• 沒有找到結果。

Aberrant glycosylation frequently occurs in cancers and plays a critical role in

cancer progression, angiogenesis and metastasis. Common feature of tumors is the

overexpression of truncated O-glycans, such as the disaccharide Thomsen–Friedenreich

antigen (T antigen), the monosaccharide GalNAc (also known as Tn) and their

sialylated forms (ST and STn). Specifically, STn expression modulates malignant

phenotypes in gastric and breast cancer cells, such as increased migration and

invasion.[6,7,34] Altered expression of glycans can be attributed to abnormal

expression of glycosyltransferases. In previous studies, GALNT2 altered the expression

of Tn antigen on EGFR in oral squamous cell carcinoma and hepatocellular carcinoma

and Tn antigen on MET in gastric cancer. [14,15] GALNT10 modified O-glycosylation

of EGFR and subsequent phosphorylation of Akt in HBV-associated HCC.[35]

Besides, GALNT2 could modify O-glycosylation and activation of IGF-1R, and then

affect the malignant phenotypes of neuroblastoma cells.[36] However, no studies

investigate about the role of GALNT2 in modifying O-glycosylation of EGFR in gastric

cancer. Here, we demonstrated that knockdown of GALNT2 decreased the Tn antigen

on EGFR in gastric cancer. Since alterations in glycosylation can significantly impact

overall glycoprotein charge and conformation and therefore readily alter its biological

activity. For example, aberrant glycosylation of receptor tyrosine kinases (RTKs) could

modulate their activities and signalings. [7] Receptor tyrosine kinase (RTK) -targeted

therapeutic agents are constantly being developed and have been shown to be effective in

various clinical trials. Trastuzumab, an inhibitor of ERBB2, has been approved for the

treatment of gastric cancer.[37] So investigating the roles of other RTKs in gastric cancer

will provide potential targets for the future treatments. In our previous study, we revealed

that GALNT2 knockdown increased EGFR phosphorylation. Constitutive activation of

the EGFR, which can be resulted from ligand depend or ligand independent pathway is

common in cancers [38] Here, we showed that knockdown of GALNT2 enhanced EGFR

phosphorylation in the presence of EGF. On the other hand, without EGF, GALNT2

didn’t affect EGFR phosphorylation. This may indicate EGFR activation modulated by

GALNT2 may dependent on the presence of ligand. Previous researches showed

glycosylation of EGFR might affect their bind affinity to EGF [39,40]. The mechanism

of how GALNT2 affects EGFR phosphorylation needs further study.

Recent studies suggested that GALNT2 regulated the malignant phenotypes by

modifying EGFR glycosylation and phosphorylation in oral squamous carcinoma and

hepatocellular carcinoma.[14,15] This study demonstrated the migration and invasion

abilities of AGS cells enhanced by GALNT2 would be suppressed by inhibiting EGFR

phosphorylation. Subsequently, inhibition of EGFR phosphorylation decreased

expression of phospho-Akt. Previous studies revealed that EGFR promoted gastric

cancer migration and invasion via Akt-activation.[27,28] In this study, cell viability,

migration and invasion abilities of AGS cells were significantly suppressed by inhibiting

Akt phosphorylation. These findings reveal that EGFR-Akt activation plays an important

role in gastric cancer progression.

In present study, cell viability during day1-5 was not affected by GALNT2 and

EGFR activation but Akt activation. In contrast to our previous study, knockdown of

GALNT2 enhanced cell viability.[33] This may be because this difference was

significant at day 6 in previous study, but we only assessed cell viability for 5 days.

Current studies about the effect of gefitinib on cell growth inhibition are controversial.

Gefitinib inhibited proliferation in prostate cancer and lung cancer.[41] However, it was

ineffective against most EGFR wild-type non-small cell lung cancer, but combined

EGFR siRNA or Akt inhibitior showed synergistic growth inhibition. [42,43]

However, there is no research about correlation of GALNT2 and phospho-EGFR in

clinical samples. This is the first study to investigate this relationship, but result showed

p-EGFR positively correlated with GALNT2, which was opposite to in vitro experiments.

The possible reasons may be as followed: first, the limitation of IHC analysis of

detection of two proteins simultaneously, so two sections were used to detect expression

of GALNT2 and phospho-EGFR respectively, which might result in scoring at different

cells. Although, we minimize this limitation by sectioning serially, but the final slide for

evaluated might not from the serial section. Immunofluorescence may serve as method to

solving this problem by double labeling of different proteins in the same slide.[44,45]

Second, downregulation of GALNT2 enhances the malignancy in gastric cancer may

attribute to multiple pathways, not all the patients undergo increasing EGFR

phosphorylation, so it needs further investigation of subgroup analysis, and it might

demonstrate clinical significance.

EGFR becomes activated upon phosphorylated, so increasing numbers of studies on

correlation of p-EGFR with clinical outcomes. Patients with p-EGFR positive exhibited

poorer overall survival.[30,31], but Nieto et al. showed p-EGFR didn’t serve as a

prognostic marker for overall survival.[29] The prognostic role of p-EGFR in gastric

cancer remains unknown. In present study revealed there were no difference in

clinico-pathalogical parameters, such as tumor size, cell differentiation, TNM stage and

progression-free survival between p-EGFR positive and negative. The plausible

explanations for controversial findings are as followed. First, scoring criteria and

threshold for positive and negative applied in each papers are not equal. Second, there

are many phosphorylation sites of EGFR, such as pY845. pY1068 and pY1173 etc., and

initiating distinct downstream signaling pathways. Recent studies detected not the same

site, and it might result in different outcomes. Third, gefitinib inhibit overall

phosphorylation of EGFR, not site specific, so other tyrosine sites may play a more

critical role in enhancing malignancy in gastric cancer. In addition, distinct molecular

drivers and tumor biology, and thus different treatment targets and predictive biomarkers

are implicated in each subtype of gastric cancer, such as intestinal versus diffuse type.

For example, EGFR, HER2 and MET overexpression are more prevalent in intestinal

type, but loss of E-cadherin and FGFR overexpression are more common in diffuse

type.[46,47] In this study, only intestinal type samples were collected for analyzing.

Thus, we will collect diffuse type samples to investigate if p-EGFR serves as a

prognostic marker in this subtype.

In conclusion, our findings of GALNT2 regulating malignancy by modifying

O-glycosylation and phosphorylation of EGFR brings new insights into new therapeutic

strategies. However, the prognostic role of pEGFR needs further investigation.

相關文件