Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Oncotarget ; 11(26): 2484-2492, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32655835

ABSTRACT

Signaling pathways have a key role in driving the uncontrolled development of familial adenomatous polyposis (FAP)- associated and sporadic desmoid tumors (DTs). The relationship between the Wnt/b-catenin signaling pathway and DTs has been extensively studied, but no reliable biomarkers able to detect their histological subtype have been identified for the accurate diagnosis. In this study we studied the differences in miRNA expression between sporadic (20 patients) and FAP-associated DTs (7 patients) using microarray confirmed by quantitative PCR (qPCR). The analysis showed 19 dysregulated miRNAs. Among them miR-133b levels were significantly lower in FAP-associated DT than in sporadic DT. Therefore, two mRNAs, associated to miR-133b and ß-catenin expression, the SIRT1 and ELAVL1were analyzed. The qPCR analysis showed that SIRT1 mRNA levels were significantly up-regulated in FAP-associated DT than in sporadic DT, whereas no differences in ELAVL1 expression was observed between these two DT types. In addition, a negative correlation was observed between miR-133b and SIRT1 in FAP-associated DTs, but not in sporadic DTs. The miR-133b-SIRT1-ß-catenin axis may represent a novel mechanism underlying progression of FAP-associated DT.

2.
Nutr Res ; 60: 68-76, 2018 12.
Article in English | MEDLINE | ID: mdl-30527261

ABSTRACT

Although the cross-kingdom transfer of vegetable miRNAs (miRNAs) in mammalian species, including humans, is still controversial, recent studies have rejected this theory. Based on these recent studies, we hypothesized that artichoke-derived miRNAs (cca-miRNAs) are not adsorbed into human intestinal cells after cooking and in vitro digestion. In order to test this hypothesis, we evaluated miRNA (cca-miRNAs) in the edible part of globe artichokes (head portion), after cooking and digestion by an in vitro digestion system. The cca-miRNA levels were analyzed by real-time PCR (RT-qPCR), and those that withstood cooking and digestion conditions were further analyzed for their bioavailability using an in vitro system (Caco-2/TC7 cell clone). We detected 20 cca-miRNAs after cooking, 5 of which were statistically down-regulated in comparison with uncooked samples. Only 4 cca-miRNAs were found after in vitro digestion. By using scanning electron microscopy (SEM), we also evaluated the extracellular vesicles (EVs) in homogenized artichoke as possible miRNA transporters. However, approximately 81% were degraded after cooking, while the remaining EVs had changed shape from round to elliptical. Finally, we detected no cell-free cca-miRNAs, miRNAs bound to protein complex, and no cca-miRNAs encapsulated in EVs inside Caco-2 cells or in basolateral medium after bioavailability experiments. In conclusion, the data from the present study agrees with recent findings that the human small intestine does not uptake dietary miRNAs from raw or cooked artichoke heads.


Subject(s)
Cooking , Cynara scolymus/chemistry , Intestinal Absorption , Intestine, Small/metabolism , MicroRNAs/pharmacokinetics , Vegetables/chemistry , Biological Availability , Biological Transport , Caco-2 Cells , Cells , Digestion , Extracellular Vesicles , Humans , Inflorescence , MicroRNAs/metabolism , Real-Time Polymerase Chain Reaction
3.
Int J Mol Sci ; 19(5)2018 May 19.
Article in English | MEDLINE | ID: mdl-29783729

ABSTRACT

Chlorogenic acid (CGA) is a polyphenol present in many human dietary foods. Several studies indicated a beneficial role of CGA in the prevention of cancer and an enhancement of chemotherapy when combined with CGA in the treatment of human hepatocarcinoma (HCC). Drug toxicity, resistance and subsequent disease progression represent a problem in HCC management, although treatment with the multikinase inhibitor Regorafenib improved overall survival. This study focused on the evaluation of the effects of combined treatment using both low Regorafenib concentrations and CGA as natural compound in HCC cells. The analysis of cell proliferation by Ki67 staining and cell cycle progression showed that CGA enhanced Regorafenib-mediated cell growth inhibition. Moreover, CGA potentiated the apoptotic effect of Regorafenib by the activation of the pro-apoptotic Annexin V, Bax and Caspase 3/7 and the inhibition of anti-apoptotic Bcl2 and Bcl-xL. Combined treatments were also effective in inhibiting cell motility. The mechanisms underlying the positive effects of combining CGA and Regorafenib were also addressed and an increased inhibition of MAPK (mitogen-activated protein kinase)and PI3K/Akt/mTORC (phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling was observed. Overall, these data demonstrated that co-treatment with Regorafenib and CGA enhanced Regorafenib action, reducing its cytotoxicity in HCC cells. In conclusion, this drug combination could be considered as a safe and more effective approach in HCC therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chlorogenic Acid/pharmacology , Drug Synergism , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Hep G2 Cells , Humans , MAP Kinase Signaling System , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
4.
Cell Oncol (Dordr) ; 41(3): 283-296, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29470830

