Share this post on:

Xpression of CTGF in NPC. After Title Loaded From File examination by NimbleGen DNA methylation microarray, we did not find any methylation modification in CTGF promoter regionCTGF in NPCin 17 NPC samples and 3 NPs (Figure 6), which suggested that reduced expression of CTGF in NPC was not related to its promoter methylation.DiscussionCTGF plays dual roles as oncogene and tumor suppressor in different cancer types [6?4], which may be attributed to tissuespecific patterns of expression in different tissues and organs in tumourigenesis. However, its roles and molecular mechanisms linking the initiation and development of NPC are not well understood [12]. In this study, we first found that CTGF expression was decreased in NPCs compared to normal nasopharyngx (NP) tissues by Title Loaded From File microarray examination. This result strongly supported Lee et al’s microarray data (GSE2370). Further, we confirmed CTGF mRNA was weakly expressed in NPC cell lines compared to NP69 cell line or in NPC tissues compared to NPs by qPCR. These results were consistent with our microarray data, suggesting that downregulated CTGF is involved in promoting NPC pathogenesis. We used immunohistochemistry to further examine the expression level of CTGF protein in NPC tissues and noncancerous tissues. We observed that cytoplasmic CTGF expression was markedly decreased in cancer tissues compared to normal epithelium. These results were not only consistent with our previous investigation [12], but also hinted that decreased expression of CTGF was involved in the stages of NPC initiation. In previous studies of other tumor types, different expression patterns of CTGF correlated with both favorable and unfavorable tumor progression. Elevated expression of CTGF was positively associated with progression and poor prognosis in melanoma, papillary thyroid carcinoma, esophageal squamous cell carcinoma, gastric cancer, and cervical tumors [18?2]. Conversely, reduced CTGF expression was favorable for tumor progression and prognosis, in oral squamous cell carcinoma, ovarian cancer, and lung adenocarcinomas [23?5]. In this study, we found that attenuated CTGF expression was negatively associated with T, N classification, and clinical stages of NPC patients. The results suggested the downregulated expression of CTGF promoted NPC pathogenesis. To specifically determine the contributions of CTGF in the regulation of NPC phenotypes, we modulated its expression in 6?0B cell lines. We found that stably decreased expression of CTGF by shRNA conferred 6?0B cells with higher expression of proliferation marker protein PCNA, cell proliferation, colony formation, G1/S cell cycle transition, migration and invasion in vitro. Similar results were observed after transiently suppressing CTGF expression by siRNA transfection in NPC 6?0B and HONE1 cells. The biological functions of CTGF found in this study provided a mechanistic basis for the pathological and clinical observations. We examined key cell cycle regulators of the G1-S transition and observed that CCND1, pRb, and E2F1 were upregulated while p15 and p21 were downregulated after stable CTGF knockdown in 6?0B cells. Further, we found that CTGF suppression-induced expression of genes is associated with cell migration and invasion. MMP2, MMP9, and EMT-marker genes including Snail, Ncadherin, and Vimentin were highly upregulated while EMT-marked gene E-cadherin was weakly expressed in shRNA treated 6?0B cells. However, CTGF suppression did not lead to any change from epithelial to.Xpression of CTGF in NPC. After examination by NimbleGen DNA methylation microarray, we did not find any methylation modification in CTGF promoter regionCTGF in NPCin 17 NPC samples and 3 NPs (Figure 6), which suggested that reduced expression of CTGF in NPC was not related to its promoter methylation.DiscussionCTGF plays dual roles as oncogene and tumor suppressor in different cancer types [6?4], which may be attributed to tissuespecific patterns of expression in different tissues and organs in tumourigenesis. However, its roles and molecular mechanisms linking the initiation and development of NPC are not well understood [12]. In this study, we first found that CTGF expression was decreased in NPCs compared to normal nasopharyngx (NP) tissues by microarray examination. This result strongly supported Lee et al’s microarray data (GSE2370). Further, we confirmed CTGF mRNA was weakly expressed in NPC cell lines compared to NP69 cell line or in NPC tissues compared to NPs by qPCR. These results were consistent with our microarray data, suggesting that downregulated CTGF is involved in promoting NPC pathogenesis. We used immunohistochemistry to further examine the expression level of CTGF protein in NPC tissues and noncancerous tissues. We observed that cytoplasmic CTGF expression was markedly decreased in cancer tissues compared to normal epithelium. These results were not only consistent with our previous investigation [12], but also hinted that decreased expression of CTGF was involved in the stages of NPC initiation. In previous studies of other tumor types, different expression patterns of CTGF correlated with both favorable and unfavorable tumor progression. Elevated expression of CTGF was positively associated with progression and poor prognosis in melanoma, papillary thyroid carcinoma, esophageal squamous cell carcinoma, gastric cancer, and cervical tumors [18?2]. Conversely, reduced CTGF expression was favorable for tumor progression and prognosis, in oral squamous cell carcinoma, ovarian cancer, and lung adenocarcinomas [23?5]. In this study, we found that attenuated CTGF expression was negatively associated with T, N classification, and clinical stages of NPC patients. The results suggested the downregulated expression of CTGF promoted NPC pathogenesis. To specifically determine the contributions of CTGF in the regulation of NPC phenotypes, we modulated its expression in 6?0B cell lines. We found that stably decreased expression of CTGF by shRNA conferred 6?0B cells with higher expression of proliferation marker protein PCNA, cell proliferation, colony formation, G1/S cell cycle transition, migration and invasion in vitro. Similar results were observed after transiently suppressing CTGF expression by siRNA transfection in NPC 6?0B and HONE1 cells. The biological functions of CTGF found in this study provided a mechanistic basis for the pathological and clinical observations. We examined key cell cycle regulators of the G1-S transition and observed that CCND1, pRb, and E2F1 were upregulated while p15 and p21 were downregulated after stable CTGF knockdown in 6?0B cells. Further, we found that CTGF suppression-induced expression of genes is associated with cell migration and invasion. MMP2, MMP9, and EMT-marker genes including Snail, Ncadherin, and Vimentin were highly upregulated while EMT-marked gene E-cadherin was weakly expressed in shRNA treated 6?0B cells. However, CTGF suppression did not lead to any change from epithelial to.

Share this post on:

Author: bet-bromodomain.