128 results on '"Guidalberto Manfioletti"'
Search Results
2. HMGA1 positively regulates the microtubule-destabilizing protein stathmin promoting motility in TNBC cells and decreasing tumour sensitivity to paclitaxel
- Author
-
Michela Sgubin, Silvia Pegoraro, Ilenia Pellarin, Gloria Ros, Riccardo Sgarra, Silvano Piazza, Gustavo Baldassarre, Barbara Belletti, and Guidalberto Manfioletti
- Subjects
Cytology ,QH573-671 - Abstract
Abstract High Mobility Group A1 (HMGA1) is an architectural chromatin factor involved in the regulation of gene expression and a master regulator in Triple Negative Breast Cancer (TNBC). In TNBC, HMGA1 is overexpressed and coordinates a gene network that controls cellular processes involved in tumour development, progression, and metastasis formation. Here, we find that the expression of HMGA1 and of the microtubule-destabilizing protein stathmin correlates in breast cancer (BC) patients. We demonstrate that HMGA1 depletion leads to a downregulation of stathmin expression and activity on microtubules resulting in decreased TNBC cell motility. We show that this pathway is mediated by the cyclin-dependent kinase inhibitor p27kip1 (p27). Indeed, the silencing of HMGA1 expression in TNBC cells results both in an increased p27 protein stability and p27-stathmin binding. When the expression of both HMGA1 and p27 is silenced, we observe a significant rescue in cell motility. These data, obtained in cellular models, were validated in BC patients. In fact, we find that patients with high levels of both HMGA1 and stathmin and low levels of p27 have a statistically significant lower survival probability in terms of relapse-free survival (RFS) and distant metastasis-free survival (DMFS) with respect to the patient group with low HMGA1, low stathmin, and high p27 expression levels. Finally, we show in an in vivo xenograft model that depletion of HMGA1 chemo-sensitizes tumour cells to paclitaxel, a drug that is commonly used in TNBC treatments. This study unveils a new interaction among HMGA1, p27, and stathmin that is critical in BC cell migration. Moreover, our data suggest that taxol-based treatments may be more effective in reducing the tumour burden when tumour cells express low levels of HMGA1.
- Published
- 2022
- Full Text
- View/download PDF
3. HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Rossella Zanin, Silvia Pegoraro, Gloria Ros, Yari Ciani, Silvano Piazza, Fleur Bossi, Roberta Bulla, Cristina Zennaro, Federica Tonon, Dejan Lazarevic, Elia Stupka, Riccardo Sgarra, and Guidalberto Manfioletti
- Subjects
HMGA1 ,FOXM1 ,Triple-negative breast cancer ,Gene network ,VEGFA ,Angiogenesis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Breast cancer is the most common malignancy in women worldwide. Among the breast cancer subtypes, triple-negative breast cancer (TNBC) is the most aggressive and the most difficult to treat. One of the master regulators in TNBC progression is the architectural transcription factor HMGA1. This study aimed to further explore the HMGA1 molecular network to identify molecular mechanisms involved in TNBC progression. Methods RNA from the MDA-MB-231 cell line, silenced for HMGA1 expression, was sequenced and, with a bioinformatic analysis, molecular partners HMGA1 could cooperate with in regulating common downstream gene networks were identified. Among the putative partners, the FOXM1 transcription factor was selected. The relationship occurring between HMGA1 and FOXM1 was explored by qRT-PCR, co-immunoprecipitation and protein stability assays. Subsequently, the transcriptional activity of HMGA1 and FOXM1 was analysed by luciferase assay on the VEGFA promoter. The impact on angiogenesis was assessed in vitro, evaluating the tube formation ability of endothelial cells exposed to the conditioned medium of MDA-MB-231 cells silenced for HMGA1 and FOXM1 and in vivo injecting MDA-MB-231 cells, silenced for the two factors, in zebrafish larvae. Results Here, we discover FOXM1 as a novel molecular partner of HMGA1 in regulating a gene network implicated in several breast cancer hallmarks. HMGA1 forms a complex with FOXM1 and stabilizes it in the nucleus, increasing its transcriptional activity on common target genes, among them, VEGFA, the main inducer of angiogenesis. Furthermore, we demonstrate that HMGA1 and FOXM1 synergistically drive breast cancer cells to promote tumor angiogenesis both in vitro in endothelial cells and in vivo in a zebrafish xenograft model. Moreover, using a dataset of breast cancer patients we show that the co-expression of HMGA1, FOXM1 and VEGFA is a negative prognostic factor of distant metastasis-free survival and relapse-free survival. Conclusions This study reveals FOXM1 as a crucial interactor of HMGA1 and proves that their cooperative action supports breast cancer aggressiveness, by promoting tumor angiogenesis. Therefore, the possibility to target HMGA1/FOXM1 in combination should represent an attractive therapeutic option to counteract breast cancer angiogenesis.
- Published
- 2019
- Full Text
- View/download PDF
4. HMGA2 Antisense Long Non-coding RNAs as New Players in the Regulation of HMGA2 Expression and Pancreatic Cancer Promotion
- Author
-
Gloria Ros, Silvia Pegoraro, Paolo De Angelis, Riccardo Sgarra, Silvia Zucchelli, Stefano Gustincich, and Guidalberto Manfioletti
- Subjects
natural antisense non-coding RNAs ,HMGA ,cancer ,gene expression regulation ,FANTOM5 ,HMGA2-AS1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Natural antisense long non-coding RNAs (lncRNAs) are regulatory RNAs transcribed from the opposite strand of either protein coding or non-coding genes, able to modulate their own sense gene expression. Hence, their dysregulation can lead to pathologic processes. Cancer is a complex class of diseases determined by the aberrant expression of a variety of factors, among them, the oncofetal chromatin architectural proteins High Mobility Group A (HMGA) modulate several cancer hallmarks. Thus, we decided to investigate the presence of natural antisense lncRNAs in HMGA1 and HMGA2 loci, and their possible involvement in gene expression regulation.Methods: We used FANTOM5 data resources, FANTOM-CAT genome browser and Zenbu visualization tool, which employ 1,829 human CAGE and RNA-sequencing libraries, to determine expression, ontology enrichment, and dynamic regulation of natural antisense lncRNAs in HMGA1 and HMGA2 loci. We then performed qRT-PCR in different cancer cell lines to validate the existence of HMGA2-AS1 transcripts. We depleted HMGA2-AS1 transcripts with siRNAs and investigated HMGA2 expression by qRT-PCR and western blot analyses. Moreover, we evaluated cell viability and migration by MTS and transwell assays, and EMT markers by qRT-PCR and immunofluorescence. Furthermore, we used bioinformatics approaches to evaluate HMGA2 and HMGA2-AS1 correlation and overall survival in tumor patients.Results: We found the presence of a promoter-associated lncRNA (CATG00000088127.1) in the HMGA1 gene and three antisense genes (RPSAP52, HMGA2-AS1, and RP11-366L20.3) in the HMGA2 gene. We studied the uncharacterized HMGA2-AS1 transcripts, validating their existence in cancer cell lines and observing a positive correlation between HMGA2 and HMGA2-AS1 expression in a cancer-derived patient dataset. We showed that HMGA2-AS1 transcripts positively modulate HMGA2 expression and migration properties of PANC1 cells through HMGA2. In addition, Kaplan-Meier analysis showed that high level of HMGA2-AS1 is a negative prognostic factor in pancreatic cancer patients.Conclusions: Our results describe novel antisense lncRNAs associated with HMGA1 and HMGA2 genes. In particular, we demonstrate that HMGA2-AS1 is involved in the regulation of its own sense gene expression, mediating tumorigenesis. Thus, we highlight a new layer of complexity in the regulation of HMGA2 expression, providing new potential targets for cancer therapy.
- Published
- 2020
- Full Text
- View/download PDF
5. Editorial: Hormone Receptors and Breast Cancer
- Author
-
Antonio Brunetti and Guidalberto Manfioletti
- Subjects
estrogen receptors ,steroid receptors ,tyrosine kinase receptors ,hormone resistance ,triple negative breast cancer ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Published
- 2019
- Full Text
- View/download PDF
6. Transcriptional Regulation of Glucose Metabolism: The Emerging Role of the HMGA1 Chromatin Factor
- Author
-
Eusebio Chiefari, Daniela P. Foti, Riccardo Sgarra, Silvia Pegoraro, Biagio Arcidiacono, Francesco S. Brunetti, Manfredi Greco, Guidalberto Manfioletti, and Antonio Brunetti
- Subjects
HMGA1 ,glucose homeostasis ,insulin resistance ,type 2 diabetes ,glucose metabolism ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
HMGA1 (high mobility group A1) is a nonhistone architectural chromosomal protein that functions mainly as a dynamic regulator of chromatin structure and gene transcription. As such, HMGA1 is involved in a variety of fundamental cellular processes, including gene expression, epigenetic regulation, cell differentiation and proliferation, as well as DNA repair. In the last years, many reports have demonstrated a role of HMGA1 in the transcriptional regulation of several genes implicated in glucose homeostasis. Initially, it was proved that HMGA1 is essential for normal expression of the insulin receptor (INSR), a critical link in insulin action and glucose homeostasis. Later, it was demonstrated that HMGA1 is also a downstream nuclear target of the INSR signaling pathway, representing a novel mediator of insulin action and function at this level. Moreover, other observations have indicated the role of HMGA1 as a positive modulator of the Forkhead box protein O1 (FoxO1), a master regulatory factor for gluconeogenesis and glycogenolysis, as well as a positive regulator of the expression of insulin and of a series of circulating proteins that are involved in glucose counterregulation, such as the insulin growth factor binding protein 1 (IGFBP1), and the retinol binding protein 4 (RBP4). Thus, several lines of evidence underscore the importance of HMGA1 in the regulation of glucose production and disposal. Consistently, lack of HMGA1 causes insulin resistance and diabetes in humans and mice, while variations in the HMGA1 gene are associated with the risk of type 2 diabetes and metabolic syndrome, two highly prevalent diseases that share insulin resistance as a common pathogenetic mechanism. This review intends to give an overview about our current knowledge on the role of HMGA1 in glucose metabolism. Although research in this field is ongoing, many aspects still remain elusive. Future directions to improve our insights into the pathophysiology of glucose homeostasis may include epigenetic studies and the use of “omics” strategies. We believe that a more comprehensive understanding of HMGA1 and its networks may reveal interesting molecular links between glucose metabolism and other biological processes, such as cell proliferation and differentiation.
- Published
- 2018
- Full Text
- View/download PDF
7. The Architectural Chromatin Factor High Mobility Group A1 Enhances DNA Ligase IV Activity Influencing DNA Repair.
- Author
-
Ilenia Pellarin, Laura Arnoldo, Silvia Costantini, Silvia Pegoraro, Gloria Ros, Carlotta Penzo, Gianluca Triolo, Francesca Demarchi, Riccardo Sgarra, Alessandro Vindigni, and Guidalberto Manfioletti
- Subjects
Medicine ,Science - Abstract
The HMGA1 architectural transcription factor is an oncogene overexpressed in the vast majority of human cancers. HMGA1 is a highly connected node in the nuclear molecular network and the key aspect of HMGA1 involvement in cancer development is that HMGA1 simultaneously confers cells multiple oncogenic hits, ranging from global chromatin structural and gene expression modifications up to the direct functional alterations of key cellular proteins. Interestingly, HMGA1 also modulates DNA damage repair pathways. In this work, we provide evidences linking HMGA1 with Non-Homologous End Joining DNA repair. We show that HMGA1 is in complex with and is a substrate for DNA-PK. HMGA1 enhances Ligase IV activity and it counteracts the repressive histone H1 activity towards DNA ends ligation. Moreover, breast cancer cells overexpressing HMGA1 show a faster recovery upon induction of DNA double-strand breaks, which is associated with a higher survival. These data suggest that resistance to DNA-damaging agents in cancer cells could be partially attributed to HMGA1 overexpression thus highlighting the relevance of considering HMGA1 expression levels in the selection of valuable and effective pharmacological regimens.
