Despite the fact that Alzheimer’s (AD) and Parkinson’s disease (PD) are the two most common neurodegenerative diseases, their pathogenesis still remains largely not well understood. However, advances during the last few decades in the characterizations of rare mendelian mutations and common risk genes as well as the role of epigenetics have greatly enhanced our understanding for the role of heritability and environmental factors in the development of AD and PD. This paper reviews key findings along their molecular mechanisms while tries to provide an overview of the overall contribution of genetics and epigenetics in the two diseases. Moreover, recent findings on neuronal genetic instability are also mentioned and, finally, a model that tries to integrate the effects of genetics, epigenetics, environment and pathological processes is proposed supporting the hypothesis that few ‘seeding’ pathogenic foci are sufficient to produce generalized pathology.
Introduction and Insight from Twin Studies.
Alzheimer’s (AD) and Parkinson’s disease (PD) are the two most common neurodegenerative diseases affecting about 1% and 0.5-1% in 65-69 years old and up to 50% and 4% respectively in older groups.[1-3] The pathogenesis of the two diseases is heterogeneous and not well understood although the long-standing involvement of specific protein misfolding and aggregation is well characterised in each case. AD presents as an insidious and progressive deterioration of global cognitive functions and is histopathologically recognized by the accumulation of extracellular amyloid-β-containing (Aβ) plaques and intraneuronal deposits of hyperphosphorylated τ protein (pτ) in the form of neurofibrillary tangles (NFTs). Both soluble/prefibrillar and fibrillar Aβ together with pτ are highly implicated in axonal transport failure, deranged dendritic signalling, excitotoxicity and ultimately cell death.[4,5] Similarly, in PD, which predominantly but not solely affects the dopaminergic system leading to a wide range of motor and neuropsychiatric symptoms, α-synuclein-containing intraneuronal Lewis bodies (LBs) are considered hallmark of the disease,[6,7] while the role of deranged protein handling, ubiquitin proteasome system (UPS) and mitochondrial dysfunction, and increased oxidative stress are considered to be pivotal.[8,9]
Nonetheless, even if age and environment are important factors, the role of genetics has been and is still extensively investigated. The recognition of rare family clusters of early-onset AD and PD cases versus the common late-onset sporadic cases has led to a ‘genetic-dichotomy model’ according to which EOAD is associated with fully penetrant rare mutations while LOAD with common genetic polymorphisms that modify the susceptibility risk of individuals.[10,11] However, an appreciation of their overall contribution in the two diseases can be better achieved by looking into twin studies. Such studies in AD have indicated that genetics contribute 48-60% in it’s development.[12-14] On the other hand, cross sectional PD studies have surprisingly revealed very low concordance rates in monozygotic twins and overall heritability[15,16] and only a longitudinal study gave a heritability estimate of 40%[17] denoting the importance of environment and variability in disease onset. However, this does not exclude important genetic contributions, as genetic polymorphisms with low penetrance and gene-environment interactions can still be widely implicated.[18] Evermore, current lines of thought suggest that plastic and rigid epigenetic differences could actually account for environmental or even inherited factors.[19-21] Therefore, aim of this essay is to investigate the aetiological roles of genetics and epigenetics in AD and PD.
Genetic factors in AD
Both the amyloid cascade and other derived hypotheses underline the central role of Aβ in AD pathogenesis.[4,5] Aβ is a 38-43 residue peptide physiologically produced by the sequential action of β- and γ-secretases on the Amyloid Precursor Protein (APP);[22] and 85% of familial EOAD cases seem to be caused by autosomal dominant mutations in the genes encoding APP and presenilins 1 and 2, which contribute to the γ-secretase complex.[23] It is generally accepted that these mutations increase the levels of pro-fibrilar/toxic Αβ peptides (Figure 1) although upregulation of inflammatory pathways could be still implicated.[24]
Figure 1. Mendelian mutations and risk polymorphisms directly linked to the amyloidogenic pathway. The amyloid precursor protein (APP) can be processed in two ways. In the non-amyloidogenic patway APP is cleaved by α-secretases at a cleavage site within the Aβ sequence (purple) producing so non-plaque-forming peptides with possibly neuroprotective effects. In the amyloidogenic pathway, APP is first cleaved by β-secretase and then by the γ-secretase complex which is formed by presinilins (PSEN) 1 and 2, nicastrin and either anterior pharynx 1 A or B. γ-Secretase first cleaves at a site close to the intracellular domain and then continues to trim the residual intramembrane fragments producing the Aβ peptide whose length can vary, although at physiological conditions Aβ40 predominates. The effect of different mendelian/fully penetrant mutations and that of a protective APP polymorphism is shown in the diagram. Effectively pathogenenic genotypes can i) increase APP expression and hence Aβ synthesis, ii) promote the amyloidogenic pathway and overall Aβ synthesis or iii) promote pathogenic Aβ profiles. Based on [25-28]. Interestingly, gene knockdown strategies employing siRNA seem to have some positive effect. Targeting presinilin 1, downregulates Aβ42 in vitro[116] and conditional inactivation in mice can actually temporarily rescue cognitive impairment.[117] On the other hand, targeting APP can produce adverse phenotypes and cognitive problems in mice.[118]
Moreover, potentially loss-of- function mutations in the SORL1 gene were lately found in 5 unexplained autosomal dominant EOAD cases although incomplete penetrance was not excluded.[29] SORL1 promotes neuronal APP recycling and actually expression of SORL1 is shown to be inversely related with Aβ synthesis.[30] Additionally, unexplained familial EOAD cases have also been found to be associated with genetic copy number variations (CNVs) such as duplications of the APP locus.[31,32] See Figure 1. Interestingly, trisomy 21, which involves the APP gene, is known to lead to AD pathological changes by middle age[35] and the only person with Down’s known to have escaped AD, had only two copies of APP.[36] Evermore, CNVs affecting other genes possibly related with AD pathology have also been recently associated with EOAD[33,34] (see Table 1) and even if extremely rare and not yet proved causal, their further investigation is important for understanding AD pathogenesis and possible therapeutic targets.
However, although some fully penetrant mutations can explain some LOAD cases, overall they still account for less than 1% of all cases.[37,38] Hence the role of common low-penetrant polymorphisms must be more significant. The gene most consistently identified with risk factor genotypes is APOE, which has three isoforms (ε2-4) and normally promotes Aβ degredation.[39] It is commonly reported that ε4 homozygocity confers a 15-fold increase in risk and up to 20 years earlier disease onset, while ε2 is protective.[41-44] However, most studies are cross-sectional and a recent longitudinal study showed that ε4 carriers despite being more likely to develop mild cognitive impairment, the transition-likelihood to AD is not different,[45] suggesting that ε4 may not actually affect life-time risk but just decrease age of onset, as is the case in familial AD.[46] See also[47,48]. Additionally, very young ε4 carriers seem to have (developmentally?) decreased entorhinal cortex volumes,[49] limiting perhaps the ‘threshold’ for manifesting the disease in susceptible individuals (see Figure 2). The second most important determinant is a locus upstream of BIN1,[50-52] which modulates BIN1 expression and BIN1-related τ-mediated neurotoxicity.[53] SORL1 polymorphisms affecting its expression are also associated with increased AD risk.[30,54-57] Many other genetic polymorphisms have been associated with increased AD risk (see [58]) although their quantitative impact and mode of contribution may need further investigations as shown by the case of APOE.Alzheimer’s and Parkinson’s diseases are two complex and very heterogeneous diseases with regards their underlying genetic aetiology, onset and pathology. The existence of rare fully penetrant mutations sufficient to cause each disease and that of common genetic polymorphisms conferring increased risk or modulating other aspects of the disease, have revealed many key molecular pathways and processes that are involved – to different extents – in the manifestation of both AD and PD. Even though, the quantitative contribution of each factor may not be clear, future approaches analyzing genetic variations within the context of molecular pathways, measurable biomarkers, epistatic and environmental interactions, genomic instability and mosaicism, may lead to a better characterization of the role that genetics play in the aetiology of PD and AD. However, the picture would be incomplete without integrating the findings from epigenetics. As summarized in Figure 4, it is probably the interplay between all these that ultimately determines the disease outcome, and evermore, the appreciation of the contribution of possibly multiple pathologic loops in AD and PD pathogenesis may inform future multiple target therapies.
Finally, a new paradigm shift has occurred with the realisation that up to 40% of human neurons have CNVs[59] while 10% are aneuploid and that actually neuronal DNA replication and increased aneuploidy states (e.g. 10-fold increase of chromosome 21-specific aneuploidy) precede neuronal death in AD.[60-62] What is more, Suberbielle et al. showed recently that neuronal activity is normally associated with double-strand breaks (DSBs) in neurons of young adult mice and that transgenic mice models expressing human APP not only had increased neuronal DSBs at baseline but Αβ further amplified and prolonged DSBs related to increases in neuronal activity. The ultimate consequence of this increase in DSBs in AD is not yet known but could contribute in cell deregulation and death. All these definitely paint a very complex and incomplete picture of AD genetics and furthermore emphasize the possibility that a two-way process, in addition to interactions between hundreds or thousands of different inherited and acquired polymorphisms within certain cell populations, determines the disease outcome.
Genetic factors in PD
Similar to AD, the central role of α–synuclein in PD was further recognized when uncommon mutations and CNVs (duplications/triplications) of the SNCA gene were found to cause autosomal dominant PD in both familial and sporadic cases, showing that altered function and increased expression are sufficient to cause PD.[63-68] Interestingly, PD with dementia is more common with SNCA duplications and early onset aggressive PD or dementia with LBs more common with triplications[65] revealing that the PD phenotypes may be actually a part of a clinicopathological continuum with not distinct boundaries. Nonetheless, the most commonly affected gene, accounting for 6% of familial and 2% of sporadic cases (depending on ethnicity) is LRRK2.[69] Its function is not well understood but it seems to interact with α-synuclein, affect LB formation[70] and misregulate autophagy,[71] explaining perhaps the great pathohistological heterogeneity of pτ aggregations or LB absence in some carriers.[69] Rare gene mutations causing autosomal recessive forms have also been found and seem to be associated with mitochondrial dysfunction and increased oxidative stress (DJ1, PINK1, parkin); or disregulation of the ubiquitin-proteosome system (parkin and UCHL1).[65] See Figure 3. Finally, there are several mutations that either cause very atypical PD or other pathologies too (e.g. ATP13A2, ATXN2, ATXN3, FBXO7)[72] raising questions about current PD classifications.
Although mendelian mutations could explain a few sporadic cases, common low-risk polymorphisms are also implicated in PD as in AD. Not surprisingly, locus variability of the SNCA promoter,[73] or SNCA itself,[74,75] which correlates with altered α-synuclein expression,[76,77] is associated with increased risk; accounting for 3% and 12% of cases (>10% α-synyclein expression translates in 40% increase in risk).
