Review Article
No access
Published Online: 1 June 2017

The Role of NMDA Receptors in Neural Stem Cell Proliferation and Differentiation

Publication: Stem Cells and Development
Volume 26, Issue Number 11

Abstract

N-methyl-d-aspartate receptors (NMDARs) are expressed abundantly in the brain and play an important role in the regulation of neuronal development, learning, memory, neurodegenerative diseases, and neurogenesis, all of which are extensively discussed in reviews published elsewhere. While the number of research articles on the developmental and neurological cues that trigger NMDAR localization across a developing neuron and the role of the NMDAR in embryonic and adult stem cell proliferation and differentiation have been growing dramatically over the past few years, there is not a single review on this latter general topic. In the proposed review, we will summarize the current understanding of NMDARs in stem cell biology and their involvement in pathophysiological processes of neuron development, especially during early neuronal development (immature neurons) and differentiation.

Get full access to this article

View all available purchase options and get full access to this article.

References

1.
Sheng M, Cummings J, Roldan LA, Jan YN and Jan LY. (1994). Changing subunit composition of heteromeric NMDA receptors during development of rat cortex. Nature 368:144–147.
2.
Iulianella A, Sharma M, Durnin M, Vanden Heuvel GB and Trainor PA. (2008). Cux2 (Cutl2) integrates neural progenitor development with cell-cycle progression during spinal cord neurogenesis. Development 135:729–741.
3.
Englund C, Fink A, Lau C, Pham D, Daza RA, Bulfone A, Kowalczyk T and Hevner RF. (2005). Pax6, Tbr2, and Tbr1 are expressed sequentially by radial glia, intermediate progenitor cells, and postmitotic neurons in developing neocortex. J Neurosci 25:247–251.
4.
Gotz M, Stoykova A and Gruss P. (1998). Pax6 controls radial glia differentiation in the cerebral cortex. Neuron 21:1031–1044.
5.
Williams K, Russell SL, Shen YM and Molinoff PB. (1993). Developmental switch in the expression of NMDA receptors occurs in vivo and in vitro. Neuron 10:267–278.
6.
Spalloni A, Nutini M and Longone P. (2013).Role of the N-methyl-d-aspartate receptors complex in amyotrophic lateral sclerosis. Biochim Biophys Acta 1832:312–322.
7.
Reiner A, Albin RL, Anderson KD, D'Amato CJ, Penney JB and Young AB. (1988). Differential loss of striatal projection neurons in Huntington disease. Proc Natl Acad Sci U S A 85:5733–5737.
8.
Tang TT, Yang F, Chen BS, Lu Y, Ji Y, Roche KW and Lu B. (2009). Dysbindin regulates hippocampal LTP by controlling NMDA receptor surface expression. Proc Natl Acad Sci 106:21395–21400.
9.
Majed HH, Chandran S, Niclou SP, Nicholas RS, Wilkins A, Wing MG, Rhodes KE, Spillantini MG and Compston A. (2006) A novel role for Sema3A in neuroprotection from injury mediated by activated microglia. J Neurosci 26:1730–1738.
10.
Berry N, Gursel DB and Boockvar JA. (2011). Direct conversion of human fibroblasts to functional neurons in one step. Neurosurgery 69:N18.
11.
Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Südhof TC and Wernig M. (2010). Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463:1035–1041.
12.
Perrier AL, Tabar V, Barberi T, Rubio ME, Bruses J, Topf N, Harrison NL and Studer L. (2004) Derivation of midbrain dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci U S A 101:12543–12548.
13.
Haubensak W, Attardo A, Denk W and Huttner WB. (2004). Neurons arise in the basal neuroepithelium of the early mammalian telencephalon: a major site of neurogenesis. Proc Natl Acad Sci U S A 101:3196–3201.
14.
Mizutani K, Yoon K, Dang L, Tokunaga A and Gaiano N. (2007). Differential Notch signalling distinguishes neural stem cells from intermediate progenitors. Nature 449:351–355.
15.
Nelson BR, Hodge RD, Bedogni F and Hevner RF. (2013). Dynamic interactions between intermediate neurogenic progenitors and radial glia in embryonic mouse neocortex: potential role in Dll1-Notch signaling. J Neurosci 33:9122–9139.
16.
Toresson H, Mata de Urquiza A, Fagerström C, Perlmann T and Campbell K. (1999). Retinoids are produced by glia in the lateral ganglionic eminence and regulate striatal neuron differentiation. Development 126:1317–1326.
17.
