Upregulation of β2-microglobulin expression in progressive human oral squamous cell carcinoma

  • Authors:
    • Qian Jiang
    • Sdek Patima
    • Dong-Xia Ye
    • Hong-Ya Pan
    • Pin Zhang
    • Zhi-Yuan Zhang
  • View Affiliations

  • Published online on: December 30, 2011     https://doi.org/10.3892/or.2011.1613
  • Pages: 1058-1064
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate β2-microglobulin (β2-M) expression in normal oral mucosa and progressive oral squamous cell carcinoma (OSCC) and to assess the clinical significance of β2-microglobulin expression. The study included 10 cases of normal oral mucosa epithelium specimens, 55 cases of primary OSCC specimens, and 25 cases of OSCC metastasis specimens. Immunohistochemistry was used to determine β2-M expression, and its correlation with clinicopathological factors in progressive OSCC was evaluated. Immunohistochemistry showed that strong β2-M expression was significantly asscociated with tumor size (T3, T4 vs. T1, T2; P=0.001), positive node status (N positive vs. N negative; P=0.000) and advanced clinical stage (Ⅲ, Ⅳ vs. Ⅰ, Ⅱ, P=0.000) in primary OSCC lesions. Compared to primary OSCC lesions, the frequency of β2-M expression was significantly increased in metastatic OSCC lesions (P=0.02). In addition, in vitro results from Western blotting showed increased β2-M expression in the two OSCC lines studied. Therefore, we speculate that the up-regulation of β2-M expression may contribute to the oncogenesis of human oral mucosa, tumor invasion and metastasis.

Introduction

Oral squamous cell carcinoma (OSCC) is the sixth most common cancer in the world (1). Postoperative quality of life for patients with OSCC has improved in recent years (2). However, the 5-year survival rate has not improved significantly. Furthermore, 30–40% of patients without evidence of nodal disease at resection eventually die from metastatic spread (3). The identification of biomarkers for evaluating the progression of OSCC is therefore urgent.

It has been suggested that β2-microglobulin (β2-M) expression in tissues may be involved in OSCC progression and metastasis (4). β2-M is a non-glycosylated protein with a molecular mass of 11,800 Da and is synthesized by all nucleated cells (5). It is present on the surface of all nucleated cells except for red blood cells (6). β2-M forms the β chain of the major histocompatibility complex (MHC) class I molecule [also known as human leukocyte antigens (HLAs) in humans] and has a 7-stranded β-pleated structure, which is believed to function in antigen presentation to cytotoxic (CD8+) T lymphocytes (7). Upon recognition of foreign peptide antigens on cell surfaces, T cells actively bind and lyse antigen-presenting cancer cells. In β2-M-deficient mice, antibody (Ab) responses are defective, and natural killer (NK) cells with increased sensitivity attack cells lacking the MHC class I molecule (8,9). In addition to the roles in immunity, the level of β2-M is associated with proliferation, apoptosis and metastasis in several cancer types (10,11), and is a predictor of survival in patients with certain types of cancer (12). β2-M was found to promote the growth of human renal cell carcinoma through the activation of the protein kinase A, cyclic AMP-responsive element-binding protein, and vascular endothelial growth factor axis (11). Overexpression of β2-M in human prostate cancer cell lines leads to inhibition of tumor growth in vivo and using the β2-M Ab to interrupt β2-M signaling in human prostate cancer cell lines inhibits cancer cell growth and induces cell apoptosis (13).

The aim of this study was to investigate β2-M expression in normal oral mucosa and progressive OSCC and to assess the clinical significance of β2-M expression. The results of our study may contribute to a better understanding of the clinical significance of alterations in β2-M expression and may lead to further insights into the mechanisms to control progression and metastatic spread of tumor cells in OSCC patients.

Materials and methods

Cell cultures

Normal human oral keratinocytes (NHOKs) and human immortalized oral epithelial cells (HIOECs) (14,15) were cultured in defined keratinocyte medium-SFM (cat. no. 10744; Gibco, USA). CAL27 was purchased from ATCC (Manassas, VA). The OSC-4 cells were from Kochi Medical School, Japan. The CAL27 cells were cultured in DMEM (Invitrogen) with supplements (10% fetal bovine serum, 1% glutamine and 1% penicillin-streptomycin). The OSC-4 cells were cultured in RPMI-1640 (Invitrogen) with the same supplements.

Western blotting

Protein extracts were prepared from 1×106 cells using standard procedures. Cell lysates containing 20 μg protein were subjected to Western blot analysis. The primary Ab was monoclonal mouse anti-β2-M (sc-13565, 1:1000; Santa Cruz Biotechnology Inc.), and tubulin was detected as input control using monoclonal mouse anti-tubulin (T9026, 1:50,000; Sigma), Blots were developed with Immobilon Western Chemiluminescent HRP Substrate (Millipore, USA).

Tissue specimens

Tissue specimens were obtained from the files of the Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China. All tissue samples had been fixed in 10% buffered formalin and embedded in paraffin wax. For primary OSCC lesions obtained from 50 untreated patients, who underwent surgery between 2008 and 2009, clinicopathological data, including gender, age, tumor site, primary tumor stage (T), lymph node status (N) and tumor-node-metastasis (M) were obtained from the patient clinical records and pathological reports (Table I). Clinical stage was determined according to the 2002 American Joint Committee on Cancer (AJCC) staging system. Histopathological diagnosis and grading were confirmed using haematoxylin and eosin-stained sections according to the criteria mentioned in ‘Histological Typing of Tumors of the Upper Respiratory Tract and Ear’, WHO, 2nd edition. All data were re-examined independently by two of the authors. Metastatic OSCC lesions from 25 patients were obtained prior to biotherapy or chemotherapy between 2008 and 2009, and data including gender, age, and metastatic type was collected. (Table II). Analyses of the tissue samples are documented in Tables III-V. Histologically normal oral mucosa samples were obtained from 10 patients who underwent dental extractions. The human studies were approved by the institutional ethics committee.

Table I

Profiles of the patients with primary oral squamous cell carcinoma.

Table I

Profiles of the patients with primary oral squamous cell carcinoma.

NoGenderAge (years)Location stagePathological gradeTNMClinical stageSmoking Alcohol consumptionβ2-microglobulin
1M54PalateG1T3N1M0IIIYesHomogeneous
2F63GingivaeG2T3N1M0IIINoHomogeneous
3M48TongueG2T2N1M0IIIYesHomogeneous
4F54TongueG2T2N1M0IIINoHomogeneous
5F62Floor of the mouthG1T4N1M0IVYesHomogeneous
6M35TongueG2T4N1M0IVNoHomogeneous
7F71TongueG3T3N1M0IIINoHomogeneous
8M55TongueG2T4N1M0IVNoHomogeneous
9F64GingivaeG1T3N0M0IIIYesHeterogeneous
10M65TongueG2T2N0M0IINoNegative
11M64TongueG2T3N0M0IIIYesHeterogeneous
12F54TongueG2T2N0M0IINoNegative
13F53TongueG1T1N0M0INoNegative
14M57TongueG2T2N1M0IIIYesHeterogeneous
15M40BuccalG2T3N1M0IIINoHomogeneous
16M51TongueG2T4N2M0IVNoHomogeneous
17M44TongueG2T4N1M0IVNoHomogeneous
18F67GingivaeG1T3N1M0IIIYesHomogeneous
19F73Floor of the mouthG2T4N1M0IVYesHomogeneous
20M63TongueG2T2N1M0IIINoHomogeneous
21M58TongueG2T2N1M0IIIYesHomogeneous
22F65GingivaeG1T3N1M0IIIYesHomogeneous
23M58BuccalG2T4N1M0IVNoHomogeneous
24F53PalateG2T2N1M0IIINoHeterogeneous
25M54Floor of the mouthG3T4N0M0IVNoHeterogeneous
26M59TongueG1T1N1M0IIIYesHeterogeneous
27M60TongueG2T2N0M0IINoNegative
28F50TongueG1T4N0M0IVNoHomogeneous
29M51BuccalG1T2N0M0IINoHeterogeneous
30M42PalateG3T2N0M0IIYesHeterogeneous
31M65TongueG2T2N0M0IINoNegative
32M76TongueG2T3N0M0IIIYesHeterogeneous
33F34TongueG2T2N0M0IIYesNegative
34F44TongueG1T1N0M0IYesNegative
35F68BuccalG2T2N1M0IIIYesHeterogeneous
36M60BuccalG2T3N1M0IIINoHomogeneous
37M68TongueG2T4N2M0IVNoHomogeneous
38M61GingivaeG2T4N1M0IVNoHomogeneous
39F66GingivaeG2T3N0M0IIIYesNegative
40F65BuccalG2T4N1M0IVYesHomogeneous
41M65TongueG2T2N1M0IIINoHomogeneous
42M58TongueG2T2N0M0IIYesHomogeneous
43F72GingivaeG2T3N1M0IIIYesHomogeneous
44M74BuccalG3T4N1M0IVYesHomogeneous
45M60PalateG2T2N0M0IINoHeterogeneous
46F73TongueG3T4N0M0IVNoHeterogeneous
47M49TongueG2T1N1M0IIIYesHeterogeneous
48M54TongueG2T2N0M0IIYesNegative
49F67BuccalG3T4N0M0IVNoHeterogeneous
50F52TongueG1T2N0M0IINoHeterogeneous

