Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Int J Mol Sci ; 23(19)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36232715

ABSTRACT

PAUF, a tumor-promoting protein secreted by cancer cells, exerts paracrine effects on immune cells through TLR4 receptors expressed on immune cell surfaces. This study aimed to investigate if PAUF elicits autocrine effects on pancreatic cancer (PC) cells through TLR4, a receptor that is overexpressed on PC cells. In this study, TLR4 expression was detected in PC cells only, but not normal pancreatic cells. The migration of TLR4 high-expressing PC cells (i.e., BxPC-3) was reduced by a selective TLR4 inhibitor, in a dose-dependent manner. Using TLR4 overexpressed and knockout PC cell lines, we observed direct PAUF-TLR4 binding on the PC cell surfaces, and that PAUF-induced cancer migration may be mediated exclusively through the TLR4 receptor. Further experiments showed that PAUF signaling was passed down through the TLR4/MyD88 pathway without the involvement of the TLR4/TRIF pathway. TLR4 knockout also downregulated PC membrane PD-L1 expression, which was not influenced by PAUF. To the best of our knowledge, TLR4 is the first receptor identified on cancer cells that mediates PAUF's migration-promoting effect. The results of this study enhanced our understanding of the mechanism of PAUF-induced tumor-promoting effects and suggests that TLR4 expression on cancer cells may be an important biomarker for anti-PAUF treatment.


Subject(s)
Intercellular Signaling Peptides and Proteins , Myeloid Differentiation Factor 88 , NF-kappa B p50 Subunit , Pancreatic Neoplasms , Toll-Like Receptor 4 , Adaptor Proteins, Vesicular Transport/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Movement/genetics , Cell Movement/physiology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lectins/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Pancreatic Neoplasms
2.
Biochem Biophys Res Commun ; 511(1): 122-128, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30771899

ABSTRACT

Although our previous studies have showed that a novel oncogene, cancer upregulated gene (CUG)2 induced epithelial-mesenchymal transition (EMT), the detailed molecular mechanism remains unknown. Because several lines of evidence documented that Yes-Associated Protein (YAP)1 is closely associated with cancer stem cell (CSC)-like phenotypes including EMT, stemness, and drug resistance, we wondered if YAP1 is involved in CUG2-induced EMT. We herein found that the overexpression of CUG2 increased YAP1 expression at the transcriptional as well as protein levels. Chromatin immunoprecipitation assay revealed that the elevated YAP1 transcripts are attributed to c-Jun and AP2 bindings to the YAP1 promoter. Akt and MAPK kinases including ERK, JNK, and p38 MAPK enhanced the level of YAP1 protein. In spite of a close relationship between ß-catenin and YAP1, not ß-catenin but NEK2 played the role in increasing YAP1 expression. Silencing YAP1 inhibited CUG2-induced cell migration and invasion. N-cadherin and vimentin expressions were decreased during YAP1 knockdown. The suppression of YAP1 diminished TGF-ß transcriptional activity and expression as well as phosphorylation level of Smad2 and Twist protein. Conversely, LY2109761 or Smad2 siRNA treatment reduced YAP1 protein levels, indicating a close interplay between YAP1 and TGF-ß signaling. Taken together, we suggest that CUG2 induces up-regulation of YAP1 expression, leading to enhancing CUG2-induced EMT via a close crosstalk between YAP1 and TGF-ß signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chromosomal Proteins, Non-Histone/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Transcription Factors/genetics , A549 Cells , Humans , Lung Neoplasms/pathology , Up-Regulation , YAP-Signaling Proteins
3.
Biochem Biophys Res Commun ; 514(4): 1278-1284, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31113615

