Enrichment of single neurons and defined brain regions from human brain tissue samples for...

download Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteome analysis

of 13

Transcript of Enrichment of single neurons and defined brain regions from human brain tissue samples for...

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    1/13

    T R A N S L A T I O N A L N E U R O S C I E N C E S - O R I G I N A L A R T I C L E

    Enrichment of single neurons and defined brain regionsfrom human brain tissue samples for subsequent proteome

    analysis

    Mariana Molina2,6 • Simone Steinbach1 • Young Mok Park4,5 • Su Yeong Yun4,5 •

    Ana Tereza Di Lorenzo Alho2,7 • Helmut Heinsen8 • Lea. T. Grinberg2,3,9 •

    Katrin Marcus1 • Renata E. Paraizo Leite2,3 • Caroline May1

    Received: 25 November 2014 / Accepted: 11 June 2015 / Published online: 30 June 2015   Springer-Verlag Wien 2015

    Abstract   Brain function in normal aging and neurologi-

    cal diseases has long been a subject of interest. With cur-rent technology, it is possible to go beyond descriptive

    analyses to characterize brain cell populations at the

    molecular level. However, the brain comprises over 100

    billion highly specialized cells, and it is a challenge to

    discriminate different cell groups for analyses. Isolating

    intact neurons is not feasible with traditional methods, such

    as tissue homogenization techniques. The advent of laser

    microdissection techniques promises to overcome previous

    limitations in the isolation of specific cells. Here, we pro-

    vide a detailed protocol for isolating and analyzing neurons

    from postmortem human brain tissue samples. We describe

    a workflow for successfully freezing, sectioning and

    staining tissue for laser microdissection. This protocol was

    validated by mass spectrometric analysis. Isolated neurons

    can also be employed for western blotting or PCR. This

    protocol will enable further examinations of brain cell-

    specific molecular pathways and aid in elucidating distinct

    brain functions.

    Keywords   Neurons    Brain    Laser microdissection 

    Substantia nigra

    Introduction

    Neurodegenerative diseases cause selective vulnerability of 

    specific cell populations (Mattson and Magnus  2006).

    Molecular analysis of single populations of affected cells

    (e.g. neurons) will lead to a better understanding of disease

    mechanisms and may therefore enable the identification of targets for early-onset diagnostics and disease treatment

    (Liao et al.  2004). However, the heterogeneity and com-

    plexity of brain tissue make it challenging to isolate

    specific brain cells. Neurons are not easily detached from

    their parenchyma, are highly branched and interact in

    networks. Recently, laser microdissection (LMD) hasM. Molina, S. Steinbach, R. E. P. Leite and C. May contributed

    equally.

    &   Katrin Marcus

    [email protected]

    &   Caroline May

    [email protected]

    1 Medizinisches Proteom-Center, Ruhr-Universität Bochum,

    ZKF, Universitätsstraße 150, 44801 Bochum, Germany

    2 Physiopathology in Aging Lab/Brazilian Aging Brain Study

    Group-LIM22, University of Sao Paulo Medical School,

    São Paulo, Brazil

    3 Discipline of Geriatrics, University of Sao Paulo Medical

    School, São Paulo, Brazil

    4 Center for Cognition and Sociality, Institute for Basic

    Science, Ochang, Korea

    5 Mass Spectrometry Research Center, Ochang, Korea

    6 Discipline of Pathophysiology, University of Sao Paulo

    Medical School, São Paulo, Brazil

    7 Instituto do Cérebro, Hospital Israelita Albert Einstein,

    São Paulo, Brazil

    8 Bavarian Julius-Maximilians-Universität, Würzburg,

    Germany

    9 Department of Neurology, Memory and Aging Center,

    University of California, San Francisco, USA

     1 3

    J Neural Transm (2015) 122:993–1005

    DOI 10.1007/s00702-015-1414-4

    http://crossmark.crossref.org/dialog/?doi=10.1007/s00702-015-1414-4&domain=pdfhttp://crossmark.crossref.org/dialog/?doi=10.1007/s00702-015-1414-4&domain=pdf

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    2/13

    emerged as a promising alternative for isolating neurons

    for genomics and transcriptomics (Boone et al.  2013;

    Kumar et al.   2013; Majer et al.   2012; Moulédous et al.

    2003; Friedrich et al. 2012; Decarlo et al. 2011; Simunovic

    et al.  2009; Cantuti-Castelvetri et al.  2007; Elstner et al.

    2011). For example, Simunovic et al. (2009) made a gene

    expression profile of dopamine neurons isolated by LMD

    for a comparison study of idiopathic PD and control sub- jects. This allowed for identification of different genes

    associated with PD, such as, e.g. PARK. Alternatively,

    LMD can also be a promising alternative for proteomic

    studies. Using this technique, the tissue is first cut into thin

    sections and mounted onto glass slides. With the aid of a

    microscope, the cells or regions of interest are dissected

    with a laser and collected in a tube (Fig. 1). An interme-

    diate staining step between tissue mounting and dissection

    enables cells to be distinguished by molecular features

    rather than by morphological characteristics. Furthermore,

    even smaller structures, such as inclusion bodies, can be

    isolated for analyses (Hashimoto et al. 2012; Minjarez et al.2013). Despite the numerous advantages offered by LMD,

    this method has yet to be broadly incorporated into neu-

    roscience proteomic studies due to technical limitations. In

    this study, using the human substantia nigra as an example,

    we provide a detailed workflow for human brain studies

    using LMD that overcomes technical limitations for pro-

    teomic analysis. This approach can be used for highly

    specific studies of the neuronal proteome, facilitating the

    further understanding of complex brain function and cir-

    cuitry. Specifically, LMD enables the researcher to char-

    acterize proteome changes, which would be masked in

    global analyses, making LMD an essential tool for neu-

    ronal proteomic studies.

    Methods

    Ethical statement

    All the protocols were approved by the ethics committee of 

    the University of São Paulo and the Ruhr-University

    Bochum, as well as the Brazilian federal research ethics

    committee. Informed written consent was obtained from

    the next of kin.

    Subjects

    Postmortem human brains of elderly subjects were supplied

    by the Brain Bank of the Brazilian Brain Aging Study

    Group (BBBABSG) at the University of Sao Paulo Medical

    School (USPMS). The BBBABSG collects brains fromdeceased individuals aged C50 years. The methods used

    by the BBBABSG have been previously reported (Grinberg

    et al. 2007).

    Defining the optimal anatomical plane for neuron

    collection

    Two brains of control human subjects were fixed by

    immersion in 10 % formalin for at least 4 weeks. There-

    after, the brainstem was severed by a horizontal cut ventral

    to the superior colliculi. The formalin-fixed brainstems

    were dehydrated in a graded series of ethanol solutions,

    embedded in celloidin, and serially sectioned at 350 lm

    thickness in either the horizontal or the sagittal plane

    (Heinsen et al.  2000; Theofilas et al. 2014). The sections

    were stained with gallocyanin (Nissl staining) and mounted

    as previously described in detail (Heinsen et al. 2000). The

    cell arrangement and density of cells were compared in the

    substantia nigra  in each section.

    Freezing for laser microdissection

    Three midbrains were collected. Each midbrain was

    mediosagittally sectioned, resulting in three right and three

    left half-midbrains. Each one of the six half-midbrain

    samples was frozen in one of the following combinations

    of temperature: (I) placement at  -20   C for 30 min, fol-

    lowed by   -80   C storage; (II) placement and storage at

    -80   C; and (III) snap freezing in liquid nitrogen, followed

    by   -80   C storage (Table 1), resulting in 2 samples of 

    each frozen method. One slice of 10  lm was obtained from

    each sample and stained with cresyl violet. Three inde-

    pendent investigators blinded to the congelation method

    Fig. 1   Principles of laser microdissection. The isolation and collec-

    tion of specific cells is based on a laser. First, a laser microdissection

    is performed. Second, a laser pulse catapults the selected sample into

    a collection device. Using this technique, it is possible to achieve

    contact-free isolation of specific areas, single cells and even specific

    cell compartments

    994 M. Molina et al.

     1 3

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    3/13

    qualitatively analyzed the slices based on the morphology

    of neurons and quality of the tissue after sectioning and

    staining optimization.

    Sectioning and staining the tissue for laser

    microdissection

    Two frozen midbrains were sectioned in sagittal plane atthe level of the substantia nigra. Sectioning was performed

    with a Cryostat Microm HM550 (Thermo Scientific,

    Dreieich, Germany) with a fixed knife holder (Leica

    Biosystems, Nußloch GmbH, Nußloch, Germany) at

    -10   C object temperature and  -20   C chamber temper-

    ature. Before sectioning, tissue was left in the cryostat for

    15 min for warming to-20   C. The sections (5, 10, 20 and

    30  lm) were placed on special membrane slides for LMD

    (1.0 PEN membrane slides, Carl Zeiss Microscopy GmbH,

    Göttingen, Germany). To distinguish the cells, sections

    were stained with cresyl violet according to a protocol from

    Carl Zeiss Microscopy GmbH (Carl Zeiss MicroImaging,LCM Protocols—Protein Handling for LC/MS), with slight

    modifications. Cresyl violet stains the nuclei deep violet

    and the cytoplasm weak purple. This staining technique is

    recommended by Zeiss for laser microdissection intended

    for subsequent proteomic approaches. Briefly, 1 g of cresyl

    violet (cresyl violet acetate, Sigma Life Sciences, St. Louis,

    USA) was diluted in 100 ml of 50 % ethanol, with stirring

    overnight on a stirring plate. The following day, the solu-

    tion was filtered to remove undissolved particles. All

    solutions were freshly prepared and used precooled (4   C).

    A summary of the staining protocol is provided in Table 2.

    Laser microdissection

    In the LMD machine, a laser pulse transports the selected

    sample out of the slide into a collection device (usually a

    microtube cap, which may be filled with a solution). With

    this approach, we tested two different solutions for wet

    collection: ultrapure water (TKA-Gene PURE, Thermo

    Scientific, Baltimore, USA) and 50 % acetonitrile (Bio-

    solve BV, Valkenswaard, Netherlands) diluted in water.

    LMD was performed with a PALM Micro Beam

    instrument (P.A.L.M.-System LCM, Carl Zeiss Micro-

    scopy GmbH) with non-adhesive tubes (MicroTube 500,

    Carl Zeiss Microscopy GmbH). The microtube cap was

    filled with 50  ll of one of the solutions. After finishing

    LMD, the tube was handled, closed and stored upside-

    down at   -80   C. Because we intended to perform mass

    spectrometry, 2.5 ll of 2 % RapidGestTM SF Surfactant

    (Waters GmbH, Milford, MA, USA) was carefully addedbefore storage for a final concentration of 0.1 %.

    The LMD was tested on 5, 10, 20 and 30  lm thick 

    sections to collect single cells as well as whole brain

    regions of the tissue sections.

    Before placing a sample cap over the PEN membrane

    slide, the regions/cells to be collected were marked; if all of 

    the desired samples could not be collected from one slide,

    the next slide was also used to prevent unnecessary evap-

    oration of the solution in the tube cap. The selection of 

    optimal thickness was based on the ability to catapult the

    region/cells, preventing falling apart or attachment to the

    membrane, and the ability to establish the laser parametersthat would enable an effective catapult process within a

    short time.

    Sample preparation and sample digestion for mass

    spectrometric analysis

    For sample digestion, the tubes containing the collected

    tissue were incubated upside-down in an ultrasonic bath for

    1 min, and then, the samples were centrifuged briefly. This

    step was repeated. Next, the samples were incubated at

    95   C for 5 min in a thermomixer and centrifuged again.

    One microliter of 250 mM 1,4-dithiothreitol (DTT,

    AppliChem GmbH, Darmstadt, Germany) was added to

    each sample, and the mixture was incubated for 30 min at

    60   C. The samples were then incubated with 1.4 ll of 

    0.55 M iodoacetamide (AppliChem) at room temperature

    for 30 min in the dark. Digestion was initiated by addition

    of 1:4 trypsin in water (Promega, Mannheim, Germany) at

    37   C for 4 h and stopped with 3.25  ll of 10 % TFA for

    30 min at 37   C. The samples were then centrifuged for

    15 min at 14,000 rpm and 4   C. The resulting supernatant

    was transferred into a new sample cap and dried with

    SpeedDry (RVC 2-25 CDplus, Martin Christ

    Gefriertrocknungsanlagen GmbH, Osterode, Germany).

    Finally, 30  ll of 0.1 % TFA was added to each sample.

    Protein concentration determination with amino

    acid analysis

    After sample digestion, protein concentrations were

    determined by amino acid analysis as described by Plum

    et al. (Plum et al.   2013). For this process, 5  ll of each

    digested sample was dried completely in a glass tube. Next,

    Table 1   Overview of the freezing procedures

    Case Side Procedure

    1 Left   -20   C/ -80   C

    Right   -80   C

    2 Left   -20   C/ -80   C

    Right Liquid nitrogen/  -80   C

    3 Left   -80   C

    Right Liquid nitrogen/  -80   C

    Enrichment of single neurons and defined brain regions from human brain tissue samples for …   995

     1 3

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    4/13

    the samples were dissolved by the addition of 10 ll of 

    20 mM HCl. According to the manufacturer’s instructions,

    an amino acid analysis was performed using the Acquity

    HPLC and AccQ-Tag Ultra system (Waters GmbH). For

    derivatization and to allow the conversion of primary and

    secondary amines into stable derivatives, the samples were

    incubated with 10 ll of AccQ-Tag reagent and 30  ll of 

    internal norvaline standard (final concentration: 10 pmol/ 

    ll) for 10 min. Amino acid derivatives were separated on

    an AccQ-Tag Ultra RP column and detected by an Acquity

    UPLC-TUV detector (Waters GmbH). The amino acids

    were quantified using 10 pmol/ ll amino acid standards.

    Mass spectrometric analysis

    The LC–MS/MS analysis was performed on an UltiMate

    3000 RSLC nano LC system (Dionex, Idstein, Germany)

    coupled to the Q Exactive system (Thermo Fisher Scien-

    tific, Bremen, Germany). Sample loading on the trap col-

    umn (100  lm  9  2 cm, particle size 5  lm, pore size 100 Å,

    C18, Thermo Scientific) was performed by an autosampler

    at a flow rate of 30  ll/min at 60   C with 0.1 % TFA. After

    a washing step, the sample was loaded onto an analytical

    C18 column (75 lm  9   50 cm, particle size 2 lm, pore

    size 100 Å, Thermo Scientific). For peptide separation, a

    linear gradient of 4–40 % running buffer B (84 % ACN,

    0.1 % FA; running buffer A: 0.1 % FA) was conducted for

    95 min, followed by a washing step at 95 % B for 5 min

    and an equilibration step from 95 to 4 % B. The connection

    between the HPLC system and Q Exactive was online, and

    electrospray ionization was performed. The ion spray

    voltage was set to 1600 V (?) and the capillary temperature

    to 250   C. The scan range was defined as 350–1400  m/z

    for the full scan mode, and for SIM scans, an Orbitrap res-

    olution of 70,000 (at 200  m/z) was set. Internal recalibration

    useda target AGC of3e6,fill timeof 80 msand the lock mass

    of polydimethylcyclosiloxane (m/z  445.120). The settings

    for the initiation of MS/MS analysis of  m/z  values were as

    follows: dynamic exclusion list: 30 s, top 10 ions (charge,

    ?2, ?3, ?4).

    Fragments for MS/MS analysis were generated using

    high-energy collision-induced dissociation (HCD). For ion

    dissociation, a normalized collision energy (NCE) of 27 %,

    isolation window of 2.2 m/z   and a fixed first mass of 

    130  m/z   were used. In addition, the following fragment

    analysis was performed in an Orbitrap analyzer with a

    resolution of 35,000 at 200 m/z, target of 1e6 and fill time

    of 120 ms.

    The resulting RAW data of the LC–MS/MS analysis

    was interpreted by Proteome Discoverer (version

    1.4.0.288) using the UniProt database [UniProt/SwissProt-

    Release 2013_05 of 01.05.2013; 541,561 (http://www.uni

    prot.org)] as a protein sequence reference. The taxonomy

    was set to Homo sapiens and a precursor mass tolerance of 

    5 ppm and fragment mass tolerance of 20 amu were

    applied. The target FDR was set to 0.01. Additionally, the

    following dynamic modifications were assumed: oxidation

    and carbamidomethylation. The resulting MGF files were

    analyzed with Protein Inference Algorithms software (PIA;

    version 10.0-rc2-dev,   http://www.ruhr-uni-bochum.de/ 

    mpc/software/PIA/index.html,   https://github.com/mpc-

    bioinformatics/pia) and Ingenuity Pathway Analysis

    software (IPA; version 18488943) (http://www.ingenuity.

    com/products/ipa) using the core analysis tool.

    Results

    In this paper, we describe an approach for studying single

    human neuron populations using LMD technology. This

    approach is effective for proteomic application. Although

    we focused on the   substantia nigra   of control human

    brains, this approach is also suitable for other cell regions

    and single cell types in humans and animal models.

    Definition of the optimal anatomical plane

    for neuron collection within the substantia nigra

    Sagittal planes were selected to allow recognition of the

    layers of the substantia nigra pars compacta  and neurons.

    Furthermore, the sagittal plane is the most appropriate

    plane of section because the long axis of a majority of 

    nigral neuronal perikarya is arranged in the rostro-caudal

    plane (Fig. 2).

    Table 2   Cresyl violet staining

    procedure  Step Solution Procedure Duration Temperature

    1. 70 % ethanol Incubation 2 min 4   C

    2. 1 % cresyl violet Incubation 30 s RT

    3. – Discard remaining solution

    4. 70 % ethanol Dipping 3–5  9  1 s 4   C

    5. 100 % ethanol Dipping 1  9  s 4   C

    6. – Air dry 1–2 min RT

    996 M. Molina et al.

     1 3

    http://www.uniprot.org/http://www.uniprot.org/http://www.ruhr-uni-bochum.de/mpc/software/PIA/index.htmlhttp://www.ruhr-uni-bochum.de/mpc/software/PIA/index.htmlhttps://github.com/mpc-bioinformatics/piahttps://github.com/mpc-bioinformatics/piahttp://www.ingenuity.com/products/ipahttp://www.ingenuity.com/products/ipahttp://www.ingenuity.com/products/ipahttp://www.ingenuity.com/products/ipahttps://github.com/mpc-bioinformatics/piahttps://github.com/mpc-bioinformatics/piahttp://www.ruhr-uni-bochum.de/mpc/software/PIA/index.htmlhttp://www.ruhr-uni-bochum.de/mpc/software/PIA/index.htmlhttp://www.uniprot.org/http://www.uniprot.org/

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    5/13

    Freezing protocol for tissues used for LMD

    processing

    We tested multiple freezing protocols to preserve tissue

    morphology and integrity. A comparison of the protocols is

    shown in Fig. 3. Direct storage of the tissue at-80   C and

    preliminary freezing at  -20   C before storage at  -80   C

    lead to tissue disruption, which adversely affected tissuemorphology and integrity. In contrast, a quick submersion

    in liquid nitrogen prior to storage at -80   C resulted in a

    more integrated tissue sample. Therefore, this protocol was

    used.

    Testing of section thickness for neuron analysis

    using LMD

    Tissue section thickness is a crucial factor in achievinghigh quality and repeatable samples when using LMD for

    400705948_93_40_2.5_SubnigCompIntermedVentralTierventral

    pars compacta

    pars diffusa

    Fig. 2   Optimal plane for

    isolation of neurons from

    substantia nigra   tissue sections.

    Sagittal sections of  substantia

    nigra tissue are optimal for the

    isolation of neurons because the

    different tiers of the  substantia

    nigra pars compacta   are easily

    identified. The  inset  shows

    neurons in the  substantia nigra

     pars diffusa  from a 400  lmthick gallocyanin-stained

    section (Heinsen et al.  2000)

    Fig. 3   Optimal freezing procedure for laser microdissection. The

    freezing process for LMD processing was optimized.  a  The resulting

    tissue after freezing the sample at -20   C before storage at -80   C is

    shown.  b  Visualizes the tissue after it was stored directly at -80   C.

    c The results of tissue preparation using a quick incubation with liquid

    nitrogen before the tissues were stored at  -80   C are shown. Tissue

    morphology and integrity was highly affected when the tissue was

    stored directly at   -80   C (b) and when the tissue was stored

    preliminary at  -20   C before it was stored at  -80   C (a). The best

    storage results and intact tissue were obtained when the tissue was

    immersed for a few seconds in liquid nitrogen before storage at

    -80   C (c)

    Enrichment of single neurons and defined brain regions from human brain tissue samples for …   997

     1 3

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    6/13

    sample collection. Therefore, we tested four different

    thicknesses (5, 10, 20 and 30  lm). Using the ‘‘Cutting’’

    and ‘‘RoboLPC’’ settings of the LMD, we compared the

    results of each tissue thickness. Figure 4   shows that the

    accuracy of laser cutting decreased with increasing thick-ness. Based on these results, we used 10–20  lm sections

    for selected regions and 5–10  lm sections for single cell

    isolations because a higher accuracy was needed.

    Staining procedure for LMD

    Cresyl violet is a common stain for neurons (Mouledous

    et al.  2002). Therefore, this method was tested for LMD

    application. This staining provided easy and fast handling

    of the samples, and it did not show any complications in

    background quality or optical resolution. These results

    supported our use of cresyl violet as the staining choice forLMD processing.

    Adjustments for laser microdissection

    LMD is a process that depends on several parameters, each

    of which must be optimized for each tissue type and

    sample. The settings are influenced by the tissue type, the

    cells or regions to be isolated, the thickness of the tissue

    and the applied magnitude. Therefore, it is essential to

    investigate these settings for each sample collection. Wang

    et al. (2009) described that a higher magnitude or thicker

    section requires more energy for cutting.

     Isolation of cells using LMD

    Single cells from the 5 and 10  lm sections were dissected,

    without difficulty, using a thin cutting line (indicating the

    best laser energy setting), and laser energy adjustments

    were achieved quickly. The catapult method was efficient

    for all attempts.

     Isolation of regions using LMD

    Energy setting adjustments took longer and resulted in

    broader cutting lines for LMD of single cells from 20 lm

    sections, and some of the catapult sections were not suc-cessful. However, 20  lm sections enabled effective and

    specific capture of isolated entire brain regions, such as the

    substantia nigra.

    Area selection for mass spectrometry analysis

    The area that must be isolated for a proper mass spectro-

    metric analysis must be investigated. The required area

    depends on the tissue and cell type and tissue thickness. At

    Fig. 4   Optimal section thickness for cutting single cells and regions.

    Section thickness is essential for optimal neuron isolation using laser

    microdissection. Therefore, four different thicknesses were tested:

    5  lm (a), 10  lm (b), 20  lm (c) and 30  lm (d). For each thickness, a

    cutting line and square were cut. Additionally, a square was

    catapulted after cutting to ensure that accurate laser microdissection

    processing was possible. An increase of section thickness decreased

    cutting accuracy. Using 5  lm sections, a thin fine cutting line was

    possible and catapulting of the sample occurred without difficulties.

    However, the cutting line broadened and a dark edge appeared with

    increasing section thickness (20  lm). Neither cutting nor catapulting

    of the sample was possible in 30  lm sections. Testing revealed that

    cutting conditions were excellent in 5 and 10  lm sections

    998 M. Molina et al.

     1 3

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    7/13

    least 100 ng protein amount must be employed for a good

    mass spectrometric analysis. Therefore, a region of 

    25,000,000  lm2 was sufficient to analyze the entire   sub-

    stantia nigra, in sections that had a thickness of 20 lm, by

    mass spectrometry. A total of 2500 neurons

    (*1,500,000  lm2) were isolated in 10 lm thick sectionsfor mass spectrometric analysis of single neurons.

    Isolation of tissue sample using LMD

    Cells for proteomic analysis must be isolated in an opti-

    mized solution. We tested two different solutions (water

    and 50 % acetonitrile) using the entire   substantia nigra.

    Mass spectrometric analysis revealed equal amounts of 

    identified proteins after isolation in water as well as 50 %

    acetonitrile. We selected water as the collecting buffer

    because no relevant differences in the amount of identified

    protein groups were observed.

    Sample preparation protocol for mass spectrometry

    The presented protocol performed a standard digestion

    with trypsin to investigate the  substantia nigra   and a

    number of identified proteins, as well as analyze their

    locations and types within this tissue. We performed a

    successful mass spectrometric analysis using this protocol

    and identified 1144 proteins within the  substantia nigra

    in 500,000,000  lm3 tissue. The left part of Fig. 5a, c

    summarizes the results of this analysis, which was per-

    formed to provide an initial overview of the  substantia

      P  r  o  t  e  i  n 

      t  y  p  e

      L  o  c  a   l  i  z  a  t  i  o

      n

    Tissue   Neurons

    a b

    c d

    Fig. 5   Summary of mass spectrometry results.   Substantia nigra

    tissue isolated by laser microdissection was analyzed successfullyusing mass spectrometry, which revealed the first overview of the

    substantia nigra   proteome. A total of 1144 proteins were identified

    (out of 500,000,000  lm3) in the entire  substantia nigra  tissue, which

    were investigated according to their location (a) and type (c). Both

    factors are of special interest to reveal membrane-specific proteins

    that greatly impact cell homeostasis and brain function. We revealed

    that 15 % of all identified proteins were located on the plasma

    membrane and that 13 % of proteins were associated with transporter(11 %) or channel proteins (2 %). Mass spectrometric analysis of 

    15,000,000  lm3 isolated neurons located in the   substantia nigra

    resulted in the same distributions (b). Proteins that were associated

    with transporter (12 %) and channel proteins (1 %) composed 13 %

    of the total amount of identified protein groups (d)

    Enrichment of single neurons and defined brain regions from human brain tissue samples for …   999

     1 3

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    8/13

    nigra   proteome. The greatest quantities of proteins were

    located in the cytoplasm (66 %) (Fig. 5a). Proteins that

    were associated with the plasma membrane made up

    15 % of the total identified protein groups. We also

    analyzed the distribution of protein types (Fig. 5c). In

    total, 13 % of identified proteins were successfully shown

    to be associated with transporter (11 %) and channel

    proteins (2 %).Mass spectrometric analysis of isolated neurons located

    in the   substantia nigra   (15,000,000  lm3) resulted in the

    same distributions (right part of Fig. 5b, c) of 303 identi-

    fied proteins. A total of 71 % of proteins were located in

    the cytoplasm. Fourteen percent of proteins were associ-

    ated with the plasma membrane (Fig. 5b). Proteins that

    were associated with transporter (12 %) and channel pro-

    teins (1 %) comprises 13 % of the total identified proteins

    (Fig. 5d). These results indicate that our digestion protocol

    is suitable for proteomic brain investigations.

    Discussion

    A combined technique of single cell dissection and sub-

    sequent molecular analysis could facilitate better under-

    standing of the disease mechanisms of neurodegenerative

    diseases such as Alzheimer’s and Parkinson’s disease.

    Common features of many neurodegenerative diseases are

    protein aggregation and the selective degeneration of a

    particular group of neurons (Dickson  2007; Duyckaerts

    et al.   2009). Therefore, the molecular analysis of single

    neuronal cell populations is expected to yield a better

    understanding of disease mechanisms. Although several

    groups in the past few years have used proteomics and

    other molecular biology techniques to study the brain and

    the mechanisms underlying neurodegenerative diseases,

    many questions still remain unanswered (Dumont et al.

    2006; Kitsou et al.   2008; Werner et al.   2008; He et al.

    2006). Changes in specific neuronal populations are com-

    monly masked in global analyses, where whole brains or

    large sections rather than specific cell groups are homog-

    enized for analysis.

    LMD was developed less than 20 years ago and enables

    the isolation of specific cells within a tissue (Emmert-Buck 

    et al. 1996). In combination with new analytical methods

    allowing for the analysis of small sample volumes, new

    insights into the pathological cascade, therapy options, and

    neuroprotective agents, preclinical/clinical biomarkers are

    possible. However, the combination of these recent tech-

    niques can have various limitations depending on the aims

    of the study.

    In the present study, by focusing on the substantia nigra

    of elderly control human brains, we aimed to overcome

    several technical hurdles described below.

    Optimized freezing process for preserving tissue

    morphology and tissue integrity

    The use of fresh tissue is mandatory for proteomics with

    LMD. Fresh tissue has no changes in RNA (Goldsworthy

    et al. 1999) or proteins (Rekhter and Chen 2001) compared

    with formalin-fixed paraffin-embedded tissue, which con-

    tains protein cross-linking products. Additionally, paraffinis not compatible with LC–MS/MS and requires a

    deparaffination step, resulting in protein loss (Hood et al.

    2006). Therefore, a freezing procedure for LMD workflows

    had to be established. We tested multiple freezing proto-

    cols, and the best results were achieved when the sample

    was incubated for a short time in liquid nitrogen before

    storage at   -80   C. Slow freezing promotes ice crystal

    formation of water within the tissue, which expands and

    disrupts the tissue. Therefore, it is important to freeze the

    tissue rapidly to prevent water crystallization. The use of 

    liquid nitrogen allows water to transform into a vitreous

    form that does not expand.

    Optimal plane for analyzing neurons

    within the substantia nigra

    Because there is a lack of validated normative data from

    morphological studies of the substantia nigra, especially for

    older individuals, we utilized a tissue-processing method

    based on celloidin mounting to establish an optimal plane for

    collecting neurons (Cabello et al. 2002). Cytoarchitectoni-

    cally, thesubstantia nigra is dividedinto three parts: the pars

    compacta, pars diffusa, and pars reticulata (Braak andBraak 1986). The neurons containing neuromelanin are in the pars

    compacta, which consists of layers of medium to large

    neurons (Braak and Braak   1986). To improve sampling

    strategy, theneuroanatomyof the region wasconsidered, and

    a sagittal plane was selected to allow recognition of the

    layers and neurons for single cell analysis. Further, the

    sagittal plane is the most appropriate plane of section

    because the long axis of a majority of nigral neuronal peri-

    karya is arranged in the rostro-caudal plane (Fig. 2).

    Optimal section thickness for neuron analysis using

    LMD

    Sectioning is a crucial step for LMD-based studies. In

    particular, section thickness is essential for good and

    repeatable LMD processing. Testing of four different

    thicknesses (5, 10, 20 and 30  lm) revealed that the accu-

    racy of laser cutting decreases with increasing thickness

    (Fig. 4). These results agree with those of Wang and

    coworkers. These authors needed higher energies for 20  lm

    sections than for 10  lm sections, and to make multiple cuts

    1000 M. Molina et al.

     1 3

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    9/13

    to sever the tissue. However, they used this cutting proce-

    dure for cell groups rather than single cells (Wang et al.

    2009), indicating that the cutting accuracy for 5 and 10 lm

    sections is excellent and that, even very small single cells

    can be isolated without damaging the tissue (Fig. 4).

    Optimal staining procedure

    Staining is a necessary technique for distinguishing dif-

    ferent tissue patterns and cell types, and is an important

    part of LMD experiments. To obtain a better signal and

    minimize background, the sample can be incubated with a

    higher concentration of staining solution; however, this

    procedure may negatively affect the proteomic analysis

    (Gutstein and Morris   2007). The literature contains

    numerous staining protocols for different stains in the

    context of LMD processing. Common stains include

    hematoxylin and eosin (De Souza et al. 2004; Dos Santos

    et al. 2007), toluidine blue (Sridharan and Shankar 2012;

    Kulkarni et al. 2013; Kirana et al. 2009; Lawrie et al. 2001;Mouledous et al.  2002) and cresyl violet (Boone et al.

    2013; Aaltonen et al. 2011). Immunohistochemical staining

    is also popular for LMD processing because it can distin-

    guish single cell types (e.g. astrocytes from small neurons)

    or cellular subtypes (e.g. dopaminergic from GABAergic

    neurons). The choice of staining procedure depends not

    only on sample tissue but also on the additional sample-

    processing procedures and the study aims. Previous

    investigations have shown that hematoxylin and eosin

    staining is not compatible with proteomic analysis. For

    instance, Gutstein and coworkers tested different conven-

    tional stainings on brain tissue, and showed that hema-

    toxylin and eosin is not compatible with 2D gel analysis

    (Gutstein and Morris   2007). Several other groups have

    confirmed the negative influence of hematoxylin/eosin

    staining on the proteome (Sitek et al. 2005; Craven et al.

    2002; Craven and Banks   2001). However, this stain has

    shown no effects on samples for RNA and DNA investi-

    gations (Burgemeister et al. 2003). Toluidine blue is often

    used for the DNA and RNA analysis of LMD samples

    (Kulkarni et al. 2013) and can also be used for proteomic

    analysis (Lawrie et al.   2001; Mouledous et al.   2002). In

    contrast, Craven and coworkers investigated kidney sam-

    ples with toluidine blue and identified detrimental effects

    on protein recovery in 2D gel analysis (Craven et al. 2002).

    Cresyl violet is especially common for staining neurons. It

    is a cationic solution (Mouledous et al. 2002) and binds to

    acidic components of the neuronal cytoplasm, especially

    ribosomes, which are present in large numbers in neurons

    (Burnet et al.  2004; Eltoum et al.   2002). Clément-Ziza

    et al. (2008) improved the cresyl violet staining procedure

    for RNA analysis by replacing all solvents with ethanol,

    which prevents sample degradation. Compared with other

    common stains, cresyl violet provides the best contrast

    between stained and unstained tissue (Ginsberg and Che

    2004). The staining of frozen tissue did not show any

    complications. The tissue background was transparent and

    resulted in an excellent optical resolution. Based on the

    literature described above all further experiments were

    performed on cresyl violet-stained samples.

    Optimized laser microdissection

    Further, LMD performance is influenced by temperature

    and humidity, which due to weather variability changes the

    energy settings daily. Therefore, it is especially important

    to optimize the sample amplitude and thickness for each

    sample. The use of a higher magnitude and thinner section

    is advisable for single cells or small areas to ensure that no

    surrounding tissue is collected.

    Furthermore, the tissue amounts required to achieve the

    amount of proteins needed for mass spectrometric analysis

    strongly depends on the tissue type. Therefore, it isessential to investigate these parameters during pre-trials.

    Optimized sample preparation protocol for mass

    spectrometry

    Sufficient tissue of at least 100 ng must be collected to

    guarantee a mass spectrometric analysis of high perfor-

    mance. This study achieved proper mass spectrometric

    analysis using 500,000,000  lm3 of   substantia nigra   tissue

    and 15,000,000 lm3 of isolated neurons located in the

    substantia nigra. However, the required area depends on

    the tissue and cell type, and it must be investigated for each

    tissue type individually. Therefore, preliminary tests are

    essential at the start of a study. For example, to define the

    amount of neurons that are needed for mass spectrometric

    analysis, 5000 neurons were set as starting point for

    investigations, revealing that a total of 2500 neurons

    (*1,500,000  lm2) in 10  lm thick sections are sufficient

    for mass spectrometric analysis of single neurons. How-

    ever, the numbers of identified proteins for isolated neurons

    could be enhanced if more neurons are isolated.

    Further, it is very important to establish a specific and

    reproducible sample preparation protocol to achieve com-

    parative proteome analyses. In this context, the digestion

    step is crucial. Tryptic digestion is commonly used in

    proteomic research, and this technique produced good

    results for LMD processing. Using this protocol, proteins

    could be identified that are associated with the  substantia

    nigra as well as Parkinson’s disease (Spillantini et al. 1997;

    Bonifati et al. 2003; Damier et al. 1999). For example,  a-

    synuclein was identified with sequence coverage of 

    46.43 %. Nine unique peptides of DJ-1 were identified, a

    protein that is associated with familial PD, resulting in

    Enrichment of single neurons and defined brain regions from human brain tissue samples for …   1001

     1 3

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    10/13

    sequence coverage of 58.20 %. Further, a dopamine

    transporter {uniprot ID Q01959 [UniProt/SwissProt-Re-

    lease 2013_05 of 01.05.2013; 541,561 (http://www.uniprot.

    org)]} was also detected.

    Tryptic digestion should be strongly considered in

    studies concerning membrane associated proteins. Mem-

    brane proteins, which are present in the plasma membrane

    and subcellular compartments, strongly impact signalingprocesses. Therefore, they are highly relevant for cell

    homeostasis and brain function, and are highly relevant in

    proteomic research, especially of transporter, receptor and

    channel proteins. However, different digestion enzymes or

    chemical cleavage should be tested depending on the

    interest of the study. For example, cyanogen bromide is a

    commonly used chemical substance. Helling et al. (2012)

    used this type of sample preparation for phosphoproteome

    analysis of the cytochrome c oxidase membrane protein

    complex and improved the mass spectrometric analysis of 

    integral membrane proteins. The use of cyanogen bromide

    identified six new phosphorylation sites that were not foundwhen the samples were digested with trypsin. Chy-

    motrypsin is another possible chemical for digestion.

    (Fischer et al. 2006) tested different digestion protocols to

    investigate the membrane proteome of   Corynebacterium

    glutamicum. A digestion mixture containing trypsin and

    chymotrypsin increased the identification of proteins that

    contained large hydrophobic domains. In addition, predi-

    gestion of the sample with trypsin decreased the amount of 

    soluble proteins that could mask low abundant membrane

    proteins (Fischer et al. 2006). Nevertheless, solvents may

    also impact digestion and optimization may be a key factor

    to improve the digestion protocol (Russell et al. 2001).

    Pitfalls for LMD-based proteomics

    Although LMD is a promising tool for proteomic studies of 

    brain tissues and especially of different neuronal cell types,

    there are some pitfalls that have to be considered con-

    cerning LMD processing. First, to obtain an adequate mass

    spectrometric analysis, frozen-fresh tissue must be used.

    The use of formalin-fixed paraffin-embedded tissue can

    negatively influence the results. During the formalin fixa-

    tion process, proteins undergo degradation and cross-link-

    ing (Azizadeh et al. 2015). These modifications can hinder

    accurate mass spectrometric analysis.

    Concerning frozen-fresh tissue, it has to be ensured that

    the samples are always stored on ice to prevent protein

    degradation or modifications. Cooling of samples during

    LMD process is not possible. Therefore, only one section

    of tissue should be processed at a time.

    To prevent disturbances during mass spectrometry, a

    sample cap should be used that can be filled with a col-

    lecting buffer instead of silicon filled caps.

    In addition, the tissue sample adhered to the slide must

    be completely dry to guarantee catapulting of the sample.

    The cutting line of the laser must be thin and accurate to

    avoid contamination, and the catapulting energy must be

    high enough to ensure that the tissue sample reaches the

    sample cap.

    Outlook

    Compared with traditional cell-isolation strategies, LMD

    offers outstanding possibilities for understanding brain

    function under normal and diseased conditions, as well as

    during the aging process. Changes in specific neuronal

    populations, which are normally masked in global

    Sampling

    Freezing and Storage

    (liquid nitrogen -80°C)

    Cryostate secons (in sagial orientaon)

    20 µm ckness (whole ssue analysis )

    5-10 µm ckness (single cells analysis)

    Staining

    (Cresyl violet)

    Laser microdissecon

    Sample preparaon for mass spectrometric

    analysis

    (trypc digest)

    Mass spectrometric analysis

    Fig. 6  Summary of the laser microdissection workflow. This figure

    demonstrates the resulting laser microdissection workflow of our

    study for human  substantia nigra  tissue

    1002 M. Molina et al.

     1 3

    http://www.uniprot.org/http://www.uniprot.org/http://www.uniprot.org/http://www.uniprot.org/

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    11/13

    analyses, can be analyzed in a highly specific manner,

    enabling insights into questions such as why specific

    neurons are affected in Parkinson’s disease or why the

    clinical symptoms of Alzheimer’s disease can greatly

    vary among people with similar neuropathological char-

    acteristics. The answers to these questions are integral in

    developing clinical biomarkers, therapeutic interventions,

    and potential neuroprotective agents. LMD, in combina-tion with modern analysis techniques such as mass spec-

    trometry may address some of these questions. Further,

    the possibility to combine LMD with other fractionation

    strategies could reveal a deeper insight in molecular

    processes. To continue with the example of substantia

    nigra analysis, a purification and enrichment of neu-

    romelanin granula or mitochondria out of laser

    microdissected neurons (Plum et al.  2014) could give a

    better understanding of molecular processes in cells.

    LMD has many advantages and is simple to execute. We

    demonstrated that this approach (summarized in Fig. 6) in

    combination with modern analysis techniques can be usedto characterize the neuronal proteome, and help to reveal

    the pathophysiological mechanisms within healthy and

    neurodegenerative affected brains.

    Acknowledgments   This work was supported by WTZ Brasilien, a

    project of the BMBF, Germany (01DN14023), Conselho Nacional de

    Desenvolvimento Cientı́fico e Tecnológico (CNPQ), Fundação de

    Amparo à   Pesquisa do Estado de São Paulo (Fapesp), P.U.R.E.

    (Protein Unit for Research in Europe), a project of Nordrhein-West-

    falen, a federal German state, and the HUPO Brain Proteome Project.

    The authors gratefully thank the Brazilian Brain Bank in São Paulo

    for providing tissues and Ulrich Sauer, Volker Wollscheid, Lukas

    Baran, Ulrike Weber and Gabrielle Friedemann from Carl Zeiss

    Microscopy GmbH, Munique, Germany, for technical support. Fur-

    thermore, we would like to thank Pascal C. Rauher for providing

    Fig. 1.

    Conflict of interest   There are no conflicts of interest to report.

    References

    Aaltonen KE, Ebbesson A, Wigerup C, Hedenfalk I (2011) Laser

    capture microdissection (LCM) and whole genome amplification

    (WGA) of DNA from normal breast tissue—optimization for

    genome wide array analyses. BMC Res Notes 4:69. doi:10.1186/ 

    1756-0500-4-69Azizadeh O, Atkinson MJ, Tapio S (2015) Qualitative and quanti-

    tative proteomic analysis of formalin-fixed paraffin-embedded

    (FFPE) tissue. Methods Mol Biol 1295:109–115. doi:10.1007/ 

    978-1-4939-2550-6_10

    Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger

    E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van Dongen JW,

    Vanacore N, van Swieten JC, Brice A, Meco G, van Duijn CM,

    Oostra BA, Heutink P (2003) Mutations in the DJ-1 gene

    associated with autosomal recessive early-onset parkinsonism.

    Science 299(5604):256–259. doi:10.1126/science.1077209

    Boone DR, Sell SL, Hellmich HL (2013) Laser capture microdissec-

    tion of enriched populations of neurons or single neurons for

    gene expression analysis after traumatic brain injury. J Vis Exp.

    doi:10.3791/50308

    Braak H, Braak E (1986) Nuclear configuration and neuronal types of 

    the nucleus niger in the brain of the human adult. Hum

    Neurobiol 5(2):71–82

    Burgemeister R, Gangnus R, Haar B, Schütze K, Sauer U (2003) High

    quality RNA retrieved from samples obtained by using LMPC

    (laser microdissection and pressure catapulting) technology.

    Pathol Res Pract 199(6):431–436. doi:10.1078/0344-0338-00442

    Burnet PWJ, Eastwood SL, Harrison PJ (2004) Laser-assisted

    microdissection: methods for the molecular analysis of psychi-

    atric disorders at a cellular resolution. Biol Psychiatry

    55(2):107–111. doi:10.1016/s0006-3223(03)00642-5

    Cabello CR, Thune JJ, Pakkenberg H, Pakkenberg B (2002) Ageing

    of substantia nigra in humans: cell loss may be compensated by

    hypertrophy. Neuropathol Appl Neurobiol 28(4):283–291

    Cantuti-Castelvetri I, Keller-McGandy C, Bouzou B, Asteris G, Clark 

    TW, Frosch MP, Standaert DG (2007) Effects of gender on

    nigral gene expression and parkinson disease. Neurobiol Dis

    26(3):606–614. doi:10.1016/j.nbd.2007.02.009

    Clément-Ziza M, Munnich A, Lyonnet S, Jaubert F, Besmond C

    (2008) Stabilization of RNA during laser capture microdissec-

    tion by performing experiments under argon atmosphere or using

    ethanol as a solvent in staining solutions. RNA

    14(12):2698–2704. doi:10.1261/rna.1261708

    Craven RA, Banks RE (2001) Laser capture microdissection and

    proteomics: possibilities and limitation. Proteomics

    1(10):1200–1204. doi:10.1002/1615-9861(200110)1:10\1200:

    AID-PROT1200[3.0.CO;2-Q

    Craven RA, Totty N, Harnden P, Selby PJ, Banks RE (2002) Laser

    capture microdissection and two-dimensional polyacrylamide

    gel electrophoresis: evaluation of tissue preparation and sample

    limitations. Am J Pathol 160(3):815–822. doi:10.1016/S0002-

    9440(10)64904-8

    Damier P, Hirsch EC, Agid Y, Graybiel AM (1999) The substantia

    nigra of the human brain—II. Patterns of loss of dopamine-

    containing neurons in Parkinson’s disease. Brain

    122:1437–1448. doi:10.1093/brain/122.8.1437

    De Souza AI, McGregor E, Dunn MJ, Rose ML (2004) Preparation of 

    human heart for laser microdissection and proteomics. Pro-

    teomics 4(3):578–586. doi:10.1002/pmic.200300660

    Decarlo K, Emley A, Dadzie OE, Mahalingam M (2011) Laser

    capture microdissection: methods and applications. Methods

    Mol Biol 755:1–15. doi:10.1007/978-1-61779-163-5_1

    Dickson DW (2007) Linking selective vulnerability to cell death

    mechanisms in Parkinson’s disease. Am J Pathol 170(1):16–19.

    doi:10.2353/ajpath.2007.061011

    Dos Santos A, Thiers V, Sar S, Derian N, Bensalem N, Yilmaz F, Bralet

    MP, Ducot B, Bréchot C, Demaugre F (2007) Contribution of laser

    microdissection-based technology to proteomic analysis in hepato-

    cellular carcinoma developing on cirrhosis. Proteomics Clin Appl

    1(6):545–554. doi:10.1002/prca.200600474

    Dumont D, Noben JP, Verhaert P, Stinissen P, Robben J (2006) Gel-

    free analysis of the human brain proteome: application of liquidchromatography and mass spectrometry on biopsy and autopsy

    samples. Proteomics 6(18):4967–4977. doi:10.1002/pmic.

    200600080

    Duyckaerts C, Delatour B, Potier MC (2009) Classification and basic

    pathology of Alzheimer disease. Acta Neuropathol 118(1):5–36.

    doi:10.1007/s00401-009-0532-1

    Elstner M, Morris CM, Heim K, Bender A, Mehta D, Jaros E,

    Klopstock T, Meitinger T, Turnbull DM, Prokisch H (2011)

    Expression analysis of dopaminergic neurons in Parkinson’s

    disease and aging links transcriptional dysregulation of energy

    metabolism to cell death. Acta Neuropathol 122(1):75–86.

    doi:10.1007/s00401-011-0828-9

    Enrichment of single neurons and defined brain regions from human brain tissue samples for …   1003

     1 3

    http://dx.doi.org/10.1186/1756-0500-4-69http://dx.doi.org/10.1186/1756-0500-4-69http://dx.doi.org/10.1007/978-1-4939-2550-6_10http://dx.doi.org/10.1007/978-1-4939-2550-6_10http://dx.doi.org/10.1126/science.1077209http://dx.doi.org/10.3791/50308http://dx.doi.org/10.1078/0344-0338-00442http://dx.doi.org/10.1016/s0006-3223(03)00642-5http://dx.doi.org/10.1016/j.nbd.2007.02.009http://dx.doi.org/10.1261/rna.1261708http://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1016/S0002-9440(10)64904-8http://dx.doi.org/10.1016/S0002-9440(10)64904-8http://dx.doi.org/10.1093/brain/122.8.1437http://dx.doi.org/10.1002/pmic.200300660http://dx.doi.org/10.1007/978-1-61779-163-5_1http://dx.doi.org/10.2353/ajpath.2007.061011http://dx.doi.org/10.1002/prca.200600474http://dx.doi.org/10.1002/pmic.200600080http://dx.doi.org/10.1002/pmic.200600080http://dx.doi.org/10.1007/s00401-009-0532-1http://dx.doi.org/10.1007/s00401-011-0828-9http://dx.doi.org/10.1007/s00401-011-0828-9http://dx.doi.org/10.1007/s00401-009-0532-1http://dx.doi.org/10.1002/pmic.200600080http://dx.doi.org/10.1002/pmic.200600080http://dx.doi.org/10.1002/prca.200600474http://dx.doi.org/10.2353/ajpath.2007.061011http://dx.doi.org/10.1007/978-1-61779-163-5_1http://dx.doi.org/10.1002/pmic.200300660http://dx.doi.org/10.1093/brain/122.8.1437http://dx.doi.org/10.1016/S0002-9440(10)64904-8http://dx.doi.org/10.1016/S0002-9440(10)64904-8http://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1002/1615-9861(200110)1:10%3c1200:AID-PROT1200%3e3.0.CO;2-Qhttp://dx.doi.org/10.1261/rna.1261708http://dx.doi.org/10.1016/j.nbd.2007.02.009http://dx.doi.org/10.1016/s0006-3223(03)00642-5http://dx.doi.org/10.1078/0344-0338-00442http://dx.doi.org/10.3791/50308http://dx.doi.org/10.1126/science.1077209http://dx.doi.org/10.1007/978-1-4939-2550-6_10http://dx.doi.org/10.1007/978-1-4939-2550-6_10http://dx.doi.org/10.1186/1756-0500-4-69http://dx.doi.org/10.1186/1756-0500-4-69

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    12/13

    Eltoum IA, Siegal GP, Frost AR (2002) Microdissection of histologic

    sections: past, present, and future. Adv Anat Pathol

    9(5):316–322

    Emmert-Buck MR, Bonner RF, Smith PD, Chuaqui RF, Zhuang Z,

    Goldstein SR, Weiss RA, Liotta LA (1996) Laser capture

    microdissection. Science 274(5289):998–1001

    Fischer F, Wolters D, Rögner M, Poetsch A (2006) Toward the

    complete membrane proteome: high coverage of integral mem-

    brane proteins through transmembrane peptide detection. Mol

    Cell Proteomics 5(3):444–453. doi:10.1074/mcp.M500234-

    MCP200

    Friedrich B, Euler P, Ziegler R, Kuhn A, Landwehrmeyer BG, Luthi-

    Carter R, Weiller C, Hellwig S, Zucker B (2012) Comparative

    analyses of Purkinje cell gene expression profiles reveal shared

    molecular abnormalities in models of different polyglutamine

    diseases. Brain Res 1481:37–48. doi:10.1016/j.brainres.2012.08.

    005

    Ginsberg SD, Che S (2004) Combined histochemical staining, RNA

    amplification, regional, and single cell cDNA analysis within the

    hippocampus. Lab Invest 84(8):952–962. doi:10.1038/labinvest.

    3700110

    Goldsworthy SM, Stockton PS, Trempus CS, Foley JF, Maronpot RR

    (1999) Effects of fixation on RNA extraction and amplification

    from laser capture microdissected tissue. Mol Carcinog

    25(2):86–91

    Grinberg LT, Ferretti RE, Farfel JM, Leite R, Pasqualucci CA,

    Rosemberg S, Nitrini R, Saldiva PH, Filho WJ, Group BABS

    (2007) Brain bank of the Brazilian aging brain study group—a

    milestone reached and more than 1,600 collected brains. Cell

    Tissue Bank 8(2):151–162. doi:10.1007/s10561-006-9022-z

    Gutstein HB, Morris JS (2007) Laser capture sampling and analytical

    issues in proteomics. Expert Rev Proteomics 4(5):627–637.

    doi:10.1586/14789450.4.5.627

    Hashimoto M, Bogdanovic N, Nakagawa H, Volkmann I, Aoki M,

    Winblad B, Sakai J, Tjernberg LO (2012) Analysis of microdis-

    sected neurons by 18O mass spectrometry reveals altered protein

    expression in Alzheimer’s disease. J Cell Mol Med

    16(8):1686–1700. doi:10.1111/j.1582-4934.2011.01441.x

    He S, Wang Q, He J, Pu H, Yang W, Ji J (2006) Proteomic analysis

    and comparison of the biopsy and autopsy specimen of human

    brain temporal lobe. Proteomics 6(18):4987–4996. doi:10.1002/ 

    pmic.200600078

    HeinsenH, Arzberger T, Schmitz C (2000) Celloidin mounting(embedding

    withoutinfiltration)—a new,simpleand reliable method forproducing

    serial sections of high thickness through complete human brains and

    its application to stereological and immunohistochemical investiga-

    tions. J Chem Neuroanat 20(1):49–59

    Helling S, Hüttemann M, Kadenbach B, Ramzan R, Vogt S, Marcus

    K (2012) Discovering the phosphoproteome of the hydrophobic

    cytochrome c oxidase membrane protein complex. Methods Mol

    Biol 893:345–358. doi:10.1007/978-1-61779-885-6_21

    Hood BL, Conrads TP, Veenstra TD (2006) Unravelling the proteome

    of formalin-fixed paraffin-embedded tissue. Brief Funct Geno-

    mic Proteomic 5(2):169–175. doi:10.1093/bfgp/ell017Kirana C, Ward T, Jordan TW, Rawson P, Royds J, Shi HJ, Stubbs R,

    Hood K (2009) Compatibility of toluidine blue with laser

    microdissection and saturation labeling DIGE. Proteomics

    9(2):485–490. doi:10.1002/pmic.200800197

    Kitsou E, Pan S, Zhang J, Shi M, Zabeti A, Dickson DW, Albin R,

    Gearing M, Kashima DT, Wang Y, Beyer RP, Zhou Y, Pan C,

    Caudle WM (2008) Identification of proteins in human substan-

    tia nigra. Proteomics Clin Appl 2(5):776–782. doi:10.1002/prca.

    200800028

    Kulkarni BB, Powe DG, Hopkinson A, Dua HS (2013) Optimised

    laser microdissection of the human ocular surface epithelial

    regions for microarray studies. BMC Ophthalmol 13:62. doi:10.

    1186/1471-2415-13-62

    Kumar A, Gibbs JR, Beilina A, Dillman A, Kumaran R, Trabzuni D,

    Ryten M, Walker R, Smith C, Traynor BJ, Hardy J, Singleton

    AB, Cookson MR (2013) Age-associated changes in gene

    expression in human brain and isolated neurons. Neurobiol

    Aging 34(4):1199–1209. doi:10.1016/j.neurobiolaging.2012.10.

    021

    Lawrie LC, Curran S, McLeod HL, Fothergill JE, Murray GI (2001)

    Application of laser capture microdissection and proteomics in

    colon cancer. Mol Pathol 54(4):253–258

    Liao L, Cheng D, Wang J, Duong DM, Losik TG, Gearing M, Rees

    HD, Lah JJ, Levey AI, Peng J (2004) Proteomic characterization

    of postmortem amyloid plaques isolated by laser capture

    microdissection. J Biol Chem 279(35):37061–37068. doi:10.

    1074/jbc.M403672200

    Majer A, Medina SJ, Niu Y, Abrenica B, Manguiat KJ, Frost KL,

    Philipson CS, Sorensen DL, Booth SA (2012) Early mechanisms

    of pathobiology are revealed by transcriptional temporal dynam-

    ics in hippocampal CA1 neurons of prion infected mice. PLoS

    Pathog 8(11):e1003002. doi:10.1371/journal.ppat.1003002

    Mattson MP, Magnus T (2006) Ageing and neuronal vulnerability.

    Nat Rev Neurosci 7(4):278–294

    Minjarez B, Valero Rustarazo ML, Sanchez del Pino MM, González-

    Robles A, Sosa-Melgarejo JA, Luna-Muñoz J, Mena R, Luna-

    Arias JP (2013) Identification of polypeptides in neurofibrillary

    tangles and total homogenates of brains with Alzheimer’s

    disease by tandem mass spectrometry. J Alzheimers Dis

    34(1):239–262. doi:10.3233/JAD-121480

    Mouledous L, Hunt S, Harcourt R, Harry JL, Williams KL, Gutstein

    HB (2002) Lack of compatibility of histological staining

    methods with proteomic analysis of laser-capture microdissected

    brain samples. J Biomol Tech 13(4):258–264

    Moulédous L, Hunt S, Harcourt R, Harry JL, Williams KL, Gutstein

    HB (2003) Proteomic analysis of immunostained, laser-capture

    microdissected brain samples. Electrophoresis 24(1–2):296–302.

    doi:10.1002/elps.200390026

    Plum S, Helling S, Theiss C, Leite RE, May C, Jacob-Filho W,

    Eisenacher M, Kuhlmann K, Meyer HE, Riederer P, Grinberg

    LT, Gerlach M, Marcus K (2013) Combined enrichment of 

    neuromelanin granules and synaptosomes from human substantia

    nigra pars compacta tissue for proteomic analysis. J Proteomics

    94:202–206. doi:10.1016/j.jprot.2013.07.015

    Plum S, Steinbach S, Abel L, Marcus K, Helling S, May C (2014)

    Proteomics in neurodegenerative diseases: methods for obtaining

    a closer look at the neuronal proteome. Proteomics Clin Appl.

    doi:10.1002/prca.201400030

    Rekhter MD, Chen J (2001) Molecular analysis of complex tissues is

    facilitated by laser capture microdissection: critical role of 

    upstream tissue processing. Cell Biochem Biophys

    35(1):103–113. doi:10.1385/CBB:35:1:103

    Russell WK, Park ZY, Russell DH (2001) Proteolysis in mixed

    organic-aqueous solvent systems: applications for peptide mass

    mapping using mass spectrometry. Anal Chem73(11):2682–2685

    Simunovic F, Yi M, Wang Y, Macey L, Brown LT, Krichevsky AM,

    Andersen SL, Stephens RM, Benes FM, Sonntag KC (2009)

    Gene expression profiling of substantia nigra dopamine neurons:

    further insights into Parkinson’s disease pathology. Brain 132(Pt

    7):1795–1809. doi:10.1093/brain/awn323

    Sitek B, Lüttges J, Marcus K, Klöppel G, Schmiegel W, Meyer HE,

    Hahn SA, Stühler K (2005) Application of fluorescence difference

    gel electrophoresis saturation labelling for the analysis of microdis-

    sected precursor lesions of pancreatic ductal adenocarcinoma.

    Proteomics 5(10):2665–2679. doi:10.1002/pmic.200401298

    1004 M. Molina et al.

     1 3

    http://dx.doi.org/10.1074/mcp.M500234-MCP200http://dx.doi.org/10.1074/mcp.M500234-MCP200http://dx.doi.org/10.1016/j.brainres.2012.08.005http://dx.doi.org/10.1016/j.brainres.2012.08.005http://dx.doi.org/10.1038/labinvest.3700110http://dx.doi.org/10.1038/labinvest.3700110http://dx.doi.org/10.1007/s10561-006-9022-zhttp://dx.doi.org/10.1586/14789450.4.5.627http://dx.doi.org/10.1111/j.1582-4934.2011.01441.xhttp://dx.doi.org/10.1002/pmic.200600078http://dx.doi.org/10.1002/pmic.200600078http://dx.doi.org/10.1007/978-1-61779-885-6_21http://dx.doi.org/10.1093/bfgp/ell017http://dx.doi.org/10.1002/pmic.200800197http://dx.doi.org/10.1002/prca.200800028http://dx.doi.org/10.1002/prca.200800028http://dx.doi.org/10.1186/1471-2415-13-62http://dx.doi.org/10.1186/1471-2415-13-62http://dx.doi.org/10.1016/j.neurobiolaging.2012.10.021http://dx.doi.org/10.1016/j.neurobiolaging.2012.10.021http://dx.doi.org/10.1074/jbc.M403672200http://dx.doi.org/10.1074/jbc.M403672200http://dx.doi.org/10.1371/journal.ppat.1003002http://dx.doi.org/10.3233/JAD-121480http://dx.doi.org/10.1002/elps.200390026http://dx.doi.org/10.1016/j.jprot.2013.07.015http://dx.doi.org/10.1002/prca.201400030http://dx.doi.org/10.1385/CBB:35:1:103http://dx.doi.org/10.1093/brain/awn323http://dx.doi.org/10.1002/pmic.200401298http://dx.doi.org/10.1002/pmic.200401298http://dx.doi.org/10.1093/brain/awn323http://dx.doi.org/10.1385/CBB:35:1:103http://dx.doi.org/10.1002/prca.201400030http://dx.doi.org/10.1016/j.jprot.2013.07.015http://dx.doi.org/10.1002/elps.200390026http://dx.doi.org/10.3233/JAD-121480http://dx.doi.org/10.1371/journal.ppat.1003002http://dx.doi.org/10.1074/jbc.M403672200http://dx.doi.org/10.1074/jbc.M403672200http://dx.doi.org/10.1016/j.neurobiolaging.2012.10.021http://dx.doi.org/10.1016/j.neurobiolaging.2012.10.021http://dx.doi.org/10.1186/1471-2415-13-62http://dx.doi.org/10.1186/1471-2415-13-62http://dx.doi.org/10.1002/prca.200800028http://dx.doi.org/10.1002/prca.200800028http://dx.doi.org/10.1002/pmic.200800197http://dx.doi.org/10.1093/bfgp/ell017http://dx.doi.org/10.1007/978-1-61779-885-6_21http://dx.doi.org/10.1002/pmic.200600078http://dx.doi.org/10.1002/pmic.200600078http://dx.doi.org/10.1111/j.1582-4934.2011.01441.xhttp://dx.doi.org/10.1586/14789450.4.5.627http://dx.doi.org/10.1007/s10561-006-9022-zhttp://dx.doi.org/10.1038/labinvest.3700110http://dx.doi.org/10.1038/labinvest.3700110http://dx.doi.org/10.1016/j.brainres.2012.08.005http://dx.doi.org/10.1016/j.brainres.2012.08.005http://dx.doi.org/10.1074/mcp.M500234-MCP200http://dx.doi.org/10.1074/mcp.M500234-MCP200

  • 8/17/2019 Enrichment of single neurons and defined brain regions from human brain tissue samples for subsequent proteom…

    13/13

    Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R,

    Goedert M (1997) Alpha-synuclein in Lewy bodies. Nature

    388(6645):839–840. doi:10.1038/42166

    Sridharan G, Shankar AA (2012) Toluidine blue: a review of its

    chemistry and clinical utility. J Oral Maxillofac Pathol

    16(2):251–255. doi:10.4103/0973-029X.99081

    Theofilas P, Polichiso L, Wang X, Lima LC, Alho AT, Leite RE,

    Suemoto CK, Pasqualucci CA, Jacob-Filho W, Heinsen H,

    Grinberg LT, Group BABS (2014) A novel approach for

    integrative studies on neurodegenerative diseases in human

    brains. J Neurosci Methods 226:171–183. doi:10.1016/j.jneu

    meth.2014.01.030

    Wang WZ, Oeschger FM, Lee S, Molnár Z (2009) High quality RNA

    from multiple brain regions simultaneously acquired by laser

    capture microdissection. BMC Mol Biol 10:69. doi:10.1186/ 

    1471-2199-10-69

    Werner CJ, Heyny-von Haussen R, Mall G, Wolf S (2008) Proteome

    analysis of human substantia nigra in Parkinson’s disease.

    Proteome Sci 6:8. doi:10.1186/1477-5956-6-8

    Enrichment of single neurons and defined brain regions from human brain tissue samples for …   1005

     1 3

    http://dx.doi.org/10.1038/42166http://dx.doi.org/10.4103/0973-029X.99081http://dx.doi.org/10.1016/j.jneumeth.2014.01.030http://dx.doi.org/10.1016/j.jneumeth.2014.01.030http://dx.doi.org/10.1186/1471-2199-10-69http://dx.doi.org/10.1186/1471-2199-10-69http://dx.doi.org/10.1186/1477-5956-6-8http://dx.doi.org/10.1186/1477-5956-6-8http://dx.doi.org/10.1186/1471-2199-10-69http://dx.doi.org/10.1186/1471-2199-10-69http://dx.doi.org/10.1016/j.jneumeth.2014.01.030http://dx.doi.org/10.1016/j.jneumeth.2014.01.030http://dx.doi.org/10.4103/0973-029X.99081http://dx.doi.org/10.1038/42166