Wang TF, Ding CN, Wang GS, Luo SC, Lin YL, Ruan Y, Hevner R, Rubenstein JL and Hsueh YP. (2004). Identification of Tbr-1/CASK complex target genes in neurons. J Neurochem 91:1483–1492.
18.
Muth-Köhne E, Pachernegg S, Karus M, Faissner A and Hollmann M. (2010). Expression of NMDA receptors and Ca2+-impermeable AMPA receptors requires neuronal differentiation and allows discrimination between two different types of neural stem cells. Cell Physiol Biochem 26:935–946.
19.
Komuro H and Kumada T. (2005). Ca2+ transients control CNS neuronal migration. Cell Calcium 37:387–393.
20.
Schwab MH, Bartholomae A, Heimrich B, Feldmeyer D, Druffel-Augustin S, Goebbels S, Naya FJ, Zhao S, Frotscher M, Tsai MJ and Nave KA. (2000). Neuronal basic helix-loop-helix proteins (NEX and BETA2/Neuro D) regulate terminal granule cell differentiation in the hippocampus. J Neurosci 20:3714–3724.
21.
Hodge RD and Hevner RF. (2011). Expression and actions of transcription factors in adult hippocampal neurogenesis. Dev Neurobiol 71:680–689.
22.
Takita M, Kaneko H, Suzuki SS and Akamatsu M. (1997). Lasting effect of NO on glutamate release in rat striatum revealed by continuous brain dialysis. Neuroreport 8:567–570.
23.
Behar TN, Scott CA, Greene CL, Wen X, Smith SV, Maric D, Liu QY, Colton CA and Barker JL. (1999). kGlutamate acting at NMDA receptors stimulates embryonic cortical neuronal migration. J Neurosci 19:4449–4461.
24.
Tremblay M, Fugère V, Tsui J, Schohl A, Tavakoli A, Travençolo BA, Costa Lda F and Ruthazer ES. (2009). Regulation of radial glial motility by visual experience. J Neurosci 29:14066–14076.
25.
López T, López-Colomé AM and Ortega A. (1997). NMDA receptors in cultured radial glia. FEBS Lett 405:245–248.
26.
Horak M, Petralia RS, Kaniakova M and Sans N. (2014). ER to synapse trafficking of NMDA receptors. Front Cell Neurosci 8:394.
27.
Zhang Y and Hu W. (2012). NFkappaB signaling regulates embryonic and adult neurogenesis. Front Biol (Beijing) 7(4): p. 10.1007/s11515-012-1233-z.
28.
Nickols JC, Valentine W, Kanwal S and Carter BD. (2003). Activation of the transcription factor NF-kappaB in Schwann cells is required for peripheral myelin formation. Nat Neurosci 6:161–167.
29.
Kumar DU, Nagaraj R and Devaraj H. (2012). Immunolocalization of Notch1, Hes1, and Nf-Kb in the murine hippocampal subgranular zone (SGZ): possible role of the Notch pathway in the maintenance of the sgz neural stem cell population. Neurophysiology 44:208–215.
30.
O'Neill LA and Kaltschmidt C. (1997). NF-kappa B: a crucial transcription factor for glial and neuronal cell function. Trends Neurosci 20:252–258.
31.
Minatohara K, Ichikawa SH, Seki T, Fujiyoshi Y and Doi T. (2013). Ligand binding of PDZ domains has various roles in the synaptic clustering of SAP102 and PSD-95. Neurosci Lett 533:44–49.
32.
Sans N, Wang PY, Du Q, Petralia RS, Wang YX, Nakka S, Blumer JB, Macara IG and Wenthold RJ. (2005). mPins modulates PSD-95 and SAP102 trafficking and influences NMDA receptor surface expression. Nat Cell Biol 7:1179–1190.
33.
Lau CG and Zukin RS. (2007). NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders. Nat Rev Neurosci 8:413–426.
34.
Sans N, Petralia RS, Wang YX, Blahos J II, Hell JW and Wenthold RJ. (2000). A developmental change in NMDA receptor-associated proteins at hippocampal synapses. J Neurosci 20:1260–1271.
35.
Elias GM, Elias LA, Apostolides PF, Kriegstein AR and Nicoll RA. (2008). Differential trafficking of AMPA and NMDA receptors by SAP102 and PSD-95 underlies synapse development. Proc Natl Acad Sci U S A 105:20953–20958.
36.
Fourie C, Li D and Montgomery JM. (2014). The anchoring protein SAP97 influences the trafficking and localisation of multiple membrane channels. Biochim Biophys Acta 1838:589–594.
37.
Jeyifous O, Waites CL, Specht CG, Fujisawa S, Schubert M, Lin EI, Marshall J, Aoki C, de Silva T, et al. (2009). SAP97 and CASK mediate sorting of NMDA receptors through a previously unknown secretory pathway. Nat Neurosci 12:1011–1019.
38.
Howard MA, Elias GM, Elias LA, Swat W and Nicoll RA. (2010). The role of SAP97 in synaptic glutamate receptor dynamics. Proc Natl Acad Sci U S A 107:3805–3810.
39.
Venere M, Han YG, Bell R, Song JS, Alvarez-Buylla A and Blelloch R. (2012). Sox1 marks an activated neural stem/progenitor cell in the hippocampus. Development 139:3938–3949.
