Review Topical Sections

Expression and function of nicotinic acetylcholine receptors in stem cells

  • Received: 15 June 2016 Accepted: 11 July 2016 Published: 17 July 2016
  • Nicotinic acetylcholine receptors are prototypical ligand gated ion channels typically found in muscular and neuronal tissues. Functional nicotinic acetylcholine receptors, however, have also recently been identified on other cell types, including stem cells. Activation of these receptors by the binding of agonists like choline, acetylcholine, or nicotine has been implicated in many cellular changes. In regards to stem cell function, nicotinic acetylcholine receptor activation leads to changes in stem cell proliferation, migration and differentiation potential. In this review we summarize the expression and function of known nicotinic acetylcholine receptors in different classes of stem cells including: pluripotent stem cells, mesenchymal stem cells, periodontal ligament derived stem cells, and neural progenitor cells and discuss the potential downstream effects of receptor activation on stem cell function.

    Citation: Carlos M. Carballosa, Jordan M. Greenberg, Herman S. Cheung. Expression and function of nicotinic acetylcholine receptors in stem cells[J]. AIMS Bioengineering, 2016, 3(3): 245-263. doi: 10.3934/bioeng.2016.3.245

    Related Papers:

  • Nicotinic acetylcholine receptors are prototypical ligand gated ion channels typically found in muscular and neuronal tissues. Functional nicotinic acetylcholine receptors, however, have also recently been identified on other cell types, including stem cells. Activation of these receptors by the binding of agonists like choline, acetylcholine, or nicotine has been implicated in many cellular changes. In regards to stem cell function, nicotinic acetylcholine receptor activation leads to changes in stem cell proliferation, migration and differentiation potential. In this review we summarize the expression and function of known nicotinic acetylcholine receptors in different classes of stem cells including: pluripotent stem cells, mesenchymal stem cells, periodontal ligament derived stem cells, and neural progenitor cells and discuss the potential downstream effects of receptor activation on stem cell function.


