Open access

Cerebrospinal Fluid Based Diagnosis in Alzheimer’s Disease

Written By

Inga Zerr, Lisa Kaerst, Joanna Gawinecka and Daniela Varges

Submitted: 31 May 2011 Published: 06 September 2011

DOI: 10.5772/16684

Chapter metrics overview

2,174 Chapter Downloads

View Full Metrics

1. Introduction

The Alzheimer’s disease (AD) is the most frequent form of dementia worldwide. The major neuropathological hallmarks of the disease are loss of neurons and synapse, senile plaques (extracellular aggregates primarily composed of ß-amyloid; Aß) and neurofibrillary tangles (aggregates of hyperphosphorylated forms of the microtubule-associated tau protein) throughout cortical and limbic regions of the brain. The definite diagnosis still requires histopathological conformation according to the criteria, however, in recent years substantial progress has been made in the area of early biomarker development. The use of cerebrospinal fluid as a testing platform is very promising because the CSF protein composition can reflect the pathological processes of the brain and because it is easily accessible by a lumbar puncture. Some proteins and peptides such as ß-amyloid 1-42 (Aβ1-42), ß-amyloid 1-40 (Aβ1-40), total tau (tau) and hyperphosphorylated tau (p-tau) have been reported to meet the criteria for a biomarker. Another series of publications reported transthyretin, isoprostane, BACE1 activity and other proteins and enzymes as a potential biomarkers in AD. Whereas the biomarkers mentioned first have been studied extensively and were suggested to be included into clinical AD criteria, less information is available on the others. This review will focus on the importance of CSF based biomarkers in AD, covers the data available from the literature and highlights their role in the differential diagnosis of dementia.

Advertisement

2. Why to use CSF as a testing platform in dementia? ─ Neuroanatomy driven approach

Cerebrospinal fluid (CSF) is the main component of the brain extracellular space and participates in the exchange of many biochemical products in the central nervous system (CNS). Consequently, CSF contains a dynamic and complex mixture of proteins, which reflects physiological or pathological state of CNS. Changes in CSF proteome have been described in various neurodegenerative disorders. These alterations are discussed to reflect pathological changes in the brain and thus contribute to a better understanding of the pathophysiology of the underlying disorder (Gawinecka et al. 2010).

CSF analysis is extremely important to identify autoimmune disorders and inflammatory conditions, which might lead to dementia. Although changes are non-specific, like pleocytosis, elevated protein content, increased albumin ratio and oligoclonal IgG synthesis within the central nervous system, their presence clearly differentiate inflammatory and autoimmune disease from neurodegenerative dementia.

Advertisement

3. How to select a biomarker ─ a concept-of-pathogenesis-driven approach

Several potential biomarkers in the CSF and blood have been already suggested. Some of them like Aβ1-42 and tau (and its phosphorylated form) became important biomarker in dementia diagnosis. The advantage of these biomarkers is their clear link to the pathological process and abnormalities, which are detected in the brain of AD patients (Aß and amyloid hypothesis as well as tau pathology).

3.1. Amyloid hypothesis

The amyloid core of senile or neuritic plaques contains an amyloid-like substance formed by peptides, which originate from proteolytic cleavage of the membrane-associated precursor protein (amyloid precursor protein, APP). They are generated by a sequential cleavage of amyloid precursor protein (APP) by β- and γ-secretase. Aβs are small hydrophobic peptides existing mainly in two lengths: Aβ1-40 and Aβ1-42. It was initially assumed that the production of Aβs occurs only under pathological conditions. Later, Aβ was shown to be constitutively released from APP and secreted to blood and CSF. In AD, Aβ peptides are involved in pathological processes and accumulate in the brain as amyloid or senile plaques. There is clear correlation between Aβ levels in CSF and plaque depositions in the brain coupled with the concept of casual involvement of APP and Aβs in the pathogenesis of AD.

The concentration of Aβs is thought to reflect disease-associated changes and is widely applied as a diagnostic biomarker of AD (Gawinecka et al. 2010).

Table 1 gives an overview of publications related to this topic from past 5 years. The vast majority of publications is related to the detection of abnormal levels of Aß1-42 in AD as compared to other dementia, however, some data are also available for Aß1-40. Recently, a ratio between both peptides has been suggested as a potential biomarker in AD (Table 2). Aß1-42 level is decreased in patients with AD, but might also decrease in other dementia, too. Test sensitivity for Aß1-42 alone is given from 60 to 96%, depending on the design of the study.

In MCI, Aß42 level is lower in patients with a subsequent AD diagnosis (De Meyer et al. 2010; Diniz et al. 2008; Mattsson et al. 2009; Stefani et al. 2006), which leads to the conclusion that this parameter might also serve as an preclinical (potential predictive?) biomarker (Stefani et al. 2006) for cognitive decline.

Aß42 level is decreased in other conditions, including prion diseases, Parkinson’s disease (Siderowf et al. 2010) and DLB. In PD, decreased levels correlate well with cognitive decline, in contrast to tau/p-tau ratio (see below) (Siderowf et al. 2010). According to some studies which used patients with non-AD dementia as controls, this marker is highly sensitive for detection of dementia, but it seems that it does not allow to discriminate between various dementia types because of limited specificity (Formichi et al. 2006; Gloeckner et al. 2008).

One approach to improve test sensitivity and specificity was to calculate an Aß42/40 ratio, which is significantly decreased in AD patients. It seems also to discriminate between different dementia including AD and non-AD (vascular, mixed, FTD, alcohol toxic and controls) (Lewczuk et al. 2004). However, the significance of this finding has still to be proven on higher numbers of patients in a prospective study.

Patients (n) Sensitivity (%) Specificity (%) Reference
Aß1-42 MCI -"/ AD (422)
controls (429)
68 93 (Diniz et al. 2008)
Aß1-42 PD (109)
AD (20)
controls (36)
n.a. (Alves et al. 2010)
Aß1-42 AD (131)
controls (72)
92 89 (Sunderland et al. 2003)
Aß1-42 PD (45) n.a. (Siderowf et al. 2010)
Aß1-42 MCI n.a. (Okonkwo et al. 2011)
Aß1-42 AD (33)
ARCD* (20)
controls (50)
70-84 80-85 (Kapaki et al. 2005)
Aß1-42 MCI (750)
AD (529)
controls (304)
79 65 (Mattsson et al. 2009)
Aß1-42 autoptic AD (68)
MCI (57)
94 n.a. (De Meyer et al. 2010)
Aß1-42 AD "/85 "/85 (Slats et al. 2010)
Aß1-42 mild AD (100)
MCI (196)
controls (114)
96 77 (Shaw et al. 2009)
Aß1-40 AD (82)
DLB (44)
controls (71)
AD vs controls
97
AD vs controls
83
(Mollenhauer et al. 2011)
Aß1-40 DLB (21)
AD (23)
PDD (21)
81 71 (Bibl et al. 2006a)
Aß1-40


Aß1-42
AD (23)
NPH (13)
DLB (23)
CJD (18)
DLB (23)
FTD (10)
controls (19)
61 78 (Gloeckner et al. 2008)

Table 1.

*ARCD = alcohol related cognitive disorder

Aß40 and Aß42 in dementia diagnosis

Diagnosis (n) Sensitivity (%) Specificity (%) Reference
Aß1-42/ Aß1-40 MCI (65) 86 60 (Brys et al. 2009)
Aß1-42/ Aß1-40 AD (22) 95 88 (Lewczuk et al. 2004)
Aß1-42/ Aß1-40 AD (157) 59 88 (Shoji and Kanai 2001)
Aß1-42/ Aß1-40 AD (69) (Spies et al. 2010)
Aß1-42/ Aß1-40 AD (18) AD vs control: 100
AD vs DLB: 100
AD vs both groups: 100
AD vs control: 93
AD vs DLB: 68
AD vs both groups: 77
(Bibl et al. 2006b)
Aß1-42/ Aß1-40 AD (109) AD vs control: 79
AD vs all: 70
AD vs control: 71
AD vs all: 71
(Brettschneider et al. 2006)

Table 2.

Aß1-42/ Aß1-40 ratio as potential biomarkers in AD

3.2. Tau hypothesis

Intracellular neurofibrillary tangles (NFT), which are neuronal inclusions consisting of abnormal cytoskeletal elements of hyperphosphorylated tau protein are another characteristic pathological feature of AD. These tangles are found throughout the neocortex, in the nucleus basalis Meynert, thalamus, and in the mammillary bodies. Tau protein is a microtubule-associated protein (MAP), which interacts with tubulin and promotes microtubule assembly and stability; it is also involved in neurogenesis, axonal maintenance and axonal transport. There are six different tau isoforms present in the human adult brain, which are generated by an alternative mRNA splicing from a single gene (Goedert et al. 1989). Tau is a phosphoprotein, with 79 putative serine or threonine phosphorylation sites on the longest tau isoform. The hyperphosphorylated tau has a reduced affinity for microtubules and reduced ability to promote their assembly (Lindwall and Cole 1984). In AD, tau detaches from microtubules and aggregates in paired helical filaments (PHFs). Tau isolated from these aggregates is found to be about 4 times more phoshorylated than tau isolated from nondemented individuals (Alonso et al. 2001; Kopke et al. 1993, Gawinecka and Zerr 2010).

The elevated CSF level of nonphosphorylated and phosphorylated tau is one of AD hallmarks (Andreasen et al. 1999; Arai et al. 1997; Galasko et al. 1997; Ishiguro et al. 1999; Itoh et al. 2001; Mecocci et al. 1998). Since the first description of this abnormality and availability of an ELISA test, extensive research has been conducted. Data on tau level in AD and other dementia are given in Table 3. Again, elevated levels can be observed in other conditions than AD too, the test sensitivity and specificity seems to be above 80% in majority of the cases (Table 3). However, it has to be kept in mind that tau levels increase in CSF with age and this physiological finding hast to be kept in mind when cut-off level are established (Figure 1). In pathological conditions, total tau levels in MCI patients indicate increased AD risk (Hertze et al. 2010; Mattsson et al. 2009; Pauwels et al. 2009) and increased level correlates well with disease severity (Buchhave et al. 2009; Stefani et al. 2006). Some studies even demonstrated that extremely high tau levels might be indicator of poor prognosis (Snider et al. 2009).

Patients (n) Sensitivity (%) Specificity (%) Reference
MCI (166) 78 83 (Hertze et al. 2010)
AD (131) 92 89 (Sunderland et al. 2003)
AD, CJD, LBD, FTD, VD 73-91 74-98 (van Harten et al. 2011)
AD (33)
ARCD* (20)
88-94 95-96 (Kapaki et al. 2005)
AD
NPH
91-93 78-96 (Kapaki et al. 2007)
MCI (750)
AD (529)
86 56 (Mattsson et al. 2009)
AD, other dementia, psychiatric (219) 88 80 (Ibach et al. 2006)
early AD (269)
mild AD (468)
late AD (495)
n.a. (Stefani et al. 2006)
MCI -"/ AD 82 87 (Pauwels et al. 2009)
mild AD (100)
MCI (192)
autoptic AD (56)
controls (114)
70 92 (Shaw et al. 2009)
MCI -"/ AD (422)
controls (420)
68 93 (Diniz et al. 2008)
DLB (34)
AD (31)
other dementia (4)
85 95 (Kasuga et al. 2010)

Table 3.

*ARCD = alcohol related cognitive disorder

Total tau level in cerebrospinal fluid in AD and other dementia

Regarding phosphorylated tau level in CSF, a recent metaanalysis on 51 publications from the area revealed that p-tau contributed to the separation of MCI from healthy individuals with a sensitivity of 80% and specificity of 84% (Mitchell 2009). CSF p-tau is a good diagnostic biomarker of AD too, with test sensitivity mostly >80% (see Table 4). MCI patients with low Aß1-42 and high p-tau levels are at a clear AD risk (Hertze et al. 2010). AD patients with higher p-tau level have greater hippocampal atrophy, poorer neuropsychological test results and it is also indicator of disease progression (Henneman et al. 2009). Whereas p-tau levels indicate AD or development of AD with good accuracy, unfortunately, this biomarker is also less adequate in separating AD from other dementias (Mitchell 2009).

Due to these considerations, several studies tried to analyse the diagnostic potential of p-tau/total tau ratio. Some of them reported test sensitivity between 88-96% with a specificity of 60-100%. While its ratio is promising, again, it has to be analysed in a prospective setting since the numbers of analysed patients so far are too low for any definite conclusions (Buerger et al. 2006; Hu et al. 2002; Kapaki et al. 2007).

Figure 1.

Total tau level in cerebrospinal fluid stratified by age in healthy controls (Gloeckner et al. 2008)

Patients (n) Sensitivity (%) Specificity (%) Reference
p-tau AD
neurological controls (46)
"/85 79-91 (Scheurich et al. 2010)
p-tau AD (94)
MCI (166)
depression (29)
controls (38)
"/85 79-91 (Hertze et al. 2010)
p-tau AD (49) 46 94 (Snider et al. 2009)
p-tau AD (251)
controls (122)
62 93 (Formichi et al. 2006)
p-tau AD, CJD, LBD, FTD, vascular dementia 79-88 78-83 (van Harten et al. 2011)
p-tau AD, NPH, controls 89 87 (Kapaki et al. 2007)
p-tau AD, other dementia, psychiatric, controls 89 87 (Ibach et al. 2006)
p-tau AD (31)
MCI (25)
72-81 78-88 (Henneman et al. 2009)
p-tau autoptic AD (68)
MCI (57)
AD 94
MCI 100
(De Meyer et al. 2010)
p-tau mild AD (100)
MCI (196)
autoptic AD (56)
controls (114)
68 73 (Shaw et al. 2009)
p-tau MCI (750)
AD (529)
Controls (304)
84 47 (Mattsson et al. 2009)
p-tau/total tau AD (52)
controls (56)
non AD (37)
vascular dementia (46)
96 94 (Hu et al. 2002)
p-tau/total tau AD (67)
NPH (18)
controls (72)
88-93 60-100 (Kapaki et al. 2007)
p-tau/total tau AD (37)
CJD (21)
controls (10)
91 97 (Buerger et al. 2006)
p-tau/total tau AD (71)
FTD (18)
CJD (20)
controls (43)
ratio AD: 1,27 (mean)
ratio FTD: 1,13 (mean)
ratio CJD: 0,05 (mean)
ratio controls: 1,7 (mean)
(Riemenschneider et al. 2003)
p-tau/total tau CJD (21)
AD (49)
neurol. controls (164)
86 90 (Bahl et al. 2009)

Table 4.

Phosphorylated tau level in cerebrospinal fluid in AD and other dementia

In general, many studies on Aß1-42, total tau level and its phosphorylated isoform have been performed. In most studies, the number of patients per group is limited and various criteria and diagnostic techniques have been applied. Of importance, a recent multicenter study demonstrated once again that CSF Aß1-42, total tau and p-tau identify incipient AD with good sensitivity and specificity, however, the data are less reliable than reported from single-center studies (Mattsson et al. 2009). Thus, improvements are necessary, with respect to standardisation protocols between centers, but also with respect to identification of further disease- specific biomarkers in biological fluids.

3.3. The role of ApoE

The presence of the apolipoprotein E allele is a well documented risk factor for AD (Lamb et al. 1998; Saunders et al. 1993) and is associated with a decreased age of clinical onset, with a higher stage of ß-amyloid deposition and neurofibrillary change formation, severe disease course, higher brain atrophy and a more rapid disease course (Ohm et al. 1999). The ApoE polymorphism includes three common alleles (ε2, ε3, ε4) at a single gene locus resulting in six ApoE genotypes ε2/ε2, ε3/ε3, ε4/ε4, ε2/ε3, ε2/ε4, ε3/ε4. The ApoE ε4 allele is an established risk factor for AD (Saunders et al. 1993) and there is a large body of evidence for a role of ApoE in the pathogenesis of AD (Davidson et al. 2006; Varges et al. 2011).

CSF marker Patients (n)
[diagnosis confirmed]
Influence of ApoE ε4 allele Reference
Aß1-42 82 dose-dependent reduction (Galasko et al. 1998)
tau heterozygous: elevation homozygous: reduction
Aß1-42 50 dose-dependent reduction (Riemenschneider et al. 2000)
Aß1-42 84 dose-dependent reduction (Hulstaert et al. 1999)
Aß1-42 73 more reduced levels in ε4 carriers (Smach et al. 2008)
tau no influence
Aß1-42 60 no influence (Ewers et al. 2008)
Aß1-42 50 no influence (Engelborghs et al. 2007)
tau no influence
tau 19 dose-dependent elevation (Golombowski et al. 1997)
Aß1-42 121 [41 NP] more reduced levels in ε4 carriers (Tapiola et al. 2000)
tau more elevated levels in ε4 carriers
Aß1-42 563 more reduced levels in ε4 carriers (Prince et al. 2004)
Aß1-42 150 more reduced levels in ε4 carriers (Sunderland et al. 2004)

Table 5.

Influence of the ApoE ε4 allele on CSF markers in AD (modified from Varges et al. 2011)

The ApoE ε4 allele status is important to be analysed in the context of CSF biomarker. Some studies report no association between ApoE ε4 allele and tau level, whereas others show higher tau level among ApoE ε4 carriers when compared to non-carriers among AD patients. The situation seems to be clearer for ß-amyloid 1-42: several studies report correlations between Aß1-42 levels and the ApoE ε4 allele (Table 5) (modified from Varges et al. 2011). Although the pathological links are not clearly identify at the moment, it is apparent that the ApoE genotype has to be taken into consideration.

Advertisement

4. Clinical criteria for AD

CSF biomarkers play an important role in clinical diagnosis and differential diagnosis of neurodegenerative dementia. A lot of research in this area has been already conducted. Recently, these markers which are associated with disease pathology in the brain namely Aß1-42 as a parameter of the amyloid cascade and tau and its phosphorylated isoforms have been suggested to be included into diagnostic criteria for AD. The typical AD signature comprises low CSF Aß1-42 levels and high total tau/p-tau level and it was suggested as a parameter of one of the supportive features at the same level as structural and functional brain imaging for probable AD diagnosis (Dubois et al. 2007).

Advertisement

5. Conclusions

The PubMed search on 31.1.2011 using keywords cerebrospinal fluid and Alzheimer reveals 2336 hits. Although not all are dealing directly with biomarker discovery and identification of novel proteins for diagnosis, they are linked to the topic and demonstrate once again the importance of the area. Adequate biomarker, which can be easily analysed in CSF and, even better, blood, will have a great potential for clinical and also preclinical diagnosis of the disease. In neurodegenerative disorders, we have to meet the problem of early disease diagnosis, because it is likely that neuroprotective and other pharmacological strategies will allow better treatment response in earlier disease stages.

Advertisement

Acknowledgments

This manuscript is supported by the BMBF grant within German Network for Degenerative Dementia, KNDD-2, 2011-2013, Determinants for disease progression in AD

References

  1. 1. Alonso A. Zaidi T. Novak M. Grundke-Iqbal I. Iqbal K. 2001 Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments.Proc Natl Acad Sci U S A 98 12 6923 6928
  2. 2. Alves G. Bronnick K. Aarsland D. Blennow K. Zetterberg H. Ballard C. et al. 2010 CSF amyloid-beta and tau proteins, and cognitive performance, in early and untreated Parkinson’s disease: the Norwegian ParkWest study.J Neurol Neurosurg Psychiatry 81 10 1080 1086
  3. 3. Andreasen N. Minthon L. Clarberg A. Davidsson P. Gottfries J. Vanmechelen E. et al. 1999Sensitivity, specificity, and stability of CSF-tau in AD in a community-based patient sample. Neurology 53 7 1488 1494
  4. 4. Arai H. Morikawa Y. Higuchi M. Matsui T. Clark C. M. Miura M. et al. 1997 Cerebrospinal fluid tau levels in neurodegenerative diseases with distinct tau-related pathology.Biochem Biophys Res Commun 236 262 264
  5. 5. Bahl JM, Heegaard NH, Falkenhorst G, Laursen H, Hogenhaven H, Molbak K et al. 2009 Neurobiol Aging The diagnostic efficiency of biomarkers in sporadic Creutzfeldt-Jakob disease compared to Alzheimer’s disease 11 30 1834 1841
  6. 6. Bibl M. Mollenhauer B. Esselmann H. Lewczuk P. Klafki H. W. Sparbier K. et al. 2006a CSF amyloid-beta-peptides in Alzheimer’s disease, dementia with Lewy bodies and Parkinson’s disease dementia.Brain 129(Pt 5):1177 EOF 87 EOF
  7. 7. Bibl M. Mollenhauer B. Esselmann H. Lewczuk P. Trenkwalder C. Brechlin P. et al. 2006b CSF diagnosis of Alzheimer’s disease and dementia of Lewy bodies.J Neural Transm 113 11 1771 1778
  8. 8. Brettschneider J. Petzold A. Schottle D. Claus A. Riepe M. Tumani H. 2006 The neurofilament heavy chain (NfH) in the cerebrospinal fluid diagnosis of Alzheimer’s disease. Dement Geriatr Cogn Disord 21(5-6):291 EOF 295 EOF
  9. 9. Brys M. Pirraglia E. Rich K. Rolstad S. Mosconi L. Switalski R. et al. 2009 Prediction and longitudinal study of CSF biomarkers in mild cognitive impairmentNeurobiol Aging 30 5 682 690
  10. 10. Buchhave P. Blennow K. Zetterberg H. Stomrud E. Londos E. Andreasen N. et al. 2009Longitudinal study of CSF biomarkers in patients with Alzheimer’s disease. PLoS One 4(7):e6294.
  11. 11. Buerger K. Otto M. Teipel S. J. Zinkowski R. Blennow K. De Bernardis J. et al. 2006Dissociation between CSF total tau and tau protein phosphorylated at threonine 231 in Creutzfedt-Jakob disease. Neurobiol Aging 27 1 10 15
  12. 12. Davidson Y. Gibbons L. Purandare N. Byrne J. Hardicre J. Wren J. et al. 2006 Apolipoprotein E epsilon4 allele frequency in vascular dementia. Dementia and Geriatric Cognitive Disorders 22 1 15 19
  13. 13. De Meyer G. Shapiro F. Vanderstichele H. Vanmechelen E. Engelborghs S. De Deyn P. P. et al. 2010Diagnosis-independent Alzheimer disease biomarker signature in cognitively normal elderly people. Arch Neurol 67 8 949 956
  14. 14. Diniz BS, Pinto Junior JA, Forlenza OV. 2008 Do CSF total tau, phosphorylated tau, and beta-amyloid 42 help to predict progression of mild cognitive impairment to Alzheimer’s disease? A systematic review and meta-analysis of the literature. World J Biol Psychiatry 9 3 172 182
  15. 15. Dubois B. Feldman H. H. Jacova C. De Kosky S. T. Barberger-Gateau P. Cummings J. et al. 2007 Research criteria for the diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA criteriaLancet Neurol 6 734 746
  16. 16. Engelborghs S, Sleegers K, Cras P. 2007. Brain No association of CSF biomarkers with APOEepsilon4, plaque and tangle burden in definite Alzheimer’s disease. 130 2320 2326
  17. 17. Ewers M. Zhong Z. Buerger K. 2008 Increased CSF BACE 1 activity is associated with ApoE-epsilon 4 genotype in subjects with mild cognitive impairment and Alzheimer’s disease.Brain 131 1252 1258
  18. 18. Battisti C. Radi E. Federico A. 2006 Cerebrospinal fluid tau, A beta, and phosphorylated tau protein for the diagnosis of Alzheimer’s disease. 208 1 39 46
  19. 19. Galasko D. Chang L. Motter R. 1998 High cerebrospinal fluid tau and low amyloid beta42 levels in the clinical diagnosis of Alzheimer disease and relation to apolipoprotein E genotype.Arch Neurol 55 937 945
  20. 20. Galasko D. Clark C. Chang L. Miller B. Green R. C. Motter R. et al. 1997 Assessment of CSF levels of tau protein in mildly demented patients with Alzheimer’s disease Neurology 48 3 632 635
  21. 21. Gawinecka J. Zerr I. 2010 Cerebrospinal fluid biomarkers in human prion diseasesFuture Neurol. 5 2 301 316
  22. 22. Gloeckner S. F. Meyne F. Wagner F. Heinemann U. Krasnianski A. Meissner B. et al. 2008 Quantitative analysis of transthyretin, tau and amyloid-beta in patients with dementia.J Alzheimers Dis 14 1 17 25
  23. 23. Goedert M. Spillantini M. G. Potier M. C. Ulrich J. Crowther R. A. 1989 Cloning and sequencing of the cDNA encoding an isoform of microtubule-associated protein tau containing four tandem repeats: differential expression of tau protein mRNAs in human brain.Embo J 8 2 393 399
  24. 24. Golombowski S. Mueller-Spahn F. Romig H. 1997 Dependence of cerebrospinal fluid Tau protein levels on apolipoprotein E4 allele frequency in patients with Alzheimer’s disease.Neurosci Lett 225 213 215
  25. 25. Henneman W. J. Vrenken H. Barnes J. Sluimer I. C. Verwey N. A. MA Blankenstein et. al 2009 Baseline CSF p-tau levels independently predict progression of hippocampal atrophy in Alzheimer disease Neurology 73 12 935 940
  26. 26. Hertze J. Minthon L. Zetterberg H. Vanmechelen E. Blennow K. Hansson O. 2010 Evaluation of CSF biomarkers as predictors of Alzheimer’s disease: a clinical follow-up study of 4.7 yearsJ Alzheimers Dis 21 4 1119 1128
  27. 27. Hu Y. Y. He S. S. Wang X. Duan Q. H. Grundke-Iqbal I. Iqbal K. et al. 2002 Levels of nonphosphorylated and phosphorylated tau in cerebrospinal fluid of Alzheimer’s disease patients: an ultrasensitive bienzyme-substrate-recycle enzyme-linked immunosorbent assay.Am J Pathol 160 4 1269 1278
  28. 28. Hulstaert F. Blennow K. Ivanoiu A. Schoonderwaldt H. C. Riemenschneider M. De Deyn P. P. et al. 1999Improved discrimination of AD patients using beta-amyloid(1-42) and tau levels in CSF. Neurology 52 8 1555 1562
  29. 29. Ibach B. Binder H. Dragon M. Poljansky S. Haen E. Schmitz E. et al. 2006 Cerebrospinal fluid tau and beta-amyloid in Alzheimer patients, disease controls and an age-matched random sample.Neurobiol Aging 27 9 1202 1211
  30. 30. Ishiguro K. Ohno H. Arai H. Yamaguchi H. Urakami K. Park J. M. et al. 1999 Phosphorylated tau in human cerebrospinal fluid is a diagnostic marker for Alzheimer’s disease.Neurosci Lett 270 2 91 94
  31. 31. Itoh N. Arai H. Urakami K. Ishiguro K. Ohno H. Hampel H. et al. 2001Large-scale, multicenter study of cerebrospinal fluid tau protein phosphorylated at serine 199 for the antemortem diagnosis of Alzheimer’s disease. Ann Neurol 50 2 150 156
  32. 32. Kapaki E. Liappas I. Paraskevas G. P. Theotoka I. Rabavilas A. 2005 The diagnostic value of tau protein, beta-amyloid (1-42) and their ratio for the discrimination of alcohol-related cognitive disorders from Alzheimer’s disease in the early stages.Int J Geriatr Psychiatry 20 8 722 729
  33. 33. Kapaki E. N. Paraskevas G. P. Tzerakis N. G. Sfagos C. Seretis A. Kararizou E. et al. 2007Cerebrospinal fluid tau, phospho-tau181 and beta-amyloid1-42 in idiopathic normal pressure hydrocephalus: a discrimination from Alzheimer’s disease. Eur J Neurol 14 2 168 173
  34. 34. Kasuga K. Tokutake T. Ishikawa A. Uchiyama T. Tokuda T. Onodera O. et al. 2010 Differential levels of alpha-synuclein, beta-amyloid42 and tau in CSF between patients with dementia with Lewy bodies and Alzheimer’s disease.J Neurol Neurosurg Psychiatry 81 6 608 610
  35. 35. Kopke E. Tung Y. C. Shaikh S. Alonso A. C. Iqbal K. Grundke-Iqbal I. 1993 Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease.J Biol Chem 268 32 24374 24384
  36. 36. Lamb H. Christie J. Singleton A. B. 1998 Apolipoprotein E and alpha-1 antichymotrypsin polymorphism genotyping in Alzheimer’s disease and in dementia with Lewy bodies. Distinctions between diseases. Neurology 50 388 391
  37. 37. Lewczuk P. Esselmann H. Otto M. Maler J. M. Henkel M. K. Eikenberg O. et al. 2004Neurochemical diagnosis of Alzheimer’s dementia by CSF Aß 42, Aß 42/ Aß40 ratio and total tau. Neurobiol Aging 25 273 281
  38. 38. Lindwall G. Cole R. D. 1984Phosphorylation affects the ability of tau protein to promote microtubule assembly. J Biol Chem 259 8 5301 5305
  39. 39. Mattsson N. Zetterberg H. Hansson O. Andreasen N. Parnetti L. Jonsson M. et al. 2009 CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairmentJama 302 4 385 393
  40. 40. Mecocci P. Cherubini A. Bregnocchi M. Chionne F. Cecchetti R. Lowenthal D. T. et al. 1998Tau protein in cerebrospinal fluid: a new diagnostic and prognostic marker in Alzheimer disease? Alzheimer Disease and Associated Disorders 12 211 214
  41. 41. Mitchell AJ. 2009 CSF phosphorylated tau in the diagnosis and prognosis of mild cognitive impairment and Alzheimer’s disease: a meta-analysis of 51 studiesJ Neurol Neurosurg Psychiatry 80 9 966 975