- Published
- 2016
- Full Text
- View/download PDF
8. Expression and functional characterization of Xhmg-at-hook genes in Xenopus laevis.
- Author
-
Simone Macrì, Riccardo Sgarra, Gloria Ros, Elisa Maurizio, Salvina Zammitti, Ornella Milani, Marco Onorati, Robert Vignali, and Guidalberto Manfioletti
- Subjects
Medicine ,Science - Abstract
High Mobility Group A proteins (HMGA1 and HMGA2) are architectural nuclear factors involved in development, cell differentiation, and cancer formation and progression. Here we report the cloning, developmental expression and functional analysis of a new multi-AT-hook factor in Xenopus laevis (XHMG-AT-hook) that exists in three different isoforms. Xhmg-at-hook1 and 3 isoforms, but not isoform 2, are expressed throughout the entire development of Xenopus, both in the maternal and zygotic phase. Localized transcripts are present in the animal pole in the early maternal phase; during the zygotic phase, mRNA can be detected in the developing central nervous system (CNS), including the eye, and in the neural crest. We show evidence that XHMG-AT-hook proteins differ from typical HMGA proteins in terms of their properties in DNA binding and in protein/protein interaction. Finally, we provide evidence that they are involved in early CNS development and in neural crest differentiation.
- Published
- 2013
- Full Text
- View/download PDF
9. Identification and characterization of new molecular partners for the protein arginine methyltransferase 6 (PRMT6).
- Author
-
Alessandra Lo Sardo, Sandro Altamura, Silvia Pegoraro, Elisa Maurizio, Riccardo Sgarra, and Guidalberto Manfioletti
- Subjects
Medicine ,Science - Abstract
PRMT6 is a protein arginine methyltransferase that has been implicated in transcriptional regulation, DNA repair, and human immunodeficiency virus pathogenesis. Only few substrates of this enzyme are known and therefore its cellular role is not well understood. To identify in an unbiased manner substrates and potential regulators of PRMT6 we have used a yeast two-hybrid approach. We identified 36 new putative partners for PRMT6 and we validated the interaction in vivo for 7 of them. In addition, using invitro methylation assay we identified 4 new substrates for PRMT6, extending the involvement of this enzyme to other cellular processes beyond its well-established role in gene expression regulation. Holistic approaches create molecular connections that allow to test functional hypotheses. The assembly of PRMT6 protein network allowed us to formulate functional hypotheses which led to the discovery of new molecular partners for the architectural transcription factor HMGA1a, a known substrate for PRMT6, and to provide evidences for a modulatory role of HMGA1a on the methyltransferase activity of PRMT6.
- Published
- 2013
- Full Text
- View/download PDF
10. Correction: Identification and Characterization of New Molecular Partners for the Protein Arginine Methyltransferase 6 (PRMT6).
- Author
-
Alessandra Lo Sardo, Sandro Altamura, Silvia Pegoraro, Elisa Maurizio, Riccardo Sgarra, and Guidalberto Manfioletti
- Subjects
Medicine ,Science - Published
- 2013
- Full Text
- View/download PDF
11. Proteomic tools to study phosphorylation of intrinsically disordered proteins
- Author
-
Barbara Spolaore, Luca Secco, Giulia Rocca, Guidalberto Manfioletti, Giorgio Arrigoni, and Riccardo Sgarra
- Subjects
Molecular Biology ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
12. Mechanism of Action of Lactic Acid on Histones in Cancer
- Author
-
Riccardo Sgarra, Sabrina Battista, Laura Cerchia, Guidalberto Manfioletti, and Monica Fedele
- Subjects
Physiology ,Clinical Biochemistry ,General Earth and Planetary Sciences ,Cell Biology ,Molecular Biology ,Biochemistry ,General Environmental Science - Published
- 2023
- Full Text
- View/download PDF
13. HMGA1 Regulates the Expression of Replication-Dependent Histone Genes and Cell-Cycle in Breast Cancer Cells
- Author
-
Sara Petrosino, Sabrina Pacor, Silvia Pegoraro, Virginia Anna Gazziero, Giulia Canarutto, Silvano Piazza, Guidalberto Manfioletti, and Riccardo Sgarra
- Subjects
Inorganic Chemistry ,Organic Chemistry ,TNBC ,HMGA1 ,RD-HIST ,NPAT ,cell-cycle ,epirubicin ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
Breast cancer (BC) is the primary cause of cancer mortality in women and the triple-negative breast cancer (TNBC) is the most aggressive subtype characterized by poor differentiation and high proliferative properties. High mobility group A1 (HMGA1) is an oncogenic factor involved in the onset and progression of the neoplastic transformation in BC. Here, we unraveled that the replication-dependent-histone (RD-HIST) gene expression is enriched in BC tissues and correlates with HMGA1 expression. We explored the role of HMGA1 in modulating the RD-HIST genes expression in TNBC cells and show that MDA-MB-231 cells, depleted of HMGA1, express low levels of core histones. We show that HMGA1 participates in the activation of the HIST1H4H promoter and that it interacts with the nuclear protein of the ataxia-telangiectasia mutated locus (NPAT), the coordinator of the transcription of the RD-HIST genes. Moreover, we demonstrate that HMGA1 silencing increases the percentage of cells in G0/G1 phase both in TNBC and epirubicin resistant TNBC cells. Moreover, HMGA1 silencing causes an increase in epirubicin IC50 both in parental and epirubicin resistant cells thus suggesting that targeting HMGA1 could affect the efficacy of epirubicin treatment.
- Published
- 2022
- Full Text
- View/download PDF
14. Heterogeneity of triple-negative breast cancer: understanding the Daedalian labyrinth and how it could reveal new drug targets
- Author
-
Alberto Zambelli, Riccardo Sgarra, Rita De Sanctis, Elisa Agostinetto, Armando Santoro, Guidalberto Manfioletti, Zambelli, Alberto, Sgarra, Riccardo, De Sanctis, Rita, Agostinetto, Elisa, Santoro, Armando, and Manfioletti, Guidalberto
- Subjects
Pharmacology ,epigenetics ,TNBC subtypes ,Clinical Biochemistry ,TNBC subtype ,Triple Negative Breast Neoplasms ,MAPK ,ADC ,BRCAness ,PI3K-AKT-mTOR ,TROP-2 ,androgen receptor ,immunotherapy ,tumor heterogeneity ,Ecosystem ,Humans ,Tumor Microenvironment ,BRCAne ,Drug Discovery ,Molecular Medicine ,epigenetic ,Human - Abstract
Introduction Triple-negative breast cancer (TNBC) is considered the most aggressive breast cancer subtype with the least favorable outcomes. However, recent research efforts have generated an enhanced knowledge of the biology of the disease and have provided a new, more comprehensive understanding of the multifaceted ecosystem that underpins TNBC. Areas covered In this review, the authors illustrate the principal biological characteristics of TNBC, the molecular driver alterations, targetable genes, and the biomarkers of immune engagement that have been identified across the subgroups of TNBC. Accordingly, the authors summarize the landscape of the innovative and investigative biomarker-driven therapeutic options in TNBC that emerge from the unique biological basis of the disease. Expert opinion The therapeutic setting of TNBC is rapidly evolving. An enriched understanding of the tumor spatial and temporal heterogeneity and the surrounding microenvironment of this complex disease can effectively support the development of novel and tailored opportunities of treatment.
- Published
- 2022
- Full Text
- View/download PDF
15. The Epithelial–Mesenchymal Transition at the Crossroads between Metabolism and Tumor Progression
- Author
-
Monica Fedele, Riccardo Sgarra, Sabrina Battista, Laura Cerchia, Guidalberto Manfioletti, Fedele, M., Sgarra, R., Battista, S., Cerchia, L., and Manfioletti, G.
- Subjects
Transcription Factor ,Drug Resistance ,tumor progression ,Review ,Breast cancer ,Neoplasms ,Tumor Microenvironment ,Molecular Targeted Therapy ,Biology (General) ,Cancer ,Epithelial–mesenchymal transition (EMT) ,Lung cancer ,Metabolic rewiring ,Metabolism ,Thyroid cancer ,Tumor progression ,Warburg effect ,Animals ,Biomarkers ,Biomarkers, Tumor ,Disease Management ,Disease Progression ,Disease Susceptibility ,Drug Resistance, Neoplasm ,Gene Expression Regulation, Neoplastic ,Humans ,Metabolic Networks and Pathways ,Neoplastic Stem Cells ,Organ Specificity ,Oxidative Stress ,RNA Interference ,RNA, Long Noncoding ,Signal Transduction ,Transcription Factors ,Energy Metabolism ,Epithelial-Mesenchymal Transition ,epithelial–mesenchymal transition (EMT) ,Spectroscopy ,Tumor ,General Medicine ,Computer Science Applications ,Chemistry ,Long Noncoding ,Human ,QH301-705.5 ,metabolic rewiring ,Catalysis ,Inorganic Chemistry ,cancer ,Physical and Theoretical Chemistry ,Molecular Biology ,QD1-999 ,Neoplastic ,Animal ,Organic Chemistry ,Metabolic Networks and Pathway ,Oxidative Stre ,Biomarker ,Gene Expression Regulation ,Neoplasm ,RNA ,Neoplastic Stem Cell ,metabolism - Abstract
The transition between epithelial and mesenchymal phenotype is emerging as a key determinant of tumor cell invasion and metastasis. It is a plastic process in which epithelial cells first acquire the ability to invade the extracellular matrix and migrate into the bloodstream via transdifferentiation into mesenchymal cells, a phenomenon known as epithelial–mesenchymal transition (EMT), and then reacquire the epithelial phenotype, the reverse process called mesenchymal–epithelial transition (MET), to colonize a new organ. During all metastatic stages, metabolic changes, which give cancer cells the ability to adapt to increased energy demand and to withstand a hostile new environment, are also important determinants of successful cancer progression. In this review, we describe the complex interaction between EMT and metabolism during tumor progression. First, we outline the main connections between the two processes, with particular emphasis on the role of cancer stem cells and LncRNAs. Then, we focus on some specific cancers, such as breast, lung, and thyroid cancer.