    加载中
    [1] Albuquerque EX, Pereira EF, Alkondon M, et al. (2009) Mammalian nicotinic acetylcholine receptors: from structure to function. Physiol Rev 89: 73–120. doi: 10.1152/physrev.00015.2008
    [2] Nemecz A, Prevost MS, Menny A, et al. (2016) Emerging Molecular Mechanisms of Signal Transduction in Pentameric Ligand-Gated Ion Channels. Neuron 90: 452–470. doi: 10.1016/j.neuron.2016.03.032
    [3] Wu J, Lukas RJ (2011) Naturally-expressed nicotinic acetylcholine receptor subtypes. Biochem Pharmacol 82: 800–807. doi: 10.1016/j.bcp.2011.07.067
    [4] Wang XJ, Liu YF, Wang QY, et al. (2010) Functional expression of alpha 7 nicotinic acetylcholine receptors in human periodontal ligament fibroblasts and rat periodontal tissues. Cell Tissue Res 340: 347–355. doi: 10.1007/s00441-010-0949-9
    [5] Ishizuka T, Goshima H, Ozawa A, et al. (2012) Effect of nicotine on the proliferation and differentiation of mouse induced pluripotent stem cells. Curr Med Chem 19: 5164–5169. doi: 10.2174/092986712803530494
    [6] Kim SY, Kang KL, Lee JC, et al. (2012) Nicotinic acetylcholine receptor alpha7 and beta4 subunits contribute nicotine-induced apoptosis in periodontal ligament stem cells. Mol Cells 33: 343–350. doi: 10.1007/s10059-012-2172-x
    [7] Lax P, Fucile S, Eusebi F (2002) Ca(2+) permeability of human heteromeric nAChRs expressed by transfection in human cells. Cell Calcium 32: 53–58. doi: 10.1016/S0143-4160(02)00076-3
    [8] Gerzanich V, Wang F, Kuryatov A, et al. (1998) alpha 5 Subunit alters desensitization, pharmacology, Ca++ permeability and Ca++ modulation of human neuronal alpha 3 nicotinic receptors. J Pharmacol Exp Ther 286: 311–320.
    [9] Kalamida D, Poulas K, Avramopoulou V, et al. (2007) Muscle and neuronal nicotinic acetylcholine receptors. Structure, function and pathogenicity. FEBS J 274: 3799–3845.
    [10] Lin W, Hirata N, Sekino Y, et al. (2012) Role of alpha7-nicotinic acetylcholine receptor in normal and cancer stem cells. Curr Drug Targets 13: 656–665.
    [11] Shen JX, Qin D, Wang H, et al. (2013) Roles of nicotinic acetylcholine receptors in stem cell survival/apoptosis, proliferation and differentiation. Curr Mol Med 13: 1455–1464. doi: 10.2174/15665240113139990074
    [12] Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. (1998) Embryonic stem cell lines derived from human blastocysts. Science 282: 1145–1147.
    [13] Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126: 663–676. doi: 10.1016/j.cell.2006.07.024
    [14] Schmidt R, Plath K (2012) The roles of the reprogramming factors Oct4, Sox2 and Klf4 in resetting the somatic cell epigenome during induced pluripotent stem cell generation. Genome Biol 13: 251. doi: 10.1186/gb-2012-13-10-251
    [15] Kim K, Doi A, Wen B, et al. (2010) Epigenetic memory in induced pluripotent stem cells. Nature 467: 285–290. doi: 10.1038/nature09342
    [16] Serobyan N, Jagannathan S, Orlovskaya I, et al. (2007) The cholinergic system is involved in regulation of the development of the hematopoietic system. Life Sci 80: 2352–2360. doi: 10.1016/j.lfs.2007.04.017
    [17] Ben-Yehudah A, Campanaro BM, Wakefield LM, et al. (2013) Nicotine exposure during differentiation causes inhibition of N-myc expression. Respir Res 14: 119. doi: 10.1186/1465-9921-14-119
    [18] Landgraf D, Barth M, Layer PG, et al. (2010) Acetylcholine as a possible signaling molecule in embryonic stem cells: studies on survival, proliferation and death. Chem Biol Interact 187: 115–119. doi: 10.1016/j.cbi.2010.03.007
    [19] Ishizuka T, Ozawa A, Goshima H, et al. (2012) Involvement of nicotinic acetylcholine receptor in the proliferation of mouse induced pluripotent stem cells. Life Sci 90: 637–648. doi: 10.1016/j.lfs.2012.03.014
    [20] Moser N, Mechawar N, Jones I, et al. (2007) Evaluating the suitability of nicotinic acetylcholine receptor antibodies for standard immunodetection procedures. J Neurochem 102: 479–492. doi: 10.1111/j.1471-4159.2007.04498.x
    [21] Yu J, Huang NF, Wilson KD, et al. (2009) nAChRs mediate human embryonic stem cell-derived endothelial cells: proliferation, apoptosis, and angiogenesis. PLoS One 4: e7040. doi: 10.1371/journal.pone.0007040
