Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2015

Open Access 01-12-2015 | Research article

Human neural stem cells alleviate Alzheimer-like pathology in a mouse model

Authors: Il-Shin Lee, Kwangsoo Jung, Il-Sun Kim, Haejin Lee, Miri Kim, Seokhwan Yun, Kyujin Hwang, Jeong Eun Shin, Kook In Park

Published in: Molecular Neurodegeneration | Issue 1/2015

Login to get access

Abstract

Background

Alzheimer’s disease (AD) is an inexorable neurodegenerative disease that commonly occurs in the elderly. The cognitive impairment caused by AD is associated with abnormal accumulation of amyloid-β (Aβ) and hyperphosphorylated tau, which are accompanied by inflammation. Neural stem cells (NSCs) are self-renewing, multipotential cells that differentiate into distinct neural cells. When transplanted into a diseased brain, NSCs repair and replace injured tissues after migration toward and engraftment within lesions. We investigated the therapeutic effects in an AD mouse model of human NSCs (hNSCs) that derived from an aborted human fetal telencephalon at 13 weeks of gestation. Cells were transplanted into the cerebral lateral ventricles of neuron-specific enolase promoter-controlled APPsw-expressing (NSE/APPsw) transgenic mice at 13 months of age.

Results

Implanted cells extensively migrated and engrafted, and some differentiated into neuronal and glial cells, although most hNSCs remained immature. The hNSC transplantation improved spatial memory in these mice, which also showed decreased tau phosphorylation and Aβ42 levels and attenuated microgliosis and astrogliosis. The hNSC transplantation reduced tau phosphorylation via Trk-dependent Akt/GSK3β signaling, down-regulated Aβ production through an Akt/GSK3β signaling-mediated decrease in BACE1, and decreased expression of inflammatory mediators through deactivation of microglia that was mediated by cell-to-cell contact, secretion of anti-inflammatory factors generated from hNSCs, or both. The hNSC transplantation also facilitated synaptic plasticity and anti-apoptotic function via trophic supplies. Furthermore, the safety and feasibility of hNSC transplantation are supported.

Conclusions

These findings demonstrate the hNSC transplantation modulates diverse AD pathologies and rescue impaired memory via multiple mechanisms in an AD model. Thus, our data provide tangible preclinical evidence that human NSC transplantation could be a safe and versatile approach for treating AD patients.
Appendix
Available only for authorised users
Literature
3.
go back to reference Heneka MT, O’Banion MK, Terwel D, Kummer MP. Neuroinflammatory processes in Alzheimer’s disease. J Neural Transm. 2010;117(8):919–47.CrossRefPubMed Heneka MT, O’Banion MK, Terwel D, Kummer MP. Neuroinflammatory processes in Alzheimer’s disease. J Neural Transm. 2010;117(8):919–47.CrossRefPubMed
4.
go back to reference Aisen PS, Cummings J, Schneider LS. Symptomatic and nonamyloid/tau based pharmacologic treatment for Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2(3):a006395.PubMedCentralCrossRefPubMed Aisen PS, Cummings J, Schneider LS. Symptomatic and nonamyloid/tau based pharmacologic treatment for Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2(3):a006395.PubMedCentralCrossRefPubMed
5.
go back to reference Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M. Alzheimer’s disease: clinical trials and drug development. Lancet Neurol. 2010;9(7):702–16.CrossRefPubMed Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M. Alzheimer’s disease: clinical trials and drug development. Lancet Neurol. 2010;9(7):702–16.CrossRefPubMed
7.
11.
go back to reference Svendsen CN, ter Borg MG, Armstrong RJ, Rosser AE, Chandran S, Ostenfeld T, et al. A new method for the rapid and long term growth of human neural precursor cells. J Neurosci Methods. 1998;85(2):141–52.CrossRefPubMed Svendsen CN, ter Borg MG, Armstrong RJ, Rosser AE, Chandran S, Ostenfeld T, et al. A new method for the rapid and long term growth of human neural precursor cells. J Neurosci Methods. 1998;85(2):141–52.CrossRefPubMed
12.
go back to reference Kim HT, Kim IS, Lee IS, Lee JP, Snyder EY, Park KI. Human neurospheres derived from the fetal central nervous system are regionally and temporally specified but are not committed. Exp Neurol. 2006;199(1):222–35.CrossRefPubMed Kim HT, Kim IS, Lee IS, Lee JP, Snyder EY, Park KI. Human neurospheres derived from the fetal central nervous system are regionally and temporally specified but are not committed. Exp Neurol. 2006;199(1):222–35.CrossRefPubMed
13.