40.
Kim DS, Lee DR, Kim HS, Yoo JE, Jung SJ, Lim BY, Jang J, Kang HC, You S, et al. (2012). highly pure and expandable PSA-NCAM-positive neural precursors from human ESC and iPSC-derived neural rosettes. PLoS One 7:e39715.
41.
Park D, Xiang AP, Mao FF, Zhang L, Di CG, Liu XM, Shao Y, Ma BF, Lee JH, et al. (2010). Nestin is required for the proper self-renewal of neural stem cells. Stem Cells 28:2162–2171.
42.
Walker TL, Yasuda T, Adams DJ and Bartlett PF. (2007). The doublecortin-expressing population in the developing and adult brain contains multipotential precursors in addition to neuronal-lineage cells. J Neurosci 27:3734–3742.
43.
Sakamoto K, Hiraiwa M, Saito M, Nakahara T, Sato Y, Nagao T and Ishii K. (2010). Protective effect of all-trans retinoic acid on NMDA-induced neuronal cell death in rat retina. Eur J Pharmacol 635:56–61.
44.
Bading H, Ginty DD and Greenberg ME. (1993). Regulation of gene expression in hippocampal neurons by distinct calcium signaling pathways. Science 260:181–186.
45.
Ghosh A and Greenberg ME. (1995). Calcium signaling in neurons: molecular mechanisms and cellular consequences. Science 268:239–247.
46.
Wang PY, Petralia RS, Wang YX, Wenthold RJ and Brenowitz SD. (2011). Functional NMDA receptors at axonal growth cones of young hippocampal neurons. J Neurosci 31:9289–9297.
47.
Martel MA, Wyllie DJ and Hardingham GE. (2009). In developing hippocampal neurons, NR2B-containing N-methyl-D-aspartate receptors (NMDARs) can mediate signaling to neuronal survival and synaptic potentiation, as well as neuronal death. Neuroscience 158:334–343.
48.
Luk KC, Kennedy TE and Sadikot AF. (2003). Glutamate promotes proliferation of striatal neuronal progenitors by an NMDA receptor-mediated mechanism. J Neurosci 23:2239–2250.
49.
Vastagh C, Gardoni F, Bagetta V, Stanic J, Zianni E, Giampà C, Picconi B, Calabresi P and Di Luca M. (2012). N-methyl-D-aspartate (NMDA) receptor composition modulates dendritic spine morphology in striatal medium spiny neurons. J Biol Chem 287:18103–18114.
50.
Logan SM, Partridge JG, Matta JA, Buonanno A and Vicini S. (2007). Long-lasting NMDA receptor-mediated EPSCs in mouse striatal medium spiny neurons. J Neurophysiol 98:2693–2704.
51.
Peng TI, Jou MJ, Sheu SS and Greenamyre JT. (1998). Visualization of NMDA receptor-induced mitochondrial calcium accumulation in striatal neurons. Exp Neurol 149:1–12.
52.
Marco S, Giralt A, Petrovic MM, Pouladi MA, Martínez-Turrillas R, Martínez-Hernández J, Kaltenbach LS, Torres-Peraza J, Graham RK, et al. (2013). Suppressing aberrant GluN3A expression rescues synaptic and behavioral impairments in Huntington's disease models. Nat Med 19:1030–1038.
53.
Fan MM and Raymond LA (2007). N-methyl-D-aspartate (NMDA) receptor function and excitotoxicity in Huntington's disease. Prog Neurobiol 81:272–293.
54.
Krapivinsky G, Krapivinsky L, Manasian Y, Ivanov A, Tyzio R, Pellegrino C, Ben-Ari Y, Clapham DE and Medina I. (2003). The NMDA receptor is coupled to the ERK pathway by a direct interaction between NR2B and RasGRF1. Neuron 40:775–784.
55.
Koponen S, Kurkinen K, Akerman KE, Mochly-Rosen D, Chan PH and Koistinaho J. (2003). Prevention of NMDA-induced death of cortical neurons by inhibition of protein kinase Czeta. J Neurochem 86:442–450.
56.
Liu Y, Wong TP, Aarts M, Rooyakkers A, Liu L, Lai TW, Wu DC, Lu J, Tymianski M, Craig AM and Wang YT. (2007). NMDA receptor subunits have differential roles in mediating excitotoxic neuronal death both in vitro and in vivo. J Neurosci 27:2846–2857.
57.
Luscher C and Malenka RC. (2012). NMDA receptor-dependent long-term potentiation and long-term depression (LTP/LTD). Cold Spring Harb Perspect Biol 4(6): p. a005710.
58.
Peng Y, Zhao J, Gu QH, Chen RQ, Xu Z, Yan JZ, Wang SH, Liu SY, Chen Z and Lu W. (2010). Distinct trafficking and expression mechanisms underlie LTP and LTD of NMDA receptor-mediated synaptic responses. Hippocampus 20:646–658.
59.
Hunt DL and Castillo PE. (2012). Synaptic plasticity of NMDA receptors: mechanisms and functional implications. Curr Opin Neurobiol 22:496–508.
60.
Paoletti P, Bellone C and Zhou Q. (2013). NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci 14:383–400.
61.
