93 results on '"Riganti C"'
Search Results
2. Selective delivery of pentamidine toward cancer cells by self-assembled nanoparticles
- Author
-
Andreana, I., Gazzano, E., Gianquinto, E., Piatti, G., Bincoletto, V., Kryza, D., Lollo, G., Spyrakis, F., Riganti, C., Arpicco, S., and Stella, B.
- Published
- 2022
- Full Text
- View/download PDF
3. Exploring the mechanisms of metabolic adaptations to cardiotoxic doxorubicin
- Author
-
Guerra, G, primary, Russo, M, additional, Priolo, R, additional, Riganti, C, additional, Reano, S, additional, Filigheddu, N, additional, Hirsch, E, additional, and Ghigo, A, additional
- Published
- 2024
- Full Text
- View/download PDF
4. Cardiometabolic rewiring in the context of doxorubicin-induced cardiotoxicity: fuel preference changes from fatty acids to glucose oxidation
- Author
-
Russo, M, primary, Guerra, G, additional, Priolo, R, additional, Riganti, C, additional, Reano, S, additional, Filigheddu, N, additional, Hirsch, E, additional, and Ghigo, A, additional
- Published
- 2023
- Full Text
- View/download PDF
5. Tuning of surface chemical and optical properties of nanodiamonds for biosensing and drug delivery applications
- Author
-
Aprà, Pietro, primary, Arpicco, S., additional, Bincoletto, V., additional, Bernardi, E., additional, Moreva, E., additional, Genovese, M., additional, Kopecka, J., additional, Losero, E., additional, Mino, L., additional, Olivero, P., additional, Riganti, C., additional, Stella, C., additional, Sturari, S., additional, Traina, P., additional, Varzi, V., additional, Zanelli, G., additional, and Picollo, F., additional
- Published
- 2023
- Full Text
- View/download PDF
6. Hazards assessment of micro- and nanoplastics and associated additives/contaminants to human health
- Author
-
Katsumiti, Alberto, Rodríguez-Llopis, Isabel, Boland, S, Devineau, S, Miremont, D, Ramsperger, A.F.R.M, Löder, MGJ, Laforsch, C, Drobne, D, Michelini, S, Repar, N, Hoekstra, L, Duivenvoorde, L, van der Zande, M, Tamargo, A, Di Maira, G, Tedesco, E, Benetti, F, Antonello, G, Fenoglio, I, Riganti, C, Bergamaschi, E, Jacobsen, N.R, Kazour, M, Peijnenburg, W, Kühnel, D, Haugen, Ø. P, Wallin, H, Narui, S, and Afanou, A.J.K
- Subjects
hazard assessment ,microplastics ,exposure ,toxicity ,human health ,nanoplastics ,plastic particles - Abstract
Summary of objectives, test materials, target systems,strategies, and expected outcomes of Work Package 3 of the PlasticsFatE Project PlasticsFatE has received funding from the European Union’s Horizon 2020 Research and Innovation programme, under the Grant Agreement number 965367
- Published
- 2022
- Full Text
- View/download PDF
7. IMMUNE SUPPRESSION OF BONE MARROW Vγ9Vδ2 T CELLS BY BONE MARROW STROMAL CELLS (BMSC) IN MULTIPLE MYELOMA
- Author
-
Giannotta, C, Castella, B, Tripoli, E, Riganti, C, Salaroglio, Ic, D'Agostino, M, and Massaia., M
- Published
- 2022
8. Sdox, a H2S releasing anthracycline, with a safer profile than doxorubicin toward vasculature
- Author
-
Durante, M., Frosini, M., Chiaino, E., Fusi, F., Gamberucci, A., Gorelli, B., Chegaev, K., Riganti, C., and Saponara, S.
- Subjects
Pharmacology ,Myocytes ,Physiology ,A7r5 cells ,Doxorubicin ,EA.hy926 cells ,Rat aorta ring ,Vascular toxicity ,Muscle, Smooth, Vascular ,Myocytes, Smooth Muscle ,Anthracyclines ,Antibiotics, Antineoplastic ,Antineoplastic ,Smooth Muscle ,Antibiotics ,Vascular ,Molecular Medicine ,Muscle ,Smooth - Published
- 2022
9. Safety and Efficacy of Magnet Use to Temporarily Inhibit Inappropriate Subcutaneous Implantable Cardioverter Defibrillator Therapy in Emergency Situations: A Case Report
- Author
-
Santomauro Maurizio, Riganti Carla, Santomauro Mario Alberto, Viggiano Aniello, Castellano Gaetano, Iovino Gianluigi, Rapacciuolo Antonio, Fiore Francesco, Cacciatore Francesco, and Esposito Giovanni
- Subjects
subcutaneous implantable cardioverter defibrillator ,inappropriate shock ,clinical magnet ,inhibition shock therapy ,magnet interference ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Introduction: The subcutaneous implantable cardioverter defibrillator (S-ICD) represents a major advancement in ICD technology. Inappropriate shocks (IAS) occur in more than 3.1% of the population with S-ICD each year and are usually followed by admission to the emergency department (ED). In this setting, the disabling of IAS is mandatory during a pseudo-electrical storm (ES). This report describes the strategies that can be followed in order to temporarily inhibit IAS in critical care settings with the use of magnets.
- Published
- 2022
- Full Text
- View/download PDF
10. Ketogal Safety Profile in Human Primary Colonic Epithelial Cells and in Mice
- Author
-
Antonio Calignano, Roberto Russo, Monica Tizzano, Maria Grazia Rimoli, Stefania Albrizio, Barbara Rolando, Chiara Riganti, Elena Gazzano, Salvatore Magliocca, Chiara Fogliano, Bice Avallone, Mariarosaria Cuozzo, Federica Sodano, Claudia Cristiano, Sodano, F., Avallone, B., Tizzano, M., Fogliano, C., Rolando, B., Gazzano, E., Riganti, C., Magliocca, S., Cuozzo, M., Albrizio, S., Calignano, A., Cristiano, C., Russo, R., and Rimoli, M. G.
- Subjects
histological evaluation ,Pharmaceutical Science ,ketorolac ,ketogal ,Pharmacology ,medicine.disease_cause ,Article ,Pharmacy and materia medica ,Oral administration ,Drug Discovery ,medicine ,Mitochondrial oxida-tive stress ,mitochondrial oxidative stress ,Chemistry ,Colonic cytotoxicity ,Histological evaluation ,Ketogal ,Ketorolac ,Prodrug ,In vitro ,Small intestine ,RS1-441 ,body regions ,medicine.anatomical_structure ,colonic cytotoxicity ,Toxicity ,Molecular Medicine ,Medicine ,Ex vivo ,Oxidative stress ,medicine.drug - Abstract
In our previous studies, a ketorolac–galactose conjugate (ketogal) showed prolonged anti-inflammatory and analgesic activity, causing less gastric ulcerogenic effect and renal toxicity than its parent drug ketorolac. In order to demonstrate the safer profile of ketogal compared to ketorolac, histopathological changes in the small intestine and liver using three staining techniques before and after repeated oral administration in mice with ketorolac or an equimolecular dose of its galactosylated prodrug ketogal were assessed. Cytotoxicity and oxidative stress parameters were evaluated and compared in ketorolac- and ketogal-treated Human Primary Colonic Epithelial cells at different concentrations and incubation times. Evidence of mitochondrial oxidative stress was found after ketorolac treatment, this was attributable to altered mitochondrial membrane depolarization and oxidative stress parameters. No mitochondrial damage was observed after ketogal treatment. In ketorolac-treated mice, severe subepithelial vacuolation and erosion with inflammatory infiltrates and edematous area in the intestinal tissues were noted, as well as alterations in sinusoidal spaces and hepatocytes with foamy cytoplasm. In contrast, treatment with ketogal provided a significant improvement in the morphology of both organs. The prodrug clearly demonstrated a safer profile than its parent drug both in vitro and ex vivo, confirming that ketogal is a strategic alternative to ketorolac.
- Published
- 2021
11. Blood-brain barrier permeability increases with the differentiation of glioblastoma cells in vitro.
- Author
-
Digiovanni S, Lorenzati M, Bianciotto OT, Godel M, Fontana S, Akman M, Costamagna C, Couraud PO, Buffo A, Kopecka J, Riganti C, and Salaroglio IC
- Subjects
- Humans, Coculture Techniques, Cell Line, Tumor, STAT3 Transcription Factor metabolism, Permeability, Glioblastoma metabolism, Blood-Brain Barrier metabolism, Blood-Brain Barrier drug effects, Cell Differentiation physiology, Cell Differentiation drug effects, Brain Neoplasms metabolism
- Abstract
Background: Glioblastoma multiforme (GBM) is an aggressive tumor, difficult to treat pharmacologically because of the blood-brain barrier (BBB), which is rich in ATP-binding cassette (ABC) transporters and tight junction (TJ) proteins. The BBB is disrupted within GBM bulk, but it is competent in brain-adjacent-to-tumor areas, where eventual GBM foci can trigger tumor relapse. How GBM cells influence the permeability of BBB is poorly investigated., Methods: To clarify this point, we co-cultured human BBB models with 3 patient-derived GBM cells, after separating from each tumor the stem cell/neurosphere (SC/NS) and the differentiated/adherent cell (AC) components. Also, we set up cultures of BBB cells with the conditioned medium of NS or AC, enriched or depleted of IL-6. Extracellular cytokines were measured by protein arrays and ELISA. The intracellular signaling in BBB cells was measured by immunoblotting, in the presence of STAT3 pharmacological inhibitor or specific PROTAC. The competence of BBB was evaluated by permeability assays and TEER measurement., Results: The presence of GBM cells or their conditioned medium increased the permeability to doxorubicin, mitoxantrone and dextran-70, decreased TEER, down-regulated ABC transporters and TJ proteins at the transcriptional level. These effects were higher with AC or their medium than with NS. The secretome analysis identified IL-6 as significantly more produced by AC than by NS. Notably, AC-conditioned medium treated with an IL-6 neutralizing antibody reduced the BBB permeability to NS levels, while NS-conditioned medium enriched with IL-6 increased BBB permeability to AC levels. Mechanistically, IL-6 released by AC GBM cells activated STAT3 in BBB cells. In turn, STAT3 down-regulated ABC transporter and TJ expression, increased permeability and decreased TEER. The same effects were obtained in BBB cells treated with STA-21, a pharmacological inhibitor of STAT3, or with a PROTAC targeting STAT3., Conclusions: Our work demonstrates for the first time that the degree of GBM differentiation influences BBB permeability. The crosstalk between GBM cells that release IL-6 and BBB cells that respond by activating STAT3, controls the expression of ABC transporters and TJ proteins on BBB. These results may pave the way for novel therapeutic tools to tune BBB permeability and improve drug delivery to GBM., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Increasing membrane polyunsaturated fatty acids sensitizes non-small cell lung cancer to anti-PD-1/PD-L1 immunotherapy.
- Author
-
La Vecchia S, Fontana S, Salaroglio IC, Anobile DP, Digiovanni S, Akman M, Jafari N, Godel M, Costamagna C, Corbet C, Kopecka J, and Riganti C
- Subjects
- Humans, Animals, Programmed Cell Death 1 Receptor metabolism, Programmed Cell Death 1 Receptor antagonists & inhibitors, Letrozole pharmacology, Letrozole therapeutic use, Mice, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Cell Line, Tumor, Immunotherapy methods, Sterol Regulatory Element Binding Protein 1 metabolism, Sterol Regulatory Element Binding Protein 1 genetics, Stearoyl-CoA Desaturase metabolism, Stearoyl-CoA Desaturase genetics, Xenograft Model Antitumor Assays, Female, Docosahexaenoic Acids pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms immunology, Lung Neoplasms genetics, Lung Neoplasms pathology, B7-H1 Antigen metabolism, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Fatty Acids, Unsaturated pharmacology
- Abstract
Immune checkpoints inhibitors (ICIs) as anti-PD-1/anti-PD-L1 have been approved as first-line treatment in patients with non-small cell lung cancer (NSCLC), but only 25 % of patients achieve durable response. We previously unveiled that estrogen receptor α transcriptionally up-regulates PD-L1 and aromatase inhibitors such as letrozole increase the efficacy of pembrolizumab. Here we investigated if letrozole may have additional immune-sensitizing mechanisms. We found that higher the level of PD-L1 in NSCLC, higher the activation of SREBP1c that transcriptionally increases fatty acid synthase and stearoyl-CoA desaturase enzymes, increasing the amount of polyunsaturated fatty acids (PUFAs). Letrozole further up-regulated SREBP1c-mediated transcription of lipogenic genes, and increased the amount of PUFAs, thereby leading to greater membrane fluidity and reduced binding between PD-L1 and PD-1. The same effects were observed upon supplementation with ω3-PUFA docosahexaenoic acid (DHA) that enhanced the efficacy of pembrolizumab in humanized NSCLC immune-xenografts. We suggest that PUFA enrichment in membrane phospholipids improves the efficacy of ICIs. We propose to repurpose letrozole or DHA as new immune-sensitizing agents in NSCLC., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. An intrinsic mechanism of metabolic tuning promotes cardiac resilience to stress.
- Author
-
Sorge M, Savoré G, Gallo A, Acquarone D, Sbroggiò M, Velasco S, Zamporlini F, Femminò S, Moiso E, Morciano G, Balmas E, Raimondi A, Nattenberg G, Stefania R, Tacchetti C, Rizzo AM, Corsetto P, Ghigo A, Turco E, Altruda F, Silengo L, Pinton P, Raffaelli N, Sniadecki NJ, Penna C, Pagliaro P, Hirsch E, Riganti C, Tarone G, Bertero A, and Brancaccio M
- Subjects
- Animals, Humans, Mice, Lipid Metabolism, Stress, Physiological, Oxidation-Reduction, Myocardium metabolism, Trimetazidine pharmacology, Mitochondria metabolism, Myocytes, Cardiac metabolism, Reactive Oxygen Species metabolism
- Abstract
Defining the molecular mechanisms underlying cardiac resilience is crucial to find effective approaches to protect the heart. A physiologic level of ROS is produced in the heart by fatty acid oxidation, but stressful events can boost ROS and cause mitochondrial dysfunction and cardiac functional impairment. Melusin is a muscle specific chaperone required for myocardial compensatory remodeling during stress. Here we report that Melusin localizes in mitochondria where it binds the mitochondrial trifunctional protein, a key enzyme in fatty acid oxidation, and decreases it activity. Studying both mice and human induced pluripotent stem cell-derived cardiomyocytes, we found that Melusin reduces lipid oxidation in the myocardium and limits ROS generation in steady state and during pressure overload and doxorubicin treatment, preventing mitochondrial dysfunction. Accordingly, the treatment with the lipid oxidation inhibitor Trimetazidine concomitantly with stressful stimuli limits ROS accumulation and prevents long-term heart dysfunction. These findings disclose a physiologic mechanism of metabolic regulation in the heart and demonstrate that a timely restriction of lipid metabolism represents a potential therapeutic strategy to improve cardiac resilience to stress., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. A novel combinatory treatment against a CDDP-resistant non-small cell lung cancer based on a Ruthenium(II)-cyclopentadienyl compound.
- Author
-
Salaroglio IC, Stefanova D, Teixeira RG, Oliveira NFB, Ahmed A, Fusi F, Tzankova V, Yordanov Y, Machuqueiro M, Saponara S, Valente A, and Riganti C
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Nude, Apoptosis drug effects, Xenograft Model Antitumor Assays, Ruthenium chemistry, Ruthenium pharmacology, Mice, DNA Damage drug effects, Organometallic Compounds pharmacology, Organometallic Compounds therapeutic use, Female, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Cisplatin pharmacology, Cisplatin therapeutic use, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Drug Resistance, Neoplasm drug effects, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
The therapeutic approach to many solid tumors, including non-small cell lung cancer (NSCLC), is mainly based on the use of platinum-containing anticancer agents and is often characterized by acquired or intrinsic resistance to the drug. Therefore, the search for safer and more effective drugs is still an open challenge. Two organometallic ruthenium(II)-cyclopentadienyl compounds [Ru(η
5 -C5 H4 CHO)(Me2 bipy)(PPh3 )]+ (RT150) and [Ru(η5 -C5 H4 CH2 OH)(Me2 bipy)(PPh3 )][CF3 SO3 ] (RT151) were tested against a panel of cisplatin-resistant NSCLC cell lines and xenografts. They were more effective than cisplatin in inducing oxidative stress and DNA damage, affecting the cell cycle and causing apoptosis. Importantly, they were found to be inhibitors of drug efflux transporters. Due to this property, the compounds significantly increased the retention and cytotoxicity of cisplatin within NSCLC cells. Notably, they did not display high toxicity in vitro against non-transformed cells (red blood cells, fibroblasts, bronchial epithelial cells, cardiomyocytes, and endothelial cells). Both compounds induced vasorelaxation and reduced endothelial cell migration, suggesting potential anti-angiogenic properties. RT151 confirmed its efficacy against NSCLC xenografts resistant to cisplatin. Either alone or combined with low doses of cisplatin, RT151 showed a good biodistribution profile in the liver, kidney, spleen, lung, and tumor. Hematochemical analysis and post-mortem organ pathology confirmed the safety of the compound in vivo, also when combined with cisplatin. To sum up, we have confirmed the effectiveness of a novel class of drugs against cisplatin-resistant NSCLC. Additionally, the compounds have a good biocompatibility and safety profile., Competing Interests: Declaration of Competing Interest The Authors declare that they have not conflicts of interest. The Authors declare that they do not have used any artificial intelligence-based tools during the writing process., (Copyright © 2024. Published by Elsevier Ltd.)- Published
- 2024
- Full Text
- View/download PDF
15. Reduction of inappropriate shock rate through signal filtering (smart-pass) in patients with implantable subcutaneous cardioverter-defibrillator: a systematic review and meta-analysis.
- Author
-
Santomauro M, Petretta M, Riganti C, Santomauro MA, De Luca M, Santomauro A, and Cittadini A
- Abstract
Subcutaneous implantable cardioverter-defibrillators (S-ICD) are effective in protecting patients against sudden death but expose them to a higher risk of inappropriate shock (IAS). We performed a systematic search of studies published between January 2010 and December 2019 assessing IAS due to cardiac oversensing by the selection process (PRISMA) and identified 17 eligible articles. Fifteen studies were observational, and two studies were retrospective. For the meta-analysis, the final population included 6111 patients: 3356 without SMART pass (SP) filter (group 1) and 2755 with SP filter (group 2). 1614 shocks (appropriate shocks plus IAS) were registered (1245 in group 1 and 369 in group 2). The random effects meta-analysis estimated an overall IAS rate of 7.78% (95% confidence interval: 4.93-10.64) with substantial variability between studies (I square=96.05%, p<0.001). The IAS rate was 10.75% (95% confidence interval: 8.49-13.02) for group 1 and 3.61% (95% confidence interval: 1.36-5.86) for group 2 (p<0.001). Third-generation S-ICD technology with SP filters reduced the risk of cardiac signal-related IAS.
- Published
- 2024
- Full Text
- View/download PDF
16. Correction: TFEB controls sensitivity to chemotherapy and immuno-killing in non-small cell lung cancer.
- Author
-
Akman M, Monteleone C, Doronzo G, Godel M, Napoli F, Merlini A, Campani V, Nele V, Balmas E, Chontorotzea T, Fontana S, Digiovanni S, Alice Barbu F, Astanina E, Jafari N, Chiara Salaroglio I, Kopecka J, De Rosa G, Mohr T, Bertero A, Righi L, Novello S, Vittorio Scagliotti G, Bussolino F, and Riganti C
- Published
- 2024
- Full Text
- View/download PDF
17. TFEB controls sensitivity to chemotherapy and immuno-killing in non-small cell lung cancer.
- Author
-
Akman M, Monteleone C, Doronzo G, Godel M, Napoli F, Merlini A, Campani V, Nele V, Balmas E, Chontorotzea T, Fontana S, Digiovanni S, Barbu FA, Astanina E, Jafari N, Salaroglio IC, Kopecka J, De Rosa G, Mohr T, Bertero A, Righi L, Novello S, Scagliotti GV, Bussolino F, and Riganti C
- Subjects
- Humans, Mice, Animals, Female, Immunotherapy methods, Cell Line, Tumor, Male, Retrospective Studies, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Lung Neoplasms drug therapy, Lung Neoplasms immunology, Lung Neoplasms pathology, Lung Neoplasms genetics, Lung Neoplasms metabolism
- Abstract
Background: In non-small cell lung cancer (NSCLC) the efficacy of chemo-immunotherapy is affected by the high expression of drug efflux transporters as ABCC1 and by the low expression of ABCA1, mediating the isopentenyl pyrophosphate (IPP)-dependent anti-tumor activation of Vγ9Vδ2 T-lymphocytes. In endothelial cells ABCA1 is a predicted target of the transcription factor EB (TFEB), but no data exists on the correlation between TFEB and ABC transporters involved in the chemo-immuno-resistance in NSCLC., Methods: The impact of TFEB/ABCC1/ABCA1 expression on NSCLC patients' survival was analyzed in the TCGA-LUAD cohort and in a retrospective cohort of our institution. Human NSCLC cells silenced for TFEB (shTFEB) were analyzed for ABC transporter expression, chemosensitivity and immuno-killing. The chemo-immuno-sensitizing effects of nanoparticles encapsulating zoledronic acid (NZ) on shTFEB tumors and on tumor immune-microenvironment were evaluated in Hu-CD34
+ mice by single-cell RNA-sequencing., Results: TFEBlow ABCA1low ABCC1high and TFEBhigh ABCA1high ABCC1low NSCLC patients had the worst and the best prognosis, respectively, in the TCGA-LUAD cohort and in a retrospective cohort of patients receiving platinum-based chemotherapy or immunotherapy as first-line treatment. By silencing shTFEB in NSCLC cells, we demonstrated that TFEB was a transcriptional inducer of ABCA1 and a repressor of ABCC1. shTFEB cells had also a decreased activity of ERK1/2/SREBP2 axis, implying reduced synthesis and efflux via ABCA1 of cholesterol and its intermediate IPP. Moreover, TFEB silencing reduced cholesterol incorporation in mitochondria: this event increased the efficiency of OXPHOS and the fueling of ABCC1 by mitochondrial ATP. Accordingly, shTFEB cells were less immuno-killed by the Vγ9Vδ2 T-lymphocytes activated by IPP and more resistant to cisplatin. NZ, which increased IPP efflux but not OXPHOS and ATP production, sensitized shTFEB immuno-xenografts, by reducing intratumor proliferation and increasing apoptosis in response to cisplatin, and by increasing the variety of anti-tumor infiltrating cells (Vγ9Vδ2 T-lymphocytes, CD8+ T-lymphocytes, NK cells)., Conclusions: This work suggests that TFEB is a gatekeeper of the sensitivity to chemotherapy and immuno-killing in NSCLC, and that the TFEBlow ABCA1low ABCC1high phenotype can be predictive of poor response to chemotherapy and immunotherapy. By reshaping both cancer metabolism and tumor immune-microenvironment, zoledronic acid can re-sensitize TFEBlow NSCLCs, highly resistant to chemo- and immunotherapy., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
18. Targeting pentamidine towards CD44-overexpressing cells using hyaluronated lipid-polymer hybrid nanoparticles.
- Author
-
Andreana I, Chiapasco M, Bincoletto V, Digiovanni S, Manzoli M, Ricci C, Del Favero E, Riganti C, Arpicco S, and Stella B
- Subjects
- Humans, Drug Carriers chemistry, Cell Line, Tumor, Cell Survival drug effects, Drug Liberation, Lipids chemistry, Drug Delivery Systems, Hyaluronic Acid chemistry, Polylactic Acid-Polyglycolic Acid Copolymer chemistry, Hyaluronan Receptors metabolism, Nanoparticles chemistry, Nanoparticles administration & dosage, Pentamidine chemistry, Pentamidine administration & dosage
- Abstract
Biodegradable nanocarriers possess enormous potential for use as drug delivery systems that can accomplish controlled and targeted drug release, and a wide range of nanosystems have been reported for the treatment and/or diagnosis of various diseases and disorders. Of the various nanocarriers currently available, liposomes and polymer nanoparticles have been extensively studied and some formulations have already reached the market. However, a combination of properties to create a single hybrid system can give these carriers significant advantages, such as improvement in encapsulation efficacy, higher stability, and active targeting towards specific cells or tissues, over lipid or polymer-based platforms. To this aim, this work presents the formulation of poly(lactic-co-glycolic) acid (PLGA) nanoparticles in the presence of a hyaluronic acid (HA)-phospholipid conjugate (HA-DPPE), which was used to anchor HA onto the nanoparticle surface and therefore create an actively targeted hybrid nanosystem. Furthermore, ionic interactions have been proposed for drug encapsulation, leading us to select the free base form of pentamidine (PTM-B) as the model drug. We herein report the preparation of hybrid nanocarriers that were loaded via ion-pairing between the negatively charged PLGA and HA and the positively charged PTM-B, demonstrating an improved loading capacity compared to PLGA-based nanoparticles. The nanocarriers displayed a size of below 150 nm, a negative zeta potential of -35 mV, a core-shell internal arrangement and high encapsulation efficiency (90%). Finally, the ability to be taken up and exert preferential and receptor-mediated cytotoxicity on cancer cells that overexpress the HA specific receptor (CD44) has been evaluated. Competition assays supported the hypothesis that PLGA/HA-DPPE nanoparticles deliver their cargo within cells in a CD44-dependent manner., (© 2024. Controlled Release Society.)
- Published
- 2024
- Full Text
- View/download PDF
19. MiR-155 deficiency and hypoxia results in metabolism switch in the leukemic B-cells.
- Author
-
Golovina E, Heizer T, Daumova L, Bajecny M, Fontana S, Griggio V, Jones R, Coscia M, Riganti C, and Savvulidi Vargova K
- Abstract
Hypoxia represents one of the key factors that stimulates the growth of leukemic cells in their niche. Leukemic cells in hypoxic conditions are forced to reprogram their original transcriptome, miRNome, and metabolome. How the coupling of microRNAs (miRNAs)/mRNAs helps to maintain or progress the leukemic status is still not fully described. MiRNAs regulate practically all biological processes within cells and play a crucial role in the development/progression of leukemia. In the present study, we aimed to uncover the impact of hsa-miR-155-5p (miR-155, MIR155HG) on the metabolism, proliferation, and mRNA/miRNA network of human chronic lymphocytic leukemia cells (CLL) in hypoxic conditions. As a model of CLL, we used the human MEC-1 cell line where we deleted mature miR-155 with CRISPR/Cas9. We determined that miR-155 deficiency in leukemic MEC-1 cells results in lower proliferation even in hypoxic conditions in comparison to MEC-1 control cells. Additionally, in MEC-1 miR-155 deficient cells we observed decreased number of populations of cells in S phase. The miR-155 deficiency under hypoxic conditions was accompanied by an increased apoptosis. We detected a stimulatory effect of miR-155 deficiency and hypoxia at the level of gene expression, seen in significant overexpression of EGLN1, GLUT1, GLUT3 in MEC-1 miR-155 deficient cells. MiR-155 deficiency and hypoxia resulted in increase of glucose and lactate uptake. Pyruvate, ETC and ATP were reduced. To conclude, miR-155 deficiency and hypoxia affects glucose and lactate metabolism by stimulating the expression of glucose transporters as GLUT1, GLUT3, and EGLN1 [Hypoxia-inducible factor prolyl hydroxylase 2 (HIF-PH2)] genes in the MEC-1 cells., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
20. Dysregulation of FLVCR1a-dependent mitochondrial calcium handling in neural progenitors causes congenital hydrocephalus.
- Author
-
Bertino F, Mukherjee D, Bonora M, Bagowski C, Nardelli J, Metani L, Zanin Venturini DI, Chianese D, Santander N, Salaroglio IC, Hentschel A, Quarta E, Genova T, McKinney AA, Allocco AL, Fiorito V, Petrillo S, Ammirata G, De Giorgio F, Dennis E, Allington G, Maier F, Shoukier M, Gloning KP, Munaron L, Mussano F, Salsano E, Pareyson D, di Rocco M, Altruda F, Panagiotakos G, Kahle KT, Gressens P, Riganti C, Pinton PP, Roos A, Arnold T, Tolosano E, and Chiabrando D
- Subjects
- Animals, Humans, Mice, Inositol 1,4,5-Trisphosphate Receptors metabolism, Inositol 1,4,5-Trisphosphate Receptors genetics, Neurogenesis genetics, Calcium metabolism, Hydrocephalus metabolism, Hydrocephalus genetics, Hydrocephalus pathology, Membrane Transport Proteins metabolism, Membrane Transport Proteins genetics, Mitochondria metabolism, Neural Stem Cells metabolism, Neural Stem Cells pathology, Receptors, Virus metabolism, Receptors, Virus genetics
- Abstract
Congenital hydrocephalus (CH), occurring in approximately 1/1,000 live births, represents an important clinical challenge due to the limited knowledge of underlying molecular mechanisms. The discovery of novel CH genes is thus essential to shed light on the intricate processes responsible for ventricular dilatation in CH. Here, we identify FLVCR1 (feline leukemia virus subgroup C receptor 1) as a gene responsible for a severe form of CH in humans and mice. Mechanistically, our data reveal that the full-length isoform encoded by the FLVCR1 gene, FLVCR1a, interacts with the IP3R3-VDAC complex located on mitochondria-associated membranes (MAMs) that controls mitochondrial calcium handling. Loss of Flvcr1a in mouse neural progenitor cells (NPCs) affects mitochondrial calcium levels and energy metabolism, leading to defective cortical neurogenesis and brain ventricle enlargement. These data point to defective NPCs calcium handling and metabolic activity as one of the pathogenetic mechanisms driving CH., Competing Interests: Declaration of interests E.T., V.F., D.Chiabrando, S.P., F.B., and A.L.A. are inventors in a patent filed by the University of Torino, not related to the research reported here., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Dual Inhibitors of P-gp and Carbonic Anhydrase XII (hCA XII) against Tumor Multidrug Resistance with Piperazine Scaffold.
- Author
-
Braconi L, Riganti C, Parenti A, Cecchi M, Nocentini A, Bartolucci G, Menicatti M, Contino M, Colabufo NA, Manetti D, Romanelli MN, Supuran CT, and Teodori E
- Subjects
- Humans, Doxorubicin pharmacology, Doxorubicin chemistry, Piperazine chemistry, Piperazine pharmacology, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents chemical synthesis, HT29 Cells, Structure-Activity Relationship, Cell Line, Tumor, Molecular Structure, A549 Cells, Drug Resistance, Neoplasm drug effects, Drug Resistance, Multiple drug effects, ATP Binding Cassette Transporter, Subfamily B, Member 1 antagonists & inhibitors, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Piperazines pharmacology, Piperazines chemistry, Piperazines chemical synthesis, Carbonic Anhydrase Inhibitors pharmacology, Carbonic Anhydrase Inhibitors chemistry, Carbonic Anhydrase Inhibitors chemical synthesis, Carbonic Anhydrases metabolism
- Abstract
A new series of piperazine derivatives were synthesized and studied with the aim of obtaining dual inhibitors of P-glycoprotein (P-gp) and carbonic anhydrase XII (hCA XII) to synergistically overcome the P-gp-mediated multidrug resistance (MDR) in cancer cells expressing the two proteins, P-gp and hCA XII. Indeed, these hybrid compounds contain both P-gp and hCA XII binding groups on the two nitrogen atoms of the heterocyclic ring. All compounds showed good inhibitory activity on each protein (P-gp and hCA XII) studied individually, and many of them showed a synergistic effect in the resistant HT29/DOX and A549/DOX cell lines which overexpress both the target proteins. In particular, compound 33 displayed the best activity by enhancing the cytotoxicity and intracellular accumulation of doxorubicin in HT29/DOX and A549/DOX cells, thus resulting as promising P-gp-mediated MDR reverser with a synergistic mechanism. Furthermore, compounds 13 , 27 and 32 induced collateral sensitivity (CS) in MDR cells, as they were more cytotoxic in resistant cells than in the sensitive ones; their CS mechanisms were extensively investigated.
- Published
- 2024
- Full Text
- View/download PDF
22. N -Adamantyl-1-alkyl-4-oxo-1,4-dihydroquinoline-3-carboxamide Derivatives as Fluorescent Probes to Detect Microglia Activation through the Imaging of Cannabinoid Receptor Subtype 2 (CB2R).
- Author
-
Intranuovo F, Majellaro M, Mastropasqua F, Delre P, Abatematteo FS, Mangiatordi GF, Stefanachi A, Brea J, Loza MI, Riganti C, Ligresti A, Kumar P, Esposito D, Cristino L, Nicois A, González L, Perrone MG, Colabufo NA, Sotelo E, Abate C, and Contino M
- Subjects
- Humans, Animals, Quinolines chemistry, Quinolines chemical synthesis, Adamantane analogs & derivatives, Adamantane chemistry, Adamantane chemical synthesis, Adamantane pharmacology, Ligands, Structure-Activity Relationship, Receptor, Cannabinoid, CB2 metabolism, Fluorescent Dyes chemistry, Fluorescent Dyes chemical synthesis, Microglia metabolism
- Abstract
Cannabinoid receptor subtype 2 (CB2R) is emerging as a pivotal biomarker to identify the first steps of inflammation-based diseases such as cancer and neurodegeneration. There is an urgent need to find specific probes that may result in green and safe alternatives to the commonly used radiative technologies, to deepen the knowledge of the CB2R pathways impacting the onset of the above-mentioned pathologies. Therefore, based on one of the CB2R pharmacophores, we developed a class of fluorescent N -adamantyl-1-alkyl-4-oxo-1,4-dihydroquinoline-3-carboxamide derivatives spanning from the green to the near-infrared (NIR) regions of the light spectrum. Among the synthesized fluorescent ligands, the green-emitting compound 55 exhibited a favorable binding profile (strong CB2R affinity and high selectivity). Notably, this ligand demonstrated versatility as its use was validated in different experimental settings such as flow cytometry saturation, competitive fluorescence assays, and in vitro microglia cells mimicking inflammation states where CB2R are overexpressed.
- Published
- 2024
- Full Text
- View/download PDF
23. Designing functionalized nanodiamonds with hyaluronic acid-phospholipid conjugates for enhanced cancer cell targeting and fluorescence imaging capabilities.
- Author
-
Sturari S, Andreana I, Aprà P, Bincoletto V, Kopecka J, Mino L, Zurletti B, Stella B, Riganti C, Arpicco S, and Picollo F
- Subjects
- Humans, Cell Line, Tumor, Phospholipids chemistry, Optical Imaging, Neoplasms diagnostic imaging, Neoplasms pathology, Neoplasms metabolism, Nanodiamonds chemistry, Hyaluronic Acid chemistry, Hyaluronan Receptors metabolism
- Abstract
Nanomedicine aims to develop smart approaches for treating cancer and other diseases to improve patient survival and quality of life. Novel nanoparticles as nanodiamonds (NDs) represent promising candidates to overcome current limitations. In this study, NDs were functionalized with a 200 kDa hyaluronic acid-phospholipid conjugate (HA/DMPE), enhancing the stability of the nanoparticles in water-based solutions and selectivity for cancer cells overexpressing specific HA cluster determinant 44 (CD44) receptors. These nanoparticles were characterized by diffuse reflectance Fourier-transform infrared spectroscopy, Raman spectroscopy, and photoluminescence spectroscopy, confirming the efficacy of the functionalization process. Scanning electron microscopy was employed to evaluate the size distribution of the dry particles, while dynamic light scattering and zeta potential measurements were utilized to evaluate ND behavior in a water-based medium. Furthermore, the ND biocompatibility and uptake mediated by CD44 receptors in three different models of human adenocarcinoma cells were assessed by performing cytofluorimetric assay and confocal microscopy. HA-functionalized nanodiamonds demonstrated the advantage of active targeting in the presence of cancer cells expressing CD44 on the surface, suggesting higher drug delivery to tumors over non-tumor tissues. Even CD44-poorly expressing cancers could be targeted by the NDs, thanks to their good passive diffusion within cancer cells.
- Published
- 2024
- Full Text
- View/download PDF
24. Unravelling the metabolic rewiring in the context of doxorubicin-induced cardiotoxicity: Fuel preference changes from fatty acids to glucose oxidation.
- Author
-
Guerra G, Russo M, Priolo R, Riganti C, Reano S, Filigheddu N, Hirsch E, and Ghigo A
- Subjects
- Animals, Class Ib Phosphatidylinositol 3-Kinase metabolism, Glycolysis drug effects, Autophagy drug effects, Male, Signal Transduction drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology, Citric Acid Cycle drug effects, Mice, Inbred C57BL, Heart Diseases chemically induced, Heart Diseases metabolism, Heart Diseases pathology, Heart Diseases prevention & control, Heart Diseases physiopathology, Mitochondria, Heart metabolism, Mitochondria, Heart drug effects, Mitochondria, Heart pathology, Mitochondria, Heart enzymology, Mice, Knockout, Disease Models, Animal, Reactive Oxygen Species metabolism, Glucose Transporter Type 4 metabolism, Antibiotics, Antineoplastic toxicity, Antibiotics, Antineoplastic adverse effects, Doxorubicin toxicity, Oxidation-Reduction, Glucose metabolism, Cardiotoxicity, Fatty Acids metabolism, Energy Metabolism drug effects
- Abstract
Doxorubicin (DOX) is a highly effective chemotherapeutic agent whose clinical use is hindered by the onset of cardiotoxic effects, resulting in reduced ejection fraction within the first year from treatment initiation. Recently it has been demonstrated that DOX accumulates within mitochondria, leading to disruption of metabolic processes and energetic imbalance. We previously described that phosphoinositide 3-kinase γ (PI3Kγ) contributes to DOX-induced cardiotoxicity, causing autophagy inhibition and accumulation of damaged mitochondria. Here we intend to describe the maladaptive metabolic rewiring occurring in DOX-treated hearts and the contribution of PI3Kγ signalling to this process. Metabolomic analysis of DOX-treated WT hearts revealed an accumulation of TCA cycle metabolites due to a cycle slowdown, with reduced levels of pyruvate, unchanged abundance of lactate and increased Acetyl-CoA production. Moreover, the activity of glycolytic enzymes was upregulated, and fatty acid oxidation downregulated, after DOX, indicative of increased glucose oxidation. In agreement, oxygen consumption was increased in after pyruvate supplementation, with the formation of cytotoxic ROS rather than energy production. These metabolic changes were fully prevented in KD hearts. Interestingly, they failed to increase glucose oxidation in response to DOX even with autophagy inhibition, indicating that PI3Kγ likely controls the fuel preference after DOX through an autophagy-independent mechanism. In vitro experiments showed that inhibition of PI3Kγ inhibits pyruvate dehydrogenase (PDH), the key enzyme of Randle cycle regulating the switch from fatty acids to glucose usage, while decreasing DOX-induced mobilization of GLUT-4-carrying vesicles to the plasma membrane and limiting the ensuing glucose uptake. These results demonstrate that PI3Kγ promotes a maladaptive metabolic rewiring in DOX-treated hearts, through a two-pronged mechanism controlling PDH activation and GLUT-4-mediated glucose uptake., (Copyright © 2024.)
- Published
- 2024
- Full Text
- View/download PDF
25. Preclinical efficacy of carfilzomib in BRAF-mutant colorectal cancer models.
- Author
-
Maione F, Oddo D, Galvagno F, Falcomatà C, Pandini M, Macagno M, Pessei V, Barault L, Gigliotti C, Mira A, Corti G, Lamba S, Riganti C, Castella B, Massaia M, Rad R, Saur D, Bardelli A, and Di Nicolantonio F
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Endoplasmic Reticulum Stress drug effects, Endoplasmic Reticulum Stress genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Autophagy drug effects, Mice, Inbred C57BL, Colorectal Neoplasms genetics, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Proto-Oncogene Proteins B-raf genetics, Oligopeptides pharmacology, Oligopeptides therapeutic use, Mutation
- Abstract
Serine/threonine-protein kinase B-raf (BRAF) mutations are found in 8-15% of colorectal cancer patients and identify a subset of tumors with poor outcome in the metastatic setting. We have previously reported that BRAF-mutant human cells display a high rate of protein production, causing proteotoxic stress, and are selectively sensitive to the proteasome inhibitors bortezomib and carfilzomib. In this work, we tested whether carfilzomib could restrain the growth of BRAF-mutant colorectal tumors not only by targeting cancer cells directly, but also by promoting an immune-mediated antitumor response. In human and mouse colorectal cancer cells, carfilzomib triggered robust endoplasmic reticulum stress and autophagy, followed by the emission of immunogenic-damage-associated molecules. Intravenous administration of carfilzomib delayed the growth of BRAF-mutant murine tumors and mobilized the danger-signal proteins calreticulin and high mobility group box 1 (HMGB1). Analyses of drug-treated samples revealed increased intratumor recruitment of activated cytotoxic T cells and natural killers, concomitant with the downregulation of forkhead box protein P3 (Foxp3)
+ T-cell surface glycoprotein CD4 (CD4)+ T cells, indicating that carfilzomib promotes reshaping of the immune microenvironment of BRAF-mutant murine colorectal tumors. These results will inform the design of clinical trials in BRAF-mutant colorectal cancer patients., (© 2024 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2024
- Full Text
- View/download PDF
26. RICTOR/mTORC2 downregulation in BRAF V600E melanoma cells promotes resistance to BRAF/MEK inhibition.
- Author
-
Ponzone L, Audrito V, Landi C, Moiso E, Levra Levron C, Ferrua S, Savino A, Vitale N, Gasparrini M, Avalle L, Vantaggiato L, Shaba E, Tassone B, Saoncella S, Orso F, Viavattene D, Marina E, Fiorilla I, Burrone G, Abili Y, Altruda F, Bini L, Deaglio S, Defilippi P, Menga A, Poli V, Porporato PE, Provero P, Raffaelli N, Riganti C, Taverna D, Cavallo F, and Calautti E
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Down-Regulation, Gene Expression Regulation, Neoplastic, Mutation, Proteomics methods, Xenograft Model Antitumor Assays, MAP Kinase Kinase Kinases antagonists & inhibitors, Drug Resistance, Neoplasm genetics, Mechanistic Target of Rapamycin Complex 2 metabolism, Mechanistic Target of Rapamycin Complex 2 genetics, Melanoma genetics, Melanoma drug therapy, Melanoma metabolism, Melanoma pathology, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Rapamycin-Insensitive Companion of mTOR Protein metabolism, Rapamycin-Insensitive Companion of mTOR Protein genetics
- Abstract
Background: The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM., Methods: After analyzing The Cancer Genome Atlas MM patients' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAF
V600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo., Results: Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi., Conclusions: Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
27. The highly selective and oral phosphoinositide 3-kinase delta (PI3K-δ) inhibitor roginolisib induces apoptosis in mesothelioma cells and increases immune effector cell composition.
- Author
-
Kalla C, Ott G, Finotello F, Niewola-Staszkowska K, Conza GD, Lahn M, van der Veen L, Schüler J, Falkenstern-Ge R, Kopecka J, and Riganti C
- Abstract
Targeting aberrantly expressed kinases in malignant pleural mesothelioma (MPM) is a promising therapeutic strategy. We here investigated the effect of the novel and highly selective Phosphoinositide 3-kinase delta (PI3K-δ) inhibitor roginolisib (IOA-244) on MPM cells and on the immune cells in MPM microenvironment. To this aim, we analyzed the expression of PI3K-δ by immunohistochemistry in specimens from primary MPM, cell viability and death in three different MPM cell lines treated with roginolisib alone and in combination with ipatasertib (AKT inhibitor) and sapanisertib (mTOR inhibitor). In a co-culture model of patient-derived MPM cells, autologous peripheral blood mononuclear cells and fibroblasts, the tumor cell viability and changes in immune cell composition were investigated after treatment of roginolisib with nivolumab and cisplatin. PI3K-δ was detected in 66/89 (74%) MPM tumors and was associated with reduced overall survival (12 vs. 25 months, P=0.0452). Roginolisib induced apoptosis in MPM cells and enhanced the anti-tumor efficacy of AKT and mTOR kinase inhibitors by suppressing PI3K-δ/AKT/mTOR and ERK1/2 signaling. Furthermore, the combination of roginolisib with chemotherapy and immunotherapy re-balanced the immune cell composition, increasing effector T-cells and reducing immune suppressive cells. Overall, roginolisib induces apoptosis in MPM cells and increases the antitumor immune cell effector function when combined with nivolumab and cisplatin. These results provide first insights on the potential of roginolisib as a therapeutic agent in patients with MPM and its potential in combination with established immunotherapy regimen., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Claudia Kalla, German Ott and Roger Falkenstern-Ge received Roginolisib from iOnctura SA and grants from iOnctura SA, Charles River Germany GmbH and Robert Bosch Stiftung (grant project 704 to CK, GO and RG-G); Francesca Finotello received grants from iOnctura SA; Karolina Niewola-Staszkowska, Giusy di Conza, Michael Lahn and Lars van der Veen are employees of iOnctura SA; Michael Lahn and Lars van der Veen are the stock owner of Roginolisib; Julia Schueler received funding from Charles River Germany GmbH; Joanna Kopecka received funding from Fondazione Cassa di Rispoarmio di Torino (grant 2021); Chiara Riganti received Roginolisib from iOnctura SA and grants from from iOnctura SA, and Italian Association for Cancer Research (IG 21480)., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
28. Enhancing pancreatic ductal adenocarcinoma (PDAC) therapy with targeted carbon nano-onion (CNO)-mediated delivery of gemcitabine (GEM)-derived prodrugs.
- Author
-
Bartkowski M, Bincoletto V, Salaroglio IC, Ceccone G, Arenal R, Nervo S, Rolando B, Riganti C, Arpicco S, and Giordani S
- Subjects
- Humans, Gemcitabine, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Onions, Hyaluronic Acid, Cell Line, Tumor, Prodrugs, Carcinoma, Pancreatic Ductal drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Nanotechnology's potential in revolutionising cancer treatments is evident in targeted drug delivery systems (DDSs) engineered to optimise therapeutic efficacy and minimise toxicity. This study examines a novel nanocarrier constructed with carbon nano-onions (CNOs), engineered and evaluated for its ability to selectively target cancer cells overexpressing the hyaluronic acid receptor; CD44. Our results highlighted that the CNO-based nanocarrier coupled with hyaluronic acid as the targeting agent demonstrated effective uptake by CD44+ PANC-1 and MIA PaCa-2 cells, while avoiding CD44- Capan-1 cells. The CNO-based nanocarrier also exhibited excellent biocompatibility in all tested pancreatic ductal adenocarcinoma (PDAC) cells, as well as healthy cells. Notably, the CNO-based nanocarrier was successfully loaded with chemotherapeutic 4-(N)-acyl- sidechain-containing prodrugs derived from gemcitabine (GEM). These prodrugs alone exhibited remarkable efficacy in killing PDAC cells which are known to be GEM resistant, and their efficacy was amplified when combined with the CNO-based nanocarrier, particularly in targeting GEM-resistant CD44+ PDAC cells. These findings demonstrate the potential of CNOs as promising scaffolds in advancing targeted DDSs, signifying the translational potential of carbon nanoparticles for cancer therapy., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
29. Flvcr1a deficiency promotes heme-based energy metabolism dysfunction in skeletal muscle.
- Author
-
Mistretta M, Fiorito V, Allocco AL, Ammirata G, Hsu MY, Digiovanni S, Belicchi M, Napoli L, Ripolone M, Trombetta E, Mauri P, Farini A, Meregalli M, Villa C, Porporato PE, Miniscalco B, Crich SG, Riganti C, Torrente Y, and Tolosano E
- Subjects
- Mice, Animals, Heme metabolism, Mice, Knockout, Muscle, Skeletal metabolism, Energy Metabolism, Cell Differentiation physiology, Membrane Transport Proteins metabolism, Satellite Cells, Skeletal Muscle metabolism
- Abstract
The definition of cell metabolic profile is essential to ensure skeletal muscle fiber heterogeneity and to achieve a proper equilibrium between the self-renewal and commitment of satellite stem cells. Heme sustains several biological functions, including processes profoundly implicated with cell metabolism. The skeletal muscle is a significant heme-producing body compartment, but the consequences of impaired heme homeostasis on this tissue have been poorly investigated. Here, we generate a skeletal-muscle-specific feline leukemia virus subgroup C receptor 1a (FLVCR1a) knockout mouse model and show that, by sustaining heme synthesis, FLVCR1a contributes to determine the energy phenotype in skeletal muscle cells and to modulate satellite cell differentiation and muscle regeneration., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
30. FLVCR1a Controls Cellular Cholesterol Levels through the Regulation of Heme Biosynthesis and Tricarboxylic Acid Cycle Flux in Endothelial Cells.
- Author
-
Manco M, Ammirata G, Petrillo S, De Giorgio F, Fontana S, Riganti C, Provero P, Fagoonee S, Altruda F, and Tolosano E
- Subjects
- Mice, Animals, Membrane Transport Proteins metabolism, Cell Membrane metabolism, Mice, Knockout, Heme metabolism, Endothelial Cells metabolism, Citric Acid Cycle
- Abstract
Feline leukemia virus C receptor 1a (FLVCR1a), initially identified as a retroviral receptor and localized on the plasma membrane, has emerged as a crucial regulator of heme homeostasis. Functioning as a positive regulator of δ-aminolevulinic acid synthase 1 (ALAS1), the rate-limiting enzyme in the heme biosynthetic pathway, FLVCR1a influences TCA cycle cataplerosis, thus impacting TCA flux and interconnected metabolic pathways. This study reveals an unexplored link between FLVCR1a, heme synthesis, and cholesterol production in endothelial cells. Using cellular models with manipulated FLVCR1a expression and inducible endothelial-specific Flvcr1a -null mice, we demonstrate that FLVCR1a-mediated control of heme synthesis regulates citrate availability for cholesterol synthesis, thereby influencing cellular cholesterol levels. Moreover, alterations in FLVCR1a expression affect membrane cholesterol content and fluidity, supporting a role for FLVCR1a in the intricate regulation of processes crucial for vascular development and endothelial function. Our results underscore FLVCR1a as a positive regulator of heme synthesis, emphasizing its integration with metabolic pathways involved in cellular energy metabolism. Furthermore, this study suggests that the dysregulation of heme metabolism may have implications for modulating lipid metabolism. We discuss these findings in the context of FLVCR1a's potential heme-independent function as a choline importer, introducing additional complexity to the interplay between heme and lipid metabolism.
- Published
- 2024
- Full Text
- View/download PDF
31. p140Cap modulates the mevalonate pathway decreasing cell migration and enhancing drug sensitivity in breast cancer cells.
- Author
-
Centonze G, Natalini D, Grasso S, Morellato A, Salemme V, Piccolantonio A, D'Attanasio G, Savino A, Bianciotto OT, Fragomeni M, Scavuzzo A, Poncina M, Nigrelli F, De Gregorio M, Poli V, Arina P, Taverna D, Kopecka J, Dupont S, Turco E, Riganti C, and Defilippi P
- Subjects
- Humans, Female, Mevalonic Acid metabolism, Cholesterol metabolism, Cell Movement, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Hydroxymethylglutaryl-CoA Reductase Inhibitors therapeutic use
- Abstract
p140Cap is an adaptor protein involved in assembling multi-protein complexes regulating several cellular processes. p140Cap acts as a tumor suppressor in breast cancer (BC) and neuroblastoma patients, where its expression correlates with a better prognosis. The role of p140Cap in tumor metabolism remains largely unknown. Here we study the role of p140Cap in the modulation of the mevalonate (MVA) pathway in BC cells. The MVA pathway is responsible for the biosynthesis of cholesterol and non-sterol isoprenoids and is often deregulated in cancer. We found that both in vitro and in vivo, p140Cap cells and tumors show an increased flux through the MVA pathway by positively regulating the pace-maker enzyme of the MVA pathway, the 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), via transcriptional and post-translational mechanisms. The higher cholesterol synthesis is paralleled with enhanced cholesterol efflux. Moreover, p140Cap promotes increased cholesterol localization in the plasma membrane and reduces lipid rafts-associated Rac1 signalling, impairing cell membrane fluidity and cell migration in a cholesterol-dependent manner. Finally, p140Cap BC cells exhibit decreased cell viability upon treatments with statins, alone or in combination with chemotherapeutic at low concentrations in a synergistic manner. Overall, our data highlight a new perspective point on tumor suppression in BC by establishing a previously uncharacterized role of the MVA pathway in p140Cap expressing tumors, thus paving the way to the use of p140Cap as a potent biomarker to stratify patients for better tuning therapeutic options., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
32. Mitochondria Transplantation Mitigates Damage in an In Vitro Model of Renal Tubular Injury and in an Ex Vivo Model of DCD Renal Transplantation.
- Author
-
Rossi A, Asthana A, Riganti C, Sedrakyan S, Byers LN, Robertson J, Senger RS, Montali F, Grange C, Dalmasso A, Porporato PE, Palles C, Thornton ME, Da Sacco S, Perin L, Ahn B, McCully J, Orlando G, and Bussolati B
- Subjects
- Humans, Swine, Animals, Kidney metabolism, Mitochondria metabolism, Mitochondria pathology, Kidney Transplantation, Reperfusion Injury, Acute Kidney Injury prevention & control, Acute Kidney Injury metabolism
- Abstract
Objectives: To test whether mitochondrial transplantation (MITO) mitigates damage in 2 models of acute kidney injury (AKI)., Background: MITO is a process where exogenous isolated mitochondria are taken up by cells. As virtually any morbid clinical condition is characterized by mitochondrial distress, MITO may find a role as a treatment modality in numerous clinical scenarios including AKI., Methods: For the in vitro experiments, human proximal tubular cells were damaged and then treated with mitochondria or placebo. For the ex vivo experiments, we developed a non-survival ex vivo porcine model mimicking the donation after cardiac death renal transplantation scenario. One kidney was treated with mitochondria, although the mate organ received placebo, before being perfused at room temperature for 24 hours. Perfusate samples were collected at different time points and analyzed with Raman spectroscopy. Biopsies taken at baseline and 24 hours were analyzed with standard pathology, immunohistochemistry, and RNA sequencing analysis., Results: In vitro, cells treated with MITO showed higher proliferative capacity and adenosine 5'-triphosphate production, preservation of physiological polarization of the organelles and lower toxicity and reactive oxygen species production. Ex vivo, kidneys treated with MITO shed fewer molecular species, indicating stability. In these kidneys, pathology showed less damage whereas RNAseq analysis showed modulation of genes and pathways most consistent with mitochondrial biogenesis and energy metabolism and downregulation of genes involved in neutrophil recruitment, including IL1A, CXCL8, and PIK3R1., Conclusions: MITO mitigates AKI both in vitro and ex vivo., Competing Interests: J.R. and R.S.S. are co-founders of Rametrix Technologies, on the board of which J.R. serves as President and CEO, whereas R.S.S. as Chief Technology Officer. The remaining authors report no conflicts of interest., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
33. Cytotoxic pathways activated by multifunctional thiosemicarbazones targeting sigma-2 receptors in breast and lung carcinoma cells.
- Author
-
Kopecka J, Barbanente A, Vitone D, Arnesano F, Margiotta N, Berchialla P, Niso M, Riganti C, and Abate C
- Subjects
- Humans, Apoptosis, Lung metabolism, Cell Line, Tumor, Receptors, sigma metabolism, Thiosemicarbazones pharmacology, Antineoplastic Agents therapeutic use, Lung Neoplasms drug therapy, Carcinoma
- Abstract
Background: Multifunctional thiosemicarbazones (TSCs) able to bind sigma receptors and chelate metals are considered as a promising avenue for the treatment of pancreatic cancer due to the encouraging results obtained on in vitro and in vivo models. Here, we assessed the biochemical mechanism of these TSCs also on lung (A549) and breast (MCF7) cancer cells., Methods: The density of sigma-2 receptors in normal (BEAS-2B and MCF10A) and in lung and breast (A549 and MCF7) cancer cells was evaluated by flow cytometry. In these cells, cytotoxicity (MTT assay) and activation of ER- and mitochondria-dependent cell death pathways (by spectrofluorimetric assays to measure Caspases 3/7/9; qRT-PCR detection of GRP78, ATF6, IRE1, PERK; MitoSOX, DCFDA-AM and JC-1 staining), induced by the TSCs FA4, MLP44, PS3 and ACThio1, were evaluated., Results: FA4 and PS3 exerted more potent cytotoxicity than MLP44 and ACThio1 in all cancer cell lines, where the density of sigma-2 receptors was higher than in normal cells. Remarkably, FA4 promoted ER- and mitochondria-dependent cell death pathways in both cell models, whereas the other TSCs had variable, cell-dependent effects on the activation of the two proapoptotic pathways., Conclusions: Our data suggest that FA4 is a promising compound that deserves to be further studied for lung and breast cancer treatment. However, the other multifunctional TSCs also hold promise for the development of therapies towards a personalized medicine approach. Indeed, the presence of the sigma-2 receptor-targeting moiety would lead to a more specific tumor delivery embracing the characteristics of individual tumor types., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
34. Identification of organizational barriers to HPV vaccination uptake in medical students in southern Italy: a cross-sectional study.
- Author
-
Sorrentino M, Mercogliano M, Esposito F, Lamberti AM, Buonocore G, Riganti C, Triassi M, and Palladino R
- Subjects
- Female, Humans, Male, Cross-Sectional Studies, Health Knowledge, Attitudes, Practice, Vaccination, Italy, Students, Medical, Papillomavirus Infections complications
- Abstract
Introduction: Despite proven efficacy, HPV vaccination coverage is still suboptimal. Factors influencing vaccination uptake are education attainment, socio-economic position, and knowledge about HPV. This study aimed to assess HPV vaccination uptake and its correlates among medical students and identify logistic-organizational barriers, knowledge, and attitudes with regard towards HPV vaccination to improve current public health vaccination strategies. Medical students, with their acquired biological knowledge, were selected as a low-risk groups for HPV vaccination uptake. This cross-sectional study was conducted using a validated questionnaire., Methods: Students in their the first 3 years of study students were preferentially invited. Eventually, the invitation was extended to every medical student. Logistic multivariable regression was used to assess determinants of HPV vaccination uptake. Additional analysis explored determinants of knowledge of and attitude toward HPV vaccination. Finally, a sensitive analysis was conducted to further assess the effect of knowledge and attitude on the HPV vaccination rate., Results: A total of 882 medical students participated, with 74.5% enrolled in the first 3 years of their training. HPV vaccination uptake was 55.5%, ranging from 78.5% for females to 16.5% for males. Male sex and increasing age were consistently associated with a lower vaccination uptake (males sex: OR 0.03, CI 0.02-0.05; age: OR 0.77, CI 0.68-0.88), whereasilst progress in their academic career was associated with a to higher likelihood of being vaccinated (6th year: OR 3.45, CI 1.24-9.57). These associations were confirmed when considering the knowledge of and attitude towards HPV. Additionally also, an active outreach from healthcare institutions was associated with a higher likelihood of receiving HPV vaccination (OR 1.70, CI 1.09-2.65., Conclusion: HPV vaccination in medical students was higher than in the general population; however, it was still suboptimal. An active and up-to-date call strategy and extending the free-of-charge offer are essential measures for to improvinge vaccination uptake. The findings support the need to improve public health strategies and increase awareness and knowledge ofregarding HPV vaccination., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Sorrentino, Mercogliano, Esposito, Lamberti, Buonocore, Riganti, Triassi and Palladino.)
- Published
- 2023
- Full Text
- View/download PDF
35. 4-(3-Phenyl-4-(3,4,5-trimethoxybenzoyl)-1 H -pyrrol-1-yl)benzenesulfonamide, a Novel Carbonic Anhydrase and Wnt/β-Catenin Signaling Pathway Dual-Targeting Inhibitor with Potent Activity against Multidrug Resistant Cancer Cells.
- Author
-
Masci D, Puxeddu M, Di Magno L, D'Ambrosio M, Parisi A, Nalli M, Bai R, Coluccia A, Sciò P, Orlando V, D'Angelo S, Biagioni S, Urbani A, Hamel E, Nocentini A, Filiberti S, Turati M, Ronca R, Kopecka J, Riganti C, Fionda C, Bordone R, Della Rocca G, Canettieri G, Supuran CT, Silvestri R, and La Regina G
- Subjects
- Humans, Structure-Activity Relationship, Drug Resistance, Multiple, Wnt Signaling Pathway, Drug Resistance, Neoplasm, Carbonic Anhydrase Inhibitors pharmacology, Carbonic Anhydrase IX, Molecular Structure, Benzenesulfonamides, Carbonic Anhydrases metabolism, Neoplasms
- Abstract
We synthesized new pyrrole and indole derivatives as human carbonic anhydrase (hCA) inhibitors with the potential to inhibit the Wnt/β-catenin signaling pathway. The presence of both N 1-(4-sulfonamidophenyl) and 3-(3,4,5-trimethoxyphenyl) substituents was essential for strong hCA inhibitors. The most potent hCA XII inhibitor 15 ( K
i = 6.8 nM) suppressed the Wnt/β-catenin signaling pathway and its target genes MYC, Fgf20, and Sall4 and exhibited the typical markers of apoptosis, cleaved poly(ADP-ribose)polymerase, and cleaved caspase-3. Compound 15 showed strong inhibition of viability in a panel of cancer cells, including colorectal cancer and triple-negative breast cancer cells, was effective against the NCI/ADR-RES DOX-resistant cell line, and restored the sensitivity to doxorubicin (DOX) in HT29/DX and MDCK/P-gp cells. Compound 15 is a novel dual-targeting compound with activity against hCA and Wnt/β-catenin. It thus has a broad targeting spectrum and is an anticancer agent with specific potential in P-glycoprotein overexpressing cell lines.- Published
- 2023
- Full Text
- View/download PDF
36. Tetrazole and oxadiazole derivatives as bioisosteres of tariquidar and elacridar: New potent P-gp modulators acting as MDR reversers.
- Author
-
Braconi L, Dei S, Contino M, Riganti C, Bartolucci G, Manetti D, Romanelli MN, Perrone MG, Colabufo NA, Guglielmo S, and Teodori E
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2, Drug Resistance, Neoplasm, Structure-Activity Relationship, Neoplasm Proteins, Drug Resistance, Multiple, Tetrazoles pharmacology, Amides pharmacology, Antineoplastic Agents pharmacology
- Abstract
New 2,5- and 1,5-disubstituted tetrazoles, and 2,5-disubstituted-1,3,4-oxadiazoles were synthesized as tariquidar and elacridar derivatives and studied as multidrug resistance (MDR) reversers. Their behaviour on the three ABC transporters P-gp, MRP1 and BCRP was investigated. All compounds inhibited the P-gp transport activity in MDCK-MDR1 cells overexpressing P-gp, showing EC
50 values even in the low nanomolar range (compounds 15, 22). Oxadiazole derivatives were able to increase the antiproliferative effect of doxorubicin in MDCK-MDR1 and in HT29/DX cells confirming their nature of P-gp modulators, with derivative 15 being the most potent in these assays. Compound 15 also displayed a dual inhibitory effect showing good activities towards both P-gp and BCRP. A computational study suggested a common interaction pattern on P-gp for most of the potent compounds. The bioisosteric substitution of the amide group of lead compounds allowed identifying a new set of potent oxadiazole derivatives that modulate MDR through inhibition of the P-gp efflux activity. If compared to previous amide derivatives, the introduction of the heterocycle rings greatly enhances the activity on P-gp, introduces in two compounds a moderate inhibitory activity on MRP1 and maintains in some cases the effect on BCRP, leading to the unveiling of dual inhibitor 15., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
37. Fighting Multidrug Resistance with Ruthenium-Cyclopentadienyl Compounds: Unveiling the Mechanism of P-gp Inhibition.
- Author
-
Teixeira RG, Salaroglio IC, Oliveira NFB, Sequeira JGN, Fontrodona X, Romero I, Machuqueiro M, Tomaz AI, Garcia MH, Riganti C, and Valente A
- Subjects
- Humans, Cisplatin pharmacology, Molecular Docking Simulation, Ruthenium Compounds pharmacology, Cell Line, Tumor, Drug Resistance, Multiple, Drug Resistance, Neoplasm, Antineoplastic Agents pharmacology, Ruthenium pharmacology, Carcinoma, Non-Small-Cell Lung, Lung Neoplasms
- Abstract
The search for more effective and selective drugs to overcome cancer multidrug resistance is urgent. As such, a new series of ruthenium-cyclopentadienyl ("RuCp") compounds with the general formula [Ru(η
5 -C5 H4 R)(4,4'-R'-2,2'-bipy)(PPh3 )] were prepared and fully characterized. All compounds were evaluated toward non-small cell lung cancer cells with different degrees of cisplatin sensitivity (A549, NCI-H2228, Calu-3, and NCI-H1975), showing better cytotoxicity than the first-line chemotherapeutic drug cisplatin. Compounds 2 and 3 (R' = -OCH3 ; R = CHO ( 2 ) or CH2 OH ( 3 )) further inhibited the activity of P-gp and MRP1 efflux pumps by impairing their catalytic activity. Molecular docking calculations identified the R-site P-gp pocket as the preferred one, which was further validated using site-directed mutagenesis experiments in P-gp. Altogether, our results unveil the first direct evidence of the interaction between P-gp and "RuCp" compounds in the modulation of P-gp activity and establish them as valuable candidates to circumvent cancer MDR.- Published
- 2023
- Full Text
- View/download PDF
38. Anti-tumor activity of selinexor in combination with antineoplastic agents in chronic lymphocytic leukemia.
- Author
-
Vitale C, Griggio V, Todaro M, Riganti C, Jones R, Boccellato E, Perutelli F, Arruga F, Vaisitti T, Efremov DG, Deaglio S, Landesman Y, Bruno B, and Coscia M
- Subjects
- Animals, Mice, Cell Line, Tumor, Hydrazines pharmacology, Hydrazines therapeutic use, Drug Combinations, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Despite recent relevant therapeutic progresses, chronic lymphocytic leukemia (CLL) remains an incurable disease. Selinexor, an oral inhibitor of the nuclear export protein XPO1, is active as single agent in different hematologic malignancies, including CLL. The purpose of this study was to evaluate the anti-tumor effects of selinexor, used in combination with chemotherapy drugs (i.e. fludarabine and bendamustine) or with the PI3Kδ inhibitor idelalisib in CLL. Our results showed a significant decrease in CLL cell viability after treatment with selinexor-containing drug combinations compared to each single compound, with demonstration of synergistic cytotoxic effects. Interestingly, this drug synergism was exerted also in the presence of the protective effect of stromal cells. From the molecular standpoint, the synergistic cytotoxic activity of selinexor plus idelalisib was associated with increased regulatory effects of this drug combination on the tumor suppressors FOXO3A and IkBα compared to each single compound. Finally, selinexor was also effective in potentiating the in vivo anti-tumor effects of the PI3Kδ inhibitor in mice treated with the drug combination compared to single agents. Our data provide preclinical evidence of the synergism and potential efficacy of a combination treatment targeting XPO1 and PI3Kδ in CLL., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
39. Autocrine 17-β-Estradiol/Estrogen Receptor-α Loop Determines the Response to Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer.
- Author
-
Anobile DP, Salaroglio IC, Tabbò F, La Vecchia S, Akman M, Napoli F, Bungaro M, Benso F, Aldieri E, Bironzo P, Kopecka J, Passiglia F, Righi L, Novello S, Scagliotti GV, and Riganti C
- Subjects
- Humans, Female, Male, Animals, Mice, Receptors, Estrogen metabolism, Immune Checkpoint Inhibitors therapeutic use, Estrogen Receptor alpha genetics, B7-H1 Antigen antagonists & inhibitors, Estradiol pharmacology, Estradiol therapeutic use, Estrogens, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Antineoplastic Agents, Immunological therapeutic use
- Abstract
Purpose: The response to immune checkpoint inhibitors (ICI) often differs between genders in non-small cell lung cancer (NSCLC), but metanalyses results are controversial, and no clear mechanisms are defined. We aim at clarifying the molecular circuitries explaining the differential gender-related response to anti-PD-1/anti-PD-L1 agents in NSCLC., Experimental Design: We prospectively analyzed a cohort of patients with NSCLC treated with ICI as a first-line approach, and we identified the molecular mechanisms determining the differential efficacy of ICI in 29 NSCLC cell lines of both genders, recapitulating patients' phenotype. We validated new immunotherapy strategies in mice bearing NSCLC patient-derived xenografts and human reconstituted immune system ("immune-PDXs")., Results: In patients, we found that estrogen receptor α (ERα) was a predictive factor of response to pembrolizumab, stronger than gender and PD-L1 levels, and was directly correlated with PD-L1 expression, particularly in female patients. ERα transcriptionally upregulated CD274/PD-L1 gene, more in females than in males. This axis was activated by 17-β-estradiol, autocrinely produced by intratumor aromatase, and by the EGFR-downstream effectors Akt and ERK1/2 that activated ERα. The efficacy of pembrolizumab in immune-PDXs was significantly improved by the aromatase inhibitor letrozole, which reduced PD-L1 and increased the percentage of antitumor CD8+T-lymphocytes, NK cells, and Vγ9Vδ2 T-lymphocytes, producing durable control and even tumor regression after continuous administration, with maximal benefit in 17-β-estradiol/ERα highfemale immune-xenografts., Conclusions: Our work unveils that 17-β-estradiol/ERα status predicts the response to pembrolizumab in patients with NSCLC. Second, we propose aromatase inhibitors as new gender-tailored immune-adjuvants in NSCLC. See related commentary by Valencia et al., p. 3832., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
40. The role of Extracellular Vesicles in glycolytic and lipid metabolic reprogramming of cancer cells: Consequences for drug resistance.
- Author
-
Polónia B, Xavier CPR, Kopecka J, Riganti C, and Vasconcelos MH
- Subjects
- Humans, Cell Communication, Drug Resistance, Neoplasm, Lipids therapeutic use, Extracellular Vesicles pathology, Neoplasms metabolism
- Abstract
In order to adapt to a higher proliferative rate and an increased demand for energy sources, cancer cells rewire their metabolic pathways, a process currently recognized as a hallmark of cancer. Even though the metabolism of glucose is perhaps the most discussed metabolic shift in cancer, lipid metabolic alterations have been recently recognized as relevant players in the growth and proliferation of cancer cells. Importantly, some of these metabolic alterations are reported to induce a drug resistant phenotype in cancer cells. The acquisition of drug resistance traits severely hinders cancer treatment, being currently considered one of the major challenges of the oncological field. Evidence suggests that Extracellular Vesicles (EVs), which play a crucial role in intercellular communication, may act as facilitators of tumour progression, survival and drug resistance by modulating several aspects involved in the metabolism of cancer cells. This review aims to gather and discuss relevant data regarding metabolic reprograming in cancer, particularly involving the glycolytic and lipid alterations, focusing on its influence on drug resistance and highlighting the relevance of EVs as intercellular mediators of this process., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
41. APE-1/Ref-1 Inhibition Blocks Malignant Pleural Mesothelioma Cell Proliferation and Migration: Crosstalk between Oxidative Stress and Epithelial Mesenchymal Transition (EMT) in Driving Carcinogenesis and Metastasis.
- Author
-
Ramundo V, Zanirato G, Palazzo ML, Riganti C, and Aldieri E
- Subjects
- Animals, Epithelial-Mesenchymal Transition, Oxidative Stress, Cell Proliferation, Carcinogenesis, Hyperplasia, Endonucleases, Mesothelioma, Malignant, Hominidae
- Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer associated with asbestos exposure. MPM pathogenesis has been related both to oxidative stress, evoked by and in response to asbestos fibers exposure, and epithelial mesenchymal transition (EMT), an event induced by oxidative stress itself and related to cancer proliferation and metastasis. Asbestos-related primary oxidative damage is counteracted in the lungs by various redox-sensitive factors, often hyperactivated in some cancers. Among these redox-sensitive factors, Apurinic-apyrimidinic endonuclease 1 (APE-1)/Redox effector factor 1 (Ref-1) has been demonstrated to be overexpressed in MPM and lung cancer, but the molecular mechanism has not yet been fully understood. Moreover, asbestos exposure has been associated with induced EMT events, via some EMT transcription factors, such as Twist, Zeb-1 and Snail-1, in possible crosstalk with oxidative stress and inflammation events. To demonstrate this hypothesis, we inhibited/silenced Ref-1 in MPM cells; as a consequence, both EMT (Twist, Zeb-1 and Snail-1) markers and cellular migration/proliferation were significantly inhibited. Taken as a whole, these results show, for the first time, crosstalk between oxidative stress and EMT in MPM carcinogenesis and invasiveness, thus improving the knowledge to better address a preventive and therapeutic approach against this aggressive cancer.
- Published
- 2023
- Full Text
- View/download PDF
42. Use of Enzymatically Activated Carbon Monoxide Donors for Sensitizing Drug-Resistant Tumor Cells.
- Author
-
Sodano F, Rolando B, Lazzarato L, Costamagna C, Failla M, Riganti C, and Chegaev K
- Subjects
- Humans, Charcoal, Mitochondria metabolism, Apoptosis, Signal Transduction, Carbon Monoxide pharmacology, Carbon Monoxide chemistry, Organometallic Compounds pharmacology, Organometallic Compounds chemistry
- Abstract
The application of gaseous signaling molecules like NO, H
2 S or CO to overcome the multidrug resistance in cancer treatment has proven to be a viable therapeutic strategy. The development of CO-releasing molecules (CORMs) in a controlled manner and in targeted tissues remains a challenge in medicinal chemistry. In this paper, we describe the design, synthesis and chemical and enzymatic stability of a novel non-metal CORM ( 1 ) able to release intracellularly CO and, simultaneously, facilitate fluorescent degradation of products under the action of esterase. The toxicity of 1 against different human cancer cell lines and their drug-resistant counterparts, as well as the putative mechanism of toxicity were investigated. The drug-resistant cancer cell lines efficiently absorbed 1 and 1 was able to restore their sensitivity vs. chemotherapeutic drugs by causing a CO-dependent mitochondrial oxidative stress that culminated in mitochondrial-dependent apoptosis. These results demonstrate the importance of CORMs in cases where conventional chemotherapy fails and thus open the horizons towards new combinatorial strategies to overcome multidrug resistance.- Published
- 2023
- Full Text
- View/download PDF
43. Long-Chain Acyl Coenzyme A Dehydrogenase, a Key Player in Metabolic Rewiring/Invasiveness in Experimental Tumors and Human Mesothelioma Cell Lines.
- Author
-
Pouliquen DL, Ortone G, Rumiano L, Boissard A, Henry C, Blandin S, Guette C, Riganti C, and Kopecka J
- Abstract
Cross-species investigations of cancer invasiveness are a new approach that has already identified new biomarkers which are potentially useful for improving tumor diagnosis and prognosis in clinical medicine and veterinary science. In this study, we combined proteomic analysis of four experimental rat malignant mesothelioma (MM) tumors with analysis of ten patient-derived cell lines to identify common features associated with mitochondrial proteome rewiring. A comparison of significant abundance changes between invasive and non-invasive rat tumors gave a list of 433 proteins, including 26 proteins reported to be exclusively located in mitochondria. Next, we analyzed the differential expression of genes encoding the mitochondrial proteins of interest in five primary epithelioid and five primary sarcomatoid human MM cell lines; the most impressive increase was observed in the expression of the long-chain acyl coenzyme A dehydrogenase (ACADL). To evaluate the role of this enzyme in migration/invasiveness, two epithelioid and two sarcomatoid human MM cell lines derived from patients with the highest and lowest overall survival were studied. Interestingly, sarcomatoid vs. epithelioid cell lines were characterized by higher migration and fatty oxidation rates, in agreement with ACADL findings. These results suggest that evaluating mitochondrial proteins in MM specimens might identify tumors with higher invasiveness. Data are available via ProteomeXchange with the dataset identifier PXD042942.
- Published
- 2023
- Full Text
- View/download PDF
44. Development of Fluorescent 4-[4-(3 H -Spiro[isobenzofuran-1,4'-piperidin]-1'-yl)butyl]indolyl Derivatives as High-Affinity Probes to Enable the Study of σ Receptors via Fluorescence-Based Techniques.
- Author
-
Abatematteo FS, Majellaro M, Montsch B, Prieto-Díaz R, Niso M, Contino M, Stefanachi A, Riganti C, Mangiatordi GF, Delre P, Heffeter P, Sotelo E, and Abate C
- Subjects
- Ligands, Fluorescence, Coloring Agents, Receptors, sigma
- Abstract
Sigma (σ) receptor subtypes, σ
1 and σ2 , are targets of wide pharmaceutical interest. The σ2 receptor holds promise for the development of diagnostics and therapeutics against cancer and Alzheimer's disease. Nevertheless, little is known about the mechanisms activated by the σ2 receptor. To contribute to the exploitation of its therapeutic potential, we developed novel specific fluorescent ligands. Indole derivatives bearing the N- butyl-3 H -spiro[isobenzofuran-1,4'-piperidine] portion were functionalized with fluorescent tags. Nanomolar-affinity fluorescent σ ligands, spanning from green to red to near-infrared emission, were obtained. Compounds 19 (σ pan affinity) and 29 (σ2 selective), which displayed the best compromise between pharmacodynamic and photophysical properties, were investigated in flow cytometry, confocal, and live cell microscopy, demonstrating their specificity for the σ2 receptor. To the best of our knowledge, these are the first red-emitting fluorescent σ2 ligands, validated as powerful tools for the study of σ2 receptors via fluorescence-based techniques.- Published
- 2023
- Full Text
- View/download PDF
45. Editorial: Novel therapeutic approaches to target drug resistant tumors.
- Author
-
Riganti C, Kowalski K, and Kopecka J
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
46. Corrigendum to 'Glabratephrin reverses doxorubicin resistance in triple negative breast cancer by inhibiting P-glycoprotein' [Pharmacol. Res. 175 (2022) 105975].
- Author
-
Abd-Ellatef GEF, Gazzano E, El-Desoky AH, Hamed AR, Kopecka J, Belisario DC, Costamagna C, Marie MAS, Fahmy SR, Abdel-Hamid AZ, and Riganti C
- Published
- 2023
- Full Text
- View/download PDF
47. The multiple combination of Paclitaxel, Ramucirumab and Elacridar reverses the paclitaxel-mediated resistance in gastric cancer cell lines.
- Author
-
Schirizzi A, Contino M, Carrieri L, Riganti C, De Leonardis G, Scavo MP, Perrone MG, Miciaccia M, Kopecka J, Refolo MG, Lotesoriere C, Depalo N, Rizzi F, Giannelli G, Messa C, and D'Alessandro R
- Abstract
Introduction: Paclitaxel (PTX) interferes with microtubule architecture by binding to β-tubulin, thereby blocking progression at the G2/M phase and inducing apoptosis. This study aimed to investigate molecular processes underlying PTX-mediated resistance in gastric cancer (GC) cells., Methods: PTX-mediated resistance involves many processes, and in this work some of the factors involved in the resistance mechanism were identified by comparing two GC lines with PTX induced resistance to their sensitive counterparts., Results: Thus, the key feature of PTX-resistant cells was the overexpression of pro-angiogenic factors such as VEGFA, VEGFC, and Ang2, known to support tumor cell growth. A second relevant change detected in PTX-resistant lines was the elevated level of TUBβIII, a tubulin isoform that opposes microtubule stabilization. A third identified factor contributing to PTX-resistance was P-glycoprotein (P-gp), a transporter responsible for chemotherapy efflux from the cells, highly expressed in PTX-resistant lines., Discussion: These findings were in line with a greater sensitivity of resistant cells to treatment with both Ramucirumab and Elacridar. Ramucirumab significantly reduced the expression of angiogenic molecules and TUBβIII, while Elacridar restored the access of chemotherapy, recovering its anti-mitotic and pro-apoptotic effects. Finally, this study highlighted the role played by exosomes in spreading factors responsible for resistance in the tumor microenvironment., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Schirizzi, Contino, Carrieri, Riganti, De Leonardis, Scavo, Perrone, Miciaccia, Kopecka, Refolo, Lotesoriere, Depalo, Rizzi, Giannelli, Messa and D’Alessandro.)
- Published
- 2023
- Full Text
- View/download PDF
48. N-adamantyl-anthranil amide derivatives: New selective ligands for the cannabinoid receptor subtype 2 (CB2R).
- Author
-
Graziano G, Delre P, Carofiglio F, Brea J, Ligresti A, Kostrzewa M, Riganti C, Gioè-Gallo C, Majellaro M, Nicolotti O, Colabufo NA, Abate C, Loza MI, Sotelo E, Mangiatordi GF, Contino M, Stefanachi A, and Leonetti F
- Subjects
- Humans, Molecular Docking Simulation, Endocannabinoids, Anti-Inflammatory Agents, Receptors, Cannabinoid, Receptor, Cannabinoid, CB2, Ligands, Amides, Cannabinoids pharmacology
- Abstract
Cannabinoid type 2 receptor (CB2R) is a G-protein-coupled receptor that, together with Cannabinoid type 1 receptor (CB1R), endogenous cannabinoids and enzymes responsible for their synthesis and degradation, forms the EndoCannabinoid System (ECS). In the last decade, several studies have shown that CB2R is overexpressed in activated central nervous system (CNS) microglia cells, in disorders based on an inflammatory state, such as neurodegenerative diseases, neuropathic pain, and cancer. For this reason, the anti-inflammatory and immune-modulatory potentials of CB2R ligands are emerging as a novel therapeutic approach. The design of selective ligands is however hampered by the high sequence homology of transmembrane domains of CB1R and CB2R. Based on a recent three-arm pharmacophore hypothesis and latest CB2R crystal structures, we designed, synthesized, and evaluated a series of new N-adamantyl-anthranil amide derivatives as CB2R selective ligands. Interestingly, this new class of compounds displayed a high affinity for human CB2R along with an excellent selectivity respect to CB1R. In this respect, compounds exhibiting the best pharmacodynamic profile in terms of CB2R affinity were also evaluated for the functional behavior and molecular docking simulations provided a sound rationale by highlighting the relevance of the arm 1 substitution to prompt CB2R action. Moreover, the modulation of the pro- and anti-inflammatory cytokines production was also investigated to exert the ability of the best compounds to modulate the inflammatory cascade., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Mabel Loza and Pepo Brea reports financial support was provided by Agencia Estatal de Investigación. Mabel Loza and Pepo Brea reports financial support was provided by Xunta de Galicia Consellería de Sanidade. Eddy Sotelo reports financial support was provided by Galician Government., (Copyright © 2023 Elsevier Masson SAS. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
49. A harmonized protocol for an international multicenter prospective study of nanotechnology workers: the NanoExplore cohort.
- Author
-
Guseva Canu I, Plys E, Velarde Crézé C, Fito C, Hopf NB, Progiou A, Riganti C, Sauvain JJ, Squillacioti G, Suarez G, Wild P, and Bergamaschi E
- Subjects
- Humans, Prospective Studies, Nanotechnology, Biological Monitoring, Multicenter Studies as Topic, Occupational Exposure analysis, Nanostructures toxicity
- Abstract
Nanotechnology applications are fast-growing in many industrial fields. Consequently, health effects of engineered nanomaterials (ENMs) should be investigated. Within the EU-Life project NanoExplore, we developed a harmonized protocol of an international multicenter prospective cohort study of workers in ENM-producing companies. This article describes the development of the protocol, sample size calculation, data collection and management procedures and discusses its relevance with respect to research needs. Within this protocol, workers' ENM exposure will be assessed over four consecutive working days during the initial recruitment campaign and the subsequent follow-up campaigns. Biomonitoring using noninvasive sampling of exhaled breath condensate (EBC), exhaled air, and urine will be collected before and after 4-day exposure monitoring. Both exposure and effect biomarkers, will be quantified along with pulmonary function tests and diagnosed diseases reported using a standardized epidemiological questionnaire available in four languages. Until now, this protocol was implemented at seven companies in Switzerland, Spain and Italy. The protocol is well standardized, though sufficiently flexible to include company-specific conditions and occupational hygiene measures. The recruitment, to date, of 140 participants and collection of all data and samples, enabled us launching the first international cohort of nanotechnology workers. All companies dealing with ENMs could join the NanoExplore Consortium, apply this harmonized protocol and enter in the cohort, concieved as an open cohort. Its protocol meets all requirements of a hypotheses-driven prospective study, which will assess and reassess effects of ENM exposure on workers' health by updating the follow-up of the cohort. New hypothesis could be also considered.
- Published
- 2023
- Full Text
- View/download PDF
50. Novel Lymphocyte-Independent Antitumor Activity by PD-1 Blocking Antibody against PD-1+ Chemoresistant Lung Cancer Cells.
- Author
-
Rotolo R, Leuci V, Donini C, Galvagno F, Massa A, De Santis MC, Peirone S, Medico G, Sanlorenzo M, Vujic I, Gammaitoni L, Basiricò M, Righi L, Riganti C, Salaroglio IC, Napoli F, Tabbò F, Mariniello A, Vigna E, Modica C, D'Ambrosio L, Grignani G, Taulli R, Hirsch E, Cereda M, Aglietta M, Scagliotti GV, Novello S, Bironzo P, and Sangiolo D
- Subjects
- Humans, Mice, Animals, Cisplatin pharmacology, Cisplatin therapeutic use, Neoplasm Recurrence, Local, Lymphocytes metabolism, Cell Line, Tumor, Lung Neoplasms pathology, Carcinoma, Non-Small-Cell Lung pathology
- Abstract
Purpose: Antibodies against the lymphocyte PD-1 (aPD-1) receptor are cornerstone agents for advanced non-small cell lung cancer (NSCLC), based on their ability to restore the exhausted antitumor immune response. Our study reports a novel, lymphocyte-independent, therapeutic activity of aPD-1 against NSCLC, blocking the tumor-intrinsic PD-1 receptors on chemoresistant cells., Experimental Design: PD-1 in NSCLC cells was explored in vitro at baseline, including stem-like pneumospheres, and following treatment with cisplatin both at transcriptional and protein levels. PD-1 signaling and RNA sequencing were assessed. The lymphocyte-independent antitumor activity of aPD-1 was explored in vitro, by PD-1 blockade and stimulation with soluble ligand (PD-L1s), and in vivo within NSCLC xenograft models., Results: We showed the existence of PD-1+ NSCLC cell subsets in cell lines and large in silico datasets (Cancer Cell Line Encyclopedia and The Cancer Genome Atlas). Cisplatin significantly increased PD-1 expression on chemo-surviving NSCLC cells (2.5-fold P = 0.0014), while the sequential treatment with anti-PD-1 Ab impaired their recovery after chemotherapy. PD-1 was found to be associated with tumor stemness features. PD-1 expression was enhanced in NSCLC stem-like pneumospheres (P < 0.0001), significantly promoted by stimulation with soluble PD-L1 (+27% ± 4, P < 0.0001) and inhibited by PD-1 blockade (-30% ± 3, P < 0.0001). The intravenous monotherapy with anti-PD-1 significantly inhibited tumor growth of NSCLC xenografts in immunodeficient mice, without the contribution of the immune system, and delayed the occurrence of chemoresistance when combined with cisplatin., Conclusions: We report first evidence of a novel lymphocyte-independent activity of anti-PD-1 antibodies in NSCLC, capable of inhibiting chemo-surviving NSCLC cells and exploitable to contrast disease relapses following chemotherapy. See related commentary by Augustin et al., p. 505., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.