005;50:427-434. https://doi.org/10.1002/glia.20207
  12. Eng LF, Ghirnikar RS. GFAP and astrogliosis. Brain Pathol. 1994;4:229-237. https://doi.org/10.1111/j.1750-3639.1994.tb00838.x
  13. Kernie SG, Erwin TM, Parada LF. Brain remodeling due to neuronal and astrocytic proliferation after controlled cortical injury in mice. J Neurosci Res. 2001;66:317-326. https://doi.org/10.1002/jnr.10013
  14. Duan CL, Liu CW, Shen SW, Yu Z, Mo JL, Chen XH, Sun FY. Striatal astrocytes transdifferentiate into functional mature neurons following ischemic brain injury. Glia. 2015;63:1660-1670. https://doi.org/10.1002/glia.22837
  15. Gotz M, Sirko S, Beckers J, Irmler M. Reactive astrocytes as neural stem or progenitor cells: in vivo lineage, in vitro potential, and genome-wide expression analysis. Glia. 2015;63:1452-1468. https://doi.org/10.1002/glia.22850
  16. Magnusson JP, Goritz C, Tatarishvili J, Dias DO, Smith EM, Lindvall O, Kokaia Z, Frisen J. A latent neurogenic program in astrocytes regulated by Notch signaling in the mouse. Science. 2014;346:237-241. https://doi.org/10.1126/science.346.6206.237
  17. Sirko S, Behrendt G, Johansson PA, Tripathi P, Costa M, Bek S, Heinrich C, Tiedt S, Colak D, Dichgans M, Fischer IR, Plesnila N, Staufenbiel M, Haass C, Snapyan M, Saghatelyan A, Tsai LH, Fischer A, Grobe K, Dimou L, et al. Reactive glia in the injured brain acquire stem cell properties in response to sonic hedgehog. [corrected]. Cell Stem Cell. 2013;12:426-439. https://doi.org/10.1016/j.stem.2013.01.019
  18. Shimada IS, LeComte MD, Granger JC, Quinlan NJ, Spees JL. Self-renewal and differentiation of reactive astrocyte-derived neural stem/progenitor cells isolated from the cortical peri-infarct area after stroke. J Neurosci. 2012;32:7926-7940. https://doi.org/10.1523/JNEUROSCI.4303-11.2012
  19. Buffo A, Rite I, Tripathi P, Lepier A, Colak D, Horn AP, Mori T, Gotz M. Origin and progeny of reactive gliosis: a source of multipotent cells in the injured brain. Proc Natl Acad Sci U S A. 2008;105:3581-3586. https://doi.org/10.1073/pnas.0709002105
  20. Zappone MV, Galli R, Catena R, Meani N, De Biasi S, Mattei E, Tiveron C, Vescovi AL, Lovell-Badge R, Ottolenghi S, Nicolis SK. Sox2 regulatory sequences direct expression of a (beta)-geo transgene to telencephalic neural stem cells and precursors of the mouse embryo, revealing regionalization of gene expression in CNS stem cells. Development. 2000;127:2367-2382. https://doi.org/10.1242/dev.127.11.2367
  21. Bani-Yaghoub M, Tremblay RG, Lei JX, Zhang D, Zurakowski B, Sandhu JK, Smith B, Ribecco-Lutkiewicz M, Kennedy J, Walker PR, Sikorska M. Role of Sox2 in the development of the mouse neocortex. Dev Biol. 2006;295:52-66. https://doi.org/10.1016/j.ydbio.2006.03.007
  22. Grazioli S, Pugin J. Mitochondrial damage-associated molecular patterns: from inflammatory signaling to human diseases. Front Immunol. 2018;9:832. https://doi.org/10.3389/fimmu.2018.00832
  23. Schaefer L. Complexity of danger: the diverse nature of damage-associated molecular patterns. J Biol Chem. 2014;289:35237-35245. https://doi.org/10.1074/jbc.R114.619304
  24. Le Feuvre RA, Brough D, Touzani O, Rothwell NJ. Role of P2X7 receptors in ischemic and excitotoxic brain injury in vivo. J Cereb Blood Flow Metab. 2003;23:381-384. https://doi.org/10.1097/01.WCB.0000048519.34839.97
  25. Zheng LM, Zychlinsky A, Liu CC, Ojcius DM, Young JD. Extracellular ATP as a trigger for apoptosis or programmed cell death. J Cell Biol. 1991;112:279-288. https://doi.org/10.1083/jcb.112.2.279
  26. Yang H, An J, Choi I, Lee K, Park SM, Jou I, Joe EH. Region-specific astrogliosis: differential vessel formation contributes to different patterns of astrogliosis in the cortex and striatum. Mol Brain. 2020;13:103. https://doi.org/10.1186/s13041-020-00642-0
  27. Jeong HK, Ji KM, Min KJ, Choi I, Choi DJ, Jou I, Joe EH. Astrogliosis is a possible player in preventing delayed neuronal death. Mol Cells. 2014;37:345-355. https://doi.org/10.14348/MOLCELLS.2014.0046
  28. Jeong HK, Ji KM, Kim B, Kim J, Jou I, Joe EH. Inflammatory responses are not sufficient to cause delayed neuronal death in ATPinduced acute brain injury. PLoS One. 2010;5:e13756. https://doi.org/10.1371/journal.pone.0013756
  29. Suzuki S, Namiki J, Shibata S, Mastuzaki Y, Okano H. The neural stem/progenitor cell marker nestin is expressed in proliferative endothelial cells, but not in mature vasculature. J Histochem Cytochem. 2010;58:721-730. https://doi.org/10.1369/jhc.2010.955609
  30. Lendahl U, Zimmerman LB, McKay RD. CNS stem cells express a new class of intermediate filament protein. Cell. 1990;60:585-595. https://doi.org/10.1016/0092-8674(90)90662-x
  31. Olsen ML, Sontheimer H. Functional implications for Kir4.1 channels in glial biology: from K+ buffering to cell differentiation. J Neurochem. 2008;107:589-601. https://doi.org/10.1111/j.1471-4159.2008.05615.x
  32. Higashimori H, Sontheimer H. Role of Kir4.1 channels in growth control of glia. Glia. 2007;55:1668-1679. https://doi.org/10.1002/glia.20574
  33. Lan JY, Williams C, Levin M, Black LD 3rd. Depolarization of cellular resting membrane potential promotes neonatal cardiomyocyte proliferation in vitro. Cell Mol Bioeng. 2014;7:432-445. https://doi.org/10.1007/s12195-014-0346-7
  34. Frederiksen K, McKay RD. Proliferation and differentiation of rat neuroepithelial precursor cells in vivo. J Neurosci. 1988;8:1144-1151. https://doi.org/10.1523/JNEUROSCI.08-04-01144.1988
  35. Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003;17:126-140. https://doi.org/10.1101/gad.224503
  36. Goldshmit Y, Frisca F, Pinto AR, Pebay A, Tang JK, Siegel AL, Kaslin J, Currie PD. Fgf2 improves functional recovery-decreasing gliosis and increasing radial glia and neural progenitor cells after spinal cord injury. Brain Behav. 2014;4:187-200. https://doi.org/10.1002/brb3.172
  37. Catterall WA. Voltage-gated calcium channels. Cold Spring Harb Perspect Biol. 2011;3:a003947. https://doi.org/10.1101/cshperspect.a003947
  38. Man JHK, Groenink L, Caiazzo M. Cell reprogramming approaches in gene- and cell-based therapies for Parkinson's disease. J Control Release. 2018;286:114-124. https://doi.org/10.1016/j.jconrel.2018.07.017