ABSTRACT

PURPOSE: Emerging evidence indicates that combining Sorafenib with vitamin K1 (VK1) may result in a synergistic inhibition of hepatocellular carcinoma (HCC) cell migration and proliferation. Despite this synergy, its benefits may be limited due to drug resistance resulting from cross-talk with the tumor microenvironment. Insulin-like growth factor-1 (IGF1) signaling acts as an important modulator of HCC cell growth, motility and drug resistance. Therefore, we aimed to explore the effects of Sorafenib in combination with VK1 and/or IGF1-R antagonists on HCC cells. METHODS: Scratch wound migration assays were performed to assess the motility of HCC-derived PLC/PRF/5, HLF and Hep3B cells. The synergistic, additive or antagonistic effects of Sorafenib, VK1 and IGF1-R antagonists on HCC cell motility were assessed using CompuSyn software. The effects mediated by these various compounds on HCC cytoskeleton organization were evaluated using DyLight 554 Phalloidin staining. Proliferation and migration-associated signaling pathways were analyzed in PLC/PRF/5 cells using Erk1/2 and Akt activation kits and Western blotting (Mek, JNK, Akt, Paxillin and p38), respectively. RESULTS: The effects of the IGF1-R antagonists GSK1838705A and OSI-906 on HCC cell migration inhibition after Sorafenib and/or VK1 administration, individually or in combination, were evaluated. We found a synergistic effect in PLC/PRF/5, HLF and Hep3B cells for combinations of fixed doses of GSK1838705A or OSI-906 together with different doses of Sorafenib and/or VK1. The levels of synergy were found to be stronger at higher Sorafenib and/or VK1 concentrations and lower or absent at lower concentrations, with some variation among the different cell lines tested. In addition, we found that in PLC/PRF/5 and HLF cells IGF1-R blockage strongly enhanced the reduction and redistribution of F-actin induced by Sorafenib and/or VK1 through alterations in the phosphorylation levels of some of the principal proteins involved in the MAPK signaling cascade, which is essential for cell migration. CONCLUSIONS: Our results indicate that modulation of the efficacy of Sorafenib through combinations with VK1 and/or IGF1-R antagonists results in synergistic inhibition of HCC cell migration.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Movement/drug effects , Insulin-Like Growth Factor I/antagonists & inhibitors , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Vitamin K 1/pharmacology , Actin Cytoskeleton/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Imidazoles/pharmacology , Liver Neoplasms/pathology , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrazines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sorafenib , Vitamin K 1/therapeutic use
5.
Oncotarget ; 8(61): 103465-103476, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262576

ABSTRACT

The recent RESORCE trial showed that treatment with Regorafenib after Sorafenib failure provided a significant improvement in overall survival in HCC patients. Preclinical and clinical trial data showed that Regorafenib is a more potent drug than Sorafenib. In this study we aimed at improving Regorafenib actions and at reducing its toxicity, by targeting parallel pathways or by combination with Vitamins K (VKs). We investigated the effects of Regorafenib administrated at low concentrations and in combination with either VK1 and/or with GSK1838705A or OSI-906, two IGF1-R inhibitors, on HCC cell growth and motility. Our results showed that both IGF1-R inhibitors potentiated the antiproliferative and pro-apoptotic effects of Regorafenib and/or VK1 in HCC cell lines. Moreover we provide evidence that the combined treatment with IG1-R antagonists and Regorafenib (and/or VK1) also caused a significant reduction and depolymerization of actin resulting in synergistic inhibition exerted on cell migration. Thus, simultaneous blocking of MAPK and PI3K/Akt cascades with IGF1-R inhibitors plus Regorafenib could represent a more potent approach for HCC treatment.