go back to reference Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, et al. Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci U S A. 2000;97(26):14720–5.PubMedCentralCrossRefPubMed Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, et al. Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci U S A. 2000;97(26):14720–5.PubMedCentralCrossRefPubMed
14.
go back to reference Piper DR, Mujtaba T, Keyoung H, Roy NS, Goldman SA, Rao MS, et al. Identification and characterization of neuronal precursors and their progeny from human fetal tissue. J Neurosci Res. 2001;66(3):356–68.PubMedCentralCrossRefPubMed Piper DR, Mujtaba T, Keyoung H, Roy NS, Goldman SA, Rao MS, et al. Identification and characterization of neuronal precursors and their progeny from human fetal tissue. J Neurosci Res. 2001;66(3):356–68.PubMedCentralCrossRefPubMed
15.
go back to reference Tamaki SJ, Jacobs Y, Dohse M, Capela A, Cooper JD, Reitsma M, et al. Neuroprotection of host cells by human central nervous system stem cells in a mouse model of infantile neuronal ceroid lipofuscinosis. Cell Stem Cell. 2009;5(3):310–9.CrossRefPubMed Tamaki SJ, Jacobs Y, Dohse M, Capela A, Cooper JD, Reitsma M, et al. Neuroprotection of host cells by human central nervous system stem cells in a mouse model of infantile neuronal ceroid lipofuscinosis. Cell Stem Cell. 2009;5(3):310–9.CrossRefPubMed
16.
go back to reference Mine Y, Tatarishvili J, Oki K, Monni E, Kokaia Z, Lindvall O. Grafted human neural stem cells enhance several steps of endogenous neurogenesis and improve behavioral recovery after middle cerebral artery occlusion in rats. Neurobiol Dis. 2013;52:191–203.CrossRefPubMed Mine Y, Tatarishvili J, Oki K, Monni E, Kokaia Z, Lindvall O. Grafted human neural stem cells enhance several steps of endogenous neurogenesis and improve behavioral recovery after middle cerebral artery occlusion in rats. Neurobiol Dis. 2013;52:191–203.CrossRefPubMed
17.
go back to reference Pluchino S, Gritti A, Blezer E, Amadio S, Brambilla E, Borsellino G, et al. Human neural stem cells ameliorate autoimmune encephalomyelitis in non-human primates. Ann Neurol. 2009;66(3):343–54.CrossRefPubMed Pluchino S, Gritti A, Blezer E, Amadio S, Brambilla E, Borsellino G, et al. Human neural stem cells ameliorate autoimmune encephalomyelitis in non-human primates. Ann Neurol. 2009;66(3):343–54.CrossRefPubMed
18.
go back to reference Horie N, Pereira MP, Niizuma K, Sun G, Keren-Gill H, Encarnacion A, et al. Transplanted stem cell-secreted vascular endothelial growth factor effects poststroke recovery, inflammation, and vascular repair. Stem Cells. 2011;29(2):274–85.CrossRefPubMed Horie N, Pereira MP, Niizuma K, Sun G, Keren-Gill H, Encarnacion A, et al. Transplanted stem cell-secreted vascular endothelial growth factor effects poststroke recovery, inflammation, and vascular repair. Stem Cells. 2011;29(2):274–85.CrossRefPubMed
19.
go back to reference Andres RH, Horie N, Slikker W, Keren-Gill H, Zhan K, Sun G, et al. Human neural stem cells enhance structural plasticity and axonal transport in the ischaemic brain. Brain. 2011;134(Pt 6):1777–89.PubMedCentralCrossRefPubMed Andres RH, Horie N, Slikker W, Keren-Gill H, Zhan K, Sun G, et al. Human neural stem cells enhance structural plasticity and axonal transport in the ischaemic brain. Brain. 2011;134(Pt 6):1777–89.PubMedCentralCrossRefPubMed
20.