ABSTRACT

Our previous study reported that cancer upregulated gene (CUG)2, a novel oncogene, induces both faster cell migration and anti-cancer drug resistance. We thus wonder whether CUG2 also induces stemness, a characteristic of cancer stem cells (CSCs) and further examine the molecular mechanism of this phenotype. To test that CUG2 induces stemness, we examined expression of stemness-related factors. Overexpression of CUG2 enhanced expression levels of stemness-related factors in human lung carcinoma A549 and immortalized bronchial BEAS-2B cells. Consequently, CUG2 increased cellular spherical cluster forming ability. Overexpression of CUG2 also induced tumor formation in xenotransplanted nude mice whereas transplantation of control cells failed to, implying that CUG2 possesses malignant tumorigenic potential. We paid attention to nucleophosmin (NPM1) for its known interaction with CUG2. Suppression of NPM1 hindered the CUG2-mediated stemness-like phenotypes and diminished TGF-ß transcriptional activity and signaling. TGF-ß increased stemness-like phenotypes in the control cells whereas TGF-ß inhibitor blocked induction of the phenotypes, indicating that NPM1 is required for CUG2-mediated stemness-like phenotypes through TGF-ß signaling. Furthermore, the suppression of Smad- and non-Smad-dependent TGF-ß signaling pathways also prevented CUG2 from inducing stemness-like phenotypes. Altogether, we suggest that the novel CUG2 oncogene promotes cellular transformation and stemness, mediated by nuclear NPM1 protein and TGF-ß signaling.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , A549 Cells , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nucleophosmin , Phenotype
4.
Biochem Biophys Res Commun ; 493(4): 1498-1503, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28988106

ABSTRACT

Pancreatic adenocarcinoma upregulated factor (PAUF) overexpressed in pancreatic ductal adenocarcinoma (PDAC) plays a major role in tumor progression and metastasis by autocrine and paracrine manners. However, underlying molecular mechanism of PAUF functioning in pancreatic cancer are not fully understood yet. The objective of this study was to evaluate the potential of demilune cell and parotid protein 1 (DCPP1) as a putative mouse ortholog of human PAUF by sequence alignment and functional studies. Overexpression of mouse DCPP1 in Chinese hamster ovary (CHO) cells or pancreatic cancer cells increased cell proliferation, migration, invasion, and adhesion ability in vitro. Treatment of human pancreatic cancer cells with recombinant mouse DCPP1 elevated cell growth, motility, invasiveness, and adhesiveness. Mouse DCPP1 exerted its function on pancreatic cancer cells by activating intracellular signaling pathways involved in aggressive cancer phenotype of human pancreatic cancer cells. Moreover, subcutaneous injection of mice with DCPP1-overexpressing CHO cells increased tumor sizes. Taken together, we conclude that mouse DCPP1 is a multifunctional promoter of tumor growth through functional activation of pancreatic cancer cells, suggesting it to be an ortholog of human PAUF.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Lectins/physiology , Pancreatic Neoplasms/physiopathology , Pregnancy Proteins/physiology , Animals , CHO Cells , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cricetulus , Gene Knockdown Techniques , Humans , Intercellular Signaling Peptides and Proteins , Lectins/antagonists & inhibitors , Lectins/genetics , Mice , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pregnancy Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Microenvironment , Up-Regulation
5.
Biochem Biophys Res Commun ; 461(3): 543-8, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25912875

ABSTRACT

Nucleotide excision repair (NER) is the sole mechanism of UV-induced DNA lesion repair in mammals. A single round of NER requires multiple components including seven core NER factors, xeroderma pigmentosum A-G (XPA-XPG), and many auxiliary effector proteins including ATR serine/threonine kinase. The XPA protein helps to verify DNA damage and thus plays a rate-limiting role in NER. Hence, the regulation of XPA is important for the entire NER kinetic. We found that NDR1, a novel XPA-interacting protein, modulates NER by modulating the UV-induced DNA-damage checkpoint. In quiescent cells, NDR1 localized mainly in the cytoplasm. After UV irradiation, NDR1 accumulated in the nucleus. The siRNA knockdown of NDR1 delayed the repair of UV-induced cyclobutane pyrimidine dimers in both normal cells and cancer cells. It did not, however, alter the expression levels or the chromatin association levels of the core NER factors following UV irradiation. Instead, the NDR1-depleted cells displayed reduced activity of ATR for some set of its substrates including CHK1 and p53, suggesting that NDR1 modulates NER indirectly via the ATR pathway.


Subject(s)
DNA Damage , DNA Repair , Protein Serine-Threonine Kinases/physiology , Ultraviolet Rays , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/metabolism , Gene Silencing , Humans , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering
6.
Biochem Biophys Res Commun ; 459(2): 313-318, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25727013

