Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cancer Res ; 82(6): 1038-1054, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34654724

ABSTRACT

Dissemination of ovarian cancer cells can lead to inoperable metastatic lesions in the bowel and omentum that cause patient death. Here we show that LRRC15, a type-I 15-leucine-rich repeat-containing membrane protein, highly overexpressed in ovarian cancer bowel metastases compared with matched primary tumors and acts as a potent promoter of omental metastasis. Complementary models of ovarian cancer demonstrated that LRRC15 expression leads to inhibition of anoikis-induced cell death and promotes adhesion and invasion through matrices that mimic omentum. Mechanistically, LRRC15 interacted with ß1-integrin to stimulate activation of focal adhesion kinase (FAK) signaling. As a therapeutic proof of concept, targeting LRRC15 with the specific antibody-drug conjugate ABBV-085 in both early and late metastatic ovarian cancer cell line xenograft models prevented metastatic dissemination, and these results were corroborated in metastatic patient-derived ovarian cancer xenograft models. Furthermore, treatment of 3D-spheroid cultures of LRRC15-positive patient-derived ascites with ABBV-085 reduced cell viability. Overall, these data uncover a role for LRRC15 in promoting ovarian cancer metastasis and suggest a novel and promising therapy to target ovarian cancer metastases.Significance: This study identifies that LRRC15 activates ß1-integrin/FAK signaling to promote ovarian cancer metastasis and shows that the LRRC15-targeted antibody-drug conjugate ABBV-085 suppresses ovarian cancer metastasis in preclinical models.


Subject(s)
Immunoconjugates , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Cell Adhesion , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacology , Integrins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
5.
Gynecol Oncol ; 154(3): 495-504, 2019 09.
Article in English | MEDLINE | ID: mdl-31204077

ABSTRACT

OBJECTIVE: This study is designed to identify genes and pathways that could promote metastasis to the bowel in high-grade serous ovarian cancer (OC) and evaluate their associations with clinical outcomes. METHODS: We performed RNA sequencing of OC primary tumors (PTs) and their corresponding bowel metastases (n = 21 discovery set; n = 18 replication set). Differentially expressed genes (DEGs) were those expressed at least 2-fold higher in bowel metastases (BMets) than PTs in at least 30% of patients (P < .05) with no increased expression in paired benign bowel tissue and were validated with quantitative reverse transcription PCR. Using an independent OC cohort (n = 333), associations between DEGs in PTs and surgical and clinical outcomes were performed. Immunohistochemistry and mouse xenograft studies were performed to confirm the role of LRRC15 in promoting metastasis. RESULTS: Among 27 DEGs in the discovery set, 21 were confirmed in the replication set: SFRP2, Col11A1, LRRC15, ADAM12, ADAMTS12, MFAP5, LUM, PLPP4, FAP, POSTN, GRP, MMP11, MMP13, C1QTNF3, EPYC, DIO2, KCNA1, NETO1, NTM, MYH13, and PVALB. Higher expression of more than half of the genes in the PT was associated with an increased requirement for bowel resection at primary surgery and an inability to achieve complete cytoreduction. Increased expression of LRRC15 in BMets was confirmed by immunohistochemistry and knockdown of LRRC15 significantly inhibited tumor progression in mice. CONCLUSIONS: We identified 21 genes that are overexpressed in bowel metastases among patients with OC. Our findings will help select potential molecular targets for the prevention and treatment of malignant bowel obstruction in OC.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/secondary , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Cohort Studies , Female , Gene Knockdown Techniques , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , RNA, Neoplasm/genetics , Transcriptome , Up-Regulation
6.
Sci Rep ; 8(1): 17717, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30531928

ABSTRACT

15-hydroxyprostaglandin dehydrogenase (15-PGDH), the rate-limiting enzyme in prostaglandin E2 degradation, is decreased in gastric cancers and microRNA (miR)-21 is one of the regulators. We investigated the expression and regulation of 15-PGDH in eary gastric carcinogenesis utilizing endoscopic submucosal dissection (ESD) and gastric cancer cell lines. Expression of 15-PGDH and cyclooxygenase-2 as well as the promoter methylation of 15-PGDH were evaluted. CRISPR, miR-21 transfection, proliferation and apoptosis assays were also done. We observed significant decreases in 15-PGDH expression but no promoter methylation was detected in any ESDs. 15-PGDH suppression by CRISPR induced enhanced growth kinetics. miR-21, which was detected in high level in gastric tumors from the TGCA data, caused increased proliferation, decreased apoptosis. miR-21 overexpression was confirmed with CISH and RT-PCR in the ESDs. Loss of 15-PGDH occurs at the very early stage of gastric adenocarcinoma by miR-21. H. pylori infection may affect miR-21 up regulation. Maintaining 15-PGDH enzyme activity could be a new strategic measure in preventing gastric cancer especially tubular adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Hydroxyprostaglandin Dehydrogenases/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cyclooxygenase 2/genetics , Dinoprostone/genetics , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic , Helicobacter Infections/genetics , Humans , Methylation , Promoter Regions, Genetic/genetics , Prospective Studies , Retrospective Studies , Transfection/methods
7.
Oncogene ; 37(27): 3715-3728, 2018 07.
Article in English | MEDLINE | ID: mdl-29628506

ABSTRACT

To elucidate the underlying oncogenic mechanism of zinc finger protein 746 (ZNF746), current study was conducted in colorectal cancers (CRCs). Herein, ZNF746 was overexpressed in HCT116, SW620, and SW480 cells, which was supported by CRC tissue microarray and TCGA analysis. Also, DNA microarray revealed the differentially expressed gene profile particularly related to cell cycle genes and c-Myc in ZNF746 depleted HCT116 cells. Furthermore, ZNF746 enhanced the stability of c-Myc via their direct binding through nuclear colocalization by immunoprecipitation and immunofluorescence, while ZNF746 and c-Myc exist mainly in nucleoplasm. Conversely, ZNF746 depletion attenuated phosphorylation of c-Myc (S62) and glycogen synthase kinase 3ß (GSK3ß) (S9) and also activated p-c-Myc (T58), which was reversed by GSK3 inhibitors such as SB-216763 and Enza. Also, c-Myc degradation by ZNF746 depletion was blocked by knockdown of F-box/WD repeat-containing protein 7 (FBW7) ubiquitin ligase or proteosomal inhibitor MG132. Additionally, the growth of ZNF746 depleted HCT116 cancer cells was retarded with decreased expression of ZNF746 and c-Myc. Overall, these findings suggest that ZNF746 promotes CRC progression via c-Myc stability mediated by GSK3 and FBW7.


Subject(s)
Colorectal Neoplasms/pathology , F-Box-WD Repeat-Containing Protein 7/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Repressor Proteins/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Cysteine Proteinase Inhibitors/pharmacology , Disease Progression , F-Box-WD Repeat-Containing Protein 7/genetics , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/genetics , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Indoles/pharmacology , Leupeptins/pharmacology , Male , Maleimides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Phosphorylation/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Transplantation, Heterologous
8.
Cancer Lett ; 412: 88-98, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29024811

ABSTRACT

Here the underlying role of CNOT2, a subunit of CCR4-NOT complex, was elucidated in cancer progression. CNOT2 was overexpressed in HIT-T15, ASPC-1, BXPC-3, PC-3, LNCaP, MCF-7 and MDA-MB-231 cell lines, which was confirmed by Tissue array in various human tumor tissues. Also, CNOT2 depletion suppressed proliferation and colony formation of MDA-MB-231 cells. Of note, microarray revealed decreased expression of CNOT2, VEGF-A, HIF2 alpha (<0.5 fold) and increased expression of UMOD1, LOC727847, MMP4, hCG and other genes (>2.0 fold) in CNOT2 depleted MDA-MB-231 cells compared to untreated control. Consistently, downregulation of VEGF, CNOT2 and HIF2 alpha was verified in CNOT2 depleted MDA-MB-231 cells by RT-qPCR. Additionally, CNOT2 depletion inhibited VEGF induced tube formation in HUVECs and reduced neovascularization in CAM assay. Furthermore, the growth of CNOT2 depleted MDA-MB-231 cells was significantly reduced in Balb/c nude mice along with decreased expression of VEGF and PCNA by immunohistochemistry compared to untreated control group. Overall, our findings provide evidences that CNOT2 promotes proliferation and angiogenesis via VEGF signaling in MDA-MB-231 breast cancer cells as a potent molecular target for breast cancer treatment.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Neovascularization, Pathologic/etiology , Repressor Proteins/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Breast Neoplasms/blood supply , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, Inbred BALB C
9.
Sci Rep ; 7: 41977, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28169314

ABSTRACT

Defective autophagy and deranged metabolic pathways are common in cancer; pharmacologic targeting of these two pathways could provide a viable therapeutic option. However, how these pathways are regulated by limited availability of growth factors is still unknown. Our study shows that HSulf-1 (endosulfatase), a known tumor suppressor which attenuates heparin sulfate binding growth factor signaling, also regulates interplay between autophagy and lipogenesis. Silencing of HSulf-1 in OV202 and TOV2223 cells (ovarian cancer cell lines) resulted in increased lipid droplets (LDs), reduced autophagic vacuoles (AVs) and less LC3B puncta. In contrast, HSulf-1 proficient cells exhibit more AVs and reduced LDs. Increased LDs in HSulf-1 depleted cells was associated with increased ERK mediated cPLA2S505 phosphorylation. Conversely, HSulf-1 expression in SKOV3 cells reduced the number of LDs and increased the number of AVs compared to vector controls. Furthermore, pharmacological (AACOCF3) and ShRNA mediated downregulation of cPLA2 resulted in reduced LDs, and increased autophagy. Finally, in vivo experiment using OV202 Sh1 derived xenograft show that AACOCF3 treatment effectively attenuated tumor growth and LD biogenesis. Collectively, these results show a reciprocal regulation of autophagy and lipid biogenesis by HSulf-1 in ovarian cancer.


Subject(s)
Autophagy , Gene Expression Regulation, Neoplastic/drug effects , Lipid Droplets/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Sulfotransferases/metabolism , Animals , Antineoplastic Agents/pharmacology , Arachidonic Acids/pharmacology , Carboplatin/pharmacology , Drug Combinations , Enzyme Inhibitors/pharmacology , Female , Humans , Lipid Droplets/drug effects , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Phospholipases A2, Cytosolic/antagonists & inhibitors , RNA, Small Interfering/genetics , Signal Transduction , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Phytother Res ; 30(1): 90-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26555861

ABSTRACT

In the present study, the underlying apoptotic mechanism of sanggenol L was elucidated in ovarian cancer cells. Sanggenol L showed cytotoxic and antiproliferative effect in A2780, SKOV-3, and OVCAR-3 ovarian cancer cells in a concentration-dependent fashion. Consistently, sanggenol L increased sub-G1 phase population and early and late apoptotic portion in ovarian cancer cells. Also, sanggenol L activated caspase9/3, suppressed the phosphorylation of IκBα and p65 NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), attenuated the expression of Cyclin D1, and cleaved poly(adenosine diphosphate ribose -ribose) polymerase in SKOV-3, A2780, and OVCAR-3 cells. Furthermore, sanggenol L blocked nuclear translocation of NF-κB and also attenuated the expression of NF-κB related genes such as c-Myc, Cyclin D1, and Bcl-X L, Bcl-2, in lipopolysaccharide-treated SKOV-3 cells. Overall, our findings for the first time suggest that sanggenol L induces apoptosis via caspase activation and inhibition of NF-κB/IκBα phosphorylation as a potent chemotherapeutic agent for ovarian cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Flavanones/pharmacology , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Caspases/metabolism , Cell Line, Tumor/drug effects , Cyclin D1/metabolism , Female , Humans , I-kappa B Proteins/metabolism , Morus/chemistry , NF-KappaB Inhibitor alpha , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
11.
Br J Pharmacol ; 173(6): 1033-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26661339

