19 results on '"Silvia Pietrobono"'
Search Results
2. Targeting non-canonical activation of GLI1 by the SOX2-BRD4 transcriptional complex improves the efficacy of HEDGEHOG pathway inhibition in melanoma
- Author
-
Eugenio Gaudio, Fabrizio Manetti, Sinforosa Gagliardi, Elena Petricci, Silvia Pietrobono, Laura Carrassa, Mariapaola Zitani, Nikolai Makukhin, Adam G. Bond, Barbara Stecca, Martin E. Fernandez-Zapico, Francesca Migliorini, Francesco Bertoni, Alessio Ciulli, and Brooke D. Paradise
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,GLI1 ,Cell Cycle Proteins ,Guanidines ,Hedgehog pathway ,Mice ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Molecular Targeted Therapy ,Melanoma ,SOX2-BRD4 transcriptional complex ,biology ,integumentary system ,Drug Synergism ,Dipeptides ,Smoothened Receptor ,Hedgehog signaling pathway ,030220 oncology & carcinogenesis ,embryonic structures ,Female ,Heterocyclic Compounds, 3-Ring ,Cell signalling ,Signal Transduction ,BRD4 ,Mice, Nude ,GLI1, Hedgehog pathway, melanoma, SOX2-BRD4 transcriptional complex, acylguanidine ,Zinc Finger Protein GLI1 ,Article ,03 medical and health sciences ,SOX2 ,Cell Line, Tumor ,Spheroids, Cellular ,Genetics ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Molecular Biology ,Hedgehog ,Cell Proliferation ,SOXB1 Transcription Factors ,acylguanidine ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,biology.protein ,Cancer research ,Smoothened ,Transcription Factors - Abstract
Despite the development of new targeted and immune therapies, the prognosis of metastatic melanoma remains bleak. Therefore, it is critical to better understand the mechanisms controlling advanced melanoma to develop more effective treatment regimens. Hedgehog/GLI (HH/GLI) signaling inhibitors targeting the central pathway transducer Smoothened (SMO) have shown to be clinical efficacious in skin cancer; however, several mechanisms of non-canonical HH/GLI pathway activation limit their efficacy. Here, we identify a novel SOX2-BRD4 transcriptional complex driving the expression of GLI1, the final effector of the HH/GLI pathway, providing a novel mechanism of non-canonical SMO-independent activation of HH/GLI signaling in melanoma. Consistently, we find a positive correlation between the expression of GLI1 and SOX2 in human melanoma samples and cell lines. Further, we show that combined targeting of canonical HH/GLI pathway with the SMO inhibitor MRT-92 and of the SOX2-BRD4 complex using a potent Proteolysis Targeted Chimeras (PROTACs)-derived BRD4 degrader (MZ1), yields a synergistic anti-proliferative effect in melanoma cells independently of their BRAF, NRAS, and NF1 mutational status, with complete abrogation of GLI1 expression. Combination of MRT-92 and MZ1 strongly potentiates the antitumor effect of either drug as single agents in an orthotopic melanoma model. Together, our data provide evidence of a novel mechanism of non-canonical activation of GLI1 by the SOX2-BRD4 transcriptional complex, and describe the efficacy of a new combinatorial treatment for a subset of melanomas with an active SOX2-BRD4-GLI1 axis.
- Published
- 2021
3. Predictive Biomarkers for a Personalized Approach in Resectable Pancreatic Cancer
- Author
-
Valeria Merz, Domenico Mangiameli, Camilla Zecchetto, Alberto Quinzii, Silvia Pietrobono, Carlo Messina, Simona Casalino, Marina Gaule, Camilla Pesoni, Pasquale Vitale, Chiara Trentin, Michela Frisinghelli, Orazio Caffo, and Davide Melisi
- Subjects
predictive factors ,target therapy ,molecular profiling ,microbiota ,tumor microenvironment ,Surgery ,neoadjuvant therapy ,preoperative treatment ,resectable pancreatic cancer - Abstract
The mainstay treatment for patients with immediate resectable pancreatic cancer remains upfront surgery, which represents the only potentially curative strategy. Nevertheless, the majority of patients surgically resected for pancreatic cancer experiences disease relapse, even when a combination adjuvant therapy is offered. Therefore, aiming at improving disease free survival and overall survival of these patients, there is an increasing interest in evaluating the activity and efficacy of neoadjuvant and perioperative treatments. In this view, it is of utmost importance to find biomarkers able to select patients who may benefit from a preoperative therapy rather than upfront surgical resection. Defined genomic alterations and a dynamic inflammatory microenvironment are the major culprits for disease recurrence and resistance to chemotherapeutic treatments in pancreatic cancer patients. Signal transduction pathways or tumor immune microenvironment could predict early recurrence and response to chemotherapy. In the last decade, distinct molecular subtypes of pancreatic cancer have been described, laying the bases to a tailored therapeutic approach, started firstly in the treatment of advanced disease. Patients with homologous repair deficiency, in particular with mutant germline BRCA genes, represent the first subgroup demonstrating to benefit from specific therapies. A fraction of patients with pancreatic cancer could take advantage of genome sequencing with the aim of identifying possible targetable mutations. These genomic driven strategies could be even more relevant in a potentially curative setting. In this review, we outline putative predictive markers that could help in the next future in tailoring the best therapeutic strategy for pancreatic cancer patients with a potentially curable disease.
