• Title/Summary/Keyword: metastasis suppressor

Search Result 73, Processing Time 0.023 seconds

Targeting the Transforming Growth Factor-β Signaling in Cancer Therapy

  • Sheen, Yhun Yhong;Kim, Min-Jin;Park, Sang-A;Park, So-Yeon;Nam, Jeong-Seok
    • Biomolecules & Therapeutics
    • /
    • v.21 no.5
    • /
    • pp.323-331
    • /
    • 2013
  • TGF-${\beta}$ pathway is being extensively evaluated as a potential therapeutic target. The transforming growth factor-${\beta}$ (TGF-${\beta}$) signaling pathway has the dual role in both tumor suppression and tumor promotion. To design cancer therapeutics successfully, it is important to understand TGF-${\beta}$ related functional contexts. This review discusses the molecular mechanism of the TGF-${\beta}$ pathway and describes the different ways of tumor suppression and promotion by TGF-${\beta}$. In the last part of the review, the data on targeting TGF-${\beta}$ pathway for cancer treatment is assessed. The TGF-${\beta}$ inhibitors in pre-clinical studies, and Phase I and II clinical trials are updated.

Altered Distribution and Expression Pattern of E-cadherin in Hepatocellular Carcinomas: Correlations with Prognosis and Clinical Features

  • Jiang, Xue-Mei;Zhang, Ju-Bo;Xiong, Ju;Huang, Xiao-Xi;Ren, Zheng-Gang
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.13 no.12
    • /
    • pp.6455-6461
    • /
    • 2012
  • Objective: E-cadherin has been identified as a tumor suppressor in many types of carcinoma. However, some studies recently suggested that the role and expression of E-cadherin might be more complex and diverse. In the present study, we evaluated the prognostic value of E-cadherin expression with reference to levels in membranes and cytoplasm, and the membrane/cytoplasm ratio, in hepatocellular carcinomas (HCCs) after curative hepatectomy. Methods: The expression of E-cadherin was assessed by immunohistochemistry in HCC tissue microarrays from 125 patients, and its prognostic values and other clinicopathlogical data were retrospectively analyzed. Patients were followed for a median period of 43.7 months (range 1 to 126 months). Results: Univariate analysis demonstrated that a high membrane/cytoplasm (M/C) ratio of E-cadherin expression was associated with poor overall survival (OS) (P =0.001) and shorter time to recurrence (TTR) (P=0.038), as well as tumor size, intrahepatic metastasis, and TNM stage. In contrast, neither membrane nor cytoplasmic expression of E-cadherin was related with OS and TTR. Furthermore, multivariate analysis confirmed the M/C ratio to be an independent predictor of OS (P=0.031). ${\chi}^2$ tests additionally showed that the M/C ratio of E-cadherin expression was related with early stage recurrence (P=0.012), rather than later stage recurrence. Conclusion: The M/C ratio of E-cadherin expression is a strong predictor of postoperative survival and is associated with early stage recurrence in patients with HCC.

Evaluation of KiSS1 as a Prognostic Biomarker in North Indian Breast Cancer Cases

  • Singh, Richa;Bhatt, Madan Lal Brahma;Singh, Saurabh Pratap;Kumar, Vijay;Goel, Madhu Mati;Mishra, Durga Prasad;Kumar, Rajendra
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.4
    • /
    • pp.1789-1795
    • /
    • 2016
  • Background: Breast cancer is the commonest female cancer worldwide and its propensity to metastasize negatively impacts on therapeutic outcome. Several clinicopathological parameters with prognostic/predictive significance have been associated with metastatic suppressor expression levels. The role of metastatic suppressor gene (MSG) KiSS1 in breast cancer remains unclear. Our goal was to investigate the possible clinical significance of KiSS1 breast cancer. Materials and Methods: The study was conducted on 87 histologically proven cases of breast cancer and background normal tiisue. Quantitative reverse transcriptase polymerase chain reaction (qRT PCR) and immunohistochemistry (IHC) were used to investigate KiSS1 at gene and protein levels, respectively, for correlation with several patient characteristics including age, family history, hormonal receptor status, stage, tumor size, nodal involvement and metastatic manifestation and finally with median overall survival (OS). Results: Our study revealed (i) KiSS1 levels were generally elevated in breast cancer vs normal tissue (P < 0.05). (ii) however, a statistically significant lower expression of KiSS1 was observed in metastatic vs non metastatic cases (P = 0.04). (iii) KiSS1 levels strongly correlated with T,N,M category, histological grade and advanced stage (p<0.001) but not other studied parameters. (iv) Lastly, a significant correlation between expression of KiSS1 and median OS was found (P = 0.04). Conclusions: Conclusively, less elevated KiSS1 expression is a negative prognostic factor for OS, advancing tumor stage, axillary lymph node status, metastatic propensity and advancing grade of the breast cancer patient. Patients with negative KiSS1 expression may require a more intensive therapeutic strategy.

SMAD4 Controls Cancer Cell Metabolism by Regulating Methylmalonic Aciduria Cobalamin Deficiency (cbl) B Type

  • Song, Kyoung;Lee, Hun Seok;Jia, Lina;Chelakkot, Chaithanya;Rajasekaran, Nirmal;Shin, Young Kee
    • Molecules and Cells
    • /
    • v.45 no.6
    • /
    • pp.413-424
    • /
    • 2022
  • Suppressor of mothers against decapentaplegic homolog (SMAD) 4 is a pluripotent signaling mediator that regulates myriad cellular functions, including cell growth, cell division, angiogenesis, apoptosis, cell invasion, and metastasis, through transforming growth factor β (TGF-β)-dependent and -independent pathways. SMAD4 is a critical modulator in signal transduction and functions primarily as a transcription factor or cofactor. Apart from being a DNA-binding factor, the additional SMAD4 mechanisms in tumor suppression remain elusive. We previously identified methyl malonyl aciduria cobalamin deficiency B type (MMAB) as a critical SMAD4 binding protein using a proto array analysis. This study confirmed the interaction between SMAD4 and MMAB using bimolecular fluorescence complementation (BiFC) assay, proximity ligation assay (PLA), and conventional immunoprecipitation. We found that transient SMAD4 overexpression down-regulates MMAB expression via a proteasome-dependent pathway. SMAD4-MMAB interaction was independent of TGF-β signaling. Finally, we determined the effect of MMAB downregulation on cancer cells. siRNA-mediated knockdown of MMAB affected cancer cell metabolism in HeLa cells by decreasing ATP production and glucose consumption as well as inducing apoptosis. These findings suggest that SMAD4 controls cancer cell metabolism by regulating MMAB.

Expression Pattern of KLF6 in Korean Gastric Cancers (한국인 위암에서 KLF6 단백 발현 양상)

  • Cho Young Gu;Kim Chang Jae;Park Cho Hyun;Kim Su Young;Nam Suk Woo;Lee Sug Hyung;Yoo Nam Jin;Lee Jung Young;Park Won Sang
    • Journal of Gastric Cancer
    • /
    • v.5 no.1
    • /
    • pp.34-39
    • /
    • 2005
  • Purpose: KLF6, a member of the KLF family, is a ubiquitous zinc finger tumor suppressor protein that is mutated in several human cancers. Our aim was to determine whether the expression pattern of KLF6 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, 85 paraffin-embedded tissues containing gastric cancer areas were cored 3 times and transferred to the recipient master block. The expression pattern of KLF6 was examined on tissue microarray slides by using immunohistochemistry and was compared with pathologic parameters, including histologic type, depth of invasion, lymph node metastasis, and peritoneal dissemination. Results: The KLF6 protein was expressed on superficial and foveolar epithelial cells in the gastric mucosa. We found loss of KLF6 expression in 28 ($32.9\%$) of the 85 gastric cancer tissues. There was a significant correlation between loss of KLF6 expression and lymph-node metastasis. However, other pathologic parameters, such as histologic type, depth of invasion, and peritoneal dissemination, were not statistically associated with loss of KLF6 expression. Conclusion: Our findings suggest that loss of KLF6 expression may contribute to abnormal regulation of gastrointestinal epithelial cell growth and differentiation and to the development and/or progression of Korean gastric cancer.

  • PDF

PU.1 Is Identified as a Novel Metastasis Suppressor in Hepatocellular Carcinoma Regulating the miR-615-5p/IGF2 Axis

  • Song, Li-Jie;Zhang, Wei-Jie;Chang, Zhi-Wei;Pan, Yan-Feng;Zong, Hong;Fan, Qing-Xia;Wang, Liu-Xing
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.9
    • /
    • pp.3667-3671
    • /
    • 2015
  • Invasion and metastasis is the major cause of tumor recurrence, difficulty for cure and low survival rate. Excavating key transcription factors, which can regulate tumor invasion and metastasis, are crucial to the development of therapeutic strategies for cancers. PU.1 is a master hematopoietic transcription factor and a vital regulator in life. Here, we report that, compared to adjacent non-cancerous tissues, expression of PU.1 mRNA in metastatic hepatocellular carcinoma (HCC), but not primary HCC, was significantly down-regulated. In addition, levels of PU.1 mRNA in metastatic hepatoma cell lines MHCC97L and MHCC97H were much lower than in non-metastatic Hep3B cells. Transwell invasion assays after PU.1 siRNA transfection showed that the invasion of hepatoma cell lines was increased markedly by PU.1 knockdown. Oppositely, overexpression of PU.1 suppressed the invasion of these cells. However, knockdown and overexpression of PU.1 did not influence proliferation. Finally, we tried to explore the potential mechanism of PU.1 suppressing hepatoma cell invasion. ChIP-qPCR analysis showed that PU.1 exhibited a high binding capacity with miR-615-5p promoter sequence. Overexpression of PU.1 caused a dramatic increase of pri-, pre- and mature miR-615-5p, as well as a marked decrease of miR-615-5p target gene IGF2. These data indicate that PU.1 inhibits invasion of human HCC through promoting miR-615-5p and suppressing IGF2. These findings improve our understanding of PU.1 regulatory roles and provided a potential target for metastatic HCC diagnosis and therapy.

Characterization of Functional Domains in NME1L Regulation of NF-κB Signaling

  • You, Dong-Joo;Park, Cho Rong;Mander, Sunam;Ahn, Curie;Seong, Jae Young;Hwang, Jong-Ik
    • Molecules and Cells
    • /
    • v.39 no.5
    • /
    • pp.403-409
    • /
    • 2016
  • NME1 is a well-known metastasis suppressor which has been reported to be downregulated in some highly aggressive cancer cells. Although most studies have focused on NME1, the NME1 gene also encodes the protein (NME1L) containing N-terminal 25 extra amino acids by alternative splicing. According to previous studies, NME1L has potent anti-metastatic activity, in comparison with NME1, by interacting with $IKK{\beta}$ and regulating its activity. In the present study, we tried to define the role of the N-terminal 25 amino acids of NME1L in $NF-{\kappa}B$ activation signaling. Unfortunately, the sequence itself did not interact with $IKK{\beta}$, suggesting that it may be not enough to constitute the functional structure. Further construction of NME1L fragments and biochemical analysis revealed that N-terminal 84 residues constitute minimal structure for homodimerization, $IKK{\beta}$ interaction and regulation of $NF-{\kappa}B$ signaling. The inhibitory effect of the fragment on cancer cell migration and $NF-{\kappa}B$-stimulated gene expression was equivalent to that of whole NME1L. The data suggest that the N-terminal 84 residues may be a core region for the anti-metastatic activity of NME1L. Based on this result, further structural analysis of the binding between NME1L and $IKK{\beta}$ may help in understanding the anti-metastatic activity of NME1L and provide direction to NME1L and $IKK{\beta}$-related anti-cancer drug design.

RECK Gene Promoter rs10814325 Polymorphism in Egyptian Patients with Hepatocellular Carcinoma on Top of Chronic Hepatitis C Viral Infection

  • Fakhry, Amal Bahgat;Ahmed, Asmaa Ismail;AbdelAlim, Mahmoud Abdo;Ramadan, Dalia Ibrahim
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.5
    • /
    • pp.2383-2388
    • /
    • 2016
  • Background: The reversion-inducing-cysteine-rich protein with Kazal motifs (RECK) gene is a novel transformation suppressor gene linked to several malignancies. Objective: To analyze any association between RECK gene rs10814325 single-nucleotide polymorphism (SNP) and HCC susceptibility with various clinicopathological and laboratory data. Materials and Methods: RECK gene rs10814325 SNP was estimated, using real-time PCR, in 30 HCC patients on top of HCV infection, 30 HCV related cirrhotic patients and 30 healthy controls. Results: No special pattern of association could be detected on comparing the RECK gene rs10814325 genotypes(P=0.5), or alleles(P=0.49) among the studied groups. HCC patients with TT genotype had younger age (mean of $54.1{\pm}6.0$ years vs $60.6{\pm}10.2$ years for TC/CC genotypes, P=0.035). Abdominal distension was significantly greater in TT genotype patients (75% vs 30%for TC/CC genotypes, P=0.045). The TT genotype was present in 75% of patients with lymph node metastasis. Serum GGT levels were higher in TT genotype patients [80 (48.5-134.8) IU/L vs 40 (33-87.5) IU/L for TC/CCgenotypes], and lower limb edema was observed in 60% for TT vs 20% for TC/CCgenotypes, but both just failed to reach significance (p=0.05 and p=0.06 respectively). Conclusions: RECK gene rs10814325 T>C could not be considered a risk factor for HCC development on top of HCV, but may be related to the disease progression and metastasis.

Overexpression of RUNX3 Inhibits Malignant Behaviour of Eca109 Cells in Vitro and Vivo

  • Chen, Hua-Xia;Wang, Shuai;Wang, Zhou;Zhang, Zhi-Ping;Shi, Shan-Shan
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.4
    • /
    • pp.1531-1537
    • /
    • 2014
  • Runt-related transcription factor 3 (RUNX3) is a tumor suppressor gene whose reduced expression may play an important role in the development and progression of esophageal squamous cell cancer (ESCC). The aim of this study was to investigate the clinical relevance of RUNX3 in ESCC patients and effects of overexpression on biological behaviour of Eca109 cells in vitro and in vivo. Immunohistochemistry was performed to detect the clinical relevance of RUNX3 and lymph node metastasis in 80 ESCC tissues and 40 non-cancerous tissues using the SP method. RT-PCR and Western blotting were applied to assess the RUNX3 level and verify the Eca109 cell line with stable overexpression. Localization of RUNX3 proteins was performed by cell immunofluorescence. CCK-8 and Scrape motility assays were used to determine proliferation and migration and the TUNEL assay to analyze cell apoptosis. Invasive potential was assessed in cell transwell invasion experiments. In nude mice, tumorigenesis in vivo was determined. Results showed decreased expression of RUNX3 in esophageal tissue to be significantly related to lymph node metastasis (LNM) (P<0.01). In addition, construction of a recombinant lentiviral vector and transfection into the human ESCC cell line Eca109 demonstrated that overexpression could inhibit cell proliferation, migration and invasion, and induce apoptosis. The in vivo experiments in mice showed tumorigenicity and invasiveness to be significantly reduced. Taken together, our studies indicate that underexpression of RUNX3 in human ESCC tissue is significantly correlated with progression. Restoration of RUNX3 expression significantly inhibits ESCC cells proliferation, migration, invasion and tumorigenesis.

Interactions between Filamin A and MMP-9 Regulate Proliferation and Invasion in Renal Cell Carcinoma

  • Sun, Guo-Gui;Wei, Cui-Da;Jing, Shao-Wu;Hu, Wan-Ning
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.8
    • /
    • pp.3789-3795
    • /
    • 2014
  • This study aimed to analyze the expression, clinical significance of filamin A (FLNA) in renal cell carcinoma (RCC) and biological effects in a cell line by regulating FLNA expression. Immunohistochemistry and Western blotting were used to analyze FLNA protein expression in 70 cases of RCC and normal tissues to study the relationship with clinical factors. FLNA lentiviral and empty vectors were transfected into RCC to study the influence of up-regulated expression of FLNA. FLNA siRNA was transiently transfected into ACHN kidney carcinoma cells by a liposome-mediated method and protein was detected by Western blotting. The level of expression was found to be significantly lower in RCC than normal tissues (p<0.05). No correlation was noted with gender, age, tumor size or pathological types (p>0.05), but links with lymph node metastasis, clinic stage and histological grade were noted (p<0.05). Loss of FLNA expression correlated significantly with poor overall survival time by Kaplan-Meier analysis (p<0.05). Results for biological function showed that ACHN cells transfected with FLNA had a lower survival fraction, significant decrease in migration and invasion, higher cell apoptosis, higher percentage of the G0/G1 phases, and lower MMP-9 protein expression compared with ACHN cells untransfected with FLNA (p<0.05). However, renal 786-0 cells transfected with FLNA siRNA had a higher survival fraction, significant increase in migration and invasion, and higher MMP-9 protein expression compared (p<0.05). In conclusion, FLNA expression was decreased in RCC and correlated significantly with lymph node metastasis, clinic stage, histological grade and poor overall survival, suggesting that FLNA may play important roles as a a tumor suppressor in RCC by promoting degradation of MMP-9.