Table II

Profiles of the patients with metastatic oral squamous cell carcinoma.

Table II

Profiles of the patients with metastatic oral squamous cell carcinoma.

NoGenderAge (years)LocationType β2-microglobulin
1M58TongueLymph nodeHomogeneous
2M64BuccalLymph nodeHeterogeneous
3F65TongueLymph nodeHomogeneous
4F67TongueLymph nodeHomogeneous
5F67TongueLymph nodeHomogeneous
6M56TongueLymph nodeHomogeneous
7M54TongueLymph nodeHomogeneous
8F60TongueLymph nodeHomogeneous
9M61TongueLymph nodeHomogeneous
10F80BuccalLymph nodeHomogeneous
11F81BuccalLymph nodeHomogeneous
12F86TongueLymph nodeHomogeneous
13M88TongueLymph nodeHomogeneous
14F49BuccalLymph nodeHeterogeneous
15F49BuccalLymph nodeHomogeneous
16M43TongueLymph nodeHomogeneous
17M67TongueLymph nodeHomogeneous
18M78TongueLymph nodeHomogeneous
19M86BuccalLymph nodeHomogeneous
20M38TongueLymph nodeHomogeneous
21M67BuccalLymph nodeHomogeneous
22M88TongueLymph nodeHomogeneous
23F55TongueLymph nodeHomogeneous
24M49TongueLymph nodeHomogeneous
25M67TongueLymph nodeHomogeneous

Table III

β2-microglobulin antigen expression in normal oral mucosa epithelial and oral squamous cell carcinoma specimens.

Table III

β2-microglobulin antigen expression in normal oral mucosa epithelial and oral squamous cell carcinoma specimens.

Staining patternNormal oral mucosa epithelial specimens n (%)Oral squamous cell carcinoma specimens n (%)
Homogeneous0 (0)46 (61.3)
Heterogeneous10 (100)20 (26.7)
Negative0 (0)9 (12.0)
Total10 (100)75 (100.0)

[i] P=0.031.

Table V

Profile of β2-microglobulin antigen expression in primary oral squamous cell carcinoma and metastases.

Table V

Profile of β2-microglobulin antigen expression in primary oral squamous cell carcinoma and metastases.

Primary OSCCMetastases


Staining patternn (%)n (%)
Homogeneous26 (52)20 (80)
Heterogeneous15 (30)5 (20)
Negative9 (18)0 (0)
Total50 (100)25 (100)

[i] P=0.027

Immunohistochemical staining

Formalin-fixed, paraffin-embedded tissue sections were dewaxed with xylene and rehydrated by passage through decreasing concentrations of ethanol (100–80%). Endogenous peroxidase activity was blocked by a 20-min incubation at room temperature with 3% H2O. The sections were heated using a water bath at 100°C with 0.01 M citrate buffer solution (pH, 6.0) for 20 min, and incubated with an optimal amount of affinity-purified monoclonal mouse anti-human β2-M (sc-13565, 1:50; Santa Cruz Biotechnology) overnight at 4°C. Sections were stained with liquid DAB substrate-chromogen, and counterstained with hematoxylin. Negative controls were carried out by omitting the primary Ab. The percentage of stained tumor cells in each lesion was enumerated independently by two investigators who had no knowledge of the patient characteristics. Variations in the percentage of stained cells as counted were within a 10% range. We scored the staining results according the report of Kageshita et al (16). Briefly, OSCC lesions consisting of >75% immunostained OSCC cells within the entire lesion were scored as homogeneously positive, those having 25–75% immunostained OSCC cells were heterogeneously positive, and those with <25% immunostained OSCC cells were negative.

Statistical analysis

Several clinicopathological factors were evaluated in the primary OSCC lesions, including gender, age (≤61 years vs. >61 years), T stage (T1, T2 vs. T3, T4), N status (negative vs. positive) and clinical stage (stage I, II vs. stage III, IV). Pearson Chi-square test, Continuity Correction test and Fisher's exact test were used to evaluate the correlation between the clinicopathological variables and the β2-M staining score using SPSS software v13.0 (SPSS Inc., USA). Differences in the β2-M staining score between primary OSCC samples and metastatic OSCC samples were also analyzed using the Chi-square test. A P-value <0.05 was considered to denote significant difference.

Results

Expression of β2-M in primary cultured NHOKs and HIOECs and the OSCC cell lines

We compared the expression levels of β2-M in NHOKs and HIOECs and in the two OSCC cancer cell lines (OSC and CAL27) by Western blotting. NHOKs were isolated and cultured as described (14). HIOECs were established by overexpression of HPV16 E6 and E7 protein (14). Western blot analysis revealed that β2-M protein expression was increased in the OSC and CAL27 cells compared to the NHOKs and HIOECs (Fig. 1).

Expression of β2-M in normal oral mucosa epithelial and OSCC tissue specimens

We performed immunohistochemical staining using normal oral mucosa and OSCC tissue specimens. Ten human normal oral mucosa samples and 75 human OSCC lesions (50 primary OSCC and 25 metastatic OSCC samples) were included. In the human normal oral mucosa, a faint but consistent staining was observed, mainly in the plasma membrane in oral mucosa epithelial cells. Stromal cells such as fibroblasts and fibrocytes were not stained by the anti-β2-M Ab (Fig. 2A). Most of the OSCC (88%) tissue sections showed distinct homogeneous (Fig. 2B) or heterogeneous staining (Fig. 2C), mainly in the cytoplasm and cytoplasmic membrane of tumor epithelial cells. However, in a few primary OSCC tissues, no staining or staining with weak intensity for β2-M was noted in the cytoplasm and cytoplasmic membrane of tumor epithelial cells (Fig. 2D). Compared with normal oral mucosa specimens, the frequency of β2-M expression was significantly increased in OSCC (P=0.031) (Table III).

Association of β2-M expression with various clinicopathological features in primary OSCC tissues

Of the 50 primary OSCC samples, 26 (52%) exhibited a homogeneous distribution of β2-M staining, and 15 (30%) exhibited a heterogeneous distribution within the OSCC cells, while 9 (18%) were negative for β2-M staining (Table IV). Of the 23 patients classified as T1, T2 in 50 primary OSCC cases, 9 (39.1%) showed heterogenous staining and 8 (34.8%) showed negative staining, while only 6 (26.1%) exhibited homogeneous staining. In contrast, of the 27 patients classified as T3, T4, 20 (74.1%) presented with homogeneous staining, whereas only 6 (22.2%) showed heterogenous staining and 1 (3.7%) showed negative staining. Compared with primary OSCC of T1, T2 stage, the intensity of β2-M expression was significantly increased in the primary OSCC specimens of T3, T4. Up-regulation of β2-M expression was also associated with lymph node invasion of OSCC. β2-M expression was significantly increased in N-positive patients compared to N-negative patients (82.8 vs. 9.5%, P=0.000). Regarding clinical stage, in highly malignant stages (III, IV) 67.6% of samples showed homogeneous staining whereas in low malignant stages (I, II), only 1% of samples showed homogeneous staining. These data suggest that the staining scores for β2-M were significantly associated with large tumor size (T3, T4 vs. T1, T2, P=0.001), positive nodal status (N-positive vs. N-negative, P=0.000), and advanced clinic stage (III, IV vs. I, II, P=0.000) (Table IV). In contrast, there were no correlations between β2-M expression and gender, age, smoking and alcohol consumption, and pathologic grade.

Table IV

Association of β2-microglobulin antigen expression with clinicopathological characteristics in primary OSCC lesions.

Table IV

Association of β2-microglobulin antigen expression with clinicopathological characteristics in primary OSCC lesions.

β2-microglobulin staining pattern

NHomogeneous n (%)Heterogeneous n (%)Negative n (%)P-value
Gender
 Female1910 (52.6)4 (21.1)5 (26.3)0.368
 Male3116 (51.6)11 (35.5)4 (12.9)
Age (years)
 ≤612914 (48.3)9 (31.0)6 (20.7)0.748
 >612112 (57.1)6 (28.6)3 (14.3)
Smoking and alcohol consumption
 No2816 (57.1)7 (25.0)5 (17.9)0.652
 Yes2210 (45.5)8 (36.4)4 (18.2)
Tumor size
 ≤4 cm (T1+T2)236 (26.1)9 (39.1)8 (34.8)0.001
 >4 cm (T3+T4)2720 (74.1)6 (22.2)1 (3.7)
Lymph nodes
 Negative (0)212 (9.5)10 (47.6)9 (42.9)0.000
 Positive (1–2)2924 (82.8)5 (17.2)0 (0.0)
Clinical stage
 Early (I, II)131 (7.7)4 (30.8)8 (61.5)0.000
 Advanced (III, IV)3725 (67.6)11 (29.7)1 (2.7)
Pathological grade
 G1115 (45.5)4 (36.4)2 (18.2)
 G23319 (57.6)7 (21.2)7 (21.2)0.237
 G332 (33.3)4 (66.7)0 (0.0)

[i] The percentage of immunoreactive cells in the entire lesion was evaluated microscopically and scored according to the method described by Kageshita et al (16): homogeneous, >75% cells stained in the entire lesion; heterogeneous, 25–75% cells stained; negative, <25% cells stained.

Up-regulation of β2-M expression in OSCC tissues correlates with tumor metastasis

Intensity of β2-M staining in OSCC metastatic lesions was significantly stronger than that in the primary OSCC lesions (Fig. 3). Ninenty-two percent of metastatic lesions exhibited a homogeneous distribution of β2-M expression while 52% of primary OSCC lesions exhibited homogeneous staining for β2-M (P=0.027) (Table V).

Discussion

In the present study, β2-M expression in OSCC lesions was evaluated and correlated with tumor progression and metastasis in OSCC patients. The results showed that β2-M expression was up-regulated in OSCC cell lines and OSCC lesions, and was associated with OSCC progression, invasion and metastasis. Consistent with our results, it was previously found that the suppression of β2-M expression using small interfering RNA (siRNA) was sufficient to decrease cell migration and invasion in vitro (4). The results of our and other research studies (4), indicate that OSCC lesions should be included in the spectrum of tumors with increased levels of β2-M expression.

Recent studies have used a wide range of experimental approaches to assess the mitogenic role of β2-M in malignancies. (1719). These studies have provided strong evidence to show that β2-M acts similarly to a prototypical oncogenic factor capable of stimulating growth and progression of various types of cancers, including breast cancer (17), prostate cancer (18), lung cancer (19), gastrointestinal (20), nasopharyngeal cancers (21), multiple myeloma (22), and particularly, lymphocytic malignancies (23), such as non-Hodgkin's lymphoma and multiple myeloma. Similar studies have also reported that β2-M is a growth-promoting factor contributing to the growth and progression of renal cell carcinoma (24,25).

However, previous studies have shown that β2-M/MHC class I can serve as important signal-transducing molecules in regulating tumor immunity and progression (26). Increased susceptibility to tumor formation was noted in β2-M gene-knockout mice, which suggests potential regulation of cancer growth by β2-M (26). Loss of β2-M expression is clinically important as it has been described in various patient-derived tumor cells, such as in melanomas (27) and cervical carcinoma (28). The possible explanation is that β2-M is expressed at a constant level on the cell surface. When expression of the β2-M molecule is below a normal level, defects in the β2-M/MHC class I signaling pathway may result in tumor immune escape. When expression of the β2-M molecule is higher than normal, β2-M promotes tumorigenesis and metastasis as an oncogene.

In some cancers, immunohistochemical evidence suggests that absence of functional (β2-M-associated) HLA class I molecules may be due to a mutational loss of β2-M (29); and in other cancers, a decreased level or the loss of β2-M in tumor cells was found due to the loss of the β2-M locus, or promoter methylation (30,31). Under these conditions, loss of β2-M prevents the synthesis of wild-type β2-M protein, which may lead to alterations in MHC class I surface expression. In our study, was not observed loss of β2-M in the OSCC lesions. In contrast, levels of β2-M expression were up-regulated in progressive OSCC lesions. The balance of β2-M expression at the cell surface was disturbal, which contributed to human cancer growth. Therefore, β2-M has a wider function than just a housekeeping gene or the role on stabilization and presentation of MHC class I molecular in cells.

Recently, marked antitumor activity has been observed by down-regulation of β2-M levels using either sequence-specific siRNA or antibodies in cases of both solid tumors and blood malignancies (13). In prostate cancer and renal cancer, growth inhibition of tumors was observed when patients were treated with anti-β2-M polyclonal or monoclonal antibodies (32), and in myeloma and other hematological malignancies, tumor cell apopotosis was observed using monoclonal β2-M Ab and sequence-specific siRNA to β2-M (33). Thus, β2-M, as an oncogenic factor in various cancer types, appears to be an excellent new target for interrupting human cancer growth. In our study, the association of β2-M expression with progression and metastasis of OSCC lesions was statistically significant. Whether we can inhibit OSCC progression, invasion or migration by using a similar anti-β2-M polyclonal or monoclonal antibody needs further study.

Acknowledgements

This study was supported by grants from the Doctoral Innovation Foundations of Shanghai Jiao Tong University School (no. BXJ 0922) and National Nature Science Foundation of China (no. 30630065 and 30973344).

Abbreviations:

OSCC

oral squamous cell carcinoma

MHC

major histocompatibility complex

HLAs

human leukocyte antigens

NK

natural killer

HIOECs

human immortalized oral epithelial cells

References

1 

Imai T, Toyota M, Suzuki H, et al: Epigenetic inactivation of RASSF2 in oral squamous cell carcinoma. Cancer Sci. 99:958–966. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Scully C and Bagan JV: Recent advances in Oral Oncology 2007: imaging, treatment and treatment outcomes. Oral Oncol. 44:211–215. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Cai ZG, Shi XJ, Gao Y, Wei MJ, Wang CY and Yu GY: Beta-catenin expression pattern in primary oral squamous cell carcinoma. Chin Med J (Engl). 121:1866–1870. 2008.PubMed/NCBI

4 

Chen CH, Su CY, Chien CY, et al: Overexpression of beta2-microglobulin is associated with poor survival in patients with oral cavity squamous cell carcinoma and contributes to oral cancer cell migration and invasion. Br J Cancer. 99:1453–1461. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Shi C, Zhu Y, Su Y, Chung LW and Cheng T: Beta2-microglobulin: emerging as a promising cancer therapeutic target. Drug Discov Today. 14:25–30. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Winchester JF, Salsberg JA and Levin NW: Beta-2 microglobulin in ESRD: an in-depth review. Adv Ren Replace Ther. 10:279–309. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Margalit A, Sheikhet HM, Carmi Y, et al: Induction of antitumor immunity by CTL epitopes genetically linked to membrane-anchored beta2-microglobulin. J Immunol. 176:217–224. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Christianson GJ, Brooks W, Vekasi S, et al: Beta 2-microglobulin-deficient mice are protected from hypergammaglobulinemia and have defective antibody responses because of increased IgG catabolism. J Immunol. 159:4781–4792. 1997.PubMed/NCBI

9 

Hoglund P, Glas R, Menard C, et al: Beta2-microglobulin-deficient NK cells show increased sensitivity to MHC class I-mediated inhibition, but self tolerance does not depend upon target cell expression of H-2Kb and Db heavy chains. Eur J Immunol. 28:370–378. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Huang WC, Wu D, Xie Z, et al: Beta2-microglobulin is a signaling and growth-promoting factor for human prostate cancer bone metastasis. Cancer Res. 66:9108–9116. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Nomura T, Huang WC, Zhau HE, et al: Beta2-microglobulin promotes the growth of human renal cell carcinoma through the activation of the protein kinase A, cyclic AMP-responsive element-binding protein, and vascular endothelial growth factor axis. Clin Cancer Res. 12:7294–7305. 2006. View Article : Google Scholar

12 

Tsimberidou AM, Kantarjian HM, Wen S, et al: The prognostic significance of serum beta2 microglobulin levels in acute myeloid leukemia and prognostic scores predicting survival: analysis of 1,180 patients. Clin Cancer Res. 14:721–730. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Huang WC, Havel JJ, Zhau HE, et al: Beta2-microglobulin signaling blockade inhibited androgen receptor axis and caused apoptosis in human prostate cancer cells. Clin Cancer Res. 14:5341–5347. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Sdek P, Zhang ZY, Cao J, Pan HY, Chen WT and Zheng JW: Alteration of cell-cycle regulatory proteins in human oral epithelial cells immortalized by HPV16 E6 and E7. Int J Oral Maxillofac Surg. 35:653–657. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Zhang L, Yang X, Zhong LP, et al: Decreased expression of Annexin A1 correlates with pathologic differentiation grade in oral squamous cell carcinoma. J Oral Pathol Med. 38:362–370. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Kageshita T, Hirai S, Ono T, Hicklin DJ and Ferrone S: Down-regulation of HLA class I antigen-processing molecules in malignant melanoma: association with disease progression. Am J Pathol. 154:745–754. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Klein B, Levin I, Kfir B, Mishaeli M, Shapira J and Klein T: The significance of soluble interleukin-2, soluble interleukin-2 receptors, soluble ICAM-1 and beta 2-microglobulin in breast cancer patients. Tumour Biol. 16:290–296. 1995. View Article : Google Scholar : PubMed/NCBI

18 

Freeman MR: Beta2 microglobulin: a surprising therapeutic target for prostate cancer and renal cell carcinoma. J Urol. 178:10–11. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Nissen MH, Bjerrum OJ, Plesner T, Wilken M and Rorth M: Modification of beta-2-microglobulin in sera from patients with small cell lung cancer: evidence for involvement of a serine protease. Clin Exp Immunol. 67:425–432. 1987.PubMed/NCBI

20 

Kaplan B, Martin BM, Livoff A, Yeremenko D, Livneh A and Cohen HIL: Gastrointestinal beta2microglobulin amyloidosis in hemodialysis patients: biochemical analysis of amyloid proteins in small formalin-fixed paraffin-embedded tissue specimens. Mod Pathol. 18:1610–1617. 2005.

