Open Access

Proteomic profiling of biomarkers by MALDI‑TOF mass spectrometry for the diagnosis of tracheobronchial stenosis after tracheobronchial tuberculosis

  • Authors:
    • Bihao Peng
    • Xiaojian Qiu
    • Zhiwu Dong
    • Jie Zhang
    • Yinghua Pei
    • Ting Wang
  • View Affiliations

  • Published online on: November 19, 2020     https://doi.org/10.3892/etm.2020.9495
  • Article Number: 63
  • Copyright: © Peng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tracheobronchial tuberculosis (TB) leads to airway stenosis, irreversible airway damage and even death. The present study aimed to identify biomarkers for the diagnosis of tracheobronchial stenosis (TBS) secondary to tracheobronchial TB. A cohort was recruited, including patients with TBS after tracheobronchial TB, TBS after tracheal intubation or tracheotomy (TIT) and no stenosis of early‑stage lung cancer,. Proteomic profiling was performed to gain insight into the mechanisms of the pathological processes. Differentially expressed proteins in the serum and bronchial alveolar lavage fluid (BALF) from patients were detected by matrix‑assisted laser desorption ionization time‑of‑flight mass spectrometry (MALDI‑TOF MS). Subsequently, ELISA was performed to validate the changes of protein levels in an additional cohort. MALDI‑TOF MS revealed that 8 peptides in the serum, including myeloid‑associated differentiation marker, keratin type I cytoskeletal 18, fibrinogen α‑chain, angiotensinogen (AGT), apolipoprotein A‑I (APOAI), clusterin and two uncharacterized peptides, and nine peptides in BALF, including argininosuccinate lyase, APOAI, AGT and five uncharacterized peptides, were differentially expressed (molecular‑weight range, 1,000‑10,000 Da) in the TB group compared with the TIT group. The ELISA results indicated that the changes in the protein levels had a similar trend as those identified by proteomic profiling. In conclusion, the present study identified proteins that may serve as potential biomarkers and provide novel insight into the molecular mechanisms underlying TBS after tracheobronchial TB.

Introduction

Benign airway stenosis may be congenital or acquired, and the causes of acquired airway stenosis include trauma, surgery, tracheobronchial TB, tracheal intubation, tracheotomy, stent insertion and inhalation injury. Airway injury results in edema and erythema of the mucosal layer, which persists as long as the pathogenic factors are present. As a consequence, the ring cartilage is exposed and inflamed, collagen and scar tissue are produced and the ring contracts, resulting in the formation of a hard stenosis covered with epithelial cells (1). Tracheobronchial TB refers to Mycobacterium tuberculosis infection of the mucosa, submucosa, smooth muscle, cartilage and the outer membrane of trachea and bronchus (2). With an increasingly mobile population and the continuing human immunodeficiency virus acquired immunodeficiency syndrome pandemic, the numbers of patients with TB are gradually increasing and the incidence of tracheobronchial TB in patients with active TB ranged from 6.4 to 40% in Iran, 2014(3). Of all patients with tracheobronchial TB in India (2014), >90% suffered from airway stenosis to varying degrees (4). A previous study demonstrated that an age of >45 years, fibrostenotic subtype and anti-TB chemotherapy are independent clinical predictors of persistent airway stenosis (5). However, only a small number of clinical biomarkers have been identified for the diagnosis of tracheobronchial stenosis (TBS) after tracheobronchial TB. Currently, TBS is confirmed by clinical manifestations, chest CT (6) and bronchoscopy (7); however, there is a lack of rapid and effective laboratory indicators. As it is difficult to differentiate TBS after TB from other lung diseases, including TBS after tracheal intubation or tracheotomy (TIT), the condition is frequently diagnosed at a late stage and numerous patients have missed the optimal time window for treatment (8). With early diagnosis, clinicians are able to provide appropriate and timely therapy, preventing the development of airway stenosis and improving the patients' prognosis (9).

Proteomics generally refers to large-scale studies of all of the proteins in cells, tissues or organisms, or in specialized samples, including body fluids (10). Proteomics consists of overall protein profiling, including expression levels, structure, interactions and modifications after translation; therefore, it represents a noninvasive method to diagnose various diseases (11). In the last few decades, proteomics has provided extensive data and insight into the pathophysiological mechanisms of numerous human diseases, including carcinoma, infection and coronary heart disease, as well as the identification of novel biomarkers and therapeutic targets for clinical diagnostics and treatments (12). Proteomics primarily involves two-dimensional gel electrophoresis, western blotting, immunoprecipitation and mass spectrometry (MS) (13).

MS is used to identify chemical compounds by ionizing them into charged molecules and separating the ions by their mass-to-charge ratio (m/z) (14). Since the introduction of the application of matrix-assisted laser desorption/ionization (MALDI) in the 1990s, MS has been widely used to profile large biological molecules (15). Large biological molecules, including DNA, proteins and sugars, are ionized by gaining or losing a proton without affecting the characteristics of the sample. The sample to be detected is dispersed into matrix material located on a metal plate and the sample and the matrix are irradiated by a pulsed laser and vaporized. The target molecules are then ionized and ready for acceleration by MS. Time-of-flight (TOF) MS examines the time it takes for ions to cover fixed distances with velocities determined by their m/z (16). In theory, ions with small m/z values or with more charges will reach the detector earlier; thus, an m/z value is determined by the time of flight and the molecule is identified accordingly. Considering MALDI-TOF MS has high-throughput capability and can be completely automated, it is ideal for proteomics research (17,18).

Proteomic changes in airway stenosis evoked by tracheobronchial TB have remained largely undefined. The aim of the present study was to compare protein expression profiles in the sera and bronchoalveolar lavage fluids (BALFs) to clarify the pathogenesis of TBS after tracheobronchial TB.

Patients and methods

Patients

A total of 91 patients with TBS after TB, TBS after TIT and early-stage lung cancer (ESLC) were enrolled at the Department of Respiratory Diseases, Beijing Tian Tan Hospital, Capital Medical University (Beijing, China) from January 2014 to October 2018. Patients enrolled between January 2014 to October 2015 were subjected to MS analysis and patients enrolled between November 2015 to October 2018 to ELISA. Patients with TBS after TB, TBS after TIT and ESLC had been confirmed by chest CT scan and clinical testing, including reverse-transcription PCR assay for TB and tumor markers of CEA, CA15-3, CA19-9, CA125, neuron-specific enolase and cytokeratin fraction 21-1. All of the patients provided written informed consent to participate in the present study and for use of their medical data according to the provisions of the Declaration of Helsinki. The protocol of the present study was approved by the Institutional Ethics Committee of Beijing Tian Tan Hospital. The serum and BALF samples from 31 patients were subjected to MALDI-TOF MS and their characteristics are presented in Table SI. The serum samples from the other 60 patients (20 with TB, 20 with TIT and 20 with ESLC) were used for validation with ELISAs (Table SII). There were no significant differences in age and sex between groups.

Sample collection

Serum samples were collected, processed and stored in according to a technique similar to a standard protocol (19). Blood samples were obtained from fasted patients in the morning (collected in vacuum tubes without preservatives or anticoagulant) and allowed to clot or to sediment at room temperature (~25˚C) for 2 h to isolate the serum. Serum samples were then separated by centrifugation at 3,000 x g at 23˚C for 15 min. Samples were immediately divided into aliquots of 50 µl and stored at -80˚C until further analysis. The cryopreservation period for serum samples was <24 months.

BALF samples were collected and processed according to a standard protocol (19). Samples were separated by centrifugation at 4,000 x g for 15 min at 4˚C. Supernatants were aliquoted into four 1 ml tubes and the precipitate was added into one 1ml tube. The samples were stored at -80˚C until use. The cryopreservation period for BALF samples was <24 months.

Weak cation exchange magnetic bead (MB-WCX) fractionation and MALDI-TOF MS analysis

All of the samples were fractionated using MB-WCX (Bruker Corporation), according to the manufacturer's protocol. The suspension was prepared with a magnetic bead-weak cation exchange MB-WCX kit (Bruker Corporation) and mixed by shaking. Following eluting and shocking, the magnetic beads were separated from the protein and the eluted peptide samples were transferred to a clean 0.5 ml tube for further MS analysis. A total of 5 µl of hydroxy-cyanocinnamic acid substrate solution (0.4 g/l; MilliporeSigma; dissolved in acetone and ethanol) and 0.8-1.2 µl of eluate were mixed. Subsequently, 0.8-1.2 µl of the mixture was applied to a metal target plate and dried at room temperature. Finally, air-dried target plates were measured immediately using a calibrated Autoflex III MALDI-TOF MS (Bruker Corporation) with Flex-Control software (version 3.0; Bruker Corporation), according to optimized measuring protocols (20). Peptides with molecular weights ranging from 1,000 to 10,000 kDa were collected with a laser intensity of 5,000 W/nm2. Peptide mass fingerprints were obtained by combining 50 individual MS signal scans. ClinProTools software (version 2.2; Bruker Corporation) was used to subtract the baseline, normalize spectra (using the total ion current) and determine peak m/z values and intensities in the mass range of 1,000-10,000 kDa. A signal-to-noise (S/N) ratio of >5 was selected to guarantee for clear acquisitions. An S/N of >3 was used for limit of detection and an S/N of >5 was used for quantification. Spectra were aligned with a mass shift of ≤0.1%. The peak area was measured for quantification. Based on the macroscopic results of the multivariate statistical analysis, the tendency of the relative integrals of each metabolite to change in the three groups was further analyzed by box-and-whisker plots.

ELISA

ELISA was performed to confirm the changes in the levels of myeloid-associated differentiation marker (MYADM), keratin, type I cytoskeletal 18 (KRT18), fibrinogen α-chain (FGA), angiotensinogen (AGT), apolipoprotein A-I (APOAI) and clusterin (CLU) in serum from another cohort of patients who were not subjected to MS analysis. This cohort included 20 patients with TB, 20 with TIT and 20 with ESLC. KRT18 (cat. no. MA1-19041; 1:5,000), FGA (cat. no. MA1-20038; 1:5,000), AGT (cat. no. MA5-29009; 1:10,000) and APOAI (cat. no. MIA1402; 1:5,000) antibodies were purchased from Thermo Fisher Scientific, Inc. MYADM (cat. no. ABIN6059266; 1:5,000) and CLU (cat. no. ABIN6574258; the microtiter plate provided in this kit was pre-coated with antibodies specific to CLU, 1:50) antibodies were purchased from Antibodies-Online. Polystyrene ELISA plates (Nunc Maxisorp; Bioon) were coated with capture antibodies (1:50) in bicarbonate buffer at 4˚C overnight. After the coating solution was removed, the plates were washed three times with 200 µl PBS containing 0.05% Tween-20 (PBST). The coated wells were then blocked for 1 h with 5% non-fat dry milk in PBST at room temperature. Equal amounts of samples were added to each well and the plates were incubated for 1 h at 37˚C. The samples were then removed and the plates were washed three times with PBST. Horseradish peroxidase-conjugated antibodies (Thermo Fisher Scientific, Inc.; cat. nos. 32230 and 65-6120; dilutions,1:5,000 and 1:3,000, respectively) in bicarbonate buffer were added and the plates were incubated at 37˚C for 45 min. After washing with PBST, 3,3',5,5'-Tetramethylbenzidine substrate was added to each well, followed by incubation at 37˚C for 15 min. The reaction was stopped by adding 50 µl 1 mol/l H2SO4. The absorbance was read by an ELISA reader at 450 nm (with a reference wavelength of 620 nm). All samples were tested in duplicate.

Statistical analysis

Values are expressed as the mean ± standard deviation. Multiple comparisons were performed using one-way ANOVA with Fisher's least-significant differences (LSD) test as the post-hoc test. P<0.05 was considered to indicate a statistically significant difference. Genetic algorithm (GA), supervised neural networks (SNN) and quick classifier (QC) algorithms were applied to identify differentially expressed peptides distinguishing TBS after TB or TIT and ESLC.

Results

General information

Serum and BALF proteomic profiles of TBS after TB, TBS after TIT and ESLC groups were compared. There were no significant differences in age, BALF return rate, protein abundance (estimated by the rate of albumin in BALF/serum) between the three groups (Table SI). There were more female than male patients in the TB group (Table SI). Fr Fractionation of serum and BALF samples by MB-WCX and analysis by MALDI-TOF MS was performed to obtain the proteomic profiles of patients with TB (red), patients with TIT (green) and patints with ESLC (blue) with proteins ranging from 1,000 to 10,000 kDa (Fig. 1).

Differences in serum and BALF peptides

Using ClinProTools software to analyze serum samples, 155 peptide peaks were identified at an S/N threshold of 3 within the mass range analyzed (1,000-10,000 kDa). A fold change of >1.5 and P<0.05 as determined by one-way ANOVA were set as the threshold for differentially expressed proteins and Fisher's LSD test was used to determine differences between pairs of groups. A total of 5 proteins, including CLU, AGT, FGA and two undefined peptides (Fisher's LSD 1-2 or 2-1) differed significantly between the TB and TIT groups and 5 of the peaks differed significantly between the TB and ESLC groups. Furthermore, one of the peaks differed significantly between the TIT and ESLC groups (Table IA). In BALF samples, using the same thresholds as above, 9 peptide peaks were identified. A total of 5 peptides were differentially expressed between the TB and TIT groups, 7 peptides differed between the TB and ESLC groups and 4 peaks differed significantly between the TIT and ESLC samples (Table IB).

Table I

Mean levels of differentially expressed peptides in serum and BALF among the three groups.

Table I

Mean levels of differentially expressed peptides in serum and BALF among the three groups.

A, Serum
Mass (m/z)P-valueFisher's LSDaTBTITESLC (mean)
1,532.240.0115431-2; 1-315.63±8.976.68±2.908.14±2.98
6,816.510.0199532-15.26±1.957.75±3.819.96±3.73
1,180.930.0223591-314.01±8.006.80±2.909.47±2.80
4,185.050.0306281-247.16±36.4624.49±3.067.37±36.78
2,660.560.0392982-130.28±27.3047.49±41.5572.25±26.88
6,844.580.0400572-1; 2-39.85±5.5810.26±9.3515.18±5.65
4,419.720.0425673-135.41±9.4374.00±19.6049.82±53.13
4,300.830.0482043-135.85±15.1558.70±10.4753.15±21.94
B, BALF
Mass (m/z)P-valueFisher's LSDaTBTITESLC (mean)
6,029.680.0154651-2; 1-381.86±51.1147.49±19.6929.78±52.77
4,200.650.0156673-1; 3-211.41±10.6632.95±3.896.15±31.95
4,186.060.0194573-1; 3-220.21±20.9556.60±7.8610.66±55.13
6,701.810.0268342-1; 2-317.96±11.3211.61±28.4831.22±5.37
7,929.010.0304481-2; 1-3607.65±529.57205.62±454.00226.75±214.01
5,763.150.0323191-253.34±35.0037.48±13.7426.71±21.31
5,278.10.0323771-2; 1-3101.01±65.4353.93±57.9448.80±37.76
4,169.490.0406763-1; 3-213.57±13.2656.60±5.5610.66±28.43
4,533.750.0422051-311.78±6.375.76±4.347.03±3.05

[i] aComparisons: 1, TB; 2, ESLC; 3, TIT were statistically significant at a P<0.05. Data are presented as mean ± SD. BALF, bronchial alveolar lavage fluid; m/z, mass-to-charge ratio; LSD, least-significant differences test; TB, tuberculosis; TIT, tracheal intubation and tracheotomy; ESLC, early-stage lung cancer.

Based on the macroscopic results of the multivariate statistical analysis, the tendency of the relative integrals of each metabolite to change in the three groups was further analyzed by box-and-whisker plots. These plots illustrated the progressive changes in metabolite levels in the TB group or TIT group relative to the ESLC group (Fig. 2).

Model building

The GA, SNN and QC model-building functions embedded in ClinPro Tools 2.2 software were used to establish a cross-validated classification model to distinguish TB, TIT and ESCL groups in terms of serum and BALF proteins. The peaks that were selected by these three algorithms are presented in Table II.

Table II

Peak selection of the algorithm models.

Table II

Peak selection of the algorithm models.

 Serum peak selection (m/z)BALF peak selection (m/z)
AlgorithmTB vs. TITTIT vs. ESLCTB vs. ESLCTB vs. TITTIT vs. ESLCTB vs. ESLC
SNN1531, 4420, 3859, 9010, 4435, 5544, 4153, 4185, 5506, 4644, 4467, 7481, 1312, 5716, 8618, 7356, 7029, 3588, 9539, 575315395266, 4300, 839, 41697769, 4170, 4534, 4136, 2022, 8621, 3276, 3086, 6960, 2423, 1722, 5279, 6831, 3475, 2711, 4941, 3905, 8323, 3012, 3463, 5218, 2316, 2090, 142753836030, 3463, 3905, 1942, 5384, 1743, 6811, 3012, 2316, 922, 5699, 7621, 5279, 7542
QC855, 1050, 1066, 1150, 1180839, 855, 861, 975, 1011, 1050, 1066, 1072, 1251, 1450, 1523, 1539, 2379, 2448, 2462, 2473839, 2022, 2037, 2044, 2052, 2065, 2660, 2674, 3508, 3736, 4128, 4152, 4169, 4186, 4300, 7067, 9539833, 1312, 1330, 1449, 2316, 3475, 4186, 4200, 4216, 4534, 5279, 7929, 8584, 8621817, 833, 839, 8456030
GA1180, 3904, 3764, 2022, 46443314, 1251, 6888, 3486, 34013508, 1180, 7067, 4152, 44674186, 2723, 6029, 4941, 69606644, 4136, 839, 5383, 20325763, 2066, 2754, 3841, 5421

[i] SNN, supervised neural network; QC, quick classifier; GA, genetic algorithm; BALF, bronchial alveolar lavage fluid; m/z, mass-to-charge ratio; TB, tuberculosis; TIT, tracheal intubation and tracheotomy; ESLC, early-stage lung cancer.

Identification of peptides by MALDI-TOF MS

To determine which peptides in the MS profiles are possible biomarkers, the current study attempted to identify those peptides with higher concentrations. The peptides in the Fisher model were purified and analyzed by MALDI-TOF MS and 6 serum peptides and 4 BALF peptides were successfully identified. A total of 8 peptides in serum, including MYADM (m/z 1,180.93), KRT18 (4,300.83), FGA (2,660.56), AGT (4,185.05), APOAI (4,419.72), CLU (1,532.24) and two undefined peptides (6,816.51 and 6,844.58) were successfully sequenced (Table III). In BALF samples, 9 peptides, including argininosuccinate lyase (ASL; 4,200.65), APOAI (two sequences: 4,169.49 and 4,533.75), AGT (4,186.06) and five undefined peptides (6,029.68, 5,763.15, 7,929.01, 6,701.81 and 5,278.1) were sequenced (Table III). AGT and APOAI simultaneously appeared in both serum and BALF samples. APOAI was identified in both serum and BALF samples by its sequence KAKPALEDLRQGLLPVLE SFKVSFLSALEEYTKKLNTQ, while AGT was confirmed as the sequence KPEVLEVTLNRPFLFAVY DQSATALHFLGRVANPLSTA. Compared with the TIT group, AGT and CLU were significantly upregulated and FGA was downregulated in serum from patients with TB, implying their potential roles in the pathogenesis of the disease.

Table III

List of detected sequences.

Table III

List of detected sequences.

A, Serum
Mass (M+H; kDa)Protein symbolProtein nameSequence
1,180.93MYADMMyeloid-associated differentiation marker M.PVTVTRTTITT.T
4,300.83KRT18Keratin, type I cytoskeletal 18 K.NREELDKYWSQQIEESTTVVTTQSAEVGAAETTLTELR.R
2,660.56FGAIsoform 1 of fibrinogen α-chain A.DEAGSEADHEGTHSTKRGHAKSRPV.R
6,816.51-N/AN/A
6,844.58-N/AN/A
4,185.05AGTAngiotensinogen precursor N.KPEVLEVTLNRPFLFAVYDQSATALHFLGRVANPLSTA.-
4,419.72APOAIApolipoprotein A-I precursor S.EKAKPALEDLRQGLLPVLESFKVSFLSALEEYTKKLNTQ.-
1,532.24CLUClusterin precursor R.RPHFFFPKSRIV.R
B, BALF
Mass (M+H; kDa)Protein symbolProtein nameSequence
6,029.68-N/AN/A
5,763.15-N/AN/A
4,200.65ASL Argininosuccinatelyase D.FVAEFLFWASLCM*THLSRM*AEDLILYCTKEFSFVQ.L
7,929.01-N/AN/A
6,701.81-N/AN/A
4,169.49APOAIApolipoprotein A-I precursor K.AKPALEDLRQGLLPVLESFKVSFLSALEEYTKKLNTQ.-
4,186.06AGTAngiotensinogen precursor N.KPEVLEVTLNRPFLFAVYDQSATALHFLGRVANPLSTA.-
5,278.10-N/AN/A
4,533.75APOAIApolipoprotein A-I precursor L.SEKAKPALEDLRQGLLPVLESFKVSFLSALEEYTKKLNTQ.-

[i] M+H, protonated molecular ion; N/A, not available; BALF, bronchial alveolar lavage fluid.

Validation of differentially expressed peptides in serum by ELISA

To validate the results of MALDI-TOF MS, ELISA was applied for the quantification of the differentially expressed peptides in sera from 20 patients with TB, 20 patients with TIT and 20 patients with ESCL. Fold changes in the protein levels are presented in Fig. 3. The results indicated that MYADM, AGT and CLU were upregulated in the TB group as compared with those in the TIT and ESLC groups, while KRT18, FGA and APOAI were downregulated in the TB group compared with those in the TIT and ESLC groups. Notably, patients with ESLC had relatively high serum FGA levels and low APOAI levels compared with those in the TIT group (Fig. 3). There was an indication that these proteins may serve as potential markers to distinguish patients with TBS after TB from patients with TBS due to other causes.

Discussion

Patients with tracheobronchial TB frequently encounter TBS, which may lead to pulmonary complications and death. In the clinic, prevention of TBS is critical to reduce the risk of death. Bronchoscopic or surgical interventions are used to restore airway patency once significant stenosis occurs (2). Although certain studies focused on the biomarkers for pulmonary TB, the diagnosis of TBS after TB with non-invasive clinical tests appears to be particularly important (21,22). The present study aimed to identify putative biomarkers of TBS after tracheobronchial TB and the results revealed that MYADM, AGT, CLU and FGA in serum may serve as biomarkers for the diagnosis of TBS after tracheobronchial TB.

In the present study, MYADM was significantly upregulated in the serum of patients with TB compared with that in the ESLC group and downregulated in the serum of patients with TIT compared with that in the ESLC and TB groups. Full-length MYADM complementary (c)DNA was identified in a human bone marrow cDNA library in 2001 and was mapped to chromosome 19q 13.33-q 13.4(23). MYADM is expressed in specialized domains on the membrane surface of human endothelial and epithelial cells and binds with actin (24). Additionally, MYADM colocalizes with Rac family small GTPase1 protein in membrane rafts during cell migration (25). Knockdown of MYADM with small interfering (si)RNA led to enhanced cell permeability, intercellular adhesion molecule-1 expression and leukocyte adhesion, which are characteristics of the inflammatory phenotype in endothelial cells (24). Notably, upregulation of MYADM specifically correlates with carotid neointimal formation, which promoted aortic artery smooth muscle cell (SMC) differentiation in a rat carotid artery balloon-injury model (26). Airway SMCs are recognized as a primary pathological determinant in lung diseases, including asthma and emphysema (27). Their dynamic and multifunctional behavior contributes to inflammation, fibroproliferation, abnormal wound healing, airway remodeling and hypertrophic scar formation and, therefore, narrowing of the airway lumen, leading to irreversible airway stenosis (28).

In the present study, FGA was downregulated in the serum of patients with TB compared with that in patients in the TIT and ESLC groups. Patients with aspirin-exacerbated respiratory disease, which is associated with asthma severity, have also been reported to exhibit reduced serum FGA levels (29).

The present results demonstrated that AGT levels were higher in sera in patients with TB compare to sera from patients in the TIT and ESLC groups; however, AGT levels in BALFs from patients with TB were lower compared with the BALFs from patients in the TIT group. AGT levels in BALFs were lower in patients with ESLC than in those with TB and TIT. AGT is thought to be involved in tumor angiogenesis and metastasis, as lung tumor growth and metastasis were attenuated in AGT (+/-) mice (30). The mechanisms of AGT-modulated airway stenosis require further elucidation.

The present results indicated that CLU was upregulated in the sera of patients with TB and it has been previously reported to be differentially expressed in plasma from patients with active TB complicated with diabetes (31). Furthermore, CLU levels were decreased in BALF from patients with pulmonary fibrosis and facilitated epithelial cell regeneration during lung repair (32,33). CLU is a critical glycoprotein with key roles in homeostasis, inhibition of cell death and promotion of pro-survival signaling pathways (34). It is involved in several physiological and pathological states. Genetic variation of CLU is associated with Alzheimer's disease (AD) (35). Notably, CLU serves as a chaperon of amyloid β and has attracted considerable attention in the field of AD research (36). CLU was indicated to be elevated in several tumor types. CLU confers chemoresistance to numerous cancer types, including primary breast cancer (37), pancreatic cancer (38), lung cancer (39), osteosarcoma (40) and prostate cancer (41). Accordingly, custirsen, a second-generation antisense oligonucleotide, was designed to inhibit the expression of CLU.

In the present study, serum APOAI was indicated to be downregulated in patients with TB. This result is in concordance with those of previous studies. APOAI is the major protein component of high-density lipoprotein that has a key role in lipid metabolism and serves as a biomarker for cardiovascular diseases (42). Serum APOAI levels were negatively correlated with the onset of cerebral infarction in patients with carotid artery stenosis (43). Of note, a recent study demonstrated that APOAI, -AⅡ and -AⅣ were decreased in patients with active TB (44). Furthermore, rifampicin treatment led to an increase in the level of APOAI in patients with TB; however, there was no statistical significance (45). Serum APOAI protein, quantified by isobaric tags for relative and absolute quantitation labeling coupled with two-dimensional liquid chromatography tandem MS, was markedly upregulated in patients cured of TB compared with that in untreated TB patients (46). APOAI is expressed in the lung. Of note, APOAI-/- mice presented with increased active lung inflammation and airway hyperresponsiveness (47). APOAI mimetics reduced the responses in the mouse model of asthma (48). Further studies are required to explore the putative implications of APOAI in the pathology and treatment of the disease.

The epithelial-mesenchymal transition is recognized as a key process in the initiation of cancer and it is accompanied by downregulation of KRT8/KRT18, which was indicated to be barely expressed in two intestinal cell lines, murine CT26 and rat IEC-6 cells (49). In IEC-6 cells, the KRT18 promoter was determined to be hypermethylated and in CT26 cells, a 9-amino acid in-frame insertion in the gene was identified. Matrigel assays indicated that restoration of KRT18 reduced CT26 cell invasion (49). The results demonstrated that the restoration of KRT18 reduced E-cadherin expression in IEC-6 cells; however, reduced expression was not observed in CT26 cells. Furthermore, KRT18 expression was modulated by early growth response 1 (EGR1) in non-small-cell lung carcinoma. Robust EGR1 expression and regulation of KRT18 led to reduced cell mobility and migration, and activated tumor cell apoptosis (50). Additionally, KRT18 was indicated to be associated with cancer cell metastasis. siRNA-mediated knockdown of KRT18 increased the migration and invasion of HepG2 and Eca109 cells (51). However, the roles and mechanisms of KRT18 in TB and tracheobronchial TB-induced TBS remain to be elucidated.

In conclusion, in the present study, MALDI-TOF MS was used to analyze proteins in the serum and BALF from patients with TB, ESLC and TIT to identify potential biomarkers for TBS secondary to tracheobronchial TB. Several candidate proteins, including MYADM, APOAI and KRT18, should be further examined for their roles in the pathophysiology of TBS. These proteins are involved in cell migration, permeability, adhesion, invasion, differentiation, neointima formation and tumor metastasis, which indicates a combination of pathological changes during TBS. These candidate proteins may be promising diagnostic tools and therapeutic targets and provide additional insight into the mechanisms underlying TBS after tracheobronchial TB.

