• 제목/요약/키워드: tumor-migration

검색결과 372건 처리시간 0.028초

EZH2-Mediated microRNA-139-5p Regulates Epithelial-Mesenchymal Transition and Lymph Node Metastasis of Pancreatic Cancer

  • Ma, Jin;Zhang, Jun;Weng, Yuan-Chi;Wang, Jian-Cheng
    • Molecules and Cells
    • /
    • 제41권9호
    • /
    • pp.868-880
    • /
    • 2018
  • Pancreatic cancer (PC) is one of the most aggressive cancers presenting with high rates of invasion and metastasis, and unfavorable prognoses. The current study aims to investigate whether EZH2/miR-139-5p axis affects epithelial-mesenchymal transition (EMT) and lymph node metastasis (LNM) in PC, and the mechanism how EZH2 regulates miR-139-5p. Human PC and adjacent normal tissues were collected to determine expression of EZH2 and miR-139-5p, and their relationship with clinicopathological features of PC. Human PC cell line was selected, and treated with miR-139-5p mimics/inhibitors, EZH2 vector or shEZH2 in order to validate the regulation of EZH2-mediated miR-139-5p in PC cells. Dual-luciferase report gene assay and chromatin immunoprecipitation assay were employed to identify the relationship between miR-139-5p and EZH2. RT-qPCR and Western blot analysis were conducted to determine the expression of miR-139-5p, EZH2 and EMT-related markers and ZEB1/2. Tumor formation ability and in vitro cell activity were also analyzed. Highly-expressed EZH2 and poorly-expressed miR-139-5p were detected in PC tissues, and miR-139-5p and EZH2 expressions were associated with patients at Stage III/IV, with LNM and highly-differentiated tumors. EZH2 suppressed the expression of miR-139-5p through up-regulating Histone 3 Lysine 27 Trimethylation (H3K27me3). EMT, cell proliferation, migration and invasion were impeded, and tumor formation and LNM were reduced in PC cells transfected with miR-139-5p mimics and shEZH2. MiR-139-5p transcription is inhibited by EZH2 through up-regulating H3K27me3, thereby down-regulation of EZH2 and up-regulation of miR-139-5p impede EMT and LNM in PC. In addition, the EZH2/miR-139-5p axis presents as a promising therapeutic strategy for the treatment of PC.

Expression Pattern of EphB2 in Gastric Cancer (위암에시 EphB2 단백의 발현 양상)

  • Song, Jae-Hwi;Kim, Chang-Jae;Cho, Young-Gu;Park, Cho-Hyun;Nam, Suk-Woo;Yoo, Nam-Jin;Lee, Jung-Young;Park, Won-Sang
    • Journal of Gastric Cancer
    • /
    • 제6권1호
    • /
    • pp.25-30
    • /
    • 2006
  • Purpose: The EphB2 receptor, a member of the receptor tyrosine kinase family, is a target gene of the Wnt signaling pathway and may achieve a tumor suppressor function through regulation of cell growth and migration. Our aim was to determine whether an altered expression of EphB2 might be associated with gastric cancer development and, if so, to determine to which pathologic parameter it is linked. Materials and Methods: For the construction of the gastric cancer tissue microarray, 83 paraffin-embedded tissues containing gastric cancer areas were cored 3 times and transferred to the recipient master block. The expression patterns of EphB2 were examined on tissue microarray slides by using immunohistochemistry and were compared using pathologic parameters, including histological type, depth of invasion, lymph node metastatsis, and peritoneal dissemination. Results: The EphB2 protein was expressed in the normal gastric mucosal epithelium, especially in the bottom of the mucosa. We found loss of EphB2 expression in 30 (36.1%) of the 83 gastric cancer tissues. Statistically, loss of EphB2 expression was more common in gastric cancer with lymph-node metastasis. There was no significant correlation between EphB2 expression and depth of invasion, histologic type, or peritoneal dissemination. Conclusion: Our findings suggest that loss of EphB2 expression may represent a critical step in gastric carcinogenesis.