- Published
- 2022
- Full Text
- View/download PDF
4. p38 MAPK-dependent phosphorylation of transcription factor SOX2 promotes an adaptive response to BRAF inhibitors in melanoma cells
- Author
-
Silvia Pietrobono, Raffaella De Paolo, Domenico Mangiameli, Andrea Marranci, Ilaria Battisti, Cinzia Franchin, Giorgio Arrigoni, Davide Melisi, Laura Poliseno, and Barbara Stecca
- Subjects
BRAF inhibitor ,SOX2 ,drug resistance ,p38 MAPK ,Proto-Oncogene Proteins B-raf ,MAP Kinase Signaling System ,SOXB1 Transcription Factors ,Cell Biology ,Biochemistry ,p38 Mitogen-Activated Protein Kinases ,Adenosine Triphosphate ,Drug Resistance, Neoplasm ,Cell Line, Tumor ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Melanoma ,Protein Kinase Inhibitors ,Zebrafish - Abstract
Despite recent advances in the development of BRAF kinase inhibitors (BRAFi) for BRAF-mutant melanomas, development of resistance remains a major clinical problem. In addition to genetic alterations associated with intrinsic resistance, several adaptive response mechanisms are known to be rapidly activated to allow cell survival in response to treatment, limiting efficacy. A better understanding of the mechanisms driving resistance is urgently needed to improve the success of BRAF-targeted therapies and to make therapeutic intervention more durable. In this study, we identify the mitogen-activated protein kinase (MAPK) p38 as a novel mediator of the adaptive response of melanoma cells to BRAF-targeted therapy. Our findings demonstrate that BRAFi leads to an early increase in p38 activation, which promotes phosphorylation of the transcription factor SOX2 at Ser251, enhancing SOX2 stability, nuclear localization, and transcriptional activity. Furthermore, functional studies show that SOX2 depletion increases sensitivity of melanoma cells to BRAFi, whereas overexpression of a phosphomimetic SOX2-S251E mutant is sufficient to drive resistance and desensitize melanoma cells to BRAFi in vitro and in a zebrafish xenograft model. We also found that SOX2 phosphorylation at Ser251 confers resistance to BRAFi by binding to the promoter and increasing transcriptional activation of the ATP-binding cassette drug efflux transporter ABCG2. In summary, we unveil a p38/SOX2-mediated mechanism of adaptive response to BRAFi, which provides prosurvival signals to melanoma cells against the cytotoxic effects of BRAFi prior to acquiring resistance.
- Published
- 2022
5. Exceptional Clinical Response to Alectinib in Pancreatic Acinar Cell Carcinoma With a Novel ALK-KANK4 Gene Fusion
- Author
-
Marina Gaule, Camilla Pesoni, Alberto Quinzii, Camilla Zecchetto, Simona Casalino, Valeria Merz, Serena Contarelli, Silvia Pietrobono, Elena Vissio, Luca Molinaro, Enrica Manzin, Roberta Volpatto, Giorgio Vellani, and Davide Melisi
- Subjects
Male ,Cancer Research ,Carcinoma, Acinar Cell ,Carbazoles ,Case Reports ,Middle Aged ,Ankyrin Repeat ,Pancreatic Neoplasms ,Treatment Outcome ,Oncology ,ALK ,Piperidines ,Humans ,Anaplastic Lymphoma Kinase ,Gene Fusion - Published
- 2022
6. MAPK15 Controls Hedgehog Signaling in Medulloblastoma Cells by Regulating Primary Ciliogenesis
- Author
-
Cristina Zanini, Mario Chiariello, Francesco Imperatore, Silvia Pietrobono, Barbara Stecca, and Lorenzo Franci
- Subjects
Medulloblastoma ,cancer stem cells ,Cancer Research ,biology ,Cell growth ,GLI1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,MAP Kinases ,medicine.disease ,Hedgehog signaling pathway ,Article ,Cell biology ,primary cilia ,Oncology ,Cancer stem cell ,Ciliogenesis ,biology.protein ,medicine ,MAPK15 ,Hedgehog ,RC254-282 ,medullo-spheres - Abstract
Simple Summary In eukaryotes, MAPK15 controls the assembly of primary cilia, which are microtubule-based cell surface organelles necessary for sensing and processing developmental signals as well as for transducing tumorigenic Hedgehog signaling in medulloblastomas and basal cell carcinomas. The aim of this study was to evaluate the role of MAPK15 in regulating Hedgehog signaling in medulloblastoma cells. Indeed, we demonstrated strict dependency on this kinase of medulloblastoma ciliogenesis and Hedgehog signaling, which resulted in a reduced cancer stem cell compartment. Based on the scarce therapeutic options available for medulloblastoma patients, our data support the possibility of exploiting novel pharmacological approaches targeting this often-underestimated MAP kinase. Abstract In medulloblastomas, genetic alterations resulting in over-activation and/or deregulation of proteins involved in Hedgehog (HH) signaling lead to cellular transformation, which can be prevented by inhibition of primary ciliogenesis. Here, we investigated the role of MAPK15 in HH signaling and, in turn, in HH-mediated cellular transformation. We first demonstrated, in NIH3T3 mouse fibroblasts, the ability of this kinase of controlling primary ciliogenesis and canonical HH signaling. Next, we took advantage of transformed human medulloblastoma cells belonging to the SHH-driven subtype, i.e., DAOY and ONS-76 cells, to ascertain the role for MAPK15 in HH-mediated cellular transformation. Specifically, medullo-spheres derived from these cells, an established in vitro model for evaluating progression and malignancy of putative tumor-initiating medulloblastoma cells, were used to demonstrate that MAPK15 regulates self-renewal of these cancer stem cell-like cells. Interestingly, by using the HH-related oncogenes SMO-M2 and GLI2-DN, we provided evidences that disruption of MAPK15 signaling inhibits oncogenic HH overactivation in a specific cilia-dependent fashion. Ultimately, we show that pharmacological inhibition of MAPK15 prevents cell proliferation of SHH-driven medulloblastoma cells, overall suggesting that oncogenic HH signaling can be counteracted by targeting the ciliary gene MAPK15, which could therefore be considered a promising target for innovative “smart” therapies in medulloblastomas.