21 

Lee JK, Tsai SC, Hsieh JF, Ho YJ, Sun SS and Kao CH: Beta-2-microglobulin (beta 2M) as a tumor marker in nasopharyngeal carcinoma. Anticancer Res. 20:4765–4768. 2000.PubMed/NCBI

22 

Kim JE, Yoo C, Lee DH, Kim SW, Lee JS and Suh C: Serum albumin level is a significant prognostic factor reflecting disease severity in symptomatic multiple myeloma. Ann Hematol. 89:391–397. 2009. View Article : Google Scholar

23 

Delgado J, Pratt G, Phillips N, et al: Beta2-microglobulin is a better predictor of treatment-free survival in patients with chronic lymphocytic leukaemia if adjusted according to glomerular filtration rate. Br J Haematol. 145:801–805. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Federico M, Guglielmi C, Luminari S, et al: Prognostic relevance of serum beta2 microglobulin in patients with follicular lymphoma treated with anthracycline-containing regimens. A GISL study. Haematologica. 92:1482–1488. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Albitar M, Vose JM, Johnson MM, et al: Clinical relevance of soluble HLA-I and beta2-microglobulin levels in non-Hodgkin's lymphoma and Hodgkin's disease. Leuk Res. 31:139–145. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Sousa AO, Mazzaccaro RJ, Russell RG, et al: Relative contributions of distinct MHC class I-dependent cell populations in protection to tuberculosis infection in mice. Proc Natl Acad Sci USA. 97:4204–4208. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Hicklin DJ, Wang Z, Arienti F, Rivoltini L, Parmiani G and Ferrone S: Beta2-Microglobulin mutations, HLA class I antigen loss, and tumor progression in melanoma. J Clin Invest. 101:2720–2729. 1998. View Article : Google Scholar : PubMed/NCBI

28 

Mehta AM, Jordanova ES, Kenter GG, Ferrone S and Fleuren GJ: Association of antigen processing machinery and HLA class I defects with clinicopathological outcome in cervical carcinoma. Cancer Immunol Immunother. 57:197–206. 2008. View Article : Google Scholar : PubMed/NCBI

29 

del Campo AB, Aptsiauri N, Mendez R, et al: Efficient recovery of HLA class I expression in human tumor cells after beta2-microglobulin gene transfer using adenoviral vector: implications for cancer immunotherapy. Scand J Immunol. 70:125–135. 2009.PubMed/NCBI

30 

Koene GJ, Arts-Hilkes YH, van der Ven KJ, et al: High level of chromosome 15 aneuploidy in head and neck squamous cell carcinoma lesions identified by FISH analysis: limited value of beta2-microglobulin LOH analysis. Tissue Antigens. 64:452–461. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Feenstra M, Veltkamp M, van Kuik J, et al: HLA class I expression and chromosomal deletions at 6p and 15q in head and neck squamous cell carcinomas. Tissue Antigens. 54:235–245. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Yang J, Qian J, Wezeman M, et al: Targeting beta2-microglobulin for induction of tumor apoptosis in human hematological malignancies. Cancer Cell. 10:295–307. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Yang J, Zhang X, Wang J, et al: Anti-beta2-microglobulin monoclonal antibodies induce apoptosis in myeloma cells by recruiting MHC class I to and excluding growth and survival cytokine receptors from lipid rafts. Blood. 110:3028–3035. 2007. View Article : Google Scholar

Related Articles

Journal Cover

April 2012
Volume 27 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang Q, Patima S, Ye D, Pan H, Zhang P and Zhang Z: Upregulation of β2-microglobulin expression in progressive human oral squamous cell carcinoma. Oncol Rep 27: 1058-1064, 2012
APA
Jiang, Q., Patima, S., Ye, D., Pan, H., Zhang, P., & Zhang, Z. (2012). Upregulation of β2-microglobulin expression in progressive human oral squamous cell carcinoma. Oncology Reports, 27, 1058-1064. https://doi.org/10.3892/or.2011.1613
MLA
Jiang, Q., Patima, S., Ye, D., Pan, H., Zhang, P., Zhang, Z."Upregulation of β2-microglobulin expression in progressive human oral squamous cell carcinoma". Oncology Reports 27.4 (2012): 1058-1064.
Chicago
Jiang, Q., Patima, S., Ye, D., Pan, H., Zhang, P., Zhang, Z."Upregulation of β2-microglobulin expression in progressive human oral squamous cell carcinoma". Oncology Reports 27, no. 4 (2012): 1058-1064. https://doi.org/10.3892/or.2011.1613