  • PDF

Identification and Functional Characterization of Two Noncoding RNAs Transcribed from Putative Active Enhancers in Hepatocellular Carcinoma

  • Lee, Ye-Eun;Lee, Jiyeon;Lee, Yong Sun;Jang, Jiyoung Joan;Woo, Hyeonju;Choi, Hae In;Chai, Young Gyu;Kim, Tae-Kyung;Kim, TaeSoo;Kim, Lark Kyun;Choi, Sun Shim
    • Molecules and Cells
    • /
    • 제44권9호
    • /
    • pp.658-669
    • /
    • 2021
  • Enhancers have been conventionally perceived as cis-acting elements that provide binding sites for trans-acting factors. However, recent studies have shown that enhancers are transcribed and that these transcripts, called enhancer RNAs (eRNAs), have a regulatory function. Here, we identified putative eRNAs by profiling and determining the overlap between noncoding RNA expression loci and eRNA-associated histone marks such as H3K27ac and H3K4me1 in hepatocellular carcinoma (HCC) cell lines. Of the 132 HCC-derived noncoding RNAs, 74 overlapped with the eRNA loci defined by the FANTOM consortium, and 65 were located in the proximal regions of genes differentially expressed between normal and tumor tissues in TCGA dataset. Interestingly, knockdown of two selected putative eRNAs, THUMPD3-AS1 and LINC01572, led to downregulation of their target mRNAs and to a reduction in the proliferation and migration of HCC cells. Additionally, the expression of these two noncoding RNAs and target mRNAs was elevated in tumor samples in the TCGA dataset, and high expression was associated with poor survival of patients. Collectively, our study suggests that noncoding RNAs such as THUMPD3-AS1 and LINC01572 (i.e., putative eRNAs) can promote the transcription of genes involved in cell proliferation and differentiation and that the dysregulation of these noncoding RNAs can cause cancers such as HCC.

Structural Disorganization of Intestinal Tumor Spheroid by Microbial Ribotoxins (방사선 모사 미생물 유래 리보솜 스트레스에 의한 대장암 스페로이드 구조 결함 유발)

  • Kim, Juil;Kim, Joongkon;Yu, Mira;Moon, Yuseok
    • Microbiology and Biotechnology Letters
    • /
    • 제47권1호
    • /
    • pp.164-171
    • /
    • 2019
  • Radiation therapy has many side effects, such as digestive mucosal ulcers, without regard to its efficacy. The purpose of this study is to address an alternative method to replace the limitation of radiation therapy using radiomimetic microbial ribotoxins. In the evaluation of cancer therapy, we analyzed the formation of colorectal cancer (CRC) cell spheroids, which can take into account the heterogeneous cellular constitution, tumor stem cells, and the surrounding microenvironment. Ribotoxic stress interfered with the spheroid structure composed of relatively small clusters. Spheroids under ribotoxic stress were structurally sparse and their shrinkage was very slow. In the control group, the clusters of strongly aggregated cells were resistant to physical stress, but the ribotoxic stress-exposed spheroids were easily broken up by the physical stress. Moreover, the ribosome-insulted CRC cells slowly migrated to form clusters and the cell-cell junctional points in the ribosome-insulted spheroids were rarer than those in the control CRC spheroid. Moreover, levels of the cell-to-cell junctional protein E-cadherin were suppressed by ribotoxic stress in both allograft and xenograft spheroids. In conclusion, the radiomimetic microbial ribotoxins induced structural defects in CRC cell spheroids via retardation of migration and cell-cell junction in the formation of three-dimensional structures, and provides a basis for the mechanism of pharmacological radiomimetic anticancer actions as an alternate to radiotherapy against cancer.

Negative Regulation of Tumor Suppressor p53 at the Promoter Regions of Oncogenic SETDB1 and FosB Genes (암종양유전자 SETDB1과 FosB 발현에 대한 p53의 음성 조절기작)

  • Yun, Hyeon Ji;Na, Han-Heom;Kim, Keun-Cheol
    • Journal of Life Science
    • /
    • 제30권12호
    • /
    • pp.1070-1077
    • /
    • 2020
  • Treatment with anticancer drugs changes the expression of multiple genes related to cell proliferation, migration, and drug resistance. These changes in gene expression may be connected to regulatory networks for each other. This study showed that doxorubicin treatment induces the expression of oncogenic FosB and decreases the expression of oncogenic SETDB1 in A549 and H1299 human lung cancer cells, which are different in tumor suppressor p53 status. However, a small difference was detected in the quantitative expression of those proteins in the two kinds of cells. To examine the potential regulation of SETDB1 and FosB by p53, we predicted putative p53 binding sites on the genomic DNA of SETDB1 and FosB using a TF motif binding search program. These putative p53 binding sites were identified as 18 sites in the promoter regions of SETDB1 and 21 sites in the genomic DNA of FosB. A luciferase assay confirmed that p53 negatively regulated the promoter activities of SETDB1 and FosB. Furthermore, the results of RT-PCR, western blot, qPCR, and immunostaining experiments indicated that the transfection of exogenous p53 decreases the expression of SETDB1 and FosB in H1299 cells. This indicates that p53 negatively regulates the expression of SETDB1 and FosB at the transcriptional level. Collectively, the downregulation of SETDB1 and FosB by p53 may provide functional networks for apoptosis and for the survival of cancer cells during anticancer drug treatment.

Molecular Analysis of the Interaction between Human PTPN21 and the Oncoprotein E7 from Human Papillomavirus Genotype 18

  • Lee, Hye Seon;Kim, Min Wook;Jin, Kyeong Sik;Shin, Ho-Chul;Kim, Won Kon;Lee, Sang Chul;Kim, Seung Jun;Lee, Eun-Woo;Ku, Bonsu
    • Molecules and Cells
    • /
    • 제44권1호
    • /
    • pp.26-37
    • /
    • 2021
  • Human papillomaviruses (HPVs) cause cellular hyperproliferation-associated abnormalities including cervical cancer. The HPV genome encodes two major viral oncoproteins, E6 and E7, which recruit various host proteins by direct interaction for proteasomal degradation. Recently, we reported the structure of HPV18 E7 conserved region 3 (CR3) bound to the protein tyrosine phosphatase (PTP) domain of PTPN14, a well-defined tumor suppressor, and found that this intermolecular interaction plays a key role in E7-driven transformation and tumorigenesis. In this study, we carried out a molecular analysis of the interaction between CR3 of HPV18 E7 and the PTP domain of PTPN21, a PTP protein that shares high sequence homology with PTPN14 but is putatively oncogenic rather than tumor-suppressive. Through the combined use of biochemical tools, we verified that HPV18 E7 and PTPN21 form a 2:2 complex, with a dissociation constant of 5 nM and a nearly identical binding manner with the HPV18 E7 and PTPN14 complex. Nevertheless, despite the structural similarities, the biological consequences of the E7 interaction were found to differ between the two PTP proteins. Unlike PTPN14, PTPN21 did not appear to be subjected to proteasomal degradation in HPV18-positive HeLa cervical cancer cells. Moreover, knockdown of PTPN21 led to retardation of the migration/invasion of HeLa cells and HPV18 E7-expressing HaCaT keratinocytes, which reflects its protumor activity. In conclusion, the associations of the viral oncoprotein E7 with PTPN14 and PTPN21 are similar at the molecular level but play different physiological roles.

Combination Therapy of the Active KRAS-Targeting Antibody inRas37 and a PI3K Inhibitor in Pancreatic Cancer

  • Lee, Ji Eun;Woo, Min Gyu;Jung, Kyung Hee;Kang, Yeo Wool;Shin, Seung-Min;Son, Mi Kwon;Fang, Zhenghuan;Yan, Hong Hua;Park, Jung Hee;Yoon, Young-Chan;Kim, Yong-Sung;Hong, Soon-Sun
    • Biomolecules & Therapeutics
    • /
    • 제30권3호
    • /
    • pp.274-283
    • /
    • 2022
  • KRAS activating mutations, which are present in more than 90% of pancreatic cancers, drive tumor dependency on the RAS/mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/AKT signaling pathways. Therefore, combined targeting of RAS/MAPK and PI3K/AKT signaling pathways may be required for optimal therapeutic effect in pancreatic cancer. However, the therapeutic efficacy of combined MAPK and PI3K/AKT signaling target inhibitors is unsatisfactory in pancreatic cancer treatment, because it is often accompanied by MAPK pathway reactivation by PI3K/AKT inhibitor. Therefore, we developed an inRas37 antibody, which directly targets the intra-cellularly activated GTP-bound form of oncogenic RAS mutation and investigated its synergistic effect in the presence of the PI3K inhibitor BEZ-235 in pancreatic cancer. In this study, inRas37 remarkably increased the drug response of BEZ-235 to pancreatic cancer cells by inhibiting MAPK reactivation. Moreover, the co-treatment synergistically inhibited cell proliferation, migration, and invasion and exhibited synergistic anticancer activity by inhibiting the MAPK and PI3K pathways. The combined administration of inRas37and BEZ-235 significantly inhibited tumor growth in mouse models. Our results demonstrated that inRas37 synergistically increased the antitumor activity of BEZ-235 by inhibiting MAPK reactivation, suggesting that inRas37 and BEZ-235 co-treatment could be a potential treatment approach for pancreatic cancer patients with KRAS mutations.

P3H4 promotes renal cell carcinoma progression and suppresses antitumor immunity via regulating GDF15-MMP9-PD-L1 axis

  • Tian, Shuo;Huang, Yan;Lai, Dong;Wang, Hanfeng;Du, Songliang;Shen, Donglai;Chen, Weihao;Xuan, Yundong;Lu, Yongliang;Feng, Huayi;Zhang, Xiangyi;Zhao, Wenlei;Wang, Chenfeng;Wang, Tao;Wu, Shengpan;Huang, Qingbo;Niu, Shaoxi;Wang, Baojun;Ma, Xin;Zhang, Xu
    • Advances in nano research
    • /
    • 제12권6호
    • /
    • pp.639-652
    • /
    • 2022
  • The prolyl 3-hydroxylase family member 4 (P3H4), is associated with post-translational modification of fibrillar collagens and aberrantly activated in cancer leading to tumor progression. However, its role in clear cell renal cell carcinoma (ccRCC) is still unknown. Here we reported that P3H4 was highly expressed in renal cancer tissues and significantly positive correlated with poor prognosis. Knockdown of P3H4 inhibited the proliferation, migration and metastasis of renal cancer cells in vitro and in vivo, and also, overexpression of it enhanced the oncogenic process. Mechanistically, P3H4 depletion decreased the levels of GDF15-MMP9 axis and repressed its downstream signaling. Further functional studies revealed that inhibition of GDF15 suppressed renal cancer cell growth and GDF15 recombinant human protein (rhGDF15) supplementation effectively rescued the inhibitory effect induced by P3H4 knockdown. Moreover, decreased levels of MMP9 caused by inhibition of P3H4-GDF15 signaling constrained the expression of PD-L1 and suppression of P3H4 accordingly promoted anti-tumor immunity via stimulating the infiltration of CD4+ and CD8+ T cells in syngeneic mice model. Taken together, our findings firstly demonstrated that P3H4 promotes ccRCC progression by activating GDF15-MMP9-PD-L1 axis and targeting P3H4-GDF15-MMP9 signaling pathway can be a novel strategy of controlling ccRCC malignancy.

Accelerating Effects of Quercetin on the $TNF-{\alpha}-Induced$ Apoptosis in MC3T3-E1 Osteoblastic Cells

  • Choi, Yong-Sung;Chung, Song-Woo;Jeon, Young-Mi;Kim, Jong-Ghee;Lee, Jeong-Chae
    • Natural Product Sciences
    • /
    • 제11권3호
    • /
    • pp.139-144
    • /
    • 2005
  • Bioflavone quercetin is believed to play an important role preventing bone loss by affecting osteoclastogenesis and regulating many systemic and local factors including hormones and cytokines. This study examined how quercetin acts on tumor necrosis factor-alpha ($TNF-{\alpha}$)-mediated apoptosis in MC3T3-E1 osteoblastic cells. Apoptosis assays revealed the dose-dependent acceleration of quercetin on $TNF-{\alpha}-induced$ apoptosis in MC3T3-E1 cells, which was demonstrated by the increased number of positively stained cells in the trypan blue staining and TUNEL assay, and the migration of many cells to the $sub-G_0/G_1$ phase in flow cytometric analysis. In particular, quercetin treatment alone increased the expression of p53 and p21 proteins in the cells. Consequently, this study showed that quercetin accelerates the $TNF-{\alpha}-induced$ apoptosis in MC3T3-E1 osteoblastic cells.

Cell Cycle Arrest of Human Lung Carcinoma A549 Cells by an Aqueous Extract from the Roots of Platycodon grandiflorum (길경 수용액 추출물에 의한 인체 폐암세포의 성장억제 기전 연구)

  • Kang Rak Won;Lee Jae Hun;Kam Cheol Woo;Choi Byung Tae;Choi Yung Hyun;Park Dong Il
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • 제17권1호
    • /
    • pp.183-189
    • /
    • 2003
  • Platycodi Radix, the root of Platycodon grandiflorum, commonly known as Doraji, is used as a traditional oriental medicine. Extracts from the roots of P. grandiflorum have been reported to have wide ranging health benefits. We investigated the effects of an aqueous extract from the roots of P. grandiflorum (AEPG) on the cell proliferation of human lung carcinoma A549 cells in order to understand its anti-proliferative mechanism. AEPG treatment resulted in the inhibition of cell proliferation in a concentration-dependent manner. This anti-proliferative effect of A549 cells by AEPG treatment was associated with morphological changes such as membrane shrinking, cell rounding up and inhibition of cell migration. DNA flow cytometric histograms showed that populations of both Sand G2/M phase of the cell cycle were increased by AEPG treatment in a concentration-dependent manner. AEPG treatment induced a marked accumulation of tumor suppressor p53 and a concomitant induction of cyclin-dependent kinase (Cdk) inhibitor p21 and p27. In addition, SSS treatment resulted in down-regulation of Cdk2 and Cdk4 expression. The present results indicated that AEPG-induced inhibition of lung cancer cell proliferation is associated with the blockage of S to G2/M phase progression the induction of apoptosis. Taken together, these findings suggest that P. grandiflorum has strong potential for development as an agent for prevention against human lung cancer.