6.
Oncotarget ; 8(26): 41866-41875, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28418912

ABSTRACT

Desmoid tumors (DT) are rare, benign, fibroblastic neoplasm with challenging histological diagnosis. DTs can occur sporadically or associated with the familial adenomatous polyposis coli (FAP). Most sporadic DTs are associated with ß-catenin gene (CTNNB1) mutations, while mutated APC gene causes FAP disease. microRNAs (miRNAs) are involved in many human carcinogenesis.The miRNA profile was analyzed by microarray in formalin-fixed, paraffin-embedded (FFPE) specimens of 12 patients (8 sporadic, 4 FAP-associated) and 4 healthy controls. One hundred and one mRNAs resulted dysregulated, of which 98 in sporadic DTs and 8 in FAP-associated DTs, 5 were shared by both tumors. Twenty-six miRNAs were then validated by RT-qPCR in 23 sporadic and 7 FAP-associated DT samples matched with healthy controls. The qPCR method was also used to evaluate the CTNNB1 mutational status in sporadic DTs. The correlation between sporadic DTs and miRNA expression showed that miR-21-3p increased in mutated versus wild-type DTs, while miR-197-3p was decreased. The mRNA expression of Tetraspanin3 and Serpin family A member 3, as miR-21-3p targets, and L1 Cell Adhesion Molecule, as miR-197-3p target, was also evaluate. CTNNB1 mutations associated to miRNA dysregulation could affect the genesis and the progression of this disease and help histological diagnosis of sporadic DTs.


Subject(s)
Abdominal Neoplasms/genetics , Adenomatous Polyposis Coli/genetics , Fibromatosis, Aggressive/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Mutation , beta Catenin/genetics , Abdominal Neoplasms/metabolism , Abdominal Neoplasms/pathology , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adolescent , Adult , Aged , DNA Mutational Analysis , Female , Fibromatosis, Aggressive/metabolism , Fibromatosis, Aggressive/pathology , Gene Expression Profiling , Genes, APC , Humans , Immunohistochemistry , Male , Middle Aged , RNA Interference , RNA, Messenger/genetics , Serpins/genetics , Tetraspanins/genetics , Transcriptome , Tumor Burden , Young Adult , beta Catenin/metabolism
7.
PLoS One ; 11(3): e0152181, 2016.
Article in English | MEDLINE | ID: mdl-27008165

ABSTRACT

Environmental chemicals, such as heavy metals, affect female reproductive function. A biological sensor of the signals of many toxic chemical compounds seems to be the aryl hydrocarbon receptor (AHR). Previous studies demonstrated the environmental of heavy metals in Taranto city (Italy), an area that has been influenced by anthropogenic factors such as industrial activities and waste treatments since 1986. However, the impact of these elements on female fertility in this geographic area has never been analyzed. Thus, in the present study, we evaluated the AHR pathway, sex steroid receptor pattern and apoptotic process in granulosa cells (GCs) retrieved from 30 women, born and living in Taranto, and 30 women who are living in non-contaminated areas (control group), who were undergoing in vitro fertilization (IVF) protocol. In follicular fluids (FFs) of both groups the toxic and essential heavy metals, such as chromiun (Cr), Manganese (Mn), iron (Fe), cobalt (Co), nickel (Ni), copper (Cu), zinc (Zn), cadmium (Cd) and lead (Pb), were also analyzed. Higher levels of Cr, Fe, Zn and Pb were found in the FFs of the women from Taranto as compared to the control group, as were the levels of AHR and AHR-dependent cytochrome P450 1A1 and 1B1; while CYP19A1 expression was decreased. The anti-apoptotic process found in the GCs of women fromTaranto was associated with the highest levels of progesterone receptor membrane component 1 (PGRMC1), a novel progesterone receptor, the expression of which is subjected to AHR activated by its highest affinity ligands (e.g., dioxins) or indirectly by other environmental pollutants, such as heavy metals. In conclusion, decreased production of estradiol and decreased number of retrieved mature oocytes found in women from Taranto could be due to chronic exposure to heavy metals, in particular to Cr and Pb.


