114 results on '"Cristiano Simone"'
Search Results
2. The novel SMYD3 inhibitor EM127 impairs DNA repair response to chemotherapy-induced DNA damage and reverses cancer chemoresistance
- Author
-
Paola Sanese, Katia De Marco, Martina Lepore Signorile, Francesca La Rocca, Giovanna Forte, Marialaura Latrofa, Candida Fasano, Vittoria Disciglio, Elisabetta Di Nicola, Antonino Pantaleo, Giusy Bianco, Vito Spilotro, Claudia Ferroni, Matilde Tubertini, Nicoletta Labarile, Lucia De Marinis, Raffaele Armentano, Gianluigi Gigante, Valerio Lantone, Giuliano Lantone, Marina Naldi, Manuela Bartolini, Greta Varchi, Alberto Del Rio, Valentina Grossi, and Cristiano Simone
- Subjects
Cancer ,SMYD3 ,Drug resistance ,Chemosensitivity ,DNA damage response ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background SMYD3 has been found implicated in cancer progression. Its overexpression correlates with cancer growth and invasion, especially in gastrointestinal tumors. SMYD3 transactivates multiple oncogenic mechanisms, favoring cancer development. Moreover, it was recently shown that SMYD3 is required for DNA restoration by promoting homologous recombination (HR) repair. Methods In cellulo and in vivo models were employed to investigate the role of SMYD3 in cancer chemoresistance. Analyses of SMYD3-KO cells, drug-resistant cancer cell lines, patients’ residual gastric or rectal tumors that were resected after neoadjuvant therapy and mice models were performed. In addition, the novel SMYD3 covalent inhibitor EM127 was used to evaluate the impact of manipulating SMYD3 activity on the sensitization of cancer cell lines, tumorspheres and cancer murine models to chemotherapeutics (CHTs). Results Here we report that SMYD3 mediates cancer cell sensitivity to CHTs. Indeed, cancer cells lacking SMYD3 functions showed increased responsiveness to CHTs, while restoring its expression promoted chemoresistance. Specifically, SMYD3 is essential for the repair of CHT-induced double-strand breaks as it methylates the upstream sensor ATM and allows HR cascade propagation through CHK2 and p53 phosphorylation, thereby promoting cancer cell survival. SMYD3 inhibition with the novel compound EM127 showed a synergistic effect with CHTs in colorectal, gastric, and breast cancer cells, tumorspheres, and preclinical colorectal cancer models. Conclusions Overall, our results show that targeting SMYD3 may be an effective therapeutic strategy to overcome chemoresistance.
- Published
- 2024
- Full Text
- View/download PDF
3. Uncoupling p38α nuclear and cytoplasmic functions and identification of two p38α phosphorylation sites on β-catenin: implications for the Wnt signaling pathway in CRC models
- Author
-
Martina Lepore Signorile, Candida Fasano, Giovanna Forte, Katia De Marco, Paola Sanese, Vittoria Disciglio, Elisabetta Di Nicola, Antonino Pantaleo, Cristiano Simone, and Valentina Grossi
- Subjects
p38α ,Wnt/β-catenin pathway ,Colorectal cancer ,Chromatin-associated kinase ,Biotechnology ,TP248.13-248.65 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Activation of the Wnt pathway has been linked to colorectal cancer (CRC). Previous reports suggest that Wnt3a can activate p38. Besides, p38α feeds into the canonical Wnt/β-catenin pathway by inhibiting GSK3β through phosphorylation. Recently, we identified p38α as a new druggable member of β-catenin chromatin-associated kinase complexes in CRC. Methods The functional relationship between p38α and β-catenin was characterized in CRC cells, patient-derived CRC stem cells, patient-derived tumor intestinal organoids, and in vivo models (C57BL/6-APCMin/+ mice). The role of p38α in β-catenin transcriptional activity was assessed by pharmacological inhibition with ralimetinib. Results We used the GSK3β inhibitor TWS-119, which promotes the activation of Wnt signaling, to uncouple p38α nuclear/cytoplasmatic functions in the Wnt pathway. Upon GSK3β inhibition, nuclear p38α phosphorylates β-catenin at residues S111 and T112, allowing its binding to promoter regions of Wnt target genes and the activation of a transcriptional program implicated in cancer progression. If p38α is pharmacologically inhibited in addition to GSK3β, β-catenin is prevented from promoting target gene transcription, which is expected to impair carcinogenesis. Conclusions p38α seems to play a dual role as a member of the β-catenin destruction complex and as a β-catenin chromatin-associated kinase in CRC. This finding may help elucidate mechanisms contributing to human colon tumor pathogenesis and devise new strategies for personalized CRC treatment.
- Published
- 2023
- Full Text
- View/download PDF
4. Corrigendum to ‘Coinheritance of germline mutations in APC and MUTYH genes defines the clinical outcome of adenomatous polyposis syndromes’ [Gene Dis (10) (2023), 1187–1189]
- Author
-
Giovanna Forte, Filomena Cariola, Antonia Lucia Buonadonna, Anna Filomena Guglielmi, Andrea Manghisi, Katia De Marco, Valentina Grossi, Candida Fasano, Martina Lepore Signorile, Paola Sanese, Rosanna Bagnulo, Nicoletta Resta, Vittoria Disciglio, and Cristiano Simone
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2024
- Full Text
- View/download PDF
5. In Silico Deciphering of the Potential Impact of Variants of Uncertain Significance in Hereditary Colorectal Cancer Syndromes
- Author
-
Candida Fasano, Martina Lepore Signorile, Katia De Marco, Giovanna Forte, Vittoria Disciglio, Paola Sanese, Valentina Grossi, and Cristiano Simone
- Subjects
hereditary colorectal polyposis syndromes ,hereditary nonpolyposis colorectal cancer ,variants of uncertain significance ,in silico prediction tools ,protein stability ,protein functions ,Cytology ,QH573-671 - Abstract
Colorectal cancer (CRC) ranks third in terms of cancer incidence worldwide and is responsible for 8% of all deaths globally. Approximately 10% of CRC cases are caused by inherited pathogenic mutations in driver genes involved in pathways that are crucial for CRC tumorigenesis and progression. These hereditary mutations significantly increase the risk of initial benign polyps or adenomas developing into cancer. In recent years, the rapid and accurate sequencing of CRC-specific multigene panels by next-generation sequencing (NGS) technologies has enabled the identification of several recurrent pathogenic variants with established functional consequences. In parallel, rare genetic variants that are not characterized and are, therefore, called variants of uncertain significance (VUSs) have also been detected. The classification of VUSs is a challenging task because each amino acid has specific biochemical properties and uniquely contributes to the structural stability and functional activity of proteins. In this scenario, the ability to computationally predict the effect of a VUS is crucial. In particular, in silico prediction methods can provide useful insights to assess the potential impact of a VUS and support additional clinical evaluation. This approach can further benefit from recent advances in artificial intelligence-based technologies. In this review, we describe the main in silico prediction tools that can be used to evaluate the structural and functional impact of VUSs and provide examples of their application in the analysis of gene variants involved in hereditary CRC syndromes.
- Published
- 2024
- Full Text
- View/download PDF
6. Corrigendum to ‘A novel STK11 gene mutation (c.388dupG, p.Glu130Glyfs∗33) in a Peutz-Jeghers family and evidence of higher gastric cancer susceptibility associated with alterations in STK11 region aa 107–170’ [Gene Dis (9) (2022) 288–291]
- Author
-
Giovanna Forte, Filomena Cariola, Katia De Marco, Andrea Manghisi, Filomena Anna Guglielmi, Raffaele Armentano, Giuseppe Lippolis, Pietro Giorgio, Cristiano Simone, and Vittoria Disciglio
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2024
- Full Text
- View/download PDF
7. Coinheritance of germline mutations in APC and MUTYH genes defines the clinical outcome of adenomatous polyposis syndromes
- Author
-
Giovanna Forte, Filomena Cariola, Antonia Lucia Buonadonna, Anna Filomena Guglielmi, Andrea Manghisi, Katia De Marco, Valentina Grossi, Candida Fasano, Martina Lepore Signorile, Paola Sanese, Rosanna Bagnulo, Nicoletta Resta, Vittoria Disciglio, and Cristiano Simone
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2023
- Full Text
- View/download PDF
8. The chromatin remodeling factors EP300 and TRRAP are novel SMYD3 interactors involved in the emerging ‘nonmutational epigenetic reprogramming’ cancer hallmark
- Author
-
Candida Fasano, Martina Lepore Signorile, Elisabetta Di Nicola, Antonino Pantaleo, Giovanna Forte, Katia De Marco, Paola Sanese, Vittoria Disciglio, Valentina Grossi, and Cristiano Simone
- Subjects
Gastrointestinal cancer cell lines ,Hallmarks of cancer ,'nonmutational epigenetic reprogramming' cancer hallmark ,In silico tripeptide screening ,SMYD3 ,SMYD3 interactome ,Biotechnology ,TP248.13-248.65 - Abstract
SMDY3 is a histone-lysine N-methyltransferase involved in several oncogenic processes and is believed to play a major role in various cancer hallmarks. Recently, we identified ATM, BRCA2, CHK2, MTOR, BLM, MET, AMPK, and p130 as direct SMYD3 interactors by taking advantage of a library of rare tripeptides, which we first tested for their in vitro binding affinity to SMYD3 and then used as in silico probes to systematically search the human proteome. Here, we used this innovative approach to identify further SMYD3-interacting proteins involved in crucial cancer pathways and found that the chromatin remodeling factors EP300 and TRRAP interact directly with SMYD3, thus linking SMYD3 to the emerging ‘nonmutational epigenetic reprogramming’ cancer hallmark. Of note, we validated these interactions in gastrointestinal cancer cell lines, including HCT-116 cells, which harbor a C-terminal truncating mutation in EP300, suggesting that EP300 binds to SMYD3 via its N-terminal region. While additional studies are required to ascertain the functional mechanisms underlying these interactions and their significance, the identification of two novel SMYD3 interactors involved in epigenetic cancer hallmark pathways adds important pieces to the puzzle of how SMYD3 exerts its oncogenic role.
