Journal Home > Volume 3 , Issue 1

Alzheimer’s disease (AD) is the most prevalent type of dementia, and its neuropathology is characterized by the deposition of insoluble β-amyloid peptides and intracellular neurofibrillary tangles and the loss of diverse neurons. Although much is known about the neurobiology of AD, few treatments are available to arrest or slow the illness. There is an urgent need for novel therapeutic approaches for AD. We reviewed the recent improvements in the neurorastorlogy strategies for AD, including medicine, bioengineering and neuromodulation and clinical cell therapy. We emphasized that cell therapy may be an promising treatment for AD.


menu
Abstract
Full text
Outline
About this article

Clinical neurorestorative progress in Alzheimer’s disease

Show Author's information Liyan Qiao1( )Hongyun Huang2,3Dafin F Muresanu4,5
Department of Neurology, Tsinghua University Yuquan Hospital,
Beijing Rehabilitation Hospital of Capital Medical University,
Beijing Hongtianji Neuroscience Academy, Beijing, People’s Republic of China
Department of Clinical Neurosciences, Iuliu Haţieganu University of Medicine and Pharmacy,
RoNeuro Institute for Neurological Research and Diagnosis, Cluj-Napoca, Romania

Abstract

Alzheimer’s disease (AD) is the most prevalent type of dementia, and its neuropathology is characterized by the deposition of insoluble β-amyloid peptides and intracellular neurofibrillary tangles and the loss of diverse neurons. Although much is known about the neurobiology of AD, few treatments are available to arrest or slow the illness. There is an urgent need for novel therapeutic approaches for AD. We reviewed the recent improvements in the neurorastorlogy strategies for AD, including medicine, bioengineering and neuromodulation and clinical cell therapy. We emphasized that cell therapy may be an promising treatment for AD.

Keywords: Alzheimer’s disease, cell therapy, neurorastorlogy, medicine

References(86)

1.
Victor M, Ropper AH. Degenerative diseases of the nervous system. In: Ropper AH, Samuels MA. Adams and Victor’s Principles of Neurology. 1106–1174.
2.
Alzheimer’s Association. 2014 Alzheimer’s disease facts and figures. Alzheimers Dement. 2014;10(2):e47–e92.
3.
Drachman DA. The amyloid hypothesis, time to move on: Amyloid is the downstream result, not cause, of Alzheimer’s disease. Alzheimers Dement. 2014;10(3):372–380.
4.
Hyman BT, Phelps CH, Beach TG, et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease. Alzheimers Dement. 2012;8(1):1–13.
5.
Nelson PT, Alafuzoff I, Bigio EH, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol. 2012;71(5):362–381.
6.
Parsons CG, Danysz W, Dekundy A, Pulte I. Memantine and cholinesterase inhibitors: complementary mechanisms in the treatment of Alzheimer’s disease. Neurotox Res. 2013;24(3):358–369.
7.
Muoio V, Persson PB, Sendeski MM. The neurovascular unit – concept review. Acta Physiol (Oxf). 2014;210(4):790–798.
8.
Bell RD, Zlokovic BV. Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer’s disease. Acta Neuropathol. 2009; 118(1):103–113.
9.
Baloyannis SJ, Baloyannis IS. The vascular factor in Alzheimer’s disease: a study in Golgi technique and electron microscopy. J Neurol Sci. 2012;322(1–2):117–121.
10.
Panza F, Frisardi V, Solfrizzi V, et al. Interacting with γ-secretase for treating Alzheimer’s disease: from inhibition to modulation. Curr Med Chem. 2011;18(35):5430–5447.
11.
Crump CJ, Johnson DS, Li YM. Development and mechanism of γ-secretase modulators for Alzheimer’s disease. Biochemistry. 2013;52(19):3197–3216.
12.
Doody RS, Raman R, Farlow M, et al; Alzheimer’s Disease Cooperative Study Steering Committee; Semagacestat Study Group. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med. 2013;369(4):341–350.
13.
Imbimbo BP, Giardina GA. γ-secretase inhibitors and modulators for the treatment of Alzheimer’s disease: disappointments and hopes. Curr Top Med Chem. 2011;11(12):1555–1570.
14.
Green RC, Schneider LS, Amato DA, et al; Tarenflurbil Phase 3 Study Group. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA. 2009;302(23):2557–2564.
15.
Wisniewski T, Goñi F. Immunotherapy for Alzheimer’s disease. Biochem Pharmacol. 2014;88(4):499–507.
16.
Lambracht-Washington D, Rosenberg RN. Anti-amyloid beta to tau – based immunization: Developments in immunotherapy for Alzheimer disease. Immunotargets Ther. 2013;2013(2):105–114.
17.
Lambracht-Washington D, Rosenberg RN. Advances in the development of vaccines for Alzheimer’s disease. Discov Med. 2013;15(84):319–326.
18.
Honig LS. Translational research in neurology: dementia. Arch Neurol. 2012;69(8):969–977.
19.
Loeffler DA. Intravenous immunoglobulin and Alzheimer’s disease: what now? J Neuroinflammation. 2013;10(1):70.
20.
Townsend KP, Praticò D. Novel therapeutic opportunities for Alzheimer’s disease: focus on nonsteroidal anti-inflammatory drugs. FASEB J. 2005;19(12):1592–1601.
21.
ADAPT-FS Research Group. Follow-up evaluation of cognitive function in the randomized Alzheimer’s Disease Anti-inflammatory Prevention Trial and its Follow-up Study. Alzheimers Dement. Epub July 8, 2014.
22.
Rockenstein E, Torrance M, Mante M, et al. Cerebrolysin decreases amyloid-beta production by regulating amyloid protein precursor maturation in a transgenic model of Alzheimer’s disease. J Neurosci Res. 2006;83(7):1252–1261.
23.
Ubhi K, Rockenstein E, Doppler E, et al. Neurofibrillary and neurodegenerative pathology in APP-transgenic mice injected with AAV2-mutant TAU: neuroprotective effects of Cerebrolysin. Acta Neuropathol. 2009;117(6):699–712.
24.
Ruether E, Husmann R, Kinzler E, et al. A 28-week, double-blind, placebo-controlled study with Cerebrolysin in patients with mild to moderate Alzheimer’s disease. Int Clin Psychopharmacol. 2001;16(5):253–263.
25.
Panisset M, Gauthier S, Moessler H, Windisch M; Cerebrolysin Study Group. Cerebrolysin in Alzheimer’s disease: a randomized, double-blind, placebo-controlled trial with a neurotrophic agent. J Neural Transm. 2002;109(7–8):1089–1104.
26.
Muresanu DF, Rainer M, Moessler H. Improved global function and activities of daily living in patients with AD: a placebo-controlled clinical study with the neurotrophic agent Cerebrolysin. J Neural Transm Suppl. 2002;(62):277–285.
27.
Alvarez XA, Cacabelos R, Laredo M, et al. A 24-week, double-blind, placebo-controlled study of three dosages of Cerebrolysin in patients with mild to moderate Alzheimer’s disease. Eur J Neurol. 2006;13(1):43–54.
28.
Alvarez XA, Sampedro C, Cacabelos R, et al. Reduced TNF-α and increased IGF-I levels in the serum of Alzheimer’s disease patients treated with the neurotrophic agent cerebrolysin. Int J Neuropsychopharmacol. 2009;12(7):867–872.
29.
Álvarez XA, Fuentes P. Cerebrolysin in Alzheimer’s Disease. Drugs Today (Barc). 2011;47(7):487–513.
30.
Plosker GL, Gauthier S. Cerebrolysin: a review of its use in dementia. Drugs Aging. 2009;26(11):893–915.
31.
Álvarez XA, Fuentes P. Cerebrolysin in Alzheimer’s disease. Drugs Today (Barc). 2011;47(7):487–513.
32.
Allegri RF, Guekht A. Cerebrolysin improves symptoms and delays progression in patients with Alzheimer’s disease and vascular dementia. Drugs Today (Barc). 2012;48 Suppl A:S25–S41.
33.
Iadecola C. The overlap between neurodegenerative and vascular factors in the pathogenesis of dementia. Acta Neuropathol. 2010;120(3):287–296.
34.
Muresanu DF, Popa-Wagner A, Stan A, Buga AM, Popescu BO. The vascular component of Alzheimer’s disease. Curr Neurovasc Res. 2014;11(2):168–176.
35.
Chen N, Yang M, Guo J, Zhou M, Zhu C, He L. Cerebrolysin for vascular dementia. Cochrane Database Syst Rev. 2013;1:CD008900.
36.
Blurton-Jones M, Spencer B, Michael S, et al. Neural stem cells genetically-modified to express neprilysin reduce pathology in Alzheimer transgenic models. Stem Cell Res Ther. 2014;5(2):46.
37.
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.
38.
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.
39.
Wu L, Sluiter AA, Guo HF, et al. Neural stem cells improve neuronal survival in cultured postmortem brain tissue from aged and Alzheimer patients. J Cell Mol Med. 2008;12(5A):1611–1621.
40.
Cenciarelli C, Budoni M, Mercanti D, et al. In vitro analysis of mouse neural stem cells genetically modified to stably express human NGF by a novel multigenic viral expression system. Neurol Res. 2006;28(5):505–512.
41.
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.
42.
Li LY, Li JT, Wu QY, et al. Transplantation of NGF-gene-modified bone marrow stromal cells into a rat model of Alzheimer’ disease. J Mol Neurosci. 2008;34(2):157–163.
43.
Liang J, Wu S, Zhao H, et al. Human umbilical cord mesenchymal stem cells derived from Wharton’s jelly differentiate into cholinergic-like neurons in vitro. Neurosci Lett. 2013;532:59–63.
44.
Tuszynski MH, Thal L, Pay M, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med. 2005;11(5):551–555.
45.
Nagahara AH, Tuszynski MH. Potential therapeutic uses of BDNF in neurological and psychiatric disorders. Nat Rev Drug Discov. 2011; 10(3):209–219.
46.
Ziabreva I, Perry E, Perry R, et al. Altered neurogenesis in Alzheimer’s disease. J Psychosom Res. 2006;61(3):311–316.
47.
Sanchez-Ramos J, Cimino C, Avila R, et al. Pilot study of granulocyte-colony stimulating factor for treatment of Alzheimer’s disease. J Alzheimers Dis. 2012;31(4):843–855.
48.
Laske C, Stellos K, Stransky E, et al. Decreased plasma and cerebrospinal fluid levels of stem cell factor in patients with early Alzheimer’s disease. J Alzheimers Dis. 2008;15(3):451–460.
49.
Lovell MA, Geiger H, Van Zant GE, Lynn BC, Markesbery WR. Isolation of neural precursor cells from Alzheimer’s disease and aged control postmortem brain. Neurobiol Aging. 2006;27(7):909–917.
50.
Seo SW, Lee JII, Kim CH, et al. A phase I trial of parenchymal injection of umbilical cord stem cells in Alzheimer’s disease. Alzheimers Dement. 2013;9(Suppl 4):S291.
51.
Laxton AW, Tang-Wai DF, McAndrews MP, et al. A phase I trial of deep brain stimulation of memory circuits in Alzheimer’s disease. Ann Neurol. 2010;68(4):521–534.
52.
Fontaine D, Deudon A, Lemaire JJ, et al. Symptomatic treatment of memory decline in Alzheimer’s disease by deep brain stimulation: a feasibility study. J Alzheimers Dis. 2013;34(1):315–323.
53.
Lefaucheur JP, André-Obadia N, Antal A, et al. Evidence-based guidelines on the therapeutic use of repetitive transcranial magnetic stimulation (rTMS). Clin Neurophysiol. Epub June 5, 2014.
54.
Rabey JM, Dobronevsky E, Aichenbaum S, Gonen O, Marton RG, Khaigrekht M. Repetitive transcranial magnetic stimulation combined with cognitive training is a safe and effective modality for the treatment of Alzheimer’s disease: a randomized, double-blind study. J Neural Transm. 2013;120(5):813–819.
55.
Eliasova I, Anderkova L, Marecek R, Rektorova I. Non-invasive brain stimulation of the right inferior frontal gyrus may improve attention in early Alzheimer’s disease: A pilot study. J Neurol Sci. Epub August 29, 2014.
56.
Demars M, Hu YS, Gadadhar A, Lazarov O. Impaired neurogenesis is an early event in the etiology of familial Alzheimer’s disease in transgenic mice. J Neurosci Res. 2010;88(10):2103–2117.
57.
Rodríguez JJ, Jones VC, Verkhratsky A. Impaired cell proliferation in the subventricular zone in an Alzheimer’s disease model. Neuroreport. 2009;20(10):907–912.
58.
Haughey NJ, Nath A, Chan SL, Borchard AC, Rao MS, Mattson MP. Disruption of neurogenesis by amyloid beta-peptide, and perturbed neural progenitor cell homeostasis, in models of Alzheimer’s disease. J Neurochem. 2002;83(6):1509–1524.
59.
Hayashi Y, Kashiwagi K, Ohta J, Nakajima M, Kawashima T, Yoshikawa K. Alzheimer amyloid protein precursor enhances proliferation of neural stem cells from fetal rat brain. Biochem Biophys Res Commun. 1994;205(1):936–943.
60.