Cousins SL, Papadakis M, Rutter AR and Stephenson FA. (2008). Differential interaction of NMDA receptor subtypes with the post-synaptic density-95 family of membrane associated guanylate kinase proteins. J Neurochem 104:903–913.
62.
Rumbaugh G and Vicini S. (1999). Distinct synaptic and extrasynaptic NMDA receptors in developing cerebellar granule neurons. J Neurosci 19:10603–10610.
63.
Pickard L, Noël J, Henley JM, Collingridge GL and Molnar E. (2000). Developmental changes in synaptic AMPA and NMDA receptor distribution and AMPA receptor subunit composition in living hippocampal neurons. J Neurosci 20:7922–7931.
64.
Zhou X, Hollern D, Liao J, Andrechek E and Wang H. (2013). NMDA receptor-mediated excitotoxicity depends on the coactivation of synaptic and extrasynaptic receptors. Cell Death Dis 4: e560.
65.
Rao A and Craig AM. (1997). Activity regulates the synaptic localization of the NMDA receptor in hippocampal neurons. Neuron 19:801–812.
66.
Berg LK, Larsson M, Morland C and Gundersen V. (2013). Pre- and postsynaptic localization of NMDA receptor subunits at hippocampal mossy fibre synapses. Neuroscience 230:139–150.
67.
Liu XB, Murray KD and Jones EG. (2004). Switching of NMDA receptor 2A and 2B subunits at thalamic and cortical synapses during early postnatal development. J Neurosci 24:8885–8895.
68.
von Engelhardt J, Coserea I, Pawlak V, Fuchs EC, Köhr G, Seeburg PH and Monyer H. (2007). Excitotoxicity in vitro by NR2A- and NR2B-containing NMDA receptors. Neuropharmacology 53:10–17.
69.
Liu SB and Zhao MG (2013). Neuroprotective effect of estrogen: role of nonsynaptic NR2B-containing NMDA receptors. Brain Res Bull 93:27–31.
70.
Habas A, Kharebava G, Szatmari E and Hetman M. (2006). NMDA neuroprotection against a phosphatidylinositol-3 kinase inhibitor, LY294002 by NR2B-mediated suppression of glycogen synthase kinase-3beta-induced apoptosis. J Neurochem 96:335–348.
71.
Harris AZ and Pettit DL. (2007). Extrasynaptic and synaptic NMDA receptors form stable and uniform pools in rat hippocampal slices. J Physiol 584(Pt 2):509–519.
72.
Li XJ, Du ZW, Zarnowska ED, Pankratz M, Hansen LO, Pearce RA and Zhang SC. (2005). Specification of motoneurons from human embryonic stem cells. Nat Biotechnol 23:215–221.
73.
Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto H, Chung W, Croft GF, Saphier G, Leibel R, et al. (2008). Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 321:1218–1221.
74.
Kim C, Wong J, Wen J, Wang S, Wang C, Spiering S, Kan NG, Forcales S, Puri PL, et al. (2013). Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature 494:105–110.
75.
Han DW, Tapia N, Hermann A, Hemmer K, Höing S, Araúzo-Bravo MJ, Zaehres H, Wu G, Frank S, et al. (2012). Direct reprogramming of fibroblasts into neural stem cells by defined factors. Cell Stem Cell 10:465–472.
76.
Lujan E, Chanda S, Ahlenius H, Südhof TC and Wernig M. (2012). Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells. Proc Natl Acad Sci U S A 109:2527–2532.
77.
Karow M, Sánchez R, Schichor C, Masserdotti G, Ortega F, Heinrich C, Gascón S, Khan MA, Lie DC, et al. (2012). Reprogramming of pericyte-derived cells of the adult human brain into induced neuronal cells. Cell Stem Cell 11:471–476.
78.
Corti S, Nizzardo M, Simone C, Falcone M, Donadoni C, Salani S, Rizzo F, Nardini M, Riboldi G, et al. (2012). Direct reprogramming of human astrocytes into neural stem cells and neurons. Exp Cell Res 318:1528–1541.
79.
Sheng C, Zheng Q, Wu J, Xu Z, Wang L, Li W, Zhang H, Zhao XY, Liu L, et al. (2012). Direct reprogramming of Sertoli cells into multipotent neural stem cells by defined factors. Cell Res 22:208–218.
80.
Momčilović O, Liu Q, Swistowski A, Russo-Tait T, Zhao Y, Rao MS and X Zeng. (2014). Genome wide profiling of dopaminergic neurons derived from human embryonic and induced pluripotent stem cells. Stem Cells Dev 23:406–420.
81.
Fietz SA, Lachmann R, Brandl H, Kircher M, Samusik N, Schröder R, Lakshmanaperumal N, Henry I, Vogt J, et al. (2012). Transcriptomes of germinal zones of human and mouse fetal neocortex suggest a role of extracellular matrix in progenitor self-renewal. Proc Natl Acad Sci U S A 109:11836–11841.
82.
Myers RA, Casals F, Gauthier J, Hamdan FF, Keebler J, Boyko AR, Bustamante CD, Piton AM, Spiegelman D, et al. (2011). A population genetic approach to mapping neurological disorder genes using deep resequencing. PLoS Genet 7:e1001318.
83.
