Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 11636, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406187

ABSTRACT

A large number of basic researches and observational studies suggested the cancer preventive activity of vitamin E, but large-scale human intervention trials have yielded disappointing results and actually showed a higher incidence of prostate cancer although the mechanisms underlying the increased risk remain largely unknown. Here we show through in vitro and in vivo studies that vitamin E produces a marked inductive effect on carcinogen-bioactivating enzymes and a pro-oxidant status promoting both DNA damage and cell transformation frequency. First, we found that vitamin E in the human prostate epithelial RWPE-1 cell line has the remarkable ability to upregulate the expression of various phase-I activating cytochrome P450 (CYP) enzymes, including activators of polycyclic aromatic hydrocarbons (PAHs), giving rise to supraphysiological levels of reactive oxygen species. Furthermore, our rat model confirmed that vitamin E in the prostate has a powerful booster effect on CYP enzymes associated with the generation of oxidative stress, thereby favoring lipid-derived electrophile spread that covalently modifies proteins. We show that vitamin E not only causes DNA damage but also promotes cell transformation frequency induced by the PAH-prototype benzo[a]pyrene. Our findings might explain why dietary supplementation with vitamin E increases the prostate cancer risk among healthy men.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Cytochrome P-450 Enzyme System/metabolism , Dietary Supplements/toxicity , Neoplasms, Experimental/chemically induced , Prostatic Neoplasms/chemically induced , Vitamin E/toxicity , 3T3 Cells , Animals , Benzo(a)pyrene/toxicity , Carcinogens/toxicity , Cell Line , Cell Transformation, Neoplastic/genetics , DNA Damage/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lipid Peroxidation/drug effects , Male , Mice , Micronuclei, Chromosome-Defective/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oxidative Stress/drug effects , Prostate/cytology , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Rats , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , Vitamin E/administration & dosage
2.
Cell Death Differ ; 26(9): 1845-1858, 2019 09.
Article in English | MEDLINE | ID: mdl-30622304

ABSTRACT

Current literature agrees on the notion that efficient DNA repair favors longevity across evolution. The DNA damage response machinery activates inflammation and type I interferon signaling. Both pathways play an acknowledged role in the pathogenesis of a variety of age-related diseases and are expected to be detrimental for human longevity. Here, we report on the anti-inflammatory molecular make-up of centenarian's fibroblasts (low levels of IL-6, type 1 interferon beta, and pro-inflammatory microRNAs), which is coupled with low level of DNA damage (measured by comet assay and histone-2AX activation) and preserved telomere length. In the same cells, high levels of the RNAseH2C enzyme subunit and low amounts of RNAseH2 substrates, i.e. cytoplasmic RNA:DNA hybrids are present. Moreover, RNAseH2C locus is hypo-methylated and RNAseH2C knock-down up-regulates IL-6 and type 1 interferon beta in centenarian's fibroblasts. Interestingly, RNAseH2C locus is hyper-methylated in vitro senescent cells and in tissues from atherosclerotic plaques and breast tumors. Finally, extracellular vesicles from centenarian's cells up-regulate RNAseH2C expression and dampen the pro-inflammatory phenotype of fibroblasts, myeloid, and cancer cells. These data suggest that centenarians are endowed with restrained DNA damage-induced inflammatory response, that may facilitate their escape from the deleterious effects of age-related chronic inflammation.


Subject(s)
Cellular Senescence/genetics , DNA Damage/genetics , Extracellular Vesicles/metabolism , Fibroblasts/metabolism , Inflammation/metabolism , Longevity/genetics , Ribonuclease H/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , DNA Damage/drug effects , Extracellular Vesicles/immunology , Female , Fibroblasts/enzymology , Genetic Loci , Humans , Inflammation/genetics , Interferon-beta/metabolism , Interleukin-6/metabolism , Longevity/physiology , Male , Methylation , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Plaque, Atherosclerotic/chemistry , Plaque, Atherosclerotic/genetics , Ribonuclease H/genetics , Telomere Homeostasis/genetics , Young Adult
3.
Nutrients ; 10(9)2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30135355

ABSTRACT

We conducted an in vitro study combining a rexinoid, 6-OH-11-O-hydroxyphenanthrene (IIF), and epigallocatechin-3-gallate (EGCG), which is the main catechin of green tea, on BE(2)-C, a neuroblastoma cell line representative of the high-risk group of patients. Neuroblastoma is the most common malignancy of childhood: high-risk patients, having N-MYC over-expression, undergo aggressive therapy and show high mortality or an increased risk of secondary malignancies. Retinoids are used in neuroblastoma therapy with incomplete success: the association of a second molecule might improve the efficacy. BE(2)-C cells were treated by EGCG and IIF, individually or in combination: cell viability, as evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, was reduced, EGCG+IIF being the most effective treatment. Apoptosis occurred and the EGCG+IIF treatment decreased N-MYC protein expression and molecular markers of invasion (MMP-2, MMP-9 and COX-2). Zymography demonstrated nearly 50% inhibition of MMP activity. When BE(2)-C cells were grown in non-adherent conditions to enrich the tumor-initiating cell population, BE(2)-C-spheres were obtained. After 48 h and 72 h treatment, EGCG+IIF limited BE(2)-C-sphere formation and elicited cell death with a reduction of N-MYC expression. We concluded that the association of EGCG to IIF might be applied without toxic effects to overcome the incomplete success of retinoid treatments in neuroblastoma patients.


Subject(s)
Catechin/analogs & derivatives , Neoplastic Stem Cells/drug effects , Neuroblastoma/pathology , Phenanthrenes/pharmacology , Apoptosis/drug effects , Catechin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Neoplastic Stem Cells/metabolism , Tea/chemistry
4.
Biosci Rep ; 37(3)2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28465354

ABSTRACT

Epidermal growth factor receptor (EGFR) expression is an important marker in breast carcinoma pathology and is considered a pivotal molecule for cancer cell proliferation, invasion and metastasis. We investigated the effects of epigallocatechin-3-gallate (EGCG), the most active green tea catechin, in combination with 6-OH-11-O-hydroxyphenanthrene (IIF), a synthetic retinoid X receptor-γ (RXRγ) agonist, on three breast carcinoma cell lines: MCF-7, MCF-7TAM and MDA-MB-231. EGFR and AKT activation and molecular markers of cell motility and migration (CD44, extracellular matrix metalloproteinase (MMP) inducer (EMMPRIN), MMP-2, MMP-9 and tissue inhibitor of metalloproteinases (TIMPs)) were studied after EGCG and IIF treatments. The EGCG + IIF treatment was the most active in down-regulating EGFR phosphorylation at Tyr1068 in all the investigated cell lines; p473AKT was also down-regulated in MCF-TAM cells. EGCG + IIF was also the most active treatment in reducing the expression of markers of invasion and migration in all the three cell lines: CD44, EMMPRIN, MMP-2 and -9 expression decreased, whereas TIMPs were up-regulated. Zymography and scratch assay also confirmed the reduced invasion tendency. We considered that EGCG and IIF treatments could alter the molecular network based on EGFR, CD44 and EMMPRIN expression interdependence and reduced the migration tendency in MCF-7, MCF-7TAM and MDA-MB-231 cells. These events only occurred in association with AKT inactivation in MCF-7TAM cells. In conclusion, the combination of EGCG and IIF significantly attenuated the invasive behaviour of breast carcinoma cells.


Subject(s)
Breast Neoplasms/drug therapy , Catechin/analogs & derivatives , Cell Movement/drug effects , ErbB Receptors/antagonists & inhibitors , Neoplasm Invasiveness/pathology , Phenanthrenes/pharmacology , Basigin/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catechin/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism
5.
Naunyn Schmiedebergs Arch Pharmacol ; 389(11): 1171-1182, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27449069

ABSTRACT

Dynamin GTPase (Dyn) plays a critical role in membrane-remodelling events underlying endocytosis. Studies in Drosophila identified a functional interaction between the Dyn homologue, encoded by the shibire (shi) gene, and Abnormal wing discs (Awd), a nucleoside diphosphate kinase (NDPK) that is the homologue of group I Nme human genes. These Drosophila studies showed that awd mutations enhance mutant shi phenotype and thus indicated the existence of a highly specific interaction between these genes. Furthermore, in human cells, it has been shown that Nme proteins promote Dyn activity in different membrane compartments through spatially controlled supply of GTP. Interestingly, Awd and Nme proteins have been detected in the extracellular environment. While no role has been inferred to extracellular Awd, presence of Nme1 in cancer patient serum is an unfavourable prognostic marker. In the present work, we used Drosophila and human cell line models to investigate the shuttling Awd/Nme1 proteins between intracellular and extracellular spaces. By using classic and reverse genetic approaches, we show that downregulation of Shi/Dyn1 activity enhances extracellular Awd/Nme1 in both Drosophila and human colon cell lines. We extended our analyses to colon cancer cell lines and found that knocking down Dyn1, besides to raise Nme1 extracellular amount, downregulates expression of molecular components that play key roles in tumour invasion. Interestingly, in vivo analyses of Drosophila larval adipocytes show that the conditional block of Shi activity greatly reduces intracellular amount of Awd confirming that Shi plays a key role in controlling the balance between intracellular and extracellular Awd.


Subject(s)
Colonic Neoplasms/enzymology , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Dynamin I/metabolism , Dynamins/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Nucleoside-Diphosphate Kinase/metabolism , Adipocytes/enzymology , Animals , Animals, Genetically Modified , Colonic Neoplasms/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Dynamin I/genetics , Dynamins/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genotype , HT29 Cells , Humans , Larva/enzymology , Mutation , NM23 Nucleoside Diphosphate Kinases/genetics , Nucleoside-Diphosphate Kinase/genetics , Phenotype , RNA Interference , Transfection
6.
Future Oncol ; 12(14): 1699-711, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27173589

ABSTRACT

AIM: To investigate mechanisms by which doxorubicin (DOX) and cisplatin (CIS) cause human ovarian stroma injury. PATIENTS & METHODS: Stromal cells from human cryopreserved ovarian tissue were cultured in the presence of 1 µM DOX and 10 µM CIS. Ovarian damage induced by treatments was evaluated by 'Live/Dead' and sulforhodamine-B assays, the expression of different apoptosis markers. RESULTS: Stromal cell growth was inhibited by DOX and CIS, and this effect was accompanied by apoptosis through mitochondrial pathway activation: Bax, cleaved-caspase 9, cleaved-PARP1 induction and Akt1, Bcl2, phospho-44/42-MAPK/ERK1/2 reduction were observed. CONCLUSION: DOX and CIS induced apoptosis in human ovarian stromal cells. Knowledge of mechanisms by which the drugs act is important to identify possible ways to counteract side effects of chemotherapy on ovaries.


Subject(s)
Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Cisplatin/adverse effects , Doxorubicin/adverse effects , Ovary/drug effects , Adult , Blotting, Western , Cell Survival/drug effects , Cryopreservation , Female , Flow Cytometry , Humans , Immunohistochemistry , Microscopy, Electron, Transmission , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/drug effects
7.
World J Stem Cells ; 8(3): 62-72, 2016 Mar 26.
Article in English | MEDLINE | ID: mdl-27022437

ABSTRACT

The recapitulation of primary tumour heterogenity and the existence of a minor sub-population of cancer cells, capable of initiating tumour growth in xenografts on serial passages, led to the hypothesis that cancer stem cells (CSCs) exist. CSCs are present in many tumours, among which is breast cancer. Breast CSCs (BCSCs) are likely to sustain the growth of the primary tumour mass, as well as to be responsible for disease relapse and metastatic spreading. Consequently, BCSCs represent the most significant target for new drugs in breast cancer therapy. Both the hypoxic condition in BCSCs biology and pro-inflammatory cytokine network has gained increasing importance in the recent past. Breast stromal cells are crucial components of the tumours milieu and are a major source of inflammatory mediators. Recently, the anti-inflammatory role of some nuclear receptors ligands has emerged in several diseases, including breast cancer. Therefore, the use of nuclear receptors ligands may be a valid strategy to inhibit BCSCs viability and consequently breast cancer growth and disease relapse.

8.
J Cell Physiol ; 231(7): 1534-41, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26553365

ABSTRACT

The hypoxic environment is a crucial component of the cancer stem cell niche and it is capable of eliciting stem cell features in cancer cells. We previously reported that SNAI2 up-regulates the expression of Carbonic Anhydrase iso-enzyme 9 (CA9) in hypoxic MCF7 cells. Here we show that SNAI2 down-regulates miR34a expression in hypoxic MCF7 cell-derived mammospheres. Next, we report on the capability of miR34a to decrease CA9 mRNA stability and CA9 protein expression. We also convey that the over-expression of cloned CA9-mRNA-3'UTR increases the mRNA half-life and protein levels of two miR34a targets JAGGED1 and NOTCH3. The data here reported shows that the SNAI2-dependent down-regulation of miR34a substantially contributes to the post-transcriptional up-regulation of CA9, and that CA9-mRNA-3'UTR acts as an endogenous microRNA sponge. We conclude that CA9/miR34 interplay shares in the hypoxic regulation of mammospheres and therefore, may play a relevant role in the hypoxic breast cancer stem cell niche.


Subject(s)
Antigens, Neoplasm/biosynthesis , Breast Neoplasms/genetics , Carbonic Anhydrases/biosynthesis , Cell Hypoxia/genetics , MicroRNAs/genetics , Antigens, Neoplasm/genetics , Breast Neoplasms/pathology , Calcium-Binding Proteins/biosynthesis , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Jagged-1 Protein , MCF-7 Cells , Membrane Proteins/biosynthesis , MicroRNAs/biosynthesis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptor, Notch3 , Receptors, Notch/biosynthesis , Serrate-Jagged Proteins , Snail Family Transcription Factors , Transcription Factors/biosynthesis
9.
Curr Cancer Drug Targets ; 16(4): 373-85, 2016.
Article in English | MEDLINE | ID: mdl-26278714

ABSTRACT

Molecules with synergistic effects often enhance the benefits of cancer therapy. We observed that the major catechin of green tea, (-)-Epigallocatechin-3-gallate (EGCG), induced retinoid X receptor-γ (RXRγ) expression in the SK-Ch-A1 cholangiocarcinoma cell line and in two colon carcinoma cell lines (LoVo and the derivative multi-drug resistant LoVoMDR). On this basis, we analyzed the effects of EGCG in combination with an RXRγ ligand, 6-OH-11-O-hydroxyphenantrene (IIF), or with a ligand of retinoic acid receptor, all-trans-retinoic acid (RA). IIF alone and in combination with EGCG activated the retinoic X response elements and induced the germ cell nuclear factor. In parallel, EGCG induced 67 kDa laminin receptor expression alone and in combination with IIF. We observed a synergistic growth inhibition with EGCG and IIF in combination at lower doses. These effects were accompanied by apoptosis activation through the mitochondrial pathway. Moreover, in LoVo cell line we observed an induction of Forkhead box O3 expression, another molecule involved in apoptosis activation. Finally, metalloproteinase activity and extracellular matrix metalloproteinase inducer (EMMPRIN) expression were inhibited and tumor cell invasion was strongly reduced in the SK-Ch-A1 cell line after treatment with EGCG and IIF. In conclusion, the use of specific RXR ligands in combination with catechins could open a new perspective in gastrointestinal tumor chemoprevention.


Subject(s)
Apoptosis/drug effects , Catechin/analogs & derivatives , Gastrointestinal Neoplasms/drug therapy , Phenanthrenes/pharmacology , Retinoid X Receptor gamma/metabolism , Catechin/pharmacology , Cell Line, Tumor , Drug Synergism , Forkhead Box Protein O3/metabolism , Gastrointestinal Neoplasms/metabolism , Humans , Ligands , Metalloproteases/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Tea/metabolism , Tretinoin/metabolism
10.
Biomed Res Int ; 2014: 853086, 2014.
Article in English | MEDLINE | ID: mdl-25013807

ABSTRACT

(-)-Epigallocatechin-3-gallate (EGCG) and chemotherapeutic agents cotreatment can improve cytotoxicity against cancer cells. We showed that EGCG and the rexinoid 6-OH-11-O-hydroxyphenanthrene (IIF), given together, were cytotoxic toward MCF-7, MCF-7TAM, and MDA-MB-231, three breast carcinoma cell lines showing different molecular characteristics. Cell growth arrest and apoptosis were greater after EGCG and IIF cotreatment than after individual administration. Cytotoxicity was related to upregulation of 67-kDa laminin receptor (LR67), one of the principal molecular targets of EGCG, and activation of the nuclear retinoic X receptors (RXRs) pathway. Furthermore, the transcription factor Forkhead box O3 (Foxo3a), a protein able to trigger apoptosis through upregulation of genes necessary for cell death, was activated. EGCG and IIF cotreatment produced a significant nuclear import of Foxo3a from the cytoplasm in MCF-7, MCF-7TAM, and MDA-MB-231 cells. In MCF-7TAM cells only, Foxo3a nuclear localization was associated with p473AKT downregulation. For the first time we showed that when EGCG and IIF, two harmless molecules, were given together, they might increase cytotoxicity in three breast carcinoma cell lines, two of them being representative of poorly responsive breast carcinoma types.


Subject(s)
Breast Neoplasms/drug therapy , Catechin/analogs & derivatives , Phenanthrenes/administration & dosage , Retinoid X Receptors/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Catechin/administration & dosage , Cell Proliferation/drug effects , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , MCF-7 Cells , Retinoid X Receptors/genetics
11.
J Cell Physiol ; 229(11): 1595-606, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24604522

ABSTRACT

Cancer stem cells (CSCs) are affected by the local micro-environment, the niche, in which inflammatory stimuli and hypoxia act as steering factors. Here, two nuclear receptors (NRs) agonists, i.e. pioglitazone (PGZ), a ligand of peroxisome proliferator activated receptor-γ, and 6-OH-11-O-hydroxyphenanthrene (IIF), a ligand of retinoid X receptors, were investigated for their capability to interference with the cross-talk between breast CSCs and the niche compartment. We found that IIF potentiates the ability of PGZ to hamper the mammospheres-forming capability of human breast tumours and MCF7 cancer cells, reducing the expression of CSCs regulatory genes (Notch3, Jagged1, SLUG, Interleukin-6, Apolipoprotein E, Hypoxia inducible factor-1α and Carbonic anhydrase IX). Notably, these effects are not observed in normal-MS obtained from human breast tissue. Importantly, NRs agonists abolish the capability of hypoxic MCF7 derived exosomes to induce a pro-inflammatory phenotype in mammary glands fibroblasts. Moreover, NRs agonist also directly acts on breast tumour associated fibroblasts to downregulate nuclear factor-κB pathway and metalloproteinases (MMP2 and MMP9) expression and activity. In conclusion, NRs agonists disrupt the inflammatory cross-talk of the hypoxic breast CSCs niche.


Subject(s)
Breast Neoplasms/pathology , Inflammation/pathology , Neoplastic Stem Cells/pathology , PPAR gamma/metabolism , Receptor Cross-Talk , Retinoid X Receptors/metabolism , Stem Cell Niche , Antigens, Neoplasm/metabolism , Apolipoproteins E/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Carbonic Anhydrase IX , Carbonic Anhydrases/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Exosomes/drug effects , Exosomes/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Ligands , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Models, Biological , NF-kappa B/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Phenanthrenes/pharmacology , Pioglitazone , Receptor Cross-Talk/drug effects , Retinoid X Receptors/agonists , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Stem Cell Niche/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Thiazolidinediones/pharmacology
12.
Pancreas ; 43(1): 53-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24201777

ABSTRACT

OBJECTIVES: Resistance to gemcitabine is one of the main causes of treatment failure in pancreatic cancer. Compelling evidences have shown the involvement of nuclear factor κB (NF-κB) activation in such phenomenon. The protease inhibitor gabexate mesilate has been shown to inhibit NF-κB. We here investigated if combined treatment with this drug could improve gemcitabine antitumoral efficacy in pancreatic cancer cells. METHODS: The effect of gabexate mesilate and gemcitabine, both used at concentrations achievable in human plasma, was assessed on in vitro pancreatic cancer cell growth, invasion, and tumor angiogenesis. The molecular mechanism at the basis of these effects was also investigated. RESULTS: Gabexate mesilate significantly increased gemcitabine anti-invasive and antiangiogenic efficacy. This effect was related to inhibition of gemcitabine-induced NF-κB activation by gabexate mesilate, which prevented RelA/p65 nuclear translocation and resulted in metalloproteinase 2, metalloproteinase 9, vascular endothelial growth factor, and interleukin 8 down-regulation. Combined treatment with gabexate mesilate also inhibited gemcitabine-induced extracellular-regulated kinase 1/2 and AKT activation by increased expression of Raf kinase inhibitor protein and phosphatase and tensin homolog. CONCLUSIONS: Combined treatment with gabexate mesilate sensitizes pancreatic cancer cells to gemcitabine by inhibition of the NF-κB pathway. The efficacy of this therapeutic strategy in pancreatic cancer patients remains to be established and deserves future clinical investigation.


Subject(s)
Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Gabexate/pharmacology , NF-kappa B/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blotting, Western , Cell Line , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Down-Regulation/drug effects , Drug Evaluation, Preclinical/methods , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gabexate/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serine Proteinase Inhibitors/administration & dosage , Serine Proteinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Gemcitabine
13.
Am J Pathol ; 183(5): 1688-1697, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036252

ABSTRACT

Cancer stem cell survival relies on the activation of inflammatory pathways, which is speculatively triggered by cell autonomous mechanisms or by microenvironmental stimuli. Here, we observed that hypoxic bone marrow stroma-derived transforming growth factor-ß 1 promotes the growth of human breast cancer stem cells as mammospheres. The ensuing Slug-dependent serine 139 phosphorylation of the DNA damage sensor H2AX in breast cancer stem cells induces tumor necrosis factor-α and IL-8 mRNAs, whose stability is enhanced by cytoplasmic ß-catenin. ß-Catenin also up-regulates and binds miR-221, reducing the stability of the miR-221 targets Rad51 and ERα mRNAs. Our data show that the Slug/ß-catenin-dependent activation of DNA damage signaling triggered by the hypoxic microenvironment sustains the proinflammatory phenotype of breast cancer stem cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Inflammation/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Transcription Factors/metabolism , beta Catenin/metabolism , Autocrine Communication/drug effects , Autocrine Communication/genetics , Breast Neoplasms/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cytokines/genetics , Cytokines/metabolism , DNA Damage/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , Inflammation/genetics , MCF-7 Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Models, Biological , Neoplastic Stem Cells/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Serine/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Snail Family Transcription Factors , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/metabolism
14.
PLoS One ; 8(1): e54968, 2013.
Article in English | MEDLINE | ID: mdl-23372804

ABSTRACT

AIMS: Cancer stem cell biology is tightly connected to the regulation of the pro-inflammatory cytokine network. The concept of cancer stem cells "inflammatory addiction" leads to envisage the potential role of anti-inflammatory molecules as new anti-cancer targets. Here we report on the relationship between nuclear receptors activity and the modulation of the pro-inflammatory phenotype in breast cancer stem cells. METHODS: Breast cancer stem cells were expanded as mammospheres from normal and tumor human breast tissues and from tumorigenic (MCF7) and non tumorigenic (MCF10) human breast cell lines. Mammospheres were exposed to the supernatant of breast tumor and normal mammary gland tissue fibroblasts. RESULTS: In mammospheres exposed to the breast tumor fibroblasts supernatant, autocrine tumor necrosis factor-α signalling engenders the functional interplay between peroxisome proliferator activated receptor-α and hypoxia inducible factor-1α (PPARα/HIF1α). The two proteins promote mammospheres formation and enhance each other expression via miRNA130b/miRNA17-5p-dependent mechanism which is antagonized by PPARγ. Further, the PPARα/HIF1α interplay regulates the expression of the pro-inflammatory cytokine interleukin-6, the hypoxia survival factor carbonic anhydrase IX and the plasma lipid carrier apolipoprotein E. CONCLUSION: Our data demonstrate the importance of exploring the role of nuclear receptors (PPARα/PPARγ) in the regulation of pro-inflammatory pathways, with the aim to thwart breast cancer stem cells functioning.


Subject(s)
Apolipoproteins E/metabolism , Breast Neoplasms/metabolism , Carbonic Anhydrases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplastic Stem Cells/metabolism , PPAR alpha/metabolism , Apolipoproteins E/genetics , Autocrine Communication , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , PPAR alpha/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , Protein Binding , Spheroids, Cellular , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
15.
Food Chem ; 138(2-3): 1521-30, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23411276

ABSTRACT

Cytotoxic effects of the combination of the food components vitexin-2-O-xyloside (X), raphasatin (4-methylsulphanyl-3-butenyl isothiocyanates; G) and (-)-epigallocatechin-3-gallate (E) were investigated in colon (LoVo and CaCo-2) and breast (MDA-MB-231 and MCF-7) cancer cells. Breast cancer cells were more resistant than colon cells to X, G and E inhibition. On the contrary, marked synergistic effects among X, G and E on cell growth were found in both colon cancer cells. Further analysis revealed a G0/G1 arrest of the phase cell progression and apoptosis, linked to modulation of Bax, Bcl2, caspase-9 and poly(ADP-ribose) polymerase as well as Reactive Oxygen Species (ROS) generation in both colon cancer cells, whereas apoptosis and ROS were not significantly detected in normal human lymphocytes. We conclude that the X, G and E mixture might act by mitochondrial pathway activation of apoptosis, possibly elicited by ROS and the mixture may be effective in the chemoprevention of colon cancer.


Subject(s)
Apigenin/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Colonic Neoplasms/physiopathology , Isothiocyanates/pharmacology , Caspase 3/metabolism , Catechin/pharmacology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Drug Synergism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
16.
Nanomedicine ; 9(5): 675-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23219878

ABSTRACT

Colitis is the term used for chronic inflammatory bowel diseases at substantially increased risk of developing a form of colorectal cancer (CRC) known as colitis-associated cancer. In our study we synthesized core-shell polymeric micelles obtained by self-assembly of block copolymers for high efficiency delivery of anti-inflammatory and anti-cancer compounds to colonocytes and colon mucosa. We achieved an efficient intracellular delivery of these hydrophobic compounds (prednisone, retinoic acid and doxorubicin) to cultured colonocytes without cellular toxicity. The efficacy of retinoic acid and doxorubicin administration was significantly increased using these nanosized carriers. Moreover, these polymeric micelles have been shown to overcome the multidrug resistance efflux mechanism effectively delivering doxorubicin to multidrug-resistant colon cancer cells. These nanocarriers are also suitable for selective in vivo delivery of lipophilic drugs by enema administration to the inflamed colon tissue, specifically targeting the inflamed mucosa. FROM THE CLINICAL EDITOR: This team of investigators studied polymeric micelles as highly efficient drug delivery systems enabling intracellular delivery of hydrophobic compounds (prednisone, retinoic acid, and doxorubicin) to cultured colonocytes without cellular toxicity, also demonstrating beneficial in vivo effects.


Subject(s)
Colitis/drug therapy , Colon/drug effects , Colonic Neoplasms/drug therapy , Polymers/administration & dosage , Animals , Colitis/complications , Colitis/pathology , Colon/cytology , Colonic Neoplasms/complications , Colonic Neoplasms/pathology , Doxorubicin/administration & dosage , Drug Delivery Systems , Drug Resistance, Multiple/drug effects , Humans , Mice , Micelles , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polymers/chemistry , Prednisone/administration & dosage , Tretinoin/administration & dosage
17.
J Mater Chem B ; 1(24): 3083-3098, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-32261012

ABSTRACT

Many whole cell-based assays in use today rely on flat, two-dimensional (2D) glass or plastic substrates that may not produce results characteristic of in vivo conditions. In this study, a three-dimensional (3D) cell-based assay scaffold was fabricated using a gas-in-foam templating technique. The scaffold was made of poly(vinyl alcohol), a water-soluble synthetic polymer with excellent film-forming, emulsifying, and biocompatible properties widely used in the biomedical field. The preliminary rheological studies on the solution of PVA and surfactant permitted us to disclose the significant physical parameters that influence the morphology of the ensuing materials. The scaffolds obtained were subjected to detailed analysis by light microscopy, Scanning Electron Microscopy (SEM), computed X-ray microtomography (µCT), infrared spectroscopy, and mechanical testing. Morphological investigations showed that the produced scaffolds are characterised by average void and interconnect diameters lying in the range of 200-300 and 30-150 µm, respectively, suitable for cell infiltration. Two different cross-linking procedures were adopted in order to modulate the mechanical properties of the PVA scaffolds. One made use of a bi-epoxide (PEGDGE), the other was based on glutaraldehyde (GA). The efficiency in terms of cross-linking density of the two procedures resulted in very different mechanical properties. Furthermore, in this article it is demonstrated how PVA foams can be processed into uniform, porous films suitable to be integrated with multi-well 2D culture plates in order to create a 3D analogue. The PEGDGE cross-linked scaffold was tested on C3A cells, a human hepatocyte cell line, representing an appropriate model for liver toxicity studies. Proliferation and cytotoxicity assays indicated good cell viability throughout the culture time, which was also confirmed by SEM analysis. Typical hepatic functions such as albumin and urea production and induction of Cyp3A4 enzyme activity following drug administration were satisfactory, thus proving the efficiency of this construct in maintaining specific liver functions.

18.
Toxicol In Vitro ; 26(1): 142-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22056261

ABSTRACT

Natural derivatives of vitamin A, including all-trans-retinoic acid (ATRA), commonly known as retinoids, currently produce favorable results in the treatment of many types of tumors. The rexinoid 6-OH-11-O-hydroxyphenantrene (IIF) is a synthetic derivative of ATRA. Previous in vitro and in vivo studies demonstrated that IIF is able to induce growth inhibition of various cancer cells and is a potent apoptosis-inducing agent with clinical potential. Osteosarcoma (OS) is the most common type of bone cancer, characterized by a rising aggressiveness. Recent evidences suggest that mesenchymal stem cells (MSC) may favour tumor growth and progression. Thus, it is important to investigate whether a compound with potential anti-tumoral properties such as IIF affects not only tumor cells but also MSC. The current study is an attempt to understand the mode of the potential cytotoxicity of IIF on OS cells and MSC. The response to IIF treatment of osteosarcoma SaOS-2, MG63, and U2OS cells and of bone marrow-derived MSC was the subject of investigation. The results showed that IIF significantly inhibited cell growth in OS cell lines and MSC in both a time- and dose-dependent manner, as evaluated by methylene blue assay. This was also associated with altered cell morphology and an increase in cell death with the involvement of apoptosis as demonstrated by NucleoCounter, Hoechst 33342 staining and FACS analysis. No cell death and apoptosis was found in U2OS cells. Analysis of cells treated with 20 and 40µM IIF for 24h by western blot suggests the activation of initiator caspase 9, indicating the involvement of caspases in inducing apoptosis. Furthermore, IIF upregulated the expression of the pro-apoptotic protein Bax and downregulated the anti-apoptotic protein Bcl2. For the first time, our results collectively provide an evidence for cell growth inhibition and activation of apoptosis in human OS cells and MSC by IIF. These results confirm that IIF may be an effective compound for anticancer treatment, including that of OS.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Mesenchymal Stem Cells/drug effects , Tretinoin/analogs & derivatives , Apoptosis/drug effects , Cell Line, Tumor , Cells, Cultured , Humans , Mesenchymal Stem Cells/pathology , Osteosarcoma , Tretinoin/pharmacology
19.
Biosci Rep ; 31(2): 99-108, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20446926

ABSTRACT

The activation of the EGFR (epidermal growth factor receptor) signalling pathway is one of the key mechanisms underlying the development of resistance to tamoxifen in breast cancer patients. As EGCG [(-)-epigallocatechin-3-gallate], the most active catechin present in green tea, has been shown to down-regulate EGFR, we studied the effects of 10-100 µg/ml EGCG treatment on growth and invasion in a breast carcinoma cell line resistant to tamoxifen [MCF-7Tam (MCF-7 breast carcinoma cell line resistant to tamoxifen) cells] and parental MCF-7. A dose-dependent down-regulation of EGFR mRNA expression and protein level occurred after 50 µg/ml EGCG treatment of MCF-7Tam cells. EGFR molecules on the plasma membrane surface of MCF-7Tam cells significantly decreased. EGFR phosphorylation (Tyr-992, Tyr-1045 and Tyr-1068) was higher in MCF-7Tam than in MCF-7 and it was reduced by EGCG treatment. ERK (extracellular regulated kinase) and phospho-ERK p42/44 were also down-regulated by EGCG treatment and in vitro cell growth and invasion decreased. MMP-2 (matrix metalloproteinase-2) and MMP-9, which are implicated in cell invasion and metastasis, and EMMPRIN (extracellular matrix metalloproteinase inducer), a glycoprotein able to activate MMPs, were significantly reduced after 50 µg/ml EGCG treatment. In keeping with this, TIMP-1 (tissue inhibitor of metalloproteinases-1) and TIMP-2, which down-regulate MMPs, increased after EGCG treatment. Altogether, the present data demonstrated that EGCG could attenuate the tamoxifen-resistant phenotype of MCF-7Tam cells. EGCG could stop MCF-7Tam cell growth and in vitro invasion through down-regulation of EGFR and other molecules implicated in aggressive biological behaviour. The present data support the hypothesis that EGCG is an interesting molecule to be investigated in tamoxifen-resistant breast carcinoma.


Subject(s)
Basigin/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Catechin/analogs & derivatives , ErbB Receptors/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Basigin/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Catechin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , RNA, Messenger/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tamoxifen/pharmacology , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/drug effects , Tissue Inhibitor of Metalloproteinase-2/genetics
20.
Cancer Lett ; 297(1): 65-74, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20510503

ABSTRACT

Nuclear retinoid X receptors (RXRs) and peroxisome proliferator-activated receptors (PPARs are potential candidates as drug target for cancer prevention and treatment. We investigated if the rexinoid 6-OH-11-O-hydroxyphenantrene (IIF) potentiates the antitumoral properties of PPARgamma ligands as ciglitazone and pioglitazone, on two colon cancer cell lines: HCA-7 and HCT-116. Drugs inhibited cell growth and induced apoptosis synergistically. The combination resulted in a decrease of cyclooxigenase-2, metalloproteinases-2 and -9 expression level and activity while PPARgamma, RXRgamma and tissue inhibitors of metalloproteinase-1 and -2 expression were increased. Finally, IIF potentiated PPAR transcriptional activity by enhancement of peroxisome proliferator response elements transactivation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , PPAR gamma/agonists , Retinoid X Receptor gamma/agonists , Apoptosis/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Drug Synergism , HCT116 Cells , Humans , Ligands , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , PPAR gamma/genetics , PPAR gamma/metabolism , Pioglitazone , Response Elements/drug effects , Retinoid X Receptor gamma/metabolism , Thiazolidinediones/pharmacology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Transcriptional Activation/drug effects , Tretinoin/analogs & derivatives , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...