Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
BMB Rep ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38649146

ABSTRACT

The aim of the present study is to provide a rational background for silencing the V-set and transmembrane domain containing 2 like (VSTM2L) in consort with recognising soluble VSTM2L against cholangiocarcinoma. A therapeutic target against cholangiocarcinoma was selected using iterative patient partitioning (IPP) calculation, and it was verified by in vitro and in silico analyses. VSTM2L was selected as a potential therapeutic target against cholangiocarcinoma. Silencing the VSTM2L expression significantly attenuated the viability and survival of cholangiocarcinoma cells through blockade of the intracellular signalling pathway. In silico analysis showed that VSTM2L affected the positive regulation of cell growth in cholangiocarcinoma. Liptak's z value revealed that the expression of VSTM2L worsened the prognosis of cholangiocarcinoma patients. In addition, soluble VSTM2L was significantly detected in the whole blood of cholangiocarcinoma patients compared with that of healthy donors. Our report reveals that VSTM2L might be the potential therapeutic target and a soluble prognostic biomarker against cholangiocarcinoma.

3.
J Med Food ; 26(8): 595-604, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37594560

ABSTRACT

Acorn (Quercus acutissima CARR.) has been used in traditional food and medicinal ethnopharmacology in Asia, and it has shown multifarious functions such as antidementia, antiobesity, and antiasthma functions. However, there is limited scientific evidence about the efficacy of acorn for ameliorating skin problems. Treatment with ethanol-extracted acorns (EeA's) ablated the expression of inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX2), monocyte chemoattractant protein-1 (MCP-1), and interleukin (IL)-8 stimulated by tumor necrosis factor (TNF)-α in human adult low calcium high temperature (HaCaT) cells under sublethal dosages. In addition, treatment with EeA dose dependently inhibited the ex vivo hyper keratin formation induced by TNF-α in HaCaT cells in conjunction with the blockade of cytokeratin-1 (CK-1) and cytokeratin-5 (CK-5) expression. Moreover, EeA treatment stimulated the expression of hyaluronic acid (HA) expression in human fibroblasts in a dose-dependent manner. Linoleamide was identified as the functional component of EeA using preparative high-performance liquid chromatography and ultra high performance liquid chromatography-mass spectrometry-mass spectrometry analysis, and the anti-inflammatory features and enhanced HA expression were verified. Collectively, these results suggest the efficacy of EeA supplementation in improving skin problems via anti-inflammation and upregulating HA production.


Subject(s)
Hyaluronic Acid , Quercus , Adult , Humans , Keratinocytes , HaCaT Cells , Ethanol
4.
BMB Rep ; 56(11): 594-599, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37357538

ABSTRACT

A number of therapeutic drugs have been developed from functional chemicals found in plants. Knowledge of plants used for medicinal purposes has historically been transmitted by word of mouth or through literature. The aim of the present study is to provide a systemic platform for the development of lead compounds against breast cancer based on a traditional medical text. To verify our systematic approach, integrating processes consisted of text mining of traditional medical texts, 3-D virtual docking screening, and in vitro and in vivo experimental validations were demonstrated. Our text analysis system identified rutin as a specific phytochemical traditionally used for cancer treatment. 3-D virtual screening predicted that rutin could block EGFR signaling. Thus, we validated significant anticancer effects of rutin against breast cancer cells through blockade of EGFR signaling pathway in vitro. We also demonstrated in vivo anti-cancer effects of rutin using the breast cancer recurrence in vivo models. In summary, our innovative approach might be proper for discovering new phytochemical lead compounds designing for blockade of malignant neoplasm including breast cancer. [BMB Reports 2023; 56(11): 594-599].


Subject(s)
Breast Neoplasms , Plants, Medicinal , Humans , Female , Breast Neoplasms/drug therapy , Plants, Medicinal/chemistry , Phytochemicals , Signal Transduction , ErbB Receptors
5.
Cell Mol Life Sci ; 79(10): 537, 2022 Oct 02.
Article in English | MEDLINE | ID: mdl-36183290

ABSTRACT

Contact-based pericellular interactions play important roles in cancer progression via juxtacrine signaling pathways. The present study revealed that hypoxia-inducible factor-1α (HIF-1α), induced even in non-hypoxic conditions by cell-to-cell contact, was a critical cue responsible for the malignant characteristics of glioblastoma multiforme (GBM) cells through Notch1 signaling. Densely cultured GBM cells showed enhanced viability and resistance to temozolomide (TMZ) compared to GBM cells at a low density. Ablating Notch1 signaling by a γ-secretase inhibitor or siRNA transfection resensitized resistant GBM cells to TMZ treatment and decreased their viability under dense culture conditions. The expression of HIF-1α was significantly elevated in highly dense GBM cells even under non-hypoxic conditions. Atypical HIF-1α expression was associated with the Notch1 signaling pathway in both GBM and glioblastoma stem cells (GSC). Proteasomal degradation of HIF-1α was prevented by binding with Notch1 intracellular domain (NICD), which translocated to the nuclei of GBM cells. Silencing Notch1 signaling using a doxycycline-inducible Notch1 RNA-interfering system or treatment with chetomin, a HIF pathway inhibitor, retarded tumor development with a significant anti-cancer effect in a murine U251-xenograft model. Using GBM patient tissue microarray analysis, a significant increase in HIF-1α expression was identified in the group with Notch1 expression compared to the group without Notch1 expression among those with positive HIF-1α expression. Collectively, these findings highlight the critical role of cell-to-cell contact-dependent signaling in GBM progression. They provide a rationale for targeting HIF-1α signaling even in a non-hypoxic microenvironment.


Subject(s)
Glioblastoma , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Doxycycline , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , Signal Transduction , Temozolomide , Tumor Microenvironment
6.
Int J Oncol ; 61(4)2022 10.
Article in English | MEDLINE | ID: mdl-36043525

ABSTRACT

Despite the high expression of neuropilin­1 (NRP­1) in human glioblastoma (GB), the understanding of its function as a co­receptor of vascular endothelial growth factor receptors (VEGFRs) in angiogenesis is currently limited. Therefore, the aim of the present study was to elucidate the non­classical function of NRP­1 expression in human GB. Expression patterns of NRP­1 and VEGF­A were determined by sandwich ELISA, western blot analysis, or immunohistochemistry. Differential dependency of GB cells following ablation of VEGF­A signaling was validated in vitro and in vivo. Cellular mechanism responsible for distinct response to VEGF­A signaling was evaluated by western blotting and immunoprecipitation analysis. Prognostic implications were assessed using IHC analysis. GB cells exhibited differing sensitivity to silencing of vascular endothelial growth factor (VEGF)­A signaling, which resulted in a distinct expression pattern of wild­type or chondroitin­sulfated NRP­1. VEGF­A­sensitive GB exhibited the physical interaction between wild­type NRP­1 and FMS related receptor tyrosine kinase 1 (Flt­1) whereas VEGF­A­resistant GB exhibited chondroitin­sulfated NRP­1 without interaction with Flt­1. Eliminating the chondroitin sulfate modification in NRP­1 led to re­sensitization to VEGF­A signaling, and chondroitin sulfate modification was found to be associated with an adverse prognosis in patients with GB. The present study identified the distinct functions of NRP­1 in VEGF­A signaling in accordance with its unique expression type and interaction with Flt­1. The present research is expected to provide a strong basis for targeting VEGF­A signaling in patients with GB, with variable responses.


Subject(s)
Glioblastoma , Neuropilin-1 , Vascular Endothelial Growth Factor A , Chondroitin Sulfates , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Neuropilin-1/genetics , Neuropilin-1/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Anticancer Res ; 42(8): 3789-3798, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35896263