ABSTRACT

Pancreatic adenocarcinoma upregulated factor (PAUF), a novel oncogene, plays a crucial role in the development of pancreatic cancer, including its metastasis and proliferation. Therefore, PAUF-expressing pancreatic cancer cells could be important targets for oncolytic virus-mediated treatment. Panc-1 cells expressing PAUF (Panc-PAUF) showed relative resistance to parvovirus H-1 infection compared with Panc-1 cells expressing an empty vector (Panc-Vec). Of interest, expression of type I IFN-α receptor (IFNAR) was higher in Panc-PAUF cells than in Panc-Vec cells. Increased expression of IFNAR in turn increased the activation of Stat1 and Tyk2 in Panc-PAUF cells compared with that in Panc-Vec cells. Suppression of Tyk2 and Stat1, which are important downstream molecules for IFN-α signaling, sensitized pancreatic cancer cells to parvovirus H-1-mediated apoptosis. Further, constitutive suppression of PAUF sensitized Bxpc3 pancreatic cancer cells to parvovirus H-1 infection. Taken together, these results suggested that PAUF conferred resistance to pancreatic cancer cells against oncolytic parvovirus H-1 infection through IFNAR-mediated signaling.


Subject(s)
H-1 parvovirus , Lectins/metabolism , Oncolytic Virotherapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Receptor, Interferon alpha-beta/metabolism , Apoptosis , Cell Line, Tumor , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins , Lectins/antagonists & inhibitors , Lectins/genetics , Pancreatic Neoplasms/genetics , Parvoviridae Infections/genetics , Parvoviridae Infections/metabolism , Parvoviridae Infections/pathology , RNA, Small Interfering/genetics , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , TYK2 Kinase/antagonists & inhibitors , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
7.
Biochem Biophys Res Commun ; 454(1): 144-50, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25450371

ABSTRACT

Pancreatic adenocarcinoma up-regulated factor (PAUF) is expressed in pancreatic ductal adenocarcinoma (PDAC) and plays an important role in tumor progression and metastasis. Here we evaluate the anti-tumor efficacy of a human monoclonal antibody against PAUF, PMAb83, to provide a therapeutic intervention to treat the disease. PMAb83 reduced tumor growth and distant metastasis in orthotopically xenografted mice of human PDAC cells. PMAb83 treatment retarded proliferation along with weakened aggressiveness traits of the carcinoma cells. AKT/ß-catenin signaling played a role in the carcinoma cell proliferation and the treated xenograft tumors exhibited reduced levels of ß-catenin and cyclin D1. Moreover PMAb83 abrogated the PAUF-induced angiogenic responses of endothelial cells, reducing the density of CD31(+) vessels in the treated tumors. In combination with gemcitabine, PMAb83 conferred enhanced survival of xenografted mice by about twofold compared to gemcitabine alone. Taken together, our findings show that PMAb83 treatment decreases the aggressiveness of carcinoma cells and suppresses tumor vascularization, which culminates in mitigated tumor growth and metastasis with improved survival in PDAC mouse models.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Lectins/antagonists & inhibitors , Lectins/immunology , Pancreatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Combined Modality Therapy , Cyclin D1/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Progression , Endothelial Cells/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , beta Catenin/metabolism , Gemcitabine
8.
Cytotherapy ; 16(10): 1419-30, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24950680

ABSTRACT

BACKGROUND AIMS: Interleukin-21 (IL-21) can enhance the effector function of natural killer (NK) cells but also limits their proliferation when continuously combined with IL-2/IL-15. Paradoxically, membrane-bound (mb)-IL-21 has been shown to improve human NK cell proliferation when cultured with IL-2/mb-IL-15. To clarify the role of IL-21, we investigated the effect of the timing of IL-21 addition to NK cell culture. METHODS: IL-2/IL-15-activated NK cells were additionally treated with IL-21 according to the following schedules; (i) control (without IL-21); (ii) first week (day 0 to day 7); (iii) intermittent (the first 3 days of each week for 7 weeks); (iv) after 1 week (day 8 to day 14); and (v) continuous (day 0 to day 49). The expression of NK receptors, granzyme B, perforin, CD107a, interferon-γ, telomere length and NK cell death were measured by flow cytometry. RESULTS: Compared with the control (2004.2-fold; n = 10 healthy donors) and intermittent groups (2063.9-fold), a strong proliferative response of the NK cells on day 42 was identified in the "first week" group (3743.8-fold) (P < 0.05). NK cells treated with IL-21 in the "first week" group showed cytotoxicity similar to that in control cells. On day 28, there was a significant increase in cytotoxicity of "first week" NK cells that received IL-21 treatment for an additional 2 days compared with the "first week" NK cells (P < 0.05). CONCLUSIONS: These data suggest that controlling temporal exposure of IL-21 during NK cell proliferation can be a critical consideration to improve the yields and cytotoxicity of NK cells.