- Published
- 2021
7. The Role of Glycosylation in Melanoma Progression
- Author
-
Chiara De Vellis, Silvia Pietrobono, and Barbara Stecca
- Subjects
Glycosylation ,QH301-705.5 ,Review ,Biology ,Malignant transformation ,Metastasis ,chemistry.chemical_compound ,sialylation ,Polysaccharides ,medicine ,melanoma ,metastasis ,Animals ,Humans ,Epigenetics ,Biology (General) ,neoplasms ,Fucosylation ,immune evasion ,glycan branching ,Melanoma ,Cancer ,General Medicine ,medicine.disease ,carbohydrates (lipids) ,Cell Transformation, Neoplastic ,chemistry ,Cancer research ,fucosylation ,Skin cancer - Abstract
Malignant melanoma is the most aggressive form of skin cancer, which originates from the malignant transformation of melanocytes, the melanin-producing cells of the skin. Melanoma progression is typically described as a stepwise process in which metastasis formation ensues late during disease. A large body of evidence has shown that the accumulation of genetic and epigenetic alterations drives melanoma progression through the different steps. Mortality in melanoma is associated with metastatic disease. Accordingly, early-stage melanoma can be cured in the majority of cases by surgical excision, while late-stage melanoma is a highly lethal disease. Glycosylation is a post-translational modification that involves the transfer of glycosyl moieties to specific amino acid residues of proteins to form glycosidic bonds through the activity of glycosyltransferases. Aberrant glycosylation is considered a hallmark of cancer as it occurs in the majority of tumor types, including melanoma. The most widely occurring glycosylation changes in melanoma are represented by sialylation, fucosylation, and N- and I-glycan branching. In this review, we discuss the role of glycosylation in melanoma and provide insights on the mechanisms by which aberrant glycosylation promotes melanoma progression through activation of invasion and metastasis, immune evasion and cell proliferation.
- Published
- 2021
8. A Novel atTAK Against Hepatocellular Carcinoma: Overcoming Resistance to Sorafenib
- Author
-
Davide Melisi and Silvia Pietrobono
- Subjects
Male ,DFS, disease-free survival ,Drug Resistance ,IC50, half maximal inhibitory concentration ,RC799-869 ,DMSO, dimethyl sulfoxide ,Mice ,GSEA, gene set enrichment analysis ,SR, sorafenib-resistant ,GEO, Gene Expression Omnibus ,NF-κB, nuclear factor kappa B ,Original Research ,Liver Neoplasms ,Gastroenterology ,RNA-Binding Proteins ,IP, immunoprecipitation ,RIP, RNA immunoprecipitation ,Hep G2 Cells ,Diseases of the digestive system. Gastroenterology ,Sorafenib ,TAK1, transforming growth factor-β–activated kinase 1 ,MAP Kinase Kinase Kinases ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Editorial ,Hepatocellular carcinoma ,qPCR, quantitative polymerase chain reaction ,EMT, epithelial-mesenchymal transition ,NASH, nonalcoholic steatohepatitis ,IHC, immunohistochemistry ,medicine.drug ,Carcinoma, Hepatocellular ,TAK1 ,CHX, cycloheximide ,Cell Line, Tumor ,medicine ,Animals ,Humans ,neoplasms ,Cell Proliferation ,Hepatology ,business.industry ,F-Box Proteins ,Phenylurea Compounds ,Carcinoma ,TNF-α, tumor necrosis factor-alpha ,Ubiquitination ,Membrane Proteins ,Hepatocellular ,Hepatocellular Carcinoma ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,SRRSH, Sir Run Run Shaw Hospital ,Drug Resistance, Neoplasm ,siRNA, small interfering RNA ,Mutation ,Proteolysis ,Cancer research ,Neoplasm ,TCGA, The Cancer Genome Atlas ,NAFLD, nonalcoholic fatty liver disease ,RT, real-time ,business ,Ubiquitin-Mediated Proteolysis ,HCC, hepatocellular carcinoma ,OXO, 5z-7-oxozeaenol ,DAPI, 4′,6-diamidino-2-phenylindole - Abstract
Background & Aims Identifying novel and actionable targets in hepatocellular carcinoma (HCC) remains an unmet medical need. TAK1 was originally identified as a transforming growth factor-β–activated kinase and was further proved to phosphorylate and activate numerous downstream targets and promote cancer progression. However, the role of TAK1 in developed HCC progression and targeted therapy resistance is poorly understood. Methods The expression of TAK1 or MTDH in HCC cell lines, tumor tissues, and sorafenib-resistant models was analyzed by in silico analysis, quantitative real-time polymerase chain reaction, Western blotting, and immunohistochemistry. In vivo and in vitro experiments were introduced to examine the function of TAK1 or MTDH in HCC and sorafenib resistance using small interfering RNA and pharmacologic inhibitors in combination with or without sorafenib. Co-immunoprecipitation and RNA immunoprecipitation were carried out to determine the binding between TAK1 and FBXW2 or between MTDH and FBXW2 mRNA. Protein half-life and in vitro ubiquitination experiment was performed to validate whether FBXW2 regulates TAK1 degradation. Results Our findings unraveled the clinical significance of TAK1 in promoting HCC and sorafenib resistance. We identified a novel E3 ubiquitin ligase, FBXW2, targeting TAK1 for K48-linked polyubiquitylation and subsequent degradation. We also found that MTDH contributes to TAK1 up-regulation in HCC and sorafenib resistance through binding to FBXW2 mRNA and accelerates its degradation. Moreover, combination of TAK1 inhibitor and sorafenib suppressed the growth of sorafenib-resistant HCCLM3 xenograft in mouse models. Conclusions These results revealed novel mechanism underlying TAK1 protein degradation and highlighted the therapeutic value of targeting TAK1 in suppressing HCC and overcoming sorafenib resistance., Graphical abstract
- Published
- 2021
9. Abstract 1064: Targeting stromal autotaxin synergizes with TGF beta inhibition in pancreatic cancer
- Author
-
Fabio Sabbadini, Monica Bertolini, Domenico Mangiameli, Serena Contarelli, Zoë Johnson, Michael M. Lahn, Silvia Pietrobono, and Davide Melisi
- Subjects
Cancer Research ,Oncology - Abstract
The TGFβ receptor inhibitor galunisertib (GAL) has been demonstrated as an effective strategy for the treatment of pancreatic cancer (PC) patients, in part through the modulation of stromal microenvironment. However, alternative pathways driving stromal paracrine signals might impair its effect. Autotaxin (ATX) is an enzyme that converts lysophosphatidylcholine into its bioactive lipid, lysophosphatidic acid (LPA), which in turn promotes inflammation and fibrosis, and contributes to treatment resistance. IOA-289 is a novel ATX inhibitor in clinical development for the treatment of tumors burdened with a high degree of stromal involvement. Here, we hypothesized that ATX could sustain an adaptive response upon inhibition of TGFβ receptor signaling, impairing GAL antitumor activity.Among ten different murine PC orthotopic models, mice bearing RC416 tumors had the highest plasma levels of TGFβ1 as well as high infiltration of cancer associated fibroblasts (CAFs) and fibrosis as measured by Masson’s staining. In coculture models of RC416 with murine pancreatic stellate cells (mPSC4), we measured a significant overexpression of ATX upon treatment with galunisertib mainly by mPSC4, which showed a skewing toward an inflammatory iCAF phenotype through the upregulation of CXCL1 and LY6C1 and the down-regulation of myCAF markers including ACTA2 and Taglin. In the RC416-mPSC4 coculture model, anti-tumor activity was achieved with a combination of gemcitabine plus GAL plus IOA-289, whereas these treatments were ineffective in single culture conditions. Mice bearing RC416 tumors reached a statistically significantly longer survival following treatment with gemcitabine plus IOA-289 or gemcitabine plus GAL [median overall survival (mOS)= 35 days, p Citation Format: Fabio Sabbadini, Monica Bertolini, Domenico Mangiameli, Serena Contarelli, Zoë Johnson, Michael M. Lahn, Silvia Pietrobono, Davide Melisi. Targeting stromal autotaxin synergizes with TGF beta inhibition in pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1064.