Mao Y, Ge X, Frank CL, Madison JM, Koehler AN, Doud MK, Tassa C, Berry EM, Soda T, et al. (2009). Disrupted in schizophrenia 1 regulates neuronal progenitor proliferation via modulation of GSK3beta/beta-catenin signaling. Cell 136:1017–1031.
84.
Wei J, Graziane NM, Wang H, Zhong P, Wang Q, Liu W, Hayashi-Takagi A, Korth C, Sawa A, Brandon NJ and Yan Z. (2014). Regulation of N-methyl-D-aspartate receptors by disrupted-in-schizophrenia-1. Biol Psychiatry 75:414–424.
85.
Hahn CG, Wang HY, Cho DS, Talbot K, Gur RE, Berrettini WH, Bakshi K, Kamins J, Borgmann-Winter KE, et al. (2006). Altered neuregulin 1-erbB4 signaling contributes to NMDA receptor hypofunction in schizophrenia. Nat Med 12:824–828.
86.
Bjarnadottir M, Misner DL, Haverfield-Gross S, Bruun S, Helgason VG, Stefansson H, Sigmundsson A, Firth DR, Nielsen B, et al. (2007). Neuregulin1 (NRG1) signaling through Fyn modulates NMDA receptor phosphorylation: differential synaptic function in NRG1+/- knock-outs compared with wild-type mice. J Neurosci 27:4519–4529.
87.
Pandya CD and Pillai A. (2014). TrkB interacts with ErbB4 and regulates NRG1-induced NR2B phosphorylation in cortical neurons before synaptogenesis. Cell Commun Signal 12:47.
88.
Jeans A, Malins R, Padamsey Z, Reinhart M and Emptage N. (2011). Increased expression of dysbindin-1A leads to a selective deficit in NMDA receptor signaling in the hippocampus. Neuropharmacology 61:1345–1353.
89.
Nutini M, Frazzini V, Marini C, Spalloni A, Sensi SL and Longone P. (2011). Zinc pre-treatment enhances NMDAR-mediated excitotoxicity in cultured cortical neurons from SOD1(G93A) mouse, a model of amyotrophic lateral sclerosis. Neuropharmacology 60:1200–1208.
90.
Zheng M, Liao M, Cui T, Tian H, Fan DS and Wan Q. (2012). Regulation of nuclear TDP-43 by NR2A-containing NMDA receptors and PTEN. J Cell Sci 125(Pt 6):1556–1567.
91.
Donnelly CJ, Zhang PW, Pham JT, Haeusler AR, Mistry NA, Vidensky S, Daley EL, Poth EM, Hoover B, et al. (2013). RNA Toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 80:415–428.
92.
Kaniakova M, et al. Key amino acid residues within the third membrane domains of NR1 and NR2 subunits contribute to the regulation of the surface delivery of N-methyl-D-aspartate receptors. J Biol Chem 287:26423–26434.
93.
Kaniakova M, Krausova B, Vyklicky V, Korinek M, Lichnerova K, Vyklicky L and Horak M. (2012). Human induced pluripotent stem cell-derived models to investigate human cytomegalovirus infection in neural cells. PLoS One 7:e49700.
94.
Liu Q, Fischer U, Wang F and Dreyfuss G. (1997). The spinal muscular atrophy disease gene product, SMN, and its associated protein SIP1 are in a complex with spliceosomal snRNP proteins. Cell 90:1013–1021.
95.
Ebert AD, Yu J, Rose Jr FF, Mattis VB, Lorson CL, Thomson JA and Svendsen CN. (2009). Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 457:277–280.
96.
Fischer U, Liu Q and Dreyfuss G. (1997). The SMN-SIP1 complex has an essential role in spliceosomal snRNP biogenesis. Cell 90:1023–1029.
97.
Biondi O, Branchu J, Sanchez G, Lancelin C, Deforges S, Lopes P, Pariset C, Lécolle S, Côté J, Chanoine C and Charbonnier F. (2010). In vivo NMDA receptor activation accelerates motor unit maturation, protects spinal motor neurons, and enhances SMN2 gene expression in severe spinal muscular atrophy mice. J Neurosci 30:11288–11299.
98.
Majumder S, Varadharaj S, Ghoshal K, Monani U, Burghes AH and Jacob ST. (2004). Identification of a novel cyclic AMP-response element (CRE-II) and the role of CREB-1 in the cAMP-induced expression of the survival motor neuron (SMN) gene. J Biol Chem 279:14803–14811.
99.
Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N, Sangar S, Li Y, Mu Y, Chen G, et al. (2011). Modelling schizophrenia using human induced pluripotent stem cells. Nature 473:221–225.
100.
Niemitz E. (2014). iPSC models of ALS. Nat Genet 46:532–532.
101.
The Hd iPsc Consortium. (2012). Induced pluripotent stem cells from patients with Huntington's disease show CAG-repeat-expansion-associated phenotypes. Cell Stem Cell 11:264–278.
102.
Briggs JA, Sun J, Shepherd J, Ovchinnikov DA, Chung TL, Nayler SP, Kao LP, Morrow CA, Thakar NY, et al. (2013). Integration-free induced pluripotent stem cells model genetic and neural developmental features of down syndrome etiology. Stem Cells 31:467–478.