- Published
- 2022
16. HMGA1 positively regulates the microtubule-destabilizing protein stathmin promoting motility in TNBC cells and decreasing tumour sensitivity to paclitaxel
- Author
-
Michela Sgubin, Silvia Pegoraro, Ilenia Pellarin, Gloria Ros, Riccardo Sgarra, Silvano Piazza, Gustavo Baldassarre, Barbara Belletti, Guidalberto Manfioletti, Sgubin, Michela, Pegoraro, Silvia, Pellarin, Ilenia, Ros, Gloria, Sgarra, Riccardo, Piazza, Silvano, Baldassarre, Gustavo, Belletti, Barbara, and Manfioletti, Guidalberto
- Subjects
Cancer Research ,Paclitaxel ,Immunology ,Microtubule ,Triple Negative Breast Neoplasms ,Cell Biology ,Microtubules ,Cellular and Molecular Neuroscience ,HMGA1a Protein ,Humans ,Neoplasm Recurrence, Local ,Stathmin ,Neoplasm Recurrence ,Local ,Human - Abstract
High Mobility Group A1 (HMGA1) is an architectural chromatin factor involved in the regulation of gene expression and a master regulator in Triple Negative Breast Cancer (TNBC). In TNBC, HMGA1 is overexpressed and coordinates a gene network that controls cellular processes involved in tumour development, progression, and metastasis formation. Here, we find that the expression of HMGA1 and of the microtubule-destabilizing protein stathmin correlates in breast cancer (BC) patients. We demonstrate that HMGA1 depletion leads to a downregulation of stathmin expression and activity on microtubules resulting in decreased TNBC cell motility. We show that this pathway is mediated by the cyclin-dependent kinase inhibitor p27kip1 (p27). Indeed, the silencing of HMGA1 expression in TNBC cells results both in an increased p27 protein stability and p27-stathmin binding. When the expression of both HMGA1 and p27 is silenced, we observe a significant rescue in cell motility. These data, obtained in cellular models, were validated in BC patients. In fact, we find that patients with high levels of both HMGA1 and stathmin and low levels of p27 have a statistically significant lower survival probability in terms of relapse-free survival (RFS) and distant metastasis-free survival (DMFS) with respect to the patient group with low HMGA1, low stathmin, and high p27 expression levels. Finally, we show in an in vivo xenograft model that depletion of HMGA1 chemo-sensitizes tumour cells to paclitaxel, a drug that is commonly used in TNBC treatments. This study unveils a new interaction among HMGA1, p27, and stathmin that is critical in BC cell migration. Moreover, our data suggest that taxol-based treatments may be more effective in reducing the tumour burden when tumour cells express low levels of HMGA1.
- Published
- 2021
17. Expression and function of the homeodomain-containing protein Hex in thyroid cells.
- Author
-
Lucia Pellizzari, Angela D'Elia, Alessandra Rustighi, Guidalberto Manfioletti, Gianluca Tell, and Giuseppe Damante
- Published
- 2000
- Full Text
- View/download PDF
18. Gene network analysis using SWIM reveals interplay between the transcription factor-encoding genes HMGA1, FOXM1, and MYBL2 in triple-negative breast cancer
- Author
-
Paola Paci, Silvia Pegoraro, Guidalberto Manfioletti, Federica Conte, Giulia Fiscon, Fiscon, G., Pegoraro, S., Conte, F., Manfioletti, G., and Paci, P.
- Subjects
Gene regulatory network ,Datasets as Topic ,Cell Cycle Proteins ,Triple Negative Breast Neoplasms ,Biochemistry ,Structural Biology ,Cell Cycle Protein ,Ductal ,Databases, Genetic ,Data Mining ,Gene Regulatory Networks ,Breast ,HMGA1a Protein ,Triple-negative breast cancer ,correlation network ,network medicine ,0303 health sciences ,Tumor ,030302 biochemistry & molecular biology ,Carcinoma, Ductal, Breast ,Phenotype ,Gene Expression Regulation, Neoplastic ,Trans-Activator ,Multigene Family ,triple-negative breast cancer ,Female ,Human ,Protein Binding ,Signal Transduction ,Triple Negative Breast Neoplasm ,Biophysics ,Computational biology ,Biology ,correlation networks ,Cell Line ,Databases ,03 medical and health sciences ,Breast cancer ,Atlases as Topic ,Genetic ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Molecular Biology ,Gene ,030304 developmental biology ,Neoplastic ,Forkhead Box Protein M1 ,Gene Expression Profiling ,Trans-Activators ,Carcinoma ,Cell Biology ,medicine.disease ,Gene expression profiling ,Gene Expression Regulation ,FOXM1 - Abstract
Among breast cancer subtypes, triple-negative breast cancer (TNBC) is the most aggressive with the worst prognosis and the highest rates of metastatic disease. To identify TNBC gene signatures, we applied the network-based methodology implemented by the SWIM software to gene expression data of TNBC patients in The Cancer Genome Atlas (TCGA) database. SWIM enables to predict key (switch) genes within the co-expression network, whose perturbations in expression pattern and abundance may contribute to the (patho)biological phenotype. Here, SWIM analysis revealed an interesting interplay between the genes encoding the transcription factors HMGA1, FOXM1, and MYBL2, suggesting a potential cooperation among these three switch genes in TNBC development. The correlative nature of this interplay in TNBC was assessed by in vitro experiments, demonstrating how they may actually modulate the expression of each other.
- Published
- 2021
- Full Text
- View/download PDF
19. Therapeutic potential of parkin as a tumor suppressor via transcriptional control of cyclins in glioblastoma cell and animal models
- Author
-
Lígia Ramos dos Santos, Frédéric Checler, Aurore Bernardin, Lila Rouland, Guidalberto Manfioletti, Cristine Alves da Costa, Ahmed Idbaih, Eric Duplan, Karen S. Katula, Institut de pharmacologie moléculaire et cellulaire (IPMC), Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), University of North Carolina [Greensboro] (UNCG), University of North Carolina System (UNC), Università degli studi di Trieste = University of Trieste, Institut du Cerveau = Paris Brain Institute (ICM), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Gestionnaire, HAL Sorbonne Université 5, Rouland, L, Duplan, E, Ramos Dos Santos, L, Bernardin, A, Katula, K, Manfioletti, G, Idbaih, A, Checler, F, Alves da Costa, C., Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), University of Trieste, Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Male ,Cyclin A ,Proliferation ,Cyclin B ,Medicine (miscellaneous) ,Gene Expression ,01 natural sciences ,030218 nuclear medicine & medical imaging ,Mice ,0302 clinical medicine ,Transcriptional regulation ,Genes, Tumor Suppressor ,parkin ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,transcription factor ,Cyclin ,Mice, Knockout ,biology ,medicine.diagnostic_test ,Chemistry ,Brain Neoplasms ,Cell Cycle ,Cell cycle ,Flow Cytometry ,Gene Expression Regulation, Neoplastic ,Models, Animal ,Cyclins ,Glioblastoma ,Parkin ,Transcription factor ,Female ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,cyclins ,Research Paper ,Ubiquitin-Protein Ligases ,proliferation ,010402 general chemistry ,Flow cytometry ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,RNA, Messenger ,[SDV.NEU] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Cell Proliferation ,glioblastoma ,0104 chemical sciences ,MicroRNAs ,Tumor progression ,biology.protein ,Cancer research - Abstract
International audience; Parkin (PK) is an E3-ligase harboring tumor suppressor properties that has been associated to various cancer types including glioblastoma (GBM). However, PK is also a transcription factor (TF), the contribution of which to GBM etiology remains to be established. Methods: The impact of PK on GBM cells proliferation was analyzed by real-time impedance measurement and flow cytometry. Cyclins A and B proteins, promoter activities and mRNA levels were measured by western blot, luciferase assay and quantitative real-time PCR. Protein-protein and protein-promoter interactions were performed by co-immunoprecipitation and by ChIP approaches. The contribution of endogenous PK to tumor progression in vivo was performed by allografts of GL261 GBM cells in wild-type and PK knockout mice. Results: We show that overexpressed and endogenous PK control GBM cells proliferation by modulating the S and G2/M phases of the cell cycle via the trans-repression of cyclin A and cyclin B genes. We establish that cyclin B is regulated by both E3-ligase and TF PK functions while cyclin A is exclusively regulated by PK TF function. PK invalidation leads to enhanced tumor progression in immunocompetent mice suggesting an impact of PK-dependent tumor environment to tumor development. We show that PK is secreted by neuronal cells and recaptured by tumor cells. Recaptured PK lowered cyclins levels and decreased GBM cells proliferation. Further, PK expression is decreased in human GBM biopsies and its expression is inversely correlated to both cyclins A and B expressions. Conclusion: Our work demonstrates that PK tumor suppressor function contributes to the control of tumor by its cellular environment. It also shows a key role of PK TF function in GBM development via the control of cyclins in vitro and in vivo. It suggests that therapeutic strategies aimed at controlling PK shuttling to the nucleus may prove useful to treat GBM.
- Published
- 2021
- Full Text
- View/download PDF
20. Epithelial–Mesenchymal Transition (EMT) 2021
- Author
-
Monica Fedele, GUIDALBERTO MANFIOLETTI, Manfioletti, Guidalberto, and Fedele, Monica
- Subjects
Epithelial Cell ,Epithelial-Mesenchymal Transition ,Cell Transdifferentiation ,Epithelial Cells ,Mesenchymal Stem Cells ,Organic Chemistry ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy - Abstract
Epithelial–mesenchymal transition (EMT) is a transdifferentiation process wherein epithelial cells acquire characteristics typical of mesenchymal cells [...]
- Published
- 2022
- Full Text
- View/download PDF
21. High Mobility Group A (HMGA): Chromatin Nodes Controlled by a Knotty miRNA Network
- Author
-
Rossella Zanin, Riccardo Sgarra, Gloria Ros, Daniela D'Angelo, Michela Sgubin, Sabrina Battista, Guidalberto Manfioletti, Silvia Pegoraro, Sara Petrosino, Sgarra, R, Pegoraro, S, D'Angelo, D, Ros, G, Zanin, R, Sgubin, M, Petrosino, S, Battista, S, and Manfioletti, G
- Subjects
0301 basic medicine ,High mobility group A ,miRNA ,post-transcriptional regulation ,cancer ,[object Object] ,Computational biology ,Review ,Biology ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,microRNA ,Gene expression ,Animals ,Humans ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Post-transcriptional regulation ,Gene ,Spectroscopy ,HMGA Proteins ,Organic Chemistry ,HMGA ,General Medicine ,Chromatin ,Computer Science Applications ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,High-mobility group ,lcsh:Biology (General) ,lcsh:QD1-999 ,030220 oncology & carcinogenesis ,Cancer cell - Abstract
High mobility group A (HMGA) proteins are oncofoetal chromatin architectural factors that are widely involved in regulating gene expression. These proteins are unique, because they are highly expressed in embryonic and cancer cells, where they play a relevant role in cell proliferation, stemness, and the acquisition of aggressive tumour traits, i.e., motility, invasiveness, and metastatic properties. The HMGA protein expression levels and activities are controlled by a connected set of events at the transcriptional, post-transcriptional, and post-translational levels. In fact, microRNA (miRNA)-mediated RNA stability is the most-studied mechanism of HMGA protein expression modulation. In this review, we contribute to a comprehensive overview of HMGA-targeting miRNAs; we provide detailed information regarding HMGA gene structural organization and a comprehensive evaluation and description of HMGA-targeting miRNAs, while focusing on those that are widely involved in HMGA regulation; and, we aim to offer insights into HMGA-miRNA mutual cross-talk from a functional and cancer-related perspective, highlighting possible clinical implications.