go back to reference Cummings BJ, Uchida N, Tamaki SJ, Salazar DL, Hooshmand M, Summers R, et al. Human neural stem cells differentiate and promote locomotor recovery in spinal cord-injured mice. Proc Natl Acad Sci U S A. 2005;102(39):14069–74.PubMedCentralCrossRefPubMed Cummings BJ, Uchida N, Tamaki SJ, Salazar DL, Hooshmand M, Summers R, et al. Human neural stem cells differentiate and promote locomotor recovery in spinal cord-injured mice. Proc Natl Acad Sci U S A. 2005;102(39):14069–74.PubMedCentralCrossRefPubMed
21.
go back to reference Blurton-Jones M, Kitazawa M, Martinez-Coria H, Castello NA, Muller FJ, Loring JF, et al. Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci U S A. 2009;106(32):13594–9.PubMedCentralCrossRefPubMed Blurton-Jones M, Kitazawa M, Martinez-Coria H, Castello NA, Muller FJ, Loring JF, et al. Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci U S A. 2009;106(32):13594–9.PubMedCentralCrossRefPubMed
22.
go back to reference Ager RR, Davis JL, Agazaryan A, Benavente F, Poon WW, LaFerla FM, et al. Human neural stem cells improve cognition and promote synaptic growth in two complementary transgenic models of Alzheimer’s disease and neuronal loss. Hippocampus. 2015;25(7):813–26.CrossRefPubMed Ager RR, Davis JL, Agazaryan A, Benavente F, Poon WW, LaFerla FM, et al. Human neural stem cells improve cognition and promote synaptic growth in two complementary transgenic models of Alzheimer’s disease and neuronal loss. Hippocampus. 2015;25(7):813–26.CrossRefPubMed
23.
go back to reference Zhang W, Wang GM, Wang PJ, Zhang Q, Sha SH. Effects of neural stem cells on synaptic proteins and memory in a mouse model of Alzheimer’s disease. J Neurosci Res. 2014;92(2):185–94.CrossRefPubMed Zhang W, Wang GM, Wang PJ, Zhang Q, Sha SH. Effects of neural stem cells on synaptic proteins and memory in a mouse model of Alzheimer’s disease. J Neurosci Res. 2014;92(2):185–94.CrossRefPubMed
24.
go back to reference Zhang W, Wang PJ, Sha HY, Ni J, Li MH, Gu GJ. Neural stem cell transplants improve cognitive function without altering amyloid pathology in an APP/PS1 double transgenic model of Alzheimer’s disease. Mol Neurobiol. 2014;50(2):423–37.CrossRefPubMed Zhang W, Wang PJ, Sha HY, Ni J, Li MH, Gu GJ. Neural stem cell transplants improve cognitive function without altering amyloid pathology in an APP/PS1 double transgenic model of Alzheimer’s disease. Mol Neurobiol. 2014;50(2):423–37.CrossRefPubMed
25.
go back to reference Ryu JK, Cho T, Wang YT, McLarnon JG. Neural progenitor cells attenuate inflammatory reactivity and neuronal loss in an animal model of inflamed AD brain. J Neuroinflammation. 2009;6:39.PubMedCentralCrossRefPubMed Ryu JK, Cho T, Wang YT, McLarnon JG. Neural progenitor cells attenuate inflammatory reactivity and neuronal loss in an animal model of inflamed AD brain. J Neuroinflammation. 2009;6:39.PubMedCentralCrossRefPubMed
26.
go back to reference Hwang DY, Cho JS, Lee SH, Chae KR, Lim HJ, Min SH, et al. Aberrant expressions of pathogenic phenotype in Alzheimer’s diseased transgenic mice carrying NSE-controlled APPsw. Exp Neurol. 2004;186(1):20–32.CrossRefPubMed Hwang DY, Cho JS, Lee SH, Chae KR, Lim HJ, Min SH, et al. Aberrant expressions of pathogenic phenotype in Alzheimer’s diseased transgenic mice carrying NSE-controlled APPsw. Exp Neurol. 2004;186(1):20–32.CrossRefPubMed
28.
go back to reference Plattner F, Angelo M, Giese KP. The roles of cyclin-dependent kinase 5 and glycogen synthase kinase 3 in tau hyperphosphorylation. J Biol Chem. 2006;281(35):25457–65.CrossRefPubMed Plattner F, Angelo M, Giese KP. The roles of cyclin-dependent kinase 5 and glycogen synthase kinase 3 in tau hyperphosphorylation. J Biol Chem. 2006;281(35):25457–65.CrossRefPubMed
29.