Furthermore, variability of the microtubule-associated protein tau (MAPT ) gene, could also contribute towards 8% of cases,[74] even though mendelian mutations do not cause PD but other neurodegenerative diseases. Interestingly, common HLA variations also seem to be strongly implicated (underlining the role of neuroinflammation),[79] but while variation in the HLA region several other genes have been proposed, they do not have as strong associations.[71] Finally, on another note, it has been suggested by in silico analysis that somatic transpositions, which are common in the brain, have the potential to alter biosynthesis of metabolites involved in PD, but further research is needed to characterise their role in PD pathogenesis.[80]
The role of the mother
One of the most interesting recent findings is that Αβ burden is higher in people with maternal history of AD than those with paternal or none,[81] and actually normal individuals with maternal history also exhibit more atrophic changes.[82] One possible explanation is that AD risk is associated with mitochondrial-DNA (mtDNA; always inherited maternally) and supporting this, normal individuals with maternal history have increased markers of oxidative stress[83] and certain mitochondrial haplotypes are also associated with AD biomarkers.[84] Similarly, the role of mitochondria in PD has already been stressed, and a study in a family with strong maternal inheritance profile, showed that maternal descendants had more abnormal mitochondrial morphologic features and increased reactive oxygen species as shown in vitro by cell lines expressing donor mtDNA.[85] Additionally, several studies have shown the existence of protective and risk mtDNA haplogroups[86-89] emphasizing the role of mitochondrial dysfunction and oxidative stress in PD pathogenesis. An alternative explanation could be parental epigenetic imprinting of pathogenic genes but there is no current evidence.
The Heritability Gap
Despite the abovementioned evidence, there is still a big gap from what would be expected and by regarding AD and PD as complex and largely heterogeneous diseases one can postulate that the effect of rare and structural variants and gene-gene (epistasis) or gene-environment interactions is “beyond the reach of the conventional single-variant association testing procedures”[90] which most-times consider clinical diagnosis as outcome against single variations even though it is known that histopathology and hence the contribution of different pathological pathways varies between individuals. Therefore it could be argued that future analyses should focus more on ‘network-centric approaches’ that consider gene-gene interactions, molecular pathways[90,91] and disease-biomarkers as outcomes too.
The Role of Epigenetics
The epigenome refers to all mechanisms of “mitotically or meiotically heritable changes in gene expression that do not involve alterations in the DNA sequence”[21] such as methylation, histone modification and regulatory RNA. Such changes can be inherited, erased or modified and can be cell-specific. Generally three types of epigenetic variation are recognize as the epigenotype is strictly determined or just facilitated by the genotype, or is genotype-independent/stochastic.[20] Hence, while monozygotic twins are epigenetically identical early in life, by age 50 a 30-35% concordance drift occurs with respect to methylation and histone modification.[92] Consequently, epigenetics integrate the contribution of both inherited and environmental factors and epigenetic regulation or deregulation could determine the outcome of complex diseases, especially when age is a major risk factor as in PD and AD.
With regards to AD, it has been shown that normal ageing is correlated with demethylation of the APP promoter probably underlying increased Αβ deposition in the ageing brain.[93] Evermore, very recently Iwata et al.[94] revealed aberrant CpG methylation within the APP and MAPT genes in the AD brain, which in vitro resulted in increased protein expression. In particular, they calculated that the temporal cortex hosted 2-5% of very abnormally methylated cells, which – by considering the prion-like behavior of Αβ and τ–[95] could then serve as “seed clones for aggregated protein production.”[94] Generally, previous studies have shown that AD is associated with abnormal methylation patterns[96] and reduced epigenetic markers and regulators,[97] suggesting that AD is associated with epigenetic deregulation. Interestingly, it was found that methylation levels of MTHFR, involved in methylation homeostasis, and APOE decreased in ageing controls whereas they increased in AD brains.[96] One genome-wide DNA methylation study identified about 1000 possible risk sites.[98] Furthermore, AD is associated with altered miRNA levels which could up-regulate APP and beta-secretase.[99,100]
These epigenetic differences could be explained either as the result of inheritance and environmental factors or as part of AD pathogenesis. Supporting the former, neonatal but not adult Pb-exposure of rats results in increased APP and Αβ expression in old age,[101] while folate deficiency (which affects methylation homeostasis) is associated with PSEN1 over-expression in ε3 and ε4 (but not ε2) transgenic mice, while folate-deficient ε4-mice exhibit increased Αβ synthesis as well.[102] Additionally, diabetic brains show increased expression of certain histone deacetylases (HDACs), and diabetic mice show increased HDAC-mediated susceptibility to Aβ-induced synaptic impairments, explaining diabetes as AD risk factor.[103] On the other hand, Aβ addition in endothelial cells seems to induce global DNA hypomethylation and further suppress neprilysin, which facilitates Aβ degradation.
Similarly, samples from sporadic parkinsonian brains show reduced methylation in SNCA intron 1 compared to controls, and it has been shown that such hypomethylation increases α-synuclein expression.[104] Moreover, there is some evidence that α-synuclein induces global DNA hypomethylation (involving SNCA too) by sequestering DNA methyltransferase 1 (involved in DNA methylation)[105] and furthermore was shown to have toxic effects by binding directly to histones preventing their acetylation.[106] Interestingly, methylation markers correlate with both APP and α-synuclein and increased methylation potential is associated with better cognitive status in PD patients.[107] However, at the same time α-sunuclein-mediated acetylation inhibition actually down-regulates specific apoptotic pathways conferring so a neuroprotective effect.[108] Hence potential therapeutic targets should be very specific. Nevertheless, well-characterized toxins causing parkinsonism (paraquat, rotenone, dieldrin) and even L-Dopa also modulate histone acetylation.[109, 110]
Finally, sex hormone imprinting could account for the differential susceptibility of males and females (AD is more common in women,[1] while PD in men[111]). For instance, it has been shown in mice PD models that the neonatal exposure to endogenous sex hormones determines not only adult susceptibility of neurons but also whether estrogen could have a protective or detrimental effect.[112] Such sexual dimorphisms possibly at the epigenetic level could also explain the possibility that certain genetic polymorphisms (e.g. in SORL1) confer a female-specific increased risk for AD.[113, 114]
It is evident that epigenetic imprinting and deregulation due to environmental factors cannot only increase Αβ and α-synuclein expression in susceptible cells, but the latter can further deregulate the epigenome in in AD and PD and further propagate the pathology via prion-like mechanisms in other cells both at the protein[89] and then at the epigenetic levels too.[95]
Concluding remarks
Alzheimer’s and Parkinson’s diseases are two complex and very heterogeneous diseases with regards their underlying genetic aetiology, onset and pathology. The existence of rare fully penetrant mutations sufficient to cause each disease and that of common genetic polymorphisms conferring increased risk or modulating other aspects of the disease, have revealed many key molecular pathways and processes that are involved – to different extents – in the manifestation of both AD and PD. Even though, the quantitative contribution of each factor may not be clear, future approaches analyzing genetic variations within the context of molecular pathways, measurable biomarkers, epistatic and environmental interactions, genomic instability and mosaicism, may lead to a better characterization of the role that genetics play in the aetiology of PD and AD. However, the picture would be incomplete without integrating the findings from epigenetics. As summarized in Figure 4, it is probably the interplay between all these that ultimately determines the disease outcome, and evermore, the appreciation of the contribution of possibly multiple pathologic loops in AD and PD pathogenesis may inform future multiple target therapies.
Figure 4. Interplay between genotype, epigenotype, environmental factors and pathology in complex diseases. Inherited or acquired, structural or sequence changes in a cell genotype can lead to altered gene regulation and protein expression. Similarly, gene regulation and protein expression can be further altered by global or specific epigenetic changes, which can be inherited, induced by early or late environmental factors and comorbidities or happen in a stochastic manner. Gene-gene interactions (epistasis) may play a role. All these deregulate several molecular pathways – possibly to different extends between different cells or individuals – and ultimately lead to the manifestation of pathological changes. Then these pathological changes can further alter the genotype (e.g. DND replication, failure of DNA repair, DNA breaks etc), further deregulate the cell epigenome and propagate the pathology to other cells by proteins with prion-like properties or even by inducing a cytotoxic environment (e.g. inflammation, oxidative stress). These pathological positive-feedback loops not only make it possible that a critical number of “seeding” cells with increased susceptibility can act as primary pathological foci and ultimately alter the susceptibility of neighboring cells but could also partially explain the histopathological variability observed, as different pathways may predominate in different cells or different individuals according to their prior risk factors.
References
- 1. Gao S, Hendrie HC, Hall KS, Hui S. The relationships between age, sex, and the incidence of dementia and Alzheimer disease: a meta-analysis. Arch Gen Psychiatry 1998;55:809-15.
- 2. Hy LX, Keller DM. Prevalence of AD among whites: a summary by levels of severity. Neurology 2000;55:198-204.
- 3. Tanner CM, Goldman SM. Epidemiology of Parkinson's disease. Neurol Clin 1996;14:317-35.
- 4. Ittner LM, Gotz J. Amyloid-beta and tau--a toxic pas de deux in Alzheimer's disease. Nat Rev Neurosci 2011;12:65-72.
- 5. Dong S, Duan Y, Hu Y, Zhao Z. Advances in the pathogenesis of Alzheimer's disease: a re-evaluation of amyloid cascade hypothesis. Transl Neurodegener 2012;1:18,9158-1-18.
- 6. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging 2003;24:197-211.
- 7. Burke RE, Dauer WT, Vonsattel JP. A critical evaluation of the Braak staging scheme for Parkinson's disease. Ann Neurol 2008;64:485-91.
- 8. Hauser DN, Hastings TG. Mitochondrial dysfunction and oxidative stress in Parkinson's disease and monogenic parkinsonism. Neurobiol Dis 2013;51:35-42.
- 9. Perfeito R, Cunha-Oliveira T, Rego AC. Revisiting oxidative stress and mitochondrial dysfunction in the pathogenesis of Parkinson disease--resemblance to the effect of amphetamine drugs of abuse. Free Radic Biol Med 2012;53:1791-806.
- 10. Tanzi RE. A genetic dichotomy model for the inheritance of Alzheimer's disease and common age-related disorders. J Clin Invest 1999;104:1175-9.
- 11. Lucking CB, Durr A, Bonifati V, Vaughan J, De Michele G, Gasser T, et al. Association between early-onset Parkinson's disease and mutations in the parkin gene. N Engl J Med 2000;342:1560-7.
- 12. Bergem AL, Engedal K, Kringlen E. The role of heredity in late-onset Alzheimer disease and vascular dementia. A twin study. Arch Gen Psychiatry 1997;54:264-70.
- 13. Pedersen NL, Gatz M, Berg S, Johansson B. How heritable is Alzheimer's disease late in life? Findings from Swedish twins. Ann Neurol 2004;55:180-5.
- 14. Ashford JW, Mortimer JA. Non-familial Alzheimer's disease is mainly due to genetic factors. J Alzheimers Dis 2002;4:169-77.
- 15. Tanner CM, Ottman R, Goldman SM, Ellenberg J, Chan P, Mayeux R, et al. Parkinson disease in twins: an etiologic study. JAMA 1999;281:341-6.
- 16. Wirdefeldt K, Gatz M, Schalling M, Pedersen NL. No evidence for heritability of Parkinson disease in Swedish twins. Neurology 2004;63:305-11.
- 17. Wirdefeldt K, Gatz M, Reynolds CA, Prescott CA, Pedersen NL. Heritability of Parkinson disease in Swedish twins: a longitudinal study. Neurobiol Aging 2011;32:1923.e1,1923.e8.
- 18. Simon DK, Lin MT, Pascual-Leone A. "Nature versus nurture" and incompletely penetrant mutations. J Neurol Neurosurg Psychiatry 2002;72:686-9.
- 19. Petronis A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature 2010;465:721-7.
- 20. Richards EJ. Inherited epigenetic variation--revisiting soft inheritance. Nat Rev Genet 2006;7:395-401.
- 21. Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet 2007;8:253-62.
- 22. Thinakaran G, Koo EH. Amyloid precursor protein trafficking, processing, and function. J Biol Chem 2008;283:29615-9.
- 23. Raux G, Guyant-Marechal L, Martin C, Bou J, Penet C, Brice A, et al. Molecular diagnosis of autosomal dominant early onset Alzheimer's disease: an update. J Med Genet 2005;42:793-5.
- 24. Beglopoulos V, Sun X, Saura CA, Lemere CA, Kim RD, Shen J. Reduced beta-amyloid production and increased inflammatory responses in presenilin conditional knock-out mice. J Biol Chem 2004;279:46907-14.
- 25. Kojro E, Fahrenholz F. The non-amyloidogenic pathway: structure and function of alpha-secretases. Subcell Biochem 2005;38:105-27.
- 26. Chavez-Gutierrez L, Bammens L, Benilova I, Vandersteen A, Benurwar M, Borgers M, et al. The mechanism of gamma-Secretase dysfunction in familial Alzheimer disease. EMBO J 2012;31:2261-74.
- 27. Dimitrov M, Alattia JR, Lemmin T, Lehal R, Fligier A, Houacine J, et al. Alzheimer's disease mutations in APP but not gamma-secretase modulators affect epsilon-cleavage-dependent AICD production. Nat Commun 2013;4:2246.
- 28. Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, et al. A mutation in APP protects against Alzheimer's disease and age-related cognitive decline. Nature 2012;488:96-9.
- 29. Pottier C, Hannequin D, Coutant S, Rovelet-Lecrux A, Wallon D, Rousseau S, et al. High frequency of potentially pathogenic SORL1 mutations in autosomal dominant early-onset Alzheimer disease. Mol Psychiatry 2012;17:875-9.
- 30. Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T, Zou F, et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet 2007;39:168-77.
- 31. Rovelet-Lecrux A, Hannequin D, Raux G, Le Meur N, Laquerriere A, Vital A, et al. APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat Genet 2006;38:24-6.
- 32. McNaughton D, Knight W, Guerreiro R, Ryan N, Lowe J, Poulter M, et al. Duplication of amyloid precursor protein (APP), but not prion protein (PRNP) gene is a significant cause of early onset dementia in a large UK series. Neurobiol Aging 2012;33:426.e13,426.e21.
- 33. Hooli BV, Kovacs-Vajna ZM, Mullin K, Blumenthal MA, Mattheisen M, Zhang C, et al. Rare autosomal copy number variations in early-onset familial Alzheimer's disease. Mol Psychiatry 2013.
- 34. Rovelet-Lecrux A, Legallic S, Wallon D, Flaman JM, Martinaud O, Bombois S, et al. A genome-wide study reveals rare CNVs exclusive to extreme phenotypes of Alzheimer disease. Eur J Hum Genet 2012;20:613-7.
- 35. Mann DM. The pathological association between Down syndrome and Alzheimer disease. Mech Ageing Dev 1988;43:99-136.
- 36. Prasher VP, Farrer MJ, Kessling AM, Fisher EM, West RJ, Barber PC, et al. Molecular mapping of Alzheimer-type dementia in Down's syndrome. Ann Neurol 1998;43:380-3.
- 37. Campion D, Dumanchin C, Hannequin D, Dubois B, Belliard S, Puel M, et al. Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet 1999;65:664-70.
- 38. Cruchaga C, Haller G, Chakraverty S, Mayo K, Vallania FL, Mitra RD, et al. Rare variants in APP, PSEN1 and PSEN2 increase risk for AD in late-onset Alzheimer's disease families. PLoS One 2012;7:e31039.
- 39. Jiang Q, Lee CY, Mandrekar S, Wilkinson B, Cramer P, Zelcer N, et al. ApoE promotes the proteolytic degradation of Abeta. Neuron 2008;58:681-93.
- 40. Holtzman DM, Bales KR, Tenkova T, Fagan AM, Parsadanian M, Sartorius LJ, et al. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2000;97:2892-7.
- 41. Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA 1997;278:1349-56.
- 42. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 1993;261:921-3.
- 43. Kuusisto J, Koivisto K, Kervinen K, Mykkanen L, Helkala EL, Vanhanen M, et al. Association of apolipoprotein E phenotypes with late onset Alzheimer's disease: population based study. BMJ 1994;309:636-8.
- 44. West HL, Rebeck GW, Hyman BT. Frequency of the apolipoprotein E epsilon 2 allele is diminished in sporadic Alzheimer disease. Neurosci Lett 1994;175:46-8.
- 45. Brainerd CJ, Reyna VF, Petersen RC, Smith GE, Kenney AE, Gross CJ, et al. The apolipoprotein E genotype predicts longitudinal transitions to mild cognitive impairment but not to Alzheimer's dementia: findings from a nationally representative study. Neuropsychology 2013;27:86-94.
- 46. Pastor P, Roe CM, Villegas A, Bedoya G, Chakraverty S, Garcia G, et al. Apolipoprotein Eepsilon4 modifies Alzheimer's disease onset in an E280A PS1 kindred. Ann Neurol 2003;54:163-9.
- 47. Meyer MR, Tschanz JT, Norton MC, Welsh-Bohmer KA, Steffens DC, Wyse BW, et al. APOE genotype predicts when--not whether--one is predisposed to develop Alzheimer disease. Nat Genet 1998;19:321-2.
- 48. Breitner JC, Wyse BW, Anthony JC, Welsh-Bohmer KA, Steffens DC, Norton MC, et al. APOE-epsilon4 count predicts age when prevalence of AD increases, then declines: the Cache County Study. Neurology 1999;53:321-3.
- 49. Shaw P, Lerch JP, Pruessner JC, Taylor KN, Rose AB, Greenstein D, et al. Cortical morphology in children and adolescents with different apolipoprotein E gene polymorphisms: an observational study. Lancet Neurol 2007;6:494-500.
- 50. Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease. Nat Genet 2009;41:1088-93.
- 51. Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease. Nat Genet 2009;41:1094-9.
- 52. Seshadri S, Fitzpatrick AL, Ikram MA, DeStefano AL, Gudnason V, Boada M, et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA 2010;303:1832-40.
- 53. Chapuis J, Hansmannel F, Gistelinck M, Mounier A, Van Cauwenberghe C, Kolen KV, et al. Increased expression of BIN1 mediates Alzheimer genetic risk by modulating tau pathology. Mol Psychiatry 2013;18:1225-34.
- 54. Reitz C, Cheng R, Rogaeva E, Lee JH, Tokuhiro S, Zou F, et al. Meta-analysis of the association between variants in SORL1 and Alzheimer disease. Arch Neurol 2011;68:99-106.
- 55. Miyashita A, Koike A, Jun G, Wang LS, Takahashi S, Matsubara E, et al. SORL1 is genetically associated with late-onset Alzheimer's disease in Japanese, Koreans and Caucasians. PLoS One 2013;8:e58618.
- 56. Bettens K, Brouwers N, Engelborghs S, De Deyn PP, Van Broeckhoven C, Sleegers K. SORL1 is genetically associated with increased risk for late-onset Alzheimer disease in the Belgian population. Hum Mutat 2008;29:769-70.
- 57. Wen Y, Miyashita A, Kitamura N, Tsukie T, Saito Y, Hatsuta H, et al. SORL1 is genetically associated with neuropathologically characterized late-onset Alzheimer's disease. J Alzheimers Dis 2013;35:387-94.
- 58. ALZGENE TOP RESULTS [homepage on the Internet]. 2011 ;cited 2013 . Available from: http://www.alzgene.org/TopResults.asp.
- 59. McConnell MJ, Lindberg MR, Brennand KJ, Piper JC, Voet T, Cowing-Zitron C, et al. Mosaic copy number variation in human neurons. Science 2013;342:632-7.
- 60. Yurov YB, Vorsanova SG, Iourov IY. The DNA replication stress hypothesis of Alzheimer's disease. ScientificWorldJournal 2011;11:2602-1.
- 61. Iourov IY, Vorsanova SG, Liehr T, Yurov YB. Aneuploidy in the normal, Alzheimer's disease and ataxia-telangiectasia brain: differential expression and pathological meaning. Neurobiol Dis 2009;34:212-20.
- 62. Yang Y, Geldmacher DS, Herrup K. DNA replication precedes neuronal cell death in Alzheimer's disease. J Neurosci 2001;21:2661-8.
- 63. Polymeropoulos MH, Higgins JJ, Golbe LI, Johnson WG, Ide SE, Di Iorio G, et al. Mapping of a gene for Parkinson's disease to chromosome 4q21-q23. Science 1996;274:1197-9.
- 64. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science 1997;276:2045-7.
- 65. Nuytemans K, Meeus B, Crosiers D, Brouwers N, Goossens D, Engelborghs S, et al. Relative contribution of simple mutations vs. copy number variations in five Parkinson disease genes in the Belgian population. Hum Mutat 2009;30:1054-61.
- 66. Athanassiadou A, Voutsinas G, Psiouri L, Leroy E, Polymeropoulos MH, Ilias A, et al. Genetic analysis of families with Parkinson disease that carry the Ala53Thr mutation in the gene encoding alpha-synuclein. Am J Hum Genet 1999;65:555-8.
- 67. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. alpha-Synuclein locus triplication causes Parkinson's disease. Science 2003;302:841.
- 68. Zarranz JJ, Alegre J, Gomez-Esteban JC, Lezcano E, Ros R, Ampuero I, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004;55:164-73.
- 69. Brice A. How much does dardarin contribute to Parkinson's disease? Lancet 2005;365:363-4.