Supplementary Material

General characteristics of the patients whose serum and BALF samples were examined by mass spectrometry.
General characteristics of the patients whose serum samples were examined by ELISAs.

Acknowledgements

Not applicable.

Funding

The current work was supported by the Capital Health Development Research Project (grant no. 2016-2-2048).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

BP, XQ and JZ conceptualized and designed the current study and acquired, analyzed and interpreted data. ZD analyzed and interpreted data. BP and JZ drafted the manuscript. JZ critically revised the manuscript for important intellectual content. YP and TW acquired data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All of the patients provided written informed consent to participate in the current study and for use of their medical data according to the provisions of the Declaration of Helsinki. The protocol of the current study was approved by the Institutional Ethics Committee of Beijing Tian Tan Hospital, Capital Medical University (Beijing, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hirshoren N and Eliashar R: Wound-healing modulation in upper airway stenosis-Myths and facts. Head Neck. 31:111–126. 2009.PubMed/NCBI View Article : Google Scholar

2 

Siow WT and Lee P: Tracheobronchial tuberculosis: A clinical review. J Thorac Dis. 9:E71–E77. 2017.PubMed/NCBI View Article : Google Scholar

3 

Nemati A, Safavi E, GhasemiEsfe M, Anaraki MZ, Firoozbakhsh S, Khalilzadeh O and Anvari M: Fistula formation between the right and left main bronchus caused by endobronchial tuberculosis. Am J Med Sci. 343:330–331. 2012.PubMed/NCBI View Article : Google Scholar

4 

Kashyap S, Mohapatra PR and Saini V: Endobronchial tuberculosis. Indian J Chest Dis Allied Sci. 45:247–256. 2003.PubMed/NCBI

5 

Um SW, Yoon YS, Lee SM, Yim JJ, Yoo CG, Chung HS, Kim YW, Han SK, Shim YS and Kim DK: Predictors of persistent airway stenosis in patients with endobronchial tuberculosis. Int J Tuberc Lung Dis. 12:57–62. 2008.PubMed/NCBI

6 

Shahzad T and Irfan M: Endobronchial tuberculosis-a review. J Thorac Dis. 8:3797–3802. 2016.PubMed/NCBI View Article : Google Scholar

7 

Mondoni M, Repossi A, Carlucci P, Centanni S and Sotgiu G: Bronchoscopic techniques in the management of patients with tuberculosis. Int J Infect Dis. 64:27–37. 2017.PubMed/NCBI View Article : Google Scholar

8 

Pathak V, Shepherd RW and Shojaee S: Tracheobronchial tuberculosis. J Thorac Dis. 8:3818–3825. 2016.PubMed/NCBI View Article : Google Scholar

9 

England K, Masini T and Fajardo E: Detecting tuberculosis: Rapid tools but slow progress. Public Health Action. 9:80–83. 2019.PubMed/NCBI View Article : Google Scholar

10 

Fan NJ, Gao CF and Wang XL: Tubulin beta chain, filamin A alpha isoform 1, and cytochrome b-c1 complex subunit 1 as serological diagnostic biomarkers of esophageal squamous cell carcinoma: A proteomics study. OMICS. 17:215–223. 2013.PubMed/NCBI View Article : Google Scholar

11 

Sobsey CA, Ibrahim S, Richard VR, Gaspar V, Mitsa G, Lacasse V, Zahedi RP, Batist G and Borchers CH: Targeted and untargeted proteomics approaches in biomarker development. Proteomics. 20(e1900029)2020.PubMed/NCBI View Article : Google Scholar

12 

Venkatesh A, Gil C, Fuentes M, LaBaer J and Srivastava S: A Perspective on Proteomics of Infectious Diseases. Proteomics Clin Appl. 12(e1700139)2018.PubMed/NCBI View Article : Google Scholar

13 

Dong W, Qiu C, Gong D, Jiang X, Liu W, Liu W, Zhang L and Zhang W: Proteomics and bioinformatics approaches for the identification of plasma biomarkers to detect Parkinson's disease. Exp Ther Med. 18:2833–2842. 2019.PubMed/NCBI View Article : Google Scholar

14 

Beavis RC and Chait BT: Rapid, sensitive analysis of protein mixtures by mass spectrometry. Proc Natl Acad Sci USA. 87:6873–6877. 1990.PubMed/NCBI View Article : Google Scholar

15 

Stoeckli M, Farmer TB and Caprioli RM: Automated mass spectrometry imaging with a matrix-assisted laser desorption ionization time-of-flight instrument. J Am Soc Mass Spectrom. 10:67–71. 1999.PubMed/NCBI View Article : Google Scholar

16 

Peng Y, Zhang Q, Xu C and Shi W: MALDI-TOF MS for the rapid identification and drug susceptibility testing of filamentous fungi. Exp Ther Med. 18:4865–4873. 2019.PubMed/NCBI View Article : Google Scholar

17 

Cho YT, Su H, Wu WJ, Wu DC, Hou MF, Kuo CH and Shiea J: Biomarker Characterization by MALDI-TOF/MS. Adv Clin Chem. 69:209–254. 2015.PubMed/NCBI View Article : Google Scholar

18 

Davies HA: The ProteinChip System from Ciphergen: A new technique for rapid, micro-scale protein biology. J Mol Med (Berl). 78(B29)2000.PubMed/NCBI

19 

Deng BG, Yao JH, Liu QY, Feng XJ, Liu D, Zhao L, Tu B and Yang F: Comparative serum proteomic analysis of serum diagnosis proteins of colorectal cancer based on magnetic bead separation and maldi-tof mass spectrometry. Asian Pac J Cancer Prev. 14:6069–6075. 2013.PubMed/NCBI View Article : Google Scholar

20 

Malys BJ, Piotrowski ML and Owens KG: Diagnosing and correcting mass accuracy and signal intensity error due to initial ion position variations in a MALDI TOFMS. J Am Soc Mass Spectrom. 29:422–434. 2018.PubMed/NCBI View Article : Google Scholar

21 

Cho Y, Park Y, Sim B, Kim J, Lee H, Cho SN, Kang YA and Lee SG: Identification of serum biomarkers for active pulmonary tuberculosis using a targeted metabolomics approach. Sci Rep. 10(3825)2020.PubMed/NCBI View Article : Google Scholar

22 

Isa F, Collins S, Lee MH, Decome D, Dorvil N, Joseph P, Smith L, Salerno S, Wells MT, Fischer S, et al: Mass spectrometric identification of urinary biomarkers of pulmonary tuberculosis. EBioMedicine. 31:157–165. 2018.PubMed/NCBI View Article : Google Scholar

23 

Cui W, Yu L, He H, Chu Y, Gao J, Wan B, Tang L and Zhao S: Cloning of human myeloid-associated differentiation marker (MYADM) gene whose expression was up-regulated in NB4 cells induced by all-trans retinoic acid. Mol Biol Rep. 28:123–138. 2001.PubMed/NCBI View Article : Google Scholar

24 

Aranda JF, Reglero-Real N, Marcos-Ramiro B, Ruiz-Sáenz A, Fernández-Martín L, Bernabé-Rubio M, Kremer L, Ridley AJ, Correas I, Alonso MA, et al: MYADM controls endothelial barrier function through ERM-dependent regulation of ICAM-1 expression. Mol Biol Cell. 24:483–494. 2013.PubMed/NCBI View Article : Google Scholar

25 

Aranda JF, Reglero-Real N, Kremer L, Marcos-Ramiro B, Ruiz-Sáenz A, Calvo M, Enrich C, Correas I, Millán J and Alonso MA: MYADM regulates Rac1 targeting to ordered membranes required for cell spreading and migration. Mol Biol Cell. 22:1252–1262. 2011.PubMed/NCBI View Article : Google Scholar

26 

Sun L, Bai Y, Zhao R, Sun T, Cao R, Wang F, He G, Zhang W, Chen Y, Ye P, et al: Oncological miR-182-3p, a novel smooth muscle cell phenotype modulator, evidences from model rats and patients. Arterioscler Thromb Vasc Biol. 36:1386–1397. 2016.PubMed/NCBI View Article : Google Scholar

27 

Panettieri RA Jr: Airway smooth muscle: An immunomodulatory cell. J Allergy Clin Immunol. 110 (Suppl):S269–S274. 2002.PubMed/NCBI View Article : Google Scholar

28 

Ozier A, Allard B, Bara I, Girodet PO, Trian T, Marthan R and Berger P: The pivotal role of airway smooth muscle in asthma pathophysiology. J Allergy (Cairo). 2011(742710)2011.PubMed/NCBI View Article : Google Scholar

29 

Kim HJ, Park JS, Heo JS, Moon KY and Park CS: Plasma apolipoprotein H levels are different between aspirin induced respiratory diseases and aspirin tolerant asthma. Pulm Pharmacol Ther. 27:184–189. 2014.PubMed/NCBI View Article : Google Scholar

30 

Choi JH, Nguyen MP, Lee D, Oh GT and Lee YM: Hypoxia-induced endothelial progenitor cell function is blunted in angiotensinogen knockout mice. Mol Cells. 37:487–496. 2014.PubMed/NCBI View Article : Google Scholar

31 

Zhang X, Ma A, Sun S and Sun Y: Proteomic analysis of plasma in adult active pulmonary tuberculosis patients with diabetes mellitus. Clin Lab. 61:1481–1490. 2015.PubMed/NCBI View Article : Google Scholar

32 

Peix L, Evans IC, Pearce DR, Simpson JK, Maher TM and McAnulty RJ: Diverse functions of clusterin promote and protect against the development of pulmonary fibrosis. Sci Rep. 8(1906)2018.PubMed/NCBI View Article : Google Scholar

33 

Habiel DM, Camelo A, Espindola M, Burwell T, Hanna R, Miranda E, Carruthers A, Bell M, Coelho AL, Liu H, et al: Divergent roles for clusterin in lung injury and repair. Sci Rep. 7(15444)2017.PubMed/NCBI View Article : Google Scholar

34 

Wilson MR and Zoubeidi A: Clusterin as a therapeutic target. Expert Opin Ther Targets. 21:201–213. 2017.PubMed/NCBI View Article : Google Scholar

35 

Nordestgaard LT, Tybjærg-Hansen A, Rasmussen KL, Nordestgaard BG and Frikke-Schmidt R: Genetic variation in clusterin and risk of dementia and ischemic vascular disease in the general population: Cohort studies and meta-analyses of 362,338 individuals. BMC Med. 16(39)2018.PubMed/NCBI View Article : Google Scholar

36 

Foster EM, Dangla-Valls A, Lovestone S, Ribe EM and Buckley NJ: Clusterin in Alzheimer's Disease: Mechanisms, genetics, and lessons from other pathologies. Front Neurosci. 13(164)2019.PubMed/NCBI View Article : Google Scholar

37 

Wang Y, Wang X, Zhao H, Liang B and Du Q: Clusterin confers resistance to TNF-alpha-induced apoptosis in breast cancer cells through NF-kappaB activation and Bcl-2 overexpression. J Chemother. 24:348–357. 2012.PubMed/NCBI View Article : Google Scholar

38 

Tang Y, Liu F, Zheng C, Sun S and Jiang Y: Knockdown of clusterin sensitizes pancreatic cancer cells to gemcitabine chemotherapy by ERK1/2 inactivation. J Exp Clin Cancer Res. 31(73)2012.PubMed/NCBI View Article : Google Scholar

39 

Panico F, Rizzi F, Fabbri LM, Bettuzzi S and Luppi F: Clusterin (CLU) and lung cancer. Adv Cancer Res. 105:63–76. 2009.PubMed/NCBI View Article : Google Scholar

40 

Ma J, Gao W and Gao J: sCLU as prognostic biomarker and therapeutic target in osteosarcoma. Bioengineered. 10:229–239. 2019.PubMed/NCBI View Article : Google Scholar

41 

Bertacchini J, Mediani L, Beretti F, Guida M, Ghalali A, Brugnoli F, Bertagnolo V, Petricoin E, Poti F, Arioli J, et al: Clusterin enhances AKT2-mediated motility of normal and cancer prostate cells through a PTEN and PHLPP1 circuit. J Cell Physiol. 234:11188–11199. 2019.PubMed/NCBI View Article : Google Scholar

42 

Charlton-Menys V and Durrington P: Apolipoproteins AI and B as therapeutic targets. J Intern Med. 259:462–472. 2006.PubMed/NCBI View Article : Google Scholar

43 

Dong Z, Guo Q, Sun L, Li F, Zhao A, Liu J, Qu P, Zhu Q, Xiao C, Niu F, et al: Serum lipoprotein and RBC rigidity index to predict cerebral infarction in patients with carotid artery stenosis. J Clin Lab Anal. 32(e22356)2018.PubMed/NCBI View Article : Google Scholar

44 

Mateos J, Estévez O, González-Fernández Á, Anibarro L, Pallarés Á, Reljic R, Mussá T, Gomes-Maueia C, Nguilichane A, Gallardo JM, et al: Serum proteomics of active tuberculosis patients and contacts reveals unique processes activated during Mycobacterium tuberculosis infection. Sci Rep. 10(3844)2020.PubMed/NCBI View Article : Google Scholar

45 

Albanna AS, Bachmann K, White D, Valiquette C and Menzies D: Serum lipids as biomarkers for therapeutic monitoring of latent tuberculosis infection. Eur Respir J. 42:547–550. 2013.PubMed/NCBI View Article : Google Scholar

46 

Wang C, Wei LL, Shi LY, Pan ZF, Yu XM, Li TY, Liu CM, Ping ZP, Jiang TT, Chen ZL, et al: Screening and identification of five serum proteins as novel potential biomarkers for cured pulmonary tuberculosis. Sci Rep. 5(15615)2015.PubMed/NCBI View Article : Google Scholar

47 

Wang W, Xu H, Shi Y, Nandedkar S, Zhang H, Gao H, Feroah T, Weihrauch D, Schulte ML, Jones DW, et al: Genetic deletion of apolipoprotein A-I increases airway hyperresponsiveness, inflammation, and collagen deposition in the lung. J Lipid Res. 51:2560–2570. 2010.PubMed/NCBI View Article : Google Scholar

48 

Yao X, Dai C, Fredriksson K, Dagur PK, McCoy JP, Qu X, Yu ZX, Keeran KJ, Zywicke GJ, Amar MJ, et al: 5A, an apolipoprotein A-I mimetic peptide, attenuates the induction of house dust mite-induced asthma. J Immunol. 186:576–583. 2011.PubMed/NCBI View Article : Google Scholar

49 

Kwan R, Looi KS and Omary MB: Absence of keratins 8 and 18 in rodent epithelial cell lines associates with keratin gene mutation and DNA methylation: Cell line selective effects on cell invasion. Exp Cell Res. 335:12–22. 2015.PubMed/NCBI View Article : Google Scholar

50 

Zhang H, Chen X, Wang J, Guang W, Han W, Zhang H, Tan X and Gu Y: EGR1 decreases the malignancy of human non-small cell lung carcinoma by regulating KRT18 expression. Sci Rep. 4(5416)2014.PubMed/NCBI View Article : Google Scholar

51 

Wang X, Lao Y, Xu N, Xi Z, Wu M, Wang H, Li X, Tan H, Sun M and Xu H: Oblongifolin C inhibits metastasis by up-regulating keratin 18 and tubulins. SciRep. 5(10293)2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Peng B, Qiu X, Dong Z, Zhang J, Pei Y and Wang T: Proteomic profiling of biomarkers by MALDI‑TOF mass spectrometry for the diagnosis of tracheobronchial stenosis after tracheobronchial tuberculosis. Exp Ther Med 21: 63, 2021
APA
Peng, B., Qiu, X., Dong, Z., Zhang, J., Pei, Y., & Wang, T. (2021). Proteomic profiling of biomarkers by MALDI‑TOF mass spectrometry for the diagnosis of tracheobronchial stenosis after tracheobronchial tuberculosis. Experimental and Therapeutic Medicine, 21, 63. https://doi.org/10.3892/etm.2020.9495
MLA
Peng, B., Qiu, X., Dong, Z., Zhang, J., Pei, Y., Wang, T."Proteomic profiling of biomarkers by MALDI‑TOF mass spectrometry for the diagnosis of tracheobronchial stenosis after tracheobronchial tuberculosis". Experimental and Therapeutic Medicine 21.1 (2021): 63.
Chicago
Peng, B., Qiu, X., Dong, Z., Zhang, J., Pei, Y., Wang, T."Proteomic profiling of biomarkers by MALDI‑TOF mass spectrometry for the diagnosis of tracheobronchial stenosis after tracheobronchial tuberculosis". Experimental and Therapeutic Medicine 21, no. 1 (2021): 63. https://doi.org/10.3892/etm.2020.9495