• Title/Summary/Keyword: p-cadherin

Search Result 95, Processing Time 0.037 seconds

LINC00174 Facilitates Proliferation and Migration of Colorectal Cancer Cells via MiR-3127-5p/ E2F7 Axis

  • Ma, Yuhong;Li, Yuzhen;Tang, Yuanyuan;Tang, Ning;Wang, Dengke;Li, Xiaofei
    • Journal of Microbiology and Biotechnology
    • /
    • v.31 no.8
    • /
    • pp.1098-1108
    • /
    • 2021
  • The literature indicates that LINC00174 promotes the growth of colorectal cancer (CRC) cells, but its research needs to be enriched. We tried to explore the function and mechanism of LINC00174 in CRC cell proliferation and migration. Bioinformatics analysis predicted the binding relationship and expressions of lncRNA, miRNA and mRNA. Clinical study analyzes the relationship between LINC00174 and clinical data characteristics of CRC patients. The expressions of LINC00174, miR-3127-5p and E2F7 were verified by RT-qPCR, and the combination of the two was verified by dual luciferase analysis and RNA immunoprecipitation as needed. Western blot was used to detect the expression of EMT-related protein and E2F7 protein. Functional experiments were used to evaluate the function of the target gene on CRC cells. LINC00174 was up-regulated in CRC clinical samples and cells and was related to the clinical characteristics of CRC patients. High-expression of LINC00174, contrary to the effect of siLINC00174, promoted cell viability, proliferation, migration and invasion, up-regulated the expressions of N-Cadherin, Vimentin, E2F7, and inhibited the expression of E-Cadherin. MiR-3127-5p was one of the targeted miRNAs of LINC00174 and was down-regulated in CRC samples. In addition, miR-3127-5p mimic partially reversed the malignant phenotype of CRC cells induced by LINC00174. Besides, E2F7 was a target gene of miR-3127-5p, and LINC00174 repressed miR-3127-5p to regulate E2F7. Our research reveals that LINC00174 affected the biological characteristics of CRC cells through regulated miR-3127-5p/ E2F7 axis.

Lack of Association between the CDH1 -160C>A Polymorphism and Risk of Gastrointestinal Cancer - a Meta-Analysis

  • Sahami-Fard, Mohammad Hossein;Yazd, Ehsan Farashahi;Khazaei, Zahra;Neamatzadeh, Hossein
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.5
    • /
    • pp.2415-2421
    • /
    • 2016
  • E-cadherin (CDH1) genetic variations alter gene transcriptional activity of epithelial cells in vitro and may cause susceptibility to various cancers. Associations of CDH1 -160C>A polymorphism with various cancers have been widely reported. However, the results are controversial and inconsistent. To derive a more accurate estimation of the relationship, a meta-analysis was performed with regard to gastrointestinal (GI) cancer risk. Eligible studies were identified through a search of PubMed database until December 2015. Associations between the CDH1 -160C>A polymorphism and GI cancer risk was considered by odds ratios (ORs) together with their 95% confidence intervals (CIs). A total of 31 studies including 11,606 cases and 12,655 controls were involved in this meta-analysis. Overall, this meta-analysis showed no association between CDH1 -160C>A polymorphism and GI cancer risk (A vs. C: OR = 1.08, 95%CI = 0.98-1.18, P = 0.086;CA vs. CC: OR = 1.09, 95%CI = 0.97-1.22, P = 0.118; AA vs. CC: OR = 1.10, 95%CI = 0.89-1.35, P = 0.356; AA vs. CC + CA: OR = 1.06, 95%CI = 0.96-1.18, P = 0.207; CA+AA vs. CC: OR = 1.01, 95%CI = 0.84-1.22, P = 0.89). In subgroup analysis, similar results were found. In conclusion, this meta-analysis has demonstrated that there is a lack of association of the CDH1-160C>A polymorphism with GI cancer susceptibility.

Effect of β-glucan on Epithelial Inflammation Induced by Atopic Dermatitis through Endocannabinoid System Activity (β-glucan의 Endocannabinoid system 활성을 통한 아토피피부염 유발 상피 염증 억제 효과)

  • Seo, Il Bok;Ahn, Sang Hyun;Kim, Ki Bong
    • The Journal of Pediatrics of Korean Medicine
    • /
    • v.34 no.4
    • /
    • pp.22-30
    • /
    • 2020
  • Objectives This study was conducted to confirm the inhibitory effect of β-glucan on epithelial inflammation induced by atopic dermatitis through Endocannabinoid system (ECS) activity. Methods Six-week-old NC/Nga mice were divided into a control group (Ctrl), atopic dermatitis elicitation group (ADE), and a β-glucan-treated group (β-glucan treatment after atopy dermatitis elicitation, β-GT). After 3 weeks, CB1, CB2, and GPR55 were observed to confirm the regulation of ECS activity, and filaggrin in the stratum corneum and Kallikrein-related peptidase (KLK) 7 in the stratum corneum and protease activated receptor (PAR)-2 were observed to confirm the inhibition of the inflammation, Phosphorylated extracellular signal-related kinase (p-ERK), Phosphorylated mammalian target of rapamycin (p-mTOR), and E-Cadherin were observed to confirm microenvironmental regulation. Results β-GT was significantly increased in CB1, CB2, and GPR55 positive reactions compared to that of the ADE. In positive reaction of the filaggrin in the stratum corneum, β-GT was significantly increased than that of the ADE. For KLK7 positive and PAR2 positive, β-GT was significantly reduced compared to the ADE. The p-ERK-positive and p-mTOR-positive reactions were significantly reduced in β-GT than in ADE. E-cadherin positive reaction was significantly increased in β-GT than in ADE (All p < 0.01). Conclusions It was confirmed that β-glucan has the effect of inhibiting the epithelium induced by atopic dermatitis through the ECS activity.

Megakaryocyte-Derived IL-8 Acts as a Paracrine Factor for Prostate Cancer Aggressiveness through CXCR2 Activation and Antagonistic AR Downregulation

  • Sadan, Dahal;Prakash, Chaudhary;Yi-Sook, Jung;Jung-Ae, Kim
    • Biomolecules & Therapeutics
    • /
    • v.31 no.2
    • /
    • pp.210-218
    • /
    • 2023
  • Prostate cancer is the fifth leading cause of cancer-related mortality in men, primarily because of treatment resistance, recurrence, and metastasis. In the present study, we investigated the role of paracrine interleukin-8 (IL-8) in the antagonistic expression of IL-8 and androgen receptor (AR), and the contribution of IL-8 to prostate cancer aggressiveness. In hormone-responsive LNCaP cells that do not express IL-8, recombinant IL-8 treatment significantly increased expressions of IL-8, CXC chemokine receptor 2 (CXCR2), matrix metalloproteinase (MMP)-2/9, Snail, and vimentin. IL-8 treatment significantly decreased AR and E-cadherin expression. IL-8-induced gene expression changes were suppressed by navarixin, a CXCR1/2 inhibitor, and gallein, a Gβγ inhibitor. In PC-3 androgen-refractory prostate cancer cells, IL-8 knockdown reduced expressions of CXCR2, MMP-2/9, Snail, and vimentin, and increased AR and E-cadherin expressions at the mRNA and protein levels. Co-culture with MEG-01 human megakaryocytic cells secreting high levels of IL-8 induced gene expression changes in both LNCaP and PC-3 cells, similar to those induced by IL-8 treatment. The altered gene expressions were accompanied by significant activation of transcription factor Snail in LNCaP and PC-3 cells. Treatment with the CXCR blocker navarixin inhibited the invasion of PC-3 cells but not LNCaP cells. However, invasion induced by MEG-01 was inhibited by navarixin in both LNCaP and PC-3 cells. The collective findings demonstrate that IL-8 enhances CXCR2 expression, which antagonistically regulates AR expression. More importantly, through changes in IL-8/CXCR2-regulated gene expression, IL-8 induces antiandrogen therapy resistance and epithelial-mesenchymal transition in prostate cancer.

Correlation of Overexpression of Nestin with Expression of Epithelial-Mesenchymal Transition-Related Proteins in Gastric Adenocarcinoma

  • Liu, Jin-Kai;Chen, Wan-Cheng;Ji, Xiao-Zhen;Zheng, Wen-Hong;Han, Wei;An, Jing
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.7
    • /
    • pp.2777-2783
    • /
    • 2015
  • Background: Nestin is associated with neoplastic transformation. However, the mechanisms by which nestin contributes regarding invasion and malignancy of gastric adenocarcinoma (GAC) remain unknown. Recent studies have shown that the epithelial-mesenchymal transition (EMT) is important in invasion and migration of cancer cells. In the present study, we aimed to investigate the expression of nestin and its correlation with EMT-related proteins in GAC. Materials and Methods: The expression of nestin and EMT-related proteins was examined in GAC specimens and cell lines by immunohistochemistry and Western blotting. Clinicopathological features and survival outcomes were retrospectively analyzed. Results: Positive nestin immunostaining was most obviously detected in the cytoplasm, nucleus or both cytoplasm and nucleus of tumor cells in 19.2% (24/125) of GAC tissues, which was significantly higher than that in normal gastric mucosa tissues (1.7%, 1/60) (p=0.001). Nestin expression was closely related to several clinicopathological factors and EMT-related proteins (E-cadherin, vimentin and Snail) and displayed a poor prognosis. Interestingly, simultaneous cytoplasmic and nuclear nestin expression correlated with EMT-related proteins (E-cadherin, vimentin and Snail) (p<0.05) and lymph node metastasis (p=0.041) and a shorter survival time (p<0.05), but this was not the case with cytoplasmic or nuclear nestin expression. Conclusions: Nestin, particularly expression in both cytoplasm and nucleus, might be involved in regulating EMT and malignant progression in GAC, with potential as an unfavorable indicator in tumor diagnosis and a target for clinical therapy.

Loss of FAT Atypical Cadherin 4 Expression Is Associated with High Pathologic T Stage in Radically Resected Gastric Cancer

  • Jung, Hae Yoen;Cho, Hyundeuk;Oh, Mee-Hye;Lee, Ji-Hye;Lee, Hyun Ju;Jang, Si-Hyong;Lee, Moon Soo
    • Journal of Gastric Cancer
    • /
    • v.15 no.1
    • /
    • pp.39-45
    • /
    • 2015
  • Purpose: Recent studies have revealed recurrent alterations in the cell adhesion gene FAT4, a candidate tumor suppressor gene, in cancer. FAT atypical cadherin 4 (FAT4) is a transmembrane receptor involved in the Hippo signaling pathway, which is involved in the control of organ size. Here, we investigated the loss of FAT4 expression and its association with clinicopathological risk factors in gastric cancer. Materials and Methods: We assessed the expression of FAT4 by using immunohistochemistry on three tissue microarrays containing samples from 136 gastric cancer cases, radically resected in the Soonchunhyang University Cheonan Hospital between July 2006 and June 2008. Cytoplasmic immunoexpression of FAT4 was semi-quantitatively scored using the H-score system. An H-score of ${\geq}10$ was considered positive for FAT4 expression. Results: Variable cytoplasmic expressions of FAT4 were observed in gastric cancers, with 33 cases (24.3%) showing loss of expression (H-score <10). Loss of FAT4 expression was associated with an increased rate of perineural invasion (H-score <10 vs. ${\geq}10$, 36.4% vs. 16.5%, P=0.015), high pathologic T stage (P=0.015), high tumor-node-metastasis stage (P=0.017), and reduced disease-free survival time (H-score <10 vs. ${\geq}10$, mean survival $62.7{\pm}7.3$ months vs. $79.1{\pm}3.1$ months, P=0.025). However, no association was found between the loss of FAT4 expression and tumor size, gross type, histologic subtype, Lauren classification, lymphovascular invasion, or overall survival. Conclusions: Loss of FAT4 expression appears to be associated with invasiveness in gastric cancer.

3-Deazaneplanocin A is a Promising Therapeutic Agent for Ovarian Cancer Cells

  • Shen, Liang;Cui, Jing;Pang, Ying-Xin;Ma, Yan-Hui;Liu, Pei-Shu
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.5
    • /
    • pp.2915-2918
    • /
    • 2013
  • Background: Recent studies have shown that 3-deazaneplanocin A (DZNep), a well-known histone methyltransferase inhibitor, disrupts polycomb-repressive complex 2 (PRC2), and induces apoptosis, while inhibiting proliferation and metastasis, in cancer cells, including acute myeloid leukemia, breast cancer and glioblastoma. However, little is known about effects of DZNep on ovarian cancer cells. Materials and Methods: We here therefore studied DZNep-treated A2780 ovarian cancer cells in vitro. Proliferation of ovarian cancer cells under treatment of DZNep was assessed by MTT and apoptosis by flow cytometry. Cell wound healing was applied to detect the migration. Finally, we used q-PCR to assess the migration-related gene, E-cadherin. Results: DZNep could inhibit the proliferation of A2780 and induce apoptosis Furthermore, it inhibited migration and increased the expression of E-cadherin (P<0.05). Conclusion: DZNep is a promising therapeutic agent for ovarian cancer cells, with potential to inhibite proliferation, induce apoptosis and decrease migration.

Anticancer effects of D-pinitol in human oral squamous carcinoma cells

  • Shin, Hyun-Chul;Bang, Tea-Hyun;Kang, Hae-Mi;Park, Bong-Soo;Kim, In-Ryoung
    • International Journal of Oral Biology
    • /
    • v.45 no.4
    • /
    • pp.152-161
    • /
    • 2020
  • D-pinitol is an analog of 3-methoxy-D-chiro-inositol found in beans and plants. D-pinitol has anti-inflammatory, antidiabetic, and anticancer effects. Additionally, D-pinitol induces apoptosis and inhibits metastasis in breast and prostate cancers. However, to date, no study has investigated the anticancer effects of D-pinitol in oral cancer. Therefore, in this study, whether the anticancer effects of D-pinitol induce apoptosis, inhibit the epithelial-to-mesenchymal transition (EMT), and arrest cell cycle was investigated in squamous epithelial cells. D-pinitol decreased the survival and cell proliferation rates of CAL-27 and Ca9-22 oral squamous carcinoma cells in a concentration- and time-dependent manner. Evidence of apoptosis, including nuclear condensation, poly (ADP-ribose) polymerase, and caspase-3 fragmentation, was also observed. D-pinitol inhibited the migration and invasion of both cell lines. In terms of EMT-related proteins, E-cadherin was increased, whereas N-cadherin, Snail, and Slug were decreased. D-pinitol also decreased the expression of cyclin D1, a protein involved in the cell cycle, but increased the expression of p21, a cyclin-dependent kinase inhibitor. Hence, D-pinitol induces apoptosis and cell cycle arrest in CAL-27 and Ca9-22 cells, demonstrating an anticancer effect by decreasing the EMT.

Inhibitory Effects of Flavonoids on Growth of HT-29 Human Colon Cancer Cells (Flavonoid의 HT-29 대장암세포 증식 억제 효과)

  • Cho, Young;Choi, Mi-Yong
    • Journal of the Korean Society of Food Science and Nutrition
    • /
    • v.44 no.3
    • /
    • pp.338-346
    • /
    • 2015
  • This study was performed to elucidate the anti-proliferative and apoptotic mechanism of flavonoids in HT-29 human colon cancer cells. We investigated the anti-proliferative activity of flavonoids in HT-29 human colon cancer cells via cell viability assay (MTT assay), caspase-3 activity, RT-PCR, and western blotting. We cultured HT-29 cells in the presence of various flavonoids (apigenin, rutin, naringenin, and myricetin) at a concentration of $100{\mu}M$. In the MTT assay, naringenin showed the strongest effect on cell viability in HT-29 colon cancer cells. Caspase-3 activity, a marker of apoptosis, significantly increased upon naringenin treatment. For RT-PCR, myricetin significantly increased Bax protein levels, naringenin increased p53 protein levels, and rutin reduced expression of the anti-apoptotic protein Bcl-2. Western blotting of HT-29 colon cancer cells showed that myricetin increased cleaved caspase-3 protein levels, naringenin significantly increased poly (ADP-ribose) polymerase protein levels, and rutin increased E-cadherin protein levels. These results indicate that flavonoid exerts anticancer effects on human colon HT-29 cells through a caspase-dependent apoptotic pathway.

Hypermethylation Status of E-Cadherin Gene in Gastric Cancer Patients in a High Incidence Area

  • Rashid, Haroon;Alam, Khursheed;Afroze, Dil;Yousuf, Adfar;Banday, Manzoor;Kawoosa, Fizalah
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.6
    • /
    • pp.2757-2760
    • /
    • 2016
  • Gastric cancer (GC) is the fourth most prevalant cancer and the second leading cause of cancer-related mortality worldwide. As in other cancers gastric carcinogenesis is multifactorial involving environmental, genetic and epigenetic components. Epigenetic silencing due to hypermethylation of tumour suppressor genes is one of the key events in gastric carcinogenesis. This study was aimed to analyse the hypermethylation status of the E-Cadherin (CDH1) gene promoter in GCs in the ethnic Kashmiri population. In this study a total of 80 GC patients were recruited. Hypermethylation in tumour tissue was detected by methylation specific PCR (MS-PCR). Hypermethylation of CDH1 promoter was observed in 52 (65%) of gastric carcinoma cases which was significantly much higher than adjacent normal tissue [$p{\leq}0.0001$]. Further the frequency of CDH1 promoter methylation was significantly different with intestinal and diffuse types of gastric cancer [55.7% vs 82.1%; p<0.05]. Moreover females and cases with lymph node invasion had higher frequencies of CDH1 hypermethylation [$P{\leq}0.05$]. Thus the current data indicate a vital role of epigenetic alteration of CDH1 in the causation and development of gastric cancer, particularly of diffuse type, in our population.