Subject(s)
Environmental Pollutants/adverse effects , Granulosa Cells/drug effects , Receptors, Aryl Hydrocarbon/drug effects , Adult , Apoptosis/drug effects , Aromatase/analysis , Blotting, Western , Case-Control Studies , Female , Follicular Fluid/chemistry , Granulosa Cells/chemistry , Humans , Italy , Membrane Proteins/analysis , Metals, Heavy/analysis , Receptors, Aryl Hydrocarbon/physiology , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Progesterone/analysis
8.
J Exp Clin Cancer Res ; 34: 90, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26329608

ABSTRACT

BACKGROUND: Blood platelet numbers are correlated with growth and aggressiveness of several tumor types, including hepatocellular carcinoma (HCC). We previously found that platelet lysates (hPLs) both stimulated HCC cell growth and migration, and antagonized the growth-inhibitory and apoptotic effects of Regorafenib, multikinase growth inhibitor, on HCC cell lines. We evaluated the effects of human insulin-like growth factor-1 (IGF1), a mitogen contained in platelets, on the Regorafenib-mediated growth inhibition. METHODS: An Elisa kit was used to evaluate hPL IGF1 concentrations. The effects of IGF1 on cell proliferation were assessed with MTT assay and analysis of cell cycle progression. Apoptosis assays, scratch assay and Transwell assay were performed to measure apoptosis, cell migration and invasion respectively. Western blots were performed by standard protocols. RESULTS: IGF1 antagonized growth inhibition exerted by Regorafenib on HCC cell lines. Moreover the mitogen blocked Regorafenib-induced apoptosis and decreased the rate of cell migration and invasion. The IGF1 effects were in turn antagonized by actions of a potent IGF1 receptor inhibitor, GSK1838705A, showing that the IGF1 receptor was involved in the mechanisms of IGF1-mediated blocking of Regorafenib action. GSK1838705A also partially blocked the effects of hPLs in antagonizing Regorafenib-mediated growth inhibition, showing that IGF1 was an important component of hPL actions. CONCLUSIONS: These results show that IGF1 antagonized Regorafenib-mediated growth, migration and invasion inhibition, as well as the drug-mediated induction of apoptosis in HCC cells and reinforce the idea that microenvironmental factors can influence cancer drug actions.


Subject(s)
Antineoplastic Agents/pharmacology , Insulin-Like Growth Factor I/pharmacology , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Apoptosis/drug effects , Cell Movement , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Neoplasm Invasiveness , Signal Transduction
9.
Cancer Chemother Pharmacol ; 75(6): 1237-1245, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25907508

ABSTRACT

PURPOSE: Blood platelet numbers are correlated to growth and aggressiveness of several tumor types, including hepatocellular carcinoma (HCC). We previously found that platelet lysates (hPLs) also stimulated growth and migration, and antagonized the growth-inhibitory and apoptotic effects of both Sorafenib and Regorafenib, two multikinase inhibitors, on three HCC cell lines. In this study, in vitro function of human epidermal growth factor (EGF) with and without Sorafenib or Regorafenib was investigated. METHODS: An ELISA kit was used to evaluate the EGF concentrations in hPLs. In vitro function of EGF was assessed with proliferation MTT test. Apoptosis assay, scratch assays, and Transwell assays were performed for apoptosis, invasion, and migration, respectively. MAPK Activation Kit was used to explore MAPK phosphorylation. RESULTS: EGF antagonized the growth inhibition of Regorafenib on three HCC cell lines. Regorafenib-mediated growth inhibition was blocked by 70 % when the cells were pre-treated with EGF. EGF also blocked Regorafenib-induced apoptosis, as well as Regorafenib-induced decreases in cell migration and invasion. The EGF effects were in turn antagonized by concomitant addition to the cultures of EGF receptor antagonist Erlotinib, showing that the EGF receptor was involved in the mechanisms of EGF-mediated blocking of Regorafenib effects. Erlotinib also partially blocked the effects of hPLs in antagonizing Regorafenib-mediated growth inhibition, showing that EGF was an important component of hPL actions. CONCLUSIONS: All these results show that EGF antagonized Regorafenib-mediated growth and migration inhibition and apoptosis induction in HCC cells and reinforce the idea that microenvironment can influence cancer drug actions.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Epidermal Growth Factor/metabolism , Liver Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Erlotinib Hydrochloride , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phosphorylation/drug effects , Quinazolines/pharmacology , Sorafenib
10.
BMC Cancer ; 14: 351, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24885890

ABSTRACT

BACKGROUND: Platelets are frequently altered in hepatocellular carcinoma (HCC) patients. Platelet lysates (hPL) can enhance HCC cell growth and decrease apoptosis. The aims were to evaluate whether hPL can modulate the actions of sorafenib or regorafenib, two clinical HCC multikinase antagonists. METHODS: Several human HCC cell lines were grown in the presence and absence of sorafenib or regorafenib, with or without hPL. Growth was measured by MTT assay, apoptosis was assessed by Annexin V and by western blot, and autophagy and MAPK growth signaling were also measured by western blot, and migration and invasion were measured by standard in vitro assays. RESULTS: Both sorafenib and regorafenib-mediated inhibition of cell growth, migration and invasion were all antagonized by hPL. Drug-mediated apoptosis and decrease in phospho-ERK levels were both blocked by hPL, which also increased anti-apoptotic phospho-STAT, Bax and Bcl-xL levels. Preliminary data, obtained with epidermal growth factor (EGF) and insulin-like growth factor-I (IGF-I), included in hPL, revealed that these factors were able to antagonized sorafenib in a proliferation assay, in particular when used in combination. CONCLUSIONS: Platelet factors can antagonize sorafenib or regorafenib-mediated growth inhibition and apoptosis in HCC cells. The modulation of platelet activity or numbers has the potential to enhance multikinase drug actions.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Platelets/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Activation , Epidermal Growth Factor/blood , Extracellular Signal-Regulated MAP Kinases/metabolism , Hep G2 Cells , Humans , Insulin-Like Growth Factor I/metabolism , Liver Neoplasms/pathology , Neoplasm Invasiveness , Niacinamide/pharmacology , Phosphorylation , Signal Transduction/drug effects , Sorafenib , Time Factors
11.
Anticancer Agents Med Chem ; 14(8): 1154-60, 2014.
Article in English | MEDLINE | ID: mdl-24934165

ABSTRACT

PURPOSE: Platelet extracts can stimulate cell growth and contribute to tumor biology. It was recently shown that they stimulate the growth of hepatocellular carcinoma (HCC) cells and decrease apoptosis. Doxorubicin is a commonly used HCC chemotherapy that increases apoptosis. We therefore examined the effects of platelet lysates (hPL) on doxorubicin-mediated HCC cell growth inhibition and apoptosis induction. METHODS: Three human HCC cell lines, PLC/PRF/5, Hep3B and HepG2 cells, were grown in culture and growth was measured by the MTT assay and apoptosis was measured using Muse Annexin V assay kit. Cells were also probed by Western blot. RESULTS: hPL decreased doxorubicin-mediated growth inhibition and apoptosis induction in all three cell lines. When doxorubicin and hPL were added at separate time intervals, protection by hPL was also observed. WB showed that hPL caused prolonged and increased levels of phospho-JNK and phospho-p38. Furthermore, a p38 inhibitor abrogated the modulating effects of hPL on both growth and apoptosis, indicating its importance in mediating hPL actions. WBs also showed that hPL decreased doxorubicin-induced markers of apoptosis. CONCLUSIONS: hPL modulate the actions of the cancer chemotherapeutic agent, doxorubicin. Platelets are part of the complex microenvironmental milieu and their effects may contribute to a modulation of chemotherapy actions. Conversely, drugs that alter platelet levels or degranulation could potentially augment doxorubicin actions on HCC cells.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Blood Platelets/metabolism , Carcinoma, Hepatocellular/pathology , Cell Extracts/pharmacology , Doxorubicin/pharmacology , Liver Neoplasms/pathology , Apoptosis , Autophagy , Cell Line, Tumor , Cell Proliferation , Humans , Imidazoles/pharmacology , MAP Kinase Kinase 4/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
BMC Cancer ; 14: 43, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24468103

ABSTRACT

BACKGROUND: Thrombocytopenia has been reported to be associated with small size HCCs, and thrombocytosis to be associated with large size HCCs. The aim was to examine the effects of platelets in relation to HCC cell growth. METHODS: The effects of time-expired pooled normal human platelets were examined on human HCC cell line growth and invasion. RESULTS: Blood platelet numbers increased with increasing HCC tumor size and portal vein invasion. Platelet extracts enhanced cell growth in 4 human HCC cell lines, as well as cell migration, medium AFP levels and decreased apoptosis. Cell invasion was significantly enhanced, using a Matrigel-coated trans-well membrane and 3D (Real-Time Imaging) invasion assay. Western blots showed that platelets caused enhanced phospho-ERK and phospho-JNK signaling and anti-apoptotic effect with increase of Bcl-xL (anti-apoptotic marker) and decrease of Bid (pro-apoptotic marker) levels. Their growth effects were blocked by a JNK inhibitor. CONCLUSIONS: Platelets stimulated growth and invasion of several HCC cell lines in vitro, suggesting that platelets or platelet growth factors could be a potential pharmacological target.


Subject(s)
Blood Platelets/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Signal Transduction , Spheroids, Cellular , Tumor Cells, Cultured
13.
Cancer Chemother Pharmacol ; 72(4): 869-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23959464

ABSTRACT

PURPOSE: Multikinase growth inhibitors inhibit their target kinases with varying potency. Patients often require lower doses or therapy breaks due to drug toxicities. To evaluate the effects of drug withdrawal on hepatocellular carcinoma cells after incubation with growth-inhibitory concentrations of regorafenib, cell growth, migration and invasion, and signaling were examined. METHODS: Cell proliferation, motility, and invasion were analyzed by MTT, wound healing, and invasion assays, respectively, and MAPK pathway protein markers were analyzed by Western blot. RESULTS: After regorafenib removal, cell growth, migration, and invasion recovered. Repeated drug exposure resulted in changes in cell growth patterns. Recovery could be blocked by sub-growth-inhibitory concentrations of either doxorubicin or vitamin K1. Recovery of growth was associated with increased phospho-JNK, phospho-p38, and phospho-STAT3 levels. The recovery of growth, migration, and signaling were blocked by a JNK inhibitor. CONCLUSIONS: Removal of regorafenib from growth-inhibited cells resulted in a JNK-dependent recovery of growth and migration.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Antineoplastic Agents/administration & dosage , Blotting, Western , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Hep G2 Cells , Humans , Liver Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Neoplasm Invasiveness , Phenylurea Compounds/administration & dosage , Pyridines/administration & dosage , Vitamin K 1/administration & dosage , Vitamin K 1/pharmacology
14.
J Cell Physiol ; 228(2): 292-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22777740

ABSTRACT

To evaluate the growth-inhibitory properties of the potent multi-kinase antagonist Regorafenib (Fluoro-Sorafenib), which was synthesized as a more potent Sorafenib, a Raf inhibitor and to determine whether similar mechanisms were involved, human hepatoma cell lines were grown in the presence or absence of Regorafanib and examined for growth inhibition. Western blots were performed for Raf targets, apoptosis, and autophagy. Regorafenib inhibited growth of human Hep3B, PLC/PRF/5, and HepG2 cells in a concentration- and time-dependent manner. Multiple signaling pathways were altered, including MAP kinases phospho-ERK and phospho-JNK and its target phospho-c-Jun. There was evidence for apoptosis by FACS, cleavage of caspases and increased Bax levels; as well as induction of autophagy, as judged by increased Beclin-1 and LC3 (II) levels. Prolonged drug exposure resulted in cell quiescence. Full growth recovery occurred after drug removal, unlike with doxorubicin chemotherapy. Regorafenib is a potent inhibitor of cell growth. Cells surviving Regorafenib treatment remain viable, but quiescent and capable of regrowth following drug removal. The reversibility of tumor cell growth suppression after drug removal may have clinical implications.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Liver Neoplasms/metabolism , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Signal Transduction/drug effects
15.
J Cell Physiol ; 228(6): 1344-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23169148

ABSTRACT

Sorafenib was shown in clinical trial to enhance survival in hepatocellular carcinoma (HCC) patients, but with minimal tumor shrinkage. To correlate several indices of HCC growth at various drug concentrations, HCC cells were grown in various low concentrations of two multikinase inhibitors, regorafenib (Stivarga) and sorafenib (Nexavar) and their effects were examined on alpha-fetoprotein (AFP), cell growth, migration, and invasion. In two AFP positive human HCC cell lines, AFP was inhibited at 0.1-1 µM drug concentrations. Cell migration and invasion were also inhibited at similar low drug concentrations. However, 10-fold higher drug concentrations were required to inhibit cell growth in both AFP positive and negative cells. To investigate this concentration discrepancy of effects, cells were then grown for prolonged times and sub-cultured in low drug concentrations and then their growth was re-tested. The growth in these drug-exposed cells was found to be slower than cells without prior drug exposure and they were also more sensitive to subsequent drug challenge. Evidence was also found for changes in cell signaling pathways in these slow-growth cells. Low multikinase inhibitor concentrations thus modulate several aspects of HCC cell biology.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/enzymology , Cell Movement/drug effects , Cell Proliferation/drug effects , Liver Neoplasms/enzymology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , alpha-Fetoproteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplasm Invasiveness , Niacinamide/pharmacology , Signal Transduction/drug effects , Sorafenib , Time Factors , alpha-Fetoproteins/genetics
16.
Cancer Biol Ther ; 12(6): 531-8, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21734462

ABSTRACT

Sorafenib is an FDA-approved agent for treatment of human hepatocellular carcinoma (HCC), but tumor shrinkage is minor. We therefore developed a strategy to combine K vitamins with sorafenib to treat HCC, and found that this combination enhanced sorafenib-induced HCC cell growth inhibition. To explore the mechanisms involved, we examined the role of Raf kinase, since both vitamins K and sorafenib were reported to inhibit tumor cell growth via Raf signaling pathway. We found that whereas lower concentration of vitamin K1 (25 µM) or sorafenib (2.5 µM) alone slightly induced c-Raf phosphorylation at both Ser-43 and Ser-259, combination vitamin K1 plus sorafenib resulted in strong c-Raf phosphorylation at these two serine residues. A Raf kinase activity assay confirmed that combination vitamin K1 plus sorafenib had a synergistic inhibitory effect on it. Since c-Raf phosphorylation at Ser-43 and Ser-259 can be regulated by either PKA or Akt kinase, we examined the effects of both vitamin K1 and sorafenib on their phosphorylation. Although vitamin K1 or sorafenib alone induced PKA phosphorylation, no enhanced phosphorylation effects on PKA were found using this combination. However, vitamin K1 enhanced sorafenib-induced c-Met phosphorylation at Tyr-1349, a DEP-1 protein phosphatase acting site, and consequently induced phosphorylation of PI3K-Akt. Both PI3K inhibitor Ly294002 as well as dominate negative Akt plasmid transfection antagonized vitamin K1 plus sorafenib actions on c-Raf phosphorylation and cell growth inhibition, suggesting that c-Met-PI3K-Akt signaling pathway mediated inhibitory c-Raf phosphorylation may play a central role in vitamin K1 plus sorafenib synergy in inhibiting HCC cell growth.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Liver Neoplasms, Experimental/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/administration & dosage , Carcinoma, Hepatocellular , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diethylnitrosamine , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , MAP Kinase Kinase Kinases/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphorylation , Pyridines/administration & dosage , Rats , Rats, Inbred F344 , Signal Transduction , Sorafenib , Vitamin K 1/administration & dosage
17.
Fertil Steril ; 96(1): 102-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21561608

ABSTRACT

OBJECTIVE: To determine whether decreased estrogen receptor alpha (ER-α) expression in endometriotic lesions could be balanced by an increased expression of estrogen receptor-related receptors (ERRs). To evaluate whether ERR-α expression is influenced by hormonal change in fertile and menopausal women. DESIGN: Prospective controlled study. SETTING: University Hospital, Department of Gynecology. PATIENT(S): Twenty-five women: 20 women of reproductive age with (n = 10) and without (control; n = 10) endometriosis and 5 menopausal women. INTERVENTION(S): Real-time polymerase chain reaction (qPCR). Immunohistochemistry. MAIN OUTCOME MEASURE(S): The ER and ERR expression levels were studied by reverse transcriptase-qPCR, ELISA, and immunohistochemistry using endometriotic and normal endometrial tissues. The ERR-α protein distribution was performed by immunohistochemistry in fertile and menopausal women. RESULT(S): Increased levels of ER-ß were associated with ER-α, ERR-α, and ERR-γ reductions in ectopic tissue but not in eutopic and normal endometria. Similar levels of ERR-ß were found in women with and without endometriosis. The ERR-α expression was similar in proliferative and secretory endometrial samples, whereas a down-regulation of this receptor was found in atrophic tissue. CONCLUSION(S): Our data confirm the up-regulation of ER-ß as the principal receptor involved in the progression of human endometriosis. In addition, we found that ERR-α seems to be unresponsive to hormonal changes during the menstrual cycle.


Subject(s)
Endometriosis/metabolism , Estrogen Receptor beta/physiology , Ovarian Cysts/metabolism , Adult , Disease Progression , Endometriosis/pathology , Estrogen Receptor beta/biosynthesis , Female , Humans , Male , Ovarian Cysts/pathology , Prospective Studies , Up-Regulation/physiology , Young Adult
18.
Anticancer Res ; 30(12): 5251-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21187522

ABSTRACT

BACKGROUND/AIM: The crucial role of KRAS status in new colorectal cancer target therapy raises the issue regarding which testing method to use. This study analysed 112 formalin fixed, paraffin-embedded (FFPE) metastatic tissue samples using three different commercially available kits. PATIENTS AND METHODS: A group of 40 KRAS wild-type (wt), 40 codon 12-mutated and 32 codon-13 mutated samples, previously evaluated by real-time PCR (TheraScreen kit), used as reference method, were analysed by Ampli-set-K-RAS and K-RAS StripAssay kit (herein called kit A and B, respectively) based on two different technologies. RESULTS: The sensitivity of both kits was 92.5% for wt samples, 100% and 95.0% for kit A and B, respectively for samples mutated in codon 12. The specificity was 100% for both kits for all groups of samples. After a minor modification of the kit A method, its specificity reached 100%. CONCLUSION: of low cost and easy to use, kit A may be suitable for use in a routine diagnostic setting.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Genes, ras , Reagent Strips , Codon , Colorectal Neoplasms/pathology , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Genotype , Humans , Mutation , Neoplasm Metastasis , Paraffin Embedding , Polymerase Chain Reaction/methods , Polymorphism, Restriction Fragment Length , Sensitivity and Specificity
20.
Reprod Biomed Online ; 19(6): 808-15, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20031021

ABSTRACT

The aim of this study was to evaluate the factors predisposing to implants of endometriotic lesions in normal ovarian cortexes of women with and without endometriosis by assessing the expression of pro-apoptotic and anti-apoptotic factors and follicular density. Ovarian biopsies were performed during laparoscopy in 18 patients with endometrioma and in 10 healthy women. Detection of apoptosis was performed with terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling assay. p53 and BCL2 proteins were assessed by immunohistochemistry. Quantitative real-time polymerase chain reaction was performed to evaluate BAX , BAK , BCL2 , BCL-XL , survivin and beta-actin ( ACTB ) expression. The p53 protein was positive in a significantly higher number of secondary follicles, whereas the B-cell chronic lymphocytic leukaemia/lymphoma 2 (BCL2) protein was positive in all follicles in unaffected tissue of endometriotic women, compared with the controls. Overexpression of the BCL2 and survivin genes and a decreased BAX and BAK gene expression were observed in the endometriotic group although only the difference in survivin expression was significant (P = 0.016). The BCL2 / BAX ratio showed an increased value in the ovarian cortex in controls compared with endometriosis patients. In conclusion, the reduction of apoptosis in unaffected tissue in women with endometriosis suggests that they may be predisposed to develop endometriosis.


Subject(s)
Apoptosis/physiology , Endometriosis/pathology , Ovary/pathology , Actins/metabolism , Adult , Endometriosis/metabolism , Female , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Ovarian Follicle/metabolism , Ovary/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Survivin , Tumor Suppressor Protein p53/biosynthesis , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...