  42. 42. Mollenhauer B. Esselmann H. Trenkwalder C. Schulz-Schaeffer W. Kretzschmar H. Otto M. et al. 2011 CSF amyloid-beta peptides in neuropathologically diagnosed dementia with Lewy bodies and Alzheimer’s Disease.J Alzheimers Dis 24 2 383 391
  43. 43. Okonkwo OC, Mielke MM, Griffith HR, Moghekar AR, O’Brien RJ, Shaw LM et al. 2011 Cerebrospinal fluid profiles and prospective course and outcome in patients with amnestic mild cognitive impairmentArch Neurol 68 1 113 119
  44. 44. Pauwels E. K. Volterrani D. Mariani G. 2009 Biomarkers for Alzheimer’s diseaseDrug News Perspect 22 3 151 160
  45. 45. Prince JA, Zetterberg H, Andreasen N. 2004. APOE E4 allele is associated with reduced cerebrospinal fluid levels of Aß42. Neurology 62 2116 2118
  46. 46. Riemenschneider M. Schmolke M. Lautenschlager 2000 Cerebrospinal beta-amyloid (1-42) in early Alzheimer’s disease: association with apolipoprotein E genotype and cognitive decline. Neurosci Lett 284 85 88
  47. 47. Riemenschneider M. Wagenpfeil S. Vanderstichele H. Otto M. Wiltfang J. Kretzschmar H. et al. 2003Phospho-tau/total tau ration in cerebrospinal fluid discriminates Creutzfeldt-Jakob disease from other dementias. Molecular Psychiatry 8 343 347
  48. 48. Saunders AM, Strittmatter WJ, Schmechel DE. 1993 Association of apolipoprotein E allele epsilon 4 with late onset familial and sporadic Alzheimer’s disease. Neurology 43 1467 1472
  49. 49. Scheurich A. Urban P. P. Koch-Khoury N. Fellgiebel A. 2010 CSF phospho-tau is independent of age, cognitive status and gender of neurological patientsJ Neurol 257 4 609 614
  50. 50. Shaw L. M. Vanderstichele H. Knapik-Czajka M. Clark C. M. Aisen P. S. Petersen R. C. et al. 2009Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann Neurol 65 4 403 413
  51. 51. Shoji M. Kanai M. 2001 Cerebrospinal fluid Abeta40 and Abeta42: Natural course and clinical usefulness.J Alzheimers Dis 3 3 313 321
  52. 52. Siderowf A. Xie S. X. Hurtig H. Weintraub D. Duda J. Chen-Plotkin A. et al. 2010CSF amyloid {beta} 1-42 predicts cognitive decline in Parkinson disease. Neurology 75 12 1055 1061
  53. 53. Slats D. Spies P. E. MJ Sjogren Verhey. F. R. MM Verbeek Olde. Rikkert M. G. 2010 Cerebrospinal fluid biomarkers in diagnosing Alzheimer’s disease in clinical practice: an illustration with 3 case reports. CaseRep Neurol 2 1 5 11
  54. 54. MA Smach Charfeddine. B. Lammouchi T. 2008 CSF beta-amyloid 1-42 and tau in Tunisian patients with Alzheimer’s disease: the effect of APOE epsilon4 allele.Neurosci Lett 440 145 149
  55. 55. Snider B. J. Fagan A. M. Roe C. Shah A. R. Grant E. A. Xiong C. et al. 2009 Cerebrospinal fluid biomarkers and rate of cognitive decline in very mild dementia of the Alzheimer typeArch Neurol 66 5 638 645
  56. 56. Spies P. E. Slats D. Sjögren J. M. Kremer B. P. Verhey F. R. Rikkert M. G. et al. 2010 The cerebrospinal fluid amyloid beta42/40 ratio in the differentiation of Alzheimer’s disease from non-Alzheimer’s dementia.Curr Alzheimer Res 7 5 470 476
  57. 57. Stefani A. Martorana A. Bernardini S. Panella M. Mercati F. Orlacchio A. et al. 2006 CSF markers in Alzheimer disease patients are not related to the different degree of cognitive impairmentJ Neurol Sci 251(1-2):124 EOF
  58. 58. Sunderland T, Linker G, Mirza N, Putnam KT, Friedman DL, Kimmel LH et al. 2003. Decreased beta-amyloid1-42 and increased tau levels in cerebrospinal fluid of patients with Alzheimer disease. Jama 16 289 2094 2103
  59. 59. Sunderland T. Mirza N. Putnam K. T. 2004Cerebrospinal fluid ß-amyloid 1-42 and tau in control subjects at risk for Alzheimer’s disease: the effect of ApoE E4 allele. Biol Psychiatry 56 670 676
  60. 60. Tapiola T. Pirttilä T. Mehta P. D. 2000 Relationship between apoE genotype and CSF beta-amyloid (1-42) and tau in patients with probable and definite Alzheimer’s disease.Neurobiol Aging 21 735 740
  61. 61. van Harten AC, Kester MI, Visser PJ, Blankenstein MA, Pijnenburg YA, van der Flier WM et al. 2011Tau and p-tau as CSF biomarkers in dementia: a meta-analysis. Clin Chem Lab Med 49 3 353 366
  62. 62. Varges D. Jung K. Gawinecka J. Heinemann U. Schmitz M. von Ahsen. N. et al. 2011Amyloid-b 1-42 levels are modified by apolipoprotein E e4 in Creutzfeldt-Jakob disease in a similar manner as in Alzheimer’s disease. J Alzheimers Dis 23 4 717 726

Written By

Inga Zerr, Lisa Kaerst, Joanna Gawinecka and Daniela Varges

Submitted: 31 May 2011 Published: 06 September 2011