go back to reference Kaytor MD, Orr HT. The GSK3 beta signaling cascade and neurodegenerative disease. Curr Opin Neurobiol. 2002;12(3):275–8.CrossRefPubMed Kaytor MD, Orr HT. The GSK3 beta signaling cascade and neurodegenerative disease. Curr Opin Neurobiol. 2002;12(3):275–8.CrossRefPubMed
30.
go back to reference Esposito G, De Filippis D, Carnuccio R, Izzo AA, Iuvone T. The marijuana component cannabidiol inhibits beta-amyloid-induced tau protein hyperphosphorylation through Wnt/beta-catenin pathway rescue in PC12 cells. J Mol Med. 2006;84(3):253–8.CrossRefPubMed Esposito G, De Filippis D, Carnuccio R, Izzo AA, Iuvone T. The marijuana component cannabidiol inhibits beta-amyloid-induced tau protein hyperphosphorylation through Wnt/beta-catenin pathway rescue in PC12 cells. J Mol Med. 2006;84(3):253–8.CrossRefPubMed
31.
go back to reference Hsia AY, Masliah E, McConlogue L, Yu GQ, Tatsuno G, Hu K, et al. Plaque-independent disruption of neural circuits in Alzheimer’s disease mouse models. Proc Natl Acad Sci U S A. 1999;96(6):3228–33.PubMedCentralCrossRefPubMed Hsia AY, Masliah E, McConlogue L, Yu GQ, Tatsuno G, Hu K, et al. Plaque-independent disruption of neural circuits in Alzheimer’s disease mouse models. Proc Natl Acad Sci U S A. 1999;96(6):3228–33.PubMedCentralCrossRefPubMed
32.
go back to reference Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, et al. A specific amyloid-beta protein assembly in the brain impairs memory. Nature. 2006;440(7082):352–7.CrossRefPubMed Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, et al. A specific amyloid-beta protein assembly in the brain impairs memory. Nature. 2006;440(7082):352–7.CrossRefPubMed
33.
go back to reference Billings LM, Oddo S, Green KN, McGaugh JL, LaFerla FM. Intraneuronal Abeta causes the onset of early Alzheimer’s disease-related cognitive deficits in transgenic mice. Neuron. 2005;45(5):675–88.CrossRefPubMed Billings LM, Oddo S, Green KN, McGaugh JL, LaFerla FM. Intraneuronal Abeta causes the onset of early Alzheimer’s disease-related cognitive deficits in transgenic mice. Neuron. 2005;45(5):675–88.CrossRefPubMed
34.
go back to reference Zhang H, Gao Y, Dai Z, Meng T, Tu S, Yan Y. IGF-1 reduces BACE-1 expression in PC12 cells via activation of PI3-K/Akt and MAPK/ERK1/2 signaling pathways. Neurochem Res. 2011;36(1):49–57.CrossRefPubMed Zhang H, Gao Y, Dai Z, Meng T, Tu S, Yan Y. IGF-1 reduces BACE-1 expression in PC12 cells via activation of PI3-K/Akt and MAPK/ERK1/2 signaling pathways. Neurochem Res. 2011;36(1):49–57.CrossRefPubMed
35.
go back to reference Ly PT, Wu Y, Zou H, Wang R, Zhou W, Kinoshita A, et al. Inhibition of GSK3beta-mediated BACE1 expression reduces Alzheimer-associated phenotypes. J Clin Invest. 2013;123(1):224–35.PubMedCentralCrossRefPubMed Ly PT, Wu Y, Zou H, Wang R, Zhou W, Kinoshita A, et al. Inhibition of GSK3beta-mediated BACE1 expression reduces Alzheimer-associated phenotypes. J Clin Invest. 2013;123(1):224–35.PubMedCentralCrossRefPubMed
36.
go back to reference Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12(9):1005–15.PubMed Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12(9):1005–15.PubMed
38.
go back to reference Lee IS, Jung K, Kim IS, Park KI. Amyloid-beta oligomers regulate the properties of human neural stem cells through GSK-3beta signaling. Exp Mol Med. 2013;45:e60.PubMedCentralCrossRefPubMed Lee IS, Jung K, Kim IS, Park KI. Amyloid-beta oligomers regulate the properties of human neural stem cells through GSK-3beta signaling. Exp Mol Med. 2013;45:e60.PubMedCentralCrossRefPubMed
39.