- Published
- 2023
- Full Text
- View/download PDF
9. A novel STK11 gene mutation (c.388dupG, p.Glu130Glyfs∗33) in a Peutz-Jeghers family and evidence of higher gastric cancer susceptibility associated with alterations in STK11 region aa 107-170
- Author
-
Giovanna Forte, Filomena Cariola, Katia De Marco, Andrea Manghisi, Filomena Anna Guglielmi, Raffaele Armentano, Giuseppe Lippolis, Pietro Giorgio, Cristiano Simone, and Vittoria Disciglio
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2022
- Full Text
- View/download PDF
10. CD90 is regulated by notch1 and hallmarks a more aggressive intrahepatic cholangiocarcinoma phenotype
- Author
-
Serena Mancarella, Grazia Serino, Isabella Gigante, Antonio Cigliano, Silvia Ribback, Paola Sanese, Valentina Grossi, Cristiano Simone, Raffaele Armentano, Matthias Evert, Diego F. Calvisi, and Gianluigi Giannelli
- Subjects
Intrahepatic Cholangiocarcinoma ,THY1/CD90 ,NOTCH pathway inhibition ,Xenograft models ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Intrahepatic Cholangiocarcinoma (iCCA) is characterized by a strong stromal reaction playing a role in tumor progression. Thymus cell antigen 1 (THY1), also called Cluster of Differentiation 90 (CD90), is a key regulator of cell–cell and cell–matrix interaction. In iCCA, CD90 has been reported to be associated with a poor prognosis. In an iCCA PDX model, we recently found that CD90 was downregulated in mice treated with the Notch γ-secretase inhibitor Crenigacestat. The study aims to investigate the role of CD90 in relation to the NOTCH pathway. Methods THY1/CD90 gene and protein expression was evaluated in human iCCA tissues and xenograft models by qRT-PCR, immunohistochemistry, and immunofluorescence. Notch1 inhibition was achieved by siRNA. THY1/CD90 functions were investigated in xenograft models built with HuCCT1 and KKU-M213 cell lines, engineered to overexpress or knockdown THY1, respectively. Results CD90 co-localized with EPCAM, showing its epithelial origin. In vitro, NOTCH1 silencing triggered HES1 and THY1 down-regulation. RBPJ, a critical transcriptional regulator of NOTCH signaling, exhibited putative binding sites on the THY1 promoter and bound to the latter, implying CD90 as a downstream NOTCH pathway effector. In vivo, Crenigacestat suppressed iCCA growth and reduced CD90 expression in the PDX model. In the xenograft model, Crenigacestat inhibited tumor growth of HuCCT1 cells transfected to overexpress CD90 and KKU-M213 cells constitutively expressing high levels of CD90, while not affecting the growth of HuCCT1 control cells and KKU-M213 depleted of CD90. In an iCCA cohort, patients with higher expression levels of NOTCH1/HES1/THY1 displayed a significantly shorter survival. Conclusions iCCA patients with higher NOTCH1/HES1/THY1 expression have the worst prognosis, but they are more likely to benefit from Notch signaling inhibition. These findings represent the scientific rationale for testing NOTCH1 inhibitors in clinical trials, taking the first step toward precision medicine for iCCA.
- Published
- 2022
- Full Text
- View/download PDF
11. Identifying novel SMYD3 interactors on the trail of cancer hallmarks
- Author
-
Candida Fasano, Martina Lepore Signorile, Katia De Marco, Giovanna Forte, Paola Sanese, Valentina Grossi, and Cristiano Simone
- Subjects
Gastrointestinal cancer cell lines ,Hallmarks of cancer ,In silico tripeptide screening ,SMYD3 ,SMYD3 interactome ,Biotechnology ,TP248.13-248.65 - Abstract
SMYD3 overexpression in several human cancers highlights its crucial role in carcinogenesis. Nonetheless, SMYD3 specific activity in cancer development and progression is currently under debate. Taking advantage of a library of rare tripeptides, which we first tested for their in vitro binding affinity to SMYD3 and then used as in silico probes, we recently identified BRCA2, ATM, and CHK2 as direct SMYD3 interactors. To gain insight into novel SMYD3 cancer-related roles, here we performed a comprehensive in silico analysis to cluster all potential SMYD3-interacting proteins identified by screening the human proteome for the previously tested tripeptides, based on their involvement in cancer hallmarks. Remarkably, we identified mTOR, BLM, MET, AMPK, and p130 as new SMYD3 interactors implicated in cancer processes. Further studies are needed to characterize the functional mechanisms underlying these interactions. Still, these findings could be useful to devise novel therapeutic strategies based on the combined inhibition of SMYD3 and its newly identified molecular partners. Of note, our in silico methodology may be useful to search for unidentified interactors of other proteins of interest.
- Published
- 2022
- Full Text
- View/download PDF
12. SMYD3 Modulates AMPK-mTOR Signaling Balance in Cancer Cell Response to DNA Damage
- Author
-
Martina Lepore Signorile, Paola Sanese, Elisabetta Di Nicola, Candida Fasano, Giovanna Forte, Katia De Marco, Vittoria Disciglio, Marialaura Latrofa, Antonino Pantaleo, Greta Varchi, Alberto Del Rio, Valentina Grossi, and Cristiano Simone
- Subjects
SMYD3 ,AMPK ,mTOR ,DNA damage ,gastrointestinal cancer ,breast cancer ,Cytology ,QH573-671 - Abstract
Cells respond to DNA damage by activating a complex array of signaling networks, which include the AMPK and mTOR pathways. After DNA double-strand breakage, ATM, a core component of the DNA repair system, activates the AMPK-TSC2 pathway, leading to the inhibition of the mTOR cascade. Recently, we showed that both AMPK and mTOR interact with SMYD3, a methyltransferase involved in DNA damage response. In this study, through extensive molecular characterization of gastrointestinal and breast cancer cells, we found that SMYD3 is part of a multiprotein complex that is involved in DNA damage response and also comprises AMPK and mTOR. In particular, upon exposure to the double-strand break-inducing agent neocarzinostatin, SMYD3 pharmacological inhibition suppressed AMPK cascade activation and thereby promoted the mTOR pathway, which reveals the central role played by SMYD3 in the modulation of AMPK-mTOR signaling balance during cancer cell response to DNA double-strand breaks. Moreover, we found that SMYD3 can methylate AMPK at the evolutionarily conserved residues Lys411 and Lys424. Overall, our data revealed that SMYD3 can act as a bridge between the AMPK and mTOR pathways upon neocarzinostatin-induced DNA damage in gastrointestinal and breast cancer cells.
- Published
- 2023
- Full Text
- View/download PDF
13. SMYD3 Modulates the HGF/MET Signaling Pathway in Gastric Cancer
- Author
-
Katia De Marco, Martina Lepore Signorile, Elisabetta Di Nicola, Paola Sanese, Candida Fasano, Giovanna Forte, Vittoria Disciglio, Antonino Pantaleo, Greta Varchi, Alberto Del Rio, Valentina Grossi, and Cristiano Simone
- Subjects
MET ,SMYD3 ,gastric cancer ,stemness activity ,Cytology ,QH573-671 - Abstract
Gastric cancer (GC) is the third most deadly cancer worldwide. Considerable efforts have been made to find targetable drivers in order to improve patient outcomes. MET is one of the most important factors involved in GC initiation and progression as it plays a major role in GC invasiveness and is related to cancer stemness. Unfortunately, treatment strategies targeting MET are still limited, with a proportion of patients responding to therapy but later developing resistance. Here, we showed that MET is a molecular partner of the SMYD3 methyltransferase in GC cells. Moreover, we found that SMYD3 pharmacological inhibition affects the HGF/MET downstream signaling pathway. Extensive cellular analyses in GC models indicated that EM127, a novel active site-selective covalent SMYD3 inhibitor, can be used as part of a synergistic approach with MET inhibitors in order to enhance the targeting of the HGF/MET pathway. Importantly, our data were confirmed in a 3D GC cell culture system, which was used as a surrogate to evaluate stemness characteristics. Our findings identify SMYD3 as a promising therapeutic target to impair the HGF/MET pathway for the treatment of GC.
- Published
- 2023
- Full Text
- View/download PDF
14. Pharmacological targeting of the novel β-catenin chromatin-associated kinase p38α in colorectal cancer stem cell tumorspheres and organoids
- Author
-
Martina Lepore Signorile, Valentina Grossi, Simone Di Franco, Giovanna Forte, Vittoria Disciglio, Candida Fasano, Paola Sanese, Katia De Marco, Francesco Claudio Susca, Laura Rosa Mangiapane, Annalisa Nicotra, Gabriella Di Carlo, Francesco Dituri, Gianluigi Giannelli, Giuseppe Ingravallo, Gianluca Canettieri, Giorgio Stassi, and Cristiano Simone
- Subjects
Cytology ,QH573-671 - Abstract
Abstract The prognosis of locally advanced colorectal cancer (CRC) is currently unsatisfactory. This is mainly due to drug resistance, recurrence, and subsequent metastatic dissemination, which are sustained by the cancer stem cell (CSC) population. The main driver of the CSC gene expression program is Wnt signaling, and previous reports indicate that Wnt3a can activate p38 MAPK. Besides, p38 was shown to feed into the canonical Wnt/β-catenin pathway. Here we show that patient-derived locally advanced CRC stem cells (CRC-SCs) are characterized by increased expression of p38α and are “addicted” to its kinase activity. Of note, we found that stage III CRC patients with high p38α levels display reduced disease-free and progression-free survival. Extensive molecular analysis in patient-derived CRC-SC tumorspheres and APCMin/+ mice intestinal organoids revealed that p38α acts as a β-catenin chromatin-associated kinase required for the regulation of a signaling platform involved in tumor proliferation, metastatic dissemination, and chemoresistance in these CRC model systems. In particular, the p38α kinase inhibitor ralimetinib, which has already entered clinical trials, promoted sensitization of patient-derived CRC-SCs to chemotherapeutic agents commonly used for CRC treatment and showed a synthetic lethality effect when used in combination with the MEK1 inhibitor trametinib. Taken together, these results suggest that p38α may be targeted in CSCs to devise new personalized CRC treatment strategies.
- Published
- 2021
- Full Text
- View/download PDF
15. Correction: CD90 is regulated by notch1 and hallmarks a more aggressive intrahepatic cholangiocarcinoma phenotype
- Author
-
Serena Mancarella, Grazia Serino, Isabella Gigante, Antonio Cigliano, Silvia Ribback, Paola Sanese, Valentina Grossi, Cristiano Simone, Raffaele Armentano, Matthias Evert, Diego F. Calvisi, and Gianluigi Giannelli
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
16. Short Linear Motifs in Colorectal Cancer Interactome and Tumorigenesis
- Author
-
Candida Fasano, Valentina Grossi, Giovanna Forte, and Cristiano Simone
- Subjects
short linear motifs ,protein–protein interactions ,SLiM-based small molecules ,colorectal cancer interactome ,targeted therapy ,cancer driver protein interactome ,Cytology ,QH573-671 - Abstract
Colorectal tumorigenesis is driven by alterations in genes and proteins responsible for cancer initiation, progression, and invasion. This multistage process is based on a dense network of protein–protein interactions (PPIs) that become dysregulated as a result of changes in various cell signaling effectors. PPIs in signaling and regulatory networks are known to be mediated by short linear motifs (SLiMs), which are conserved contiguous regions of 3–10 amino acids within interacting protein domains. SLiMs are the minimum sequences required for modulating cellular PPI networks. Thus, several in silico approaches have been developed to predict and analyze SLiM-mediated PPIs. In this review, we focus on emerging evidence supporting a crucial role for SLiMs in driver pathways that are disrupted in colorectal cancer (CRC) tumorigenesis and related PPI network alterations. As a result, SLiMs, along with short peptides, are attracting the interest of researchers to devise small molecules amenable to be used as novel anti-CRC targeted therapies. Overall, the characterization of SLiMs mediating crucial PPIs in CRC may foster the development of more specific combined pharmacological approaches.
- Published
- 2022
- Full Text
- View/download PDF
17. Targeting SMYD3 to Sensitize Homologous Recombination-Proficient Tumors to PARP-Mediated Synthetic Lethality
- Author
-
Paola Sanese, Candida Fasano, Giacomo Buscemi, Cinzia Bottino, Silvia Corbetta, Edoardo Fabini, Valentina Silvestri, Virginia Valentini, Vittoria Disciglio, Giovanna Forte, Martina Lepore Signorile, Katia De Marco, Stefania Bertora, Valentina Grossi, Ummu Guven, Natale Porta, Valeria Di Maio, Elisabetta Manoni, Gianluigi Giannelli, Manuela Bartolini, Alberto Del Rio, Giuseppina Caretti, Laura Ottini, and Cristiano Simone
- Subjects
Molecular Biology ,Cell Biology ,Cancer ,Science - Abstract
Summary: SMYD3 is frequently overexpressed in a wide variety of cancers. Indeed, its inactivation reduces tumor growth in preclinical in vivo animal models. However, extensive characterization in vitro failed to clarify SMYD3 function in cancer cells, although confirming its importance in carcinogenesis. Taking advantage of a SMYD3 mutant variant identified in a high-risk breast cancer family, here we show that SMYD3 phosphorylation by ATM enables the formation of a multiprotein complex including ATM, SMYD3, CHK2, and BRCA2, which is required for the final loading of RAD51 at DNA double-strand break sites and completion of homologous recombination (HR). Remarkably, SMYD3 pharmacological inhibition sensitizes HR-proficient cancer cells to PARP inhibitors, thereby extending the potential of the synthetic lethality approach in human tumors.
