Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Yonsei Med J ; 59(5): 595-601, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29869457

ABSTRACT

PURPOSE: C-end rule (CendR) peptides are found to enhance the penetration of chemotherapeutic agents into tumor cells, while GX1 is a peptide that homes to gastric cancer (GC) vasculature. This study aimed to synthesize a novel peptide GX1-RPAKPAR (GXC) and to explore the effect of GXC on sensitizing GC cells to chemotherapeutic agents. MATERIALS AND METHODS: Intracellular Adriamycin concentration analysis was applied to conform whether GXC peptide increases the penetration of chemotherapeutic agents into GC cells in vitro. The effect of GXC peptide on sensitizing GC cells to chemotherapeutics was validated by apoptosis assay and in vitro/vivo drug sensitivity assay. The specificity of GXC to GC tissue was validated by ex vivo fluorescence imaging. RESULTS: In vitro, administration of GXC significantly increased Adriamycin concentrations inside SGC-7901 cells, and enhanced the efficacy of chemotherapeutic agents by decreasing the IC50 value. In vivo, FITC-GXC specifically accumulated in GC tissue. Moreover, systemic co-injection with GXC peptide and Adriamycin statistically improved the therapeutic efficacy in SGC-7901 xenograft models, surprisingly, without obviously increasing side effects. CONCLUSION: These results demonstrated that co-administration of the novel peptide GXC with chemotherapeutic agents may be a potential way to enhance the efficacy of anticancer drugs in GC treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Peptides/pharmacology , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Humans , Peptides/pharmacokinetics , Stomach Neoplasms/pathology
2.
Cancer Sci ; 109(4): 1044-1054, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29450946

ABSTRACT

Multidrug resistance (MDR) significantly restricts the clinical efficacy of gastric cancer (GC) chemotherapy, and it is critical to search novel targets to predict and overcome MDR. Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) has been proved to be correlated with drug resistance in several cancers. The present study revealed that LRIG1 was overexpressed in chemosensitive GC tissues and decreased expression of LRIG1 predicted poor survival in GC patients. We observed that upregulation of LRIG1 enhanced chemosensitivity in GC cells. Interestingly, miR-20a, which was overexpressed in GC MDR cell lines and tissues, was identified to regulate LRIG1 expression by directly targeting its 3' untranslated region. We also found that inhibition of miR-20a suppressed GC MDR, and upregulation showed opposite effects. Moreover, we demonstrated that the miR-20a/LRIG1 axis regulated GC cell MDR through epidermal growth factor receptor (EGFR)-mediated PI3K/AKT and MAPK/ERK signaling pathways. Finally, LRIG1 expression in human GC tissues is inversely correlated with miR-20a and EGFR. Taken together, the newly identified miR-20a/LRIG1/EGFR link provides insight into the MDR process of GC, and targeting this axis represents a novel potential therapeutic strategy to block GC chemoresistance.


Subject(s)
Down-Regulation/genetics , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Leucine/genetics , Membrane Glycoproteins/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , 3' Untranslated Regions/genetics , Cell Line, Tumor , Cell Proliferation/genetics , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Phosphatidylinositol 3-Kinases
4.
Cancer Biol Ther ; 16(8): 1241-51, 2015.
Article in English | MEDLINE | ID: mdl-26147747

ABSTRACT

Epigenetic changes play significant roles in cancer development. UHRF1, an epigenetic regulator, has been shown to be overexpressed and to coordinate tumor suppressor gene (TSG) silencing in several cancers. In a previous study, we found that UHRF1 promoted gastric cancer (GC) invasion and metastasis. However, the role and underlying mechanism of UHRF1 in GC carcinogenesis remain largely unknown. In the present study, we investigated UHRF1 expression and function in GC proliferation and explored its downstream regulatory mechanism. The results demonstrated that UHRF1 overexpression was an independent and significant predictor of GC prognosis. Downregulation of UHRF1 suppressed GC proliferation and growth in vitro and in vivo, and UHRF1 upregulation showed opposite effects. Furthermore, downregulation of UHRF1 reactivated 7 TSGs, including CDX2, CDKN2A, RUNX3, FOXO4, PPARG, BRCA1 and PML, via promoter demethylation. These results provide insight into the GC proliferation process, and suggest that targeting UHRF1 represents a new therapeutic approach to block GC development.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Genes, Tumor Suppressor , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Aged , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line, Tumor , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Promoter Regions, Genetic , Stomach Neoplasms/mortality , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays
5.
Tumour Biol ; 35(7): 6999-7007, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24748206

ABSTRACT

Previous studies have indicated that heat shock protein 27 (HSP27) had high correlation with the development and progression in several tumors. However, the roles of HSP27 in esophageal squamous cell carcinoma (ESCC) were uncertain. The aim in this study is to investigate the potential roles of HSP27 in the metastasis of ESCC. The expression of HSP27 in ESCC tissues and four human esophageal cancer cell lines were examined by immunohistochemistry and Western blotting, respectively. Wound healing assays, transwell assays, and in vivo assays were used to identify the differences of metastasis potential between normal and HSP27 overexpressed cells. HSP27 expression was downregulated in cancer tissue compared to the matched normal tissue. And the positive staining was mainly located in the cytoplasm. Statistical analyses showed that the expression of HSP27 in ESCC was significantly correlated with the tumor differentiation (P = 0.023), the patient's TNM stage (P = 0.013), lymph metastasis (P = 0.020), and distant metastasis (P = 0.017). HSP27 expression was significantly lower in highly metastatic cells than the less ones. The metastatic potentials of EC9706-H and EC109-H cells were higher than EC9706-L and EC109-L cells. In vitro and in vivo assays showed that overexpression of HSP27 in highly metastatic cells dramatically decreased their metastatic capacity. This study indicated that the expression level of HSP27 may be inversely correlated with the metastasis behavior of ESCC, and HSP27 may play an important role in this progression. HSP27 may be a potential molecular target for the therapy and prognosis of patients with ESCC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , HSP27 Heat-Shock Proteins/biosynthesis , Neoplasm Invasiveness/genetics , Aged , Animals , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Gene Expression Regulation, Neoplastic , HSP27 Heat-Shock Proteins/genetics , Humans , Lymphatic Metastasis/genetics , Male , Mice , Middle Aged , Neoplasm Staging , Prognosis
6.
FASEB J ; 27(12): 4929-39, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23982143

ABSTRACT

Epigenetic changes play significant roles in the development of cancer. UHRF1, as an epigenetic regulator, has been shown to be overexpressed and to coordinate tumor suppressor gene silencing in several cancers. However, the role and underlying mechanism of UHRF1 in gastric cancer (GC) progression remain largely unknown. In this study, we investigated the expression and function of UHRF1 in GC metastasis and explored its upstream regulatory mechanisms at the microRNA level. UHRF1 was overexpressed in GC tissues, especially in metastatic ones, and a high level of UHRF1 expression predicted poor survival. The down-regulation of UHRF1 suppressed GC invasion and metastasis in vitro and in vivo. We identified and verified miR-146a and miR-146b as direct upstream regulators of UHRF1. Furthermore, the restoration of miR-146a/b dramatically reduced the expression of UHRF1 through the direct targeting of its 3'-UTR, and this effect in turn reactivated the slit homologue 3 (Slit3), cadherin 4 (CDH4), and runt-related transcription factor 3 (RUNX3) genes via promoter demethylation. Finally, analyses of miR-146a/b and UHRF1 levels in human GC tissues revealed that miR-146a/b correlated inversely with UHRF1 expression. These findings describe a new mechanism for the regulation of UHRF1 and aberrant DNA hypermethylation in GC. The newly identified miR-146a/b/UHRF1 axis provides insight into the GC metastasis process, and targeting this novel axis represents a therapeutic approach to blocking GC metastasis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Stomach Neoplasms/metabolism , 3' Untranslated Regions , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , DNA Methylation , Down-Regulation , HEK293 Cells , Humans , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Stomach Neoplasms/pathology , Transcription, Genetic , Ubiquitin-Protein Ligases
7.
ACS Macro Lett ; 2(3): 256-259, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-35581892

ABSTRACT

A new approach to polymer cross-linking is investigated using a cascading cycloreversion of a maleimide-furan adduct and a double 1,3-dipolar cycloaddition between a sydnone and maleimide. The cross-linking proceeds quantitatively above 63 °C, despite the polymer possessing no observable glass transition temperature. The resulting polymer network possesses a high thermal stability (>300 °C) due to the irreversibility of the sydnone-maleimide cycloaddition, which releases CO2 during the cross-linking, driving the reaction. The rigid three-dimensional structure of the bis-maleimide-sydnone cycloadduct produced local free volumes in films, decreasing the dielectric constant of the material.

8.
J Phys Condens Matter ; 22(16): 165903, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-21386426

ABSTRACT

We discuss the potential functionalities of a mixed tellurite-chalcogenide glass where alternate oxygen atoms in a TeO(2) vitreous network are substituted with either sulfur or selenium atoms. We calculate the microscopic linear and nonlinear polarizabilities of chains with a finite number (n) of TeOX (X = S or Se) units from ab initio quantum chemistry computations and apply rotational averaging to the results. We find that the rotationally averaged linear polarizability, α(av), increases linearly with the number of units, while the second hyper-polarizability, γ(av), grows as a power law with an exponent that changes from 1.2 to 1.9 and 2.4 when going from pure tellurite to S substitution and Se substitution, respectively. We then estimate the density of the resulting glass and local field corrections to obtain the macroscopic linear and nonlinear susceptibilities for a glass made from chains of n = 5. We find that the extra Te-S (or Te-Se) bonds in the mixed tellurite-chalcogenide system enhance the linear refractive index n(0) and the third-order susceptibility χ((3)) by factors of ∼ 1.3 (or 1.4) and ∼ 8 (or 14), respectively, over the base tellurite glasses (X = O).

SELECTION OF CITATIONS
SEARCH DETAIL