- Published
- 2020
- Full Text
- View/download PDF
22. Targeting the intrinsically disordered architectural High Mobility Group A (HMGA) oncoproteins in breast cancer: learning from the past to design future strategies
- Author
-
Gloria Ros, Silvia Pegoraro, Sara Petrosino, Alberto Zambelli, Riccardo Sgarra, Michela Sgubin, Guidalberto Manfioletti, Pegoraro, S., Ros, G., Sgubin, M., Petrosino, S., Zambelli, A., Sgarra, R., and Manfioletti, G.
- Subjects
0301 basic medicine ,Clinical Biochemistry ,Triple Negative Breast Neoplasms ,03 medical and health sciences ,stemness ,0302 clinical medicine ,Breast cancer ,breast cancer ,anticancer therapies ,chemoresistance ,High Mobility Group A ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,skin and connective tissue diseases ,Pharmacology ,HMGA Proteins ,business.industry ,HMGA ,anticancer therapie ,Breast cancer subtype ,medicine.disease ,030104 developmental biology ,High-mobility group ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Female ,business - Abstract
Introduction: Triple-negative breast cancer (TNBC) is the most difficult breast cancer subtype to treat because of its heterogeneity and lack of specific therapeutic targets. High Mobility Group A (HMGA) proteins are chromatin architectural factors that have multiple oncogenic functions in breast cancer, and they represent promising molecular therapeutic targets for this disease. Areas covered: We offer an overview of the strategies that have been exploited to counteract HMGA oncoprotein activities at the transcriptional and post-transcriptional levels. We also present the possibility of targeting cancer-associated factors that lie downstream of HMGA proteins and discuss the contribution of HMGA proteins to chemoresistance. Expert opinion: Different strategies have been exploited to counteract HMGA protein activities; these involve interfering with their nucleic acid binding properties and the blocking of HMGA expression. Some approaches have provided promising results. However, some unique characteristics of the HMGA proteins have not been exploited; these include their extensive protein-protein interaction network and their intrinsically disordered status that present the possibility that HMGA proteins could be involved in the formation of proteinaceous membrane-less organelles (PMLO) by liquid-liquid phase separation. These unexplored characteristics could open new pharmacological avenues to counteract the oncogenic contributions of HMGA proteins.
- Published
- 2020
23. HMGA2 Antisense Long Non-coding RNAs as New Players in the Regulation of HMGA2 Expression and Pancreatic Cancer Promotion
- Author
-
Stefano Gustincich, Gloria Ros, Guidalberto Manfioletti, Paolo De Angelis, Riccardo Sgarra, Silvia Zucchelli, Silvia Pegoraro, Ros, G, Pegoraro, S, De Angelis, P, Sgarra, R, Zucchelli, S, Gustincich, S, and Manfioletti, G
- Subjects
0301 basic medicine ,Cancer Research ,Small interfering RNA ,Computational biology ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,HMGA1 Gene ,03 medical and health sciences ,0302 clinical medicine ,natural antisense non-coding RNAs ,Gene expression ,Sense (molecular biology) ,medicine ,cancer ,HMGA ,FANTOM5 ,HMGA2-AS1 ,gene expression regulation ,Gene ,Original Research ,Regulation of gene expression ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Carcinogenesis - Abstract
Background: Natural antisense long non-coding RNAs (lncRNAs) are regulatory RNAs transcribed from the opposite strand of either protein coding or non-coding genes, able to modulate their own sense gene expression. Hence, their dysregulation can lead to pathologic processes. Cancer is a complex class of diseases determined by the aberrant expression of a variety of factors, among them, the oncofetal chromatin architectural proteins High Mobility Group A (HMGA) modulate several cancer hallmarks. Thus, we decided to investigate the presence of natural antisense lncRNAs in HMGA1 and HMGA2 loci, and their possible involvement in gene expression regulation. Methods: We used FANTOM5 data resources, FANTOM-CAT genome browser and Zenbu visualization tool, which employ 1,829 human CAGE and RNA-sequencing libraries, to determine expression, ontology enrichment, and dynamic regulation of natural antisense lncRNAs in HMGA1 and HMGA2 loci. We then performed qRT-PCR in different cancer cell lines to validate the existence of HMGA2-AS1 transcripts. We depleted HMGA2-AS1 transcripts with siRNAs and investigated HMGA2 expression by qRT-PCR and western blot analyses. Moreover, we evaluated cell viability and migration by MTS and transwell assays, and EMT markers by qRT-PCR and immunofluorescence. Furthermore, we used bioinformatics approaches to evaluate HMGA2 and HMGA2-AS1 correlation and overall survival in tumor patients. Results: We found the presence of a promoter-associated lncRNA (CATG00000088127.1) in the HMGA1 gene and three antisense genes (RPSAP52, HMGA2-AS1, and RP11-366L20.3) in the HMGA2 gene. We studied the uncharacterized HMGA2-AS1 transcripts, validating their existence in cancer cell lines and observing a positive correlation between HMGA2 and HMGA2-AS1 expression in a cancer-derived patient dataset. We showed that HMGA2-AS1 transcripts positively modulate HMGA2 expression and migration properties of PANC1 cells through HMGA2. In addition, Kaplan-Meier analysis showed that high level of HMGA2-AS1 is a negative prognostic factor in pancreatic cancer patients. Conclusions: Our results describe novel antisense lncRNAs associated with HMGA1 and HMGA2 genes. In particular, we demonstrate that HMGA2-AS1 is involved in the regulation of its own sense gene expression, mediating tumorigenesis. Thus, we highlight a new layer of complexity in the regulation of HMGA2 expression, providing new potential targets for cancer therapy.
- Published
- 2020
- Full Text
- View/download PDF
24. High Mobility Group A (HMGA) proteins: Molecular instigators of breast cancer onset and progression
- Author
-
Antonio Brunetti, Guidalberto Manfioletti, Daniela Foti, Silvia Pegoraro, Eusebio Chiefari, Gloria Ros, Carlotta Penzo, Riccardo Sgarra, Sgarra, R, Pegoraro, S, Ros, G, Penzo, C, Chiefari, E, Foti, D, Brunetti, A, and Manfioletti, G.
- Subjects
0301 basic medicine ,Cancer Research ,Transcription, Genetic ,Antineoplastic Agents ,Breast Neoplasms ,Disease ,Computational biology ,03 medical and health sciences ,Breast cancer ,HMGA2 ,Cancer heterogeneity ,Biomarkers, Tumor ,Genetics ,medicine ,Animals ,Humans ,HMGA ,HMGA Proteins ,biology ,Chromatin Assembly and Disassembly ,Prognosis ,medicine.disease ,Chromatin ,Architectural transcription factors ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,030104 developmental biology ,High-mobility group ,Oncology ,biology.protein ,Female ,Architectural transcription factor ,Signal Transduction - Abstract
Cancer heterogeneity is one of the factors that constitute an obstacle towards an efficient targeting of this multifaceted disease. Molecular information can help in classifying cancer subtypes and in providing clinicians with novel targeted therapeutic opportunities. In this regard, classification of breast cancer into intrinsic subtypes based on molecular profiling represents a valuable prototype. The High Mobility Group A (HMGA) chromatin architectural factors (HMGA1a, HMGA1b, and HMGA2) have a relevant and causal role in breast cancer onset and development, by influencing virtually all cancer hallmarks. The regulation of HMGA expression is under the control of major pathways involved in cell proliferation and survival, as well as in other cancer-related processes, thereby suggesting, for the HMGA members, a high degree of homology and overlapping activities. Despite of this evidence, HMGA proteins display also specific functions. In this review, we provide an overview of (i) the pathways involved in HMGA transcriptional and post-transcriptional regulation, (ii) the utilization of HMGA as molecular markers, and (iii) the biological role of HMGA in the context of breast cancer. We focus on the potential significance of HMGA in governing the onset and development of this tumour, as well as on the potential of these factors as novel specific targets for preventing and treating strategies. The emerging picture is a highly interconnected triad of proteins that could mutually influence each other, either in a competitive or cooperative manner, and that, in our opinion, should be considered as a unified and integrated protein system.
- Published
- 2018
- Full Text
- View/download PDF
25. The High Mobility Group A1 (HMGA1) Chromatin Architectural Factor Modulates Nuclear Stiffness in Breast Cancer Cells
- Author
-
Enrico Pobega, Carlotta Penzo, Hernán Morales-Navarrete, Silvia Pegoraro, Riccardo Maraspini, Luisa Ulloa Severino, Elena Ambrosetti, Pietro Parisse, Riccardo Sgarra, Loredana Casalis, Beatrice Senigagliesi, Sara Petrosino, Guidalberto Manfioletti, Senigagliesi, Beatrice, Penzo, C, Severino, Lu, Maraspini, R, Petrosino, Sara, Morales-Navarrete, H, Pobega, E, Ambrosetti, E, Parisse, P, Pegoraro, S, Manfioletti, G, Casalis, L, and Sgarra, R
- Subjects
nuclear stiffness ,HMGA1 ,Heterochromatin ,Gene Expression ,Breast Neoplasms ,Kaplan-Meier Estimate ,nuclear stiffne ,histone H1 ,Catalysis ,Article ,Inorganic Chemistry ,Histones ,lcsh:Chemistry ,Histone H1 ,Cell Line, Tumor ,Humans ,cancer ,Neoplastic transformation ,atomic force microscopy (AFM) ,Physical and Theoretical Chemistry ,Phosphorylation ,chromatin ,mass spectrometry ,Stimulated emission depletion (STED) microscopy ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,Cell Nucleus ,HMGA Proteins ,biology ,Chemistry ,Organic Chemistry ,Cell Cycle ,General Medicine ,Prognosis ,Computer Science Applications ,Cell biology ,Chromatin ,Histone ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer cell ,biology.protein ,Nuclear lamina ,Female ,Protein Binding - Abstract
Plasticity is an essential condition for cancer cells to invade surrounding tissues. The nucleus is the most rigid cellular organelle and it undergoes substantial deformations to get through environmental constrictions. Nuclear stiffness mostly depends on the nuclear lamina and chromatin, which in turn might be affected by nuclear architectural proteins. Among these is the HMGA1 (High Mobility Group A1) protein, a factor that plays a causal role in neoplastic transformation and that is able to disentangle heterochromatic domains by H1 displacement. Here we made use of atomic force microscopy to analyze the stiffness of breast cancer cellular models in which we modulated HMGA1 expression to investigate its role in regulating nuclear plasticity. Since histone H1 is the main modulator of chromatin structure and HMGA1 is a well-established histone H1 competitor, we correlated HMGA1 expression and cellular stiffness with histone H1 expression level, post-translational modifications, and nuclear distribution. Our results showed that HMGA1 expression level correlates with nuclear stiffness, is associated to histone H1 phosphorylation status, and alters both histone H1 chromatin distribution and expression. These data suggest that HMGA1 might promote chromatin relaxation through a histone H1-mediated mechanism strongly impacting on the invasiveness of cancer cells.
- Published
- 2019
- Full Text
- View/download PDF
26. Semaphorin-7A on Exosomes: A Promigratory Signal in the Glioma Microenvironment
- Author
-
Anna Bartolini, Carla Di Loreto, Daniela Cesselli, Federica Caponnetto, Miran Skrap, Ivana Manini, Riccardo Sgarra, Guidalberto Manfioletti, Tamara Ius, Antonio Paolo Beltrami, Maria Elisabetta Ruaro, Manini, I, Ruaro, Me, Sgarra, R, Bartolini, Anna, Caponnetto, EDMEA FRANCESCA ADELAIDE, Ius, T, Skrap, M, DI LORETO, Carla, Beltrami, Ap, Manfioletti, G, and Cesselli, D.
