Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(3)2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35158962

ABSTRACT

Although fluoropyrimidines were introduced as anticancer agents over 60 years ago, they are still the backbone of many combination chemotherapy regimens for the treatment of solid cancers. Like other chemotherapeutic agents, the therapeutic efficacy of fluoropyrimidines can be affected by drug resistance and severe toxicities; thus, novel therapeutic approaches are required to potentiate their efficacy and overcome drug resistance. In the last 20 years, the deregulation of epigenetic mechanisms has been shown to contribute to cancer hallmarks. Histone modifications play an important role in directing the transcriptional machinery and therefore represent interesting druggable targets. In this review, we focused on histone deacetylase inhibitors (HDACis) that can increase antitumor efficacy and overcome resistance to fluoropyrimidines by targeting specific genes or proteins. Our preclinical data showed a strong synergistic interaction between HDACi and fluoropyrimidines in different cancer models, but the clinical studies did not seem to confirm these observations. Most likely, the introduction of increasingly complex preclinical models, both in vitro and in vivo, cannot recapitulate human complexity; however, our analysis of clinical studies revealed that most of them were designed without a mechanistic approach and, importantly, without careful patient selection.

2.
Mol Oncol ; 15(4): 1005-1023, 2021 04.
Article in English | MEDLINE | ID: mdl-33331136

ABSTRACT

Acquired resistance to platinum (Pt)-based therapies is an urgent unmet need in the management of epithelial ovarian cancer (EOC) patients. Here, we characterized by an unbiased proteomics method three isogenic EOC models of acquired Pt resistance (TOV-112D, OVSAHO, and MDAH-2774). Using this approach, we identified several differentially expressed proteins in Pt-resistant (Pt-res) compared to parental cells and the chaperone HSP90 as a central hub of these protein networks. Accordingly, up-regulation of HSP90 was observed in all Pt-res cells and heat-shock protein 90 alpha isoform knockout resensitizes Pt-res cells to cisplatin (CDDP) treatment. Moreover, pharmacological HSP90 inhibition using two different inhibitors [17-(allylamino)-17-demethoxygeldanamycin (17AAG) and ganetespib] synergizes with CDDP in killing Pt-res cells in all tested models. Mechanistically, genetic or pharmacological HSP90 inhibition plus CDDP -induced apoptosis and increased DNA damage, particularly in Pt-res cells. Importantly, the antitumor activities of HSP90 inhibitors (HSP90i) were confirmed both ex vivo in primary cultures derived from Pt-res EOC patients ascites and in vivo in a xenograft model. Collectively, our data suggest an innovative antitumor strategy, based on Pt compounds plus HSP90i, to rechallenge Pt-res EOC patients that might warrant further clinical evaluation.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Drug Resistance, Neoplasm , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Platinum/therapeutic use , Animals , Benzoquinones , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Humans , Lactams, Macrocyclic , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Proteomics , Triazoles , Xenograft Model Antitumor Assays
3.
J Exp Clin Cancer Res ; 39(1): 213, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33032653

ABSTRACT

BACKGROUND: Despite the introduction of several novel therapeutic approaches that improved survival, metastatic castration-resistant prostate cancer (mCRPC) remains an incurable disease. Herein we report the synergistic antitumor interaction between two well-known drugs used for years in clinical practice, the antiepileptic agent with histone deacetylase inhibitory activity valproic acid and the cholesterol lowering agent simvastatin, in mCRPC models. METHODS: Synergistic anti-tumor effect was assessed on PC3, 22Rv1, DU145, DU145R80, LNCaP prostate cancer cell lines and EPN normal prostate epithelial cells, by calculating combination index (CI), caspase 3/7 activation and colony formation assays as well as on tumor spheroids and microtissues scored with luminescence 3D-cell viability assay. Cancer stem cells (CSC) compartment was studied evaluating specific markers by RT-PCR, western blotting and flow cytometry as well as by limiting dilution assay. Cholesterol content was evaluated by 1H-NMR. Overexpression of wild-type YAP and constitutively active YAP5SA were obtained by lipofectamine-based transfection and evaluated by immunofluorescence, western blotting and RT-PCR. 22Rv1 R_39 docetaxel resistant cells were selected by stepwise exposure to increasing drug concentrations. In vivo experiments were performed on xenograft models of DU145R80, 22Rv1 parental and docetaxel resistant cells, in athymic mice. RESULTS: We demonstrated the capacity of the combined approach to target CSC compartment by a novel molecular mechanism based on the inhibition of YAP oncogene via concurrent modulation of mevalonate pathway and AMPK. Because both CSCs and YAP activation have been associated with chemo-resistance, we tested if the combined approach can potentiate docetaxel, a standard of care in mCRCP treatment. Indeed, we demonstrated, both in vitro and in vivo models, the ability of valproic acid/simvastatin combination to sensitize mCRPC cells to docetaxel and to revert docetaxel-resistance, by mevalonate pathway/YAP axis modulation. CONCLUSION: Overall, mCRPC progression and therapeutic resistance driven by CSCs via YAP, can be tackled by the combined repurposing of two generic and safe drugs, an approach that warrants further clinical development in this disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Docetaxel/administration & dosage , Drug Resistance, Neoplasm , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Simvastatin/administration & dosage , Tumor Cells, Cultured , Valproic Acid/administration & dosage , Xenograft Model Antitumor Assays
4.
J Exp Clin Cancer Res ; 38(1): 317, 2019 Jul 18.
Article in English | MEDLINE | ID: mdl-31319863

ABSTRACT

BACKGROUND: Molecular markers for prostate cancer (PCa) are required to improve the early definition of patient outcomes. Atypically large extracellular vesicles (EVs), referred as "Large Oncosomes" (LO), have been identified in highly migratory and invasive PCa cells. We recently developed and characterized the DU145R80 subline, selected from parental DU145 cells as resistant to inhibitors of mevalonate pathway. DU145R80 showed different proteomic profile compared to parental DU145 cells, along with altered cytoskeleton dynamics and a more aggressive phenotype. METHODS: Immunofluorescence staining and western blotting were used to identify blebbing and EVs protein cargo. EVs, purified by gradient ultra-centrifugations, were analyzed by tunable resistive pulse sensing and multi-parametric flow cytometry approach coupled with high-resolution imaging technologies. LO functional effects were tested in vitro by adhesion and invasion assays and in vivo xenograft model in nude mice. Xenograft and patient tumor tissues were analyzed by immunohistochemistry. RESULTS: We found spontaneous blebbing and increased shedding of LO from DU145R80 compared to DU145 cells. LO from DU145R80, compared to those from DU145, carried increased amounts of key-molecules involved in PCa progression including integrin alpha V (αV-integrin). By incubating DU145 cells with DU145R80-derived LO we demonstrated that αV-integrin on LO surface was functionally involved in the increased adhesion and invasion of recipient cells, via AKT. Indeed either the pre-incubation of LO with an αV-integrin blocking antibody, or a specific AKT inhibition in recipient cells are able to revert the LO-induced functional effects. Moreover, DU145R80-derived LO also increased DU145 tumor engraftment in a mice model. Finally, we identified αV-integrin positive LO-like structures in tumor xenografts as well as in PCa patient tissues. Increased αV-integrin tumor expression correlated with high Gleason score and lymph node status. CONCLUSIONS: Overall, this study is the first to demonstrate the critical role of αV-integrin positive LO in PCa aggressive features, adding new insights in biological function of these large EVs and suggesting their potential use as PCa prognostic markers.


Subject(s)
Extracellular Vesicles/pathology , Integrin alphaV/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Extracellular Vesicles/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Transplantation , Prostatic Neoplasms/metabolism , Proteomics/methods , Up-Regulation
5.
J Cell Physiol ; 234(6): 9077-9092, 2019 06.
Article in English | MEDLINE | ID: mdl-30362533

ABSTRACT

Although platinum-based chemotherapy remains the standard-of-care for most patients with advanced non-small-cell lung cancer (NSCLC), acquired resistance occurs frequently predicting poor prognosis. To examine the mechanisms underlying platinum resistance, we have generated and characterized by proteomic approach the resistant A549 CDDP-resistant (CPr-A549) and their parental A549 cells, identifying 15 proteins differentially expressed (13 upregulated and 2 downregulated in CPr-A549). In details, we highlighted a coherent network of proteins clustering together and involved in altered protein folding and endoplasmic reticulum stress, correlated with epithelial to mesenchymal transition process and cancer stem cell markers, where vimentin played a hierarchical role, ultimately resulting in increased aggressive features. By using publicly available databases we showed that the modulated proteins could contribute to NSCLC carcinogenesis and correlate with NSCLC patients prognosis and survival probability, suggesting that they can be used as novel potential prognostic/predictive biomarkers or therapeutic targets to overcome platinum-resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Proteomics , Vimentin/metabolism , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Databases, Protein , Endoplasmic Reticulum Stress , Epithelial-Mesenchymal Transition , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Neoplastic Stem Cells/metabolism , Protein Folding , Protein Interaction Maps , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Recent Pat Anticancer Drug Discov ; 13(2): 184-200, 2018.
Article in English | MEDLINE | ID: mdl-29189178

ABSTRACT

BACKGROUND: Modifications of lipid metabolism have been progressively accepted as a hallmark of tumor cells and in particular, an elevated lipogenesis has been described in various types of cancers. OBJECTIVE: Important or deregulated activity of the mevalonate pathway has been demonstrated in different tumors and a wide range of studies have suggested that tumor cells are more dependent on the unceasing availability of mevalonate pathway metabolites than their non-malignant complements. METHODS: This study provides an overview of the state of the art of statins treatment on human cancer. RESULTS: In recent times, various actions have been proposed for statins in different physiological and pathological conditions beyond anti-inflammation and neuroprotection activity. Statins have been shown to act through mevalonate-dependent and -independent mechanisms able to affect several tissue functions and modulating specific signal transduction pathways that could account for statin pleiotropic effect. Based on their characteristics, statins represent ideal candidates for repositioning in cancer therapy. CONCLUSION: In this review article, we provide an overview of the current preclinical and clinical status of statins as antitumor agents. In addition, we evaluated various patents that describe the role of mevalonate pathway inhibitors and methods to determine if cancer cells are sensitive to statins treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Drug Repositioning/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Mevalonic Acid/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Drug Delivery Systems/trends , Drug Repositioning/trends , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Mevalonic Acid/metabolism , Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Amino Acids ; 49(3): 517-528, 2017 03.
Article in English | MEDLINE | ID: mdl-27761756

ABSTRACT

Vorinostat demonstrated preclinical and clinical efficacy in human cancers and is the first histone deacetylase inhibitor (HDACi) approved for cancer treatment. Tissue transglutaminase (TG2) is a multifunctional enzyme that catalyzes a Ca2+ dependent transamidating reaction resulting in covalent cross-links between proteins. TG2 acts also as G-protein in trans-membrane signaling and as a cell surface adhesion mediator. TG2 up-regulation has been demonstrated in several cancers and its expression levels correlate with resistance to chemotherapy and metastatic potential. We demonstrated that the anti-proliferative effect of the HDACi vorinostat is paralleled by the induction of TG2 mRNA and protein expression in cancer cells but not in ex vivo treated peripheral blood lymphocytes. This effect was also shared by other pan-HDACi and resulted in increased TG2 transamidating activity. Notably, high TG2 basal levels in a panel of cancer cell lines correlated with lower vorinostat antiproliferative activity. Notably, in TG2-knockdown cancer cells vorinostat anti-proliferative and pro-apoptotic effects were enhanced, whereas in TG2-full-length transfected cells were impaired, suggesting that TG2 could represent a mechanism of intrinsic or acquired resistance to vorinostat. In fact, co-treatment of tumor cells with inhibitors of TG2 transamidating activity potentiated the antitumor effect of vorinostat. Moreover, vorinostat-resistant MCF7 cells selected by stepwise increasing concentrations of the drug, significantly overexpressed TG2 protein compared to parental cells, and co-treatment of these cells with TG2 inhibitors reversed vorinostat-resistance. Taken together, our data demonstrated that TG2 is involved in the resistance of cancer cells to vorinostat, as well as to other HDACi.


Subject(s)
Antineoplastic Agents/pharmacology , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Transglutaminases/genetics , A549 Cells , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , HCT116 Cells , HT29 Cells , Histone Deacetylases/metabolism , Humans , Isoxazoles/pharmacology , MCF-7 Cells , Protein Glutamine gamma Glutamyltransferase 2 , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transglutaminases/antagonists & inhibitors , Transglutaminases/metabolism , Vorinostat
8.
Mol Oncol ; 10(8): 1344-62, 2016 10.
Article in English | MEDLINE | ID: mdl-27499265

ABSTRACT

Peroxisome proliferator-activated receptor-γ (PPARγ) is a transcription factor of the nuclear hormone receptor superfamily implicated in a wide range of processes, including tumorigenesis. Its role in colorectal cancer (CRC) is still debated; most reports support that PPARγ reduced expression is associated with poor prognosis. We employed 2-Dimensional Differential InGel Electrophoresis (2-D DIGE) followed by Liquid Chromatography (LC)-tandem Mass Spectrometry (MS/MS) to identify differentially expressed proteins and the molecular pathways underlying PPARγ expression in CRC progression. We identified several differentially expressed proteins in HT29 and HCT116 CRC cells and derived clones either silenced or overexpressing PPARγ, respectively. In Ingenuity Pathway Analysis (IPA) they showed reciprocal relation with PPARγ and a strong relationship with networks linked to cell death, growth and survival. Interestingly, five of the identified proteins, ezrin (EZR), isoform C of prelamin-A/C (LMNA), alpha-enolase (ENOA), prohibitin (PHB) and RuvB-like 2 (RUVBL2) were shared by the two cell models with opposite expression levels, suggesting a possible regulation by PPARγ. mRNA and western blot analysis were undertaken to obtain a technical validation and confirm the expression trend observed by 2-D DIGE data. We associated EZR upregulation with increased cell surface localization in PPARγ-overexpressing cells by flow cytometry and immunofluorescence staining. We also correlated EZR and PPARγ expression in our series of CRC specimens and the expression profiling of all five proteins levels in the publicly available colon cancer genomic data from Oncomine and Cancer Genome Atlas (TCGA) colon adenocarcinoma (COAD) datasets. In summary, we identified a panel of proteins correlated with PPARγ expression that could be associated with CRC unveiling new pathways to be investigated for the selection of novel potential prognostic/predictive biomarkers and/or therapeutic targets.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Silencing , PPAR gamma/metabolism , Protein Interaction Maps , Proteomics/methods , Blotting, Western , Computational Biology , Cytoskeletal Proteins/metabolism , Databases as Topic , Electrophoresis, Gel, Two-Dimensional , HCT116 Cells , HT29 Cells , Humans , Immunoblotting , Mass Spectrometry , Phenotype , Prohibitins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Subcellular Fractions/metabolism
9.
Int J Pharm ; 511(1): 127-139, 2016 Sep 10.
Article in English | MEDLINE | ID: mdl-27374195

ABSTRACT

With the aim to develop alternative therapeutic tools for the treatment of resistant cancers, here we propose targeted Pluronic(®) P123/F127 mixed micelles (PMM) delivering niclosamide (NCL) as a repositioning strategy to treat multidrug resistant non-small lung cancer cell lines. To build multifunctional PMM for targeting and imaging, Pluronic(®) F127 was conjugated with biotin, while Pluronic(®) P123 was fluorescently tagged with rhodamine B, in both cases at one of the two hydroxyl end groups. This design intended to avoid any interference of rhodamine B on biotin exposition on PMM surface, which is a key fundamental for cell trafficking studies. Biotin-decorated PMM were internalized more efficiently than non-targeted PMM in A549 lung cancer cells, while very low internalization was found in NHI3T3 normal fibroblasts. Biotin-decorated PMM entrapped NCL with good efficiency, displayed sustained drug release in protein-rich media and improved cytotoxicity in A549 cells as compared to free NCL (P<0.01). To go in depth into the actual therapeutic potential of NCL-loaded PMM, a cisplatin-resistant A549 lung cancer cell line (CPr-A549) was developed and its multidrug resistance tested against common chemotherapeutics. Free NCL was able to overcome chemoresistance showing cytotoxic effects in this cell line ascribable to nucleolar stress, which was associated to a significant increase of the ribosomal protein rpL3 and consequent up-regulation of p21. It is noteworthy that biotin-decorated PMM carrying NCL at low doses demonstrated a significantly higher cytotoxicity than free NCL in CPr-A549. These results point at NCL-based regimen with targeted PMM as a possible second-line chemotherapy for lung cancer showing cisplatin or multidrug resistance.


Subject(s)
Biotin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms , Niclosamide/administration & dosage , Poloxalene/administration & dosage , Poloxamer/administration & dosage , A549 Cells , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Biotin/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/physiology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Micelles , NIH 3T3 Cells , Niclosamide/metabolism , Poloxalene/metabolism , Poloxamer/metabolism , Ribosomal Protein L3
10.
Oncotarget ; 6(28): 25076-92, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26312765

ABSTRACT

In this study, we have characterized the role of annexin A1 (ANXA1) in the acquisition and maintenance of stem-like/aggressive features in prostate cancer (PCa) cells comparing zoledronic acid (ZA)-resistant DU145R80 with their parental DU145 cells. ANXA1 is over-expressed in DU145R80 cells and its down-regulation abolishes their resistance to ZA. Moreover, ANXA1 induces DU145 and DU145R80 invasiveness acting through formyl peptide receptors (FPRs). Also, ANXA1 knockdown is able to inhibit epithelial to mesenchymal transition (EMT) and to reduce focal adhesion kinase (FAK) and metalloproteases (MMP)-2/9 expression in PCa cells. DU145R80 show a cancer stem cell (CSC)-like signature with a high expression of CSC markers including CD44, CD133, NANOG, Snail, Oct4 and ALDH7A1 and CSC-related genes as STAT3. Interestingly, ANXA1 knockdown induces these cells to revert from a putative prostate CSC to a more differentiated phenotype resembling DU145 PCa cell signature. Similar results are obtained concerning some drug resistance-related genes such as ATP Binding Cassette G2 (ABCG2) and Lung Resistant Protein (LRP). Our study provides new insights on the role of ANXA1 protein in PCa onset and progression.


Subject(s)
Annexin A1/metabolism , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Diphosphonates/pharmacology , Drug Resistance, Neoplasm , Imidazoles/pharmacology , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Annexin A1/genetics , Biomarkers, Tumor/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Shape/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , Signal Transduction/drug effects , Transfection , Zoledronic Acid
11.
Oncotarget ; 6(7): 5324-41, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25481874

ABSTRACT

Proteomic analysis identified differentially expressed proteins between zoledronic acid-resistant and aggressive DU145R80 prostate cancer (PCa) cells and their parental DU145 cells. Ingenuity Pathway Analysis (IPA) showed a strong relationship between the identified proteins within a network associated with cancer and with homogeneous cellular functions prevalently related with regulation of cell organization, movement and consistent with the smaller and reduced cell-cell contact morphology of DU145R80 cells. The identified proteins correlated in publically available human PCa genomic data with increased tumor expression and aggressiveness. DU145R80 exhibit also a clear increase of alpha-v-(αv) integrin, and of urokinase receptor (uPAR), both included within the same network of the identified proteins. Interestingly, the actin-rich structures localized at the cell periphery of DU145R80 cells are rich of Filamin A, one of the identified proteins and uPAR which, in turn, co-localizes with αv-integrin, in podosomes and/or invadopodia. Notably, the invasive feature of DU145R80 may be prevented by blocking anti-αv antibody. Overall, we unveil a signaling network that physically links the interior of the nucleus via the cytoskeleton to the extracellular matrix and that could dictate PCa aggressiveness suggesting novel potential prognostic markers and therapeutic targets for PCa patients.


Subject(s)
Bone Density Conservation Agents/pharmacology , Diphosphonates/pharmacology , Drug Resistance, Neoplasm , Imidazoles/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics/methods , Signal Transduction/drug effects , Blotting, Western , Cell Movement , Cell Proliferation , Electrophoresis, Gel, Two-Dimensional , Gene Ontology , Gene Regulatory Networks , Humans , Male , Neoplasm Invasiveness , Prostatic Neoplasms/drug therapy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured , Zoledronic Acid
12.
Molecules ; 18(3): 2549-62, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23442931

ABSTRACT

The need to explore new alternative therapeutic strategies and chemoprevention methods for hepatocellular carcinoma is growing significantly. Selenium is a trace element that plays a critical role in physiological processes, and is used in cancer chemoprevention. The aim of this work was to test in vitro the effect of sodium selenite on the human hepatoma cell lines, HepG2 and Huh7, to assess its effect on the expression of GPX1, SELK and SELENBP1 and also to evaluate its action on inflammation determinants such as cytokines. Our results show that: (i) the increase observed for the GPX1 and SELK expression is correlated with an increase in the sodium selenite concentration, also evidencing an inverse association between the levels of these two proteins and SELENBP1; (ii) the selenium concentrations evaluated in protein extracts increase in proportional way with the selenite concentrations used in the treatment, suggesting that other selenoproteins can also be modulated and should be evaluated in further studies, and (iii) some cytokines, VEGF and three pro-inflammatory cytokines, i.e., IL-6, IL-8, and IL-17, decreased with an increasing selenite concentration. Finally, interactomic studies show that GPX1 and SELK, and the four pro-inflammatory cytokines are functionally correlated evidencing a putative anti-inflammatory role for the selenite.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cytokines/metabolism , Liver Neoplasms/metabolism , Selenoproteins/metabolism , Sodium Selenite/pharmacology , Cell Line, Tumor , Glutathione Peroxidase/metabolism , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Selenium-Binding Proteins/metabolism , Glutathione Peroxidase GPX1
13.
Proteomics ; 11(18): 3725-42, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21761561

ABSTRACT

Several solid tumors are characterized by poor prognosis and few effective treatment options, other than palliative chemotherapy in the recurrent/metastatic setting. Epidermal growth factor receptor (EGFR) has been considered an important anticancer target because it is involved in the development and progression of several solid tumors; however, only a subset of patients show a clinically meaningful response to EGFR inhibition, particularly to EGFR tyrosine kinase inhibitors such as gefitinib. We have recently demonstrated synergistic antitumor effect of the histone deacetylase inhibitor vorinostat combined with gefitinib. To further characterize the interaction between these two agents, cellular extracts from Hep-2 cancer cells that were untreated or treated for 24 h with either vorinostat or gefitinib alone or with a vorinostat/gefitinib combination were analyzed using 2-D DIGE. Software analysis using DeCyder was performed, and numerous differentially expressed protein spots were visualized between the four examined settings. Using MALDI-TOF MS and ESI-Ion trap MS/MS, several differentially expressed proteins were identified; some of these were validated by Western blotting. Finally, a pathway analysis of experimental data performed using MetaCore highlighted a relevant relationship between the identified proteins and additional potential effectors. In conclusion, we performed a comprehensive analysis of proteins regulated by vorinostat and gefitinib, alone and in combination, providing a useful insight into their mechanisms of action as well as their synergistic interaction.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Quinazolines/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Drug Synergism , Gefitinib , Gene Expression Regulation, Neoplastic , Humans , Mass Spectrometry/methods , Peptide Mapping , Protein Isoforms/chemistry , Proteomics , Software , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Two-Dimensional Difference Gel Electrophoresis , Vorinostat
14.
FASEB J ; 24(8): 2881-92, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20354138

ABSTRACT

Medulloblastoma is the most common malignant brain tumor in children, and despite improvements in the overall survival rate, it still lacks an effective treatment. Src plays an important role in cancer, and recently high Src activity was documented in medulloblastoma. In this report, we examined the effects of novel pyrazolo-[3,4-d]-pyrimidine derivative Src inhibitors in medulloblastoma. By MTS assay, we showed that the pyrimidine derivatives indicated as S7, S29, and SI163 greatly reduce the growth rate of medulloblastoma cells by inhibiting Src phosphorylation, compared with HT22 non-neoplastic nerve cells. These compounds also halt cells in the G(2)/M phase, and this effect likely occurs through the regulation of cdc2 and CDC25C phosphorylation, as shown by Western blot. Moreover, the exposure to pyrimidine derivatives induces apoptosis, assayed by the supravital propidium iodide assay, through modulation of the apoptotic proteins Bax and Bcl2, and inhibits tumor growth in vivo in a mouse model. Notably, S7, S29, and SI163 show major inhibitory effects on medulloblastoma cell growth compared with the chemotherapeutic agents cisplatin and etoposide. In conclusion, our results suggest that S7, S29, and SI163 could be novel attractive candidates for the treatment of medulloblastoma or tumors characterized by high Src activity.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Medulloblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , src-Family Kinases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Humans , Medulloblastoma/pathology , Protein Kinase Inhibitors/chemistry , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
15.
Extremophiles ; 14(2): 193-204, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20049620

ABSTRACT

The gene encoding a novel alcohol dehydrogenase that belongs to the short-chain dehydrogenases/reductases (SDRs) superfamily was identified in the aerobic thermoacidophilic crenarchaeon Sulfolobus acidocaldarius strain DSM 639. The saadh gene was heterologously overexpressed in Escherichia coli, and the protein (SaADH) was purified to homogeneity and characterized. SaADH is a tetrameric enzyme consisting of identical 28,978-Da subunits, each composed of 264 amino acids. The enzyme has remarkable thermophilicity and thermal stability, displaying activity at temperatures up to 75 degrees C and a 30-min half-inactivation temperature of ~90 degrees C, and shows good tolerance to common organic solvents. SaADH has a strict requirement for NAD(H) as the coenzyme, and displays a preference for the reduction of alicyclic, bicyclic and aromatic ketones and alpha-keto esters, but is poorly active on aliphatic, cyclic and aromatic alcohols, and shows no activity on aldehydes. The enzyme catalyses the reduction of alpha-methyl and alpha-ethyl benzoylformate, and methyl o-chlorobenzoylformate with 100% conversion to methyl (S)-mandelate [17% enantiomeric excess (ee)], ethyl (R)-mandelate (50% ee), and methyl (R)-o-chloromandelate (72% ee), respectively, with an efficient in situ NADH-recycling system which involves glucose and a thermophilic glucose dehydrogenase. This study provides further evidence supporting the critical role of the D37 residue in discriminating NAD(H) from NAD(P)H in members of the SDR superfamily.


Subject(s)
Alcohol Oxidoreductases/chemistry , Archaeal Proteins/chemistry , Sulfolobus acidocaldarius/enzymology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Base Sequence , Cloning, Molecular , DNA Primers/genetics , DNA, Archaeal/genetics , Enzyme Stability , Escherichia coli/genetics , Genes, Archaeal , Hydrogen-Ion Concentration , Kinetics , NAD/metabolism , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Sulfolobus acidocaldarius/genetics , Thermodynamics
16.
Appl Environ Microbiol ; 74(13): 3949-58, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18456852

ABSTRACT

The gene encoding a novel alcohol dehydrogenase (ADH) that belongs to the short-chain dehydrogenase/reductase (SDR) superfamily was identified in the extremely thermophilic, halotolerant gram-negative eubacterium Thermus thermophilus HB27. The T. thermophilus ADH gene (adh(Tt)) was heterologously overexpressed in Escherichia coli, and the protein (ADH(Tt)) was purified to homogeneity and characterized. ADH(Tt) is a tetrameric enzyme consisting of identical 26,961-Da subunits composed of 256 amino acids. The enzyme has remarkable thermophilicity and thermal stability, displaying activity at temperatures up to approximately 73 degrees C and a 30-min half-inactivation temperature of approximately 90 degrees C, as well as good tolerance to common organic solvents. ADH(Tt) has a strict requirement for NAD(H) as the coenzyme, a preference for reduction of aromatic ketones and alpha-keto esters, and poor activity on aromatic alcohols and aldehydes. This thermophilic enzyme catalyzes the following reactions with Prelog specificity: the reduction of acetophenone, 2,2,2-trifluoroacetophenone, alpha-tetralone, and alpha-methyl and alpha-ethyl benzoylformates to (S)-(-)-1-phenylethanol (>99% enantiomeric excess [ee]), (R)-alpha-(trifluoromethyl)benzyl alcohol (93% ee), (S)-alpha-tetralol (>99% ee), methyl (R)-(-)-mandelate (92% ee), and ethyl (R)-(-)-mandelate (95% ee), respectively, by way of an efficient in situ NADH-recycling system involving 2-propanol and a second thermophilic ADH. This study further supports the critical role of the D37 residue in discriminating NAD(H) from NADP(H) in members of the SDR superfamily.


Subject(s)
Alcohol Dehydrogenase/isolation & purification , Alcohol Dehydrogenase/metabolism , Thermus thermophilus/enzymology , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/genetics , Amino Acid Sequence , Enzyme Stability , Escherichia coli/enzymology , Escherichia coli/genetics , Hot Temperature , Kinetics , Molecular Sequence Data , NAD/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Stereoisomerism , Substrate Specificity , Thermus thermophilus/genetics
17.
Nucleic Acids Res ; 36(10): 3235-43, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18417534

ABSTRACT

The Mini-Chromosome Maintenance (MCM) proteins are candidates of replicative DNA helicase in eukarya and archaea. Here we report a 2.8 A crystal structure of the N-terminal domain (residues 1-268) of the Sulfolobus solfataricus MCM (Sso MCM) protein. The structure reveals single-hexameric ring-like architecture, at variance from the protein of Methanothermobacter thermoautotrophicus (Mth). Moreover, the central channel in Sso MCM seems significantly narrower than the Mth counterpart, which appears to more favorably accommodate single-stranded DNA than double-stranded DNA, as supported by DNA-binding assays. Structural analysis also highlights the essential role played by the zinc-binding domain in the interaction with nucleic acids and allows us to speculate that the Sso MCM N-ter domain may function as a molecular clamp to grasp the single-stranded DNA passing through the central channel. On this basis possible DNA unwinding mechanisms are discussed.


Subject(s)
Bacterial Proteins/chemistry , DNA Helicases/chemistry , Sulfolobus solfataricus/enzymology , Amino Acid Sequence , Archaeal Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , DNA Helicases/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Methanobacteriaceae/enzymology , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry , Sequence Deletion , Structural Homology, Protein , Zinc/chemistry
18.
Biochem J ; 408(1): 87-95, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17683280

ABSTRACT

To protect their genetic material cells adopt different mechanisms linked to DNA replication, recombination and repair. Several proteins function at the interface of these DNA transactions. In the present study, we report on the identification of a novel archaeal DNA helicase. BlastP searches of the Sulfolobus solfataricus genome database allowed us to identify an open reading frame (SSO0112, 875 amino acid residues) having sequence similarity with the human RecQ5beta. The corresponding protein, termed Hel112 by us, was produced in Escherichia coli in soluble form, purified to homogeneity and characterized. Gel-filtration chromatography and glycerol-gradient sedimentation analyses revealed that Hel112 forms monomers and dimers in solution. Biochemical characterization of the two oligomeric species revealed that only the monomeric form has an ATP-dependent 3'-5' DNA-helicase activity, whereas, unexpectedly, both the monomeric and dimeric forms possess DNA strand-annealing capability. The Hel112 monomeric form is able to unwind forked and 3'-tailed DNA structures with high efficiency, whereas it is almost inactive on blunt-ended duplexes and bubble-containing molecules. This analysis reveals that S. solfataricus Hel112 shares some enzymatic features with the RecQ-like DNA helicases and suggests potential cellular functions of this protein.


Subject(s)
DNA Helicases/metabolism , DNA/metabolism , Sulfolobus solfataricus/enzymology , Adenosine Triphosphate/metabolism , Catalysis , DNA Helicases/classification , Dimerization , Hydrolysis , Protein Binding , Substrate Specificity
19.
J Biol Chem ; 282(17): 12574-82, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17337732

ABSTRACT

Mini-chromosome maintenance (MCM) proteins form ring-like hexameric complexes that are commonly believed to act as the replicative DNA helicase at the eukaryotic/archaeal DNA replication fork. Because of their simplified composition with respect to the eukaryotic counterparts, the archaeal MCM complexes represent a good model system to use in analyzing the structural/functional relationships of these important replication factors. In this study the domain organization of the MCM-like protein from Sulfolobus solfataricus (Sso MCM) has been dissected by trypsin partial proteolysis. Three truncated derivatives of Sso MCM corresponding to protease-resistant domains were produced as soluble recombinant proteins and purified: the N-terminal domain (N-ter, residues 1-268); a fragment comprising the AAA+ and C-terminal domains (AAA+-C-ter, residues 269-686); and the C-terminal domain (C-ter, residues 504-686). All of the purified recombinant proteins behaved as monomers in solution as determined by analytical gel filtration chromatography, suggesting that the polypeptide chain integrity is required for stable oligomerization of Sso MCM. However, the AAA+-C-ter derivative, which includes the AAA+ motor domain and retains ATPase activity, was able to form dimers in solution when ATP was present, as analyzed by size exclusion chromatography and glycerol gradient sedimentation analyses. Interestingly, the AAA+-C-ter protein could displace oligonucleotides annealed to M13 single-stranded DNA although with a reduced efficiency in comparison with the full-sized Sso MCM. The implications of these findings for understanding the DNA helicase mechanism of the MCM complex are discussed.


Subject(s)
Adenosine Triphosphatases/metabolism , Archaeal Proteins/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Models, Biological , Sulfolobus solfataricus/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Archaeal Proteins/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Protein Structure, Tertiary/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Structure-Activity Relationship , Sulfolobus solfataricus/genetics
20.
Extremophiles ; 11(2): 277-82, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17082970

ABSTRACT

The hyper-thermophilic archaeon Sulfolobus solfataricus possesses two functional DNA polymerases belonging to the B-family (Sso DNA pol B1) and to the Y-family (Sso DNA pol Y1). Sso DNA pol B1 recognizes the presence of uracil and hypoxanthine in the template strand and stalls synthesis 3-4 bases upstream of this lesion ("read-ahead" function). On the other hand, Sso DNA pol Y1 is able to synthesize across these and other lesions on the template strand. Herein we report evidence that Sso DNA pol B1 physically interacts with DNA pol Y1 by surface plasmon resonance measurements and immuno-precipitation experiments. The region of DNA pol B1 responsible for this interaction has been mapped in the central portion of the polypeptide chain (from the amino acid residue 482 to 617), which includes an extended protease hyper-sensitive linker between the N- and C-terminal modules (amino acid residues Asn482-Ala497) and the alpha-helices forming the "fingers" sub-domain (alpha-helices R, R' and S). These results have important implications for understanding the polymerase-switching mechanism on the damaged template strand during genome replication in S. solfataricus.


Subject(s)
Archaeal Proteins/metabolism , DNA Replication/physiology , DNA-Directed DNA Polymerase/metabolism , Genome, Archaeal/physiology , Sulfolobus solfataricus/enzymology , Archaeal Proteins/chemistry , DNA Damage , DNA-Directed DNA Polymerase/chemistry , Protein Binding , Protein Structure, Secondary , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...