Open Access

Proteomic analysis of serum deprivation in tongue squamous cell carcinoma

  • Authors:
    • Junfeng Zhang
    • Wei Dong
    • Yufen Meng
    • Miao Jiang
    • Zhen Zhan
  • View Affiliations

  • Published online on: October 17, 2017     https://doi.org/10.3892/mmr.2017.7807
  • Pages: 9323-9330
  • Copyright: © Zhang 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

The occurrence of tongue squamous cell carcinoma (TSCC) is closely correlated with serum components; however, the detailed mechanism remains to be fully elucidated. Proteomic analysis contributed to the discovery of potential biomarkers and provided an insight into TSCC at a molecular level. The present study investigated the effect of serum deprivation on the Tca‑8113 TSCC cell line through protein profiling using two‑dimensional gel electrophoresis and mass spectrometry, with the aim of improving TSCC diagnosis. The results showed that the Tca‑8113 cells maintained proliferative capacity and resisted apoptosis following serum deprivation. A total of 43 proteins were upregulated and 45 were downregulated following serum deprivation for 24 h, compared with untreated controls (0 h). The upregulated caspase-7, heat shock protein 27 and Annexin A1, and the downregulated peroxiredoxin‑6 and heat shock protein 70, were selected for verification using reverse transcription‑polymerase chain reaction analysis following serum deprivation for 16 h. The results indicated that reactive oxygen species may be important in serum deprivation‑induced oxidative stress.

Introduction

Tongue squamous cell carcinoma (TSCC) has a particularly high mortality rate among malignancies, predominantly due to the difficulty of early detection, and its incidence is increasing rapidly according to the American Cancer Society (1,2). Studies have shown that TSCC has a poorer prognosis and higher recurrence rate, compared with several other types of cancer due to metastasis being common (3). Clinical results suggest that a lack of available biomarkers result in the majority of patients with TSCC not being diagnosed until an advanced stage. Therefore, understanding the molecular basis of tongue carcinogenesis may provide potential molecular markers for more rapid diagnosis of TSCC. Several biological molecules have been reported in the saliva and serum, and histological analysis has indicated that they are useful for screening and early detection of several types of cancer (46).

Serum is important for supporting cell viability and pH maintenance (7), whereas saliva is important in oral hygiene, food digestion and the repair of mucosal damage, as demonstrated by animal behavioural experiments and in vivo analysis of patients (8). Studies have suggested that saliva and serum are functionally similar and reflect the physiological state of the body (911), and that saliva is a non-invasive sample for potential clinical diagnosis. In the present study, serum deprivation experiments were performed on the Tca-8113 TSCC cell line to screen for potential biomarkers suitable for TSCC diagnosis. This was based on previous observations that serum deprivation synchronises proliferating cells at the G0 phase, induces apoptosis and stimulates the fibrotic response (12).

The results of the present study showed that the Tca-8113 cells resisted serum deprivation-induced apoptosis. Two-dimensional gel electrophoresis (2-DE) and mass spectrometry-based protein identification were used for the proteomic analysis of serum-deprived Tca-8113 cells. The results provided insight for potential diagnostic biomarkers or drug targets for the treatment of TSCC.

Materials and methods

Cell culture and MTT assay

The cells were routinely cultured, and MTT (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) assays were performed as previously described (13). Briefly, the TSCC Tca-811 cell line, provided by the Ninth People's Hospital of Shanghai (Shanghai, China) was maintained by routine culturing in RPMI-1640 medium with 10% fetal bovine serum (FBS) (both from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in a 37°C incubator with 5% CO2. The cells were digested with 0.25% trypsin and seeded into 96-well plates (cat no. 3524; Corning Incorporated, Corning, NY, USA) at a density of 1×104 cells/well and cultured for 18–24 h to 90% confluence. Serum deprivation was performed following washing twice with PBS. The cells were cultured for 12, 24, 36, 48 and 72 h following serum deprivation in the media at 37°C, with 10% serum as a control. Following cell culture, the culture medium were discarded, 200 µl of 0.5 mg/ml MTT in PBS was added to wells, and the cells were cultured for 4 h at 37°C. Following removal of the supernatants, 200 µl of DMSO was added and cells were shaken lightly for 15 min. The optical density value was measured using a microplate spectrophotometer (POWERWAVE 340; BioTek Instruments, Inc., Winooski, VT, USA) at 490 nm. All experiments were repeated four times and the mean ± standard deviation of results were calculated.

Proteomic analysis

The proteomic experiments were performed as described previously (14). The steps were as follows: i) At least 1.0×108 Tca-8113 cells were harvested and lysed for sample preparation, and the bicinchoninic acid method was used for measurement of protein concentration; ii) each 80 µg sample was subjected to 2-DE on an Ettan IPGphor II apparatus (GE Healthcare Life Sciences, Chalfont, UK) in triplicate. Silver-stained gels were scanned at 300 dpi resolution, and differential analysis was performed to assign spots. Student's t-test was used for filtering, and spots with a fold-change >1.2 were considered differentially expressed; iii) selected spots were subjected to MALDI-TOF mass spectrometry and tandem TOF/TOF mass spectrometry. The identified peptides were used for Basic Local Alignment Search Tool searches of the International Protein Index rat sequence database (http://www.matrixscience.com). The protein number, protein name, sequence coverage, score and Mr/pI were obtained and included in the analysis.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

The serum-deprivation treated Tca-8113 cells were prepared for one-step RT-qPCR SYBR Green assay as previous described (15). Briefly, RNA was extracted using the TRIzol LS reagent and the cDNA Synthesis kit (both from Gibco; Thermo Fisher Scientific, Inc.) was used for cDNA synthesis. A total of 1 µl reverse transcriptase product was used to amplify the six genes (Annexin A1, peroxiredoxin 6, heat shock protein 27, heat shock protein 70, caspase-7 and GAPDH). Primers (Table I) were designed based on the sequences of the five differentially expressed proteins identified in the proteomic analysis and GAPDH gene was used as reference. The primers were synthesized by Sangon Biotech (Shanghai) Co., Ltd. (Shanghai, China). The SYBR Green Real-Time PCR Master Mix (TOYOBO; Toyobo Life Science, Inc., Osaka, Japan) was used and the PCR cycling parameters were as follows: 94°C for 30 sec, followed by 35 cycles of 60°C for 30 sec and 72°C for 30 sec. A MyCycler thermal cycler (Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used for the PCR analysis. Each gene was quantified by 2−ΔΔCq method (16) and the relative expression level was calculated by the ratio of each targeted gene to GAPDH gene.

Table I.

Reverse transcription-polymerase chain reaction primer sequences for the six genes.

Table I.

Reverse transcription-polymerase chain reaction primer sequences for the six genes.

GenePrimer sequenceLength (bp)
Annexin A1F: 5′-AAGACTTGGCTGATTCAGATGC-3′124
R: 5′-AACACTCTGCGAAGTTGTGGATA-3′
Peroxiredoxin 6F: 5′-CGCATCCGTTTCCACGACTT-3′271
R: 5′-TGGCAAGCTCCCGATTCCTAT-3′
Heat shock protein 27F: 5′-AGTGGTCGCAGTGGTTAGGC-3′186
R: 5′-GGTTGACATCCAGGGACACG-3′
Heat shock protein 70F: 5′-CTGCTGCGACAGTCCACTACC-3′177
R: 5′-TCGGCTCCGCTCTGAGATT-3′
Caspase-7F: 5′-GAAGAGGCTCCTGGTTTGTG-3′106
R: 5′-CTGGCAACTCTGTCATTCACC-3′
GAPDHF: 5′-GGGAAACTGTGGCGTGAT-3′299
R: 5′-GAGTGGGTGTCGCTGTTGA-3′

[i] GAPDH, glyceraldehyde phosphate dehydrogenase; F, forward; R, reverse.

Statistical analysis

Data are reported as the mean ± standard deviation, and Student's t-tests were performed using the SPSS 16.0 software package (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Serum deprivation inhibits cell proliferation

Serum deprivation inhibited the proliferation of cells, as exhibited in Fig. 1. When the Tca-8113 cells were subjected to serum deprivation for 12, 24, 36, 48 or 72 h, the proliferative activity was measured using an MTT assay. No significant differences were observed over time in the serum-deprived group (P>0.05), however, proliferation continued to increase in the cells cultured in 10% FBS medium (P<0.05).

Identification of differentially expressed proteins using proteomic analysis

As proliferation was inhibited in the Tca-8113 cells subjected to serum deprivation for 24 h, the time-points of 0 h (control) and 24 h were selected for the comparison of triplicate samples using 2-DE and mass spectrometry. Representative 2-DE images are shown in Fig. 2. A total of 1,237 protein spots were detected on the 2-DE gel, with 43 significantly upregulated and 45 significantly downregulated at 24 h, compared with levels at 0 h (P<0.05; average spot intensity difference >1.2-fold). Of these 88 potential differentially expressed proteins, 35 were identified using MALDI-TOF/MS, whereas the remaining 53 did not yield suitably complete polypeptide fragments or were too low in abundance for reliable identification. The results of the spot ID, average spot intensity, protein number, protein name, sequence coverage, score and Mr/pI are exhibited in Table II. The present study focussed on five differentially expressed proteins: Caspase-7 (spot 70), heat shock protein 27 (HSP 27; spot 390), Annexin A1 (spot 560), peroxiredoxin-6 (spot 405) and HSP 70 (spot 1194; Fig. 3). Notably, all are involved in mitochondrial oxidative stress, consistent with a previous conclusion that serum deprivation can induce apoptosis (7).

Table II.

List of differentially expressed proteins during serum-deprivation treatment.

Table II.

List of differentially expressed proteins during serum-deprivation treatment.

Percentage of total volume of all spotsa

Spot IDControl (0 h)Serum-deprivation (24 h)Protein numberProtein nameSequence coverage (%)ScoreMr/pI
Upregulated
  590.000±0.0000.086±0.005gi|2250700KNP-I α protein6213823,806/8.61
  4890.000±0.0000.002±0.001gi|4505773Prohibitin5819729,843/5.57
  4970.000±0.0000.153±0.005gi|28931β-subunit (AA 1–312)42  9534,026/4.90
  5660.001±0.0010.110±0.015gi|194376310Actin, β, partial3417438,950/5.19
  3430.007±0.0040.214±0.030gi|194375974 Peroxiredoxin-25719320,209/8.90
  700.016±0.0100.156±0.016gi|20150093Chain B, crystal structure of caspase-7 complexed with xiap6314312,244/5.73
  2200.010±0.0030.090±0.004gi|55959240Nicotinamide nucleotide adenylyltransferase 16613218,413/8.99
  1030.078±0.0230.417±0.035gi|1362965T cell receptor Mb11 β chain4810915,697/6.56
  3760.076±0.0240.399±0.017gi|5822016Chain C, erythropoietin complexed with extracellular domains of erythropoietin receptor6616518,655/9.28
  5490.065±0.0190.250±0.024gi|21667979Oxysterol binding protein-related protein 3 isoform 2d4012335,786/5.11
  3900.456±0.0371.119±0.024gi|662841Heat shock protein 273911122,427/7.83
  5600.439±0.0351.011±0.071gi|4502101Annexin A15221538,918/6.57
  3920.856±0.0671.734±0.087gi|1871210T-complex protein 1, β subunit (TCP-1-β)6315422,924/5.88
Downregulated
  3581.744±0.0431.021±0.067gi|2392338Glyoxalase-I4212120,861/5.12
  7750.852±0.0300.456±0.067gi|301030821c-Myc promoter-binding protein 14210036,874/6.17
  11840.416±0.0070.174±0.012gi|194388088Heat shock 70 kDa protein 1A/1B3420564,170/5.39
  11620.120±0.0040.050±0.011gi|194384236Heat shock 70 kDa protein 1A/1B19  8760,091/5.39
  11940.774±0.0280.311±0.029gi|62897129Heat shock 70 kDa protein 8 isoform 1 variant17  7871,083/5.28
  8851.450±0.0870.476±0.039gi|92911770 XTP3TPA-transactivated protein 15014623,896/6.42
  7140.822±0.1560.233±0.076gi|296211572Tubulin α-1B chain isoform 23011046,797/4.96
  2040.327±0.0350.057±0.018gi|56605994CDGSH iron-sulfur domain-containing protein 28217715,497/9.66
  10770.055±0.0050.005±0.003gi|32189394ATP synthase subunit β, mitochondrial precursor4620856,525/5.26
  1980.161±0.0220.000±0.000gi|57162210CDC14 cell division cycle 14 homolog B (S. cerevisiae)4712631,655/8.33
  2060.107±0.0120.000±0.000gi|51479152ATP synthase subunit d, mitochondrial isoform b48  8915,820/6.60
  2450.052±0.0100.000±0.000gi|119595056Glutathione S-transferase pi, isoform CRA_c24  4119,577/4.84
  3140.054±0.0090.000±0.000gi|306517833MHC class I antigen4810921,098/6.64
  4050.356±0.0060.000±0.000gi|4758638 Peroxiredoxin-64113325,133/6.00
  4190.267±0.0340.000±0.000gi|3329384Translation initiation factor 4e5513127,481/10.00
  7720.076±0.0040.000±0.000gi|7705592Mediator of RNA polymerase II transcription subunit 316120115,966/8.72
  8450.131±0.0030.000±0.000gi|89574029Mitochondrial ATP synthase, H+ transporting F1 complex β subunit3913448,083/4.95
  10000.235±0.0470.000±0.000gi|48146259CCT23115957,766/6.01
  10060.173±0.0350.000±0.000gi|220702506Chain A, TapasinERP57 HETERODIMER3417454,541/5.61
  10800.052±0.0030.000±0.000gi|169404695Chain A, pyruvate kinase M2, phosphotyrosine binding protein2915357,091/8.00
  11890.063±0.0100.000±0.000gi|14718539HIC-315  8265,103/5.89
  12220.186±0.0050.000±0.000gi|386758GRP78 precursor, partial3216972,185/5.03
  12300.035±0.0010.000±0.000gi|15293595Seven transmembrane helix receptor43  8524,812/8.87

a Data are presented as the mean ± standard deviation. Mr/pI, molecular mass/isoelectric point.

RT-PCR verification

The present study performed RT-PCR analysis to investigate the expression of caspase-7, HSP 27, Annexin A1, peroxiredoxin-6 and HSP 70 in the Tca-8113 cells subjected to serum deprivation at two time-points (0 and 16 h). The stably expressed GAPDH mRNA was selected as an internal reference gene as the expression of cytoskeletal proteins was significantly disrupted. For example, the expression of β-actin was increased at the protein (Table II) and mRNA levels (data not shown), whereas the expression of tubulin was decreased (Table II). As shown in Fig. 4, the expression patterns of the five proteins were similar to the results obtained using 2-DE.

Discussion

Serum deprivation is widely used for cell cycle arrest and apoptosis, and various molecular mechanisms may be involved in the modulation of serum deprivation-induced apoptosis (17). However, in the present study, the effects of serum deprivation on cell proliferation and apoptosis were not observed in the Tca-8113 cells, even following a long duration (72 h) of treatment (Fig. 1). The proteomic analysis showed that a 24 h period of serum deprivation induced significant expression changes of numerous mitochondrial proteins, including prohibitin, peroxiredoxin, glyoxalase, caspase and ATP synthase (Table II). This is consistent with a previous study, in which mitochondrial function was readily disrupted by alterations of the cultivation environment through serum deprivation, which affects mitochondrial viability in several fibroblast lines (18). However, the detailed molecular mechanism linking culture conditions and mitochondrial activity remains to be fully elucidated. The results of the present study provided novel insight into the mechanism by which Tca-8113 cells subjected to serum deprivation avoid apoptosis.

Although the mechanism by which serum deprivation stimulates reactive oxygen species (ROS) production remains to be elucidated, studies have shown that ROS resulting from serum deprivation can induce programmed cell death (7,19). Oxidative stress has a wide range of effects on cellular proliferation, differentiation, migration and other physiological processes (20). Several antioxidant proteins are involved in alleviating oxidative stress by removing ROS. The present study found that peroxiredoxin-2 (Prx2) was upregulated, whereas the associated Prx6 was downregulated following serum deprivation. Prx enzymes have six isoforms, and they are located at sites of energy production and cellular motion, where ROS can be produced in abundance, particularly in mitochondria and at the plasma membrane. The peroxiredoxins are known for their function in removing endogenous peroxides. Therefore, the upregulation of peroxiredoxins is linked to high levels of oxidative stress (21). Prx2 is the major Prx enzyme in the cytoplasm, where it reduces peroxynitrite and hydroperoxides via a rapid, diffusion-controlled reaction (22). Prx2 is also important in the mechanism of H2O2-induced redox signalling (23). The overexpression of Prx2 has been associated with the progression of squamous cell carcinoma, and it may be an underlying biomarker for radiotherapy in colorectal cancer (24) lung cancer (25) and bladder cancer (26). Similarly, the expression of Prx6 has been associated with high rates of metastasis of breast cancer cell lines, and the overexpression of Prx6 occurs in malignant mesothelioma, breast cancer, oesophageal carcinoma and oligodendroglioma (27). A previous study revealed a marked upregulation of Prx2 following demethylation in gefitinib-resistant A549 cells (28). The present results suggested that Tca-8113 cells may resist the serum deprivation-induced apoptosis by upregulating Prx2, which may serve as a potent target for assisting in anti-angiogenic drug treatment in human TSCC.

Annexin A1 (ANXA1) is a well-characterised apoptosis-associated membrane protein, which is regulated by the epidermal growth factor receptor, insulin receptor, TRPM7 channel kinase 1, protein kinase C and protein kinase A. ANXA1 has also been linked with carcinogenesis and metastasis in various types of tumour (29). As an essential protein in tumourigenesis and apoptosis, the overexpression of ANXA1 is linked to carcinogenesis and high pathological differentiation grade (30,31). There is accumulating evidence suggesting that ANXA1 may be regulated by pro-inflammatory proteins, including lipopolysaccharide and interleukin-6, and it is reported to act as a brake to control the inflammatory response (32). Kang et al found that ANXA1 was positively upregulated during serum deprivation-induced autophagic degradation in HCT116 cells, indicating a potential role in autophagy by downregulating the inflammatory response (33). The overexpression of ANXA1 can also inhibit the aberrant dysregulation of cytoskeletal proteins and proliferation of pulmonary microvascular endothelial cells (34), which is consistent with the observed morphology, static proliferation and upregulation of actin in the serum deprived Tca-8113 cells in the present study. The results of the present study showed that ANXA1 was upregulated at the mRNA and protein levels following serum deprivation, suggesting that autophagy, and not apoptosis, was a factor in Tca-8113 cells; autophagy protected the cells against serum deprivation by promoting cell survival.

Autophagy and apoptosis are the two major cell death pathways, and are tightly regulated at the genetic level to ensure that tissue and organ development proceeds correctly, in addition to other pathophysiological processes (35). A hallmark of autophagy is lysosomal degradation, which performs an adaptive function to protect organisms against infections, cancer and ageing (36). Apoptosis is a programmed process for killing and removing certain cells during morphological development, which also assists in maintaining tissue homeostasis in multicellular organisms. The aberrant regulation of apoptosis has been consistently linked with human proliferative diseases, including malignant tumours (37). Apoptosis involves a large number of molecules, including cysteine proteases in the form of caspases, which are synthesised as pro-forms and become activated by cleavage at aspartate residues. Initiator caspases, including caspase-1, 2, 4, 5, 8, 9, 10, 11 and 12, integrate molecular signals and activate downstream effector caspases, including caspase-3, 6, 7 and 14. As caspases can cleave and activate each other, triggering of the caspase cascade initiates a rapidly amplified signal, which ensures accurate apoptotic cell death. Caspases are also able to cleave several other substrates, including nuclear lamins and cytoskeletal proteins, thereby inducing typical morphological characteristics associated with apoptosis. Caspase-7 is most closely linked with caspase-3, and these enzymes are activated by mitochondria-induced apoptosis.

Effector caspases are in charge of initiating DNA fragmentation, cell shrinkage and membrane blebbing (38). Brentnall et al (39) showed that caspase-9 and caspase-3 are involved in remodeling of mitochondria and ROS production. Caspase-7 has no significant effect on sensitivity to intrinsic cell death, but is involved in ROS production and cell detachment. In the present study, serum deprivation upregulated the level of caspase-7 in Tca-8113 cells, which underwent shrinkage and membrane blebbing, suggesting that serum deprivation stimulated TSCC differentiation and inhibited cancer growth.

HSPs are also closely linked with stress responses, including the response to serum deprivation. HSP70 is a major stress-inducible protein, which can fulfil multiple roles depending on its location. The upregulation of intracellular HSP70 is involved in the stress associated with recovery from clinical treatment, including radiotherapy or chemotherapy, whereas extracellular HSP70 stimulates the innate immune system to initiate antitumour immunity (40). In the present study, the serum deprivation-induced downregulation of HSP70 may assist TSCCs to escape and evade attacks from natural killer (NK) cells, however, the downregulation of major histocompatibility complex-I can compensate for this and ensure activation of the NK cells. HSP27 is primarily involved in proteasome-mediated protein degradation and the regulation of apoptosis. A previous study revealed that HSP27 mediates apoptotic programming in pancreatic cancer cells (41), and clinical immunohistochemical evidence demonstrates that the overexpression of HSP27 causes high-grade differentiation in TSCCs (42). Therefore, the serum deprivation-induced upregulation of HSP27 may be correlated with the morphology and differentiation of TSCCs, consistent with the expression of caspase-7.

In conclusion, the present investigative preclinical study found that Tca-8113 TSCC cells resisted apoptosis induced by serum deprivation. The changes in proteins were primarily involved in the oxidative stress process in the mitochondrion, which indicated that ROS may be important in serum deprivation-induced oxidative stress. In the future, further detailed investigations on the signalling pathway and clinical verification are required.

Acknowledgements

The authors would like to thank the Natural Science Foundation of China (grant nos. 81473593 and 81473458), the Jiangsu Qing Lan Project (grant no. QL-2014) for financial support, and the Priority Academic Program Development of Jiangsu Higher Education Institutions (Integration of Chinese and Western Medicine; grant no. PAPD-2014) for funding.

References

1 

Ching CT, Sun TP, Huang SH, Hsiao CS, Chang CH, Huang SY, Chen YJ, Cheng CS, Shieh HL and Chen CY: A preliminary study of the use of bioimpedance in the screening of squamous tongue cancer. Int J Nanomed. 5:213–220. 2010. View Article : Google Scholar

2 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Layland MK, Sessions DG and Lenox J: The influence of lymph node metastasis in the treatment of squamous cell carcinoma of the oral cavity, oropharynx, larynx, and hypopharynx: N0 versus N+. Laryngoscope. 115:629–639. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Korostoff A, Reder L, Masood R and Sinha UK: The role of salivary cytokine biomarkers in tongue cancer invasion and mortality. Oral Oncol. 4:282–287. 2011. View Article : Google Scholar

5 

Faratzis G, Tsiambas E, Rapidis AD, Machaira A, Xiromeritis K and Patsouris E: VEGF and ki 67 expression in squamous cell carcinoma of the tongue: An immunohistochemical and computerized image analysis study. Oral Oncol. 45:584–588. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Patel RS, Clark JR, Dirven R, Wyten R, Gao K and O'Brien CJ: Prognostic factors in the surgical treatment of patients with oral carcinoma. Anz J Surg. 79:19–22. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Lee SB, Kim JJ, Kim TW, Kim BS, Lee MS and Yoo YD: Serum deprivation-induced reactive oxygen species production is mediated by Romo1. Apoptosis. 15:204–218. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Grossman N, Binyamin LA and Bodner L: Effect of rat salivary glands extracts on the proliferation of cultured skin cells-a wound healing model. Cell Tissue Bank. 4:205–212. 2004. View Article : Google Scholar

9 

De Giuseppe R, Cossellu G, Vigna L, Dicorato F, De Vita C, Venturelli G, Bamonti F, Maiavacca R and Farronato G: Correlation between salivary and serum oxidized LDL levels: A pilot study on overweight/obese subjects. J Oral Pathol Med. 44:884–887. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Hayes LD, Sculthorpe N, Herbert P, Baker JS, Hullin DA, Kilduff LP and Grace FM: Poor levels of agreement between serum and saliva testosterone measurement following exercise training in aging men. Aging Male. 18:67–70. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Seethalakshmi C, Koteeswaran D and Chiranjeevi V: Correlation of serum and salivary biochemical parameters in end stage renal disease patients undergoing hemodialysis in pre and post-dialysis state. J Clin Diagn Res. 12:CC12–CC14. 2014.

12 

Boraldi F, Annovi G, Paolinelli-Devincenzi C, Tiozzo R and Quaglino D: The effect of serum withdrawal on the protein profile of quiescent human dermal fibroblasts in primary cell culture. Proteomics. 8:66–82. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Li WZ, Wang XY, Li ZG, Zhang JH and Ding YQ: Celecoxib enhances the inhibitory effect of cisplatin on Tca8113 cells in human tongue squamous cell carcinoma in vivo and in vitro. J Oral Pathol Med. 7:579–584. 2010.

14 

Wu J, Wang F, Gong Y, Li D, Sha J, Huang X and Han X: Proteomic analysis of changes induced by nonylphenol in Sprague-Dawley rat Sertoli cells. Chem Res Toxicol. 22:668–675. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Cheuk BL and Cheng SW: Annexin A1 expression in atherosclerotic carotid plaques and its relationship with plaque characteristics. Eur J Vasc Endovasc Surg. 41:364–371. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Yuan F, Cheng Q, Li G and Tong T: Nucleostemin knockdown sensitizes hepatocellular carcinoma cells to ultraviolet and serum starvation-induced apoptosis. PLoS One. 10:e01416782015. View Article : Google Scholar : PubMed/NCBI

18 

Takeda K, Akagi S, Takahashi S, Onishi A, Hanada H and Pinkert CA: Mitochondrial activity in response to serum starvation in bovine (Bos taurus) cell culture. Cloning Stem Cells. 4:223–229. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Liu J and Du L: PERK pathway is involved in oxygen-glucose-serum deprivation-induced NF-kB activation via ROS generation in spinal cord astrocytes. Biochem Biophys Res Commun. 467:197–203. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Gerrits EG, Alkhalaf A, Landman GW, van Hateren KJ, Groenier KH, Struck J, Schulte J, Gans RO, Bakker SJ, Kleefstra N and Bilo HJ: Serum peroxiredoxin 4: A marker of oxidative stress associated with mortality in type 2 diabetes (ZODIAC-28). PLoS One. 2:e897192014. View Article : Google Scholar

21 

Ding Y, Yamada S, Wang KY, Shimajiri S, Guo X, Tanimoto A, Murata Y, Kitajima S, Watanabe T, Izumi H, et al: Overexpression of peroxiredoxin 4 protects against high-dose streptozotocin-induced diabetes by suppressing oxidative stress and cytokines in transgenic mice. Antioxid Redox Signal. 13:1477–1490. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Manta B, Hugo M, Ortiz C, Ferrer-Sueta G, Trujillo M and Denicola A: The peroxidase and peroxynitrite reductase activity of human erythrocyte peroxiredoxin 2. Arch Biochem Biophys. 484:146–154. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Martinez A, Peluffo G, Petruk AA, Hugo M, Piñeyro D, Demicheli V, Moreno DM, Lima A, Batthyány C, Durán R, et al: Structural and molecular basis of the peroxynitrite-mediated nitration and inactivation of trypanosoma cruzi iron-superoxide dismutases (Fe-SODs) A and B: Disparate susceptibilities due to the repair of Tyr35 radical by Cys83 in Fe-SODB through intramolecular electron transfer. J Biol Chem. 289:12760–12778. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Cerda MB, Lloyd R, Batalla M, Giannoni F, Casal M and Policastro L: Silencing peroxiredoxin-2 sensitizes human colorectal cancer cells to ionizing radiation and oxaliplatin. Cancer Lett. 388:312–319. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Rostila A, Puustinen A, Toljamo T, Vuopala K, Lindström I, Nyman TA, Oksa P, Vehmas T and Anttila SL: Peroxiredoxins and tropomyosins as plasma biomarkers for lung cancer and asbestos exposure. Lung Cancer. 77:450–459. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Shiota M, Yokomizo A, Kashiwagi E, Takeuchi A, Fujimoto N, Uchiumi T and Naito S: Peroxiredoxin 2 in the nucleus and cytoplasm distinctly regulates androgen receptor activity in prostate cancer cells. Free Radic Biol Med. 51:78–87. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Zhang B, Wang Y and Su Y: Peroxiredoxins, a novel target in cancer radiotherapy. Cancer Lett. 286:154–160. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Kwon T, Jin KR, Lee JC, Park YH, Shin HJ, Cho S, Kang YK, Kim BY, Yoon DY and Yu DY: An important role for peroxiredoxin II in survival of A549 lung cancer cells resistant to gefitinib. Exp Mol Med. 47:e1652015. View Article : Google Scholar : PubMed/NCBI

29 

Sobral-Leite M, Wesseling J, Smit VT, Nevanlinna H, van Miltenburg MH, Sanders J, Hofland I, Blows FM, Coulson P and Patrycja G: Annexin A1 expression in a pooled breast cancer series: association with tumor subtypes and prognosis. BMC Med. 13:1562015. View Article : Google Scholar : PubMed/NCBI

30 

Lin CY, Jeng YM, Chou HY, Hsu HC, Yuan RH, Chiang CP and Kuo MY: Nuclear localization of annexin A1 is a prognostic factor in oral squamous cell carcinoma. J Surg Oncol. 97:544–550. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Faria PC, Sena AA, Nascimento R, Carvalho WJ, Loyola AM, Silva SJ, Durighetto AF, Oliveira AD, Oliani SM and Goulart LR: Expression of annexin A1 mRNA in peripheral blood from oral squamous cell carcinoma patients. Oral Oncol. 46:25–30. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Vago JP, Nogueira CR, Tavares LP, Soriani FM, Lopes F, Russo RC, Pinho V, Teixeira MM and Sousa LP: Annexin A1 modulates natural and glucocorticoid-induced resolution of inflammation by enhancing neutrophil apoptosis. J Leukoc Biol. 92:249–258. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Kang JH, Li M, Chen X and Yin XM: Proteomics analysis of starved cells revealed Annexin A1 as an important regulator of autophagic degradation. Biochem Biophys Res Commun. 407:581–586. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Yi B, Jing Z, Wang G, Qian G and Lu K: Annexin A1 protein regulates the expression of PMVEC cytoskeletal proteins in CBDL rat serum-induced pulmonary microvascular remodeling. J Transl Med. 11:982013. View Article : Google Scholar : PubMed/NCBI

35 

Thorburn A: Apoptosis and autophagy: Regulatory connections between two supposedly different processes. Apoptosis. 1:1–9. 2008. View Article : Google Scholar

36 

Levine B and Kroemer G: Autophagy in the pathogenesis of disease. Cell. 132:27–42. 2008. View Article : Google Scholar : PubMed/NCBI

37 

PLOS ONE Staff, . Correction: Association of genetic markers in the BCL-2 family of apoptosis-related genes with endometrial cancer risk in a Chinese population. PLoS One. 10:e01176322015. View Article : Google Scholar : PubMed/NCBI

38 

Coutinho-Camillo CM and Soares FA: CASP7 (caspase 7, apoptosis-related cysteine peptidase). Atlas Genetics Cytogenetics Oncology Haematology. 3:160–163. 2015.

39 

Brentnall M, Rodriguez-Menocal L, Guevara RL, Cepero E and Boise LH: Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 14:322013. View Article : Google Scholar : PubMed/NCBI

40 

Multhoff G, Pockley AG, Schmid TE and Schilling D: The role of heat shock protein 70 (Hsp70) in radiation-induced immunomodulation. Cancer Lett. 368:179–184. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Yang G, Ziesch A, Hocke S, Kampmann E, Ochs S, De Toni EN, Göke B and Gallmeier E: Overexpression of heat shock protein 27 (HSP27) increases gemcitabine sensitivity in pancreatic cancer cells through S-phase arrest and apoptosis. J Cell Mol Med. 2:340–350. 2015.

42 

Zhang NN, Sheng SH, Chen D, et al: Expression and significance of HSP27 in tongue squamous cell carcinoma. Beijing J Stomatol. 3:146–148. 2010.

Related Articles

Journal Cover

December-2017
Volume 16 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang J, Dong W, Meng Y, Jiang M and Zhan Z: Proteomic analysis of serum deprivation in tongue squamous cell carcinoma. Mol Med Rep 16: 9323-9330, 2017
APA
Zhang, J., Dong, W., Meng, Y., Jiang, M., & Zhan, Z. (2017). Proteomic analysis of serum deprivation in tongue squamous cell carcinoma. Molecular Medicine Reports, 16, 9323-9330. https://doi.org/10.3892/mmr.2017.7807
MLA
Zhang, J., Dong, W., Meng, Y., Jiang, M., Zhan, Z."Proteomic analysis of serum deprivation in tongue squamous cell carcinoma". Molecular Medicine Reports 16.6 (2017): 9323-9330.
Chicago
Zhang, J., Dong, W., Meng, Y., Jiang, M., Zhan, Z."Proteomic analysis of serum deprivation in tongue squamous cell carcinoma". Molecular Medicine Reports 16, no. 6 (2017): 9323-9330. https://doi.org/10.3892/mmr.2017.7807