- 70. Guerreiro PS, Huang Y, Gysbers A, Cheng D, Gai WP, Outeiro TF, et al. LRRK2 interactions with alpha-synuclein in Parkinson's disease brains and in cell models. J Mol Med (Berl) 2013;91:513-22.
- 71. Ray S, Liu M. Current understanding of LRRK2 in Parkinson's disease: biochemical and structural features and inhibitor design. Future Med Chem 2012;4:1701-13.
- 72. Houlden H, Singleton AB. The genetics and neuropathology of Parkinson's disease. Acta Neuropathol 2012;124:325-38.
- 73. Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioannidis JP, Kruger R, et al. Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease. JAMA 2006;296:661-70.
- 74. Edwards TL, Scott WK, Almonte C, Burt A, Powell EH, Beecham GW, et al. Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann Hum Genet 2010;74:97-109.
- 75. Simon-Sanchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, et al. Genome-wide association study reveals genetic risk underlying Parkinson's disease. Nat Genet 2009;41:1308-12.
- 76. Fuchs J, Tichopad A, Golub Y, Munz M, Schweitzer KJ, Wolf B, et al. Genetic variability in the SNCA gene influences alpha-synuclein levels in the blood and brain. FASEB J 2008;22:1327-34.
- 77. Mata IF, Shi M, Agarwal P, Chung KA, Edwards KL, Factor SA, et al. SNCA variant associated with Parkinson disease and plasma alpha-synuclein level. Arch Neurol 2010;67:1350-6.
- 78. Farrer MJ. Genetics of Parkinson disease: paradigm shifts and future prospects. Nat Rev Genet 2006;7:306-18.
- 79. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson's disease. Nat Genet 2010;42:781-5.
- 80. Abrusan G. Somatic transposition in the brain has the potential to influence the biosynthesis of metabolites involved in Parkinson's disease and schizophrenia. Biol Direct 2012;7:41,6150-7-41.
- 81. Honea RA, Vidoni ED, Swerdlow RH, Burns JM, Alzheimer's Disease Neuroimaging Initiative. Maternal family history is associated with Alzheimer's disease biomarkers. J Alzheimers Dis 2012;31:659-68.
- 82. Honea RA, Swerdlow RH, Vidoni ED, Burns JM. Progressive regional atrophy in normal adults with a maternal history of Alzheimer disease. Neurology 2011;76:822-9.
- 83. Mosconi L, Glodzik L, Mistur R, McHugh P, Rich KE, Javier E, et al. Oxidative stress and amyloid-beta pathology in normal individuals with a maternal history of Alzheimer's. Biol Psychiatry 2010;68:913-21.
- 84. Ridge PG, Koop A, Maxwell TJ, Bailey MH, Swerdlow RH, Kauwe JS, et al. Mitochondrial haplotypes associated with biomarkers for Alzheimer's disease. PLoS One 2013;8:e74158.
- 85. Swerdlow RH, Parks JK, Davis JN,2nd, Cassarino DS, Trimmer PA, Currie LJ, et al. Matrilineal inheritance of complex I dysfunction in a multigenerational Parkinson's disease family. Ann Neurol 1998;44:873-81.
- 86. Huerta C, Castro MG, Coto E, Blazquez M, Ribacoba R, Guisasola LM, et al. Mitochondrial DNA polymorphisms and risk of Parkinson's disease in Spanish population. J Neurol Sci 2005;236:49-54.
- 87. Autere J, Moilanen JS, Finnila S, Soininen H, Mannermaa A, Hartikainen P, et al. Mitochondrial DNA polymorphisms as risk factors for Parkinson's disease and Parkinson's disease dementia. Hum Genet 2004;115:29-35.
- 88. Ghezzi D, Marelli C, Achilli A, Goldwurm S, Pezzoli G, Barone P, et al. Mitochondrial DNA haplogroup K is associated with a lower risk of Parkinson's disease in Italians. Eur J Hum Genet 2005;13:748-52.
- 89. Pyle A, Foltynie T, Tiangyou W, Lambert C, Keers SM, Allcock LM, et al. Mitochondrial DNA haplogroup cluster UKJT reduces the risk of PD. Ann Neurol 2005;57:564-7.
- 90. Okser S, Pahikkala T, Aittokallio T. Genetic variants and their interactions in disease risk prediction - machine learning and network perspectives. BioData Min 2013;6:5,0381-6-5.
- 91. Srinivasan BS, Doostzadeh J, Absalan F, Mohandessi S, Jalili R, Bigdeli S, et al. Whole genome survey of coding SNPs reveals a reproducible pathway determinant of Parkinson disease. Hum Mutat 2009;30:228-3.
- 92. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A 2005;102:10604-9.
- 93. Tohgi H, Utsugisawa K, Nagane Y, Yoshimura M, Genda Y, Ukitsu M. Reduction with age in methylcytosine in the promoter region -224 approximately -101 of the amyloid precursor protein gene in autopsy human cortex. Brain Res Mol Brain Res 1999;70:288-92.
- 94. Iwata A, Nagata K, Hatsuta H, Takuma H, Bundo M, Iwamoto K, et al. Altered CpG methylation in sporadic Alzheimer's disease is associated with APP and MAPT dysregulation. Hum Mol Genet 2013.
- 95. Fernandez-Borges N, Erana H, Elezgarai SR, Harrathi C, Gayosso M, Castilla J. Infectivity versus Seeding in Neurodegenerative Diseases Sharing a Prion-Like Mechanism. Int J Cell Biol 2013;2013:583498.
- 96. Wang SC, Oelze B, Schumacher A. Age-specific epigenetic drift in late-onset Alzheimer's disease. PLoS One 2008;3:e2698.
- 97. Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD, Rogers J. Epigenetic changes in Alzheimer's disease: decrements in DNA methylation. Neurobiol Aging 2010;31:2025-37.
- 98. Bakulski KM, Dolinoy DC, Sartor MA, Paulson HL, Konen JR, Lieberman AP, et al. Genome-wide DNA methylation differences between late-onset Alzheimer's disease and cognitively normal controls in human frontal cortex. J Alzheimers Dis 2012;29:571-88.
- 99. Hebert SS, Horre K, Nicolai L, Bergmans B, Papadopoulou AS, Delacourte A, et al. MicroRNA regulation of Alzheimer's Amyloid precursor protein expression. Neurobiol Dis 2009;33:422-8.
- 100. Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, et al. Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward regulation of beta-secretase. Nat Med 2008;14:723-30.
- 101. Basha MR, Wei W, Bakheet SA, Benitez N, Siddiqi HK, Ge YW, et al. The fetal basis of amyloidogenesis: exposure to lead and latent overexpression of amyloid precursor protein and beta-amyloid in the aging brain. J Neurosci 2005;25:823-9.
- 102. Chan A, Tchantchou F, Rogers EJ, Shea TB. Dietary deficiency increases presenilin expression, gamma-secretase activity, and Abeta levels: potentiation by ApoE genotype and alleviation by S-adenosyl methionine. J Neurochem 2009;110:831-6.
- 103. Wang J, Gong B, Zhao W, Tang C, Varghese M, Nguyen T, et al. Epigenetic mechanisms linking diabetes and synaptic impairments. Diabetes 2013.
- 104. Jowaed A, Schmitt I, Kaut O, Wullner U. Methylation regulates alpha-synuclein expression and is decreased in Parkinson's disease patients' brains. J Neurosci 2010;30:6355-9.
- 105. Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci U S A 2009;106:13010-5.
- 106. Kontopoulos E, Parvin JD, Feany MB. Alpha-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity. Hum Mol Genet 2006;15:3012-23.
- 107. Obeid R, Schadt A, Dillmann U, Kostopoulos P, Fassbender K, Herrmann W. Methylation status and neurodegenerative markers in Parkinson disease. Clin Chem 2009;55:1852-60.
- 108. Jin H, Kanthasamy A, Ghosh A, Yang Y, Anantharam V, Kanthasamy AG. alpha-Synuclein negatively regulates protein kinase Cdelta expression to suppress apoptosis in dopaminergic neurons by reducing p300 histone acetyltransferase activity. J Neurosci 2011;31:2035-51.
- 109. Nicholas AP, Lubin FD, Hallett PJ, Vattem P, Ravenscroft P, Bezard E, et al. Striatal histone modifications in models of levodopa-induced dyskinesia. J Neurochem 2008;106:486-94.
- 110. Song C, Kanthasamy A, Anantharam V, Sun F, Kanthasamy AG. Environmental neurotoxic pesticide increases histone acetylation to promote apoptosis in dopaminergic neuronal cells: relevance to epigenetic mechanisms of neurodegeneration. Mol Pharmacol 2010;77:621-32.
- 111. Wooten GF, Currie LJ, Bovbjerg VE, Lee JK, Patrie J. Are men at greater risk for Parkinson's disease than women? J Neurol Neurosurg Psychiatry 2004;75:637-9.
- 112. Murray HE, Pillai AV, McArthur SR, Razvi N, Datla KP, Dexter DT, et al. Dose- and sex-dependent effects of the neurotoxin 6-hydroxydopamine on the nigrostriatal dopaminergic pathway of adult rats: differential actions of estrogen in males and females. Neuroscience 2003;116:213-22.
- 113. Cellini E, Tedde A, Bagnoli S, Pradella S, Piacentini S, Sorbi S, et al. Implication of sex and SORL1 variants in italian patients with Alzheimer disease. Arch Neurol 2009;66:1260-6.
- 114. Olgiati P, Politis A, Albani D, Rodilossi S, Polito L, Zisaki A, et al. Effects of SORL1 gene on Alzheimer's disease. Focus on gender, neuropsychiatric symptoms and pro-inflammatory cytokines. Curr Alzheimer Res 2013;10:154-6.
- 115. Gerrish A, Russo G, Richards A, Moskvina V, Ivanov D, Harold D, et al. The role of variation at AbetaPP, PSEN1, PSEN2, and MAPT in late onset Alzheimer's disease. J Alzheimers Dis 2012;28:377-8.
- 116. Kandimalla RJ, Wani WY, Binukumar BK, Gill KD. siRNA against presenilin 1 (PS1) down regulates amyloid β42 production in IMR-32 cells. J Biomed Sci. 2012 Jan 3;19:2. doi: 10.1186/1423-0127-19-2.
- 117. Saura CA, Chen G, Malkani S, Choi SY, Takahashi RH, Zhang D, Gouras GK, Kirkwood A, Morris RG, Shen J. Conditional inactivation of presenilin 1 prevents amyloid accumulation and temporarily rescues contextual and spatial working memory impairments in amyloid precursor protein transgenic mice. J Neurosci. 2005 Jul 20;25(29):6755-64.
- 118. Senechal Y, Kelly PH, Cryan JF, Natt F, Dev KK. Amyloid precursor protein knockdown by siRNA impairs spontaneous alternation in adult mice. J Neurochem. 2007 Sep;102(6):1928-40. Epub 2007 Jun 1.
No comments