- Published
- 2022
- Full Text
- View/download PDF
10. ST3GAL1 is a target of the SOX2-GLI1 transcriptional complex and promotes melanoma metastasis through AXL
- Author
-
Brooke D. Paradise, Lorenzo Tofani, Li Ying, Jaime I. Davila, Giulia Anichini, Silvia Pietrobono, Jann N. Sarkaria, Fabrizio Manetti, Martin E. Fernandez-Zapico, Ryan M. Carr, Barbara Stecca, Hu Li, Alexander Meves, Sara Pepe, Cheng Zhang, Cesare Sala, Luciana L. Almada, Ilaria Battisti, and Giorgio Arrigoni
- Subjects
0301 basic medicine ,Glycosylation ,Skin Neoplasms ,Transcription, Genetic ,Nude ,General Physics and Astronomy ,Receptor tyrosine kinase ,Metastasis ,Mice ,0302 clinical medicine ,lcsh:Science ,Animals ,Gene Expression Regulation, Neoplastic ,Humans ,Melanoma ,Mice, Nude ,Multiprotein Complexes ,Proto-Oncogene Proteins ,Receptor Protein-Tyrosine Kinases ,SOXB1 Transcription Factors ,Sialyltransferases ,Xenograft Model Antitumor Assays ,Zinc Finger Protein GLI1 ,Cancer ,Multidisciplinary ,Molecular medicine ,biology ,030220 oncology & carcinogenesis ,Transcription ,beta-Galactoside alpha-2,3-Sialyltransferase ,Science ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Genetic ,SOX2 ,GLI1 ,medicine ,Gene silencing ,neoplasms ,Neoplastic ,General Chemistry ,medicine.disease ,Axl Receptor Tyrosine Kinase ,030104 developmental biology ,Gene Expression Regulation ,biology.protein ,Cancer research ,lcsh:Q - Abstract
Understanding the molecular events controlling melanoma progression is of paramount importance for the development of alternative treatment options for this devastating disease. Here we report a mechanism regulated by the oncogenic SOX2-GLI1 transcriptional complex driving melanoma invasion through the induction of the sialyltransferase ST3GAL1. Using in vitro and in vivo studies, we demonstrate that ST3GAL1 drives melanoma metastasis. Silencing of this enzyme suppresses melanoma invasion and significantly reduces the ability of aggressive melanoma cells to enter the blood stream, colonize distal organs, seed and survive in the metastatic environment. Analysis of glycosylated proteins reveals that the receptor tyrosine kinase AXL is a major effector of ST3GAL1 pro-invasive function. ST3GAL1 induces AXL dimerization and activation that, in turn, promotes melanoma invasion. Our data support a key role of the ST3GAL1-AXL axis as driver of melanoma metastasis, and highlight the therapeutic potential of targeting this axis to treat metastatic melanoma., Understanding the molecular events controlling melanoma progression are necessary to find improved therapeutics. Here, the authors report oncogenic SOX2-GLI1 transcriptional complex to drive melanoma invasion through the induction of the sialyltransferase ST3GAL1, and report ST3GAL1-AXL axis as driver of melanoma metastasis.
- Published
- 2020
- Full Text
- View/download PDF
11. SOX2 as a novel contributor of oxidative metabolism in melanoma cells
- Author
-
Elena Andreucci, Silvia Pietrobono, Jessica Ruzzolini, Lido Calorini, Francesca Bianchini, Alessio Biagioni, Barbara Stecca, and Silvia Peppicelli
- Subjects
0301 basic medicine ,SOX2 ,lcsh:Medicine ,Oxidative phosphorylation ,Biochemistry ,Oxidative Phosphorylation ,Metastasis ,Tumor extracellular acidosis ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Humans ,HIF1α ,lcsh:QH573-671 ,Molecular Biology ,Melanoma ,Chemistry ,lcsh:Cytology ,Research ,SOXB1 Transcription Factors ,lcsh:R ,Cancer ,Cell Biology ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Warburg effect ,Gene Expression Regulation, Neoplastic ,Melanoma, Tumor extracellular acidosis, SOX2, HIF1α, Oxidative metabolism ,030104 developmental biology ,HIF1A ,Phenotype ,Anaerobic glycolysis ,Cancer cell ,Cancer research ,Oxidative metabolism ,Extracellular Space ,Oxidation-Reduction - 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. Electronic supplementary material The online version of this article (10.1186/s12964-018-0297-z) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
12. The Multifaceted Role of TGF-β in Gastrointestinal Tumors
- Author
-
Monica Bertolini, Serena De Matteis, Davide Melisi, Fabio Sabbadini, Serena Contarelli, Domenico Mangiameli, and Silvia Pietrobono
- Subjects
TGF-β ,Cancer Research ,Tumor microenvironment ,immune plasticity ,gastrointestinal tumors ,medicine.medical_treatment ,EMT ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Review ,SMAD ,Biology ,targeted therapy ,medicine.disease_cause ,Paracrine signalling ,Cytokine ,Oncology ,Tumor progression ,medicine ,Cancer research ,Signal transduction ,Carcinogenesis ,RC254-282 ,Transforming growth factor - Abstract
Simple Summary The transforming growth factor β signaling pathway elicits a broad range of physiological re-sponses, and its misregulation has been related to cancer. The secreted cytokine TGFβ exerts a tumor-suppressive effect that counteracts malignant transformation. However, once tumor has developed, TGFβ can support tumor progression regulating epithelial to mesenchymal transition, invasion and metastasis, stimulating fibrosis, angiogenesis and immune suppression. Here we review the dichotomous role of TGF-β in the progression of gastrointestinal tumors, as well as its intricate crosstalk with other signaling pathways. We also discuss about the therapeutic strate-gies that are currently explored in clinical trials to counteract TGF-β functions. Abstract Transforming growth factor-beta (TGF-β) is a secreted cytokine that signals via serine/threonine kinase receptors and SMAD effectors. Although TGF-β acts as a tumor suppressor during the early stages of tumorigenesis, it supports tumor progression in advanced stages. Indeed, TGF-β can modulate the tumor microenvironment by modifying the extracellular matrix and by sustaining a paracrine interaction between neighboring cells. Due to its critical role in cancer development and progression, a wide range of molecules targeting the TGF-β signaling pathway are currently under active clinical development in different diseases. Here, we focused on the role of TGF-β in modulating different pathological processes with a particular emphasis on gastrointestinal tumors.