Information & Authors

Information

Published In

cover image Stem Cells and Development
Stem Cells and Development
Volume 26Issue Number 11June 1, 2017
Pages: 798 - 807
PubMed: 28381110

History

Published in print: June 1, 2017
Published online: 1 June 2017
Published ahead of print: 10 May 2017
Published ahead of production: 5 April 2017
Accepted: 5 April 2017
Received: 15 November 2016

Permissions

Request permissions for this article.

Topics

Authors

Affiliations

Adri Chakraborty
Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania.
Suzanne Murphy
Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania.
Natalia Coleman
Biology, New Jersey City University, Jersey City, New Jersey.

Notes

Address correspondence to:Dr. Natalia ColemanBiologyNew Jersey City UniversityJersey City, NJ 07305-1596E-mail: [email protected]

Author Disclosure Statement

No competing financial interests exist.

Metrics & Citations

Metrics

Citations

Export citation

Select the format you want to export the citations of this publication.

View Options

Get Access

Access content

To read the fulltext, please use one of the options below to sign in or purchase access.

Society Access

If you are a member of a society that has access to this content please log in via your society website and then return to this publication.

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

View options

PDF/EPUB

View PDF/ePub

Full Text

View Full Text

Media

Figures

Other

Tables

Share

Share

Copy the content Link

Share on social media

Back to Top