- Published
- 2020
- Full Text
- View/download PDF
18. FOXO3 on the Road to Longevity: Lessons From SNPs and Chromatin Hubs
- Author
-
Paola Sanese, Giovanna Forte, Vittoria Disciglio, Valentina Grossi, and Cristiano Simone
- Subjects
Biotechnology ,TP248.13-248.65 - Abstract
Health span is driven by a precise interplay between genes and the environment. Cell response to environmental cues is mediated by signaling cascades and genetic variants that affect gene expression by regulating chromatin plasticity. Indeed, they can promote the interaction of promoters with regulatory elements by forming active chromatin hubs.FOXO3 encodes a transcription factor with a strong impact on aging and age-related phenotypes, as it regulates stress response, therefore affecting lifespan. A significant association has been shown between human longevity and several FOXO3 variants located in intron 2. This haplotype block forms a putative aging chromatin hub in which FOXO3 has a central role, as it modulates the physical connection and activity of neighboring genes involved in age-related processes.Here we describe the role of FOXO3 and its single-nucleotide polymorphisms (SNPs) in healthy aging, with a focus on the enhancer region encompassing the SNP rs2802292, which upregulates FOXO3 expression and can promote the activity of the aging hub in response to different stress stimuli. FOXO3 protective effect on lifespan may be due to the accessibility of this region to transcription factors promoting its expression. This could in part explain the differences in FOXO3 association with longevity between genders, as its activity in females may be modulated by estrogens through estrogen receptor response elements located in the rs2802292-encompassing region. Altogether, the molecular mechanisms described here may help establish whether the rs2802292 SNP can be taken advantage of in predictive medicine and define the potential of targeting FOXO3 for age-related diseases. Keywords: FOXO3, Longevity, Aging, SNP, Chromatin hub
- Published
- 2019
- Full Text
- View/download PDF
19. Germline pathogenic variant in PIK3CA leading to symmetrical overgrowth with marked macrocephaly and mild global developmental delay
- Author
-
Marcella Zollino, Carlotta Ranieri, Valentina Grossi, Chiara Leoni, Serena Lattante, Daniela Mazzà, Cristiano Simone, and Nicoletta Resta
- Subjects
germline variant ,macrocephaly ,overgrowth ,PI3K/AKT/mTOR pathway ,PIK3CA ,Genetics ,QH426-470 - Abstract
Abstract Background Activating pathogenic variants in PIK3CA gene usually occur at a mosaic status and underlie a variety of segmental overgrowth phenotypes. Germline variants in PIK3CA have been rarely reported, described in a total of 12 patients with macrocephaly to date. Clinical and prognostic features of these germline variants have not been described in detail yet. Methods Targeted deep sequencing by custom panel of the 21 genes involved in the PI3K/AKT/mTOR pathway was performed in a 13‐year‐old boy with macrocephaly and physical overgrowth. PI3K/AKT/mTOR pathway analysis was performed in fibroblasts by Western blot. The effects of miransertib (AKT inhibitor) and rapamycin (mTOR inhibitor) were assessed. Results A de novo pathogenic variant (c.1090G>C; p.Gly364Arg) in PIK3CA gene was detected in a non‐mosaic status in peripheral blood cells, buccal smears, and skin fibroblasts. Increased levels of phosphorylated AKT residues were observed in fibroblasts, rescued by miransertib. Conclusion Germline variants in PIK3CA are associated to a mild phenotype characterized by overgrowth, severe macrocephaly, mild intellectual disability, and few dysmorphic features. Investigations of PI3K/AKT/mTOR pathway should be performed in patients with severe macrocephaly and unspecific physical overgrowth. Longitudinal studies to assess prognosis and cancer predisposition are recommended.
- Published
- 2019
- Full Text
- View/download PDF
20. FOXO3a from the Nucleus to the Mitochondria: A Round Trip in Cellular Stress Response
- Author
-
Candida Fasano, Vittoria Disciglio, Stefania Bertora, Martina Lepore Signorile, and Cristiano Simone
- Subjects
FOXO3a ,transcription factors ,cellular homeostasis ,stress response ,nuclear/mitochondrial crosstalk ,Cytology ,QH573-671 - Abstract
Cellular stress response is a universal mechanism that ensures the survival or negative selection of cells in challenging conditions. The transcription factor Forkhead box protein O3 (FOXO3a) is a core regulator of cellular homeostasis, stress response, and longevity since it can modulate a variety of stress responses upon nutrient shortage, oxidative stress, hypoxia, heat shock, and DNA damage. FOXO3a activity is regulated by post-translational modifications that drive its shuttling between different cellular compartments, thereby determining its inactivation (cytoplasm) or activation (nucleus and mitochondria). Depending on the stress stimulus and subcellular context, activated FOXO3a can induce specific sets of nuclear genes, including cell cycle inhibitors, pro-apoptotic genes, reactive oxygen species (ROS) scavengers, autophagy effectors, gluconeogenic enzymes, and others. On the other hand, upon glucose restriction, 5′-AMP-activated protein kinase (AMPK) and mitogen activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) -dependent FOXO3a mitochondrial translocation allows the transcription of oxidative phosphorylation (OXPHOS) genes, restoring cellular ATP levels, while in cancer cells, mitochondrial FOXO3a mediates survival upon genotoxic stress induced by chemotherapy. Interestingly, these target genes and their related pathways are diverse and sometimes antagonistic, suggesting that FOXO3a is an adaptable player in the dynamic homeostasis of normal and stressed cells. In this review, we describe the multiple roles of FOXO3a in cellular stress response, with a focus on both its nuclear and mitochondrial functions.
- Published
- 2019
- Full Text
- View/download PDF
21. Updates from the Intestinal Front Line: Autophagic Weapons against Inflammation and Cancer
- Author
-
Cristiano Simone, Alessia Peserico, Federica Madia, and Valentina Grossi
- Subjects
autophagy ,colorectal cancer ,inflammation ,chemotherapy ,p38a ,Cytology ,QH573-671 - Abstract
The intestine lies at the interface between the organism and its environment and responds to infection/inflammation in a multi-leveled manner, potentially leading to chronic inflammatory pathologies and cancer formation. Indeed, the immune response at the intestinal epithelium has been found to be involved in the origin and development of colorectal cancer, which is the third most commonly diagnosed neoplastic disease. Among the mechanisms induced upon inflammation, autophagy appears as a defensive strategy for the clearance of invading microbes and intracellular waste components. Autophagy has also been found to play an important role in colorectal cancer, where it seems to have a pro-survival or pro-death function depending on the stage of the neoplastic process. In this paper we discuss the dual role of autophagy in colorectal cancer and review evidence showing that modulation of autophagy affects the immune response and cancer biology. The study of key players involved in autophagy might contribute to the design of new approaches for colorectal cancer, consisting in combined therapies capable of modifying cancer-specific metabolism rather than simply evoking a generic apoptotic and/or autophagic response, thus enhancing the efficacy of currently used drugs and treatments.
- Published
- 2012
- Full Text
- View/download PDF
22. Clinical and functional characterization of a novel mutation in lamin a/c gene in a multigenerational family with arrhythmogenic cardiac laminopathy.
- Author
-
Cinzia Forleo, Monica Carmosino, Nicoletta Resta, Alessandra Rampazzo, Rosanna Valecce, Sandro Sorrentino, Massimo Iacoviello, Francesco Pisani, Giuseppe Procino, Andrea Gerbino, Arnaldo Scardapane, Cristiano Simone, Martina Calore, Silvia Torretta, Maria Svelto, and Stefano Favale
- Subjects
Medicine ,Science - Abstract
Mutations in the lamin A/C gene (LMNA) were associated with dilated cardiomyopathy (DCM) and, recently, were related to severe forms of arrhythmogenic right ventricular cardiomyopathy (ARVC). Both genetic and phenotypic overlap between DCM and ARVC was observed; molecular pathomechanisms leading to the cardiac phenotypes caused by LMNA mutations are not yet fully elucidated. This study involved a large Italian family, spanning 4 generations, with arrhythmogenic cardiomyopathy of different phenotypes, including ARVC, DCM, system conduction defects, ventricular arrhythmias, and sudden cardiac death. Mutation screening of LMNA and ARVC-related genes PKP2, DSP, DSG2, DSC2, JUP, and CTNNA3 was performed. We identified a novel heterozygous mutation (c.418_438dup) in LMNA gene exon 2, occurring in a highly conserved protein domain across several species. This newly identified variant was not found in 250 ethnically-matched control subjects. Genotype-phenotype correlation studies suggested a co-segregation of the LMNA mutation with the disease phenotype and an incomplete and age-related penetrance. Based on clinical, pedigree, and molecular genetic data, this mutation was considered likely disease-causing. To clarify its potential pathophysiologic impact, functional characterization of this LMNA mutant was performed in cultured cardiomyocytes expressing EGFP-tagged wild-type and mutated LMNA constructs, and indicated an increased nuclear envelope fragility, leading to stress-induced apoptosis as the main pathogenetic mechanism. This study further expands the role of the LMNA gene in the pathogenesis of cardiac laminopathies, suggesting that LMNA should be included in mutation screening of patients with suspected arrhythmogenic cardiomyopathy, particularly when they have ECG evidence for conduction defects. The combination of clinical, genetic, and functional data contribute insights into the pathogenesis of this form of life-threatening arrhythmogenic cardiac laminopathy.
- Published
- 2015
- Full Text
- View/download PDF
23. Molecular and Functional Characterization of Three Different Postzygotic Mutations in PIK3CA-Related Overgrowth Spectrum (PROS) Patients: Effects on PI3K/AKT/mTOR Signaling and Sensitivity to PIK3 Inhibitors.
- Author
-
Daria C Loconte, Valentina Grossi, Cristina Bozzao, Giovanna Forte, Rosanna Bagnulo, Alessandro Stella, Patrizia Lastella, Mario Cutrone, Francesco Benedicenti, Francesco C Susca, Margherita Patruno, Dora Varvara, Aldo Germani, Luciana Chessa, Nicola Laforgia, Romano Tenconi, Cristiano Simone, and Nicoletta Resta
- Subjects
Medicine ,Science - Abstract
PIK3CA-related overgrowth spectrum (PROS) include a group of disorders that affect only the terminal portion of a limb, such as type I macrodactyly, and conditions like fibroadipose overgrowth (FAO), megalencephaly-capillary malformation (MCAP) syndrome, congenital lipomatous asymmetric overgrowth of the trunk, lymphatic, capillary, venous, and combined-type vascular malformations, epidermal nevi, skeletal and spinal anomalies (CLOVES) syndrome and Hemihyperplasia Multiple Lipomatosis (HHML). Heterozygous postzygotic PIK3CA mutations are frequently identified in these syndromes, while timing and tissue specificity of the mutational event are likely responsible for the extreme phenotypic variability observed.We carried out a combination of Sanger sequencing and targeted deep sequencing of genes involved in the PI3K/AKT/mTOR pathway in three patients (1 MCAP and 2 FAO) to identify causative mutations, and performed immunoblot analyses to assay the phosphorylation status of AKT and P70S6K in affected dermal fibroblasts. In addition, we evaluated their ability to grow in the absence of serum and their response to the PI3K inhibitors wortmannin and LY294002 in vitro.Our data indicate that patients' cells showed constitutive activation of the PI3K/Akt pathway. Of note, PI3K pharmacological blockade resulted in a significant reduction of the proliferation rate in culture, suggesting that inhibition of PI3K might prove beneficial in future therapies for PROS patients.
- Published
- 2015
- Full Text
- View/download PDF
24. c-MYC Protein Stability Is Sustained by MAPKs in Colorectal Cancer
- Author
-
Martina Lepore Signorile, Valentina Grossi, Candida Fasano, Giovanna Forte, Vittoria Disciglio, Paola Sanese, Katia De Marco, Francesca La Rocca, Raffaele Armentano, Anna Valentini, Gianluigi Giannelli, and Cristiano Simone
- Subjects
Cancer Research ,Oncology ,p38α ,MAPKs ,c-MYC ,protein stabilization ,kinase activity ,CRC - Abstract
c-MYC is one of the most important factors involved in colorectal cancer (CRC) initiation and progression; indeed, it is found to be upregulated in up to 80% of sporadic cases. During colorectal carcinogenesis, c-MYC is maintained upregulated through β-catenin-mediated transcriptional activation and ERK-mediated post-translational stabilization. Our data demonstrate that p38α, a kinase involved in CRC metabolism and survival, contributes to c-Myc protein stability. Moreover, we show that p38α, like ERK, stabilizes c-MYC protein levels by preventing its ubiquitination. Of note, we found that p38α phosphorylates c-MYC and interacts with it both in vitro and in cellulo. Extensive molecular analyses in the cellular and in vivo models revealed that the p38α kinase inhibitors, SB202190 and ralimetinib, affect c-MYC protein levels. Ralimetinib also exhibited a synthetic lethality effect when used in combination with the MEK1 inhibitor trametinib. Overall, our findings identify p38α as a promising therapeutic target, acting directly on c-MYC, with potential implications for countering c-MYC-mediated CRC proliferation, metastatic dissemination, and chemoresistance.
- Published
- 2022
- Full Text
- View/download PDF
25. Discovery of an Allosteric Ligand Binding Site in SMYD3 Lysine Methyltransferase
- Author
-
Doreen Dobritzsch, Daniela Cederfeldt, Daniele Tedesco, Martin J. Talu, Alberto Del Rio, Xavier Barril, Marina Naldi, Giovanna Forte, Paola Sanese, Edoardo Fabini, Vladimir O. Talibov, Elisabetta Manoni, Martina Lepore Signorile, Manuela Bartolini, Moira Rachman, Edward A. FitzGerald, U. Helena Danielson, Cristiano Simone, Filip Mihalic, Talibov, Vladimir O, Fabini, Edoardo, FitzGerald, Edward A, Tedesco, Daniele, Cederfeldt, Daniela, Talu, Martin J, Rachman, Moira M, Mihalic, Filip, Manoni, Elisabetta, Naldi, Marina, Sanese, Paola, Forte, Giovanna, Lepore Signorile, Martina, Barril, Xavier, Simone, Cristiano, Bartolini, Manuela, Dobritzsch, Doreen, Del Rio, Alberto, and Danielson, U Helena
- Subjects
Methyltransferase ,Lysine ,Drug Evaluation, Preclinical ,Druggability ,epigenetic enzyme ,Plasma protein binding ,Ligands ,01 natural sciences ,Biochemistry ,Piperidines ,biophysical methods ,chemistry.chemical_classification ,SMYD3 ,Full Paper ,biology ,epigenetic enzymes ,Stereoisomerism ,Full Papers ,Ligand (biochemistry) ,Hsp90 ,Biofysik ,3. Good health ,Molecular Medicine ,Allosteric Site ,Protein Binding ,Allosteric regulation ,Biophysics ,Molecular Dynamics Simulation ,biophysical method ,010402 general chemistry ,Cell Line, Tumor ,Surface plasmon resonance ,Humans ,HSP90 Heat-Shock Proteins ,diperodon ,Molecular Biology ,Binding Sites ,Lysine methyl transferase ,010405 organic chemistry ,screening ,Organic Chemistry ,Histone-Lysine N-Methyltransferase ,0104 chemical sciences ,SMYD3 biology ,Kinetics ,Enzyme ,chemistry ,biology.protein ,ligand discovery - Abstract
SMYD3 is a multifunctional epigenetic enzyme with lysine methyltransferase activity and various interaction partners. It is implicated in the pathophysiology of cancers but with an unclear mechanism. To discover tool compounds for clarifying its biochemistry and potential as a therapeutic target, a set of drug‐like compounds was screened in a biosensor‐based competition assay. Diperodon was identified as an allosteric ligand; its R and S enantiomers were isolated, and their affinities to SMYD3 were determined (K D=42 and 84 μM, respectively). Co‐crystallization revealed that both enantiomers bind to a previously unidentified allosteric site in the C‐terminal protein binding domain, consistent with its weak inhibitory effect. No competition between diperodon and HSP90 (a known SMYD3 interaction partner) was observed although SMYD3–HSP90 binding was confirmed (K D=13 μM). Diperodon clearly represents a novel starting point for the design of tool compounds interacting with a druggable allosteric site, suitable for the exploration of noncatalytic SMYD3 functions and therapeutics with new mechanisms of action., From a distance: SMYD3 lysine methyltransferase is an epigenetic enzyme with multiple cellular functions and the ability to recognize broad range of substrates from histones to cytosolic proteins. An SPR biosensors‐based biophysical strategy for screening SMYD3 ligands was developed and revealed an allosteric binding site. Subsequent crystallographic studies provided a structural description of the novel binding site of SMYD3.
- Published
- 2021
- Full Text
- View/download PDF
26. Colorectal Cancer Chemoprevention: A Dream Coming True?
- Author
-
Martina Lepore Signorile, Valentina Grossi, Candida Fasano, and Cristiano Simone
- Subjects
Inorganic Chemistry ,Organic Chemistry ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
Colorectal cancer (CRC) is one of the deadliest forms of cancer worldwide. CRC development occurs mainly through the adenoma-carcinoma sequence, which can last decades, giving the opportunity for primary prevention and early detection. CRC prevention involves different approaches, ranging from fecal occult blood testing and colonoscopy screening to chemoprevention. In this review, we discuss the main findings gathered in the field of CRC chemoprevention, focusing on different target populations and on various precancerous lesions that can be used as efficacy evaluation endpoints for chemoprevention. The ideal chemopreventive agent should be well tolerated and easy to administer, with low side effects. Moreover, it should be readily available at a low cost. These properties are crucial because these compounds are meant to be used for a long time in populations with different CRC risk profiles. Several agents have been investigated so far, some of which are currently used in clinical practice. However, further investigation is needed to devise a comprehensive and effective chemoprevention strategy for CRC.
- Published
- 2023
- Full Text
- View/download PDF
27. First Description of a Frameshift PAX5 Germline Variant in Two Siblings with B-Cell Precursor Acute Lymphoblastic Leukemia
- Author
-
Laura Rachele Bettini, Grazia Fazio, Claudia Saitta, Sonia Palamini, Chiara Buracchi, Stefano Rebellato, Nicola Santoro, Cristiano Simone, Andrea Biondi, and Giovanni Cazzaniga
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
28. Discovery of the 4-aminopiperidine-based compound EM127 for the site-specific covalent inhibition of SMYD3
- Author
-
Marco Daniele Parenti, Marina Naldi, Elisabetta Manoni, Edoardo Fabini, Daniela Cederfelt, Vladimir O. Talibov, Valeria Gressani, Ummu Guven, Valentina Grossi, Candida Fasano, Paola Sanese, Katia De Marco, Alexander A. Shtil, Alexander V. Kurkin, Andrea Altieri, U. Helena Danielson, Giuseppina Caretti, Cristiano Simone, Greta Varchi, Manuela Bartolini, and Alberto Del Rio
- Subjects
Pharmacology ,Histones ,Cell Line, Tumor ,Organic Chemistry ,Drug Discovery ,Humans ,Female ,Breast Neoplasms ,General Medicine ,Histone-Lysine N-Methyltransferase - Abstract
Recent findings support the hypothesis that inhibition of SMYD3 methyltransferase may be a therapeutic avenue for some of the deadliest cancer types. Herein, active site-selective covalent SMYD3 inhibitors were designed by introducing an appropriate reactive cysteine trap into reversible first-generation SMYD3 inhibitors. The 4-aminopiperidine derivative EM127 (11C) bearing a 2-chloroethanoyl group as reactive warhead showed selectivity for Cys186, located in the substrate/histone binding pocket. Selectivity towards Cys186 was retained even at high inhibitor/enzyme ratio, as shown by mass spectrometry. The mode of interaction with the SMYD3 substrate/histone binding pocket was revealed by crystallographic studies. In enzymatic assays, 11C showed a stronger SMYD3 inhibitory effect compared to the reference inhibitor EPZ031686. Remarkably, 11C attenuated the proliferation of MDA-MB-231 breast cancer cell line at the same low micromolar range of concentrations that reduced SMYD3 mediated ERK signaling in HCT116 colorectal cancer and MDA-MB-231 breast cancer cells. Furthermore, 11C (5 μM) strongly decreased the steady-state mRNA levels of genes important for tumor biology such as cyclin dependent kinase 2, c-MET, N-cadherin and fibronectin 1, all known to be regulated, at least in part, by SMYD3. Thus, 11C is as a first example of second generation SMYD3 inhibitors; this agent represents a covalent and a site specific SMYD3 binder capable of potent and prolonged attenuation of methyltransferase activity.
- Published
- 2022
29. Identification and Somatic Characterization of the Germline
- Author
-
Vittoria, Disciglio, Paola, Sanese, Candida, Fasano, Claudio, Lotesoriere, Anna Maria, Valentini, Giovanna, Forte, Martina, Lepore Signorile, Katia, De Marco, Valentina, Grossi, Ivan, Lolli, Filomena, Cariola, and Cristiano, Simone
- Subjects
Germ Cells ,Esophageal Neoplasms ,PTEN Phosphohydrolase ,Humans ,Breast Neoplasms ,Female ,Adenocarcinoma ,Hamartoma Syndrome, Multiple - Abstract
Genetic variants located in non-coding regions can affect processes that regulate protein expression, functionally contributing to human disease. Germline heterozygous mutations in the non-coding region of the
- Published
- 2021
30. Playing on the Dark Side: SMYD3 Acts as a Cancer Genome Keeper in Gastrointestinal Malignancies
- Author
-
Cristiano Simone, Paola Sanese, and Candida Fasano
- Subjects
Cancer Research ,SMYD3 ,Cell cycle checkpoint ,gastrointestinal tumors ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Synthetic lethality ,Review ,Cell cycle ,Biology ,medicine.disease_cause ,medicine.disease ,DNA damage response ,synthetic lethality ,homologous recombination repair ,PARP1 ,Oncology ,Tumor progression ,Cancer cell ,medicine ,Cancer research ,SMYD3 inhibition ,Carcinogenesis ,cell cycle checkpoints ,RC254-282 - Abstract
Simple Summary The activity of SMYD3 in promoting carcinogenesis is currently under debate. Growing evidence seems to confirm that SMYD3 overexpression correlates with poor prognosis, cancer growth and invasion, especially in gastrointestinal tumors. In this review, we dissect the emerging role played by SMYD3 in the regulation of cell cycle and DNA damage response by promoting homologous recombination (HR) repair and hence cancer cell genomic stability. Considering the crucial role of PARP1 in other DNA repair mechanisms, we also discuss a recently evaluated synthetic lethality approach based on the combined use of SMYD3 and PARP inhibitors. Interestingly, a significant proportion of HR-proficient gastrointestinal tumors expressing high levels of SMYD3 from the PanCanAtlas dataset seem to be eligible for this innovative strategy. This promising approach could be taken advantage of for therapeutic applications of SMYD3 inhibitors in cancer treatment. Abstract The SMYD3 methyltransferase has been found overexpressed in several types of cancers of the gastrointestinal (GI) tract. While high levels of SMYD3 have been positively correlated with cancer progression in cellular and advanced mice models, suggesting it as a potential risk and prognosis factor, its activity seems dispensable for autonomous in vitro cancer cell proliferation. Here, we present an in-depth analysis of SMYD3 functional role in the regulation of GI cancer progression. We first describe the oncogenic activity of SMYD3 as a transcriptional activator of genes involved in tumorigenesis, cancer development and transformation and as a co-regulator of key cancer-related pathways. Then, we dissect its role in orchestrating cell cycle regulation and DNA damage response (DDR) to genotoxic stress by promoting homologous recombination (HR) repair, thereby sustaining cancer cell genomic stability and tumor progression. Based on this evidence and on the involvement of PARP1 in other DDR mechanisms, we also outline a synthetic lethality approach consisting of the combined use of SMYD3 and PARP inhibitors, which recently showed promising therapeutic potential in HR-proficient GI tumors expressing high levels of SMYD3. Overall, these findings identify SMYD3 as a promising target for drug discovery.
- Published
- 2021
31. Gastric polyposis and desmoid tumours as a new familial adenomatous polyposis clinical variant associated with APC mutation at the extreme 3′-end
- Author
-
Alessandro Stella, Martina Lepore Signorile, Ivan Lolli, Valentina Grossi, Candida Fasano, Claudio Lotesoriere, Filomena Cariola, Nicoletta Resta, Vittoria Disciglio, Cristiano Simone, Giovanna Forte, and Paola Sanese
- Subjects
Adult ,Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Adenomatous Polyposis Coli Protein ,medicine.disease_cause ,Germline ,Familial adenomatous polyposis ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Stomach Neoplasms ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Gastric Polyposis ,Gene ,Germ-Line Mutation ,Genetics (clinical) ,Mutation ,gastric polyposis ,business.industry ,Gastric Polyposis and Desmoid FAP ,Genotype-Phenotype Correlations ,FAP ,Middle Aged ,medicine.disease ,Phenotype ,APC C-terminus ,digestive system diseases ,Pedigree ,APC ,Fibromatosis, Aggressive ,030104 developmental biology ,Adenomatous Polyposis Coli ,030220 oncology & carcinogenesis ,Familial Adenomatous Polyposis ,Cancer research ,Female ,Desmoid tumours ,business - Abstract
Germline mutations of the APC gene, which encodes a multidomain protein of 2843 amino acid residues, cause familial adenomatous polyposis (FAP). Three FAP clinical variants are correlated with the location of APC mutations: (1) classic FAP with profuse polyposis (>1000 adenomas), associated with mutations from codon 1250 to 1424; (2) attenuated FAP (APC extremities (before codon 157 and after codon 1595); (3) classic FAP with intermediate colonic polyposis (100–1000 adenomas), associated with mutations located in the remaining part of APC. In an effort to decipher the clinical phenotype associated with APC C-terminal germline truncating mutations in patients with FAP, after screening APC mutations in one family whose members (n=4) developed gastric polyposis, colon oligo-polyposis and desmoid tumours, we performed a literature meta-analysis of clinically characterised patients (n=97) harbouring truncating mutations in APC C-terminus. The APC distal mutations identified in this study cluster with a phenotype characterised by colon oligo-polyposis, diffuse gastric polyposis and desmoid tumours. In conclusion, we describe a novel FAP clinical variant, which we propose to refer to as Gastric Polyposis and Desmoid FAP, that may require tailored management.
- Published
- 2019
- Full Text
- View/download PDF
32. A novel
- Author
-
Giovanna, Forte, Filomena, Cariola, Katia, De Marco, Andrea, Manghisi, Filomena Anna, Guglielmi, Raffaele, Armentano, Giuseppe, Lippolis, Pietro, Giorgio, Cristiano, Simone, and Vittoria, Disciglio
- Published
- 2021
33. From Genetics to Histomolecular Characterization: An Insight into Colorectal Carcinogenesis in Lynch Syndrome
- Author
-
Cristiano Simone, Gabriella Di Carlo, Giuseppe Ingravallo, Antonio Pisani, Martina Lepore Signorile, and Vittoria Disciglio
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Adenoma ,Colorectal cancer ,QH301-705.5 ,Carcinogenesis ,Review ,MLH1 ,medicine.disease_cause ,DNA Mismatch Repair ,Models, Biological ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,PMS2 ,medicine ,Humans ,Genetic Predisposition to Disease ,Physical and Theoretical Chemistry ,Biology (General) ,early detection ,Molecular Biology ,QD1-999 ,neoplasms ,Spectroscopy ,Germ-Line Mutation ,MMR genes ,business.industry ,Organic Chemistry ,General Medicine ,medicine.disease ,Colorectal Neoplasms, Hereditary Nonpolyposis ,Lynch syndrome ,digestive system diseases ,Computer Science Applications ,CRC ,MSH6 ,Chemistry ,Phenotype ,MSH2 ,030220 oncology & carcinogenesis ,Cancer research ,030211 gastroenterology & hepatology ,Female ,business - Abstract
Lynch syndrome is a hereditary cancer-predisposing syndrome caused by germline defects in DNA mismatch repair (MMR) genes such as MLH1, MSH2, MSH6, and PMS2. Carriers of pathogenic mutations in these genes have an increased lifetime risk of developing colorectal cancer (CRC) and other malignancies. Despite intensive surveillance, Lynch patients typically develop CRC after 10 years of follow-up, regardless of the screening interval. Recently, three different molecular models of colorectal carcinogenesis were identified in Lynch patients based on when MMR deficiency is acquired. In the first pathway, adenoma formation occurs in an MMR-proficient background, and carcinogenesis is characterized by APC and/or KRAS mutation and IGF2, NEUROG1, CDK2A, and/or CRABP1 hypermethylation. In the second pathway, deficiency in the MMR pathway is an early event arising in macroscopically normal gut surface before adenoma formation. In the third pathway, which is associated with mutations in CTNNB1 and/or TP53, the adenoma step is skipped, with fast and invasive tumor growth occurring in an MMR-deficient context. Here, we describe the association between molecular and histological features in these three routes of colorectal carcinogenesis in Lynch patients. The findings summarized in this review may guide the use of individualized surveillance guidelines based on a patient’s carcinogenesis subtype.
- Published
- 2021
34. Correspondence on 'Clinical spectrum of MTOR-related hypomelanosis of Ito with neurodevelopmental abnormalities,' by Carmignac et al
- Author
-
Martina Lepore Signorile, Olga Calabrese, Diana Carli, Valentina Grossi, Carlotta Ranieri, Alessandro Mussa, K Rossi, Cristiano Simone, Nicoletta Resta, Licia Lugli, and Marilidia Piglionica
- Subjects
Hypopigmentation ,medicine.medical_specialty ,Mosaicism ,business.industry ,TOR Serine-Threonine Kinases ,MEDLINE ,Dermatology ,Human genetics ,Humans ,Medicine ,business ,Genetics (clinical) ,PI3K/AKT/mTOR pathway - Published
- 2021
- Full Text
- View/download PDF
35. APC Splicing Mutations Leading to In-Frame Exon Skipping are Rare Events in FAP Pathogenesis and Define the Clinical Outcome
- Author
-
Vittoria Disciglio, Filomena Cariola, Candida Fasano, Giovanna Forte, Cristiano Simone, Martina Lepore Signorile, Katia De Marco, Valentina Grossi, and Paola Sanese
- Subjects
Pathogenesis ,congenital, hereditary, and neonatal diseases and abnormalities ,business.industry ,allergology ,RNA splicing ,Frame (networking) ,Rare events ,medicine ,Bioinformatics ,medicine.disease ,business ,Exon skipping ,Familial adenomatous polyposis - Abstract
Familial adenomatous polyposis (FAP) is caused by germline mutations in the tumor suppressor gene APC. To date, nearly 2000 APC mutations have been described in FAP, most of which are predicted to result in truncated protein products. Mutations leading to aberrant APC splicing have rarely been reported. Here, we characterized a novel germline heterozygous splice donor site mutation in APC exon 12 (NM_000038.5: c.1621_1626+7del) leading to exon 12 skipping in an Italian family with the attenuated FAP (AFAP) phenotype. Moreover, we performed a literature me-ta-analysis of APC splicing mutations. We found that 123 unique APC splice site mutations, in-cluding the one described here, have been reported in FAP patients, 69 of which have been char-acterized at the mRNA level. Among these, only a small proportion (9/69) results in an in-frame protein, with 4 mutations causing skipping of exon 12 and/or 13 with loss of armadillo repeat 2 (ARM2) and 3 (ARM3), and 5 mutations leading to skipping of exon 5, 7, 8, and (partially) 9 with loss of regions not encompassing known functional domains. The APC splicing mutations considered in this study cluster with the AFAP phenotype and delineate a novel molecular mechanism of pathogenesis in FAP disease.
- Published
- 2021
- Full Text
- View/download PDF
36. Pharmacological targeting of the novel β-catenin chromatin-associated kinase p38α in colorectal cancer stem cell tumorspheres and organoids
- Author
-
Gianluca Canettieri, Katia De Marco, Martina Lepore Signorile, Cristiano Simone, Gabriella Di Carlo, Valentina Grossi, Francesco Dituri, Giuseppe Ingravallo, Candida Fasano, Annalisa Nicotra, Giovanna Forte, Paola Sanese, Gianluigi Giannelli, Simone Di Franco, Giorgio Stassi, Francesco Susca, Laura Rosa Mangiapane, Vittoria Disciglio, Lepore Signorile, Martina, Grossi, Valentina, Di Franco, Simone, Forte, Giovanna, Disciglio, Vittoria, Fasano, Candida, Sanese, Paola, De Marco, Katia, Susca, Francesco Claudio, Mangiapane, Laura Rosa, Nicotra, Annalisa, Di Carlo, Gabriella, Dituri, Francesco, Giannelli, Gianluigi, Ingravallo, Giuseppe, Canettieri, Gianluca, Stassi, Giorgio, and Simone, Cristiano
- Subjects
Cancer Research ,Settore MED/06 - Oncologia Medica ,post-translational ,Immunology ,Population ,Synthetic lethality ,Article ,Cellular and Molecular Neuroscience ,Cancer stem cell ,chromatin ,colorectal neoplasms ,humans ,mitogen-activated protein kinase 14 ,neoplastic stem cells ,organoids ,prognosis ,protein processing, post-translational ,beta catenin ,Medicine ,Kinase activity ,colon cancer, p38, cancer stem cells ,lcsh:QH573-671 ,education ,Trametinib ,Settore MED/04 - Patologia Generale ,education.field_of_study ,business.industry ,lcsh:Cytology ,Cancer stem cells ,Wnt signaling pathway ,protein processing ,Cell Biology ,Colorectal cancer ,digestive system diseases ,Settore BIO/12 - Biochimica Clinica E Biologia Molecolare Clinica ,Catenin ,Cancer research ,Stem cell ,Settore MED/46 - Scienze Tecniche Di Medicina Di Laboratorio ,business ,Protein Processing, Post-Translational ,Post-translational modifications - Abstract
The prognosis of locally advanced colorectal cancer (CRC) is currently unsatisfactory. This is mainly due to drug resistance, recurrence, and subsequent metastatic dissemination, which are sustained by the cancer stem cell (CSC) population. The main driver of the CSC gene expression program is Wnt signaling, and previous reports indicate that Wnt3a can activate p38 MAPK. Besides, p38 was shown to feed into the canonical Wnt/β-catenin pathway. Here we show that patient-derived locally advanced CRC stem cells (CRC-SCs) are characterized by increased expression of p38α and are “addicted” to its kinase activity. Of note, we found that stage III CRC patients with high p38α levels display reduced disease-free and progression-free survival. Extensive molecular analysis in patient-derived CRC-SC tumorspheres and APCMin/+ mice intestinal organoids revealed that p38α acts as a β-catenin chromatin-associated kinase required for the regulation of a signaling platform involved in tumor proliferation, metastatic dissemination, and chemoresistance in these CRC model systems. In particular, the p38α kinase inhibitor ralimetinib, which has already entered clinical trials, promoted sensitization of patient-derived CRC-SCs to chemotherapeutic agents commonly used for CRC treatment and showed a synthetic lethality effect when used in combination with the MEK1 inhibitor trametinib. Taken together, these results suggest that p38α may be targeted in CSCs to devise new personalized CRC treatment strategies.
- Published
- 2021
37. Targeting SMYD3 to Sensitize Homologous Recombination-Proficient Tumors to PARP-Mediated Synthetic Lethality
- Author
-
Silvia Corbetta, Vittoria Disciglio, Martina Lepore Signorile, Alberto Del Rio, Natale Porta, Valentina Grossi, Stefania Bertora, Valeria Di Maio, Manuela Bartolini, Valentina Silvestri, Giovanna Forte, Cristiano Simone, Paola Sanese, Giuseppina Caretti, Katia De Marco, Giacomo Buscemi, Laura Ottini, Cinzia Bottino, Candida Fasano, Virginia Valentini, Elisabetta Manoni, Gianluigi Giannelli, Ummu Guven, Edoardo Fabini, Sanese, Paola, Fasano, Candida, Buscemi, Giacomo, Bottino, Cinzia, Corbetta, Silvia, Fabini, Edoardo, Silvestri, Valentina, Valentini, Virginia, Disciglio, Vittoria, Forte, Giovanna, Lepore Signorile, Martina, De Marco, Katia, Bertora, Stefania, Grossi, Valentina, Guven, Ummu, Porta, Natale, Di Maio, Valeria, Manoni, Elisabetta, Giannelli, Gianluigi, Bartolini, Manuela, Del Rio, Alberto, Caretti, Giuseppina, Ottini, Laura, and Simone, Cristiano
- Subjects
0301 basic medicine ,Mutant ,RAD51 ,02 engineering and technology ,Synthetic lethality ,medicine.disease_cause ,Article ,03 medical and health sciences ,medicine ,carcinogenesi ,lcsh:Science ,Molecular Biology ,Cancer ,SMYD3 ,Multidisciplinary ,Chemistry ,cancer ,cell biology ,molecular biology ,Cell Biology ,021001 nanoscience & nanotechnology ,medicine.disease ,Synthetic Lethality ,030104 developmental biology ,PARP-dependent DNA damage ,Cancer cell ,Cancer research ,Phosphorylation ,lcsh:Q ,0210 nano-technology ,Carcinogenesis ,Homologous recombination - Abstract
Summary SMYD3 is frequently overexpressed in a wide variety of cancers. Indeed, its inactivation reduces tumor growth in preclinical in vivo animal models. However, extensive characterization in vitro failed to clarify SMYD3 function in cancer cells, although confirming its importance in carcinogenesis. Taking advantage of a SMYD3 mutant variant identified in a high-risk breast cancer family, here we show that SMYD3 phosphorylation by ATM enables the formation of a multiprotein complex including ATM, SMYD3, CHK2, and BRCA2, which is required for the final loading of RAD51 at DNA double-strand break sites and completion of homologous recombination (HR). Remarkably, SMYD3 pharmacological inhibition sensitizes HR-proficient cancer cells to PARP inhibitors, thereby extending the potential of the synthetic lethality approach in human tumors., Graphical Abstract, Highlights • SMYD3 phosphorylation by ATM favors the formation of HR complexes during DSB response • SMYD3 mediates DSB repair by promoting RAD51 recruitment at DNA damage sites • SMYD3 inhibition triggers a compensatory PARP-dependent DNA damage response • Co-targeting SMYD3/PARP leads to synthetic lethality in HR-proficient cancer cells, Molecular Biology; Cell Biology; Cancer
- Published
- 2020
38. Author response for 'Functional evidence of <scp>mTORβ</scp> splice variant involvement in the pathogenesis of congenital heart defects'
- Author
-
Romina Ficarella, Carlotta Ranieri, Antonella Turchiano, Martina Lepore Signorile, Valentina Grossi, Mattia Gentile, Cristiano Simone, Gabriele Scalzo, Giovanna Maria Ventola, Nicoletta Resta, Daria Carmela Loconte, Francesco Susca, Paolo Volpe, Angela Cordella, and Emanuela Ponzi
- Subjects
Pathogenesis ,Genetics ,business.industry ,Alternative splicing ,Medicine ,business - Published
- 2020
- Full Text
- View/download PDF
39. Functional evidence of mTORβ splice variant involvement in the pathogenesis of congenital heart defects
- Author
-
Paolo Volpe, Mattia Gentile, Gabriele Scalzo, Angela Cordella, Giovanna Maria Ventola, Carlotta Ranieri, Antonella Turchiano, Daria Carmela Loconte, Francesco Susca, Martina Lepore Signorile, Cristiano Simone, Nicoletta Resta, Valentina Grossi, Romina Ficarella, and Emanuela Ponzi
- Subjects
0301 basic medicine ,Heart Defects, Congenital ,030105 genetics & heredity ,Pathogenesis ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,Downregulation and upregulation ,Genetics ,Medicine ,Humans ,Genetic Predisposition to Disease ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase B ,Pathological ,Gene ,Genetics (clinical) ,PI3K/AKT/mTOR pathway ,Mitogen-Activated Protein Kinase Kinases ,business.industry ,TOR Serine-Threonine Kinases ,Alternative splicing ,Infant ,Up-Regulation ,030104 developmental biology ,Chromosomes, Human, Pair 1 ,Mutation ,Cancer research ,Female ,business ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
mTOR dysregulation has been described in pathological conditions, such as cardiovascular and overgrowth disorders. Here we report on the first case of a patient with a complex congenital heart disease and an interstitial duplication in the short arm of chromosome 1, encompassing part of the mTOR gene. Our results suggest that an intragenic mTOR microduplication might play a role in the pathogenesis of non-syndromic congenital heart defects (CHDs) due to an upregulation of mTOR/Rictor and consequently an increased phosphorylation of PI3K/AKT and MEK/ERK signaling pathways in patient-derived amniocytes. This is the first report which shows a causative role of intragenic mTOR microduplication in the etiology of an isolated complex CHD.
- Published
- 2020
40. FOXO3 on the Road to Longevity: Lessons From SNPs and Chromatin Hubs
- Author
-
Valentina Grossi, Giovanna Forte, Paola Sanese, Cristiano Simone, and Vittoria Disciglio
- Subjects
Aging ,Review Article ,Biochemistry ,0302 clinical medicine ,FHRE, Forkhead response element ,Structural Biology ,PJS, Peutz-Jeghers syndrome ,TAD, Topologically associated domain ,media_common ,SNP, Single nucleotide polymorphism ,Genetics ,0303 health sciences ,FOXO3 ,SOD2, Superoxide dismutase 2 ,Longevity ,LD, Linkage disequilibrium ,Phenotype ,HSE, Heat shock element ,ER, Estrogen receptor ,Computer Science Applications ,Chromatin ,ACH, Active chromatin hub ,030220 oncology & carcinogenesis ,FOXO3, Forkhead box 3 ,HPS, Hamartomatous polyposis syndrome ,GPx, Glutathione peroxidase ,SNV, Single nucleotide variant ,GWAS, Genome-wide association study ,Biotechnology ,media_common.quotation_subject ,lcsh:Biotechnology ,Biophysics ,SNP ,Biology ,03 medical and health sciences ,lcsh:TP248.13-248.65 ,Chromatin hub ,Enhancer ,Gene ,Transcription factor ,030304 developmental biology ,5′UTR, Five prime untranslated region ,3C, Chromosome conformation capture ,HSF1, Heat shock factor 1 ,Promoter ,PHTS, PTEN hamartoma tumor syndrome ,IGF-1, Insulin growth factor-1 ,ERE, Estrogen-responsive element ,ROS, Reactive oxygen species - Abstract
Health span is driven by a precise interplay between genes and the environment. Cell response to environmental cues is mediated by signaling cascades and genetic variants that affect gene expression by regulating chromatin plasticity. Indeed, they can promote the interaction of promoters with regulatory elements by forming active chromatin hubs. FOXO3 encodes a transcription factor with a strong impact on aging and age-related phenotypes, as it regulates stress response, therefore affecting lifespan. A significant association has been shown between human longevity and several FOXO3 variants located in intron 2. This haplotype block forms a putative aging chromatin hub in which FOXO3 has a central role, as it modulates the physical connection and activity of neighboring genes involved in age-related processes. Here we describe the role of FOXO3 and its single-nucleotide polymorphisms (SNPs) in healthy aging, with a focus on the enhancer region encompassing the SNP rs2802292, which upregulates FOXO3 expression and can promote the activity of the aging hub in response to different stress stimuli. FOXO3 protective effect on lifespan may be due to the accessibility of this region to transcription factors promoting its expression. This could in part explain the differences in FOXO3 association with longevity between genders, as its activity in females may be modulated by estrogens through estrogen receptor response elements located in the rs2802292-encompassing region. Altogether, the molecular mechanisms described here may help establish whether the rs2802292 SNP can be taken advantage of in predictive medicine and define the potential of targeting FOXO3 for age-related diseases., Graphical Abstract Unlabelled Image
- Published
- 2019
41. Identification and Somatic Characterization of the Germline PTEN Promoter Variant rs34149102 in a Family with Gastrointestinal and Breast Tumors
- Author
-
Vittoria Disciglio, Paola Sanese, Candida Fasano, Claudio Lotesoriere, Anna Maria Valentini, Giovanna Forte, Martina Lepore Signorile, Katia De Marco, Valentina Grossi, Ivan Lolli, Filomena Cariola, and Cristiano Simone
- Subjects
Genetics ,PTEN promoter ,PTEN hamartoma tumor syndrome (PHTS) ,gastroesophageal junction adenocarcinoma ,breast cancer ,Genetics (clinical) - Abstract
Genetic variants located in non-coding regions can affect processes that regulate protein expression, functionally contributing to human disease. Germline heterozygous mutations in the non-coding region of the PTEN gene have been previously identified in patients with PTEN hamartoma tumor syndrome (PHTS) diagnosed with breast, thyroid, and/or endometrial cancer. In this study, we report a PTEN promoter variant (rs34149102 A allele) that was identified by direct sequencing in an Italian family with a history of gastroesophageal junction (GEJ) adenocarcinoma and breast cancer. In order to investigate the putative functional role of the rs34149102 A allele variant, we evaluated the status of PTEN alterations at the somatic level. We found that PTEN protein expression was absent in the GEJ adenocarcinoma tissue of the index case. Moreover, we detected the occurrence of copy number loss involving the PTEN rs34149102 major C allele in tumor tissue, revealing that the second allele was somatically inactivated. This variant is located within an active regulatory region of the PTEN core promoter, and in silico analysis suggests that it may affect the binding of the nuclear transcription factor MAZ and hence PTEN expression. Overall, these results reveal the functional role of the PTEN promoter rs34149102 A allele variant in the modulation of PTEN protein expression and highlight its contribution to hereditary cancer risk.
- Published
- 2022
- Full Text
- View/download PDF
42. The longevity SNP rs2802292 uncovered: HSF1 activates stress-dependent expression of FOXO3 through an intronic enhancer
- Author
-
Martina Lepore Signorile, Rosanna Bagnulo, Giovanna Forte, Paola Sanese, Alessia Peserico, Daria Carmela Loconte, Rosaura Lovaglio, Francesco Susca, Nicoletta Resta, Cristiano Simone, Tugsan Tezil, Valentina Grossi, and Candida Fasano
- Subjects
Transcriptional Activation ,0301 basic medicine ,Enhancer Elements ,Cell Survival ,DNA repair ,Cells ,Physiological ,Longevity ,Locus (genetics) ,Single-nucleotide polymorphism ,Biology ,Stress ,Polymorphism, Single Nucleotide ,Cell Line ,longevity ,snp ,HSF1-FOXO3A ,Promoter Regions ,03 medical and health sciences ,Heat Shock Transcription Factors ,Genetic ,Stress, Physiological ,Cellular stress response ,Genetics ,Humans ,Polymorphism ,Promoter Regions, Genetic ,Enhancer ,Transcription factor ,Cells, Cultured ,5' Untranslated Regions ,Alleles ,Binding Sites ,Forkhead Box Protein O3 ,Introns ,Enhancer Elements, Genetic ,Cultured ,Gene regulation, Chromatin and Epigenetics ,Single Nucleotide ,Isogenic human disease models ,Cell biology ,030104 developmental biology ,FOXO3 - Abstract
The HSF and FOXO families of transcription factors play evolutionarily conserved roles in stress resistance and lifespan. In humans, the rs2802292 G-allele at FOXO3 locus has been associated with longevity in all human populations tested; moreover, its copy number correlated with reduced frequency of age-related diseases in centenarians. At the molecular level, the intronic rs2802292 G-allele correlated with increased expression of FOXO3, suggesting that FOXO3 intron 2 may represent a regulatory region. Here we show that the 90-bp sequence around the intronic single nucleotide polymorphism rs2802292 has enhancer functions, and that the rs2802292 G-allele creates a novel HSE binding site for HSF1, which induces FOXO3 expression in response to diverse stress stimuli. At the molecular level, HSF1 mediates the occurrence of a promoter–enhancer interaction at FOXO3 locus involving the 5′UTR and the rs2802292 region. These data were confirmed in various cellular models including human HAP1 isogenic cell lines (G/T). Our functional studies highlighted the importance of the HSF1-FOXO3-SOD2/CAT/GADD45A cascade in cellular stress response and survival by promoting ROS detoxification, redox balance and DNA repair. Our findings suggest the existence of an HSF1-FOXO3 axis in human cells that could be involved in stress response pathways functionally regulating lifespan and disease susceptibility.
- Published
- 2018
- Full Text
- View/download PDF
43. In vitro efficacy of ARQ 092, an allosteric AKT inhibitor, on primary fibroblast cells derived from patients with PIK3CA-related overgrowth spectrum (PROS)
- Author
-
Andrea D. Praticò, S. Di Tommaso, Angelo Selicorni, Giovanni Abbadessa, Andrea Bartuli, Cristiano Simone, Daria Carmela Loconte, Y. Yu, Francesco Susca, Rosanna Bagnulo, A De Luisi, Carlotta Ranieri, Martino Ruggieri, Valentina Grossi, M. Lerone, Nicoletta Resta, Alessia Peserico, Daniela Melis, Brian Schwartz, Giovanna Forte, and Paola Sanese
- Subjects
Male ,0301 basic medicine ,Cell ,Aminopyridines ,Target therapy ,Wortmannin ,chemistry.chemical_compound ,0302 clinical medicine ,Child ,Growth Disorders ,Genetics (clinical) ,Kinase ,Imidazoles ,Oncogene Protein v-akt ,medicine.anatomical_structure ,Child, Preschool ,PI3K/AKT/mTOR pathway ,Phosphorylation ,Original Article ,Female ,PIK3CA mutations ,Signal Transduction ,Adolescent ,Class I Phosphatidylinositol 3-Kinases ,Primary Cell Culture ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Allosteric Regulation ,Mosaic neurocutaneous disorders ,PI3K/AKT/mTOR inhibitors ,PROS ,Rapamycin ,Fibroblasts ,Humans ,Mutation ,Proto-Oncogene Proteins c-akt ,Genetics ,medicine ,Preschool ,Fibroblast ,Protein kinase B ,business.industry ,Molecular medicine ,030104 developmental biology ,chemistry ,Cancer research ,business ,030217 neurology & neurosurgery - Abstract
Postzygotic mutations of the PIK3CA [phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha] gene constitutively activate the PI3K/AKT/mTOR pathway in PIK3CA-related overgrowth spectrum (PROS) patients, causing congenital mosaic tissue overgrowth that even multiple surgeries cannot solve. mTOR inhibitors are empirically tested and given for compassionate use in these patients. PROS patients could be ideal candidates for enrolment in trials with PI3K/AKT pathway inhibitors, considering the “clean” cellular setting in which a unique driver, a PIK3CA mutation, is present. We aimed to assess the effects of blocking the upstream pathway of mTOR on PROS patient-derived cells by using ARQ 092, a potent, selective, allosteric, and experimental orally bioavailable and highly selective AKT-inhibitor with activity and long-term tolerability, currently under clinical development for treatment of cancer and Proteus syndrome. Cell samples (i.e., primary fibroblasts) were derived from cultured tissues obtained from six PROS patients [3 boys, 3 girls; aged 2 to 17 years] whose spectrum of PIK3A-related overgrowth included HHML [hemihyperplasia multiple lipomatosis; n = 1], CLOVES [congenital lipomatosis, overgrowth, vascular malformations, epidermal nevi, spinal/skeletal anomalies, scoliosis; n = 1], and MCAP [megalencephaly capillary malformation syndrome; n = 4]. We performed the following: (a) a deep sequencing assay of PI3K/AKT pathway genes in the six PROS patients’ derived cells to identify the causative mutations and (b) a pathway analysis to assess the phosphorylation status of AKT [Ser473 and Thr308] and its downstream targets [pAKTS1 (Thr246), pRPS6 (Ser235/236), and pRPS6Kβ1 (Ser371)]. The anti-proliferative effect of ARQ 092 was tested and compared to other PI3K/AKT/mTOR inhibitors [i.e., wortmannin, LY249002, and rapamycin] in the six PROS patient-derived cells. Using ARQ 092 to target AKT, a critical node connecting PI3K and mTOR pathways, we observed the following: (1) strong anti-proliferative activity [ARQ 092 at 0.5, 1, and 2.5 μM blunted phosphorylation of AKT and its downstream targets (in the presence or absence of serum) and inhibited proliferation after 72 h; rapamycin at 100 nM did not decrease AKT phosphorylation] and (2) less cytotoxicity as compared to rapamycin and wortmannin. We demonstrated the following: (a) that PROS cells are dependent on AKT; (b) the advantage of inhibiting the pathway immediately downstream of PI3K to circumventing problems depending on multiple classes a PI3K kinases; and (c) that PROS patients benefit from inhibition of AKT rather than mTOR. Clinical development of ARQ 092 in PROS patients is on going in these patients. Electronic supplementary material The online version of this article (10.1007/s10048-018-0540-1) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
44. Spectrum of Germline Pathogenic Variants in BRCA1/2 Genes in the Apulian Southern Italy Population: Geographic Distribution and Evidence for Targeted Genetic Testing
- Author
-
Cristiano Simone, Simona De Summa, Maria Iole Natalicchio, Stefania Tommasi, Paolo Tarantino, Nicoletta Resta, Elisabetta De Matteis, Mariapia Caputo, Angelo Paradiso, Antonino Pantaleo, Margherita Patruno, Rosanna Bagnulo, Brunella Pilato, Maria Digennaro, and Silvia Costanzo
- Subjects
Proband ,Cancer Research ,endocrine system diseases ,Population ,Biology ,hereditary breast and ovarian cancer ,Article ,Germline ,southern Italy ,geographic distribution ,medicine ,skin and connective tissue diseases ,education ,Gene ,RC254-282 ,Genetic testing ,Genetics ,education.field_of_study ,medicine.diagnostic_test ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,BRCA1 ,BRCA2 ,Geographic distribution ,Oncology ,Shared database ,cardiovascular system ,Apulian population ,Founder effect - Abstract
Simple Summary BRCA1 and BRCA2 are two major high-penetrance breast/ovarian cancer predisposition genes, whose mutations can lead to high risk and early onset of breast and ovarian cancer. Numerous studies are focused on spectrum and prevalence of BRCA1/2 mutations worldwide. This is the first study that exclusively focused on native Apulian probands. We found that ten recurrent BRCA1/2 pathogenic variants account for more than half of the patients with proven HBOC syndrome from Apulia. Besides BRCA1 c.5266dupC, which is present in significant numbers in every Apulian province, the other PVs occur at a high frequency in some areas and not others. In-depth knowledge of the mutation spectrum of the target population and of the relatively small number of recurrent mutations is crucial to develop a specific cost-effective strategy for mutation screening and a program for breast–ovarian cancer control and prevention through more liberal, yet rational, genetic testing and counseling. Abstract BRCA1/2-associated hereditary breast and ovarian cancer is the most common form of hereditary breast and ovarian cancer and occurs in all ethnicities and racial populations. Different BRCA1/BRCA2 pathogenic variants (PVs) have been reported with a wide variety among populations. In this study, we retrospectively analyzed prevalence and geographic distribution of pathogenic germline BRCA1/2 variants in families from Apulia in southern Italy and evaluated the genotype–phenotype correlations. Data were collected from Oncogenetic Services present in Apulian hospitals and a shared database was built containing Apulian native probands (n = 2026) that had undergone genetic testing from 2004 to 2019. PVs were detected in 499 of 2026 (24.6%) probands and 68.5% of them (342 of 499) were in the BRCA1 gene. We found 65 different PVs in BRCA1 and 46 in BRCA2. There were 10 most recurrent PVs and their geographical distribution appears to be significantly specific for each province. We have assumed that these PVs are related to the historical and geopolitical changes that occurred in Apulia over time and/or to a “founder effect”. Broader knowledge of BRCA1/2 prevalence and recurring PVs in specific geographic areas could help establish more flexible genetic testing strategies that may enhance our ability to detect high-risk subjects.
- Published
- 2021
- Full Text
- View/download PDF
45. SMYD3: An oncogenic driver targeting epigenetic regulation and signaling pathways
- Author
-
Cinzia Bottino, Alessia Peserico, Cristiano Simone, and Giuseppina Caretti
- Subjects
0301 basic medicine ,Epigenetic inhibitors ,KMT ,Lysine methylation ,SMYD3 ,Cancer Research ,Methyltransferase ,Review ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Epigenetics ,Cancer ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,030104 developmental biology ,Histone ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,Carcinogenesis ,Reprogramming - Abstract
SMYD3 is a member of the SMYD lysine methylase family and plays an important role in the methylation of various histone and non-histone targets. Aberrant SMYD3 expression contributes to carcinogenesis and SMYD3 upregulation was proposed as a prognostic marker in various solid cancers. Here we summarize SMYD3-mediated regulatory mechanisms, which are implicated in the pathophysiology of cancer, as drivers of distinct oncogenic pathways. We describe SMYD3-dependent mechanisms affecting cancer progression, highlighting SMYD3 interplay with proteins and RNAs involved in the regulation of cancer cell proliferation, migration and invasion. We also address the effectiveness and mechanisms of action for the currently available SMYD3 inhibitors. The findings analyzed herein demonstrate that a complex network of SMYD3-mediated cytoplasmic and nuclear interactions promote oncogenesis across different cancer types. These evidences depict SMYD3 as a modulator of the transcriptional response and of key signaling pathways, orchestrating multiple oncogenic inputs and ultimately, promoting transcriptional reprogramming and tumor transformation. Further insights into the oncogenic role of SMYD3 and its targeting of different synergistic oncogenic signals may be beneficial for effective cancer treatment.
