Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Mol Med (Berl) ; 98(5): 761-773, 2020 05.
Article in English | MEDLINE | ID: mdl-32318747

ABSTRACT

Tight junction protein 1 (TJP1) is a membrane-associated cytosolic protein important for cell-cell communication in intercellular barriers in epithelial and non-epithelial cells. Here, we explored the functional involvement of TJP1 in non-epithelial tumors such as soft tissue sarcoma, especially in leiomyosarcoma (LMS). TJP1 expression in soft tissue sarcoma was analyzed in normal and tumor tissues as well as from public datasets such as the TCGA provisional dataset, in which TJP1 expression was compared with other subtypes such as undifferentiated sarcomas, and myxofibrosarcomas. SK-LMS-1 cell lines with reduced TJP1 expression showed attenuated anchorage-independent colony formation as well as reduced intercellular aggregation on non-coated culture plates compared with control as well as parental SK-LMS-1 cells. Transcriptome profiling following TJP1 knockdown in SK-LMS-1 cells suggested the involvement of several signaling pathways, including NF-κB pathway and growth factor receptor signaling. In addition, TJP1 downregulation induced enhanced response against anti-cancer agents, doxorubicin and gefitinib. Taken together, these results suggest that TJP1 contributes to sarcoma genesis and might be useful therapeutic target. KEY MESSAGES: • TJP1 expression at RNA level higher in tumor than in normal tissues of sarcoma. • Targeting TJP1 attenuates cell-cell aggregation and anchorage-independent growth. • Targeting TJP1 is beneficial in anti-cancer therapy in LMS.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Communication/drug effects , Cell Communication/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Leiomyosarcoma/genetics , Zonula Occludens-1 Protein/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Gene Editing , Gene Expression Profiling , Gene Silencing , Humans , Janus Kinases/metabolism , Leiomyosarcoma/drug therapy , Leiomyosarcoma/metabolism , Leiomyosarcoma/pathology , NF-kappa B/metabolism , RNA, Small Interfering/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Transcriptome , Zonula Occludens-1 Protein/antagonists & inhibitors
2.
J Clin Med ; 8(8)2019 08 06.
Article in English | MEDLINE | ID: mdl-31390831

ABSTRACT

Corneal dystrophies (CDs) are a diverse group of inherited disorders with a heterogeneous genetic background. Here, we report the identification of a novel ZNF143 heterozygous missense mutation in three individuals of the same family with clinical and pathological features that are consistent with endothelial CD. Ophthalmologic examination revealed diffuse corneal clouding and edema with decreased endothelial cell density. Pathological findings showed increased corneal thickness due to edema of basal epithelial cells and stroma, and abnormal metaplastic endothelium with stratified epithelium-like changes. Patients' metaplastic corneal endothelial cells expressed predominantly cytokerain 7, cytokeratin 19, and E-cadherin. Although Sanger sequencing did not detect any mutation associated with endothelial CDs, whole exome sequencing identified the ZNF143 c.937G>C p.(Asp313His) mutation as a candidate gene for our patients' endothelial CD. In-vitro functional studies demonstrated that mutant ZNF143 promoted the mesenchymal-to-epithelial transition; it upregulated the expression of genes associated with epithelialization in human corneal endothelial cells. Additionally, proinflammatory cytokine responsive genes were significantly enriched after mutant ZNF143 transfection, which may contribute to the severe phenotype of the three patients. These findings link a mutation in ZNF143 with endothelial CD for the first time.

3.
Cells ; 8(4)2019 03 30.
Article in English | MEDLINE | ID: mdl-30935019

ABSTRACT

Autophagy is a cellular process that disrupts and uses unnecessary or malfunctioning components for cellular homeostasis. Evidence has shown a role for autophagy in tumor cell survival, but the molecular determinants that define sensitivity against autophagic regulation in cancers are not clear. Importantly, we found that breast cancer cells with low expression levels of a zinc-finger protein, ZNF143 (MCF7 sh-ZNF143), showed better survival than control cells (MCF7 sh-Control) under starvation, which was compromised with chloroquine, an autophagy inhibitor. In addition, there were more autophagic vesicles in MCF7 sh-ZNF143 cells than in MCF7 sh-Control cells, and proteins related with the autophagic process, such as Beclin1, p62, and ATGs, were altered in cells with less ZNF143. ZNF143 knockdown affected the stability of p53, which showed a dependence on MG132, a proteasome inhibitor. Data from proteome profiling in breast cancer cells with less ZNF143 suggest a role of NAD(P)H quinone dehydrogenase 1(NQO1) for p53 stability. Taken together, we showed that a subset of breast cancer cells with low expression of ZNF143 might exhibit better survival via an autophagic process by regulating the p53⁻Beclin1 axis, corroborating the necessity of blocking autophagy for the best therapy.


Subject(s)
Beclin-1/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Signal Transduction , Stress, Physiological , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis , Autophagy , Breast Neoplasms/ultrastructure , Cell Line, Tumor , Cell Survival , Disease-Free Survival , Female , Humans , Protein Stability , Vacuoles/metabolism , Vacuoles/ultrastructure
4.
J Cell Mol Med ; 23(6): 4043-4053, 2019 06.
Article in English | MEDLINE | ID: mdl-30933430

ABSTRACT

Several studies have shown that expression of zinc-finger protein 143 (ZNF143) is closely related to tumour progression including colon cancer. However, it remains unclear how ZNF143 expression is related to tumour progression within the tumour microenvironment. Here, we investigated whether ZNF143 expression affects the tumour microenvironment and tumour progression by screening molecules secreted by colon cancer cells stably expressing short-hairpin RNAs against ZNF143 or control RNAs. We observed that secretion of interleukin (IL)-8 was increased when ZNF143 expression was reduced in two colon cancer cell lines. The mRNA and protein levels of IL-8 were increased in cells following ZNF143 knockdown, and this effect was reversed when ZNF143 expression was restored. The Janus tyrosine kinase/signal transducer and activator of transcription (JAK/STAT) and extracellular signal-regulated kinase pathways were also shown to contribute to IL-8 expression in ZNF143-knockdown cells. The expression levels of ZNF143 and IL-8 were inversely correlated with three-dimensionally grown spheroids and colon cancer tissues. THP-1 cells were differentiated when cells were incubated with condition media from colon cancer cell with less ZNF143, drastically. Loss of ZNF143 may contribute to the development of colon cancer by regulating intracellular and intercellular signalling for cell plasticity and the tumour microenvironment respectively.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Interleukin-8/genetics , Trans-Activators/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Disease Progression , Extracellular Signal-Regulated MAP Kinases/genetics , HCT116 Cells , HT29 Cells , Humans , Transcription, Genetic/genetics , Tumor Microenvironment/genetics
5.
BMB Rep ; 50(12): 621-627, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29065970

ABSTRACT

We previously reported the involvement of zinc-finger protein 143 (ZNF143) on cancer cell motility in colon cancer cells. Here, ZNF143 was further characterized in breast cancer. Immunohistochemistry was used to determine the expression of ZNF143 in normal tissues and in tissues from metastatic breast cancer at various stages. Notably, ZNF143 was selectively expressed in duct and gland epithelium of normal breast tissues, which decreased when the tissue became malignant. To determine the molecular mechanism how ZNF143 affects breast cancer progression, it was knocked down by infecting benign breast cancer cells with shorthairpin (sh) RNA-lentiviral particles against ZNF143 (MCF7 sh-ZNF143). MCF7 sh-ZNF143 cells showed different cell-cell contacts and actin filament (F-actin) structures when compared with MCF7 sh-Control cells. In migration and invasion assays, ZNF143 knockdown induced increased cellular motility in breast carcinoma cells. This was reduced by the recovery of ZNF143 expression. Taken together, these results suggest that ZNF143 expression contributes to breast cancer progression. [BMB Reports 2017; 50(12): 621-627].


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Trans-Activators/metabolism , Cell Movement/genetics , Female , Humans , MCF-7 Cells , Real-Time Polymerase Chain Reaction , Trans-Activators/genetics
6.
Eur J Pharmacol ; 769: 257-65, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26607468

ABSTRACT

The proto-oncogene c-Myc has been implicated in a variety of cellular processes, such as proliferation, differentiation and apoptosis. Several c-Myc targets have been studied; however, selective regulation of c-Myc is not easy in cancer cells. Herein, we attempt to identify chemical compounds that induce cell death in c-Myc-overexpressing cells (STF-cMyc and STF-Control) by conducting MTS assays on approximately 4000 chemical compounds. One compound, C604, induced cell death in STF-cMyc cells but not STF-Control cells. Apoptotic proteins, including caspase-3 and poly(ADP-ribose) polymerase (PARP), were cleaved in C604-treated STF-cMyc cells. In addition, SW620, HCT116 and NCI-H23 cells, which exhibit higher basal levels of c-Myc, underwent apoptotic cell death in response to C604, suggesting a role for C604 as an inducer of apoptosis in cancer cells with c-Myc amplification. C604 induced cell cycle arrest at the G2/M phase in cells, which was not affected by apoptotic inhibitors. Interestingly, C604 induced accumulation of c-Myc and Cdc25A proteins. In summary, a chemical compound was identified that may induce cell death in cancer cells with c-Myc amplification specifically through an apoptotic pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Proto-Oncogene Proteins c-myc/genetics , Pyrimidines/pharmacology , Thiazoles/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression , Humans , M Phase Cell Cycle Checkpoints/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Mas
7.
BMB Rep ; 48(2): 115-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25027596

ABSTRACT

Tight junction protein 1 (TJP1), a component of tight junction, has been reported to play a role in protein networks as an adaptor protein, and TJP1 expression is altered during tumor development. Here, we found that TJP1 expression was increased at the RNA and protein levels in TGF-ß-stimulated lung cancer cells, A549. SB431542, a type-I TGF-ß receptor inhibitor, as well as SB203580, a p38 kinase inhibitor, significantly abrogated the effect of TGF-ß on TJP1 expression. Diphenyleneiodonium, an NADPH oxidase inhibitor, also attenuated TJP1 expression in response to TGF-ß in lung cancer cells. When TJP1 expression was reduced by shRNA lentiviral particles in A549 cells (A549-sh TJP1), wound healing was much lower than in cells infected with control viral particles. Taken together, these data suggest that TGF-ß enhances TJP1 expression, which may play a role beyond structural support in tight junctions during cancer development.


Subject(s)
Gene Expression Regulation/drug effects , Transforming Growth Factor beta/pharmacology , Zonula Occludens-1 Protein/metabolism , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Dioxoles/pharmacology , Humans , Imidazoles/pharmacology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Zonula Occludens-1 Protein/antagonists & inhibitors , Zonula Occludens-1 Protein/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Mol Carcinog ; 53 Suppl 1: E161-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24009065

ABSTRACT

To investigate the role of zinc-finger protein 143 in cancer cells, we stably introduced ZNF143 expression knockdown by infecting colon cancer cells with short hairpin (sh) RNA-lentiviral particles against ZNF143 (HCT116 sh-ZNF143). Compared to sh-control cells, HCT116 sh-ZNF143 cells showed faster wound healing, increased migration through Transwell chambers, and increased invasion through Matrigel in Transwell chambers. ZNF143 knockdown increased transcriptional expression of ZEB1. Additionally, ZNF143 regulated E-cadherin transcriptional expression. Small interfering-RNA-mediated silencing of ZEB1 expression affected motility in HCT116 sh-ZNF143 cells. These data suggest that ZNF143 is involved in cellular motility through a ZEB1-E-cadherin-linked pathway in colon cancer cells.


Subject(s)
Cadherins/metabolism , Cell Movement , Colonic Neoplasms/pathology , Homeodomain Proteins/metabolism , Trans-Activators/physiology , Transcription Factors/metabolism , Apoptosis , Blotting, Western , Cadherins/genetics , Cell Adhesion , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Flow Cytometry , Homeodomain Proteins/genetics , Humans , Immunoenzyme Techniques , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factors/genetics , Tumor Cells, Cultured , Wound Healing , Zinc Finger E-box-Binding Homeobox 1
9.
Mol Cells ; 32(5): 415-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21909943

ABSTRACT

Previously, we reported that the expression of zinc-finger protein 143 (ZNF143) was induced by insulin-like growth factor-1 (IGF-1) via reactive oxygen species (ROS)- and phosphatidylinositide-3-kinase (PI3-kinase)-linked pathways in colon cancer cells. Here, we investigated whether GAIP-interacting protein, C-terminus (GIPC), a binding partner of IGF-1R, is involved in ZNF143 expression through IGF-1 and IGF-1R signaling in colon cancer cells. The knockdown of GIPC in colon cancer cells reduced ZNF143 expression in response to IGF-1. IGF-1 signaling through its receptor, leading to the phosphorylation and activation of the PI3-kinase-Akt pathway and mitogenactivated protein kinases (MAPKs) was unaffected by the knockdown of GIPC, indicating the independence of the GIPC-linked pathway from PI3-kinase- and MAPK-linked signaling in IGF-1-induced ZNF143 expression. In accordance with previous results in breast cancer cells (Choi et al., 2010), the knockdown of GIPC reduced ROS production in response to IGF-1 in colon cancer cells. Furthermore, the knockdown of GIPC reduced the expression of Rad51, which is regulated by ZNF143, in response to IGF-1 in colon cancer cells. Taken together, these data suggest that GIPC is involved in IGF-1 signaling leading to ZNF143 expression through the regulation of ROS production, which may play a role for colon cancer tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colonic Neoplasms/metabolism , Insulin-Like Growth Factor I/metabolism , Trans-Activators/biosynthesis , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , HCT116 Cells , Humans , MAP Kinase Kinase 4/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rad51 Recombinase/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction , Trans-Activators/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Exp Mol Med ; 42(10): 696-702, 2010 Oct 31.
Article in English | MEDLINE | ID: mdl-20733350

ABSTRACT

Expression of zinc-finger protein 143 (ZNF143), a human homolog of the Xenopus transcriptional activator protein Staf, is induced by various DNA-damaging agents including etoposide, doxorubicin, and gamma-irradiation. ZNF143 binds to cisplatin-modified DNA, and its levels are increased in cancer cells that are resistant to anticancer drugs, including cisplatin, suggesting that it plays a role in carcinogenesis and cancer cell survival. However, the mechanism of ZNF143 induction in cancer cells remains unclear. Both insulin-like growth factor-1 (IGF-1) and its receptor (IGF-1R) have been reported to be overexpressed in cancer cells and to be related to anticancer drug resistance, but the identity of the relevant signaling mediators is still being investigated. In the present study, we observed that IGF-1 was able to induce ZNF143 expression in HCT116 human colon cancer cells and that wortmannin, an inhibitor of phosphatidylinositide 3- kinase (PI3-kinase), inhibited this induction, as did diphenyleneiodonium (DPI), an NADPH oxidase inhibitor, and monodansylcardavarine (MDC), a receptor internalization inhibitor. Treatment with MDC decreased the IGF-1-stimulated generation of reactive oxygen species. Taken together, these data suggest that IGF-1 induces ZNF143 expression in cancer cells via PI3-kinase and reactive oxygen species generation during receptor internalization.


Subject(s)
Colonic Neoplasms/metabolism , Insulin-Like Growth Factor I/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Reactive Oxygen Species/metabolism , Trans-Activators/biosynthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , HCT116 Cells , Humans , Signal Transduction , Trans-Activators/genetics
11.
Cancer Lett ; 294(2): 254-63, 2010 Aug 28.
Article in English | MEDLINE | ID: mdl-20206441

ABSTRACT

Insulin-like growth factor-1 (IGF-1)/IGF-1 receptor signaling participates in a variety of cellular processes, including cell survival, growth, and proliferation. Increased expression of IGF-1R and activation of its downstream signaling components have been implicated in human cancers. Although a regulatory role for IGF-1R has been established, the relationship between IGF-1R and its binding partner, GAIP-interacting protein C-terminus (GIPC), in terms of promoting cell proliferation, remains unclear. We found that siRNA-mediated silencing of GIPC expression decreased IGF-1-mediated IGF-1R phosphorylation and cellular proliferation in breast cancer models. IGF-1-mediated cellular proliferation was also inhibited by N-acetylcysteine, which implicates reactive oxygen species generation. siRNA-mediated silencing of GIPC expression also decreased IGF-1-mediated reactive oxygen species generation. Taken together, these data suggest that GIPC contributes to IGF-1-induced cancer cell proliferation via the regulation of reactive oxygen species production.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Insulin-Like Growth Factor I/metabolism , Reactive Oxygen Species/metabolism , Receptor, IGF Type 1/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Insulin-Like Growth Factor I/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptor, IGF Type 1/antagonists & inhibitors , Signal Transduction/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...