Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Br J Cancer ; 118(2): 200-212, 2018 01.
Article in English | MEDLINE | ID: mdl-29161243

ABSTRACT

BACKGROUND: Platinum-based drugs such as Cisplatin are commonly employed for cancer treatment. Despite an initial therapeutic response, Cisplatin treatment often results in the development of chemoresistance. To identify novel approaches to overcome Cisplatin resistance, we tested Cisplatin in combination with K+ channel modulators on colorectal cancer (CRC) cells. METHODS: The functional expression of Ca2+-activated (KCa3.1, also known as KCNN4) and voltage-dependent (Kv11.1, also known as KCNH2 or hERG1) K+ channels was determined in two CRC cell lines (HCT-116 and HCT-8) by molecular and electrophysiological techniques. Cisplatin and several K+ channel modulators were tested in vitro for their action on K+ currents, cell vitality, apoptosis, cell cycle, proliferation, intracellular signalling and Platinum uptake. These effects were also analysed in a mouse model mimicking Cisplatin resistance. RESULTS: Cisplatin-resistant CRC cells expressed higher levels of KCa3.1 and Kv11.1 channels, compared with Cisplatin-sensitive CRC cells. In resistant cells, KCa3.1 activators (SKA-31) and Kv11.1 inhibitors (E4031) had a synergistic action with Cisplatin in triggering apoptosis and inhibiting proliferation. The effect was maximal when KCa3.1 activation and Kv11.1 inhibition were combined. In fact, similar results were produced by Riluzole, which is able to both activate KCa3.1 and inhibit Kv11.1. Cisplatin uptake into resistant cells depended on KCa3.1 channel activity, as it was potentiated by KCa3.1 activators. Kv11.1 blockade led to increased KCa3.1 expression and thereby stimulated Cisplatin uptake. Finally, the combined administration of a KCa3.1 activator and a Kv11.1 inhibitor also overcame Cisplatin resistance in vivo. CONCLUSIONS: As Riluzole, an activator of KCa3.1 and inhibitor of Kv11.1 channels, is in clinical use, our results suggest that this compound may be useful in the clinic to improve Cisplatin efficacy and overcome Cisplatin resistance in CRC.


Subject(s)
Cisplatin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , ERG1 Potassium Channel/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Apoptosis/drug effects , Benzothiazoles/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacokinetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Drug Synergism , ERG1 Potassium Channel/metabolism , HCT116 Cells , HT29 Cells , Humans , Inhibitory Concentration 50 , Mice , Potassium Channel Blockers/pharmacology , Pyrazoles/pharmacology , Riluzole/pharmacology
2.
Blood ; 117(3): 902-14, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21048156

ABSTRACT

Bone marrow mesenchymal cells (MSCs) can protect leukemic cells from chemotherapy, thus increasing their survival rate. We studied the potential molecular mechanisms underlying this effect in acute lymphoblastic leukemia (ALL) cells. Coculture of ALL cells with MSCs induced on the lymphoblast plasma membrane the expression of a signaling complex formed by hERG1 (human ether-à-go-go-related gene 1) channels, the ß(1)-integrin subunit, and the chemokine receptor CXC chemokine receptor-4. The assembly of such a protein complex activated both the extracellular signal-related kinase 1/2 (ERK1/2) and the phosphoinositide 3-kinase (PI3K)/Akt prosurvival signaling pathways. At the same time, ALL cells became markedly resistant to chemotherapy-induced apoptosis. hERG1 channel function appeared to be important for both the initiation of prosurvival signals and the development of drug resistance, because specific channel blockers decreased the protective effect of MSCs. NOD/SCID mice engrafted with ALL cells and treated with channel blockers showed reduced leukemic infiltration and had higher survival rates. Moreover, hERG1 blockade enhanced the therapeutic effect produced by corticosteroids. Our findings provide a rationale for clinical testing of hERG1 blockers in the context of antileukemic therapy for patients with ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Coculture Techniques , Doxorubicin/pharmacology , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Female , Humans , Integrin beta1/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Multiprotein Complexes/metabolism , Piperidines/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prednisone/pharmacology , Pyridines/pharmacology , RNA Interference , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Biomolecules ; 13(12)2023 12 06.
Article in English | MEDLINE | ID: mdl-38136626

ABSTRACT

Oxygen level is a key regulator of organogenesis and its modification in postnatal life alters the maturation process of organs, including the intestine, which do not completely develop in utero. The ß3-adrenoreceptor (ß3-AR) is expressed in the colon and has an oxygen-dependent regulatory mechanism. This study shows the effects of the ß3-AR agonist BRL37344 in a neonatal model of hyperoxia-driven colonic injury. For the first 14 days after birth, Sprague-Dawley rat pups were exposed to ambient oxygen levels (21%) or hyperoxia (85%) and treated daily with BRL37344 at 1, 3, 6 mg/kg or untreated. At the end of day 14, proximal colon samples were collected for analysis. Hyperoxia deeply influences the proximal colon development by reducing ß3-AR-expressing cells (27%), colonic length (26%) and mucin production (47%), and altering the neuronal chemical coding in the myenteric plexus without changes in the neuron number. The administration of BRL37344 at 3 mg/kg, but not at 1 mg/kg, significantly prevented these alterations. Conversely, it was ineffective in preventing hyperoxia-induced body weight loss. BRL37344 at 6 mg/kg was toxic. These findings pave the way for ß3-AR pharmacological targeting as a therapeutic option for diseases caused by hyperoxia-impaired development, typical prematurity disorders.


Subject(s)
Ethanolamines , Hyperoxia , Rats , Animals , Ethanolamines/pharmacology , Rats, Sprague-Dawley , Adrenergic Agonists , Receptors, Adrenergic, beta-3 , Oxygen
4.
Cancers (Basel) ; 13(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207243

ABSTRACT

Soft tissue sarcomas (STSs) are a heterogeneous group of rare tumors. Although constituting only 1% of all human malignancies, STSs represent the second most common type of solid tumors in children and adolescents and comprise an important group of secondary malignancies. Over 100 histologic subtypes have been characterized to date (occurring predominantly in the trunk, extremity, and retroperitoneum), and many more are being discovered due to molecular profiling. STS mortality remains high, despite adjuvant chemotherapy. New prognostic stratification markers are needed to help identify patients at risk of recurrence and possibly apply more intensive or novel treatments. Recent scientific advancements have enabled a more precise molecular characterization of sarcoma subtypes and revealed novel therapeutic targets and prognostic/predictive biomarkers. This review aims at providing a comprehensive overview of the most relevant cellular, molecular and metabolic biomarkers for STS, and highlight advances in STS-related biomarker research.

5.
Bioconjug Chem ; 21(8): 1432-8, 2010 Aug 18.
Article in English | MEDLINE | ID: mdl-20669938

ABSTRACT

Immunotherapy of tumors and of melanoma in particular has a long history, and recently this therapeutic approach found a reliable scientific rationale. This biological therapy aims to teach the patient's immune system to recognize the antigens expressed on tumor cells and destroy them, leaving normal cells intact. The success of this therapy highly depends on the selection of target antigens that are essential for tumors growth and progression. The overexpression of GM(3) ganglioside 1 and especially the expression of its metabolite GM(3) lactone 2 characterize murine and human melanomas, playing an important role in tumor progression and making such self-antigens potential targets for the immunotherapy of these neoplasms. Although more immunogenic than its precursor, GM(3) lactone 2 is unsuitable to be used in immunotherapy as a melanoma-associated antigen (MAA) because it is unstable under physiological conditions. We designed and synthesized the hydrolytically stable mimetic 3, which is remarkably simpler than the native lactone 2; after conjugation of 3 to the protein carrier keyhole-limpet hemocyanin (KLH), the obtained glycoprotein 5 was used as the immunogen in vivo to successfully elicit specific antimelanoma antibodies. In fact, no appreciable binding to GM(1) was observed. Capitalizing on the stability and on the reduced structural complexity of mimetic 3, the immunostimulant 5 we report represents a new promising synthetic glycoconjugate for the immunotherapy of melanoma.


Subject(s)
Antibodies/immunology , G(M3) Ganglioside/analogs & derivatives , Melanoma/immunology , Molecular Mimicry/immunology , Animals , Antibodies/chemistry , Antibody Specificity , Antigen-Antibody Reactions , Carbohydrate Conformation , Computer Simulation , G(M3) Ganglioside/chemistry , G(M3) Ganglioside/genetics , G(M3) Ganglioside/immunology , Hemocyanins/chemistry , Hemocyanins/immunology , Humans , Immunotherapy , Melanoma/genetics , Melanoma/therapy , Mice , Molecular Mimicry/genetics
6.
Adv Exp Med Biol ; 674: 23-32, 2010.
Article in English | MEDLINE | ID: mdl-20549937

ABSTRACT

Protein-protein interactions between integrins and ion channels consist in a complicated bidirectional talk, not yet understood in detail, which triggers a downstream signaling network. Such a coordinated process occurs in discrete, localized microcompartments and involves different membrane and cytoplasmic proteins. Since the early nineties, when the first functional association between integrins and ion channels was characterized, the number of similar examples is constantly increasing. Identifying the components of this pathway has general importance for cell physiology and will eventually lead to fully understand the role of ion channels in the physiological processes typically controlled by integrin receptors, such as cell adhesion, migration and proliferation. Here, we detail the main experimental methods currently available to study these processes and discuss their advantages and disadvantages. Biochemical copurification and genetic interaction studies, as well as high-throughput screening, can be performed to initially identify the interacting proteins. Successively, in vitro binding assays such as pull-down and immunoprecipitation-based techniques allow to verify and better characterize these partnerships, possibly in combination with mass spectrometry methods. When transient interactions are involved, more sophisticated techniques, such as photoaffinity labelingprocedures, are necessary to detect the multiprotein complexes by having them covalently bound together as they interact. To provide even more thorough analyses of the formation, function and composition of protein complexes, other technologies such as confocal microscopy, fluorescence resonance energy transfer microscopy and site directed mutagenesis (possibly in murine models) have to be performed. The progressive accumulation of data defining novel protein-protein interactions has been considerably accelerated by the identification of specific sequence motifs that regulate integrin binding to other proteins as well as integrin recognition sequences in the ligand. Moreover, the availability of protein tagging strategies and the increased sensitivity of mass spectrometry-based methods for protein identification have also contributed important tools. In the near future, the coupling of traditional techniques with proteomic approaches is likely to offer invaluable help in unraveling integrin-ion channel interactions, thus elucidating the biological implication of these complexes.


Subject(s)
Biochemistry/methods , Integrins/metabolism , Ion Channels/metabolism , Multiprotein Complexes/metabolism , Animals , Humans , Integrins/chemistry , Ion Channels/chemistry , Multiprotein Complexes/chemistry
7.
Data Brief ; 26: 104406, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31508470

ABSTRACT

Macrolide antibiotics, such as Clarithromycin (Cla), have been proven to exert anti-tumour activity in several preclinical models of different types of cancer. Cla can exert its anti-tumour effects through different mechanisms, e.g. by blocking the autophagic flux, inducing apoptosis or inhibiting tumour-induced angiogenesis. The clinical benefit of Cla in treating various tumours in combination with conventional treatment was confirmed in extensive clinical studies in patients suffering from non-small cell lung cancer, breast cancer, multiple myeloma and other haematological malignancies. Data regarding the anti-cancer effect of Cla on Colorectal Cancer (CRC) are still lacking. This article shares data on the in vivo efficacy of Cla in two xenograft models of CRC. Our results show that Cla treatment reduces tumour growth and increases the overall survival in CRC mouse xenograft models. Moreover, the Western blot analysis of autophagic and apoptotic markers suggests that the anti-tumour effects of Cla are related to a modulation of both cellular processes. The data suggest that it will worth consider Cla as treatment option for CRC patients.

8.
ACS Med Chem Lett ; 10(4): 656-660, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30996813

ABSTRACT

In recent years, a few successful attempts were made to repurpose the clinically approved antiarthritic gold drug, Auranofin (AF), as an anticancer agent. The present study shows that the iodido(triethylphosphine)gold(I) complex, (Et 3 PAuI hereafter)-an AF analogue where the thiosugar ligand is simply replaced by one iodide ligand-manifests a solution chemistry resembling that of AF and exerts similar cytotoxic and proapoptotic effects on A2780 human ovarian cancer cells in vitro. However, when evaluated in a preclinical orthotopic model of ovarian cancer, Et 3 PAuI produces a far superior anticancer action than AF inducing a nearly complete tumor remission. The highly promising in vivo performances here documented for Et 3 PAuI warrant its further evaluation as a drug candidate for ovarian cancer treatment.

9.
Sci Rep ; 9(1): 8586, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31197180

ABSTRACT

The definition of the gene expression profile of genes encoding Ion Channels and Transporters (ICT-GEP) represents a novel and attracting aspect in cancer. We determined the ICT-GEP of Follicular Lymphoma (FL), and compared it with that of the more aggressive Diffuse Large B Cell Lymphoma (DLBCL). cDNA microarray data were collected both from patients enrolled for this study, and from public datasets. In FL the ICT-GEP indicated the overexpression of both the K+ channel encoding gene KCNN4, and SLC2A1, which encodes the Glut1 glucose transporter. SLC2A1 turned out to represent the hub of a functional network, connecting channels and transporters in FL. Relapsed FL patients were characterised by 38 differentially expressed ICT genes, among which ATP9A, SLC2A1 and KCNN4 were under-expressed, indicating a down-regulation of both excitability and glycolysis. A completely different profile of K+ channel encoding genes emerged in DLBCL accompanied by the over-expression of the fatty acid transporter-encoding gene SLC27A1 as well as of the metabolism regulator NCoR1. This indicates a change in excitability and a shift towards an oxidative metabolism in DLBCL. Overall, the ICT-GEP may contribute to identifying novel lymphoma biomarkers related to excitability and metabolic pathways, with particular relevance for drug resistant, relapsed FL.


Subject(s)
Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Ion Channels/genetics , Lymphoma, Follicular/genetics , Lymphoma, Follicular/metabolism , Membrane Transport Proteins/genetics , Aged , Cohort Studies , Databases, Genetic , Female , Gene Regulatory Networks , Humans , Ion Channels/metabolism , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Membrane Transport Proteins/metabolism , Middle Aged
10.
Mol Biol Cell ; 16(6): 2972-83, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15800067

ABSTRACT

Adhesive receptors of the integrin family are primarily involved in cell-extracellular matrix adhesion. Additionally, integrins trigger multiple signaling pathways that are involved in cell migration, proliferation, survival, and differentiation. We previously demonstrated that the activation of integrins containing the beta(1) subunit leads to a selective increase in potassium currents carried by the human ether-a-go-go-related gene (hERG) channels in neuroblastoma and leukemia cells; this current activation modulates adhesion-dependent differentiation in these cells. We hypothesized that the cross-talk between integrins and hERG channels could be traced back to the assembly of a macromolecular signaling complex comprising the two proteins. We tested this hypothesis in both SH-SY5Y neuroblastoma cells and in human embryonic kidney 293 cells stably transfected with hERG1 and, therefore, expressing only the full-length hERG1 protein on the plasma membrane. The beta(1) integrin and hERG1 coprecipitate in these cells and colocalize in both intracellular and surface membrane compartments. The two proteins also coprecipitate with caveolin-1, suggesting the localization of the complex in lipid rafts/caveolae. hERG1-transfected cells undergo an activation of hERG currents after beta(1) integrin-mediated adhesion to fibronectin; concomitant with this activation, the focal adhesion kinase associates with the hERG1 protein and becomes tyrosine phosphorylated. Using hERG1-specific inhibitors, we show that the tyrosine phosphorylation of focal adhesion kinase is strictly dependent on hERG channel activity. Similarly, the activity of the small GTPase Rac1 turned out to be dependent on hERG currents. On the whole, these data indicate that the hERG1 protein associates with beta(1) integrins and modulates adhesion receptor signaling.


Subject(s)
Cell Adhesion , Ether-A-Go-Go Potassium Channels/metabolism , Integrin beta Chains/metabolism , Signal Transduction , Cell Culture Techniques , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , ERG1 Potassium Channel , Fibronectins/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Integrin beta Chains/analysis , Intracellular Membranes/metabolism , Kidney/cytology , Kidney/embryology , Models, Biological , Neuroblastoma/pathology , Patch-Clamp Techniques , Phosphorylation , Precipitin Tests , Transfection , rac1 GTP-Binding Protein/metabolism
11.
Cell Cycle ; 17(3): 288-297, 2018.
Article in English | MEDLINE | ID: mdl-29160745

ABSTRACT

The purpose of this research has been deciphering the Warburg paradox, the biochemical enigma unsolved since 1923. We solved it by demonstrating that its specific character, i.e. the forced aerobic lactate exportation, represents a crucial metabolic device to counteract the cytotoxic effect produced by an excess of pyruvate at the connection of glycolysis with the Krebs cycle. This solution was verified by exposing cancer cells of different histogenesis to pyruvate concentrations higher than the physiological ones, after showing that these concentrations are totally innocuous when injected into mice. The mechanism of the pyruvate cytotoxicity relies on the saturation of the respiratory chain, leading to a negative shift of the cytosolic NADP/NADPH ratio and the consequent restriction of the purine synthesis and the related cell apoptosis. The reducing equivalents generated by glycolysis and by cytosolic metabolism compete each other for their disposal trough the respiratory chain; this makes it that the cytotoxicity of pyruvate is inversely related to the mitochondrial number and efficiency of various cell types. Thus, the cytotoxicity is high in anaplastic cancer stem cells, whose mitochondria are extremely few and immature (cristae-poor); on the contrary, no inhibition is brought about in adult differentiated cells, physiologically rich of mature mitochondria. All this generates the pyruvate anticancer selectivity, together with the lack of a general toxicity, making pyruvate represent an ideal candidate for a radical non toxical anticancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Glycolysis/drug effects , Animals , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Tumor Hypoxia/drug effects
12.
Oncotarget ; 9(79): 34972-34989, 2018 Oct 09.
Article in English | MEDLINE | ID: mdl-30405887

ABSTRACT

Modern molecular imaging techniques have greatly improved tumor detection and post-treatment follow-up of cancer patients. In this context, antibody-based imaging is rapidly becoming the gold standard, since it combines the unique specificity of antibodies with the sensitivity of the different imaging technologies. The aim of this study was to generate and characterize antibodies in single chain Fragment variable (scFv) format directed to an emerging cancer biomarker, the human ether-à-go-go-related gene-1 (hERG1) potassium channel, and to obtain a proof of concept for their potential use for in vivo molecular imaging. The anti-hERG1scFv was generated from a full length monoclonal antibody and then mutagenized, substituting a Phenylalanine residue in the third framework of the VH domain with a Cysteine residue. The resulting scFv-hERG1-Cys showed much higher stability and protein yield, increased affinity and more advantageous binding kinetics, compared to the "native" anti-hERG1scFv. The scFv-hERG1-Cys was hence chosen and characterized: it showed a good binding to the native hERG1 antigen expressed on cells, was stable in serum and displayed a fast pharmacokinetic profile once injected intravenously in nude mice. The calculated half-life was 3.1 hours and no general toxicity or cardiac toxic effects were detected. Finally, the in vivo distribution of an Alexa Fluor 750 conjugated scFv-hERG1-Cys was evaluated both in healthy and tumor-bearing nude mice, showing a good tumor-to-organ ratio, ideal for visualizing hERG1-expressing tumor masses in vivo. In conclusion, the scFv-hERG1-Cys possesses features which make it a suitable tool for application in cancer molecular imaging.

13.
Sci Signal ; 10(473)2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28377405

ABSTRACT

Ion channels regulate cell proliferation, differentiation, and migration in normal and neoplastic cells through cell-cell and cell-extracellular matrix (ECM) transmembrane receptors called integrins. K+ flux through the human ether-à-go-go-related gene 1 (hERG1) channel shapes action potential firing in excitable cells such as cardiomyocytes. Its abundance is often aberrantly high in tumors, where it modulates integrin-mediated signaling. We found that hERG1 interacted with the ß1 integrin subunit at the plasma membrane of human cancer cells. This interaction was not detected in cardiomyocytes because of the presence of the hERG1 auxiliary subunit KCNE1 (potassium voltage-gated channel subfamily E regulatory subunit 1), which blocked the ß1 integrin-hERG1 interaction. Although open hERG1 channels did not interact as strongly with ß1 integrins as did closed channels, current flow through hERG1 channels was necessary to activate the integrin-dependent phosphorylation of Tyr397 in focal adhesion kinase (FAK) in both normal and cancer cells. In immunodeficient mice, proliferation was inhibited in breast cancer cells expressing forms of hERG1 with impaired K+ flow, whereas metastasis of breast cancer cells was reduced when the hERG1/ß1 integrin interaction was disrupted. We conclude that the interaction of ß1 integrins with hERG1 channels in cancer cells stimulated distinct signaling pathways that depended on the conformational state of hERG1 and affected different aspects of tumor progression.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Integrin beta1/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Disease Progression , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/genetics , Fluorescence Resonance Energy Transfer , HCT116 Cells , HEK293 Cells , Humans , Immunoblotting , Integrin beta1/chemistry , Integrin beta1/genetics , Mice, Nude , Mice, SCID , Microscopy, Confocal , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , Protein Conformation , Transplantation, Heterologous
14.
Cancer Res ; 64(2): 606-11, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14744775

ABSTRACT

The acquisition of the capacity to invade surrounding tissues confers a more malignant phenotype to tumor cells and is necessary for the establishment of metastases. The understanding of the molecular mechanisms underlying cell invasion in human solid tumors such as colorectal cancers could provide not only more sensitive prognostic analyses but also novel molecular targets for cancer therapy. We report in this article that K(+) ion channels belonging to the HERG family are important determinants for the acquisition of an invasive phenotype in colorectal cancers. The herg1 gene and HERG1 protein are expressed in many colon cancer cell lines, and the activity of HERG channels modulates colon cancer cell invasiveness. Moreover, the amount of HERG1 protein expressed on the plasma membrane is directly related to the invasive phenotype of colon cancer cells. Finally, both the herg1 gene and HERG1 protein were expressed in a high percentage of primary human colorectal cancers, with the highest incidence occurring in metastatic cancers, whereas no expression could be detected either in normal colonic mucosa or in adenomas.


Subject(s)
Cation Transport Proteins , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic/genetics , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Trans-Activators , Cell Line, Tumor , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Immunohistochemistry , Neoplasm Invasiveness , Patch-Clamp Techniques , Potassium Channels/physiology , Transcriptional Regulator ERG
15.
J Inorg Biochem ; 163: 318-322, 2016 10.
Article in English | MEDLINE | ID: mdl-27381671

ABSTRACT

The trinuclear platinum cluster [Pt3(µ-PBut2)3(CO)3]CF3SO3 (I) was designed featuring the presence of a nearly equilateral platinum triangle bridged by three di-tert-butylphosphide ligands; in addition, each platinum center bears a terminal carbonyl ligand. This triplatinum cluster was initially developed in view of applications in the field of cluster-containing innovative materials. Yet, due to the large success of platinum complexes in cancer treatment, we also decided to explore its cytotoxic and anticancer properties. Accordingly, the solubility profile of this compound in several solvents was preliminarily investigated, revealing a conspicuous solubility in DMSO and DMSO/buffer mixtures; this makes the biological testing of I amenable. UV-Vis measurements showed that the triplatinum cluster is stable for several hours under a variety of conditions, within aqueous environments. No measurable reactivity was observed for I toward two typical model proteins, i.e. lysozyme and cytochrome c. On the contrary, a significant reactivity was evidenced when reacting I with small sulfur-containing ligands. In particular, a pronounced reactivity with reduced glutathione and cysteine emerged from ESI-MS experiments, proving complete formation of I-GSH and I-Cys derivatives, with the loss of a single carbonyl ligand. Starting from these encouraging results, the cytotoxic potential of I was assayed in vitro against a panel of representative cancer cell lines, and potent cytotoxic properties were disclosed. Of particular interest is the finding that the triplatinum species manifests potent antiproliferative properties toward Triple Negative Breast Cancer Cells, often refractory to most anticancer drugs. Owing to the reported encouraging results, a more extensive biological and pharmacological evaluation of this Pt cluster is now warranted to better elucidate its mode of action.


Subject(s)
Antineoplastic Agents , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Organoplatinum Compounds , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Drug Screening Assays, Antitumor , Female , HL-60 Cells , Humans , MCF-7 Cells , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology
16.
J Neurosci ; 22(9): 3414-25, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11978818

ABSTRACT

Native rat lactotrophs express thyrotrophin-releasing hormone-dependent K+ currents consisting of fast and slow deactivating components that are both sensitive to the class III anti-arrhythmic drugs that block the eag-related gene (ERG) K+ current (I(ERG)). Here we describe in MMQ prolactin-releasing pituitary cells the isolation of the slowly deactivating long-lasting component (I(ERGS)), which, unlike the fast component (I(ERGF)), is insensitive to verapamil 2 microm but sensitive to a novel scorpion toxin (ErgTx-2) that hardly affects I(ERGF). The time constants of I(ERGS) activation, deactivation, and recovery from inactivation are more than one order of magnitude greater than in I(ERGF), and the voltage-dependent inactivation is left-shifted by approximately 25 mV. The very slow MMQ firing frequency (approximately 0.2 Hz) investigated in perforated patch is increased approximately four times by anti-arrhythmic agents, by ErgTx-2, and by the abrupt I(ERGS) deactivation. Prolactin secretion in the presence of anti-arrhythmics is three- to fourfold higher in comparison with controls. We provide evidence from I(ERGS) and I(ERGF) simulations in a firing model cell to indicate that only I(ERGS) has an accommodating role during the experimentally observed very slow firing. Thus, we suggest that I(ERGS) potently modulates both firing and prolactin release in lactotroph cells.


Subject(s)
Cation Transport Proteins , Membrane Transport Proteins , Pituitary Gland/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Potassium/metabolism , Prolactin/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , Brain/metabolism , Calcium Channel Blockers/pharmacology , Cell Line , Computer Simulation , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Models, Neurological , Patch-Clamp Techniques , Pituitary Gland/cytology , Pituitary Gland/drug effects , Potassium Channels/drug effects , Potassium Channels/genetics , RNA/genetics , RNA/metabolism , Rats , Scorpion Venoms/isolation & purification , Scorpion Venoms/pharmacology , Tretinoin/metabolism , Verapamil/pharmacology
17.
J Comp Neurol ; 491(2): 157-74, 2005 Oct 17.
Article in English | MEDLINE | ID: mdl-16127690

ABSTRACT

Voltage-dependent K+ channels are the main determinants in controlling cellular excitability within the central nervous system. Among voltage-dependent K+ channels, the ERG subfamily is deeply involved in the control of cellular excitability, both in mammals and in invertebrates. ERG channels are encoded by different genes: the erg1 gene, which can generate two alternative transcripts (erg1a and erg1b), erg2 and erg3. The aim of the present study was to determine the expression pattern and cellular localization of ERG proteins (ERG1, ERG2, and ERG3) in the mouse CNS, differentiating, for the first time, the ERG1A and ERG1B isoforms. To this purpose, novel specific antibodies were raised against the various channel proteins and their specificity and immunoreactivity tested. It emerged that: 1) all the erg genes were indeed translated in neuronal tissue; 2) ERG proteins distribution in the mouse CNS often overlapped, and only in specific areas each ERG protein showed a distinct pattern of expression; and 3) ERG proteins were generally expressed in neuronal soma, but dendritic and/or white matter labeling could be detected in specific areas. The finding that ERG proteins often have an overlapping expression suggests that neuronal ERG currents could be determined, at least in part, by heterotetrameric ERG channels. This suggestion is demonstrated to occur for ERG1A/ERG1B by showing that the two isoforms coassemble in mouse brain.


Subject(s)
Brain/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Neurons/metabolism , Spinal Cord/metabolism , Animals , Brain/cytology , ERG1 Potassium Channel , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Protein Isoforms , Spinal Cord/cytology , Tissue Distribution
18.
FEBS Lett ; 547(1-3): 20-6, 2003 Jul 17.
Article in English | MEDLINE | ID: mdl-12860380

ABSTRACT

The scorpion toxin peptide BeKm-1 was synthesised by fluorenylmethoxycarbonyl solid phase chemistry and folded by air oxidation. The peptide's effects on heterologous human ether-a-go-go-related gene potassium current (I(HERG)) in HEK293 cells were assessed using 'whole-cell' patch clamp. Blockade of I(HERG) by BeKm-1 was concentration-dependent, temperature-dependent, and rapid in onset and reversibility. Blockade also exhibited inverse voltage dependence, inverse dependence on duration of depolarisation, and reverse use- and frequency-dependence. Blockade by BeKm-1 and recombinant ergtoxin, another scorpion toxin known to block HERG, differed in their recovery from HERG current inactivation elicited by strong depolarisation and in their ability to block HERG when the channels were already activated. We conclude that synthetic BeKm-1 toxin blocks HERG preferentially through a closed (resting) state channel blockade mechanism, although some open channel blockade also occurs.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Scorpion Venoms/pharmacology , Trans-Activators , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Kinetics , Patch-Clamp Techniques , Potassium Channel Blockers/chemical synthesis , Potassium Channels/drug effects , Potassium Channels/genetics , Protein Folding , Recombinant Proteins/antagonists & inhibitors , Scorpion Venoms/chemical synthesis , Scorpion Venoms/chemistry , Thermodynamics , Transcriptional Regulator ERG , Transfection
19.
Gene Expr Patterns ; 3(6): 767-76, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14643686

ABSTRACT

Deciphering the expression pattern of K+ channel encoding genes during development can help in the understanding of the establishment of cellular excitability and unravel the molecular mechanisms of neuromuscular diseases. We focused our attention on genes belonging to the erg family, which is deeply involved in the control of neuromuscular excitability in Drosophila flies and possibly other organisms. Both in situ hybridisation and RNase Protection Assay experiments were used to study the expression pattern of mouse (m)erg1, m-erg2 and m-erg3 genes during mouse embryo development, to allow the pattern to be compared with their expression in the adult. M-erg1 is first expressed in the heart and in the central nervous system (CNS) of embryonic day 9.5 (E9.5) embryos; the gene appears in ganglia of the peripheral nervous system (PNS) (dorsal root (DRG) and sympathetic (SCG) ganglia, mioenteric plexus), in the neural layer of retina, skeletal muscles, gonads and gut at E13.5. In the adult m-erg1 is expressed in the heart, various structures of the CNS, DRG and retina. M-erg2 is first expressed at E9.5 in the CNS, thereafter (E13.5) in the neural layer of retina, DRG, SCG, and in the atrium. In the adult the gene is present in some restricted areas of the CNS, retina and DRG. M-erg3 displayed an expression pattern partially overlapping that of m-erg1, with a transitory expression in the developing heart as well. A detailed study of the mouse adult brain showed a peculiar expression pattern of the three genes, sometimes overlapping in different encephalic areas.


Subject(s)
Mice/embryology , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Animals , Brain/embryology , Brain/metabolism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice/genetics , Mice/metabolism , Mice, Inbred C57BL , Potassium Channels/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism
20.
Br J Pharmacol ; 139(5): 887-98, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12839862

ABSTRACT

1. Pharmacological blockade of the Human ether-a-go-go related gene (HERG) potassium channel is commonly linked with acquired long QT syndrome and associated proarrhythmia. The objectives of this study were (i) to identify and characterise any inhibitory action on HERG of the selective-serotonin re-uptake inhibitor fluvoxamine, (ii) to then determine whether fluvoxamine shared the consensus molecular determinants of HERG blockade of those drugs so far tested. 2. Heterologous HERG potassium current (I(HERG)) was measured at 37 degrees C, using the whole-cell patch-clamp technique, from a mammalian cell line (Human embryonic kidney 293) expressing HERG channels. I(HERG) tails, following repolarisation from +20 to -40 mV, were blocked by fluvoxamine with an IC(50) of 3.8 micro M. 3. Blockade of wild-type HERG was of extremely rapid onset (within 10 ms) and showed voltage dependence, with fluvoxamine also inducing a leftward shift in voltage-dependent activation of I(HERG). Characteristics of block were consistent with a component of closed channel (or extremely rapidly developing open channel) blockade and dependence on open and inactivated channel states. The attenuated-inactivation mutation S631A partially reduced the blocking effect of fluvoxamine. 4. The S6 mutations, Y652A and F656A, and the pore helix mutant S631A only partially attenuated blockade by fluvoxamine at concentrations causing profound blockade of wild-type HERG. 5. All HERG-blocking pharmaceuticals studied to date have been shown to block F656 mutant channels with over 100-fold reduced potency compared to their blockade of the wild-type channel. Fluvoxamine is therefore quite distinct in this regard from previously studied agents.


Subject(s)
Amino Acid Substitution/genetics , Cation Transport Proteins , DNA-Binding Proteins , Fluvoxamine/pharmacology , Mutation , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Trans-Activators , Cell Line , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Fluvoxamine/chemistry , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Phenylalanine/genetics , Potassium Channel Blockers/chemistry , Potassium Channels/metabolism , Transcriptional Regulator ERG , Tyrosine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL