ArticlePDF Available

Novel diagnostic cerebrospinal fluid protein biomarkers for pathologic subtypes of frontotemporal dementia identified by proteomics

Authors:

Abstract and Figures

Introduction: Reliable cerebrospinal fluid (CSF) biomarkers enabling identification of frontotemporal dementia (FTD) and its pathologic subtypes are lacking. Methods: Unbiased high-resolution mass spectrometry-based proteomics was applied on CSF of FTD patients with TAR DNA-binding protein 43 (TDP-43, FTD-TDP, n = 12) or tau pathology (FTD-tau, n = 8), and individuals with subjective memory complaints (SMC, n = 10). Validation was performed by applying enzyme-linked immunosorbent assay (ELISA) or enzymatic assays, when available, in a larger cohort (FTLD-TDP, n = 21, FTLD-tau, n = 10, SMC, n = 23) and in Alzheimer's disease (n = 20), dementia with Lewy bodies (DLB, n = 20), and vascular dementia (VaD, n = 18). Results: Of 1914 identified CSF proteins, 56 proteins were differentially regulated (fold change >1.2, P < .05) between the different patient groups: either between the two pathologic subtypes (10 proteins), or between at least one of these FTD subtypes and SMC (47 proteins). We confirmed the differential expression of YKL-40 by ELISA in a partly independent cohort. Furthermore, enzyme activity of catalase was decreased in FTD subtypes compared with SMC. Further validation in a larger cohort showed that the level of YKL-40 was twofold increased in both FTD pathologic subtypes compared with SMC and that the levels in FTLD-tau were higher compared to Alzheimer's dementia (AD), DLB, and VaD patients. Clinical validation furthermore showed that the catalase enzyme activity was decreased in the FTD subtypes compared to SMC, AD and DLB. Discussion: We identified promising CSF biomarkers for both FTD differential diagnosis and pathologic subtyping. YKL-40 and catalase enzyme activity should be validated further in similar pathology defined patient cohorts for their use for FTD diagnosis or treatment development.
Content may be subject to copyright.
CSF Biomarkers
Novel diagnostic cerebrospinal fluid biomarkers for pathologic subtypes
of frontotemporal dementia identified by proteomics
Charlotte E. Teunissen
a,
*, Naura Elias
a
, Marleen J. A. Koel-Simmelink
a
, Sisi Durieux-Lu
a
,
Arjan Malekzadeh
a
, Thang V. Pham
b
, Sander R. Piersma
b
, Tommaso Beccari
c
,
Lieke H. H. Meeter
d
, Elise G. P. Dopper
d,e
, John C. van Swieten
d,e
, Connie R. Jimenez
b,
*,
Yolande A. L. Pijnenburg
e,
*
a
Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam,
The Netherlands
b
OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
c
Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
d
Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
e
Alzheimer Center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
Abstract Introduction: Reliable cerebrospinal fluid (CSF) biomarkers enabling identification of frontotem-
poral dementia (FTD) and its pathologic subtypes are lacking.
Methods: Unbiased high-resolution mass spectrometry–based proteomics was applied on CSF of FTD
patients with TAR DNA-binding protein 43 (TDP-43, FTD-TDP, n 512) or tau pathology (FTD-tau,
n58), and individuals with subjective memory complaints (SMC, n 510). Validation was performed
by applying enzyme-linked immunosorbent assay (ELISA) or enzymatic assays, when available, in a
larger cohort (FTLD-TDP, n 521, FTLD-tau, n 510, SMC, n 523) and in Alzheimer’s disease
(n 520), dementia with Lewy bodies (DLB, n 520), and vascular dementia (VaD, n 518).
Results: Of 1914 identified CSF proteins, 56 proteins were differentially regulated (fold change
.1.2, P,.05) between the different patient groups: either between the two pathologic subtypes
(10 proteins), or between at least one of these FTD subtypes and SMC (47 proteins). We confirmed
the differential expression of YKL-40 by ELISA in a partly independent cohort. Furthermore,
enzyme activity of catalase was decreased in FTD subtypes compared with SMC. Further validation
in a larger cohort showed that the level of YKL-40 was twofold increased in both FTD pathologic
subtypes compared with SMC and that the levels in FTLD-tau were higher compared to Alzheimer’s
dementia (AD), DLB, and VaD patients. Clinical validation furthermore showed that the catalase
enzyme activity was decreased in the FTD subtypes compared to SMC, AD and DLB.
Discussion: We identified promising CSF biomarkers for both FTD differential diagnosis and path-
ologic subtyping. YKL-40 and catalase enzyme activity should be validated further in similar pathol-
ogy defined patient cohorts for their use for FTD diagnosis or treatment development.
Ó2016 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an
open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/
4.0/).
Keywords: Biomarkers; Cerebrospinal fluid; Proteomics; Frontotemporal dementia; Pathology; TDP-43; Tau; Differential
diagnosis
1. Introduction
Frontotemporal dementia (FTD) is the second most prev-
alent dementia of patients aged ,65 years that clinically
presents with either behavior and personality changes or
*Shared senior authors.
*Corresponding author. Tel.: 131-20-4443680; Fax: 131-20-4443895.
E-mail address: c.teunissen@vumc.nl
http://dx.doi.org/10.1016/j.dadm.2015.12.004
2352-8729/ Ó2016 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-94
language disturbance. The disease is often misdiagnosed in
the early stage, either as a psychiatric disorder or as a
different type of dementia such as Alzheimer’s dementia
(AD). The pathology is characterized by two main distinct
subtypes, i.e., tau pathology accounting for roughly one
half of cases and TAR DNA-binding protein 43 (TDP-43)
pathology for the other half [1,2]. The clinical spectrum of
FTD does not correlate with the distinct pathologies,
except when the underlying pathology of FTD is predicted
by the presence of an autosomal dominant mutation, which
is found in only 20%–30% of the patients [3]. Mutations
in the C9orf72 and GRN genes correspond to TDP-43
pathology, and mutations in the microtubule-associated pro-
tein tau (MAPT) to tau pathology, but an autosomal domi-
nant family history is found in only 20%–30% of the
patients. In addition, FTD with amyotrophic lateral sclerosis
or motor neuron disease is almost always associated with un-
derlying TDP-43 pathology [4]. Correct diagnosis and sub-
typing is very relevant to determine patient management
plans and boost therapy development, especially to develop
treatments targeting either tau or TDP-43 pathologic
mechanisms.
Thus far, no reliable biomarker or set of biomarker with
both high sensitivity and high specificity is available for
FTD, let alone its pathologic subtypes. The cerebrospinal
fluid (CSF) biomarkers for AD, i.e., (p)Tau and amyloid
b-42 (Ab42), appear to be of limited value for the diagnosis
of clinical FTD [5,6], although a prognostic value of tau in
diagnosed FTD patients has been reported [7]. A reduced
CSF P-tau-181-to-tau ratio has recently been found to iden-
tify patients with TDP-43 pathology at a sensitivity and
specificity of each 82% [8], which awaits independent vali-
dation.
A good technology to identify multiple novel bio-
markers in body fluids is mass spectrometry–based prote-
omics [9]. So far, no comprehensive discovery at the
protein level has been performed as previous proteomics
studies used low resolution methods for profiling of a
limited set of abundant CSF peptides or proteins in clini-
cally defined FTD patient groups [10–14]. A recent
immunoassay-based proteomics study focused on an anal-
ysis of 151 biomarkers for different pathologic subtypes
of FTD. This has yielded several proteins that discrimi-
nated FTD-TDP-43 and FTD-tau, including interleukins
(IL-23 and IL-7), which combined had an 86% sensitivity
and 78% specificity [15].
In the present study, we aimed to identify novel
pathology-specific biomarkers for FTD by in-depth protein
profiling of antemortem collected CSF of FTD patients
with known underlying pathology. We applied unbiased
CSF proteomics methods [16] in patients with confirmed
tau or TDP-43 pathology, either by genetic testing or post-
mortem analysis, and controls with subjective memory com-
plaints (SMC). We validated the findings using alternative
assays in a largely independent cohort and in patients with
other dementias.
2. Methods
2.1. Patients
Patients with established FTD subtypes and SMC were
included from the biobanks of the Amsterdam Dementia
Cohort and from the Erasmus MC. The method to define
the pathology to is outlined in Table 1, which shows that
the majority was based on postmortem examination. FTD
pathology was reviewed according to international criteria
[17]. Pathologic examination was performed according to
protocolized procedures by the Dutch brain bank, including
specific immunostaining for TDP-43 pathology and tau pa-
thology. Genetic testing was performed for mutations in
the MAPT and progranulin genes and for the hexanucleotide
repeat at C9orf72. The discovery cohort contained 30 pa-
tients, the validation cohort 53 patients, and 17 of the 53 pa-
tients in the validation cohort overlapped with the discovery
cohort, as outlined in Table 1.
All subjects underwent extensive dementia screening at
baseline, including physical and neurologic examination,
mini-mental state examination (MMSE), neuropsychological
investigation, electroencephalogram,magnetic resonance im-
aging, and laboratory tests, including lumbar puncture. De-
mentia diagnoses were made by consensus in a
multidisciplinary meeting according to standard criteria
[18,19]. Probable AD was diagnosed according to the
criteria of the National Institute of Neurological and
Communicative Disorders and Stroke-Alzheimer’s Disease
and Related Disorders association [20], and all patients met
the core clinical National Institute on Aging-Alzheimer’s As-
sociation criteria [21]. Definite FTD was diagnosed using the
criteria of Rascovsky et al. [2]. Control groups of subjects
with SMC consisted of individuals who presented with cogni-
tive complaints, but cognitive and laboratory investigations,
were normal and criteria for mild cognitive impairment, de-
mentia, or any other neurologic or psychiatric disorders
known to cause cognitive complaints were not met. CSF bio-
markers abeta (1–42), tTau, and pTau were not used for the
clinical diagnosis of any of the patients. Groups were matched
for age and gender. Patient characteristics of the discovery and
validation cohorts are presented in Table 1.
The study was performed according to the ethical
principles of the Declaration of Helsinki and was appro-
ved by the local ethics committee. We obtained written
informed consent from all subjects participating in the study.
2.2. CSF biobanking and proteomics analysis
CSF and blood were collected and stored at 280Cin
polypropylene tubes (Sarstedt, N
umbrecht, Germany) after
centrifugation within 1 hourafter withdrawal according to in-
ternational biobanking consensus guidelines optimized for
CSF proteomics [22].CSFAb42, total tau, and p-tau were
measured with commercially available enzyme-linked immu-
nosorbent assays (ELISAs; INNOTEST Fujirebio, Ghent,
Belgium) on a routine basis as described previously [23].
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-94 87
Proteomics analysis was essentially performed as de-
scribed previously [16] and details are presented in
Supplementary File 1. Because this is a typical proteomics
discovery study, where the candidates are to be validated,
we did not apply a multiple testing correction and applied
aPvalue of .05.
2.3. Independent assay validation
The following assays were used: human adipocyte fatty
acid binding protein (FABP4) ELISA kit was obtained
from BioVendor (Karasek, Czech Republic). The MicroVue
YKL-40 enzyme immunoassay kit was from Quidel Corpo-
ration (San Diego, USA). Complement factor D (CFD)
Quantikine ELISA and interleukin 1 receptor accessory pro-
tein (IL1RAP) DuoSet ELISA were from R&D Systems
(Abingdon, UK). Human apolipoprotein L1 (APOL1)
ELISA was from Proteintech (Manchester, UK). Catalase
enzyme activity was measured using an EnzyChrom
Assay from BioAssay Systems (Hayward, CA, USA). Total
b-hexosaminidase (HexA) activity was measured using
3-mM 4-methylumbelliferyl-2-acetamido-2-deoxy-b-D-
glucopyranoside (4MUGlcNAc) as a substrate in 0.1-M cit-
ric acid/0.2-M disodium phosphate buffer. b-HexA was
measured using 3-mM 4-Methylumbelliferyl 6-Sulfo-2-
acetamido-2-deoxy-b-D-glucopyranoside in 0.1 M citric
acid/0.2 M disodium phosphate buffer. a-Galactosidase A
(GLA) was measured using 4.5-mM 4-methylumbelliferyl-
a-D-galactoside in 0.2-M Na/acetate buffer.
Candidate biomarkers for validation were selected based
on the following criteria: (1) fold change .1.2 and Pvalue
,.05; (2) mean spectral count in all patients in one of the
patients groups .2; and (3) number of identified peptide se-
quences covering at least 20% of the protein. We searched
for availability of ELISAs and selected those with a detailed
validation report (i.e., recovery, linearity, and coefficients of
variation (CV) presented of individual samples). We briefly
tested the analytical performance of all assays for analysis of
CSF as described in Supplementary File 1. We, furthermore,
performed extensive validation of the FABP4 and YKL-40
assays according to the immunoassay validation SOP devel-
oped by the BIOMARKAPD project [24], the results of
which are also presented in Supplementary File 1. Precision
data of all assays are presented in Table 2.
2.4. Statistics
Statistics were performed in SPSS (version 20). None of
the validation results had a normal distribution, as tested by
the Shapiro-Wilk test. Differences in mean values between
clinical groups obtained by ELISA were analyzed with
one-way analysis of variance on the ranked values, correcting
for age as indicated in the results. Spearman correlation
Table 1
Characteristics of included patient populations
Characteristics Age (y)
Sex
(female/
male)
MMSE
score
Disease
duration (y) n
Amsterdam/
Rotterdam Method pathology definition
FTD discovery cohort
SMC 60.9 (15.7) 5/5 10 5/5
TDP-43-FTD 60.9 (5.7) 5/7 12 3/9 PGRN mutation (n 52)/pathology
confirmed (n 510)
Tau-FTD 53.4 (6.7)*3/5 8 2/6 MAPT mutation (n 57)/Pathology
confirmed (n 51)
FTD validation cohort
SMC 60.7 (10.1) 12/9 27.3 (2.9) 23 20/3
TDP-43-FTD 60.5 (5.8) 10/12 26.2 (3.1) 3.3 (2.5) (n 518) 20 12/8 PGRN mutation (n 52)/C9orf72
hexanucleotide repeat (n 52)/
FTD-MND (n 52)/pathology
confirmed (n 514)
Tau-FTD 55.2 (13.6)*6/3 NA 2.9 (1.0) (n 58) 10 4/6 MAPT (n 56)/pathology
confirmed (n 54)
Overlap discovery and validation cohort
SMC 60.1 (19.2) 5/2 7 5/2
FTD-TDP-43 59.9 (4.7) 2/5 7 4/3 GRN (n 52)/pathology
confirmed (n 55)
FTD-Tau 51.5 (6.3) 1/2 3 0/3 MAPT (n 53)
Dementia validation cohort
Alzheimer’s disease (AD) 63.7 (6.9) 8/12 19.7 (6.9) 20
Dementia with Lewy bodies (DLB) 62.9 (5.1) 3/17 23.2 (4.8) 20
Vascular Dementia (VaD) 63.6 (5.4) 3/15 22.3 (4.8) 18
Abbreviations: MMSE, mini-mental state examination; FTD, frontotemporal dementia; TDP-43, TAR DNA-binding protein 43; SMC, subjective memory
complaints; MAPT, microtubule-associated protein tau; NA, not available; SD, standard deviation.
NOTE. Mean values (SD) are given.
*Significantly different between Tau-FTD and the other two groups (P,.05).
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-9488
analyses were performed to study correlations between
markers or demographics. A Pvalue ,.05 was considered
significant.
3. Results
3.1. Identification of discriminatory CSF biomarkers and
functions
To facilitate in-depth coverage of the CSF proteome, CSF
samples were subjected to high-abundant protein depletion
followed by SDS-PAGE (sodium dodecyl sulfate polyacryl-
amide gel electrophoresis) fractionation, in-gel tryptic
digestion, and nanoLC-MS/MS analysis. In total, 1914 pro-
teins were identified in CSF (Supplementary Table 1). The
beta binomial test for comparison of the protein spectral
counts in the three patient groups yielded 56 differentially
regulated proteins (Tables 3–7). Supplementary Fig. 1 shows
a heat map of the relative concentrations of these proteins in
each individual patient sample.
3.1.1. FTD-tau versus FTD-TDP-43
Ten proteins were differentially regulated between FTD-
tau and FTD-TDP-43 patients (Table 3). The largest negative
fold change was observed for KLK7 and F13A1 (,22.0
fold), and the largest positive fold change was observed
for UBL3 (.2.1-fold increase in FTD-TDP-43 compared
with FTD-tau).
3.1.2. FTD-tau versus both SMC and TDP-43
In total, six proteins were differentially regulated be-
tween FTD-tau patients compared with both SMC and
FTD-TDP-43 patients (Table 4). The proteins APOL1 and
N-Myc downstream regulated gene (NDRG)4 had the high-
est positive fold change (.3.5 higher spectral counts in
FTD-tau compared with both SMC and TDP-43).
3.1.3. FTD-tau versus SMC
In total, 25 proteins were differentially regulated between
FTD-tau patients compared with SMC only (Table 5). The
proteins IL1RAP, OLFML1 had the highest negative fold
change (,22.0 times decreased in FTD-tau compared
with SMC). The proteins MAT2A, CA1, CAT, and
S100A7 had the highest positive fold change (.2.4-fold in-
crease in FTD-tau compared with SMC).
3.1.4. FTD-TDP-43 versus SMC
In total, 10 CSF proteins were differentially regulated
(P,.05) between SMC and FTD-TDP-43 patients
(Table 6). Of these, GLA and LAMTOR had the largest
negative fold change (,22.5-fold decrease in FTD-TDP-
43 compared with SMC).
3.1.5. Both FTD-tau and FTD-TDP-43 versus SMC
In total, five proteins were differentially regulated be-
tween SMC and FTD-tau or FTD-TDP-43 patients
(Table 6). Among these, SPTBN5 had the largest negative
fold change (.2.5-fold decrease in both FTD subtypes
compared with SMC).
3.2. Validation by independent assays
Validation was performed in a small subset of biomarker
candidates for which independent assays were available
Table 3
Proteins that were differentially regulated between TDP-43-FTD and Tau-
FTD
Gene
name
tau versus
SMC, fold
change
P
value
TDP-43
versus
SMC
fold change Pvalue
Tau versus
TDP-43,
fold
change
P
value
KLK7 22.064 .029
F13A1 22.003 .026
HSPA8 21.634 .036
FSCN1 21.491 .032
CLIC4 21.481 .030
ABI3BP 21.268 .044
MOG 1.295 .042
HEXA 1.309 .009
CHID1 1.553 .035
UBL3 2.168 .045
Abbreviations: TDP-43, TAR DNA-binding protein 43; FTD, frontotem-
poral dementia; SMC, subjective memory complaints.
Table 4
Proteins that were differentially regulated between FTD-tau and both FTD-
TDP-43 and SMC
Gene
name
SMC
versus tau,
fold
change
P
value
SMC versus
TDP-43,
fold
change
P
value
TDP-43
versus tau,
fold
change
P
value
MYOC 21.817 .011 22.054 .016
SHBG 21.420 .026 21.527 .008
FCGBP 21.335 .002 21.427 .049
IGFALS 1.267 .005 1.299 .011
NDRG4 3.459 .000 2.729 .000
APOL1 5.127 .001 2.595 .009
Abbreviations: FTD, frontotemporal dementia; SMC, subjective memory
complaints; TDP-43, TAR DNA-binding protein 43.
Table 2
Assay precision of all assays included in the validation
Marker
Intra-assay
CV (%) Stdev
Inter-assay
CV (%) Stdev
APOL1 2.55 0.219 1.6–5.1 2.5
Catalase 10.33 0.17 34 0.1
FABP4 1.9–3.0 0.5 5.7–8.8 1.8
CFD 3.2 2.1 4.9 2.8
IL-RAcP 3.82 3.99 4.14 0.017
YKL-40 3.1–3.7 0.3 3.1–10.9 4.4
b-Hexosaminidase-A 3.24 0.73 3.15 0.70
a-Galactosidase A 5.68 2.43 5.73 1.41
Abbreviations: CV, coefficients of variation; FABP4, fatty acid binding
protein; CFD, Complement factor D; Stdev, standard deviation.
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-94 89
commercially or via collaboration as specified in the method
section.
Three of six tested ELISA assays met our validation
criteria for reliable detection in CSF. These were FABP4,
YKL-40, complement factor D, IL1RAP, and APOL1. No
assays to analyze the concentration were available, but
enzyme activity assays were available for catalase, total
HexA, b-HexA, and GLA.
The results in Fig. 1 show that the levels of FABP4 were
comparable between the FTD-patient groups and SMC.
The levels of the YKL-40 were twofold increased in both
FTD pathologic subtypes compared with SMC (Fig. 2),
which confirmed the increase in FTD-TDP-43 and FTD-
tau compared with SMC of the discovery results.
The complement factor D levels were comparable be-
tween the FTD patient groups and SMC (Fig. 3).
The catalase enzyme activities were decreased in both the
FTD-TDP-43 and FTD-tau groups compared with the con-
trols (Fig. 4), which is a reverse relation compared to the in-
crease observed in the protein concentration by proteomics.
The total HexA and GLA activity was not significantly
changed in any of the groups (data now shown).
3.3. Correlations with demographics and clinical
characteristics and between validated biomarkers
CSF YKL-40 was positively correlated with age. We,
therefore, included age as covariate in further analyses of
Table 6
Proteins that were differentially regulated between TDP-43-FTD and SMC
Gene name
SMC
versus tau,
fold
change
P
value
SMC versus
TDP-43,
fold
change
P
value
TDP-43
versus
tau, fold
change
P
value
GLA 25.512 .026
LAMTOR2 22.547 .035
MPZ 21.813 .043
TMEM132B 21.792 .017
IMPA1 21.480 .024
DLD 21.469 .045
GDA 21.205 .034
CPVL 21.351 .013
CTSL1 1.215 .014
ST6GAL2 1.611 .042
Abbreviations: FTD, frontotemporal dementia; SMC, subjective memory
complaints; TDP-43, TAR DNA-binding protein 43.
Table 7
Proteins that were differentially regulated between FTD and SMC (no
specific pathologic subtype)
Gene
name
SMC
versus
tau, fold
change
P
value
SMC versus
TDP-43,
fold
change
P
value
TDP-43
versus
tau, fold
change
P
value
SPTBN5 22.482 .040 22.947 .045
RAD23B 21.901 .016 21.539 .044
AP2B1 21.371 .031 21.306 .039
CFD 21.284 .033 21.223 .037
YKL-40 1.243 .020 1.276 .009
Abbreviations: FTD, frontotemporal dementia; SMC, subjective memory
complaints; TDP-43, TAR DNA-binding protein 43.
SMC TDP43 Tau
0
1
2
3
4
FABP4 (ng/mL)
Fig. 1. FABP4 levels in CSF of different patient groups as measured by
ELISA. Abbreviations: FABP4, fatty acid binding protein; CSF, cerebrospi-
nal fluid; ELISA, enzyme-linked immunosorbent assay; SMC, subjective
memory complaints; TDP-43, TAR DNA-binding protein 43.
Table 5
Proteins that were differentially regulated between Tau-FTD and SMC
Gene
name
SMC
versus
tau, fold
change
P
value
SMC versus
TDP-43,
fold
change
P
value
TDP-43
versus tau,
fold
change
P
value
IL1RAP 22.533 .034
OLFML1 22.013 .023
ISLR2 21.969 .014
CNTNAP3 21.850 .039
TENM2 21.655 .012
GALNS 21.462 .041
F5 21.424 .037
QDPR 21.368 .028
PCMT1 21.307 .049
CTSH 21.307 .018
OLFML3 21.277 .027
NOV 21.266 .011
LCAT 1.347 .032
FSTL5 1.406 .045
CDH15 1.519 .019
CASP14 1.660 .043
ACAN 1.671 .026
FABP4 1.710 .047
FAH 1.731 .018
VCL 1.856 .036
LRP8 1.901 .013
MAT2A 2.471 .022
CA1 3.283 .036
CAT 4.473 .031
S100A7 4.591 .007
Abbreviations: FTD, frontotemporal dementia; SMC, subjective memory
complaint; TDP-43, TAR DNA-binding protein 43; FABP4, fatty acid bind-
ing protein.
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-9490
this marker. CSF YKL-40 was positively correlated with
t-tau and negatively to catalase enzyme activity levels
(data not shown, Table 8). There were no correlations with
MMSE or difference in levels between females and males.
3.4. Validation in other dementia types
We next continued validation of the proteins for which a
differential expression was confirmed, i.e., YKL-40 and
catalase, by analysis of the levels in other common dementia
subtypes, AD, DLB, and vascular dementia (VaD).
The levels of YKL-40 (Fig. 2) were higher in FTD-tau
compared with AD (P,.001), dementia with Lewy bodies
(DLB, P,.001), and VaD (P5.001).
The levels of catalase (Fig. 4) were lower in FTD-tau or
FTD-TDP-43 compared with AD (P,.001), DLB
(P,.05), and in the FTD-tau group compared with VaD
(P5.05).
3.5. Serum analysis
We analyzed YKL-40 concentrations in paired serum of
the FTD patients available in the Amsterdam cohort (12
FTD-TDP-43, 4 FTD-Tau, and 18 SMC). There was no cor-
relation between serum and CSF levels of YKL-40 and no
differences in serum YKL-40 levels between the patient
groups (data not shown).
4. Discussion
In this study, we identified 56 candidate biomarkers that
were differentially regulated between the pathologic sub-
types of FTD or between FTD and SMC, in CSF of homoge-
neous patient groups. The result could be confirmed for one
of five biomarkers for which a robust ELISA was available,
***
***
***
***
***
**
*
**
*
Fig. 2. YKL-40 levels in CSF of different patient groups as measured by
ELISA. ***P,.001. Abbreviations: CSF, cerebrospinal fluid; ELISA,
enzyme-linked immunosorbent assay; SMC, subjective memory complaint;
TDP-43, TAR DNA-binding protein 43; AD, Alzheimer’s dementia; DLB,
dementia with Lewy bodies; VaD, vascular dementia.
SMC TDP43 Tau
50
0
100
150
CFD (ng/mL)
Fig. 3. Complement factor D levels in CSF of different patient groups as
measured by ELISA. Abbreviations: CSF, cerebrospinal fluid; ELISA,
enzyme-linked immunosorbent assay; SMC, subjective memory com-
plaints; TDP-43, TAR DNA-binding protein 43.
**
**
***
*
*
***
*
**
*
*
*
Fig. 4. Catalase enzyme activity in CSF of different patient groups.
*P,.05, **P,.01, ***P,.001. Abbreviations: CSF, cerebrospinal fluid;
SMC, subjective memory complaints; TDP-43, TAR DNA-binding protein
43; AD, Alzheimer’s dementia; DLB, dementia with Lewy bodies; VaD,
vascular dementia.
Table 8
Correlation between CSF biomarkers and demographics in all groups*
Demographics
CSF biomarker
YKL-40 Catalase
Age 0.300** 0.093
Disease duration*20.133 20.222
Abeta (1–42) 20.100 20.058
t-Tau 0.397*** 0.044
p-Tau 0.197* 0.084
YKL-40 x 20.240*
Abbreviation: CSF, cerebrospinal fluid.
NOTE. Bivariate Spearman correlation coefficients were calculated
(n 588–101). *P,.05; **P,.01; ***P,.001.
*n 526 (YKL-40) and 19 (catalase).
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-94 91
i.e., YKL-40. Moreover, enzyme activity of one of three
tested proteins (catalase) was confirmed to be differentially
regulated between the FTD subtypes and controls.
The strongest increase in the proteomics results (fold
change .3) was observed for S100A7, GLA, APOL1,
NDRG4, CAT, and CA1 (Tables 3–7). Each of these
specific proteins have not yet been related to FTD, and we
here provide some more background information on these
proteins.
S100A7, also called psoriasin, is increased in several can-
cers and has multiple functions, including inflammatory
roles [25].
GLA and APOL1 are lysosomal proteins. They have not
yet been related to FTD pathology but can be very promising
as they confirm the previously revealed important role of the
autophagy/lysosome system in the etiology of FTD. For
instance, the GRN and CHMP2B genes that are directly
related to FTD are associated with the autophagy/lysosomal
pathway [26,27]. Similarly, the discovered genetic risk
factors for FTD, TMEM106b [28,29],RAB38, and CTCS
[30] are all associated with lysosomal pathways. These
data support further research into the role of the identified
CSF proteins in the FTD pathology.
Catalase and CA1 are relevant proteins for oxidative
stress, but it remains to be determined if the increase is
real and not an artifact, as a recent proteomics study sug-
gested these two, besides hemoglobin and peroxiredoxin,
as suitable biomarkers for blood contaminated CSF [31].
NDRG4 levels were increased specifically in the tau
group. The NDRG4 protein expression is upregulated in
aggressive meningioma [32]. In contrast, the levels were
decreased in glioblastoma or colorectal cancer [33].
NDRG4 is specifically expressed in the brain and heart
and plays a possible role in neuronal differentiation. Interest-
ingly, the NDRG4 messenger RNA expression was
decreased in Alzheimer brain tissue [34].
We validated a subset of biomarkers by immunoassays
as independent methods. YKL-40 has already been proven
to have a role in dementia, i.e., in Alzheimer’s dementia
[35,36], and also in early stages of multiple sclerosis
[37]. Our data expand findings in a recent report showing
increased levels in clinically defined FTD patients and a
positive correlation with CSF t-tau [38]. The 1.2-fold in-
crease in CSF levels in AD is of similar magnitude as
earlier reports, and lack of significant difference in our
cohort is likely due to the smaller number of AD patients
included in our study [39]. The elevation observed in other
diseases and the correlation with t-tau, albeit moderate,
indicate that YKL-40 cannot function as a single
pathology-specific biomarker and is probably a sensitive
biomarker for astrogliosis [37]. Interestingly, neuroinflam-
mation is one of the explanatory mechanisms through
which the progranulin haploinsufficiency can cause FTD
[40]. Further studies should define the role of neuroinflam-
mation and, in particular, astrogliosis, in FTD in more
depth [40–43].
Lumbar puncture may be perceived as inconvenient but
gets common practice in the dementia field and risk compli-
cations as severe headache of 0.9%, typical postlumbar
puncture headache of 9%, and back pain of 17% is generally
accepted in this population [44]. Unfortunately, blood levels
of YKL-40 were similar in FTD groups and SMC. This was,
however, not surprising, as blood levels of these biomarkers
have been related to inflammation in several other diseases,
including cardiovascular disease and diabetes, which could
mask brain-related alterations [45–47].
The discovery results on catalase were validated by an ac-
tivity assay, due to the lack of assays to analyze the concen-
trations. It remains to be determined if the activity of this
enzyme correlates to its concentrations, but the decreased
activity observed in FTD is at least encouraging to perform
future validation studies, including studies on the effects of
blood contamination on the activity.
A major strength of the study was the study design, i.e.,
the start with pathologically homogeneous patient groups
that were defined either by postmortem evaluation or genetic
subtyping and the use of an unbiased, well-validated method
for CSF proteomics [48]. Because clinical FTD is heteroge-
neous, which does not correlate strongly to its pathologic
subtypes [49], cohorts with known pathologic subtypes are
very relevant to increase knowledge on pathways, reflected
in biomarkers changes, and therapy development. Another
strength was the stringent screening of ELISA assays as in-
dependent methods and validation in independent cohort
including other dementia types. The validation was success-
ful for 20% of the biomarkers for which a CSF-validated
assay was available, which may forecast a similar success
rate for the remainder-identified proteins. The commercial
availability and shown quality of these assays have impor-
tant implications. First, these can be easily applied for inde-
pendent replication of our study by other research groups,
and second, implementation in clinical practice will be
feasible.
A weakness of this study is that we did not further differ-
entiate the pathologic subtypes, as the TDP-43 can be subdi-
vided into at least four distinct pathologic subtypes,
depending on the cellular location of the TDP-43 inclusions
(e.g., cytoplasmic or axonal) or stage of the pathology
[1,50]. Further subtyping would have led to lower number
of patients per subgroup, due to limited availability.
Nevertheless, the here identified biomarkers can be further
studied in relation to these different pathologies.
So far, there has been a lack of differential diagnostic bio-
markers of the pathologic subtypes of FTD. An interesting
recent report showed that a reduced ratio (,0.37) of p-tau
to t-tau could be a biomarker to identify FTD-TDP-43
from FTD-tau, AD, and healthy seniors with 82% sensitivity
and 82% specificity in cohorts of similar size as ours [8].An
imaging study showed that white matter volume could
discriminate between these pathologic FTD subtypes [51].
Thus, studies focusing on pathologic subtypes of FTD are
just emerging, and it will be interesting to evaluate the
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-9492
additional value combining different CSF biomarkers and
different biomarker modalities for accurate diagnosis.
Taken together, YKL-40 should be studied further for its
use in diagnosis of FTD, as it discriminated both FTD sub-
types from SMC. Our study presented promising new candi-
date biomarkers for a correct and differential diagnosis of the
FTD subtypes, which is important for inclusion of specific
patients for either TDP-43 or tau-targeted treatments.
Acknowledgments
The Association for Frontotemporal Degeneration and
Alzheimer’s Drug Discovery Foundation, and the ZonMw
Memorabel program project “PRODIA,” as part of the
Deltaplan Dementie, are acknowledged for their grant sup-
port of this study.
Supplementary data
Supplementary data related to this article can be found at
http://dx.doi.org/10.1016/j.dadm.2015.12.004.
RESEARCH IN CONTEXT
1. Systematic review: The authors reviewed the litera-
ture using traditional (e.g., PubMed) sources and
citations within articles. Key words were frontotem-
poral dementia (FTD) and biomarkers and cerebro-
spinal fluid (CSF).
2. Interpretation: Our findings led to the discovery and
validation of several novel CSF biomarkers for the
different pathologic subtypes of FTD. Validation of
inflammatory biomarkers underscores the involve-
ment of these pathways in FTD.
3. Future directions: Future studies should aim at (1) in-
dependent multicenter validation of the biomarkers;
(2) establish the pathways role of these identified
key biomarkers for lysosomal and inflammatory
pathways in FTD pathology; and (3) validation of
biomarkers for which methods are to be developed.
References
[1] Irwin DJ, Trojanowski JQ, Grossman M. Cerebrospinal fluid bio-
markers for differentiation of frontotemporal lobar degeneration
from Alzheimer’s disease. Front Aging Neurosci 2013;5:6.
[2] Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH,
Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behav-
ioural variant of frontotemporal dementia. Brain 2011;134:2456–77.
[3] Seelaar H, Rohrer JD, Pijnenburg YA, Fox NC, van Swieten JC. Clin-
ical, genetic and pathological heterogeneity of frontotemporal demen-
tia: A review. J Neurol Neurosurg Psychiatry 2011;82:476–86.
[4] Ng AS, Rademakers R, Miller BL. Frontotemporal dementia: A bridge
between dementia and neuromuscular disease. Ann N Y Acad Sci
2015;1338:71–93.
[5] Pijnenburg YA, Janssen JC, Schoonenboom NS, Petzold A, Mulder C,
Stigbrand T, et al. CSF neurofilaments in frontotemporal dementia
compared with early onset Alzheimer’s disease and controls. Dement
Geriatr Cogn Disord 2007;23:225–30.
[6] Schoonenboom NS, Reesink FE, Verwey NA, Kester MI,
Teunissen CE, van de Ven PM, et al. Cerebrospinal fluid markers for
differential dementia diagnosis in a large memory clinic cohort.
Neurology 2012;78:47–54.
[7] Borroni B, Benussi A, Cosseddu M, Archetti S, Padovani A. Cerebro-
spinal fluid tau levels predict prognosis in non-inherited frontotempo-
ral dementia. Neurodegener Dis 2014;13:224–9.
[8] Hu WT, Watts K, Grossman M, Glass J, Lah JJ, Hales C, et al. Reduced
CSF p-Tau181 to tau ratio is a biomarker for FTLD-TDP. Neurology
2013;81:1945–52.
[9] Pham TV, Piersma SR, Oudgenoeg G, Jimenez CR. Label-free mass
spectrometry-based proteomics for biomarker discovery and valida-
tion. Expert Rev Mol Diagn 2012;12:343–59.
[10] Davidsson P, Sjogren M, Andreasen N, Lindbjer M, Nilsson CL, West-
man-Brinkmalm A, et al. Studies of the pathophysiological mecha-
nisms in frontotemporal dementia by proteome analysis of CSF
proteins. Brain Res Mol Brain Res 2002;109:128–33.
[11] Mattsson N, Ruetschi U, Pijnenburg YA, Blankenstein MA,
Podust VN, Li S, et al. Novel cerebrospinal fluid biomarkers of axonal
degeneration in frontotemporal dementia. Mol Med Rep 2008;
1:757–61.
[12] Ruetschi U, Zetterberg H, Podust VN, Gottfries J, Li S, Hviid SA, et al.
Identification of CSF biomarkers for frontotemporal dementia using
SELDI-TOF. Exp Neurol 2005;196:273–81.
[13] Schweitzer K, Decker E, Zhu L, Miller RE, Mirra SS, Spina S, et al.
Aberrantly regulated proteins in frontotemporal dementia. Biochem
Biophys Res Commun 2006;348:465–72.
[14] Simonsen AH, McGuire J, Podust VN, Hagnelius NO, Nilsson TK,
Kapaki E, et al. A novel panel of cerebrospinal fluid biomarkers for
the differential diagnosis of Alzheimer’s disease versus normal aging
and frontotemporal dementia. Dement Geriatr Cogn Disord 2007;
24:434–40.
[15] Hu WT, Chen-Plotkin A, Grossman M, Arnold SE, Clark CM,
Shaw LM, et al. Novel CSF biomarkers for frontotemporal lobar de-
generations. Neurology 2010;75:2079–86.
[16] Fratantoni SA, Piersma SR, Jimenez CR. Comparison of the perfor-
mance of two affinity depletion spin filters for quantitative proteomics
of CSF: Evaluation of sensitivity and reproducibility of CSF analysis
using GeLC-MS/MS and spectral counting. Proteomics Clin Appl
2010;4:613–7.
[17] Cairns NJ, Bigio EH, Mackenzie IR, Neumann M, Lee VM,
Hatanpaa KJ, et al. Neuropathologic diagnostic and nosologic criteria
for frontotemporal lobar degeneration: Consensus of the Consortium
for Frontotemporal Lobar Degeneration. Acta Neuropathol 2007;
114:5–22.
[18] Roman GC, Tatemichi TK, Erkinjuntti T, Cummings JL, Masdeu JC,
Garcia JH, et al. Vascular dementia: Diagnostic criteria for research
studies. Report of the NINDS-AIREN International Workshop.
Neurology 1993;43:250–60.
[19] McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H,
et al. Diagnosis and management of dementia with Lewy bodies: Third
report of the DLB Consortium. Neurology 2005;65:1863–72.
[20] McKhann G, Drachman D, Folstein M, Katzman R, Price D,
Stadlan EM. Clinical diagnosis of Alzheimer’s disease: Report of
the NINCDS-ADRDAWork Group under the auspices of Department
of Health and Human Services Task Force on Alzheimer’s Disease.
Neurology 1984;34:939–44.
[21] McKhann GM, Knopman DS, Chertkow H, Hyman BT,
Jack CR Jr, Kawas CH, et al. The diagnosis of dementia due to
Alzheimer’s disease: Recommendations from the National Institute
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-94 93
on Aging-Alzheimer’s Association workgroups on diagnostic
guidelines for Alzheimer’s disease. Alzheimers Dement 2011;
7:263–9.
[22] Teunissen CE, Petzold A, Bennett JL, Berven FS, Brundin L,
Comabella M, et al. A consensus protocol for the standardization of
cerebrospinal fluid collection and biobanking. Neurology 2009;
73:1914–22.
[23] Mulder C, Verwey NA, van der Flier WM, Bouwman FH, Kok A, Van
Elk EJ, et al. Amyloid-beta(1-42), total tau, and phosphorylated tau as
cerebrospinal fluid biomarkers for the diagnosis of Alzheimer disease.
Clin Chem 2010;56:248–53.
[24] Andreasson U, Perret-Liaudet A, van Waalwijk van Doorn LJ,
Blennow K, Chiasserini D, Engelborghs S, et al. A practical guide to
immunoassay method validation. Front Neurol 2015;6:179.
[25] Jia J, Duan Q, Guo J, Zheng Y. Psoriasin, a multifunctional player in
different diseases. Curr Protein Pept Sci 2014;15:836–42.
[26] Smith KR, Damiano J, Franceschetti S, Carpenter S, Canafoglia L,
Morbin M, et al. Strikingly different clinicopathological phenotypes
determined by progranulin-mutation dosage. Am J Hum Genet
2012;90:1102–7.
[27] Urwin H, Authier A, Nielsen JE, Metcalf D, Powell C, Froud K, et al.
Disruption of endocytic trafficking in frontotemporal dementia with
CHMP2B mutations. Hum Mol Genet 2010;19:2228–38.
[28] Brady OA, Zheng Y, Murphy K, Huang M, Hu F. The frontotemporal
lobar degeneration risk factor, TMEM106B, regulates lysosomal
morphology and function. Hum Mol Genet 2013;22:685–95.
[29] Van Deerlin VM, Sleiman PM, Martinez-Lage M, Chen-Plotkin A,
Wang LS, Graff-Radford NR, et al. Common variants at 7p21 are asso-
ciated with frontotemporal lobar degeneration with TDP-43 inclu-
sions. Nat Genet 2010;42:234–9.
[30] Ferrari R, Hernandez DG, Nalls MA, Rohrer JD, Ramasamy A,
Kwok JB, et al. Frontotemporal dementia and its subtypes: A
genome-wide association study. Lancet Neurol 2014;13:686–99.
[31] You JS, Gelfanova V, Knierman MD, Witzmann FA, Wang M, Hale JE.
The impact of blood contamination on the proteome of cerebrospinal
fluid. Proteomics 2005;5:290–6.
[32] Kotipatruni RP, Ren X, Thotala D, Jaboin JJ. NDRG4 is a novel onco-
genic protein and p53 associated regulator of apoptosis in malignant
meningioma cells. Oncotarget 2015;6:17594–604.
[33] Li S, Yang B, Li G, He S, Li Y. Downregulation of N-Myc
downstream-regulated gene 4 influences patient survival in gliomas.
Brain Tumor Pathol 2013;30:8–14.
[34] Zhou RH, Kokame K, Tsukamoto Y, Yutani C, Kato H, Miyata T.
Characterization of the human NDRG gene family: A newly identified
member, NDRG4, is specifically expressed in brain and heart. Geno-
mics 2001;73:86–97.
[35] Craig-Schapiro R, Perrin RJ, Roe CM, Xiong C, Carter D, Cairns NJ,
et al. YKL-40: A novel prognostic fluid biomarker for preclinical Alz-
heimer’s disease. Biol Psychiatry 2010;68:903–12.
[36] Olsson B, Hertze J, Lautner R, Zetterberg H, Nagga K, Hoglund K,
et al. Microglial markers are elevated in the prodromal phase of Alz-
heimer’s disease and vascular dementia. J Alzheimers Dis 2013;
33:45–53.
[37] Comabella M, Fernandez M, Martin R, Rivera-Vallve S, Borras E,
Chiva C, et al. Cerebrospinal fluid chitinase 3-like 1 levels are associ-
ated with conversion to multiple sclerosis. Brain 2010;133:1082–93.
[38] Alcolea D, Carmona-Iragui M, Suarez-Calvet M, Sanchez-
Saudinos MB, Sala I, Anton-Aguirre S, et al. Relationship between
b-Secretase, inflammation and core cerebrospinal fluid biomarkers
for Alzheimer’s disease. J Alzheimers Dis 2014;42:157–67.
[39] Alcolea D, Vilaplana E, Pegueroles J, Montal V, Sanchez-Juan P, Gon-
zalez-Suarez A, et al. Relationship between cortical thickness and ce-
rebrospinal fluid YKL-40 in predementia stages of Alzheimer’s
disease. Neurobiol Aging 2015;36:2018–23.
[40] Galimberti D, Scarpini E. Genetics and biology of Alzheimer’s disease
and frontotemporal lobar degeneration. Int J Clin Exp Med 2010;
3:129–43.
[41] Arnold SE, Han LY, Clark CM, Grossman M, Trojanowski JQ. Quan-
titative neurohistological features of frontotemporal degeneration.
Neurobiol Aging 2000;21:913–9.
[42] Rentzos M, Paraskevas GP, Kapaki E, Nikolaou C, Zoga M,
Rombos A, et al. Interleukin-12 is reduced in cerebrospinal fluid of pa-
tients with Alzheimer’s disease and frontotemporal dementia. J Neurol
Sci 2006;249:110–4.
[43] Sjogren M, Folkesson S, Blennow K, Tarkowski E. Increased intra-
thecal inflammatory activity in frontotemporal dementia: Pathophysi-
ological implications. J Neurol Neurosurg Psychiatry 2004;
75:1107–11.
[44] Duits FH, Martinez-Lage P, Paquet C, Engelborghs S, Lleo A,
Hausner L, et al. Performance and complications of lumbar puncture
in memory clinics: Results of the multicenter lumbar puncture feasi-
bility study. Alzheimers Dement 2016;12:154–63.
[45] Rathcke CN, Vestergaard H. YKL-40—An emerging biomarker in car-
diovascular disease and diabetes. Cardiovasc Diabetol 2009;8:61.
[46] Thumser AE, Moore JB, Plant NJ. Fatty acid binding proteins: Tissue-
specific functions in health and disease. Curr Opin Clin Nutr Metab
Care 2014;17:124–9.
[47] Djousse L, Gaziano JM. Plasma levels of FABP4, but not FABP3, are
associated with increased risk of diabetes. Lipids 2012;47:757–62.
[48] Piersma SR, Fiedler U, Span S, Lingnau A, Pham TV, Hoffmann S,
et al. Workflow comparison for label-free, quantitative secretome pro-
teomics for cancer biomarker discovery: Method evaluation, differen-
tial analysis, and verification in serum. J Proteome Res 2010;
9:1913–22.
[49] Josephs KA, Hodges JR, Snowden JS, Mackenzie IR, Neumann M,
Mann DM, et al. Neuropathological background of phenotypical vari-
ability in frontotemporal dementia. Acta Neuropathol 2011;
122:137–53.
[50] Mackenzie IR, Neumann M, Bigio EH, Cairns NJ, Alafuzoff I, Kril J,
et al. Nomenclature and nosology for neuropathologic subtypes of
frontotemporal lobar degeneration: An update. Acta Neuropathol
2010;119:1–4.
[51] McMillan CT, Irwin DJ, Avants BB, Powers J, Cook PA, Toledo JB,
et al. White matter imaging helps dissociate tau from TDP-43 in fron-
totemporal lobar degeneration. J Neurol Neurosurg Psychiatry 2013;
84:949–55.
C.E. Teunissen et al. / Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 2 (2016) 86-9494
... The lack of studies aiming to develop TDP-43 disease-specific in vivo biofluid biomarkers by searching for common changes in TDP-43 pathology relevant patient groups is striking [138]. Especially in FTD, the clinical spectrum does not correlate with the distinct pathologies, except when the underlying pathology of FTD is predicted by the presence of an autosomal dominant mutation, which is found in only 20%-30% of the patients [139]. ...
... A recent proteomic study compared CSF samples from FTD patients with TDP-43 or Tau proteinopathy, confirmed by autopsy or genetic testing to a group of patients with subjective memory complaint [138]. For the discovery experiments, only a small number of samples (n = 8 for Tau and n = 12 for TDP-43) were investigated. ...
Article
Full-text available
TDP-43 is the primary or secondary pathological hallmark of neurodegenerative diseases, such as amyotrophic lateral sclerosis, half of frontotemporal dementia cases, and limbic age-related TDP-43 encephalopathy, which clinically resembles Alzheimer’s dementia. In such diseases, a biomarker that can detect TDP-43 proteinopathy in life would help to stratify patients according to their definite diagnosis of pathology, rather than in clinical subgroups of uncertain pathology. Therapies developed to target pathological proteins that cause the disease a biomarker to detect and track the underlying pathology would greatly enhance such undertakings. This article reviews the latest developments and outlooks of deriving TDP-43-specific biomarkers from the pathophysiological processes involved in the development of TDP-43 proteinopathy and studies, using biosamples from clinical entities associated with TDP-43 pathology to investigate biomarker candidates.
... In line with these findings, our bootstrap classification exercises identified a combination of 61 proteins demarcating FTD patients and non-demented controls with limited accuracy (AUC: 0.61) and large confidence intervals, underpinning the insufficient diagnostic accuracy and further supporting the lack of specific plasma protein signals specifically associated to FTD. These results contrast with previous unbiased proteomics studies performed in CSF samples, including ours, in which several CSF FTD biomarker candidates were identified [31,32]. Whether such discrepancies are driven by the different technologies (i.e. ...
... Throughout literature, it has been challenging to identify and validate protein alterations between both pathological subtypes. For CSF, previous proteomic studies reported several differentially regulated proteins [31] or a biomarker panel enabling sensitive differentiation between TDP and Tau pathology [56], although independent multicenter validation and replication on different platforms is still needed. The lack of a biomarker (panel) for FTD subtypes with feasibility in clinical practice thus far, could have several possible explanations. ...
Article
Full-text available
Background Frontotemporal dementia (FTD) is caused by frontotemporal lobar degeneration (FTLD), characterized mainly by inclusions of Tau (FTLD-Tau) or TAR DNA binding43 (FTLD-TDP) proteins. Plasma biomarkers are strongly needed for specific diagnosis and potential treatment monitoring of FTD. We aimed to identify specific FTD plasma biomarker profiles discriminating FTD from AD and controls, and between FTD pathological subtypes. In addition, we compared plasma results with results in post-mortem frontal cortex of FTD cases to understand the underlying process. Methods Plasma proteins (n = 1303) from pathologically and/or genetically confirmed FTD patients (n = 56; FTLD-Tau n = 16; age = 58.2 ± 6.2; 44% female, FTLD-TDP n = 40; age = 59.8 ± 7.9; 45% female), AD patients (n = 57; age = 65.5 ± 8.0; 39% female), and non-demented controls (n = 148; 61.3 ± 7.9; 41% female) were measured using an aptamer-based proteomic technology (SomaScan). In addition, exploratory analysis in post-mortem frontal brain cortex of FTD (n = 10; FTLD-Tau n = 5; age = 56.2 ± 6.9, 60% female, and FTLD-TDP n = 5; age = 64.0 ± 7.7, 60% female) and non-demented controls (n = 4; age = 61.3 ± 8.1; 75% female) were also performed. Differentially regulated plasma and tissue proteins were identified by global testing adjusting for demographic variables and multiple testing. Logistic lasso regression was used to identify plasma protein panels discriminating FTD from non-demented controls and AD, or FTLD-Tau from FTLD-TDP. Performance of the discriminatory plasma protein panels was based on predictions obtained from bootstrapping with 1000 resampled analysis. Results Overall plasma protein expression profiles differed between FTD, AD and controls (6 proteins; p = 0.005), but none of the plasma proteins was specifically associated to FTD. The overall tissue protein expression profile differed between FTD and controls (7-proteins; p = 0.003). There was no difference in overall plasma or tissue expression profile between FTD subtypes. Regression analysis revealed a panel of 12-plasma proteins discriminating FTD from AD with high accuracy (AUC: 0.99). No plasma protein panels discriminating FTD from controls or FTD pathological subtypes were identified. Conclusions We identified a promising plasma protein panel as a minimally-invasive tool to aid in the differential diagnosis of FTD from AD, which was primarily associated to AD pathophysiology. The lack of plasma profiles specifically associated to FTD or its pathological subtypes might be explained by FTD heterogeneity, calling for FTD studies using large and well-characterize cohorts.
... We hypothesized the relatively fewer changes in FTLD-TDP result from the higher pathological heterogeneity of FTLD-TDP, which can be further classified into four (or five) subtypes according to the cortical layer localization of TDP43 aggregates [31,32]. Conceptually similar findings were reported by our group when comparing the ante mortem cerebrospinal fluid (CSF) proteome of FTLD-TDP and FTLD-tau individuals with that of NHC, the latter comparison yielding many more differentially regulated proteins than the former [35]. To date, all CNS tissue proteomic studies investigated FTLD-TDP cases as one group, so that our findings cannot be corroborated yet [36][37][38][39]. ...
Article
Full-text available
Background: Frontotemporal lobar degeneration (FTLD) is characterized pathologically by neuronal and glial inclusions of hyperphosphorylated tau or by neuronal cytoplasmic inclusions of TDP43. This study aimed at deciphering the molecular mechanisms leading to these distinct pathological subtypes. Methods: To this end, we performed an unbiased mass spectrometry-based proteomic and systems-level analysis of the middle frontal gyrus cortices of FTLD-tau (n = 6), FTLD-TDP (n = 15), and control patients (n = 5). We validated these results in an independent patient cohort (total n = 24). Results: The middle frontal gyrus cortex proteome was most significantly altered in FTLD-tau compared to controls (294 differentially expressed proteins at FDR = 0.05). The proteomic modifications in FTLD-TDP were more heterogeneous (49 differentially expressed proteins at FDR = 0.1). Weighted co-expression network analysis revealed 17 modules of co-regulated proteins, 13 of which were dysregulated in FTLD-tau. These modules included proteins associated with oxidative phosphorylation, scavenger mechanisms, chromatin regulation, and clathrin-mediated transport in both the frontal and temporal cortex of FTLD-tau. The most strongly dysregulated subnetworks identified cyclin-dependent kinase 5 (CDK5) and polypyrimidine tract-binding protein 1 (PTBP1) as key players in the disease process. Dysregulation of 9 of these modules was confirmed in independent validation data sets of FLTD-tau and control temporal and frontal cortex (total n = 24). Dysregulated modules were primarily associated with changes in astrocyte and endothelial cell protein abundance levels, indicating pathological changes in FTD are not limited to neurons. Conclusions: Using this innovative workflow and zooming in on the most strongly dysregulated proteins of the identified modules, we were able to identify disease-associated mechanisms in FTLD-tau with high potential as biomarkers and/or therapeutic targets.
... Increased levels of GST pi (π), encoded by GSTP1, were found in an AD mouse model [72]. Moreover, increased activity of both catalase and GSTs has been found in CSF [73][74][75] and blood samples [76] of patients with different dementia types. Previously, the GSTP1 rs1695 polymorphism was associated with increased AD risk in different studies and one meta-analysis [58,77,78]. ...
Article
Full-text available
Oxidative stress and neuroinflammation are important processes involved in Alzheimer’s disease (AD) and mild cognitive impairment (MCI). Numerous risk factors, including genetic background, can affect the complex interplay between those mechanisms in the aging brain and can also affect typical AD hallmarks: amyloid plaques and neurofibrillary tangles. Our aim was to evaluate the association of polymorphisms in oxidative stress- and inflammation-related genes with cerebrospinal fluid (CSF) biomarker levels and cognitive test results. The study included 54 AD patients, 14 MCI patients with pathological CSF biomarker levels, 20 MCI patients with normal CSF biomarker levels and 62 controls. Carriers of two polymorphic IL1B rs16944 alleles had higher CSF Aβ1–42 levels (p = 0.025), while carriers of at least one polymorphic NFE2L2 rs35652124 allele had lower CSF Aβ1–42 levels (p = 0.040). Association with IL1B rs16944 remained significant in the AD group (p = 0.029). Additionally, MIR146A rs2910164 was associated with Aβ42/40 ratio (p = 0.043) in AD. Significant associations with cognitive test scores were observed for CAT rs1001179 (p = 0.022), GSTP1 rs1138272 (p = 0.005), KEAP1 rs1048290 and rs9676881 (both p = 0.019), as well as NFE2L2 rs35652124 (p = 0.030). In the AD group, IL1B rs1071676 (p = 0.004), KEAP1 rs1048290 and rs9676881 (both p = 0.035) remained associated with cognitive scores. Polymorphisms in antioxidative and inflammation genes might be associated with CSF biomarkers and cognitive test scores and could serve as additional biomarkers contributing to early diagnosis of dementia.
... The patterns of up-and downregulated proteins can provide useful information about the mechanisms that might contribute to the disease progression, or provide protection against cognitive decline. The analysis of CSF proteome has been useful to identify candidate proteins and biological pathways involved in the disease pathophysiology 9,[20][21][22] , and to identify biological subtypes that may respond differently to treatments 19 . ...
Preprint
Full-text available
Providing an accurate prognosis for individual dementia patients remains a challenge since they greatly differ in rates of cognitive decline. In this study, we used machine learning techniques to identify cerebrospinal fluid (CSF) biomarkers that predict the rate of cognitive decline. First, longitudinal follow-up data of 210 dementia patients were used to create fast and slow progression groups. Secondly, we trained random forest classifiers on CSF proteomic profiles and obtained a well-performing prediction model for the progression group (ROC-AUC = 0.82). As a third step, Shapley values and Gini feature importance measures were used to interpret the model performance and identify top biomarker candidates. Lastly, we explored the progression biomarker potential for each of the 20 top candidates in internal sensitivity analyses. TNFRSF4 and TGF β-1 emerged as the top markers, being lower in fast-progressing patients compared to slow-progressing patients. Proteins of which a low concentration was associated with fast progression were enriched for cell signalling and immune response pathways, which could indicate a lack of a protective response in these individuals. None of our top markers stood out as strong individual predictors of subsequent cognitive decline. This could be explained by small effect sizes per protein and biological heterogeneity among dementia patients. Taken together, this study presents a novel progression biomarker identification framework and protein leads for personalised prediction of cognitive decline in dementia.
... 20,21 Conceptually similar ndings were reported by our group when comparing the ante mortem cerebrospinal uid (CSF) proteome of FTLD-TDP and FTLD-tau individuals with that of NHC, the latter comparison yielding many more differentially regulated proteins than the former. 24 To date all CNS tissue proteomic studies investigated FTLD-TDP cases as one group, so that our ndings cannot be corroborated yet. [25][26][27][28] Several tau-related proteins were dysregulated in the FTLD-tau dataset compared to NHC, including PTBP1, a splicing regulator which represses the splicing of MAPT exon 10. 29 This is relevant to FTLD-tau pathology as alternative splicing of exon 10 results in tau isoforms containing either three or four microtubule-binding repeats (3R-tau and 4R-tau, respectively). ...
Preprint
Full-text available
Background Frontotemporal lobar degeneration (FTLD) is characterized pathologically by neuronal and glial inclusions of hyperphosphorylated tau or by neuronal cytoplasmic inclusions of TDP43. This study aimed at deciphering the molecular mechanisms leading to these distinct pathological subtypes. Methods To this end, we performed an unbiased mass spectrometry-based proteomic and systems-level analysis of middle frontal gyrus cortices of FTLD-tau (n = 6), FTLD-TDP (n = 15), and control patients (n = 5). We validated these results in an independent patient cohort (total n = 24). Results The middle frontal gyrus cortex proteome was most significantly altered in FTLD-tau compared to controls (294 differentially expressed proteins at FDR = 0.05). The proteomic modifications in FTLD-TDP were more heterogeneous (49 differentially expressed proteins at FDR = 0.1). Weighted co-expression network analysis revealed 17 modules of co-regulated proteins, 13 of which were dysregulated in FTLD-tau. These modules included proteins associated with oxidative phosphorylation, scavenger mechanisms, chromatin regulation and clathrin-mediated transport in both the frontal and temporal cortex of FTLD-tau. The most strongly dysregulated subnetworks identified Cyclin-Dependent Kinase 5 (CDK5) and Polypyrimidine Tract Binding Protein 1 (PTBP1) as key players in the disease process. Dysregulation of 9 of these modules was confirmed in independent validation datasets of FLTD-tau and control temporal and frontal cortex (total n = 24). Dysregulated modules were primarily associated with changes in astrocyte and endothelial cell protein expression levels, indicating pathological changes in FTD are not limited to neurons. Conclusions Using this innovative workflow and zooming in on the most strongly dysregulated proteins of the identified modules, we were able to identify disease-associated mechanisms in FTLD-tau with high potential as biomarkers and/or therapeutic targets.
... First, some of the CSF proteins measured and known to be changed in AD (for example, NPTXR 39 or CHI3L1 (ref. 54 )) were not dysregulated in this study. This indicates that our findings are to some extent dependent on the technology used. ...
Article
Full-text available
Development of disease-modifying therapies against Alzheimer’s disease (AD) requires biomarkers reflecting the diverse pathological pathways specific for AD. We measured 665 proteins in 797 cerebrospinal fluid (CSF) samples from patients with mild cognitive impairment with abnormal amyloid (MCI(Aβ+): n = 50), AD-dementia (n = 230), non-AD dementias (n = 322) and cognitively unimpaired controls (n = 195) using proximity ligation-based immunoassays. Here we identified >100 CSF proteins dysregulated in MCI(Aβ+) or AD compared to controls or non-AD dementias. Proteins dysregulated in MCI(Aβ+) were primarily related to protein catabolism, energy metabolism and oxidative stress, whereas those specifically dysregulated in AD dementia were related to cell remodeling, vascular function and immune system. Classification modeling unveiled biomarker panels discriminating clinical groups with high accuracies (area under the curve (AUC): 0.85–0.99), which were translated into custom multiplex assays and validated in external and independent cohorts (AUC: 0.8–0.99). Overall, this study provides novel pathophysiological leads delineating the multifactorial nature of AD and potential biomarker tools for diagnostic settings or clinical trials.
... We observed that CSF YKL-40 levels were associated with tau but not Aβ pathology, indicating that YKL-40 levels in the CSF reflect an astrocyte response to tau tangles deposition in AD. In vivo studies suggest that CSF YKL-40 levels are elevated in AD [12,56] and other tauopathies [18,57,58], as well as correlate with CSF tau levels [17][18][19][20]. Accordingly, recent post-mortem studies reported astrocyte overexpression of YKL-40 in AD and non-AD Fig. 2 Sensitivity analyses testing the associations of Aβ-PET and tau-PET with reactive astrocyte biomarkers using plasma GFAP instead of CSF GFAP. ...
Article
Full-text available
Astrocytes can adopt multiple molecular phenotypes in the brain of Alzheimer’s disease (AD) patients. Here, we studied the associations of cerebrospinal fluid (CSF) glial fibrillary acidic protein (GFAP) and chitinase-3-like protein 1 (YKL-40) levels with brain amyloid-β (Aβ) and tau pathologies. We assessed 121 individuals across the aging and AD clinical spectrum with positron emission tomography (PET) brain imaging for Aβ ([18F]AZD4694) and tau ([18F]MK-6240), as well as CSF GFAP and YKL-40 measures. We observed that higher CSF GFAP levels were associated with elevated Aβ-PET but not tau-PET load. By contrast, higher CSF YKL-40 levels were associated with elevated tau-PET but not Aβ-PET burden. Structural equation modeling revealed that CSF GFAP and YKL-40 mediate the effects of Aβ and tau, respectively, on hippocampal atrophy, which was further associated with cognitive impairment. Our results suggest the existence of distinct astrocyte biomarker signatures in response to brain Aβ and tau accumulation, which may contribute to our understanding of the complex link between reactive astrogliosis heterogeneity and AD progression.
Article
Full-text available
Background: Neuroinflammation has been shown to be an important pathophysiological disease mechanism in frontotemporal dementia (FTD). This includes activation of microglia, a process that can be measured in life through assaying different glia-derived biomarkers in cerebrospinal fluid. However, only a few studies so far have taken place in FTD, and even fewer focusing on the genetic forms of FTD. Methods: We investigated the cerebrospinal fluid concentrations of TREM2, YKL-40 and chitotriosidase using immunoassays in 183 participants from the Genetic FTD Initiative (GENFI) study: 49 C9orf72 (36 presymptomatic, 13 symptomatic), 49 GRN (37 presymptomatic, 12 symptomatic) and 23 MAPT (16 presymptomatic, 7 symptomatic) mutation carriers and 62 mutation-negative controls. Concentrations were compared between groups using a linear regression model adjusting for age and sex, with 95% bias-corrected bootstrapped confidence intervals. Concentrations in each group were correlated with the Mini-Mental State Examination (MMSE) score using non-parametric partial correlations adjusting for age. Age-adjusted z-scores were also created for the concentration of markers in each participant, investigating how many had a value above the 95th percentile of controls. Results: Only chitotriosidase in symptomatic GRN mutation carriers had a concentration significantly higher than controls. No group had higher TREM2 or YKL-40 concentrations than controls after adjusting for age and sex. There was a significant negative correlation of chitotriosidase concentration with MMSE in presymptomatic GRN mutation carriers. In the symptomatic groups, for TREM2 31% of C9orf72, 25% of GRN, and 14% of MAPT mutation carriers had a concentration above the 95th percentile of controls. For YKL-40 this was 8% C9orf72, 8% GRN and 0% MAPT mutation carriers, whilst for chitotriosidase it was 23% C9orf72, 50% GRN, and 29% MAPT mutation carriers. Conclusions: Although chitotriosidase concentrations in GRN mutation carriers were the only significantly raised glia-derived biomarker as a group, a subset of mutation carriers in all three groups, particularly for chitotriosidase and TREM2, had elevated concentrations. Further work is required to understand the variability in concentrations and the extent of neuroinflammation across the genetic forms of FTD. However, the current findings suggest limited utility of these measures in forthcoming trials.
Article
Full-text available
Background: The differential diagnosis of frontotemporal dementia (FTD) is still a challenging task due to its symptomatic overlap with other neurological diseases and the lack of biofluid-based biomarkers. Objective: To investigate the diagnostic potential of a combination of novel biomarkers in cerebrospinal fluid (CSF) and blood. Methods: We included 135 patients from the Centre for Memory Disturbances, University of Perugia, with the diagnoses FTD (n = 37), mild cognitive impairment due to Alzheimer's disease (MCI-AD, n = 47), Lewy body dementia (PDD/DLB, n = 22), and cognitively unimpaired patients as controls (OND, n = 29). Biomarker levels of neuronal pentraxin-2 (NPTX2), neuronal pentraxin receptor, neurofilament light (NfL) and glial fibrillary acidic protein (GFAP) were measured in CSF, as well as NfL and GFAP in serum. We assessed biomarker differences by analysis of covariance and generalized linear models (GLM). We performed receiver operating characteristics analyses and Spearman correlation to determine biomarker associations. Results: CSF NPTX2 and serum GFAP levels varied most between diagnostic groups. The combination of CSF NPTX2, serum NfL and serum GFAP differentiated FTD from the other groups with good accuracy FTD versus MCI-AD: area under the curve (AUC [95% CI] = 0.89 [0.81-0.96]; FTD versus PDD/DLB: AUC = 0.82 [0.71-0.93]; FTD versus OND: AUC = 0.80 [0.70-0.91]). CSF NPTX2 and serum GFAP correlated positively only in PDD/DLB (ρ= 0.56, p < 0.05). NPTX2 and serum NfL did not correlate in any of the diagnostic groups. Serum GFAP and serum NfL correlated positively in all groups (ρ= 0.47-0.74, p < 0.05). Conclusion: We show the combined potential of CSF NPTX2, serum NfL, and serum GFAP to differentiate FTD from other neurodegenerative disorders.
Article
Full-text available
Frontotemporal lobar degeneration (FTLD) is the umbrella term encompassing a heterogeneous group of pathological disorders. With recent discoveries, the FTLDs have been show to classify nicely into three main groups based on the major protein deposited in the brain: FTLD-tau, FTLD-TDP and FTLD-FUS. These pathological groups, and their specific pathologies, underlie a number of well-defined clinical syndromes, including three frontotemporal dementia (FTD) variants [behavioral variant frontotemporal dementia (bvFTD), progressive non-fluent aphasia, and semantic dementia (SD)], progressive supranuclear palsy syndrome (PSPS) and corticobasal syndrome (CBS). Understanding the neuropathological background of the phenotypic variability in FTD, PSPS and CBS requires large clinico-pathological studies. We review current knowledge on the relationship between the FTLD pathologies and clinical syndromes, and pool data from a number of large clinico-pathological studies that collectively provide data on 544 cases. Strong relationships were identified as follows: FTD with motor neuron disease and FTLD-TDP; SD and FTLD-TDP; PSPS and FTLD-tau; and CBS and FTLD-tau. However , the relationship between some of these clinical diagnoses and specific pathologies is not so clear cut. In addition, the clinical diagnosis of bvFTD does not have a strong relationship to any FTLD subtype or specific pathology and therefore remains a diagnostic challenge. Some evidence suggests improved clinicopathological association of bvFTD by further refining clinical characteristics. Unlike FTLD-tau and FTLD-TDP, FTLD-FUS has been less well characterized, with only 69 cases reported. However, there appears to be some associations between clinical phenotypes and FTLD-FUS pathologies. Clinical diagnosis is therefore promising in predicting molecular pathology.
Article
Full-text available
Biochemical markers have a central position in the diagnosis and management of patients in clinical medicine, and also in clinical research and drug development, also for brain disorders, such as Alzheimer's disease. The enzyme-linked immunosorbent assay (ELISA) is frequently used for measurement of low-abundance biomarkers. However, the quality of ELISA methods varies, which may introduce both systematic and random errors. This urges the need for more rigorous control of assay performance, regardless of its use in a research setting, in clinical routine, or drug development. The aim of a method validation is to present objective evidence that a method fulfills the requirements for its intended use. Although much has been published on which parameters to investigate in a method validation, less is available on a detailed level on how to perform the corresponding experiments. To remedy this, standard operating procedures (SOPs) with step-by-step instructions for a number of different validation parameters is included in the present work together with a validation report template, which allow for a well-ordered presentation of the results. Even though the SOPs were developed with the intended use for immunochemical methods and to be used for multicenter evaluations, most of them are generic and can be used for other technologies as well.
Article
Full-text available
Aggressive meningiomas exhibit high levels of recurrence, morbidity and mortality. When surgical and radiation options are exhausted, there is need for novel molecularly-targeted therapies. We have recently identified NDRG4 overexpression in aggressive meningiomas. NDRG4 is a member of the N-Myc Downstream Regulated Gene (NDRG) family of the alpha/beta hydrolase superfamily. We have demonstrated that NDRG4 downregulation results in decreased cell proliferation, migration and invasion. In follow up to our prior studies; here we demonstrate that the predominant form of cell death following NDRG4 silencing is apoptosis, utilizing Annexin-V flow cytometry assay. We show that apoptosis caused by p53 upregulation, phosphorylation at Ser15, BAX activation, Bcl-2 and BcL-xL downregulation, mitochondrial cytochrome c release and execution of caspases following NDRG4 depletion. Sub-cellular distribution of BAX and cytochrome c indicated mitochondrial-mediated apoptosis. In addition, we carried out the fluorescence cytochemical analysis to confirm mitochondrial-mediated apoptosis by changes in mitochondrial membrane potential (Ψm), using JC-1 dye. Immunoprecipitation and immunofluorescence confirmed binding of NDRG4 to p53. In addition, we demonstrate that apoptosis is mitochondrial and p53 dependent. The proapoptotic effect of p53 was verified by the results in which a small molecule compound PFT-α, an inhibitor of p53 phosphorylation, is greatly protected against targeting NDRG4 induced apoptosis. These findings bring novel insight to the roles of NDRG4 in meningioma progression. A better understanding of this pathway and its role in meningioma carcinogenesis and cell biology is promising for the development of novel therapeutic targets for the management of aggressive meningiomas.
Article
Based on the recent literature and collective experience, an international consortium developed revised guidelines for the diagnosis of behavioural variant frontotemporal dementia. The validation process retrospectively reviewed clinical records and compared the sensitivity of proposed and earlier criteria in a multi-site sample of patients with pathologically verified frontotemporal lobar degeneration. According to the revised criteria, 'possible' behavioural variant frontotemporal dementia requires three of six clinically discriminating features (disinhibition, apathy/inertia, loss of sympathy/empathy, perseverative/ compulsive behaviours, hyperorality and dysexecutive neuropsychological profile). 'Probable' behavioural variant frontotemporal dementia adds functional disability and characteristic neuroimaging, while behavioural variant frontotemporal dementia 'with definite frontotemporal lobar degeneration' requires histopathological confirmation or a pathogenic mutation. Sixteen brain banks contributed cases meeting histopathological criteria for frontotemporal lobar degeneration and a clinical diagnosis of behavioural variant frontotemporal dementia, Alzheimer's disease, dementia with Lewy bodies or vascular dementia at presentation. Cases with predominant primary progressive aphasia or extra-pyramidal syndromes were excluded. In these autopsy-confirmed cases, an experienced neurologist or psychiatrist ascertained clinical features necessary for making a diagnosis according to previous and proposed criteria at presentation. Of 137 cases where features were available for both proposed and previously established criteria, 118 (86%) met 'possible' criteria, and 104 (76%) met criteria for 'probable' behavioural variant frontotemporal dementia. In contrast, 72 cases (53%) met previously established criteria for the syndrome (P 5 0.001 for comparison with 'possible' and 'probable' criteria). Patients who failed to meet revised criteria were significantly older and most had atypical presentations with marked memory impairment. In conclusion, the revised criteria for behavioural variant fronto-temporal dementia improve diagnostic accuracy compared with previously established criteria in a sample with known fronto-temporal lobar degeneration. Greater sensitivity of the proposed criteria may reflect the optimized diagnostic features, less restrictive exclusion features and a flexible structure that accommodates different initial clinical presentations. Future studies will be needed to establish the reliability and specificity of these revised diagnostic guidelines.
Article
Alzheimer's disease is the most common form of dementia in the elderly. Owing to a lack of accurate easily determined clinical criteria, neuropathological findings remain decisive for establishing the diagnosis. However, full utilization of all the presently available methods, such as history taking, physical examination, neurological and mental status, psychological testing, laboratory examinations, additional procedures involving the use of diagnostic equipment, can result in a diagnostic accuracy of about 90%. The very wide spectrum of possible differential diagnostic possibilities can be greatly reduced by the judicious use of a few selective examinations. Here, the possibilities available in the doctor's office are better than ever before. Of decisive importance for the patient is to consider the possibility of dementia, and to exclude treatable causes as soon as possible.
Article
Introduction: Lumbar puncture (LP) is increasingly performed in memory clinics. We investigated patient-acceptance of LP, incidence of and risk factors for post-LP complications in memory clinic populations. Methods: We prospectively enrolled 3868 patients (50% women, age 66 ± 11 years, mini mental state examination 25 ± 5) at 23 memory clinics. We used logistic regression analysis using generalized estimated equations to investigate risk factors for post-LP complications, such as typical postlumbar puncture headache (PLPH) and back pain. Results: A total of 1065 patients (31%) reported post-LP complaints; 589 patients (17%) reported back pain, 649 (19%) headache, of which 296 (9%) reported typical PLPH. Only few patients needed medical intervention: 11 (0.3%) received a blood patch, 23 (0.7%) were hospitalized. The most important risk factor for PLPH was medical history of headache. An atraumatic needle and age >65 years were preventive. Gender, rest after LP, or volume of cerebrospinal fluid had no effect. Conclusions: The overall risk of complications is relatively low. If risk factors shown in this study are taken into account, LPs can be safely performed in memory clinics.