You are on page 1of 7

Acta Neuropathol (2011) 122:543549 DOI 10.

1007/s00401-011-0878-z

REVIEW

Opportunities and challenges in developing Alzheimer disease therapeutics


Khalid Iqbal Inge Grundke-Iqbal

Received: 5 August 2011 / Revised: 17 September 2011 / Accepted: 17 September 2011 / Published online: 30 September 2011 Springer-Verlag 2011

Abstract Alzheimer disease (AD) is a chronic, progressive disorder with an average disease progression of 710 years. However, the histopathological hallmark lesions of this disease, the extracellular Ab plaques and the intraneuronal neurobrillary tangles, start as early as childhood in the affected individuals. AD is multifactorial and probably involves many different etiopathogenic mechanisms. Thus, while AD offers a wide window of opportunity that practically includes the whole life span of the affected individuals, and numerous therapeutic targets, the multifactorial nature of this disease also makes the selection of the therapeutic targets an immensely challenging task. In addition to b-amyloidosis and neurobrillary degeneration, the AD brain also is compromised in its ability to regenerate by enhancing neurogenesis and neuronal plasticity. An increasing number of preclinical studies in transgenic mouse models of AD show that enhancement of neurogenesis and neuronal plasticity can reverse cognitive impairment. Development of both drugs that can inhibit neurodegeneration and drugs that can increase the regenerative capacity of the brain by enhancing neurogenesis and neuronal plasticity are required to control AD. Keywords Alzheimer disease Abnormally hyperphosphorylated tau Neurogenesis Neuronal plasticity Ciliary neurotrophic factor

Introduction Alzheimer disease (AD) is the single major cause of dementia in the middle- to old-aged individuals. Currently, over 35 million people worldwide are suffering from AD and this number is projected to triple by 2050 if no drug is developed that can prevent or inhibit this disease. AD is multifactorial and probably involves several different etiopathogenic mechanisms [42, 43]. The familial form of AD, which accounts for \1% of all cases, is caused by certain point mutations in b-amyloid precursor protein, presenilin 1 or presenilin 2 genes [7]. The exact causes of the sporadic form of AD, which accounts for over 99% of the cases, are not yet understood. Individuals who inherit one or two APOE4 alleles carry a *3.5-fold or*10-fold risk, respectively, of coming down with AD [20]. Histopathologically the familial and the sporadic forms of AD are indistinguishable from each other and are characterized by neurodegeneration of the brain, especially the hippocampus and the rest of the neocortex that is associated with numerous intraneuronal neurobrillary tangles and the extracellular deposits of b-amyloid as cores of neuritic (senile) plaques. Although the discoveries of Ab, which is seen both as plaque core b-amyloid and as congophilic angiopathy [33, 60] and of abnormal hyperphosphorylation of tau as the protein subunit of paired helical laments (PHF)/neurobrillary tangles [35, 44] were made in around the same period, the immense popularity of the Amyloid Cascade Hypothesis, according to which b-amyloid is the primary cause of neurodegeneration and dementia in AD [36, 37] resulted in Ab as the focus of a large majority of studies on biology and drug development of AD. However, to date, Ab-based therapeutics of AD have been unsuccessful. While on one hand

This article is dedicated to the celebration of Prof. Kurt Jellingers 80th birthday, which was on May 28th. K. Iqbal (&) I. Grundke-Iqbal Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314-6399, USA e-mail: khalid.iqbal.ibr@gmail.com

123

544

Acta Neuropathol (2011) 122:543549

it is truly a great setback in the development of diseasemodifying drugs, it has increased awareness of the involvement of several different etiopathogenic mechanisms and stimulated research on non-Ab-based therapeutic approaches to this disease.