Subject(s)
Interleukins/pharmacology , Killer Cells, Natural/drug effects , Adult , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Flow Cytometry , Humans , Interferon-gamma/metabolism , Interleukin-15/pharmacology , Interleukin-2/pharmacology , K562 Cells , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Perforin/metabolism , Telomere Homeostasis/drug effects , Telomere Homeostasis/immunology , Time Factors
9.
Carcinogenesis ; 34(3): 694-702, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23222813

ABSTRACT

Collagen triple helix repeat containing-1 (CTHRC1) is a secreted protein involved in vascular remodeling, bone formation and developmental morphogenesis. CTHRC1 has recently been shown to be expressed in human cancers such as breast cancer and melanoma. In this study, we show that CTHRC1 is highly expressed in human pancreatic cancer tissues and plays a role in the progression and metastasis of the disease. CTHRC1 promoted primary tumor growth and metastatic spread of cancer cells to distant organs in orthotopic xenograft tumor mouse models. Overexpression of CTHRC1 in cancer cells resulted in increased motility and adhesiveness, whereas these cellular activities were diminished by down-regulation of the protein. CTHRC1 activated several key signaling molecules, including Src, focal adhesion kinase, paxillin, mitogen-activated protein kinase kinase (MEK), extracellular signal-regulated kinase and Rac1. Treatment with chemical inhibitors of Src, MEK or Rac1 and expression of dominant-negative Rac1 attenuated CTHRC1-induced cell migration and adhesion. Collectively, our results suggest that CTHRC1 has a role in pancreatic cancer progression and metastasis by regulating migration and adhesion activities of cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Adhesion , Cell Movement , Extracellular Matrix Proteins/physiology , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Pancreatic Ductal/secondary , Case-Control Studies , Cell Line, Tumor , Disease Progression , Enzyme Activation , Epithelium/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/pathology , Paxillin/metabolism , Signal Transduction , Up-Regulation , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
10.
Cancers (Basel) ; 15(13)2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37444482

ABSTRACT

[BACKGROUND] Collagen triple helix repeat containing-1 (CTHRC1) is a secreted protein that contributes to the progression of various cancers, including pancreatic cancer. The higher expression of CTHRC1 in tumor tissues is associated with poorer survival outcomes. However, its specific roles in tumor extracellular matrix (ECM) remodeling remain unclear. Our study aims to investigate the influences of CTHRC1 on pancreatic stellate cells (PSCs), a main source of ECM production in pancreatic cancer. [METHODS AND RESULTS] The analyses of the publicly available pancreatic cancer patient data revealed that CTHRC1 is mainly expressed in cancer stroma and highly correlated with ECM-related genes. An in vitro study showed that more than 40% of these genes can be upregulated by CTHRC1. CTHRC1 specifically activated PSC into myofibroblast-like cancer-associated fibroblasts (myCAFs), which are characterized by a significantly upregulated POSTN gene expression. Periostin (coded by the POSTN gene) has a central role in the CTHRC1-PSCs-cancer metastasis axis. Furthermore, CTHRC1 promoted pancreatic cancer cell proliferation through PSC activation to a greater extent than via direct stimulation. Proof-of-concept experiments showed that the long-term (4-week) inhibition of CTHRC1 led to significant tumor suppression and ECM reduction, and also resulted in an unexpected shift in the CAF subtype from myCAFs to inflammatory CAFs (iCAFs). [CONCLUSION] PSC activation was demonstrated to be the key molecular mechanism responsible for the tumor-promoting effects of CTHRC1, and CTHRC1 has a critical role in CAF subtype differentiation and tumor microenvironment (TME) remodeling. The inhibition of CTHRC1 as a therapeutic strategy for the treatment of pancreatic cancer warrants further investigation.