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system ,Motility ,glioblastoma microenvironment ,exosomes ,Semaphorin 7A ,integrin β1/FAK signalling ,motility ,Biology ,lcsh:RC254-282 ,Article ,Focal adhesion ,03 medical and health sciences ,0302 clinical medicine ,Semaphorin ,Glioma ,medicine ,exosome ,fungi ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Microvesicles ,In vitro ,Cell biology ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Stem cell ,Function (biology) - Abstract
Exosomes are one of the most important mediators of the cross talk occurring between glioma stem cells (GSCs) and the surrounding microenvironment. We have previously shown that exosomes released by patient-derived glioma-associated stem cells (GASC) are able to increase, in vitro, the aggressiveness of both GSC and glioblastoma cell lines. To understand which molecules are responsible for this tumour-supporting function, we performed a descriptive proteomic analysis of GASC-exosomes and identified, among the others, Semaphorin7A (SEMA7A). SEMA7A was described as a promigratory cue in physiological and pathological conditions, and we hypothesised that it could modulate GSC migratory properties. Here, we described that SEMA7A is exposed on GASC-exosomes&rsquo, surface and signals to GSC through Integrin &beta, 1. This interaction activates focal adhesion kinase into GSC and increases their motility, in our patient-based in vitro model. Our findings suggest SEMA7A-&beta, 1-integrin as a new target to disrupt the communication between GSCs and the supporting microenvironment.
- Published
- 2019
27. Additional file 11: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Subjects
endocrine system ,skin and connective tissue diseases - Abstract
Figure S8. (a-c) Kaplan Meier curves of DMFS in a cohort of breast cancer patients stratified by HMGA1 (a), FOXM1 (b) and VEGFA (c) expression. (d-f) Kaplan Meier curves of RFS in a cohort of breast cancer patients stratified by HMGA1 (d), FOXM1 (e) and VEGFA (f) expression. (PDF 387 kb)
- Published
- 2019
- Full Text
- View/download PDF
28. HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Fleur Bossi, Silvano Piazza, Elia Stupka, Rossella Zanin, Riccardo Sgarra, Roberta Bulla, Guidalberto Manfioletti, Federica Tonon, Dejan Lazarevic, Cristina Zennaro, Yari Ciani, Gloria Ros, Silvia Pegoraro, Zanin, R, Pegoraro, S, Ros, G, Ciani, Y, Piazza, S, Bossi, F, Bulla, R, Zennaro, C, Tonon, F, Lazarevic, D, Stupka, E, Sgarra, R, and Manfioletti, G
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cancer Research ,Transcription, Genetic ,Angiogenesis ,Triple Negative Breast Neoplasms ,0302 clinical medicine ,HMGA1a Protein ,Promoter Regions, Genetic ,Zebrafish ,Triple-negative breast cancer ,Tube formation ,biology ,Protein Stability ,Gene network ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Prognosis ,Gene Expression Regulation, Neoplastic ,Vascular endothelial growth factor A ,Oncology ,030220 oncology & carcinogenesis ,Female ,VEGFA ,HMGA1 ,lcsh:RC254-282 ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene Silencing ,Transcription factor ,Cell Nucleus ,Sequence Analysis, RNA ,Gene Expression Profiling ,Research ,Forkhead Box Protein M1 ,FOXM1 ,Endothelial Cells ,medicine.disease ,Survival Analysis ,HEK293 Cells ,030104 developmental biology ,Culture Media, Conditioned ,biology.protein ,Cancer research - Abstract
Background Breast cancer is the most common malignancy in women worldwide. Among the breast cancer subtypes, triple-negative breast cancer (TNBC) is the most aggressive and the most difficult to treat. One of the master regulators in TNBC progression is the architectural transcription factor HMGA1. This study aimed to further explore the HMGA1 molecular network to identify molecular mechanisms involved in TNBC progression. Methods RNA from the MDA-MB-231 cell line, silenced for HMGA1 expression, was sequenced and, with a bioinformatic analysis, molecular partners HMGA1 could cooperate with in regulating common downstream gene networks were identified. Among the putative partners, the FOXM1 transcription factor was selected. The relationship occurring between HMGA1 and FOXM1 was explored by qRT-PCR, co-immunoprecipitation and protein stability assays. Subsequently, the transcriptional activity of HMGA1 and FOXM1 was analysed by luciferase assay on the VEGFA promoter. The impact on angiogenesis was assessed in vitro, evaluating the tube formation ability of endothelial cells exposed to the conditioned medium of MDA-MB-231 cells silenced for HMGA1 and FOXM1 and in vivo injecting MDA-MB-231 cells, silenced for the two factors, in zebrafish larvae. Results Here, we discover FOXM1 as a novel molecular partner of HMGA1 in regulating a gene network implicated in several breast cancer hallmarks. HMGA1 forms a complex with FOXM1 and stabilizes it in the nucleus, increasing its transcriptional activity on common target genes, among them, VEGFA, the main inducer of angiogenesis. Furthermore, we demonstrate that HMGA1 and FOXM1 synergistically drive breast cancer cells to promote tumor angiogenesis both in vitro in endothelial cells and in vivo in a zebrafish xenograft model. Moreover, using a dataset of breast cancer patients we show that the co-expression of HMGA1, FOXM1 and VEGFA is a negative prognostic factor of distant metastasis-free survival and relapse-free survival. Conclusions This study reveals FOXM1 as a crucial interactor of HMGA1 and proves that their cooperative action supports breast cancer aggressiveness, by promoting tumor angiogenesis. Therefore, the possibility to target HMGA1/FOXM1 in combination should represent an attractive therapeutic option to counteract breast cancer angiogenesis. Electronic supplementary material The online version of this article (10.1186/s13046-019-1307-8) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
29. Additional file 5: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Figure S2. (a) Boxplots showing the expression levels of FOXM1 mRNA in breast cancer samples with high and low expression of HMGA1 (b) Schematic representation of the pGL3-5BS reporter vector. The 5 binding elements of FOXM1 are represented with white boxes. (c) Western blot analysis of HEK293T transfected with pEGFP-FOXM1 (600 ng) and increasing amounts (200, 400 and 600 ng) of pEGFP-HMGA1, using an α-GFP as primary antibody. pRL-CMV Renilla luciferase expression vector was used to normalize for transfection efficiencies. (d) Confirmation of gene silencing. qRT-PCR of HMGA1 and FOXM1 levels after 72 h of HMGA1 (grey bar), FOXM1 (light blue bar) and HMGA1/FOXM1 (purple bar) silencing in MDA-MB-231 cell line. GAPDH was used for normalization. The data are compared to siCTRL and are presented as the mean ± SD (n = 3), ***p
- Published
- 2019
- Full Text
- View/download PDF
30. Additional file 4: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Table S4. List of differentially expressed genes after HMGA1-silencing at 24 (a) and 72 (b) hours in common with FOXM1-gene network. (PDF 35 kb)
- Published
- 2019
- Full Text
- View/download PDF
31. Additional file 2: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Figure S1. Clustering of gene expression data showing expression levels at different time points before and after silencing of HMGA1. Color intensity corresponds to the row Z-Score. (PDF 1107 kb)
- Published
- 2019
- Full Text
- View/download PDF
32. Additional file 6: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Figure S3. (a) Representative immunofluorescence images of the translocation of FOXM1 (green) and its colocalization with the Subunit β5 of the proteasome (red) in MDA-MB-231 control cells versus cells silenced for HMGA1. Images were taken at 60X magnification. (b) and (c) Representative immunofluorescence images of the translocation of FOXM1 (green) and its colocalization with the Subunit β2 (b) and β5 (c) of the proteasome (red) in MDA-MB-157 control cells versus cells silenced for HMGA1. Images were taken at 60X magnification. (d) Representative images of FOXM1-GFP after HMGA1 silencing and pEGFP-FOXM1 transfection in MDA-MB-231 cells. White arrows indicate the translocation of FOXM1 after HMGA1 silencing. (e) Representative images of FOXM1-GFP after HMGA1 silencing and pEGFP-FOXM1 transfection in HEK293T cells. White arrows indicate the cellular localization of FOXM1 after HMGA1 silencing. (PDF 1197 kb)
- Published
- 2019
- Full Text
- View/download PDF
33. Additional file 7: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Figure S4. (a) Boxplots showing the expression levels of VEGFA mRNA in breast cancer samples. The samples were stratified based on HMGA1 (left) and FOXM1 (right) mRNA expression levels. (b) and (c) Luciferase assays on HEK293T cells transiently co-transfected with the luciferase reporter plasmid pGL4.10-VEGFprom (− 1000–1) with increasing quantities of either the expression plasmid pEGFP-FOXM1 (b) or pEGFP-HMGA1 (c). pRL-CMV Renilla luciferase expression vector was included to normalize for transfection efficiencies. Values are reported as relative luciferase activity comparing to cells transfected with the expression control vector pEGFP. The data are represented as the mean ± SD (n = 3). **p
- Published
- 2019
- Full Text
- View/download PDF
34. Additional file 8: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Figure S5. (a) Schematic representation of the bioinformatic analysis of the 1000 bp VEGFA promoter sequence cloned upstream the luciferase sequence in pGL4.10-VEGFprom (− 1000–1) used in reporter experiments. FOXM1 binding sites are represented with light blue ovals, whereas the AT-enriched sequences bound by HMGA1 are figured as grey boxes. In detailed, we found several AT-rich sequences in the region from – 979 to 907 bp, from − 641 to − 521 bp, from − 355 to − 322 bp, from − 169 to − 75 bp, where the AT stretches are particularly long, and from − 17 to − 13 bp from the TSS of the VEGFA promoter. In addition, we found 9 putative FOXM1 binding sites from − 993 to − 922 bp, from − 643 to – 638 bp, from − 326 to − 322 bp, from − 124 to − 104 bp, where it is located the FOXM1 preferential binding sequence TAAACA, and from − 17 to − 13 bp from the TSS of the VEGFA promoter. (b) Schematic representation of deletion reporter vectors pGL4.10-VEGF (− 1000–500) and pGL4.10-VEGF (− 500–1) obtained from pGL4.10-VEGFprom (− 1000–1). (c) Luciferase assay on HEK293T cells transiently co-transfected with the luciferase reporter plasmid pVEGFprom (− 1000–1), the deletion mutants pVEGFprom (− 1000–500) or pVEGFprom (− 500–1) with the expression plasmids pEGFP-HMGA1 and pEGFP-FOXM1. pRL-CMV Renilla luciferase expression vector was included to normalize for transfection efficiencies. Values are reported as relative luciferase activity comparing to cells transfected with the expression plasmid pEGFP. The data are represented as the mean ± SD (n = 3). NS: not significant; two-tailed Student’s t-test. An example of western blot validations is reported. (d) and (e) Representative images of western blot validations of experiments presented in Fig. 5b and d respectively. (PDF 2000 kb)
- Published
- 2019
- Full Text
- View/download PDF
35. Additional file 9: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Figure S6. HUVEC cells were treated with MDA-MB-231 cells supernatants, who had been previously silenced for HMGA1, FOXM1 or co-silenced for HMGA1 and FOXM1. Serum-free medium and normal human serum (NHS) were used as negative and positive controls respectively. (a) The proliferation of HUVEC cells was investigated by the positivity to the Ki67 marker and expressed in terms of relative % of fluorescence respect to negative control (CTRL-). The data are represented as the mean ± SD (n > 3); **p 3). *p
- Published
- 2019
- Full Text
- View/download PDF
36. HMGA1 Modulates Gene Transcription Sustaining a Tumor Signalling Pathway Acting on the Epigenetic Status of Triple-Negative Breast Cancer Cells
- Author
-
Rossella Zanin, Riccardo Sgarra, Carlotta Penzo, Laura Arnoldo, Jacek R. Wiśniewski, Gloria Ros, Sara Petrosino, Guidalberto Manfioletti, Silvia Pegoraro, Penzo, C, Arnoldo, L, Pegoraro, S, Petrosino, S, Ros, G, Zanin, R, Wiśniewski, Jr, Manfioletti, G, and Sgarra, R
- Subjects
Cancer Research ,Biology ,CBP ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,breast cancer ,histone H3 ,Histone H2B ,Histone code ,Epigenetics ,High Mobility Group A ,Transcription factor ,030304 developmental biology ,0303 health sciences ,TNBC ,epigenetic ,RSK2 ,histone H2B ,HMGA ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Chromatin ,Cell biology ,Histone ,Oncology ,030220 oncology & carcinogenesis ,biology.protein - Abstract
Chromatin accessibility plays a critical factor in regulating gene expression in cancer cells. Several factors, including the High Mobility Group A (HMGA) family members, are known to participate directly in chromatin relaxation and transcriptional activation. The HMGA1 oncogene encodes an architectural chromatin transcription factor that alters DNA structure and interacts with transcription factors favouring their landing onto transcription regulatory sequences. Here, we provide evidence of an additional mechanism exploited by HMGA1 to modulate transcription. We demonstrate that, in a triple-negative breast cancer cellular model, HMGA1 sustains the action of epigenetic modifiers and in particular it positively influences both histone H3S10 phosphorylation by ribosomal protein S6 kinase alpha-3 (RSK2) and histone H2BK5 acetylation by CREB-binding protein (CBP). HMGA1, RSK2, and CBP control the expression of a set of genes involved in tumor progression and epithelial to mesenchymal transition. These results suggest that HMGA1 has an effect on the epigenetic status of cancer cells and that it could be exploited as a responsiveness predictor for epigenetic therapies in triple-negative breast cancers.