Challenges To date therapeutic attempts, which included inhibition of Ab production, its aggregation as well as removal from the brain, have all been unsuccessful. Based on what is known about AD and Ab to date, there could be four major reasons for the failure of the Ab-based therapeutics: First, b-amyloid could be a non-deleterious marker and not a cause of the disease. It is well established that as many as 30% of the normal elderly have as much b-amyloid plaque load as typical cases of AD, and the number of plaques in AD does not correlate with the degree of dementia [5, 21, 50, 75]. Only some of the presenilin-1 and presenilin-2 mutations that produce AD result in increased brain levels of Ab; some of the AD-causing mutations either result in no change or a decrease in brain Ab levels [69, 73]. While in cultured cells and in experimental animals Ab has been found to be neurotoxic, these ndings were made with either treatment or overexpression with a very high non-physiological concentration of Ab. Although a lot has been learned about Ab during the last *25 years, there is still not any conclusive evidence and, thus, agreement on what form, state, cellular/extra cellular location, if and how Ab causes AD. Another possibility is that inhibition or removal of Ab alone is not enough to inhibit AD. Both in cultured cells and in vivo in transgenic mice studies have shown that Ab neurotoxicity requires tau [72, 74]. Thus, Ab-based therapy with a concomitant tau-based therapy might be required for successful treatment of AD. Second, the Ab-based therapeutics employed so far were not potent enough to ameliorate the disease. In the case of Flurizan (Myriad Genetics, USA), a c-secretase inhibitor, the drug had no serious side effects but failed in Phase III clinical trials. Samagucestat (Eli Lilly & Company, USA), a potent c-secretase inhibitor, made AD patients worse as well as increased the risk for skin cancer, probably due to non-selectivity of this drug to c-secretase activities towards other substrate proteins; there are about 50 other proteins including NOTCH which are c-secretase substrates. Alzhamed (Neurochem, Inc., Canada), an Ab aggregation inhibitor, Tramiprosate, had no serious side effects and failed in Phase III clinical trials. Ab vaccine (Elan Corporation, Ireland) successfully removed Ab plaques from brain parenchyma but increased congophilic angiopathy and in around 5% of the subjects caused meningoencephalitis and the Phase III clinical trial had to be halted. However, the treated patients failed to show any inhibition of cognitive deterioration. Development of an Ab vaccine that does not produce congophilic angiopathy and meningoencephalitis is eagerly awaited. Unlike active, the passive immunization using a monoclonal antibody to Ab,

Opportunities AD is a chronic, progressive, neurodegenerative disease with an average progression of 710 years. However, the histopathological hallmarks of this disease, the neurobrillary tangles of abnormally hyperphosphorylated tau and Ab plaques, are known to occur many years before the clinical expression of the disease [12]. A recent study by Braak and Tredici [13] have shown that neurobrillary degeneration of abnormally hyperphosphorylated tau occurs as early as in early childhood and starts from select subcortical nuclei. Neurodegeneration of the AD type probably occurs throughout the life of an individual and clinically manifests when it crosses a certain threshold. In the familial form of AD, which is caused by certain mutations, this process is mostly more accelerated than in the sporadic form and, thus, results in dementia at an earlier age. In the case of Down syndrome, a developmental disease with severe mental retardation, which is caused by an extra copy of chromosome 21, in the fourth decade of life without fail these affected individuals develop AD histopathology, i.e. numerous plaques and tangles in the forebrain. It is possible that, like Down syndrome, AD is a developmental disorder, the clinical phenotype of which does not become apparent until middle- to old-age. Thus, AD offers for therapeutic treatment a window of opportunity that extends practically the whole life span of the affected individuals. There are at least ve subgroups of sporadic AD. These subgroups, each of which displays different clinical proles, were identied based on the CSF levels of Ab142, total tau, and ubiquitin [42]. Though AD is histopathologically characterized by the presence of numerous Ab plaques and neurobrillary tangles of abnormally hyperphosphorylated tau, each of these lesions can result from different etiological factors and upstream molecular mechanisms. For instance, dysregulation of a-, b-, or csecretase activity can all lead to b-amyloidosis [19, 65, 67]. The abnormal hyperphosphorylation of tau that leads to its aggregation into paired helical laments that form neurobrillary tangles and neuropil threads can be generated by several different combinations of proline-directed protein kinases (PDPKs) and non-PDPKS [89]. These reports are consistent with the involvement of several different etiopathogenic mechanisms of AD. Thus, AD offers a large number of therapeutic targets.

123

Acta Neuropathol (2011) 122:543549

545

Bapineuzumab, failed to show any clinical improvement in a large Phase II clinical trial carried out by Elan Corp. Third, all Ab-based therapies were tested in mild to moderate cases of AD which was too late to see any inhibition of cognitive decline. AD is a chronic, progressive, neurodegenerative disease where the pathology starts decades before the onset of any clinically detectable signs. Principally, the earlier the better and the easier it is to treat a disease. However, given the fact that AD is a chronic, progressive, neurodegenerative disease where the pathology starts several decades before the clinical onset of the disease, it is unlikely that the Ab drugs were unsuccessful because clinically diagnosed mild to moderate and not predromal state patients were treated. Fourth, the Ab-based drugs might be effective only towards a small subgroup of this multifactorial disease. There are at least ve subgroups of AD and in one of these ve subgroups, called HARO, the CSF Ab levels are elevated whereas in the remaining four subgroups, AELO, ATEO, LEBALO and ATURO, it is the opposite [42]. If Ab-based therapies are effective only towards a specic small subgroup of AD, it will be difcult to see any positive outcome without stratifying patients into various subgroups. The multifactorial nature and the likely involvement of several different etiopathogenic mechanisms pose the most difcult challenge for the development of AD therapeutics. To develop rational therapeutic strategies and drugs, biomarkers and procedures to identify various subgroups as well as determination of the etiopathogenesis of each subgroup are required (Fig. 1).
Fig. 1 Multifactorial nature of Alzheimer disease and involvement of several different disease mechanisms. APP, b-amyloid precursor protein; PS1, presenilin 1; PS2, presenilin 2; Inam, inammation; SETa, inhibitor-2 of protein phosphatase 2A; TBD, to be determined

Neuroregeneration, a therapeutic strategy Independent of the various etiopathogenic mechanisms involved in AD, they all cause neurodegeneration. Thus, a successful therapeutic strategy for AD may include both inhibition of neurodegeneration as well as stimulation of regeneration of the affected areas of the brain. The latter can be achieved by drugs that can promote both neurogenesis and neuronal plasticity. Several lines of evidence are consistent with the involvement of neurogenesis in memory in the adult brain. In particular, adult-born hippocampal neurons have been implicated in complex forms of spatial or associative memories [2, 3, 53, 57]. Dysregulation of neurotrophic activities, either due to age, genetic background or other unknown factors, has been implicated in neurodegeneration and mood disorders [38]. There is an imbalance between neurogenesis and neurodegeneration in AD and other neurodegenerative disorders [30, 41, 68]. Several studies have suggested that age-associated decline in neurogenesis might contribute to a pathological condition and the associated learning and memory decline in AD [46, 55] and in transgenic mouse models of this disease [26, 28, 39, 40, 88]. The neurogenic decline and associated cognitive impairment happen prior to the formation of any Ab plaques or neurobrillary tangles in 3xTg-AD mice, suggesting that the down regulation of neurogenesis could be a component of the primary pathology caused by the expressions of mutated human APP, presenilin 1 and tau in these animals [9]. Neuronal survival during maturation is believed to depend on the surrounding microenvironment.

123

546 Fig. 2 Pathogenesis of Alzheimer disease and the two major therapeutic strategies

Acta Neuropathol (2011) 122:543549

The microenvironment of the dentate gyrus (DG) in neurodegenerative conditions apparently becomes adverse for maintaining greater levels of neurogenesis [34, 87]. In AD, the DG neuroproliferation is increased [45] but the newly generated neurons apparently do not mature [55]. Both newly born immature and mature neurons are believed to have an inherent advantage to be recruited into patterns of new memory networks [48] and are necessary for complex forms of hippocampal-mediated learning [3, 29]. The hippocampus is particularly vulnerable to neurodegeneration and hippocampal-dependent memory impairment is reported as the earliest symptom of dementia [6]. Thus, considering the regenerative ability of the brain, treatments promoting neuronal differentiation enriching the biochemical brain milieu could be a successful therapy for AD and related neurodegenerative disorders [8, 9, 18, 49, 54, 88]. In AD, the most signicant correlate to the severity of cognitive impairment is the synaptic loss in the frontal cortex and the limbic system [24, 25, 59, 82]. In the mature brain, neurogenesis is believed to play an important role in maintaining synaptic plasticity and memory formation in the hippocampus [86]. Both AD as well as transgenic mouse models of AD show signicant alterations in the process of neurogenesis in the hippocampus [17, 2628, 45, 88, 90]. Thus, alterations in synaptic plasticity in AD might not only involve direct damage to the synapses, but also interference with neurogenesis. Neurogenesis in the aging brain can be promoted by increasing the level of pro-neurogenic factors like neurosteroids [47, 61], cell-cycle regulators [62], NMDA receptor antagonists [63], and growth factors [1, 4, 46, 71, 83]. Neurotrophins and neurokines have been shown to be involved in the promotion of survival of subsets of neurons

vulnerable in neurodegenerative diseases [23, 76, 78, 85]. Several different approaches have been employed to enhance neurogenesis and/or neuronal plasticity to improve cognition in different animal models of AD. These strategies included direct implantation of neural stem cells in the brain of 3xTgAD mice [10]; stimulation of hematopoietic stem cell production by subcutaneous administration of granulocyte colony stimulating factor in Tg2576 and Tg-APP/PS1 mice [77, 84]; intraperitoneal administration of macrophage colony stimulating factor in Tg-APP/PS1 mice [11]; delivery of CNTF by implantation of recombinant cells secreting the neurotrophic factor encapsulated in alginate polymers [32]; and the entorhinal administration of the brain-derived neurotrophic factor in several animal models of AD [64]; the neuroprotective effect observed in this latter study was through amyloid-independent mechanisms (Fig. 2). Growth factors such as insulin-like growth factor (IGF-1) [56], epidermal growth factor (EGF), and broblast growth factor (FGF-2) [46] or a reduction of corticosteroids level by adrenalectomy [14] can at least partially negate the effect of age on the rate of neural stem proliferation. This environmentdependent positive regulation of neurogenesis supports the idea that the age-associated loss of new neurons is not an irreversible mechanism which, if triggered by appropriate signals, can be reactivated in the senescent brain.

Enhancement of neurogenesis and neuronal plasticity with ciliary neurotrophic factor peptidergic drugs Ciliary neurotrophic factor (CNTF) promotes neurogenesis both in hippocampus and subventricular zone [31, 91]. In

123

Acta Neuropathol (2011) 122:543549

547

the brain CNTF is expressed in subsets of astrocytes in the neurogenic regions, whereas its receptor, CNTFRa, seems to be expressed mostly in progenitor cells and neurons of the hippocampus and various other areas of the brain, including motorcortex and cerebellum [31, 52, 80]. CNTF belongs to the IL-6 family of cytokines which also includes IL-11, leukemia inhibitor factor (LIF), oncostatin-M, cardiotrophin-1, and cardiotrophin-like cytokine [79, 81]. CNTF signaling occurs through the formation of a tripartite complex of CNTFRa, the LIFb receptor (LIFR) and glycoprotein 130 (gp130). CNTF and LIF both signal through tyrosine phosphorylation of the signal transducers and activators of transcription (STAT) proteins by the membrane-associated Janus kinase (JAK) [22]. Upon injury of the brain, the expression of both CNTF and CNTFRa increases [51, 52, 58]. Like other neurotrophins [70], the therapeutic potential of exogenous CNTF is eclipsed by its short half-life when administered peripherally, requiring an invasive mode of administration with unpredictable pharmacokinetics [16]. Moreover, the clinical use of CNTF, due to its serious side effects, i.e. anorexia, skeletal muscle loss, hyperalgesia, cramps and muscle pain, has not materialized. In our laboratory, employing neutralizing antibodies to CNTF, we identied the amino acid residues 146156 as an active region of this neurotrophic factor [15, 18]. Peripheral administration of this 11-mer CNTF peptide, named Peptide 6, for 30 days enhanced dentate gyrus neurogenesis and neuronal plasticity in normal adult C57BL6 mice [18]. This peptide, Peptide 6, induced proliferation and increased survival and maturation of neural progenitor cells into neurons in the dentate gyrus. Furthermore, Peptide 6 increased the MAP2 and synaptophysin immunoreactivity in the dentate gyrus. The 30-day treatment with a slow release bolus of the peptide implanted subcutaneously improved reference memory of the mice in the Morris water maze. Peptide 6 had a plasma half-life of over 6 h, was bloodbrain barrier permeable, and acted by competitively inhibiting the LIF signaling. Like AD, several transgenic mouse models of this disease show failed hippocampal neurogenesis and cognitive impairment. The triple transgenic AD (3xTg0-AD) mouse represents one of the most biologically relevant animal models of AD described so far [66]. The 3xTg-AD mice harbor three AD-related genetic loci: human PS1M146V, human APPSWE, and human tauP301L. These mice develop b-amyloid plaques and neurobrillary tangle-like pathologies in a progressive and age-dependent manner, starting at around 12 months but show cognitive impairment as early as around 5 months. Treatment of 6- to 7-month-old 3xTgAD mice with intraperitoneal administration of Peptide 6 for 6 weeks restored cognition by enhancing dentate gyrus neurogenesis and neuronal plasticity in these animals [9].

Interestingly, the treatment with Peptide 6 had no detectable effect on Ab and tau pathologies, which at this age in these mice is seen as intraneuronal accumulation of Ab and tau and not as plaques and tangles. In subsequent studies we narrowed down the minimal active region of Peptide 6 to 4 amino acids, D G G L [8]. The neurogenic and neurotrophic activities of this tetrapeptide, Peptide 6c, are preserved when it is carboxy adamantylated to enhance its lipophilicity [54]. Thus, preclinical studies clearly suggest enhancement of neurogenesis and neuronal plasticity as a promising approach to restore cognition in AD and related neurodegenerative cognitive disorders.
Acknowledgments We are grateful to Janet Murphy for secretarial assistance. Studies from our lab described in this article were supported in part by NIH grants AG019158, AG028538, Alzheimers Association grant IIRG-06-25836, a research grant from EVER Neuropharma, Unteract, Austria, and by the New York State Ofce of People with Developmental Disabilities.

References
1. Aberg MA, Aberg ND, Hedbacker H, Oscarsson J, Eriksson PS (2000) Peripheral infusion of IGF-I selectively induces neurogenesis in the adult rat hippocampus. J Neurosci 20:28962903 2. Abrous DN, Wojtowics JM (2008) Neurogenesis and hippocampal memory system, in adult neurogenesis. Cold Spring Harbor Press, New York 3. Aimone JB, Wiles J, Gage FH (2006) Potential role for adult neurogenesis in the encoding of time in new memories. Nat Neurosci 9:723727 4. Anderson MF, Aberg MA, Nilsson M, Eriksson PS (2002) Insulin-like growth factor-I and neurogenesis in the adult mammalian brain. Brain Res Dev Brain Res 134:115122 5. Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT (1992) Neurobrillary tangles but not senile plaques parallel duration and severity of Alzheimers disease. Neurology 42:631639 6. Belleville S, Sylvain-Roy S, de Boysson C, Menard MC (2008) Characterizing the memory changes in persons with mild cognitive impairment. Prog Brain Res 169:365375 7. Bird TD (2008) Genetic aspects of Alzheimer disease. Genet Med 10:231239 8. Blanchard J, Chohan MO, Li B, Liu F, Iqbal K, Grundke-Iqbal I (2010) Benecial effect of a CNTF tetrapeptide on adult hippocampal neurogenesis, neuronal plasticity and spatial memory in mice. J Alzheimers Dis 21:11851195 9. Blanchard J, Wanka L, Tung YC et al (2010) Pharmacologic reversal of neurogenic and neuroplastic abnormalities and cognitive impairments without affecting Abeta and tau pathologies in 3xTg-AD mice. Acta Neuropathol 120:605621 10. Blurton-Jones M, Kitazawa M, Martinez-Coria H et al (2009) Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci USA 106:1359413599 11. Boissonneault V, Filali M, Lessard M, Relton J, Wong G, Rivest S (2009) Powerful benecial effects of macrophage colonystimulating factor on beta-amyloid deposition and cognitive impairment in Alzheimers disease. Brain 132:10781092 12. Braak H, Braak E (1997) Frequency of stages of Alzheimerrelated lesions in different age categories. Neurobiol Aging 18:351357

123

548 13. Braak H, Del Tredici K (2011) The pathological process underlying Alzheimers disease in individuals under thirty. Acta Neuropathol 121:171181 14. Cameron HA, McKay RD (1999) Restoring production of hippocampal neurons in old age. Nat Neurosci 2:894897 15. Chen H, Tung YC, Li B, Iqbal K, Grundke-Iqbal I (2007) Trophic factors counteract elevated FGF-2-induced inhibition of adult neurogenesis. Neurobiol Aging 28:11481162 16. Chen ZY, Cao L, Wang LM et al (2001) Development of neurotrophic molecules for treatment of neurodegeneration. Curr Protein Pept Sci 2:261276 17. Chevallier NL, Soriano S, Kang DE, Masliah E, Hu G, Koo EH (2005) Perturbed neurogenesis in the adult hippocampus associated with presenilin-1 A246E mutation. Am J Pathol 167:151159 18. Chohan MO, Li B, Blanchard J et al (2011) Enhancement of dentate gyrus neurogenesis, dendritic and synaptic plasticity and memory by a neurotrophic peptide. Neurobiol Aging 32:14201434 19. Chow VW, Mattson MP, Wong PC, Gleichmann M (2010) An overview of APP processing enzymes and products. Neuromolecular Med 12:112 20. Corder EH, Saunders AM, Strittmatter WJ et al (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimers disease in late onset families. Science 261:921923 21. Crystal HA, Dickson DW, Sliwinski MJ et al (1993) Pathological markers associated with normal aging and dementia in the elderly. Ann Neurol 34:566573 22. Davis S, Aldrich TH, Stahl N et al (1993) LIFR beta and gp130 as heterodimerizing signal transducers of the tripartite CNTF receptor. Science 260:18051808 23. Dawbarn D, Allen SJ (2003) Neurotrophins and neurodegeneration. Neuropathol Appl Neurobiol 29:211230 24. DeKosky ST, Scheff SW (1990) Synapse loss in frontal cortex biopsies in Alzheimers disease: correlation with cognitive severity. Ann Neurol 27:457464 25. DeKosky ST, Scheff SW, Styren SD (1996) Structural correlates of cognition in dementia: quantication and assessment of synapse change. Neurodegeneration 5:417421 26. Demars M, Hu YS, Gadadhar ALazarov O (2010) Impaired neurogenesis is an early event in the etiology of familial Alzheimers disease in transgenic mice. J Neurosci Res 88:21032117 27. Dong H, Goico B, Martin M, Csernansky CA, Bertchume AC, sernansky JG (2004) Modulation of hippocampal cell proliferation, memory, and amyloid plaque deposition in APPsw (Tg2576) mutant mice by isolation stress. Neuroscience 127:601609 28. Donovan MH, Yazdani U, Norris RD, Games D, German DC, Eisch AJ (2006) Decreased adult hippocampal neurogenesis in the PDAPP mouse model of Alzheimers disease. J Comp Neurol 495:7083 29. Dupret D, Revest JM, Koehl M et al (2008) Spatial relational memory requires hippocampal adult neurogenesis. PLoS One 3:e1959 30. Durany N, Michel T, Kurt J, Cruz-Sanchez FF, Cervas-Navarro J, Riederer P (2000) Brain-derived neurotrophic factor and neurotrophin-3 levels in Alzheimers disease brains. Int J Dev Neurosci 18:807813 31. Emsley JG, Hagg T (2003) Endogenous and exogenous ciliary neurotrophic factor enhances forebrain neurogenesis in adult mice. Exp Neurol 183:298310 32. Garcia P, Youssef I, Utvik JK et al (2010) Ciliary neurotrophic factor cell-based delivery prevents synaptic impairment and improves memory in mouse models of Alzheimers disease. J Neurosci 30:75167527 33. Glenner GG, Wong CW (1984) Alzheimers disease: initial report of the purication and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun 120:885890

Acta Neuropathol (2011) 122:543549 34. Grote HE, Hannan AJ (2007) Regulators of adult neurogenesis in the healthy and diseased brain. Clin Exp Pharmacol Physiol 34:533545 35. Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI (1986) Abnormal phosphorylation of the microtubuleassociated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci USA 83:49134917 36. Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimers disease: progress and problems on the road to therapeutics. Science 297:353356 37. Hardy JA, Higgins GA (1992) Alzheimers disease: the amyloid cascade hypothesis. Science 256:184185 38. Hashimoto K, Shimizu EIyo M (2004) Critical role of brainderived neurotrophic factor in mood disorders. Brain Res Brain Res Rev 45:104114 39. Haughey NJ, Liu D, Nath A, Borchard AC, Mattson MP (2002) Disruption of neurogenesis in the subventricular zone of adult mice, and in human cortical neuronal precursor cells in culture, by amyloid beta-peptide: implications for the pathogenesis of Alzheimers disease. Neuromolecular Med 1:125135 40. Haughey NJ, Nath A, Chan SL, Borchard AC, Rao MS, Mattson MP (2002) Disruption of neurogenesis by amyloid beta-peptide, and perturbed neural progenitor cell homeostasis, in models of Alzheimers disease. J Neurochem 83:15091524 41. Hock C, Heese K, Hulette C, Rosenberg C, Otten U (2000) Region-specic neurotrophin imbalances in Alzheimer disease: decreased levels of brain-derived neurotrophic factor and increased levels of nerve growth factor in hippocampus and cortical areas. Arch Neurol 57:846851 42. Iqbal K, Flory M, Khatoon S et al (2005) Subgroups of Alzheimers disease based on cerebrospinal uid molecular markers. Ann Neurol 58:748757 43. Iqbal K, Grundke-Iqbal I (2010) Alzheimers disease, a multifactorial disorder seeking multitherapies. Alzheimers Dementia 6:420424 44. Iqbal K, Grundke-Iqbal I, Zaidi T et al (1986) Defective brain microtubule assembly in Alzheimers disease. Lancet 2:421426 45. Jin K, Galvan V, Xie L et al (2004) Enhanced neurogenesis in Alzheimers disease transgenic (PDGF-APPSw, Ind) mice. Proc Natl Acad Sci USA 101:1336313367 46. Jin K, Xie L, Childs J et al (2003) Cerebral neurogenesis is induced by intranasal administration of growth factors. Ann Neurol 53:405409 47. Karishma KK, Herbert J (2002) Dehydroepiandrosterone (DHEA) stimulates neurogenesis in the hippocampus of the rat, promotes survival of newly formed neurons and prevents corticosterone-induced suppression. Eur J Neurosci 16:445453 48. Kee N, Teixeira CM, Wang AH, Frankland PW (2007) Preferential incorporation of adult-generated granule cells into spatial memory networks in the dentate gyrus. Nat Neurosci 10:355362 49. Kimura T, Hong Nguyen PT, Ho SA, Tran AH, Ono T, Nishijo H (2009) T-817MA, a neurotrophic agent, ameliorates the decits in adult neurogenesis and spatial memory in rats infused i.c.v. with amyloid-beta peptide. Br J Pharmacol 157:451463 50. Knopman DS, Parisi JE, Salviati A et al (2003) Neuropathology of cognitively normal elderly. J Neuropathol Exp Neurol 62:10871095 51. Kordower JH, Yaping C, Maclennan AJ (1997) Ciliary neurotrophic factor receptor alpha-immunoreactivity in the monkey central nervous system. J Comp Neurol 377:365380 52. Lee MY, Deller T, Kirsch M, Frotscher M, Hofmann HD (1997) Differential regulation of ciliary neurotrophic factor (CNTF) and CNTF receptor alpha expression in astrocytes and neurons of the fascia dentata after entorhinal cortex lesion. J Neurosci 17:11371146

123

Acta Neuropathol (2011) 122:543549 53. Leuner B, Gould E, Shors TJ (2006) Is there a link between adult neurogenesis and learning? Hippocampus 16:216224 54. Li B, Wanka L, Blanchard J et al (2010) Neurotrophic peptides incorporating adamantane improve learning and memory, promote neurogenesis and synaptic plasticity in mice. FEBS Lett 584:33593365 55. Li B, Yamamori H, Tatebayashi Y et al (2008) Failure of neuronal maturation in Alzheimer disease dentate gyrus. J Neuropathol Exp Neurol 67:7884 56. Lichtenwalner RJ, Forbes ME, Bennett SA, Lynch CD, Sonntag WE, Riddle DR (2001) Intracerebroventricular infusion of insulin-like growth factor-I ameliorates the age-related decline in hippocampal neurogenesis. Neuroscience 107:603613 57. Lledo PM, Alonso M, Grubb MS (2006) Adult neurogenesis and functional plasticity in neuronal circuits. Nat Rev Neurosci 7:179193 58. MacLennan AJ, Vinson EN, Marks L, McLaurin DL, Pfeifer M, Lee N (1996) Immunohistochemical localization of ciliary neurotrophic factor receptor alpha expression in the rat nervous system. J Neurosci 16:621630 59. Masliah E, Mallory M, Hansen L, DeTeresa R, Alford M, Terry R (1994) Synaptic and neuritic alterations during the progression of Alzheimers disease. Neurosci Lett 174:6772 60. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K (1985) Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci USA 82:42454249 61. Mayo W, George O, Darbra S et al (2003) Individual differences in cognitive aging: implication of pregnenolone sulfate. Prog Neurobiol 71:4348 62. Molofsky AV, He S, Bydon M, Morrison SJ, Pardal R (2005) Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16Ink4a and p19Arf senescence pathways. Genes Dev 19:14321437 63. Nacher J, Alonso-Llosa G, Rosell DR, McEwen BS (2003) NMDA receptor antagonist treatment increases the production of new neurons in the aged rat hippocampus. Neurobiol Aging 24:273284 64. Nagahara AH, Merrill DA, Coppola G et al (2009) Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimers disease. Nat Med 15:331337 65. Nathalie P, Jean-Noel O (2008) Processing of amyloid precursor protein and amyloid peptide neurotoxicity. Curr Alzheimer Res 5:9299 66. Oddo S, Caccamo A, Shepherd JD et al (2003) Triple-transgenic model of Alzheimers disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron 39:409421 67. Palop JJ, Mucke L (2010) Amyloid-beta-induced neuronal dysfunction in Alzheimers disease: from synapses toward neural networks. Nat Neurosci 13:812818 68. Phillips W, Michell AW, Barker RA (2006) Neurogenesis in diseases of the central nervous system. Stem Cells Dev 15:359379 69. Pimplikar SW, Nixon RA, Robakis NK, Shen J, Tsai LH (2010) Amyloid-independent mechanisms in Alzheimers disease pathogenesis. J Neurosci 30:1494614954 70. Price RD, Milne SA, Sharkey J, Matsuoka N (2007) Advances in small molecules promoting neurotrophic function. Pharmacol Ther 115:292306 71. Rai KS, Hattiangady B, Shetty AK (2007) Enhanced production and dendritic growth of new dentate granule cells in the middleaged hippocampus following intracerebroventricular FGF-2 infusions. Eur J Neurosci 26:17651779

549 72. Rapoport M, Dawson HN, Binder LI, Vitek MP, Ferreira A (2002) Tau is essential to beta-amyloid-induced neurotoxicity. Proc Natl Acad Sci USA 99:63646369 73. Robakis NK (2010) Mechanisms of AD neurodegeneration may be independent of Abeta and its derivatives. Neurobiol Aging 32:372379 74. Roberson ED, Scearce-Levie K, Palop JJ et al (2007) Reducing endogenous tau ameliorates amyloid beta-induced decits in an Alzheimers disease mouse model. Science 316:750754 75. Rodrigue KM, Kennedy KM, Park DC (2009) Beta-amyloid deposition and the aging brain. Neuropsychol Rev 19:436450 76. Rosenblad C (2004) Growth factor treatment of neurodegenerative disorders: new developments pave the way for clinical success. IDrugs 7:243248 77. Sanchez-Ramos J, Song S, Sava V et al (2009) Granulocyte colony stimulating factor decreases brain amyloid burden and reverses cognitive impairment in Alzheimers mice. Neuroscience 163:5572 78. Schulte-Herbruggen O, Braun A, Rochlitzer S, Jockers-Scherubl MC, Hellweg R (2007) Neurotrophic factorsa tool for therapeutic strategies in neurological, neuropsychiatric and neuroimmunological diseases? Curr Med Chem 14:23182329 79. Senaldi G, Varnum BC, Sarmiento U et al (1999) Novel neurotrophin-1/B cell-stimulating factor-3: a cytokine of the IL-6 family. Proc Natl Acad Sci USA 96:1145811463 80. Sendtner M, Carroll P, Holtmann B, Hughes RA, Thoenen H (1994) Ciliary neurotrophic factor. J Neurobiol 25:14361453 81. Shi Y, Wang W, Yourey PA et al (1999) Computational EST database analysis identies a novel member of the neuropoietic cytokine family. Biochem Biophys Res Commun 262:132138 82. Terry RD, Masliah E, Salmon DP et al (1991) Physical basis of cognitive alterations in Alzheimers disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol 30:572580 83. Trejo JL, Carro E, Torres-Aleman I (2001) Circulating insulinlike growth factor I mediates exercise-induced increases in the number of new neurons in the adult hippocampus. J Neurosci 21:16281634 84. Tsai KJ, Tsai YC, Shen CK (2007) G-CSF rescues the memory impairment of animal models of Alzheimers disease. J Exp Med 204:12731280 85. Tuszynski MH, Thal L, Pay M et al (2005) A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med 11:551555 86. van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, Gage FH (2002) Functional neurogenesis in the adult hippocampus. Nature 415:10301034 87. Verret L, Jankowsky JL, Xu GM, Borchelt DR, Rampon C (2007) Alzheimers-type amyloidosis in transgenic mice impairs survival of newborn neurons derived from adult hippocampal neurogenesis. J Neurosci 27:67716780 88. Wang JM, Singh C, Liu L et al (2010) Allopregnanolone reverses neurogenic and cognitive decits in mouse model of Alzheimers disease. Proc Natl Acad Sci USA 107:64986503 89. Wang JZ, Grundke-Iqbal I, Iqbal K (2007) Kinases and phosphatases and tau sites involved in Alzheimer neurobrillary degeneration. Eur J Neurosci 25:5968 90. Wen PH, Hof PR, Chen X et al (2004) The presenilin-1 familial Alzheimer disease mutant P117L impairs neurogenesis in the hippocampus of adult mice. Exp Neurol 188:224237 91. Yang P, Arnold SA, Habas A, Hetman M, Hagg T (2008) Ciliary neurotrophic factor mediates dopamine D2 receptor-induced CNS neurogenesis in adult mice. J Neurosci 28:22312241

123

You might also like