ABSTRACT

BACKGROUND AND PURPOSE: The TNF-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent due to its remarkable ability to selectively kill tumour cells. However, because most tumours exhibit resistance to TRAIL-induced apoptosis, the development of combination therapies to overcome resistance to TRAIL is required for effective cancer therapy. EXPERIMENTAL APPROACH: Cell viability and possible synergy between the plant pyranocoumarin decursin and TRAIL was measured by MTT assay and calcusyn software. Reactive oxygen species (ROS) and apoptosis were measured using dichlorodihydrofluorescein and annexin/propidium iodide in cell flow cytometry. Changes in protein levels were assessed with Western blotting. KEY RESULTS: Combining decursin and TRAIL markedly decreased cell viability and increased apoptosis in TRAIL-resistant non-small-cell lung cancer (NSCLC) cell lines. Decursin induced expression of the death receptor 5 (DR5). Inhibition of DR5 attenuated apoptotic cell death in decursin + TRAIL treated NSCLC cell lines. Interestingly, induction of DR5 and CCAAT/enhancer-binding protein homologues protein by decursin was mediated through selective induction of the pancreatic endoplasmic reticulum kinase (PERK)/activating transcription factor 4 (ATF4) branch of the endoplasmic reticulum stress response pathway. Furthermore, enhancement of PERK/ATF4 signalling by decursin was mediated by ROS generation in NSCLC cell lines, but not in normal human lung cells. Decursin also markedly down-regulated expression of survivin and Bcl-xL in TRAIL-resistant NSCLC cells. CONCLUSIONS AND IMPLICATIONS: ROS generation by decursin selectively activated the PERK/ATF4 axis of the endoplasmic reticulum stress signalling pathway, leading to enhanced TRAIL sensitivity in TRAIL-resistant NSCLC cell lines, partly via up-regulation of DR5.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzopyrans/pharmacology , Butyrates/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Activating Transcription Factor 4/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Humans , Oxidative Stress/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , eIF-2 Kinase/metabolism
12.
Cell Physiol Biochem ; 37(5): 1881-9, 2015.
Article in English | MEDLINE | ID: mdl-26584287

ABSTRACT

BACKGROUND/AIMS: Though CCR4-NOT2 (CNOT2), one of CCR4-NOT complex subunits, was known to be involved in metastasis and apoptosis through transcription and mRNA degradation, its other biological function is poorly understood so far. The aim of this study is to elucidate the molecular role of CNOT2 in the differentiation process of 3T3-L1 preadipocytes. METHODS AND RESULTS: CNOT2 was overexpressed during the differentiation process of 3T3-L1 preadipocytes. Consistently, mRNA levels of CNOT2, adiponectin, adiponectin 2, PPARx03B3; and CEBPα were enhanced in 3T3-L1 adipocytes. Conversely, CNOT2 depletion by siRNA transfection also reversed the activation of PPARx03B3; and CEBPα and inhibition of GSK3α/ß and ß-catenin at the protein level in 3T3-L1 preadipocytes. Immunofluorescence assay revealed that CNOT2 was colocalized with PPARx03B3;, but not with CEBPα in 3T3-L1 adipocyte. Consistently, IP western blots revealed that CNOT2 interacted with PPARx03B3; in 3T3-L1 adipocyte. CONCLUSION: Our findings demonstrate that CNOT2 promotes the differentiation of 3T3-L1 preadipocytes via upregulation of PPARx03B3;, and CEBPα and inhibition of GSK3α/ß and ß-catenin signaling as a potent molecular target for obesity.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , PPAR gamma/metabolism , Repressor Proteins/metabolism , beta Catenin/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis , Adiponectin/genetics , Adiponectin/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Down-Regulation , Glycogen Synthase Kinase 3 beta , Mice , Microscopy, Fluorescence , PPAR gamma/genetics , Phosphorylation , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Up-Regulation
13.
Phytother Res ; 29(8): 1225-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26014377

ABSTRACT

Although gallotannin contained in several medicinal plants was known to have multi-biological activities, such as antioxidant, antiinflammatory, antimicrobial, immunomodulatory, and antitumor effects, the underlying apoptotic mechanism of gallotannin is not fully understood so far. Thus, in the present study, the apoptotic mechanism of gallotannin was elucidated in DU145, PC-3, and M2182 prostate cancer cells in association with myeloid cell leukemia 1 (Mcl-1) signaling. Gallotannin exerted dose-dependent cytotoxicity in DU145, PC-3, and M2182 prostate cancer cells. Also, gallotannin showed apoptotic morphological features and increased the number of terminal deoxynucleotidyl transferase dUTP nick end labeling positive cells and sub-G1 accumulation in three prostate cancer cell lines. Consistently, gallotannin cleaved poly (ADP-ribose) polymerase (PARP) and attenuated the expression of procaspases 9 and 3 in three prostate cancer cell lines. Furthermore, gallotannin attenuated the expression of survival genes such as Mcl-1, B-cell lymphoma 2, and B-cell lymphoma 2 extra large in three prostate cancer cell lines. Interestingly, overexpression of Mcl-1 reversed the ability of gallotannin to cleave PARP and increase sub-G1 population in three prostate cancer cell lines. Conversely, silencing of Mcl-1 enhanced apoptosis by gallotannin in three prostate cancer cell lines by FACSCalibur (Becton Dickinson, Franklin Lakes, NJ, USA). Taken together, our findings demonstrate that inhibition of Mcl-1 and activation of caspases are critically involved in gallotannin-induced apoptosis in prostate cancer cells.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Hydrolyzable Tannins/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Prostatic Neoplasms/pathology , Cell Line, Tumor/drug effects , Humans , Male , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
14.
Phytother Res ; 29(7): 1026-31, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25892665

ABSTRACT

This study was designed to investigate the antitumor mechanism of Phytol in hepatocellular carcinomas including Huh7 and HepG2 cells in association with caspase dependent apoptosis and epithelial mesenchymal transition (EMT) signaling. Phytol significantly suppressed the viability of Huh7 and HepG2 cells. Also, Phytol significantly increased the sub G1 population and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling (TUNEL) positive cells in a concentration dependent manner in Huh7 and HepG2 cells. Consistently, Phytol cleaved poly (adenosine diphosphate-ribose) polymerase (PARP), activated caspase-9/3, and Bax attenuated the expression of survival genes such as Bcl-2, Mcl-1, and c-Myc in Huh7 and HepG2 cells. Of note, Phytol also suppressed typical morphology change of EMT such as loss of cell adhesion and formation of fibroblast like mesenchymal cells in HepG2 cells. Furthermore, Phytol also reversed the loss of E-cadherin and overexpression of p-smad2/3, alpha-smooth muscle actin, and Snail induced by EMT promoter transforming growth factor beta1 in HepG2 cells. Overall, our findings suggest that Phytol exerts antitumor activity via apoptosis induction through activation of caspas-9/3 and inhibition of EMT in hepatocellular carcinoma cells as a potent anticancer candidate for liver cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/pathology , Phytol/pharmacology , Apoptosis/drug effects , Cadherins/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Hep G2 Cells , Humans , In Situ Nick-End Labeling , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
15.
Br J Pharmacol ; 172(14): 3565-78, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25825203

ABSTRACT

BACKGROUND AND PURPOSE: Epigallocatechin-3-gallate (EGCG) is a component of green tea known to have chemo-preventative effects on several cancers. However, EGCG has limited clinical application, which necessitates the development of a more effective EGCG prodrug as an anticancer agent. EXPERIMENTAL APPROACH: Derivatives of EGCG were evaluated for their stability and anti-tumour activity in human chronic myeloid leukaemia (CML) K562 and KBM5 cells. KEY RESULTS: EGCG-mono-palmitate (EGCG-MP) showed most prolonged stability compared with other EGCG derivatives. EGCG-MP exerted greater cytotoxicity and apoptosis in K562 and KBM5 cells than the other EGCG derivatives. EGCG-MP induced Src-homology 2 domain-containing tyrosine phosphatase 1 (SHP-1) leading decreased oncogenic protein BCR-ABL and STAT3 phosphorylation in CML cells, compared with treatment with EGCG. Furthermore, EGCG-MP reduced phosphorylation of STAT3 and survival genes in K562 cells, compared with EGCG. Conversely, depletion of SHP-1 or application of the tyrosine phosphatase inhibitor pervanadate blocked the ability of EGCG-MP to suppress phosphorylation of BCR-ABL and STAT3, and the expression of survival genes downstream of STAT3. In addition, EGCG-MP treatment more effectively suppressed tumour growth in BALB/c athymic nude mice compared with untreated controls or EGCG treatment. Immunohistochemistry revealed increased caspase 3 and SHP-1 activity and decreased phosphorylation of BCR-ABL in the EGCG-MP-treated group relative to that in the EGCG-treated group. CONCLUSIONS AND IMPLICATIONS: EGCG-MP induced SHP-1-mediated inhibition of BCR-ABL and STAT3 signalling in vitro and in vivo more effectively than EGCG. This derivative may be a potent chemotherapeutic agent for CML treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction , Animals , Catechin/pharmacology , Cell Cycle/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Phytother Res ; 29(6): 844-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25753585

ABSTRACT

Galbanic acid (GBA), a major compound of Ferula assafoetida, was known to have cytotoxic, anti-angiogenic and apoptotic effects in prostate cancer and murine Lewis lung cancer cells; the underling apoptotic mechanism of GBA still remains unclear so far. Thus, in the present study, the apoptotic mechanism of GBA was investigated mainly in H460 non-small cell lung carcinoma (NSCLC) cells because H460 cells were most susceptible to GBA than A549, PC-9 and HCC827 NSCLC cells. Galbanic acid showed cytotoxicity in wild EGFR type H460 and A549 cells better than other mutant type PC-9 and HCC827 NSCLC cells. Also, GBA significantly increased the number of Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) positive cells and sub G1 population in H460 cells. Western blotting revealed that GBA cleaved poly (ADP-ribose) polymerase (PARP), activated Bax and caspase 9, attenuated the expression of Bcl-2, Bcl-x(L), and Myeloid cell leukemia 1 (Mcl-1) in H460 cells. However, interestingly, overexpression of Mcl-1 blocked the ability of GBA to exert cytotoxicity, activate caspase9 and Bax, cleave PARP, and increase sub G1 accumulation in H460 cells. Overall, these findings suggest that GBA induces apoptosis in H460 cells via caspase activation and Mcl-1 inhibition in H460 cells as a potent anticancer agent for NSCLC treatment.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Caspases/metabolism , Coumarins/pharmacology , Lung Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor/drug effects , Ferula/chemistry , Humans , Poly(ADP-ribose) Polymerases/metabolism
17.
Oncotarget ; 6(7): 4992-5004, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25669977

ABSTRACT

The heparan sulfate mimetic PG545 has been shown to exert anti-angiogenic and anti-metastatic activity in vitro and in vivo cancer models. Although much of this activity has been attributed to inhibition of heparanase and heparan sulfate-binding growth factors, it was hypothesized that PG545 may additionally disrupt Wnt signaling, an important pathway underlying the malignancy of pancreatic cancer. We show that PG545, by directly interacting with Wnt3a and Wnt7a, inhibits Wnt/ß-catenin signaling leading to inhibition of proliferation in pancreatic tumor cell lines. Additionally, we demonstrate for the first time that the combination of PG545 with gemcitabine has strong synergistic effects on viability, motility and apoptosis induction in several pancreatic cell lines. In an orthotopic xenograft mouse model, combination of PG545 with gemcitabine efficiently inhibited tumor growth and metastasis compared to single treatment alone. Also, PG545 treatment alone decreased the levels of ß-catenin and its downstream targets, cyclin D1, MMP-7 and VEGF which is consistent with our in vitro data. Collectively, our findings suggest that PG545 exerts anti-tumor activity by disrupting Wnt/ß-catenin signaling and combination with gemcitabine should be considered as a novel therapeutic strategy for pancreatic cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Saponins/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Biomimetic Materials/administration & dosage , Biomimetic Materials/pharmacology , Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Drug Synergism , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Random Allocation , Saponins/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine
18.
Cancer Metab ; 2: 13, 2014.
Article in English | MEDLINE | ID: mdl-25225614

ABSTRACT

BACKGROUND: Loss of the endosulfatase HSulf-1 is common in ovarian cancer, upregulates heparin binding growth factor signaling and potentiates tumorigenesis and angiogenesis. However, metabolic differences between isogenic cells with and without HSulf-1 have not been characterized upon HSulf-1 suppression in vitro. Since growth factor signaling is closely tied to metabolic alterations, we determined the extent to which HSulf-1 loss affects cancer cell metabolism. RESULTS: Ingenuity pathway analysis of gene expression in HSulf-1 shRNA-silenced cells (Sh1 and Sh2 cells) compared to non-targeted control shRNA cells (NTC cells) and subsequent Kyoto Encyclopedia of Genes and Genomics (KEGG) database analysis showed altered metabolic pathways with changes in the lipid metabolism as one of the major pathways altered inSh1 and 2 cells. Untargeted global metabolomic profiling in these isogenic cell lines identified approximately 338 metabolites using GC/MS and LC/MS/MS platforms. Knockdown of HSulf-1 in OV202 cells induced significant changes in 156 metabolites associated with several metabolic pathways including amino acid, lipids, and nucleotides. Loss of HSulf-1 promoted overall fatty acid synthesis leading to enhance the metabolite levels of long chain, branched, and essential fatty acids along with sphingolipids. Furthermore, HSulf-1 loss induced the expression of lipogenic genes including FASN, SREBF1, PPARγ, and PLA2G3 stimulated lipid droplet accumulation. Conversely, re-expression of HSulf-1 in Sh1 cells reduced the lipid droplet formation. Additionally, HSulf-1 also enhanced CPT1A and fatty acid oxidation and augmented the protein expression of key lipolytic enzymes such as MAGL, DAGLA, HSL, and ASCL1. Overall, these findings suggest that loss of HSulf-1 by concomitantly enhancing fatty acid synthesis and oxidation confers a lipogenic phenotype leading to the metabolic alterations associated with the progression of ovarian cancer. CONCLUSIONS: Taken together, these findings demonstrate that loss of HSulf-1 potentially contributes to the metabolic alterations associated with the progression of ovarian pathogenesis, specifically impacting the lipogenic phenotype of ovarian cancer cells that can be therapeutically targeted.

19.
J Nat Prod ; 77(1): 63-9, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24328151

ABSTRACT

The underlying antimetastatic mechanism of anethole (1) still remains unclear in association with the molecules of the epithelial to mesenchymal transition (EMT). Herein, the role of the EMT molecules was elucidated in terms of the antimetastatic activity of 1 using DU145 cells. Anethole significantly inhibited the adhesion of DU145 cells to vitronectin-coated plates, as well as migration in a wound-healing assay and invasion using a Boyden chamber. Also, anethole suppressed the expression of MMP-9 in DU145 cells by zymography, ELISA, and RT-PCR. Consistently, the silencing of MMP-9 enhanced the activity of 1 to upregulate the expression of E-cadherin and to attenuate the expression of Vimentin in DU145 cells. Compound 1 enhanced E-cadherin, which is an epithelial marker and attenuated the expression of Vimentin, Twist, and Snail as mesenchymal molecules at the mRNA level. Consistently, anethole upregulated E-cadherin and downregulated the expression of Vimentin, Twist and PI3K, and AKT at the protein level in DU145 cells. Conversely, the antimetastatic effects of 1 to inhibit invasion and the expression of MMP-9 and upregulate E-cadherin were reversed by the EMT inducer TGF-ß in DU145 cells. Overall, the present findings suggest that anethole exerts antimetastatic activity via regulation of crosstalk between EMT molecules and MMP-9 on the basis of the in vitro data obtained.


Subject(s)
Anisoles/pharmacology , Antineoplastic Agents/pharmacology , Matrix Metalloproteinase 9/metabolism , Allylbenzene Derivatives , Anisoles/chemistry , Biomarkers , Cadherins/metabolism , Epithelial-Mesenchymal Transition , Humans , Male , Matrix Metalloproteinase 9/genetics , Mesoderm/metabolism , Molecular Structure , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/pharmacology , Twist-Related Protein 1/drug effects , Vimentin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...