- Published
- 2019
37. Additional file 3: of HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1
- Author
-
Zanin, Rossella, Pegoraro, Silvia, Ros, Gloria, Yari Ciani, Piazza, Silvano, Bossi, Fleur, Bulla, Roberta, Zennaro, Cristina, Tonon, Federica, Lazarevic, Dejan, Stupka, Elia, Sgarra, Riccardo, and Guidalberto Manfioletti
- Abstract
Tables S2. and S3. GSEA analysis on 24 and 72â h siHMGA1-regulated genes. (PDF 72 kb)
- Published
- 2019
- Full Text
- View/download PDF
38. Retraction for Berlingieri et al., 'Inhibition of HMGI-C Protein Synthesis Suppresses Retrovirally Induced Neoplastic Transformation of Rat Thyroid Cells'
- Author
-
Roberta Visconti, Maria Teresa Berlingieri, Massimo Santoro, Guidalberto Manfioletti, Vincenzo Giancotti, Alfredo Fusco, and Antonella Bandiera
- Subjects
Rat Thyroid ,HMGA2 Protein ,Cancer research ,Neoplastic transformation ,Cell Biology ,Biology ,Molecular Biology ,Retraction - Published
- 2018
39. Transcriptional Regulation of Glucose Metabolism: The Emerging Role of the HMGA1 Chromatin Factor
- Author
-
Antonio Brunetti, Francesco Saverio Brunetti, Daniela Foti, Eusebio Chiefari, Riccardo Sgarra, Biagio Arcidiacono, Silvia Pegoraro, Manfredi Greco, Guidalberto Manfioletti, Chiefari, E, Foti, Dp, Sgarra, R, Pegoraro, S, Arcidiacono, B, Brunetti, F, Greco, M, Manfioletti, G, and Brunetti, A.
- Subjects
0301 basic medicine ,HMGA1 ,glucose homeostasis ,glucose metabolism ,insulin resistance ,type 2 diabetes ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,FOXO1 ,Review ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,HMGA1 Gene ,03 medical and health sciences ,Insulin resistance ,Endocrinology ,medicine ,Glucose homeostasis ,Epigenetics ,lcsh:RC648-665 ,biology ,Insulin ,medicine.disease ,Cell biology ,Insulin receptor ,030104 developmental biology ,biology.protein ,glucose homeostasi - Abstract
HMGA1 (high mobility group A1) is a nonhistone architectural chromosomal protein that functions mainly as a dynamic regulator of chromatin structure and gene transcription. As such, HMGA1 is involved in a variety of fundamental cellular processes, including gene expression, epigenetic regulation, cell differentiation and proliferation, as well as DNA repair. In the last years, many reports have demonstrated a role of HMGA1 in the transcriptional regulation of several genes implicated in glucose homeostasis. Initially, it was proved that HMGA1 is essential for normal expression of the insulin receptor (INSR), a critical link in insulin action and glucose homeostasis. Later, it was demonstrated that HMGA1 is also a downstream nuclear target of the INSR signaling pathway, representing a novel mediator of insulin action and function at this level. Moreover, other observations have indicated the role of HMGA1 as a positive modulator of the Forkhead box protein O1 (FoxO1), a master regulatory factor for gluconeogenesis and glycogenolysis, as well as a positive regulator of the expression of insulin and of a series of circulating proteins that are involved in glucose counterregulation, such as the insulin growth factor binding protein 1 (IGFBP1), and the retinol binding protein 4 (RBP4). Thus, several lines of evidence underscore the importance of HMGA1 in the regulation of glucose production and disposal. Consistently, lack of HMGA1 causes insulin resistance and diabetes in humans and mice, while variations in the HMGA1 gene are associated with the risk of type 2 diabetes and metabolic syndrome, two highly prevalent diseases that share insulin resistance as a common pathogenetic mechanism. This review intends to give an overview about our current knowledge on the role of HMGA1 in glucose metabolism. Although research in this field is ongoing, many aspects still remain elusive. Future directions to improve our insights into the pathophysiology of glucose homeostasis may include epigenetic studies and the use of “omics” strategies. We believe that a more comprehensive understanding of HMGA1 and its networks may reveal interesting molecular links between glucose metabolism and other biological processes, such as cell proliferation and differentiation.
- Published
- 2018
40. The binding landscape of a partially-selective isopeptidase inhibitor with potent pro-death activity, based on the bis(arylidene)cyclohexanone scaffold
- Author
-
Carlotta Penzo, Guidalberto Manfioletti, Andrea Sgorbissa, Fabio Benedetti, Federico Berti, Federico Casarsa, Sonia Ciotti, Claudio Brancolini, Andrea Tomasella, Riccardo Sgarra, Ciotti, Sonia, Sgarra, Riccardo, Sgorbissa, Andrea, Penzo, Carlotta, Tomasella, Andrea, Casarsa, Federico, Benedetti, Fabio, Berti, Federico, Manfioletti, Guidalberto, and Brancolini, Claudio
- Subjects
Proteomics ,0301 basic medicine ,Unsaturated Keton ,Cancer Research ,Programmed cell death ,Biological Thiols ,Immunology ,Isopeptidase Inhibitor ,Biotin ,Article ,Mass Spectrometry ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Ubiquitin ,Chemical Proteomics ,Carbon-Nitrogen Lyases ,Humans ,Cysteine ,lcsh:QH573-671 ,Biological Thiol ,Cancer ,Bioconjugation ,Chemical Proteomic ,Cell Biology ,biology ,Cyclohexanones ,Chemistry ,lcsh:Cytology ,Metabolism ,Cofilin ,Small molecule ,Promiscuity ,Pull-down ,Bioconjugate ,030104 developmental biology ,Biochemistry ,030220 oncology & carcinogenesis ,Unfolded protein response ,biology.protein - Abstract
Diaryldienone derivatives with accessible β-carbons show strong anti-neoplastic properties, related to their ability to make covalent adducts with free thiols by Michael addition, and low toxicity in vivo. Accumulation of poly-ubiquitylated proteins, activation of the unfolded protein response (UPR) and induction of cell death are universal hallmarks of their activities. These compounds have been characterized as inhibitors of isopeptidases, a family of cysteine-proteases, which de-conjugate ubiquitin and ubiquitin-like proteins from their targets. However, it is unclear whether they can also react with additional proteins. In this work, we utilized the biotin-conjugated diaryldienone-derivative named 2c, as a bait to purify novel cellular targets of these small molecules. Proteomic analyses have unveiled that, in addition to isopeptidases, these inhibitors can form stable covalent adducts with different intracellular proteins, thus potentially impacting on multiple functions of the cells, from cytoskeletal organization to metabolism. These widespread activities can explain the ability of diaryldienone derivatives to efficiently trigger different cell death pathways.
- Published
- 2018
41. HMGA1 regulates the Plasminogen activation system in the secretome of breast cancer cells
- Author
-
Cinzia Franchin, Silvano Piazza, Giulia Resmini, Carlotta Penzo, Silvia Pegoraro, Giorgio Arrigoni, Serena Rizzo, Yari Ciani, Guidalberto Manfioletti, Rossella Zanin, Riccardo Sgarra, Resmini, G, Rizzo, S, Franchin, C, Zanin, R, Penzo, C, Pegoraro, S, Ciani, Y, Piazza, S, Arrigoni, G, Sgarra, R, and Manfioletti, G.
- Subjects
0301 basic medicine ,MOBILITY GROUP A1 ,INVASION ,lcsh:Medicine ,Triple Negative Breast Neoplasms ,Metastasis ,0302 clinical medicine ,iTRAQ LC-MS/MS ,TRANSCRIPTION ,HMGA ,lcsh:Science ,Plasminogen activation system ,HMGA Proteins ,Multidisciplinary ,Cell biology ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Female ,EXPRESSION ,LARGE GENE LISTS ,PROTEINS ,Biology ,Article ,03 medical and health sciences ,Paracrine signalling ,breast cancer ,Cell Line, Tumor ,medicine ,Gene silencing ,Humans ,Autocrine signalling ,MESENCHYMAL TRANSITION ,HEK 293 cells ,lcsh:R ,Plasminogen ,medicine.disease ,TRANSFORMATION ,secretome ,030104 developmental biology ,Secretory protein ,HEK293 Cells ,RHIZOBIUM-LEGUMINOSARUM ,chromatin ,Cancer cell ,Immunology ,METASTASIS ,lcsh:Q ,Plasminogen activator - Abstract
Cancer cells secrete proteins that modify the extracellular environment acting as autocrine and paracrine stimulatory factors and have a relevant role in cancer progression. The HMGA1 oncofetal protein has a prominent role in controlling the expression of an articulated set of genes involved in various aspect of cancer cell transformation. However, little is known about its role in influencing the secretome of cancer cells. Performing an iTRAQ LC–MS/MS screening for the identification of secreted proteins, in an inducible model of HMGA1 silencing in breast cancer cells, we found that HMGA1 has a profound impact on cancer cell secretome. We demonstrated that the pool of HMGA1–linked secreted proteins has pro–migratory and pro-invasive stimulatory roles. From an inspection of the HMGA1–dependent secreted factors it turned out that HMGA1 influences the presence in the extra cellular milieu of key components of the Plasminogen activation system (PLAU, SERPINE1, and PLAUR) that has a prominent role in promoting metastasis, and that HMGA1 has a direct role in regulating the transcription of two of them, i.e. PLAU and SERPINE1. The ability of HMGA1 to regulate the plasminogen activator system may constitute an important mechanism by which HMGA1 promotes cancer progression.
- Published
- 2017
42. The HMGA gene family in chordates: evolutionary perspectives from amphioxus
- Author
-
Matteo Bozzo, Paola Ramoino, Guidalberto Manfioletti, Mario Pestarino, Riccardo Sgarra, Robert Vignali, Simona Candiani, Daniela Calzia, Simone Macrì, Thurston C. Lacalli, Bozzo, M, Macrì, S, Calzia, D, Sgarra, Riccardo, Manfioletti, Guidalberto, Ramoino, P, Lacalli, T, Vignali, R, Pestarino, M, and Candiani, S.
- Subjects
0301 basic medicine ,Photoreceptors ,animal structures ,Amphioxus ,Development ,Evodevo ,HMGA ,Non-histone chromosomal proteins ,Pharyngeal skeleton ,Lamellar Body ,Evolution ,Chordates ,Cellular differentiation ,Amphioxus, Development, Evodevo, HMGA, Non-histone chromosomal proteins, Photoreceptors ,Chordate ,Evolution, Molecular ,03 medical and health sciences ,0302 clinical medicine ,Microscopy, Electron, Transmission ,biology.animal ,amphioxus, developmental biology, neurobiology ,Genetics ,Animals ,Gene family ,Neoplastic transformation ,Amino Acid Sequence ,Cloning, Molecular ,HMGA Proteins ,Phylogeny ,Lancelets ,biology ,neurobiology ,Vertebrate ,biology.organism_classification ,030104 developmental biology ,Neurula ,Evolutionary biology ,Sequence Alignment ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
High mobility group A proteins of vertebrates, HMGA1 and 2, are chromatin architectural factors involved in development, cell differentiation, and neoplastic transformation. Here, we characterize an amphioxus HMGA gene ortholog and analyze its expression. As a basal chordate, amphioxus is well placed to provide insights into the evolution of the HMGA gene family, particularly in the transition from invertebrates to vertebrates. Our phylogenetic analysis supports the basal position of amphioxus, echinoderm, and hemichordate HMGA sequences to those of vertebrate HMGA1 and HMGA2. Consistent with this, the genomic landscape around amphioxus HMGA shares features with both. Whole mount in situ hybridization shows that amphioxus HMGA mRNA is detectable from neurula stage onwards in both nervous and non-nervous tissues. This correlates with protein expression monitored immunocytochemically using antibodies against human HMGA2 protein, revealing especially high levels of expression in cells of the lamellar body, the amphioxus homolog of the pineal, suggesting that the gene may have, among its many functions, an evolutionarily conserved role in photoreceptor differentiation.
- Published
- 2017
43. Translating Proteomic Into Functional Data: An High Mobility Group A1 (HMGA1) Proteomic Signature Has Prognostic Value in Breast Cancer
- Author
-
Vincenzo Giancotti, Silvia Pegoraro, Carlotta Penzo, Angela Amato, Elisa Maurizio, Jacek R. Wiśniewski, Yari Ciani, Guidalberto Manfioletti, Silvano Piazza, Alberto Zambelli, Riccardo Sgarra, Laura Arnoldo, Maurizio, Elisa, Wiśniewski, Jr, Ciani, Yari, Amato, A, Arnoldo, Laura, Penzo, Carlotta, Pegoraro, Silvia, Giancotti, V, Zambelli, A, Piazza, S, Manfioletti, Guidalberto, and Sgarra, Riccardo
- Subjects
Proteomics ,0301 basic medicine ,Proteome ,TRIP13 ,Blotting, Western ,Kinesins ,Breast Neoplasms ,Cell Cycle Proteins ,Kaplan-Meier Estimate ,proteomic and genomic data ,Computational biology ,Biology ,Bioinformatics ,Biochemistry ,Disease-Free Survival ,Mass Spectrometry ,Analytical Chemistry ,Metastasis ,Translational Research, Biomedical ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,LRRC59 ,medicine ,Humans ,Neoplastic transformation ,HMGA1a Protein ,HMGA ,Molecular Biology ,Triple-negative breast cancer ,Regulation of gene expression ,Reverse Transcriptase Polymerase Chain Reaction ,Research ,HMGA, triple negative breast cancer, KIFC1, LRRC59, TRIP13, proteomic and genomic data ,Membrane Proteins ,Prognosis ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,triple negative breast cancer ,Multivariate Analysis ,ATPases Associated with Diverse Cellular Activities ,RNA Interference ,Carrier Proteins ,KIFC1 - Abstract
Cancer is a very heterogeneous disease, and biological variability adds a further level of complexity, thus limiting the ability to identify new genes involved in cancer development. Oncogenes whose expression levels control cell aggressiveness are very useful for developing cellular models that permit differential expression screenings in isogenic contexts. HMGA1 protein has this unique property because it is a master regulator in breast cancer cells that control the transition from a nontumorigenic epithelial-like phenotype toward a highly aggressive mesenchymal-like one. The proteins extracted from HMGA1-silenced and control MDA-MB-231 cells were analyzed using label-free shotgun mass spectrometry. The differentially expressed proteins were cross-referenced with DNA microarray data obtained using the same cellular model and the overlapping genes were filtered for factors linked to poor prognosis in breast cancer gene expression meta-data sets, resulting in an HMGA1 protein signature composed of 21 members (HRS, HMGA1 reduced signature). This signature had a prognostic value (overall survival, relapse-free survival, and distant metastasis-free survival) in breast cancer. qRT-PCR, Western blot, and immunohistochemistry analyses validated the link of three members of this signature (KIFC1, LRRC59, and TRIP13) with HMGA1 expression levels both in vitro and in vivo and wound healing assays demonstrated that these three proteins are involved in modulating tumor cell motility. Combining proteomic and genomic data with the aid of bioinformatic tools, our results highlight the potential involvement in neoplastic transformation of a restricted list of factors with an as-yet-unexplored role in cancer. These factors are druggable targets that could be exploited for the development of new, targeted therapeutic approaches in triple-negative breast cancer.
- Published
- 2016
44. The Architectural Chromatin Factor High Mobility Group A1 Enhances DNA Ligase IV Activity Influencing DNA Repair
- Author
-
Francesca Demarchi, Guidalberto Manfioletti, Ilenia Pellarin, Riccardo Sgarra, Silvia Costantini, Laura Arnoldo, Alessandro Vindigni, Gloria Ros, Carlotta Penzo, Gianluca Triolo, Silvia Pegoraro, Pellarin, Ilenia, Arnoldo, Laura, Costantini, S, Pegoraro, Silvia, Ros, Gloria, Penzo, Carlotta, Triolo, G, Demarchi, F, Sgarra, Riccardo, Vindigni, A, and Manfioletti, Guidalberto
- Subjects
0301 basic medicine ,DNA Repair ,lcsh:Medicine ,Biochemistry ,Substrate Specificity ,Ligases ,Histones ,DNA Ligase ATP ,0302 clinical medicine ,Cancer epigenetics ,HMGA1a Protein ,Post-Translational Modification ,Phosphorylation ,Ligation Assay ,lcsh:Science ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,Multidisciplinary ,biology ,DNA repair protein XRCC4 ,Recombinant Proteins ,Chromatin ,Enzymes ,Nucleic acids ,Architectural transcription factors ,030220 oncology & carcinogenesis ,MCF-7 Cells ,DNA mismatch repair ,Comet Assay ,Research Article ,DNA repair ,Research and Analysis Methods ,Non-Homologous End Joining ,03 medical and health sciences ,breast cancer ,Cell Line, Tumor ,DNA-binding proteins ,Genetics ,Humans ,Molecular Biology Techniques ,Molecular Biology ,Ku Autoantigen ,DNA double-stranded breaks ,Architectural transcription factors, DNA repair, DNA double-stranded breaks, breast cancer ,DNA ligase ,Molecular Biology Assays and Analysis Techniques ,lcsh:R ,HMGA2 Protein ,Biology and Life Sciences ,Proteins ,DNA ,Proliferating cell nuclear antigen ,030104 developmental biology ,chemistry ,Microscopy, Fluorescence ,biology.protein ,Cancer research ,Enzymology ,DNA damage ,lcsh:Q ,Nucleotide excision repair - Abstract
The HMGA1 architectural transcription factor is an oncogene overexpressed in the vast majority of human cancers. HMGA1 is a highly connected node in the nuclear molecular network and the key aspect of HMGA1 involvement in cancer development is that HMGA1 simultaneously confers cells multiple oncogenic hits, ranging from global chromatin structural and gene expression modifications up to the direct functional alterations of key cellular proteins. Interestingly, HMGA1 also modulates DNA damage repair pathways. In this work, we provide evidences linking HMGA1 with Non-Homologous End Joining DNA repair. We show that HMGA1 is in complex with and is a substrate for DNA-PK. HMGA1 enhances Ligase IV activity and it counteracts the repressive histone H1 activity towards DNA ends ligation. Moreover, breast cancer cells overexpressing HMGA1 show a faster recovery upon induction of DNA double-strand breaks, which is associated with a higher survival. These data suggest that resistance to DNA-damaging agents in cancer cells could be partially attributed to HMGA1 overexpression thus highlighting the relevance of considering HMGA1 expression levels in the selection of valuable and effective pharmacological regimens.
- Published
- 2016
45. HMGA Interactome: New Insights from Phage Display Technology
- Author
-
Elisa Maurizio, Erika Malini, Riccardo Sgarra, Guidalberto Manfioletti, Sara Bembich, Paolo Edomi, Malini, E., Maurizio, Elisa, Bembich, S., Sgarra, Riccardo, Edomi, Paolo, and Manfioletti, Guidalberto
- Subjects
CHROMATIN ,GENE EXPRESSION ,DNA, Complementary ,Phage display ,Molecular Sequence Data ,Biology ,Biochemistry ,Interactome ,Mice ,Open Reading Frames ,Peptide Library ,Transcription (biology) ,TAF3 ,P150/CAF-1 ,Protein Interaction Mapping ,Animals ,Humans ,Amino Acid Sequence ,Chromatin Assembly Factor-1 ,Sequence Deletion ,Regulation of gene expression ,Genetics ,TATA-Binding Protein Associated Factors ,High Mobility Group Proteins ,HMGA ,Recombinant Proteins ,Chromatin ,Cell biology ,HEK293 Cells ,High-mobility group ,Mutation - Abstract
High mobility group A proteins (HMGA1 and HMGA2) are architectural factors involved in chromatin remodelling and regulation of gene expression. HMGA are highly expressed during embryogenesis and in cancer cells and are involved in development and cell differentiation as well as cancer formation and progression. These factors, by binding to DNA and interacting with other nuclear proteins, can organize macromolecular complexes involved in transcription, chromatin dynamics, RNA processing, and DNA repair. The identification of protein partners for HMGA has greatly contributed to our understanding of their multiple functions. He we report the identification of HMGA molecular partners using a gene fragment library in a phage display screening. Using an ORF-enriched cDNA library, we have isolated several HMGA1 interacting clones and for two of them, TBP associated factor 3 (TAF3) and chromatin assembly factor 1 p150/CAF-1, have demonstrated an in vivo association with HMGA1. The identification of these new partners suggests that HMGA can also influence general aspects of transcription and once more underlines their involvement in chromatin remodelling and dynamics.