ABSTRACT

BACKGROUND/AIM: Cholangiocarcinoma remains one of the most dangerous types of cancer. Eriodictyol is a well-known flavonoid having effective bioactivity against various malignant tumor types. However, the anticancer effect of eriodictyol against cholangiocarcinoma remains ambiguous. Thus, the aim of the present study was to investigate the effects of eriodictyol on human cholangiocarcinoma. MATERIALS AND METHODS: The biological effects of eriodictyol were validated by viability assay, colony formation and western blot analysis. The significance of heme oxygenase 1 (HMOX1) expression in cholangio-carcinoma was demonstrated using bioinformatics analysis and knockdown of HMOX1 by transfection with short interfering (si)-RNA. RESULTS: Eriodictyol highly reduced the in vitro viability of SNU-308, SNU-478, SNU-1079, and SNU-1196 cholangiocarcinoma cells compared with that of 293T cells, in a dose-dependent manner. The anticancer effect of eriodictyol was achieved by caspase-3-mediated apoptosis. In particular, eriodictyol increased HMOX1 expression, which resulted in attenuation of cholangiocarcinoma cell proliferation. In contrast, ablating HMOX1 expression by si-RNA transfection against HMOX1 made cholangiocarcinoma cells insensitive to the antiproliferative effect of eriodictyol treatment. CONCLUSION: These results collectively indicate that eriodictyol acts as an anticancer agent via regulation of HMOX1 expression against human cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/metabolism , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Flavanones , Heme Oxygenase-1/genetics , Humans , RNA, Small Interfering/genetics
8.
Cell Oncol (Dordr) ; 45(2): 275-291, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35357655

ABSTRACT

BACKGROUND: Cholangiocarcinoma is a devastating cancer with a poor prognosis. Previous reports have presented conflicting results on the role of transforming growth factor-ß-induced protein (TGFBI) in malignant cancers. Currently, our understanding of the role of TGFBI in cholangiocarcinoma is ambiguous. The aim of the present study was to investigate the role of TGFBI in human cholangiocarcinoma. METHODS: Iterative patient partitioning (IPP) scoring and consecutive elimination methods were used to select prognostic biomarkers. mRNA and protein expression levels were determined using Gene Expression Omnibus (GEO), Western blot and ELISA analyses. Biological activities of selected biomarkers were examined using both in vitro and in vivo assays. Prognostic values were assessed using Kaplan-Meier and Liptak's z score analyses. RESULTS: TGFBI was selected as a candidate cholangiocarcinoma biomarker. GEO database analysis revealed significantly higher TGFBI mRNA expression levels in cholangiocarcinoma tissues compared to matched normal tissues. TGFBI protein was specifically detected in a soluble form in vitro and in vivo. TGFBI silencing evoked significant anti-cancer effects in vitro. Soluble TGFBI treatment aggravated the malignancy of cholangiocarcinoma cells both in vitro and in vivo through activation of the integrin beta-1 (ITGB1) dependent PPARγ signalling pathway. High TGFBI expression was associated with a poor prognosis in patients with cholangiocarcinoma. CONCLUSIONS: Our data suggest that TGFBI may serve as a promising prognostic biomarker and therapeutic target for cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cholangiocarcinoma/genetics , Extracellular Matrix Proteins , Humans , Integrin beta1 , PPAR gamma , RNA, Messenger/genetics , Transforming Growth Factor beta/metabolism
9.
Biochem Biophys Res Commun ; 599: 100-105, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35180468

ABSTRACT

BACKGROUND: Luffa cylindrica stem sap (LuCS) has been ethnopharmacologically used as a cosmetic ingredients to improve the facial condition in Asians, but there is no scientific proof about the advantages of LuCS as a supplement for skin elasticity inducer. PURPOSE: Presently, we have validated the beneficial effect of LuCS in human preadipocyte and fibroblast. METHODS: In vitro activities of LuCS on expression of cellular elastin and collagen type I were validated using Western blot analysis in human fibroblasts. Effect of LuCS on preadipocyte development was performed using MDI medium containing isobutyl-methylxanthine, dexamethasone, and insulin and then evaluated using oil red O staining. RESULTS: Treatment of LuCS stimulated the expression of cellular elastin and type I procollagen in human skin fibroblasts. Exposure to LuCS induced lipid accumulation of preadipocytes via activation of CEBP/α signaling pathway in preadipocytes. Expression of collagen I, elastin, or CEBP/α mRNA was decreased by age. 3-bromo-3-methylisoxazol-5-amine enhanced the synthesis of cellular lipid in preadipocytes. CONCLUSIONS: Collectively, these results suggest the rationale of LuCS treatment in enhancing the skin condition.


Subject(s)
Adipocytes/drug effects , Fibroblasts/drug effects , Luffa/chemistry , Plant Extracts/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Drug Evaluation, Preclinical/methods , Elastin/genetics , Elastin/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Lipid Metabolism/drug effects , Mice , Plant Extracts/chemistry , Procollagen/genetics , Procollagen/metabolism
10.
Biochem Biophys Res Commun ; 580: 41-47, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34619551

ABSTRACT

Luffa cylindrica stem sap (LuCS) has been traditionally used as a facial cosmetic supplement to enhance the skin condition of Asians. However, LuCS has yet to be described and there is no solid scientific evidence regarding the use of LuCS as an anti-wrinkle agent. In the present study, we have evaluated the functional effect of LuCS and its underlying mechanisms based on scientific evidence. Treatment with LuCS stimulated the growth and migration of human skin fibroblasts. LuCS treatment activated EGFR signaling via the enhanced expression of EGFR and down-regulation of PPARγ in human skin fibroblasts. Exposure to LuCS induced the synthesis of cellular type I procollagen and elastin in consort with the down-regulation of various proteinases including MMP-1, -2 and -9 in human skin fibroblasts. LuCS treatment also reversed the skin damage induced by UV-A irradiation in human skin fibroblasts. 3-bromo-3-methylisoxazol-5-amine was identified as the functional component using UPLC-MS-MS analysis and increased production of cellular type I procollagen. Collectively, these results suggest the efficacy of LuCS supplementation in improving the skin condition via anti-wrinkle effect.


Subject(s)
Fibroblasts/drug effects , Luffa , Plant Extracts/pharmacology , Protective Agents/pharmacology , Skin Aging/drug effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Fibroblasts/cytology , Fibroblasts/radiation effects , Humans , Luffa/chemistry , Plant Extracts/chemistry , Plant Stems/chemistry , Protective Agents/chemistry
11.
Cell Oncol (Dordr) ; 43(5): 821-834, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32483746

ABSTRACT

PURPOSE: Pancreatic cancer is one of the most aggressive cancers. Preclinical and clinical data indicate that Notch 1 ligand jagged1 (JAG1) plays a pro-oncogenic role in several malignant cancers. As yet, however, the role of JAG1 in pancreatic cancer is poorly understood. The objective of the present study was to investigate JAG1 as a therapeutic target in human pancreatic cancer. METHODS: Expression levels of Notch signaling molecules were assessed using GEO datasets and Western blot analysis, respectively. Anti-tumor effects following JAG1 silencing were evaluated using in vitro and in vivo assays. Prognostic implications were assessed using GEO datasets. RESULTS: Using GEO datasets and Western blot analysis we detected significantly higher JAG1 mRNA and protein expression levels in pancreatic cancer compared to normal pancreatic tissues. JAG1 silencing significantly restrained the growth, migration and invasion of pancreatic cancer cells through the induction of apoptosis and blockade of various kinases independent of the Notch1 pathway. Combined JAG1 silencing and gemcitabine treatment showed synergistic anti-viability effects in human pancreatic cancer cells. JAG1 silencing also resulted in significant anti-cancer effects in vivo and high JAG1 expression was found to be associated with an adverse prognosis in pancreatic cancer patients. CONCLUSIONS: From our data we conclude that JAG1 may be a promising therapeutic target in pancreatic cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Jagged-1 Protein/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Jagged-1 Protein/antagonists & inhibitors , Jagged-1 Protein/metabolism , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/drug therapy , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Gemcitabine
12.
Anticancer Res ; 40(3): 1335-1344, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32132030

ABSTRACT

BACKGROUND/AIM: The prognosis of pancreatic cancer has not improved due to its migratory feature and refractory potential to chemo-resistance with absence of effective diagnosis. Despite continuous efforts, its underlying mechanisms of malignant nature remain ambiguous. The objective of this study was to investigate delta-like 1 (DLL1) as a tumor suppressor in the metastasic ability of human pancreatic cancer cells. MATERIALS AND METHODS: Cellular expression of DLL1 was demonstrated using the GEO public database and western blot analysis. The biological function of DLL1 was validated by biological behavior analysis. Prognosis to DLL1 expression was demonstrated using analysis of the GEO public database. RESULTS: Analysis using the GEO database and western blotting showed higher DLL1 mRNA and protein expression levels in pancreatic cancer compared to those in normal pancreas. DLL1 was uniquely expressed in seven human pancreatic cancer cell lines compared to human pancreatic duct epithelial H6c7 cells. Ablation of DLL1 expression stimulated migration and invasion by activating Src and p38 phosphorylation, but not viability and chemo-resistance of human pancreatic cancer cells. In addition, expression of DLL1 was correlated with migratory features of pancreatic cancer in vivo. Moreover, high DLL1 expression was associated with a favorable prognosis in pancreatic cancer patients. CONCLUSION: DLL1 is a potent suppressor of pancreatic cancer metastasis. Understanding correlation between expression and function of DLL1 might contribute to our knowledge of the complicated mechanism of pancreatic cancer metastasis.


Subject(s)
Calcium-Binding Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , Pancreatic Neoplasms/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prognosis , Signal Transduction , Survival Rate
13.
Cancers (Basel) ; 12(2)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979216

ABSTRACT

Even though the tumour suppressive role of PTEN is well-known, its prognostic implications are ambiguous. The objective of this study was to further explore the function of PTEN expression in human pancreatic cancer. The expression of PTEN has been dominant in various human cancers including pancreatic cancer when compared with their matched normal tissues. The pancreatic cancer cells have been divided into PTEN blockade-susceptible and PTEN blockade-impassible groups dependent on targeting PTEN by altering intracellular signaling. The expression of PTEN has led to varying clinical outcomes of pancreatic cancer based on GEO Series (GSE) data analysis and Liptak's z analysis. Differential dependency to PTEN blockade has been ascertained based on the expression of polo-like kinase1 PLK1 in pancreatic cancer cells. The prognostic value of PTEN also depends on PLK1 expression in pancreatic cancer. Collectively, the present study provides a rationale for targeting PTEN as a promising therapeutic strategy dependent on PLK1 expressions using a companion biomarker discovery platform.

14.
Anticancer Res ; 39(12): 6685-6691, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31810933

ABSTRACT

BACKGROUND/AIM: No effective therapeutics have yet been developed for pancreatic cancer. 2-Methoxy-4-vinyl phenol (2M4VP), a member of the class of phenols, has been demonstrated to have anti-inflammatory properties and cause cell cycle arrest making it an attractive candidate drug for the treatment of pancreatic cancer. MATERIALS AND METHODS: The effects of 2M4VP were examined in Panc-1 and SNU-213 human pancreatic cancer cells. RESULTS: 2M4VP had anticancer effects on pancreatic cancer cell lines, Panc-1 and SNU-213. 2M4VP reduced the viability of Panc-1 cells by inhibiting the expression of the cell nuclear antigen (PCNA) protein. 2M4VP also suppressed the migratory activity of both cell lines. In addition, treatment with 2M4VP effectively decreased the phosphorylation of Focal Adhesion Kinase (FAK) and AKT. CONCLUSION: 2M4VP might be used as a pancreatic cancer treatment supplement.


Subject(s)
Cell Movement/drug effects , Focal Adhesion Kinase 1/antagonists & inhibitors , Guaiacol/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Vinyl Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Focal Adhesion Kinase 1/metabolism , Guaiacol/pharmacology , Hepatocyte Growth Factor/metabolism , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
15.
Anticancer Res ; 39(11): 5963-5971, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31704821

ABSTRACT

BACKGROUND/AIM: The aim of this study was to investigate matrix metalloproteinase 11 (MMP11) as a promising biomarker in human pancreatic cancer. MATERIALS AND METHODS: A consecutive eliminating method was used to select biomarker candidates in pancreatic cancer. mRNA and protein expression levels of candidates were determined in tissues and whole blood samples of healthy donors and pancreatic cancer patients. The prognostic value of MMP11 was determined using various data-sets and Liptak's Z analysis. RESULTS: Analysis using Gene Expression Omnibus (GEO) database showed significantly higher MMP11 mRNA expression in pancreatic cancer tissues compared to that in various normal tissues. MMP11 protein was specifically expressed in pancreatic cancer tissues, but not in various normal or other cancer tissues. Secreted MMP11 levels could be measured using easily accessible techniques and whole blood samples of pancreatic cancer. In addition, high levels of MMP11 were associated with poor prognosis of pancreatic cancer patients. CONCLUSION: MMP11 may be a promising prognostic biomarker for pancreatic cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Matrix Metalloproteinase 11/metabolism , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Proliferation , Follow-Up Studies , Humans , Matrix Metalloproteinase 11/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/enzymology , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Phytomedicine ; 58: 152762, 2019 May.
Article in English | MEDLINE | ID: mdl-31005717

ABSTRACT

BACKGROUND: We have previously reported the functional anti-cancer effects of the products of enzymatic hydrolysis of Citrus unshiu peel (εCUP) and fermented extraction of Citrus unshiu peel (ƒCUP) in human pancreatic cancer. Despite their different characteristics and effects, the underlying mechanism remains elusive. PURPOSE: In this study, we further demonstrate the impact of ingredient contents of Citrus unshiu peel on the cancer's natural features. METHODS: Anti-pancreatic cancer activities following combined treatment of naringenin and hesperetin were demonstrated in vitro and in vivo experiments. RESULTS: Combined treatment with naringenin and hesperetin inhibited the growth of human pancreatic cancer cells (εCUP mimic condition, p < 0.001 for Miapaca-2 cells) through induction of caspase-3 cleavage compared to separate treatment with naringenin or hesperetin. Combined treatment with naringenin and hesperetin also inhibited the migration (εCUP mimic condition, p < 0.001 for Panc-1 cells) of human pancreatic cancer cells. The εCUP mimic condition had the most effective anti-cancer features; in contrast, which had no inhibitory effect on growth and migration of normal cells (HUVECs and Detroit551 cells). In addition, εCUP mimic condition inhibited the phosphorylation of focal adhesion kinase (FAK) and p38 signaling compared with separate treatment with naringenin or hesperetin. Of note, εCUP mimic condition showed a prominent anti-growth effect (p < 0.001) compared with control or ƒCUP mimic condition in vivo xenograft models. CONCLUSION: These results suggest that combined treatment with naringenin and hesperetin might be a promising anti-cancer strategy for pancreatic cancers without eliciting toxicity on normal cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Focal Adhesion Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Citrus/chemistry , Down-Regulation/drug effects , Flavanones/administration & dosage , Hesperidin/administration & dosage , Humans , Mice, Inbred BALB C , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Xenograft Model Antitumor Assays
17.
Cancer Lett ; 456: 1-12, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30902562

ABSTRACT

Pancreatic cancer remains one of the most dangerous cancers with a grave prognosis. We have reported that dual specificity phosphatise 28 (DUSP28) could be secreted in pancreatic cancer cells. However, its biological function is poorly understood. Here, we distinguish the function of scattered DUSP28 in human pancreatic cancer. DUSP28 was specifically secreted to cultured medium in metastatic pancreatic cancer cells. Treatment with recombinant DUSP28 significantly increased the migration, invasion, and viability of metastatic pancreatic cancer cells through the activation of CREB, AKT, and ERK1/2 signaling pathways. In addition, administration of recombinant DUSP28 elicited pro-angiogenic effects in human umbilical vein endothelial cells. Injection of recombinant DUSP28 also produced tumor growth in vivo. Of interest, DUSP28 formed an autocrine loop with integrin α1 (ITGα1) by transcriptional regulation and recombinant DUSP28 acted as an oncogenic reagent through the interaction with ITGα1. Notably, scattered DUSP28 could be detected in whole blood samples of pancreatic cancer patients by accessible immunoassay. These results provide the basis for DUSP28 as a promising therapeutic target and a biomarker for metastatic pancreatic cancer.


Subject(s)
Autocrine Communication , Cell Movement , Dual-Specificity Phosphatases/metabolism , Pancreatic Neoplasms/enzymology , Paracrine Communication , Animals , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Integrin alpha1/genetics , Integrin alpha1/metabolism , MAP Kinase Signaling System , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Physiologic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Burden
18.
Sci Rep ; 9(1): 2793, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808960

ABSTRACT

The prognosis of pancreatic cancer remains dismal despite continuous and considerable efforts. Integrins (ITGs) are highly expressed in various malignant cancers. However, very few studies investigated the role of integrin α3 (ITGα3) in malignant cancers. Here, we determined the functional role of ITGα3 in pancreatic cancer. Analysis of public microarray databases and Western blot analysis indicated a unique expression of ITGα3 in human pancreatic cancer. Silencing ITGα3 expression significantly inhibited the viability and migration of human pancreatic cancer cells. Notably, ablation of ITGα3 expression resulted in a significant decrease of epidermal growth factor receptor (EGFR) expression compared with transfection of control-siRNA through an increased number of leucine-rich repeats and immunoglobulin-like domain protein 1 (LRIG1) expression. In addition, ablating ITGα3 inhibited tumour growth via blockade of EGFR signalling in vivo. Furthermore, the highly expressed ITGα3 led to a poor prognosis of pancreatic cancer patients. Our results provide novel insights into ITGα3-induced aggressive pancreatic cancer.


Subject(s)
Integrin alpha3/metabolism , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Survival , Databases, Genetic , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Integrin alpha3/chemistry , Integrin alpha3/genetics , Kaplan-Meier Estimate , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Up-Regulation
19.
J Med Food ; 21(1): 5-12, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29346059

ABSTRACT

Pancreatic cancer is one of the most dangerous cancers with high mortality rates. Despite continuous efforts, there has been limited improvement in its prognosis. In this study, we prepared fermented extract of Citrus unshiu peel (fCUP) from the by-product after juice processing and then examined the anticancer effects of fCUP on human pancreatic cancer cells. Treatment with fCUP inhibited the growth of human pancreatic cancer cells through induction of caspase-3 cleavage both in vitro and in vivo. Treatment with fCUP also blocked the migration of human pancreatic cancer cells through activation of intracellular signaling pathways such as MKK3/6 and P38. In contrast, treatment with fCUP did not inhibit growth and migration of human umbilical vein endothelial cells. In addition, we found that fCUP mainly consisted of aboriginal compounds, narirutin and hesperidin, as well as newly generated compounds, naringenin and hesperetin. In silico analysis showed that naringenin and hesperetin were the unique modules related to anticancer effect. Furthermore, fCUP exhibited the anticancer effects in in vivo xenograft models. Collectively, these results suggest that fCUP might have the potential to be developed into an effective anticancer drug for pancreatic cancers without causing adverse side-effects.


Subject(s)
Citrus/chemistry , Pancreatic Neoplasms/drug therapy , Plant Extracts/administration & dosage , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Fermentation , Fruit/chemistry , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/physiopathology , Plant Extracts/chemistry , Signal Transduction/drug effects , Waste Products/analysis , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Sci Rep ; 7(1): 12760, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28986588

ABSTRACT

Pancreatic cancer remains one of the most deadly cancers with a grave prognosis. Despite continuous efforts to improve remedial values, limited progress has been made. We have reported that dual specificity phosphatase 28 (DUSP28) has a critical role of chemo-resistance and migration in pancreatic cancers. However, its mechanism remains unclear. Here, we further clarify the function of DUSP28 in pancreatic cancers. Analysis using a public microarray database and in vitro assay indicated a critical role of platelet derived growth factor A (PDGF-A) in pancreatic cancer malignancy. PDGF-A was positively regulated by DUSP28 expression at the mRNA and protein levels. Enhanced DUSP28 sensitized pancreatic cancer cells to exogenous PDGF-A treatment in migration, invasion, and proliferation. Transfection with siRNA targeting DUSP28 blunted the influence of administered PDGF-A by inhibition of phosphorylation of FAK, ERK1/2, and p38 signalling pathways. In addition, DUSP28 and PDGF-A formed an acquired autonomous autocrine-signaling pathway. Furthermore, targeting DUSP28 inhibited the tumor growth and migratory features through the blockade of PDGF-A expression and intracellular signaling in vivo. Our results establish novel insight into DUSP28 and PDGF-A related autonomous signaling pathway in pancreatic cancer.


Subject(s)
Autocrine Communication , Dual-Specificity Phosphatases/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Platelet-Derived Growth Factor/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/genetics , Platelet-Derived Growth Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...