128 results on '"Pillozzi S"'
Search Results
2. 36P Expression of germinal center B cell- and Th17 cell-related transcripts are prognostic of soft-tissue sarcoma patient outcomes
- Author
-
Petroni, G., primary, Scolari, F., additional, Caliman, E., additional, Scanferla, R., additional, Scoccianti, G., additional, Palomba, A., additional, Campanacci, D.A., additional, Antonuzzo, L., additional, and Pillozzi, S., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Neutrophil-to-eosinophil ratio (NER) predicts the efficacy of avelumab in patients with advanced Urothelial Carcinoma (aUC) enrolled in the MALVA study (Meet-URO 25)
- Author
-
Gambale, E., primary, MARUZZO, M., additional, MESSINA, C., additional, DE GENNARO AQUINO, I., additional, VASCOTTO, I.A., additional, ROSSI, V., additional, BIMBATTI, D., additional, CAVASIN, N., additional, MESSINA, M., additional, MENNITTO, A., additional, REBUZZI, S.E., additional, MERCINELLI, C., additional, FANELLI, M., additional, SORARù, M., additional, SCOLARI, F., additional, MELA, M.M., additional, GALLI, L., additional, DONI, L., additional, PILLOZZI, S., additional, and ANTONUZZO, L., additional
- Published
- 2023
- Full Text
- View/download PDF
4. Neoadjuvant versus adjuvant chemotherapy in muscle-invasive bladder cancer: The RealBLADDER study. Results on behalf of the GOIRC
- Author
-
Gambale, E., primary, GIORGIONE, R., additional, VASCOTTO, I.A., additional, DE GENNARO AQUINO, I., additional, SCOLARI, F., additional, MELA, M.M., additional, CATALANO, M., additional, ROVIELLO, G., additional, DONI, L., additional, GALLI, L., additional, BALDAZZI, V., additional, ATZORI, F., additional, MASINI, C., additional, SCAGLIARINI, S., additional, PILLOZZI, S., additional, BASSO, U., additional, and ANTONUZZO, L., additional
- Published
- 2023
- Full Text
- View/download PDF
5. 111P A novel immune cell signature for predicting pleomorphic sarcoma prognosis
- Author
-
Gambale, E., primary, Scolari, F., additional, Scoccianti, G., additional, Caliman, E., additional, Palomba, A., additional, Antonuzzo, L., additional, Campanacci, D.A., additional, and Pillozzi, S., additional
- Published
- 2023
- Full Text
- View/download PDF
6. 110P Characterization of miRNA-mRNA network in soft tissue sarcoma
- Author
-
Pillozzi, S., primary, Scolari, F., additional, Scoccianti, G., additional, Boddi, A., additional, Palchetti, I., additional, Antonuzzo, L., additional, and Campanacci, D.A., additional
- Published
- 2023
- Full Text
- View/download PDF
7. 81P Soft tissue solitary fibrous tumor: Analysis of a single center series with a comparison between three prognostic scores
- Author
-
Scoccianti, G., primary, Palomba, A., additional, Caporalini, C., additional, Nozzoli, F., additional, Scanferla, R., additional, Scolari, F., additional, Pillozzi, S., additional, and Campanacci, D.A., additional
- Published
- 2023
- Full Text
- View/download PDF
8. Data describing the effects of the Macrolide Antibiotic Clarithromycin on preclinical mouse models of Colorectal Cancer
- Author
-
Petroni, G, Stefanini, M, Pillozzi, S, Crociani, O, Becchetti, A, Arcangeli, A, Petroni G., Stefanini M., Pillozzi S., Crociani O., Becchetti A., Arcangeli A., Petroni, G, Stefanini, M, Pillozzi, S, Crociani, O, Becchetti, A, Arcangeli, A, Petroni G., Stefanini M., Pillozzi S., Crociani O., Becchetti A., and Arcangeli A.
- 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.
- Published
- 2019
9. ROS1 rearrangements are uncommon in biliary tract cancers
- Author
-
Mazzoni F., Petreni P., Vasile E., Panebianco M., Casadei Gardini A., Negri F., Lunghi A., Pillozzi S., Vivaldi C., Gervasi E., Frassineti G. L., Messerini L., Jocolle G., Bisagni A., Antonuzzo L., Rossi G., Mazzoni, F., Petreni, P., Vasile, E., Panebianco, M., Casadei Gardini, A., Negri, F., Lunghi, A., Pillozzi, S., Vivaldi, C., Gervasi, E., Frassineti, G. L., Messerini, L., Jocolle, G., Bisagni, A., Antonuzzo, L., and Rossi, G.
- Subjects
Molecular target ,Fluorescence in situ hybridization ,Biliary tract cancers ,Biomarker ,Immuno‑ histochemistry ,ROS1 rearrangements ,Articles - Abstract
Biliary tract cancers (BTCs) are a pool of diseases with poor prognosis and there is no orphan drug available. Currently, no molecular targets have been tested as druggable oncogenic drivers. C‑ros oncogene 1 (ROS1) rearrangements have been previously described in various tumors, including BTCs; however, data regarding their incidence and biological significance are controversial. Therefore, a retrospective multi‑ center study was performed to assess the incidence of ROS1 rearrangements in BTCs by means of immunohistochemistry and fluorescence in situ hybridization (FISH). The present study failed to demonstrate ROS1 expression in a multicenter series of 150 cases with BTCs and revealed that D4D6 was the most specific clone compared with other ROS1 primary antibodies, namely PA1‑30318 and EPMGHR2. Notably, nega‑ tive results obtained with D4D6 completely matched to data sorted out by FISH analysis, thus confirming a lack of ROS1 gene rearrangements in BTCs and false positive results when PA1‑30318 and EPMGHR2 clones were used. These results suggest that ROS1 rearrangements may not be targets for molecular therapy of BTCs with specific inhibitors.
- Published
- 2020
10. Anticancer effects against colorectal cancer models of chloro(triethylphosphine)gold(I) encapsulated in PLGA–PEG nanoparticles
- Author
-
Menconi, A., Marzo, T., Massai, L., Pratesi, A., Severi, M., Petroni, G., Antonuzzo, L., Messori, L., Pillozzi, S., and Cirri, D.
- Subjects
Anticancer complexes ,Auranofin ,Colorectal cancer ,PLGA–PEG nanoparticles - Published
- 2021
11. P228 - Neoadjuvant versus adjuvant chemotherapy in muscle-invasive bladder cancer: The RealBLADDER study. Results on behalf of the GOIRC
- Author
-
Gambale, E., GIORGIONE, R., VASCOTTO, I.A., DE GENNARO AQUINO, I., SCOLARI, F., MELA, M.M., CATALANO, M., ROVIELLO, G., DONI, L., GALLI, L., BALDAZZI, V., ATZORI, F., MASINI, C., SCAGLIARINI, S., PILLOZZI, S., BASSO, U., and ANTONUZZO, L.
- Published
- 2023
- Full Text
- View/download PDF
12. P216 - Neutrophil-to-eosinophil ratio (NER) predicts the efficacy of avelumab in patients with advanced Urothelial Carcinoma (aUC) enrolled in the MALVA study (Meet-URO 25)
- Author
-
Gambale, E., MARUZZO, M., MESSINA, C., DE GENNARO AQUINO, I., VASCOTTO, I.A., ROSSI, V., BIMBATTI, D., CAVASIN, N., MESSINA, M., MENNITTO, A., REBUZZI, S.E., MERCINELLI, C., FANELLI, M., SORARù, M., SCOLARI, F., MELA, M.M., GALLI, L., DONI, L., PILLOZZI, S., and ANTONUZZO, L.
- Published
- 2023
- Full Text
- View/download PDF
13. 1199P MAVERIC: Phase II randomized study of everolimus as maintenance therapy for metastatic neuroendocrine neoplasms (mNEN) with pulmonary or gastroenteropancreatic (GEP) origin. Results on behalf of the GOIRC
- Author
-
Antonuzzo, L., Pillozzi, S., Marconcini, R., Spada, F., Gelsomino, F., Amoroso, V., cosso, F., messerini, L., Zepponi, F., Scolari, F., Pellegrini, E., Sparano, C., Lavacchi, D., Brugia, M., Giommoni, E., Massaro, G., Milione, M., Di Costanzo, F., Boni, L., and Fazio, N.
- Published
- 2023
- Full Text
- View/download PDF
14. 1677P Pancreatic enzyme replacement therapy improves survival in patients receiving nab-paclitaxel plus gemcitabine as first-line treatment for advanced pancreatic adenocarcinoma
- Author
-
Lavacchi, D., Winchler, C., Giommoni, E., Vascotto, I.A., Romano, S., Scolari, F., Taddei, A., Pillozzi, S., Galli, A., and Antonuzzo, L.
- Published
- 2023
- Full Text
- View/download PDF
15. Differential expression of hERG1A and hERG1B genes in pediatric acute lymphoblastic leukemia identifies different prognostic subgroups
- Author
-
Pillozzi, S, Accordi, B, Rebora, P, Serafin, V, Valsecchi, M G, Basso, G, and Arcangeli, A
- Published
- 2014
- Full Text
- View/download PDF
16. HERG potassium channels are constitutively expressed in primary human acute myeloid leukemias and regulate cell proliferation of normal and leukemic hemopoietic progenitors
- Author
-
Pillozzi, S, Brizzi, MF, Balzi, M, Crociani, O, Cherubini, A, Guasti, L, Bartolozzi, B, Becchetti, A, Wanke, E, Bernabei, PA, Olivotto, M, Pegoraro, L, and Arcangeli, A
- Published
- 2002
- Full Text
- View/download PDF
17. The combined activation of K Ca 3.1 and inhibition of K v 11.1/hERG1 currents contribute to overcome Cisplatin resistance in colorectal cancer cells
- Author
-
Pillozzi, S, D'Amico, M, Bartoli, G, Gasparoli, L, Petroni, G, Crociani, O, Marzo, T, Guerriero, A, Messori, L, Severi, M, Udisti, R, Wulff, H, Chandy, KG, Becchetti, A, and Arcangeli, A
- Subjects
preclinical mouse models ,Riluzole ,Cisplatin uptake ,SKA-31 ,E4031 - 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 Ca 2+ -activated (K Ca 3.1, also known as KCNN4) and voltage-dependent (K v 11.1, 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 K Ca 3.1 and K v 11.1 channels, compared with Cisplatin-sensitive CRC cells. In resistant cells, K Ca 3.1 activators (SKA-31) and K v 11.1 inhibitors (E4031) had a synergistic action with Cisplatin in triggering apoptosis and inhibiting proliferation. The effect was maximal when K Ca 3.1 activation and K v 11.1 inhibition were combined. In fact, similar results were produced by Riluzole, which is able to both activate K Ca 3.1 and inhibit K v 11.1. Cisplatin uptake into resistant cells depended on K Ca 3.1 channel activity, as it was potentiated by K Ca 3.1 activators. K v 11.1 blockade led to increased K Ca 3.1 expression and thereby stimulated Cisplatin uptake. Finally, the combined administration of a K Ca 3.1 activator and a K v 11.1 inhibitor also overcame Cisplatin resistance in vivo.Conclusions:As Riluzole, an activator of K Ca 3.1 and inhibitor of K v 11.1 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.
- Published
- 2018
18. The combined activation of KCa3.1 and inhibition of Kv11.1/hERG1 currents contribute to overcome Cisplatin resistance in colorectal cancer cells
- Author
-
Pillozzi, S, D'Amico, M, Bartoli, G, Gasparoli, L, Petroni, G, Crociani, O, Marzo, T, Guerriero, A, Messori, L, Severi, M, Udisti, R, Wulff, H, Chandy, K, Becchetti, A, Arcangeli, A, Pillozzi, S, D'Amico, M, Bartoli, G, Gasparoli, L, Petroni, G, Crociani, O, Marzo, T, Guerriero, A, Messori, L, Severi, M, Udisti, R, Wulff, H, Chandy, K, Becchetti, A, and Arcangeli, A
- 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
- Published
- 2018
19. Ion Channels in Hematopoietic and Mesenchymal Stem Cells
- Author
-
Pillozzi, S, Becchetti, A, PILLOZZI, S, BECCHETTI, ANDREA, Pillozzi, S, Becchetti, A, PILLOZZI, S, and BECCHETTI, ANDREA
- Abstract
Hematopoietic stem cells (HSCs) reside in bone marrow niches and give rise to hematopoietic precursor cells (HPCs). These have more restricted lineage potential and eventually differentiate into specific blood cell types. Bone marrow also contains mesenchymal stromal cells (MSCs), which present multilineage differentiation potential toward mesodermal cell types. In bone marrow niches, stem cell interaction with the extracellular matrix is mediated by integrin receptors. Ion channels regulate cell proliferation and differentiation by controlling intracellular Ca2+, cell volume, release of growth factors, and so forth. Although little evidence is available about the ion channel roles in true HSCs, increasing information is available about HPCs and MSCs, which present a complex pattern of K+ channel expression. K+ channels cooperate with Ca2+ and Cl- channels in regulating calcium entry and cell volume during mitosis. Other K+ channels modulate the integrin-dependent interaction between leukemic progenitor cells and the niche stroma. These channels can also regulate leukemia cell interaction with MSCs, which also involves integrin receptors and affects the MSC-mediated protection from chemotherapy. Ligand-gated channels are also implicated in these processes. Nicotinic acetylcholine receptors regulate cell proliferation and migration in HSCs and MSCs and may be implicated in the harmful effects of smoking.
- Published
- 2012
20. Deregulation of Ion Channel and Transporter Encoding Genes in Pediatric Gliomas
- Author
-
Masselli, M, Laise, P, Tonini, G, Fanelli, D, Pillozzi, S, Cetica, V, DA ROS, M, Sardi, I, Buccoliero, A, Arico', M, Genitori, L, Becchetti, A, Arcangeli, A, MASSELLI, M, LAISE, P, TONINI, G, FANELLI, D, PILLOZZI, S, CETICA, V, SARDI, I, BUCCOLIERO, AM, ARICO', M, GENITORI, L, ARCANGELI, A., BECCHETTI, ANDREA, Masselli, M, Laise, P, Tonini, G, Fanelli, D, Pillozzi, S, Cetica, V, DA ROS, M, Sardi, I, Buccoliero, A, Arico', M, Genitori, L, Becchetti, A, Arcangeli, A, MASSELLI, M, LAISE, P, TONINI, G, FANELLI, D, PILLOZZI, S, CETICA, V, SARDI, I, BUCCOLIERO, AM, ARICO', M, GENITORI, L, ARCANGELI, A., and BECCHETTI, ANDREA
- Abstract
Brain tumors, including the majority gliomas, are the leading cause of cancer-related death in children. World Health Organization has divided pediatric brain tumors into different grades and, based upon cDNA microarray data identifying gene expression profiles (GEPs), it has become evident in the last decade that the various grades involve different types of genetic alterations. However, it is not known whether ion channel and transporter genes, intimately involved in brain functioning, are associated with such GEPs. We determined the GEPs in an available cohort of 10 pediatric brain tumors initially by comparing the data obtained from four primary tumor samples and corresponding short-term cultures. The correspondence between the two types of samples was statistically significant. We then performed bioinformatic analyses on those samples (a total of nine) which corresponded to tumors of glial origin, either tissues or cell cultures, depending on the best "RNA integrity number." We used R software to evaluate the genes which were differentially expressed (DE) in gliomas compared with normal brain. Applying a p-value below 0.01 and fold change ≥4, led to identification of 2284 DE genes. Through a Functional Annotation Analysis (FAA) using the NIH-DAVID software, the DE genes turned out to be associated mainly with: immune/inflammatory response, cell proliferation and survival, cell adhesion and motility, neuronal phenotype, and ion transport. We have shown that GEPs of pediatric brain tumors can be studied using either primary tumor samples or short-term cultures with similar results. From FAA, we concluded that, among DE genes, pediatric gliomas show a strong deregulation of genes related to ion channels and transporters. © 2012 Masselli, Laise, Tonini, Fanelli, Pillozzi, Cetica, Da Ros, Sardi, Buccoliero, Aricò, Genitori, Becchetti and Arcangeli.
- Published
- 2012
21. Macrolide antibiotics exert antileukemic effects by modulating the autophagic flux through inhibition of hERG1 potassium channels
- Author
-
Pillozzi, S, Masselli, M, Gasparoli, L, D'Amico, M, Polletta, L, Veltroni, M, Favre, C, Basso, G, Becchetti, A, Arcangeli, A, Arcangeli, A., BECCHETTI, ANDREA, Pillozzi, S, Masselli, M, Gasparoli, L, D'Amico, M, Polletta, L, Veltroni, M, Favre, C, Basso, G, Becchetti, A, Arcangeli, A, Arcangeli, A., and BECCHETTI, ANDREA
- Published
- 2016
22. Macrolide antibiotics exert antileukemic effects by modulating the autophagic flux through inhibition of hERG1 potassium channels
- Author
-
Pillozzi, S, primary, Masselli, M, additional, Gasparoli, L, additional, D'Amico, M, additional, Polletta, L, additional, Veltroni, M, additional, Favre, C, additional, Basso, G, additional, Becchetti, A, additional, and Arcangeli, A, additional
- Published
- 2016
- Full Text
- View/download PDF
23. NAMI-A is highly cytotoxic toward leukaemia cell lines: evidence of inhibition of KCa3.1 channels
- Author
-
Pillozzi, S, Gasparoli, L, Stefanini, M, Ristori, M, D'Amico, M, Alessio, E, Scaletti, F, Becchetti, A, Arcangeli, A, Messori, L, Messori, L., BECCHETTI, ANDREA, Pillozzi, S, Gasparoli, L, Stefanini, M, Ristori, M, D'Amico, M, Alessio, E, Scaletti, F, Becchetti, A, Arcangeli, A, Messori, L, Messori, L., and BECCHETTI, ANDREA
- Abstract
We report here that the established anticancer ruthenium(iii) complex NAMI-A induces potent and selective cytotoxic effects in a few leukaemia cell lines. These results sound very surprising after 20 years of intense studies on NAMI-A, commonly considered as a "non-cytotoxic" antimetastatic agent. In addition, evidence is given for selective inhibition of KCa 3.1 channels. The implications of these findings are discussed. © 2014 the Partner Organisations.
- Published
- 2014
24. Glutamine depletion by crisantaspase hinders the growth of human hepatocellular carcinoma xenografts
- Author
-
Chiu, M, primary, Tardito, S, additional, Pillozzi, S, additional, Arcangeli, A, additional, Armento, A, additional, Uggeri, J, additional, Missale, G, additional, Bianchi, M G, additional, Barilli, A, additional, Dall'Asta, V, additional, Campanini, N, additional, Silini, E M, additional, Fuchs, J, additional, Armeanu-Ebinger, S, additional, and Bussolati, O, additional
- Published
- 2014
- Full Text
- View/download PDF
25. HERG K+ channels and β1 integrins interact through the assembly of a macromolecular complex
- Author
-
Cherubini, A., Pillozzi, S., Hofmann, G., Crociani, O., Guasti, L., Lastraioli, E., Polvani, S., Masi, A., Becchetti, A., Wanke, E., Olivotto, M., and Arcangeli, A.
- Subjects
Cell differentiation ,herg gene ,HERG protein ,Integrins ,PKA ,Cell Line ,ERG1 Potassium Channel ,Ether-A-Go-Go Potassium Channels ,Humans ,Integrin beta1 ,Macromolecular Substances ,Multigene Family ,Potassium Channels ,Transcriptional Regulator ERG ,Tumor Cells, Cultured ,Cation Transport Proteins ,DNA-Binding Proteins ,Potassium Channels, Voltage-Gated ,Trans-Activators - Published
- 2002
26. Targeting ion channels in leukemias: a new challenge for treatment
- Author
-
Arcangeli, A, Pillozzi, S, Becchetti, A, BECCHETTI, ANDREA, Arcangeli, A, Pillozzi, S, Becchetti, A, and BECCHETTI, ANDREA
- Abstract
Leukemias, as other cancers, bear several genetic alterations of tumor-related genes, such as point mutations, translocations, epigenetic modifications, often accompanied by gene amplification or inactivation. The identification of tumor-related genes provides considerable insight into the biology of leukemias and opens the way to more specific pharmacological treatments. These genes comprise several ion channels and pumps, as the transport mechanisms associated with volume control, proliferation and apoptosis are often altered in cancers. In leukemic cells, such changes are observed as early as the stem cell stage. Ion channels can regulate other malignant features, such as lack of differentiation, increased migratory and invasive phenotype and chemoresistance. The role of certain voltagegated K+ channels, such as Kv11.1 (also known as hERG1) can be largely attributed to modulation of cell adhesion to the extracellular matrix (ECM). Kv11.1 exerts pleiotropic regulatory effects by forming multiprotein membrane complexes with integrin receptors in both acute myeloid leukemias (AML) and acute lymphoblastic leukemias (ALL). By recruiting growth factor and chemokine receptors, these complexes form signaling hubs that control neoplastic progression. Work in mice shows that blocking Kv11.1 has a protective effect in acute leukemias. Ion channels are most promising targets for anti-leukemic therapy, because of their accessibility from the extracellular side and the thorough understanding of their pharmacology. In ALL cells, Kv11.1 inhibitors abrogate the protective effect of bone marrow stromal cells and enhance the cytotoxicity of some common antileukemic drugs. Hence, ion channel modulators could overcome chemoresistance in acute leukemias, a major hindrance to therapeutic success. © 2012 Bentham Science Publishers.
- Published
- 2012
27. Ion Channels: Novel Functional Hubs in Leukemia
- Author
-
Ozdemir, O, Arcangeli, A, Pillozzi, S, Becchetti, A, BECCHETTI, ANDREA, Ozdemir, O, Arcangeli, A, Pillozzi, S, Becchetti, A, and BECCHETTI, ANDREA
- Published
- 2011
28. New insights into the regulation of ion channels by integrins
- Author
-
Becchetti, A, Pillozzi, S, Morini, R, Nesti, E, Arcangeli, A, Becchetti, A, Pillozzi, S, Morini, R, Nesti, E, and Arcangeli, A
- Abstract
By controlling cell adhesion to the extracellular matrix, integrin receptors regulate processes as diverse as cell migration, proliferation, differentiation, apoptosis, and synaptic stability. Because the underlying mechanisms are generally accompanied by changes in transmembrane ion flow, a complex interplay occurs between integrins, ion channels, and other membrane transporters. This reciprocal interaction regulates bidirectional signal transduction across the cell surface and may take place at all levels of control, from transcription to direct conformational coupling. In particular, it is becoming increasingly clear that integrin receptors form macromolecular complexes with ion channels. Besides contributing to the membrane localization of the channel protein, the integrin/channel complex can regulate a variety of downstream signaling pathways, centered on regulatory proteins like tyrosine kinases and small GTPases. In turn, the channel protein usually controls integrin activation and expression. We review some recent advances in the field, with special emphasis on hematology and neuroscience. Some oncological implications are also discussed. © 2010 Elsevier Inc.
- Published
- 2010
29. New insights into the regulation of ion channels by integrins
- Author
-
Kwang Jeon, Becchetti, A, Pillozzi, S, Morini, R, Nesti, E, Arcangeli, A, Kwang Jeon, Becchetti, A, Pillozzi, S, Morini, R, Nesti, E, and Arcangeli, A
- Abstract
By controlling cell adhesion to the extracellular matrix, integrin receptors regulate processes as diverse as cell migration, proliferation, differentiation, apoptosis, and synaptic stability. Because the underlying mechanisms are generally accompanied by changes in transmembrane ion flow, a complex interplay occurs between integrins, ion channels, and other membrane transporters. This reciprocal interaction regulates bidirectional signal transduction across the cell surface and may take place at all levels of control, from transcription to direct conformational coupling. In particular, it is becoming increasingly clear that integrin receptors form macromolecular complexes with ion channels. Besides contributing to the membrane localization of the channel protein, the integrin/channel complex can regulate a variety of downstream signaling pathways, centered on regulatory proteins like tyrosine kinases and small GTPases. In turn, the channel protein usually controls integrin activation and expression. We review some recent advances in the field, with special emphasis on hematology and neuroscience. Some oncological implications are also discussed. © 2010 Elsevier Inc.
- Published
- 2010
30. Method and kit for the prevention and/or the monitoring of chemoresistance of leukaemia forms
- Author
-
Arcangeli, A, Becchetti, A, Pillozzi, S, Masselli, M, De Lorenzo, E, De Lorenzo, E., BECCHETTI, ANDREA, Arcangeli, A, Becchetti, A, Pillozzi, S, Masselli, M, De Lorenzo, E, De Lorenzo, E., and BECCHETTI, ANDREA
- Published
- 2009
31. New compositions for the treatment of chemoresistant and/or potentially chemoresistant leukaemias
- Author
-
Arcangeli, A, Becchetti, A, Pillozzi, S, Masselli, M, De Lorenzo, E, BECCHETTI, ANDREA, De Lorenzo, E., Arcangeli, A, Becchetti, A, Pillozzi, S, Masselli, M, De Lorenzo, E, BECCHETTI, ANDREA, and De Lorenzo, E.
- Published
- 2009
32. Targeting ion channels in cancer: A novel frontier in antineoplastic therapy
- Author
-
Arcangeli, A, Crociani, O, Lastraioli, E, Masi, A, Pillozzi, S, Becchetti, A, BECCHETTI, ANDREA, Arcangeli, A, Crociani, O, Lastraioli, E, Masi, A, Pillozzi, S, Becchetti, A, and BECCHETTI, ANDREA
- Abstract
Targeted therapy is considerably changing the treatment and prognosis of cancer. Progressive understanding of the molecular mechanisms that regulate the establishment and progression of different tumors is leading to ever more specific and efficacious pharmacological approaches. In this picture, ion channels represent an unexpected, but very promising, player. The expression and activity of different channel types mark and regulate specific stages of cancer progression. Their contribution to the neoplastic phenotype ranges from control of cell proliferation and apoptosis, to regulation of invasiveness and metastatic spread. As is being increasingly recognized, some of these roles can be attributed to signaling mechanisms independent of ion flow. Evidence is particularly extensive for K+ channels. Their expression is altered in many primary human cancers, especially in early stages, and they frequently exert pleiotropic effects on the neoplastic cell physiology. For instance, by regulating membrane potential they can control Ca2+ fluxes and thus the cell cycle machinery. Their effects on mitosis can also depend on regulation of cell volume, usually in cooperation with chloride channels. However, ion channels are also implicated in late neoplastic stages, by stimulating angiogenesis, mediating the cell-matrix interaction and regulating cell motility. Not surprisingly, the mechanisms of these effects are manifold. For example, intracellular signaling cascades can be triggered when ion channels form protein complexes with other membrane proteins such as integrins or growth factor receptors. Altered channel expression can be exploited for diagnostic purposes or for addressing traceable or cytotoxic compounds to specific neoplastic tissue. What is more, recent evidence indicates that blocking channel activity impairs the growth of some tumors, both in vitro and in vivo. This opens a new field for medicinal chemistry studies, which can avail of the many available tools, su
- Published
- 2009
33. Identification of a posttranslational mechanism for the regulation of hERG1 K+ channel expression and hERG1 current density in tumor cells
- Author
-
Guasti, L, Crociani, O, Redaelli, E, Pillozzi, S, Polvani, S, Masselli, M, Mello, T, Galli, A, Amedei, A, Wymore, R, Wanke, E, Arcangeli, A, Wymore, RS, Arcangeli, A., REDAELLI, ELISA, WANKE, ENZO, Guasti, L, Crociani, O, Redaelli, E, Pillozzi, S, Polvani, S, Masselli, M, Mello, T, Galli, A, Amedei, A, Wymore, R, Wanke, E, Arcangeli, A, Wymore, RS, Arcangeli, A., REDAELLI, ELISA, and WANKE, ENZO
- Abstract
A common feature of tumor cells is the aberrant expression of ion channels on their plasma membrane. The molecular mechanisms regulating ion channel expression in cancer cells are still poorly known. K+ channels that belong to the human ether-a-go-go-related gene 1 (herg1) family are frequently misexpressed in cancer cells compared to their healthy counterparts. We describe here a posttranslational mechanism for the regulation of hERG1 channel surface expression in cancer cells. This mechanism is based on the activity of hERG1 isoforms containing the USO exon. These isoforms (i) are frequently overexpressed in human cancers, (ii) are retained in the endoplasmic reticulum, and (iii) form heterotetramers with different proteins of the hERG family. (iv) The USO-containing heterotetramers are retained intracellularly and undergo ubiquitin-dependent degradation. This process results in decreased hERG1 current (IhERG1) density. We detailed such a mechanism in heterologous systems and confirmed its functioning in tumor cells that endogenously express hERG1 proteins. The silencing of USO-containing hERG1 isoforms induces a higher IhERG1 density in tumors, an effect that apparently regulates neurite outgrowth in neuroblastoma cells and apoptosis in leukemia cells. Copyright © 2008, American Society for Microbiology. All Rights Reserved.
- Published
- 2008
34. Hepatocyte growth factor receptor c-MET is associated with FAS and when activated enhances drug-induced apoptosis in pediatric B acute lymphoblastic leukemia with TEL-AML1 translocation
- Author
-
Accordi, B, Pillozzi, S, Dell'Orto, M, Cazzaniga, G, Arcangeli, A, Kronnie, G, Basso, G, Accordi, Benedetta, Pillozzi, Serena, Dell'Orto, Marta Campo, Cazzaniga, Giovanni, Arcangeli, Annarosa, Kronnie, Geertruy Te, Basso, Giuseppe, Accordi, B, Pillozzi, S, Dell'Orto, M, Cazzaniga, G, Arcangeli, A, Kronnie, G, Basso, G, Accordi, Benedetta, Pillozzi, Serena, Dell'Orto, Marta Campo, Cazzaniga, Giovanni, Arcangeli, Annarosa, Kronnie, Geertruy Te, and Basso, Giuseppe
- Abstract
Expression of c-MET, the HGF (hepatocyte growth factor) tyrosine kinase receptor, was investigated in pediatric B-acute lymphoblastic leukemia (ALL) patients. c-MET was found to be expressed in normal B cells and in B-ALL patients with the t(12;21) TEL-AML1 translocation, but it is not expressed in the most part of B-ALL without the t(12;21). We also found that c-MET, related to proliferation and protection from apoptosis, is associated with the pro-apoptotic protein FAS in TEL-AML1 B-ALL cells and in normal B lymphocytes. The possible role of this protein complex in drug-induced apoptosis was thus investigated in REH TEL-AML1 B-ALL cell line. REH cells prestimulated with HGF and treated with doxorubicin had shown a higher apoptotic rate than non-HGF-prestimulated ones (p = 0.03). REH cells stimulated with IL-3 and treated with doxorubicin did not undergo apoptosis more than nonstimulated cells, demonstrating that increased proliferation in itself is not directly related to the higher apoptotic sensitivity observed with HGF stimulation. These results indicate that c-MET activation enhances specifically FAS-mediated apoptosis in TEL-AML1 ALL cells and, considering that the c-MET/FAS complex is present only in normal B lymphocytes and in TEL-AML1 leukemias, this implies that it may have an important contribution in cellular homeostasis and in high sensitivity of TEL-AML1 ALL to chemotherapeutic regimens.
- Published
- 2007
35. Developmentally regulated expression of the mouse homologues of the potassium channel encoding genes m-erg1, m-erg2 and m-erg3
- Author
-
Polvani, S, Masi, A, Pillozzi, S, Gragnani, L, Crociani, O, Olivotto, M, Becchetti, A, Wanke, E, Arcangeli, A, Arcangeli, A., BECCHETTI, ANDREA, WANKE, ENZO, Polvani, S, Masi, A, Pillozzi, S, Gragnani, L, Crociani, O, Olivotto, M, Becchetti, A, Wanke, E, Arcangeli, A, Arcangeli, A., BECCHETTI, ANDREA, and WANKE, ENZO
- 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. (C) 2003 Elsevier B.V. All rights reserved.
- Published
- 2003
36. HERG K+ channels and beta1 integrins interact through the assembly of a macromolecular complex
- Author
-
Cherubini, A, Pillozzi, S, Hofmann, G, Crociani, O, Guasti, L, Lastraioli, E, Polvani, S, Masi, A, Becchetti, A, Wanke, E, Olivotto, M, Arcangeli, A, BECCHETTI, ANDREA, WANKE, ENZO, Arcangeli, A., Cherubini, A, Pillozzi, S, Hofmann, G, Crociani, O, Guasti, L, Lastraioli, E, Polvani, S, Masi, A, Becchetti, A, Wanke, E, Olivotto, M, Arcangeli, A, BECCHETTI, ANDREA, WANKE, ENZO, and Arcangeli, A.
- Published
- 2002
37. Irresponsiveness of two retinoblastoma cases to conservative therapy correlates with up- regulation of hERG1 channels and of the VEGF-A pathway
- Author
-
La Torre Agostino, Franchi Alessandro, Pollazzi Liliana, Tamburini Angela, Pillozzi Serena, Fortunato Pina, and Arcangeli Annarosa
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Treatment strategies for Retinoblastoma (RB), the most common primary intraocular tumor in children, have evolved over the past few decades and chemoreduction is currently the most popular treatment strategy. Despite success, systemic chemotherapeutic treatment has relevant toxicity, especially in the pediatric population. Antiangiogenic therapy has thus been proposed as a valuable alternative for pediatric malignancies, in particolar RB. Indeed, it has been shown that vessel density correlates with both local invasive growth and presence of metastases in RB, suggesting that angiogenesis could play a pivotal role for both local and systemic invasive growth in RB. We present here two cases of sporadic, bilateral RB that did not benefit from the conservative treatment and we provide evidence that the VEGF-A pathway is significantly up-regulated in both RB cases along with an over expression of hERG1 K+ channels. Case presentation Two patients showed a sporadic, bilateral RB, classified at Stage II of the Reese-Elsworth Classification. Neither of them got benefits from conservative treatment, and the two eyes were enucleated. In samples from both RB cases we studied the VEGF-A pathway: VEGF-A showed high levels in the vitreous, the vegf-a, flt-1, kdr, and hif1-α transcripts were over-expressed. Moreover, both the transcripts and proteins of the hERG1 K+ channels turned out to be up-regulated in the two RB cases compared to the non cancerous retinal tissue. Conclusions We provide evidence that the VEGF-A pathway is up-regulated in two particular aggressive cases of bilateral RB, which did not experience any benefit from conservative treatment, showing the overexpression of the vegf-a, flt-1, kdr and hif1-α transcripts and the high secretion of VEGF-A. Moreover we also show for the first time that the herg1 gene transcripts and protein are over expressed in RB, as occurs in several aggressive tumors. These results further stress the relevance of the VEGF-A pathway in RB and the correlation with hERG1, making aggressive and recurrent RB cases good candidates for antiangiogenesis therapies based on the targeting of VEGF-A.
- Published
- 2010
- Full Text
- View/download PDF
38. Irresponsiveness of two retinoblastoma cases to conservative therapy correlates with up- regulation of hERG1 channels and of the VEGF-A pathway.
- Author
-
Fortunato P, Pillozzi S, Tamburini A, Pollazzi L, Franchi A, La Torre A, Arcangeli A, Fortunato, Pina, Pillozzi, Serena, Tamburini, Angela, Pollazzi, Liliana, Franchi, Alessandro, La Torre, Agostino, and Arcangeli, Annarosa
- Abstract
Background: Treatment strategies for Retinoblastoma (RB), the most common primary intraocular tumor in children, have evolved over the past few decades and chemoreduction is currently the most popular treatment strategy. Despite success, systemic chemotherapeutic treatment has relevant toxicity, especially in the pediatric population. Antiangiogenic therapy has thus been proposed as a valuable alternative for pediatric malignancies, in particolar RB. Indeed, it has been shown that vessel density correlates with both local invasive growth and presence of metastases in RB, suggesting that angiogenesis could play a pivotal role for both local and systemic invasive growth in RB. We present here two cases of sporadic, bilateral RB that did not benefit from the conservative treatment and we provide evidence that the VEGF-A pathway is significantly up-regulated in both RB cases along with an over expression of hERG1 K+ channels.Case Presentation: Two patients showed a sporadic, bilateral RB, classified at Stage II of the Reese-Elsworth Classification. Neither of them got benefits from conservative treatment, and the two eyes were enucleated. In samples from both RB cases we studied the VEGF-A pathway: VEGF-A showed high levels in the vitreous, the vegf-a, flt-1, kdr, and hif1-α transcripts were over-expressed. Moreover, both the transcripts and proteins of the hERG1 K+ channels turned out to be up-regulated in the two RB cases compared to the non cancerous retinal tissue.Conclusions: We provide evidence that the VEGF-A pathway is up-regulated in two particular aggressive cases of bilateral RB, which did not experience any benefit from conservative treatment, showing the overexpression of the vegf-a, flt-1, kdr and hif1-α transcripts and the high secretion of VEGF-A. Moreover we also show for the first time that the herg1 gene transcripts and protein are over expressed in RB, as occurs in several aggressive tumors. These results further stress the relevance of the VEGF-A pathway in RB and the correlation with hERG1, making aggressive and recurrent RB cases good candidates for antiangiogenesis therapies based on the targeting of VEGF-A. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
39. Data describing the effects of the Macrolide Antibiotic Clarithromycin on preclinical mouse models of Colorectal Cancer
- Author
-
Serena Pillozzi, Matteo Stefanini, Giulia Petroni, Andrea Becchetti, Olivia Crociani, Annarosa Arcangeli, Petroni, G, Stefanini, M, Pillozzi, S, Crociani, O, Becchetti, A, and Arcangeli, A
- Subjects
Colorectal cancer ,Angiogenesis ,medicine.drug_class ,Antibiotics ,lcsh:Computer applications to medicine. Medical informatics ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,BIO/09 - FISIOLOGIA ,Clarithromycin ,Biochemistry, Genetics and Molecular Biology ,Autophagy ,Medicine ,lcsh:Science (General) ,Multiple myeloma ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,integumentary system ,business.industry ,Xenograft ,Macrolides ,Cancer ,food and beverages ,medicine.disease ,Apoptosis ,Cancer research ,lcsh:R858-859.7 ,lipids (amino acids, peptides, and proteins) ,Macrolide ,business ,030217 neurology & neurosurgery ,medicine.drug ,lcsh:Q1-390 - 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. Keywords: Macrolides, Colorectal cancer, Xenograft, Autophagy
- Published
- 2019
40. The combined activation of KCa3.1 and inhibition of Kv11.1/hERG1 currents contribute to overcome Cisplatin resistance in colorectal cancer cells
- Author
-
Massimo D'Amico, Luca Gasparoli, Mirko Severi, Tiziano Marzo, Giulia Petroni, Olivia Crociani, Gianluca Bartoli, Luigi Messori, Angela Guerriero, Heike Wulff, Serena Pillozzi, Annarosa Arcangeli, Andrea Becchetti, K. George Chandy, Roberto Udisti, Pillozzi, S, D'Amico, M, Bartoli, G, Gasparoli, L, Petroni, G, Crociani, O, Marzo, T, Guerriero, A, Messori, L, Severi, M, Udisti, R, Wulff, H, Chandy, K, Becchetti, A, Arcangeli, A, and Lee Kong Chian School of Medicine (LKCMedicine)
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,ERG1 Potassium Channel ,preclinical mouse models ,Cell ,Drug Resistance ,Apoptosis ,Mice ,BIO/09 - FISIOLOGIA ,Riluzole ,SKA-31 ,E4031 ,Cisplatin uptake ,Cancer ,Tumor ,Chemistry ,Cell Cycle ,Drug Synergism ,Cell cycle ,Intermediate-Conductance Calcium-Activated Potassium Channels ,Colo-Rectal Cancer ,medicine.anatomical_structure ,5.1 Pharmaceuticals ,Public Health and Health Services ,Development of treatments and therapeutic interventions ,Colorectal Neoplasms ,HT29 Cells ,medicine.drug ,medicine.medical_specialty ,Cell Survival ,Oncology and Carcinogenesis ,Cell Line ,03 medical and health sciences ,KCNN4 ,Inhibitory Concentration 50 ,In vivo ,Internal medicine ,medicine ,Potassium Channel Blockers ,Animals ,Humans ,Medicine [Science] ,Benzothiazoles ,Oncology & Carcinogenesis ,Cisplatin ,Activator (genetics) ,HCT116 Cells ,030104 developmental biology ,Cell culture ,Cancer research ,Pyrazoles ,Neoplasm ,cisplatin, KCa, hERG, colorectal cancer, chemoresistance ,Digestive Diseases - 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. Published version
- Published
- 2018
41. The conformational state of hERG1 channels determines integrin association, downstream signaling, and cancer progression
- Author
-
Maria Felice Brizzi, Massimo D'Amico, F. Zanieri, Davide Ricci, Alessio Masi, Laura Carraresi, Virginia Morello, Silvia Crescioli, Stefano Coppola, Olivia Crociani, Franco Quercioli, Serena Pillozzi, Antonella Fiore, Sagar S. Manoli, Paola Defilippi, Raffaella Mercatelli, Luca Gasparoli, Matteo Stefanini, Andrea Becchetti, Thomas Schmidt, Annarosa Arcangeli, Giulia Petroni, Mauro Rinaldi, Becchetti, A, Crescioli, S, Zanieri, F, Petroni, G, Mercatelli, R, Coppola, S, Gasparoli, L, D’Amico, M, Pillozzi, S, Crociani, O, Stefanini, M, Fiore, A, Carraresi, L, Morello, V, Manoli, S, Brizzi, M, Ricci, D, Rinaldi, M, Masi, A, Schmidt, T, Quercioli, F, Defilippi, P, and Arcangeli, A
- Subjects
0301 basic medicine ,Protein Conformation ,Nude ,Animals ,Cell Line, Tumor ,Disease Progression ,Ether-A-Go-Go Potassium Channels ,Fluorescence Resonance Energy Transfer ,HCT116 Cells ,HEK293 Cells ,Humans ,Immunoblotting ,Integrin beta1 ,Mice, Nude ,Mice, SCID ,Microscopy, Confocal ,Neoplasms ,Protein Binding ,Transplantation, Heterologous ,Signal Transduction ,Biochemistry ,Mice ,0302 clinical medicine ,BIO/09 - FISIOLOGIA ,Microscopy ,Heterologous ,Tumor ,biology ,Chemistry ,potassium channels ,Potassium channel ,Cell biology ,Confocal ,030220 oncology & carcinogenesis ,Signal transduction ,Integrin ,SCID ,Cell Line ,Focal adhesion ,03 medical and health sciences ,cell signaling ,cancer ,Molecular Biology ,Ion channel ,Transplantation ,HEK 293 cells ,Cell Biology ,030104 developmental biology ,Tumor progression ,Immunology ,Cancer cell ,hERG1, ion channels, integrin, proliferation, migration, neoplasia, cancer ,biology.protein - 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-a-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 beta(1) 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 beta(1) integrin-hERG1 interaction. Although open hERG1 channels did not interact as strongly with beta(1) integrins as did closed channels, current flow through hERG1 channels was necessary to activate the integrin-dependent phosphorylation of Tyr(397) 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/beta(1) integrin interaction was disrupted. We conclude that the interaction of beta(1) 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.
- Published
- 2017
42. Water-soluble Ru(II)- and Ru(III)-halide-PTA complexes (PTA=1,3,5-triaza-7-phosphaadamantane): Chemical and biological properties
- Author
-
Federica Scaletti, Enzo Alessio, Serena Pillozzi, Luigi Messori, Gianni Balducci, Federica Battistin, Annarosa Arcangeli, Battistin, Federica, Scaletti, F., Balducci, Gabriele, Pillozzi, S., Arcangeli, A., Messori, L, and Alessio, Enzo
- Subjects
Ruthenium complexes ,Oligonucleotides ,Adamantane ,Filaggrin Proteins ,Ligands ,01 natural sciences ,Medicinal chemistry ,Biochemistry ,chemistry.chemical_compound ,Coordination Complexes ,Oligonucleotide interaction ,Imidazole ,Solubility ,Aqueous solution ,Chemistry ,Protein interaction ,Imidazoles ,Cytochromes c ,Hydrogen-Ion Concentration ,Ruthenium ,PTA ,Protons ,Bromides ,Indazoles ,Stereochemistry ,Cell Survival ,chemistry.chemical_element ,Protonation ,Antineoplastic Agents ,010402 general chemistry ,Inorganic Chemistry ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Organophosphorus Compounds ,Chlorides ,Cell Line, Tumor ,Organometallic Compounds ,Molecule ,Humans ,Reactivity (chemistry) ,Indazole ,010405 organic chemistry ,In vitro antiproliferative activity ,Ruthenium complexe ,Water ,Ribonuclease, Pancreatic ,HCT116 Cells ,0104 chemical sciences - Abstract
Four structurally related Ru(II)-halide-PTA complexes, of general formula trans- or cis-[Ru(PTA)4X2] (PTA = 1,3,5-triaza-7-phosphaadamantane, X = Cl (1, 2), Br (3, 4), were prepared and characterized. Whereas compounds 1 and 2 are known, the corresponding bromo derivatives 3 and 4 are new. The Ru(III)-PTA compound trans-[RuCl4(PTAH)2]Cl (5, PTAH = PTA protonated at one N atom), structurally similar to the well-known Ru(III) anticancer drug candidates (Na)trans-[RuCl4(ind)2] (NKP-1339, ind = indazole) and (Him)trans-[RuCl4(dmso-S)(im)] (NAMI-A, im = imidazole), was also prepared and similarly investigated. Notably, the presence of PTA confers to all complexes an appreciable solubility in aqueous solutions at physiological pH. The chemical behavior of compounds 1–5 in water and in physiological buffer, their interactions with two model proteins – cytochrome c and ribonuclease A – as well as with a single strand oligonucleotide (5′-CGCGCG-3′), and their in vitro cytotoxicity against a human colon cancer cell line (HCT-116) and a myeloid leukemia (FLG 29.1) were investigated. Upon dissolution in the buffer, sequential halide replacement by water molecules was observed for complexes 1–4, with relatively slow kinetics, whereas the Ru(III) complex 5 is more inert. All tested compounds manifested moderate antiproliferative properties, the cis compounds 2 and 4 being slightly more active than the trans ones (1 and 3). Mass spectrometry experiments evidenced that all complexes exhibit a far higher reactivity towards the reference oligonucleotide than towards model proteins. The chemical and biological profiles of compounds 1–5 are compared to those of established ruthenium drug candidates in clinical development.
- Published
- 2016
43. Chemotherapy resistance in acute lymphoblastic leukemia requires hERG1 channels and is overcome by hERG1 blockers
- Author
-
Massimo D'Amico, Dario Campana, Marinella Veltroni, Emanuele Cilia, Olivia Crociani, Andrea Becchetti, Annarosa Arcangeli, Serena Pillozzi, Amedeo Amedei, Emanuele De Lorenzo, Giuseppe Basso, Benedetta Accordi, Marika Masselli, Pillozzi, S, Masselli, M, De Lorenzo, E, Accordi, B, Cilia, E, Crociani, O, Amedei, A, Veltroni, M, D'Amico, M, Basso, G, Becchetti, A, Campana, D, and Arcangeli, A
- Subjects
Receptors, CXCR4 ,ALL, ERG, potassium channels, doxorubicine, chemotherapeutics ,Pyridines ,Blotting, Western ,Immunology ,Antineoplastic Agents ,Mice, SCID ,Biochemistry ,Mice ,Chemokine receptor ,Piperidines ,BIO/09 - FISIOLOGIA ,Mice, Inbred NOD ,Acute lymphocytic leukemia ,Potassium Channel Blockers ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Protein kinase B ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,business.industry ,Integrin beta1 ,Lymphoblast ,Cell Membrane ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Xenograft Model Antitumor Assays ,Coculture Techniques ,Ether-A-Go-Go Potassium Channels ,medicine.anatomical_structure ,Doxorubicin ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,Cancer research ,Prednisone ,Female ,RNA Interference ,Bone marrow ,Signal transduction ,business ,Signal Transduction - 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 β1-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.
- Published
- 2011
44. Hepatocyte Growth Factor Receptor c-MET Is Associated with FAS and When Activated Enhances Drug-induced Apoptosis in Pediatric B Acute Lymphoblastic Leukemia with TEL-AML1 Translocation
- Author
-
Giovanni Cazzaniga, Serena Pillozzi, Marta Campo Dell'Orto, Annarosa Arcangeli, Geertruy te Kronnie, Giuseppe Basso, Benedetta Accordi, Accordi, B, Pillozzi, S, Dell'Orto, M, Cazzaniga, G, Arcangeli, A, Kronnie, G, and Basso, G
- Subjects
C-Met ,Cellular homeostasis ,Apoptosis ,Bone Marrow Cells ,Biochemistry ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,fas Receptor ,Molecular Biology ,Interleukin 3 ,Antibiotics, Antineoplastic ,Proto-Oncogene Proteins c-ets ,Proto-Oncogene Proteins c-et ,biology ,Apoptosi ,Cell Biology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Proto-Oncogene Proteins c-met ,Repressor Protein ,Fas receptor ,Repressor Proteins ,Protein Transport ,chemistry ,Doxorubicin ,Hepatocyte Growth Factor Receptor ,Core Binding Factor Alpha 2 Subunit ,Immunology ,Cancer research ,biology.protein ,Bone Marrow Cell ,RNA ,Interleukin-3 ,Hepatocyte growth factor ,Human ,medicine.drug - Abstract
Expression of c-MET, the HGF (hepatocyte growth factor) tyrosine kinase receptor, was investigated in pediatric B-acute lymphoblastic leukemia (ALL) patients. c-MET was found to be expressed in normal B cells and in B-ALL patients with the t(12;21) TEL-AML1 translocation, but it is not expressed in the most part of B-ALL without the t(12;21). We also found that c-MET, related to proliferation and protection from apoptosis, is associated with the pro-apoptotic protein FAS in TEL-AML1 B-ALL cells and in normal B lymphocytes. The possible role of this protein complex in drug-induced apoptosis was thus investigated in REH TEL-AML1 B-ALL cell line. REH cells prestimulated with HGF and treated with doxorubicin had shown a higher apoptotic rate than non-HGF-prestimulated ones (p = 0.03). REH cells stimulated with IL-3 and treated with doxorubicin did not undergo apoptosis more than nonstimulated cells, demonstrating that increased proliferation in itself is not directly related to the higher apoptotic sensitivity observed with HGF stimulation. These results indicate that c-MET activation enhances specifically FAS-mediated apoptosis in TEL-AML1 ALL cells and, considering that the c-MET/FAS complex is present only in normal B lymphocytes and in TEL-AML1 leukemias, this implies that it may have an important contribution in cellular homeostasis and in high sensitivity of TEL-AML1 ALL to chemotherapeutic regimens.
- Published
- 2007
45. VEGFR-1 (FLT-1), β1 integrin, and hERG K+ channel for a macromolecular signaling complex in acute myeloid leukemia: role in cell migration and clinical outcome
- Author
-
Maria Felice Brizzi, Serena Pillozzi, Benedetta Bartolozzi, Luigi Pegoraro, Roberto Caporale, Venere Basile, Vieri Boddi, P. A. Bernabei, Andrea Becchetti, Annarosa Arcangeli, Pillozzi, S, Brizzi, M, Bernabei, P, Bartolozzi, B, Caporale, L, Basile, V, Boddi, V, Pegoraro, L, Becchetti, A, and Arcangeli, A
- Subjects
Adult ,Male ,Cell Survival ,Immunology ,Population ,Integrin ,Biology ,Biochemistry ,Immunoenzyme Techniques ,chemistry.chemical_compound ,Cell Movement ,BIO/09 - FISIOLOGIA ,hemic and lymphatic diseases ,Cell Adhesion ,Tumor Cells, Cultured ,medicine ,Humans ,Neoplasm Invasiveness ,Phosphorylation ,RNA, Small Interfering ,education ,Aged ,education.field_of_study ,Vascular Endothelial Growth Factor Receptor-1 ,Integrin beta1 ,Myeloid leukemia ,Cell migration ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,Gene Expression Regulation, Neoplastic ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Leukemia ,chemistry ,Leukemia, Myeloid ,Acute Disease ,Cancer research ,biology.protein ,Female ,potassium channel, leukaemia, AML, VEGF, integrins, macromolecular complex ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Leukemia cell motility and transendothelial migration into extramedullary sites are regulated by angiogenic factors and are considered unfavorable prognostic factors in acute leukemias. We have studied cross talk among (1) the vascular endothelial growth factor receptor-1, FLT-1; (2) the human eag-related gene 1 (hERG1) K+ channels; and (3) integrin receptors in acute myeloid leukemia (AML) cells. FLT-1, hERG1, and the β1 integrin were found to form a macromolecular signaling complex. The latter mostly recruited the hERG1B isoform of hERG1 channels, and its assembly was necessary for FLT-1 signaling activation and AML cell migration. Both effects were inhibited when hERG1 channels were specifically blocked. A FLT-1/hERG1/ β1 complex was also observed in primary AML blasts, obtained from a population of human patients. The co-expression of FLT-1 and hERG1 conferred a pro-migratory phenotype to AML blasts. Such a phenotype was also observed in vivo. The hERG1-positive blasts were more efficient in invading the peripheral circulation and the extramedullary sites after engraftment into immunodeficient mice. Moreover, hERG1 expression in leukemia patients correlated with a higher probability of relapse and shorter survival periods. We conclude that in AML, hERG1 channels mediate the FLT-1-dependent cell migration and invasion, and hence confer a greater malignancy. © 2007 by The American Society of Hematology.
- Published
- 2007
46. New pyrimido-indole compound CD-160130 preferentially inhibits the KV11.1B isoform and produces antileukemic effects without cardiotoxicity
- Author
-
Massimo D'Amico, John S. Mitcheson, Luca Gasparoli, Andrea Becchetti, Rawan Khuwaileh, Wolfgang Tiedke, Serena Pillozzi, Marika Masselli, Rachel E. Caves, Giuseppe Basso, Kenneth Mugridge, Annarosa Arcangeli, Alessandro Pratesi, Gasparoli, L, D'Amico, M, Masselli, M, Pillozzi, S, Caves, R, Khuwaileh, R, Tiedke, W, Mugridge, K, Pratesi, A, Mitcheson, J, Basso, G, Becchetti, A, and Arcangeli, A
- Subjects
Male ,Gene isoform ,ERG1 Potassium Channel ,Indoles ,Stromal cell ,Kir2.1 ,Chronic lymphocytic leukemia ,Guinea Pigs ,Antineoplastic Agents ,HL-60 Cells ,CHO Cells ,Pyrimidinones ,Pharmacology ,Cardiotoxins ,MSC ,Mice ,Cricetulus ,BIO/09 - FISIOLOGIA ,In vivo ,Cell Line, Tumor ,Cricetinae ,Leukemia, B-Cell ,medicine ,Animals ,Humans ,Protein Isoforms ,cancer ,hERG ,Kv1.5 ,Cardiotoxicity ,ECG ,Chemistry ,Kv1.3 ,Cardiac action potential ,medicine.disease ,Xenograft Model Antitumor Assays ,Ether-A-Go-Go Potassium Channels ,Leukemia ,HEK293 Cells ,Pyrimidines ,Treatment Outcome ,medicine.anatomical_structure ,Molecular Medicine ,Female ,KCa3.1 ,Bone marrow ,CLL ,potassium channel - Abstract
KV11.1 (hERG1) channels are often overexpressed in human cancers. In leukemias, KV11.1 regulates pro-survival signals that promote resistance to chemotherapy, raising the possibility that inhibitors of KV11.1 could be therapeutically beneficial. However, because of the role of KV11.1 in cardiac repolarization, blocking these channels may cause cardiac arrhythmias. We show that CD-160130, a novel pyrimido-indole compound, blocksKV11.1 channels with a higher efficacy for the KV 11.1 isoform B, in which the IC50 (1.8 μM) was approximately 10-fold lower than observed in KV 11.1 isoform A. At this concentration, CD-160130 also had minor effects on Kir 2.1, KV 1.3,KV1.5, and KCa 3.1. In vitro, CD-160130 induced leukemia cell apoptosis, and could overcome bone marrow mesenchymal stromal cell (MSC)-induced chemoresistance. This effect was caused by interference with the survival signaling pathways triggered by MSCs. In vivo, CD-160130 produced an antileukemic activity, stronger than that caused by cytarabine. Consistent with its atypical target specificity, CD-160130 did not bind to the main binding site of the arrhythmogenicKV11.1 blockers (the Phe656 pore residue). Importantly, in guinea pigs CD-160130 produced neither alteration of the cardiac action potential shape in dissociated cardiomyocytes nor any lengthening of the QT interval in vivo. Moreover, CD-160130 had no myelotoxicity on human bone marrow-derived cells. Therefore, CD-160130 is a promising first-in-class compound to attempt oncologic therapy without cardiotoxicity, based on targeting KV11.1. Because leukemia and cardiac cells tend to express different ratios of the A and B KV 11.1 isoforms, the pharmacological properties of CD-160130 may depend, at least in part, on isoform specificity.
- Published
- 2015
47. HERG potassium channels are constitutively expressed in primary human acute myeloid leukemias and regulate cell proliferation of normal and leukemic hemopoietic progenitors
- Author
-
Luigi Pegoraro, Alessia Cherubini, Massimo Olivotto, Annarosa Arcangeli, Leonardo Guasti, Enzo Wanke, Andrea Becchetti, Maria Felice Brizzi, P A Bernabei, Serena Pillozzi, Manuela Balzi, Olivia Crociani, Benedetta Bartolozzi, Pillozzi, S, Brizzi, M, Balzi, M, Crociani, O, Cherubini, A, Guasti, L, Bartolozzi, B, Becchetti, A, Wanke, E, Bernabei, P, Olivotto, M, Pegoraro, L, and Arcangeli, A
- Subjects
ERG1 Potassium Channel ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Patch-Clamp Techniques ,Potassium Channels ,hERG ,HERG inhibitors ,HERG ,Antigens, CD34 ,Biology ,hemopoietic progenitor ,Immunoenzyme Techniques ,Transcriptional Regulator ERG ,AML ,hemic and lymphatic diseases ,Precursor cell ,Sulfanilamides ,Potassium Channel Blockers ,Tumor Cells, Cultured ,medicine ,Humans ,Cation Transport Proteins ,CD34(+) ,Voltage-gated ion channel ,Cell growth ,Hematology ,Hematopoietic Stem Cells ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,Potassium channel ,DNA-Binding Proteins ,Leukemia ,Oncology ,Leukemia, Myeloid ,Potassium Channels, Voltage-Gated ,Acute Disease ,Cancer cell ,Immunology ,Trans-Activators ,Cancer research ,biology.protein ,Benzimidazoles ,Stem cell ,Cell Division - Abstract
An important target in the understanding of the pathogenesis of acute myeloid leukemias (AML) relies on deciphering the molecular features of normal and leukemic hemopoietic progenitors. In particular, the analysis of the mechanisms involved in the regulation of cell proliferation is decisive for the establishment of new targeted therapies. To gain further insight into this topic we report herein a novel approach by analyzing the role of HERG K(+) channels in the regulation of hemopoietic cell proliferation. These channels, encoded by the human ether-a-gò-gò-related gene (herg), belong to a family of K(+) channels, whose role in oncogenesis has been recently demonstrated. We report here that herg is switched off in normal peripheral blood mononuclear cells (PBMNC) as well as in circulating CD34(+) cells, however, it is rapidly turned on in the latter upon induction of the mitotic cycle. Moreover, hergappears to be constitutively activated in leukemic cell lines as well as in the majority of circulating blasts from primary AML. Evidence is also provided that HERG channel activity regulates cell proliferation in stimulated CD34(+) as well as in blast cells from AML patients. These results open new perspectives on the pathogenetic role of HERG K(+) channels in leukemias.
- Published
- 2002
48. NAMI-A is highly cytotoxic toward leukaemia cell lines: evidence of inhibition of KCa3.1 channels
- Author
-
Luca Gasparoli, Matteo Stefanini, Serena Pillozzi, Federica Scaletti, Enzo Alessio, Mirco Ristori, Andrea Becchetti, Luigi Messori, Annarosa Arcangeli, Massimo D'Amico, Pillozzi, S, Gasparoli, L, Stefanini, M, Ristori, M, D'Amico, M, Alessio, E, Scaletti, F, Becchetti, A, Arcangeli, A, Messori, L, Serena, Pillozzi, Luca, Gasparoli, Matteo, Stefanini, Mirco, Ristori, Massimo, D'Amico, Alessio, Enzo, Federica, Scaletti, Andrea, Becchetti, Annarosa, Arcangeli, and Luigi, Messori
- Subjects
Cell Survival ,Leukaemia cell ,Antineoplastic Agents ,Apoptosis ,inorganic medicinal chemistry ,Selective inhibition ,Pharmacology ,KCa channel ,Inorganic Chemistry ,chemistry.chemical_compound ,antimetastatic ,BIO/09 - FISIOLOGIA ,Antimetastatic Agent ,Cell Line, Tumor ,medicine ,Organometallic Compounds ,Potassium Channel Blockers ,Cytotoxic T cell ,NAMI-A ,Humans ,Dimethyl Sulfoxide ,ruthenium ,Leukemia ,KCa channels ,Cell Proliferation ,Cisplatin ,Chemistry ,Cell Cycle ,leukemia ,ion channels ,Intermediate-Conductance Calcium-Activated Potassium Channels ,Potassium channel ,Ruthenium Compounds ,KCa3.1 ,medicine.drug - Abstract
We report here that the established anticancer ruthenium(iii) complex NAMI-A induces potent and selective cytotoxic effects in a few leukaemia cell lines. These results sound very surprising after 20 years of intense studies on NAMI-A, commonly considered as a "non-cytotoxic" antimetastatic agent. In addition, evidence is given for selective inhibition of KCa 3.1 channels. The implications of these findings are discussed. © 2014 the Partner Organisations.
- Published
- 2014
49. Differential expression of hERG1A and hERG1B genes in pediatric acute lymphoblastic leukemia identifies different prognostic subgroups
- Author
-
Paola Rebora, Serena Pillozzi, Annarosa Arcangeli, Benedetta Accordi, Serafin, Giuseppe Basso, Maria Grazia Valsecchi, Pillozzi, S, Accordi, B, Rebora, P, Serafin, V, Valsecchi, M, Basso, G, and Arcangeli, A
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Messenger ,Real-Time Polymerase Chain Reaction ,Pediatrics ,Precursor Cell Lymphoblastic Leukemia Lymphoma ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Neoplasm Recurrence ,Pediatric Acute Lymphoblastic Leukemia ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Differential expression ,Child ,Preschool ,Gene ,Letter to the Editor ,030304 developmental biology ,0303 health sciences ,Acute lymphoblastic leukemia ,hERG1A , hERG1B ,business.industry ,Reverse Transcriptase Polymerase Chain Reaction ,RNA ,Infant ,hemic and immune systems ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Child, Preschool ,Ether-A-Go-Go Potassium Channels ,Female ,Neoplasm Recurrence, Local ,Prognosis ,RNA, Messenger ,Real-time polymerase chain reaction ,Local ,030220 oncology & carcinogenesis ,Immunology ,business - Abstract
Differential expression of hERG1A and hERG1B genes in pediatric acute lymphoblastic leukemia identifies different prognostic subgroups
- Published
- 2014
50. Macrolide antibiotics exert antileukemic effects by modulating the autophagic flux through inhibition of hERG1 potassium channels
- Author
-
Marika Masselli, Lucia Polletta, Andrea Becchetti, Giuseppe Basso, Marinella Veltroni, Luca Gasparoli, Claudio Favre, Massimo D'Amico, Annarosa Arcangeli, Serena Pillozzi, Pillozzi, S, Masselli, M, Gasparoli, L, D'Amico, M, Polletta, L, Veltroni, M, Favre, C, Basso, G, Becchetti, A, and Arcangeli, A
- Subjects
0301 basic medicine ,ERG1 Potassium Channel ,medicine.drug_class ,Antineoplastic Agents ,HL-60 Cells ,Tumor cells ,Pharmacology ,Biology ,Macrolide Antibiotics ,03 medical and health sciences ,0302 clinical medicine ,BIO/09 - FISIOLOGIA ,Autophagy ,Tumor Cells, Cultured ,medicine ,Humans ,Letter to the Editor ,Early Detection of Cancer ,Leukemia ,ether-a-go-go-realted gene 1 ,Translational biology ,Hematology ,clarithromycin ,Potassium channel ,Anti-Bacterial Agents ,030104 developmental biology ,Oncology ,erythromycin ,Apoptosis ,030220 oncology & carcinogenesis ,Acute Disease ,Immunology ,Macrolides ,Flux (metabolism) ,Signal Transduction - Abstract
Macrolide antibiotics exert antileukemic effects by modulating the autophagic flux through inhibition of hERG1 potassium channels
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.