- Published
- 2011
- Full Text
- View/download PDF
46. A simple discontinuous buffer system for increased resolution and speed in gel electrophoretic analysis of DNA sequence.
- Author
-
Piero Carninci, Stefano Gustincich, S. Bottega, C. Patrosso, G. Del Sal, Guidalberto Manfioletti, and Claudio Schneider
- Published
- 1990
- Full Text
- View/download PDF
47. Malignant Ectomesenchymoma: Genetic Profile Reflects Rhabdomyosarcomatous Differentiation
- Author
-
Raphael Sciot, Agnieszka Wozniak, Ivo De Wever, Guidalberto Manfioletti, Maria Debiec-Rychter, Elisabetta Magrini, Giovanni Tallini, and Guiseppe Floris
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Marker chromosome ,Biology ,Pathology and Forensic Medicine ,Malignant transformation ,Diagnosis, Differential ,medicine ,Humans ,Mesenchymoma ,Paired Box Transcription Factors ,Rhabdomyosarcoma, Embryonal ,HMGA1a Protein ,Ganglioneuroma ,PAX3 Transcription Factor ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Chromosome Aberrations ,medicine.diagnostic_test ,Gene Expression Profiling ,HMGA2 Protein ,Infant ,PAX7 Transcription Factor ,Karyotype ,Cell Biology ,medicine.disease ,Malignant ectomesenchymoma ,Genital Neoplasms, Male ,Scrotum ,Embryonal rhabdomyosarcoma ,Fluorescence in situ hybridization ,Comparative genomic hybridization - Abstract
Malignant ectomesenchymoma (MEM) represents a heterogeneous group of tumors, most likely originating from pluripotent primitive neural crest cells. In this report, we present an 8-month-old infant boy with an MEM on the left scrotum. Retrospective review of the incision biopsy showed the presence of a few ganglion cells in an otherwise classic embryonal rhabdomyosarcoma (RMS), whereas in the resection specimen after chemotherapy the combined RMS and ganglioneuroma components were very obvious. Cytogenetic analysis of the residual lesion showed an abnormal karyotype, 49, XY, +2, -6, +11, +20, +mar, with a hyperploidy in a subset of cells. By fluorescence in situ hybridization analysis, the marker chromosome was identified as originating from chromosome 6, and the tumor cells were negative for PAX3/PAX7 disrupting translocations specific for alveolar RMS. Gains of chromosomes 2, 11, and 20, found in the current case, are a common finding in embryonal RMS. These gains probably reflect the myogenic differentiation of MEM and support the genetic link between these 2 neoplasms. In addition to the conventional cytogenetics, array comparative genomic hybridization analysis was performed on the primary and residual tumors. The genomic profiles of both specimens were basically the same including the presence of 2 distinctive chromosome 6p21.32-p21.2 and 6p11.2 amplification regions in the primary tumor, which vanished in the postchemotherapy specimen. The pretreatment biopsy exhibited strong expression of HMGA1 and HMGA2 proteins in immunohistochemistry, with the shift toward the loss of expression of both genes in the posttreatment tumoral tissue. This finding supports the oncogenic properties of the HMGA family of proteins and their role in the process of malignant transformation.
- Published
- 2007
- Full Text
- View/download PDF
48. The second AT-hook of the architectural transcription factor HMGA2 is determinant for nuclear localization and function
- Author
-
Giacomo Cattaruzzi, Alessandra Rustighi, Sandro Altamura, Guidalberto Manfioletti, Michela A. Tessari, Carlo Pucillo, Vincenzo Giancotti, Cattaruzzi, G, Altamura, S, Tessari, Ma, Rustighi, Alessandra, Giancotti, V, Pucillo, C, and Manfioletti, Guidalberto
- Subjects
alpha Karyopherins ,HMGA2 ,Molecular Sequence Data ,Active Transport, Cell Nucleus ,Biology ,Cell Line ,Mice ,Cricetinae ,Genetics ,HMGA2, AT-hook ,Animals ,Humans ,Neoplastic transformation ,Amino Acid Sequence ,nuclear localization ,Nuclear protein ,HMGA Proteins ,AT-hook ,Molecular Biology ,Sequence Deletion ,Cell Nucleus ,Amino Acids, Basic ,HMGA2 Protein ,HMGA ,Alpha Karyopherins ,AT-Hook Motifs ,Cell biology ,gene expression ,chromatin ,Nuclear transport ,Nuclear localization sequence ,Transcription Factors - Abstract
High Mobility Group A (HMGA) is a family of architectural nuclear factors which play an important role in neoplastic transformation. HMGA proteins are multifunctional factors that associate both with DNA and nuclear proteins that have been involved in several nuclear processes including transcription. HMGA localization is exclusively nuclear but, to date, the mechanism of nuclear import for these proteins remains unknown. Here, we report the identification and characterization of a nuclear localization signal (NLS) for HMGA2, a member of the HMGA family. The NLS overlaps with the second of the three AT-hooks, the DNA-binding domains characteristic for this group of proteins. The functionality of this NLS was demonstrated by its ability to target a heterologous beta-galactosidase/green fluorescent protein fusion protein to the nucleus. Mutations to alanine of basic residues within the second AT-hook resulted in inhibition of HMGA2 nuclear localization and impairment of its function in activating the cyclin A promoter. In addition, HMGA2 was shown to directly interact with the nuclear import receptor importin-alpha2 via the second AT-hook. HMGA proteins are overexpressed and rearranged in a variety of tumors; our findings can thus help elucidating their role in neoplastic transformation.
- Published
- 2007
- Full Text
- View/download PDF
49. A novel mechanism of post-translational modulation of HMGA functions by the histone chaperone nucleophosmin
- Author
-
Antonio Brunetti, Laura Arnoldo, Stefania Iiritano, Riccardo Sgarra, Biagio Arcidiacono, Eusebio Chiefari, Ilenia Pellarin, Guidalberto Manfioletti, Silvia Pegoraro, Arnoldo, Laura, Sgarra, Riccardo, Chiefari, Eusebio, Iiritano, Stefania, Arcidiacono, Biagio, Pegoraro, Silvia, Pellarin, Ilenia, Brunetti, Antonio, and Manfioletti, Guidalberto
- Subjects
Transcription, Genetic ,Molecular Sequence Data ,Regulatory Sequences, Nucleic Acid ,Article ,DNA-binding ,Chaperones ,Humans ,Histone Chaperones ,Amino Acid Sequence ,HMGA ,RNA, Small Interfering ,HMGA Proteins ,Promoter Regions, Genetic ,Genetics ,Nucleophosmin ,Multidisciplinary ,Binding Sites ,biology ,Nuclear Proteins ,Promoter ,DNA ,Hep G2 Cells ,Chromatin ,Receptor, Insulin ,Cell biology ,Protein Structure, Tertiary ,Insulin-Like Growth Factor Binding Protein 1 ,Histone ,Regulatory sequence ,Chaperone (protein) ,biology.protein ,Multidisciplinary, HMGA, Chaperones, Chromatin, DNA-binding ,RNA Interference ,Protein Processing, Post-Translational ,Protein Binding - Abstract
High Mobility Group A are non-histone nuclear proteins that regulate chromatin plasticity and accessibility, playing an important role both in physiology and pathology. Their activity is controlled by transcriptional, post-transcriptional and post-translational mechanisms. In this study we provide evidence for a novel modulatory mechanism for HMGA functions. We show that HMGAs are complexed in vivo with the histone chaperone nucleophosmin (NPM1), that this interaction requires the histone-binding domain of NPM1 and that NPM1 modulates both DNA-binding affinity and specificity of HMGAs. By focusing on two human genes whose expression is directly regulated by HMGA1, the Insulin receptor (INSR) and the Insulin-like growth factor-binding protein 1 (IGFBP1) genes, we demonstrated that occupancy of their promoters by HMGA1 was NPM1-dependent, reflecting a mechanism in which the activity of these cis-regulatory elements is directly modulated by NPM1 leading to changes in gene expression. HMGAs need short stretches of AT-rich nucleosome-free regions to bind to DNA. Therefore, many putative HMGA binding sites are present within the genome. Our findings indicate that NPM1, by exerting a chaperoning activity towards HMGAs, may act as a master regulator in the control of DNA occupancy by these proteins and hence in HMGA-mediated gene expression.
- Published
- 2015
50. A novel HMGA1-CCNE2-YAP axis regulates breast cancer aggressiveness
- Author
-
Yari Ciani, Gloria Ros, Silvano Piazza, Silvia Pegoraro, Riccardo Sgarra, Guidalberto Manfioletti, Pegoraro, Silvia, Ros, Gloria, Ciani, Yari, Sgarra, Riccardo, Piazza, Silvano, and Manfioletti, Guidalberto
- Subjects
Hippo pathway ,Breast Neoplasms ,Cell Cycle Proteins ,Transfection ,CDK inhibitor ,Cell Movement ,Cyclin-dependent kinase ,Cell Line, Tumor ,Cyclins ,CDK inhibitors ,Cyclin E2 ,Oncogene/tumour suppressor ,YAP/TAZ ,Oncology ,medicine ,Humans ,Neoplasm Invasiveness ,Neoplastic transformation ,HMGA1a Protein ,Phosphorylation ,Transcription factor ,Tissue homeostasis ,Hippo signaling pathway ,biology ,Kinase ,Nuclear Proteins ,Cancer ,medicine.disease ,Cell biology ,HEK293 Cells ,biology.protein ,Cancer research ,Female ,Acyltransferases ,Signal Transduction ,Transcription Factors ,Research Paper - Abstract
High Mobility Group A1 (HMGA1) is an architectural chromatin factor that promotes neoplastic transformation and progression. However, the mechanism by which HMGA1 exerts its oncogenic function is not fully understood. Here, we show that cyclin E2 (CCNE2) acts downstream of HMGA1 to regulate the motility and invasiveness of basal-like breast cancer cells by promoting the nuclear localization and activity of YAP, the downstream mediator of the Hippo pathway. Mechanistically, the activity of MST1/2 and LATS1/2, the core kinases of the Hippo pathway, are required for the HMGA1- and CCNE2-mediated regulation of YAP localization. In breast cancer patients, high levels of HMGA1 and CCNE2 expression are associated with the YAP/TAZ signature, supporting this connection. Moreover, we provide evidence that CDK inhibitors induce the translocation of YAP from the nucleus to the cytoplasm, resulting in a decrease in its activity. These findings reveal an association between HMGA1 and the Hippo pathway that is relevant to stem cell biology, tissue homeostasis, and cancer.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.