Wen PH, Shao X, Shao Z, et al. Overexpression of wild type but not an FAD mutant presenilin-1 promotes neurogenesis in the hippocampus of adult mice. Neurobiol Dis. 2002;10(1):8–19.
61.
Chevallier NL, Soriano S, Kang DE, Masliah E, Hu G, Koo EH. Perturbed neurogenesis in the adult hippocampus associated with presenilin-1 A246E mutation. Am J Pathol. 2005;167(1):151–159.
62.
Zhang X, Jin G, Tian M, Qin J, Huang Z. The denervated hippocampus provides proper microenvironment for the survival and differentiation of neural progenitors. Neurosci Lett. 2007;414(2):115–120.
63.
Oh SH, Kim HN, Park HJ, Shin JY, Lee PH. Mesenchymal stem cells increase hippocampal neurogenesis and neuronal differentiation by enhancing the Wnt signaling pathway in Alzheimer’s disease model. Cell Transplant. March 7, 2014.
64.
Zilka N, Zilkova M, Kazmerova Z, Sarissky M, Cigankova V, Novak M. Mesenchymal stem cells rescue the Alzheimer’s disease cell model from cell death induced by misfolded truncated tau. Neuroscience. 2011;193:330–337.
65.
Shin JY, Park HJ, Kim HN, et al. Mesenchymal stem cells enhance autophagy and increase β-amyloid clearance in Alzheimer disease models. Autophagy. 2014;10(1):32–44.
66.
Zilka N, Zilkova M, Kazmerova Z, Sarissky M, Cigankova V, Novak M. Mesenchymal stem cells rescue the Alzheimer’s disease cell model from cell death induced by misfolded truncated tau. Neuroscience. 2011;193:330–337.
67.
Lee JK, Jin HK, Bae JS. Bone marrow-derived mesenchymal stem cells attenuate amyloid β-induced memory impairment and apoptosis by inhibiting neuronal cell death. Curr Alzheimer Res. 2010;7(6):540–548.
68.
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.
69.
Bae JS, Jin HK, Lee JK, Richardson JC, Carter JE. Bone marrow-derived mesenchymal stem cells contribute to the reduction of amyloid-β 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.
70.
Park D, Yang G, Bae DK, et al. Human adipose tissue-derived mesenchymal stem cells improve cognitive function and physical activity in ageing mice. J Neurosci Res. 2013;91(5):660–670.
71.
Kim S, Chang KA, Kim Ja, et al. The preventive and therapeutic effects of intravenous human adipose-derived stem cells in Alzheimer’s disease mice. PLoS One. 2012;7(9):e45757.
72.
Xuan AG, Luo M, Ji WD, Long DH. Effects of engrafted neural stem cells in Alzheimer’s disease rats. Neurosci Lett. 2009;450(2):167–171.
73.
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.
74.
Fujiwara N, Shimizu J, Takai K, et al. Restoration of spatial memory dysfunction of human APP transgenic mice by transplantation of neuronal precursors derived from human iPS cells. Neurosci Lett. 2013;557 Pt B:129–134.
75.
Ooi L, Sidhu K, Poljak A, et al. Induced pluripotent stem cells as tools for disease modelling and drug discovery in Alzheimer’s disease. J Neural Transm. 2013;120(1):103–111.
76.
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.
77.
Lovell MA, Geiger H, Van Zant GE, Lynn BC, Markesbery WR. Isolation of neural precursor cells from Alzheimer’s disease and aged control postmortem brain. Neurobiol Aging. 2006;27(7):909–917.
78.
Choi SS, Lee SR, Kim SU, Lee HJ. Alzheimer’s disease and stem cell therapy. Exp Neurobiol. 2014;23(1):45–52.
79.
Chang KA, Kim JA, Kim S, Jet al. Therapeutic potentials of neural stem cells treated with fluoxetine in Alzheimer’s disease. Neurochem Int. 2012;61(6):885–891.
80.
Chadwick W, Mitchell N, Caroll J, et al. Amitriptyline-mediated cognitive enhancement in aged 3× Tg Alzheimer’s disease mice is associated with neurogenesis and neurotrophic activity. PLoS One. 2011;6(6):e21660.
81.
Ridwan S, Bauer H, Frauenknecht K, Hefti K, von Pein H, Sommer CJ. Distribution of the hematopoietic growth factor G-CSF and its receptor in the adult human brain with specific reference to Alzheimer’s disease. J Anat. 2014;224(4):377–391.
82.
Prakash A, Medhi B, Chopra K. Granulocyte colony stimulating factor (GCSF) improves memory and neurobehavior in an amyloid-β induced experimental model of Alzheimer’s disease. Pharmacol Biochem Behav. 2013;110:46–57.
83.
Tsai KJ, Tsai YC, Shen CK. G-CSF rescues the memory impairment of animal models of Alzheimer’s disease. J Exp Med. 2007;204(6):1273–1280.
84.
Sanchez-Ramos J, Song S, Sava V, et al. Granulocyte colony stimulating factor decreases brain amyloid burden and reverses cognitive impairment in Alzheimer’s mice. Neuroscience. 2009;163(1):55–72.
85.
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-1α in an Alzheimer’s disease mouse model. Stem Cells. 2011;29(7):1075–1089.
86.
Parthsarathy V, Hölscher C. Chronic treatment with the GLP1 analogue liraglutide increases cell proliferation and differentiation into neurons in an AD mouse model. PLoS One. 2013;8(3):e58784.
Publication history
Copyright
Rights and permissions

Publication history

Published: 15 December 2014
Issue date: December 2015

Copyright

© 2015 The Author(s).

Rights and permissions

© 2015 Qiao 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