    [22] Ruiz JP, Pelaez D, Dias J, et al. (2012) The effect of nicotine on the mechanical properties of mesenchymal stem cells. Cell Health Cytoskelet 4: 29–35.
    [23] Zhao Z, Reece EA (2005) Nicotine-induced embryonic malformations mediated by apoptosis from increasing intracellular calcium and oxidative stress. Birth Defects Res B Dev Reprod Toxicol 74: 383–391. doi: 10.1002/bdrb.20052
    [24] Zdravkovic T, Genbacev O, LaRocque N, et al. (2008) Human embryonic stem cells as a model system for studying the effects of smoke exposure on the embryo. Reprod Toxicol 26: 86–93. doi: 10.1016/j.reprotox.2008.07.004
    [25] Hurlin PJ (2005) N-Myc functions in transcription and development. Birth Defects Res C Embryo Today 75: 340–352. doi: 10.1002/bdrc.20059
    [26] Lodish HF (2008) Molecular cell biology. New York: W.H. Freeman.
    [27] Niwa H, Miyazaki J, Smith AG (2000) Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet 24: 372–376. doi: 10.1038/74199
    [28] Yamashita J, Itoh H, Hirashima M, et al. (2000) Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature 408: 92–96. doi: 10.1038/35040568
    [29] Pittenger MF, Mackay AM, Beck SC, et al. (1999) Multilineage potential of adult human mesenchymal stem cells. Science 284: 143–147. doi: 10.1126/science.284.5411.143
    [30] Narbona-Carceles J, Vaquero J, Suarez-Sancho S, et al. (2014) Bone marrow mesenchymal stem cell aspirates from alternative sources: is the knee as good as the iliac crest? Injury 45 Suppl 4: S42–47.
    [31] Priya N, Sarcar S, Majumdar AS, et al. (2014) Explant culture: a simple, reproducible, efficient and economic technique for isolation of mesenchymal stromal cells from human adipose tissue and lipoaspirate. J Tissue Eng Regen Med 8: 706–716. doi: 10.1002/term.1569
    [32] Vangsness CT Jr., Sternberg H, Harris L (2015) Umbilical cord tissue offers the greatest number of harvestable mesenchymal stem cells for research and clinical application: a literature review of different harvest sites. Arthroscopy 31: 1836–1843. doi: 10.1016/j.arthro.2015.03.014
    [33] Devine SM, Cobbs C, Jennings M, et al. (2003) Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood 101: 2999–3001. doi: 10.1182/blood-2002-06-1830
    [34] Olson AL, McNiece IK (2015) Novel clinical uses for cord blood derived mesenchymal stromal cells. Cytotherapy 17: 796–802. doi: 10.1016/j.jcyt.2015.03.612
    [35] Hoogduijn MJ, Cheng A, Genever PG (2009) Functional nicotinic and muscarinic receptors on mesenchymal stem cells. Stem Cells Dev 18: 103–112. doi: 10.1089/scd.2008.0032
    [36] Schraufstatter IU, DiScipio RG, Khaldoyanidi SK (2009) Alpha 7 subunit of nAChR regulates migration of human mesenchymal stem cells. J Stem Cells 4: 203–215.
    [37] Zablotni A, Dakischew O, Trinkaus K, et al. (2015) Regulation of acetylcholine receptors during differentiation of bone mesenchymal stem cells harvested from human reaming debris. Int Immunopharmacol 29: 119–126. doi: 10.1016/j.intimp.2015.07.021
    [38] Michailovici I, Harrington HA, Azogui HH, et al. (2014) Nuclear to cytoplasmic shuttling of ERK promotes differentiation of muscle stem/progenitor cells. Development 141: 2611–2620. doi: 10.1242/dev.107078
    [39] Frescaline G, Bouderlique T, Huynh MB, et al. (2012) Glycosaminoglycans mimetics potentiate the clonogenicity, proliferation, migration and differentiation properties of rat mesenchymal stem cells. Stem Cell Res 8: 180–192. doi: 10.1016/j.scr.2011.09.005
    [40] Improgo MR, Scofield MD, Tapper AR, et al. (2010) The nicotinic acetylcholine receptor CHRNA5/A3/B4 gene cluster: dual role in nicotine addiction and lung cancer. Prog Neurobiol 92: 212–226. doi: 10.1016/j.pneurobio.2010.05.003
    [41] Ng TK, Carballosa CM, Pelaez D, et al. (2013) Nicotine alters MicroRNA expression and hinders human adult stem cell regenerative potential. Stem Cells Dev 22: 781–790. doi: 10.1089/scd.2012.0434
    [42] Huang CY, Pelaez D, Dominguez-Bendala J, et al. (2009) Plasticity of stem cells derived from adult periodontal ligament. Regen Med 4: 809–821. doi: 10.2217/rme.09.55
    [43] Liu J, Yu F, Sun Y, et al. (2015) Concise reviews: Characteristics and potential applications of human dental tissue-derived mesenchymal stem cells. Stem Cells 33: 627–638. doi: 10.1002/stem.1909
    [44] Menicanin D, Mrozik KM, Wada N, et al. (2014) Periodontal-ligament-derived stem cells exhibit the capacity for long-term survival, self-renewal, and regeneration of multiple tissue types in vivo. Stem Cells Dev 23: 1001–1011. doi: 10.1089/scd.2013.0490
    [45] Moshaverinia A, Xu X, Chen C, et al. (2014) Application of stem cells derived from the periodontal ligament or gingival tissue sources for tendon tissue regeneration. Biomaterials 35: 2642–2650. doi: 10.1016/j.biomaterials.2013.12.053
    [46] Zhou Z, Li B, Dong Z, et al. (2013) Nicotine deteriorates the osteogenic differentiation of periodontal ligament stem cells through alpha7 nicotinic acetylcholine receptor regulating Wnt pathway. PLoS ONE 8: e83102. doi: 10.1371/journal.pone.0083102
    [47] Criado M, Valor LM, Mulet J, et al. (2012) Expression and functional properties of alpha7 acetylcholine nicotinic receptors are modified in the presence of other receptor subunits. J Neurochem 123: 504–514. doi: 10.1111/j.1471-4159.2012.07931.x
    [48] Shu Y, Yang H, Hallberg E, et al. (1997) Molecular genetic analysis of Rts1p, a B' regulatory subunit of Saccharomyces cerevisiae protein phosphatase 2A. Mol Cell Biol 17: 3242–3253. doi: 10.1128/MCB.17.6.3242
    [49] Fridman JS, Lowe SW (2003) Control of apoptosis by p53. Oncogene 22: 9030–9040. doi: 10.1038/sj.onc.1207116
    [50] Damarla M, Parniani AR, Johnston L, et al. (2014) Mitogen-activated protein kinase-activated protein kinase 2 mediates apoptosis during lung vascular permeability by regulating movement of cleaved caspase 3. Am J Respir Cell Mol Biol 50: 932–941. doi: 10.1165/rcmb.2013-0361OC
    [51] Liu N, Shi S, Deng M, et al. (2011) High levels of beta-catenin signaling reduce osteogenic differentiation of stem cells in inflammatory microenvironments through inhibition of the noncanonical Wnt pathway. J Bone Miner Res 26: 2082–2095. doi: 10.1002/jbmr.440
    [52] Roy NS, Wang S, Jiang L, et al. (2000) In vitro neurogenesis by progenitor cells isolated from the adult human hippocampus. Nat Med 6: 271–277. doi: 10.1038/73119
    [53] Roy NS, Benraiss A, Wang S, et al. (2000) Promoter-targeted selection and isolation of neural progenitor cells from the adult human ventricular zone. J Neurosci Res 59: 321–331.
    [54] Shamloo A, Heibatollahi M, Mofrad MR (2015) Directional migration and differentiation of neural stem cells within three-dimensional microenvironments. Integr Biol (Camb) 7: 335–344. doi: 10.1039/C4IB00144C
    [55] Takarada T, Nakamichi N, Kitajima S, et al. (2012) Promoted neuronal differentiation after activation of alpha4/beta2 nicotinic acetylcholine receptors in undifferentiated neural progenitors. PLoS One 7: e46177. doi: 10.1371/journal.pone.0046177
    [56] Liu F, Xuan A, Chen Y, et al. (2014) Combined effect of nerve growth factor and brainderived neurotrophic factor on neuronal differentiation of neural stem cells and the potential molecular mechanisms. Mol Med Rep 10: 1739–1745.
    [57] Cheng Y, Zhang J, Deng L, et al. (2015) Intravenously delivered neural stem cells migrate into ischemic brain, differentiate and improve functional recovery after transient ischemic stroke in adult rats. Int J Clin Exp Pathol 8: 2928–2936.
    [58] Ivanov VN, Hei TK (2014) Radiation-induced glioblastoma signaling cascade regulates viability, apoptosis and differentiation of neural stem cells (NSC). Apoptosis 19: 1736–1754. doi: 10.1007/s10495-014-1040-x
    [59] Chang YJ, Su HL, Hsu LF, et al. (2015) Isolation of Human Neural Stem Cells from the Amniotic Fluid with Diagnosed Neural Tube Defects. Stem Cells Dev 24: 1740–1750. doi: 10.1089/scd.2014.0516
    [60] Wang C, Luan Z, Yang Y, et al. (2015) High purity of human oligodendrocyte progenitor cells obtained from neural stem cells: suitable for clinical application. J Neurosci Methods 240: 61–66. doi: 10.1016/j.jneumeth.2014.10.017
    [61] Weaver CL, Cui XT (2015) Directed Neural Stem Cell Differentiation with a Functionalized Graphene Oxide Nanocomposite. Adv Healthc Mater 4: 1408–1416. doi: 10.1002/adhm.201500056