- Published
- 2020
46. FOXO3a from the Nucleus to the Mitochondria: A Round Trip in Cellular Stress Response
- Author
-
Stefania Bertora, Cristiano Simone, Martina Lepore Signorile, Candida Fasano, and Vittoria Disciglio
- Subjects
Cellular homeostasis ,FOXO3A ,stress ,nucleus-mitochondria ,Review ,Mitochondrion ,Mitogen-activated protein kinase kinase ,nuclear/mitochondrial crosstalk ,cellular homeostasis ,Stress, Physiological ,Cellular stress response ,transcription factors ,Animals ,Humans ,FOXO3a ,Protein kinase A ,Transcription factor ,lcsh:QH301-705.5 ,Cellular compartment ,Cell Nucleus ,Kinase ,Chemistry ,Forkhead Box Protein O3 ,General Medicine ,stress response ,Cell biology ,Mitochondria ,lcsh:Biology (General) - Abstract
Cellular stress response is a universal mechanism that ensures the survival or negative selection of cells in challenging conditions. The transcription factor Forkhead box protein O3 (FOXO3a) is a core regulator of cellular homeostasis, stress response, and longevity since it can modulate a variety of stress responses upon nutrient shortage, oxidative stress, hypoxia, heat shock, and DNA damage. FOXO3a activity is regulated by post-translational modifications that drive its shuttling between different cellular compartments, thereby determining its inactivation (cytoplasm) or activation (nucleus and mitochondria). Depending on the stress stimulus and subcellular context, activated FOXO3a can induce specific sets of nuclear genes, including cell cycle inhibitors, pro-apoptotic genes, reactive oxygen species (ROS) scavengers, autophagy effectors, gluconeogenic enzymes, and others. On the other hand, upon glucose restriction, 5′-AMP-activated protein kinase (AMPK) and mitogen activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) -dependent FOXO3a mitochondrial translocation allows the transcription of oxidative phosphorylation (OXPHOS) genes, restoring cellular ATP levels, while in cancer cells, mitochondrial FOXO3a mediates survival upon genotoxic stress induced by chemotherapy. Interestingly, these target genes and their related pathways are diverse and sometimes antagonistic, suggesting that FOXO3a is an adaptable player in the dynamic homeostasis of normal and stressed cells. In this review, we describe the multiple roles of FOXO3a in cellular stress response, with a focus on both its nuclear and mitochondrial functions.
- Published
- 2019
47. APC Splicing Mutations Leading to In-Frame Exon 12 or Exon 13 Skipping Are Rare Events in FAP Pathogenesis and Define the Clinical Outcome
- Author
-
Martina Lepore Signorile, Filomena Cariola, Katia De Marco, Giovanna Forte, Paola Sanese, Vittoria Disciglio, Valentina Grossi, Cristiano Simone, and Candida Fasano
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,lcsh:QH426-470 ,Tumor suppressor gene ,Biology ,medicine.disease_cause ,Article ,Germline ,Familial adenomatous polyposis ,splicing ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Germline mutation ,familial adenomatous polyposis ,Genetics ,medicine ,Genetics (clinical) ,Mutation ,medicine.disease ,Exon skipping ,APC ,lcsh:Genetics ,030104 developmental biology ,030220 oncology & carcinogenesis ,RNA splicing ,FAP pathogenesis ,exon skipping - Abstract
Familial adenomatous polyposis (FAP) is caused by germline mutations in the tumor suppressor gene APC. To date, nearly 2000 APC mutations have been described in FAP, most of which are predicted to result in truncated protein products. Mutations leading to aberrant APC splicing have rarely been reported. Here, we characterized a novel germline heterozygous splice donor site mutation in APC exon 12 (NM_000038.5: c.1621_1626+7del) leading to exon 12 skipping in an Italian family with the attenuated FAP (AFAP) phenotype. Moreover, we performed a literature meta-analysis of APC splicing mutations. We found that 119 unique APC splicing mutations, including the one described here, have been reported in FAP patients, 69 of which have been characterized at the mRNA level. Among these, only a small proportion (9/69) results in an in-frame protein, with four mutations causing skipping of exon 12 or 13 with loss of armadillo repeat 2 (ARM2) and 3 (ARM3), and five mutations leading to skipping of exon 5, 7, 8, or (partially) 9 with loss of regions not encompassing known functional domains. The APC splicing mutations causing skipping of exon 12 or 13 considered in this study cluster with the AFAP phenotype and reveal a potential molecular mechanism of pathogenesis in FAP disease.
- Published
- 2021
- Full Text
- View/download PDF
48. Chasing the FOXO3: Insights into Its New Mitochondrial Lair in Colorectal Cancer Landscape
- Author
-
Candida Fasano, Valentina Grossi, Martina Lepore Signorile, Cristiano Simone, Valentina Celestini, and Paola Sanese
- Subjects
0301 basic medicine ,Cancer Research ,Mitochondrial DNA ,Cellular homeostasis ,colorectal cancer ,Review ,Mitochondrion ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,precision cancer medicine ,medicine ,post-translational modifications ,Transcription factor ,Cancer ,chemoresistance ,cancer-related signaling pathways ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,CRC ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,mitochindrial ,Cancer cell ,FOXO3A ,Signal transduction ,Carcinogenesis ,mitochondrial FOXO3a ,metformin - Abstract
Colorectal cancer (CRC) poses a formidable challenge in terms of molecular heterogeneity, as it involves a variety of cancer-related pathways and molecular changes unique to an individual’s tumor. On the other hand, recent advances in DNA sequencing technologies provide an unprecedented capacity to comprehensively identify the genetic alterations resulting in tumorigenesis, raising the hope that new therapeutic approaches based on molecularly targeted drugs may prevent the occurrence of chemoresistance. Regulation of the transcription factor FOXO3a in response to extracellular cues plays a fundamental role in cellular homeostasis, being part of the molecular machinery that drives cells towards survival or death. Indeed, FOXO3a is controlled by a range of external stimuli, which not only influence its transcriptional activity, but also affect its subcellular localization. These regulation mechanisms are mediated by cancer-related signaling pathways that eventually drive changes in FOXO3a post-translational modifications (e.g., phosphorylation). Recent results showed that FOXO3a is imported into the mitochondria in tumor cells and tissues subjected to metabolic stress and cancer therapeutics, where it induces expression of the mitochondrial genome to support mitochondrial metabolism and cell survival. The current review discusses the potential clinical relevance of multidrug therapies that drive cancer cell fate by regulating critical pathways converging on FOXO3a.
- Published
- 2019
49. Characterization of a rare variant (c.2635-2AG) of the MSH2 gene in a family with Lynch syndrome
- Author
-
Floranna Guglielmi, Claudio Lotesoriere, Candida Fasano, Ivan Lolli, Anna Maria Valentini, Andrea Manghisi, Cristiano Simone, L. Russo, Vittoria Disciglio, Giovanna Forte, Filomena Cariola, Antonio Di Carlo, and Maria Lucia Caruso
- Subjects
0301 basic medicine ,Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Clinical Biochemistry ,nutritional and metabolic diseases ,Biology ,medicine.disease ,MLH1 ,digestive system diseases ,Lynch syndrome ,Pathology and Forensic Medicine ,MSH6 ,03 medical and health sciences ,030104 developmental biology ,Germline mutation ,Oncology ,MSH2 ,medicine ,PMS2 ,DNA mismatch repair ,Msh2 gene ,neoplasms - Abstract
Introduction: Lynch syndrome is caused by germline mutations in one of the mismatch repair genes ( MLH1, MSH2, MSH6, and PMS2) or in the EPCAM gene. Lynch syndrome is defined on the basis of clinical, pathological, and genetic findings. Accordingly, the identification of predisposing genes allows for accurate risk assessment and tailored screening protocols. Case Description: Here, we report a family case with three family members manifesting the Lynch syndrome phenotype, all of which harbor the rare variant c.2635-2A>G affecting the splice site consensus sequence of intron 15 of the MSH2 gene. This mutation was previously described only in one family with Lynch syndrome, in which mismatch repair protein expression in tumor tissues was not assessed. In this study, we report for the first time the molecular characterization of the MSH2 c.2635-2A>G variant through in silico prediction analysis, microsatellite instability, and mismatch repair protein expression experiments on tumor tissues of Lynch syndrome patients. The potential effect of the splice site variant was revealed by three splicing prediction bioinformatics tools, which suggested the generation of a new cryptic splicing site. The potential pathogenic role of this variant was also revealed by the presence of microsatellite instability and the absence of MSH2/MSH6 heterodimer protein expression in the tumor cells of cancer tissues of the affected family members. Conclusions: We provide compelling evidence in favor of the pathogenic role of the MSH2 variant c.2635-2A>G, which could induce an alteration of the canonical splice site and consequently an aberrant form of the protein product (MSH2).
- Published
- 2018
50. A SMYD3 Small-Molecule Inhibitor Impairing Cancer Cell Growth
- Author
-
Alessia Peserico, Arménio Jorge Moura Barbosa, Paola Sanese, Mary Pat Moyer, Edoardo Fabini, Valeria Di Virgilio, Alberto Del Rio, Aldo Germani, Carlo Bertucci, Cristiano Simone, Giuseppina Caretti, Raffaella Fittipaldi, and Greta Varchi
- Subjects
Methyltransferase ,Physiology ,Cell growth ,Colorectal cancer ,Drug discovery ,Clinical Biochemistry ,Druggability ,Cancer ,Cell Biology ,Biology ,medicine.disease ,medicine.disease_cause ,Cell biology ,Cancer cell ,medicine ,Carcinogenesis - Abstract
SMYD3 is a histone lysine methyltransferase that plays an important role in transcriptional activation as a member of an RNA polymerase complex, and its oncogenic role has been described in different cancer types. We studied the expression and activity of SMYD3 in a preclinical model of colorectal cancer (CRC) and found that it is strongly upregulated throughout tumorigenesis both at the mRNA and protein level. Our results also showed that RNAi-mediated SMYD3 ablation impairs CRC cell proliferation indicating that SMYD3 is required for proper cancer cell growth. These data, together with the importance of lysine methyltransferases as a target for drug discovery, prompted us to carry out a virtual screening to identify new SMYD3 inhibitors by testing several candidate small molecules. Here we report that one of these compounds (BCI-121) induces a significant reduction in SMYD3 activity both in vitro and in CRC cells, as suggested by the analysis of global H3K4me2/3 and H4K5me levels. Of note, the extent of cell growth inhibition by BCI-121 was similar to that observed upon SMYD3 genetic ablation. Most of the results described above were obtained in CRC; however, when we extended our observations to tumor cell lines of different origin, we found that SMYD3 inhibitors are also effective in other cancer types, such as lung, pancreatic, prostate, and ovarian. These results represent the proof of principle that SMYD3 is a druggable target and suggest that new compounds capable of inhibiting its activity may prove useful as novel therapeutic agents in cancer treatment.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.