Journal Home > Volume 3 , Issue 1

Alzheimer’s disease (AD) causes degeneration of brain neurons and leads to memory loss and cognitive impairment. Since current therapeutic strategies cannot cure the disease, stem cell therapy represents a powerful tool for the treatment of AD. We first review the advances in molecular pathogenesis and animal models of AD and then discuss recent clinical studies using small molecules and immunoglobulins to target amyloid-beta plaques for AD therapy. Finally, we discuss stem cell therapy for AD using neural stem cells, olfactory ensheathing cells, embryonic stem cells, and mesenchymal stem cell from bone marrow, umbilical cord, and umbilical cord blood. In particular, patient-specific induced pluripotent stem cells are proposed as a future treatment for AD.


menu
Abstract
Full text
Outline
About this article

Research progress in animal models and stem cell therapy for Alzheimer’s disease

Show Author's information Fabin Han1,2( )Wei Wang1Chao Chen1
Centre for Stem Cells and Regenerative Medicine,
Department of Neurology, Liaocheng People’s Hospital/The Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, People’s Republic of China

Abstract

Alzheimer’s disease (AD) causes degeneration of brain neurons and leads to memory loss and cognitive impairment. Since current therapeutic strategies cannot cure the disease, stem cell therapy represents a powerful tool for the treatment of AD. We first review the advances in molecular pathogenesis and animal models of AD and then discuss recent clinical studies using small molecules and immunoglobulins to target amyloid-beta plaques for AD therapy. Finally, we discuss stem cell therapy for AD using neural stem cells, olfactory ensheathing cells, embryonic stem cells, and mesenchymal stem cell from bone marrow, umbilical cord, and umbilical cord blood. In particular, patient-specific induced pluripotent stem cells are proposed as a future treatment for AD.

Keywords: olfactory ensheathing cell, neural stem cell, mesenchymal stem cell, amyloid-beta plaque, neurofibrillary tangle, induced pluripotent stem cell

References(95)