    [62] Marei HE, Lashen S, Farag A, et al. (2015) Human olfactory bulb neural stem cells mitigate movement disorders in a rat model of Parkinson's disease. J Cell Physiol 230: 1614–1629. doi: 10.1002/jcp.24909
    [63] Zhongling F, Gang Z, Lei Y (2009) Neural stem cells and Alzheimer's disease: challenges and hope. Am J Alzheimers Dis Other Demen 24: 52–57. doi: 10.1177/1533317508327587
    [64] Campbell NR, Fernandes CC, Halff AW, et al. (2010) Endogenous signaling through alpha7-containing nicotinic receptors promotes maturation and integration of adult-born neurons in the hippocampus. J Neurosci 30: 8734–8744. doi: 10.1523/JNEUROSCI.0931-10.2010
    [65] Adams CE, Broide RS, Chen Y, et al. (2002) Development of the alpha7 nicotinic cholinergic receptor in rat hippocampal formation. Brain Res Dev Brain Res 139: 175–187. doi: 10.1016/S0165-3806(02)00547-3
    [66] Quik M, Polonskaya Y, Gillespie A, et al. (2000) Localization of nicotinic receptor subunit mRNAs in monkey brain by in situ hybridization. J Comp Neurol 425: 58–69.
    [67] Lee H, Park JR, Yang J, et al. (2014) Nicotine inhibits the proliferation by upregulation of nitric oxide and increased HDAC1 in mouse neural stem cells. In Vitro Cell Dev Biol Anim 50: 731–739. doi: 10.1007/s11626-014-9763-0
    [68] He N, Wang Z, Wang Y, et al. (2013) ZY-1, a novel nicotinic analog, promotes proliferation and migration of adult hippocampal neural stem/progenitor cells. Cell Mol Neurobiol 33: 1149–1157. doi: 10.1007/s10571-013-9981-0
    [69] Balaraman S, Winzer-Serhan UH, Miranda RC (2012) Opposing actions of ethanol and nicotine on microRNAs are mediated by nicotinic acetylcholine receptors in fetal cerebral cortical-derived neural progenitor cells. Alcohol Clin Exp Res 36: 1669–1677. doi: 10.1111/j.1530-0277.2012.01793.x
    [70] Visvader JE, Lindeman GJ (2008) Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8: 755–768. doi: 10.1038/nrc2499
    [71] Hirata N, Sekino Y, Kanda Y (2010) Nicotine increases cancer stem cell population in MCF-7 cells. Biochem Biophys Res Commun 403: 138–143. doi: 10.1016/j.bbrc.2010.10.134
    [72] Scherl C, Schafer R, Schlabrakowski A, et al. (2016) Nicotinic Acetylcholine Receptors in Head and Neck Cancer and Their Correlation to Tumor Site and Progression. ORL J Otorhinolaryngol Relat Spec 78: 151–158. doi: 10.1159/000445781
    [73] Tu CC, Huang CY, Cheng WL, et al. (2016) The alpha7-nicotinic acetylcholine receptor mediates the sensitivity of gastric cancer cells to taxanes. Tumour Biol 37: 4421–4428. doi: 10.1007/s13277-015-4260-y
    [74] Chen CM, Chou HC, Huang LT (2015) Maternal nicotine exposure during gestation and lactation induces kidney injury and fibrosis in rat offspring. Pediatr Res 77: 56–63. doi: 10.1038/pr.2014.148
    [75] Dahlstrom A, Ebersjo C, Lundell B (2008) Nicotine in breast milk influences heart rate variability in the infant. Acta Paediatr 97: 1075–1079. doi: 10.1111/j.1651-2227.2008.00785.x
    [76] Paccola CC, Miraglia SM (2016) Prenatal and lactation nicotine exposure affects Sertoli cell and gonadotropin levels in rats. Reproduction 151: 117–133. doi: 10.1530/REP-15-0135
    [77] Maritz GS (2002) Maternal nicotine exposure during gestation and lactation of rats induce microscopic emphysema in the offspring. Exp Lung Res 28: 391–403. doi: 10.1080/01902140290092010
    [78] Maritz GS, Dennis H (1998) Maternal nicotine exposure during gestation and lactation interferes with alveolar development in the neonatal lung. Reprod Fertil Dev 10: 255–261. doi: 10.1071/R98036
    [79] Wong MK, Holloway AC, Hardy DB (2016) Nicotine Directly Induces Endoplasmic Reticulum Stress Response in Rat Placental Trophoblast Giant Cells. Toxicol Sci 151: 23–34.
    [80] Wong MK, Nicholson CJ, Holloway AC, et al. (2015) Maternal nicotine exposure leads to impaired disulfide bond formation and augmented endoplasmic reticulum stress in the rat placenta. PLoS ONE 10: e0122295. doi: 10.1371/journal.pone.0122295
  • Reader Comments
  • © 2016 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(5200) PDF downloads(1249) Cited by(2)

Article outline

Figures and Tables

Figures(1)  /  Tables(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog