Skip to main content
Biomarker Insights logoLink to Biomarker Insights
. 2009 Oct 28;4:135–164. doi: 10.4137/bmi.s2965

An Optimal Protocol to Analyze the Rat Spinal Cord Proteome

F Gil-Dones 1, S Alonso-Orgaz 1, G Avila 2, T Martin-Rojas 1, V Moral-Darde 3, G Barroso 3, F Vivanco 4,5, J Scott-Taylor 2, MG Barderas 1
PMCID: PMC2796866  PMID: 20029654

Abstract

Since the function of the spinal cord depends on the proteins found there, better defing the normal Spinal Cord Proteome is an important and challenging task. Although brain and cerebrospinal fluid samples from patients with different central nervous system (CNS) disorders have been studied, a thorough examination of specific spinal cord proteins and the changes induced by injury or associated to conditions such as neurodegeneration, spasticity and neuropathies has yet to be performed. In the present study, we aimed to describe total protein content in the spinal cord of healthy rats, employing different proteomics tools. Accordingly, we have developed a fast, easy, and reproducible sequential protocol for protein extraction from rat spinal cords. We employed conventional two dimensional electrophoresis (2DE) in different pH ranges (eg. 4–7, 3–11 NL) combined with identification by mass spectrometry (MALDI-TOF/TOF), as well as first dimension protein separation combined with Liquid Chromatography Mass Spectrometry/Mass Spectrometry (LC-MS/MS) to maximise the benefits of this technology. The value of these techniques is demonstrated here by the identification of several proteins known to be associated with neuroglial structures, neurotransmission, cell survival and nerve growth in the central nervous system. Furthermore this study identified many spinal proteins that have not previously been described in the literature and which may play an important role as either sensitive biomarkers of dysfunction or of recovery after Spinal Cord Injury.

Keywords: proteomics, two dimensional electrophoresis (2-DE), Liquid Chromatography Mass Spectrometry/Mass Spectrometry (LC-MS MS), spinal cord, proteome

Introduction

Spinal cord injury (SCI) has a significant disabling and lifelong effect on many people and as such, it represents a major challenge for successful health care management. SCI is a devastating neurotrauma insult that can lead to the loss of sensory and motor function below the level of injury.1, 2 The progressive pathological changes initiated by SCI include complex and evolving molecular cascades whose interrelationships are not fully understood, and many molecules involved in these processes remain to be discovered.37 To date, brain and cerebrospinal fluid samples from patients with different central nervous system (CNS) disorders have been studied extensively using different biochemical assays.812 However, relatively few studies have focused on spinal cord protein content, and the changes induced after spinal neurotrama or in association with symptoms such as spasticity or neuropathic pain. Indeed, recent studies have been conducted to screen for a wide range of proteins following SCI using comparative proteomic technologies.1317

The tremendous advances in molecular biology, mainly in the field of genomics and proteomics, open the possibility to understand the mechanisms underlying many neuropathologies. After genomics, proteomics is often considered the next logical step to study biological systems, with the added capacity to describe the spatiotemporal differences in protein expression, both in normal and pathological tissue.1820 The proteome represents all the proteins expressed by a genome, cell, tissue or organism at a given time under defined physiological conditions. Since most physiological body functions reflect the integrity of their proteins, understanding the complex biological processes active in the spinal cord during pathological conditions like SCI requires the key proteins involved at an early stage of the neurotrauma21, 22 (acute phase) and during injury progression to be identified.

Proteomic analysis is now a key biomedical tool to establish protein maps that can assist in biomarker discovery and in the identification of therapeutic targets. In this respect, an important and challenging task is to develop protocols designed to extend our knowledge of the spinal cord (SC) protein profile that combine mass spectrometry with two dimensional gels (2-DE). Until now most studies have focussed on one protein or on a small number of proteins using standard techniques such as Western blotting, immunohistochemistry or RT-PCR, which fail to provide complete information regarding the general physiological state of the SC. In contrast, proteomic analysis is useful as multiple molecules can be assayed simultaneously using separation techniques combined with the powerful new mass spectrometry technologies, such as MALDI-TOF/TOF (Matrix Assisted Laser Desorption Ionization-Time of Flight/Time of Flight Mass Spectrometry), SELDI-TOF (Surface Enhanced Laser Desorption Ionization Time Of Flight Mass Spectrometry), Protein Arrays, LCM (Laser Capture Microdissection), MS-Imaging, LC-MS (Liquid Chromatography Mass Spectrometry), TOF-SIMS (Time of Flight Secondary Ion Mass Spectrometry).2329

However, the development of global protein analysis using proteomic technologies needs to address several limitations and challenges. An important tool applied to study the proteome is 2-DE, whereby proteins are first separated by isoelectric focusing (IEF) and then based on their molecular weight by SDS-PAGE (sodium dodecyl sulphate polyacrylamide gel electrophoresis).3032 However, this technique presents some important limitations that could be resolved by the application of other proteomics tools such as LC-MS/MS.33 In addition, there is a need to develop efficient protocols to extract most of the proteins present in the spinal cord, given the limitations of each technique and the complexity of the proteome.

In this technical report, we present a fast, easy and reproducible protocol to extract SC proteins and analyze its proteome (Fig. 1). The aim of this study is to describe the majority of the proteins extracted from the rat SC proteome by employing conventional 2-DE spot maps over different pH ranges and MALDI-TOF/TOF for their identification, in combination with LC-MS/MS to maximise the utility of this technology. The application of this newly developed optimal protein extraction protocol compatible with 2-DE and LC-MS/MS will permit future translational studies to identify the main pathophysiological mechanisms associated with SCI.

Figure 1.

Figure 1.

The proteomic platforms used in this study and a flowchart demonstrating the strategy for the rat spinal cord analysis. Schematic illustration of the proteomics methods used to characterise the rat spinal cord proteome.

Materials and Methods

Collection of rat spinal cords

Thoracico-lumbar spinal cord tissue was obtained from 12 week old male adult Wistar rats (n = 6: Harlan SA, Milano, Italy) weighing between 300–400 g sacrificed with an intraperitoneal overdose of Sodium Pentobarbital (Dolethal, Norman SA). Shortly afterwards, the spinal cord tissue was extracted using hydraulic pressure applied to the caudal vertebral canal, whereupon the tissue was cleaned with a saline solution (0.9%). The thoracico-lumbar segments were carefully dissected out and then frozen and stored at −20 °C until analyses.

Rat spinal cord processing: protein extraction

After removal from −20 °C storage, the tissue was maintained at 4 °C in PBS solution and all the following steps in the protocol were performed at 4 °C (Fig. 2).

Figure 2.

Figure 2.

The protocol to extract proteins from the rat spinal cord. A) After surgery the spinal cord tissue was washed in saline buffer to eliminate blood contaminants and tissue was homogenized (Buffer 1) and later a new extraction of proteins was realized using buffer 2. Supernatant A, containing most of the soluble proteins and supernatant B, containing membrane and hydrophobic proteins were analysed separately in 2-DE in order to check the efficiency of the protein extraction protocol. B) Supernatant A and B were mixed and analysed by 2-DE.

Firstly, the tissue was washed 3 times in PBS to remove blood contaminants and it was then ground into a powder with a mortar in Liquid Nitrogen. This powder (0.3 g) was resuspended in 300 μL of protein extraction buffer 1 (Tris 10 mM [pH 7.5], 500 mM NaCl 0.1%, Triton x-100, 1% β-mercaptoethanol and 1 mM PMSF).34 The homogenate was sonicated for 5 minutes and centrifuged at 21,000 g (5840 R Eppendorf) for 15 minutes at 4 °C to precipitate the membrane and tissue debris. The supernatant (supernatant A), containing most of the soluble proteins was collected and stored at 4 °C. The pellet was then dissolved in a buffer containing 7 M Urea, 2 M Thiourea, 5% CHAPS,35, 36 and it was again centrifuged at 21,000g to obtain a second supernatant separated from the pellet of tissue debris (Supernatant B), mainly composed of membrane proteins. The tissue debris was then resuspended in protein loading buffer (Tris 0.5 M [pH 8.0], SDS 10%, Glycerol, β-mercaptoethanol and bromophenol blue 0.02%) and the protein concentration was determined by the Bradford-Lowry method using the Bio-Rad protein assay commercial Kit.37 Finally, the protein composition was analyzed by resolving 25 μg of total protein content from each sample by SDS-PAGE 12% (Acrylamide/Bisacrylamide 30%/0.8% v/v).

Two-dimensional electrophoresis (2-DE)

All chemicals and instruments used for 2-DE gels have been described previously.35, 36 Both the soluble and hydrophobic protein extracts were mixed and dialysed against 2 mM Tris buffer using Mini dialysis Kit 1 kDa cut-off (GE Healthcare). Subsequently, 300 μg of each protein extract was cleaned with the 2 D Clean up Kit (GE-Healthcare) and resuspended in rehydration buffer (7 M Urea, 2 M Thiourea, 4% CHAPS, 1%–2% Ampholites and 1% TBP: Bio-Rad). Isoelectric focusing (IEF) was performed in an IPGphor unit (GE Healthcare). The strips (17 cm and pH 4–7: Bio-Rad, or 24 cm pH 3–11 NL—non-lineal: GE Healthcare) were actively rehydrated at 20 °C for 12 h at 50 V to enhance protein uptake, and the voltage was then increased according to the following program: 500 V for 30 minutes, 1000 V for 1 h, 1000–2000 V in 1 h (gradient), 2000–5000 V in 2 h (gradient), 5000–8000 V in 1 h (gradient), 8000 V to a total 88,000 V/h.

Subsequently, the strips IEF were equilibrated as described previously35, 36 and the second dimension (SDS-PAGE) was run according to Laemmli’s method,38 using a Protean II system (Bio-Rad) at 1 W/gel at 20 °C overnight. Gels were fixed and stained by Silver Staining (GE Healthcare, according to the manufacturer’s instructions) and they were then scanned with a GS-800 Calibrated Densitometer (Bio-Rad). Evaluation of the 2-DE gels was performed using PDQuest 2DE Gel Analysis Software version 8.0.1 (Bio-Rad). Reproducibility was tested comparing the variation within the different gels in the same group using the same software.

In gel digestion

Spots (200) were manually excised, automatically digested with “Ettan Digester” (GE Healthcare) and identified at the HNP Proteomic Unit according to Schevchenko et al39 with minor modifications.40 Gel plugs were reduced with 10 mM dithiothreitol (Sigma Aldrich) in 50 mM ammonium bicarbonate (99% purity; Scharlau) and by alkylation with 55 mM iodoacetamide (Sigma Aldrich) in 50 mM ammonium bicarbonate. The gel fragments were then rinsed with 50 mM ammonium bicarbonate in 50%. Methanol (gradient, HPLC grade, Scharlau) and acetonitrile (gradient, HPLC grade, Scharlau), and they were dried in a Speedvac. Modified porcine trypsin (sequencing grade; Promega, Madison, WI, USA) was added to the dry gel pieces at a final concentration of 20 ng/μl in 20 mM ammonium bicarbonate and the digestion proceeded at 37 °C overnight. Finally, 70% aqueous acetonitrile and 0.1% formic acid (99.5% purity; Sigma Aldrich) was added for peptide extraction.

Protein identification by MALDI-TOF/TOF

An aliquot of each digestion was mixed with an aliquot of the matrix solution (3 mg/mL α-cyano-4-Hydroxycinnamic acid: Sigma Aldrich) in 30% ACN, 15% 2-propanol and 0.1% TFA. This mixture was pipetted directly onto the stainless steel sample plate of the mass spectrometer (384 Opti-TOF 123 × 81 mm MALDI: Applied Biosystem) and dried at room temperature.

The MALDI-MS/MS data were obtained in an automated analysis loop using a 4800 Plus MALDI TOF/TOF Analyzer (Applied Biosystems). Spectra were acquired in the reflector positive-ion mode with a Nd:YAG laser (355 nm wavelength at a frequency of 200 Hz), and between 100 and 2000 individual spectra were averaged. The experiments were acquired in a uniform mode with a fixed laser intensity. For the MS/MS 1 kV analysis mode, precursors were accelerated to 8 kV in source 1, and they were selected at a relative resolution of 350 (FWHM) with metastable suppression. Fragment ions generated by collision with air in a CID chamber were further accelerated at 15 kV in source 2. Mass data was analysed automatically with the 4000 Series Explorer Software version 3.5.3 (Applied Biosystems). Internal calibration of MALDI-TOF mass spectra was performed using two trypsin autolysis ions with m/z = 842.510 and m/z = 2211.105. For calibration in the MS/MS mode, the fragment ion spectra obtained from Glub-fibrinopeptide were used (4700 Cal Mix, Applied Biosystems). MALDI-MS and MS/MS data were combined through the GPS Explorer Software Version 3.6 to search a nonredundant protein database (Swissprot 56.7) using the Mascot software (version 2.2, Matrix Science), employing the following parameters: 50 ppm precursor tolerance; 0.6 Da MS/MS fragment tolerance; and allowing 1 missed cleavage, carbamidomethyl cysteines and methionine oxidation as a modification. The MALDI-MS/MS spectra and database search results were manually inspected in detail using the aforementioned software.

LC-MS/MS and database searching

Sample preparation

Total spinal cord proteins (50 μg) were resolve by one dimensional (1-D) SDS-PAGE 12%. Each lane in the 1-D gel was divided into 24 gel slices that were manually excised and then digested automatically using the Ettan Digester (GE Healthcare). The digestion was performed according to Schevchenko et al39 with minor modifications40 and using Modified porcine trypsin (sequencing grade; Promega, Madison, WI, USA) diluted to a final concentration of 20 ng/μl in 20 mM ammonium bicarbonate. The gel slices were incubated with 10 mM dithiothreitol (Sigma Aldrich) in 50 mM ammonium bicarbonate (99% purity; Scharlau) for 30 minutes at 56 °C and after reduction, they were alkylated with 55 mM iodoacetamide (Sigma Aldrich) in 50 mM ammonium bicarbonate for 20 minutes at RT. Gel plugs were washed with 50 mM ammonium bicarbonate in 50% methanol (gradient, HPLC grade, Scharlau), rinsed in acetonitrile (gradient, HPLC grade, Scharlau) and dried in a Speedvac. Dry gel pieces were then embedded in sequencing grade modified porcine trypsin (20 ng/μL: Promega, Madison, WI, USA) and after digestion at 37 °C overnight, the peptides were extracted with 70% acetonitrile (ACN) in 0.1% formic acid (99.5% purity; Sigma Aldrich). Finally, the samples were dried in a speedvac and resuspended in 98% water with 0.1% formic acid (FA) and 2% ACN.

LC-MS/MS and database searching

The LC/MSMS system was comprised of a TEMPO nano LC system (Applied Biosystems) combined with a nano LC Autosampler. Each sample was injected in three replicates (3 μL) using mobile phase A (2% ACN/98% water, 0.1% FA) at a flow rate of 10 μL/minute for 10 minutes. Peptides were loaded onto a μ-Precolumn Cartridge (Acclaim Pep Map 100 C18, 5 μm, 100Å; 300 μm i.d. × 5 mm, LC Packings) to preconcentrate and desalt samples. The RPLC was performed on a C18 column (Acclaim Pep Map 100 C18, 3 μm, 100Å; NAN75-15-03-C18PM, 75 μm I.D. × 15 cm, LC Packings) using mobile phase A (2% ACN/98% water, 0.1% FA) and mobile phase B (98% ACN/2% water, 0.1% FA). Peptides were eluted at a flow rate of 300 nL/minute over the following gradient: initial conditions of 5% B that increased to 50% B over 70 minutes, 50 to 95% B for 1 minute and then 95% B for 3 minutes, returning to the initial conditions (5% B) over 2 minutes and maintaining these conditions for a further 14 minutes.

The LC-MS/MS analysis was performed on an AB/MDS Sciex 4000 Q TRAP System with NanoSprayII Source (Applied Biosystems). The TEMPO nano LC system and 4000 QTRAP were both controlled by Analyst Software v.1.4.2.

All the MS and MS/MS data were obtained in positive ion mode, with an ion spray voltage of 2800 V and a declustering of 85V. Nanoflow interface was heated at 150 °C, and the source gas 1 and curtain gas were set to 20 and 10, respectively. Nitrogen was applied as both curtain and collision gas. An Information Dependent Acquisition (IDA) method was programmed, with a full scan Enhanced MS (EMS) experiment at 4000 amu/s for ion profiling that was followed by an enhanced resolution (ER) MS experiment at 250 amu/s. The ER experiment permitted charge state recognition that was further submitted to IDA criteria to select precursor ions, and to estimate the collision energy to fragment them. These IDA criteria were set to select the 8 most intense double, triple or quadruple charged ions from 400–1200 m/z that exceed 100,000 counts for fragmentation in the LINAC collision cell. Isotopes within a 4.0 amu window and with a mass tolerance of 1,000,000 mmu were excluded. These 8 ions were submitted to 8 independent Enhanced Product Ion (EPI) MS/MS experiments at 4000 amu/s with Dynamic Fill Time (DFT). The total number of MS and MS/MS experiments per cycle was 10 (1 EMS, 1 ER and 8 EPI), resulting in a total cycle time of 5.0058 s.

Analyst software creates wiff format files including all the spectra data that were batch-processed with ProteinPilot Software 2.0.1 (Applied Biosystems/MDS Sciex). This software automatically generated peak lists that were searched against the Swissprot database version 56.7 using Paragon Algorithm (Applied Biosystems). Settings in the Paragon Algorithm included a detected protein threshold >1.0 (90%), Iodoacetamide was selected for Cys alkylation and Gel-based ID was selected as a special factor.

Results

Rat spinal cord processing and protein extraction

To describe the complete proteome of an organ or tissue, it is necessary to establish an efficient extraction protocol to maximize protein recovery. Here, we present a flowchart to explain our approach to the proteomic study of rat SC (Fig. 1) and a schedule of the consecutive extraction protocol (Fig. 2). This method was based on two consecutive steps using two distinct extraction buffers, the first of which extracted the more soluble proteins, while the second was designed to dissolve the membrane and hydrophobic proteins that were assumed to be abundant in SC tissue.

Sample preparation and conventional 2-DE

In order to reduce the presence of lipids and other interfering substances, samples were sonicated, filtered with a micro spin-filter (SIGMA) and cleaned with the Clean-up Kit (GE Healthcare). We tested different pH ranges (pH 4–7; pH 3–11 NL) in order to select that which was optimal to detect the maximal number of spots with the greatest resolution. Spinal Cord protein extracts were quantified and approximately 300 μg was loaded onto each 2-DE gel. After analysis with the PD-Quest software (Bio-Rad), around 300 spots were detected by 2-DE in the 4–7 pH range (Fig. 3A). However, these gels did not present an homogeneous spot distribution due to the fact that most of them co-localized in the same area.

Figure 3.

Figure 3.

2-DE gel images. 2-DE was performed with IPG strips at different pH ranges: A) pH 4–7 (left) and B) pH 3–11 NL (right). C) 2-DE gel performed with 3–11 NL IPG strip and 9%–16% acrylamide/bisacrylamide.

For this reason, we performed 2-DE gels with 24 cm pH 3–11 NL IPG strips. We obtained a good distribution, definition and a large number of spots under these conditions, although some streaking in the 53–96 kDa molecular weight region could be due to the high concentration of these abundant proteins. This problem did not arise in the same region of the pH 4–7 2 D gels. Hence, the use of the two types of gels with complementary pH ranges (pH 4–7 and 3–11 NL) helped improve the overall spot resolution, as reported previously.35

Thus, more than 1000 spots were detected after PD-Quest software analysis, improving the resolution and permitting the subsequent identification of the spots (Fig. 3B). Reproducibility was tested by comparing the variation within the different gels in the same group using the PD Quest 8.0 software. An analysis of 1126 spots revealed a coefficient of variation (CV) < 50% for 90.4% of the spots in same group of gels. Among these, a CV < 30% was obtained for 67.1% of the spots. These data confirmed the high reproducibility of the gels obtained with the method used.

Protein identification (MALDI-TOF/TOF)

In order to verify the effectiveness of our methodology, 200 spots were chosen at random, they were excised from the stained 2-DE gels, digested and the resultant tryptic peptides were deposited an a MALDI plaque and applied in a 4800 Plus MALDI-TOF/TOF Analyzer (Applied Biosystem). Proteins were identified by Peptide mass fingerprinting using the “MASCOT” search engine (www.matrixscience.com). All the spots were identified and they corresponded to 128 proteins (Fig. 4), as summarized in Table 1 where their molecular weight, isoelectric point, cellular sublocalization and function are shown.

Figure 4.

Figure 4.

Preparative 2-DE Gel (700 μg). Spot Map of the proteins identified. The characterization of the spots identified is shown in Table 1.

Table 1.

Spots identified with 2-DE gel (pH: 3-11 NL). The data indicates accession number, the isoelectric point (theoretical and experimental), molecular weight (theoretical and experimental), subcellular localization and recognised function.

Protein name Accession no. MALDI-TOF/Spot Nº LC-MS Q-TRAP MW Da theorical MW Da experimental pI theorical pI experimental Subcellular localization Function
Myelin basic protein S MBP_RAT Spot Nº 2 Identified 21,489.00 15 11.25 10.8 Cell mb. Structural…
Tubulin polymerization-promoting protein family TPPP3_BOVIN Spot Nº 3 Identified 18,931.00 24.5 9.18 10.8 Myelin mb. Structural…
Protein NipSnap homolog 1 NIPS1_MOUSE Spot Nº 6 No 33,342.00 35 9.48 10.8 Mit inn mb. Others.
Prohibitin-2 PHB2_MOUSE Spot Nº 8 No 33,276.00 40 9.83 10.8 Mit inn mb; Cp; N. Others.
Aspartate aminotransferase, mitochondrial AATM_RAT Spot Nº 10 Identified 47,284.00 51 9.13 10.8 Mit; Cell mb. Metabolism.
Elongation factor 1-alpha 1 EF1A1_CRIGR Spot Nº 11 No 50,109.00 56 9.10 10.8 Cp. Prot regulation.
Profilin-1 PROF1_RAT Spot Nº 13 Identified 14,948.00 13 8.46 9.6 Cp. Structural…
Peptidyl-prolyl cis-trans isomerase A PPIA_RAT Spot Nº 16 Identified 17,863.00 18 8.34 9.5 Cp. Protregulation.
Destrin OS DEST_RAT Spot Nº 17 Identified 18.522,00 20 8.19 9.2 Structural…
Cofilin-1 COF1_RAT Spot Nº 18 Identified 18,521.00 23 8.22 9.2 N; Cp. Structural…
Peroxiredoxin-1 PRDX1_RAT Spot Nº 19 Identified 22,095.00 31 8.27 9.5 Cp; Mel. Stress Resp...
Peroxiredoxin-1 PRDX1_RAT Spot Nº 20 Identified 22,095.00 31 8.27 9.2 Cp; Mel. Stress Resp...
Proteasome subunit beta type-1 PSB1_MOUSE Spot Nº 21 Identified 26,355.00 33 7.67 9.4 Cp; N. Prot regulation.
Glutathione S-transferase alpha-3 GSTA3_RAT Spot Nº 22 No 25,303.00 35 8.78 9.6 Cp. Stress Resp...
Glutathione S-transferase Mu 1 GSTM1_RAT Spot Nº 23 No 25,897.00 34.2 8.27 9.4 Cp. Stress Resp...
Proteasome subunit alpha type-7 PSA7_MOUSE Spot Nº 24 Identified 27,838.00 35.5 8.59 9.6 Cp; N. Prot regulation.
ATP synthase subunit alpha liver isoform, mitochondrial ATPA2_BOVIN Spot Nº 25 No 38,852.00 37 9.57 9.6 Mit inn mb. Metabolism.
ATP synthase subunit gamma, mitochondrial ATPG_RAT Spot Nº 26 Identified 30,172.00 38 8.87 9.5 Mit inn mb. Metabolism.
Voltage-dependent anion-selective channel protein 1 VDAC1_RABIT Spot Nº 27 Identified 30,722.00 39 8.62 9.5 Mit out mb; Cell mb. Stress Resp...
Malate dehydrogenase, mitochondrial MDHM_RAT Spot Nº 28 Identified 35,661.00 45 8.93 9.9 Mit. Metabolism.
Malate dehydrogenase, mitochondrial MDHM_RAT Spot Nº 29 Identified 35,661.00 45 8.93 9.65 Mit. Metabolism.
L-lactate dehydrogenase A chain LDHA_RAT Spot Nº 30 Identified 36,427.00 45 8.45 9.5 Cp. Metabolism.
Malate dehydrogenase, mitochondrial MDHM_RAT Spot Nº 31 Identified 35,661.00 49 8.93 9.5 Mit. Metabolism.
Malate dehydrogenase, mitochondrial MDHM_RAT Spot Nº 32 Identified 35,661.00 45 8.93 9.4 Mit. Metabolism.
Glyceraldehyde-3-phosphate dehydrogenase G3P_RAT Spot Nº 33 Identified 35,787.00 49 8.44 9.4 Cp. Metabolism.
Fructose-bisphosphate aldolase A ALDOA_RAT Spot Nº 34 Identified 39,327.00 51 8.31 9.4 Mit. Metabolism.
Cytochrome b-c1 complex subunit 2, mitochondrial QCR2_RAT Spot Nº 35 Identified 48,366.00 53 9.16 9.5 Mit inn mb. Metabolism.
2’,3’-cyclic-nucleotide 3’-phosphodiesterase CN37_RAT Spot Nº 36 Identified 47,239.00 60 9.03 9.4 Cell mb; Mel. Metabolism.
Septin-7 SEPT7_RAT Spot Nº 37 No 50,518.00 66 8.73 9.4 Cp. Structural…
Glyceraldehyde-3-phosphate dehydrogenase G3P_RAT Spot Nº 38 Identified 35,787.00 49 8.44 9.2 Cp. Metabolism.
Glyceraldehyde-3-phosphate dehydrogenase G3P_RAT Spot Nº 39 Identified 35,787.00 50 8.44 9.2 Cp. Metabolism.
Fumarate hydratase, mitochondrial FUMH_RAT Spot Nº 41 No 54,429.00 60 9.06 9.2 Mit; Cp. Metabolism.
ATP synthase subunit alpha, mitochondrial ATPA_RAT Spot Nº 42 Identified 59,717.00 75 9.22 9.1 Mit inn mb. Metabolism.
T-complex protein 1 subunit eta TCPH_PONAB Spot Nº 43 No 59,329.00 80 7.55 9.3 Cp. Prot regulation.
Phosphoglycerate kinase 1 PGK1_RAT Spot Nº 44 Identified 44,510.00 53 8.02 9.1 Cp. Metabolism.
Fumarate hydratase, mitochondrial FUMH_RAT Spot Nº 45 No 54,429.00 60 9.06 8.9 Mit; Cp. Metabolism.
Citrate synthase, mitochondrial CISY_RAT Spot Nº 46 Identified 51,833.00 53 8.53 8.88 Mit. Metabolism.
Isocitrate dehydrogenase [NAD] subunit beta, IDH3B_RAT Spot Nº 47 Identified 42,327.00 51 8.89 8.87 Mit. Metabolism.
Fructose-bisphosphate aldolase A ALDOA_RAT Spot Nº 48 Identified 39,327.00 51 8.31 8.8 Mit. Metabolism.
Glyceraldehyde-3-phosphate dehydrogenase G3P_RAT Spot Nº 49 Identified 35,787.00 49 8.44 8.9 Cp. Metabolism.
Thiosulfate sulfurtransferase THTR_RAT Spot Nº 50 Identified 33,386.00 45 7.71 9.2 Mit. Carrier.
Hydroxyacyl-coenzyme A dehydrogenase, HCDH_RAT Spot Nº 51 No 34,426.00 40 8.83 9.2 Mit. Metabolism.
Voltage-dependent anion-selective channel protein 1 VDAC1_RABIT Spot Nº 52 Identified 30,722.00 39 8.62 9.2 Mit out mb; Cell mb. Stress Resp...
Superoxide dismutase [Mn], mitochondrial SODM_RAT Spot Nº 56 Identified 24,659.00 30 8.96 8.88 Mit. Stress Resp...
Peptidyl-prolyl cis-trans isomerase A PPIA_RAT Spot Nº 57 Identified 17,863.00 18.3 8.34 8.9 Cp. Prot regulation.
Peptidyl-prolyl cis-trans isomerase A PPIA_RAT Spot Nº 58 Identified 17,863.00 18.3 8.34 8.7 Cp. Prot regulation.
Cofilin-2 OS=Homo sapiens COF2_HUMAN Spot Nº 59 Identified 18,725.00 24 7.66 8.2 N; Cp. Structural…
Superoxide dismutase [Mn], mitochondrial SODM_RAT Spot Nº 60 Identified 24,659.00 30 8.96 8.3 Mit. Stress Resp...
Adenylate kinase isoenzyme 1 KAD1_RAT Spot Nº 60 No 21,570.00 31.5 7.66 8.5 Cp. Metabolism.
Glutathione S-transferase P GSTP1_RAT Spot Nº 61 Identified 23,424.00 31.3 6.89 8.6 Stress Resp...
Peroxiredoxin-1 PRDX1_RAT Spot Nº 62 Identified 22,095.00 30 8.27 8.7 Cp; Mel. Stress Resp...
Superoxide dismutase [Mn], mitochondrial SODM_RAT Spot Nº 63 Identified 24,659.00 34 8.96 8.7 Mit. Stress Resp...
Cytochrome b-c1 complex subunit Rieske UCRI_RAT Spot Nº 64 Identified 29,427.00 35 9.04 8.7 Mit inn mb. Metabolism.
Dihydropteridine reductase DHPR_RAT Spot Nº 65 No 25,536.00 36 7.67 8.7 Stress Resp...
Electron transfer flavoprotein subunit beta ETFB_RAT Spot Nº 66 Identified 27,670.00 39 7.60 8.8 Mit. Metabolism.
Voltage-dependent anion-selective channel protein 1 VDAC1_RABIT Spot Nº 67 Identified 30,722.00 49 8.62 8.8 Mit out mb; Cell mb. Stress Resp...
Phosphoserine aminotransferase SERC_HUMAN Spot Nº 69 No 40,397.00 51 7.56 8.6 mb, mithoc Metabolism.
Isocitrate dehydrogenase [NAD] subunit beta, IDH3B_RAT Spot Nº 70 Identified 42,327.00 53 8.89 8.6 Mit. Metabolism.
Fructose-bisphosphate aldolase C ALDOC_RAT SpotNº 70 Identified 39,259.00 60 6.67 8.6 Metabolism.
Creatine kinase, ubiquitous mitochondrial KCRU_MOUSE Spot Nº 71 Identified 46,974.00 57 8.39 8.7 Mit inn mb. Metabolism.
Phosphoglycerate kinase 1 PGK1_HORSE Spot Nº 71 No 42,327.00 57 8.89 8.5 Metabolism.
Fumarate hydratase, mitochondrial FUMH_RAT Spot Nº 72 No 54,429.00 64 9.06 8.3 Mit; Cp. Metabolism.
Pyruvate kinase isozymes M1/M2 KPYM_RAT Spot Nº 73 Identified 57,781.00 90 6.63 8.2 Metabolism.
Transketolase TKT_RAT Spot Nº 74 Identified 67,601.00 84 7.23 8.2 Prot regulation.
Aconitate hydratase, mitochondrial ACON_RAT Spot Nº 75 Identified 85,380.00 80 7.87 8.2 Mit. Metabolism.
Aconitate hydratase, mitochondrial ACON_RAT Spot Nº 76 Identified 85,380.00 78 7.87 8.3 Mit. Metabolism.
Transketolase TKT_RAT Spot Nº 77 Identified 67,601.00 75 7.23 8.1 Prot regulation.
Pyruvate kinase isozymes M1/M2 KPYM_RAT Spot Nº 78 Identified 57,781.00 75 6.63 8.0 Metabolism.
Glucose-6-phosphate isomerase G6PI_RAT Spot Nº 79 No 62,787.00 65 7.38 8.1 Cp. Metabolism.
Glutamate dehydrogenase 1, mitochondrial DHE3_RAT Spot Nº 80 Identified 61,298.00 60 8.05 8.2 Mit. Metabolism.
Glutamate dehydrogenase 1, mitochondrial DHE3_RAT Spot Nº 81 Identified 61,298.00 74 8.05 7.5 Mit. Metabolism.
Vesicle-fusing ATPase NSF_MOUSE Spot Nº 82 Identified 82,561.00 52 6.52 8.2 Cp. Prot regulation.
Platelet-activating factor acetylhydrolase IB subunit LIS1_MOUSE Spot Nº 83 Identified 46,670.00 53 6.97 8.0 Cp; N. Structural…
Creatine kinase, ubiquitous mitochondrial KCRU_MOUSE Spot Nº 84 Identified 46,974.00 50 8.39 8.1 Mit inn mb. Metabolism.
Aspartate aminotransferase, cytoplasmic AATC_RAT Spot Nº 85 Identified 46,400.00 49 6.73 8.2 Cp. Metabolism.
Glutamine synthetase GLNA_RAT Spot Nº 86 Identified 42,240.00 49 6.64 8.05 Cp. Metabolism.
Fructose-bisphosphate aldolase C ALDOC_RAT Spot Nº 87 Identified 39,259.00 50 6.67 8.0 Metabolism.
Glyceraldehyde-3-phosphate dehydrogenase G3P_RAT Spot Nº 88 Identified 35,787.00 51 8.44 7.9 Cp. Metabolism.
Glyceraldehyde-3-phosphate dehydrogenase G3P_RAT Spot Nº 89 Identified 35,787.00 53 8.44 7.7 Cp. Metabolism.
Alcohol dehydrogenase [NADP+] AK1A1_RAT Spot Nº 90 Identified 36,483.00 51 6.84 7.7 Stress Resp...
Isocitrate dehydrogenase [NADP] cytoplasmic IDHC_RAT Spot Nº 92 No 46,705.00 49 6.53 7.5 Cp. Metabolism.
NAD-dependent deacetylase sirtuin-2 SIRT2_RAT Spot Nº 95 Identified 39,294.00 36 6.67 8.1 Cp. Structural…
Ribose-phosphate pyrophosphokinase 1 PRPS1_HUMAN Spot Nº 96 No 34,812.00 36 6.51 7.8 Metabolism.
Electron transfer flavoprotein subunit alpha ETFA_RAT Spot Nº 97 No 34,929.00 35.5 8.62 7.7 Mit. Metabolism.
Carbonic anhydrase 2 CAH2_RAT Spot Nº 98 No 29,096.00 35 6.89 8.3 Cp. Stress Resp...
Hydroxyacylglutathione hydrolase GLO2_RAT Spot Nº 99 Identified 28,878.00 34.1 6.46 8.2 Metabolism.
Phosphoglycerate mutase 1 PGAM1_MOUSE Spot Nº 100 No 28,786.00 34 6.67 8.25 N. Metabolism.
Triosephosphate isomerase TPIS_RAT Spot Nº 101 Identified 26,832.00 34 6.89 8.3 Metabolism.
Protein-L-isoaspartate (D-aspartate) PIMT_RAT Spot Nº 103 No 24,619.00 34 7.10 7.8 Cp. Metabolism.
Glutathione S-transferase Yb-3 GSTM4_RAT Spot Nº 104 No 25,664.00 35 6.84 7.75 Cp. Stress Resp...
GTP-binding nuclear protein Ran RAN_CANFA Spot Nº 105 No 24,408.00 32 7.01 7.8 Cp; N; Mel. Prot regulation.
Proteasome subunit alpha type-2 PSA2_RAT Spot Nº 106 No 25,909.00 28 8.39 8.0 Cp; N. Prot regulation.
Alpha-crystallin B chain CRYAB_RAT Spot Nº 109 Identified 20,076.00 18 6.76 8.2 Stress Resp...
Nucleoside diphosphate kinase B NDKB_RAT Spot Nº 110 No 17,272.00 8 6.92 8.3 Cp; Cell mb. Metabolism.
Peroxiredoxin-5, mitochondrial PRDX5_RAT Spot Nº 111, 112 Identified 22,165.00 10 8.94 7.5 Mit; Cp; Per. Stress Resp...
Peroxiredoxin-5, mitochondrial PRDX5_RAT Spot Nº 111, 112 Identified 22,165.00 11 8.94 7.0 Mit; Cp; Per. Stress Resp...
Macrophage migration inhibitory factor MIF_RAT Spot Nº 113 Identified 12,496.00 15 6.79 7.1 Cp; ES; N. Stress Resp...
Cytochrome c oxidase polypeptide 6A1, mitochondrial CX6A1_CANFA Spot Nº 114 No 2,109.00 15 6.48 7.3 Mit inn mb. Metabolism.
D-dopachrome decarboxylase OS DOPD_RAT Spot Nº 115 Identified 13,125.00 15 6.09 6.8 Cp. Stress Resp...
Histidine triad nucleotide-binding protein 1 HINT1_MOUSE Spot Nº 116, 118 Identified 13,768.00 13.5 6.38 6.8 Cp. Others.
Fatty acid-binding protein, epidermal OS FABP5_RAT Spot Nº 117 No 15,050.00 12.5 6.73 6.1 Cp. Metabolism.
Histidine triad nucleotide-binding protein 1 HINT1_MOUSE Spot Nº 116, 118 Identified 13,768.00 14 6.38 6.3 Cp. Others.
Prefoldin subunit 1 PFD1_HUMAN Spot Nº 119 Identified 14,202.00 20 6.32 6.3 ??? Prot regulation.
Profilin-2 PROF2_RAT Spot Nº 121 No 14,992.00 18.5 6.55 6.5 Cp. Structural…
Nucleoside diphosphate kinase A NDKA_RAT Spot Nº 122 Identified 17,182.00 17 5.96 4.0 Cp; N. Metabolism.
Superoxide dismutase [Cu-Zn] SODC_RAT Spot Nº 122 Identified 15,912.00 19 5.88 4.3 Cp. Stress Resp...
Gamma-synuclein SYUG_MOUSE Spot Nº 125 Identified 13,152.00 19.5 4.63 5.2 Cp. Structural…
Beta-synuclein MNME_XYLFT Spot Nº 126 Identified 14,268.00 22 4.43 4.7 Cp. Structural…
Calmodulin CALM_BOVIN Spot Nº 127 Identified 16,827.00 31 4.09 3.6 Spindle. Prot regulation.
Phosphatidylethanolamine-binding protein 1 PEBP1_RAT Spot Nº 128 Identified 20,788.00 35 5.48 4.9 Cp; Cell mb. Metabolism.
Peroxiredoxin-2 PRDX2_RAT Spot Nº 129 Identified 21,765.00 35 5.20 4.7 Cp. Stress Resp...
Peroxiredoxin-2 PRDX2_RAT Spot Nº 129 Identified 21,765.00 32 5.20 4.3 Cp. Stress Resp...
Ubiquitin carboxyl-terminal hydrolase isozyme L1 UCHL1_MOUSE Spot Nº 131 Identified 24,822.00 32 5.14 4.5 Cp. Prot regulation.
Rho GDP-dissociation inhibitor 1 GDIR1_RAT Spot Nº 132 Identified 23,393.00 36 5.12 4.3 Cp. Prot regulation.
Translationally-controlled tumor protein TCTP_MOUSE Spot Nº 133 Identified 19,450.00 38 4.76 4.2 Cp. Structural…
Lactoylglutathione lyase LGUL_RAT Spot Nº 134 Identified 20,806.00 40 5.12 4.4 Stress Resp...
14-3-3 protein gamma 1433G_HUMAN Spot Nº 135 Identified 28,235.00 40 4.80 4.5 Cp. Prot regulation.
Calretinin CALB2_RAT Spot Nº 135 Identified 31,384.00 43 4.94 4.3 Carrier.
14-3-3 protein epsilon 1433E_BOVIN Spot Nº 136 Identified 29,155.00 51 4.63 4.3 Cp; Mel. Prot regulation.
Annexin A5 ANXA5_RAT Spot Nº 137 Identified 35,722.00 53 4.93 4.4 Others.
Annexin A5 ANXA5_RAT Spot Nº 138 Identified 35,722.00 75 4.93 3.88 Others.
Ubiquitin thioesterase OTUB1 OTUB1_RAT Spot Nº 139 No 31,250.00 80 4.85 4.6 Prot regulation.
Glial fibrillary acidic protein GFAP_RAT Spot Nº 140 Identified 49,927.00 47 5.35 4.5 Cp. Structural…
40S ribosomal protein SA RSSA_RAT Spot Nº 140 Identified 32,803.00 47 4.80 4.3 Cp. Structural…
Glial fibrillary acidic protein GFAP_RAT Spot Nº 141 Identified 49,927.00 51 5.35 4.3 Cp. Structural…
Calreticulin CALR_RAT Spot Nº 142 No 47,966.00 63 4.33 4.6 ER. Prot regulation.
Rab GDP dissociation inhibitor alpha GDIA_RAT Spot Nº 143 Identified 50,504.00 64 5.00 5.0 Cp. Carrier.
Heat shock protein HSP 90-beta HS90B_RAT Spot Nº 144 Identified 83,229.00 97 4.97 5.2 Cp; Mel. Prot regulation.
Neurofilament medium polypeptide NFM_RAT Spot Nº 145 Identified 95,734.00 115 4.77 5.18 Structural…
Neurofilament medium polypeptide NFM_RAT Spot Nº 145 Identified 95,734.00 115 4.77 5.5 Structural…
Neurofilament heavy polypeptide NFH_RAT Spot Nº 146 Identified 115,308.00 160 5.74 5.2 Structural…
Gamma-enolase ENOG_RAT Spot Nº 147 Identified 47,111.00 66 5.03 5.3 Cp; Cell mb. Metabolism.
Actin, cytoplasmic 1 ACT5_CHICK Spot Nº 148 No 41,809.00 58 5.30 5.3 Cp. Structural…
Tropomodulin-2 TMOD2_MOUSE Spot Nº 149 Identified 39,487.00 57 5.28 5.6 Cp. Structural…
Tubulin alpha-1 chain (Fragment) TBA1_CHICK Spot Nº 150 Identified 50,104.00 50 4.96 6.0 Structural…
L-lactate dehydrogenase B chain LDHB_RAT Spot Nº 151 Identified 36,589.00 51 5.70 5.7 Cp. Metabolism.
Pyruvate dehydrogenase E1 component subunit beta, mitochondrial ODPB_MOUSE Spot Nº 152 Identified 38,912.00 49 6.41 6.0 Mit. Metabolism.
Pyruvate dehydrogenase E1 component subunit beta, mitochondrial ODPB_MOUSE Spot Nº 154 Identified 38,912.00 45 6.41 6.4 Mit. Metabolism.
L-lactate dehydrogenase B chain LDHB_RAT Spot Nº 155 Identified 36,589.00 45 5.70 6.8 Cp. Metabolism.
L-lactate dehydrogenase B chain LDHB_RAT Spot Nº 158 Identified 36,589.00 36 5.70 5.5 Cp. Metabolism.
Malate dehydrogenase, cytoplasmic MDHC_RAT Spot Nº 159 Identified 36,460.00 52 6.16 5.9 Cp. Metabolism.
Malate dehydrogenase, cytoplasmic MDHC_RAT Spot Nº 160 Identified 36,460.00 33 6.16 5.7 Cp. Metabolism.
Prohibitin PHB_RAT Spot Nº 162 Identified 29,802.00 34 5.57 6.2 Mit inn mb. Others.
6-phosphogluconolactonase 6PGL_RAT Spot Nº 164 No 27,217.00 34 5.54 6.7 Metabolism.
Peroxiredoxin-6 PRDX6_RAT Spot Nº 165 Identified 24,803.00 32.5 5.64 6.5 Cp; Lys. Stress Resp...
Peroxiredoxin-6 PRDX6_RAT Spot Nº 165 Identified 24,803.00 32.5 5.64 6.7 Cp; Lys. Stress Resp...
NADH dehydrogenase [ubiquinone] iron-sulfur protein 3, mitochondrial NDUS3_MOUSE Spot Nº 166 Identified 30,131.00 31.5 6.67 6.65 Mit inn mb. Metabolism.
Protein DJ-1 PARK7_RAT Spot Nº 167 Identified 19,961.00 25 6.32 6.65 N; Cp. Stress Resp...
Dynactin subunit 3 DCTN3_BOVIN Spot Nº 168 Identified 21,178.00 30 5.39 6.6 Cp. Structural…
Glutathione S-transferase A6 GSTA6_RAT Spot Nº 170 No 25,791.00 32 5.90 7.3 Cp. Stress Resp...
Thioredoxin-dependent peroxide reductase, mitochondrial PRDX3_RAT Spot Nº 171 Identified 28,277.00 32 7.14 7.6 Mit. Stress Resp...
Thioredoxin-dependent peroxide reductase, mitochondrial PRDX3_RAT Spot Nº 171 Identified 28,277.00 35.5 7.14 7.3 Mit. Stress Resp...
Protein DJ-1 PARK7_RAT Spot Nº 173 Identified 19,961.00 34 6.32 7.2 N; Cp. Stress Resp...
ATP synthase subunit d, mitochondrial ATP5H_RAT Spot Nº 175 Identified 18,752.00 34 6.17 6.9 Mit inn mb. Metabolism.
Protein-L-isoaspartate (D-aspartate) O-methyltransferase OS=Macaca fascicularis GN=PCMT1 PE=2 SV=3 PIMT_MACFA Spot Nº 180 Identified 24,622.00 34 6.23 7.0 Cp. Metabolism.
Flavin reductase BLVRB_MOUSE Spot Nº 181 No 22,183.00 35 6.49 7.15 Cp. Stress Resp...
Protein-L-isoaspartate (D-aspartate) O-methyltransferase OS=Macaca fascicularis GN=PCMT1 PE=2 SV=3 PIMT_MACFA Spot Nº 182 Identified 24,622.00 34 6.23 6.8 Cp. Metabolism.
V-type proton ATPase subunit E 1 VATE1_BOVIN Spot Nº 183 Identified 26,123.00 39 8.45 7.2 Metabolism.
Pirin PIR_RAT Spot Nº 184 No 32,158.00 40 6.22 7.5 Others.
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 10, mitochondrial NDUAA_RAT Spot Nº 187 No 40,468.00 41 7.64 6.7 Mit. Metabolism.
Elongation factor Tu, mitochondrial EFTU_RAT Spot Nº 190 Identified 49,491.00 75 7.23 7.1 Mit. Prot regulation.
Alpha-enolase ENOA_MOUSE Spot Nº 191 Identified 47,111.00 80 6.37 7.8 Cp; Cell mb. Metabolism.
Alpha-enolase ENOA_MOUSE Spot Nº 192 Identified 47,111.00 80 6.37 8.0 Cp; Cell mb. Metabolism.
Beta-centractin ACTY_MOUSE Spot Nº 193 No 42,255.00 86 5.98 7.4 Cp. Structural…
Alpha-enolase ENOA_MOUSE Spot Nº 194 Identified 47,111.00 80 6.37 6.5 Cp; Cell mb. Metabolism.
Alpha-enolase ENOA_MOUSE Spot Nº 195 Identified 47,111.00 80 6.37 6.3 Cp; Cell mb. Metabolism.
Syntaxin-binding protein 1 STXB1_BOVIN Spot Nº 197 Identified 67,526.00 82 6.49 6.8 Cp; Cell mb. Prot regulation.

Abbreviations: Cp, Cytoplasm; N, nucleus; Mit inn mb, mitochondrial inner membrane; Mit, mitochondrion; cell mb, cellular membrane; ES, extra cellular space; Mit out mb, mitochondrial outer membrane; Mel, Melanosome; Cp-sec-syn ves, Cytoplasmic-secreted-synaptic vesicle; Per mb, peroxisomal membrane; Lys, lysosome; Gol app, golgi apparatus; ER, endoplasmic reticulum; EM, extracellular matrix.

Our data show the broad range of proteins identified by 2-DE from Macrophage migration inhibitory factor 12.5 kDa up to the Neurofilament heavy polypeptide with a molecular weight of 115.31 kDa. Furthermore we identified the Myelin basic protein, as the most basic protein (pI 11.25) and Calreticulin as the most acidic (pI 4.33).

Liquid-Chromatography Mass Spectrometry (LC-MS/MS)

To improve the number of proteins identified by MALDI, a LC-MS analysis was carried out. Total rat SC protein (50 μg) was resolved by SDS-PAGE and after Coomassie staining (PageBlue Protein Staining Solution, Fermentas), the gel was divided and cut into 24 pieces, each of which was subjected to in-gel tryptic digestion. After digestion, the peptide samples were analyzed by HPLC (TEMPO, Applied Biosystem) and the peptides eluted were analyzed on a Q-TRAP ion trap MS workstation (Applied Biosystem).

These analyses identified a total of 18,734 peptides that corresponded to 41,481 spectra. After data grouping and filtration, 387 proteins were identified (cut off > 1 and 90% of confident) and their theoretical MW, pI, subcellular localization and function are shown in Table 2, excluding the proteins previously identified by 2-DE. Many acidic proteins were identified, such as Acidic leucine-rich-nuclear phosphoprotein 32 family member B with a pI of 3.87, and basic proteins such as Myelin basic protein with a pI of 11.25. The molecular weights of these proteins ranged from 299.53 kDa for the Microtubule-associated protein 1A to 7850.14 Da for the gamma-2 subunit of the Guanine nucleotide-binding protein G(I)/G(S)/G(O).

Table 2.

Proteins identified with 1-D gel and LC-MS/MS analysis.

Protein name Accession no. MW Da pI Subcellular localization Function
Slice 2, 3
Protein S100-B |P50114|S100B_MOUSEN 10728.05 4.52 Cp; N. Carrier.
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 4 |Q62425|NDUA4_MOUSE 9326.79 9.52 Mit inn mb. Metabolism.
10 kDa heat shock protein, mitochondrial s|P26772|CH10_RAT 10901.67 8.89 Mit. Prot regulation.
Cytochrome c oxidase polypeptide 7A2, mitochondrial sp|P35171|CX7A2_RAT 9352.97 10.28 Mit inn mb. Metabolism.
Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-12 |Q9DAS9|GBG12_MOUSE 7997.23 9.14 Cell mb; Cp. Carrier.
Cytochrome c oxidase subunit VIb isoform 1 |P56391|CX6B1_MOUSE 10071.45 8.96 Mit intermb sp. Metabolism.
Slice 4
ATP synthase subunit e, mitochondrial sp|P29419|ATP5I_RAT 8254.65 9.34 Mit inn mb. Metabolism.
Histone H2A type 1-A |Q96QV6|H2A1A_HUMAN 14233.51 10.86 N. Structural…
Acyl-CoA-binding protein |P11030|ACBP_RAT 10027.46 8.78 Carrier.
Dynein light chain roadblock-type 1 |P62628|DLRB1_RATE 10989.68 6.58 Cp. Structural…
Glutaredoxin-1 sp|Q9ESH6|GLRX1_RAT 11878.88 8.93 Cp. Stress Resp...
Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2 |P63213|GBG2_MOUSE 7850.14 7.78 Cell mb; Cp. Carrier.
Glycogen phosphorylase, brain form |P11216|PYGB_HUMAN 96695.96 6.40 Metabolism.
Mitochondrial import inner membrane translocase subunit Tim13 |Q9Y5L4|TIM13_HUMAN 10500.02 8.42 Mit inn mb. Prot regulation.
Neurofilament light polypeptide sp|P19527|NFL_RAT 61335.28 4.63 Structural…
Slice 5
Galectin-1 |P16045|LEG1_MOUSE 14865.85 5.32 ES. Others.
Cytochrome b-c1 complex subunit 7 |Q9D855|QCR7_MOUSE 13527.47 9.10 Mit inn mb. Metabolism.
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 5 sp|Q63362|NDUA5_RAT 13411.79 6.84 Mit inn mb. Metabolism.
Ribonuclease UK114 |P52759|UK114_RAT 14303.46 7.80 Mit; Cp; N. Prot regulation.
Myotrophin |P62775|MTPN_RAT 12860.77 5.27 Cp. Structural…
Thioredoxin |P11232|THIO_RAT 11673.47 4.80 Cp. Stress Resp...
Fatty acid-binding protein, brain |P55051|FABP7_RAT 14863.98 5.46 Cp. Carrier.
Slice 6
Cytochrome c, somatic |P62898|CYC_RAT 11605.44 9.61 Mit. Metabolism.
Glial fibrillary acidic protein lP47819 |GFAP_RAT 49957.09 5.35 Cp. Structural…
CDGSH iron sulfur domain-containing protein 1 |Q9NZ45|CISD1_HUMAN 12199.05 9.20 Mit out mb. Metabolism.
Parvalbumin alpha |P02625|PRVA_RAT 11925.52 5.00 Carrier.
Astrocytic phosphoprotein PEA-15 |Q5U318|PEA15_RAT 15040.10 4.93 Cp. Stress Resp...
Slice 7
60S acidic ribosomal protein P2 sp|P02401|RLA2_RAT 11691.96 4.44 Prot regulation.
Myosin light polypeptide 6 sp|Q64119|MYL6_RAT 16975.15 4.46 Structural…
V-type proton ATPase subunit G 2 |Q9TSV6|VATG2_PIG 13579.34 10.26 Mel. Carrier.
V-Vesicle-associated membrane protein 2 |P63045|VAMP2_RAT 12690.78 7.84 Cp-sec-syn ves. Carrier.
Ubiquitin-conjugating enzyme E2 N |Q9EQX9|UBE2N_RAT 17123.79 6.13 Prot regulation.
Histone H2A.J |A9UMV8|H2AJ_RAT 14045.45 11.05 N. Structural…
ATP synthase subunit delta, mitochondrial |P35434|ATPD_RAT 17595.07 5.16 Mit inn mb. Metabolism.
Calcineurin subunit B type 1 |P63100|CANB1_RAT 19299.91 4.64 Carrier.
Histone H2B type 2-E |Q64524|H2B2E_MOUSE 13993.26 10.31 N. Structural…
Vesicle-associated membrane protein 3 |Q4R8T0|VAMP3_MACFA 11319.13 8.89 Cell mb. Prot regulation.
Single-stranded DNA-binding protein, mitochondrial sp|P28042|SSB_RAT 17454.93 9.84 Mit. Others.
Tubulin alpha-1A chain |Q6AYZ1|TBA1C_RAT 50135.63 4.94 Structural…
Creatine kinase B-type sp|Q04447|KCRB_MOUSE; sp|P07335|KCRB_RAT 42725.27 5.39 Cp. Metabolism.
Fibrous sheath-interacting protein 1 |Q66H16|FSIP1_RAT 49568.06 5.02 Others.
Mitochondrial fission 1 protein |Q9CQ92|FIS1_MOUSE 17008.65 8.55 Mit out mb; Per mb. Stress Resp...
Slice 8
Low molecular weight phosphotyrosine protein phosphatase |Q5REM7|PPAC_PONAB 18086.50 6.29 Cp. Prot regulation.
Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 |Q9QUR7|PIN1_MOUSE 18370.46 8.93 N. Structural…
Protein S100-A16 |Q96FQ6|S10AG_HUMAN 11801.40 6.28 Carrier.
Vesicle-associated membrane protein 1 |Q63666|VAMP1_RAT 12796.81 6.24 Cp-sec-syn ves. Prot regulation.
Histone H2B type 1-K |Q8CGP1|H2B1K_MOUSE 13920.17 10.31 Nucleus. Structural…
Visinin-like protein 1 |Q5RD22|VISL1_PONAB 22338.24 5.32 Prot regulation.
Thrombospondin type-1 domain-containing protein 7B |Q6P4U0|THS7B_MOUSE 179309.14 8.01 Cell mb. Others.
Slice 9
ADP-ribosylation factor 3 |P61206|ARF3_RAT 20456.51 6.74 Gol app. Prot regulation.
Prefoldin subunit 2 |B0BN18|PFD2_RAT 16579.73 6.20 Prot regulation.
Stathmin |Q6DUB7|STMN1_PIG 17302.51 5.76 Cp. Structural…
Vesicle-associated membrane protein-associated protein B sp|A5GFS8|VAPB_PIG 27053.25 6.85 Cp ves. Prot regulation.
Nucleoside diphosphate kinase A |Q05982|NDKA_RAT 17192.74 5.96 Cp; N. Metabolism.
Ubiquitin-conjugating enzyme E2 variant 2 |Q7M767|UB2V2_RAT 16352.71 7.79 Prot regulation.
Transgelin-3 |Q5R6R2|TAGL3_PONAB 22472.64 6.84 Others.
Ubiquitin-conjugating enzyme E2 L3 |P68037|UB2L3_MOUSE 17861.58 8.68 Prot regulation.
Endothelin-1 sp|P22388|EDN1_RAT 23134.93 9.77 Sec. Stress Resp...
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13 sp|Q95KV7|NDUAD_BOVIN 16673.39 9.22 Mit inn mb; N. Stress Resp...
Slice 10
Tubulin beta chain sp|P02554|TBB_PIG 49860.95 4.78 Structural…
Actin-related protein 2/3 complex subunit 5-like protein |Q9BPX5|ARP5L_HUMAN 17010.32 6.31 Cp. Structural…
Hippocalcin-like protein 1 |P37235|HPCL1_HUMAN 22338.24 5.32 Others.
Neurocalcin-delta |Q5PQN0|NCALD_RAT 22245.23 5.23 Others.
Transcription factor BTF3 sp|Q64152|BTF3_MOUSE 22030.81 9.52 N. Others.
Ferritin heavy chain |P19132|FRIH_RAT 21126.66 5.86 Others.
Cell division control protein 42 homolog |Q8CFN2|CDC42_RAT 21258.61 6.16 Cell mb. Stress Resp...
Phospholipid hydroperoxide glutathione peroxidase, nuclear |Q91XR8|GPX42_RAT 29184.69 10.83 N. Stress Resp...
60S ribosomal protein L12 |P35979|RL12_MOUSE 17804.56 9.48 Others.
Slice 11
Ferritin light chain 1 sp|P02793|FRIL1_RAT 20748.50 5.99 Others.
Cysteine and glycine-rich protein 1 sp|P47875|CSRP1_RAT 20613.48 8.90 N. Others.
NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 10 |Q9DCS9|NDUBA_MOUSE 21023.81 8.19 Mit inn mb. Metabolism.
Tubulin beta-2C chain |Q6P9T8|TBB2C_RAT 49800.98 4.79 Structural…
Slice 12
ATP synthase subunit O, mitochondrial sp|Q06647|ATPO_RAT 23397.55 10.03 Mit inn mb. Metabolism.
Glutathione S-transferase P sp|P47954|GSTP1_CRIMI 23469.00 7.64 Stress Resp...
Adenylate kinase isoenzyme 1 sp|P39069|KAD1_RAT 21583.76 7.66 Cp. Metabolism.
Ras-related protein Rab-1B |Q9H0U4|RAB1B_HUMAN 22163.10 5.55 Cell mb; Cp. Prot regulation.
Glycolipid transfer protein |B0BNM9|GLTP_RAT 23703.65 6.90 Cp. Carrier.
Ras-related protein Rab-11B |O35509|RB11B_RAT 24488.50 5.64 Cell mb. Carrier.
Histone H2A.x sp|P27661|H2AX_MOUSE 15142.60 10.74 N. Structural…
Ras-related protein Rab-7a |P51150|RAB7A_MOUSE 23489.75 6.39 Mel. Prot regulation.
Cell cycle exit and neuronal differentiation protein 1 |Q5FVI4|CEND_RAT 15043.01 9.01 Cell mb. Structural…
UMP-CMP kinase |Q9DBP5|KCY_MOUSE 22165.33 5.68 N. Cp. Metabolism.
Apolipoprotein D |P23593|APOD_RAT 21634.86 4.93 Sec. Carrier.
Microtubule-actin cross-linking factor 1, isoform 4 |Q96PK2|MACF4_HUMAN 670150.80 5.20 Cp. Structural…
GrpE protein homolog 1, mitochondrial |Q99LP6|GRPE1_MOUSE 24307.02 8.58 Mit. Prot regulation.
Transgelin-2 |Q9WVA4|TAGL2_MOUSE 22393.42 8.41 Others.
Ras-related protein Rap-1b |Q62636|RAP1B_RAT 20824.79 5.65 Cell mb; Cp. Prot regulation.
Glutathione S-transferase P 1 |P19157|GSTP1_MOUSE 23609.18 7.69 Stress Resp...
Slice 13
Heat shock protein beta-1 |P14602|HSPB1_MOUSE 23013.85 6.12 Stress Resp...
Myelin-oligodendrocyte glycoprotein |Q63345|MOG_RAT 27881.56 8.61 Cell mb. Structural…
Glutathione S-transferase Y1 |Q00285|GSTMU_CRILO 25818.98 8.74 Cp. Stress Resp...
Tumor protein D52 |Q62393|TPD52_MOUSE 20059.41 4.87 Structural…
Osteoclast-stimulating factor 1 |Q62422|OSTF1_MOUSE 23782.74 5.46 Cp. Others.
UPF0568 protein C14orf166 homolog |Q9CQE8|CN166_MOUSE 28152.21 6.40 N; Cp. Others.
NADH dehydrogenase [ubiquinone] flavoprotein 2, mitochondrial |P19234|NDUV2_RAT 27378.34 6.23 Mit inn mb. Metabolism.
3-hydroxyacyl-CoA dehydrogenase type-2 |O02691|HCD2_BOVIN 27140.29 8.45 Mit. Others.
Glutathione S-transferase alpha I |Q08863|GSTA1_RABIT 25691.11 8.92 Cp. Stress Resp...
Ras-related protein Rab-5A |P20339|RAB5A_HUMAN 23658.68 8.23 Cell mb; Mel. Prot regulation.
Slice 14
14-3-3 protein zeta/delta |P63102|1433Z_RAT; sp|P63101|1433Z_MOUSE 27771.14 4.73 Cp; Mel. Prot regulation.
Dihydropteridine reductase |P11348|DHPR_RAT 25552.20 7.67 Stress Resp...
Succinate dehydrogenase [ubiquinone] iron-sulfur subunit, mitochondrial |P21913|DHSB_RAT 31829.94 8.96 Mit inn mb. Metabolism.
Gamma-enolase |P09104|ENOG_HUMAN 47268.58 4.91 Cp; Cell mb. Metabolism.
Tumor protein D54 |Q6PCT3|TPD54_RAT 23991.85 5.80 Prot regulation.
Coiled-coil-helix-coiled-coil-helix domain-containing protein 3, mitochondrial |Q9CRB9|CHCH3_MOUSE 26334.52 8.56 Others.
14-3-3 protein eta |P68511|1433F_RAT; sp|P68510|1433F_MOUSE; sp|P68509|1433F_BOVIN 28211.74 4.81 Cp. Carrier.
Endoplasmic reticulum protein ERp29 |P52555|ERP29_RAT 28574.83 6.23 ER. Prot regulation.
14-3-3 protein theta |Q5ZMD1|1433T_CHICK 27782.28 4.68 Cp. Prot regulation.
Proteasome subunit alpha type-5 |Q9Z2U1|PSA5_MOUSE 26411.03 4.74 Cp; N. Prot regulation.
Electron transfer flavoprotein subunit beta sp|Q68FU3|ETFB_RAT 27687.42 7.61 Mit. Metabolism.
14-3-3 protein beta/alpha |P35213|1433B_RAT 28054.39 4.81 Cp; Mel. Prot regulation.
Myelin P0 protein |Q6WEB5|MYP0_HORSE 27570.67 9.40 Cell mb. Structural…
ADP/ATP translocase 1 |P48962|ADT1_MOUSE 32904.27 9.73 Mit inn mb. Carrier.
Hypoxanthine-guanine phosphoribosyltransferase (Fragment) |P00493|HPRT_MOUSE 24081.78 5.74 Cp. Metabolism.
Acidic leucine-rich nuclear phosphoprotein 32 family member A |P49911|AN32A_RAT 28564.59 3.99 N; Cp. Stress Resp...
Cytochrome c1, heme protein, mitochondrial sp|P00125|CY1_BOVIN 35296.75 9.14 Mit inn mb. Metabolism.
Microtubule-associated protein 1A |Q9QYR6|MAP1A_MOUSE 300139.96 4.92 Structural…
N(G),N(G)-dimethylarginine dimethylaminohydrolase 2 |Q6MG60|DDAH2_RAT 29687.91 5.66 Metabolism.
Myelin proteolipid protein |P60203|MYPR_RAT 30077.17 8.71 Cell mb. Structural…
Tropomyosin alpha-3 chain |P06753|TPM3_HUMAN 32818.79 4.68 Cp. Structural…
Peflin |Q641Z8|PEF1_RAT 30012.40 5.67 Cp; Cell mb. Others.
Rap guanine nucleotide exchange factor-like 1 |Q68EF8|RPGFL_MOUSE 73695.33 5.84 Carrier.
Calcyclin-binding protein |Q6AYK6|CYBP_RAT 26541.19 7.64 N; Cp. Prot regulation.
Slice 15
Tropomyosin alpha-1 chain |P42639|TPM1_PIG 32680.56 4.69 Cp. Structural…
Pyruvate dehydrogenase E1 component subunit beta, mitochondrial |P49432|ODPB_RAT 38982.13 6.20 Mit. Metabolism.
3-hydroxyisobutyrate dehydrogenase, mitochondrial |P29266|3HIDH_RAT 35302.71 8.73 Mit.
Carbonyl reductase [NADPH] 1 |P47727|CBR1_RAT 30578.12 8.21 Cp. Stress Resp...
EF-hand domain-containing protein D2 |A5D7A0|EFHD2_BOVIN 26918.43 5.26 Others.
Charged multivesicular body protein 4b |Q9D8B3|CHM4B_MOUSE 24936.13 4.76 Cp. Prot regulation.
Elongation factor 1-beta |O70251|EF1B_MOUSE 24693.68 4.53 Prot regulation.
Clathrin light chain B |P08082|CLCB_RAT 25117.44 4.56 Cp ves. Prot regulation.
Complement component 1 Q subcomponent-binding protein, mitochondrial |O35796|C1QBP_RAT 30996.92 4.77 Mit. Others.
Methylglutaconyl-CoA hydratase, mitochondrial |Q9JLZ3|AUHM_MOUSE 33394.99 9.56 Mit. Metabolism.
Tropomyosin alpha-4 chain |P09495|TPM4_RAT 28509.70 4.66 Cp. Structural…
Coiled-coil-helix-coiled-coil-helix domain-containing protein 6 |Q91VN4|CHCH6_MOUSE 29798.81 8.41 Others.
Polymerase I and transcript release factor |Q6NZI2|PTRF_HUMAN 43476.14 5.51 Cell mb; ER; Cp; Mit; N. Others.
Drebrin-like protein |Q9JHL4|DBNL_RAT 48612.51 4.89 Cp. Structural…
Tubulin alpha-1B chain |Q6P9V9|TBA1B_RAT 50151.63 4.94 Structural…
Syntaxin-1B |P61265|STX1B_RAT 33244.69 5.25 Cell mb. Carrier.
Clathrin light chain A |P08081|CLCA_RAT 26980.50 4.41 Cp ves. Prot regulation.
Phosphoglycerate kinase 2 |Q8MIF7|PGK2_HORSE 44879.16 8.62 Cp. Metabolism.
Annexin A3 |P14669|ANXA3_RAT 36363.20 5.96 Others.
Acidic leucine-rich nuclear phosphoprotein 32 family member B |Q9EST6|AN32B_RAT 31060.63 3.87 N. Stress Resp...
Alpha-S1-casein |O62823|CASA1_BUBBU 24326.77 4.87 Sec. Carrier.
Tubulin beta chain lP02554lTBB_PIG 49860.95 4.78 Structural…
Slice 16
Apolipoprotein E |P02650|APOE_RAT 35753.46 5.23 Sec. Stress Resp...
Breast carcinoma-amplified sequence 1 homolog (Fragment) |Q3ZB98|BCAS1_RAT 58623.87 5.58 Cp. Others.
Heterogeneous nuclear ribonucleoproteins A2/B1 |O88569|ROA2_MOUSE 37402.67 8.97 N; Cp. Others.
60S acidic ribosomal protein P0 sp|P19945|RLA0_RAT 34215.47 5.91 Prot regulation.
Adaptin ear-binding coat-associated protein 1 |P69682|NECP1_RAT 29792.40 5.97 Cp ves; Cell mb. Prot regulation.
Alpha-internexin |P23565|AINX_RAT 56115.38 5.20 Structural…
Gamma-soluble NSF attachment protein |Q9CWZ7|SNAG_MOUSE 34732.33 5.31 Cell mb. Prot regulation.
Heterogeneous nuclear ribonucleoprotein H3 |P31942|HNRH3_HUMAN 36926.49 6.37 N. Others.
Elongation factor 1-delta |P57776|EF1D_MOUSE 31293.03 4.91 Prot regulation.
RNA-binding protein Musashi homolog 2 |Q96DH6|MSI2H_HUMAN 39133.53 7.71 Cp; N. Others.
Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1 |P54311|GBB1_RAT 37376.97 5.60 Carrier.
NSFL1 cofactor p47 p|O35987|NSF1C_RAT 40679.96 5.04 N; Gol app. Others.
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9, mitochondrial |Q9DC69|NDUA9_MOUSE 42509.15 9.75 Mit. Metabolism.
F-actin-capping protein subunit alpha-1 |B2GUZ5|CAZA1_RAT 32909.77 5.34 Structural…
Putative heterogeneous nuclear ribonucleoprotein A1-like protein 3 |P0C7M2|RA1L3_HUMAN 34223.28 9.23 N; Cp. Carrier.
Translocon-associated protein subunit alpha |Q9CY50|SSRA_MOUSE 32065.01 4.36 ER: Carrier.
Annexin A2 |Q07936|ANXA2_RAT 38678.24 7.55 Sec; EM; Mel. Others.
Palmitoyl-protein thioesterase 1 |P45479|PPT1_RAT 34455.01 7.09 Lys. Stress Resp...
Dihydropyrimidinase-related protein 2 |P47942|DPYL2_RAT 62277.57 5.95 Cp. Others.
Putative L-aspartate dehydrogenase |Q9DCQ2|ASPD_MOUSE 30269.63 6.45 Metabolism.
Transcriptional activator protein Pur-alpha |P42669|PURA_MOUSE 34883.73 6.07 N. Stress Resp...
Slice 17
Tubulin beta-4 chain |B4F7C2|B4F7C2_RAT 49585.77 4.78 Structural…
Tubulin alpha chain |P68370|TBA1A_RAT 50630.14 4.93 Structural…
Vimentin |P08670|VIME_HUMAN 53651.68 5.06 Structural…
Tubulin beta-3 chain |Q4QRB4|TBB3_RAT 50418.65 4.82 Structural…
Citrate synthase, mitochondrial |Q8VHF5|CISY_RAT 51866.75 8.54 Mit. Metabolism.
Reticulocalbin-2 |Q8BP92|RCN2_MOUSE 37432.96 4.27 ER. Others.
Septin-4 |Q4R4X5|SEPT4_MACFA 55147.26 5.64 Structural…
Protein kinase C and casein kinase substrate in neurons protein 1 |Q5R411|PACN1_PONAB 50921.58 5.15 Cp. Structural…
Obg-like ATPase 1 |Q9NTK5|OLA1_HUMAN 44743.57 7.64 Metabolism.
Sodium/potassium-transporting ATPase subunit beta-1 |P07340|AT1B1_RAT 35201.59 8.83 Cell mb. Carrier.
Hsc70-interacting protein |P50503|F10A1_RAT 41279.50 5.28 Cp. Prot regulation.
Dynactin subunit 2 |Q99KJ8|DCTN2_MOUSE 44116.88 5.14 Cp; Cell mb. Structural…
Hsp90 co-chaperone Cdc37 |Q61081|CDC37_MOUSE 44510.36 5.24 Cp. Prot regulation.
Creatine kinase, ubiquitous mitochondrial |P30275|KCRU_MOUSE 47003.72 8.39 Mit inn mb. Metabolism.
Slice 18
Endophilin-A1 |O35179|SH3G2_RAT 39899.28 5.26 Cp; Cell mb. Carrier.
F-box only protein 2 |Q80UW2|FBX2_MOUSE 33675.95 4.21 Prot regulation.
Septin-2 |Q91Y81|SEPT2_RAT 41592.55 6.15 Cp. Structural…
Acetyl-CoA acetyltransferase, mitochondrial |P17764|THIL_RAT 44695.00 8.92 Mit. Metabolism.
Stomatin-like protein 2 |Q99JB2|STML2_MOUSE 38413.95 8.74 Cp; Cell mb. Structural…
Fumarylacetoacetase |A5PKH3|FAAA_BOVIN 45975.54 6.67 Metabolism.
NAD-dependent deacetylase sirtuin-2 |Q5RJQ4|SIRT2_RAT 39319.27 6.67 Cp. Structural…
Acetyl-CoA acetyltransferase, cytosolic |Q5XI22|THIC_RAT 41108.41 6.86 Cp. Metabolism.
Thioredoxin-dependent peroxide reductase, mitochondrial |P20108|PRDX3_MOUSE 28127.03 7.15 Mit. Stress Resp...
Microtubule-associated protein 6 |Q63560|MAP6_RAT 100484.89 9.45 Cp; Gol app. Structural…
Macrophage-capping protein |Q6AYC4|CAPG_RAT 38798.86 6.11 N; Cp; Mel. Structural…
Tubulin alpha-1D chain |Q2HJ86|TBA1D_BOVIN 50282.78 4.91 Structural…
Heterogeneous nuclear ribonucleoprotein A3 |Q6URK4|ROA3_RAT 39651.99 9.10 N. Others.
Neuromodulin |P07936|NEUM_RAT 23603.34 4.61 Cell mb; Structural…
Proto-oncogene C-crk |Q63768|CRK_RAT 33844.72 5.39 Cp; Cell mb. Others.
Sodium/potassium-transporting ATPase subunit beta-3 |Q63377|AT1B3_RAT 31829.68 8.08 Cell mb; Mel. Others.
Slice 19
60 kDa heat shock protein, mitochondrial |P18687|CH60_CRIGR 60955.49 5.91 Mit. Stress Resp...
Tubulin alpha-1C chain |P68373|TBA1C_MOUSE 49937.37 4.96 Structural…
Peripherin |P21807|PERI_RAT 53549.76 5.37 Structural…
Myc box-dependent-interacting protein 1 |O08839|BIN1_RAT 64533.21 4.95 Cp; N. Structural…
Dihydrolipoyl dehydrogenase, mitochondrial |Q6P6R2|DLDH_RAT 54038.09 7.96 Mit. Metabolism.
Septin-8 |Q8CHH9|SEPT8_MOUSE 49812.39 5.68 Structural…
Slice 20
Stress-70 protein, mitochondrial |O35501|GRP75_CRIGR 73857.70 5.97 Mit. Prot regulation.
Actin, cytoplasmic 2 |P63259|ACTG_RAT 41792.84 5.31 Cp. Structural…
V-type proton ATPase catalytic subunit A |P50516|VATA_MOUSE 68326.08 5.42 Metabolism.
Dihydrolipoyllysine-residue acetyltransferase component of pyruvate dehydrogenase complex, mitochondrial |P08461|ODP2_RAT 67165.84 8.76 Mit. Metabolism.
78 kDa glucose-regulated protein |P06761|GRP78_RAT 72346.99 5.07 ER; Mel. Carrier.
Annexin A6 |P48037|ANXA6_RAT 75754.16 5.39 Cp; Mel. Others.
Lamin-A/C |P48678|LMNA_MOUSE 74237.57 6.54 N. Structural…
Myristoylated alanine-rich C-kinase substrate |P29966|MARCS_HUMAN 29794.51 4.32 Cp; Cell mb. Structural…
NADH-ubiquinone oxidoreductase 75 kDa subunit, mitochondrial |Q66HF1|NDUS1_RAT 79412.33 5.65 Mit inn mb. Metabolism.
Synaptotagmin-2 |P29101|SYT2_RAT 47209.57 8.18 Cp ves; syn ves. Structural…
Heat shock-related 70 kDa protein 2 |P14659|HSP72_RAT 69641.66 5.51 Stress Resp...
Heat shock 70 kDa protein 12A |Q8K0U4|HS12A_MOUSE 74978.38 6.32 Carrier.
Slice 21
Heat shock protein HSP 90-alpha sp|P46633|HS90A_CRIGR 84814.91 4.93 Stress Resp...
Mitochondrial inner membrane protein sp|Q8CAQ8|IMMT_MOUSE 83900.08 6.18 Mit inn mb. Others.
Slice 22
Spectrin alpha chain, brain |P16086|SPTA2_RAT 284637.50 5.20 Cp. Structural…
Heat shock cognate 71 kDa protein |P63018|HSP7C_RAT 70871.07 5.37 Cp. Mel. Stress Resp...
Microtubule-associated protein 2 |P11137|MAP2_HUMAN 202410.75 4.77 Cp. Structural…
Neural cell adhesion molecule 1 |P13595|NCAM1_MOUSE 94658.31 4.83 Cell mb. Structural…
Rab GDP dissociation inhibitor alpha |Q7YQM0|GDIA_PONPY 50536.64 5.00 Cp. Carrier.
Dihydropyrimidinase-related protein 5 |Q9JHU0|DPYL5_RAT 61540.39 6.60 Cp. Others.
Neurofascin |Q810U3|NFASC_MOUSE 138004.21 5.79 Cell mb. Structural…
Slice 23
Tubulin beta-2A chain |Q7TMM9|TBB2A_MOUSE 49906.97 4.78 Structural…
Tubulin alpha-1B chain |Q6P9V9|TBA1B_RAT 50151.63 4.94 Structural…
Neuroblast differentiation-associated protein AHNAK Q09666|AHNK_HUMAN 629101.22 5.80 N. Others.
Regulating synaptic membrane exocytosis protein 1 |Q86UR5|RIMS1_HUMAN 179654.84 9.62 Cell mb. Carrier.
Slice 24
Sodium/potassium-transporting ATPase subunit alpha-3 |P06687|AT1A3_RAT 111691.53 5.26 Cell mb. Carrier.
Aconitate hydratase, mitochondrial |Q9ER34|ACON_RAT 85433.44 7.87 Mit. Metabolism.
Tubulin beta-5 chain |P09653|TBB5_CHICK 49670.82 4.78 Structural…
Myelin-associated glycoprotein |P20917|MAG_MOUSE 69352.86 4.96 Cell mb. Structural…
6-phosphofructokinase type C |P47860|K6PP_RAT 85720.28 6.94 Metabolism.
Microtubule-associated protein 1B |P15205|MAP1B_RAT 269499.65 4.74 Structural…
Plasma membrane calcium-transporting ATPase 2 |P11506|AT2B2_RAT 136811.20 5.70 Cell mb. Metabolism.
Dihydrolipoyllysine-residue acetyltransferase component of pyruvate dehydrogenase complex, mitochondrial |P08461|ODP2_RAT 67165.84 8.76 Mit. Metabolism.
Microtubule-associated protein 1A |P34926|MAP1A_RAT 299530.68 4.87 Structural…
Spectrin beta chain, brain 1 |Q62261|SPTB2_MOUSE 274223.06 5.40 Cp. Structural…
Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform |Q76MZ3|2AAA_MOUSE 65322.61 5.00 Prot regulation.
Hexokinase-1 |P27595|HXK1_BOVIN 102408.01 6.29 Mit out mb. Metabolism.
Characterization and classification of the proteins identified

The proteins identified by MALDI-TOF/TOF and LC-MS/MS were characterized according to their molecular weight (MW), isoelectric point (pI), subcellular localization and recognized function. In total 367 unique proteins were identified with the different techniques employed. On the basis of Swiss-Prot and NCBI database information, the proteins were classified into six functional groups (Fig. 5A): Structural and Cell Cycle Proteins; Metabolic Proteins; Stress Response, Redox State and Apoptosis Proteins; Regulation proteins; Carriers and Other proteins. The different types of protein functions assigned to the proteins identified and the relative proportion of each group were represented (Fig. 5A represents), and a graph of the distribution of pI’s and cellular localization was generated (Fig. 5B). In addition, similar graphs were generated to represent the same features of those proteins recognized to be active in the nervous system.

Figure 5.

Figure 5.

Characterization of the spinal cord proteins identified. A) The functional grouping of all the proteins identified using 2-DE and MALDI-TOF/TOF together with LC-MS/MS are presented. B) Isoelectric point distribution and subcellular localization of the proteins identified. C) Additional classification of the proteins with recognized function in spinal cord.

Discussion

To understand the complex biological processes at play in the central nervous system the key proteins involved must be identified. The exploration of the proteome has attracted increasing interest in recent years, particularly to establish reference maps designed to assist in biomarker discovery. In this regard, defining the complete spinal cord proteome is still an important challenge. This proteome may represent a fundamental key to better understand normal spinal cord physiology, as well as providing important clues to discover the molecular basis of neurodegeneration after spinal cord injury.

In the present study, we have described the proteins present in the rat spinal cord by employing different proteomic tools. Accordingly, we have defined a fast, easy and reproducible protein extraction protocol for the spinal cord. Efficient protein extraction is an essential step in proteomic studies, and the development of this specific sequential extraction augmented the number of proteins isolated, focusing mainly on membrane and hydrophobic proteins. As expected, we identified many mitochondrial and membrane proteins, as well as many soluble proteins, further supporting the efficiency of this methodology.

One of the major problems associated with proteomic analyses are the contaminants in the sample that could interfere with the isoelectrofocusing of spinal proteins (salts, DNA, lipids …). To diminish the effect of this interference, a filter step was included before initiating the 2-DE gel protocol. We employed conventional 2-DE over different pH ranges (e.g. 4–7 and 3–11 NL) to generate different maps that could help search for potential biomarkers. Furthermore, the high degree of 2-DE gel reproducibility and the resolution obtained is necessary to generate good quality maps from the rat spinal cord and for future differential expression analyses. The gels focused with 17 cm pH 4–7 IPG strips did not resolve a large number of spots, and some proteins with a high isoelectric point were not focused correctly with a line of precipitated proteins appearing at the basic extreme of the gel. This distribution in 2-DE gels pH 4–7 could present problems for posterior spot identification, and even for future differential expression analyses between healthy individuals and patients. Accordingly, better resolution was obtained with 2-DE gels with non-linear pH3–11 24 cm IPG strips, avoiding the precipitation of basic proteins. These quality of these gels was relatively high and with a good protein spot distribution, leading to the identification of 200 different spots by MALDI-TOF/TOF.

It is important to note that 2-DE gels cannot resolve proteins below 10 kDa and above 100 kDa, including the more acidic or basic proteins. To maximize the number of proteins identified and to complement the results obtained for 2-DE MALDI-MS/MS, LC-MS/MS analyses identified a further 367 unique proteins. Interestingly both proteomic tools could detect proteins with a broad range of molecular weights and isoelectric points, reflecting the efficiency of the methods employed. We found many proteins in the rat spinal cord with theoretical isoelectric points between 4.0–6.0 and 8.0–9.5, although less were obtained between 6.5 and 7.5.

The spinal proteins were classified into 6 different functional groups: Structural and Cell Cycle Proteins (25%), Metabolic Proteins (30%), Stress Response, Redox State and Apoptosis Proteins (16%), Regulation proteins (8%), Carriers and Other proteins Structural Proteins 12%. Structural and cell cycle proteins constituted a complex and heterogeneous group of cytoskeleton proteins, such as Microtubule-associated protein 1A, myelin sheet, or extracellular matrix and attachment proteins. In addition, DNA scaffold proteins and other structural proteins implicated in mitotic division and cell cycle regulation were characterized, making up around 25% of the total proteins identified. The second category, metabolic proteins, was also very broad and it reached nearly 30% of the total protein content, mainly containing hydrolytic and glucolytic enzymes. The third group, Stress Response, Redox State and Apoptosis proteins, was also a complex group made up of different proteins implicated in stress and injury response (Heat Shock Proteins). Furthermore, we included other proteins here associated with reducing oxidative damage and apoptosis. This group contained around 12% of the total proteins identified. Regulatory proteins related to protein synthesis, including transcription and translation, protein folding and degradation, made up about 16% of the proteins identified. Protein carriers were comprised of transporters and other metabolite binding molecules that represented approximately 8% of the total. Finally, a category of proteins that could not be classified into any of the above groups was denominated as “other” and contributed up to 12% to the complete proteome described here.

The proteins identified with a recognized function in the SC were organized into four functional groups. The numerous proteins in each functional group suggests that the technique developed in this report will be extremely useful to identify possible therapeutic targets for spinal cord injury, and pathways that may arrest the development of associated pathologies such as neuropathic pain and spasticity. Furthermore this technique will be important to develop future regenerative strategies.

Structural proteins were defined that included many common neuronal and glial proteins normally present in central nervous system tissue such as: Neurofilament (NF), Glial fibrillary acidic protein (GFAP), Myelin basic protein (MBP), Myelin-associated glycoprotein (MAG), Neural cell adhesion molecule (NCAM) and Macrophage migration inhibitory factor (MIF). Several of these proteins have a clear role during acute SCI such as GFAP in gliogenesis41 or MIF in astrocyte proliferation,42 while an increase in MAG would suggest the presence of a spinal environment that is inhibitory to nerve growth.43

The second group of proteins were related to neurotransmission. Several Vesicle-associated membrane proteins (VAMPs) were identified but only some of these are thought to be upregulated in the pathological state following SCI, although similar changes may have been identified following peripheral nerve injury axotomy.44 Many others were related to glutamatergic communication such as Glutamine synthetase (GS) and Glutamate dehydrogenase (GDH). These two proteins are known to be therapeutic targets for the successful treatment of spinal cord ischemia.45

Among the proteins responsible for cell survival and combating apoptosis, the presence of Gamma-enolase, Glucose-6-phosphate isomerase, Peroxiredoxin-2 (possible anti-oxidant protein) and Protein DJ-1 in the normal spinal cord should be highlighted, as opposed to only one protein (Glyceraldehyde-3-phosphate dehydrogenase) associated with a pro-apoptotic profile. The upregulation of neuron-specific enolase has been previously described as a potential biomarker of acute SCI.46 An increase in glyceraldehyde-3-phosphate dehydrogenase in spinal cord tissue has been demonstrated after contusion injury,47 while previous proteomic analysis has highlighted the upregulation of peroxiredoxin 2 protein after experimental SCI.48

Lastly, numerous proteins associated with cell metabolism, development, and response to injury were identified, including those associated with neuronneuron interactions (Neural cell adhesion molecule 1) and neuron-glial cell interactions (Neurofascin), neurogenesis (Lyssencephaly-1 homologue A, Alpha-Internexin, Stathmin, Dihydropyrimidinase-related protein), neurite outgrowth (Neural cell adhesion molecule 1, Neurofascin), neuronal precursor proliferation (Lyssencephaly-1 homologue A), synaptogenesis and synaptic plasticity (Neurofascin and 14-3-3 protein gamma), axonal guidance (Neurofascin), axonal regeneration (Macrophage migration inhibitory factor) and myelination (Neurofascin). Significantly, the induction of a serine-threonine kinase stathmin after SCI has already been demonstrated and it was associated with an increase in glial proliferation.49

In addition, several proteins with no known spinal function were identified (following a NCBI bibliographic database search), as well as Protein S100-B that has been proposed as a marker of SCI severity46 and Ubiquitin carboxyl-terminal hydrolase isozyme L1 that may be related to axon degradation.50 An upregulation of Gamma-synuclein has been described in the SC51 and the spinal dorsal horn45 although its precise role during acute SCI is not known. Moreover, both Thioredoxin-dependent peroxide reductase and Palmitoyl-protein thioesterase 1 have been linked to the negative regulation of neuron apoptosis (Swiss-prot Database). Finally, the Platelet-activating factor acetylhydrolase IB subunit alpha may promote the proliferation of neuronal precursors (Swiss-prot Database).

Taken together these data help highlight the change in the spinal cord proteome during acute and chronic SCI, as well helping to define the different profiles associated with symptoms such as neuropathic pain, spasticity, they will serve to benchmark future neuro-regenerative therapies. Despite the promising results obtained in these studies, it will be necessary to define more of the proteins present in the spinal cord proteome. We hope that by continuing these studies and complementing them, the characterization of the complete protein profile of the rat spinal cord will be possible, and differential expression analyses can be carried out in human and/or other animal models.

Acknowledgments

We thank Carmen Bermudez and Ana Isabel Carrasco for their technical support.

This work was supported by grants from the Instituto de Salud Carlos III (FIS PI070537), Fondo de Investigación Sanitaria de Castilla la Mancha (FISCAM, PI2008/08), Fondo de Investigación Sanitaria de Castilla la Mancha (FISCAM, PI2008/28), REDES TEMATICAS DE INVESTIGACION COOPERATIVA (RD06/0014/1015).

Footnotes

Disclosures

The authors report no conflicts of interest.

References

  • 1.Bonita R. Epidemiology of Stroke. Lancet. 1992;339:342–4. doi: 10.1016/0140-6736(92)91658-u. [DOI] [PubMed] [Google Scholar]
  • 2.Alonso I, Regidor E, Rodriguez C, Gutierrez-Fisac JL. The main causes of death in Spain. Med Clin (Barc) 1996;12:441–5. [PubMed] [Google Scholar]
  • 3.Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. Cell death in models of spinal cord injury. Prog Brain Res. 2002;137:37–47. doi: 10.1016/s0079-6123(02)37006-7. [DOI] [PubMed] [Google Scholar]
  • 4.Profyris C, Cheema SS, Zang D, Azari MF, Boyle K, Petratos S. Degenerative and regenerative mechanisms governing spinal cord injury. Neurobiol Dis. 2004;3:415–36. doi: 10.1016/j.nbd.2003.11.015. [DOI] [PubMed] [Google Scholar]
  • 5.Sindrup SH, Jensen TS. Efficacy of pharmacological treatments of neuropathic pain: An update and effect related to mechanism of drug action. Pain. 1999;83:389–400. doi: 10.1016/S0304-3959(99)00154-2. [DOI] [PubMed] [Google Scholar]
  • 6.Hansson PT, Dickenson AH. Pharmacological treatment of peripheral neuropathic pain conditions based on shared commonalities despite multiple etiologies. Pain. 2005;113:251–4. doi: 10.1016/j.pain.2004.10.007. [DOI] [PubMed] [Google Scholar]
  • 7.Chen H, Lamer TJ, Rho RH, et al. Contemporary management of neuropathic pain for the primary care physician. Mayo Clin Proc. 2004;79:1533–45. doi: 10.4065/79.12.1533. [DOI] [PubMed] [Google Scholar]
  • 8.Lanzrein AS, Johnston CM, Perry VH, et al. Longitudinal study of inflammatory factors in serum, cerebrospinal fluid, and brain tissue in Alzheimer disease: interleukin-1beta, interleukin-6, interleukin-1 receptor antagonist, tumor necrosis factor-alpha, the soluble tumor necrosis factor receptors I and II and alpha1-antichymotrypsin. Alzheimer Dis Assoc Disord. 1998 Sep;12(3):215–27. doi: 10.1097/00002093-199809000-00016. [DOI] [PubMed] [Google Scholar]
  • 9.Rachel Pardes Berger, Mary Clyde Pierce, Stephen R, Wisniewski, et al. Neuron-Specific Enolase and S100B in Cerebrospinal Fluid After Severe Traumatic Brain Injury in Infants and Children. Pediatrics. 2002 Feb 2;109(1):e31. doi: 10.1542/peds.109.2.e31. [DOI] [PubMed] [Google Scholar]
  • 10.Brown P, Kenney K, Little B, et al. Gajdusek. Intracerebral distribution of infectious amyloid protein in spongiform encephalopathy. Annals of Neurology. 38(2):245–53. doi: 10.1002/ana.410380218. [DOI] [PubMed] [Google Scholar]
  • 11.Allan Butterfield D. Debra Boyd-Kimball and Alessandra Castegna. Proteomics in Alzheimer’s disease: insights into potential mechanisms of neurodegeneration. Journal of Neurochemistry. 2003;86:1313–27. doi: 10.1046/j.1471-4159.2003.01948.x. [DOI] [PubMed] [Google Scholar]
  • 12.Maja Puchadesa, Sara Folkesson Hanssona, Carol L, et al. Proteomic studies of potential cerebrospinal fluid protein markers for Alzheimer’s disease. Molecular Brain Research. 2003 Oct 21;:118, 1–2, 140–6. doi: 10.1016/j.molbrainres.2003.08.005. [DOI] [PubMed] [Google Scholar]
  • 13.Kunz S, Tegeder I, Coste O, et al. Comparative proteomic analysis of the rat spinal cord in inflammatory and neuropathic pain models. Neuroscience Letters. 2005;381:289–93. doi: 10.1016/j.neulet.2005.02.022. [DOI] [PubMed] [Google Scholar]
  • 14.Ding Q, Wu Z, Guo Y, et al. Proteome analysis of up-regulated proteins in the rat spinal cord induced by transection injury. Proteomics. 2006;6:505–18. doi: 10.1002/pmic.200500296. [DOI] [PubMed] [Google Scholar]
  • 15.Yan X, Liu T, Yang S, et al. Proteomic profiling of the insoluble pellets of the transected rat spinal cord. J Neurotrauma. 2009;26:179–93. doi: 10.1089/neu.2008.0533. [DOI] [PubMed] [Google Scholar]
  • 16.Ka Wan Li, Martin P, Hornshaw Roel C, Van der Schors. Proteomics Analysis of Rat Brain Postsynaptic Density. Implications of the Diverse Protein Functional Groups for the Integration Ofsynaptic Physiology. Journal of Biological Chemistry. 2004 Jan 9;279(2):987–1002. doi: 10.1074/jbc.M303116200. [DOI] [PubMed] [Google Scholar]
  • 17.Michael Fountoulakis, Sophia Kossida. Proteomics-driven progress in neurodegeneration research. Electrophoresis. 2006;27:1556–73. doi: 10.1002/elps.200500738. [DOI] [PubMed] [Google Scholar]
  • 18.Orth AP, Batalov S, Perrone M, Chanda SK. The promise of genomics to identify novel therapeutic targets. Expert Opin Ther Targets. 2004;8:587–96. doi: 10.1517/14728222.8.6.587. [DOI] [PubMed] [Google Scholar]
  • 19.Choudhary J, Grant SG. Proteomics in postgenomic neuroscience: the end of the beginning. Nat Neurosci. 2004;7:440–5. doi: 10.1038/nn1240. [DOI] [PubMed] [Google Scholar]
  • 20.Bermudez-Crespo J, Lopez JL. A better understanding of molecular mechanisms underlying human diseases. Proteomics Clin Appl. 2007;1:983–1003. doi: 10.1002/prca.200700086. [DOI] [PubMed] [Google Scholar]
  • 21.Pandey A, Mann M. Proteomics to study genes and genomes. Nature. 2000;405:837–46. doi: 10.1038/35015709. [DOI] [PubMed] [Google Scholar]
  • 22.Norin M, Sundstrom M. Structural proteomics: Developments in structure-to-function predictions. Trends Biotechnol. 2002;20:79–84. doi: 10.1016/s0167-7799(01)01884-4. [DOI] [PubMed] [Google Scholar]
  • 23.Matsumoto H, Komori N. Protein identification on two-dimensional gels archived nearly two decades ago by in-gel digestion and matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Anal Biochem. 1999;270:176–9. doi: 10.1006/abio.1999.4054. [DOI] [PubMed] [Google Scholar]
  • 24.Weston AD, Hood L. Systems biology, proteomics, and the future of health care: toward predictive, preventative, and personalized medicine. J Proteome Res. 2004;3:179–96. doi: 10.1021/pr0499693. [DOI] [PubMed] [Google Scholar]
  • 25.van der Greef J, Stroobant P, van der Heijden R. The role of analytical sciences in medical systems biology. Curr Opin Chem Biol. 2004;8:559–65. doi: 10.1016/j.cbpa.2004.08.013. [DOI] [PubMed] [Google Scholar]
  • 26.Ideker I. Systems biology 101—what you need to know. Nat Biotechnol. 2004;22:473–5. doi: 10.1038/nbt0404-473. [DOI] [PubMed] [Google Scholar]
  • 27.Marko-Varga G, Fehniger TE. Proteomics and disease—the challenges for technology and discovery. J Proteome Res. 2004;3:167–78. doi: 10.1021/pr049958+. [DOI] [PubMed] [Google Scholar]
  • 28.Domon B, Broder S. Implications of new proteomics strategies for biology and medicine. J Proteome Res. 2004;3:253–60. doi: 10.1021/pr034082c. [DOI] [PubMed] [Google Scholar]
  • 29.Fernie AR, Trethewey RN, Krotzky AJ, Willmitzer L. Metabolite profiling: from diagnostics to systems biology. Nat Rev Mol Cell Biol. 2004;5:763–9. doi: 10.1038/nrm1451. [DOI] [PubMed] [Google Scholar]
  • 30.Gorg A, Weiss W, Dunn MJ. Current two-dimensional electrophoresis technology for proteomics. Proteomics. 2004;4:3665–85. doi: 10.1002/pmic.200401031. [DOI] [PubMed] [Google Scholar]
  • 31.O’Farrell PH. High resolution two-dimensional electrophoresis of proteins. J Biol Chem. 1975;250:4007–21. [PMC free article] [PubMed] [Google Scholar]
  • 32.Carrette O, Burkhard PR, Sanchez JC, Hochstrasser DF. State-of-the-art two-dimensional gel electrophoresis: a key tool of proteomics research. Nat Protoc. 2006;1:812–23. doi: 10.1038/nprot.2006.104. [DOI] [PubMed] [Google Scholar]
  • 33.Rosenfeld J, Capdevielle J, Guillemot JC, Ferrara P. In-gel digestion of proteins for internal sequence analysis after one- or two-dimensional gel electrophoresis. Anal Biochem. 1992;203:173–9. doi: 10.1016/0003-2697(92)90061-b. [DOI] [PubMed] [Google Scholar]
  • 34.Barderas MG, Wigdorovitz A, Merelo F, et al. Serodiagnosis of African swine fever using the recombinant protein p30 expressed in insect larvae. Journal Virological Methods. 2000;89:129–36. doi: 10.1016/s0166-0934(00)00209-3. [DOI] [PubMed] [Google Scholar]
  • 35.Gonzalez-Barderas M, Gallego-Delgado J, Mas S, et al. Isolation of circulating human monocytes with high purity for proteomic analysis. Proteomics. 2004;4:432–7. doi: 10.1002/pmic.200300629. [DOI] [PubMed] [Google Scholar]
  • 36.Durán MC, Mas S, Martin-Ventura JL, et al. Proteomic analysis of human vessels: application to atherosclerotic plaques. Proteomics. 2003;3:973–8. doi: 10.1002/pmic.200300389. [DOI] [PubMed] [Google Scholar]
  • 37.Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248–54. doi: 10.1006/abio.1976.9999. [DOI] [PubMed] [Google Scholar]
  • 38.Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970;227:680–5. doi: 10.1038/227680a0. [DOI] [PubMed] [Google Scholar]
  • 39.Shevchenko A, Tomas H, Havlis J, Olsen JV, Mann M. In-gel digestion for mass spectrometric characterization of proteins and proteomes. Nat Protoc. 2006;1:2856–60. doi: 10.1038/nprot.2006.468. [DOI] [PubMed] [Google Scholar]
  • 40.Barderas MG, Tunon J, Darde VM, et al. Circulating human monocytes in the acute coronary syndrome express a characteristic proteomic profile. J Proteome Res. 2007;6(2):876–6. doi: 10.1021/pr0601990. [DOI] [PubMed] [Google Scholar]
  • 41.Nieto-Sampedro M. Neurite outgrowth inhibitors in gliotic tissue. Adv Exp Med Biol. 1999;468:207–24. doi: 10.1007/978-1-4615-4685-6_17. [DOI] [PubMed] [Google Scholar]
  • 42.Koda M, Nishio Y, Hashimoto M, et al. Up-regulation of macrophage migration-inhibitory factor expression after compression-induced spinal cord injury in rats. Acta Neuropathol. 2004;108:31–6. doi: 10.1007/s00401-004-0853-z. [DOI] [PubMed] [Google Scholar]
  • 43.Giger RJ, Venkatesh K, Chivatakarn O, et al. Mechanisms of CNS myelin inhibition: evidence for distinct and neuronal cell type specific receptor systems. Restor Neurol Neurosci. 2008;26:97–115. [PMC free article] [PubMed] [Google Scholar]
  • 44.Jacobsson G, Piehl F, Meister B. VAMP-1 and VAMP-2 gene expression in rat spinal motoneurones: differential regulation after neuronal injury. Eur J Neurosci. 1998;10:301–16. doi: 10.1046/j.1460-9568.1998.00050.x. [DOI] [PubMed] [Google Scholar]
  • 45.Wu GJ, Chen WF, Sung CS, et al. Preventive effects of intrathecal methylprednisolone administration on spinal cord ischemia in rats: the role of excitatory amins acid metabolizing systems. Neuroscience. 2007;147:294–303. doi: 10.1016/j.neuroscience.2007.04.040. [DOI] [PubMed] [Google Scholar]
  • 46.Cao F, Yang XF, Liu WG, et al. Elevation of neuron-specific enolase and S-100beta protein level in experimental acute spinal cord injury. J Clin Neurosci. 2008;15:541–4. doi: 10.1016/j.jocn.2007.05.014. [DOI] [PubMed] [Google Scholar]
  • 47.Wu X, Yoo S, Wrathall JR. Real-time quantitative PCR analysis of temporal-spatial alterations in gene expression after spinal cord contusion. J Neurochem. 2005;93:943–52. doi: 10.1111/j.1471-4159.2005.03078.x. [DOI] [PubMed] [Google Scholar]
  • 48.Kang SK, So HH, Moon YS, Kim CH. Proteomic analysis of injured spinal cord tissue proteins using 2-DE and MALDI-TOF MS. Proteomics. 2006;6:2797–812. doi: 10.1002/pmic.200500621. [DOI] [PubMed] [Google Scholar]
  • 49.Shen A, Liu Y, Zhao J, et al. Temporal-spatial expressions of p27kip1 and its phosphorylation on Serine-10 after acute spinal cord injury in adult rat: Implications for post-traumatic glial proliferation. Neurochem Int. 2008;52:1266–75. doi: 10.1016/j.neuint.2008.01.011. [DOI] [PubMed] [Google Scholar]
  • 50.Li GL, Farooque M, Holtz A, Olsson Y. Expression of the ubiquitin carboxyl-terminal hydrolase PGP 9.5 in axons following spinal cord compression trauma. An immunohistochemical study in the rat. APMIS. 1997;105:384–90. doi: 10.1111/j.1699-0463.1997.tb00585.x. [DOI] [PubMed] [Google Scholar]
  • 51.Willis D, Li KW, Zheng JQ, et al. Differential transport and local translation of cytoskeletal, injury-response, and neurodegeneration protein mRNAs in axons. J Neurosci. 2005;25:778–91. doi: 10.1523/JNEUROSCI.4235-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Li JY, Henning Jensen P, Dahlström A. Differential localization of alpha-, beta- and gamma-synucleins in the rat CNS. Neuroscience. 2002;113:463–78. doi: 10.1016/s0306-4522(02)00143-4. [DOI] [PubMed] [Google Scholar]

Articles from Biomarker Insights are provided here courtesy of SAGE Publications

RESOURCES