go back to reference Fricker RA, Carpenter MK, Winkler C, Greco C, Gates MA, Bjorklund A. Site-specific migration and neuronal differentiation of human neural progenitor cells after transplantation in the adult rat brain. J Neurosci. 1999;19(14):5990–6005.PubMed Fricker RA, Carpenter MK, Winkler C, Greco C, Gates MA, Bjorklund A. Site-specific migration and neuronal differentiation of human neural progenitor cells after transplantation in the adult rat brain. J Neurosci. 1999;19(14):5990–6005.PubMed
40.
go back to reference Lee JP, Jeyakumar M, Gonzalez R, Takahashi H, Lee PJ, Baek RC, et al. Stem cells act through multiple mechanisms to benefit mice with neurodegenerative metabolic disease. Nat Med. 2007;13(4):439–47.CrossRefPubMed Lee JP, Jeyakumar M, Gonzalez R, Takahashi H, Lee PJ, Baek RC, et al. Stem cells act through multiple mechanisms to benefit mice with neurodegenerative metabolic disease. Nat Med. 2007;13(4):439–47.CrossRefPubMed
41.
go back to reference Hawryluk GW, Mothe AJ, Chamankhah M, Wang J, Tator C, Fehlings MG. In vitro characterization of trophic factor expression in neural precursor cells. Stem Cells Dev. 2012;21(3):432–47.CrossRefPubMed Hawryluk GW, Mothe AJ, Chamankhah M, Wang J, Tator C, Fehlings MG. In vitro characterization of trophic factor expression in neural precursor cells. Stem Cells Dev. 2012;21(3):432–47.CrossRefPubMed
42.
go back to reference Pluchino S, Zanotti L, Brambilla E, Rovere-Querini P, Capobianco A, Alfaro-Cervello C, et al. Immune regulatory neural stem/precursor cells protect from central nervous system autoimmunity by restraining dendritic cell function. PLoS One. 2009;4(6):e5959.PubMedCentralCrossRefPubMed Pluchino S, Zanotti L, Brambilla E, Rovere-Querini P, Capobianco A, Alfaro-Cervello C, et al. Immune regulatory neural stem/precursor cells protect from central nervous system autoimmunity by restraining dendritic cell function. PLoS One. 2009;4(6):e5959.PubMedCentralCrossRefPubMed
43.
go back to reference Einstein O, Fainstein N, Vaknin I, Mizrachi-Kol R, Reihartz E, Grigoriadis N, et al. Neural precursors attenuate autoimmune encephalomyelitis by peripheral immunosuppression. Ann Neurol. 2007;61(3):209–18.CrossRefPubMed Einstein O, Fainstein N, Vaknin I, Mizrachi-Kol R, Reihartz E, Grigoriadis N, et al. Neural precursors attenuate autoimmune encephalomyelitis by peripheral immunosuppression. Ann Neurol. 2007;61(3):209–18.CrossRefPubMed
44.
go back to reference Liu J, Hjorth E, Zhu M, Calzarossa C, Samuelsson EB, Schultzberg M, et al. Interplay between human microglia and neural stem/progenitor cells in an allogeneic co-culture model. J Cell Mol Med. 2013;17(11):1434–43.PubMedCentralCrossRefPubMed Liu J, Hjorth E, Zhu M, Calzarossa C, Samuelsson EB, Schultzberg M, et al. Interplay between human microglia and neural stem/progenitor cells in an allogeneic co-culture model. J Cell Mol Med. 2013;17(11):1434–43.PubMedCentralCrossRefPubMed
45.
go back to reference Elliott E, Ginzburg I. The role of neurotrophins and insulin on tau pathology in Alzheimer’s disease. Rev Neurosci. 2006;17(6):635–42.CrossRefPubMed Elliott E, Ginzburg I. The role of neurotrophins and insulin on tau pathology in Alzheimer’s disease. Rev Neurosci. 2006;17(6):635–42.CrossRefPubMed
46.
go back to reference Matrone C, Ciotti MT, Mercanti D, Marolda R, Calissano P. NGF and BDNF signaling control amyloidogenic route and Abeta production in hippocampal neurons. Proc Natl Acad Sci U S A. 2008;105(35):13139–44.PubMedCentralCrossRefPubMed Matrone C, Ciotti MT, Mercanti D, Marolda R, Calissano P. NGF and BDNF signaling control amyloidogenic route and Abeta production in hippocampal neurons. Proc Natl Acad Sci U S A. 2008;105(35):13139–44.PubMedCentralCrossRefPubMed
47.
go back to reference Nagahara AH, Merrill DA, Coppola G, Tsukada S, Schroeder BE, Shaked GM, et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. 2009;15(3):331–7.PubMedCentralCrossRefPubMed Nagahara AH, Merrill DA, Coppola G, Tsukada S, Schroeder BE, Shaked GM, et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. 2009;15(3):331–7.PubMedCentralCrossRefPubMed
48.
go back to reference Tuszynski MH, Thal L, Pay M, Salmon DP, HS U, Bakay R, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Na Med. 2005;11(5):551–5.CrossRef Tuszynski MH, Thal L, Pay M, Salmon DP, HS U, Bakay R, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Na Med. 2005;11(5):551–5.CrossRef
49.
go back to reference Lalonde R, Fukuchi K, Strazielle C. APP transgenic mice for modelling behavioural and psychological symptoms of dementia (BPSD). Neurosci Biobehav Rev. 2012;36(5):1357–75.PubMedCentralCrossRefPubMed Lalonde R, Fukuchi K, Strazielle C. APP transgenic mice for modelling behavioural and psychological symptoms of dementia (BPSD). Neurosci Biobehav Rev. 2012;36(5):1357–75.PubMedCentralCrossRefPubMed
50.
go back to reference Baki L, Shioi J, Wen P, Shao Z, Schwarzman A, Gama-Sosa M, et al. PS1 activates PI3K thus inhibiting GSK-3 activity and tau overphosphorylation: effects of FAD mutations. EMBO J. 2004;23(13):2586–96.PubMedCentralCrossRefPubMed Baki L, Shioi J, Wen P, Shao Z, Schwarzman A, Gama-Sosa M, et al. PS1 activates PI3K thus inhibiting GSK-3 activity and tau overphosphorylation: effects of FAD mutations. EMBO J. 2004;23(13):2586–96.PubMedCentralCrossRefPubMed
51.
go back to reference Elliott E, Atlas R, Lange A, Ginzburg I. Brain-derived neurotrophic factor induces a rapid dephosphorylation of tau protein through a PI-3 Kinase signalling mechanism. Eur J Neurosci. 2005;22(5):1081–9.CrossRefPubMed Elliott E, Atlas R, Lange A, Ginzburg I. Brain-derived neurotrophic factor induces a rapid dephosphorylation of tau protein through a PI-3 Kinase signalling mechanism. Eur J Neurosci. 2005;22(5):1081–9.CrossRefPubMed
52.
go back to reference Hampton DW, Webber DJ, Bilican B, Goedert M, Spillantini MG, Chandran S. Cell-mediated neuroprotection in a mouse model of human tauopathy. J Neurosci. 2010;30(30):9973–83.CrossRefPubMed Hampton DW, Webber DJ, Bilican B, Goedert M, Spillantini MG, Chandran S. Cell-mediated neuroprotection in a mouse model of human tauopathy. J Neurosci. 2010;30(30):9973–83.CrossRefPubMed
53.
go back to reference Kern DS, Maclean KN, Jiang H, Synder EY, Sladek Jr JR, Bjugstad KB. Neural stem cells reduce hippocampal tau and reelin accumulation in aged Ts65Dn Down syndrome mice. Cell Transplant. 2011;20(3):371–9.CrossRefPubMed Kern DS, Maclean KN, Jiang H, Synder EY, Sladek Jr JR, Bjugstad KB. Neural stem cells reduce hippocampal tau and reelin accumulation in aged Ts65Dn Down syndrome mice. Cell Transplant. 2011;20(3):371–9.CrossRefPubMed
54.
go back to reference De Felice FG, Wu D, Lambert MP, Fernandez SJ, Velasco PT, Lacor PN, et al. Alzheimer’s disease-type neuronal tau hyperphosphorylation induced by A beta oligomers. Neurobiol Aging. 2008;29(9):1334–47.PubMedCentralCrossRefPubMed De Felice FG, Wu D, Lambert MP, Fernandez SJ, Velasco PT, Lacor PN, et al. Alzheimer’s disease-type neuronal tau hyperphosphorylation induced by A beta oligomers. Neurobiol Aging. 2008;29(9):1334–47.PubMedCentralCrossRefPubMed
55.
go back to reference Jin M, Shepardson N, Yang T, Chen G, Walsh D, Selkoe DJ. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108(14):5819–24.PubMedCentralCrossRefPubMed Jin M, Shepardson N, Yang T, Chen G, Walsh D, Selkoe DJ. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108(14):5819–24.PubMedCentralCrossRefPubMed
56.
go back to reference Zempel H, Thies E, Mandelkow E, Mandelkow EM. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30(36):11938–50.CrossRefPubMed Zempel H, Thies E, Mandelkow E, Mandelkow EM. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30(36):11938–50.CrossRefPubMed
57.
go back to reference Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837–42.PubMedCentralCrossRefPubMed Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837–42.PubMedCentralCrossRefPubMed
58.
go back to reference Su Y, Ryder J, Li B, Wu X, Fox N, Solenberg P, et al. Lithium, a common drug for bipolar disorder treatment, regulates amyloid-beta precursor protein processing. Biochemistry. 2004;43(22):6899–908.CrossRefPubMed Su Y, Ryder J, Li B, Wu X, Fox N, Solenberg P, et al. Lithium, a common drug for bipolar disorder treatment, regulates amyloid-beta precursor protein processing. Biochemistry. 2004;43(22):6899–908.CrossRefPubMed
59.
go back to reference Rockenstein E, Torrance M, Adame A, Mante M, Bar-on P, Rose JB, et al. Neuroprotective effects of regulators of the glycogen synthase kinase-3beta signaling pathway in a transgenic model of Alzheimer’s disease are associated with reduced amyloid precursor protein phosphorylation. J Neurosci. 2007;27(8):1981–91.CrossRefPubMed Rockenstein E, Torrance M, Adame A, Mante M, Bar-on P, Rose JB, et al. Neuroprotective effects of regulators of the glycogen synthase kinase-3beta signaling pathway in a transgenic model of Alzheimer’s disease are associated with reduced amyloid precursor protein phosphorylation. J Neurosci. 2007;27(8):1981–91.CrossRefPubMed
60.
go back to reference Qing H, He G, Ly PT, Fox CJ, Staufenbiel M, Cai F, et al. Valproic acid inhibits Abeta production, neuritic plaque formation, and behavioral deficits in Alzheimer’s disease mouse models. J Exp Med. 2008;205(12):2781–9.PubMedCentralCrossRefPubMed Qing H, He G, Ly PT, Fox CJ, Staufenbiel M, Cai F, et al. Valproic acid inhibits Abeta production, neuritic plaque formation, and behavioral deficits in Alzheimer’s disease mouse models. J Exp Med. 2008;205(12):2781–9.PubMedCentralCrossRefPubMed
61.
go back to reference Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L, Salani G, et al. Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature. 2005;436(7048):266–71.CrossRefPubMed Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L, Salani G, et al. Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature. 2005;436(7048):266–71.CrossRefPubMed
62.
go back to reference Cusimano M, Biziato D, Brambilla E, Donega M, Alfaro-Cervello C, Snider S, et al. Transplanted neural stem/precursor cells instruct phagocytes and reduce secondary tissue damage in the injured spinal cord. Brain. 2012;135(Pt 2):447–60.PubMedCentralCrossRefPubMed Cusimano M, Biziato D, Brambilla E, Donega M, Alfaro-Cervello C, Snider S, et al. Transplanted neural stem/precursor cells instruct phagocytes and reduce secondary tissue damage in the injured spinal cord. Brain. 2012;135(Pt 2):447–60.PubMedCentralCrossRefPubMed
63.
go back to reference Rota Nodari L, Ferrari D, Giani F, Bossi M, Rodriguez-Menendez V, Tredici G, et al. Long-term survival of human neural stem cells in the ischemic rat brain upon transient immunosuppression. PLoS One. 2010;5(11):e14035.PubMedCentralCrossRefPubMed Rota Nodari L, Ferrari D, Giani F, Bossi M, Rodriguez-Menendez V, Tredici G, et al. Long-term survival of human neural stem cells in the ischemic rat brain upon transient immunosuppression. PLoS One. 2010;5(11):e14035.PubMedCentralCrossRefPubMed
64.
go back to reference Wyss-Coray T, Lin C, Yan F, Yu GQ, Rohde M, McConlogue L, et al. TGF-beta1 promotes microglial amyloid-beta clearance and reduces plaque burden in transgenic mice. Nat Med. 2001;7(5):612–8.CrossRefPubMed Wyss-Coray T, Lin C, Yan F, Yu GQ, Rohde M, McConlogue L, et al. TGF-beta1 promotes microglial amyloid-beta clearance and reduces plaque burden in transgenic mice. Nat Med. 2001;7(5):612–8.CrossRefPubMed
65.
go back to reference Brionne TC, Tesseur I, Masliah E, Wyss-Coray T. Loss of TGF-beta 1 leads to increased neuronal cell death and microgliosis in mouse brain. Neuron. 2003;40(6):1133–45.CrossRefPubMed Brionne TC, Tesseur I, Masliah E, Wyss-Coray T. Loss of TGF-beta 1 leads to increased neuronal cell death and microgliosis in mouse brain. Neuron. 2003;40(6):1133–45.CrossRefPubMed
66.
go back to reference Tian L, Rauvala H, Gahmberg CG. Neuronal regulation of immune responses in the central nervous system. Trends Immunol. 2009;30(2):91–9.CrossRefPubMed Tian L, Rauvala H, Gahmberg CG. Neuronal regulation of immune responses in the central nervous system. Trends Immunol. 2009;30(2):91–9.CrossRefPubMed
67.
go back to reference Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT. Regulation of tau pathology by the microglial fractalkine receptor. Neuron. 2010;68(1):19–31.PubMedCentralCrossRefPubMed Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT. Regulation of tau pathology by the microglial fractalkine receptor. Neuron. 2010;68(1):19–31.PubMedCentralCrossRefPubMed
68.
go back to reference Sastre M, Dewachter I, Landreth GE, Willson TM, Klockgether T, van Leuven F, et al. Nonsteroidal anti-inflammatory drugs and peroxisome proliferator-activated receptor-gamma agonists modulate immunostimulated processing of amyloid precursor protein through regulation of beta-secretase. J Neurosci. 2003;23(30):9796–804.PubMed Sastre M, Dewachter I, Landreth GE, Willson TM, Klockgether T, van Leuven F, et al. Nonsteroidal anti-inflammatory drugs and peroxisome proliferator-activated receptor-gamma agonists modulate immunostimulated processing of amyloid precursor protein through regulation of beta-secretase. J Neurosci. 2003;23(30):9796–804.PubMed
69.
go back to reference Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C, et al. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain. 2005;128(Pt 6):1442–53.CrossRefPubMed Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C, et al. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain. 2005;128(Pt 6):1442–53.CrossRefPubMed
70.
go back to reference Yamasaki TR, Blurton-Jones M, Morrissette DA, Kitazawa M, Oddo S, LaFerla FM. Neural stem cells improve memory in an inducible mouse model of neuronal loss. J Neurosci. 2007;27(44):11925–33.CrossRefPubMed Yamasaki TR, Blurton-Jones M, Morrissette DA, Kitazawa M, Oddo S, LaFerla FM. Neural stem cells improve memory in an inducible mouse model of neuronal loss. J Neurosci. 2007;27(44):11925–33.CrossRefPubMed
71.
go back to reference Mastrangelo MA, Bowers WJ. Detailed immunohistochemical characterization of temporal and spatial progression of Alzheimer’s disease-related pathologies in male triple-transgenic mice. BMC Neurosci. 2008;9:81.PubMedCentralCrossRefPubMed Mastrangelo MA, Bowers WJ. Detailed immunohistochemical characterization of temporal and spatial progression of Alzheimer’s disease-related pathologies in male triple-transgenic mice. BMC Neurosci. 2008;9:81.PubMedCentralCrossRefPubMed
72.
go back to reference Lambert MP, Viola KL, Chromy BA, Chang L, Morgan TE, Yu J, et al. Vaccination with soluble Abeta oligomers generates toxicity-neutralizing antibodies. J Neurochem. 2001;79(3):595–605.CrossRefPubMed Lambert MP, Viola KL, Chromy BA, Chang L, Morgan TE, Yu J, et al. Vaccination with soluble Abeta oligomers generates toxicity-neutralizing antibodies. J Neurochem. 2001;79(3):595–605.CrossRefPubMed
Metadata
Title
Human neural stem cells alleviate Alzheimer-like pathology in a mouse model
Authors
Il-Shin Lee
Kwangsoo Jung
Il-Sun Kim
Haejin Lee
Miri Kim
Seokhwan Yun
Kyujin Hwang
Jeong Eun Shin
Kook In Park
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2015
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-015-0035-6

Other articles of this Issue 1/2015

Molecular Neurodegeneration 1/2015 Go to the issue