1.
Holtzman DM, Morris JC, Goate AM. Alzheimer’s disease: the challenge of the second century. Sci Transl Med. 2011;3(77):77sr1.
2.
Barnes DE, Yaffe K. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol. 2011;10(9):819–828.
3.
Goldstein LS. Axonal transport and neurodegenerative disease:can we see the elephant? Prog Neurobiol. 2012;99(3):186–190.
4.
Rank KB, Pauley AM, Bhattacharya K, et al. Direct interaction of soluble human recombinant tau protein with Abeta 1-42 results in tau aggregation and hyperphosphorylation by tau protein kinase II. FEBS Lett. 2002;514(2–3):263–268.
5.
Shipton OA, Leitz JR, Dworzak J, et al. Tau protein is required for amyloid {beta}-induced impairment of hippocampal long-term potentiation. J Neurosci. 2011;31(5):1688–1692.
6.
Thinakaran G, Koo EH. Amyloid precursor protein trafficking, processing, and function. J Biol Chem. 2008;283(44):29615–29619.
7.
Mattson MP. Pathways towards and away from Alzheimer’s disease. Nature. 2004;430(7000):631–639.
8.
Mandelkow EM, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med. 2012;2(7):a006247.
9.
Wang JZ, Xia YY, Grundke-Iqbal I, Iqbal K. Abnormal hyperphosphorylation of tau:sites, regulation, and molecular mechanism of neurofibrillary degeneration. J Alzheimers Dis. 2013;33 Suppl 1:S123–S139.
10.
Gunawardena S, Goldstein LS. Disruption of axonal transport and neuronal viability by amyloid precursor protein mutations in Drosophila. Neuron. 2001;32(3):389–401.
11.
Brunholz S, Sisodia S, Lorenzo A, Deyts C, Kins S, Morfini G. Axonal transport of APP and the spatial regulation of APP cleavage and function in neuronal cells. Exp Brain Res. 2012;217(3–4):353–364.
12.
Reitz C. Alzheimer’s disease and the amyloid cascade hypothesis: a critical review. Int J Alzheimers Dis. 2012;2012:369808.
13.
Johnstone EM, Chaney MO, Norris FH, Pascual R, Little SP. Conservation of the sequence of the Alzheimer’s disease amyloid peptide in dog, polar bear and five other mammals by cross-species polymerase chain reaction analysis. Brain Res Mol Brain Res. 1991;10(4):299–305.
14.
Languille S, Blanc S, Blin O, et al. The grey mouse lemur: a non-human primate model for ageing studies. Ageing Res Rev. 2012;11(1):150–162.
15.
Bons N, Rieger F, Prudhomme D, Fisher A, Krause KH. Microcebus murinus: a useful primate model for human cerebral aging and Alzheimer’s disease? Genes Brain Behav. 2006;5(2):120–130.
16.
Kraska A, Dorieux O, Picq JL, et al. Age-associated cerebral atrophy in mouse lemur primates. Neurobiol Aging. 2011;32(5):894–906.
17.
Buccafusco JJ. Estimation of working memory in macaques for studying drugs for the treatment of cognitive disorders. J Alzheimers Dis. 2008;15(4):709–720.
18.
Inestrosa NC, Reyes AE, Chacon MA, et al. Human-like rodent amyloid-beta-peptide determines Alzheimer pathology in aged wild-type Octodon degu. Neurobiol Aging. 2005;26(7):1023–1028.
19.
Castane A, Theobald DE, Robbins TW. Selective lesions of the dorsomedial striatum impair serial spatial reversal learning in rats. Behav Brain Res. 2010;210(1):74–83.
20.
Nakamura S, Murayama N, Noshita T, Annoura H, Ohno T. Progressive brain dysfunction following intracerebroventricular infusion of beta(1–42)-amyloid peptide. Brain Res. 2001;912(2):128–136.
21.
Van Dam D, De Deyn PP. Animal models in the drug discovery pipeline for Alzheimer’s disease. Br J Pharmacol. 2011;164(4):1285–1300.
22.
Li W, Wu Y, Min F, Li Z, Huang J, Huang R. A nonhuman primate model of Alzheimer’s disease generated by intracranial injection of amyloid-beta42 and thiorphan. Metab Brain Dis. 2010;25(3):277–284.
23.
Kalback W, Watson MD, Kokjohn TA, et al. APP transgenic mice Tg2576 accumulate Abeta peptides that are distinct from the chemically modified and insoluble peptides deposited in Alzheimer’s disease senile plaques. Biochemistry. 2002;41(3):922–928.
24.
Goedert M, Klug A, Crowther RA. Tau protein, the paired helical filament and Alzheimer’s disease. J Alzheimers Dis. 2006;9(Suppl 3):195–207.
25.
Chui DH, Tanahashi H, Ozawa K, et al. Transgenic mice with Alzheimer presenilin 1 mutations show accelerated neurodegeneration without amyloid plaque formation. Nat Med. 1999;5(5):560–564.
26.
Rutten BP, Van der Kolk NM, Schafer S, et al. Age-related loss of synaptophysin immunoreactive presynaptic boutons within the hippocampus of APP751SL, PS1M146L, and APP751SL/PS1M146L transgenic mice. Am J Pathol. 2005;167(1):161–173.
27.
Oddo S, Caccamo A, Shepherd JD, et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39(3):409–421.
28.
Du H, Guo L, Fang F, et al. Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer’s disease. Nat Med. 2008;14(10):1097–1105.
29.
Lopez EM, Bell KF, Ribeiro-da-Silva A, Cuello AC. Early changes in neurons of the hippocampus and neocortex in transgenic rats expressing intracellular human a-beta. J Alzheimers Dis. 2004;6(4):421–431.
30.
Vercauteren FG, Clerens S, Roy L, et al. Early dysregulation of hippocampal proteins in transgenic rats with Alzheimer’s disease-linked mutations in amyloid precursor protein and presenilin 1. Brain Res Mol Brain Res. 2004;132(2):241–259.
31.
Flood DG, Lin YG, Lang DM, et al. A transgenic rat model of Alzheimer’s disease with extracellular Abeta deposition. Neurobiol Aging. 2009;30(7):1078–1090.
32.
Leon WC, Canneva F, Partridge V, et al. A novel transgenic rat model with a full Alzheimer’s-like amyloid pathology displays pre-plaque intracellular amyloid-β-associated cognitive impairment. J Alzheimers Dis. 2010;20(1):113–126.
33.
Iijima K, Iijima-Ando K. Drosophila models of Alzheimer’s amyloidosis: the challenge of dissecting the complex mechanisms of toxicity of amyloid-beta 42. J Alzheimers Dis. 2008;15(4):523–540.
34.
Sang TK, Jackson GR. Drosophila models of neurodegenerative disease. NeuroRx. 2005;2(3):438–446.
35.
Lieschke GJ, Currie PD. Animal models of human disease: zebrafish swim into view. Nat Rev Genet. 2007;8(5):353–367.
36.
Liao HK, Wang Y, Noack Watt KE, et al. Tol2 gene trap integrations in the zebrafish amyloid precursor protein genes appa and aplp2 reveal accumulation of secreted APP at the embryonic veins. Dev Dyn. 2012;241(2):415–425.
37.
Newman M, Nornes S, Martins RN, Lardelli MT. Robust homeostasis of presenilin1 protein levels by transcript regulation. Neurosci Lett. 2012;519(1):14–19.
38.
Sundvik M, Chen YC, Panula P. Presenilin1 regulates histamine neuron development and behavior in zebrafish, Danio rerio. J Neurosci. 2013;33(4):1589–1597.
39.
Nornes S, Newman M, Verdile G, et al. Interference with splicing of presenilin transcripts has potent dominant negative effects on presenilin activity. Hum Mol Genet. 2008;17(3):402–412.
40.
Nornes S, Newman M, Wells S, Verdile G, Martins RN, Lardelli M. Independent and cooperative action of Psen2 with Psen1 in zebrafish embryos. Exp Cell Res. 2009;315(16):2791–2801.
41.
Herreman A, Hartmann D, Annaert W, et al. Presenilin 2 deficiency causes a mild pulmonary phenotype and no changes in amyloid precursor protein processing but enhances the embryonic lethal phenotype of presenilin 1 deficiency. Proc Natl Acad Sci U S A. 1999;96(21):11872–11877.
42.
Joshi P, Liang JO, DiMonte K, Sullivan J, Pimplikar SW. Amyloid precursor protein is required for convergent-extension movements during zebrafish development. Dev Biol. 2009;335(1):1–11.
43.
Abramsson A, Kettunen P, Banote RK, et al. The zebrafish amyloid precursor protein-b is required for motor neuron guidance and synapse formation. Dev Biol. 2013;381(2):377–388.
44.
Song P, Pimplikar SW. Knockdown of amyloid precursor protein in zebrafish causes defects in motor axon outgrowth. PLoS One. 2012;7(4):e34209.
45.
Nygaard HB, Strittmatter SM. Cellular prion protein mediates the toxicity of beta-amyloid oligomers: implications for Alzheimer disease. Arch Neurol. 2009;66(11):1325–1328.
46.
Dodel R, Rominger A, Bartenstein P, et al. Intravenous immunoglobulin for treatment of mild-to-moderate Alzheimer’s disease: a phase 2, randomised, double-blind, placebo-controlled, dose-finding trial. Lancet Neurol. 2013;12(3):233–243.
47.
Coric V, van Dyck CH, Salloway S, et al. Safety and tolerability of the gamma-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch Neurol. 2012;69(11):1430–1440.
48.
Kang JE, Lim MM, Bateman RJ, et al. Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science. 2009;326(5955):1005–1007.
49.
Blanchard J, Chohan MO, Li B, Liu F, Iqbal K, Grundke-Iqbal I. Beneficial effect of a CNTF tetrapeptide on adult hippocampal neurogenesis, neuronal plasticity, and spatial memory in mice. J Alzheimers Dis. 2010;21(4):1185–1195.
50.
Cardenas-Aguayo Mdel C, Kazim SF, Grundke-Iqbal I, Iqbal K. Neurogenic and neurotrophic effects of BDNF peptides in mouse hippocampal primary neuronal cell cultures. PLoS One. 2013;8(1):e53596.
51.
Cummings JL, Morstorf T, Zhong K. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther. 2014;6(4):37.
52.
Gilman S, Koller M, Black RS, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology. 2005;64(9):1553–1562.
53.
Rinne JO, Brooks DJ, Rossor MN, et al. 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer’s disease treated with bapineuzumab: a phase 2, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol. 2010;9(4):363–372.
54.
Mullane K, Williams M. Alzheimer’s therapeutics: continued clinical failures question the validity of the amyloid hypothesis – but what lies beyond? Biochem Pharmacol. 2013;85(3):289–305.
55.
Taupin P. The therapeutic potential of adult neural stem cells. Curr Opin Mol Ther. 2006;8(3):225–231.
56.
Singh C, Liu L, Wang JM, et al. Allopregnanolone restores hippocampal-dependent learning and memory and neural progenitor survival in aging 3xTgAD and nonTg mice. Neurobiol Aging. 2012;33(8):1493–1506.
57.
Shin JW, Lee JK, Lee JE, et al. Combined effects of hematopoietic progenitor cell mobilization from bone marrow by granulocyte colony stimulating factor and AMD3100 and chemotaxis into the brain using stromal cell-derived factor-1alpha in an Alzheimer’s disease mouse model. Stem Cells. 2011;29(7):1075–1089.
58.
Chang KA, Kim JA, Kim S, et al. Therapeutic potentials of neural stem cells treated with fluoxetine in Alzheimer’s disease. Neurochem Int. 2012;61(6):885–891.
59.
Taupin P. Adult neural stem cells, neurogenic niches, and cellular therapy. Stem Cell Rev. 2006;2(3):213–219.
60.
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):11925–11933.
61.
Blurton-Jones M, Kitazawa M, Martinez-Coria H, 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):13594–13599.
62.
Njie EG, Kantorovich S, Astary GW, et al. A preclinical assessment of neural stem cells as delivery vehicles for anti-amyloid therapeutics. PLoS One. 2012;7(4):e34097.
63.
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):423–437.
64.
Fan X, Sun D, Tang X, Cai Y, Yin ZQ, Xu H. Stem-cell challenges in the treatment of Alzheimer’s disease: a long way from bench to bedside. Med Res Rev. 2014;34(5):957–978.
65.
Lee HJ, Lim IJ, Park SW, Kim YB, Ko Y, Kim SU. Human neural stem cells genetically modified to express human nerve growth factor(NGF) gene restore cognition in the mouse with ibotenic acid-induced cognitive dysfunction. Cell Transplant. 2012;21(11):2487–2496.
66.
Park D, Joo SS, Kim TK, et al. Human neural stem cells overexpressing choline acetyltransferase restore cognitive function of kainic acid-induced learning and memory deficit animals. Cell Transplant. 2012;21(1):365–371.
67.
Srivastava N, Seth K, Khanna VK, Ansari RW, Agrawal AK. Long-term functional restoration by neural progenitor cell transplantation in rat model of cognitive dysfunction: co-transplantation with olfactory ensheathing cells for neurotrophic factor support. Int J Dev Neurosci. 2009;27(1):103–110.
68.
Wang G, Ao Q, Gong K, Zuo H, Gong Y, Zhang X. Synergistic effect of neural stem cells and olfactory ensheathing cells on repair of adult rat spinal cord injury. Cell Transplant. 2010;19(10):1325–1337.
69.
Sun T, Ye C, Zhang Z, Wu J, Huang H. Cotransplantation of olfactory ensheathing cells and Schwann cells combined with treadmill training promotes functional recovery in rats with contused spinal cords. Cell Transplant. 2013;22 Suppl 1:S27–S38.
70.
Huang H, Xi H, Chen L, Zhang F, Liu Y. Long-term outcome of olfactory ensheathing cell therapy for patients with complete chronic spinal cord injury. Cell Transplant. 2012;21 Suppl 1:S23–S31.
71.
Park D, Lee HJ, Joo SS, et al. Human neural stem cells over-expressing choline acetyltransferase restore cognition in rat model of cognitive dysfunction. Exp Neurol. 2012;234(2):521–526.
72.
Marei HE, Farag A, Althani A, et al. Human olfactory bulb neural stem cells expressing hNGF restore cognitive deficit in Alzheimer’s disease rat model. J Cell Physiol. 2015;230(1):116–130.
73.
Tang J, Xu H, Fan X, et al. Embryonic stem cell-derived neural precursor cells improve memory dysfunction in Abeta(1–40) injured rats. Neurosci Res. 2008;62(2):86–96.
74.
Moghadam FH, Alaie H, Karbalaie K, Tanhaei S, Nasr Esfahani MH, Baharvand H. Transplantation of primed or unprimed mouse embryonic stem cell-derived neural precursor cells improves cognitive function in Alzheimerian rats. Differentiation. 2009;78(2–3):59–68.
75.
Bae JS, Jin HK, Lee JK, Richardson JC, Carter JE. Bone marrow-derived mesenchymal stem cells contribute to the reduction of amyloid-beta deposits and the improvement of synaptic transmission in a mouse model of pre-dementia Alzheimer’s disease. Curr Alzheimer Res. 2013;10(5):524–531.
76.
Lee JK, Jin HK, Bae JS. Bone marrow-derived mesenchymal stem cells reduce brain amyloid-beta deposition and accelerate the activation of microglia in an acutely induced Alzheimer’s disease mouse model. Neurosci Lett. 2009;450(2):136–141.
77.
Lee JK, Jin HK, Endo S, Schuchman EH, Carter JE, Bae JS. Intracerebral transplantation of bone marrow-derived mesenchymal stem cells reduces amyloid-beta deposition and rescues memory deficits in Alzheimer’s disease mice by modulation of immune responses. Stem Cells. 2010;28(2):329–343.
78.
Malm TM, Koistinaho M, Parepalo M, et al. Bone-marrow-derived cells contribute to the recruitment of microglial cells in response to beta-amyloid deposition in APP/PS1 double transgenic Alzheimer mice. Neurobiol Dis. 2005;18(1):134–142.
79.
Rubio-Perez JM, Morillas-Ruiz JM. A review: inflammatory process in Alzheimer’s disease, role of cytokines. ScientificWorldJournal. 2012;2012:756357.
80.
Yang H, Xie Z, Wei L, Bi J. Systemic transplantation of human umbilical cord derived mesenchymal stem cells-educated T regulatory cells improved the impaired cognition in AbetaPPswe/PS1dE9 transgenic mice. PLoS One. 2013;8(7):e69129.
81.
Kim HJ, Lee JH, Kim SH. Therapeutic effects of human mesenchymal stem cells on traumatic brain injury in rats: secretion of neurotrophic factors and inhibition of apoptosis. J Neurotrauma. 2010;27(1):131–138.
82.
Jin K, Xie L, Mao X, et al. Effect of human neural precursor cell transplantation on endogenous neurogenesis after focal cerebral ischemia in the rat. Brain Res. 2011;1374:56–62.
83.
Kan I, Barhum Y, Melamed E, Offen D. Mesenchymal stem cells stimulate endogenous neurogenesis in the subventricular zone of adult mice. Stem Cell Rev. 2011;7(2):404–412.
84.
Yan Y, Ma T, Gong K, Ao Q, Zhang X, Gong Y. Adipose-derived mesenchymal stem cell transplantation promotes adult neurogenesis in the brains of Alzheimer’s disease mice. Neural Regen Res. 2014;9(8):798–805.
85.
Letourneau PA, Menge TD, Wataha KA, et al. Human bone marrow derived mesenchymal stem cells regulate leukocyte-endothelial interactions and activation of transcription factor NF-kappa B. J Tissue Sci Eng. 2011;Suppl 3:001.
86.
Duff K, Suleman F. Transgenic mouse models of Alzheimer’s disease: how useful have they been for therapeutic development? Brief Funct Genomic Proteomic. 2004;3(1):47–59.
87.
Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872.
88.
Yu J, Hu K, Smuga-Otto K, et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324(5928):797–801.
89.
Park I-H, Arora N, Huo H, et al. Disease-specific induced pluripotent stem cells. Cell. 2008;134(5):877–886.
90.
Zhou T, Benda C, Dunzinger S, et al. Generation of human induced pluripotent stem cells from urine samples. Nat Protoc. 2012;7(12):2080–2089.
91.
Yagi T, Ito D, Okada Y, et al. Modeling familial Alzheimer’s disease with induced pluripotent stem cells. Hum Mol Genet. 2011;20(23):4530–4539.
92.
Israel MA, Yuan SH, Bardy C, et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature. 2012;482(7384):216–220.
93.
Machairaki V, Ryu J, Peters A, et al. Induced pluripotent stem cells from familial Alzheimer’s disease patients differentiate into mature neurons with amyloidogenic properties. Stem Cells Dev. August 21, 2014. [Epub ahead of print.]
94.
Garate Z, Davis BR, Quintana-Bustamante O, Segovia JC. New frontier in regenerative medicine:site-specific gene correction in patient-specific induced pluripotent stem cells. Hum Gene Ther. 2013;24(6):571–583.
95.
Hockemeyer D, Wang H, Kiani S, et al. Genetic engineering of human pluripotent cells using TALE nucleases. Nat Biotechnol. 2011;29(8):731–734.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Published: 19 December 2014
Issue date: December 2015

Copyright

© 2015 The Author(s).

Acknowledgements

The authors are grateful to Shichao Wu and Junyu Gao for their technical assistance in preparation of the figures for the manuscript. This work was supported by the Developmental Fund of Shandong Science and Technology Department, Shandong, People’s Republic of China (2011YD18054).

Rights and permissions

© 2015 Han et al. This work is published by Dove Medical Press Limited, and licensed under Creative Commons Attribution – Non Commercial (unported, v3.0) License. The full terms of the License are available at http://creativecommons.org/licenses/by-nc/3.0/. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. Permissions beyond the scope of the License are administered by Dove Medical Press Limited. Information on how to request permission may be found at: http://www.dovepress.com/permissions.php

Return