31 results on '"Alessio Biagioni"'
Search Results
2. Editorial: CRISPR advancement in cancer research and future perspectives
- Author
-
Alessio Biagioni and Reza Mohammadinejad
- Subjects
cancer therapeutics ,gene editing ,gene therapy ,gene delivery ,CRISPR ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. Gastric Cancer Vascularization and the Contribution of Reactive Oxygen Species
- Author
-
Alessio Biagioni, Sara Peri, Giampaolo Versienti, Claudia Fiorillo, Matteo Becatti, Lucia Magnelli, and Laura Papucci
- Subjects
gastric cancer ,angiogenesis ,vasculogenic mimicry ,epithelial-to-endothelial transition ,hypoxia ,reactive oxygen species ,Microbiology ,QR1-502 - Abstract
Blood vessels are the most important way for cancer cells to survive and diffuse in the body, metastasizing distant organs. During the process of tumor expansion, the neoplastic mass progressively induces modifications in the microenvironment due to its uncontrolled growth, generating a hypoxic and low pH milieu with high fluid pressure and low nutrients concentration. In such a particular condition, reactive oxygen species play a fundamental role, enhancing tumor proliferation and migration, inducing a glycolytic phenotype and promoting angiogenesis. Indeed, to reach new sources of oxygen and metabolites, highly aggressive cancer cells might produce a new abnormal network of vessels independently from endothelial cells, a process called vasculogenic mimicry. Even though many molecular markers and mechanisms, especially in gastric cancer, are still unclear, the formation of such intricate, leaky and abnormal vessel networks is closely associated with patients’ poor prognosis, and therefore finding new pharmaceutical solutions to be applied along with canonical chemotherapies in order to control and normalize the formation of such networks is urgent.
- Published
- 2023
- Full Text
- View/download PDF
4. Extracellular Lactic Acidosis of the Tumor Microenvironment Drives Adipocyte-to-Myofibroblast Transition Fueling the Generation of Cancer-Associated Fibroblasts
- Author
-
Elena Andreucci, Bianca Saveria Fioretto, Irene Rosa, Marco Matucci-Cerinic, Alessio Biagioni, Eloisa Romano, Lido Calorini, and Mirko Manetti
- Subjects
adipocytes ,adipose-derived stem cells ,cell differentiation ,extracellular acidosis ,lactate ,myofibroblasts ,Cytology ,QH573-671 - Abstract
Lactic acidosis characterizes the tumor microenvironment (TME) and is involved in the mechanisms leading to cancer progression and dissemination through the reprogramming of tumor and local host cells (e.g., endothelial cells, fibroblasts, and immune cells). Adipose tissue also represents a crucial component of the TME which is receiving increasing attention due to its pro-tumoral activity, however, to date, it is not known whether it could be affected by the acidic TME. Now, emerging evidence from chronic inflammatory and fibrotic diseases underlines that adipocytes may give rise to pathogenic myofibroblast-like cells through the adipocyte-to-myofibroblast transition (AMT). Thus, our study aimed to investigate whether extracellular acidosis could affect the AMT process, sustaining the acquisition by adipocytes of a cancer-associated fibroblast (CAF)-like phenotype with a pro-tumoral activity. To this purpose, human subcutaneous adipose-derived stem cells committed to adipocytes (acADSCs) were cultured under basal (pH 7.4) or lactic acidic (pH 6.7, 10 mM lactate) conditions, and AMT was evaluated with quantitative PCR, immunoblotting, and immunofluorescence analyses. We observed that lactic acidosis significantly impaired the expression of adipocytic markers while inducing myofibroblastic, pro-fibrotic, and pro-inflammatory phenotypes in acADSCs, which are characteristic of AMT reprogramming. Interestingly, the conditioned medium of lactic acidosis-exposed acADSC cultures was able to induce myofibroblastic activation in normal fibroblasts and sustain the proliferation, migration, invasion, and therapy resistance of breast cancer cells in vitro. This study reveals a previously unrecognized relationship between lactic acidosis and the generation of a new CAF-like cell subpopulation from adipocytic precursor cells sustaining tumor malignancy.
- Published
- 2023
- Full Text
- View/download PDF
5. Editorial: Redox Potential and Metabolic Behavior in Gastrointestinal Cancers
- Author
-
Alessio Biagioni, Sara Peri, Nicola Schiavone, Elisa Giommoni, and Laura Papucci
- Subjects
gastric cancer ,colorectal cancer ,gastrointestinal cancers ,metabolism ,redox potential ,chronic hepatitis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2022
- Full Text
- View/download PDF
6. Extracellular Acidosis Differentially Regulates Estrogen Receptor β-Dependent EMT Reprogramming in Female and Male Melanoma Cells
- Author
-
Silvia Peppicelli, Jessica Ruzzolini, Matteo Lulli, Alessio Biagioni, Francesca Bianchini, Adele Caldarella, Chiara Nediani, Elena Andreucci, and Lido Calorini
- Subjects
extracellular acidity ,estrogen receptor β ,epithelial-to-mesenchymal transition ,NF-κB ,human melanoma ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Clinical outcomes of melanoma patients pointed out a gender disparity that supports a correlation between sex hormone activity on estrogen receptors (ER) and melanoma development and progression. Here, we found that the epithelial-to-mesenchymal transition (EMT) of melanoma cells induced by extracellular acidosis, which is a crucial hallmark of solid cancers, correlates with the expression of ERβ, the most representative ER on melanoma cells. Extracellular acidosis induces an enhanced expression of ERβ in female cells and EMT markers remain unchanged, while extracellular acidosis did not induce the expression of ERβ in male cells and EMT was strongly promoted. An inverse relationship between ERβ expression and EMT markers in melanoma cells of different sex exposed to extracellular acidosis was revealed by two different technical approaches: florescence-activated cell sorting of high ERβ expressing cell subpopulations and ERβ receptor silencing. Finally, we found that ERβ regulates EMT through NF-κB activation. These results demonstrate that extracellular acidosis drives a differential ERβ regulation in male and female melanoma cells and that this gender disparity might open new perspectives for personalized therapeutic approaches.
- Published
- 2022
- Full Text
- View/download PDF
7. An Oleocanthal-Enriched EVO Oil Extract Induces the ROS Production in Gastric Cancer Cells and Potentiates the Effect of Chemotherapy
- Author
-
Sara Peri, Jessica Ruzzolini, Silvia Urciuoli, Giampaolo Versienti, Alessio Biagioni, Elena Andreucci, Silvia Peppicelli, Francesca Bianchini, Andrea Bottari, Lido Calorini, Chiara Nediani, Lucia Magnelli, and Laura Papucci
- Subjects
gastric cancer ,chemoresistance ,nutraceuticals ,oleocanthal ,complementary therapy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Oleocanthal, a minor polar compound in extra-virgin olive (EVO) oil, contains anticancer properties, which should be encouraged in its use in oncology. Gastric Cancer (GC), a very aggressive human cancer, is often diagnosed at advanced stages, when surgery is substituted or supported by chemotherapy (CT). However, CT frequently fails due to the patient’s resistance to the treatment. Thus, the aim of this study is to verify whether an OC-enriched EVO oil extract fraction (OCF) may be useful in order to overcome a resistance to GC. We evaluated the OCF effects on an AGS gastric adenocarcinoma cell line wild type (AGS wt) and on its subpopulations resistant to 5-fluorouracil (5FUr), Paclitaxel (TAXr) or cisplatin (CISr). We found that a 60 µM dose of the OCF acts on the AGS wt, 5FUr and TAXr, leading to the cell cycle inhibition and to a ROS production, but not on CISr cells. Resistance of CISr to the OCF seems to be due to higher levels of antioxidant-enzymes that can counteract the OCF-induced ROS production. Moreover, using the OCF plus 5-fluorouracil, Paclitaxel or cisplatin, we found a potentiating effect compared with a mono-treatment in all resistant GC cells, including CISr. In conclusion, the use of the OCF in the management of GC has shown very interesting advantages, opening-up the possibility to evaluate the efficacy of the OCF in vivo, as a valid adjuvant in the treatment of resistant GC.
- Published
- 2022
- Full Text
- View/download PDF
8. EGFR/uPAR interaction as druggable target to overcome vemurafenib acquired resistance in melanoma cellsResearch in context
- Author
-
Anna Laurenzana, Francesca Margheri, Alessio Biagioni, Anastasia Chillà, Nicola Pimpinelli, Jessica Ruzzolini, Silvia Peppicelli, Elena Andreucci, Lido Calorini, Simona Serratì, Mario Del Rosso, and Gabriella Fibbi
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: BRAF inhibitor (BRAF-I) therapy for melanoma patients harboring the V600E mutation is initially highly effective, but almost all patients relapse within a few months. Understanding the molecular mechanisms behind BRAF-I responsiveness and acquired resistance is therefore an important issue. Here we assessed the role of urokinase type plasminogen activator receptor (uPAR) as a potentially valuable biomarker in the acquisition of BRAF-I resistance in V600E mutant melanoma cells. Methods: We examined uPAR and EGFR levels by real time PCR and western blot analysis. uPAR loss of function was realized by knocking down uPAR by RNAi or using M25, a peptide that uncouples uPAR-integrin interaction. We investigated uPAR-β1integrin-EGFR association by co-immunoprecipitation and confocal immuno-fluorescence analysis. Acquired resistance to BRAF-I was generated by chronic exposure of cells to vemurafenib. Findings: We proved that uPAR knockdown in combination with vemurafenib inhibits melanoma cell proliferation to greater extent than either treatment alone causing a decrease in AKT and ERK1/2 phosphorylation. Conversely, we demonstrated that uPAR enforced over-expression results in reduced sensitivity to BRAF inhibition. Moreover, by targeting uPAR and EGFR interaction with an integrin antagonist peptide we restored vemurafenib responsiveness in melanoma resistant cells. Furthermore, we found significant detectable uPAR and EGFR levels in tumor biopsies of 4 relapsed patients. Interpretation: We disclosed an unpredicted mechanism of reduced sensitiveness to BRAF inhibition, driven by elevated levels of uPAR and identified a potential therapeutic strategy to overcome acquired resistance. Funds: Associazione Italiana Ricerca sul Cancro (AIRC); Ente Cassa di Risparmio di Firenze. Keywords: Vemurafenib, Acquired resistance, Melanoma, uPAR, EGFR
- Published
- 2019
- Full Text
- View/download PDF
9. Delivery systems of CRISPR/Cas9-based cancer gene therapy
- Author
-
Alessio Biagioni, Anna Laurenzana, Francesca Margheri, Anastasia Chillà, Gabriella Fibbi, and Mario Del Rosso
- Subjects
CRISPR ,Delivery systems ,Gene-editing ,Cancer ,Gene therapy ,Biology (General) ,QH301-705.5 - Abstract
Abstract CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats) is today one of the most reliable method for gene-editing, supporting previous gene therapies technologies such as TALEN, Meganucleases and ZFNs. There is a growing up number of manuscripts reporting several successful gene-edited cancer cell lines, but the real challenge is to translate this technique to the clinical practice. While treatments for diseases based on a single gene mutation is closer, being possible to target and repair the mutant allele in a selective way generating specific guide RNAs (gRNAs), many steps need to be done to apply CRISPR to face cancer. In this review, we want to give a general overview to the recent advancements in the delivery systems of the CRISPR/Cas9 machinery in cancer therapy.
- Published
- 2018
- Full Text
- View/download PDF
10. CRISPR/Cas9 uPAR Gene Knockout Results in Tumor Growth Inhibition, EGFR Downregulation and Induction of Stemness Markers in Melanoma and Colon Carcinoma Cell Lines
- Author
-
Alessio Biagioni, Anastasia Chillà, Mario Del Rosso, Gabriella Fibbi, Francesca Scavone, Elena Andreucci, Silvia Peppicelli, Francesca Bianchini, Lido Calorini, Anna Li Santi, Pia Ragno, Francesca Margheri, and Anna Laurenzana
- Subjects
urokinase-type plasminogen activator receptor ,CRISPR ,miR146a ,melanoma ,colon cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
uPAR is a globular protein, tethered to the cell membrane by a GPI-anchor involved in several cancer-related properties and its overexpression commonly correlates with poor prognosis and metastasis. We investigated the consequences of uPAR irreversible loss in human melanoma and colon cancer cell lines, knocking out its expression by CRISPR/Cas9. We analyzed through flow cytometry, western blotting and qPCR, the modulation of the most known cancer stem cells-associated genes and the EGFR while we observed the proliferation rate exploiting 2D and 3D cellular models. We also generated uPAR “rescue” expression cell lines as well as we promoted the expression of only its 3’UTR to demonstrate the involvement of uPAR mRNA in tumor progression. Knocking out PLAUR, uPAR-encoding gene, we observed an inhibited growth ratio unexpectedly coupled with a significant percentage of cells acquiring a stem-like phenotype. In vivo experiments demonstrated that uPAR loss completely abrogates tumorigenesis despite the gained stem-like profile. Nonetheless, we proved that the reintroduction of the 3’UTR of PLAUR gene was sufficient to restore the wild-type status validating the hypothesis that such a region may act as a “molecular sponge”. In particular miR146a, by binding PLAUR 3’ UTR region might be responsible for uPAR-dependent inhibition of EGFR expression.
- Published
- 2021
- Full Text
- View/download PDF
11. Enhanced Antitumoral Activity and Photoacoustic Imaging Properties of AuNP‐Enriched Endothelial Colony Forming Cells on Melanoma
- Author
-
Paolo Armanetti, Anastasia Chillà, Francesca Margheri, Alessio Biagioni, Luca Menichetti, Giancarlo Margheri, Fulvio Ratto, Sonia Centi, Francesca Bianchini, Mirko Severi, Rita Traversi, Daniele Bani, Matteo Lulli, Tommaso Del Rosso, Alessandra Mocali, Elisabetta Rovida, Mario Del Rosso, Gabriella Fibbi, and Anna Laurenzana
- Subjects
endothelial colony forming cells ,gold nanoparticles ,melanoma ,nanomedicine ,photoacoustic imaging ,Science - Abstract
Abstract Near infrared (NIR)‐resonant gold nanoparticles (AuNPs) hold great promise in cancer diagnostics and treatment. However, translating the theranostic potential of AuNPs into clinical applications still remains a challenge due to the difficulty to improve the efficiency and specificity of tumor delivery in vivo as well as the clearance from liver and spleen to avoid off target toxicity. In this study, endothelial colony forming cells (ECFCs) are exploited as vehicles to deliver AuNPs to tumors. It is first demonstrated that ECFCs display a great capability to intake AuNPs without losing viability, and exert antitumor activity per se. Using a human melanoma xenograft mouse model, it is next demonstrated that AuNP‐loaded ECFCs retain their capacity to migrate to tumor sites in vivo 1 day after injection and stay in the tumor mass for more than 1 week. In addition, it is demonstrated that ECFC‐loaded AuNPs are efficiently cleared by the liver over time and do not elicit any sign of damage to healthy tissue.
- Published
- 2021
- Full Text
- View/download PDF
12. SOX2 as a novel contributor of oxidative metabolism in melanoma cells
- Author
-
Elena Andreucci, Silvia Pietrobono, Silvia Peppicelli, Jessica Ruzzolini, Francesca Bianchini, Alessio Biagioni, Barbara Stecca, and Lido Calorini
- Subjects
Melanoma ,Tumor extracellular acidosis ,SOX2 ,HIF1α ,Oxidative metabolism ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Deregulated metabolism is a hallmark of cancer and recent evidence underlines that targeting tumor energetics may improve therapy response and patient outcome. Despite the general attitude of cancer cells to exploit the glycolytic pathway even in the presence of oxygen (aerobic glycolysis or “Warburg effect”), tumor metabolism is extremely plastic, and such ability to switch from glycolysis to oxidative phosphorylation (OxPhos) allows cancer cells to survive under hostile microenvironments. Recently, OxPhos has been related with malignant progression, chemo-resistance and metastasis. OxPhos is induced under extracellular acidosis, a well-known characteristic of most solid tumors, included melanoma. Methods To evaluate whether SOX2 modulation is correlated with metabolic changes under standard or acidic conditions, SOX2 was silenced and overexpressed in several melanoma cell lines. To demonstrate that SOX2 directly represses HIF1A expression we used chromatin immunoprecipitation (ChIP) and luciferase assay. Results In A375-M6 melanoma cells, extracellular acidosis increases SOX2 expression, that sustains the oxidative cancer metabolism exploited under acidic conditions. By studying non-acidic SSM2c and 501-Mel melanoma cells (high- and very low-SOX2 expressing cells, respectively), we confirmed the metabolic role of SOX2, attributing SOX2-driven OxPhos reprogramming to HIF1α pathway disruption. Conclusions SOX2 contributes to the acquisition of an aggressive oxidative tumor phenotype, endowed with enhanced drug resistance and metastatic ability.
- Published
- 2018
- Full Text
- View/download PDF
13. Mature and progenitor endothelial cells perform angiogenesis also under protease inhibition: the amoeboid angiogenesis
- Author
-
Anastasia Chillà, Francesca Margheri, Alessio Biagioni, Mario Del Rosso, Gabriella Fibbi, and Anna Laurenzana
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Controlling vascular growth is a challenging aim for the inhibition of tumor growth and metastasis. The amoeboid and mesenchymal types of invasiveness are two modes of migration interchangeable in cancer cells: the Rac-dependent mesenchymal migration requires the activity of proteases; the Rho-ROCK-dependent amoeboid motility is protease-independent and has never been described in endothelial cells. Methods A cocktail of physiologic inhibitors (Ph-C) of serine-proteases, metallo-proteases and cysteine-proteases, mimicking the physiological environment that cells encounter during their migration within the angiogenesis sites was used to induce amoeboid style migration of Endothelial colony forming cells (ECFCs) and mature endothelial cells (ECs). To evaluate the mesenchymal-ameboid transition RhoA and Rac1 activation assays were performed along with immunofluorescence analysis of proteins involved in cytoskeleton organization. Cell invasion was studied in Boyden chambers and Matrigel plug assay for the in vivo angiogenesis. Results In the present study we showed in both ECFCs and ECs, a decrease of activated Rac1 and an increase of activated RhoA upon shifting of cells to the amoeboid conditions. In presence of Ph-C inhibitors both cell lines acquired a round morphology and Matrigel invasion was greatly enhanced with respect to that observed in the absence of protease inhibition. We also observed that the urokinase-plasminogen-activator (uPAR) receptor silencing and uPAR-integrin uncoupling with the M25 peptide abolished both mesenchymal and amoeboid angiogenesis of ECFCs and ECs in vitro and in vivo, indicating a role of the uPAR-integrin-actin axis in the regulation of amoeboid angiogenesis. Furthermore, under amoeboid conditions endothelial cells seem to be indifferent to VEGF stimulation, which induces an amoeboid signaling pattern also in mesenchymal conditions. Conclusion Here we first provide a data set disclosing that endothelial cells can move and differentiate into vascular structures in vitro and in vivo also in the absence of proteases activity, performing a new type of neovascularization: the “amoeboid angiogenesis”. uPAR is indispensable for ECs and ECFCs to perform an efficient amoeboid angiogenesis. Therefore, uPAR silencing or the block of its integrin-interaction, together with standard treatment against VEGF, could be a possible solution for angiogenesis inhibition.
- Published
- 2018
- Full Text
- View/download PDF
14. Editing SOX Genes by CRISPR-Cas: Current Insights and Future Perspectives
- Author
-
Ali Dehshahri, Alessio Biagioni, Hadi Bayat, E. Hui Clarissa Lee, Mohammad Hashemabadi, Hojjat Samareh Fekri, Ali Zarrabi, Reza Mohammadinejad, and Alan Prem Kumar
- Subjects
CRISPR ,SOX transcription factors ,gene editing ,cancer ,stem cells ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and its associated proteins (Cas) is an adaptive immune system in archaea and most bacteria. By repurposing these systems for use in eukaryote cells, a substantial revolution has arisen in the genome engineering field. In recent years, CRISPR-Cas technology was rapidly developed and different types of DNA or RNA sequence editors, gene activator or repressor, and epigenome modulators established. The versatility and feasibility of CRISPR-Cas technology has introduced this system as the most suitable tool for discovering and studying the mechanism of specific genes and also for generating appropriate cell and animal models. SOX genes play crucial roles in development processes and stemness. To elucidate the exact roles of SOX factors and their partners in tissue hemostasis and cell regeneration, generating appropriate in vitro and in vivo models is crucial. In line with these premises, CRISPR-Cas technology is a promising tool for studying different family members of SOX transcription factors. In this review, we aim to highlight the importance of CRISPR-Cas and summarize the applications of this novel, promising technology in studying and decoding the function of different members of the SOX gene family.
- Published
- 2021
- Full Text
- View/download PDF
15. Enhanced Vasculogenic Capacity Induced by 5-Fluorouracil Chemoresistance in a Gastric Cancer Cell Line
- Author
-
Sara Peri, Alessio Biagioni, Giampaolo Versienti, Elena Andreucci, Fabio Staderini, Giuseppe Barbato, Lisa Giovannelli, Francesco Coratti, Nicola Schiavone, Fabio Cianchi, Laura Papucci, and Lucia Magnelli
- Subjects
gastric cancer ,chemoresistance ,tumor angiogenesis ,vasculogenic mimicry ,epithelial-to-endothelial transition ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Chemotherapy is still widely used as a coadjutant in gastric cancer when surgery is not possible or in presence of metastasis. During tumor evolution, gatekeeper mutations provide a selective growth advantage to a subpopulation of cancer cells that become resistant to chemotherapy. When this phenomenon happens, patients experience tumor recurrence and treatment failure. Even if many chemoresistance mechanisms are known, such as expression of ATP-binding cassette (ABC) transporters, aldehyde dehydrogenase (ALDH1) activity and activation of peculiar intracellular signaling pathways, a common and universal marker for chemoresistant cancer cells has not been identified yet. In this study we subjected the gastric cancer cell line AGS to chronic exposure of 5-fluorouracil, cisplatin or paclitaxel, thus selecting cell subpopulations showing resistance to the different drugs. Such cells showed biological changes; among them, we observed that the acquired chemoresistance to 5-fluorouracil induced an endothelial-like phenotype and increased the capacity to form vessel-like structures. We identified the upregulation of thymidine phosphorylase (TYMP), which is one of the most commonly reported mutated genes leading to 5-fluorouracil resistance, as the cause of such enhanced vasculogenic ability.
- Published
- 2021
- Full Text
- View/download PDF
16. 5-Fluorouracil Conversion Pathway Mutations in Gastric Cancer
- Author
-
Alessio Biagioni, Fabio Staderini, Sara Peri, Giampaolo Versienti, Nicola Schiavone, Fabio Cianchi, Laura Papucci, and Lucia Magnelli
- Subjects
5 fluorouracil ,chemoresistance ,gastric cancer ,mutations ,Biology (General) ,QH301-705.5 - Abstract
To date, 5-Fluorouracil (5FU) is a major component of several chemotherapy regimens, thus its study is of fundamental importance to better understand all the causes that may lead to chemoresistance and treatment failure. Given the evident differences between prognosis in Asian and Caucasian populations, triggered by clear genetic discordances and given the extreme genetic heterogeneity of gastric cancer (GC), the evaluation of the most frequent mutations in every single member of the 5FU conversion and activation pathway might reveal several important results. Here, we exploited the cBioPortal analysis software to query a large databank of clinical and wide-genome studies to evaluate the components of the three major 5FU transformation pathways. We demonstrated that mutations in such ways were associated with a poor prognosis and reduced overall survival, often caused by a deletion in the TYMP gene and amplification in TYMS. The use of prodrugs and dihydropyrimidine dehydrogenase (DPD) inhibitors, which normally catabolizes 5FU into inactive metabolites, improved such chemotherapies, but several steps forward still need to be taken to select better therapies to target the chemoresistant pools of cells with high anaplastic features and genomic instability.
- Published
- 2020
- Full Text
- View/download PDF
17. MAP Kinases Pathways in Gastric Cancer
- Author
-
Lucia Magnelli, Nicola Schiavone, Fabio Staderini, Alessio Biagioni, and Laura Papucci
- Subjects
MAPK ,gastric cancer ,metastasis ,miRNA ,lncRNA ,epigenome ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Gastric cancer (GC) is turning out today to be one of the most important welfare issues for both Asian and European countries. Indeed, while the vast majority of the disease burden is located in China and in Pacific and East Asia, GC in European countries still account for about 100,000 deaths per year. With this review article, we aim to focus the attention on one of the most complex cellular pathways involved in GC proliferation, invasion, migration, and metastasis: the MAP kinases. Such large kinases family is to date constantly studied, since their discovery more than 30 years ago, due to the important role that it plays in the regulation of physiological and pathological processes. Interactions with other cellular proteins as well as miRNAs and lncRNAs may modulate their expression influencing the cellular biological features. Here, we summarize the most important and recent studies involving MAPK in GC. At the same time, we need to underly that, differently from cancers arising from other tissues, where MAPK pathways seems to be a gold target for anticancer therapies, GC seems to be unique in any aspect. Our aim is to review the current knowledge in MAPK pathways alterations leading to GC, including H. pylori MAPK-triggering to derail from gastric normal epithelium to GC and to encourage researches involved in MAPK signal transduction, that seems to definitely sustain GC development.
- Published
- 2020
- Full Text
- View/download PDF
18. uPAR Knockout Results in a Deep Glycolytic and OXPHOS Reprogramming in Melanoma and Colon Carcinoma Cell Lines
- Author
-
Alessio Biagioni, Anna Laurenzana, Anastasia Chillà, Mario Del Rosso, Elena Andreucci, Martina Poteti, Daniele Bani, Daniele Guasti, Gabriella Fibbi, and Francesca Margheri
- Subjects
colon cancer ,crispr ,gene-editing ,melanoma ,upar ,Cytology ,QH573-671 - Abstract
Urokinase Plasminogen Activator (uPA) Receptor (uPAR) is a well-known GPI-anchored three-domain membrane protein with pro-tumor roles largely shown in all the malignant tumors where it is over-expressed. Here we have exploited the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 gene knock out approach to investigate its role in the oxidative metabolism in human melanoma and colon cancer as the consequences of its irreversible loss. Knocking out PLAUR, a uPAR-encoding gene, in A375p, A375M6 and HCT116, which are two human melanoma and a colon carcinoma, respectively, we have observed an increased number of mitochondria in the two melanoma cell lines, while we evidenced an immature biogenesis of mitochondria in the colon carcinoma culture. Such biological diversity is, however, reflected in a significant enhancement of the mitochondrial spare respiratory capacity, fueled by an increased expression of GLS2, and in a decreased glycolysis paired with an increased secretion of lactate by all uPAR KO cells. We speculated that this discrepancy might be explained by an impaired ratio between LDHA and LDHB.
- Published
- 2020
- Full Text
- View/download PDF
19. Publisher Correction to: delivery systems of CRISPR/Cas9-based cancer gene therapy
- Author
-
Alessio Biagioni, Anna Laurenzana, Francesca Margheri, Anastasia Chillà, Gabriella Fibbi, and Mario Del Rosso
- Subjects
Biology (General) ,QH301-705.5 - Abstract
In the original version of this article [1], published on 18 December 2018, there was 1 incorrect author name.
- Published
- 2019
- Full Text
- View/download PDF
20. Identification of Novel Human Breast Carcinoma (MDA-MB-231) Cell Growth Modulators from a Carbohydrate-Based Diversity Oriented Synthesis Library
- Author
-
Elena Lenci, Riccardo Innocenti, Alessio Biagioni, Gloria Menchi, Francesca Bianchini, and Andrea Trabocchi
- Subjects
heterocycles ,sugars ,chemical diversity ,cancer ,Organic chemistry ,QD241-441 - Abstract
The application of a cell-based growth inhibition on a library of skeletally different glycomimetics allowed for the selection of a hexahydro-2H-furo[3,2-b][1,4]oxazine compound as candidate inhibitors of MDA-MB-231 cell growth. Subsequent synthesis of analogue compounds and preliminary biological studies validated the selection of a valuable hit compound with a novel polyhydroxylated structure for the modulation of the breast carcinoma cell cycle mechanism.
- Published
- 2016
- Full Text
- View/download PDF
21. uPAR Controls Vasculogenic Mimicry Ability Expressed by Drug-Resistant Melanoma Cells
- Author
-
Silvia Peppicelli, Mario Del Rosso, Francesca Bianchini, Lido Calorini, Elena Andreucci, Chiara Nediani, Jessica Ruzzolini, Gabriella Fibbi, Francesca Margheri, Simona Serratì, Livia Fucci, Anna Laurenzana, Michele Guida, and Alessio Biagioni
- Subjects
Cancer Research ,Article ,Receptors, Urokinase Plasminogen Activator ,Cell Line, Tumor ,medicine ,Humans ,Vasculogenic mimicry ,Drug resistance ,Extracellular acidosis ,Melanoma ,uPAR ,Vemurafenib ,Tube formation ,business.industry ,General Medicine ,EPH receptor A2 ,medicine.disease ,Urokinase plasminogen activator receptor (uPAR) ,Urokinase receptor ,Oncology ,Pharmaceutical Preparations ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,Skin cancer ,business ,medicine.drug - Abstract
Malignant melanoma is a highly aggressive skin cancer characterized by an elevated grade of tumor cell plasticity. Such plasticity allows adaptation of melanoma cells to different hostile conditions and guarantees tumor survival and disease progression, including aggressive features such as drug resistance. Indeed, almost 50% of melanoma rapidly develop resistance to the BRAFV600Einhibitor vemurafenib, with fast tumor dissemination, a devastating consequence for patients outcomes. Vasculogenic mimicry (VM), the ability of cancer cells to organize themselves in perfused vascular-like channels, might sustain tumor spread by providing vemurafenib-resistant cancer cells with supplementary ways to enter into circulation and disseminate. Thus, this research aims to determine if vemurafenib resistance goes with the acquisition of VM ability by aggressive melanoma cells, and identify a driving molecule for both vemurafenib resistance and VM. We used two independent experimental models of drug-resistant melanoma cells, the first one represented by a chronic adaptation of melanoma cells to extracellular acidosis, known to drive a particularly aggressive and vemurafenib-resistant phenotype, the second one generated with chronic vemurafenib exposure. By performing in vitro tube formation assay and evaluating the expression levels of the VM markers EphA2 and VE-cadherin by Western blotting and flow cytometer analyses, we demonstrated that vemurafenib-resistant cells obtained by both models are characterized by an increased ability to perform VM. Moreover, by exploiting the CRISPR-Cas9 technique and using the urokinase plasminogen activator receptor (uPAR) inhibitor M25, we identified uPAR as a driver of VM expressed by vemurafenib-resistant melanoma cells. Thus, uPAR targeting may be successfully leveraged as a new complementary therapy to inhibit VM in drug-resistant melanoma patients, to counteract the rapid progression and dissemination of the disease.
- Published
- 2022
22. Enhanced Vasculogenic Capacity Induced by 5-Fluorouracil Chemoresistance in a Gastric Cancer Cell Line
- Author
-
Fabio Cianchi, Francesco Coratti, Lucia Magnelli, Giampaolo Versienti, Elena Andreucci, Laura Papucci, Giuseppe Barbato, Sara Peri, Alessio Biagioni, Nicola Schiavone, Fabio Staderini, and Lisa Giovannelli
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Metastasis ,0302 clinical medicine ,Biology (General) ,Spectroscopy ,vasculogenic mimicry ,Neovascularization, Pathologic ,chemoresistance ,General Medicine ,Thalidomide ,Up-Regulation ,Computer Science Applications ,Chemistry ,030220 oncology & carcinogenesis ,Fluorouracil ,medicine.drug ,Paclitaxel ,QH301-705.5 ,Antineoplastic Agents ,Article ,Catalysis ,epithelial-to-endothelial transition ,Inorganic Chemistry ,03 medical and health sciences ,Downregulation and upregulation ,Stomach Neoplasms ,Cell Line, Tumor ,medicine ,Humans ,Vasculogenic mimicry ,Physical and Theoretical Chemistry ,Thymidine phosphorylase ,Molecular Biology ,QD1-999 ,Cisplatin ,Thymidine Phosphorylase ,Chemotherapy ,business.industry ,gastric cancer ,Organic Chemistry ,Endothelial Cells ,Cancer ,tumor angiogenesis ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,business - Abstract
Chemotherapy is still widely used as a coadjutant in gastric cancer when surgery is not possible or in presence of metastasis. During tumor evolution, gatekeeper mutations provide a selective growth advantage to a subpopulation of cancer cells that become resistant to chemotherapy. When this phenomenon happens, patients experience tumor recurrence and treatment failure. Even if many chemoresistance mechanisms are known, such as expression of ATP-binding cassette (ABC) transporters, aldehyde dehydrogenase (ALDH1) activity and activation of peculiar intracellular signaling pathways, a common and universal marker for chemoresistant cancer cells has not been identified yet. In this study we subjected the gastric cancer cell line AGS to chronic exposure of 5-fluorouracil, cisplatin or paclitaxel, thus selecting cell subpopulations showing resistance to the different drugs. Such cells showed biological changes, among them, we observed that the acquired chemoresistance to 5-fluorouracil induced an endothelial-like phenotype and increased the capacity to form vessel-like structures. We identified the upregulation of thymidine phosphorylase (TYMP), which is one of the most commonly reported mutated genes leading to 5-fluorouracil resistance, as the cause of such enhanced vasculogenic ability.
- Published
- 2021
23. EGFR/uPAR interaction as druggable target to overcome vemurafenib acquired resistance in melanoma cells
- Author
-
Mario Del Rosso, Elena Andreucci, Francesca Margheri, Silvia Peppicelli, Gabriella Fibbi, Nicola Pimpinelli, Simona Serratì, Alessio Biagioni, Anastasia Chillà, Anna Laurenzana, Jessica Ruzzolini, and Lido Calorini
- Subjects
0301 basic medicine ,Male ,Research paper ,Acquired resistance ,EGFR ,Melanoma ,Vemurafenib ,uPAR ,0302 clinical medicine ,skin and connective tissue diseases ,Aged, 80 and over ,Gene knockdown ,medicine.diagnostic_test ,General Medicine ,Middle Aged ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Female ,medicine.drug ,Protein Binding ,Signal Transduction ,Proto-Oncogene Proteins B-raf ,Cell Survival ,General Biochemistry, Genetics and Molecular Biology ,Receptors, Urokinase Plasminogen Activator ,03 medical and health sciences ,Western blot ,Cell Line, Tumor ,medicine ,Humans ,Protein kinase B ,neoplasms ,Aged ,Cell Proliferation ,Cell growth ,business.industry ,medicine.disease ,biological factors ,Urokinase receptor ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,business ,V600E - Abstract
Background BRAF inhibitor (BRAF-I) therapy for melanoma patients harboring the V600E mutation is initially highly effective, but almost all patients relapse within a few months. Understanding the molecular mechanisms behind BRAF-I responsiveness and acquired resistance is therefore an important issue. Here we assessed the role of urokinase type plasminogen activator receptor (uPAR) as a potentially valuable biomarker in the acquisition of BRAF-I resistance in V600E mutant melanoma cells. Methods We examined uPAR and EGFR levels by real time PCR and western blot analysis. uPAR loss of function was realized by knocking down uPAR by RNAi or using M25, a peptide that uncouples uPAR-integrin interaction. We investigated uPAR-β1integrin-EGFR association by co-immunoprecipitation and confocal immuno-fluorescence analysis. Acquired resistance to BRAF-I was generated by chronic exposure of cells to vemurafenib. Findings We proved that uPAR knockdown in combination with vemurafenib inhibits melanoma cell proliferation to greater extent than either treatment alone causing a decrease in AKT and ERK1/2 phosphorylation. Conversely, we demonstrated that uPAR enforced over-expression results in reduced sensitivity to BRAF inhibition. Moreover, by targeting uPAR and EGFR interaction with an integrin antagonist peptide we restored vemurafenib responsiveness in melanoma resistant cells. Furthermore, we found significant detectable uPAR and EGFR levels in tumor biopsies of 4 relapsed patients. Interpretation We disclosed an unpredicted mechanism of reduced sensitiveness to BRAF inhibition, driven by elevated levels of uPAR and identified a potential therapeutic strategy to overcome acquired resistance. Funds Associazione Italiana Ricerca sul Cancro (AIRC); Ente Cassa di Risparmio di Firenze.
- Published
- 2019
24. CRISPR/Cas9 uPAR Gene Knockout Results in Tumor Growth Inhibition, EGFR Downregulation and Induction of Stemness Markers in Melanoma and Colon Carcinoma Cell Lines
- Author
-
Francesca Bianchini, Mario Del Rosso, Pia Ragno, Anna Li Santi, Alessio Biagioni, Gabriella Fibbi, Anastasia Chillà, Anna Laurenzana, Elena Andreucci, Francesca Margheri, Francesca Scavone, Lido Calorini, and Silvia Peppicelli
- Subjects
PLAUR Gene ,Cancer Research ,Melanoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biology ,medicine.disease ,medicine.disease_cause ,CRISPR ,colon cancer ,melanoma ,miR146a ,urokinase-type plasminogen activator receptor ,Metastasis ,Urokinase receptor ,Downregulation and upregulation ,Oncology ,Tumor progression ,medicine ,Cancer research ,Carcinogenesis ,Gene knockout ,RC254-282 ,Original Research - Abstract
uPAR is a globular protein, tethered to the cell membrane by a GPI-anchor involved in several cancer-related properties and its overexpression commonly correlates with poor prognosis and metastasis. We investigated the consequences of uPAR irreversible loss in human melanoma and colon cancer cell lines, knocking out its expression by CRISPR/Cas9. We analyzed through flow cytometry, western blotting and qPCR, the modulation of the most known cancer stem cells-associated genes and the EGFR while we observed the proliferation rate exploiting 2D and 3D cellular models. We also generated uPAR “rescue” expression cell lines as well as we promoted the expression of only its 3’UTR to demonstrate the involvement of uPAR mRNA in tumor progression. Knocking out PLAUR, uPAR-encoding gene, we observed an inhibited growth ratio unexpectedly coupled with a significant percentage of cells acquiring a stem-like phenotype. In vivo experiments demonstrated that uPAR loss completely abrogates tumorigenesis despite the gained stem-like profile. Nonetheless, we proved that the reintroduction of the 3’UTR of PLAUR gene was sufficient to restore the wild-type status validating the hypothesis that such a region may act as a “molecular sponge”. In particular miR146a, by binding PLAUR 3’ UTR region might be responsible for uPAR-dependent inhibition of EGFR expression.
- Published
- 2021
25. Glutamine Availability Controls BCR/Abl Protein Expression and Functional Phenotype of Chronic Myeloid Leukemia Cells Endowed with Stem/Progenitor Cell Potential
- Author
-
Silvia Peppicelli, Caterina Mancini, Alessio Biagioni, Martina Poteti, Nathalie M. Mazure, Matteo Lulli, Alessandro Tubita, Elisabetta Rovida, Giulio Menegazzi, Giulia Cheloni, Ignazia Tusa, Persio Dello Sbarba, Angela Silvano, mazure, nathalie, Università degli Studi di Firenze = University of Florence (UniFI), Beth Israel Deaconess Medical Center [Boston] (BIDMC), Harvard Medical School [Boston] (HMS), Centre méditerranéen de médecine moléculaire (C3M), Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Côte d'Azur (UCA), Università degli Studi di Firenze = University of Florence [Firenze] (UNIFI), Université Nice Sophia Antipolis (... - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
[SDV.MHEP.HEM] Life Sciences [q-bio]/Human health and pathology/Hematology ,Cancer Research ,[SDV]Life Sciences [q-bio] ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Article ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,chronic myeloid leukemia ,hemic and lymphatic diseases ,medicine ,[SDV.BC.BC] Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,stem cell niche ,Progenitor cell ,glucose ,neoplasms ,RC254-282 ,030304 developmental biology ,0303 health sciences ,low oxygen ,ABL ,Glutaminase ,Chemistry ,breakpoint cluster region ,Myeloid leukemia ,Hematopoietic stem cell ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Chronic myeloid leukemia ,Glucose ,Glutamine ,Low oxygen ,Minimal residual disease ,Stem cell niche ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,glutamine ,minimal residual disease ,Stem cell - Abstract
Simple Summary In chronic myeloid leukemia (CML), a neoplasm brilliantly taken care of by a molecularly targeted therapeutic approach, the achievement of cure is nevertheless prevented by the maintenance of a small subset of treatment-resistant leukemia stem cells (LSCs), sustaining the so-called minimal residual disease of CML. The phenotypical and functional characterization of this LSC subset is, therefore, crucial to aim at the eradication of disease. Such a characterization includes the acquisition of information relative to the metabolic profile of treatment-resistant LSCs, which is functional to their maintenance in bone marrow. A number of metabolic features of LSCs were shown to determine their sensitivity or resistance to therapy. Glutamine metabolism emerged from this study as a potential target to overcome the persistence of therapy-resistant LSCs. Abstract This study was directed to characterize the role of glutamine in the modulation of the response of chronic myeloid leukemia (CML) cells to low oxygen, a main condition of hematopoietic stem cell niches of bone marrow. Cells were incubated in atmosphere at 0.2% oxygen in the absence or the presence of glutamine. The absence of glutamine markedly delayed glucose consumption, which had previously been shown to drive the suppression of BCR/Abl oncoprotein (but not of the fusion oncogene BCR/abl) in low oxygen. Glutamine availability thus emerged as a key regulator of the balance between the pools of BCR/Abl protein-expressing and -negative CML cells endowed with stem/progenitor cell potential and capable to stand extremely low oxygen. These findings were confirmed by the effects of the inhibitors of glucose or glutamine metabolism. The BCR/Abl-negative cell phenotype is the best candidate to sustain the treatment-resistant minimal residual disease (MRD) of CML because these cells are devoid of the molecular target of the BCR/Abl-active tyrosine kinase inhibitors (TKi) used for CML therapy. Therefore, the treatments capable of interfering with glutamine action may result in the reduction in the BCR/Abl-negative cell subset sustaining MRD and in the concomitant rescue of the TKi sensitivity of CML stem cell potential. The data obtained with glutaminase inhibitors seem to confirm this perspective.
- Published
- 2021
26. Cell-Mediated Release of Nanoparticles as a Preferential Option for Future Treatment of Melanoma
- Author
-
Mario Del Rosso, Anna Laurenzana, G. Fibbi, Tommaso Del Rosso, F. Margheri, Anastasia Chillà, and Alessio Biagioni
- Subjects
0301 basic medicine ,Cancer Research ,Enhanced permeability and retention effect ,Review ,Regenerative medicine ,lcsh:RC254-282 ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Therapeutic index ,medicine ,melanoma ,tumor microenvironment ,Tumor microenvironment ,business.industry ,Melanoma ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Nanomedicine ,nanoparticles ,Stem cell ,cell therapy ,business - Abstract
Targeted and immune therapies have unquestionably improved the prognosis of melanoma patients. However the treatment of this neoplasm still requires approaches with a higher therapeutic index, in order to reduce shortcomings related to toxic effects and aspecific targeting. This means developing therapeutic tools derived with high affinity molecules for tumor components differentially expressed in melanoma cells with respect to their normal counterpart. Nanomedicine has sought to address this problem owing to the high modulability of nanoparticles. This approach exploits not only the enhanced permeability and retention effect typical of the tumor microenvironment (passive targeting), but also the use of specific “molecular antennas” that recognize some tumor-overexpressed molecules (active targeting). This line of research has given rise to the so-called “smart nanoparticles,” some of which have already passed the preclinical phase and are under clinical trials in melanoma patients. To further improve nanoparticles partition within tumors, for some years now a line of thought is exploiting the molecular systems that regulate the innate tumor-homing activity of platelets, granulocytes, monocytes/macrophages, stem cells, endothelial-colony-forming cells, and red blood cells loaded with nanoparticles. This new vision springs from the results obtained with some of these cells in regenerative medicine, an approach called “cell therapy.” This review takes into consideration the advantages of cell therapy as the only one capable of overcoming the limits of targeting imposed by the increased interstitial pressure of tumors.
- Published
- 2020
27. microRNA-378a-5p iS a novel positive regulator of melanoma progression
- Author
-
Alessio Biagioni, Gabriella Fibbi, Gennaro Ciliberto, Sara Donzelli, Marianna Desideri, Cristiana Ercolani, Rita Mancini, Elisabetta Valentini, Maria Grazia Tupone, Andrea Sacconi, Giovanni Blandino, Simona D'Aguanno, Luigi Fattore, Simonetta Buglioni, Marta Di Martile, Daniela Trisciuoglio, and Donatella Del Bufalo
- Subjects
0301 basic medicine ,Cancer Research ,Cell biology ,MMP2 ,Angiogenesis ,tumor progression ,Biology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,microRNA ,melanoma ,medicine ,Skin cancer ,Molecular Biology ,Melanoma ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Urokinase receptor ,KLF9 ,030104 developmental biology ,microrna ,cell survival ,030220 oncology & carcinogenesis ,Cancer research ,HOXD10 - Abstract
Evaluating the expression levels of miR-378a-5p both in a large melanoma patient cohort from The Cancer Genome Atlas database and in melanoma patients from our Institute, we found that miR-378a-5p is upregulated in metastatic melanoma specimens. miR-378a-5p expression was also increased in melanoma cells resistant to target therapy, and decreased in response to drug treatment. We also demonstrated that overexpression of miR-378a-5p enhances in vitro cell invasion and migration, and facilitates the ability of melanoma cells to form de novo vasculogenic structures. While performing downstream targeting studies, we confirmed the ability of miR-378a-5p to modulate the expression of known target genes, such as SUFU, FUS-1, and KLF9. Luciferase-3′UTR experiments also identified STAMBP and HOXD10 as new miR-378a-5p target genes. MMP2 and uPAR, two HOXD10 target genes, were positively regulated by miR-378a-5p. Genetic and pharmacologic approaches inhibiting uPAR expression and activity evidenced that the in vitro tumor-promoting functions of miR-378a-5p, were in part mediated by uPAR. Of note miR-378a-5p was also able to increase VEGF, as well as in vitro and in vivo angiogenesis. Finally, genetic and pharmacologic modulation of Bcl-2 evidenced Bcl-2 ability to regulate miR-378a-5p expression. In conclusion, to the best of our knowledge, this is the first study demonstrating that miR-378a-5p acts as an oncogenic microRNA in melanoma.
- Published
- 2020
28. uPA/uPAR system activation drives a glycolytic phenotype in melanoma cells
- Author
-
Anna, Laurenzana, Anastasia, Chillà, Cristina, Luciani, Silvia, Peppicelli, Alessio, Biagioni, Francesca, Bianchini, Elena, Tenedini, Eugenio, Torre, Alessandra, Mocali, Lido, Calorini, Francesca, Margheri, Gabriella, Fibbi, and Mario, Del Rosso
- Subjects
cancer cell metabolism ,melanoma cells ,uPA/uPAR system ,Animals ,Cell Line, Tumor ,Down-Regulation ,Female ,Glycolysis ,HEK293 Cells ,Heterografts ,Humans ,Melanoma ,Mice ,Mice, Nude ,Mice, SCID ,Neoplasm Invasiveness ,Phenotype ,Receptors, Urokinase Plasminogen Activator ,Urokinase-Type Plasminogen Activator ,Oncology ,Cancer Research - Abstract
In this manuscript, we show the involvement of the uPA/uPAR system in the regulation of aerobic glycolysis of melanoma cells. uPAR over-expression in human melanoma cells controls an invasive and glycolytic phenotype in normoxic conditions. uPAR down-regulation by siRNA or its uncoupling from integrins, and hence from integrin-linked tyrosine kinase receptors (IL-TKRs), by an antagonist peptide induced a striking inhibition of the PI3K/AKT/mTOR/HIF1α pathway, resulting into impairment of glucose uptake, decrease of several glycolytic enzymes and of PKM2, a checkpoint that controls metabolism of cancer cells. Further, binding of uPA to uPAR regulates expression of molecules that govern cell invasion, including extracellular matrix metallo-proteinases inducer (EMPPRIN) and enolase, a glycolytyc enzyme that also serves as a plasminogen receptor, thus providing a common denominator between tumor metabolism and phenotypic invasive features. Such effects depend on the α5β1-integrin-mediated uPAR connection with EGFR in melanoma cells with engagement of the PI3K-mTOR-HIFα pathway. HIF-1α trans-activates genes whose products mediate tumor invasion and glycolysis, thus providing the common denominator between melanoma metabolism and its invasive features. These findings unveil a unrecognized interaction between the invasion-related uPAR and IL-TKRs in the control of glycolysis and disclose a new pharmacological target (i.e., uPAR/IL-TKRs axis) for the therapy of melanoma.
- Published
- 2017
29. Type II CRISPR/Cas9 approach in the oncological therapy
- Author
-
Elena Andreucci, Francesca Margheri, Anastasia Chillà, Gabriella Fibbi, Silvia Peppicelli, M. Del Rosso, Alessio Biagioni, and Anna Laurenzana
- Subjects
0301 basic medicine ,Cancer Research ,Review ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,Gene Knockout Techniques ,Plasmid ,Gene therapy ,Genome editing ,Neoplasms ,CRISPR ,Animals ,Humans ,Gene delivery ,Clinical treatment ,Gene knockout ,Genetics ,Mechanism (biology) ,Genetic Therapy ,Highly selective ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Genetic engineering ,Immune therapy ,Oncology ,Gene Expression Regulation ,CRISPR-Cas Systems - Abstract
CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) is a prokaryotic adaptable immune mechanism used by many bacteria and archaea to protect themselves from foreign nucleic acids. This complex system can recognize and cut non-self DNA in order to provide the prokaryotic organisms a strong defense against foreign viral or plasmid attacks and make the cell immune from further assaults. Today, it has been adapted to be used in vitro and in vivo in eukaryotic cells to perform a complete and highly selective gene knockout or a specific gene editing. The ease of use and the low cost are only two features that have made it very popular among the scientific community and the possibility to be used as a clinical treatment in several genetic derived pathologies has rapidly spread its fame worldwide. However, CRISPR is still not fully understood and many efforts need to be done in order to make it a real power tool for the human clinical treatment especially for oncological patients. Indeed, since cancer originates from non-lethal genetic disorders, CRISPR discovery fuels the hope to strike tumors on their roots. More than 4000 papers regarding CRISPR were published in the last ten years and only few of them take in count the possible applications in oncology. The purpose of this review is to clarify many problematics on the CRISPR usage and highlight its potential in oncological therapy.
- Published
- 2017
30. Inhibition of uPAR-TGFβ crosstalk blocks MSC-dependent EMT in melanoma cells
- Author
-
Mario Del Rosso, Silvia Peppicelli, Nicola Pimpinelli, Gabriella Fibbi, Cristina Luciani, Francesca Bianchini, Francesca Margheri, Lido Calorini, Anna Laurenzana, Alessio Biagioni, and Anastasia Chillà
- Subjects
Epithelial-Mesenchymal Transition ,Transplantation, Heterologous ,Receptors, Urokinase Plasminogen Activator ,Mice ,Transforming Growth Factor beta ,Cell Line, Tumor ,Drug Discovery ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,Melanoma ,neoplasms ,Genetics (clinical) ,Tumor microenvironment ,biology ,Chemistry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Transforming growth factor beta ,medicine.disease ,Urokinase receptor ,Tumor progression ,Culture Media, Conditioned ,Cancer cell ,Immunology ,biology.protein ,Cancer research ,Epithelial-to-mesenchymal transition (EMT) ,MSC ,TGFβ ,uPAR ,Molecular Medicine ,Drug Discovery3003 Pharmaceutical Science ,Receptors, Transforming Growth Factor beta ,Neoplasm Transplantation - Abstract
The capacity of cancer cells to undergo epithelial-to-mesenchymal transition (EMT) is now considered a hallmark of tumor progression, and it is known that interactions between cancer cells and mesenchymal stem cells (MSCs) of tumor microenvironment may promote this program. Herein, we demonstrate that MSC-conditioned medium (MSC-CM) is a potent inducer of EMT in melanoma cells. The EMT profile acquired by MSC-CM-exposed melanoma cells is characterized by an enhanced level of mesenchymal markers, including TGFβ/TGFβ-receptors system upregulation, by increased invasiveness and uPAR expression, and in vivo tumor growth. Silencing TGFβ in MSC is found to abrogate ability of MSC to promote EMT characteristics in melanoma cells, together with uPAR expression, and this finding is strengthened using an antagonist peptide of TGFβRIII, the so-called P17. Finally, we demonstrate that the uPAR antisense oligonucleotide (uPAR aODN) may inhibit EMT of melanoma cells either stimulated by exogenous TGFβ or MSC-CM. Thus, uPAR upregulation in melanoma cells exposed to MSC-medium drives TGFβ-mediated EMT. On the whole, TGFβ/uPAR dangerous liaison in cancer cell/MSC interactions may disclose a new strategy to abrogate melanoma progression.Mesenchymal stem cell (MSC)-conditioned medium induces EMT-like profile in melanoma. MSC-derived TGFβ promotes uPAR and TGFβ/TGFβ-receptor upregulation in melanoma. TGFβ gene silencing in MSCs downregulates uPAR expression and EMT in melanoma. uPAR downregulation prevents MSC-induced EMT-like profile in melanoma cells. Inhibition of the dangerous TGFβ/uPAR relationship might abrogate melanoma progression.
- Published
- 2015
31. Differential uPAR recruitment in caveolar-lipid rafts by GM1 and GM3 gangliosides regulates endothelial progenitor cells angiogenesis
- Author
-
Alessio Biagioni, Laura Papucci, Nicola Schiavone, Anastasia Chillà, Giancarlo Margheri, Riccardo D’Agostino, Tommaso Del Rosso, Mario Del Rosso, Cristina Luciani, Silvana Trigari, Simona Serratì, Elena Andreucci, Francesca Margheri, Gabriella Fibbi, and Anna Laurenzana
- Subjects
GM3 ,Angiogenesis ,Caveolin 1 ,GM1 ,Neovascularization, Physiologic ,G(M1) Ganglioside ,Biology ,Caveolae ,Receptors, Urokinase Plasminogen Activator ,Colony-Forming Units Assay ,endothelial colony-forming cells ,Cell membrane ,angiogenesis ,Membrane Microdomains ,medicine ,G(M3) Ganglioside ,Humans ,skin and connective tissue diseases ,Lipid raft ,neoplasms ,endothelial progenitor cells ,lipid rafts ,uPAR ,gangliosides ,Endothelial progenitor cells ,caveolar lipid rafts ,GM1 GM3 ,Infant, Newborn ,Original Articles ,Cell Biology ,caveolar-lipid rafts ,biological factors ,Cell biology ,Blot ,Urokinase receptor ,carbohydrates (lipids) ,Kinetics ,MAPKinases ,Phenotype ,medicine.anatomical_structure ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Signal transduction ,Signal Transduction - Abstract
Gangliosides and the urokinase plasminogen activator receptor (uPAR) tipically partition in specialized membrane microdomains called lipid-rafts. uPAR becomes functionally important in fostering angiogenesis in endothelial progenitor cells (EPCs) upon recruitment in caveolar-lipid rafts. Moreover, cell membrane enrichment with exogenous GM1 ganglioside is pro-angiogenic and opposite to the activity of GM3 ganglioside. On these basis, we first checked the interaction of uPAR with membrane models enriched with GM1 or GM3, relying on the adoption of solid-supported mobile bilayer lipid membranes with raft-like composition formed onto solid hydrophilic surfaces, and evaluated by surface plasmon resonance (SPR) the extent of uPAR recruitment. We estimated the apparent dissociation constants of uPAR-GM1/GM3 complexes. These preliminary observations, indicating that uPAR binds preferentially to GM1-enriched biomimetic membranes, were validated by identifying a pro-angiogenic activity of GM1-enriched EPCs, based on GM1-dependent uPAR recruitment in caveolar rafts. We have observed that addition of GM1 to EPCs culture medium promotes matrigel invasion and capillary morphogenesis, as opposed to the anti-angiogenesis activity of GM3. Moreover, GM1 also stimulates MAPKinases signalling pathways, typically associated with an angiogenesis program. Caveolar-raft isolation and Western blotting of uPAR showed that GM1 promotes caveolar-raft partitioning of uPAR, as opposed to control and GM3-challenged EPCs. By confocal microscopy, we have shown that in EPCs uPAR is present on the surface in at least three compartments, respectively, associated to GM1, GM3 and caveolar rafts. Following GM1 exogenous addition, the GM3 compartment is depleted of uPAR which is recruited within caveolar rafts thereby triggering angiogenesis.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.