11.
Biochem Biophys Res Commun ; 429(1-2): 45-50, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23137540

ABSTRACT

We previously showed that SIRT1 deacetylase inhibits proliferation of hepatocellular carcinoma cells expressing hepatitis B virus (HBV) X protein (HBX), by destabilization of ß-catenin. Here, we report another role for SIRT1 in HBX-mediated resistance to oxidative stress. Ectopic expression and enhanced activity of SIRT1 sensitize Hep3B cells stably expressing HBX to oxidative stress-induced apoptosis. SIRT1 mutant analysis showed that nuclear localization of SIRT1 is not required for sensitization of oxidation-mediated apoptosis. Furthermore, ectopic expression of SIRT1 and treatment with resveratrol (a SIRT1 activator) attenuated JNK phosphorylation, which is a prerequisite for resistance to oxidative stress-induced apoptosis. Conversely, suppression of SIRT1 activity with nicotinamide inhibited the effect of resveratrol on JNK phosphorylation, leading to restoration of resistance to oxidation-induced apoptosis. Taken together, these results suggest that up-regulation of SIRT1 under oxidative stress may be a therapeutic strategy for treatment of hepatocellular carcinoma cells related to HBV through inhibition of JNK activation.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Oxidative Stress , Sirtuin 1/metabolism , Trans-Activators/biosynthesis , Up-Regulation , Antioxidants/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , MAP Kinase Kinase 4/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Resveratrol , Sirtuin 1/genetics , Stilbenes/pharmacology , Viral Regulatory and Accessory Proteins
12.
Biochem Biophys Res Commun ; 423(2): 270-5, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22640743

ABSTRACT

Because we found in a recent study that pancreatic adenocarcinoma up-regulated factor (PAUF), a novel oncogene, induces a rapid proliferation of pancreatic cells by up-regulation of ß-catenin, we postulated that ß-catenin might be a target molecule for pancreatic cancer treatment. We thus speculated whether SIRT1, known to target ß-catenin in a colon cancer model, suppresses ß-catenin in those pancreatic cancer cells that express PAUF (Panc-PAUF). We further evaluated whether such suppression would lead to inhibition of the proliferation of these cells. The ectopic expression of either SIRT1 or resveratrol (an activator of SIRT1) suppressed levels of ß-catenin protein and its transcriptional activity in Panc-PAUF cells. Conversely, suppression of SIRT1 expression by siRNA enhanced ß-catenin expression and transcriptional activity. SIRT1 mutant analysis showed that nuclear localization of SIRT1 is not required for reduction of ß-catenin. Treatment with MG132, a proteasomal inhibitor, restored ß-catenin protein levels, suggesting that SIRT1-mediated degradation of ß-catenin requires proteasomal activity. It was reported that inhibition of GSK-3ß or Siah-1 stabilizes ß-catenin in colon cancer cells, but suppression of GSK-3ß or Siah-1 using siRNA in the presence of resveratrol instead diminished ß-catenin protein levels in Panc-PAUF cells. This suggests that GSK-3ß and Siah-1 are not involved in SIRT1-mediated degradation of ß-catenin in the cells. Finally, activation of SIRT1 inhibited the proliferation of Panc-PAUF cells by down-regulation of cyclin-D1, a target molecule of ß-catenin. These results suggest that SIRT1 activation may be a therapeutic strategy for treatment of pancreatic cancer cells that express PAUF via the down-regulation of ß-catenin.


Subject(s)
Cell Proliferation , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Lectins/genetics , Oncogenes , Pancreatic Neoplasms/pathology , Sirtuin 1/physiology , beta Catenin/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins , Leupeptins/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Sirtuin 1/genetics
13.
Front Pharmacol ; 13: 890614, 2022.
Article in English | MEDLINE | ID: mdl-35600865

ABSTRACT

Pancreatic adenocarcinoma up-regulated factor (PAUF) plays an important role in tumor growth, metastasis, and immune evasion in the pancreatic tumor microenvironment, and recent studies suggest an association between PAUF expression and poor prognosis in ovarian cancer patients. The current study aimed 1) to characterize the potential tumor-promoting role of PAUF in ovarian cancer, using in vitro and in vivo models, including a PAUF-knockout OVCAR-5 cell line, and 2) to explore the potential therapeutic effects of an anti-PAUF antibody for ovarian cancer. Recombinant PAUF significantly increased tumor metastatic capacity (migration, invasion, and adhesion) in all the ovarian cancer cell lines tested, except for the OVCAR-5 cell line which expresses PAUF at a much higher level than the other cells. PAUF-knockout in the OVCAR-5 cell line led to apparently delayed tumor growth in vitro and in vivo. Furthermore, the administration of an anti-PAUF antibody exhibited notable sensitizing and synchronizing effects on docetaxel in mice bearing the OVCAR-5 xenograft tumors. Taken together, this study shows that the expression level of PAUF is an independent factor determining malignant behaviors of ovarian cancer and, for the first time, it suggests that PAUF may be a promising therapeutic target for high PAUF-expressing ovarian cancer.

14.
BMB Rep ; 55(2): 98-103, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35000669

ABSTRACT

Increased mRNA levels of cancer upregulated gene (CUG)2 have been detected in many different tumor tissues using Affymetrix microarray. Oncogenic capability of the CUG2 gene has been further reported. However, the mechanism by which CUG2 overexpression promotes cancer stem cell (CSC)-like phenotypes remains unknown. With recent studies showing that pyruvate kinase muscle 2 (PKM2) is overexpressed in clinical tissues from gastric, lung, and cervical cancer patients, we hypothesized that PKM2 might play an important role in CSC-like phenotypes caused by CUG2 overexpression. The present study revealed that PKM2 protein levels and translocation of PKM2 into the nucleus were enhanced in CUG2-overexpressing lung carcinoma A549 and immortalized bronchial BEAS-2B cells than in control cells. Expression levels of c-Myc, CyclinD1, and PKM2 were increased in CUG2-overexpressing cells than in control cells. Furthermore, EGFR and ERK inhibitors as well as suppression of Yap1 and NEK2 expression reduced PKM2 protein levels. Interestingly, knockdown of ß-catenin expression failed to reduce PKM2 protein levels. Furthermore, reduction of PKM2 expression with its siRNA hindered CSC-like phenotypes such as faster wound healing, aggressive transwell migration, and increased size/number of sphere formation. The introduction of mutant S37A PKM2-green fluorescence protein (GFP) into cells without ability to move to the nucleus did not confer CSC-like phenotypes, whereas forced expression of wild-type PKM2 promoted such phenotypes. Overall, CUG2-induced increase in the expression of nuclear PKM2 contributes to CSC-like phenotypes by upregulating c-Myc and CyclinD1 as a co-activator. [BMB Reports 2022;55(2): 98-103].


Subject(s)
Carrier Proteins/genetics , Chromosomal Proteins, Non-Histone , Membrane Proteins/genetics , Neoplasms , Pyruvate Kinase , Thyroid Hormones/genetics , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic , Humans , Muscle Proteins/genetics , NIMA-Related Kinases/genetics , NIMA-Related Kinases/metabolism , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Phenotype , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Signal Transduction/genetics , Thyroid Hormone-Binding Proteins
15.
Biomolecules ; 12(1)2022 01 12.
Article in English | MEDLINE | ID: mdl-35053270

ABSTRACT

Overexpression of cancer upregulated gene (CUG) 2 induces cancer stem cell-like phenotypes, such as enhanced epithelial-mesenchymal transition, sphere formation, and doxorubicin resistance. However, the precise mechanism of CUG2-induced oncogenesis remains unknown. We evaluated the effects of overexpression of CUG2 on microRNA levels using a microRNA microarray. Levels of miR-3656 were decreased when CUG2 was overexpressed; on the basis of this result, we further examined the target proteins of this microRNA. We focused on Jumonji C domain-containing protein 5 (JMJD5), as it has not been previously reported to be targeted by miR-3656. When CUG2 was overexpressed, JMJD5 expression was upregulated compared to that in control cells. A 3' untranslated region (UTR) assay revealed that an miR-3656 mimic targeted the JMJD5 3'UTR, but the miR-3656 mimic failed to target a mutant JMJD5 3'UTR, indicating that miR-3656 targets the JMJD5 transcript. Administration of the miR-3656 mimic decreased the protein levels of JMD5 according to Western blotting. Additionally, the miR-3656 mimic decreased CUG2-induced cell migration, evasion, and sphere formation and sensitized the cells to doxorubicin. Suppression of JMJD5, with its small interfering RNA, impeded CUG2-induced cancer stem cell-like phenotypes. Thus, overexpression of CUG2 decreases miR-3656 levels, leading to upregulation of JMJD5, eventually contributing to cancer stem cell-like phenotypes.


Subject(s)
MicroRNAs , Neoplasms , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Phenotype , Signal Transduction
16.
Lab Invest ; 91(11): 1652-67, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21844869

ABSTRACT

The tight junction (TJ) protein claudin-4 is aberrantly upregulated in gastric cancer, but its clinical significance and the molecular mechanisms underlying claudin-4 overexpression in gastric cancer remain unclear. Here, we investigated its roles and epigenetic mechanisms regulating CLDN4 expression in gastric cancer. We show that increased membranous expression of claudin-4 in gastric carcinoma is associated with better patient prognosis, whereas cytoplasmic claudin-4 expression did not show a significant association with prognosis. Consistent with the correlation of increased membranous claudin-4 with favorable clinicopathological factors, claudin-4 overexpression inhibited the migration and invasion of gastric cancer cells; in contrast, it did not affect cell growth. Claudin-4 expression also increased the barrier function of TJs. Claudin-4 upregulation was strongly correlated with DNA hypomethylation in both gastric tissues and gastric cancer cells. Moreover, CLDN4 expression was repressed in normal gastric tissues in association with bivalent histone modifications, and loss of repressive histone methylations and gain of active histone modifications were associated with CLDN4 overexpression in gastric cancer cells. Interestingly, CLDN4 repression could be markedly derepressed by combined treatments that simultaneously target both histone modifications and DNA demethylation in CLDN4-hypermethylated cells, whereas concomitant changes in histone methylations and acetylations are required for CLDN4 induction in CLDN4-repressed cells with low DNA methylation. Taken together, this study reveals that membranous claudin-4 expression is associated with gastric cancer progression and that it is an independent positive prognosis marker in gastric carcinoma. Furthermore, our findings suggest that epigenetic derepression may be a possible mechanism underlying CLDN4 overexpression in gastric cancer and that claudin-4 may have potential as a promising target for the treatment of gastric cancer.


Subject(s)
Carcinoma/metabolism , Claudins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/physiology , Stomach Neoplasms/metabolism , Tight Junctions/metabolism , Analysis of Variance , Cell Line, Tumor , Cell Membrane/metabolism , Chromatin Immunoprecipitation , Claudin-4 , Cytoplasm/metabolism , DNA Methylation/physiology , Flow Cytometry , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Inhibitors
17.
Biochem Biophys Res Commun ; 408(1): 160-6, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21466784

ABSTRACT

Elafin, a serine protease inhibitor, induces the intrinsic apoptotic pathway in human melanoma cells, where its expression is transcriptionally silenced. However, it remains unknown how the elafin gene is repressed in melanoma cells. We here demonstrate that elafin expression is modulated via epigenetically regulated expression of the transcription factor Foxa2. Treatment of melanoma cells with a DNA methyltransferase inhibitor induced elafin expression, which was specifically responsible for reduced proliferation and increased apoptosis. Suppression of Foxa2 transcription, mediated by DNA hypermethylation in its promoter region, was released in melanoma cells upon treatment with the demethylating agent. Luciferase reporter assays indicated that the Foxa2 binding site in the elafin promoter was critical for the activation of the promoter. Chromatin immunoprecipitation assays further showed that Foxa2 bound to the elafin promoter in vivo. Analyses of melanoma cells with varied levels of Foxa2 revealed a correlated expression between Foxa2 and elafin and the ability of Foxa2 to induce apoptosis. Our results collectively suggest that, in melanoma cells, Foxa2 expression is silenced and therefore elafin is maintained unexpressed to facilitate cell proliferation in the disease melanoma.


Subject(s)
Apoptosis/genetics , Elafin/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-beta/genetics , Melanocytes/metabolism , Melanoma/pathology , Skin Neoplasms/pathology , Cell Line, Tumor , DNA Methylation , Gene Silencing , Humans , Melanoma/genetics , Promoter Regions, Genetic , Skin Neoplasms/genetics
18.
Genes Genomics ; 43(4): 351-359, 2021 04.
Article in English | MEDLINE | ID: mdl-33555501

ABSTRACT

BACKGROUND: The renal cell carcinoma (RCC) incidences are continuously increasing, however, their proper characterization remains difficult. Mammalian kidneys require large amounts of energy, and monocarboxylate transporter (MCT) gene family is responsible for the transport of monocarboxylic compounds across plasma membranes. OBJECTIVE: A total of 14 MCT members have been identified in humans, which show highly distinct substrate affinities and tissue distributions. To understand the yet-uncharacterized renal cancer-specific role of MCTs, we identified MCT members that are differentially regulated during the renal tumor progression. METHODS: We examined the expression level of MCT members in renal cell tumors and their relationship with survival rate of patients using a public database. Quantitative RT-PCR and northern blotting were performed to validate the expression of MCTs. Anti-MCT9 antiserum was raised in rabbit and used to examine MCT9 expression in normal and tumor tissue arrays. Effect of MCT9 overexpression on cell proliferation was measured using renal cancer cell lines. RESULTS: MCT9 was found to be abundantly and exclusively expressed in human kidney cells, and was highly downregulated in renal cancers. Kaplan-Meier plotter analysis revealed an increased survival rate of MCT9 high-expressing RCC patients. MCT9 proteins were detected in normal kidney tissue sections and their overexpression clearly attenuated renal cell proliferation. CONCLUSIONS: MCT9 was identified as a novel highly downregulated gene in renal cell cancer, and its overexpression clearly attenuated RCC cell proliferation. Thus, functional analysis of MCT9 may help in deciphering a yet-undiscovered kidney-specific energy metabolism during renal tumor progression.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Monocarboxylic Acid Transporters/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Kidney/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Monocarboxylic Acid Transporters/genetics
19.
Exp Mol Med ; 53(3): 432-445, 2021 03.
Article in English | MEDLINE | ID: mdl-33731895

