Você está na página 1de 10

J Neuropathol Exp Neurol Vol. 67, No.

10
Copyright Ó 2008 by the American Association of Neuropathologists, Inc. October 2008
pp. 923Y932

REVIEW ARTICLE

Proteomics of Human Neurodegenerative Diseases


Jing Zhang, MD, PhD, C. Dirk Keene, MD, PhD, Catherine Pan,
Kathleen S. Montine, PhD, and Thomas J. Montine, MD, PhD

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


(AD) and Parkinson disease (PD) pose serious public health
Abstract challenges that will increase in the coming decades. Many
The technology, experimental approaches, and bioinformatics that discoveries have been made in the genetic causes and risk
support proteomic research are evolving rapidly. The application of factors for several neurodegenerative diseases (4); however,
these new capabilities to the study of neurodegenerative diseases is there are relatively few proteomic studies of neurodegener-
providing insight into the biochemical pathogenesis of neurodegen- ative diseases to date. The major foci of these investigations
eration as well as fueling major efforts in biomarker discovery. have been analysis of protein from autopsy samples in studies
Here, we review the fundamentals of commonly used proteomic ap- of pathogenesis and from cerebrospinal fluid (CSF) in the
proaches and the outcomes of these investigations with autopsy and pursuit of biomarkers. It is important to be aware of some
cerebrospinal fluid samples from patients with neurodegenerative limitations of these initial proteomic studies of human neu-
diseases. rodegenerative diseases. Many are limited to comparisons
between 1 diseased group and controls and therefore cannot
Key Words: Alzheimer disease, Cerebrospinal fluid, Mass spec-
address the important issue of disease specificity. This lim-
trometry, Neurodegeneration, Parkinson disease, Proteomics.
itation has been approached by some more recent inves-
tigations reviewed later. Another limitation has been a focus
on changes observed during clinically diagnosed stages of
INTRODUCTION disease, whereas some recent studies have begun to explore
Proteomic technologies provide powerful means to sys- alterations that accompany preclinical disease stages.
tematically profile the peptide or protein constituents of com-
plex mixtures and, in some instances, to use these data to
impute protein identity, amount, or both. Although comple- FUNDAMENTALS OF PROTEOMICS
mentary to genomics, there are important differences between
Shared Features of Different Proteomic
gene expression profiling and protein profiling. Perhaps most
important is that it is now possible to interrogate essentially
Techniques
the entire transcriptome, athough there is no comparably com- Although it is difficult to summarize such a dynamic and
prehensive approach to the proteins encoded therein. This may rapidly changing scientific area, the various approaches to
be viewed by some as sufficient justification to concentrate protein profiling share common features. These include
on the transcriptome; however, the quantitative concordance sample preparation, protein or peptide separation, mass spec-
between message and corresponding protein is surprisingly trometry (MS), and bioinformatic data processing, as well as
poor (1Y3). Moreover, the complex and rich range of chem- subsequent confirmation and validation. For the sake of clar-
ical modifications that occur to proteins after translation are ity, we will use validation to mean replication of findings by
largely transparent to genomics but are coming into pro- alternative method(s) in the same samples used in profiling
gressively greater focus in proteomics. Another important and confirmation to mean replication of findings in inde-
consideration in the context of this review is the relative sta- pendent samples.
bility of protein versus messenger RNA in samples of brain Sample preparation is a major source of variation in the
obtained from autopsy. outcomes of proteomic experiments. Again, proteomics dif-
Human neurodegenerative diseases range from rare to fers from genomics in that it is not yet possible to analyze
common illnesses. Indeed, some such as Alzheimer disease virtually all proteins in a cell, tissue, or body fluid compre-
hensively, especially in samples with wide ranges of protein
concentration. Consequently, the first step in all proteomic ex-
From the Department of Pathology, University of Washington, Seattle,
Washington.
periments is preparation of a subproteome, or a subset of pro-
Send correspondence and reprint requests to: Thomas J. Montine, MD, PhD, teins, that is defined by the physical or chemical means used
Department of Pathology, University of Washington, Harborview to isolate it from all proteins in tissue or body fluid. Examples
Medical Center, Box 359791, Seattle, WA 98104; E-mail: tmontine@ to follow have generated subproteomes using gradient de-
u.washington.edu tergent extractions to enrich for hydrophobic or membrane-
This work was supported by Grants AG05136 and AG029808 from the
National Institutes of Health, by the Michael J. Fox Foundation for bound proteins, affinity purification, immunoprecipitation,
Parkinson’s Research, by the C.-M. Shaw endowment, and by the Nancy and laser capture microdissection (Fig. 1). Typical sub-
and Buster Alvord Endowment. proteomes are still complex protein mixtures that require

J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008 923

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Zhang et al J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008

the signal that is typically plotted as the total ion chromato-


gram for the first MS dimension, with the x axis being time
and the y axis being relative abundance, and mass-to-change
ratio (m/z) chromatogram for the daughter ions.
The next step is synthesis of the wealth of mass spec-
tral data obtained. A major step forward in the evolution of
proteomics was the development of bioinformatic tools to
interpret the ever increasing rich data sets generated from
advancing technology. Here, the pivotal event was the devel-
opment of search engines, such as SEQUEST and MASCOT,
which correlate actual tandem mass spectra with predicted
tandem mass spectra based on species-specific amino acid

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


sequences from protein or genomic databases (5, 6). It is
important to appreciate that these different programs do not
perform in exactly the same way and can produce different
peptide matches and levels of confidence. Moreover, the da-
tabases currently searched are incomplete and imperfect. It is
for this reason that all results require validation.
FIGURE 1. Diagram of the steps in typical proteomic experi-
ments that use liquid chromatography (LC) or 2-dimensional Specifics of Different Proteomic Techniques
gel electrophoresis (2-DE) followed by tandem mass spectrom- Classically, experiments isolate a subproteome of in-
etry (MS-MS). Protein identification (ID) is imputed by bioin- terest, separate the protein by 2-DE, excise the spot of interest
formatic tools that compare tandem MS data with protein or from the stained gel, digest the proteins contained in each
genomic databases. spot with trypsin, and then perform tandem MS with MALDI
TOF-TOF. This powerful approach is still extensively used,
although 2-DE is labor intensive and has a relatively low dy-
further separation. Classically, proteins are separated by 2- namic range. Some of these issues have been alleviated
dimensional gel electrophoresis (2-DE), and spots detected partially by advances in technology, especially the use of fluo-
on the gel by a variety of means are excised and digested rescent dyes. Proteins in a control sample can be labeled with
to peptides, typically with trypsin. Alternatively, the protein 1 fluorescent dye and proteins in an experimental sample la-
mixture in the subproteome is first digested to peptides, and beled with a different fluorescent dye. The samples are then
these are then separated by single or multidimensional liquid mixed, separated by 2-DE, and the relative fluorescent inten-
chromatography (LC). sities are measured within the spot of interest before digestion
Mass spectrometry is an elegant technique for detecting and tandem MS. In this way, analysis of fluorescent intensity
the mass of charged species. There are many excellent reviews provides relative quantification, and tandem MS provides
devoted to MS. Here we summarize the basics. The first step in protein identification for the spot of interest in the gel.
MS is to ionize and vaporize the analytes (i.e. proteins or their One response to the time and labor devoted to spot
[typically] tryptic peptide fragments) without significant frag- picking was to develop multidimensional LC systems for the
mentation. This can be achieved by a variety of methods: 2 separation of peptides that are coupled to tandem MS. In a
commonly used approaches are matrix-assisted laser desorp- typical experiment, the subproteome is first digested to pep-
tion ionization (MALDI) and electrospray ionization (ESI). tides, and these are separated by LC and then directly ionized
Matrix-assisted laser desorption ionization mixes the analyte by ESI followed by tandem MS. Alternatively, the separated
with 1 of several different molecules that facilitate ionization peptides are spotted directly onto a MALDI plate followed by
and vaporization. Generating peptide ions from ESI is com- tandem MS. An advantage of these approaches is that they
plex and is thought to involve a process called Coulombic can be much more automated than the 2-DE method; how-
fission. The peptide or protein ions are then introduced into ever, this comes with a price. The major one is that without
the mass analyzer. Again, there are different types including the gel and fluorescent dyes, there are no quantitative data.
ion trap, triple quadrupole, time-of flight (TOF), and Fourier Indeed, it is critical to appreciate that the size of the resulting
transform ion cyclotron that differ in their mechanisms of ion peptide ion peaks does not necessarily bear any quantitative
separation, mass accuracy, and resolution. It is critical to ap- resemblance to the amount of protein in the subproteome; the
preciate that some, but not all, common proteomic techniques major sources of variance are ionization efficiency and de-
use tandem MS (MS-MS) to analyze the daughter or fragment tector sensitivity. Several groups have ingeniously addressed
ions of the parent ions whose mass was determined in first MS this shortcoming of high-throughput proteomics by adapting
dimension. In these collision-induced dissociation tandem MS a classical MS technique: the stable isotope dilution assay. In
experiments, the energy applied greatly favors fragmentation general, stable isotope labeling coupled with MS is more
of the peptide (amide) bond over amino acid side chains in the quantitatively accurate than are comparisons of spot densities
parent ion, thereby generating an ensemble of daughter ions, on 2-DE. All of these techniques require the introduction of
called b ions and y ions, from which the amino acid sequence a stable isotope label, usually deuterium or 13C into samples
can be deduced (Fig. 2). Detectors and multipliers generate that are then mixed and analyzed simultaneously by MS. The

924 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008 Proteomics in Neurodegeneration

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


FIGURE 2. Typical collision-induced dissociation tandem mass spectrometry data that show total ion chromatogram (time vs
relative abundance) in the upper panel. The selected ion marked by the red line in the total ion chromatogram is subjected to
further fragmentation to yield characteristic daughter ions in the lower mass-to-change ratio (m/z) chromatogram.

isotopically labeled peptide serves as a mass signature for tially bind proteins based on hydrophobicity, charge interac-
sample source and as the foundation for estimation of relative tions, chelation, and other characteristics. In addition, SELDI
abundance, and in some instances, the absolute concentration plates can be activated with immobilized organics that can
of the ionized peptides. There are several approaches to the covalently bind antibodies, other proteins such as receptors,
introduction of isotope labels to proteins or peptides; among and even nucleic acids. After the subproteome has been in-
these are isotope-coded affinity tags (7) and isobaric tags for cubated with the desired plate, the poorly bound proteins or
relative and absolute quantification (iTRAQ) (8). Isotope- peptides are washed away, thereby acting like affinity chro-
coded affinity tag was one of the earlier approaches and was matography. The versatility of agents that can be used at this
limited to labeling cysteinyl thiolates with light (protonated) step is a clear strength of SELDI. Another advantage is that
or heavy (deuterated) probes (Fig. 3). Isobaric tags for rela- SELDI is relatively simple and quick compared with other
tive and absolute quantification now permits differential iso- proteomic techniques. A disadvantage of SELDI as it is com-
topic labeling of ?-amino lysyl groups with up to 8 different monly used is that only a single MS dimension is used and
probes. A bioinformatics approach similar to that previously there is, therefore, no attempt to impute protein identity.
described is used for imputation of protein identity from Rather, the typical experiment deals with finding those ions
peptide ion data. Specialized computer software is needed that have optimal characteristics and then using other tech-
to integrate isotopic data from multiple peptides to estimate niques to identify the protein from which the selected ions
protein concentrations. Newer methods for quantification are derived. Finally, given the nature of the SELDI experi-
without isotope labels are an area of active investigation (9). ment, it typically is not coupled with stable-isotope dilution
A third approach used in proteomics of neurodegener- techniques; therefore, reproducibility and quantification are
ative diseases is surface-enhanced laser desorption/ionization problematic (11).
(SELDI), also called SELDI-TOF. Surface-enhanced laser de- These are not the only approaches to MS. One example,
sorption/ionization is a variant of MALDI that is commonly an elegant technique of particular interest to pathologists, is
used in biomarker discovery. There are few very attractive tissue-based MS that can profile proteins in situ from different
features of SELDI (10). First, proteins or peptides can be regions within a tissue specimen (12). The methods previ-
separated or enriched through a variety of plates or chips with ously described, however, are those that have been used to
immobilized biochemically active materials that preferen- date to investigate human neurodegenerative diseases.

Ó 2008 American Association of Neuropathologists, Inc. 925

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Zhang et al J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


FIGURE 3. Two subproteomes were generated from human CSF: one was labeled with protonated isotope-coded affinity tags
(ICAT) reagent and the other with deuterated ICAT reagent. They were then combined before tandem mass spectrometry. The
mass-to-change ratio (m/z) chromatogram shows the relative abundance of ions conjugated to the light (protonated) versus
heavy (deuterated) ICAT reagent.

Identifying Protein Modifications traction protocol, another study identified 15 proteins, the
One approach to evaluating protein modifications is in levels of which differed in different AD brain regions (16).
preparation of the subproteome. For example, some investi- Again, no validation was performed. Agreement between
gators have focused on only those spots from 2-DE that have these 2 similar studies is low; only increased levels of glyc-
reactivity for protein carbonyls (vide infra). Alternatively, an eraldehyde 3-phosphate dehydrogenase (GAPDH) and syn-
antibody that recognizes only a certain posttranslational mod- aptotagmin I in AD were confirmed. A third similar study
ification may be adhered to a SELDI plate. Another approach identified significantly decreased levels of mitochondrial com-
has been to analyze tandem MS data for apparent mass shifts plex III core protein 1 (17). This finding was not validated. A
not predicted by the peptide sequence. There are several com- fourth study of frontal cortex focused on 9 antioxidant pro-
puter programs that perform such searches. Two contrasting teins and identified significantly increased peroxiredoxin 2
approaches are SEQUEST and MASCOT versus P-MOD and from AD patients compared with controls; parallel studies
InsPecT. The SEQUEST and MASCOT search for specified showed that peroxiredoxin 3 levels were decreased in frontal
mass shifts on specified amino acids, for example +16 on M cortex from Down syndrome and Pick disease but not AD
for methionine sulfoxide, across all peptide ions detected. In patients (18).
contrast, P-MOD and InsPectT search the same spectral data
in a different manner (13, 14). Here, one provides the pro- Laser Capture Microdissection of
grams with the amino acid sequence of a protein of interest, Hallmark Structures
and they list all unexpected masses and a calculated level of There have been 3 reports of proteomic characteri-
confidence. zation of hallmark structures of neurodegenerative diseases
(i.e. amyloid plaques and neurofibrillary tangles [NFTs] from
AD and Lewy bodies [LB] from patients with dementia with
PROTEOMIC STUDIES OF LBs [DLB]) that were enriched by laser capture microdis-
NEURODEGENERATIVE DISEASES FROM section from human brain samples. Amyloid plaques were
AUTOPSY BRAIN localized by thioflavin-S staining, microdissected, and then
In this section, we review proteomic studies of autopsy extracted with detergent; NFTs and LBs were identified by
samples from patients with neurodegenerative diseases. Al- immunohistochemistry (tau-2 for NFTs, >-synuclein [SNCA]
though there are several excellent proteomic investigations of for LBs) and solubilized with formic acid; all were then
corresponding animal models, we limited our comments to separated with LC followed by MS-MS. A total of 488 pro-
investigations of human tissue and grouped them by the teins were identified from laser capture microdissection of
method of subproteome preparation. amyloid plaques; several of these were validated by immu-
nohistochemistry. Of these, 26 proteins were enriched in
Homogenates of Human Brain Regions captured amyloid plaques compared with areas without am-
Four studies have used 2-DE followed by tandem MS yloid. Of particular interest was the identification of dynein
to investigate detergent-extractable proteins in brain regions heavy chain in both human postmortem amyloid plaques
of patients with AD, and in 1 case, patients with Down syn- as well as in plaques obtained from APPswe/PS1$E9 trans-
drome. One study identified 37 proteins with altered levels in genic mice (19).
AD (15); 11 of these had increased levels in AD and were Seventy-two proteins were identified in NFTs obtained
identified by investigators by tandem MS. No validation was by laser capture microdissection by 2 or more unique pep-
performed. Using a similar approach but not the same ex- tides, of which 63 had no previously known association with

926 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008 Proteomics in Neurodegeneration

NFTs (20). One of these proteins, GAPDH, localized to Chamorros from Guam who had Parkinson-dementia complex
NFTs by immunohistochemistry, was validated in the or their controls (Fig. 4). In addition to the expected increase
detergent-insoluble fraction from hippocampus of patients in abnormal frontal cortical AA peptides, tau, ubiquitin, and
with AD and was confirmed by coimmunoprecipitation with apolipoprotein E (apoE) in AD, and tau in Parkinson-
PHF-tau. Coincident with this publication was the observa- dementia complex, we identified SNCA as a major detergent-
tion by others that polymorphisms of GAPD may be genetic insoluble protein in Parkinson-dementia complex but not AD
risk factors for late-onset AD (21). This finding has been cor- despite a lack of LBs in corresponding tissue samples (34).
roborated by others, although the overall effect may be small The proteomic findings were confirmed by ELISA in frontal
(22). Others have subsequently shown that GAPDH localizes and temporal cortices in which SNCA levels in Parkinson-
to LBs (23), can be oxidized and aggregated by exposure to dementia complex patient samples were quantitatively similar
AA peptides (24), and has increased expression in the hip- to patients with DLB and echo earlier findings of abnormal
pocampus of AD patients compared with controls (25). SNCA immunohistochemical findings in some patients with

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


Traditional biochemical or histochemical methods Parkinson-dementia complex (35Y38).
previously identified approximately 70 molecular compo-
nents of LBs in both the brainstem and cerebral cortex (26);
SNCA has been identified as one of the major constituents. A Modified Proteins
recent study used laser capture microdissection of SNCA- Substantial antibody-based evidence supports accumu-
immunoreactive cerebral cortical LBs from patients with lation of proteins that are pathologically modified by oxi-
DLB coupled with LC/ESI/MS/MS. This platform identified dative or nitrative reactions in several neurodegenerative
156 protein with 2 or more unique peptides; of these, 17 had diseases, including AD. One limitation of these earlier studies
previously been associated with cerebral cortical or brainstem has been the difficulties in identification of the modified pro-
LBs (27). One particular protein, heat shock cognate 71 was teins. Proteomic techniques provide approaches to investigate
previously associated only with brainstem LBs. this facet of AD pathogenesis.
Several elegant studies have combined immuno-
chemical detection of protein carbonyls (a form of protein
Insoluble Proteins in Cerebral Cortex oxidation) in 2-DE followed by tandem MS for protein iden-
Accumulation of insoluble proteins is a defining char- tification from soluble extracts of AD cerebrum. Proteins path-
acteristic of several neurodegenerative diseases, including ologically oxidized in AD include glutamine synthase, creatine
AD and PD (28, 29). Indeed, it was the identification of kinase BB, UCHL1, >-enolase, and DRP-2/CRMP2 (39, 40).
detergent-insoluble protein such as AA and paired helical At least 3 of these oxidized proteins validated earlier results,
filament tau that ushered in the modern era of neurodegener- including those on UCHL1-validated earlier results (41).
ative disease research (30, 31). Conversion to insoluble forms A similar approach coupled immunologic detection of
disrupts protein function, can lead to further cellular damage, nitrotyrosine residues with tandem MS to identify >-enolase,
and may precede the formation of the hallmark inclusions triosephosphate isomerase, and neuropolypeptide h3 as tar-
(32). To gain insight into disease pathogenesis, we have gets for this type of pathological modification in AD cerebral
focused modern proteomic techniques on this class of cortex (40, 42). Moreover, the authors extended their inves-
pathological protein (33). We used LC-MS-MS and identified tigations to autopsy samples from individuals who died with
in the detergent-insoluble fraction of late-onset AD temporal
cortex samples 125 proteins by 2 or more unique peptides
that included several proteins critical to AA production, com-
ponents of synaptic scaffolding, and products of genes linked
to an increased risk of late-onset AD. Each of 15 candidates
were validated by Western blot including GAPDH (33), and
the levels of several are altered in hippocampi of AD patients
compared with controls (25). AA, tau, and 7 of 8 other newly
identified detergent-insoluble proteins were confirmed to be
increased in temporal cortex by Western blot, not only from
patients with late-onset AD but from patients with AD as-
sociated with mutations in PSEN1 and PSEN2. All of these
except tau were elevated in individuals with mild cognitive
impairment, whereas none except AA were elevated in aged
APPswe mice. These results extend the amyloid hypothesis
for AD to include widespread protein insolubility that is not
exclusive to AA, even early in the course of AD before the
onset of dementia.
We have used iTRAQ labeling combined with MALDI-
TOF-TOF to analyze the detergent-insoluble proteome in FIGURE 4. Experimental approach for isobaric tags for
frontal cortex from 4 groups of individuals: patients with late- relative and absolute quantification labeling of 4 samples.
onset AD and their controls from mainland United States and MS, mass spectrometry.

Ó 2008 American Association of Neuropathologists, Inc. 927

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Zhang et al J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008

amnestic mild cognitive impairment (MCI), a clinically de- Huntington disease, and prion diseases, we are unaware of any
fined condition that largely represents prodromal AD. They that has examined human brain or spinal cord from patients
showed that modified proteins in amnestic MCI partially with these diseases.
overlap with those identified in patients with dementia from
AD (43, 44).
One group used affinity-purification with the MC1 PROTEOMIC STUDIES OF HUMAN CSF
monoclonal antibody to isolate a soluble fraction of PHF-tau Cerebrospinal fluid is widely considered an imperfect
from AD brain (45). In addition to identifying a large number but relatively accessible portal into the neurochemical changes
of phosphorylated sites, they found that soluble PHF-tau is that accompany neurological diseases (52Y57). Over the past
ubiquitin conjugated within its microtubule-binding domain few years, several groups have used proteomics to charac-
at residues Lys-254, Lys-311, and Lys-353. Polyubiquitin terize the human CSF proteome and to profile CSF bio-
conjugates formed mostly at Lys-48 of ubiquitin, suggesting markers related to a few major neurodegenerative diseases,

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


a failure of the ubiquitin-proteasome system. such as AD, PD, and ALS (58Y67). Together, these studies
Another study investigated methionine oxidation to its have generated lists of protein candidates associated with
sulfoxide in neuron-specific A-III tubulin and AA peptides each disease that are altered in relative abundance within
(46). We used P-MOD to identify and map posttranslational CSF. Major discrepancies have also been noted among dif-
modifications using tandem MS data from detergent-insoluble ferent groups of investigators that are likely related to several
protein fractions from AD temporal cortex. Our results con- factors especially preparation of the subproteome.
firmed the direct observations of others by identifying me-
thionine 35 sulfoxides in AA peptides and numerous sites of
tau phosphorylation. P-MOD mapped several abundant CSF Sample Collection and Storage
methionine sulfoxides to neuron-enriched A-III tubulin but Human CSF derives from choroid plexus and the
not >-III tubulin, its heterodimeric partner. These findings extracellular fluid of the brain and spinal cord (68); the vol-
point to oxidative modification of A-III tubulin as a potential ume of approximately 140 to 150 mL in an adult is renewed
contributor to the neuronal cytoskeletal disruption in AD. every 6 to 8 hours (69, 70). It has a rostrocaudal protein
concentration gradient that is about 2.5-fold (71). Moreover,
CSF production rate varies throughout the day and is altered
Lewy Body Disorders by aging and some medications (72, 73). Thus, CSF provides
Lewy body disorders are a recent consensus classifica- a temporally limited window into the neurochemical activity
tion of diseases that share the histopathologic hallmark of LB of the CNS that is confounded by regionally varying mixture
formation (47). This includes several neurodegenerative with a transudate of plasma, circadian variation, changes with
diseases, the most common of which are PD and DLB. We age, and alteration by medications. Great care must obviously
previously described the proteomic analysis of laser capture be taken, therefore, to match fractions of CSF obtained from
microdissected LBs from cerebral cortex of patients with individuals for study.
DLB. Basso et al (48) examined protein levels in the sub- In addition to physiological variables, contamination
stantia nigra pars compacta from 4 patients who died of PD with blood during lumbar puncture is another critical factor.
and 4 controls using 2-DE followed by MALDI-TOF-TOF. The CSF protein content is approximately 1/200th that of
They identified 44 proteins in their subproteome extracted blood, yet the protein profile overlaps extensively with blood
with urea and detergent. Nine of these showed abundant (74Y76). As a result, even minute contamination with blood
changes between the 2 groups; several were mitochondrial can have large effects on the concentrations of CSF proteins.
and reduced oxygen species-scavenging proteins. A recent Several methods are available to test for trace contamination
study compared 3 subcellular fractions of frontal cortex from of blood in CSF samples, such as red blood cell density and
4 groups: control individuals and patients with PD who had measuring concentrations of exclusive blood proteins in CSF;
brainstem, limbic, or frontal cortex LB formation (49). Each one such protein is apoB (77). Because it is extremely dif-
sample was labeled with specific iTRAQ reagent before anal- ficult to obtain CSF samples without some blood contami-
ysis by MALDI TOF/TOF. A total of 1,864 nonredundant nation, we consider CSF with more than 10 red blood cells
proteins were identified; 199 of these displayed significant per microliter or an apoB serum: CSF concentration ratio less
changes in relative abundance between PD groups and con- than 6,000 unacceptably contaminated by blood (58, 61, 77).
trol. One particular mitochondrial protein, mortalin, was fur- Using relative apoB concentration is particularly helpful with
ther validated by Western blot and found to be decreased samples that have been previously sedimented.
in all PD groups as well as in a cellular model of PD (50). Although a seemingly trivial and straightforward pro-
Mortalin seems to have several activities, including molec- cedure, CSF sample storage can significantly influence
ular chaperoning, mitochondrial import, and energy gener- sample integrity. Protein degradation can occur, particularly
ation, and response to oxidative stress (51). when samples are kept at j20-C or have undergone multiple
freeze-thaw cycles; this may be especially problematic for
cystatin C (52, 78). Another unsettled issue related to CSF
Other Neurodegenerative Diseases storage is the addition of protease inhibitors (79, 80). Cur-
Although there are several excellent studies of murine rently, no recommendation has been made on this issue by
and cellular models of amyotrophic lateral sclerosis (ALS), the Human Proteome Organization.

928 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008 Proteomics in Neurodegeneration

Characterization of CSF Proteome protien VGF, transthyretin (TTR), conjugated TTR, cystatin
Several laboratories are using tandem MS to define the C, and a fragment of chromogranin B (95). Another group
human CSF proteome. Early experiments used CSF and 2-DE also has noted the potential for the relative concentrations of
and identified a few hundred proteins (81Y84). The number conjugated TTR in CSF to discriminate between AD and
of identified CSF proteins has increased exponentially since controls (96).
the application of LC-based proteomics (58, 61Y63, 85Y87).
Indeed, with improvements in chromatography and MS, the Two-Dimensional Gel Electrophoresis
human CSF proteome has been expanded to more than 2,000 Tandem MS
proteins (88). Five major studies have used 2-DE coupled with tan-
dem MS in the search for CSF biomarkers for AD. Castano
CSF Proteins in Neurodegenerative Diseases et al (97) examined pooled CSF samples obtained from neu-
Characterization of the human CSF proteome is 1 step ropathologically confirmed AD and nondemented control

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


toward identifying protein profiles that can aide in diagnosis subjects (n = 43 for both groups) and identified 5 differ-
and monitoring progression of neurodegenerative diseases. entially expressed proteins in AD versus controls. Davidsson
Several groups are pursuing this goal using a variety of et al (59) identified 6 proteins and their isoforms that were
platforms. significantly altered in CSF of AD (n = 15) patients versus
controls (n = 12). Hu et al (65) discovered 11 proteins or
Surface-Enhanced Laser Desorption/Ionization isomers that are altered significantly in AD patients (n = 2)
Several SELDI-based studies have focused on AD. versus controls (n = 4). Finehout et al (64) reported a panel of
Carrette and colleagues (52) have used SELDI to com- 23 spots (21 that have been identified) that could be used to
pare AD patients with controls and have identified several differentiate AD (n = 34) from non-AD (n = 9 normal
unique ion peaks that can differentiate disease from con- controls and n = 24 other diseases) with high sensitivity and
trols groups. Several of these putative AD biomarkers have specificity. Puchades et al (60) compared samples from AD
been subsequently purified and identified as cystatin C, A2- patients and normal control subjects (n = 7 for both groups)
microglobulin isoforms, to name a few. Notably, however, revealing 9 proteins to be significantly altered in AD. Only
cystatin C is not only prone to freeze/thawing artifacts (78), 10 proteins were identified in 2 or more of these studies.
but also seems to be nonspecific to AD as it also is altered in These include albumin precursor, apoA1, apoE, >-1A gly-
patients with Creutzfeldt-Jakob disease (89), multiple scle- coprotein, >-1 antitrypsin (or precursor), A-2 microglobulin,
rosis (90), and chronic pain (91). One study used SELDI to complement component 3, prostaglandin D synthase, retinal
investigate CSF from patients with AD or frontotemporal binding protein, and TTR.
dementia versus controls; some peaks were discriminating,
but no proteins were identified (92). Another study used Liquid Chromatography-Tandem MS
SELDI analysis of CSF from 113 patients with MCI who A third approach is multidimensional LC followed by
were followed for up to 6 years and 28 controls who were tandem MS. One group used this technology coupled with
followed for 3 years after lumbar puncture (93). These in- isotope-coded affinity tags labeling to investigate specific
vestigators discovered a panel of 17 candidate biomarkers changes in the proteome associated with aging (61) or AD
that could distinguish between patients with stable MCI and (62) and identified approximately 400 CSF proteins, many
patients with MCI who progressed to AD; however, the iden- of which had altered relative abundance in AD. The same
tity of most of these proteins is not yet known. groups subsequently used iTRAQ labeling to compare the
Two studies have reported SELDI-based identification CSF proteome of patients with AD, PD, DLB, and age-
of CSF biomarker candidates for ALS. One study identified matched controls. More than 1,500 CSF proteins were iden-
30 ion peaks with statistically significant differences between tified; of those, 136, 72, and 101 proteins seemed to be
controls and patients with ALS. Transthyretin and cystatin uniquely associated with AD, PD, and DLB, respectively
C were identified and validated to be decreased in ALS CSF, (58). Although these studies validated several of their
whereas the carboxy-terminal fragment of the neuroendo- findings, only a very small fraction of candidate biomarkers
crine protein 7B2 was increased in CSF from ALS patients has been confirmed across platforms, underscoring the need
(94). In another study, 3 candidates were identified by SELDI for testing the use of proteomics-discovered candidates by al-
as being significantly lower in CSF from patients with ALS ternative means in large-scale clinical studies.
(n = 36) compared with controls (n = 21). These findings were
validated in a separate set of samples, and 2 candidates were Multianalyte Profile
identified as cystatin C and a peptic fragment of neuro- We extended our iTRAQ proteomics discoveries by
secretory protien VGF (66). developing a multianalyte profile (MAP) for the 8 best-
We are aware of 1 study that used SELDI to investigate performing biomarker candidates and then conducted an in-
CSF from patients with frontotemporal dementia (n = 16) dependent clinical study using CSF from 95 control subjects,
versus controls (n = 12). Ten ion peaks with good signal- 48 patients with probable AD, and 40 patients with probable
to-noise ratio and resolution were significantly differentially PD. Our 8-member MAP agreed with expert diagnosis for
expressed in frontotemporal dementia; 5 were increased 95% of controls, 95% of probable PD, and 75% of probable
and 5 were decreased. Five of the ion peaks were purified AD. This MAP (in decreasing order of contribution to clas-
and identified by tandem MS as a fragment of neurosecretory sification) consisted of tau, brain-derived neurotrophic factor,

Ó 2008 American Association of Neuropathologists, Inc. 929

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Zhang et al J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008

interleukin 8, AA42, A2-microglobulin, vitamin D-binding pro- 12. Cornett DS, Reyzer ML, Chaurand P, et al. MALDI imaging mass
tein, apoAII, and apoE. This first large-scale clinical appli- spectrometry: Molecular snapshots of biochemical systems. Nat Methods
2007;4:828Y33
cation of a proteomic-discovered MAP suggests a panel of 13. Tanner S, Shu H, Frank A, et al. InsPecT: Identification of posttransla-
8 CSF proteins that are highly effective at identifying PD but tionally modified peptides from tandem mass spectra. Anal Chem 2005;
only modestly effective at identifying AD (98). The MAPs 77:4626Y39
for other candidate CSF biomarkers are pending. Finally, 14. Hansen BT, Davey SW, Ham AJ, et al. P-Mod: An algorithm and
software to map modifications to peptide sequences using tandem MS
MAPs are not limited to antibody-based platforms. For ex- data. J Proteome Res 2005;4:358Y68
ample, MS-based MAPs for biomarker confirmation or vali- 15. Schonberger SJ, Edgar PF, Kydd R, et al. Proteomic analysis of the brain
dation are under active investigation (99). This new platform in Alzheimer’s disease: Molecular phenotype of a complex disease
stands out for its precision without a requirement for process. Proteomics 2001;1:1519Y28
antibodies. 16. Shiozaki A, Tsuji T, Kohno R, et al. Proteome analysis of brain proteins
in Alzheimer’s disease: Subproteomics following sequentially extracted
protein preparation. J Alzheimers Dis 2004;6:257Y68

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


17. Kim SH, Vlkolinsky R, Cairns N, et al. Decreased levels of complex III
CONCLUSIONS core protein 1 and complex V beta chain in brains from patients with
The technology, experimental approaches, and bioinfor- Alzheimer’s disease and Down syndrome. Cell Mol Life Sci 2000;57:
1810Y16
matics that support proteomic research continue to undergo 18. Krapfenbauer K, Engidawork E, Cairns N, et al. Aberrant expression of
rapid evolution. The application of these new capabilities to peroxiredoxin subtypes in neurodegenerative disorders. Brain Res 2003;
the study of neurodegenerative diseases is providing insights 967:152Y60
into the biochemical pathogenesis of neurodegeneration as 19. Liao L, Cheng D, Wang J, et al. Proteomic characterization of
postmortem amyloid plaques isolated by laser capture microdissection.
well as fueling major efforts in the discovery of biomarkers. J Biol Chem 2004;279:37061Y68
Experience gained so far combined with expected advances in 20. Wang Q, Woltjer RL, Cimino PJ, et al. Proteomic analysis of
the near future will lead to greater throughput, accuracy, and neurofibrillary tangles in Alzheimer disease identifies GAPDH as a
reproducibility. Recent studies previously cited and additional detergent-insoluble paired helical filament tau binding protein. FASEB J
future studies will focus on the critically important issues of 2005;19:869Y71
21. Li Y, Nowotny P, Holmans P, et al. Association of late-onset
disease specificity and changes in the proteome that occur Alzheimer’s disease with genetic variation in multiple members of the
during latent and prodromal stages of disease. Ultimately, the GAPD gene family. Proc Natl Acad Sci U S A 2004;101:15688Y93
convergence of validated and confirmed data from multiple 22. Lin PI, Martin ER, Bronson PG, et al. Exploring the association of
laboratories will establish richly informative consensus sub- glyceraldehyde-3-phosphate dehydrogenase gene and Alzheimer disease.
Neurology 2006;67:64Y68
proteomes for human brain regions and CSF in health and 23. Olah J, Tokesi N, Vincze O, et al. Interaction of TPPP/p25 protein with
disease. glyceraldehyde-3-phosphate dehydrogenase and their co-localization in
Lewy bodies. FEBS Lett 2006;580:5807Y14
24. Cumming RC, Schubert D. Amyloid-beta induces disulfide bonding and
REFERENCES aggregation of GAPDH in Alzheimer’s disease. FASEB J 2005;19:
1. Ideker T, Thorsson V, Ranish JA, et al. Integrated genomic and pro- 2060Y62
teomic analyses of a systematically perturbed metabolic network. 25. Sultana R, Boyd-Kimball D, Cai J, et al. Proteomics analysis of the
Science 2001;292:929Y34 Alzheimer’s disease hippocampal proteome. J Alzheimers Dis 2007;11:
2. Johnson MD, Yu LR, Conrads TP, et al. Proteome analysis of DNA 153Y64
damage-induced neuronal death using high throughput mass spectrom- 26. Wakabayashi K, Tanji K, Mori F, et al. The Lewy body in Parkinson’s
etry. J Biol Chem 2004;279:26685Y97 disease: Molecules implicated in the formation and degradation of
3. Griffin TJ, Gygi SP, Ideker T, et al. Complementary profiling of gene alpha-synuclein aggregates. Neuropathology 2007;27:494Y506
expression at the transcriptome and proteome levels in Saccharomyces 27. Leverenz JB, Umar I, Wang Q, et al. Proteomic identification of novel
cerevisiae. Mol Cell Proteomics 2002;1:323Y33 proteins in cortical Lewy bodies. Brain Pathol (Zurich, Switzerland)
4. Tsuang DW, Bird TD. Genetics of dementia. Med Clin North Am 2002; 2007;17:139Y45
86:591Y614 28. Baba M, Nakajo S, Tu PH, et al. Aggregation of alpha-synuclein in
5. Eng JK, McCormack AL, Yates JR. An approach to correlate tandem Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy
mass-spectral data of peptides with amino-acid-sequences in a protein bodies. Am J Pathol 1998;152:879Y84
database. J Am Soc Mass Spectrom 1994;5:976Y89 29. Trojanowski JQ, Lee VM. Brain degeneration linked to Bfatal at-
6. Fenyo D, Beavis RC. A method for assessing the statistical significance tractions[ of proteins in Alzheimer’s disease and related disorders. J
of mass spectrometry-based protein identifications using general scoring Alzheimers Dis 2001;3:117Y19
schemes. Anal Chem 2003;75:768Y74 30. Glenner GG, Wong CW. Alzheimer’s disease: Initial report of the
7. Gygi SP, Rist B, Gerber SA, et al. Quantitative analysis of complex purification and characterization of a novel cerebrovascular amyloid
protein mixtures using isotope-coded affinity tags. Nat Biotechnol 1999; protein. Biochem Biophy Res Comm 1984;120:885Y90
17:994Y99 31. Lee VM, Balin BJ, Otvos L Jr, et al. A68: A major subunit of paired
8. Ross PL, Huang YN, Marchese JN, et al. Multiplexed protein quan- helical filaments and derivatized forms of normal Tau. Science 1991;
titation in Saccharomyces cerevisiae using amine-reactive isobaric tag- 251:675Y78
ging reagents. Mol Cell Proteomics 2004;3:1154Y69 32. Skovronsky DM, Lee VM, Trojanowski JQ. Neurodegenerative dis-
9. Haqqani AS, Kelly JF, Stanimirovic DB. Quantitative protein profiling eases: New concepts of pathogenesis and their therapeutic implications.
by mass spectrometry using label-free proteomics. In: Starkey M, Ann Rev Pathol 2006;1:151Y70
Elaswarapu R, eds. Genomics Protocols, vol. 439. Totowa, NJ: Humana 33. Woltjer RL, Cimino PJ, Boutte AM, et al. Proteomic determination of
Press, 2008;241Y56 widespread detergent-insolubility including AA but not tau early in the
10. Dijkstra M, Vonk RJ, Jansen RC. SELDI-TOF mass spectra: A view on pathogenesis of Alzheimer’s disease. FASEB J 2005;19:1923Y25
sources of variation. J Chromatogr B Analyt Technol Biomed Life Sci 34. Yang W, Woltjer RL, Sokal I, et al. Quantitative proteomics identifies
2007;847:12Y23 surfactant-resistant alpha-synuclein in cerebral cortex of Parkinsonism-
11. Albrethsen J. Reproducibility in protein profiling by MALDI-TOF mass dementia complex of Guam but not Alzheimer’s disease or progressive
spectrometry. Clin Chem 2007;53:852Y58 supranuclear palsy. Am J Pathol 2007;171:993Y1002

930 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008 Proteomics in Neurodegeneration

35. Winton MJ, Joyce S, Zhukareva V, et al. Characterization of tau multiplex quantitative proteomic platform in cerebrospinal fluid of
pathologies in gray and white matter of Guam parkinsonism-dementia patients with neurodegenerative disorders. J Alzheimers Dis 2006;9:
complex. Acta Neuropathol (Berl) 2006;111:401Y12 293Y348
36. Sebeo J, Hof PR, Perl DP. Occurrence of alpha-synuclein pathology in 59. Davidsson P, Westman-Brinkmalm A, Nilsson CL, et al. Proteome
the cerebellum of Guamanian patients with parkinsonism-dementia analysis of cerebrospinal fluid proteins in Alzheimer patients. Neuro-
complex. Acta Neuropathol 2004;107:497Y503 report 2002;13:611Y15
37. Forman MS, Schmidt ML, Kasturi S, et al. Tau and alpha-synuclein 60. Puchades M, Hansson SF, Nilsson CL, et al. Proteomic studies of
pathology in amygdala of Parkinsonism-dementia complex patients of potential cerebrospinal fluid protein markers for Alzheimer’s disease.
Guam. Am J Pathol 2002;160:1725Y31 Brain Res Mol Brain Res 2003;118:140Y46
38. Yamazaki M, Arai Y, Baba M, et al. Alpha-synuclein inclusions in 61. Zhang J, Goodlett DR, Peskind ER, et al. Quantitative proteomic
amygdala in the brains of patients with the parkinsonism-dementia analysis of age-related changes in human cerebrospinal fluid. Neurobiol
complex of Guam. J Neuropathol Exp Neurol 2000;59:585Y91 Aging 2005;26:207Y27
39. Castegna A, Aksenov M, Aksenova M, et al. Proteomic identification of 62. Zhang J, Goodlett DR, Quinn JF, et al. Quantitative proteomics of
oxidatively modified proteins in Alzheimer’s disease brain. Part I: cerebrospinal fluid from patients with Alzheimer disease. J Alzheimers
Creatine kinase BB, glutamine synthase, and ubiquitin carboxy-terminal Dis 2005;7:125Y33

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


hydrolase L-1. Free Radic Biol Med 2002;33:562Y71 63. Pan S, Wang Y, Quinn JF, et al. Identification of glycoproteins in human
40. Castegna A, Aksenov M, Thongboonkerd V, et al. Proteomic identi- cerebrospinal fluid with a complementary proteomic approach. J
fication of oxidatively modified proteins in Alzheimer’s disease brain. Proteome Res 2006;5:2769Y79
Part II: Dihydropyrimidinase-related protein 2, alpha-enolase and heat 64. Finehout EJ, Franck Z, Choe LH, et al. Cerebrospinal fluid proteomic
shock cognate 71. J Neurochem 2002;82:1524Y32 biomarkers for Alzheimer’s disease. Ann Neurol 2007;61:120Y29
41. Choi J, Levey AI, Weintraub ST, et al. Oxidative modifications and 65. Hu Y, Malone JP, Fagan AM, et al. Comparative proteomic analysis of
down-regulation of ubiquitin carboxyl-terminal hydrolase L1 associated intra- and interindividual variation in human cerebrospinal fluid. Mol
with idiopathic Parkinson’s and Alzheimer’s diseases. J Biol Chem Cell Proteomics 2005;4:2000Y09
2004;279:13256Y64 66. Pasinetti GM, Ungar LH, Lange DJ, et al. Identification of potential CSF
42. Butterfield DA, Sultana R. Identification of 3-Nitrotyrosine-modified biomarkers in ALS. Neurology 2006;66:1218Y22
brain proteins by redox proteomics. Methods Enzymol 2008;440: 67. Bowser R, Cudkowicz M, Kaddurah-Daouk R. Biomarkers for amyo-
295Y308 trophic lateral sclerosis. Expert Rev Mol Diagn 2006;6:387Y98
43. Sultana R, Reed T, Perluigi M, et al. Proteomic identification of nitrated 68. McComb JG. Recent research into the nature of cerebrospinal fluid
brain proteins in amnestic mild cognitive impairment: A regional study. J formation and absorption. J Neurosurg 1983;59:369Y83
Cell Mol Med 2007;11:839Y51 69. Han CY, Backous DD. Basic principles of cerebrospinal fluid meta-
44. Butterfield DA, Reed TT, Perluigi M, et al. Elevated levels of bolism and intracranial pressure homeostasis. Otolaryngol Clin North
3-nitrotyrosine in brain from subjects with amnestic mild cognitive Am 2005;38:569Y76
impairment: Implications for the role of nitration in the progression of 70. Bergsneider M. Evolving concepts of cerebrospinal fluid physiology.
Alzheimer’s disease. Brain Res 2007;1148:243Y48 Neurosurg Clin N Am 2001;12:631Y38, vii
45. Cripps D, Thomas SN, Jeng Y, et al. Alzheimer disease-specific con- 71. Huhmer AF, Biringer RG, Amato H, et al. Protein analysis in human
formation of hyperphosphorylated paired helical filament-Tau is poly- cerebrospinal fluid: Physiological aspects, current progress and future
ubiquitinated through Lys-48, Lys-11, and Lys-6 ubiquitin conjugation. challenges. Dis Markers 2006;22:3Y26
J Biol Chem 2006;281:10825Y38 72. Nilsson C, Stahlberg F, Thomsen C, et al. Circadian variation in human
46. Boutte AM, Woltjer RL, Zimmerman LJ, et al. Selectively increased cerebrospinal fluid production measured by magnetic resonance imag-
oxidative modifications mapped to detergent-insoluble forms of AA and ing. Am J Physiol 1992;262:R20YR24
beta-III tubulin in Alzheimer’s disease. FASEB J 2006;20:1473Y83 73. Preston JE. Ageing choroid plexus-cerebrospinal fluid system. Microsc
47. Lippa CF, Duda JE, Grossman M, et al. DLB and PDD boundary issues: Res Tech 2001;52:31Y37
Diagnosis, treatment, molecular pathology, and biomarkers. Neurology 74. Blennow K, Fredman P, Wallin A, et al. Protein analysis in cere-
2007;68:812Y19 brospinal fluid. II. Reference values derived from healthy individuals
48. Basso M, Giraudo S, Corpillo D, et al. Proteome analysis of human 18Y88 years of age. Eur Neurol 1993;33:129Y33
substantia nigra in Parkinson’s disease. Proteomics 2004;4:3943Y52 75. Blennow K, Fredman P, Wallin A, et al. Protein analyses in ce-
49. Shi M, Jin J, Wang Y, et al. Mortalin: A protein associated with rebrospinal fluid. I. Influence of concentration gradients for proteins
progression of Parkinson disease? J Neuropathol Exp Neurol 2008;67: on cerebrospinal fluid/serum albumin ratio. Eur Neurol 1993;33:
117Y24 126Y28
50. Jin J, Hulette C, Wang Y, et al. Proteomic identification of a stress 76. Blennow K, Fredman P, Wallin A, et al. Protein analysis in cere-
protein, mortalin/mthsp70/GRP75: Relevance to Parkinson disease. Mol brospinal fluid. III. Relation to blood-cerebrospinal fluid barrier function
Cell Proteomics 2006;5:1193Y204 for formulas for quantitative determination of intrathecal IgG production.
51. Kaul SC, Deocaris CC, Wadhwa R. Three faces of mortalin: A Eur Neurol 1993;33:134Y42
housekeeper, guardian and killer. Exp Gerontol 2007;42:263Y74 77. Osman I, Gaillard O, Meillet D, et al. A sensitive time-resolved
52. Carrette O, Demalte I, Scherl A, et al. A panel of cerebrospinal fluid immunofluorometric assay for the measurement of apolipoprotein B
potential biomarkers for the diagnosis of Alzheimer’s disease. Proteo- in cerebrospinal fluid. Application to multiple sclerosis and other
mics 2003;3:1486Y94 neurological diseases. Eur J Clin Chem Clin Biochem 1995;33:53Y58
53. Blennow K. CSF biomarkers for Alzheimer’s disease: Use in early 78. Carrette O, Burkhard PR, Hughes S, et al. Truncated cystatin C in
diagnosis and evaluation of drug treatment. Expert Rev Mol Diagn 2005; cerebrospinal fluid: Technical [corrected] artefact or biological process?
5:661Y72 Proteomics 2005;5:3060Y65
54. Davidsson P, Sjogren M. The use of proteomics in biomarker discovery 79. Hu S, Loo JA, Wong DT. Human body fluid proteome analysis.
in neurodegenerative diseases. Dis Markers 2005;21:81Y92 Proteomics 2006;6:6326Y53
55. D’Ascenzo M, Relkin NR, Lee KH. Alzheimer’s disease cerebrospinal 80. Hardt M, Thomas LR, Dixon SE, et al. Toward defining the human
fluid biomarker discovery: A proteomics approach. Curr Opin Mol Ther parotid gland salivary proteome and peptidome: Identification and
2005;7:557Y64 characterization using 2D SDS-PAGE, ultrafiltration, HPLC, and mass
56. Galasko D. Biomarkers for Alzheimer’s diseaseVclinical needs and spectrometry. Biochemistry 2005;44:2885Y99
application. J Alzheimers Dis 2005;8:339Y46 81. Davidsson P, Nilsson CL. Peptide mapping of proteins in cerebrospinal
57. Olsson A, Vanderstichele H, Andreasen N, et al. Simultaneous measure- fluid utilizing a rapid preparative two-dimensional electrophoretic
ment of {beta}-amyloid(1Y42), total tau, and phosphorylated tau procedure and matrix-assisted laser desorption/ionization mass spec-
(Thr181) in cerebrospinal fluid by the xMAP technology. Clin Chem trometry. Biochim Biophys Acta 1999;1473:391Y99
2005;51:336Y45 82. Raymackers J, Daniels A, De Brabandere V, et al. Identification of
58. Abdi F, Quinn JF, Jankovic J, et al. Detection of biomarkers with a two-dimensionally separated human cerebrospinal fluid proteins by

Ó 2008 American Association of Neuropathologists, Inc. 931

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Zhang et al J Neuropathol Exp Neurol  Volume 67, Number 10, October 2008

N-terminal sequencing, matrix-assisted laser desorption/ionization–mass 91. Mannes AJ, Martin BM, Yang HY, et al. Cystatin C as a cerebrospinal
spectrometry, nanoliquid chromatography-electrospray ionization-time fluid biomarker for pain in humans. Pain 2003;102:251Y56
of flight-mass spectrometry, and tandem mass spectrometry. Electro- 92. Simonsen AH, McGuire J, Podust VN, et al. A novel panel of
phoresis 2000;21:2266Y83 cerebrospinal fluid biomarkers for the differential diagnosis of Alzhei-
83. Sickmann A, Dormeyer W, Wortelkamp S, et al. Towards a high mer’s disease versus normal aging and frontotemporal dementia. Dement
resolution separation of human cerebrospinal fluid. J Chromatogr B Geriatr Cogn Disord 2007;24:434Y40
Analyt Technol Biomed Life Sci 2002;771:167Y96 93. Simonsen AH, McGuire J, Hansson O, et al. Novel panel of
84. Finehout EJ, Franck Z, Lee KH. Towards two-dimensional electro- cerebrospinal fluid biomarkers for the prediction of progression to
phoresis mapping of the cerebrospinal fluid proteome from a single Alzheimer dementia in patients with mild cognitive impairment. Arch
individual. Electrophoresis 2004;25:2564Y75 Neurol 2007;64:366Y70
85. Maccarrone G, Milfay D, Birg I, et al. Mining the human cerebrospinal 94. Ranganathan S, Williams E, Ganchev P, et al. Proteomic profiling of
fluid proteome by immunodepletion and shotgun mass spectrometry. cerebrospinal fluid identifies biomarkers for amyotrophic lateral scle-
Electrophoresis 2004;25:2402Y12 rosis. J Neurochem 2005;95:1461Y71
86. Ramstrom M, Ivonin I, Johansson A, et al. Cerebrospinal fluid 95. Ruetschi U, Zetterberg H, Podust VN, et al. Identification of CSF
biomarkers for frontotemporal dementia using SELDI-TOF. Experimen-

Downloaded from https://academic.oup.com/jnen/article-abstract/67/10/923/2916851 by guest on 13 May 2019


protein patterns in neurodegenerative disease revealed by liquid
chromatography-Fourier transform ion cyclotron resonance mass spec- tal Neurology 2005;196:273Y81
96. Biroccio A, Del Boccio P, Panella M, et al. Differential post-translational
trometry. Proteomics 2004;4:4010Y18
modifications of transthyretin in Alzheimer’s disease: A study of the
87. Wenner BR, Lovell MA, Lynn BC. Proteomic analysis of human cerebral spinal fluid. Proteomics 2006;6:2305Y13
ventricular cerebrospinal fluid from neurologically normal, elderly 97. Castano EM, Roher AE, Esh CL, et al. Comparative proteomics of
subjects using two-dimensional LC-MS/MS. J Proteome Res 2004;3: cerebrospinal fluid in neuropathologically-confirmed Alzheimer’s dis-
97Y103 ease and non-demented elderly subjects. Neurol Res 2006;28:155Y63
88. Xu J, Chen J, Peskind E, et al. Characterization of proteome of human 98. Zhang J, Sokal I, Peskind ER, et al. CSF multianalyte profile
cerebrospinal fluid. Int Rev Neurobiol 2006;73:29Y98 distinguishes Alzheimer and Parkinson diseases. Am J Clin Pathol
89. Sanchez JC, Guillaume E, Lescuyer P, et al. Cystatin C as a potential 2008;129:526Y29
cerebrospinal fluid marker for the diagnosis of Creutzfeldt-Jakob 99. Pan S, Rush J, Peskind ER, et al. Application of targeted quantitative
disease. Proteomics 2004;4:2229Y33 proteomics analysis in human cerebrospinal fluid using a liquid
90. Irani DN, Anderson C, Gundry R, et al. Cleavage of cystatin C in the chromatography matrix-assisted laser desorption/ionization time-of-
cerebrospinal fluid of patients with multiple sclerosis. Ann Neurol 2006; flight tandem mass spectrometer (LC MALDI TOF/TOF) platform. J
59:237Y47 Proteome Res 2008;7:720Y30

932 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.

Você também pode gostar