- Published
- 2021
- Full Text
- View/download PDF
13. Non-canonical Hedgehog Signaling Pathway in Cancer: Activation of GLI Transcription Factors Beyond Smoothened
- Author
-
Sinforosa Gagliardi, Silvia Pietrobono, and Barbara Stecca
- Subjects
0301 basic medicine ,Patched ,animal structures ,lcsh:QH426-470 ,tumor suppressor ,Review ,Biology ,03 medical and health sciences ,0302 clinical medicine ,non-canonical ,oncogene ,medicine ,Genetics ,cancer ,Hedgehog ,Transcription factor ,Genetics (clinical) ,integumentary system ,Cancer ,medicine.disease ,targeted therapy ,Hedgehog signaling pathway ,lcsh:Genetics ,030104 developmental biology ,PTCH1 ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,Molecular Medicine ,Signal transduction ,Smoothened ,GLI - Abstract
The Hedgehog-GLI (HH-GLI) pathway is a highly conserved signaling that plays a critical role in controlling cell specification, cell-cell interaction and tissue patterning during embryonic development. Canonical activation of HH-GLI signaling occurs through binding of HH ligands to the twelve-pass transmembrane receptor Patched 1 (PTCH1), which derepresses the seven-pass transmembrane G protein-coupled receptor Smoothened (SMO). Thus, active SMO initiates a complex intracellular cascade that leads to the activation of the three GLI transcription factors, the final effectors of the HH-GLI pathway. Aberrant activation of this signaling has been implicated in a wide variety of tumors, such as those of the brain, skin, breast, gastrointestinal, lung, pancreas, prostate and ovary. In several of these cases, activation of HH-GLI signaling is mediated by overproduction of HH ligands (e.g., prostate cancer), loss-of-function mutations in PTCH1 or gain-of-function mutations in SMO, which occur in the majority of basal cell carcinoma (BCC), SHH-subtype medulloblastoma and rhabdomyosarcoma. Besides the classical canonical ligand-PTCH1-SMO route, mounting evidence points toward additional, non-canonical ways of GLI activation in cancer. By non-canonical we refer to all those mechanisms of activation of the GLI transcription factors occurring independently of SMO. Often, in a given cancer type canonical and non-canonical activation of HH-GLI signaling co-exist, and in some cancer types, more than one mechanism of non-canonical activation may occur. Tumors harboring non-canonical HH-GLI signaling are less sensitive to SMO inhibition, posing a threat for therapeutic efficacy of these antagonists. Here we will review the most recent findings on the involvement of alternative signaling pathways in inducing GLI activity in cancer and stem cells. We will also discuss the rationale of targeting these oncogenic pathways in combination with HH-GLI inhibitors as a promising anti-cancer therapies.
- Published
- 2019
14. Aberrant Sialylation in Cancer: Biomarker and Potential Target for Therapeutic Intervention?
- Author
-
Silvia Pietrobono and Barbara Stecca
- Subjects
Cancer Research ,Sialyltransferase ,Angiogenesis ,Review ,sialyltransferases ,Biology ,survival ,Metastasis ,angiogenesis ,inhibitors ,medicine ,cancer ,metastasis ,RC254-282 ,immune evasion ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,invasion ,medicine.disease ,Biomarker (cell) ,carbohydrates (lipids) ,cell death ,Oncology ,Tumor progression ,Cancer research ,biology.protein ,biomarker ,Cancer biomarkers ,Signal transduction - Abstract
Simple Summary Sialylation is a post-translational modification that consists in the addition of sialic acid to growing glycan chains on glycoproteins and glycolipids. Aberrant sialylation is an established hallmark of several types of cancer, including breast, ovarian, pancreatic, prostate, colorectal and lung cancers, melanoma and hepatocellular carcinoma. Hypersialylation can be the effect of increased activity of sialyltransferases and results in an excess of negatively charged sialic acid on the surface of cancer cells. Sialic acid accumulation contributes to tumor progression by several paths, including stimulation of tumor invasion and migration, and enhancing immune evasion and tumor cell survival. In this review we explore the mechanisms by which sialyltransferases promote cancer progression. In addition, we provide insights into the possible use of sialyltransferases as biomarkers for cancer and summarize findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments. Abstract Sialylation is an integral part of cellular function, governing many biological processes including cellular recognition, adhesion, molecular trafficking, signal transduction and endocytosis. Sialylation is controlled by the levels and the activities of sialyltransferases on glycoproteins and lipids. Altered gene expression of these enzymes in cancer yields to cancer-specific alterations of glycoprotein sialylation. Mounting evidence indicate that hypersialylation is closely associated with cancer progression and metastatic spread, and can be of prognostic significance in human cancer. Aberrant sialylation is not only a result of cancer, but also a driver of malignant phenotype, directly impacting key processes such as tumor cell dissociation and invasion, cell-cell and cell-matrix interactions, angiogenesis, resistance to apoptosis, and evasion of immune destruction. In this review we provide insights on the impact of sialylation in tumor progression, and outline the possible application of sialyltransferases as cancer biomarkers. We also summarize the most promising findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments.
- Published
- 2021
- Full Text
- View/download PDF
15. Down-Regulation of SOX2 Underlies the Inhibitory Effects of the Triphenylmethane Gentian Violet on Melanoma Cell Self-Renewal and Survival
- Author
-
Gianni Gerlini, Andrea Morandi, Silvia Pietrobono, Jack L. Arbiser, Lorenzo Borgognoni, Barbara Stecca, Sinforosa Gagliardi, and Paola Chiarugi
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Skin Neoplasms ,Cell Survival ,Population ,Apoptosis ,Cell Line, Tumor ,Cell Self Renewal ,Down-Regulation ,G2 Phase Cell Cycle Checkpoints ,Gentian Violet ,Humans ,Melanoma ,Mitochondria ,Neoplastic Stem Cells ,Phosphorylation ,SOXB1 Transcription Factors ,Dermatology ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,SOX2 ,medicine ,STAT3 ,education ,Molecular Biology ,Transcription factor ,education.field_of_study ,Cell growth ,Cell Biology ,medicine.disease ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Stem cell - Abstract
Human melanomas contain a population of tumor-initiating cells that are able to maintain the growth of the tumor. We previously showed that the embryonic transcription factor SOX2 is essential for self-renewal and tumorigenicity of human melanoma-initiating cells. However, targeting a transcription factor is still challenging. Gentian violet (GV) is a cationic triphenylmethane dye with potent antifungal and antibacterial activity. Recently, a combination therapy of imiquimod and GV has shown an inhibitory effect against melanoma metastases. Whether and how GV affects melanoma cells remains unknown. Here we show that GV represses melanoma stem cell self-renewal through inhibition of SOX2. Mechanistically, GV hinders EGFR activation and inhibits the signal transducer and activator of transcription-3 [(STAT3)/SOX2] axis. Importantly, we show that GV treatment decreases STAT3 phosphorylation at residue tyrosine 705, thus preventing the translocation of STAT3 into the nucleus and its binding to SOX2 promoter. In addition, GV affects melanoma cell growth by promoting mitochondrial apoptosis and G2 cell cycle arrest. This study shows that in melanoma, GV affects both the stem cell and the tumor bulk compartments, suggesting the potential use of GV in treating human melanoma alone or in combination with targeted therapy and/or immunotherapy.
- Published
- 2016
- Full Text
- View/download PDF
16. Targeting the Oncoprotein Smoothened by Small Molecules: Focus on Novel Acylguanidine Derivatives as Potent Smoothened Inhibitors
- Author
-
Silvia Pietrobono and Barbara Stecca
- Subjects
0301 basic medicine ,hedgehog ,medicine.medical_treatment ,Vismodegib ,Review ,Sonidegib ,Targeted therapy ,03 medical and health sciences ,chemistry.chemical_compound ,smoothened ,missense mutations ,drug-resistance ,Cancer stem cell ,medicine ,cancer ,Basal cell carcinoma ,Transcription factor ,Hedgehog ,lcsh:QH301-705.5 ,General Medicine ,medicine.disease ,targeted therapy ,small molecule inhibitors ,030104 developmental biology ,chemistry ,lcsh:Biology (General) ,acylguanidine derivative ,Cancer research ,Smoothened ,medicine.drug ,GLI - Abstract
Hedgehog-GLI (HH) signaling was originally identified as a critical morphogenetic pathway in embryonic development. Since its discovery, a multitude of studies have reported that HH signaling also plays key roles in a variety of cancer types and in maintaining tumor-initiating cells. Smoothened (SMO) is the main transducer of HH signaling, and in the last few years, it has emerged as a promising therapeutic target for anticancer therapy. Although vismodegib and sonidegib have demonstrated effectiveness for the treatment of basal cell carcinoma (BCC), their clinical use has been hampered by severe side effects, low selectivity against cancer stem cells, and the onset of mutation-driven drug resistance. Moreover, SMO antagonists are not effective in cancers where HH activation is due to mutations of pathway components downstream of SMO, or in the case of noncanonical, SMO-independent activation of the GLI transcription factors, the final mediators of HH signaling. Here, we review the current and rapidly expanding field of SMO small-molecule inhibitors in experimental and clinical settings, focusing on a class of acylguanidine derivatives. We also discuss various aspects of SMO, including mechanisms of resistance to SMO antagonists.
- Published
- 2018
17. Soft Tissue Sarcoma Cancer Stem Cells: An Overview
- Author
-
Katia C. Genadry, Silvia Pietrobono, Rossella Rota, and Corinne M. Linardic
- Subjects
0301 basic medicine ,cancer stem cells ,Cancer Research ,developmental pathways ,sarcoma ,Cell of origin ,Tumor initiation ,Disease ,Review ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,stemness ,Cancer stem cell ,medicine ,Epigenetics ,Soft tissue sarcoma ,Mesenchymal stem cell ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,soft tissue sarcoma ,Cancer research ,Sarcoma ,epigenetic plasticity - Abstract
Soft tissue sarcomas (STSs) are an uncommon group of solid tumors that can arise throughout the human lifespan. Despite their commonality as non-bony cancers that develop from mesenchymal cell precursors, they are heterogeneous in their genetic profiles, histology, and clinical features. This has made it difficult to identify a single target or therapy specific to STSs. And while there is no one cell of origin ascribed to all STSs, the cancer stem cell (CSC) principle-that a subpopulation of tumor cells possesses stem cell-like properties underlying tumor initiation, therapeutic resistance, disease recurrence, and metastasis-predicts that ultimately it should be possible to identify a feature common to all STSs that could function as a therapeutic Achilles' heel. Here we review the published evidence for CSCs in each of the most common STSs, then focus on the methods used to study CSCs, the developmental signaling pathways usurped by CSCs, and the epigenetic alterations critical for CSC identity that may be useful for further study of STS biology. We conclude with discussion of some challenges to the field and future directions.
- Published
- 2018
- Full Text
- View/download PDF
18. Targeted inhibition of Hedgehog-GLI signaling by novel acylguanidine derivatives inhibits melanoma cell growth by inducing replication stress and mitotic catastrophe
- Author
-
Elena Petricci, Sinforosa Gagliardi, Cosima Leone, Fabrizio Manetti, Francesca Dapporto, Mario Chiariello, Silvia Pietrobono, Massimo Valoti, Maurizio Taddei, Roberta Santini, David Colecchia, and Barbara Stecca
- Subjects
DNA Replication ,0301 basic medicine ,Cancer Research ,Cell cycle checkpoint ,Cell Survival ,Immunology ,Mice, Nude ,Mitosis ,Vismodegib ,Apoptosis ,Guanidines ,Zinc Finger Protein GLI1 ,Article ,Inhibitory Concentration 50 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Stress, Physiological ,GLI1 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Hedgehog Proteins ,lcsh:QH573-671 ,Cell Self Renewal ,Melanoma ,Mitotic catastrophe ,Cell Proliferation ,Cell Biology ,biology ,lcsh:Cytology ,Cell growth ,Chemistry ,Cell Cycle Checkpoints ,Smoothened Receptor ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Neoplastic Stem Cells ,biology.protein ,Cancer research ,Female ,Signal transduction ,Smoothened ,DNA Damage ,Signal Transduction ,medicine.drug - Abstract
Aberrant activation of the Hedgehog (HH) signaling is a critical driver in tumorigenesis. The Smoothened (SMO) receptor is one of the major upstream transducers of the HH pathway and a target for the development of anticancer agents. The SMO inhibitor Vismodegib (GDC-0449/Erivedge) has been approved for treatment of basal cell carcinoma. However, the emergence of resistance during Vismodegib treatment and the occurrence of numerous side effects limit its use. Our group has recently discovered and developed novel and potent SMO inhibitors based on acylguanidine or acylthiourea scaffolds. Here, we show that the two acylguanidine analogs, compound (1) and its novel fluoride derivative (2), strongly reduce growth and self-renewal of melanoma cells, inhibiting the level of the HH signaling target GLI1 in a dose-dependent manner. Both compounds induce apoptosis and DNA damage through the ATR/CHK1 axis. Mechanistically, they prevent G2 to M cell cycle transition, and induce signs of mitotic aberrations ultimately leading to mitotic catastrophe. In a melanoma xenograft mouse model, systemic treatment with 1 produced a remarkable inhibition of tumor growth without body weight loss in mice. Our data highlight a novel route for cell death induction by SMO inhibitors and support their use in therapeutic approaches for melanoma and, possibly, other types of cancer with active HH signaling.
- Published
- 2018
- Full Text
- View/download PDF
19. SOX2 regulates self-renewal and tumorigenicity of human melanoma-initiating cells
- Author
-
Valentina Montagnani, Roberta Santini, Massimo D'Amico, Silvia Pietrobono, Silvia Pandolfi, Barbara Stecca, Maria Cristina Vinci, Junia Y. Penachioni, and Lorenzo Borgognoni
- Subjects
Cancer Research ,Skin Neoplasms ,Carcinogenesis ,Mice, Nude ,Apoptosis ,Biology ,medicine.disease_cause ,Zinc Finger Protein GLI1 ,Mice ,SOX2 ,GLI1 ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Melanoma ,Cell Proliferation ,Cell growth ,SOXB1 Transcription Factors ,Aldehyde Dehydrogenase ,medicine.disease ,Embryonic stem cell ,Cell culture ,embryonic structures ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Female ,Original Article ,Stem cell ,Neoplasm Transplantation ,Signal Transduction ,Transcription Factors - Abstract
Melanoma is one of the most aggressive types of human cancer, characterized by enhanced heterogeneity and resistance to conventional therapy at advanced stages. We and others have previously shown that HEDGEHOG-GLI (HH-GLI) signaling is required for melanoma growth and for survival and expansion of melanoma-initiating cells (MICs). Recent reports indicate that HH-GLI signaling regulates a set of genes typically expressed in embryonic stem cells, including SOX2 (sex-determining region Y (SRY)-Box2). Here we address the function of SOX2 in human melanomas and MICs and its interaction with HH-GLI signaling. We find that SOX2 is highly expressed in melanoma stem cells. Knockdown of SOX2 sharply decreases self-renewal in melanoma spheres and in putative melanoma stem cells with high aldehyde dehydrogenase activity (ALDH(high)). Conversely, ectopic expression of SOX2 in melanoma cells enhances their self-renewal in vitro. SOX2 silencing also inhibits cell growth and induces apoptosis in melanoma cells. In addition, depletion of SOX2 progressively abrogates tumor growth and leads to a significant decrease in tumor-initiating capability of ALDH(high) MICs upon xenotransplantation, suggesting that SOX2 is required for tumor initiation and for continuous tumor growth. We show that SOX2 is regulated by HH signaling and that the transcription factors GLI1 and GLI2, the downstream effectors of HH-GLI signaling, bind to the proximal promoter region of SOX2 in primary melanoma cells. In functional studies, we find that SOX2 function is required for HH-induced melanoma cell growth and MIC self-renewal in vitro. Thus SOX2 is a critical factor for self-renewal and tumorigenicity of MICs and an important mediator of HH-GLI signaling in melanoma. These findings could provide the basis for novel therapeutic strategies based on the inhibition of SOX2 for the treatment of a subset of human melanomas.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.