ABSTRACT

Cancer cachexia is a highly debilitating condition characterized by weight loss and muscle wasting that contributes significantly to the morbidity and mortality of pancreatic cancer. The factors that induce cachexia in pancreatic cancer are largely unknown. We previously showed that pancreatic adenocarcinoma upregulated factor (PAUF) secreted by pancreatic cancer cells is responsible for tumor growth and metastasis. Here, we analyzed the relation between pancreatic cancer-derived PAUF and cancer cachexia in mice and its clinical significance. Body weight loss and muscle weight loss were significantly higher in mice with Panc-1/PAUF tumors than in those with Panc-1/Mock tumors. Direct administration of rPAUF to muscle recapitulated tumor-induced atrophy, and a PAUF-neutralizing antibody abrogated tumor-induced muscle wasting in Panc-1/PAUF tumor-bearing mice. C2C12 myotubes treated with rPAUF exhibited rapid inactivation of Akt-Foxo3a signaling, resulting in Atrogin1/MAFbx upregulation, myosin heavy chain loss, and muscle atrophy. The neutrophil-to-lymphocyte ratio and body weight loss were significantly higher in pancreatic cancer patients with high PAUF expression than in those with low PAUF expression. Analysis of different pancreatic cancer datasets showed that PAUF expression was significantly higher in the pancreatic cancer group than in the nontumor group. Analysis of The Cancer Genome Atlas data found associations between high PAUF expression or a high DNA copy number and poor overall survival. Our data identified tumor-secreted circulating PAUF as a key factor of cachexia, causing muscle wasting in mice. Neutralizing PAUF may be a useful therapeutic strategy for the treatment of pancreatic cancer-induced cachexia.


Subject(s)
Adenocarcinoma/complications , Biomarkers, Tumor/metabolism , Cachexia/pathology , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Muscular Atrophy/pathology , Pancreatic Neoplasms/complications , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cachexia/etiology , Cachexia/metabolism , Cell Proliferation , Female , Humans , Male , Mice , Middle Aged , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Int J Cancer ; 127(6): 1308-20, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20020498

ABSTRACT

Expression of the protease inhibitor elafin is deregulated in several human cancers. However, functions of the protein in cancer are yet to be established. Here, we show that elafin elicits pro-apoptotic effects in melanoma cells but not in normal melanocytes. Elafin triggered the intrinsic apoptotic pathway as evidenced by the increased caspase 9 activity and unaltered caspase 8 activity. Caspase 9-specific siRNA, but not caspase 8-specific siRNA, dramatically abrogated elafin-induced apoptosis. Elevated level of p53 was observed, resulting in increased transcriptional activation and consequent expression of downstream effector molecules (Bax, Puma, Noxa, p21). Moreover, the apoptotic effect of elafin was inhibited by p53-specific siRNA and the p53 inhibitor pifithrin-alpha. Elafin treatment of xenograft mice of melanoma cells led to significantly smaller tumor sizes compared with those of untreated control mice. Immunohistochemical analysis revealed decreased elafin expression in melanoma tissue specimens. Western blot and reverse transcription analyses indicated transcriptional repression of the elafin gene in melanoma cells. Our results collectively indicate that elafin induces apoptosis in melanoma cells through a p53-dependent intrinsic apoptotic pathway, and that repression of elafin expression in melanoma may contribute to disease progression.


Subject(s)
Apoptosis/drug effects , Elafin/pharmacology , Melanoma/pathology , Protease Inhibitors/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Humans , Immunohistochemistry , Melanoma/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Small Interfering , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL