iTRAQ-based quantitative proteomic analysis of cerebrospinal fluid reveals NELL2 as a potential diagnostic biomarker of tuberculous meningitis

  • Authors:
    • Yongtao Yang
    • Jun Mu
    • Guanghui Chen
    • Yuan Zhan
    • Jiaju Zhong
    • Youdong Wei
    • Ke Cheng
    • Bin Qin
    • Hongmin You
    • Peng Xie
  • View Affiliations

  • Published online on: March 11, 2015     https://doi.org/10.3892/ijmm.2015.2131
  • Pages: 1323-1332
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tuberculous meningitis (TBM) is a serious complication of tuberculosis that affects the central nervous system. As TBM may result in permanent sequelae and death, rapid, accurate diagnostic tests using novel biomarkers are required for the early diagnosis and treatment of TBM. A quantitative proteomic study was therefore performed to identify differential proteins in the cerebrospinal fluid (CSF) obtained from TBM patients (n=12) and healthy controls (n=12). CSF samples were labelled with iTRAQ™ and analyzed by LC‑MS/MS. Gene ontology and Pathway analysis were conducted using DAVID bioinformatics resources. Neural epidermal growth factor‑like like 2 (NELL2) with the largest fold‑change value was selected for validation by western blotting. Proteomic phenotyping revealed over‑representation in two inflammation‑associated processes, complement and coagulation cascades as well as cell adhesion molecules. Western blotting showed a significant decrease in NELL2 levels in TBM subjects compared to healthy controls. The AUC analysis revealed NELL2 was able to distinguish TBM subjects from healthy controls with 83.3% sensitivity and 75% specificity. In conclusion, the results showed that CSF NELL2 is a potential diagnostic biomarker for TBM. Further evaluation of these findings in larger studies including anti‑tuberculosis medicated and unmedicated patient cohorts with other intracranial infectious diseases is required for clinical translation.

Introduction

Each year, almost 10 million individuals worldwide are newly diagnosed with tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (MTB) with 1.7 million mortalities owing to TB (1). Moreover, two billion individuals worldwide with latent MTB infection represent a potential risk of TB reactivation (2). The burden of disease of TB is highest in Asia, with China listed only second to India. In the latest Global Tuberculosis Report by the World Health Organization (WHO), China accounts for 12% of the TB cases worldwide. According to the National Survey of Drug-Resistant TB conducted by the Chinese Center for Disease Control, China also has a serious epidemic of drug-resistant TB (3), with 5.7% of newly diagnosed cases and 25.6% of previously treated cases being multidrug-resistant (MDR) TB.

TB affects the central nervous system (CNS); a condition termed CNS tuberculosis, and is the most severe complication of TB infection. Tuberculous meningitis (TBM) is the most common form of CNS tuberculosis. In TBM patients, MTB in Rich foci (previously seeded and formed in the meninges or brain parenchyma through primary infection) reactivate and rupture into the subarachnoid space, producing meningitis. As TBM may result in permanent sequelae and death, prompt diagnosis and treatment are required (4). Prognosis depends on the clinical stage at which TBM is diagnosed (5). Effective TBM control requires early, efficient, and accurate diagnostic biomarkers in addition to drug-resistance identification and treatment response evaluation (68). However, the currrent gold standard for TBM diagnosis, MTB isolation from cerebrospinal fluid (CSF) culture, usually takes weeks to reach a positive outcome and has a relatively low sensitivity of 25–70% (9).

Since these conventional methods do not satisfy the requirements for diagnosis of TBM, it is crucial to target complementary disease-specific biomarkers that can facilitate earlier diagnosis and treatment in TBM patients. However, most studies on TB biomarkers have focused on pulmonary TB. In previous studies it was suggested that diagnostic biomarkers for TBM, were adenosine deaminase isoenzyme-2 (ADA2) (10), adenosine deaminase (ADA) (11,12), 65-kDa heat shock protein antigen (13), CSF lactate, CSF lactate dehydrogenase (14) and arachidonate 5-lipoxygenase (15). However, none of these biomarkers have been sufficiently validated (16).

Therefore, rapid, accurate diagnostic tests using novel biomarkers for TBM remain to be developed. To that end, mass spectrometry (MS)-based quantitative proteomics has become a useful tool in identifying disease-specific biomarkers (17) by yielding information about differences across a spectrum of proteins within samples in a single experiment. Kumar et al (18) have applied an MS-based quantitative proteomic approach to compare postmortem brain tissues from confirmed cases of TBM with uninfected brain tissues and found upregulated levels of signal-regulatory protein α (SIRPA), protein disulfide isomerase family A, member 6 (PDIA6), amphiphysin (AMPH) and neurofascin (NFASC) in addition to downregulated levels of ferritin light chain (FTL). Although all the postmortem brain tissue samples were collected within 8–16 h of death with the patients’ bodies maintained at 4°C, proteins are rapidly degraded after death. Thus, there is a reasonable risk that TBM-specific proteins may have been missing or severely altered in the study of Kumar et al. As opposed to brain tissue, CSF can more easily be collected from live patients by lumbar puncture. As the CSF is in constant exchange with the brain’s interstitial fluid and thus alterations in protein expression within the brain may be closely reflected in the CSF (17,19), the CSF appears to be a more promising source of biomarkers for TBM. Among MS-based approaches, the isobaric tag for relative and absolute quantitation (iTRAQ™)-based proteomics has emerged as particularly valuable in detecting relatively scarce proteins, such as those in the CSF (18,20).

In the present study, an iTRAQ™-based quantitative proteomic approach was used to identify differential proteins in the CSF obtained from live TBM patients and healthy controls. Proteomic phenotyping of the 81 differential proteins revealed over-representation in two inflammation-associated processes, complement and coagulation cascades as well as cell adhesion molecules. Western blotting of the neuron-specific differential protein with the largest absolute fold-change value, neural epidermal growth factor-like like 2 (NELL2), showed significantly decreased NELL2 levels in TBM subjects compared to healthy controls. Area under the receiver operating characteristic curve analysis demonstrated NELL2 was able to distinguish TBM subjects from healthy controls with 83.3% sensitivity and 75% specificity, indicating that CSF NELL2 shows promise as a diagnostic biomarker for TBM.

Materials and methods

Subjects

The protocol of the present study and the procedures employed for sample collection were approved by the Ethics Committee of Chongqing Medical University and have therefore been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki and its later amendments. Written informed consent was obtained from all participants. Twelve TBM subjects were diagnosed following the standard criteria (21) and enrolled in the Department of Neurology at the First Affiliated Hospital of Chongqing Medical University. Twelve healthy control (HC) subjects were enrolled after written informed consent was obtained in the medical examination center at the First Affiliated Hospital of Chongqing Medical University. The HC subjects were required to have neither active TB nor any neurological complaints.

Protein digestion and iTRAQ™ labeling

Pooled CSF samples were generated by combining equal volumes of the 12 individual plasma samples from each group. Each sample (5 ml) was desalted and concentrated to 80 ml in a Vivaspin spin column with a molecular weight cut-off of 5 kDa (Vivascience AG, Hannover, Germany). Then, 120 μg of protein from each sample was dissolved in 30 μl of STD buffer (4% SDS, 100 mM DTT, 150 mM Tris-HCl pH 8.0), boiled in water for 5 min, cooled to room temperature, diluted with 200 μl of UA buffer (8 M Urea, 150 mM Tris-HCl pH 8.0), and transferred to 30 kDa ultrafiltration. The samples were centrifuged at 14,000 × g for 15 min and 200 μl of UA buffer was added. The samples were centrifuged again for 15 min under the same conditions. Then, 100 μl of 0.05 M iodoacetamide in UA buffer was added, and the samples were incubated for 20 min in the dark. After 10 min of centrifugation at the above conditions, the filters were washed three times with 100 μl of UA buffer. Then, 100 μl of DS buffer (50 mM triethylammoniumbicarbonate, pH 8.5) was added to the filters, and the samples were centrifuged for 10 min at the above conditions. This step was repeated twice. Subsequently, 2 μg of trypsin (Promega, Madison, WI, USA) in 40 μl of DS buffer was added to each filter, and the samples were incubated overnight at 37°C. The resulting peptides were collected by centrifugation. The filters were rinsed with 40 μl of 10X DS buffer again. iTRAQ™ labeling was performed according to the manufacturer’s instructions (Applied Biosystems, Foster City, CA, USA). As each sample consisted of a pool of CSF from 12 TBM subjects or 12 HC subjects, iTRAQ™ reagents 114 and 116 were applied to the HC sample and reagents 115 and 117 were applied to the TBM sample. The analytic processes, including plasma depletion, protein digestion, iTRAQ™ labeling, SCX fractionation and LC-MS/MS analysis, were repeated twice.

Peptide fractionation with strong cation exchange chromatography

Prior to liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) analysis, the peptides were purified from excess labeling reagent by strong cation exchange (SCX) chromatography. Briefly, the peptides were dried in a vacuum concentrator, dissolved in strong cation exchange buffer A (10 mM KH2PO4 in 25% of ACN, pH 3.0), and loaded onto a Polysulfoethyl 4.6 × 100 mm column (5 μm, 200 Å; PolyLC Inc., Columbia, MD, USA) at a flow rate of 1 ml/min. A suitable gradient elution was applied to separate peptides at a flow rate of 1 ml/min with elution buffer (10 mM KH2PO4, 500 mM KCl in 25% acetonitrile, pH 3.0). Eluted peptides were collected and desalted by an offline fraction collector. The resulting fractions were combined to 10 pools and desalted on C18 Cartridges [Empore™ SPE Cartridges C18 (standard density), bed I.D. 7 mm, volume 3 ml; Sigma, St. Louis, MO, USA]. Each final fraction was concentrated by a vacuum concentrator and resuspended in 40 μl of 0.1% (v/v) trifluoroacetic acid. All the samples were stored at −80°C until subsequent LC-MS/MS analysis.

LC-MS/MS analysis

For each fraction, 10 μl of solution was injected for nano LC-MS/MS analysis using an AB SCIEX TripleTOF 5600 MS (Toronto, Concord, ON, Canada) equipped with a splitless Eksigent nanoUltra 2D Plus nanoLC system and a cHiPLC Nanoflex microchip system (Eksigent, Dublin, CA, USA). The Nanoflex system uses replaceable microfluidic traps and columns packed with ChromXP C18 (3 μm, 120 Å) for online trapping, desalting and analytical separations. The sample was loaded, and trapping and desalting were performed at 2 μl/min for 10 min with 100% mobile phase A (2% acetonitrile/0.2% formic acid/98% water). For peptide elution, the gradient was initiated at 5% mobile phase B (98% acetonitrile/0.2% formic acid/2% water) and increased to 24% linearly in 70 min at a flow rate of 300 nl/min. The total gradient length was 120 min. MS data acquisition was performed in the information-dependent acquisition (IDA) mode. For TOF MS scans, the TripleTOF 5600 MS was operated with a resolving power of 30,000 FWHM. IDA survey scans were acquired in 250 msec with a mass range of m/z 350–1250. Specifically, 30×100 msec MS/MS were followed at a 120 cps (counts/sec) trigger with a precursor charge state of +2 to +5. Dynamic exclusion was set for 18 sec. Collision energies were calculated on-the-fly for all precursor ions using empirical equations based on mass and charge, and the Enhance iTRAQ™ function was turned on to improve the efficiency of the collision-induced dissociation.

Protein identification and quantification

All LC-MS/MS data were processed using ProteinPilot v4.2 and used to search the Uniprot_HUMAN.20140108_134997 database using the ‘thorough search’ option. The ‘iTRAQ™ 8-plex peptide labeled’ sample type and a ‘biological modification ID focus’ were selected for the analysis method. Trypsin was selected as the digestion enzyme, with cysteine alkylation by iodoacetamide as a modification. Data were normalized by ProteinPilot. The reported data were based on a false discovery rate (FDR) ≤1% confidence for protein identification. Differential proteins were analyzed by ProteinPilot for significant downregulation or upregulation. A significance threshold of 95% was selected as a criterion for each individual experiment. Peptides that matched multiple proteins were not included in the software quantification. The final ratios were calculated from the average of the ratios obtained in the two independent experiments in which the protein abundance was significant (P<0.05). In each experiment, bias correction for unequal mixing in the differently labeled samples was performed based on the assumption that most proteins do not change in expression. Thus, if the sample from each experimental condition was not combined in exactly equal amounts, this bias correction would fix the systematic error. The software identifies the median average protein ratio and corrects it to unity, and then applies this factor to all quantification results.

Gene Ontology and Pathway Analysis

Functional clustering was conducted using DAVID Bioinformatics Resources v6.7 (http://david.abcc.ncifcrf.gov/home.jsp) (22). A functional annotation analysis was performed with the Gene Ontology tool (GOTERM_CC_ALL) using the UniProt accession numbers for all the differential proteins. Only those GO terms in each gene list that yielded a P-value of <0.05 using Fisher’s exact test were considered significantly enriched. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was subsequently performed using the same software, in which a corrected P-value of <0.05 was deemed to indicate a statistically significant difference.

Western blotting validation

Western blotting was performed on the 12 individual TBM samples and 12 individual HC samples. CSF samples were separated by 10% SDS-PAGE, blocked in 5% skim milk in TBST, and transferred onto PVDF membranes. The membranes were incubated overnight at 4°C with an anti-NELL2 (diluted 1:1,000; Abcam, Cambridge, MA, USA) primary antibody. The membranes were washed and incubated with goat polyclonal to rabbit IgG horseradish peroxidase-coupled secondary antibody (diluted 1:10,000; Bio-Rad Laboratories, Hercules, CA, USA). After extensive washing, the protein bands detected by the antibodies were visualized by the enhanced chemiluminescence method and exposed to autoradiography film. After immunodetection, the membranes were stained with Ponceau S as an internal control for normalization. The western blot signals were densitometrically quantified in each sample with Quantity One (Bio-Rad Laboratories).

Statistical analysis

Statistical analysis was performed using the Statistical Package of Social Science (SPSS) for Windows v19.0. Data are expressed as the means ± SD. The Student’s t-test was applied to identify proteins with significant abundance differences between the TBM and HC subjects. Areas under the receiver operating characteristic (ROC) curve (AUC) were calculated to evaluate classifier performance. All the tests were two-tailed. The significance threshold was set at P<0.05.

Results

Subject characteristics

Demographic and clinical data obtained from TBM and HC subjects are provided in Table I. The two groups of subjects were not differentiable by any key demographic characteristics, such as age, gender or body mass index (BMI).

Table I

Demographic and clinical characteristics of the participants.

Table I

Demographic and clinical characteristics of the participants.

VariablesHCTBMP-value (HC vs. TBM)
Sample size1212
Gender (M/F)7/55/70.44
Age (years)a27.7±5.328.3±8.20.09
BMIa46.1±10.036.9±14.60.85
TB culture12/120/12
Fungal culture0/120/12

{ label (or @symbol) needed for fn[@id='tfn1-ijmm-35-05-1323'] } HC, healthy controls;TBM, patients infected with tuberculous meningitis; M, male; F, female; BMI, body mass index; TB, tuberculosis.

a Age and BMI are presented as the means ± SD.

Qualitative results

Data from the two MS repetitions identified 572 unique proteins according to the previously described parameter set. Only the 81 differential proteins with a ±1.2-fold-change and P<0.05 were selected for further analysis (Table II). The threshold criteria were selected based on literature investigating the reproducibility of iTRAQ™ quantification (23).

Table II

Differential proteins identified by iTRAQ coupled with LC-MS/MS ordered by absolute fold-change.

Table II

Differential proteins identified by iTRAQ coupled with LC-MS/MS ordered by absolute fold-change.

Accession no.Gene nameProtein namePep countCover %Biological functionFold-change (TBM/HC)
P01024C3Complement C314752.5Inflammation: acute-phase2.25
P01023A2M α-2-macroglobulin13752.5Inflammation: acute-phase5.45
C0JYY2APOBApolipoprotein B4818.8Lipid metabolic, inflammation13.55
P00738HPHaptoglobin11667.5Inflammation: acute-phase33.4195
A8K5T0CFHComplement factor H6238.7Inflammation: acute-phase3.37
A5PL27CPCeruloplasmin6641.3Inflammation: acute-phase; copper metabolism5.25
B7Z9A0GSNGelsolin4439.2Motility, signaling, apoptosis, cancer−14.60
G3V5I3SERPINA3 α-1-antichymotrypsin5547.1Inflammation: acute-phase10.19
P10645CHGAChromogranin-A2841.8Regulation of blood pressure−31.35
E9PGN7SERPING1Plasma protease C1 inhibitor2928.4Regulation of complement activation blood coagulation, fibrinolysis and the generation of kinins5.65
Q8WXD2SCG3 Secretogranin-31223.1Platelet activation platelet degranulation−7.94
P02671FGAFibrinogen α chain1417.1Coagulation, inflammation13.80
Q12860CNTN1Contactin-1614.4Notch signaling95.24
P06702S100A9Protein S100-A9970.2Inflammation: acute-phase40.93
P02647APOA1Apolipoprotein A-I6186.5Lipid metabolic, inflammation4.45
B2RMS9ITIH4Inter-α (Globulin) inhibitor H42225.7Inflammation: acute-phase; development or regeneration4.25
P36955SERPINF1Pigment epithelium-derived factor1629Cell proliferation−8.47
Q5VVQ8C4BPAComplement component 4 binding protein924.8Inflammation: acute-phase3.56
Q92876KLK6Kallikrein-61749.2Regulation of cell differentiation−30.77
P02649APOEApolipoprotein E5648.6Regulation of cell growth, lipid metabolic−3.66
Q68CK4LRG1Leucine-rich α-2-glycoprotein1223.3Inflammation: acute-phase9.29
F8VVB6NELL2Protein kinase C-binding protein1119.9Cell growth and differentiation−20.88
P16870CPECarboxypeptidase E927.9Protein modification process−72.46
Q14CA1NRCAMNRCAM protein1217Cell morphogenesis−22.47
E7EUF1ENPP2Ectonucleotide pyrophosphatase/phosphodiesterase family member 21824.6Immune system process, lipid metabolic process−15.85
Q16270IGFBP7Insulin-like growth factor-binding protein 7933.7Regulation of cell growth−43.67
B4E1Z4CFBComplement factor B4028.9Inflammation: acute-phase2.27
P02790HPXHemopexin9655.6Inflammation: acute-phase4.57
P01031C5Complement C52219Inflammation: acute-phase4.02
P05109S100A8Protein S100-A8538.7Defense response29.1
P02763ORM1α-1-acid glycoprotein6955.2Inflammation: acute-phase9.38
Q8TCF0LBP Lipopolysaccharide-binding protein310.5Negative regulation of cytokine12.13
P19021PAMPeptidyl-glycine α-amidating monooxygenase69.5Central nervous system development−22.88
D9YZU5HBBHemoglobin881.6Inflammation: acute-phase−79.36
P01871IGHMIg μ chain C region2633.2Inflammation: acute-phase13.06
F8W7I2PTGDSProstaglandin-H2 D-isomerase4445.5Lipid metabolic−18.87
O15240VGFNeurosecretory protein616.8Ovarian follicle development−14.99
D3DVW9PTPNS1Protein tyrosine phosphatase418.7Enhances cell proliferation, cell motility and invasive activity, and promotes cancer metastasis−22.27
P02748C9Complement component C92237.4Inflammation: acute-phase4.88
Q14520HABP2Hyaluronan-binding protein 2722Blood coagulation3.69
P51693APLP1Amyloid-like protein 11523.9Blood coagulation−11.06
Q9NQ79CRTAC1Cartilage acidic protein 1511.7Calcium ion binding−2.70
E9PEV0APPγ-secretase C-terminal fragment 591526.2Apoptosis, cell adhesion, endocytosis, Notch signaling pathway−18.87
A6XGL1TtrTransthyretin8085.3Transport thyroxine−11.38
P00734F2Prothrombin3845.2Blood coagulation1.85
P01008SERPINC1 Antithrombin-III4453Blood coagulation, inflammation3.13
P02741CRPC-reactive protein212.1Inflammation: acute-phase22.28
O43866CD5LCD5 antigen-like520.2Inflammation: acute-phase3.37
Q9BX83HBA1Hemoglobin α 1 globin chain643Inflammation: acute-phase−11.59
B2R4C5LYZLysozyme642.6Bacteriolytic function19.41
O95502NPTXRNeuronal pentraxin receptor1018.4Synapse remodeling−3.77
K9JA46EL52Epididymis luminal secretory protein 52310.4Neuron migration, cardiac muscle cell apoptotic process2.22
Q92859NEO1Neogenin25.5Glycolysis and gluconeogenesis−7.11
J3KPS3ALDOA Fructose-bisphosphate aldolase729.1Glycolysis and gluconeogenesis4.20
P13521SCG2 Secretogranin-236.3MAPK cascade, angiogenesis, endothelial cell migration−20.49
D3DNU8KNG1Kininogen 13246.6Blood coagulation, inflammation4.49
D3DQX7SAA1Serum amyloid A protein436.1Inflammation: acute-phase13.55
Q32Q65FGBFibrinogen β chain2546.6Coagulation, inflammation5.445
Q53FR6COMPCartilage oligomeric matrix protein variant715.6Coagulation, apoptosis−13.55
Q9NT99LRRC4BLeucine-rich repeat-containing protein 4B39.5Positive regulation of synapse assembly−12.47
P12109COL6A1Collagen α-1(VI) chain49.6Cell adhesion, blood coagulation11.48
D3DTX7COL1A1Collagen, type I, α 1, isoform CRA_a922.2Blood vessel development−1.29
Q6FHM9CD59CD59 antigen, complement regulatory protein, isoform CRA_b632.8Inflammation−17.54
Q06033ITIH3Inter-α-trypsin inhibitor heavy chain H3510.3Carbohydrate metabolic process4.25
P19022CDH2Cadherin-2312.1Blood vessel development−5.60
D6RD17IGJImmunoglobulin J chain520.3Inflammation: acute-phase14.99
P17900GM2AGanglioside GM2 activator424.9Carbohydrate metabolic process−6.98
Q86XR1PRNPMajor prion protein (fragment)413.5Neuronal development−16.45
A4D2D2PCOLCEProcollagen C-endopeptidase enhancer821.2Developmental process−10.57
Q86U78AGT Angiotensinogen1534.9Regulation of blood pressure, body fluid and electrolyte−3.02
Q53HB8MOGOligodendrocyte myelin glycoprotein variant312.7Inflammation−8.63
C9JEU5FGAFibrinogen γ chain3253.7Coagulation, inflammation12.13
P01034CST3Cystatin-C6356.9Negative regulation of blood vessel remodeling−17.70
Q7Z532OGNOsteoglycin219.1Induces bone formation−8.02
P23471PTPRZ1Receptor-type tyrosine-protein phosphatase ζ35.3Cell morphogenesis−5.44
P00558PGK1Phosphoglycerate kinase 1532.1Carbohydrate metabolic process4.33
P25311AZGP1 Zinc-α-2-glycoprotein1839.6Inflammation: acute-phase2.65
P24592IGFBP6Insulin-like growth factor-binding protein 6723.3Regulation of cell growth−4.74
P23142FBLN1Fibulin-11833Cell adhesion and migration−2.63
Q5NV91IGLV3-27V2-19 protein748.4Inflammation, complement activation8.32
P13591NCAM1Neural cell adhesion molecule 11021.5Regulation of signal−6.08
Annotation of differential proteins

Some differential proteins exhibited multiple functions and subcellular locations, which resulted in individual proteins being counted more than once. Considering that a given protein can be attributed to several functions and/or processes, we categorized differential proteins by their biological processes (Fig. 1A) and molecular functions (Fig. 1B) by pie chart. ‘Response to stimulus’, ‘biological adhesion’ and ‘biological regulation’ were the most over-represented biological processes. ‘Enzyme inhibitor activity’ was the most over-represented molecular function, distantly followed by ‘protein binding’ and ‘cell surface binding’.

Altered biological pathways by KEGG

The 81 differential proteins were analyzed for KEGG over-representation of pathways (‘proteomic phenotyping’) (24) to obtain insights into the functional differences between the CSF proteomes of TBM and HC subjects. The top three-ranking canonical KEGG pathways are listed in Table II. Inflammatory processes were the most statistically over-represented, including complement and coagulation cascades ranking first (P<0.001) and cell adhesion molecules (CAMs) ranking third (P<0.01).

Validation of NELL2 by western blotting

The neuron-specific differential protein with the largest absolute fold-change value, neural epidermal growth factor-like like 2 (NELL2), was selected for western blot validation (Table II). In accordance with iTRAQ™, NELL2 decreased significantly in TBM compared to HC subjects (Fig. 2A). ROC curves were constructed using the western blotting data, in which the AUC, sensitivity, and specificity were 0.868, 83.3 and 75%, respectively (Fig. 2B).

Discussion

In the present study, using an iTRAQ™-based quantitative proteomic approach, we compared 12 CSF samples from TBM subjects to 12 CSF samples from demographically-matched HC subjects. Among the 81 differential proteins identified, the neuron-specific protein with the largest absolute fold-change value, NELL2, was selected for western blotting validation and was found to be significantly decreased in TBM subjects compared to HC subjects. NELL2 had 83.3% sensitivity and 75% specificity and shows promise as a diagnostic biomarker for TBM.

Inflammation

KEGG analysis revealed over-representation in two inflammation-associated processes, namely complement and coagulation cascades and CAMs (Table III). As MTB infects and persists inside host macrophages, MTB infection is naturally accompanied by a host inflammatory response (25). In the initial stage, MTB activates several pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) (26), interleukin 1 (IL-1) (27), and reactive oxygen/nitrogen intermediaries (ROI/RNI) that promote MTB termination (28). Simultaneously, MTB induces the release of anti-inflammatory molecules, such as transforming growth factor-β (TGF-β) (29) and IL-10 (30), to suppress the pro-inflammatory response and maintain MTB survival. In ~90% of MTB cases, the host inflammatory response results in granuloma formation, which is sufficient to contain the infection and prevent disease. However, in 5–10% of MTB cases, the pathogen evades or subverts the host’s granulomatous inflammatory response, producing either a latent or active infection (31). Therefore, the outcome of MTB infection depends on a complex set of host-pathogen interactions, and further investigation into the differential proteins associated with the host inflammatory response can offer insight into the pathogenesis and treatment of TBM. For example, drugs that intervene in the host inflammatory response, such as TGF-β blockers, have been suggested as a method to prevent the spread of MTB infection (32).

Table III

Top ten-ranking canonical KEGG pathways associated with differential proteins.

Table III

Top ten-ranking canonical KEGG pathways associated with differential proteins.

KEGG pathwayNo. of molecules
P-value
Mapping%
Complement and coagulation cascades1016.7 2.40e−11
Prior diseases46.7 7.00e−4
Cell adhesion molecules (CAMs)58.3 10.20e−3
Glycolysis/gluconeogenesis

Two significantly altered proteins including neogenin and fructose-bisphosphate aldolase related to glycolysis/gluconeogenesis were identified. MTB prefers to utilize host fatty acids as an energy source. Thus, glycolytic/gluconeogenic proteins that are involved in fatty acid utilization would be required for the growth and persistence of MTB in vivo (33).

NELL2

TBM is characterized by meningeal inflammation, CSF flow obstruction, fibrogelatinous exudates, and arterial vasculitis that traverse the exudates, resulting in cerebral infarction (34,35). TBM sometimes results in tuberculoma formation (36), which is most commonly observed in the cerebellum, cerebrum and pons (37). In the present study, by magnetic resonance imaging (MRI), the 12 TBM subjects excibited basal meningeal enhancement consistent with TBM, seven of whom excibited cerebral infarction shown by high DWI signal intensity. These findings suggest a strong possibility of brain neuronal cell damage via vasculitis and tuberculoma formation. Considering that CSF neuron-specific enolase (NSE) is the most commonly applied biomarker of nervous tissue damage and can act as an early diagnostic marker of TBM (38,39), we selected NELL2 for western blotting validation due to its neuronal specificity and high absolute fold-change value. NELL2, also known as neural epidermal growth factor-like like 2, NEL-like protein 2, Nel-related protein 2 (NRP2), and protein kinase C-binding protein, is a cytoplasmic enzyme that is expressed almost exclusively in the nervous system (40), predominantly in neurons with the highest density in the hippocampus and cerebral cortex (41). NELL2 promotes neuronal differentiation and survival during embryonic development (42) through mitogen-activated protein kinases (MAPK) (43), stimulates motor and sensory neuron differentiation in dorsal root ganglia (44), and promotes the differentiation of neural progenitor cells into neurons (45). NELL2 knock-out mice have demonstrated learning impairment in the Morris water maze task.

The present study revealed significantly decreased levels of NELL2 in TBM patients. As neuron-produced NELL2 supports neuronal survival and differentiation and neurons are compromised in TBM, it is reasonable to surmise that the reduced NELL2 levels observed in TBM patients result from brain neuronal cell damage or death. Additional study is required to determine whether there is a TBM disease stage in which NELL2 significantly increases to compensate for brain neuronal cell damage.

There are several limitations to the present study. First, eight of the 12 TBM patients also had pulmonary TB, and another TBM patient had vertebral TB. As 25% of miliary TB patients also have TBM, presumably by transmission across the blood-brain barrier (46), future studies should include TBM patient cohorts with and without pulmonary TB, which may provide additional novel biomarkers of TBM. Second, the World Health Organization (WHO) recommends that all newly diagnosed human immunodeficiency virus (HIV) and syphilis patients should be tested for TB, as individuals with HIV or syphilis and latent TB infection are at a significantly higher risk of developing active TB (47). However, since no subjects of the present study were co-infected with HIV or syphilis, future studies should include cohorts of HIV or syphilis co-infected patients to improve our understanding of MTB host interaction in these patient populations. Third, the influence of anti-TB agents on differential protein expression was not assessed and should be taken into consideration in future studies. Fourth, in order to improve specificity for TBM, the current findings require further validation in larger sample sets that include other intracranial infectious diseases.

In conclusion, an iTRAQ™-based quantitative proteomic approach was utilized in the present study to identify differential proteins in the CSF of TBM patients and healthy controls. A total of 111 differential proteins were identified, and NELL2 was validated by western blotting as a promising diagnostic biomarker for TBM. Further evaluation of these findings in larger studies including medicated and unmedicated cohorts with other intracranial infectious diseases is required for clinical translation.

Acknowledgments

We graciously thank Dr Wei Ding (Shanghai Institute for Biological Sciences, Chinese Academy of Sciences) for his assistance with the mass spectrometric analysis and his technical assistance with the KEGG database. We also extend thanks to Dr N.D. Melgiri for editing and proofreading the manuscript. The present study was supported by the National Key Scientific Program of China (nos. 2009CB918300 and 2012CB910602) and the National Nature Science Foundation of China (no. 81371310).

References

1 

WHO global tuberculosis control report 2010. Summary. Cent Eur J Public Health. 18:2372010.

2 

Corbett EL, Watt CJ, Walker N, Maher D, Williams BG and Raviglione MC: The growing burden of tuberculosis: global trends and interactions with the HIV epidemic. Arch Intern Med. 163:1009–1021. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Zhao Y, Xu S, Wang L, Chin DP, Wang S and Jiang G: National survey of drug-resistant tuberculosis in China. N Engl J Med. 366:2161–2170. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Thwaites GE: Advances in the diagnosis and treatment of tuberculous meningitis. Curr Opin Neurol. 26:295–300. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Thwaites GE, van Toorn R and Schoeman J: Tuberculous meningitis: more questions, still too few answers. Lancet Neurol. 12:999–1010. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Rylance J, Pai M, Lienhardt C and Garner P: Priorities for tuberculosis research: a systematic review. Lancet Infect Dis. 10:886–892. 2010. View Article : Google Scholar : PubMed/NCBI

7 

McNerney R, Maeurer M, Abubakar I, et al: Tuberculosis diagnostics and biomarkers: needs, challenges, recent advances, and opportunities. J Infect Dis. 205:S147–S158. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Yew WW, Borgdorff MW and Enarson DA: New tools for global tuberculosis control: are we any closer? Int J Tuberc Lung Dis. 16:855–856. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Thwaites G, Chau TT, Mai NT, Drobniewski F, McAdam K and Farrar J: Tuberculous meningitis. J Neurol Neurosurg Psychiatry. 68:289–299. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Eintracht S, Silber E, Sonnenberg P, Koornhof HJ and Saffer D: Analysis of adenosine deaminase isoenzyme-2 (ADA2) in cerebrospinal fluid in the diagnosis of tuberculosis meningitis. J Neurol Neurosurg Psychiatry. 69:137–138. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Donald PR, Malan C, van der Walt A and Schoeman JF: The simultaneous determination of cerebrospinal fluid and plasma adenosine deaminase activity as a diagnostic aid in tuberculous meningitis. S Afr Med J. 69:505–507. 1986.PubMed/NCBI

12 

Mishra OP, Loiwal V, Ali Z, Nath G and Chandra L: Cerebrospinal fluid adenosine deaminase activity for the diagnosis of tuberculous meningitis in children. J Trop Pediatr. 42:129–132. 1996. View Article : Google Scholar : PubMed/NCBI

13 

Mudaliar AV, Kashyap RS, Purohit HJ, Taori GM and Daginawala HF: Detection of 65 kD heat shock protein in cerebrospinal fluid of tuberculous meningitis patients. BMC Neurol. 6:342006. View Article : Google Scholar : PubMed/NCBI

14 

Donald PR and Malan C: Cerebrospinal fluid lactate and lactate dehydrogenase levels as diagnostic aids in tuberculous meningitis. S Afr Med J. 67:19–20. 1985.PubMed/NCBI

15 

Kataria J, Rukmangadachar LA, Hariprasad G, O J, Tripathi M and Srinivasan A: Two dimensional difference gel electrophoresis analysis of cerebrospinal fluid in tuberculous meningitis patients. J Proteomics. 74:2194–2203. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Walzl G, Ronacher K, Hanekom W, Scriba TJ and Zumla A: Immunological biomarkers of tuberculosis. Nat Rev Immunol. 11:343–354. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Waybright TJ: Preparation of human cerebrospinal fluid for proteomics biomarker analysis. Methods Mol Biol. 1002:61–70. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Kumar GS, Venugopal AK and Mahadevan A: Quantitative proteomics for identifying biomarkers for tuberculous meningitis. Clin Proteomics. 9:122012. View Article : Google Scholar : PubMed/NCBI

19 

Tumani H, Lehmensiek V, Lehnert S, Otto M and Brettschneider J: 2D DIGE of the cerebrospinal fluid proteome in neurological diseases. Expert Rev Proteomics. 7:29–38. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Ross PL, Huang YN, Marchese JN, Williamson B, Parker K, Hattan S, et al: Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol Cell Proteomics. 3:1154–1169. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Ahuja GK, Mohan KK, Prasad K and Behari M: Diagnostic criteria for tuberculous meningitis and their validation. Tuber Lung Dis. 75:149–152. 1994. View Article : Google Scholar : PubMed/NCBI

22 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W and Lane HC: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI

23 

Brea D, Sobrino T, Blanco M, Fraga M, Agulla J and Rodriguez-Yanez M: Usefulness of haptoglobin and serum amyloid A proteins as biomarkers for atherothrombotic ischemic stroke diagnosis confirmation. Atherosclerosis. 205:561–567. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Pan C, Kumar C, Bohl S, Klingmueller U and Mann M: Comparative proteomic phenotyping of cell lines and primary cells to assess preservation of cell type-specific functions. Mol Cell Proteomics. 8:443–450. 2009. View Article : Google Scholar :

25 

Toossi Z: The inflammatory response in Mycobacterium tuberculosis infection. Arch Immunol Ther Exp (Warsz). 48:513–519. 2000.

26 

Valone SE, Rich EA, Wallis RS and Ellner JJ: Expression of tumor necrosis factor in vitro by human mononuclear phagocytes stimulated with whole Mycobacterium bovis BCG and mycobacterial antigens. Infect Immun. 56:3313–3315. 1988.PubMed/NCBI

27 

Wallis RS, Fujiwara H and Ellner JJ: Direct stimulation of monocyte release of interleukin 1 by mycobacterial protein antigens. J Immunol. 136:193–196. 1986.PubMed/NCBI

28 

Bulut Y, Michelsen KS, Hayrapetian L, Naiki Y, Spallek R and Singh M: Mycobacterium tuberculosis heat shock proteins use diverse Toll-like receptor pathways to activate pro-inflammatory signals. J Biol Chem. 280:20961–20967. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Hirsch CS, Toossi Z, Othieno C, Johnson JL, Schwander SK and Robertson S: Depressed T-cell interferon-γ responses in pulmonary tuberculosis: analysis of underlying mechanisms and modulation with therapy. J Infect Dis. 180:2069–2073. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Lin Y, Zhang M, Hofman FM, Gong J and Barnes PF: Absence of a prominent Th2 cytokine response in human tuberculosis. Infect Immun. 64:1351–1356. 1996.PubMed/NCBI

31 

Stavrum R, Stavrum AK, Valvatne H, Riley LW, Ulvestad E, Jonassen I, et al: Modulation of transcriptional and inflammatory responses in murine macrophages by the Mycobacterium tuberculosis mammalian cell entry (Mce) 1 complex. PLoS One. 6:e262952011. View Article : Google Scholar : PubMed/NCBI

32 

Sasindran SJ and Torrelles JB: Mycobacterium tuberculosis infection and inflammation: what is beneficial for the host and for the hacterium? Front Microbiol. 2:22011. View Article : Google Scholar

33 

Shi L, Sohaskey CD, Pfeiffer C, Datta P, Parks M and McFadden J: Carbon flux rerouting during Mycobacterium tuberculosis growth arrest. Mol Microbiol. 78:1199–1215. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Thwaites GE and Schoeman JF: Update on tuberculosis of the central nervous system: pathogenesis, diagnosis, and treatment. Clin Chest Med. 30:745–754. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Malhotra HS, Garg RK, Singh MK, Agarwal A and Verma R: Corticosteroids (dexamethasone versus intravenous methylprednisolone) in patients with tuberculous meningitis. Ann Trop Med Parasitol. 103:625–634. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Garg RK: Tuberculosis of the central nervous system. Postgrad Med J. 75:133–140. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Damergis JA, Leftwich EI, Curtin JA and Witorsch P: Tuberculoma of the brain. JAMA. 239:413–415. 1978. View Article : Google Scholar : PubMed/NCBI

38 

Rodriguez-Nunez A, Cid E, Rodriguez-Garcia J, Camina F, Rodriguez-Segade S and Castro-Gago M: Neuron-specific enolase, nucleotides, nucleosides, purine bases, oxypurines and uric acid concentrations in cerebrospinal fluid of children with meningitis. Brain Dev. 25:102–106. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Song TJ, Choi YC, Lee KY and Kim WJ: Serum and cerebrospinal fluid neuron-specific enolase for diagnosis of tuberculous meningitis. Yonsei Med J. 53:1068–1072. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Watanabe TK, Katagiri T, Suzuki M, Shimizu F, Fujiwara T and Kanemoto N: Cloning and characterization of two novel human cDNAs (NELL1 and NELL2) encoding proteins with six EGF-like repeats. Genomics. 38:273–276. 1996. View Article : Google Scholar : PubMed/NCBI

41 

Oyasu M, Kuroda S, Nakashita M, Fujimiya M, Kikkawa U and Saito N: Immunocytochemical localization of a neuron-specific thrombospondin-1-like protein, NELL2: light and electron microscopic studies in the rat brain. Brain Res Mol Brain Res. 76:151–160. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Tong ZB, Bondy CA, Zhou J and Nelson LM: A human homologue of mouse Mater, a maternal effect gene essential for early embryonic development. Hum Reprod. 17:903–911. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Aihara K, Kuroda S, Kanayama N, Matsuyama S, Tanizawa K and Horie M: A neuron-specific EGF family protein, NELL2, promotes survival of neurons through mitogen-activated protein kinases. Brain Res Mol Brain Res. 116:86–93. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Nelson BR, Claes K, Todd V, Chaverra M and Lefcort F: NELL2 promotes motor and sensory neuron differentiation and stimulates mitogenesis in DRG in vivo. Dev Biol. 270:322–335. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Hwangpo TA, Jordan JD, Premsrirut PK, Jayamaran G, Licht JD and Iyengar R: G Protein-regulated inducer of neurite outgrowth (GRIN) modulates Sprouty protein repression of mitogen-activated protein kinase (MAPK) activation by growth factor stimulation. J Biol Chem. 287:13674–13685. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Jain SK, Paul-Satyaseela M, Lamichhane G, Kim KS and Bishai WR: Mycobacterium tuberculosis invasion and traversal across an in vitro human blood-brain barrier as a pathogenic mechanism for central nervous system tuberculosis. J Infect Dis. 193:1287–1295. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Goswami ND, Hecker EJ, Vickery C, Ahearn MA, Cox GM and Holl DP: Geographic information system-based screening for TB, HIV, and syphilis (GIS-THIS): a cross-sectional study. PLoS One. 7:e460292012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 35 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang Y, Mu J, Chen G, Zhan Y, Zhong J, Wei Y, Cheng K, Qin B, You H, Xie P, Xie P, et al: iTRAQ-based quantitative proteomic analysis of cerebrospinal fluid reveals NELL2 as a potential diagnostic biomarker of tuberculous meningitis. Int J Mol Med 35: 1323-1332, 2015
APA
Yang, Y., Mu, J., Chen, G., Zhan, Y., Zhong, J., Wei, Y. ... Xie, P. (2015). iTRAQ-based quantitative proteomic analysis of cerebrospinal fluid reveals NELL2 as a potential diagnostic biomarker of tuberculous meningitis. International Journal of Molecular Medicine, 35, 1323-1332. https://doi.org/10.3892/ijmm.2015.2131
MLA
Yang, Y., Mu, J., Chen, G., Zhan, Y., Zhong, J., Wei, Y., Cheng, K., Qin, B., You, H., Xie, P."iTRAQ-based quantitative proteomic analysis of cerebrospinal fluid reveals NELL2 as a potential diagnostic biomarker of tuberculous meningitis". International Journal of Molecular Medicine 35.5 (2015): 1323-1332.
Chicago
Yang, Y., Mu, J., Chen, G., Zhan, Y., Zhong, J., Wei, Y., Cheng, K., Qin, B., You, H., Xie, P."iTRAQ-based quantitative proteomic analysis of cerebrospinal fluid reveals NELL2 as a potential diagnostic biomarker of tuberculous meningitis". International Journal of Molecular Medicine 35, no. 5 (2015): 1323-1332. https://doi.org/10.3892/ijmm.2015.2131